Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
1.
Neurobiol Dis ; 180: 106071, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36898613

RESUMO

The accurate regulation of the microenvironment within the nervous system is one of the key features characterizing complex organisms. To this end, neural tissue has to be physically separated from circulation, but at the same time, mechanisms must be in place to allow controlled transport of nutrients and macromolecules into and out of the brain. These roles are executed by cells of the blood-brain barrier (BBB) found at the interface of circulation and neural tissue. BBB dysfunction is observed in several neurological diseases in human. Although this can be considered as a consequence of diseases, strong evidence supports the notion that BBB dysfunction can promote the progression of brain disorders. In this review, we compile the recent evidence describing the contribution of the Drosophila BBB to the further understanding of brain disease features in human patients. We discuss the function of the Drosophila BBB during infection and inflammation, drug clearance and addictions, sleep, chronic neurodegenerative disorders and epilepsy. In summary, this evidence suggests that the fruit fly, Drosophila melanogaster, can be successfully employed as a model to disentangle mechanisms underlying human diseases.


Assuntos
Barreira Hematoencefálica , Encefalopatias , Animais , Humanos , Barreira Hematoencefálica/fisiologia , Drosophila melanogaster , Drosophila , Encéfalo
2.
Development ; 147(2)2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31862845

RESUMO

The development of tissues and organs requires close interaction of cells. To achieve this, cells express adhesion proteins such as the neural cell adhesion molecule (NCAM) or its Drosophila ortholog Fasciclin 2 (Fas2). Both are members of the Ig-domain superfamily of proteins that mediate homophilic adhesion. These proteins are expressed as isoforms differing in their membrane anchorage and their cytoplasmic domains. To study the function of single isoforms, we have conducted a comprehensive genetic analysis of Fas2 We reveal the expression pattern of all major Fas2 isoforms, two of which are GPI anchored. The remaining five isoforms carry transmembrane domains with variable cytoplasmic tails. We generated Fas2 mutants expressing only single isoforms. In contrast to the null mutation, which causes embryonic lethality, these mutants are viable, indicating redundancy among the different isoforms. Cell type-specific rescue experiments showed that glial-secreted Fas2 can rescue the Fas2 mutant phenotype to viability. This demonstrates that cytoplasmic Fas2 domains have no apparent essential functions and indicate that Fas2 has function(s) other than homophilic adhesion. In conclusion, our data suggest novel mechanistic aspects of a long-studied adhesion protein.


Assuntos
Moléculas de Adesão Celular Neuronais/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Transdução de Sinais , Animais , Adesão Celular , Moléculas de Adesão Celular Neuronais/química , Moléculas de Adesão Celular Neuronais/genética , Movimento Celular , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Glicosilfosfatidilinositóis/metabolismo , Mutação/genética , Neuroglia/metabolismo , Domínios Proteicos , Isoformas de Proteínas/metabolismo , Traqueia/embriologia , Traqueia/metabolismo
3.
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
4.
Development ; 145(18)2018 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-30242104

RESUMO

Metazoan Hedgehog (Hh) morphogens are essential regulators of growth and patterning at significant distances from their source, despite being produced as N-terminally palmitoylated and C-terminally cholesteroylated proteins, which firmly tethers them to the outer plasma membrane leaflet of producing cells and limits their spread. One mechanism to overcome this limitation is proteolytic processing of both lipidated terminal peptides, called shedding, but molecular target site requirements for effective Hh shedding remained undefined. In this work, by using Drosophila melanogaster as a model, we show that mutagenesis of the N-terminal Cardin-Weintraub (CW) motif inactivates recombinant Hh proteins to variable degrees and, if overexpressed in the same compartment, converts them into suppressors of endogenous Hh function. In vivo, additional removal of N-palmitate membrane anchors largely restored endogenous Hh function, supporting the hypothesis that proteolytic CW processing controls Hh solubilization. Importantly, we also observed that CW repositioning impairs anterior/posterior compartmental boundary maintenance in the third instar wing disc. This demonstrates that Hh shedding not only controls the differentiation of anterior cells, but also maintains the sharp physical segregation between these receiving cells and posterior Hh-producing cells.


Assuntos
Motivos de Aminoácidos/genética , Padronização Corporal/genética , Olho Composto de Artrópodes/embriologia , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Proteínas Hedgehog/genética , Asas de Animais/embriologia , Animais , Diferenciação Celular , Lipoilação/fisiologia , Palmitatos/metabolismo , Transdução de Sinais/genética
5.
Development ; 144(24): 4673-4683, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29084807

RESUMO

The development of the nervous system requires tight control of cell division, fate specification and migration. The anaphase-promoting complex/cyclosome (APC/C) is an E3 ubiquitin ligase that affects different steps of cell cycle progression, as well as having postmitotic functions in nervous system development. It can therefore link different developmental stages in one tissue. The two adaptor proteins, Fizzy/Cdc20 and Fizzy-related/Cdh1, confer APC/C substrate specificity. Here, we show that two distinct modes of APC/C function act during Drosophila eye development. Fizzy/Cdc20 controls the early growth of the eye disc anlage and the concomitant entry of glial cells onto the disc. In contrast, fzr/cdh1 acts during neuronal patterning and photoreceptor axon growth, and subsequently affects neuron-glia interaction. To further address the postmitotic role of Fzr/Cdh1 in controlling neuron-glia interaction, we identified a series of novel APC/C candidate substrates. Four of our candidate genes are required for fzr/cdh1-dependent neuron-glia interaction, including the dynein light chain Dlc90F Taken together, our data show how different modes of APC/C activation can couple early growth and neuron-glia interaction during eye disc development.


Assuntos
Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas Cdc20/metabolismo , Proteínas Cdh1/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Olho/embriologia , Neuroglia/metabolismo , Neurônios/metabolismo , Animais , Comunicação Celular/fisiologia , Ciclo Celular/fisiologia , Dineínas do Citoplasma/metabolismo , Dineínas , Células Fotorreceptoras de Invertebrados/citologia
6.
J Am Soc Nephrol ; 30(6): 1006-1019, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31097607

RESUMO

BACKGROUND: Patients with certain mutations in the gene encoding the slit diaphragm protein Nephrin fail to develop functional slit diaphragms and display severe proteinuria. Many adult-onset glomerulopathies also feature alterations in Nephrin expression and function. Nephrin signals from the podocyte slit diaphragm to the Actin cytoskeleton by recruiting proteins that can interact with C3G, a guanine nucleotide exchange factor of the small GTPase Rap1. Because Rap activity affects formation of focal adhesions, we hypothesized that Nephrin transmits signals to the Integrin receptor complex, which mediates podocyte adhesion to the extracellular matrix. METHODS: To investigate Nephrin's role in transmitting signals to the Integrin receptor complex, we conducted genetic studies in Drosophila nephrocytes and validated findings from Drosophila in a cultured human podocyte model. RESULTS: Drosophila nephrocytes form a slit diaphragm-like filtration barrier and express the Nephrin ortholog Sticks and stones (Sns). A genetic screen identified c3g as necessary for nephrocyte function. In vivo, nephrocyte-specific gene silencing of sns or c3g compromised nephrocyte filtration and caused nephrocyte diaphragm defects. Nephrocytes with impaired Sns or C3G expression displayed an altered localization of Integrin and the Integrin-associated protein Talin. Furthermore, gene silencing of c3g partly rescued nephrocyte diaphragm defects of an sns overexpression phenotype, pointing to genetic interaction of sns and c3g in nephrocytes. We also found that activated Nephrin recruited phosphorylated C3G and resulted in activation of Integrin ß1 in cultured podocytes. CONCLUSIONS: Our findings suggest that Nephrin can mediate a signaling pathway that results in activation of Integrin ß1 at focal adhesions, which may affect podocyte attachment to the extracellular matrix.


Assuntos
Regulação da Expressão Gênica/genética , Integrina beta1/metabolismo , Proteínas de Membrana/genética , Fosforilação/genética , Podócitos/metabolismo , Insuficiência Renal Crônica/genética , Animais , Células Cultivadas , Drosophila/citologia , Citometria de Fluxo , Humanos , Microscopia Eletrônica de Transmissão , Insuficiência Renal Crônica/patologia , Transdução de Sinais/genética , Estatísticas não Paramétricas
7.
Glia ; 67(1): 5-26, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30443934

RESUMO

Glial cells constitute without any dispute an essential element in providing an efficiently operating nervous system. Work in many labs over the last decades has demonstrated that neuronal function, from action potential generation to its propagation, from eliciting synaptic responses to the subsequent postsynaptic integration, is evolutionarily highly conserved. Likewise, the biology of glial cells appears conserved in its core elements and therefore, a deeper understanding of glial cells is expected to benefit from analyzing model organisms such as Drosophila melanogaster. Drosophila is particularly well suited for studying glial biology since in the fly nervous system only a limited number of glial cells exists, which can be individually identified based on position and a set of molecular markers. In combination with the well-known genetic tool box an unprecedented level of analysis is feasible, that not only can help to identify novel molecules and principles governing glial cell function but also will help to better understand glial functions first identified in the mammalian nervous system. Here we review the current knowledge on Drosophila glia to spark interest in using this system to analyze complex glial traits in the future.


Assuntos
Proteínas de Drosophila/metabolismo , Modelos Animais , Neuroglia/metabolismo , Animais , Diferenciação Celular/fisiologia , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster , Neuroglia/patologia , Neurônios/metabolismo
8.
Development ; 142(12): 2184-93, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26015542

RESUMO

In the nervous system, glial cells need to be specified from a set of progenitor cells. In the developing Drosophila eye, perineurial glia proliferate and differentiate as wrapping glia in response to a neuronal signal conveyed by the FGF receptor pathway. To unravel the underlying transcriptional network we silenced all genes encoding predicted DNA-binding proteins in glial cells using RNAi. Dref and other factors of the TATA box-binding protein-related factor 2 (TRF2) complex were previously predicted to be involved in cellular metabolism and cell growth. Silencing of these genes impaired early glia proliferation and subsequent differentiation. Dref controls proliferation via activation of the Pdm3 transcription factor, whereas glial differentiation is regulated via Dref and the homeodomain protein Cut. Cut expression is controlled independently of Dref by FGF receptor activity. Loss- and gain-of-function studies show that Cut is required for glial differentiation and is sufficient to instruct the formation of membrane protrusions, a hallmark of wrapping glial morphology. Our work discloses a network of transcriptional regulators controlling the progression of a naïve perineurial glia towards the fully differentiated wrapping glia.


Assuntos
Drosophila melanogaster/embriologia , Olho/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Neurais/citologia , Neurogênese/genética , Neuroglia/citologia , Animais , Proliferação de Células/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Ativação Enzimática , Olho/inervação , Redes Reguladoras de Genes , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transdução Genética
9.
Development ; 142(7): 1336-45, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25758464

RESUMO

Efficient neuronal conductance requires that axons are insulated by glial cells. For this, glial membranes need to wrap around axons. Invertebrates show a relatively simple extension of glial membranes around the axons, resembling Remak fibers formed by Schwann cells in the mammalian peripheral nervous system. To unravel the molecular pathways underlying differentiation of glial cells that provide axonal wrapping, we are using the genetically amenable Drosophila model. At the end of larval life, the wrapping glia differentiates into very large cells, spanning more than 1 mm of axonal length. The extension around axonal membranes is not influenced by the caliber of the axon or its modality. Using cell type-specific gene knockdown we show that the extension of glial membranes around the axons is regulated by an autocrine activation of the EGF receptor through the neuregulin homolog Vein. This resembles the molecular mechanism employed during cell-autonomous reactivation of glial differentiation after injury in mammals. We further demonstrate that Vein, produced by the wrapping glia, also regulates the formation of septate junctions in the abutting subperineurial glia. Moreover, the wrapping glia indirectly controls the proliferation of the perineurial glia. Thus, the wrapping glia appears center stage to orchestrate the development of the different glial cell layers in a peripheral nerve.


Assuntos
Axônios/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Neurregulinas/metabolismo , Neuroglia/metabolismo , Sistema Nervoso Periférico/metabolismo , Homologia de Sequência de Aminoácidos , Animais , Axônios/ultraestrutura , Barreira Hematoencefálica/metabolismo , Diferenciação Celular , Drosophila melanogaster/citologia , Drosophila melanogaster/ultraestrutura , Receptores ErbB/metabolismo , Larva/citologia , Larva/metabolismo , Larva/ultraestrutura , Neuroglia/citologia , Neuroglia/ultraestrutura , Nervos Periféricos/citologia , Nervos Periféricos/metabolismo , Nervos Periféricos/ultraestrutura , Sistema Nervoso Periférico/citologia , Sistema Nervoso Periférico/ultraestrutura , Transdução de Sinais
10.
PLoS Comput Biol ; 13(5): e1005530, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28493862

RESUMO

Imaging and analyzing the locomotion behavior of small animals such as Drosophila larvae or C. elegans worms has become an integral subject of biological research. In the past we have introduced FIM, a novel imaging system feasible to extract high contrast images. This system in combination with the associated tracking software FIMTrack is already used by many groups all over the world. However, so far there has not been an in-depth discussion of the technical aspects. Here we elaborate on the implementation details of FIMTrack and give an in-depth explanation of the used algorithms. Among others, the software offers several tracking strategies to cover a wide range of different model organisms, locomotion types, and camera properties. Furthermore, the software facilitates stimuli-based analysis in combination with built-in manual tracking and correction functionalities. All features are integrated in an easy-to-use graphical user interface. To demonstrate the potential of FIMTrack we provide an evaluation of its accuracy using manually labeled data. The source code is available under the GNU GPLv3 at https://github.com/i-git/FIMTrack and pre-compiled binaries for Windows and Mac are available at http://fim.uni-muenster.de.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Locomoção/fisiologia , Software , Algoritmos , Animais , Caenorhabditis elegans/fisiologia , Biologia Computacional
11.
Development ; 141(6): 1366-80, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24553290

RESUMO

A tight spatiotemporal control of actin polymerization is important for many cellular processes that shape cells into a multicellular organism. The formation of unbranched F-actin is induced by several members of the formin family. Drosophila encodes six formin genes, representing six of the seven known mammalian subclasses. Knittrig, the Drosophila homolog of mammalian FHOD1, is specifically expressed in the developing central nervous system midline glia, the trachea, the wing and in macrophages. knittrig mutants exhibit mild tracheal defects but survive until late pupal stages and mainly die as pharate adult flies. knittrig mutant macrophages are smaller and show reduced cell spreading and cell migration in in vivo wounding experiments. Rescue experiments further demonstrate a cell-autonomous function of Knittrig in regulating actin dynamics and cell migration. Knittrig localizes at the rear of migrating macrophages in vivo, suggesting a cellular requirement of Knittrig in the retraction of the trailing edge. Supporting this notion, we found that Knittrig is a target of the Rho-dependent kinase Rok. Co-expression with Rok or expression of an activated form of Knittrig induces actin stress fibers in macrophages and in epithelial tissues. Thus, we propose a model in which Rok-induced phosphorylation of residues within the basic region mediates the activation of Knittrig in controlling macrophage migration.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiologia , Quinases Associadas a rho/metabolismo , Animais , Movimento Celular/imunologia , Movimento Celular/fisiologia , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes de Insetos , Imunidade Celular , Macrófagos/imunologia , Macrófagos/fisiologia , Mutação , Fibras de Estresse/metabolismo , Quinases Associadas a rho/genética
12.
Development ; 141(16): 3233-42, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25063458

RESUMO

Cell migration is an important feature of glial cells. Here, we used the Drosophila eye disc to decipher the molecular network controlling glial migration. We stimulated glial motility by pan-glial PDGF receptor (PVR) activation and identified several genes acting downstream of PVR. Drosophila lox is a non-essential gene encoding a secreted protein that stiffens the extracellular matrix (ECM). Glial-specific knockdown of Integrin results in ECM softening. Moreover, we show that lox expression is regulated by Integrin signaling and vice versa, suggesting that a positive-feedback loop ensures a rigid ECM in the vicinity of migrating cells. The general implication of this model was tested in a mammalian glioma model, where a Lox-specific inhibitor unraveled a clear impact of ECM rigidity in glioma cell migration.


Assuntos
Olho Composto de Artrópodes/embriologia , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/fisiologia , Matriz Extracelular/fisiologia , Neuroglia/citologia , Proteína-Lisina 6-Oxidase/fisiologia , Animais , Animais Geneticamente Modificados , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Matriz Extracelular/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Glioblastoma/metabolismo , Humanos , Integrinas/metabolismo , Camundongos , Camundongos Nus , Dados de Sequência Molecular , Transplante de Neoplasias , Proteína-Lisina 6-Oxidase/genética , Transdução de Sinais
13.
J Exp Biol ; 220(Pt 13): 2452-2475, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28679796

RESUMO

Mapping brain function to brain structure is a fundamental task for neuroscience. For such an endeavour, the Drosophila larva is simple enough to be tractable, yet complex enough to be interesting. It features about 10,000 neurons and is capable of various taxes, kineses and Pavlovian conditioning. All its neurons are currently being mapped into a light-microscopical atlas, and Gal4 strains are being generated to experimentally access neurons one at a time. In addition, an electron microscopic reconstruction of its nervous system seems within reach. Notably, this electron microscope-based connectome is being drafted for a stage 1 larva - because stage 1 larvae are much smaller than stage 3 larvae. However, most behaviour analyses have been performed for stage 3 larvae because their larger size makes them easier to handle and observe. It is therefore warranted to either redo the electron microscopic reconstruction for a stage 3 larva or to survey the behavioural faculties of stage 1 larvae. We provide the latter. In a community-based approach we called the Ol1mpiad, we probed stage 1 Drosophila larvae for free locomotion, feeding, responsiveness to substrate vibration, gentle and nociceptive touch, burrowing, olfactory preference and thermotaxis, light avoidance, gustatory choice of various tastants plus odour-taste associative learning, as well as light/dark-electric shock associative learning. Quantitatively, stage 1 larvae show lower scores in most tasks, arguably because of their smaller size and lower speed. Qualitatively, however, stage 1 larvae perform strikingly similar to stage 3 larvae in almost all cases. These results bolster confidence in mapping brain structure and behaviour across developmental stages.


Assuntos
Comportamento Animal , Drosophila melanogaster/fisiologia , Animais , Encéfalo/citologia , Encéfalo/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Larva/crescimento & desenvolvimento , Larva/fisiologia
14.
Development ; 139(10): 1765-76, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22461565

RESUMO

The differentiation of the blood-brain barrier (BBB) is an essential process in the development of a complex nervous system and depends on alternative splicing. In the fly BBB, glial cells establish intensive septate junctions that require the cell-adhesion molecule Neurexin IV. Alternative splicing generates two different Neurexin IV isoforms: Neurexin IV(exon3), which is found in cells that form septate junctions, and Neurexin IV(exon4), which is found in neurons that form no septate junctions. Here, we show that the formation of the BBB depends on the RNA-binding protein HOW (Held out wings), which triggers glial specific splicing of Neurexin IV(exon3). Using a set of splice reporters, we show that one HOW-binding site is needed to include one of the two mutually exclusive exons 3 and 4, whereas binding at the three further motifs is needed to exclude exon 4. The differential splicing is controlled by nuclear access of HOW and can be induced in neurons following expression of nuclear HOW. Using a novel in vivo two-color splicing detector, we then screened for genes required for full HOW activity. This approach identified Cyclin-dependent kinase 12 (Cdk12) and the splicesosomal component Prp40 as major determinants in regulating HOW-dependent splicing of Neurexin IV. Thus, in addition to the control of nuclear localization of HOW, the phosphorylation of the C-terminal domain of the RNA polymerase II by Cdk12 provides an elegant mechanism in regulating timed splicing of newly synthesized mRNA molecules.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Neuroglia/metabolismo , Proteínas Nucleares/metabolismo , Splicing de RNA/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Quinases Ciclina-Dependentes/genética , Drosophila , Imuno-Histoquímica , Proteínas Nucleares/genética , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Neurooncol ; 124(3): 373-83, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26224160

RESUMO

Platelet-derived growth factor receptor (PDGFR) signaling plays an important role in the biology of malignant gliomas. To investigate mechanisms modulating PDGFR signaling in gliomagenesis, we employed a Drosophila glioma model and genetic screen to identify genes interacting with Pvr, the fly homolog of PDGFRs. Glial expression of constitutively activated Pvr (λPvr) led to glial over migration and lethality at late larval stage. Among 3316 dsRNA strains crossed against the tester strain, 128 genes shifted lethality to pupal stage, including tetraspanin 2A (tsp2A). In a second step knockdown of all Drosophila tetraspanins was investigated. Of all tetraspanin dsRNA strains only knockdown of tsp2A partially rescued the Pvr-induced phenotype. Human CD9 (TSPAN29/MRP-1), a close homolog of tsp2A, was found to be expressed in glioma cell lines A172 and U343MG as well as in the majority of glioblastoma samples (16/22, 73 %). Furthermore, in situ proximity ligation assay revealed close association of CD9 with PDGFR α and ß. In U343MG cells, knockdown of CD9 blocked PDGF-BB stimulated migration. In conclusion, modulation of PDGFR signaling by CD9 is evolutionarily conserved from Drosophila glia to human glioma and plays a role in glia migration.


Assuntos
Movimento Celular/fisiologia , Glioma/patologia , Neuroglia/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Tetraspanina 29/metabolismo , Análise de Variância , Animais , Animais Geneticamente Modificados , Evolução Biológica , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Relação Dose-Resposta a Droga , Drosophila , Proteínas de Drosophila/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neuroglia/patologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/farmacologia , Transdução de Sinais
16.
Nature ; 460(7256): 758-61, 2009 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-19597479

RESUMO

The formation of a complex nervous system requires the intricate interaction of neurons and glial cells. Glial cells generally migrate over long distances before they initiate their differentiation, which leads to wrapping and insulation of axonal processes. The molecular pathways coordinating the switch from glial migration to glial differentiation are largely unknown. Here we demonstrate that, within the Drosophila eye imaginal disc, fibroblast growth factor (FGF) signalling coordinates glial proliferation, migration and subsequent axonal wrapping. Glial differentiation in the Drosophila eye disc requires a succession from glia-glia interaction to glia-neuron interaction. The neuronal component of the fly eye develops in the peripheral nervous system within the eye-antennal imaginal disc, whereas glial cells originate from a pool of central-nervous-system-derived progenitors and migrate onto the eye imaginal disc. Initially, glial-derived Pyramus, an FGF8-like ligand, modulates glial cell number and motility. A switch to neuronally expressed Thisbe, a second FGF8-like ligand, then induces glial differentiation. This switch is accompanied by an alteration in the intracellular signalling pathway through which the FGF receptor channels information into the cell. Our findings reveal how a switch from glia-glia interactions to glia-neuron interactions can trigger formation of glial membrane around axonal trajectories. These results disclose an evolutionarily conserved control mechanism of axonal wrapping, indicating that Drosophila might serve as a model to understand glial disorders in humans.


Assuntos
Diferenciação Celular , Drosophila melanogaster/metabolismo , Olho/citologia , Fatores de Crescimento de Fibroblastos/metabolismo , Neuroglia/citologia , Neuroglia/metabolismo , Transdução de Sinais , Animais , Axônios/metabolismo , Movimento Celular , Proliferação de Células , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Olho/crescimento & desenvolvimento , Olho/inervação , Olho/metabolismo , Cobaias , Ligantes
17.
Nat Rev Neurosci ; 10(11): 769-79, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19773781

RESUMO

Neurons and glial cells show mutual interdependence in many developmental and functional aspects of their biology. To establish their intricate relationships with neurons, glial cells must migrate over what are often long distances. In the CNS glial cells generally migrate as single cells, whereas PNS glial cells tend to migrate as cohorts of cells. How are their journeys initiated and directed, and what stops the migratory phase once glial cells are aligned with their neuronal counterparts? A deeper understanding of glial migration and the underlying neuron-glia interactions may contribute to the development of therapeutics for demyelinating diseases or glial tumours.


Assuntos
Movimento Celular/fisiologia , Invertebrados , Neuroglia/citologia , Neuroglia/fisiologia , Vertebrados , Animais , Evolução Molecular , Humanos , Invertebrados/anatomia & histologia , Invertebrados/fisiologia , Neuroglia/classificação , Transdução de Sinais/fisiologia , Especificidade da Espécie , Vertebrados/anatomia & histologia , Vertebrados/fisiologia
18.
Front Cell Neurosci ; 18: 1397627, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38846639

RESUMO

The blood-brain barrier (BBB) represents a crucial interface between the circulatory system and the brain. In Drosophila melanogaster, the BBB is composed of perineurial and subperineurial glial cells. The perineurial glial cells are small mitotically active cells forming the outermost layer of the nervous system and are engaged in nutrient uptake. The subperineurial glial cells form occluding septate junctions to prevent paracellular diffusion of macromolecules into the nervous system. To address whether the subperineurial glia just form a simple barrier or whether they establish specific contacts with both the perineurial glial cells and inner central nervous system (CNS) cells, we undertook a detailed morphological analysis. Using genetically encoded markers alongside with high-resolution laser scanning confocal microscopy and transmission electron microscopy, we identified thin cell processes extending into the perineurial layer and into the CNS cortex. Interestingly, long cell processes were observed reaching the glia ensheathing the neuropil of the central brain. GFP reconstitution experiments highlighted multiple regions of membrane contacts between subperineurial and ensheathing glia. Furthermore, we identify the G-protein-coupled receptor (GPCR) Moody as negative regulator of the growth of subperineurial cell processes. Loss of moody triggered a massive overgrowth of subperineurial cell processes into the CNS cortex and, moreover, affected the polarized localization of the xenobiotic transporter Mdr65. Finally, we found that GPCR signaling, but not septate junction formation, is responsible for controlling membrane overgrowth. Our findings support the notion that the Drosophila BBB is able to bridge the communication gap between circulation and synaptic regions of the brain by long cell processes.

19.
Artigo em Inglês | MEDLINE | ID: mdl-38167424

RESUMO

Glia play a crucial role in providing metabolic support to neurons across different species. To do so, glial cells isolate distinct neuronal compartments from systemic signals and selectively transport specific metabolites and ions to support neuronal development and facilitate neuronal function. Because of their function as barriers, glial cells occupy privileged positions within the nervous system and have also evolved to serve as signaling intermediaries in various contexts. The fruit fly, Drosophila melanogaster, has significantly contributed to our understanding of glial barrier development and function. In this review, we will explore the formation of the glial sheath, blood-brain barrier, and nerve barrier, as well as the significance of glia-extracellular matrix interactions in barrier formation. Additionally, we will delve into the role of glia as signaling intermediaries in regulating nervous system development, function, and response to injury.


Assuntos
Proteínas de Drosophila , Drosophila melanogaster , Animais , Drosophila melanogaster/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo
20.
J Neurosci ; 32(22): 7466-76, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22649226

RESUMO

Kinesin heavy chain (Khc) is crucially required for axonal transport and khc mutants show axonal swellings and paralysis. Here, we demonstrate that in Drosophila khc is equally important in glial cells. Glial-specific downregulation of khc by RNA interference suppresses neuronal excitability and results in spastic flies. The specificity of the phenotype was verified by interspecies rescue experiments and further mutant analyses. Khc is mostly required in the subperineurial glia forming the blood-brain barrier. Following glial-specific knockdown, peripheral nerves are swollen with maldistributed mitochondria. To better understand khc function, we determined Khc-dependent Rab proteins in glia and present evidence that Neurexin IV, a well known blood-brain barrier constituent, is one of the relevant cargo proteins. Our work shows that the role of Khc for neuronal excitability must be considered in the light of its necessity for directed transport in glia.


Assuntos
Regulação para Baixo/fisiologia , Cinesinas/metabolismo , Neuroglia/metabolismo , Neurônios/fisiologia , Animais , Animais Geneticamente Modificados , Transporte Axonal/genética , Regulação para Baixo/genética , Drosophila , Proteínas de Drosophila/genética , Estimulação Elétrica , Cinesinas/genética , Larva , Locomoção/genética , Locomoção/fisiologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Potenciais da Membrana/genética , Técnicas de Patch-Clamp , Nervos Periféricos/citologia , Interferência de RNA/fisiologia , Proteínas rab de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA