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










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 13(2): e0191453, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29415052

RESUMO

Along the anterior-posterior axis the central nervous system is subdivided into segmental units (neuromeres) the composition of which is adapted to their region-specific functional requirements. In Drosophila melanogaster each neuromere is formed by a specific set of identified neural stem cells (neuroblasts, NBs). In the thoracic and anterior abdominal region of the embryonic ventral nerve cord segmental sets of NBs resemble the ground state (2nd thoracic segment, which does not require input of homeotic genes), and serial (segmental) homologs generate similar types of lineages. The three gnathal head segments form a transitional zone between the brain and the ventral nerve cord. It has been shown recently that although all NBs of this zone are serial homologs of NBs in more posterior segments, they progressively differ from the ground state in anterior direction (labial > maxillary > mandibular segment) with regard to numbers and expression profiles. To study the consequences of their derived characters we traced the embryonic lineages of gnathal NBs using the Flybow and DiI-labelling techniques. For a number of clonal types serial homology is rather clearly reflected by their morphology (location and projection patterns) and cell specific markers, despite of reproducible segment-specific differences. However, many lineages, particularly in the mandibular segment, show a degree of derivation that impedes their assignment to ground state serial homologs. These findings demonstrate that differences in gene expression profiles of gnathal NBs go along with anteriorly directed progressive derivation in the composition of their lineages. Furthermore, lineage sizes decrease from labial to mandibular segments, which in concert with decreasing NB-numbers lead to reduced volumes of gnathal neuromeres, most significantly in the mandibular segment.


Assuntos
Sistema Nervoso Central/citologia , Drosophila/citologia , Neurônios/citologia , Animais , Linhagem da Célula
2.
Development ; 144(1): 139-150, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27913640

RESUMO

Hox genes are known to specify motoneuron pools in the developing vertebrate spinal cord and to control motoneuronal targeting in several species. However, the mechanisms controlling axial diversification of muscle innervation patterns are still largely unknown. We present data showing that the Drosophila Hox gene Ultrabithorax (Ubx) acts in the late embryo to establish target specificity of ventrally projecting RP motoneurons. In abdominal segments A2 to A7, RP motoneurons innervate the ventrolateral muscles VL1-4, with VL1 and VL2 being innervated in a Wnt4-dependent manner. In Ubx mutants, these motoneurons fail to make correct contacts with muscle VL1, a phenotype partially resembling that of the Wnt4 mutant. We show that Ubx regulates expression of Wnt4 in muscle VL2 and that it interacts with the Wnt4 response pathway in the respective motoneurons. Ubx thus orchestrates the interaction between two cell types, muscles and motoneurons, to regulate establishment of the ventrolateral neuromuscular network.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster , Proteínas de Homeodomínio/fisiologia , Neurônios Motores/fisiologia , Músculos/embriologia , Junção Neuromuscular/embriologia , Junção Neuromuscular/genética , Fatores de Transcrição/fisiologia , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox/fisiologia , Genes de Insetos , Morfogênese/genética , Neurônios Motores/metabolismo , Desenvolvimento Muscular/genética , Músculos/metabolismo , Via de Sinalização Wnt
3.
Development ; 143(8): 1290-301, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-27095493

RESUMO

The numbers and types of progeny cells generated by neural stem cells in the developing CNS are adapted to its region-specific functional requirements. In Drosophila, segmental units of the CNS develop from well-defined patterns of neuroblasts. Here we constructed comprehensive neuroblast maps for the three gnathal head segments. Based on the spatiotemporal pattern of neuroblast formation and the expression profiles of 46 marker genes (41 transcription factors), each neuroblast can be uniquely identified. Compared with the thoracic ground state, neuroblast numbers are progressively reduced in labial, maxillary and mandibular segments due to smaller sizes of neuroectodermal anlagen and, partially, to suppression of neuroblast formation and induction of programmed cell death by the Hox gene Deformed Neuroblast patterns are further influenced by segmental modifications in dorsoventral and proneural gene expression. With the previously published neuroblast maps and those presented here for the gnathal region, all neuroectodermal neuroblasts building the CNS of the fly (ventral nerve cord and brain, except optic lobes) are now individually identified (in total 2×567 neuroblasts). This allows, for the first time, a comparison of the characteristics of segmental populations of stem cells and to screen for serially homologous neuroblasts throughout the CNS. We show that approximately half of the deutocerebral and all of the tritocerebral (posterior brain) and gnathal neuroblasts, but none of the protocerebral (anterior brain) neuroblasts, display serial homology to neuroblasts in thoracic/abdominal neuromeres. Modifications in the molecular signature of serially homologous neuroblasts are likely to determine the segment-specific characteristics of their lineages.


Assuntos
Sistema Nervoso Central/embriologia , Drosophila melanogaster/embriologia , Células-Tronco Neurais/metabolismo , Transcriptoma , Abdome/embriologia , Animais , Contagem de Células , Linhagem da Célula , Sistema Nervoso Central/citologia , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Genes de Insetos , Marcadores Genéticos , Células-Tronco Neurais/citologia , Tórax/citologia , Tórax/embriologia
4.
PLoS Genet ; 12(3): e1005961, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27015425

RESUMO

During central nervous system (CNS) development neural stem cells (Neuroblasts, NBs) have to acquire an identity appropriate to their location. In thoracic and abdominal segments of Drosophila, the expression pattern of Bithorax-Complex Hox genes is known to specify the segmental identity of NBs prior to their delamination from the neuroectoderm. Compared to the thoracic, ground state segmental units in the head region are derived to different degrees, and the precise mechanism of segmental specification of NBs in this region is still unclear. We identified and characterized a set of serially homologous NB-lineages in the gnathal segments and used one of them (NB6-4 lineage) as a model to investigate the mechanism conferring segment-specific identities to gnathal NBs. We show that NB6-4 is primarily determined by the cell-autonomous function of the Hox gene Deformed (Dfd). Interestingly, however, it also requires a non-cell-autonomous function of labial and Antennapedia that are expressed in adjacent anterior or posterior compartments. We identify the secreted molecule Amalgam (Ama) as a downstream target of the Antennapedia-Complex Hox genes labial, Dfd, Sex combs reduced and Antennapedia. In conjunction with its receptor Neurotactin (Nrt) and the effector kinase Abelson tyrosine kinase (Abl), Ama is necessary in parallel to the cell-autonomous Dfd pathway for the correct specification of the maxillary identity of NB6-4. Both pathways repress CyclinE (CycE) and loss of function of either of these pathways leads to a partial transformation (40%), whereas simultaneous mutation of both pathways leads to a complete transformation (100%) of NB6-4 segmental identity. Finally, we provide genetic evidences, that the Ama-Nrt-Abl-pathway regulates CycE expression by altering the function of the Hippo effector Yorkie in embryonic NBs. The disclosure of a non-cell-autonomous influence of Hox genes on neural stem cells provides new insight into the process of segmental patterning in the developing CNS.


Assuntos
Sistema Nervoso Central/embriologia , Proteínas de Drosophila/metabolismo , Proteínas de Homeodomínio/metabolismo , Imunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neurais/citologia , Proteínas Tirosina Quinases/metabolismo , Animais , Linhagem da Célula/genética , Ciclina E/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Imunoglobulinas/genética , Glicoproteínas de Membrana/genética , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Proteínas Tirosina Quinases/genética
5.
Biol Open ; 4(4): 420-34, 2015 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25819843

RESUMO

The clarification of complete cell lineages, which are produced by specific stem cells, is fundamental for understanding mechanisms, controlling the generation of cell diversity and patterning in an emerging tissue. In the developing Central Nervous System (CNS) of Drosophila, neural stem cells (neuroblasts) exhibit two periods of proliferation: During embryogenesis they produce primary lineages, which form the larval CNS. After a phase of mitotic quiescence, a subpopulation of them resumes proliferation in the larva to give rise to secondary lineages that build up the CNS of the adult fly. Within the ventral nerve cord (VNC) detailed descriptions exist for both primary and secondary lineages. However, while primary lineages have been linked to identified neuroblasts, the assignment of secondary lineages has so far been hampered by technical limitations. Therefore, primary and secondary neural lineages co-existed as isolated model systems. Here we provide the missing link between the two systems for all lineages in the thoracic and abdominal neuromeres. Using the Flybow technique, embryonic neuroblasts were identified by their characteristic and unique lineages in the living embryo and their further development was traced into the late larval stage. This comprehensive analysis provides the first complete view of which embryonic neuroblasts are postembryonically reactivated along the anterior/posterior-axis of the VNC, and reveals the relationship between projection patterns of primary and secondary sublineages.

6.
J Neurogenet ; 28(3-4): 171-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24913688

RESUMO

Studies performed at the level of single, identified cells in the fruitfly Drosophila have decisively contributed to our understanding of the mechanisms underlying the development and function of the nervous system. This review highlights some of the work based on single-cell analyses in the embryonic/larval CNS that sheds light on the principles underlying formation and organization of an entire segmental unit and its divergence along the anterior/posterior body axis.


Assuntos
Padronização Corporal/genética , Sistema Nervoso Central/embriologia , Proteínas de Drosophila/genética , Drosophila/genética , Genes Homeobox , Animais , Drosophila/embriologia
7.
Development ; 141(10): 2046-56, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24803653

RESUMO

The regulated head-to-tail expression of Hox genes provides a coordinate system for the activation of specific programmes of cell differentiation according to axial level. Recent work indicates that Hox expression can be regulated via RNA processing but the underlying mechanisms and biological significance of this form of regulation remain poorly understood. Here we explore these issues within the developing Drosophila central nervous system (CNS). We show that the pan-neural RNA-binding protein (RBP) ELAV (Hu antigen) regulates the RNA processing patterns of the Hox gene Ultrabithorax (Ubx) within the embryonic CNS. Using a combination of biochemical, genetic and imaging approaches we demonstrate that ELAV binds to discrete elements within Ubx RNAs and that its genetic removal reduces Ubx protein expression in the CNS leading to the respecification of cellular subroutines under Ubx control, thus defining for the first time a specific cellular role of ELAV within the developing CNS. Artificial provision of ELAV in glial cells (a cell type that lacks ELAV) promotes Ubx expression, suggesting that ELAV-dependent regulation might contribute to cell type-specific Hox expression patterns within the CNS. Finally, we note that expression of abdominal A and Abdominal B is reduced in elav mutant embryos, whereas other Hox genes (Antennapedia) are not affected. Based on these results and the evolutionary conservation of ELAV and Hox genes we propose that the modulation of Hox RNA processing by ELAV serves to adapt the morphogenesis of the CNS to axial level by regulating Hox expression and consequently activating local programmes of neural differentiation.


Assuntos
Proteínas de Drosophila/fisiologia , Drosophila melanogaster/embriologia , Proteínas ELAV/fisiologia , Genes Homeobox , Sistema Nervoso/embriologia , Processamento Pós-Transcricional do RNA , Animais , Sequência de Bases , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Dados de Sequência Molecular , Morfogênese/genética , Sistema Nervoso/metabolismo , Neurogênese/genética , Filogenia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
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
9.
Development ; 140(17): 3552-64, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23903193

RESUMO

The central nervous system of Drosophila melanogaster consists of fused segmental units (neuromeres), each generated by a characteristic number of neural stem cells (neuroblasts). In the embryo, thoracic and anterior abdominal neuromeres are almost equally sized and formed by repetitive sets of neuroblasts, whereas the terminal abdominal neuromeres are generated by significantly smaller populations of progenitor cells. Here we investigated the role of the Hox gene Abdominal-B in shaping the terminal neuromeres. We show that the regulatory isoform of Abdominal-B (Abd-B.r) not only confers abdominal fate to specific neuroblasts (e.g. NB6-4) and regulates programmed cell death of several progeny cells within certain neuroblast lineages (e.g. NB3-3) in parasegment 14, but also inhibits the formation of a specific set of neuroblasts in parasegment 15 (including NB7-3). We further show that Abd-B.r requires cooperation of the ParaHox gene caudal to unfold its full competence concerning neuroblast inhibition and specification. Thus, our findings demonstrate that combined action of Abdominal-B and caudal contributes to the size and composition of the terminal neuromeres by regulating both the number and lineages of specific neuroblasts.


Assuntos
Diferenciação Celular/fisiologia , Sistema Nervoso Central/embriologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Neurais/fisiologia , Cauda/embriologia , Fatores de Transcrição/metabolismo , Animais , Apoptose/fisiologia , Primers do DNA/genética , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Hibridização In Situ , Microscopia de Fluorescência
10.
Development ; 140(8): 1830-42, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23533181

RESUMO

The central nervous system is composed of segmental units (neuromeres), the size and complexity of which evolved in correspondence to their functional requirements. In Drosophila, neuromeres develop from populations of neural stem cells (neuroblasts) that delaminate from the early embryonic neuroectoderm in a stereotyped spatial and temporal pattern. Pattern units closely resemble the ground state and are rather invariant in thoracic (T1-T3) and anterior abdominal (A1-A7) segments of the embryonic ventral nerve cord. Here, we provide a comprehensive neuroblast map of the terminal abdominal neuromeres A8-A10, which exhibit a progressively derived character. Compared with thoracic and anterior abdominal segments, neuroblast numbers are reduced by 28% in A9 and 66% in A10 and are almost entirely absent in the posterior compartments of these segments. However, all neuroblasts formed exhibit serial homology to their counterparts in more anterior segments and are individually identifiable based on their combinatorial code of marker gene expression, position, delamination time point and the presence of characteristic progeny cells. Furthermore, we traced the embryonic origin and characterised the postembryonic lineages of a set of terminal neuroblasts, which have been previously reported to exhibit sex-specific proliferation behaviour during postembryonic development. We show that the respective sex-specific product of the gene doublesex promotes programmed cell death of these neuroblasts in females, and is needed for their survival, but not proliferation, in males. These data establish the terminal neuromeres as a model for further investigations into the mechanisms controlling segment- and sex-specific patterning in the central nervous system.


Assuntos
Padronização Corporal/fisiologia , Linhagem da Célula/fisiologia , Sistema Nervoso Central/embriologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Células-Tronco Neurais/citologia , Caracteres Sexuais , Abdome/embriologia , Animais , Apoptose/genética , Apoptose/fisiologia , Feminino , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Microscopia Confocal
11.
Development ; 139(14): 2510-22, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22675205

RESUMO

Key to understanding the mechanisms that underlie the specification of divergent cell types in the brain is knowledge about the neurectodermal origin and lineages of their stem cells. Here, we focus on the origin and embryonic development of the four neuroblasts (NBs) per hemisphere in Drosophila that give rise to the mushroom bodies (MBs), which are central brain structures essential for olfactory learning and memory. We show that these MBNBs originate from a single field of proneural gene expression within a specific mitotic domain of procephalic neuroectoderm, and that Notch signaling is not needed for their formation. Subsequently, each MBNB occupies a distinct position in the developing MB cortex and expresses a specific combination of transcription factors by which they are individually identifiable in the brain NB map. During embryonic development each MBNB generates an individual cell lineage comprising different numbers of neurons, including intrinsic γ-neurons and various types of non-intrinsic neurons that do not contribute to the MB neuropil. This contrasts with the postembryonic phase of MBNB development during which they have been shown to produce identical populations of intrinsic neurons. We show that different neuron types are produced in a lineage-specific temporal order and that neuron numbers are regulated by differential mitotic activity of the MBNBs. Finally, we demonstrate that γ-neuron axonal outgrowth and spatiotemporal innervation of the MB lobes follows a lineage-specific mode. The MBNBs are the first stem cells of the Drosophila CNS for which the origin and complete cell lineages have been determined.


Assuntos
Drosophila/citologia , Corpos Pedunculados/citologia , Neurônios/citologia , Animais , Drosophila/embriologia , Drosophila/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Corpos Pedunculados/embriologia , Corpos Pedunculados/metabolismo , Placa Neural/citologia , Placa Neural/metabolismo , Neurônios/metabolismo
12.
J Neurosci ; 31(44): 15870-83, 2011 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-22049430

RESUMO

Decisive contributions to our understanding of the mechanisms underlying the development of the nervous system have been made by studies performed at the level of single, identified cells in the fruit fly Drosophila. While all the motor neurons and glial cells in thoracic and abdominal segments of the Drosophila embryo have been individually identified, few of the interneurons, which comprise the vast majority of cells in the CNS, have been characterized at this level. We have applied a single cell labeling technique to carry out a detailed morphological characterization of the entire population of interneurons in abdominal segments A1-A7. Based on the definition of a set of spatial parameters specifying axonal projection patterns and cell body positions, we have identified 270 individual cell types as the complete hemisegmental set of interneurons and placed these in an interactive database. As well as facilitating analyses of developmental processes, this comprehensive set of data sheds light on the principles underlying the formation and organization of an entire segmental unit of the CNS.


Assuntos
Axônios/fisiologia , Sistema Nervoso Central/citologia , Interneurônios/classificação , Interneurônios/citologia , Aminoácidos/metabolismo , Animais , Animais Geneticamente Modificados , Axônios/metabolismo , Antígenos CD8/metabolismo , Contagem de Células/métodos , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Lateralidade Funcional/fisiologia , Proteínas de Fluorescência Verde/genética , Interneurônios/fisiologia , Proteínas com Homeodomínio LIM/metabolismo , Modelos Neurológicos , Vias Neurais/fisiologia , Estatística como Assunto , Fatores de Transcrição/metabolismo
13.
Gene Expr Patterns ; 10(6): 274-82, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20558325

RESUMO

The initial steps towards the generation of cell diversity in the central nervous system of the fruitfly Drosophila melanogaster take place during early phases of embryonic development when a stereotypic population of neural progenitor cells (neuroblasts and midline precursors) is formed in a precise spatial and temporal pattern, and subsequently expresses a particular sequence of genes. The clarification of the positional, temporal and molecular features of the individual progenitor cells in the nerve cord and brain as well as of their specific types of neuronal and/or glial progeny cells forms an essential basis to understand the mechanisms controlling their development. The present study contributes to this effort by tracing the expression of period and timeless, two genes that encode transcription factors with a key role in the molecular mechanism of the biological clock. Using a combination of genetic markers and immunocytochemistry with antibodies specific for period and timeless we define the number, location, origin and lineage of period cells in the nerve cord throughout embryogenesis. We also provide the first description of the expression of timeless in the embryonic central nervous system. We found a major transformation in the number and types of cells that express period and timeless takes place between embryonic and larval life.


Assuntos
Linhagem da Célula/genética , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/embriologia , Proteínas Circadianas Period/genética , Animais , Animais Geneticamente Modificados , Padronização Corporal/genética , Movimento Celular/fisiologia , Sistema Nervoso Central/citologia , Células Clonais/metabolismo , Células Clonais/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Larva/genética , Larva/metabolismo , Estágios do Ciclo de Vida/genética , Modelos Biológicos , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas Circadianas Period/metabolismo
14.
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
15.
Dev Biol ; 337(2): 415-24, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19914234

RESUMO

During development, neural progenitor cells or neuroblasts generate a great intra- and inter-segmental diversity of neuronal and glial cell types in the nervous system. In thoracic segments of the embryonic central nervous system of Drosophila, the neuroblast NB6-4t undergoes an asymmetric first division to generate a neuronal and a glial sublineage, while abdominal NB6-4a divides once symmetrically to generate only 2 glial cells. We had earlier reported a critical function for the G1 cyclin, CyclinE (CycE) in regulating asymmetric cell division in NB6-4t. Here we show that (i) this function of CycE is independent of its role in cell cycle regulation and (ii) the two functions are mediated by distinct domains at the protein level. Results presented here also suggest that CycE inhibits the function of Prospero and facilitates its cortical localization, which is critical for inducing stem cell behaviour, i.e. asymmetric cell division of NB6-4t. Furthermore our data imply that CycE is required for the maintenance of stem cell identity of most other neuroblasts.


Assuntos
Ciclo Celular , Linhagem da Célula , Ciclina E/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Proliferação de Células , Ciclina E/química , Drosophila melanogaster/metabolismo , Neurônios/metabolismo , Ligação Proteica , Transporte Proteico , Deleção de Sequência , Regulação para Cima
16.
Mech Dev ; 127(1-2): 137-45, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19799999

RESUMO

Homeotic/Hox genes are known to specify a given developmental pathway by regulating the expression of downstream effector genes. During embryonic CNS development of Drosophila, the Hox protein Abdominal-A (AbdA) is required for the specification of the abdominal NB6-4 lineage. It does so by down regulating the expression of the cell cycle regulator gene Dcyclin E (CycE). CycE is normally expressed in the thoracic NB6-4 lineage to give rise to mixed lineage of neurons and glia, while only glial cells are produced from the abdominal NB6-4 lineage due to the repression of CycE by AbdA. Here we investigate how AbdA represses the expression of CycE to define the abdominal fate of a single NB6-4 precursor cell. We analyze, both in vitro and in vivo, the regulation of a 1.9 kb CNS-specific CycE enhancer element in the abdominal NB6-4 lineage. We show that CycE is a direct target of AbdA and it binds to the CNS specific enhancer of CycE to specifically repress the enhancer activity in vivo. Our results suggest preferential involvement of a series of multiple AbdA binding sites to selectively enhance the repression of CycE transcription. Furthermore, our data suggest a complex network to regulate CycE expression where AbdA functions as a key regulator.


Assuntos
Sistema Nervoso Central/embriologia , Ciclina E/fisiologia , Drosophila melanogaster/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Linhagem da Célula , Ciclina E/antagonistas & inibidores , Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Neuroglia/metabolismo , Neurônios/metabolismo , Transcrição Gênica , Transgenes
17.
Dev Biol ; 336(2): 156-68, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19782677

RESUMO

The generation of cellular diversity in the nervous system involves the mechanism of asymmetric cell division. Besides an array of molecules, including the Par protein cassette, a heterotrimeric G protein signalling complex, Inscuteable plays a major role in controlling asymmetric cell division, which ultimately leads to differential activation of the Notch signalling pathway and correct specification of the two daughter cells. In this context, Notch is required to be active in one sibling and inactive in the other. Here, we investigated the requirement of genes previously known to play key roles in sibling cell fate specification such as members of the Notch signalling pathway, e.g., Notch (N), Delta (Dl), and kuzbanian (kuz) and a crucial regulator of asymmetric cell division, inscuteable (insc) throughout lineage progression of 4 neuroblasts (NB1-1, MP2, NB4-2, and NB7-1). Notch-mediated cell fate specification defects were cell-autonomous and were observed in all neuroblast lineages even in cells born from late ganglion mother cells (GMC) within the lineages. We also show that Dl functions non-autonomously during NB lineage progression and clonal cells do not require Dl from within the clone. This suggests that within a NB lineage Dl is dispensable for sibling cell fate specification. Furthermore, we provide evidence that kuz is involved in sibling cell fate specification in the central nervous system. It is cell-autonomously required in the same postmitotic cells which also depend on Notch function. This indicates that KUZ is required to facilitate a functional Notch signal in the Notch-dependent cell for correct cell fate specification. Finally, we show that three neuroblast lineages (NB1-1, NB4-2, and NB7-1) require insc function for sibling cell fate specification in cells born from early GMCs whereas insc is not required in cells born from later GMCs of the same lineages. Thus, there is differential requirement for insc for cell fate specification depending on the stage of lineage progression of NBs.


Assuntos
Proteínas do Citoesqueleto/fisiologia , Desintegrinas/fisiologia , Proteínas de Drosophila/fisiologia , Drosophila/embriologia , Proteínas de Membrana/fisiologia , Metaloendopeptidases/fisiologia , Neurônios/citologia , Receptores Notch/fisiologia , Animais , Sequência de Bases , Linhagem da Célula , Proteínas do Citoesqueleto/genética , Primers do DNA , Desintegrinas/genética , Proteínas de Drosophila/genética , Embrião não Mamífero/citologia , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/genética , Metaloendopeptidases/genética , Reação em Cadeia da Polimerase , Receptores Notch/genética , Transdução de Sinais
18.
Neural Dev ; 4: 30, 2009 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-19650920

RESUMO

BACKGROUND: The Drosophila embryonic central nervous system (CNS) develops from two sets of progenitor cells, neuroblasts and ventral midline progenitors, which behave differently in many respects. Neuroblasts derive from the neurogenic region of the ectoderm and form the lateral parts of the CNS. Ventral midline precursors are formed by two rows of mesectodermal cells and build the CNS midline. There is plenty of evidence that individual identities are conferred to precursor cells by positional information in the ectoderm. It is unclear, however, how far the precursors can maintain their identities and developmental properties in the absence of normal external signals. RESULTS: To separate the respective contributions of autonomous properties versus extrinsic signals during their further development, we isolated individual midline precursors and neuroectodermal precursors at the pre-mitotic gastrula stage, traced their development in vitro, and analyzed the characteristics of their lineages in comparison with those described for the embryo. Although individually cultured mesectodermal cells exhibit basic characteristics of CNS midline progenitors, the clones produced by these progenitors differ from their in situ counterparts with regard to cell numbers, expression of molecular markers, and the separation of neuronal and glial fate. In contrast, clones derived from individually cultured precursors taken from specific dorsoventral zones of the neuroectoderm develop striking similarities to the lineages of neuroblasts that normally delaminate from these zones and develop in situ. CONCLUSION: This in vitro analysis allows for the first time a comparison of the developmental capacities in situ and in vitro of individual neural precursors of defined spatial and temporal origin. The data reveal that cells isolated at the pre-mitotic and pre-delamination stage express characteristics of the progenitor type appropriate to their site of origin in the embryo. However, presumptive neuroblasts, once specified in the neuroectoderm, exhibit a higher degree of autonomy regarding generation of their lineages compared to mesectodermal midline progenitors.


Assuntos
Padronização Corporal/fisiologia , Linhagem da Célula/fisiologia , Sistema Nervoso Central/citologia , Células-Tronco Embrionárias/fisiologia , Mesoderma/citologia , Placa Neural/citologia , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Células Cultivadas , Sistema Nervoso Central/embriologia , Drosophila , Proteínas de Drosophila/metabolismo , Embrião não Mamífero , Mesoderma/embriologia , Placa Neural/embriologia
19.
Fly (Austin) ; 2(6): 316-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19077542

RESUMO

In this article we highlight some of the recently accumulating evidence showing that Hox genes are involved at different steps during the development of neural cell lineages to control segmental patterning of the CNS. In addition to their well-known early role in establishing segmental identities, Hox genes act on neural stem cells and their progeny at various stages during embryonic and postembryonic development to control proliferation, cell fate and/or apoptosis in a segment-specific manner. This leads to differential shaping of serially homologous lineages and thus to structural diversification of segmental CNS units (neuromeres) in adaptation to their specific functional tasks in processing sensory information and generation of motor patterns.


Assuntos
Drosophila melanogaster/fisiologia , Genes Homeobox/fisiologia , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia
20.
Development ; 135(20): 3435-45, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18799545

RESUMO

The generation of morphological diversity among segmental units of the nervous system is crucial for correct matching of neurons with their targets and for formation of functional neuromuscular networks. However, the mechanisms leading to segment diversity remain largely unknown. We report here that the Hox genes Ultrabithorax (Ubx) and Antennapedia (Antp) regulate segment-specific survival of differentiated motoneurons in the ventral nerve cord of Drosophila embryos. We show that Ubx is required to activate segment-specific apoptosis in these cells, and that their survival depends on Antp. Expression of the Ubx protein is strongly upregulated in the motoneurons shortly before they undergo apoptosis, and our results indicate that this late upregulation is required to activate reaper-dependent cell death. We further demonstrate that Ubx executes this role by counteracting the function of Antp in promoting cell survival. Thus, two Hox genes contribute to segment patterning and diversity in the embryonic CNS by carrying out opposing roles in the survival of specific differentiated motoneurons.


Assuntos
Proteína do Homeodomínio de Antennapedia/fisiologia , Apoptose , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Proteínas de Drosophila/fisiologia , Drosophila/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/fisiologia , Fatores de Transcrição/fisiologia , Animais , Proteína do Homeodomínio de Antennapedia/genética , Proteína do Homeodomínio de Antennapedia/metabolismo , Diferenciação Celular , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Embrião não Mamífero , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...