Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 370(1): 33-41, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22884563

RESUMO

Neural progenitor cells in the pseudostratified neuroepithelium in vertebrates undergo interkinetic nuclear migration, which results in mitotic cells localized to the apical surface. Interphase nuclei are distributed throughout the rest of the epithelium. How mitosis is coordinated with nuclear movement is unknown, and the mechanism by which the nucleus migrates apically is controversial. Using time-lapse confocal microscopy, we show that nuclei migrate apically in G2 phase via microtubules. However, late in G2, centrosomes leave the apical surface after cilia are disassembled, and mitosis initiates away from the apical surface. The mitotic cell then rounds up to the apical surface, which is an actin-dependent process. This behavior is observed in both chicken neural-tube-slice preparations and in mouse cortical slices, and therefore is likely to be a general feature of interkinetic nuclear migration. We propose a new model for interkinetic nuclear migration in which actin and microtubules are used to position the mitotic cell at the apical surface.


Assuntos
Núcleo Celular/fisiologia , Mitose/fisiologia , Modelos Biológicos , Células-Tronco Neurais/fisiologia , Células Neuroepiteliais/citologia , Animais , Centrossomo/fisiologia , Embrião de Galinha , Eletroporação , Fase G2/fisiologia , Camundongos , Microscopia Confocal , Microtúbulos/fisiologia , Células Neuroepiteliais/fisiologia , Imagem com Lapso de Tempo
2.
Dev Growth Differ ; 54(3): 306-16, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22524603

RESUMO

During interkinetic nuclear migration (INM), the nuclei in many epithelial cells migrate between the apical and basal surfaces, coordinating with the cell cycle, and undergoing cytokinesis at the apical surface. INM is observed in a wide variety of tissues and species. Recent advances in time-lapse microscopy have provided clues about the mechanisms and functions of INM. Whether actin or microtubules are responsible for nuclear migration is controversial. How mitosis is initiated during INM is poorly understood, as is the relationship between the cell cycle and nuclear movement. It is possible that the disagreements stem from differences in the tissues being studied, since epithelia undergoing INM vary greatly in terms of cell height and cell fates. In this review we examine the reports addressing the mode and mechanisms that regulate INM and suggest possible functions for this dramatic event.


Assuntos
Núcleo Celular/fisiologia , Microtúbulos/fisiologia , Mitose , Células Neuroepiteliais/citologia , Animais , Crescimento Celular , Polaridade Celular , Proliferação de Células , Forma Celular , Centrossomo/fisiologia , Células Neuroepiteliais/fisiologia , Neurogênese , Neurônios/citologia , Neurônios/fisiologia , Imagem com Lapso de Tempo
3.
Dev Dyn ; 240(9): 2084-100, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22016183

RESUMO

Migration and differentiation of cranial neural crest cells are largely controlled by environmental cues, whereas pathfinding at the trunk level is dictated by cell-autonomous molecular changes owing to early specification of the premigratory crest. Here, we investigated the migration and patterning of vagal neural crest cells. We show that (1) vagal neural crest cells exhibit some developmental bias, and (2) they take separate pathways to the heart and to the gut. Together these observations suggest that prior specification dictates initial pathway choice. However, when we challenged the vagal neural crest cells with different migratory environments, we observed that the behavior of the anterior vagal neural crest cells (somite-level 1-3) exhibit considerable migratory plasticity, whereas the posterior vagal neural crest cells (somite-level 5-7) are more restricted in their behavior. We conclude that the vagal neural crest is a transitional population that has evolved between the head and the trunk.


Assuntos
Crista Neural/embriologia , Codorniz/embriologia , Nervo Vago/embriologia , Animais , Diferenciação Celular/imunologia , Movimento Celular/genética , Movimento Celular/fisiologia , Células Cultivadas , Embrião de Galinha , Galinhas , Morfogênese/genética , Morfogênese/fisiologia , Crista Neural/metabolismo , Nervo Vago/metabolismo
4.
Cell Adh Migr ; 4(4): 567-85, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20962585

RESUMO

Neural crest cells are pluripotent cells that emerge from the neural epithelium, migrate extensively, and differentiate into numerous derivatives, including neurons, glial cells, pigment cells and connective tissue. Major questions concerning their morphogenesis include: 1) what establishes the pathways of migration and 2) what controls the final destination and differentiation of various neural crest subpopulations. These questions will be addressed in this review. Neural crest cells from the trunk level have been explored most extensively. Studies show that melanoblasts are specified shortly after they depart from the neural tube, and this specification directs their migration into the dorsolateral pathway. We also consider other reports that present strong evidence for ventrally migrating neural crest cells being similarly fate restricted. Cranial neural crest cells have been less analyzed in this regard but the preponderance of evidence indicates that either the cranial neural crest cells are not fate-restricted, or are extremely plastic in their developmental capability and that specification does not control pathfinding. Thus, the guidance mechanisms that control cranial neural crest migration and their behavior vary significantly from the trunk. The vagal neural crest arises at the axial level between the cranial and trunk neural crest and represents a transitional cell population between the head and trunk neural crest. We summarize new data to support this claim. In particular, we show that: 1) the vagal-level neural crest cells exhibit modest developmental bias; 2) there are differences in the migratory behavior between the anterior and the posterior vagal neural crest cells reminiscent of the cranial and the trunk neural crest, respectively; 3) the vagal neural crest cells take the dorsolateral pathway to the pharyngeal arches and the heart, but the ventral pathway to the peripheral nervous system and the gut. However, these pathways are not rigidly specified because of prior fate restriction. Understanding the molecular, cellular and behavioral differences between these three populations of neural crest cells will be of enormous assistance when trying to understand the evolution of the neck.


Assuntos
Morfogênese , Crista Neural/citologia , Abdome/embriologia , Abdome/inervação , Animais , Sistema Nervoso Autônomo/citologia , Sistema Nervoso Autônomo/embriologia , Diferenciação Celular , Movimento Celular , Gânglios Espinais/citologia , Gânglios Espinais/embriologia , Cabeça/embriologia , Cabeça/inervação , Pescoço/embriologia , Pescoço/inervação , Células Receptoras Sensoriais/citologia , Tórax/embriologia , Tórax/inervação
5.
Development ; 136(11): 1849-58, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19403660

RESUMO

The first neural crest cells to emigrate from the neural tube are specified as neurons and glial cells and are subsequently followed by melanocytes of the skin. We wished to understand how this fate switch is controlled. The transcriptional repressor FOXD3 is expressed exclusively in the neural/glial precursors and MITF is expressed only in melanoblasts. Moreover, FOXD3 represses melanogenesis. Here we show that avian MITF expression begins very early during melanoblast migration and that loss of MITF in melanoblasts causes them to transdifferentiate to a glial phenotype. Ectopic expression of FOXD3 represses MITF in cultured neural crest cells and in B16-F10 melanoma cells. We also show that FOXD3 does not bind directly to the MITF promoter, but instead interacts with the transcriptional activator PAX3 to prevent the binding of PAX3 to the MITF promoter. Overexpression of PAX3 is sufficient to rescue MITF expression from FOXD3-mediated repression. We conclude that FOXD3 controls the lineage choice between neural/glial and pigment cells by repressing MITF during the early phase of neural crest migration.


Assuntos
Linhagem da Célula/fisiologia , Fatores de Transcrição Forkhead/fisiologia , Melanócitos/citologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Crista Neural/fisiologia , Neuroglia/citologia , Animais , Sequência de Bases , Movimento Celular , Transdiferenciação Celular , Células Cultivadas , Embrião de Galinha , Embrião não Mamífero , Fatores de Transcrição Forkhead/genética , Melanócitos/fisiologia , Dados de Sequência Molecular , Crista Neural/citologia , Crista Neural/embriologia , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/fisiologia , Regiões Promotoras Genéticas , Codorniz
6.
Development ; 136(11): 1801-12, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19429784

RESUMO

An epithelial-mesenchymal transition (EMT) is the process whereby epithelial cells become mesenchymal cells, and is typified by the generation of neural crest cells from the neuroepithelium of the dorsal neural tube. To investigate the neural crest EMT, we performed live cell confocal time-lapse imaging to determine the sequence of cellular events and the role of cell division in the EMT. It was observed that in most EMTs, the apical cell tail is retracted cleanly from the lumen of the neuroepithelium, followed by movement of the cell body out of the neural tube. However, exceptions to this sequence include the rupture of the neural crest cell tail during retraction (junctional complexes not completely downregulated), or translocation of the cell body away from the apical surface while morphologically rounded up in M phase (no cell tail retraction event). We also noted that cell tail retraction can occur either before or after the redistribution of apical-basolateral epithelial polarity markers. Surprisingly, we discovered that when an EMT was preceded by a mitotic event, the plane of cytokinesis does not predict neural crest cell fate. Moreover, when daughter cells are separated from the adherens junctions by a parallel mitotic cleavage furrow, most re-establish contact with the apical surface. The diversity of cellular mechanisms by which neural crest cells can separate from the neural tube suggests that the EMT program is a complex network of non-linear mechanisms that can occur in multiple orders and combinations to allow neural crest cells to escape from the neuroepithelium.


Assuntos
Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Mesoderma/citologia , Crista Neural/citologia , Animais , Divisão Celular/fisiologia , Movimento Celular/fisiologia , Centrossomo/fisiologia , Embrião de Galinha , Crista Neural/embriologia , Tubo Neural/citologia , Tubo Neural/embriologia
7.
Semin Cell Dev Biol ; 20(1): 90-104, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18977309

RESUMO

Pigment patterns in the integument have long-attracted attention from both scientists and non-scientists alike since their natural attractiveness combines with their excellence as models for the general problem of pattern formation. Pigment cells are formed from the neural crest and must migrate to reach their final locations. In this review, we focus on our current understanding of mechanisms underlying the control of pigment cell migration and patterning in diverse vertebrates. The model systems discussed here - chick, mouse, and zebrafish - each provide unique insights into the major morphogenetic events driving pigment pattern formation. In birds and mammals, melanoblasts must be specified before they can migrate on the dorsolateral pathway. Transmembrane receptors involved in guiding them onto this route include EphB2 and Ednrb2 in chick, and Kit in mouse. Terminal migration depends, in part, upon extracellular matrix reorganization by ADAMTS20. Invasion of the ectoderm, especially into the feather germ and hair follicles, requires specific signals that are beginning to be characterized. We summarize our current understanding of the mechanisms regulating melanoblast number and organization in the epidermis. We note the apparent differences in pigment pattern formation in poikilothermic vertebrates when compared with birds and mammals. With more pigment cell types, migration pathways are more complex and largely unexplored; nevertheless, a role for Kit signaling in melanophore migration is clear and indicates that at least some patterning mechanisms may be highly conserved. We summarize the multiple factors thought to contribute to zebrafish embryonic pigment pattern formation, highlighting a recent study identifying Sdf1a as one factor crucial for regulation of melanophore positioning. Finally, we discuss the mechanisms generating a second, metamorphic pigment pattern in adult fish, emphasizing recent studies strengthening the evidence that undifferentiated progenitor cells play a major role in generating adult pigment cells.


Assuntos
Forma Celular , Pigmentação/fisiologia , Vertebrados/fisiologia , Envelhecimento/fisiologia , Animais , Humanos , Modelos Biológicos , Células-Tronco
8.
Commun Integr Biol ; 2(6): 489-93, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20195454

RESUMO

By developing a technique for imaging the avian neural crest epithelial-mesenchymal transition (EMT), we have discovered cellular behaviors that challenge current thinking on this important developmental event, including the probability that complete disassembly of the adherens junctions may not control whether or not a neural epithelial cell undergoes an EMT. Further, neural crest cells can adopt multiple modes of cell motility in order to emigrate from the neuroepithelium. We also gained insights into interkinetic nuclear migration (INM). For example, the movement of the nucleus from the basal to apical domain may not require microtubule motors nor an intact nuclear envelope, and the nucleus does not always need to reach the apical surface in order for cytokinesis to occur. These studies illustrate the value of live-cell imaging to elucidate cellular processes.

9.
Pigment Cell Melanoma Res ; 21(6): 598-610, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19067969

RESUMO

Melanocytes differentiate from the neural crest (NC), which is a transient population of cells that delaminates from the neural tube and migrates extensively throughout the embryo during vertebrate development. Melanoblast specification from NC precursors is a progressive process during which initially pluripotent cells become restricted to the melanogenic lineage and adopt the gene expression profile and morphology of melanocytes. This specification process is governed primarily by Wnt and BMP signaling molecules, although other signaling pathways, such as those activated by Kit and Endothelin 3, can also stimulate melanogenesis. The transcriptional repressor FoxD3 occupies a central role in melanocyte fate determination by repressing melanogenesis in premigratory NC cells and in other NC lineages.


Assuntos
Linhagem da Célula , Melanócitos/citologia , Crista Neural/embriologia , Organogênese/fisiologia , Animais , Diferenciação Celular , Humanos , Melanócitos/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Transdução de Sinais
10.
Development ; 135(24): 4113-22, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19004859

RESUMO

Neural crest cells that become pigment cells migrate along a dorsolateral route between the ectoderm and the somite, whereas most other neural crest cells are inhibited from entering this space. This pathway choice has been attributed to unique, cell-autonomous migratory properties acquired by neural crest cells when they become specified as melanoblasts. By shRNA knockdown and overexpression experiments, we investigated the roles of three transmembrane receptors in regulating dorsolateral pathfinding in the chick trunk. We show that Endothelin receptor B2 (EDNRB2) and EphB2 are both determinants in this process, and that, unlike in other species, c-KIT is not. We demonstrate that the overexpression of EDNRB2 can maintain normal dorsolateral migration of melanoblasts in the absence of EphB2, and vice versa, suggesting that changes in receptor expression levels regulate the invasion of this pathway. Furthermore, by heterotopic grafting, we show that neural crest cell populations that do not rely on the activation of these receptors can migrate dorsolaterally only if this path is free of inhibitory molecules. We conclude that the requirement for EDNRB2 and EphB2 expression by melanoblasts is to support their migration by helping them to overcome repulsive or non-permissive cues in the dorsolateral environment.


Assuntos
Crista Neural/embriologia , Receptor EphB2/fisiologia , Receptores de Endotelina/fisiologia , Animais , Animais Geneticamente Modificados , Sequência de Bases , Padronização Corporal/genética , Padronização Corporal/fisiologia , Movimento Celular , Sobrevivência Celular , Embrião de Galinha , Coturnix , Primers do DNA/genética , Regulação da Expressão Gênica no Desenvolvimento , Melanócitos/citologia , Melanócitos/fisiologia , Modelos Neurológicos , Crista Neural/citologia , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/fisiologia , RNA Interferente Pequeno/genética , Receptor EphB2/genética , Receptores de Endotelina/genética , Transdução de Sinais , Quimeras de Transplante/embriologia
11.
Dev Biol ; 313(1): 279-92, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18031721

RESUMO

The enteric nervous system (ENS) is formed from vagal and sacral neural crest cells (NCC). Vagal NCC give rise to most of the ENS along the entire gut, whereas the contribution of sacral NCC is mainly limited to the hindgut. This, and data from heterotopic quail-chick grafting studies, suggests that vagal and sacral NCC have intrinsic differences in their ability to colonize the gut, and/or to respond to signalling cues within the gut environment. To better understand the molecular basis of these differences, we studied the expression of genes known to be essential for ENS formation, in sacral NCC within the chick hindgut. Our results demonstrate that, as in vagal NCC, Sox10, EdnrB, and Ret are expressed in sacral NCC within the gut. Since we did not detect a qualitative difference in expression of these ENS genes we performed DNA microarray analysis of vagal and sacral NCC. Of 11 key ENS genes examined from the total data set, Ret was the only gene identified as being highly differentially expressed, with a fourfold increase in expression in vagal versus sacral NCC. We also found that over-expression of RET in sacral NCC increased their ENS developmental potential such that larger numbers of cells entered the gut earlier in development, thus promoting the fate of sacral NCC towards that of vagal NCC.


Assuntos
Movimento Celular , Sistema Nervoso Entérico/embriologia , Crista Neural/citologia , Proteínas Proto-Oncogênicas c-ret/metabolismo , Animais , Embrião de Galinha , Proteínas de Ligação a DNA/metabolismo , Sistema Digestório/embriologia , Sistema Digestório/inervação , Sistema Digestório/metabolismo , Embrião não Mamífero/metabolismo , Sistema Nervoso Entérico/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , Crista Neural/transplante , Análise de Sequência com Séries de Oligonucleotídeos , Codorniz , Fatores de Transcrição SOXE , Sacro/citologia , Fatores de Transcrição/metabolismo , Transplante Heterólogo
12.
Dev Dyn ; 236(1): 1-19, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16894594

RESUMO

There are two principal models to explain neural crest patterning. One assumes that neural crest cells are multipotent precursors that migrate throughout the embryo and differentiate according to cues present in the local environment. A second proposes that the neural crest is a population of cells that becomes restricted to particular fates early in its existence and migrates along particular pathways dependent on unique cell-autonomous properties. Although it is now evident that the neural crest cell population, as a whole, is actually heterogenous (composed of both multipotent and restricted progenitors), evidence supporting the model of prespecification has increased over the past few years. This review will begin by telling the story of melanoblasts: a neural crest subpopulation that is biased toward a single fate and subsequently acquires intrinsic properties that guide cells of this lineage to their final destination. The remainder of this review will explore whether this model is exclusive to melanoblasts or if it can also be used to explain the patterning of other neural crest cells like those of the sensory, sympathoadrenal, and enteric lineages.


Assuntos
Linhagem da Célula , Crista Neural/citologia , Crista Neural/crescimento & desenvolvimento , Animais , Padronização Corporal , Diferenciação Celular , Indução Embrionária , Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/fisiologia , Humanos , Modelos Biológicos , Neurônios/metabolismo
14.
Dev Dyn ; 229(1): 42-53, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14699576

RESUMO

To investigate the roles that matrix-degrading proteases may have in development of the chicken embryo, we documented the expression pattern of matrix metalloprotease-2 (MMP-2, 72-kDa type IV collagenase or gelatinase A) and perturbed its function in vitro and in vivo. MMP-2 is expressed as neural crest cells detach from the neural epithelium during an epithelial-mesenchymal transformation (EMT) but is rapidly extinguished as they disperse. It is also expressed in the sclerotome and in the dermis at the time that the EMT is initiated, and also as these cells migrate, and is down-regulated once motility has ceased. These patterns suggest that MMP-2 plays a role in cell motility during the EMT and during later morphogenesis. Inhibitors of MMPs, including BB-94 and TIMP-2 (tissue inhibitor of metalloprotease-2), prevent the EMT that generates neural crest cells, both in tissue culture and in vivo, but do not affect migration of the cells that have already detached from the neural tube. Similarly, knockdown of MMP-2 expression in the dorsal neural tube using antisense morpholino oligos perturbs the EMT, but also does not affect migration of neural crest cells after they have detached from the neural tube. On the other hand, when somites in culture are treated with TIMP-2, some mesenchymal cells are produced, suggesting that they undergo the EMT, but show greatly reduced migration through the collagen gel. MMP-2 is also expressed in mesenchyme where tissue remodeling is in progress, such as in the developing feather germs, in the head mesenchyme, in the lateral plate mesoderm, and in the limb dermis, especially in the regions where tendons are developing. Comparisons of these expression patterns in multiple embryonic tissues suggest a probable role for MMP-2 in the migration phase of the EMT, in addition to mesenchyme dispersion and tissue remodeling. Developmental Dynamics 229:42-53, 2004.


Assuntos
Embrião de Galinha/embriologia , Embrião de Galinha/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Animais , Sequência de Bases , Movimento Celular , Coturnix/embriologia , Coturnix/genética , Coturnix/metabolismo , DNA Complementar/genética , Epitélio/embriologia , Epitélio/enzimologia , Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Hibridização In Situ , Metaloproteinase 2 da Matriz/genética , Mesoderma/citologia , Mesoderma/enzimologia , Crista Neural/citologia , Crista Neural/embriologia , Crista Neural/enzimologia , Somitos/citologia , Somitos/enzimologia
15.
Dev Dyn ; 226(3): 470-7, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12619133

RESUMO

The use of antisense morpholino oligos to inhibit the translation of a target transcript has been applied recently to studies of the chicken embryo. In contrast to other developmental systems such as in frog, sea urchin, and zebrafish that permit the direct microinjection of morpholinos into a blastomere, square pulse electroporation is used to introduce fluorescently tagged morpholinos into specific populations of chick embryo cells in ovo. This article reviews the methods that have proven successful, the types of controls that are necessary when performing knockdowns of gene expression in the chick embryo, and discusses the limitations of the current technique, as well as directions for further research.


Assuntos
Eletroporação/métodos , Regulação da Expressão Gênica no Desenvolvimento , Oligonucleotídeos Antissenso/farmacocinética , Animais , Embrião de Galinha , Galinhas , Eletroquímica , Técnicas de Transferência de Genes , Mutagênese , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/toxicidade
16.
Dev Biol ; 256(1): 146-59, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12654298

RESUMO

The ADAMs are a family of proteins containing multiple functional domains. We have cloned the avian orthologue of ADAM 10 and demonstrate that it has metalloprotease activity. Chick ADAM 10 is expressed in the developing dermatome and myotome of the somite, epidermis, gut endoderm, the epithelial tissues of the kidney, liver, and heart, and in neural crest cells. The expression patterns and protein distribution of ADAM 10 suggest it may play a significant role in the morphogenesis of several epithelial tissues. When a dominant-negative metalloprotease-mutant form of ADAM 10 is expressed in the ectoderm or ADAM 10 expression is knocked down with morpholinos, morphogenesis and tissue specification are altered.


Assuntos
Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Embrião de Galinha , Clonagem Molecular , DNA Complementar/genética , Epitélio/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Coração/embriologia , Hibridização In Situ , Dados de Sequência Molecular , Morfogênese/genética , Crista Neural/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos , Somitos/metabolismo
17.
Gene Expr Patterns ; 3(1): 53-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12609603

RESUMO

We have used whole-mount in situ hybridization to investigate the patterns of c-kit and Sl expression in stage 11-22 chicken embryos. Our analysis shows that c-kit and Sl are expressed quite differently in chicken embryos compared to the reported expression patterns of these genes in embryos of other taxa. Most notably, chicken c-kit is expressed in primordial germ cells as well as in the developing somite, the apical ectodermal ridge, and in the early foregut endoderm. Sl is expressed in the lateral and intermediate mesoderm and in extraembryonic membranes. These data suggest that chicken c-kit and Sl may play novel and unexpected roles in somitogenesis, limb development, and foregut development in avian embryos.


Assuntos
Extremidades/embriologia , Proteínas Proto-Oncogênicas c-kit/genética , Somitos/metabolismo , Fator de Células-Tronco/genética , Animais , Embrião de Galinha , Proteínas Proto-Oncogênicas c-kit/biossíntese , Fator de Células-Tronco/biossíntese
18.
Development ; 129(15): 3621-32, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12117812

RESUMO

Little is known about the mechanisms that direct neural crest cells to the appropriate migratory pathways. Our aim was to determine how neural crest cells that are specified as neurons and glial cells only migrate ventrally and are prevented from migrating dorsolaterally into the skin, whereas neural crest cells specified as melanoblasts are directed into the dorsolateral pathway. Eph receptors and their ephrin ligands have been shown to be essential for migration of many cell types during embryonic development. Consequently, we asked if ephrin-B proteins participate in the guidance of melanoblasts along the dorsolateral pathway, and prevent early migratory neural crest cells from invading the dorsolateral pathway. Using Fc fusion proteins, we detected the expression of ephrin-B ligands in the dorsolateral pathway at the stage when neural crest cells are migrating ventrally. Furthermore, we show that ephrins block dorsolateral migration of early-migrating neural crest cells because when we disrupt the Eph-ephrin interactions by addition of soluble ephrin-B ligand to trunk explants, early neural crest cells migrate inappropriately into the dorsolateral pathway. Surprisingly, we discovered the ephrin-B ligands continue to be expressed along the dorsolateral pathway during melanoblast migration. RT-PCR analysis, in situ hybridisation, and cell surface-labelling of neural crest cell cultures demonstrate that melanoblasts express several EphB receptors. In adhesion assays, engagement of ephrin-B ligands to EphB receptors increases melanoblast attachment to fibronectin. Cell migration assays demonstrate that ephrin-B ligands stimulate the migration of melanoblasts. Furthermore, when Eph signalling is disrupted in vivo, melanoblasts are prevented from migrating dorsolaterally, suggesting ephrin-B ligands promote the dorsolateral migration of melanoblasts. Thus, transmembrane ephrins act as bifunctional guidance cues: they first repel early migratory neural crest cells from the dorsolateral path, and then later stimulate the migration of melanoblasts into this pathway. The mechanisms by which ephrins regulate repulsion or attraction in neural crest cells are unknown. One possibility is that the cellular response involves signalling to the actin cytoskeleton, potentially involving the activation of Cdc42/Rac family of GTPases. In support of this hypothesis, we show that adhesion of early migratory cells to an ephrin-B-derivatized substratum results in cell rounding and disruption of the actin cytoskeleton, whereas plating of melanoblasts on an ephrin-B substratum induces the formation of microspikes filled with F-actin.


Assuntos
Movimento Celular/fisiologia , Crista Neural/citologia , Crista Neural/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Adesão Celular/fisiologia , Tamanho Celular , Células Cultivadas , Embrião de Galinha , Coturnix/embriologia , Proteínas Fetais/metabolismo , Fibronectinas/metabolismo , Hibridização In Situ , Ligantes , Melanócitos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Receptor EphA4 , Receptores da Família Eph , Receptores Fc/genética , Receptores Fc/metabolismo , Proteínas Recombinantes de Fusão/metabolismo
19.
Mech Dev ; 116(1-2): 173-6, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12128219

RESUMO

To determine the possible involvement of Wnt signaling in eye development, we analyzed the expression patterns of Wnts and Wnt inhibitors in the chicken eye at stage 25, when the first wave of neural crest migration into the cornea begins, and stage 27, just prior to the second wave of neural crest migration. Wnt expression is developmentally regulated in the chicken eye, and antagonists of Wnt signaling are generally expressed in patterns that are temporally distinct from the Wnts.


Assuntos
Olho/embriologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas de Peixe-Zebra , Animais , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Crista Neural/embriologia , Transdução de Sinais , Proteínas Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA