Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 153
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Med Sci ; 15(5): 436-446, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29559832

RESUMO

Idiopathic scoliosis is one of the most common disabling pathologies of children and adolescents. Etiology and pathogenesis of idiopathic scoliosis remain unknown. To study the etiology of this disease we identified the cells' phenotypes in the vertebral body growth plates in patients with idiopathic scoliosis. Materials and methods: The cells were isolated from vertebral body growth plates of the convex and concave sides of the deformity harvested intraoperatively in 50 patients with scoliosis. Cells were cultured and identified by methods of common morphology, neuromorphology, electron microscopy, immunohistochemistry and PCR analysis. Results: Cultured cells of convex side of deformation were identified as chondroblasts. Cells isolated from the growth plates of the concave side of the deformation showed numerous features of neuro- and glioblasts. These cells formed synapses, contain neurofilaments, and expressed neural and glial proteins. Conclusion: For the first time we demonstrated the presence of cells with neural/glial phenotype in the concave side of the vertebral body growth plate in scoliotic deformity. We hypothesized that neural and glial cells observed in the growth plates of the vertebral bodies represent derivatives of neural crest cells deposited in somites due to alterations in their migratory pathway during embryogenesis. We also propose that ectopic localization of cells derived from neural crest in the growth plate of the vertebral bodies is the main etiological factor of the scoliotic disease.


Assuntos
Lâmina de Crescimento/patologia , Crista Neural/patologia , Neuroglia/patologia , Escoliose/patologia , Adolescente , Criança , Condrócitos/metabolismo , Condrócitos/patologia , Condrócitos/ultraestrutura , Desenvolvimento Embrionário/genética , Feminino , Regulação da Expressão Gênica/genética , Lâmina de Crescimento/metabolismo , Lâmina de Crescimento/ultraestrutura , Humanos , Masculino , Microscopia Eletrônica de Varredura , Crista Neural/metabolismo , Crista Neural/ultraestrutura , Neuroglia/metabolismo , Escoliose/etiologia , Escoliose/genética , Coluna Vertebral/metabolismo , Coluna Vertebral/patologia , Coluna Vertebral/ultraestrutura
2.
Methods Mol Biol ; 2179: 107-114, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32939716

RESUMO

Live embryo imaging may provide a wealth of information on intact cell and tissue dynamics, but can be technically challenging to sustain embryo orientation and health for long periods under a microscope. In this protocol, we describe an in vivo method to mount and image cell movements during the epithelial-to-mesenchymal transition (EMT) of neural crest cells within the chick dorsal neural tube. We focus on describing the collection of images and data preparation for image analysis throughout the developmental stages HH15-21 in the chick trunk. Trunk neural crest cell EMT is crucial to development of the peripheral nervous system and pigment cell patterning. The methods we describe may also be applied to other cell and tissue phenomena at various chick developmental stages with some modifications.


Assuntos
Transição Epitelial-Mesenquimal/genética , Imagem Molecular/métodos , Crista Neural/ultraestrutura , Tubo Neural/ultraestrutura , Animais , Movimento Celular/genética , Embrião de Galinha , Tubo Neural/crescimento & desenvolvimento
3.
Methods Mol Biol ; 2179: 79-106, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32939715

RESUMO

The study of cell migration has been greatly enhanced by the development of new model systems and analysis protocols to study this process in vivo. Zebrafish embryos have been a principal protagonist because they are easily accessible, genetically tractable, and optically transparent. Neural crest cells, on the other hand, are the ideal system to study cell migration. These cells migrate extensively, using different modalities of movement and sharing many traits with metastatic cancer cells. In this chapter, we present new tools and protocols that allow the study of NC development and migration in vivo.


Assuntos
Movimento Celular/genética , Biologia Molecular/métodos , Crista Neural/ultraestrutura , Proteínas de Peixe-Zebra/ultraestrutura , Animais , Desenvolvimento Embrionário/genética , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento
4.
Dev Biol ; 324(2): 236-44, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18926812

RESUMO

The induction and migration of neural crest cells (NCCs) are essential to the development of craniofacial structures and the peripheral nervous system. A critical step in the development of NCCs is the epithelial to mesenchymal transition (EMT) that they undergo in order to initiate migration. Several transcription factors are important for the NCC EMT. However, less is known about the effectors regulating changes in cell adhesion, the cytoskeleton, and cell motility associated with the EMT or about specific changes in the behavior of cells undergoing EMT in vivo. We used time-lapse imaging of NCCs in the zebrafish hindbrain to show that NCCs undergo a stereotypical series of behaviors during EMT. We find that loss of cell adhesion and membrane blebbing precede filopodial extension and the onset of migration. Live imaging of actin dynamics shows that actin localizes differently in blebs and filopodia. Moreover, we find that disruption of myosin II or Rho-kinase (ROCK) activity inhibits NCC blebbing and causes reduced NCC EMT. These data reveal roles for myosin II and ROCK in NCC EMT in vivo, and provide a detailed characterization of NCC behavior during EMT that will form a basis for further mechanistic studies.


Assuntos
Diferenciação Celular , Miosina Tipo II/metabolismo , Crista Neural/embriologia , Peixe-Zebra/embriologia , Quinases Associadas a rho/metabolismo , Actinas/metabolismo , Animais , Animais Geneticamente Modificados/embriologia , Animais Geneticamente Modificados/metabolismo , Adesão Celular , Membrana Celular/metabolismo , Movimento Celular , Citocinese , Epitélio/embriologia , Mesoderma/embriologia , Crista Neural/ultraestrutura , Pseudópodes/fisiologia , Rombencéfalo/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
5.
J Cell Biol ; 98(6): 1947-60, 1984 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-6373786

RESUMO

In the trunk region of avian embryos, neural crest cells migrate along two pathways: dorsally just under the ectoderm, and ventrally between the neural tube and the somites. Previous work from this laboratory has shown that uncoated latex beads are able to translocate along the ventral neural crest pathway after injection into young embryos; however, beads coated with fibronectin are restricted from the ventral route ( Bronner -Fraser, M.E., 1982, Dev. Biol., 91: 50-63). Here, we extend these observations to determine the effects of other macromolecules on bead distribution. The data show that laminin-coated beads, like fibronectin-coated beads, are restricted from the ventral pathway. In contrast, beads coated with type I collagen translocate ventrally after injection. Because macromolecules have characteristic charge properties, changes in surface charge caused by coating the beads may confound interpretation of the results. Electrostatic effects on bead movement were examined by coating the latex beads with polyamino acids in order to predictably alter the initial surface charge. The surface charge before injection was measured for beads coated with amino acid polymers or with various biologically important macromolecules; the subsequent translocation ability of these beads was then monitored in the embryo. Polylysine-coated beads (positively charged) were restricted from the ventral pathway as were fibronectin and laminin-coated beads, even though fibronectin and laminin beads were both negatively charged. In contrast, polytyrosine -coated beads ( neutrally charged) translocated ventrally as did negatively charged collagen-coated or uncoated beads. The results demonstrate that no correlation exists between the charge properties on the latex bead surface and their subsequent ability to translocate along the ventral pathway. Therefore, an adhesion mechanism independent of surface charge effects must explain the restriction or translocation of latex beads on a neural crest pathway.


Assuntos
Colágeno/fisiologia , Laminina/fisiologia , Crista Neural/fisiologia , Animais , Embrião de Galinha , Imunofluorescência , Látex , Microinjeções , Microscopia Eletrônica de Varredura , Microesferas , Crista Neural/ultraestrutura , Polilisina
6.
J Cell Biol ; 102(5): 1917-30, 1986 May.
Artigo em Inglês | MEDLINE | ID: mdl-2422181

RESUMO

The sequential appearance of the extracellular matrix (ECM) protein, cytotactin, was examined during development of the chicken embryo by immunohistochemical techniques. Although cytotactin was identified as a molecule that mediates glia-neuron interactions, preliminary immunohistochemical localization of the molecule suggested that it was an ECM protein with a widespread but nonetheless more restricted distribution than either fibronectin or laminin. In the present study, it was found that cytotactin is first present in the gastrulating chicken embryo. It appears later in the basement membrane of the developing neural tube and notochord in a temporal sequence beginning in the cephalic regions and proceeding caudally. Between 2 and 3 d of development, the molecule is present at high levels in the early neural crest pathways (surrounding the neural tube and somites) but, in contrast to fibronectin and laminin, is not found in the lateral plate mesoderm or ectoderm. At later times, cytotactin is expressed extensively in the central nervous system, in lesser amounts in the peripheral nervous system, and in a number of nonneural sites, most prominently in all smooth muscles and in basement membranes of lung and kidney. Cytotactin appears in adult tissues with distributions that are similar to those seen in embryonic tissues. The findings raise the possibility that certain ECM proteins contribute to pattern formation in embryogenesis as a result of their restricted expression in a spatiotemporally regulated fashion at some sites but not at others.


Assuntos
Matriz Extracelular/fisiologia , Glicoproteínas/metabolismo , Sistema Nervoso/embriologia , Fatores Etários , Animais , Membrana Basal/fisiologia , Membrana Basal/ultraestrutura , Adesão Celular , Movimento Celular , Embrião de Galinha , Matriz Extracelular/ultraestrutura , Fibronectinas/metabolismo , Imunofluorescência , Gástrula/ultraestrutura , Laminina/metabolismo , Morfogênese , Crista Neural/ultraestrutura , Tenascina , Distribuição Tecidual
7.
Methods Mol Biol ; 1976: 207-221, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30977076

RESUMO

One of the early, profound insights regarding the biology of the neural crest was the observation of its contribution to the skeletal structures of the cranium and jaws. The critical nature of these structures made the comparative analysis of the cranial neural crest and its derived structures essential investigative aims toward our understanding of the development and evolution of vertebrates and vertebrate-specific structures. Though classically applied to a relatively wide range of taxa in the nineteenth and early twentieth centuries, the application of traditional methodologies for complex comparative developmental and anatomical analyses subsequently become more limited by their time-consuming nature, resource scarcity, and a greater emphasis on the genetic and molecular regulation of patterning and morphogenesis in a select number of tractable model organisms. Recently, however, this trend has been reversed, and the value of genetic and molecular-based questions applied to non-model (unconventional) vertebrate organisms has been re-appreciated. This is particularly true of comparative investigations of cranial neural crest biology. Herein, we present methodologies for the analysis of the cranial neural crest and its structural derivatives employable in modern investigations of both model and unconventional vertebrate organisms.


Assuntos
Crista Neural/citologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Microscopia Eletrônica de Varredura , Crista Neural/ultraestrutura , Esqueleto/citologia , Esqueleto/ultraestrutura , Vertebrados
8.
Mol Neurobiol ; 55(4): 3523-3536, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28509082

RESUMO

Autophagy plays a very important role in numerous physiological and pathological events. However, it still remains unclear whether Atg7-induced autophagy is involved in the regulation of neural crest cell production. In this study, we found the co-location of Atg7 and Pax7+ neural crest cells in early chick embryo development. Upregulation of Atg7 with unilateral transfection of full-length Atg7 increased Pax7+ and HNK-1+ cephalic and trunk neural crest cell numbers compared to either Control-GFP transfection or opposite neural tubes, suggesting that Atg7 over-expression in neural tubes could enhance the production of neural crest cells. BMP4 in situ hybridization and p-Smad1/5/8 immunofluorescent staining demonstrated that upregulation of Atg7 in neural tubes suppressed the BMP4/Smad signaling, which is considered to promote the delamination of neural crest cells. Interestingly, upregulation of Atg7 in neural tubes could significantly accelerate cell progression into the S phase, implying that Atg7 modulates cell cycle progression. However, ß-catenin expression was not significantly altered. Finally, we demonstrated that upregulation of the Atg7 gene could activate autophagy as did Atg8. We have also observed that similar phenotypes, such as more HNK-1+ neural crest cells in the unilateral Atg8 transfection side of neural tubes, and the transfection with full-length Atg8-GFP certainly promote the numbers of BrdU+ neural crest cells in comparison to the GFP control. Taken together, we reveal that Atg7-induced autophagy is involved in regulating the production of neural crest cells in early chick embryos through the modification of the cell cycle.


Assuntos
Proteína 7 Relacionada à Autofagia/metabolismo , Autofagia , Crista Neural/citologia , Neurogênese , Animais , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Proteína 7 Relacionada à Autofagia/genética , Família da Proteína 8 Relacionada à Autofagia/genética , Família da Proteína 8 Relacionada à Autofagia/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Modelos Biológicos , Crista Neural/metabolismo , Crista Neural/ultraestrutura , Tubo Neural/citologia , Tubo Neural/embriologia , Tubo Neural/metabolismo , Tubo Neural/ultraestrutura , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
9.
J Histochem Cytochem ; 64(7): 412-24, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27199282

RESUMO

The appropriate projection of axons within the nervous system is a crucial component of the establishment of neural circuitry. Draxin is a repulsive axon guidance protein. Draxin has important functions in the guidance of three commissures in the central nervous system and in the migration of neural crest cells and dI3 interneurons in the chick spinal cord. Here, we report that the distribution of the draxin protein and the location of 23C10-positive areas have a strong temporal and spatial correlation. The overexpression of draxin, especially transmembrane draxin, caused 23C10-positive axon bundles to misproject in the dorsal hindbrain. In addition, the overexpression of transmembrane draxin caused abnormal formation of the ganglion crest of the IX and X cranial nerves, misprojection of some anti-human natural killer-1 (HNK-1)-stained structures in the dorsal roof of the hindbrain, and a simultaneous reduction in the efferent nerves of some motoneuron axons inside the hindbrain. Our data reveal that draxin might be involved in the fascicular projection of cranial nerves in the hindbrain.


Assuntos
Axônios/fisiologia , Nervos Cranianos/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Rombencéfalo/fisiologia , Animais , Embrião de Galinha , Vias Eferentes/fisiologia , Nervo Glossofaríngeo/fisiologia , Neurônios Motores/fisiologia , Crista Neural/fisiologia , Crista Neural/ultraestrutura , Nervo Vago/fisiologia
10.
Sci Rep ; 6: 20927, 2016 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-26887292

RESUMO

Neural crest cells (NCCs) are a population of multipotent cells that migrate extensively during vertebrate development. Alterations to neural crest ontogenesis cause several diseases, including cancers and congenital defects, such as Hirschprung disease, which results from incomplete colonization of the colon by enteric NCCs (ENCCs). We investigated the influence of the stiffness and structure of the environment on ENCC migration in vitro and during colonization of the gastrointestinal tract in chicken and mouse embryos. We showed using tensile stretching and atomic force microscopy (AFM) that the mesenchyme of the gut was initially soft but gradually stiffened during the period of ENCC colonization. Second-harmonic generation (SHG) microscopy revealed that this stiffening was associated with a gradual organization and enrichment of collagen fibers in the developing gut. Ex-vivo 2D cell migration assays showed that ENCCs migrated on substrates with very low levels of stiffness. In 3D collagen gels, the speed of the ENCC migratory front decreased with increasing gel stiffness, whereas no correlation was found between porosity and ENCC migration behavior. Metalloprotease inhibition experiments showed that ENCCs actively degraded collagen in order to progress. These results shed light on the role of the mechanical properties of tissues in ENCC migration during development.


Assuntos
Movimento Celular/fisiologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/ultraestrutura , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/ultraestrutura , Crista Neural/embriologia , Crista Neural/ultraestrutura , Animais , Embrião de Galinha , Colagenases/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestrutura , Camundongos , Microscopia de Força Atômica
11.
Anat Rec A Discov Mol Cell Evol Biol ; 282(2): 130-7, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15627983

RESUMO

In vertebrate embryos, neural crest cells emigrate out of the neural tube and contribute to the formation of a variety of neural and nonneural tissues. Some neural crest cells undergo apoptotic death during migration, but its biological significance and the underlying mechanism are not well understood. We carried out an in vitro study to examine how the morphology and survival of cranial neural crest (CNC) cells of the mouse embryo are affected when their actin cytoskeleton or anchorage-dependent cell spreading is perturbed. Disruption of actin fiber organization by cytochalasin D (1 microg/ml) and inhibition of cell attachment by matrix metalloproteinase-2 (MMP-2; 2.0 units/ml) were followed by morphologic changes and apoptotic death of cultured CNC cells. When the actin cytoskeleton was disrupted by cytochalasin D, the morphologic changes of cultured CNC cells preceded DNA fragmentation. These results indicate that the maintenance of cytoskeleton and anchorage-dependent cell spreading are required for survival of CNC cells. The spatially and temporally regulated expression of proteinases may be essential for the differentiation and migration of neural crest cells.


Assuntos
Actinas/metabolismo , Apoptose , Adesão Celular , Citoesqueleto/ultraestrutura , Crista Neural/citologia , Animais , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Citocalasina D/farmacologia , Citoesqueleto/efeitos dos fármacos , Feminino , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos ICR , Crista Neural/efeitos dos fármacos , Crista Neural/ultraestrutura , Inibidores da Síntese de Ácido Nucleico/farmacologia
12.
Int J Dev Biol ; 47(7-8): 541-53, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14756330

RESUMO

The vertebrate head is a complex assemblage of cranial specializations, including the central and peripheral nervous systems, viscero- and neurocranium, musculature and connective tissue. The primary differences that exist between vertebrates and other chordates relate to their craniofacial organization. Therefore, evolution of the head is considered fundamental to the origins of vertebrates (Gans and Northcutt, 1983). The transition from invertebrate to vertebrate chordates was a multistep process, involving the formation and patterning of many new cell types and tissues. The evolution of early vertebrates, such as jawless fish, was accompanied by the emergence of a specialized set of cells, called neural crest cells which have long held a fascination for developmental and evolutionary biologists due to their considerable influence on the complex development of the vertebrate head. Although it has been classically thought that protochordates lacked neural crest counterparts, the recent identification and characterization of amphioxus and ascidian genes homologous to those involved in vertebrate neural crest development challenges this idea. Instead it suggests thatthe neural crest may not be a novel vertebrate cell population, but could have in fact originated from the protochordate dorsal midline epidermis. Consequently, the evolution of the neural crest cells could be reconsidered in terms of the acquisition of new cell properties such as delamination-migration and also multipotency which were key innovations that contributed to craniofacial development. In this review we discuss recent findings concerning the inductive origins of neural crest cells, as well as new insights into the mechanisms patterning this cell population and the subsequent influence this has had on craniofacial evolution.


Assuntos
Evolução Biológica , Face/embriologia , Cabeça/embriologia , Arcada Osseodentária/embriologia , Crista Neural/embriologia , Animais , Movimento Celular , Variação Genética , Arcada Osseodentária/citologia , Modelos Biológicos , Crista Neural/ultraestrutura , Plasticidade Neuronal/genética , Vertebrados/embriologia , Vertebrados/genética
13.
Int J Dev Biol ; 34(3): 351-64, 1990 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-2268541

RESUMO

Initial development of astroglial phenotype has been studied in vitro in an amphibian embryo (Pleurodeles waltI), to document the differentiation potentialities acquired by neural precursor cells isolated at the early neurula stage. In particular, we sought to determine whether interactions between neuroepithelial cells and the inducing tissue, the chordamesoderm, are required beyond this stage to specify precursor cells along glial lineages. Glial cell differentiation was documented by examining the appearance of glial fibrillary acidic protein (GFAp), a specific marker of astroglial lineages. Cells expressing GFAp-immunoreactivity differentiated rapidly, after 48 hours of culture, from cultivated neural plate cells, irrespective of the presence or absence of the inducing tissue. The widespread expression of Pleurodeles GFAp protein in neural plate cultures, in which CNS precursor cells develop alone in a simple saline medium, showed that prolonged contact with chordamesodermal cells was not necessary for the emergence of the astroglial phenotype. In addition, the initial development of astroglial phenotype has been defined in vivo. The first detectable GFAp-immunoreactivity was visualized in the neural tube of stage-24 embryos, a stage corresponding to 2-3 days in culture, defining radial glial cell end-feet. Thus, dissociation and culture of neural precursor cells did not appear to modify the onset of astroglial differentiation. At stage 32, GFAp-immunoreactivity was observed over the entire length of radial glial fibers and was also evidenced in mitotic cells located in the ventricular zone, suggesting that radial glial cells were not all post-mitotic.


Assuntos
Astrócitos/fisiologia , Neurônios/fisiologia , Pleurodeles/embriologia , Animais , Especificidade de Anticorpos , Astrócitos/imunologia , Astrócitos/ultraestrutura , Biomarcadores , Diferenciação Celular , Epitélio/fisiologia , Epitélio/ultraestrutura , Proteína Glial Fibrilar Ácida/imunologia , Técnicas In Vitro , Filamentos Intermediários/imunologia , Filamentos Intermediários/fisiologia , Filamentos Intermediários/ultraestrutura , Mitose , Crista Neural/fisiologia , Crista Neural/ultraestrutura , Neurônios/imunologia , Neurônios/ultraestrutura
14.
Int J Dev Biol ; 35(4): 437-52, 1991 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-1724907

RESUMO

We have investigated the distribution of type I collagen, tenascin, and laminin in younger chick embryos than have previously been studied in detail. The initial appearance of type I collagen, but not tenascin and laminin, is exactly correlated with the beginning of neural crest migration, suggesting a role for collagen I in the migration. Light microscopy of whole mounts of 2-day-old chick embryos reveals that type I collagen is expressed in a rostral to caudal gradient; it localizes to the notochord sheath before accumulating around the neural tube and somites. Collagen I and tenascin also associate with central somite cells. Surprisingly, no extracellular matrix can be detected among the early sclerotomal cells, which suggests that little or no cell migration is involved in this epithelial-mesenchymal transformation. Electron microscopy using peroxidase antiperoxidase reveals that tenascin is present in nonstriated, 10 nm wide fibrils and in interstitial bodies, both of which have previously been reported to contain fibronectin. However, collagen I only occurs in the 10 nm fibrils and larger striated fibrils. This is the first ultrastructural study to assign tenascin to fibrils and interstitial bodies and to describe its appearance and disappearance from embryonic basement membranes. The discussion emphasizes the possible importance of type I collagen in neural crest cell migration and compares the ultrastructural associations of the ECM molecules present at this early embryonic stage.


Assuntos
Moléculas de Adesão Celular Neuronais/análise , Colágeno/análise , Proteínas da Matriz Extracelular/análise , Crista Neural/embriologia , Animais , Membrana Basal/química , Movimento Celular , Embrião de Galinha , Imuno-Histoquímica , Laminina/análise , Crista Neural/química , Crista Neural/ultraestrutura , Notocorda/química , Tenascina
15.
Integr Biol (Camb) ; 7(3): 356-63, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25710133

RESUMO

AFM-based force spectroscopy in combination with optical microscopy is a powerful tool for investigating cell mechanics and adhesion on the single cell level. However, standard setups featuring an AFM mounted on an inverted light microscope only provide a bottom view of cell and AFM cantilever but cannot visualize vertical cell shape changes, for instance occurring during motile membrane blebbing. Here, we have integrated a mirror-based sideview system to monitor cell shape changes resulting from motile bleb behavior of Xenopus cranial neural crest (CNC) cells during AFM elasticity and adhesion measurements. Using the sideview setup, we quantitatively investigate mechanical changes associated with bleb formation and compared cell elasticity values recorded during membrane bleb and non-bleb events. Bleb protrusions displayed significantly lower stiffness compared to the non-blebbing membrane in the same cell. Bleb stiffness values were comparable to values obtained from blebbistatin-treated cells, consistent with the absence of a functional actomyosin network in bleb protrusions. Furthermore, we show that membrane blebs forming within the cell-cell contact zone have a detrimental effect on cell-cell adhesion forces, suggesting that mechanical changes associated with bleb protrusions promote cell-cell detachment or prevent adhesion reinforcement. Incorporating a sideview setup into an AFM platform therefore provides a new tool to correlate changes in cell morphology with results from force spectroscopy experiments.


Assuntos
Extensões da Superfície Celular/fisiologia , Extensões da Superfície Celular/ultraestrutura , Fluidez de Membrana/fisiologia , Microscopia de Força Atômica/instrumentação , Crista Neural/fisiologia , Crista Neural/ultraestrutura , Animais , Adesão Celular/fisiologia , Células Cultivadas , Módulo de Elasticidade/fisiologia , Desenho de Equipamento , Análise de Falha de Equipamento , Dureza/fisiologia , Micromanipulação/instrumentação , Micromanipulação/métodos , Microscopia de Força Atômica/métodos , Estresse Mecânico , Xenopus laevis
16.
J Invest Dermatol ; 112(5): 796-801, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10233774

RESUMO

Previous findings indicate that the protein c-KIT and its ligand, stem cell factor (SCF) play a crucial role in the development of melanocytes from their precursors in the embryonic neural crest cells. Using a monoclonal anti-c-KIT antibody, ACK2, which is an antagonistic blocker of c-KIT function, we and colleagues demonstrated that mouse melanocytes disappeared with the injection of ACK2 during certain periods of embryonic and postnatal life. The precise mechanisms of this disappearance, however, remain unclear. Because melanocytes disappeared without any inflammation in these in vivo studies, we suspect that apoptosis was a main cause of their disappearance. In this study, to clarify the underlying mechanism, we studied whether ACK2 induces apoptosis in c-KIT-positive melanoblasts, which appear in mouse neural crest cells cultured with SCF from 9.5 d old mouse embryos. With an in situ apoptosis detection kit, a significant increase in apoptosis was detected after the removal of SCF, which further increased with the addition of ACK2 during SCF-dependent periods. The occurrence of apoptosis in the cultured cells was also demonstrated by a DNA analysis and electron microscopy. Immunohistochemical double staining confirmed that the apoptotic cells were c-KIT positive, and the electron microscopy showed that these apoptotic cells were melanocyte precursors. It was therefore demonstrated that apoptosis was induced in the SCF-dependent c-KIT-positive melanocytes in vitro when the SCF/c-KIT interaction was obstructed. These findings elucidate the mechanism of the regulation of melanocyte development, and the survival and proliferation of these precursor cells, by SCF/c-KIT interaction.


Assuntos
Anticorpos Monoclonais/farmacologia , Apoptose , Melanócitos/citologia , Crista Neural/citologia , Proteínas Proto-Oncogênicas c-kit/fisiologia , Fator de Células-Tronco/fisiologia , Células-Tronco/citologia , Animais , Ligação Competitiva , Células Cultivadas , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Imuno-Histoquímica , Melanócitos/efeitos dos fármacos , Melanócitos/metabolismo , Melanócitos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Crista Neural/efeitos dos fármacos , Crista Neural/metabolismo , Crista Neural/ultraestrutura , Proteínas Proto-Oncogênicas c-kit/imunologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura
17.
J Neuropathol Exp Neurol ; 47(6): 609-17, 1988 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3171606

RESUMO

Closure of the posterior neuropore was analyzed by means of ultrastructural cytochemistry in ten-day dysraphic mouse embryos homozygous for the mutant gene vl, and comparisons were made with normal embryos in terms of convergence, apposition and fusion of the apices of the neural folds. In abnormal embryos, regional differences in the distribution of the surface coat were comparable to those in normal embryos. However, there was an abnormally acute medial bending of the neural folds, as well as a delay in closure of the posterior neuropore. In closed areas of the abnormal embryos the dorsum also showed an erratic knot of disorganized cells. Thus, the pathogenetic mechanism in this mutant appears to involve not only a failure in apposition in open areas, as well as an inappropriate association of cells in areas which do fuse, but possibly also a failure of proper alignment of neural fold apices prior to apposition and fusion.


Assuntos
Camundongos Mutantes Neurológicos/embriologia , Crista Neural/citologia , Animais , Autorradiografia , Feminino , Camundongos , Microscopia Eletrônica , Crista Neural/ultraestrutura , Gravidez , Valores de Referência , Timidina/metabolismo , Trítio
18.
Int J Biochem Cell Biol ; 36(6): 1135-9, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15094128

RESUMO

Neural crest cells escape the neural tube by undergoing an epithelial to mesenchymal transition (EMT). This is followed by extensive migration along specific pathways that are lined with extracellular matrix (ECM). In this study, we have examined the roles of matrix receptors containing beta1 integrin subunits in neural crest cell morphogenesis using antisense morpholino oligos electroporated in ovo into avian neural crest cell precursors. Our results show that reduced levels of expression of beta1 integrin subunits in the dorsal neural tube results in an abnormal epithelial to mesenchymal transition. In approximately half of the experimental embryos, however, some neural crest cells filled with beta1 antisense are able to escape the neural tube and migrate ventrally, indicating that grossly normal migration of trunk neural crest cells can take place after beta1 integrin expression is reduced. This study shows the potential of this novel method for investigating the roles of genes that are required for the survival of early mouse embryos in later development events.


Assuntos
Integrina beta1/fisiologia , Crista Neural/embriologia , Animais , Embrião de Galinha , Integrina beta1/análise , Integrina beta1/genética , Crista Neural/anormalidades , Crista Neural/ultraestrutura , Interferência de RNA , RNA Antissenso/genética
19.
Endocrinology ; 131(5): 2090-6, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1330489

RESUMO

The expression and structure of the receptors for neuropeptide-Y (NPY) and peptide-YY (PYY) were studied in 16 human and rodent tumor cell lines derived from the neural crest by ligand binding and cross-linking techniques using [125I]Bolton-Hunter-NPY, [125I]PYY, and various forms of monoiodinated NPY and PYY. Although NPY-binding sites were observed in most of the tumor cells, PYY-binding sites were found only on the human neuroblastoma cell lines SMS-MSN, SMS-KAN, SK-N-MC, and MC-IXC and the human Ewing's sarcoma cell line SK-ES. The differential labeling of the NPY/PYY receptors on these cell lines suggests that the NPY/PYY receptors are more heterogeneous than previously described as the Y1, Y2, and Y3 receptor subtypes. Cross-linking studies demonstrate that the Y1 and Y2 receptors for NPY/PYY are structurally different (mol wt, 70 and 50 kilodaltons, respectively) and that the 70- and 50-kilodalton receptor proteins are coexpressed in certain tumor cell lines. This could explain at least in part why cell lines show a relative specificity for Y1/Y2 classification, observed as the inhibition by both C-terminal fragments and Y1-specific analogs on the NPY/PYY binding to membrane receptors. Collectively, the present study suggests further heterogeneity of the NPY/PYY receptors and the existence of multiple receptor proteins in the tumor cell lines derived from the neural crest.


Assuntos
Crista Neural/química , Peptídeos/metabolismo , Receptores de Neuropeptídeo Y/análise , Receptores de Neurotransmissores/análise , Animais , Glioma/química , Glioma/patologia , Glioma/ultraestrutura , Humanos , Radioisótopos do Iodo , Ligantes , Crista Neural/patologia , Crista Neural/ultraestrutura , Neuroblastoma/química , Neuroblastoma/patologia , Neuroblastoma/ultraestrutura , Células PC12 , Peptídeo YY , Ratos , Células Tumorais Cultivadas
20.
J Comp Neurol ; 222(4): 496-505, 1984 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-6699215

RESUMO

The origin of neural crest cells was studied histologically and ultrastructurally in caudal regions of mouse embryos at 8.5-11 days of gestation (day of vaginal plug = day 0). The neural tube of caudal regions develops in two different phases, called primary and secondary neurulation. The primary (more cranial) portion of the neural tube originates from the ectodermal neural plate, whereas the secondary (most caudal) portion originates from the tail bud. We asked in this study: Do neural crest cells of caudal regions originate exclusively from the developing primary portion of the neural tube and, subsequently, migrate into areas undergoing secondary neurulation; or do some of these cells originate from the tail bud, with the secondary portion of the neural tube, forming in situ in caudal areas? Most embryos were preserved with fixative containing cetylpyridinium chloride (CPC) to facilitate the identification of neural crest cells, which stain darkly after exposure to CPC during fixation. Our results suggest that there are two sources of neural crest cells in caudal regions: the neuroectoderm of the neural folds flanking the closing posterior neuropore, and the tail bud. Neural crest cells derived from the neuroectoderm are designated as the primary neural crest, because they form in conjunction with the primary portion of the neural tube during primary neurulation, whereas neural crest cells derived from the tail bud are designated as the secondary neural crest, because they form in conjunction with the secondary portion of the neural tube during secondary neurulation.


Assuntos
Crista Neural/fisiologia , Animais , Embrião de Mamíferos , Feminino , Idade Gestacional , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Microscopia Eletrônica , Microscopia Eletrônica de Varredura , Crista Neural/citologia , Crista Neural/ultraestrutura , Gravidez
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa