Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Dev Biol ; 464(1): 71-87, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32320685

RESUMO

Animal development and homeostasis depend on precise temporal and spatial intercellular signaling. Components shared between signaling pathways, generally thought to decrease specificity, paradoxically can also provide a solution to pathway coordination. Here we show that the Bone Morphogenetic Protein (BMP) and Wnt signaling pathways share Apcdd1 as a common inhibitor and that Apcdd1 is a taxon-restricted gene with novel domains and signaling functions. Previously, we showed that Apcdd1 inhibits Wnt signaling (Shimomura et al., 2010), here we find that Apcdd1 potently inhibits BMP signaling in body axis formation and neural differentiation in chicken, frog, zebrafish. Furthermore, we find that Apcdd1 has an evolutionarily novel protein domain. Our results from experiments and modeling suggest that Apcdd1 may coordinate the outputs of two signaling pathways that are central to animal development and human disease.


Assuntos
Padronização Corporal , Proteínas Morfogenéticas Ósseas/metabolismo , Embrião não Mamífero/embriologia , Glicoproteínas de Membrana/metabolismo , Via de Sinalização Wnt , Proteínas de Xenopus/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/genética , Glicoproteínas de Membrana/genética , Domínios Proteicos , Proteínas de Xenopus/genética , Xenopus laevis
2.
J Neurosci ; 39(47): 9316-9327, 2019 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-31578231

RESUMO

Regenerating axons often have to grow considerable distances to reestablish circuits, making functional recovery a lengthy process. One solution to this problem would be to co-opt the "temporal" guidance mechanisms that control the rate of axon growth during development to accelerate the rate at which nerves regenerate in adults. We have previously found that the loss of Limk1, a negative regulator of cofilin, accelerates the rate of spinal commissural axon growth. Here, we use mouse models to show that spinal motor axon outgrowth is similarly promoted by the loss of Limk1, suggesting that temporal guidance mechanisms are widely used during development. Furthermore, we find that the regulation of cofilin activity is an acute response to nerve injury in the peripheral nervous system. Within hours of a sciatic nerve injury, the level of phosphorylated cofilin dramatically increases at the lesion site, in a Limk1-dependent manner. This response may be a major constraint on the rate of peripheral nerve regeneration. Proof-of-principle experiments show that elevating cofilin activity, through the loss of Limk1, results in faster sciatic nerve growth, and improved recovery of some sensory and motor function.SIGNIFICANCE STATEMENT The studies shed light on an endogenous, shared mechanism that controls the rate at which developing and regenerating axons grow. An understanding of these mechanisms is key for developing therapies to reduce painful recovery times for nerve-injury patients, by accelerating the rate at which damaged nerves reconnect with their synaptic targets.


Assuntos
Fatores de Despolimerização de Actina/metabolismo , Axônios/fisiologia , Crescimento Celular , Quinases Lim/metabolismo , Neurônios Motores/fisiologia , Regeneração Nervosa/fisiologia , Fatores de Despolimerização de Actina/genética , Animais , Feminino , Quinases Lim/deficiência , Quinases Lim/genética , Masculino , Camundongos , Camundongos Transgênicos , Neurônios Motores/química , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Transdução de Sinais/fisiologia
3.
Neuron ; 94(4): 790-799.e3, 2017 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-28434801

RESUMO

Netrin1 has been proposed to act from the floor plate (FP) as a long-range diffusible chemoattractant for commissural axons in the embryonic spinal cord. However, netrin1 mRNA and protein are also present in neural progenitors within the ventricular zone (VZ), raising the question of which source of netrin1 promotes ventrally directed axon growth. Here, we use genetic approaches in mice to selectively remove netrin from different regions of the spinal cord. Our analyses show that the FP is not the source of netrin1 directing axons to the ventral midline, while local VZ-supplied netrin1 is required for this step. Furthermore, rather than being present in a gradient, netrin1 protein accumulates on the pial surface adjacent to the path of commissural axon extension. Thus, netrin1 does not act as a long-range secreted chemoattractant for commissural spinal axons but instead promotes ventrally directed axon outgrowth by haptotaxis, i.e., directed growth along an adhesive surface.


Assuntos
Orientação de Axônios/genética , Axônios/metabolismo , Fatores de Crescimento Neural/genética , Células-Tronco Neurais/metabolismo , Medula Espinal/embriologia , Proteínas Supressoras de Tumor/genética , Animais , Axônios/ultraestrutura , Fatores Quimiotáticos/genética , Fatores Quimiotáticos/metabolismo , Imageamento Tridimensional , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Microscopia Confocal , Fatores de Crescimento Neural/metabolismo , Netrina-1 , Neurogênese/genética , RNA Mensageiro/metabolismo , Medula Espinal/ultraestrutura , Proteínas Supressoras de Tumor/metabolismo
4.
PLoS One ; 8(4): e62977, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23646165

RESUMO

Axons use temporal and directional guidance cues at intermediate targets to set the rate and direction of growth towards their synaptic targets. Our recent studies have shown that disrupting the temporal guidance process, by unilaterally accelerating the rate at which spinal dI1 (commissural) axons grow, resulted in turning errors both in the ventral spinal cord and after crossing the floor plate. Here we investigate a mechanistic explanation for these defects: the accelerated dI1 axons arrive in the ventral spinal cord before necessary fasciculation cues from incoming dI1 axons from the opposite side of the spinal cord. The identification of such an interaction would support a model of selective fasciculation whereby the pioneering dI1 axons serve as guides for the processes of the bilaterally symmetrical population of dI1 neurons. To test this model, we first developed the ability to "double" in ovo electroporate the embryonic chicken spinal cord to independently manipulate the rate of growth of the two bilateral populations of dI1 axons. Second, we examined the requirement for a putative bilateral interaction by unilaterally ablating the dI1 population in cultured explants of chicken embryonic spinal cord. Surprisingly, we find no evidence for a bilateral dI1 axon interaction, rather dI1 axons appear to project independently of each other.


Assuntos
Células do Corno Anterior/citologia , Células do Corno Anterior/fisiologia , Axônios/fisiologia , Animais , Embrião de Galinha , Galinhas , Eletroporação/métodos , Fenótipo , Coloração e Rotulagem/métodos
5.
PLoS One ; 6(7): e22072, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21779375

RESUMO

Dcc is the key receptor that mediates attractive responses of axonal growth cones to netrins, a family of axon guidance cues used throughout evolution. However, a Dcc homolog has not yet been identified in the chicken genome, raising the possibility that Dcc is not present in avians. Here we show that the closely related family member neogenin may functionally substitute for Dcc in the developing chicken spinal cord. The expression pattern of chicken neogenin in the developing spinal cord is a composite of the distribution patterns of both rodent Dcc and neogenin. Moreover, whereas the loss of mouse neogenin has no effect on the trajectory of commissural axons, removing chicken neogenin by RNA interference results in a phenotype similar to the functional inactivation of Dcc in mouse. Taken together, these data suggest that the chick neogenin is functionally equivalent to rodent Dcc.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/metabolismo , Animais , Galinhas , Receptor DCC , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Membrana/genética , Camundongos , Interferência de RNA , Receptores de Superfície Celular/genética , Medula Espinal/embriologia , Medula Espinal/metabolismo , Proteínas Supressoras de Tumor/genética
6.
J Neurosci ; 30(46): 15430-40, 2010 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-21084599

RESUMO

Commissural spinal axons extend away from the roof plate (RP) in response to a chemorepellent mediated by the bone morphogenetic proteins (BMPs). Previous studies have focused on the ability of commissural axons to translate a spatial gradient of BMPs into directional information in vitro. However, a notable feature of this system in vivo is that the gradient of BMPs is thought to act from behind the commissural cell bodies, making it possible for the BMPs to have a continued effect on commissural axons as they grow away from the RP. Here, we demonstrate that BMPs activate the cofilin regulator Lim domain kinase 1 (Limk1) to control the rate of commissural axon extension in the dorsal spinal cord. By modulating Limk1 activity in both rodent and chicken commissural neurons, the rate of axon growth can either be stalled or accelerated. Altering the activation state of Limk1 also influences subsequent guidance decisions: accelerated axons make rostrocaudal projection errors while navigating their intermediate target, the floor plate. These results suggest that guidance cues can specify information about the rate of growth, to ensure that axons reach subsequent signals either at particular times or speeds during development.


Assuntos
Axônios/fisiologia , Proteínas Morfogenéticas Ósseas/fisiologia , Quinases Lim/fisiologia , Animais , Células COS , Células Cultivadas , Embrião de Galinha , Chlorocebus aethiops , Camundongos , Vias Neurais/crescimento & desenvolvimento , Ratos , Fatores de Tempo
7.
J Vis Exp ; (37)2010 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-20212425

RESUMO

Dorsal commissural axons in the vertebrate spinal cord(1) have been an invaluable model system in which to identify axon guidance signals. Here, we describe an in vitro assay, "the reorientation assay", that has been used extensively to study the effect of extrinsic and intrinsic signals on the orientation of commissural axons(2). This assay was developed by numerous people in the laboratories of Jane Dodd, Thomas Jessell and Andrew Lumsden (see acknowledgements for more details) and versions of this assay were used to demonstrate the reorientation activities of key axon guidance molecules, including the BMP chemorepellent in the roof plate(3,4) and the chemoattractive activities of Netrin1(5) and Sonic Hedgehog (Shh)(6) in the floor plate in the spinal cord. Explants comprising 2-3 segments of the dorsal two-thirds of spinal cord are dissected from embryonic day (E) 11 rats and cultured in three dimensional collagen gels(7). E11 dorsal spinal explants contain newly born commissural neurons, which can be identified by their axonal expression of the glycoprotein, Tag1(8). Over the course of 30-40 hours in culture, the commissural axon trajectory is recapitulated in these dorsal explants with a time course similar to that seen in vivo. This axonal trajectory can be challenged by placing either test tissues or a COS cell aggregate expressing a candidate signaling molecule in contact with one of the lateral edges of the dorsal explant. Commissural axons extending in the vicinity of the appended tissue will grow under the influence of both the endogenous roof plate and signals from the ectopic lateral tissue. The degree to which commissural axons are reoriented under these circumstances can be quantified. Using this assay, it is possible both to examine the sufficiency of a particular signal to reorient commissural axons(3,4) as well the necessity for this signal to direct the commissural trajectory(9).


Assuntos
Axônios/fisiologia , Medula Espinal/citologia , Animais , Axônios/efeitos dos fármacos , Células COS , Chlorocebus aethiops , Colágeno/química , Embrião de Mamíferos , Feminino , Microscopia Confocal , Gravidez , Ratos , Medula Espinal/efeitos dos fármacos , Técnicas de Cultura de Tecidos
8.
Development ; 135(6): 1119-28, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18272594

RESUMO

The finding that morphogens, signalling molecules that specify cell identity, also act as axon guidance molecules has raised the possibility that the mechanisms that establish neural cell fate are also used to assemble neuronal circuits. It remains unresolved, however, how cells differentially transduce the cell fate specification and guidance activities of morphogens. To address this question, we have examined the mechanism by which the Bone morphogenetic proteins (BMPs) guide commissural axons in the developing spinal cord. In contrast to studies that have suggested that morphogens direct axon guidance decisions using non-canonical signal transduction factors, our results indicate that canonical components of the BMP signalling pathway, the type I BMP receptors (BMPRs), are both necessary and sufficient to specify the fate of commissural neurons and guide their axonal projections. However, whereas the induction of cell fate is a shared property of both type I BMPRs, axon guidance is chiefly mediated by only one of the type I BMPRs, BMPRIB. Taken together, these results indicate that the diverse activities of BMP morphogens can be accounted for by the differential use of distinct components of the canonical BMPR complex.


Assuntos
Axônios/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/química , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Diferenciação Celular , Embrião de Galinha , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Modelos Neurológicos , Complexos Multiproteicos , Gravidez , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Medula Espinal/embriologia , Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...