Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 565(7739): 356-360, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30626971

RESUMEN

The development of neural circuits relies on axon projections establishing diverse, yet well-defined, connections between areas of the nervous system. Each projection is formed by growth cones-subcellular specializations at the tips of growing axons, encompassing sets of molecules that control projection-specific growth, guidance, and target selection1. To investigate the set of molecules within native growth cones that form specific connections, here we developed growth cone sorting and subcellular RNA-proteome mapping, an approach that identifies and quantifies local transcriptomes and proteomes from labelled growth cones of single projections in vivo. Using this approach on the developing callosal projection of the mouse cerebral cortex, we mapped molecular enrichments in trans-hemispheric growth cones relative to their parent cell bodies, producing paired subcellular proteomes and transcriptomes from single neuron subtypes directly from the brain. These data provide generalizable proof-of-principle for this approach, and reveal molecular specializations of the growth cone, including accumulations of the growth-regulating kinase mTOR2, together with mRNAs that contain mTOR-dependent motifs3,4. These findings illuminate the relationships between subcellular distributions of RNA and protein in developing projection neurons, and provide a systems-level approach for the discovery of subtype- and stage-specific molecular substrates of circuit wiring, miswiring, and the potential for regeneration.


Asunto(s)
Axones/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteoma/metabolismo , Transcriptoma/genética , Animales , Axones/enzimología , Procesos de Crecimiento Celular , Movimiento Celular , Separación Celular , Femenino , Conos de Crecimiento/enzimología , Conos de Crecimiento/metabolismo , Masculino , Ratones , Proteoma/genética , Serina-Treonina Quinasas TOR/metabolismo
2.
Mol Cell Biochem ; 444(1-2): 1-13, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29159770

RESUMEN

The JNK-interacting protein 3 (JIP3) is a molecular scaffold, expressed predominantly in neurons, that serves to coordinate the activation of the c-Jun N-terminal kinase (JNK) by binding to JNK and the upstream kinases involved in its activation. The JNK pathway is involved in the regulation of many cellular processes including the control of cell survival, cell death and differentiation. JIP3 also associates with microtubule motor proteins such as kinesin and dynein and is likely an adapter protein involved in the tethering of vesicular cargoes to the motors involved in axonal transport in neurons. We have used immunofluorescence microscopy and biochemical fractionation to investigate the subcellular distribution of JIP3 in relation to JNK and to vesicular and organelle markers in rat pheochromocytoma cells (PC12) differentiating in response to nerve growth factor. In differentiated PC12 cells, JIP3 was seen to accumulate in growth cones at the tips of developing neurites where it co-localised with both JNK and the JNK substrate paxillin. Cellular fractionation of PC12 cells showed that JIP3 was associated with a subpopulation of vesicles in the microsomal fraction, distinct from synaptic vesicles, likely to be an anterograde-directed exocytic vesicle pool. In differentiated PC12 cells, JIP3 did not appear to associate with retrograde endosomal vesicles thought to be involved in signalling axonal injury. Together, these observations indicate that JIP3 may be involved in transporting vesicular cargoes to the growth cones of PC12 cells, possibly targeting JNK to its substrate paxillin, and thus facilitating neurite outgrowth.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular , Conos de Crecimiento/enzimología , Proteínas del Tejido Nervioso/metabolismo , Neuritas/enzimología , Vesículas Secretoras/enzimología , Vesículas Sinápticas/enzimología , Animales , Células PC12 , Ratas
3.
Dev Biol ; 430(1): 41-51, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28844905

RESUMEN

Arginylation is an emerging protein modification mediated by arginyltransferase ATE1, shown to regulate embryogenesis and actin cytoskeleton, however its functions in different physiological systems are not well understood. Here we analyzed the role of ATE1 in brain development and neuronal growth by producing a conditional mouse knockout with Ate1 deletion in the nervous system driven by Nestin promoter (Nes-Ate1 mice). These mice were weaker than wild type, resulting in low postnatal survival rates, and had abnormalities in the brain that suggested defects in neuronal migration. Cultured Ate1 knockout neurons showed a reduction in the neurite outgrowth and the levels of doublecortin and F-actin in the growth cones. In wild type, ATE1 prominently localized to the growth cones, in addition to the cell bodies. Examination of the Ate1 mRNA sequence reveals the existence of putative zipcode-binding sequences involved in mRNA targeting to the cell periphery and local translation at the growth cones. Fluorescence in situ hybridization showed that Ate1 mRNA localized to the tips of the growth cones, likely due to zipcode-mediated targeting, and this localization coincided with spots of localization of arginylated ß-actin, which disappeared in the presence of protein synthesis inhibitors. We propose that zipcode-mediated co-targeting of Ate1 and ß-actin mRNA leads to localized co-translational arginylation of ß-actin that drives the growth cone migration and neurite outgrowth.


Asunto(s)
Aminoaciltransferasas/metabolismo , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Conos de Crecimiento/enzimología , Neuritas/enzimología , Proyección Neuronal , Actinas/metabolismo , Animales , Arginina/metabolismo , Encéfalo/anomalías , Encéfalo/patología , Movimiento Celular , Proteínas de Dominio Doblecortina , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Neuropéptidos/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Biomed Res Int ; 2014: 191767, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25050325

RESUMEN

A persistent inflammatory and oxidative stress is a hallmark of most chronic CNS pathologies (Alzheimer's (ALS)) as well as the aging CNS orchestrated by the proinflammatory cytokines tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL-1ß). Loss of the integrity and plasticity of neuronal morphology and connectivity comprises an early step in neuronal degeneration and ultimate decline of cognitive function. We examined in vitro whether TNFα or IL-1ß impaired morphology and motility of growth cones in spinal cord neuron cultures. TNFα and IL-1ß paralyzed growth cone motility and induced growth cone collapse in a dose-dependent manner reflected by complete attenuation of neurite outgrowth. Scavenging reactive oxygen species (ROS) or inhibiting NADPH oxidase activity rescued loss of neuronal motility and morphology. TNFα and IL-1ß provoked rapid, NOX-mediated generation of ROS in advancing growth cones, which preceded paralysis of motility and collapse of morphology. Increases in ROS intermediates were accompanied by an aberrant, nonproductive reorganization of actin filaments. These findings suggest that NADPH oxidase serves as a pivotal source of oxidative stress in neurons and together with disruption of actin filament reorganization contributes to the progressive degeneration of neuronal morphology in the diseased or aging CNS.


Asunto(s)
Conos de Crecimiento/patología , Mediadores de Inflamación/toxicidad , Interleucina-1beta/toxicidad , Parálisis/patología , Especies Reactivas de Oxígeno/toxicidad , Médula Espinal/patología , Factor de Necrosis Tumoral alfa/toxicidad , Citoesqueleto de Actina/efectos de los fármacos , Citoesqueleto de Actina/metabolismo , Animales , Cuerpo Celular/efectos de los fármacos , Cuerpo Celular/metabolismo , Embrión de Pollo , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/enzimología , Humanos , Modelos Biológicos , NADPH Oxidasas/metabolismo , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Proteína de Unión al GTP rac1/metabolismo
5.
J Biol Chem ; 288(29): 20837-20842, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23775074

RESUMEN

Negatively targeting the tumor suppressor and phosphoinositide phosphatase PTEN (phosphatase and tensin homologue) promotes axon regrowth after injury. How PTEN functions in axon guidance has remained unknown. Here we report the differential role of PTEN in chemotactic guidance of axonal growth cones. Down-regulating PTEN expression in Xenopus laevis spinal neurons selectively abolished growth cone chemorepulsion but permitted chemoattraction. These findings persisted during cAMP-dependent switching of turning behaviors. Live cell imaging using a GFP biosensor revealed rapid PTEN-dependent depression of phosphatidylinositol 3,4,5-trisphosphate levels in the growth cone induced by the repellent myelin-associated glycoprotein. Moreover, down-regulating PTEN expression blocked negative remodeling of ß1-integrin adhesions triggered by myelin-associated glycoprotein, yet permitted integrin clustering by a positive chemotropic treatment. Thus, PTEN negatively regulates growth cone phosphatidylinositol 3,4,5-trisphosphate levels and mediates chemorepulsion, whereas chemoattraction is PTEN-independent. Regenerative therapies targeting PTEN may therefore suppress growth cone repulsion to soluble cues while permitting attractive guidance, an essential feature for re-forming functional neural circuits.


Asunto(s)
Quimiotaxis , Conos de Crecimiento/enzimología , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animales , Quimiotaxis/efectos de los fármacos , Análisis por Conglomerados , AMP Cíclico/farmacología , Regulación hacia Abajo/efectos de los fármacos , Endocitosis/efectos de los fármacos , Conos de Crecimiento/efectos de los fármacos , Integrina beta1/metabolismo , Glicoproteína Asociada a Mielina/farmacología , Fosfatos de Fosfatidilinositol/metabolismo
6.
Neurosci Lett ; 547: 59-64, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23680464

RESUMEN

Growth cone motility and morphology, which are critical for axon guidance, are controlled through intracellular events such as actin cytoskeletal reorganization and vesicular trafficking. The membrane phospholipid phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] has been implicated in regulation of these cellular processes in a diverse range of cell types. The main kinases involved in the production of PI(4,5)P2 are the type I phosphatidylinositol 4-phosphate 5-kinase (PIP5K) family, which consist of three isozymes, α, ß and γ. Here, we demonstrate the involvement of PIP5Kß in growth cone dynamics. Overexpression of a lipid kinase-deficient mutant of PIP5Kß (PIP5Kß-KD) in mouse dorsal root ganglion (DRG) neurons stimulated axon elongation and increased growth cone size, whereas wild-type PIP5Kß tended to show opposite effects. Furthermore, PIP5Kß-KD inhibited growth cone collapse of DRG neurons induced by semaphorin 3A (Sema3A). These results provide evidence that PIP5Kß negatively regulates axon elongation and growth cone size and is involved in the cellular signaling pathway for Sema3A-triggered repulsion in DRG neurons.


Asunto(s)
Ganglios Espinales/enzimología , Conos de Crecimiento/enzimología , Neurogénesis/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Semaforina-3A/metabolismo , Animales , Técnica del Anticuerpo Fluorescente , Immunoblotting , Ratones , Ratones Mutantes , Neuronas/enzimología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología
7.
PLoS Biol ; 10(12): e1001439, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23226105

RESUMEN

Local mRNA translation in neurons has been mostly studied during axon guidance and synapse formation but not during initial neurite outgrowth. We performed a genome-wide screen for neurite-enriched mRNAs and identified an mRNA that encodes mitogen-activated protein kinase kinase 7 (MKK7), a MAP kinase kinase (MAPKK) for Jun kinase (JNK). We show that MKK7 mRNA localizes to the growth cone where it has the potential to be translated. MKK7 is then specifically phosphorylated in the neurite shaft, where it is part of a MAP kinase signaling module consisting of dual leucine zipper kinase (DLK), MKK7, and JNK1. This triggers Map1b phosphorylation to regulate microtubule bundling leading to neurite elongation. We propose a model in which MKK7 mRNA localization and translation in the growth cone allows for a mechanism to position JNK signaling in the neurite shaft and to specifically link it to regulation of microtubule bundling. At the same time, this uncouples activated JNK from its functions relevant to nuclear translocation and transcriptional activation.


Asunto(s)
Conos de Crecimiento/enzimología , MAP Quinasa Quinasa 7/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Neuritas/metabolismo , Transporte de ARN , Regiones no Traducidas 3'/genética , Animales , Secuencia de Bases , Diferenciación Celular , Línea Celular , Genoma/genética , Hipocampo/citología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Modelos Biológicos , Neuritas/enzimología , Fosforilación , Fosfotreonina/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Factores de Tiempo
8.
J Neurosci ; 32(19): 6587-99, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22573681

RESUMEN

Axonal outgrowth is a coordinated process of cytoskeletal dynamics and membrane trafficking; however, little is known about proteins responsible for regulating the membrane supply. LMTK1 (lemur kinase 1)/AATYK1 (apoptosis-associated tyrosine kinase 1) is a serine/threonine kinase that is highly expressed in neurons. We recently reported that LMTK1 plays a role in recycling endosomal trafficking in CHO-K1 cells. Here we explore the role of LMTK1 in axonal outgrowth and its regulation by Cdk5 using mouse brain cortical neurons. LMTK1 was expressed and was phosphorylated at Ser34, the Cdk5 phosphorylation site, at the time of axonal outgrowth in culture and colocalized with Rab11A, the small GTPase that regulates recycling endosome traffic, at the perinuclear region and in the axon. Overexpression of the unphosphorylated mutant LMTK1-S34A dramatically promoted axonal outgrowth in cultured neurons. Enhanced axonal outgrowth was diminished by the inactivation of Rab11A, placing LMTK1 upstream of Rab11A. Unexpectedly, the downregulation of LMTK1 by knockdown or gene targeting also significantly enhanced axonal elongation. Rab11A-positive vesicles were transported anterogradely more quickly in the axons of LMTK1-deficient neurons than in those of wild-type neurons. The enhanced axonal outgrowth was reversed by LMTK1-WT or the LMTK1-S34D mutant, which mimics the phosphorylated state, but not by LMTK1-S34A. Thus, LMTK1 can negatively control axonal outgrowth by regulating Rab11A activity in a Cdk5-dependent manner, and Cdk5-LMTK1-Rab11 is a novel signaling pathway involved in axonal outgrowth.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Axones/fisiología , Quinasa 5 Dependiente de la Ciclina/fisiología , Conos de Crecimiento/fisiología , Proteínas Tirosina Quinasas/fisiología , Proteínas de Unión al GTP rab/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Axones/enzimología , Células COS , Células Cultivadas , Chlorocebus aethiops , Femenino , Conos de Crecimiento/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Fosforilación/fisiología , Proteínas Tirosina Quinasas/biosíntesis , Proteínas Tirosina Quinasas/genética , Proteínas de Unión al GTP rab/antagonistas & inhibidores
9.
Mol Cell Neurosci ; 50(1): 82-92, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22521536

RESUMEN

In addition to its role as a morphogen, Sonic hedgehog (Shh) has also been shown to function as a guidance factor that directly acts on the growth cones of various types of axons. However, the noncanonical signaling pathways that mediate the guidance effects of Shh protein remain poorly understood. We demonstrate that a novel signaling pathway consisting of protein kinase Cα (PKCα) and integrin-linked kinase (ILK) mediates the negative guidance effects of high concentration of Shh on retinal ganglion cell (RGC) axons. Shh rapidly increased Ca(2+) level and activated PKCα and ILK in the growth cones of RGC axons. By in vitro kinase assay, PKCα was found to directly phosphorylate ILK on threonine-173 and -181. Inhibition of PKCα or expression of a mutant ILK with the PKCα phosphorylation sites mutated (ILK-DM), abolished the Shh-induced macropinocytosis, growth cone collapse and repulsive axon turning. In vivo, expression of a dominant negative PKCα or ILK-DM disrupted RGC axon pathfinding at the optic chiasm but not the projection toward the optic disk, supporting that this signaling pathway plays a specific role in Shh-mediated negative guidance effects.


Asunto(s)
Axones/enzimología , Proteínas Hedgehog/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Acetofenonas/farmacología , Animales , Axones/fisiología , Benzopiranos/farmacología , Calcio/metabolismo , Células Cultivadas , Embrión de Pollo , Inhibidores Enzimáticos/farmacología , Conos de Crecimiento/enzimología , Mutación , Fosforilación , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Quinasa C-alfa/genética , Proteínas Serina-Treonina Quinasas/genética , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/enzimología , Treonina
10.
J Cell Sci ; 125(Pt 12): 2918-29, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22393238

RESUMEN

There is biochemical, imaging and functional evidence that Rho GTPase signaling is a crucial regulator of actin-based structures such as lamellipodia and filopodia. However, although Rho GTPases are believed to serve similar functions in growth cones, the spatiotemporal dynamics of Rho GTPase signaling has not been examined in living growth cones in response to known axon guidance cues. Here we provide the first measurements of Cdc42 activity in living growth cones acutely stimulated with both growth-promoting and growth-inhibiting axon-guidance cues. Interestingly, we find that both permissive and repulsive factors can work by modulating Cdc42 activity, but in opposite directions. We find that the growth-promoting factors laminin and BDNF activate Cdc42, whereas the inhibitor Slit2 reduces Cdc42 activity in growth cones. Remarkably, we find that regulation of focal adhesion kinase (FAK) activity is a common upstream modulator of Cdc42 by BDNF, laminin and Slit. These findings suggest that rapid modulation of Cdc42 signaling through FAK by receptor activation underlies changes in growth cone motility in response to permissive and repulsive guidance cues.


Asunto(s)
Axones/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Animales , Axones/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Movimiento Celular , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Conos de Crecimiento/enzimología , Conos de Crecimiento/metabolismo , Humanos , Laminina/metabolismo , Transducción de Señal , Xenopus laevis , Proteína de Unión al GTP cdc42/genética
11.
Mol Brain ; 4: 40, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-22051374

RESUMEN

BACKGROUND: The small non-coding microRNAs play an important role in development by regulating protein translation, but their involvement in axon guidance is unknown. Here, we investigated the role of microRNA-134 (miR-134) in chemotropic guidance of nerve growth cones. RESULTS: We found that miR-134 is highly expressed in the neural tube of Xenopus embryos. Fluorescent in situ hybridization also showed that miR-134 is enriched in the growth cones of Xenopus spinal neurons in culture. Importantly, overexpression of miR-134 mimics or antisense inhibitors blocked protein synthesis (PS)-dependent attractive responses of Xenopus growth cones to a gradient of brain-derived neurotrophic factor (BDNF). However, miR-134 mimics or inhibitors had no effect on PS-independent bidirectional responses of Xenopus growth cones to bone morphogenic protein 7 (BMP7). Our data further showed that Xenopus LIM kinase 1 (Xlimk1) mRNA is a potential target of miR-134 regulation. CONCLUSIONS: These findings demonstrate a role for miR-134 in translation-dependent guidance of nerve growth cones. Different guidance cues may act through distinct signaling pathways to elicit PS-dependent and -independent mechanisms to steer growth cones in response to a wide array of spatiotemporal cues during development.


Asunto(s)
Quimiotaxis , Conos de Crecimiento/metabolismo , MicroARNs/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/farmacología , Quimiotaxis/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/enzimología , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Modelos Biológicos , Oligonucleótidos Antisentido/farmacología , Fosforilación/efectos de los fármacos , Xenopus , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
12.
Genes Dev ; 25(18): 1968-81, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21937714

RESUMEN

Suppression of glycogen synthase kinase 3 (GSK3) activity in neurons yields pleiotropic outcomes, causing both axon growth promotion and inhibition. Previous studies have suggested that specific GSK3 substrates, such as adenomatous polyposis coli (APC) and collapsin response mediator protein 2 (CRMP2), support axon growth by regulating the stability of axonal microtubules (MTs), but the substrate(s) and mechanisms conveying axon growth inhibition remain elusive. Here we show that CLIP (cytoplasmic linker protein)-associated protein (CLASP), originally identified as a MT plus end-binding protein, displays both plus end-binding and lattice-binding activities in nerve growth cones, and reveal that the two MT-binding activities regulate axon growth in an opposing manner: The lattice-binding activity mediates axon growth inhibition induced by suppression of GSK3 activity via preventing MT protrusion into the growth cone periphery, whereas the plus end-binding property supports axon extension via stabilizing the growing ends of axonal MTs. We propose a model in which CLASP transduces GSK3 activity levels to differentially control axon growth by coordinating the stability and configuration of growth cone MTs.


Asunto(s)
Axones/fisiología , Regulación del Desarrollo de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Conos de Crecimiento/metabolismo , Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Animales , Axones/enzimología , Citoesqueleto/metabolismo , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3/genética , Conos de Crecimiento/enzimología , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Miosina Tipo II/metabolismo , Neuronas/citología , Unión Proteica
13.
Alcohol Clin Exp Res ; 35(7): 1321-30, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21676004

RESUMEN

BACKGROUND: The effects of ethanol on development of postmitotic neurons include altered neurite outgrowth and differentiation, which may contribute to neuropathology associated with fetal alcohol spectrum disorders. We previously reported that ethanol exposure alters axon growth dynamics in dissociated cultures of rat hippocampal pyramidal neurons. Given the important regulatory role of small Rho guanosine triphosphatases (GTPases) in cytoskeletal reorganization associated with axon growth, and reports that ethanol alters whole cell Rho GTPase activity in other cell types, this study explored the hypothesis that ethanol alters Rho GTPase activity specifically in axonal growth cones. METHODS: Fetal rat hippocampal pyramidal neurons were maintained in dissociated cultures for 1 day in control medium or medium containing 11 to 43 mM ethanol. Some cultures were also treated with brain-derived neurotrophic factor (BDNF), an activator of Rac1 and Cdc42 GTPases that promotes axon extension. Levels of active Rho GTPases in growth cones were measured using in situ binding assays for GTP-bound Rac1, Cdc42, and RhoA. Axon length, growth cone area, and growth cone surface expression of tyrosine kinase B (TrkB), the receptor for BDNF, were assessed by digital morphometry and immunocytochemistry. RESULTS: Although ethanol increased the surface area of growth cones, the levels of active Rho GTPases in axonal growth cones were not affected in the absence of exogenous BDNF. In contrast, ethanol exposure inhibited BDNF-induced Rac1/Cdc42 activation in a dose-dependent manner and increased RhoA activation at the highest concentration tested. Similar TrkB expression was observed on the surface of axonal growth cones of control and ethanol-treated neurons. CONCLUSIONS: These results reveal an inhibitory effect of ethanol on growth cone signaling via small Rho GTPases during early stages of hippocampal development in vitro, and suggest a mechanism whereby ethanol may disrupt neurotrophic factor regulation of axon growth and guidance.


Asunto(s)
Axones/enzimología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Etanol/farmacología , Conos de Crecimiento/enzimología , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/metabolismo , Animales , Axones/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/antagonistas & inhibidores , Tamaño de la Célula/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Conos de Crecimiento/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Hipocampo/crecimiento & desarrollo , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
14.
Dev Cell ; 20(2): 177-91, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21316586

RESUMEN

Although a growing body of evidence supports that Wnt-Frizzled signaling controls axon guidance from vertebrates to worms, whether and how this is mediated by planar cell polarity (PCP) signaling remain elusive. We show here that the core PCP components are required for Wnt5a-stimulated outgrowth and anterior-posterior guidance of commissural axons. Dishevelled1 can inhibit PCP signaling by increasing hyperphosphorylation of Frizzled3 and preventing its internalization. Vangl2 antagonizes that by reducing Frizzled3 phosphorylation and promotes its internalization. In commissural axon growth cones, Vangl2 is predominantly localized on the plasma membrane and is highly enriched on the tips of the filopodia as well as in patches of membrane where new filopodia emerge. Taken together, we propose that the antagonistic functions of Vangl2 and Dvl1 (over Frizzled3 hyperphosphorylation and endocytosis) allow sharpening of PCP signaling locally on the tips of the filopodia to sense directional cues, Wnts, eventually causing turning of growth cones.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Polaridad Celular , Retroalimentación Fisiológica , Conos de Crecimiento/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Tipificación del Cuerpo , Membrana Celular/metabolismo , Proteínas Dishevelled , Embrión de Mamíferos/metabolismo , Activación Enzimática , Receptores Frizzled/metabolismo , Conos de Crecimiento/enzimología , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Fosforilación , Transporte de Proteínas , Seudópodos/metabolismo , Ratas , Proteína Wnt-5a
15.
Neurobiol Dis ; 41(2): 421-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20971191

RESUMEN

Axon regeneration in the adult central nervous system (CNS) is prevented by inhibitory molecules present in myelin, which bind to a receptor complex that leads to downstream RhoGTP activation and axon growth cone collapse. Here, we compared expression of Citron kinase (Citron-K), a target molecule of RhoGTP in non-regenerating dorsal root ganglion neurons (DRGN) after dorsal column (DC) injury, and in regenerating DRGN after either sciatic nerve (SN) injury or preconditioning SN+DC lesion models. We show by microarray that Citron-K mRNA levels in DRGN of a non-regenerating DC injury model were elevated 2-fold compared to those of intact control DRGN. Conversely, Citron-K levels were reduced by 2 and 2.4-fold at 10 days post lesion in the regenerating SN and preconditioning SN+DC lesion models, respectively, compared to levels in control intact DRGN. Western blotting and immunohistochemistry confirmed these observations and localised Citron-K immunostaining to both DRGN and satellite glia. In dissociated, adult rat DRG cell cultures, 80% knockdown of Citron-K, in the presence of inhibitory concentrations of CNS myelin extract (CME), promoted significant disinhibited DRGN neurite outgrowth, only when cells were stimulated with neurotrophic factors. The levels of RhoGTP remained unchanged after Citron-K knockdown in the presence of CME while enhanced cofilin levels correlated with disinhibited DRGN neurite outgrowth. This observation suggests that Citron-K plays a role in axon growth downstream of Rho activation. We conclude that Citron-K regulates actin polymerisation downstream of RhoA and may offer a potentially novel therapeutic approach for promoting CNS axon regeneration.


Asunto(s)
Axones/enzimología , Cofilina 1/metabolismo , Conos de Crecimiento/enzimología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Regeneración Nerviosa/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/fisiología , Proteína de Unión al GTP rhoA/metabolismo , Actinas/metabolismo , Animales , Axones/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Regulación hacia Abajo/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Lim/fisiología , Masculino , Regeneración Nerviosa/genética , Polimerizacion , Proteínas Serina-Treonina Quinasas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Quinasas Asociadas a rho/fisiología
16.
Mol Cell Neurosci ; 45(4): 439-48, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20692345

RESUMEN

The Ca(2+)-stimulated adenylate cyclase 1 (AC1) is a key mediator of retinotopic map refinement and is required for the retraction response of retinal growth cones to the guidance cue ephrin-A5. We show here that AC1 is dynamically expressed in subpopulations of motor neurons in the spinal cord and sensory neurons of the dorsal root ganglia during development. AC1 was first detected around E12.5 in motoneurons of the medial aspect of the lateral motor column (LMCm) and the lateral region of the medial motor column (MMCl), which project to the ventral limb and body wall musculature, respectively. Expression levels gradually increased until they reached a maximum at a time when peripheral sensory and motor axons branch and establish connections with their targets. In barrelless mice, where a mutation inactivates the AC1 gene, sensory projections to the skin in the limbs and trunk region as well as innervations of the intercostal musculature provided by MMCl axons show increased branching. These results suggest a function of AC1 in the formation of peripheral nerve trajectories such as branching and pruning, after the initial projections have been laid down.


Asunto(s)
Adenilil Ciclasas/metabolismo , Ganglios Espinales/crecimiento & desarrollo , Conos de Crecimiento/enzimología , Neurogénesis/fisiología , Adenilil Ciclasas/genética , Animales , Ganglios Espinales/enzimología , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Mutantes , Neuronas Motoras/enzimología , ARN Mensajero/análisis , Células Receptoras Sensoriales/enzimología , Piel/inervación , Médula Espinal/enzimología , Médula Espinal/crecimiento & desarrollo
17.
Neural Dev ; 5: 16, 2010 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-20569485

RESUMEN

BACKGROUND: In the developing hindbrain, cranial motor axon guidance depends on diffusible repellent factors produced by the floor plate. Our previous studies have suggested that candidate molecules for mediating this effect are Slits, Netrin-1 and Semaphorin3A (Sema3A). It is unknown to what extent these factors contribute to floor plate-derived chemorepulsion of motor axons, and the downstream signalling pathways are largely unclear. RESULTS: In this study, we have used a combination of in vitro and in vivo approaches to identify the components of floor plate chemorepulsion and their downstream signalling pathways. Using in vitro motor axon deflection assays, we demonstrate that Slits and Netrin-1, but not Sema3A, contribute to floor plate repulsion. We also find that the axon pathways of dorsally projecting branchiomotor neurons are disrupted in Netrin-1 mutant mice and in chick embryos expressing dominant-negative Unc5a receptors, indicating an in vivo role for Netrin-1. We further demonstrate that Slit and Netrin-1 signalling are mediated by Rho-kinase (ROCK) and myosin light chain kinase (MLCK), which regulate myosin II activity, controlling actin retrograde flow in the growth cone. We show that MLCK, ROCK and myosin II are required for Slit and Netrin-1-mediated growth cone collapse of cranial motor axons. Inhibition of these molecules in explant cultures, or genetic manipulation of RhoA or myosin II function in vivo causes characteristic cranial motor axon pathfinding errors, including the inability to exit the midline, and loss of turning towards exit points. CONCLUSIONS: Our findings suggest that both Slits and Netrin-1 contribute to floor plate-derived chemorepulsion of cranial motor axons. They further indicate that RhoA/ROCK, MLCK and myosin II are components of Slit and Netrin-1 signalling pathways, and suggest that these pathways are of key importance in cranial motor axon navigation.


Asunto(s)
Axones/fisiología , Nervios Craneales/embriología , Neuronas Motoras/fisiología , Miosina Tipo II/fisiología , Quinasa de Cadena Ligera de Miosina/fisiología , Factores de Crecimiento Nervioso/fisiología , Proteínas del Tejido Nervioso/fisiología , Proteínas Supresoras de Tumor/fisiología , Quinasas Asociadas a rho/fisiología , Animales , Axones/ultraestructura , Embrión de Pollo , Nervios Craneales/citología , Nervios Craneales/enzimología , Vías Eferentes/citología , Vías Eferentes/embriología , Vías Eferentes/enzimología , Conos de Crecimiento/enzimología , Conos de Crecimiento/fisiología , Conos de Crecimiento/ultraestructura , Ratones , Ratones Noqueados , Ratones Mutantes , Neuronas Motoras/citología , Neuronas Motoras/enzimología , Miosina Tipo II/metabolismo , Quinasa de Cadena Ligera de Miosina/metabolismo , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Netrina-1 , Técnicas de Cultivo de Órganos , Rombencéfalo/citología , Rombencéfalo/embriología , Rombencéfalo/enzimología , Transducción de Señal/genética , Proteínas Supresoras de Tumor/genética , Quinasas Asociadas a rho/metabolismo
18.
Neurosignals ; 18(4): 246-58, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21358174

RESUMEN

Syntrophins are scaffold proteins that can bind several signaling molecules and localize them to the plasma membrane. We demonstrate here that in neuroblastoma SH-SY5Y cells, brain-specific γ1-syntrophin binds the neurotrophic factor γ-enolase through its PDZ domain, and translocates it to the plasma membrane, as shown by immunoprecipitation, surface plasmon resonance, fluorescence colocalization and flow cytometry. Extensive colocalization of γ1-syntrophin and γ-enolase was observed in neurite growth cones in differentiated SH-SY5Y cells. Silencing of the γ1-syntrophin gene by RNA interference significantly reduced the re-distribution of γ-enolase to the plasma membrane and impaired its neurotrophic effects. We demonstrated that an intact C-terminal end of γ-enolase is essential for its γ1-syntrophin-assisted trafficking. The cleavage of two amino acids at the C-terminal end of γ-enolase by the carboxypeptidase cathepsin X prevents binding with the γ1-syntrophin PDZ domain. Collectively, these data demonstrate that γ1-syntrophin participates in γ-enolase translocation towards the plasma membrane, a pre-requisite for its neurotrophic activity. By disrupting this γ1-syntrophin-guided subcellular distribution, cathepsin X reduces γ-enolase-induced neurotrophic signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Membrana Celular/metabolismo , Conos de Crecimiento/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Regulación hacia Arriba/fisiología , Secuencia de Aminoácidos , Diferenciación Celular/fisiología , Línea Celular Tumoral , Membrana Celular/enzimología , Membrana Celular/patología , Conos de Crecimiento/enzimología , Conos de Crecimiento/patología , Humanos , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/fisiología , Neuroblastoma/enzimología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fosfopiruvato Hidratasa/fisiología , Transporte de Proteínas/fisiología
19.
J Neurosci ; 29(44): 13981-91, 2009 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-19890008

RESUMEN

Adhesion controls growth cone motility, yet the effects of axon guidance cues on adhesion site dynamics are poorly understood. Here we show that ephrin-A1 reduces retinal ganglion cell (RGC) axon outgrowth by stabilizing existing adhesions and inhibiting new adhesion assembly. Ephrin-A1 activates focal adhesion kinase (FAK) in an integrin- and Src-dependent manner and the effects of ephrin-A1 on growth cone motility require FAK activation. We also find that FAK is expressed in a high temporal to low nasal gradient in RGCs, similar to EphA receptors, and that balanced FAK activation is necessary for optimal axon outgrowth. Last, we find that FAK is required for proper topographic positioning of retinal axons along the anterior-posterior axis of the optic tectum in both Xenopus and zebrafish, a guidance decision mediated in part by A-type ephrins. Together, our data suggest that ephrin-A1 controls growth cone advance by modulating adhesive point contacts through FAK activation and that graded FAK signaling is an important component of ephrin-A-mediated retinotopic mapping.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/fisiología , Conos de Crecimiento/fisiología , Retina/fisiología , Animales , Animales Modificados Genéticamente , Adhesión Celular/fisiología , Células Cultivadas , Pollos , Activación Enzimática/fisiología , Conos de Crecimiento/enzimología , Retina/enzimología , Xenopus laevis , Pez Cebra
20.
Mol Biol Cell ; 20(24): 5166-80, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19846664

RESUMEN

The actin microstructure in dendritic spines is involved in synaptic plasticity. Inositol trisphosphate 3-kinase A (ITPKA) terminates Ins(1,4,5)P(3) signals emanating from spines and also binds filamentous actin (F-actin) through its amino terminal region (amino acids 1-66, N66). Here we investigated how ITPKA, independent of its kinase activity, regulates dendritic spine F-actin microstructure. We show that the N66 region of the protein mediates F-actin bundling. An N66 fusion protein bundled F-actin in vitro, and the bundling involved N66 dimerization. By mutagenesis we identified a point mutation in a predicted helical region that eliminated both F-actin binding and bundling, rendering the enzyme cytosolic. A fusion protein containing a minimal helical region (amino acids 9-52, N9-52) bound F-actin in vitro and in cells, but had lower affinity. In hippocampal neurons, GFP-tagged N66 expression was highly polarized, with targeting of the enzyme predominantly to spines. By contrast, N9-52-GFP expression occurred in actin-rich structures in dendrites and growth cones. Expression of N66-GFP tripled the length of dendritic protrusions, induced longer dendritic spine necks, and induced polarized actin motility in time-lapse assays. These results suggest that, in addition to its ability to regulate intracellular Ca(2+) via Ins(1,4,5)P(3) metabolism, ITPKA regulates structural plasticity.


Asunto(s)
Espinas Dendríticas/enzimología , Proteínas de Microfilamentos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Secuencia de Aminoácidos , Aminoácidos/metabolismo , Animales , Axones/enzimología , Línea Celular Tumoral , Citosol/enzimología , Recuperación de Fluorescencia tras Fotoblanqueo , Conos de Crecimiento/enzimología , Proteínas de Microfilamentos/química , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Mutación Puntual/genética , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Conejos , Ratas , Sinapsis/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA