Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Death Differ ; 16(4): 624-37, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19136940

RESUMEN

Positive alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor modulators include benzamide compounds that allosterically modulate AMPA glutamate receptors. These small molecules that cross the blood-brain barrier have been shown to act as a neuroprotectant by increasing the levels of endogenous brain-derived neurotrophic factor (BDNF). Positive AMPA receptor modulators have also been shown to increase the levels of growth-associated protein-43 (GAP-43). GAP-43 plays a major role in many aspects of neuronal function in vertebrates. The goal of this study was to determine whether GAP-43 was important in mediating the actions of positive AMPA receptor modulator (S18986) and BDNF. Using cortical cultures from GAP-43 knockout and control mice, we show that (1) GAP-43 is upregulated in response to S18986 and BDNF in control cultures; (2) this upregulation of GAP-43 is essential for mediating the neuroprotective effects of S18986 and BDNF; (3) administration of S18986 and BDNF leads to an increase in the expression of the glutamate transporters GLT-1 and GLAST that are key to limiting excitotoxic cell death and this increase in GLT-1 and GLAST expression is completely blocked in the absence of GAP-43. Taken together this study concludes that GAP-43 is an important mediator of the neurotrophic effects of S18986 and BDNF on neuronal survival and plasticity, and is essential for the success of positive AMPA receptor modulator-BDNF-based neurotrophin therapy.


Asunto(s)
Benzotiadiazinas/farmacología , Factor Neurotrófico Derivado del Encéfalo/farmacología , Corteza Cerebral/efectos de los fármacos , Proteína GAP-43/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores AMPA/agonistas , Animales , Western Blotting , Muerte Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Proteína GAP-43/genética , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo
2.
Mol Cell Neurosci ; 17(1): 54-66, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11161469

RESUMEN

GAP-43 is first expressed in proliferating neuroblasts and is required for maturation of neurons. When GAP-43 is not expressed in differentiating embryonal carcinoma P19 cells, reduced numbers of neurons were generated. Here we show that neuronal differentiation is initially disrupted at the onset of cell-cycle arrest in aggregated, proliferating neuronal precursors. The ratio of nestin:beta-tubulin-labeled progeny generated at this stage suggests that the differentiation is asymmetric. Apoptosis of immature neurons subsequently produced was also significantly induced. In vivo, too, proliferation of neuroblasts was significantly reduced in cortex of GAP-43(-/-) mice at E14.5. These data demonstrate that when GAP-43 is not expressed in proliferating neuroblasts, neural differentiation is not initiated appropriately, inducing apoptosis. Moreover, the concurrent inhibition of Ca2+-dependent adhesion between differentiating P19 cells in aggregates implicates GAP-43 in CAM-mediated signaling during neurogenesis, as has been previously shown in growth cones.


Asunto(s)
Apoptosis , Diferenciación Celular/genética , Corteza Cerebral/metabolismo , Proteína GAP-43/biosíntesis , Proteínas del Tejido Nervioso , Neuronas/metabolismo , Animales , Calcio/metabolismo , Agregación Celular/efectos de los fármacos , Agregación Celular/genética , Recuento de Células , Ciclo Celular/genética , División Celular/efectos de los fármacos , División Celular/genética , Células Cultivadas , Corteza Cerebral/embriología , Corteza Cerebral/patología , Células Clonales , Células Madre de Carcinoma Embrionario , Proteína GAP-43/deficiencia , Proteína GAP-43/genética , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/biosíntesis , Ratones , Ratones Noqueados , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Nestina , Neuronas/patología , Transducción de Señal , Tretinoina/farmacología
3.
Neuroreport ; 11(8): 1603-5, 2000 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-10852209

RESUMEN

Plasticity after microinjection of kainic acid (KA) into the adult rat whisker barrel cortex was investigated with immunohistochemical staining of phosphorylated growth-associated protein (GAP)-43. After mapping the barrel cortex with the technique of intrinsic signal optical imaging, a small volume of KA was injected into one barrel. Rats were sacrificed at 2 days, 3 days, 1 week, and 6 weeks after lesioning. GAP-43 staining demonstrated intense immunoreactivity (IR) at the injected barrel which spread to the inter-barrel septa and the surrounding barrels. Elevated IR of GAP-43 was visible 2 days after KA injection, and increased gradually at least 6 weeks following the lesion. This model has the possibility of offering a simple and reliable tool for studying cortical plasticity.


Asunto(s)
Proteína GAP-43/metabolismo , Ácido Kaínico/farmacología , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/metabolismo , Vibrisas/fisiología , Animales , Inmunohistoquímica/métodos , Inyecciones , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley , Coloración y Etiquetado
4.
Brain Res ; 860(1-2): 95-103, 2000 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-10727627

RESUMEN

C57BL/6 (B6) mice display better contextual learning than the DBA/2 (D2) mice. The possibility that GAP-43, is differentially affected as a function of strain and learning was investigated in the present study. No basal difference between C57BL/6J (B6) and DBA/2J (D2) mice in the amount of hippocampal GAP-43 was observed, but naive D2 mice have slightly lower basal levels of GAP-43 phosphorylation than do B6 mice. Interestingly, alterations in hippocampal GAP-43 protein levels and phosphorylation state in response to training for contextual learning were observed only in B6 mice. Immediate-shocked mice, serving as nonlearning controls, showed no GAP-43 alterations, nor did D2 mice subjected to either training condition. These results suggest that modulation of hippocampal GAP-43 may be important for contextual learning and that strain-specific alterations in GAP-43 may be part of a disrupted pathway in D2 mice that is essential for learning.


Asunto(s)
Condicionamiento Operante/fisiología , Miedo/fisiología , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Aprendizaje/fisiología , Ratones Endogámicos C57BL/psicología , Ratones Endogámicos DBA/psicología , Procesamiento Proteico-Postraduccional , Estimulación Acústica , Animales , Cruzamientos Genéticos , Electrochoque , Masculino , Ratones , Ratones Endogámicos C57BL/fisiología , Ratones Endogámicos DBA/fisiología , Fosforilación
5.
Brain Res ; 853(2): 384-95, 2000 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-10640639

RESUMEN

GAP-43 is expressed in proliferating neuroblasts in vivo and in vitro, but its role during early neurogenesis has not been investigated. Here we show that neuroectodermal differentiation stimulated by retinoic acid (RA) in the embryonal carcinoma (EC) line P19 is accompanied by upregulation of GAP-43 expression in neuroepithelial precursor cells. In contrast, when upregulation of GAP-43 expression was prevented in 3 independent P19 lines because of a targeted insertion into the gene, generation of neuroepithelial precursors was inhibited. Consequently, neuronal number was significantly decreased, neuronal morphology was abnormal and fewer than 20% of all neurons were able to initiate neuritogenesis. Extracellular matrix (ECM) was unable to rescue initiation of neuritogenesis in the mutant cells, however those neurites that were extended responded normally to ECM-stimulated neurite outgrowth-promoting signals. These data suggest that GAP-43 function is required for commitment to a neuronal phenotype as well as initiation of neurite extension. However, stimulation of neurite outgrowth by ECM in P19s occurs independently of GAP-43.


Asunto(s)
Diferenciación Celular/genética , Proteína GAP-43/genética , Marcación de Gen , Células Madre Neoplásicas/metabolismo , Neuronas/citología , Animales , Diferenciación Celular/efectos de los fármacos , Colágeno/metabolismo , Colágeno/farmacología , Combinación de Medicamentos , Células Madre de Carcinoma Embrionario , Matriz Extracelular/metabolismo , Proteína GAP-43/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Homocigoto , Laminina/metabolismo , Laminina/farmacología , Ratones , Microscopía por Video , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/efectos de los fármacos , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Fenotipo , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , Células Madre/citología , Células Madre/efectos de los fármacos , Transfección , Tretinoina/farmacología , Células Tumorales Cultivadas
6.
Proc Natl Acad Sci U S A ; 96(16): 9397-402, 1999 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-10430954

RESUMEN

There is strong evidence that growth-associated protein (GAP-43), a protein found only in the nervous system, regulates the response of neurons to axonal guidance signals. However, its role in complex spatial patterning in cerebral cortex has not been explored. We show that mice lacking GAP-43 expression (-/-) fail to establish the ordered whisker representation (barrel array) normally found in layer IV of rodent primary somatosensory cortex. Thalamocortical afferents to -/- cortex form irregular patches in layer IV within a poorly defined cortical field, which varies between hemispheres, rather than the stereotypic, whisker-specific, segregated map seen in normal animals. Furthermore, many thalamocortical afferents project abnormally to widely separated cortical targets. Taken together, our findings indicate a loss of identifiable whisker territories in the GAP-43 -/- mouse cortex. Here, we present a disrupted somatotopic map phenotype in cortex, in clear contrast to the blurring of boundaries within an ordered whisker map in other barrelless mutants. Our results indicate that GAP-43 expression is critical for the normal establishment of ordered topography in barrel cortex.


Asunto(s)
Mapeo Encefálico , Proteína GAP-43/fisiología , Corteza Somatosensorial/fisiología , Vibrisas/inervación , Animales , Corteza Auditiva/citología , Corteza Auditiva/fisiología , Transporte Axonal , Cruzamientos Genéticos , Complejo IV de Transporte de Electrones/análisis , Femenino , Lateralidad Funcional , Proteína GAP-43/deficiencia , Proteína GAP-43/genética , Heterocigoto , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , Fenotipo , Reacción en Cadena de la Polimerasa , Corteza Somatosensorial/citología , Conducta Estereotipada , Núcleos del Trigémino/fisiología
7.
J Neurosci ; 18(24): 10429-37, 1998 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-9852580

RESUMEN

The mechanisms whereby cell adhesion molecules (CAMs) promote axonal growth and synaptic plasticity are poorly understood. Here we show that the neurite outgrowth stimulated by NCAM-mediated fibroblast growth factor (FGF) receptor activation in cerebellar granule cells is associated with increased GAP-43 phosphorylation on serine-41. In contrast, neither NCAM nor FGF was able to stimulate neurite outgrowth in similar neurons from mice in which the GAP-43 gene had been deleted by homologous recombination. Integrin-mediated neurite outgrowth was unaffected by GAP-43 deletion. Both neurite outgrowth and rapid phosphorylation of GAP-43 in isolated growth cones required the first three Ig domains of a NCAM-Fc chimera and were stimulated maximally at 5 micrograms/ml (approximately 50 nM). Likewise, GAP-43 phosphorylation in isolated growth cones also was stimulated by an L1-Fc chimera. Both neurite outgrowth and NCAM-stimulated GAP-43 phosphorylation were inhibited by antibodies to the FGF receptor and a diacylglycerol lipase inhibitor (RHC80267) that blocks the production of arachidonic acid in response to activation of the FGF receptor. Direct activation of the FGF receptor and the arachidonic acid cascade with either basic FGF or melittin also resulted in increased GAP-43 phosphorylation. These data suggest that the stimulation of neurite outgrowth by NCAM requires GAP-43 function and that GAP-43 phosphorylation in isolated growth cones occurs via an FGF receptor-dependent increase in arachidonic acid.


Asunto(s)
Proteína GAP-43/metabolismo , Proteína GAP-43/fisiología , Conos de Crecimiento/metabolismo , Moléculas de Adhesión de Célula Nerviosa/farmacología , Neuritas/fisiología , Células 3T3 , Animales , Anticuerpos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ciclohexanonas/farmacología , Factores de Crecimiento de Fibroblastos/farmacología , Proteína GAP-43/genética , Ratones , Ratones Noqueados , Neuritas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/efectos de los fármacos , Transfección
8.
J Neurochem ; 71(5): 2104-11, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9798936

RESUMEN

Consumption of moderate quantities of ethanol during pregnancy produces deficits in long-term potentiation in the hippocampal formation of adult offspring. Protein kinase C (PKC)-mediated phosphorylation of the presynaptic protein GAP-43 is critical for the induction of long-term potentiation. We tested the hypothesis that this system is affected in fetal alcohol-exposed (FAE) rats by measuring GAP-43 phosphorylation and PKC activity in the hippocampus of adult offspring of rat dams that had consumed one of three diets throughout gestation: (a) a 5% ethanol liquid diet, which produced a maternal blood ethanol concentration of 83 mg/dl (FAE); (b) an isocalorically equivalent 0% ethanol diet (pair-fed); or (c) lab chow ad libitum. Western blot analysis using specific antibodies to PKC-phosphorylated GAP-43 revealed that FAE rats had an approximately 50% reduction in the proportion of phosphorylated GAP-43. Similarly, we found that PKC-mediated incorporation of 32P into GAP-43 was reduced by 85% in hippocampal slices from FAE rats compared with both control groups. FAE animals also showed a 50% reduction in total hippocampal PKC activity, whereas the levels of six major PKC isozymes did not change in any of the diet groups. These results suggest that GAP-43 phosphorylation deficits in rats prenatally exposed to moderate levels of ethanol are not due to alterations in the expression of either the enzyme or substrate protein, but rather to a defect in kinase activation.


Asunto(s)
Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Efectos Tardíos de la Exposición Prenatal , Proteína Quinasa C/antagonistas & inhibidores , Animales , Femenino , Hipocampo/efectos de los fármacos , Técnicas In Vitro , Isoenzimas/metabolismo , Fosforilación/efectos de los fármacos , Embarazo , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley
9.
J Neurobiol ; 35(3): 287-99, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9622012

RESUMEN

Phosphorylation of GAP-43 (neuromodulin) by protein kinase C (PKC) occurs at a single site, serine41. In vivo, phosphorylation is induced after initiation of axonogenesis and is confined to distal axons and growth cones. Within individual growth cones, phosphorylation is nonuniformly distributed. Here, we have used high-resolution video-enhanced microscopy of cultured dorsal root ganglia neurons together with immunocytochemistry with a monoclonal antibody that recognizes PKC-phosphorylated GAP-43 to correlate the distribution of phosphorylated GAP-43 with growth cone behavior. In "quiescent," nontranslocating growth cones, phosphorylated GAP-43 was confined to the proximal neurite and the central organelle-rich region, and was low in organelle-poor lamellae. However, levels in lamellae were elevated when they became motile. Conversely, levels of phosphorylated GAP-43 were low in either lamellae that were actively retracting or in the central organelle-rich region and proximal neurite of growth cones that had totally collapsed. The results suggest a mechanism whereby phosphorylation of GAP-43 by PKC, potentially in response to extracellular signals, could direct the functional behavior of the growth cone.


Asunto(s)
Proteína GAP-43/metabolismo , Neuronas/fisiología , Animales , Movimiento Celular/fisiología , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Neuronas/metabolismo , Neuronas/ultraestructura , Orgánulos/ultraestructura , Fosforilación , Ratas/embriología , Distribución Tisular
10.
Exp Neurol ; 154(2): 673-83, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9878202

RESUMEN

Tacrolimus (FK506), a widely used immunosuppressant drug, has neurite-promoting activity in cultured PC12 cells and peripheral neurons. The present study investigated whether tacrolimus affects the expression of the neuronal growth-associated protein, GAP-43, as well as functional recovery after photothrombotic spinal cord injury in the rat. In injured animals receiving tacrolimus, the number of neurons expressing GAP-43 mRNA and protein approximately doubled compared to that in injured animals receiving vehicle alone. This increase in GAP-43-positive cells was paralleled by a significant improvement in neurological function evaluated by open-field and inclined plane tests. Another FKBP-12 ligand (V-10,367) had similar effects on GAP-43 expression and functional outcome, indicating that the observed effects of tacrolimus do not involve inhibition of the phosphatase calcineurin. Thus, tacrolimus, a drug which is already approved for use in humans, as well as other FKBP-12 ligands which do not inhibit calcineurin, could potentially enhance functional outcome after CNS injury in humans.


Asunto(s)
Proteína GAP-43/genética , Inmunosupresores/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Tacrolimus/farmacología , Animales , Anticuerpos , Conducta Animal/fisiología , Calcineurina/metabolismo , Proteína GAP-43/análisis , Proteína GAP-43/inmunología , Expresión Génica/efectos de los fármacos , Hibridación in Situ , Macrófagos/fisiología , Microglía/fisiología , Neuritas/química , Neuritas/enzimología , Fosforilación , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Traumatismos de la Médula Espinal/inmunología
11.
J Neurosci ; 17(10): 3515-24, 1997 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9133376

RESUMEN

Synthesis of GAP-43 (also known as neuromodulin) in neurons is induced during axon growth, and high concentrations (estimated between 50 and 100 microM) accumulate in the growth cone. GAP-43 is tightly associated with the growth cone membrane skeleton, the structure that transduces extracellular guidance cues into alterations in morphology by spatially regulating polymerization of actin filaments, thereby causing directional changes in axon growth. GAP-43 cosediments with actin filaments, and its phosphorylation on serine 41 by PKC, too, is spatially regulated so that phosphorylated GAP-43 is found in areas where growth cones make productive, stable contacts with other cells. In contrast, unphosphorylated GAP-43, which binds calmodulin, is always found in parts of the growth cone that are retracting. Here we have used a cell-free assay to investigate how the phosphorylation status of GAP-43 affects its interactions with actin and show that both phosphorylated and unphosphorylated GAP-43 have different, independent effects on actin filament structure. Phosphorylated GAP-43 stabilizes long actin filaments (Kd = 161 nM), and antibodies to phosphorylated GAP-43 inhibit binding of actin to phalloidin, implying a lateral interaction with filaments. In contrast, unphosphorylated GAP-43 reduces filament length distribution (Kd = 1.2 microM) and increases the critical concentration for polymerization. Prebinding calmodulin potentiates this effect. The results show that spatially regulated post-translational modifications of GAP-43 within the growth cone, which can be regulated in response to extracellular signals, have the ability to directly influence the structure of the actin cytoskeleton.


Asunto(s)
Actinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C/metabolismo , Serina/metabolismo , Actinas/análisis , Animales , Unión Competitiva/fisiología , Química Encefálica , Calmodulina/metabolismo , Células Cultivadas/química , Células Cultivadas/enzimología , Células Cultivadas/ultraestructura , Citoesqueleto/química , Citoesqueleto/enzimología , Proteína GAP-43 , Ganglios Espinales/citología , Inmunohistoquímica , Cinética , Glicoproteínas de Membrana/análisis , Músculo Esquelético/química , Proteínas del Tejido Nervioso/análisis , Neuritas/química , Neuritas/enzimología , Proteínas de Neurofilamentos/análisis , Proteínas de Neurofilamentos/metabolismo , Fosforilación , Conejos , Ratas
12.
Brain Res ; 732(1-2): 36-42, 1996 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-8891266

RESUMEN

Nerve growth factor (NGF) has been shown to play important roles in neuronal survival, growth and differentiation. Recently, we have found that intracortical infusion of NGF into adult cat visual cortex can recreate ocular dominance plasticity, suggesting that NGF is also involved in activity-dependent modification of synaptic connectivity in the adult brain. To further explore the mechanisms of NGF-induced plasticity in adult visual cortex, we studied two presynaptic markers: GAP-43 and synaptophysin. Immunocytochemical staining showed that NGF-treatment of adult visual cortex selectively increased the level of the phosphorylated form of GAP-43, while the total level of GAP-43 was not changed. These results demonstrate that NGF-treatment stimulates phosphorylation processes of GAP-43 in vivo. In addition, NGF-treatment of adult visual cortex increased the level of synaptophysin immunoreactivity. Since the phosphorylated form of GAP-43 is known to be enriched in the membrane skeleton of growth cones and of developing synapses, and the phosphorylation of GAP-43 has been linked with events that underlie synaptic plasticity, and since synaptophysin is a major component of presynaptic vesicles, our results suggest that NGF-treatment of adult visual cortex modulates presynaptic terminals, possibly by inducing axonal sprouting and formation of new synapses, and that these changes may play a role in the NGF-induced functional plasticity.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Factores de Crecimiento Nervioso/farmacología , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/efectos de los fármacos , Sinaptofisina/metabolismo , Corteza Visual/fisiología , Animales , Biomarcadores , Gatos , Proteína GAP-43 , Humanos , Inmunohistoquímica , Infusiones Parenterales , Factores de Crecimiento Nervioso/administración & dosificación , Proteínas de Neurofilamentos/metabolismo , Fosforilación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Valores de Referencia , Sinapsis/efectos de los fármacos , Sinapsis/fisiología , Sinaptofisina/análisis , Corteza Visual/citología , Corteza Visual/efectos de los fármacos
13.
J Pharmacol Exp Ther ; 278(3): 1428-34, 1996 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8819530

RESUMEN

Repeated, intermittent treatment of rats with amphetamine results in a sensitization of locomotor and stereotyped behaviors that is accompanied by an enhancement in stimulus-induced dopamine release. The effects of repeated treatment with amphetamine on the phosphorylation state of neuromodulin and synapsin I, proteins involved in neurotransmitter release, were investigated. Rats were injected with 2.5 mg/kg AMPH, twice a week for 5 weeks (intermittent treatment). One week after the last injection, a challenge dose of 2.5 mg/kg AMPH was given 30 min before sacrifice. We previously reported an increase in neuromodulin phosphorylation with this sensitization paradigm. Site 3-phospho-synapsin I, site 1-phospho-synapsin I and phosphoser41-neuromodulin were detected with phosphorylation state-specific antibodies. Acute treatment with amphetamine did not increase the state of synapsin phosphorylation at either site 1 or site 3, but both site 1-phospho-synapsin I and site 3-phospho-synapsin I were increased (38% and 34%, respectively) after repeated, intermittent amphetamine. Immunoreactivity for phosphoser41-neuromodulin was increased by acute amphetamine. Site 3-phospho-synapsin I, site 1-phospho-synapsin I and phosphoser41-neuromodulin were also measured in striatum from rats receiving a different regimen in which amphetamine is given in escalating doses for 4 weeks. With this regimen, behavioral sensitization and enhanced dopamine release are exhibited in rats withdrawn 4 weeks, but not 3 days, after pretreatment. Small but significant increases in site 3-phospho-synapsin I and phosphoser41-neuromodulin were found in rats withdrawn 4 weeks from the escalating dose regimen, but not in those withdrawn 3 days. The increase in the phosphorylation state of synapsin I and neuromodulin reflect changes in the presynaptic signal transduction pathways which could play a role in the behavioral sensitization and contribute to the enhanced dopamine release reported in amphetamine-sensitized rats.


Asunto(s)
Cuerpo Estriado/metabolismo , Dextroanfetamina/administración & dosificación , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sinapsinas/metabolismo , Animales , Calmodulina/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Femenino , Proteína GAP-43 , Fosforilación , Fosfoserina/metabolismo , Ratas , Sinaptosomas/metabolismo
14.
Diabetes ; 45(2): 199-204, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8549865

RESUMEN

Experimental diabetes in the rat is associated with impaired axon regeneration. Successful regeneration depends on the construction of axonal growth cones and establishment of appropriate target connections. The growth-associated protein (GAP)-43 is a major component of the axonal growth cone, and its synthesis and axonal transport are markedly increased during regeneration. The purpose of this study was to determine the effect of experimental diabetes on the synthesis and axonal transport of GAP-43 in regenerating sciatic nerves. Rats were rendered diabetic with 50 mg/kg streptozotocin i.p. Four weeks later, the rats were anesthetized, and one sciatic nerve was crushed to induce regeneration. After 2 weeks, nerves were ligated, and 6 h later, nerve pieces proximal to the ligature and dorsal root ganglia were removed, and proteins were separated by PAGE. Western blots of gels were probed with antibody 10E8/E7 against GAP-43. The presence of GAP-43 was confirmed by immunohistochemistry of nerve sections. Densitometric analysis of the blots showed a 45% reduction in native GAP-43 immunoreactivity in nerve pieces proximal to the ligature (P < 0.05; n = 7). Northern blots of total RNA extracted from pooled dorsal root ganglia were probed with a 32P-radiolabeled cDNA probe for GAP-43. There was no significant difference in the amount of GAP-43 mRNA between diabetic and nondiabetic rats. Immunohistochemistry of sciatic nerve confirmed the reduction in GAP-43 immunoreactivity. We conclude that a defect in turnover or axonal transport of GAP-43 may contribute to the impaired peripheral nerve regeneration in diabetes.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nervio Ciático/metabolismo , Animales , Transporte Axonal , Western Blotting , Diabetes Mellitus Experimental , Femenino , Proteína GAP-43 , Ganglios Espinales/metabolismo , Compresión Nerviosa , Regeneración Nerviosa , ARN Mensajero/genética , Ratas , Ratas Wistar
15.
J Neurobiol ; 29(2): 213-32, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8821178

RESUMEN

To investigate the molecular basis for GAP-43 function in axon outgrowth, we produced a mutant, GAP-43 (Ala41), whose interaction with calmodulin in vitro was unaffected by increasing Ca2+ concentrations, and stably transfected it into GAP-43-deficient PC12B cells. Several lines that expressed wild-type or mutant protein at levels that resembled endogenous GAP-43 expression in PC12 controls were subcloned and characterized. GAP-43 (Ala41) was significantly more extractable with Nonidet P-40 and less tightly associated with the membrane skeleton than the wild-type protein. Furthermore, GAP-43 (Ala41) expression by PC12B cells profoundly affected their phenotype: First, observation of living cells using video-enhanced microscopy revealed irregular plasma membranes with numerous blebs and protrusions and neurites that appeared thin and varicose. Second, both the cells' ability to remain attached to laminin substrates and the amount of alpha 1 beta 1 integrin expressed on the cell surface was significantly decreased. Finally, peripherin transport, which is abnormal in PC12B cells, could be rescued by transfection of wild-type GAP-43 but not the GAP-43 (Ala41) mutant. The phenotypic abnormalities resemble other cell types in which membrane skeleton/plasma membrane interactions have been functionally decoupled, and our results are consistent with the notion that these interactions may be abnormal in GAP-43 (Ala41)-expressing PC12B cells, either as a direct consequence of the mutation or arising secondarily to the altered availability of calmodulin in the growing neurite.


Asunto(s)
Alanina/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Serina/metabolismo , Animales , Calcio/farmacología , Calmodulina/metabolismo , Adhesión Celular , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Proteína GAP-43 , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Integrinas/metabolismo , Microscopía por Video , Mutagénesis , Neuritas/ultraestructura , Células PC12 , Ratas , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/ultraestructura , Transfección
16.
Curr Opin Neurol ; 7(2): 153-9, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8019661

RESUMEN

Over the past several years remarkable progress has been made towards unraveling the complex mechanisms that regulate neuronal development. Because the end results of abnormalities in brain development are often developmental disabilities, it is timely to review several recent advances in the field of neurobiology. This review, with contributions from several co-authors, provides a synopsis of breakthroughs from the fields of embryology, cell biology, and molecular genetics that hold promise for exciting clinical application. This article is arranged to reflect the stages of normal development. Understanding the mechanisms underlying neuronal induction, regional specification, neuronal specification, migration, axonal growth, neurotrophic factors, and myelination should clarify the pathophysiology of numerous neurological disorders, and provide new insights into their treatment.


Asunto(s)
Diferenciación Celular/fisiología , Sistema Nervioso/citología , Animales , Axones/fisiología , Diferenciación Celular/genética , Movimiento Celular/genética , Movimiento Celular/fisiología , Humanos , Mutación , Proteínas de la Mielina/genética , Fibras Nerviosas Mielínicas/fisiología , Factores de Crecimiento Nervioso , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/trasplante
17.
J Comp Neurol ; 340(3): 361-80, 1994 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-8188856

RESUMEN

Olfactory axons have been shown to grow aberrantly and form dense collections of axons, termed neuromas, in the olfactory epithelium of rats in which the olfactory bulb was ablated. Likewise, in human olfactory mucosa, collections of neurites have been noted in a variety of disease states, including Alzheimer's disease. We report here an immunohistochemical and electron microscopic analysis of aberrant axonal growth in the rat olfactory mucosa induced by experimental lesion. In particular, we have used the monoclonal antibody 2G12, which binds to the phosphorylated form of GAP-43, as an extremely sensitive marker for neuromatous axons, because it does not label neuronal cell bodies. In unilaterally bulbectomized rats, neuromas form in posterior olfactory epithelium on the operated side. Several lines of evidence, including serial section reconstruction, indicate that olfactory axons are induced to grow back into the epithelium at a distance from their point of origin as a consequence of bulbectomy, and are accompanied by glial cells from the olfactory nerve. Avulsion of a part of the olfactory nerve has similar effects as destruction of the olfactory bulb. Intraepithelial neuromas also develop in the olfactory mucosa of rats simultaneously exposed to methyl bromide gas and injected with 3-methyl indole; this treatment severely damages the olfactory epithelium directly. Exposure to methyl bromide alone causes milder damage, and the neuromas that form are transient. The evidence indicates that neuromas form after the epithelium is directly damaged because axons are trapped in the epithelium. Both of the mechanisms identified here should be taken into account when considering the findings in the human olfactory mucosa.


Asunto(s)
Neoplasias Encefálicas/patología , Neuroma/patología , Vías Olfatorias/citología , Animales , Anticuerpos Monoclonales/inmunología , Axones/fisiología , Axones/ultraestructura , Hidrocarburos Bromados/toxicidad , Inmunohistoquímica , Masculino , Microscopía Electrónica , Bulbo Olfatorio/fisiología , Mucosa Olfatoria/efectos de los fármacos , Mucosa Olfatoria/crecimiento & desarrollo , Mucosa Olfatoria/inervación , Nervio Olfatorio/fisiología , Vías Olfatorias/crecimiento & desarrollo , Vías Olfatorias/ultraestructura , Ratas , Ratas Sprague-Dawley , Escatol/toxicidad
18.
J Neurochem ; 62(1): 291-9, 1994 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8263528

RESUMEN

Phosphorylation of the nervous system-specific protein GAP-43 in growth cones in vivo increases as the growth cones near their targets, at a time when the gangliosides GM1 and GD1a are being accumulated in the growth cone membrane, thus raising the possibility that the gangliosides could modulate GAP-43 behavior. We used a subcellular fraction of intact isolated growth cones to show that both GM1 and GD1a affected the calcium-dependent posttranslational regulation of GAP-43 in several similar ways. Both gangliosides induced rapid incorporation of phosphate into GAP-43; however, the induction was undetectable with our antibody 2G12 that is specific for kinase C-phosphorylated GAP-43. Furthermore, neither ganglioside stimulated kinase C activity in isolated growth cones, suggesting that the rapid phosphorylation may not be on Ser41, the kinase C site. However, both gangliosides did induce a slower accumulation of GAP-43 phosphorylated on Ser41, apparently by inhibiting a phosphatase. Finally, calcium-dependent proteolysis of GAP-43 was also stimulated by both GM1 and GD1a. In contrast, GD1a, but not GM1, caused the redistribution of GAP-43 into the isolated growth cone cytoskeleton. The results demonstrate that both gangliosides can modulate the calcium-dependent regulation of GAP-43.


Asunto(s)
Axones/metabolismo , Gangliósido G(M1)/farmacología , Gangliósidos/farmacología , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Prosencéfalo/metabolismo , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Axones/ultraestructura , Fraccionamiento Celular , Centrifugación por Gradiente de Densidad , Citoesqueleto/metabolismo , Proteína GAP-43 , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Serina/metabolismo
19.
Neuroscience ; 52(3): 563-74, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8095708

RESUMEN

Pharmacological and biochemical evidence implicate the Ca2+ and phospholipid-dependent protein kinase C in long-term potentiation. The in vitro hippocampal slice preparation was used to demonstrate redistribution of protein kinase C from cytosol to membrane and protein kinase C-dependent phosphorylation of the presynaptic growth-associated protein-43 substrate following long-term potentiation induction in area CA1. Protein kinase C translocation was assessed using both quantitative immunoblotting with a monoclonal antibody recognizing a common epitope in the alpha and beta isoforms of protein kinase C and Ca2+ and phospholipid-dependent phosphorylation of exogenous histone substrate. Slices examined 5 min after tetanus-induced spike potentiation showed no change in protein kinase C redistribution, whereas slices examined at 15-, 30- and 60-min intervals all showed a similar degree of protein kinase C translocation to membrane, although only at 15 min was the effect statistically significant. Additionally, an increase in protein kinase C-dependent growth-associated protein 43 phosphorylation was observed 10 min after high-frequency stimulation. The translocation of protein kinase C and phosphorylation of growth-associated protein 43 were dependent upon high-frequency (repetitive 400 Hz) afferent stimulation, as no effects were observed in slices receiving low-frequency (1 Hz) or no stimulation. The N-methyl-D-aspartate receptor antagonist, DL-2-amino-5-phosphonovaleric acid (50 microM), inhibited induction of long-term potentiation, redistribution of protein kinase C and phosphorylation of growth-associated protein 43. A significant redistribution of the predominantly presynaptic protein kinase C isoform, protein kinase C-alpha, was also detected 15 min after induction of long-term potentiation using an alpha-isoform-specific monoclonal antibody. These observations support a presynaptic role for protein kinase C and growth-associated protein 43 in the early maintenance phase of LTP, and further suggest that a retrograde messenger produced postsynaptically following N-methyl-D-aspartate receptor activation mediates these effects.


Asunto(s)
Potenciales Evocados/fisiología , Hipocampo/fisiología , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Sinapsis/fisiología , 2-Amino-5-fosfonovalerato/farmacología , Animales , Fraccionamiento Celular , Membrana Celular/enzimología , Citosol/enzimología , Estimulación Eléctrica , Activación Enzimática , Proteína GAP-43 , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Técnicas In Vitro , Glicoproteínas de Membrana/análisis , Proteínas del Tejido Nervioso/análisis , Fosfoproteínas/análisis , Fosforilación , Tractos Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
20.
J Neurobiol ; 23(8): 1037-53, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1460463

RESUMEN

In vivo, kinase C phosphorylation of the growth-associated protein GAP-43 is spatially and temporally associated with the proximity of growing axons to their targets. Here we have used dissociated dorsal root ganglia (DRG)s and an antibody specific for the phosphorylated form of GAP-43 to demonstrate that neurite regeneration in culture also begins in the absence of detectable levels of phosphorylated GAP-43. Since the beta isoform of kinase C was found to be enriched in growth cones before stably phosphorylated GAP-43 was detected, it may normally be inactive during initial neurite outgrowth; however, premature phosphorylation of GAP-43 could be stimulated in newly dissociated DRGs by plating them on cultures in which phosphorylation had already been initiated media conditioned by such cultures caused no response suggesting an effect of either cell-cell or cell-substrate contact. Increased GAP-43 phosphorylation correlated with a reduced extent of neurite outgrowth but not with the rate at which individual growth cones translocated so that motile growth cones contained very low levels of phosphorylated GAP-43, whereas stationary growth cones showed much more immunoreactivity. Downregulation of kinase C by phorbol ester prevented increased GAP-43 phosphorylation and led to growth cone collapse. Finally, phosphorylated GAP-43 was found to be differently distributed within growth cones. Increased immunoreactivity was frequently observed in the neck of the growth cone and was heterogeneously distributed in lamellae and filopodia. These results, which demonstrate the dynamic regulation of GAP-43 phosphorylation in individual growth cones, are discussed with reference to the association between changes in growth cone shape and the ability to translocate and change direction.


Asunto(s)
Ganglios Espinales/crecimiento & desarrollo , Glicoproteínas de Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Carbocianinas , División Celular , Células Cultivadas , Regulación hacia Abajo/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Proteína GAP-43 , Ganglios Espinales/metabolismo , Histocitoquímica , Isoenzimas/metabolismo , Forbol 12,13-Dibutirato/farmacología , Fosforilación , Embarazo , Proteína Quinasa C/inmunología , Proteína Quinasa C/metabolismo , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...