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










Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 24(12): e57145, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37870370

RESUMEN

α-Synuclein phosphorylation at serine-129 (pS129) is a widely used surrogate marker of pathology in Parkinson's disease and other synucleinopathies. However, we recently demonstrated that phosphorylation of S129 is also a physiological activator of synaptic transmission. In a feed-forward fashion, neuronal activity triggers reversible pS129. Here, we show that Parkinson's disease-linked missense mutations in SNCA impact activity-dependent pS129. Under basal conditions, cytosol-enriched A30P, H50Q, and G51D mutant forms of α-synuclein exhibit reduced pS129 levels in rat primary cortical neurons. A53T pS129 levels are similar to wild-type, and E46K pS129 levels are higher. A30P and E46K mutants show impaired reversibility of pS129 after stimulation. For the engineered profoundly membrane-associated α-synuclein mutant "3K" (E35K + E46K + E61K), de-phosphorylation was virtually absent after blocking stimulation, implying that reversible pS129 is severely compromised. Importantly, pS129 excess resulting from proteasome inhibition is also associated with reduced reversibility by neuronal inhibition, kinase inhibition, or phosphatase activation. Our findings suggest that perturbed pS129 dynamics are probably a shared characteristic of pathology-associated α-synuclein, with possible implications for synucleinopathy treatment and diagnosis.


Asunto(s)
Enfermedad de Parkinson , Sinucleinopatías , Animales , Ratas , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Serina/metabolismo , Fosforilación
2.
NPJ Parkinsons Dis ; 9(1): 4, 2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36646701

RESUMEN

In Parkinson's disease and other synucleinopathies, the elevation of α-synuclein phosphorylated at Serine129 (pS129) is a widely cited marker of pathology. However, the physiological role for pS129 has remained undefined. Here we use multiple approaches to show for the first time that pS129 functions as a physiological regulator of neuronal activity. Neuronal activity triggers a sustained increase of pS129 in cultured neurons (200% within 4 h). In accord, brain pS129 is elevated in environmentally enriched mice exhibiting enhanced long-term potentiation. Activity-dependent α-synuclein phosphorylation is S129-specific, reversible, confers no cytotoxicity, and accumulates at synapsin-containing presynaptic boutons. Mechanistically, our findings are consistent with a model in which neuronal stimulation enhances Plk2 kinase activity via a calcium/calcineurin pathway to counteract PP2A phosphatase activity for efficient phosphorylation of membrane-bound α-synuclein. Patch clamping of rat SNCA-/- neurons expressing exogenous wild-type or phospho-incompetent (S129A) α-synuclein suggests that pS129 fine-tunes the balance between excitatory and inhibitory neuronal currents. Consistently, our novel S129A knock-in (S129AKI) mice exhibit impaired hippocampal plasticity. The discovery of a key physiological function for pS129 has implications for understanding the role of α-synuclein in neurotransmission and adds nuance to the interpretation of pS129 as a synucleinopathy biomarker.

3.
Neurobiol Dis ; 172: 105816, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35820646

RESUMEN

Early cognitive impairment in Alzheimer's disease may result in part from synaptic dysfunction caused by the accumulation oligomeric assemblies of amyloid ß-protein (Aß). Changes in hippocampal function seem critical for cognitive impairment in early Alzheimer's disease (AD). Diffusible oligomers of Aß (oAß) have been shown to block canonical long-term potentiation (LTP) in the CA1 area of hippocampus, but whether there is also a direct effect of oAß on synaptic transmission and plasticity at synapses between mossy fibers (axons) from the dentate gyrus granule cells and CA3 pyramidal neurons (mf-CA3 synapses) is unknown. Studies in APP transgenic mice have suggested an age-dependent impairment of mossy fiber LTP. Here we report that although endogenous AD brain-derived soluble oAß had no effect on mossy-fiber basal transmission, it strongly impaired paired-pulse facilitation in the mossy fiber pathway and presynaptic mossy fiber LTP (mf-LTP). Selective activation of both ß1 and ß2 adrenergic receptors and their downstream cAMP/PKA signaling pathway prevented oAß-mediated inhibition of mf-LTP. Unexpectedly, activation of the cGMP/PKG signaling pathway also prevented oAß-impaired mf-LTP. Our results reveal certain specific pharmacological targets to ameliorate human oAß-mediated impairment at the mf-CA3 synapse.


Asunto(s)
Enfermedad de Alzheimer , Potenciación a Largo Plazo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Hipocampo/metabolismo , Humanos , Potenciación a Largo Plazo/fisiología , Ratones , Fibras Musgosas del Hipocampo/fisiología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
4.
Alzheimers Dement ; 18(6): 1186-1202, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34550630

RESUMEN

INTRODUCTION: Evidence strongly suggests that soluble oligomers of amyloid beta protein (oAß) help initiate the pathogenic cascade of Alzheimer's disease (AD). To date, there have been no validated assays specific for detecting and quantifying oAß in human blood. METHODS: We developed an ultrasensitive oAß immunoassay using a novel capture antibody (71A1) with N-terminal antibody 3D6 for detection that specifically quantifies soluble oAß in the human brain, cerebrospinal fluid (CSF), and plasma. RESULTS: Two new antibodies (71A1; 1G5) are oAß-selective, label Aß plaques in non-fixed AD brain sections, and potently neutralize the synaptotoxicity of AD brain-derived oAß. The 71A1/3D6 assay showed excellent dilution linearity in CSF and plasma without matrix effects, good spike recovery, and specific immunodepletion. DISCUSSION: We have created a sensitive, high throughput, and inexpensive method to quantify synaptotoxic oAß in human plasma for analyzing large cohorts of aged and AD subjects to assess the dynamics of this key pathogenic species and response to therapy.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Anciano , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Encéfalo/patología , Humanos , Inmunoensayo , Placa Amiloide/metabolismo
5.
Glia ; 69(3): 594-608, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32970902

RESUMEN

Fragile X syndrome (FXS) is one of the most common inherited intellectual disability (ID) disorders, in which the loss of FMRP protein induces a range of cellular signaling changes primarily through excess protein synthesis. Although neuron-centered molecular and cellular events underlying FXS have been characterized, how different CNS cell types are involved in typical FXS synaptic signaling changes and behavioral phenotypes is largely unknown. Recent evidence suggests that selective loss of astroglial FMRP is able to dysregulate glutamate uptake, increase spine density, and impair motor-skill learning. Here we investigated the effect of astroglial FMRP on synaptic signaling and FXS-related behavioral and learning phenotypes in astroglial Fmr1 cKO and cON mice in which FMRP expression is selectively diminished or restored in astroglia. We found that selective loss of astroglial FMRP contributes to cortical hyperexcitability by enhancing NMDAR-mediated evoked but not spontaneous miniEPSCs and elongating cortical UP state duration. Selective loss of astroglial FMRP is also sufficient to increase locomotor hyperactivity, significantly diminish social novelty preference, and induce memory acquisition and extinction deficits in astroglial Fmr1 cKO mice. Importantly, re-expression of astroglial FMRP is able to significantly rescue the hyperactivity (evoked NMDAR response, UP state duration, and open field test) and social novelty preference in astroglial Fmr1 cON mice. These results demonstrate a profound role of astroglial FMRP in the evoked synaptic signaling, spontaneously occurring cortical UP states, and FXS-related behavioral and learning phenotypes and provide important new insights in the cell type consideration for the FMRP reactivation strategy.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Animales , Astrocitos , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Ratones , Fenotipo , Receptores de N-Metil-D-Aspartato
6.
J Endocrinol ; 245(3): 397-410, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32240981

RESUMEN

RASGRF1 (GRF1) is a calcium-stimulated guanine-nucleotide exchange factor that activates RAS and RAC GTPases. In hippocampus neurons, it mediates the action of NMDA and calcium-permeable AMPA glutamate receptors on specific forms of synaptic plasticity, learning, and memory in both male and female mice. Recently, we showed GRF1 also regulates the HPA axis response to restraint stress, but only in female mice before puberty. In particular, we found that after 7 days of restraint stress (7DRS) (30 min/day) both elevated serum CORT levels and induction of an anxiolytic phenotype normally observed in early adolescent (EA) female mice are blocked in GRF1-knockout mice. In contrast, no effects were observed in EA male or adult females. Here, we show this phenotype is due, at least in part, to GRF1 loss in CRF cells of the paraventricular nucleus of the hypothalamus, as GRF1 knockout specifically in these cells suppressed 7DRS-induced elevation of serum CORT levels specifically in EA females, but only down to levels found in comparably stressed EA males. Nevertheless, it still completely blocked the 7DRS-induced anxiolytic phenotype observed in EA females. Interestingly, loss of GRF1 in CRF cells had no effect after only three restraint stress exposures, implying a role for GRF1 in 7DRS stress-induced plasticity of CRF cells that appears to be specific to EA female mice. Overall, these findings indicate that GRF1 in CRF cells makes a key contribution to the distinct response EA females display to repeated stress.


Asunto(s)
ras-GRF1/metabolismo , Animales , Corticosterona/sangre , Femenino , Ratones , Ratones Noqueados , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Receptores de Glutamato/metabolismo , Receptores de Glutamato/fisiología , Estrés Fisiológico , ras-GRF1/genética
7.
J Neurosci ; 37(12): 3138-3149, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28213442

RESUMEN

The principal neurons of the medial superior olive (MSO) encode cues for horizontal sound localization through comparisons of the relative timing of EPSPs. To understand how the timing and amplitude of EPSPs are maintained during propagation in the dendrites, we made dendritic and somatic whole-cell recordings from MSO principal neurons in brain slices from Mongolian gerbils. In somatic recordings, EPSP amplitudes were largely uniform following minimal stimulation of excitatory synapses at visualized locations along the dendrites. Similar results were obtained when excitatory synaptic transmission was eliminated in a low calcium solution and then restored at specific dendritic sites by pairing input stimulation and focal application of a higher calcium solution. We performed dual dendritic and somatic whole-cell recordings to measure spontaneous EPSPs using a dual-channel template-matching algorithm to separate out those events initiated at or distal to the dendritic recording location. Local dendritic spontaneous EPSP amplitudes increased sharply in the dendrite with distance from the soma (length constant, 53.6 µm), but their attenuation during propagation resulted in a uniform amplitude of ∼0.2 mV at the soma. The amplitude gradient of dendritic EPSPs was also apparent in responses to injections of identical simulated excitatory synaptic currents in the dendrites. Compartmental models support the view that these results extensively reflect the influence of dendritic cable properties. With relatively few excitatory axons innervating MSO neurons, the normalization of dendritic EPSPs at the soma would increase the importance of input timing versus location during the processing of interaural time difference cues in vivoSIGNIFICANCE STATEMENT The neurons of the medial superior olive analyze cues for sound localization by detecting the coincidence of binaural excitatory synaptic inputs distributed along the dendrites. Previous studies have shown that dendritic voltages undergo severe attenuation as they propagate to the soma, potentially reducing the influence of distal inputs. However, using dendritic and somatic patch recordings, we found that dendritic EPSP amplitude increased with distance from the soma, compensating for dendritic attenuation and normalizing EPSP amplitude at the soma. Much of this normalization reflected the influence of dendritic morphology. As different combinations of presynaptic axons may be active during consecutive cycles of sound stimuli, somatic EPSP normalization renders spike initiation more sensitive to synapse timing than dendritic location.


Asunto(s)
Dendritas/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Células Receptoras Sensoriales/fisiología , Localización de Sonidos/fisiología , Complejo Olivar Superior/fisiología , Sinapsis/fisiología , Animales , Células Cultivadas , Femenino , Gerbillinae , Masculino
8.
J Endocrinol ; 227(1): 1-12, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246084

RESUMEN

Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis has been implicated in the induction and prolongation of a variety of psychiatric disorders. As such, much effort has been made to understand the molecular mechanisms involved in its control. However, the vast majority of the studies on the HPA axis have used adult animals, and among these the majority has used males. Here we show that in knockout mice lacking the guanine nucleotide exchange factor, RasGRF1, habituation to 30 min/day of restraint stress is markedly accelerated, such that these mice do not display elevated corticosterone levels or enhanced locomotion after 7 days of stress exposure, like WT mice do. Strikingly, this phenotype is present in early-adolescent female RasGRF1 knockout mice, but not in their early-adolescent male, mid-adolescent female, adult female or adult male counterparts. Moreover, not only is there a clear response to restraint stress in early-adolescent female RasGRF1 knockout mice, their response after one, three and five exposures is magnified approximately threefold compared to WT mice. These findings imply that distinct mechanisms exist to regulate the HPA axis in early-adolescent females that involves RasGRF1. A full understanding of how RasGRF1 controls the HPA axis response to stress may be required to design effective strategies to combat stress-associated psychiatric disorders initiated in young females.


Asunto(s)
Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , ras-GRF1/metabolismo , Factores de Edad , Animales , Conducta Animal , Corticosterona/sangre , Femenino , Habituación Psicofisiológica , Sistema Hipotálamo-Hipofisario/crecimiento & desarrollo , Sistema Hipotálamo-Hipofisario/fisiología , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Sistema Hipófiso-Suprarrenal/crecimiento & desarrollo , Sistema Hipófiso-Suprarrenal/fisiología , Sistema Hipófiso-Suprarrenal/fisiopatología , Restricción Física , Caracteres Sexuales , Estrés Fisiológico , Estrés Psicológico/sangre , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Factores de Tiempo , ras-GRF1/genética
9.
Hippocampus ; 24(11): 1317-29, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24894950

RESUMEN

Hippocampal adult neurogenesis contributes to key functions of the dentate gyrus (DG), including contextual discrimination. This is due, at least in part, to the unique form of plasticity that new neurons display at a specific stage of their development when compared with the surrounding principal neurons. In addition, the contribution that newborn neurons make to dentate function can be enhanced by an increase in their numbers induced by a stimulating environment. However, signaling mechanisms that regulate these properties of newborn neurons are poorly understood. Here, we show that Ras-GRF2 (GRF2), a calcium-regulated exchange factor that can activate Ras and Rac GTPases, contributes to both of these properties of newborn neurons. Using Ras-GRF2 knockout mice and wild-type mice stereotactically injected with retrovirus containing shRNA against the exchange factor, we demonstrate that GRF2 promotes the survival of newborn neurons of the DG at approximately 1-2 weeks after their birth. GRF2 also controls the distinct form of long-term potentiation that is characteristic of new neurons of the hippocampus through its effector Erk MAP kinase. Moreover, the enhancement of neuron survival that occurs after mice are exposed to an enriched environment also involves GRF2 function. Consistent with these observations, GRF2 knockout mice display defective contextual discrimination. Overall, these findings indicate that GRF2 regulates both the basal level and environmentally induced increase of newborn neuron survival, as well as in the induction of a distinct form of synaptic plasticity of newborn neurons that contributes to distinct features of hippocampus-derived learning and memory.


Asunto(s)
Supervivencia Celular/fisiología , Giro Dentado/fisiología , Vivienda para Animales , Potenciación a Largo Plazo/fisiología , Neuronas/fisiología , Factores de Intercambio de Guanina Nucleótido ras/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Discriminación en Psicología/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Reacción Cataléptica de Congelación/fisiología , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Neuronas/efectos de los fármacos , ARN Interferente Pequeño , Percepción Espacial/fisiología , Técnicas de Cultivo de Tejidos , Factores de Intercambio de Guanina Nucleótido ras/genética
10.
J Biol Chem ; 289(23): 16551-64, 2014 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-24755227

RESUMEN

Ras-GRF1 (GRF1) and Ras-GRF2 (GRF2) constitute a family of similar calcium sensors that regulate synaptic plasticity. They are both guanine exchange factors that contain a very similar set of functional domains, including N-terminal pleckstrin homology, coiled-coil, and calmodulin-binding IQ domains and C-terminal Dbl homology Rac-activating domains, Ras-exchange motifs, and CDC25 Ras-activating domains. Nevertheless, they regulate different forms of synaptic plasticity. Although both GRF proteins transduce calcium signals emanating from NMDA-type glutamate receptors in the CA1 region of the hippocampus, GRF1 promotes LTD, whereas GRF2 promotes θ-burst stimulation-induced LTP (TBS-LTP). GRF1 can also mediate high frequency stimulation-induced LTP (HFS-LTP) in mice over 2-months of age, which involves calcium-permeable AMPA-type glutamate receptors. To add to our understanding of how proteins with similar domains can have different functions, WT and various chimeras between GRF1 and GRF2 proteins were tested for their abilities to reconstitute defective LTP and/or LTD in the CA1 hippocampus of Grf1/Grf2 double knock-out mice. These studies revealed a critical role for the GRF2 CDC25 domain in the induction of TBS-LTP by GRF proteins. In contrast, the N-terminal pleckstrin homology and/or coiled-coil domains of GRF1 are key to the induction of HFS-LTP by GRF proteins. Finally, the IQ motif of GRF1 determines whether a GRF protein can induce LTD. Overall, these findings show that for the three forms of synaptic plasticity that are regulated by GRF proteins in the CA1 hippocampus, specificity is encoded in only one or two domains, and a different set of domains for each form of synaptic plasticity.


Asunto(s)
Factor 2 Liberador de Guanina Nucleótido/metabolismo , Transducción de Señal , ras-GRF1/metabolismo , Animales , Señalización del Calcio , Hipocampo/metabolismo , Ratones
11.
Hippocampus ; 24(3): 315-25, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24174283

RESUMEN

The dentate gyrus of the hippocampus plays a pivotal role in pattern separation, a process required for the behavioral task of contextual discrimination. One unique feature of the dentate gyrus that contributes to pattern separation is adult neurogenesis, where newly born neurons play a distinct role in neuronal circuitry. Moreover,the function of neurogenesis in this brain region differs in adolescent and adult mice. The signaling mechanisms that differentially regulate the distinct steps of adult neurogenesis in adolescence and adulthood remain poorly understood. We used mice lacking RASGRF1(GRF1), a calcium-dependent exchange factor that regulates synaptic plasticity and participates in contextual discrimination performed by mice, to test whether GRF1 plays a role in adult neurogenesis.We show Grf1 knockout mice begin to display a defect in neurogenesis at the onset of adulthood (~2 months of age), when wild-type mice first acquire the ability to distinguish between closely related contexts. At this age, young hippocampal neurons in Grf1 knockout mice display severely reduced dendritic arborization. By 3 months of age, new neuron survival is also impaired. BrdU labeling of new neurons in 2-month-old Grf1 knockout mice shows they begin to display reduced survival between 2 and 3 weeks after birth, just as new neurons begin to develop complex dendritic morphology and transition into using glutamatergic excitatory input. Interestingly, GRF1 expression appears in new neurons at the developmental stage when GRF1 loss begins to effect neuronal function. In addition, we induced a similar loss of new hippocampal neurons by knocking down expression of GRF1 solely in new neurons by injecting retrovirus that express shRNA against GRF1 into the dentate gyrus. Together, these findings show that GRF1 expressed in new neurons promotes late stages of adult neurogenesis. Overall our findings show GRF1 to be an age-dependent regulator of adult hippocampal neurogenesis, which contributes to ability of mice to distinguish closely related contexts.


Asunto(s)
Envejecimiento/fisiología , Giro Dentado/metabolismo , Discriminación en Psicología/fisiología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/fisiología , ras-GRF1/fisiología , Animales , Animales Recién Nacidos , Animales Lactantes , División Celular , Supervivencia Celular/efectos de los fármacos , Giro Dentado/crecimiento & desarrollo , Quinasas Similares a Doblecortina , Fluoxetina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/análisis , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Transducción de Señal/fisiología , ras-GRF1/antagonistas & inhibidores , ras-GRF1/deficiencia , ras-GRF1/genética
12.
J Biol Chem ; 288(30): 21703-13, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23766509

RESUMEN

RAS-GRF1 is a guanine nucleotide exchange factor with the ability to activate RAS and RAC GTPases in response to elevated calcium levels. We previously showed that beginning at 1 month of age, RAS-GRF1 mediates NMDA-type glutamate receptor (NMDAR)-induction of long term depression in the CA1 region of the hippocampus of mice. Here we show that beginning at 2 months of age, when mice first acquire the ability to discriminate between closely related contexts, RAS-GRF1 begins to contribute to the induction of long term potentiation (LTP) in the CA1 hippocampus by mediating the action of calcium-permeable, AMPA-type glutamate receptors (CP-AMPARs). Surprisingly, LTP induction by CP-AMPARs through RAS-GRF1 occurs via activation of p38 MAP kinase rather than ERK MAP kinase, which has more frequently been linked to LTP. Moreover, contextual discrimination is blocked by knockdown of Ras-Grf1 expression specifically in the CA1 hippocampus, infusion of a p38 MAP kinase inhibitor into the CA1 hippocampus, or the injection of an inhibitor of CP-AMPARs. These findings implicate the CA1 hippocampus in the developmentally dependent capacity to distinguish closely related contexts through the appearance of a novel LTP-supporting signaling pathway.


Asunto(s)
Discriminación en Psicología/fisiología , Potenciación a Largo Plazo/fisiología , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , ras-GRF1/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacología , Animales , Butadienos/farmacología , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Calcio/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Imidazoles/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/metabolismo , Neuronas/fisiología , Nitrilos/farmacología , Piridinas/farmacología , Interferencia de ARN , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , ras-GRF1/genética
13.
PLoS One ; 5(7): e11732, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20661302

RESUMEN

BACKGROUND: NMDA-type glutamate receptors (NMDARs) are major contributors to long-term potentiation (LTP), a form of synaptic plasticity implicated in the process of learning and memory. These receptors consist of calcium-permeating NR1 and multiple regulatory NR2 subunits. A majority of studies show that both NR2A and NR2B-containing NMDARs can contribute to LTP, but their unique contributions to this form of synaptic plasticity remain poorly understood. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we show that NR2A and NR2B-containing receptors promote LTP differently in the CA1 hippocampus of 1-month old mice, with the NR2A receptors functioning through Ras-GRF2 and its downstream effector, Erk Map kinase, and NR2B receptors functioning independently of these signaling molecules. CONCLUSIONS/SIGNIFICANCE: This study demonstrates that NR2A-, but not NR2B, containing NMDA receptors induce LTP in pyramidal neurons of the CA1 hippocampus from 1 month old mice through Ras-GRF2 and Erk. This difference add new significance to the observation that the relative levels of these NMDAR subtypes is regulated in neurons, such that NR2A-containing receptors become more prominent late in postnatal development, after sensory experience and synaptic activity.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Potenciación a Largo Plazo/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología , Factores de Intercambio de Guanina Nucleótido ras/metabolismo , Animales , Electrofisiología , Quinasas MAP Reguladas por Señal Extracelular/genética , Potenciación a Largo Plazo/genética , Ratones , Ratones Mutantes , Modelos Biológicos , Receptores de N-Metil-D-Aspartato/genética , Transducción de Señal/genética , Factores de Intercambio de Guanina Nucleótido ras/genética
14.
Neuron ; 61(3): 351-8, 2009 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-19217373

RESUMEN

Calcium-calmodulin-dependent protein kinase II (CaMKII) is a key mediator of synaptic plasticity and learning. Global pyramidal cell glutamate stimulation induces translocation of CaMKII from dendritic shafts to spines. Here we show that local dendritic stimulation by puffing glutamate onto a region containing 7-32 synapses induces translocation of CaMKII to synapses initially at the puff site but that translocation subsequently spreads within dendrites to the distal dendrite arbor, resulting in a persistent, widespread synaptic accumulation. This locally induced propagating synaptic (L-IPS) accumulation of CaMKII requires activation of NMDA receptors and L-type Ca(2+) channels and is preceded by a Ca(2+) spike. L-IPS translocation of CaMKII alters biochemical signaling and is associated with an increase in AMPA receptor GluR1 at both stimulated and nonstimulated synapses and thus provides a molecular mechanism for heterosynaptic plasticity.


Asunto(s)
Señalización del Calcio/fisiología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dendritas/enzimología , Hipocampo/enzimología , Receptores de Glutamato/metabolismo , Sinapsis/enzimología , Animales , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Dendritas/ultraestructura , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Hipocampo/ultraestructura , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Transporte de Proteínas/fisiología , Ratas , Receptores AMPA/efectos de los fármacos , Receptores AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/ultraestructura , Potenciales Sinápticos/efectos de los fármacos , Potenciales Sinápticos/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
15.
Cell ; 119(7): 1013-26, 2004 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-15620359

RESUMEN

Formation of synaptic connections requires alignment of neurotransmitter receptors on postsynaptic dendrites opposite matching transmitter release sites on presynaptic axons. beta-neurexins and neuroligins form a trans-synaptic link at glutamate synapses. We show here that neurexin alone is sufficient to induce glutamate postsynaptic differentiation in contacting dendrites. Surprisingly, neurexin also induces GABA postsynaptic differentiation. Conversely, neuroligins induce presynaptic differentiation in both glutamate and GABA axons. Whereas neuroligins-1, -3, and -4 localize to glutamate postsynaptic sites, neuroligin-2 localizes primarily to GABA synapses. Direct aggregation of neuroligins reveals a linkage of neuroligin-2 to GABA and glutamate postsynaptic proteins, but the other neuroligins only to glutamate postsynaptic proteins. Furthermore, mislocalized expression of neuroligin-2 disperses postsynaptic proteins and disrupts synaptic transmission. Our findings indicate that the neurexin-neuroligin link is a core component mediating both GABAergic and glutamatergic synaptogenesis, and differences in isoform localization and binding affinities may contribute to appropriate differentiation and specificity.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/farmacología , Sinapsis/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo , Secuencias de Aminoácidos/genética , Animales , Células COS , Moléculas de Adhesión Celular Neuronal , Técnicas de Cocultivo , Dendritas/metabolismo , Distroglicanos/metabolismo , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Ratones , Mutación , Proteínas del Tejido Nervioso/genética , Unión Proteica , Ratas , Sinapsis/metabolismo
16.
J Neurosci Methods ; 134(2): 121-31, 2004 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-15003378

RESUMEN

We have recently applied Lindau-Neher's capacitance measurement technique to study vesicle trafficking at the calyx-type synapse in the rat medial nucleus of the trapezoid body (MNTB) in slice conditions. This application made the MNTB synapse an excellent model for the study of exocytosis and endocytosis at conventional active zones. However, the application was only made at calyces that are presumably equivalent to a single-compartment circuit because their passive current transients decayed mono-exponentially. Here, we determined whether the application could be extended to majority of calyces whose passive current transients decayed bi-exponentially. By comparison of calyces with mono- or bi-exponential decay in their passive current transients, we found similar properties in respect to: (1) the capacitance jump induced by trains of action-potential equivalent stimuli, which reflects exocytosis; (2) the size of a releasable vesicle pool; (3) the time course of the decay after the capacitance jump, which reflects endocytosis; and (4) the transient capacitance artifact observed in the presence of Cd(2+) that blocks exocytosis. These similar properties were also obtained from modeling calyces as a single- or two-compartment circuit. Thus, capacitance measurements may be extended to the majority of calyces, which may facilitate the study of rapid vesicle trafficking at conventional active zones.


Asunto(s)
Tronco Encefálico/citología , Capacidad Eléctrica , Neuronas/fisiología , Sinapsis/fisiología , Animales , Animales Recién Nacidos , Axones/metabolismo , Tronco Encefálico/fisiología , Cadmio/farmacología , Simulación por Computador , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Isoquinolinas/metabolismo , Modelos Neurológicos , Neuronas/citología , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Wistar , Sinapsis/clasificación , Sinapsis/efectos de los fármacos , Factores de Tiempo
17.
J Neurosci ; 23(10): 4017-22, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12764087

RESUMEN

The possible involvement of p38 mitogen-activated protein kinase activation in spinal cord and dorsal root ganglion (DRG) cells in the development of peripheral neuropathic pain has been explored. Ligation of the L5 spinal nerve (SNL) on one side in adult rats produces an early onset and long-lasting mechanical allodynia. This lesion results in activation of p38 in the L5 segment of the spinal cord, most prominently in the ipsilateral dorsal horn, starting soon after the lesion (<1 d) and persisting for >3 weeks. The activated p38 in the spinal cord is restricted entirely to microglia; phospho-p38 colocalizes only with the microglial marker OX-42 and not with either the neuronal marker neuronal-specific nuclear protein or the astrocyte marker GFAP. In contrast, SNL induces a delayed (>3 d) activation of p38 in the L5 DRG that occurs predominantly in neurons. Continuous injection of the p38 inhibitor 4-(4-fluorophenyl)-2-(4-methylsulfonylphenyl)-5-(4-pyridyl)-1H-imidazole (SB203580) via the intrathecal route, starting before the SNL surgery, reduces SNL-induced mechanical allodynia from day 1 to day 10, with maximal effects at early time points. Post-treatment with SB203580 starting on day 1 or on day 10 after surgery also reduces established mechanical allodynia. Because the reduction in neuropathic pain by p38 inhibition occurs before the appearance of p38 activation in DRG neurons, p38 activation in spinal cord microglia is likely to have a substantial role in the earliest phase of neuropathic pain. Coactivation of p38 in DRG neurons and spinal microglia may contribute to later phases of neuropathic pain.


Asunto(s)
Ganglios Espinales/enzimología , Ganglios Espinales/patología , Microglía/enzimología , Microglía/patología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Dolor/enzimología , Dolor/fisiopatología , Médula Espinal/enzimología , Médula Espinal/patología , Nervios Espinales/enzimología , Nervios Espinales/patología , Animales , Conducta Animal/efectos de los fármacos , Activación Enzimática , Inhibidores Enzimáticos/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/enzimología , Hiperalgesia/fisiopatología , Imidazoles/administración & dosificación , Imidazoles/uso terapéutico , Inyecciones Espinales , Ligadura , Masculino , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Dimensión del Dolor , Piridinas/administración & dosificación , Piridinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas p38 Activadas por Mitógenos
18.
Neuron ; 37(1): 121-33, 2003 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-12526778

RESUMEN

Here we describe a novel mechanism for plasma membrane insertion of the delta opioid receptor (DOR). In small dorsal root ganglion neurons, only low levels of DORs are present on the cell surface, in contrast to high levels of intracellular DORs mainly associated with vesicles containing calcitonin gene-related peptide (CGRP). Activation of surface DORs caused Ca(2+) release from IP(3)-sensitive stores and Ca(2+) entry, resulting in a slow and long-lasting exocytosis, DOR insertion, and CGRP release. In contrast, membrane depolarization or activation of vanilloid and P2Y(1) receptors induced a rapid DOR insertion. Thus, DOR activation induces a Ca(2+)-dependent insertion of DORs that is coupled to a release of excitatory neuropeptides, suggesting that treatment of inflammatory pain should include blockade of DORs.


Asunto(s)
Membrana Celular/metabolismo , Exocitosis/fisiología , Ganglios Espinales/metabolismo , Neuronas Aferentes/metabolismo , Nociceptores/metabolismo , Receptores Opioides delta/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Membrana Celular/efectos de los fármacos , Membrana Celular/ultraestructura , Exocitosis/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/ultraestructura , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/ultraestructura , Neuropéptidos/metabolismo , Nociceptores/efectos de los fármacos , Nociceptores/ultraestructura , Células PC12 , Dolor/metabolismo , Dolor/fisiopatología , Ratas , Receptores de Droga/efectos de los fármacos , Receptores de Droga/metabolismo , Receptores de Neurotransmisores/efectos de los fármacos , Receptores de Neurotransmisores/metabolismo , Receptores Opioides delta/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1 , Vesículas Secretoras/metabolismo , Vesículas Secretoras/ultraestructura
19.
Neuron ; 36(1): 57-68, 2002 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-12367506

RESUMEN

Peripheral inflammation induces p38 MAPK activation in the soma of C fiber nociceptors in the dorsal root ganglion (DRG) after 24 hr. Inflammation also increases protein, but not mRNA levels, of the heat-gated ion channel TRPV1 (VR1) in these cells, which is then transported to peripheral but not central C fiber terminals. Inhibiting p38 activation in the DRG reduces the increase in TRPV1 in the DRG and inflamed skin and diminishes inflammation-induced heat hypersensitivity without affecting inflammatory swelling or basal pain sensitivity. p38 activation in the DRG is secondary to peripheral production of NGF during inflammation and is required for NGF-induced increases in TRPV1. The activation of p38 in the DRG following retrograde NGF transport, by increasing TRPV1 levels in nociceptor peripheral terminals in a transcription-independent fashion, contributes to the maintenance of inflammatory heat hypersensitivity.


Asunto(s)
Hiperalgesia/enzimología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Neuronas Aferentes/enzimología , Receptores de Droga/deficiencia , Regulación hacia Arriba/fisiología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/enzimología , Hiperalgesia/fisiopatología , Inmunohistoquímica , Inflamación/enzimología , Inflamación/fisiopatología , Masculino , Ratones , Ratones Noqueados , Fibras Nerviosas Amielínicas/efectos de los fármacos , Fibras Nerviosas Amielínicas/enzimología , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Neuralgia/enzimología , Neuralgia/fisiopatología , Neuronas Aferentes/efectos de los fármacos , Nociceptores/efectos de los fármacos , Nociceptores/enzimología , Células del Asta Posterior/enzimología , Ratas , Ratas Sprague-Dawley , Receptores de Droga/efectos de los fármacos , Receptores de Droga/genética , Regulación hacia Arriba/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Eur J Neurosci ; 16(2): 175-85, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12169100

RESUMEN

Peripheral axotomy-induced sprouting of thick myelinated afferents (A-fibers) from laminae III-IV into laminae I-II of the spinal cord is a well-established hypothesis for the structural basis of neuropathic pain. However, we show here that the cholera toxin B subunit (CTB), a neuronal tracer used to demonstrate the sprouting of A-fibers in several earlier studies, also labels unmyelinated afferents (C-fibers) in lamina II and thin myelinated afferents in lamina I, when applied after peripheral nerve transection. The lamina II afferents also contained vasoactive intestinal polypeptide and galanin, two neuropeptides mainly expressed in small dorsal root ganglion (DRG) neurons and C-fibers. In an attempt to label large DRG neurons and A-fibers selectively, CTB was applied four days before axotomy (pre-injury-labelling), and sprouting was monitored after axotomy. We found that only a small number of A-fibers sprouted into inner lamina II, a region normally innervated by C-fibers, but not into outer lamina II or lamina I. Such sprouts made synaptic contact with dendrites in inner lamina II. Neuropeptide Y (NPY) was found in these sprouts in inner lamina II, an area very rich in Y1 receptor-positive processes. These results suggest that axotomy-induced sprouting from deeper to superficial layers is much less pronounced than previously assumed, in fact it is only marginal. This limited reorganization involves large NPY immunoreactive DRG neurons sprouting into the Y1 receptor-rich inner lamina II. Even if quantitatively small, it cannot be excluded that this represents a functional circuitry involved in neuropathic pain.


Asunto(s)
Vías Aferentes/fisiología , Fibras Nerviosas Mielínicas/fisiología , Regeneración Nerviosa/fisiología , Plasticidad Neuronal/fisiología , Células del Asta Posterior/fisiología , Raíces Nerviosas Espinales/fisiología , Vías Aferentes/ultraestructura , Animales , Células Cultivadas , Toxina del Cólera/metabolismo , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Inmunohistoquímica , Masculino , Microscopía Electrónica , Compresión Nerviosa , Fibras Nerviosas Mielínicas/ultraestructura , Fibras Nerviosas Amielínicas/fisiología , Fibras Nerviosas Amielínicas/ultraestructura , Proteínas de Neurofilamentos/metabolismo , Neuropéptido Y/metabolismo , Células del Asta Posterior/ultraestructura , Terminales Presinápticos/fisiología , Terminales Presinápticos/ultraestructura , Ratas , Ratas Sprague-Dawley , Receptores de Neuropéptido Y/metabolismo , Nervio Ciático/lesiones , Nervio Ciático/fisiología , Nervio Ciático/cirugía , Raíces Nerviosas Espinales/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...