Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Elife ; 122024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727712

RESUMEN

Vesicles within presynaptic terminals are thought to be segregated into a variety of readily releasable and reserve pools. The nature of the pools and trafficking between them is not well understood, but pools that are slow to mobilize when synapses are active are often assumed to feed pools that are mobilized more quickly, in a series. However, electrophysiological studies of synaptic transmission have suggested instead a parallel organization where vesicles within slowly and quickly mobilized reserve pools would separately feed independent reluctant- and fast-releasing subdivisions of the readily releasable pool. Here, we use FM-dyes to confirm the existence of multiple reserve pools at hippocampal synapses and a parallel organization that prevents intermixing between the pools, even when stimulation is intense enough to drive exocytosis at the maximum rate. The experiments additionally demonstrate extensive heterogeneity among synapses in the relative sizes of the slowly and quickly mobilized reserve pools, which suggests equivalent heterogeneity in the numbers of reluctant and fast-releasing readily releasable vesicles that may be relevant for understanding information processing and storage.


Asunto(s)
Hipocampo , Sinapsis , Vesículas Sinápticas , Animales , Hipocampo/fisiología , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Ratas , Exocitosis , Terminales Presinápticos/fisiología
2.
Alzheimers Dement ; 19(9): 4264-4266, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37483152

RESUMEN

N-methyl-D-aspartate (NMDA) receptor (NMDAR) dysregulation is thought to contribute to impaired cognition and neurodegeneration in a variety of brain disorders. In a recent article, Zhong et al. proposed that deficiency of the NMDAR subunit GluN3A may be a primary pathogenic factor in sporadic Alzheimer´s disease (AD) based on evidence for degenerative excitotoxicity and cognitive impairment in aging mice lacking GluN3A. Because the result appeared to be at odds with earlier work where genetic GluN3A deletion enhanced learning in younger mice, we have now compared wild-type and GluN3A knockout mice at later life stages using a congenic mouse strain. Rather than age-dependent cognitive decline or neurodegeneration, we find that the enhanced performance of young adult GluN3A knockouts in memory tasks persists during aging. In sum, our analysis does not support the hypothesis that GluN3A loss underlies cognitive impairment in AD..


Asunto(s)
Disfunción Cognitiva , Ratones , Animales , Ratones Noqueados , Disfunción Cognitiva/genética , Receptores de N-Metil-D-Aspartato/genética
3.
Cell Rep ; 42(5): 112477, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37149869

RESUMEN

Signaling via N-methyl-d-aspartate receptors (NMDARs) is critical for the maturation of glutamatergic synapses, partly through a developmental switch from immature synapses expressing primarily GluN2B- and GluN3A-containing subtypes to GluN2A-rich mature ones. This subunit switch is thought to underlie the synaptic stabilization of NMDARs necessary for neural network consolidation. However, the cellular mechanisms controlling the NMDAR exchange remain unclear. Using a combination of single-molecule and confocal imaging and biochemical and electrophysiological approaches, we show that surface GluN3A-NMDARs form a highly diffusive receptor pool that is loosely anchored to synapses. Remarkably, changes in GluN3A subunit expression selectively alter the surface diffusion and synaptic anchoring of GluN2A- but not GluN2B-NMDARs, possibly through altered interactions with cell surface receptors. The effects of GluN3A on NMDAR surface diffusion are restricted to an early time window of postnatal development in rodents, allowing GluN3A subunits to control the timing of NMDAR signaling maturation and neuronal network refinements.


Asunto(s)
Hipocampo , Receptores de N-Metil-D-Aspartato , Hipocampo/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Transducción de Señal , Encéfalo/metabolismo
4.
J Vis Exp ; (202)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38163265

RESUMEN

Behavior is shaped by actions, and actions necessitate motor skills such as strength, coordination, and learning. None of the behaviors essential for sustaining life would be possible without the ability to transition from one position to another. Unfortunately, motor skills can be compromised in a wide array of diseases. Therefore, investigating the mechanisms of motor functions at the cellular, molecular, and circuit levels, as well as understanding the symptoms, causes, and progression of motor disorders, is crucial for developing effective treatments. Mouse models are frequently employed for this purpose. This article describes a protocol that allows the monitoring of various aspects of motor performance and learning in mice using an automated tool called the Erasmus Ladder. The assay involves two phases: an initial phase where mice are trained to navigate a horizontal ladder built of irregular rungs ("fine motor learning"), and a second phase where an obstacle is presented in the path of the moving animal. The perturbation can be unexpected ("challenged motor learning") or preceded by an auditory tone ("associative motor learning"). The task is easy to conduct and is fully supported by automated software. This report shows how different readouts from the test, when analyzed with sensitive statistical methods, allow fine monitoring of mouse motor skills using a small cohort of mice. We propose that the method will be highly sensitive to evaluate motor adaptations driven by environmental modifications as well as early-stage subtle motor deficits in mutant mice with compromised motor functions.


Asunto(s)
Aprendizaje , Destreza Motora , Humanos , Ratones , Animales , Condicionamiento Clásico , Programas Informáticos
5.
Neuron ; 110(15): 2438-2454.e8, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35700736

RESUMEN

GluN3A is an atypical glycine-binding subunit of NMDA receptors (NMDARs) whose actions in the brain are mostly unknown. Here, we show that the expression of GluN3A subunits controls the excitability of mouse adult cortical and amygdalar circuits via an unusual signaling mechanism involving the formation of excitatory glycine GluN1/GluN3A receptors (eGlyRs) and their tonic activation by extracellular glycine. eGlyRs are mostly extrasynaptic and reside in specific neuronal populations, including the principal cells of the basolateral amygdala (BLA) and SST-positive interneurons (SST-INs) of the neocortex. In the BLA, tonic eGlyR currents are sensitive to fear-conditioning protocols, are subject to neuromodulation by the dopaminergic system, and control the stability of fear memories. In the neocortex, eGlyRs control the in vivo spiking of SST-INs and the behavior-dependent modulation of cortical activity. GluN3A-containing eGlyRs thus represent a novel and widespread signaling modality in the adult brain, with attributes that strikingly depart from those of conventional NMDARs.


Asunto(s)
Amígdala del Cerebelo , Neocórtex , Receptores de Glicina , Receptores de N-Metil-D-Aspartato , Amígdala del Cerebelo/metabolismo , Animales , Corteza Cerebral/metabolismo , Glicina/metabolismo , Interneuronas/metabolismo , Ratones , Neocórtex/metabolismo , Neuronas/metabolismo , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
6.
J Physiol ; 600(2): 261-276, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33942912

RESUMEN

Non-conventional N-methyl-d-aspartate receptors (NMDARs) containing GluN3A subunits have unique biophysical, signalling and localization properties within the NMDAR family, and are typically thought to counterbalance functions of classical NMDARs made up of GluN1/2 subunits. Beyond their recognized roles in synapse refinement during postnatal development, recent evidence is building a wider perspective for GluN3A functions. Here we draw particular attention to the latest developments for this multifaceted and unusual subunit: from finely timed expression patterns that correlate with plasticity windows in developing brains or functional hierarchies in the mature brain to new insight onto presynaptic GluN3A-NMDARs, excitatory glycine receptors and behavioural impacts, alongside further connections to a range of brain disorders.


Asunto(s)
Receptores de Glicina , Receptores de N-Metil-D-Aspartato , Subunidades de Proteína/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Sinapsis/metabolismo
7.
Elife ; 102021 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-34787081

RESUMEN

De novo protein synthesis is required for synapse modifications underlying stable memory encoding. Yet neurons are highly compartmentalized cells and how protein synthesis can be regulated at the synapse level is unknown. Here, we characterize neuronal signaling complexes formed by the postsynaptic scaffold GIT1, the mechanistic target of rapamycin (mTOR) kinase, and Raptor that couple synaptic stimuli to mTOR-dependent protein synthesis; and identify NMDA receptors containing GluN3A subunits as key negative regulators of GIT1 binding to mTOR. Disruption of GIT1/mTOR complexes by enhancing GluN3A expression or silencing GIT1 inhibits synaptic mTOR activation and restricts the mTOR-dependent translation of specific activity-regulated mRNAs. Conversely, GluN3A removal enables complex formation, potentiates mTOR-dependent protein synthesis, and facilitates the consolidation of associative and spatial memories in mice. The memory enhancement becomes evident with light or spaced training, can be achieved by selectively deleting GluN3A from excitatory neurons during adulthood, and does not compromise other aspects of cognition such as memory flexibility or extinction. Our findings provide mechanistic insight into synaptic translational control and reveal a potentially selective target for cognitive enhancement.


Asunto(s)
Memoria/fisiología , Biosíntesis de Proteínas/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
8.
Cereb Cortex ; 31(4): 1914-1926, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33290502

RESUMEN

GluN3A subunits endow N-Methyl-D-Aspartate receptors (NMDARs) with unique biophysical, trafficking, and signaling properties. GluN3A-NMDARs are typically expressed during postnatal development, when they are thought to gate the refinement of neural circuits by inhibiting synapse maturation, and stabilization. Recent work suggests that GluN3A also operates in adult brains to control a variety of behaviors, yet a full spatiotemporal characterization of GluN3A expression is lacking. Here, we conducted a systematic analysis of Grin3a (gene encoding mouse GluN3A) mRNA expression in the mouse brain by combining high-sensitivity colorimetric and fluorescence in situ hybridization with labeling for neuronal subtypes. We find that, while Grin3a mRNA expression peaks postnatally, significant levels are retained into adulthood in specific brain regions such as the amygdala, medial habenula, association cortices, and high-order thalamic nuclei. The time-course of emergence and down-regulation of Grin3a expression varies across brain region, cortical layer of residence, and sensory modality, in a pattern that correlates with previously reported hierarchical gradients of brain maturation and functional specialization. Grin3a is expressed in both excitatory and inhibitory neurons, with strong mRNA levels being a distinguishing feature of somatostatin interneurons. Our study provides a comprehensive map of Grin3a distribution across the murine lifespan and paves the way for dissecting the diverse functions of GluN3A in health and disease.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Neuronas/metabolismo , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Receptores de N-Metil-D-Aspartato/biosíntesis , Factores de Edad , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de N-Metil-D-Aspartato/genética
9.
Brain Struct Funct ; 224(9): 3263-3276, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31667576

RESUMEN

Most vesicles in the interior of synaptic terminals are clustered in clouds close to active zone regions of the plasma membrane where exocytosis occurs. Electron-dense structures, termed bridges, have been reported between a small minority of pairs of neighboring vesicles within the clouds. Synapsin proteins have been implicated previously, but the existence of the bridges as stable structures in vivo has been questioned. Here we use electron tomography to show that the bridges are present but less frequent in synapsin knockouts compared to wildtype. An analysis of distances between neighbors in wildtype tomograms indicated that the bridges are strong enough to resist centrifugal forces likely induced by fixation with aldehydes. The results confirm that the bridges are stable structures and that synapsin proteins are involved in formation or stabilization.


Asunto(s)
Terminales Presinápticos/ultraestructura , Sinapsinas/metabolismo , Vesículas Sinápticas/ultraestructura , Animales , Ratones Noqueados , Modelos Neurológicos , Terminales Presinápticos/metabolismo , Sinapsinas/genética , Vesículas Sinápticas/metabolismo
11.
Elife ; 82019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-31090538

RESUMEN

Synaptophysins 1 and 2 and synaptogyrins 1 and 3 constitute a major family of synaptic vesicle membrane proteins. Unlike other widely expressed synaptic vesicle proteins such as vSNAREs and synaptotagmins, the primary function has not been resolved. Here, we report robust elevation in the probability of release of readily releasable vesicles with both high and low release probabilities at a variety of synapse types from knockout mice missing all four family members. Neither the number of readily releasable vesicles, nor the timing of recruitment to the readily releasable pool was affected. The results suggest that family members serve as negative regulators of neurotransmission, acting directly at the level of exocytosis to dampen connection strength selectively when presynaptic action potentials fire at low frequency. The widespread expression suggests that chemical synapses may play a frequency filtering role in biological computation that is more elemental than presently envisioned. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Asunto(s)
Neuronas/metabolismo , Vesículas Sinápticas/metabolismo , Sinaptogirinas/deficiencia , Sinaptofisina/deficiencia , Animales , Ratones Noqueados , Transmisión Sináptica
12.
Mol Ther ; 26(8): 1965-1972, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29914757

RESUMEN

Huntington's disease (HD) is a dominantly inherited neurodegenerative disease caused by expansion of a polyglutamine tract in the huntingtin protein. HD symptoms include severe motor, cognitive, and psychiatric impairments that result from dysfunction and later degeneration of medium-sized spiny neurons (MSNs) in the striatum. A key early pathogenic mechanism is dysregulated synaptic transmission due to enhanced surface expression of juvenile NMDA-type glutamate receptors containing GluN3A subunits, which trigger the aberrant pruning of synapses formed by cortical afferents onto MSNs. Here, we tested the therapeutic potential of silencing GluN3A expression in YAC128 mice, a well-established HD model. Recombinant adeno-associated viruses encoding a short-hairpin RNA against GluN3A (rAAV-shGluN3A) were generated, and the ability of different serotypes to transduce MSNs was compared. A single injection of rAAV9-shGluN3A into the striatum of 1-month-old mice drove potent (>90%) and long-lasting reductions of GluN3A expression in MSNs, prevented dendritic spine loss and improved motor performance in YAC128 mice. Later delivery, when spine pathology is already apparent, was also effective. Our data provide proof-of-concept for GluN3A silencing as a beneficial strategy to prevent or reverse corticostriatal disconnectivity and motor impairment in HD and support the use of RNAi-based or small-molecule approaches for harnessing this therapeutic potential.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/terapia , Tratamiento con ARN de Interferencia/métodos , Receptores de N-Metil-D-Aspartato/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Silenciador del Gen , Vectores Genéticos/administración & dosificación , Humanos , Enfermedad de Huntington/genética , Ratones , Mutación , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transducción Genética , Resultado del Tratamiento
13.
Mol Neurobiol ; 55(10): 7728-7742, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29460266

RESUMEN

Rictor associates with mTOR to form the mTORC2 complex, which activity regulates neuronal function and survival. Neurodegenerative diseases are characterized by the presence of neuronal dysfunction and cell death in specific brain regions such as for example Huntington's disease (HD), which is characterized by the loss of striatal projection neurons leading to motor dysfunction. Although HD is caused by the expression of mutant huntingtin, cell death occurs gradually suggesting that neurons have the capability to activate compensatory mechanisms to deal with neuronal dysfunction and later cell death. Here, we analyzed whether mTORC2 activity could be altered by the presence of mutant huntingtin. We observed that Rictor levels are specifically increased in the striatum of HD mouse models and in the putamen of HD patients. Rictor-mTOR interaction and the phosphorylation levels of Akt, one of the targets of the mTORC2 complex, were increased in the striatum of the R6/1 mouse model of HD suggesting increased mTORC2 signaling. Interestingly, acute downregulation of Rictor in striatal cells in vitro reduced mTORC2 activity, as shown by reduced levels of phospho-Akt, and increased mutant huntingtin-induced cell death. Accordingly, overexpression of Rictor increased mTORC2 activity counteracting cell death. Furthermore, normalization of endogenous Rictor levels in the striatum of R6/1 mouse worsened motor symptoms suggesting an induction of neuronal dysfunction. In conclusion, our results suggest that increased Rictor striatal levels could counteract neuronal dysfunction induced by mutant huntingtin.


Asunto(s)
Proteína Huntingtina/metabolismo , Proteínas Mutantes/metabolismo , Degeneración Nerviosa/patología , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Animales , Muerte Celular , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Masculino , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Ratones , Actividad Motora , Neostriado/metabolismo , Neostriado/patología , Degeneración Nerviosa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
14.
Nat Rev Neurosci ; 17(10): 623-35, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27558536

RESUMEN

GluN3-containing NMDA receptors (GluN3-NMDARs) are rarer than the 'classical' NMDARs, which are composed solely of GluN1 and GluN2 subunits, and have non-conventional biophysical, trafficking and signalling properties. In the CNS, they seem to have important roles in delaying synapse maturation until the arrival of sensory experience and in targeting non-used synapses for pruning. The reactivation of GluN3A expression at inappropriate ages may underlie maladaptive synaptic rearrangements observed in addiction, neurodegenerative diseases and other major brain disorders. Here, we discuss current evidence for these and other emerging roles for GluN3-NMDARs in the physiology and pathology of the CNS.


Asunto(s)
Encefalopatías/fisiopatología , Sistema Nervioso Central/fisiología , Subunidades de Proteína/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Encefalopatías/metabolismo , Sistema Nervioso Central/citología , Sistema Nervioso Central/patología , Humanos , Modelos Neurológicos , Receptores de N-Metil-D-Aspartato/fisiología , Transducción de Señal , Sinapsis/metabolismo , Sinapsis/fisiología
15.
Neurobiol Dis ; 93: 47-56, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27072890

RESUMEN

Age-inappropriate expression of juvenile NMDA receptors (NMDARs) containing GluN3A subunits has been linked to synapse loss and death of spiny projection neurons of the striatum (SPNs) in Huntington's disease (HD). Here we show that suppressing GluN3A expression prevents a multivariate synaptic transmission phenotype that precedes morphological signs at early prodromal stages. We start by confirming that afferent fiber stimulation elicits larger synaptic responses mediated by both AMPA receptors and NMDARs in SPNs in the YAC128 mouse model of HD. We then show that the enhancement mediated by both is fully prevented by suppressing GluN3A expression. Strong fiber-stimulation unexpectedly elicited robust NMDAR-mediated electrogenic events (termed "upstates" or "NMDA spikes"), and the effective threshold for induction was more than 2-fold lower in YAC128 SPNs because of the enhanced synaptic transmission. The threshold could be restored to control levels by suppressing GluN3A expression or by applying the weak NMDAR blocker memantine. However, the threshold was not affected by preventing glutamate spillover from synaptic clefts. Instead, long-lasting NMDAR responses interpreted previously as activation of extrasynaptic receptors by spilled-over glutamate were caused by NMDA spikes occurring in voltage clamp mode as escape potentials. Together, the results implicate GluN3A reactivation in a broad spectrum of early-stage synaptic transmission deficits in YAC128 mice; question the current concept that NMDAR mislocalization is the pathological trigger in HD; and introduce NMDA spikes as a new candidate mechanism for coupling NMDARs to neurodegeneration.


Asunto(s)
Enfermedad de Huntington/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transmisión Sináptica , Animales , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Ácido Glutámico/metabolismo , Enfermedad de Huntington/genética , Memantina/farmacología , Ratones , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo , Transmisión Sináptica/efectos de los fármacos
16.
JAMA Neurol ; 72(4): 468-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25686081

RESUMEN

Huntington disease (HD) is an inherited neurodegenerative disorder with no cure or effective palliative treatment. An ideal therapy would arrest pathogenesis at early stages before neuronal damage occurs. However, although the genetic mutation that causes HD is known, the molecular chain of events that leads from the mutation to disease is not well understood. Accumulating evidence suggests that synaptic dysregulation may be involved, and the earliest known deficit is hyperfunction of glutamate-type N-methyl-d-aspartate receptors (NMDARs) in the selectively vulnerable medium spiny neurons of the striatum. A previous study found that the mutant Htt protein interferes with downregulation of juvenile NMDAR subtypes that contain GluN3A subunits by sequestering the endocytic adaptor PACSIN1 and preventing their removal from the cell surface. Loss of PACSIN1 and consequent gain of GluN3A function reactivate a synapse pruning mechanism that is important during development but harmful when active at later stages. Suppressing the GluN3A reactivation corrected the NMDAR hyperfunction and prevented the full range of HD signs and symptoms in mouse models, encouraging efforts to develop GluN3A-selective antagonists and/or explore alternative therapeutic approaches to block GluN3A expression.


Asunto(s)
Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/genética , Proteínas del Tejido Nervioso/genética , Neuronas/patología
17.
J Neurosci ; 34(28): 9213-21, 2014 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25009255

RESUMEN

Synaptic rearrangements during critical periods of postnatal brain development rely on the correct formation, strengthening, and elimination of synapses and associated dendritic spines to form functional networks. The correct balance of these processes is thought to be regulated by synapse-specific changes in the subunit composition of NMDA-type glutamate receptors (NMDARs). Among these, the nonconventional NMDAR subunit GluN3A has been suggested to play a role as a molecular brake in synaptic maturation. We tested here this hypothesis using confocal time-lapse imaging in rat hippocampal organotypic slices and assessed the role of GluN3A-containing NMDARs on spine dynamics. We found that overexpressing GluN3A reduced spine density over time, increased spine elimination, and decreased spine stability. The effect of GluN3A overexpression could be further enhanced by using an endocytosis-deficient GluN3A mutant and reproduced by silencing the adaptor protein PACSIN1, which prevents the endocytosis of endogenous GluN3A. Conversely, silencing of GluN3A reduced spine elimination and favored spine stability. Moreover, reexpression of GluN3A in more mature tissue reinstated an increased spine pruning and a low spine stability. Mechanistically, the decreased stability in GluN3A overexpressing neurons could be linked to a failure of plasticity-inducing protocols to selectively stabilize spines and was dependent on the ability of GluN3A to bind the postsynaptic scaffold GIT1. Together, these data provide strong evidence that GluN3A prevents the activity-dependent stabilization of synapses thereby promoting spine pruning, and suggest that GluN3A expression operates as a molecular signal for controlling the extent and timing of synapse maturation.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Espinas Dendríticas/fisiología , Espinas Dendríticas/ultraestructura , Hipocampo/ultraestructura , Glicoproteínas de Membrana/metabolismo , Transmisión Sináptica/fisiología , Potenciales de Acción/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Hipocampo/fisiología , Masculino , Plasticidad Neuronal/fisiología , Ratas
18.
Proc Natl Acad Sci U S A ; 110(51): 20807-12, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297929

RESUMEN

NMDA-type glutamate receptors (NMDARs) guide the activity-dependent remodeling of excitatory synapses and associated dendritic spines during critical periods of postnatal brain development. Whereas mature NMDARs composed of GluN1 and GluN2 subunits mediate synapse plasticity and promote spine growth and stabilization, juvenile NMDARs containing GluN3A subunits are thought to inhibit these processes via yet unknown mechanisms. Here, we report that GluN3A binds G protein-coupled receptor kinase-interacting protein (GIT1), a postsynaptic scaffold that assembles actin regulatory complexes, including the Rac1 guanine nucleotide exchange factor ßPIX, to promote Rac1 activation in spines. Binding to GluN3A limits the synaptic localization of GIT1 and its ability to complex ßPIX, leading to decreased Rac1 activation and reduced spine density and size in primary cultured neurons. Conversely, knocking out GluN3A favors the formation of GIT1/ßPIX complexes and increases the activation of Rac1 and its main effector p21-activated kinase. We further show that binding of GluN3A to GIT1 is regulated by synaptic activity, a response that might restrict the negative regulatory effects of GluN3A on actin signaling to inactive synapses. Our results identify inhibition of Rac1/p21-activated kinase actin signaling pathways as an activity-dependent mechanism mediating the inhibitory effects of GluN3A on spine morphogenesis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Fosfoproteínas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/fisiología , Columna Vertebral/embriología , Sinapsis/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Células Cultivadas , Activación Enzimática/fisiología , Morfogénesis/fisiología , Fosfoproteínas/genética , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Columna Vertebral/citología , Sinapsis/genética , Proteína de Unión al GTP rac1/genética
19.
Neuron ; 80(4): 1025-38, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24183704

RESUMEN

Drug-evoked synaptic plasticity in the mesolimbic dopamine (DA) system reorganizes neural circuits that may lead to addictive behavior. The first cocaine exposure potentiates AMPAR excitatory postsynaptic currents (EPSCs) onto DA neurons of the VTA but reduces the amplitude of NMDAR-EPSCs. While plasticity of AMPAR transmission is expressed by insertion of calcium (Ca(2+))-permeable GluA2-lacking receptors, little is known about the expression mechanism for altered NMDAR transmission. Combining ex vivo patch-clamp recordings, mouse genetics, and subcellular Ca(2+) imaging, we observe that cocaine drives the insertion of NMDARs that are quasi-Ca(2+)-impermeable and contain GluN3A and GluN2B subunits. These GluN3A-containing NMDARs appear necessary for the expression of cocaine-evoked plasticity of AMPARs. We identify an mGluR1-dependent mechanism to remove these noncanonical NMDARs that requires Homer/Shank interaction and protein synthesis. Our data provide insight into the early cocaine-driven reorganization of glutamatergic transmission onto DA neurons and offer GluN3A-containing NMDARs as new targets in drug addiction.


Asunto(s)
Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/fisiología , Sinapsis/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Calcio/metabolismo , Neuronas Dopaminérgicas/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Femenino , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Técnicas de Placa-Clamp , Interferencia de ARN , Receptores AMPA/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Técnicas Estereotáxicas , Transmisión Sináptica/efectos de los fármacos
20.
Nat Med ; 19(8): 1030-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23852340

RESUMEN

Huntington's disease is caused by an expanded polyglutamine repeat in the huntingtin protein (HTT), but the pathophysiological sequence of events that trigger synaptic failure and neuronal loss are not fully understood. Alterations in N-methyl-D-aspartate (NMDA)-type glutamate receptors (NMDARs) have been implicated. Yet, it remains unclear how the HTT mutation affects NMDAR function, and direct evidence for a causative role is missing. Here we show that mutant HTT redirects an intracellular store of juvenile NMDARs containing GluN3A subunits to the surface of striatal neurons by sequestering and disrupting the subcellular localization of the endocytic adaptor PACSIN1, which is specific for GluN3A. Overexpressing GluN3A in wild-type mouse striatum mimicked the synapse loss observed in Huntington's disease mouse models, whereas genetic deletion of GluN3A prevented synapse degeneration, ameliorated motor and cognitive decline and reduced striatal atrophy and neuronal loss in the YAC128 Huntington's disease mouse model. Furthermore, GluN3A deletion corrected the abnormally enhanced NMDAR currents, which have been linked to cell death in Huntington's disease and other neurodegenerative conditions. Our findings reveal an early pathogenic role of GluN3A dysregulation in Huntington's disease and suggest that therapies targeting GluN3A or pathogenic HTT-PACSIN1 interactions might prevent or delay disease progression.


Asunto(s)
Conducta Animal , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Muerte Celular/efectos de los fármacos , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Eliminación de Gen , Células HEK293 , Humanos , Enfermedad de Huntington/fisiopatología , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Ratones , Actividad Motora/efectos de los fármacos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas Mutantes/toxicidad , Neostriado/metabolismo , Neostriado/patología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Neuropéptidos/metabolismo , Fosfoproteínas/metabolismo , Unión Proteica/efectos de los fármacos , Estructura Cuaternaria de Proteína , Prueba de Desempeño de Rotación con Aceleración Constante , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA