首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
The N-methyl-D-aspartate receptor (NMDAR) ion channel plays a pivotal role in the pathology of ischemic stroke. The functional receptor consists of two GluN1 subunits (a-h) and two GluN2 subunits (A/B/C/D), the expression of which are spatially and temporally regulated in pathological and physiological conditions. While the roles of the GluN2A and GluN2B subunit in ischemic stroke have been well developed, the role of the GluN2C subunit in ischemia is not well understood. Following middle carotid artery occlusion (MCAO), GluN2C-/- male mice displayed similar volumes of infarct as wild-type (WT) mice. However, GluN2C-/- mice showed decreased cerebral edema and an enhanced rate of neurological recovery compared to WT mice. The ischemic penumbra of GluN2C-/- mice showed fewer cytoarchitectural deficits and decreased tauopathy relative to WT mice. These neuroprotective changes in GluN2C-/- mice also corresponded with decreased expression of Fyn kinase and decreased phosphorylation of GluN2B subunit at Tyr1336. Lastly, a GluN2C deficiency modified the NMDAR/pro-survival signaling axis, as shown by increased levels of nuclear CREB(P-Ser133). Thus, the GluN2C subunit enhances ischemic stroke pathology by promoting neuronal dysfunction in the penumbra region.  相似文献   

2.
NMDARs (N-methyl-D-aspartate receptors) mediate the predominantly excitatory neurotransmission in the CNS (central nervous system). Excessive release of glutamate and overactivation of NMDARs during brain ischemia and the hypoxia process are causally linked to excitotoxicity and neuronal damage. GluN3 subunits, the third member of the NMDAR family with two isoforms, GluN3A and GluN3B, have been confirmed to display an inhibitory effect on NMDAR activity. However, the effect of GluN3 subunits in brain ischemia and hypoxia is not clearly understood. In the present study, the influence of ischemia and hypoxia on GluN3 subunit expression was observed by using the 2VO (two-vessel occlusion) rat brain ischemia model and cell OGD (oxygen and glucose deprivation) hypoxia model. It was found that GluN3A protein expression in rat hippocampus and the prefrontal cortex was increased quickly after brain ischemia and remained at a high level for at least 24 h. However, the expression of the GluN3B subunit was not remarkably changed in both the animal and cell models. After OGD exposure, rat hippocampal neurons with GluN3A subunit overexpression displayed more viability than the wild-type neurons. NG108-15 cells overexpressing GluN3A presented pronounced resistance to glutamate insult. Blocking the increase of intracellular Ca2+ concentration may underlie the neuroprotective mechanism of up-regulated GluN3A subunit. Suppressing the generation of hydroxyl radicals and NO (nitric oxide) is probably also involved in the neuroprotection.  相似文献   

3.
GluN2A and GluN2B are the major subunits of functional NMDA receptors (NMDAR). Previous studies have suggested that GluN2A and GluN2B may differentially mediate NMDAR function at synaptic and extrasynaptic locations and play opposing roles in excitotoxicity, such as neurodegeneration triggered by ischemic stroke and brain injury. By using pharmacological and molecular approaches to suppress or enhance the function of GluN2A and GluN2B in cultured cortical neurons, we examined NMDAR-mediated, bidirectional regulation of prosurvival signaling (i.e. the cAMP response element-binding protein (CREB)-Bdnf cascade) and cell death. Inhibition of GluN2A or GluN2B attenuated the up-regulation of prosurvival signaling triggered by the activation of either synaptic or extrasynaptic NMDAR. Inhibition of GluN2A or GluN2B also attenuated the down-regulation of prosurvival signaling triggered by the coactivation of synaptic and extrasynaptic receptors. The effects of GluN2B on CREB-Bdnf signaling were larger than those of GluN2A. Consistently, compared with suppression of GluN2A, suppression of GluN2B resulted in more reduction of NMDA- and oxygen glucose deprivation-induced excitotoxicity as well as NMDAR-mediated elevation of intracellular calcium. Moreover, excitotoxicity and down-regulation of CREB were exaggerated in neurons overexpressing GluN2A or GluN2B. Together, we found that GluN2A and GluN2B are involved in the function of both synaptic and extrasynaptic NMDAR, demonstrating that they play similar rather than opposing roles in NMDAR-mediated bidirectional regulation of prosurvival signaling and neuronal death.  相似文献   

4.
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to debilitating cognitive deficits. Recent evidence demonstrates that glutamate receptors are dysregulated by amyloid beta peptide (Aβ) oligomers, resulting in disruption of glutamatergic synaptic transmission which parallels early cognitive deficits. Although it is well accepted that neuronal death in AD is related to disturbed intracellular Ca(2+) (Ca(2+)(i)) homeostasis, little is known about the contribution of NMDARs containing GluN2A or GluN2B subunits on Aβ-induced Ca(2+)(i) rise and neuronal dysfunction. Thus, the main goal of this work was to evaluate the role of NMDAR subunits in dysregulation of Ca(2+)(i) homeostasis induced by Aβ 1-42 preparation containing both oligomers (in higher percentage) and monomers in rat cerebral cortical neurons. The involvement of NMDARs was evaluated by pharmacological inhibition with MK-801 or the selective GluN2A and GLUN2B subunit antagonists NVP-AAM077 and ifenprodil, respectively. We show that Aβ, like NMDA, increase Ca(2+)(i) levels mainly through activation of NMDARs containing GluN2B subunits. Conversely, GluN2A-NMDARs antagonism potentiates Ca(2+)(i) rise induced by a high concentration of Aβ (1μM), suggesting that GluN2A and GluN2B subunits have opposite roles in regulating Ca(2+)(i) homeostasis. Moreover, Aβ modulate NMDA-induced responses and vice versa. Indeed, pre-exposure to Aβ (1μM) decrease NMDA-evoked Ca(2+)(I) rise and pre-exposure to NMDA decrease Aβ response. Interestingly, simultaneous addition of Aβ and NMDA potentiate Ca(2+)(I) levels, this effect being regulated by GluN2A and GluN2B subunits in opposite manners. This study contributes to the understanding of the molecular basis of early AD pathogenesis, by exploring the role of GluN2A and GluN2B subunits in the mechanism of Aβ toxicity in AD.  相似文献   

5.
The N-methyl-D-aspartate receptor (NMDAR) in adult forebrain is a heterotetramer mainly composed of two GluN1 subunits and two GluN2A and/or GluN2B subunits. The synaptic expression and relative numbers of GluN2A- and GluN2B-containing NMDARs play critical roles in controlling Ca2+-dependent signaling and synaptic plasticity. Previous studies have suggested that the synaptic trafficking of NMDAR subtypes is differentially regulated, but the precise molecular mechanism is not yet clear. In this study, we demonstrated that Bip, an endoplasmic reticulum (ER) chaperone, selectively interacted with GluN2A and mediated the neuronal activity-induced assembly and synaptic incorporation of the GluN2A-containing NMDAR from dendritic ER. Furthermore, the GluN2A-specific synaptic trafficking was effectively disrupted by peptides interrupting the interaction between Bip and GluN2A. Interestingly, fear conditioning in mice was disrupted by intraperitoneal injection of the interfering peptide before training. In summary, we have uncovered a novel mechanism for the activity-dependent supply of synaptic GluN2A-containing NMDARs, and demonstrated its relevance to memory formation.  相似文献   

6.
Curcumin is one of the major compounds contained in turmeric, the powdered rhizome of Curcuma longa. Results obtained in various experimental models indicate that curcumin has the potential to treat a large variety of neuronal diseases. Excitotoxicity, the toxicity due to pathological glutamate receptors stimulation, has been considered to be involved in several ocular pathologies including ischemia, glaucoma, and diabetic retinopathy. The NMDA receptor (NMDAR), a heteromeric ligand-gated ion channel, is composed of GluN1 and GluN2 subunits. There are four GluN2 subunits (GluN2A-D), which are major determinants of the functional properties of NMDARs. It is widely accepted that GluN2B has a pivotal role in excitotoxicity while the role of GluN2A remains controversial. We previously demonstrated that curcumin is neuroprotective against NMDA-induced excitotoxicity with a mechanism involving an increase of GluN2A subunit activity. In this paper, we investigate the mechanisms involved in curcumin-induced GluN2A increase in retinal cultures. Our results show that curcumin treatment activated CaMKII with a time-course that paralleled those of GluN2A increase. Moreover, KN-93, a CaMKII inhibitor, was able to block the effect of curcumin on GluN2A expression. Finally, in our experimental model, curcumin reduced ser/thr phosphatases activity. Using okadaic acid, a specific PP1 and PP2A blocker, we observed an increase in GluN2A levels in cultures. The ability of okadaic acid to mimic the effect of curcumin on GluN2A expression suggests that curcumin might regulate GluN2A expression through a phosphatase-dependent mechanism. In conclusion, our findings indicate curcumin modulation of CaMKII and/or ser/thr phosphatases activities as a mechanism involved in GluN2A expression and neuroprotection against excitotoxicity.  相似文献   

7.
It is currently unclear whether the GluN2 subtype influences NMDA receptor (NMDAR) excitotoxicity. We report that the toxicity of NMDAR-mediated Ca(2+) influx is differentially controlled by the cytoplasmic C-terminal domains of GluN2B (CTD(2B)) and GluN2A (CTD(2A)). Studying the effects of acute expression of GluN2A/2B-based chimeric subunits with reciprocal exchanges of their CTDs revealed that CTD(2B) enhances NMDAR toxicity, compared to CTD(2A). Furthermore, the vulnerability of forebrain neurons in?vitro and in?vivo to NMDAR-dependent Ca(2+) influx is lowered by replacing the CTD of GluN2B with that of GluN2A by targeted exon exchange in a mouse knockin model. Mechanistically, CTD(2B) exhibits stronger physical/functional coupling to the PSD-95-nNOS pathway, which suppresses protective CREB activation. Dependence of NMDAR excitotoxicity on the GluN2 CTD subtype can be overcome by inducing high levels of NMDAR activity. Thus, the identity (2A versus 2B) of the GluN2 CTD controls the toxicity dose-response to episodes of NMDAR activity.  相似文献   

8.
Conantokin-G (con-G) is a small, gamma-carboxyglutamic acid (Gla)-containing peptide that functions neurophysiologically by inhibiting the N-methyl-d-aspartate receptor (NMDAR). In the current study, the receptor binding properties of an alanine-rich, Gla-deficient con-G variant, Ala-con-G, were assessed following tracer radioiodination with 125I. Direct binding experiments with [125I]Ala-con-G yielded a single site defined by a Kd value of 516 +/- 120 nm. Displacement of [125I]Ala-con-G binding by Ala-con-G resulted in 100% displacement with an IC50 value of 564 +/- 33 nm, while heterologous displacement by con-G[S16Y], con-G, con-T, and con-R[1-17] yielded IC50 values in the range of 15-45 microm. No displacement was observed with d-gamma-con-G or con-G[L5A], analogs that are inactive at NMDARs. Specific [125I]Ala-con-G binding was displaced by NMDA and 2-amino-5-phosphopentanoic acid in a dose-dependent manner, suggesting an interaction at the glutamate binding site. The direct binding of [125I]Ala-con-G to adult rat brain sections revealed an anatomical distribution of binding sites in all regions known to contain the NR2B subunit of the NMDAR. These results constitute the only known demonstration of the direct binding of a radiolabeled conantokin to the NMDARs present in rat brain membrane preparations and rat brain sections, and suggest that radiolabeled Ala-con-G, and similar conantokin derivatives, may find utility as probes of NMDARs in a variety of systems.  相似文献   

9.
Ketamine is a NMDA receptor (NMDAR) antagonist used in pediatric anesthesia. Given the role of glutamatergic signaling during brain maturation, we studied the effects of a single ketamine injection (40 mg/kg s.c) in mouse neonates depending on postnatal age at injection (P2, P5, or P10) on cortical NMDAR subunits expression and association with Membrane‐Associated Guanylate Kinases PSD95 and SAP102. The effects of ketamine injection at P2, P5, or P10 on motor activity were compared in adulthood. Ketamine increased GluN2A and GluN2B mRNA levels in P2‐treated mice without change in proteins, while it decreased GluN2B protein in P10‐treated mice without change in mRNA. Ketamine reduced GluN2A mRNA and protein levels in P5‐treated mice without change in GluN2B and GluN1. Ketamine affected the GluN2A/PSD95 association regardless of the age at injection, while GluN2B/PSD95 association was enhanced only in P5‐treated mice. Microdissection of ketamine‐treated mouse cortex showed a decrease in GluN2A mRNA level in superficial layers (I–IV) and an increase in all subunit expressions in deep layers (V–VI) in P5‐ and P10‐treated mice, respectively. Our data suggest that ketamine impairs cortical NMDAR subunit developmental profile and delays the synaptic targeting of GluN2A‐enriched NMDAR. Ketamine injection at P2 or P10 resulted in hyperlocomotion in adult male mice in an open field, without change in females. Voluntary running‐wheel exercise showed age‐ and sex‐dependent alterations of the mouse activity, especially during the dark phase. Overall, a single neonatal ketamine exposure led to short‐term NMDAR cortical developmental profile impairments and long‐term motor activity alterations persisting in adulthood. © 2014 Wiley Periodicals, Inc. Develop Neurobiol 75: 315–333, 2015  相似文献   

10.
The NMDA receptor, which is heavily involved in several human brain diseases, is a heteromeric ligand-gated ion channel that interacts with multiple intracellular proteins through the C-termini of different subunits. GluN2A and GluN2B are the two primary types of GluN2 subunits in the forebrain. During the developmental period, there is a switch from GluN2B- to GluN2A-containing NMDA receptors in synapses. In the adult brain, GluN2A exists at synaptic sites more abundantly than GluN2B. GluN2A plays important roles not only in synaptic plasticity but also in mediating physiological functions, such as learning and memory. GluN2A has also been involved in many common human diseases, such as cerebral ischemia, seizure disorder, Alzheimer’s disease, and systemic lupus erythematosus. The following review investigates the functional and molecular properties, physiological functions, and pathophysiological roles of the GluN2A subunit.  相似文献   

11.
The N-methyl-d-aspartate receptor (NMDAR) is involved in synaptic plasticity, learning, memory, and neurological diseases like epilepsy and it is the major mediator of excitotoxicity. Functional NMDARs in the mature brain are heteromeric complexes composed of different subunits: GluN1 and GluN2. There are four different GluN2 subunits (A–D) and each of them critically determines the pharmacological and electrophysiological properties of NMDARs. GluN1 is ubiquitously expressed in the central nervous system while the highest GluN2A expression is in the hippocampus. Adenosine, an endogenous anticonvulsant, is a neuromodulator with a critical role in the regulation of neuronal activity, mediating its effect on specific receptors, among which adenosine A1 receptor is highly expressed in the hippocampus. In the present work hippocampal GluN2A expression after the convulsant drug 3-mercaptopropionic acid (MP) induced seizures and the effect of cyclopentyladenosine (CPA) given alone or prior to MP (CPA + MP) in an acute or repetitive experimental model was studied. CPA administered to rats for one or 4 days increases seizure threshold induced by MP. After one administration of MP, no significant difference in GluN2A expression was observed in CPA and CPA + MP by Western blot, although immunohistochemistry revealed an increase in CA2/3 area. However, repetitive MP administration during 4 days showed a significant increase of GluN2A expression, and the repetitive administration of CPA 30 min prior to MP caused a significant decrease of GluN2A expression with respect to MP treatment, returning to control levels. These results show that GluN2A subunit is involved in repetitive MP-induced seizures, while CPA administration displays a protective effect against it.  相似文献   

12.
Glutamate neurotransmission via the N‐methyl‐d ‐aspartate receptor (NMDAR) is thought to mediate the synaptic plasticity underlying learning and memory formation. There is increasing evidence that deficits in NMDAR function are involved in the pathophysiology of cognitive dysfunction seen in neuropsychiatric disorders and addiction. NMDAR subunits confer different physiological properties to the receptor, interact with distinct intracellular postsynaptic scaffolding and signaling molecules, and are differentially expressed during development. Despite these known differences, the relative contribution of individual subunit composition to synaptic plasticity and learning is not fully elucidated. We have previously shown that constitutive deletion of GluN2A subunit in the mouse impairs discrimination and re‐learning phase of reversal when exemplars are complex picture stimuli, but spares acquisition and extinction of non‐discriminative visually cued instrumental response. To investigate the role of GluN2A containing NMDARs in executive control, we tested GluN2A knockout (GluN2AKO), heterozygous (GluN2AHET) and wild‐type (WT) littermates on an attentional set‐shifting task using species‐specific stimulus dimensions. To further explore the nature of deficits in this model, mice were tested on a visual discrimination reversal paradigm using simplified rotational stimuli. GluN2AKO were not impaired on discrimination or reversal problems when tactile or olfactory stimuli were used, or when visual stimuli were sufficiently easy to discriminate. GluN2AKO showed a specific and significant impairment in ventromedial prefrontal cortex‐mediated set‐shifting. Together these results support a role for GluN2A containing NMDAR in modulating executive control that can be masked by overlapping deficits in attentional processes during high task demands.  相似文献   

13.
The purpose of this study was to investigate neuroprotective efficiency of N-methyl D-aspartate (NMDA) receptor (NMDAR) blockade on the neuronal damage in the less studied and allegedly less affected CA3 hippocampus and striatum in the Mongolian gerbil model of global cerebral ischemia. The common carotid arteries of gerbils were occluded for 5, 10 or 15 minutes. Gerbils were given a low dose of non-competitive NMDA antagonist (MK-801, 3 mg/kg i.p.) or saline immediately after the occlusion in normothermic conditions. Neuronal damage was examined on 4th, 14th and 28th day after reperfusion. The effect of NMDAR blockade was followed in vivo by monitoring the neurological status of whole animals or at the cellular level by standard light- and confocal microscopy on brain slices. Increased duration of cerebral ischemia resulted in a progressive loss of striatal and CA3 hippocampal neurons. The most beneficial NMDAR blockade effect was observed when the neuronal damage was most severe - on the 28th day after 15-min ischemia. As judged by morphological and neurological data, the effect of ischemia is also apparent in the presumed less vulnerable regions (CA3 and striatum) which are functionally important in stroke plasticity. So, NMDAR blockade in normothermic conditions showed neuroprotective efficiency.  相似文献   

14.
Gong B  Wang YT 《The EMBO journal》2012,31(4):783-785
EMBO J 31 4, 805–816 (2012); published online December202011Synaptic plasticity, the activity-dependent modification of synaptic strength, plays a fundamental role in learning and memory as well as in developmental maturation of neuronal circuitry. However, how synaptic plasticity is induced and regulated remains poorly understood. In this issue of The EMBO Journal, Yang and colleagues present sets of exciting data, suggesting that G-protein-coupled receptors (GPCRs) selectively execute distinct signalling pathways to differentially regulate induction thresholds of hippocampal long-term potentiation (LTP) and long-term depression (LTD), thereby governing the direction of synaptic plasticity. These results shed significant light on our current understanding of how bidirectional synaptic plasticity is regulated.Synaptic plasticity has been demonstrated at synapses in various brain regions; the most well-characterized forms are LTP and LTD at hippocampal CA1 glutamatergic synapses (Collingridge et al, 2004). In experimental models, LTP and LTD can be, respectively, induced by high-frequency stimulation (HFS) and low-frequency stimulation (LFS) via activation of the N-methyl-D-aspartic acid (NMDA) subtype ionotropic glutamate receptor (NMDAR). However, how HFS and LFS activate NMDARs and thereby lead to synaptic plasticity remains poorly understood and highly controversial. It is even more unclear how the bidirectional synaptic plasticity is produced and regulated in response to physiological or pathological changes.Functional NMDARs consist primarily of two GluN1 subunits and two GluN2 subunits, with GluN2A and GluN2B subunits being the most common NMDAR subunits found in the cortical and hippocampal regions of the adult brain (Cull-Candy et al, 2001). GluN2A and GluN2B subunits may confer distinct gating and pharmacological properties to NMDARs and couple them to distinct intracellular signalling machineries (Cull-Candy et al, 2001). Moreover, the ratio of these two subpopulations of NMDARs at the glutamatergic synapse is dynamically regulated in an activity-dependent manner (Bellone and Nicoll, 2007; Cho et al, 2009; Xu et al, 2009). Although controversial, GluN2A- and GluN2B-containing NMDARs have been suggested to have differential roles in regulating the direction of synaptic plasticity (Collingridge et al, 2004; Morishita et al, 2007). Among the factors shown to regulate NMDAR function, Src family tyrosine kinases may be the best characterized, with both Src and Fyn able to upregulate NMDAR function, and thus LTP induction (Salter and Kalia, 2004). However, if these kinases modulate NMDAR function in a NMDAR subunit-specific manner remains unknown. To explore this concept, Yang et al (2012) investigated the potential subunit-specific regulation of NMDARs by Src and Fyn using whole-cell patch clamp recording of NMDAR-mediated currents from acutely dissociated CA1 hippocampal neurons or from rat hippocampal slices. They found that intracellular perfusion of recombinant Src or Fyn increased the NMDAR-mediated currents. By applying subunit-preferential antagonists of GluN2A- or GluN2B-containing NMDARs, or by using neurons obtained from GluN2A knockout mice, they discovered that Src and Fyn differentially enhanced currents gated through GluN2A- and GluN2B-containing NMDARs, respectively.Can physiological or pathological factors differentially activate Src or Fyn, thereby exerting subunit-specific regulation of NMDAR function? To answer this question, Yang et al focused their investigation on the role of GPCRs, specifically pituitary adenylate cyclase activating peptide receptor (PAC1R) and dopamine D1 receptor (D1R), both of which have recently been shown to potentiate NMDARs through Src family kinases (Macdonald et al, 2005; Hu et al, 2010). Indeed, they found that activation of PAC1R specifically increased GluN2A-NMDAR-mediated currents without affecting currents gated through GluN2B-NMDARs, and this potentiation was prevented by the Src-specific inhibitory peptide Src(40–58) (Salter and Kalia, 2004). To rule out the contribution of Fyn, the authors developed a novel-specific Fyn inhibitory peptide Fyn(39–57), and demonstrated that it had little effect on PAC1R potentiation. In contrast, activation of D1R potentiated GluN2B- (but not GluN2A-) NMDAR-mediated currents, and this potentiation was specifically eliminated by Fyn(39–57), but not by Src(40–58). The authors further demonstrated that stimulation of PAC1Rs resulted in a selective activation of Src kinase and consequent tyrosine phosphorylation of the GluN2A subunit, whereas activation of D1Rs led to a specific increase in Fyn-mediated tyrosine phosphorylation of the GluN2B subunit. To provide convincing evidence that these subunit-differential modulations are indeed the result of tyrosine phosphorylation of the respective NMDAR subunits, the authors then performed electrophysiological experiments using neurons from two knockin mouse lines GluN2A(Y1325F) and GluN2B(Y1472F), in which the tyrosine phosphorylation residues in native GluN2A and GluN2B subunits were, respectively, replaced with non-phosphorylatable phenylalanine residues. As expected, the authors found that PAC1R-mediated potentiation of NMDA currents was lost in neurons from GluN2A(Y1325F) mice (but maintained in neurons from GluN2B(Y1472F) mice), while D1R-mediated enhancement of NMDA currents was only observed in neurons from GluN2A(Y1325F) mice. Together, as illustrated in Figure 1, the authors have made a very convincing case that PAC1R and D1R, respectively, enhance function of GluN2A- and GluN2B-containing NMDARs by differentially activating Src- and Fyn-mediated phosphorylation of respective NMDAR subunits.Open in a separate windowFigure 1GPCRs regulate the direction of synaptic plasticity via activating distinct signalling pathways. Synaptic NMDA receptors, both GluN2A- and GluN2B-containing, play key roles in the induction of various forms of synaptic plasticity at the hippocampal CA1 glutamatergic synapse. Under the basal level of GluN2A and GluN2B ratio, stimulation with a train of pulses at frequencies from 1 to 100 Hz produces a frequency and plasticity (LTD–LTP) curve, with maximum LTD and LTP being, respectively, induced at 1 and 100 Hz. Activation of PAC1R with its agonist PACAP38 activates Src and thereby results in tyrosine phosphorylation and consequent functional upregulation of GluN2A-containing NMDARs, resulting in an increase in the ratio of functional GluN2A and GluN2B. The increased ratio in turn causes a left shift of frequency and plasticity curve, favouring LTP induction. In contrast, activation of D1R by the receptor agonist SKF81297 triggers Fyn-specific tyrosine phosphorylation and functional upregulation of GluN2B, causing a reduction of GluN2A and GluN2B ratio. This decreased ratio results in a right shift of the curve, favouring LTD induction. The ability of GPCRs to differentially activate distinct downstream signalling pathways involved in synaptic plasticity suggests the potential roles of GPCRs in governing the direction of synaptic plasticity.Given the coupling of NMDARs to the induction of synaptic plasticity, it is then reasonable to ask if activation of the two GPCRs can selectively affect the induction of LTP or LTD at CA1 synapses. Yang and colleagues investigated the effects of pharmacological activation of PAC1R and D1R on the induction of LTP and LTD by recording the field excitatory postsynaptic potentials from hippocampal slices. Consistent with differential roles of NMDAR subunits in governing directions of synaptic plasticity, the authors observed that activation of PAC1Rs reduces the induction threshold of LTP, while stimulation of D1Rs favours LTD induction (Figure 1). Facilitation of LTP by PAC1R and LTD by D1R were, respectively, prevented in the brain slices obtained from GluN2A(Y1325F) and GluN2B(Y1472F) knockin mice, supporting the differential involvements of Src-mediated GluN2A phosphorylation and Fyn-mediated GluN2B phosphorylation.Taken together, the authors'' results have demonstrated that activation of PAC1R and D1R can control the direction of synaptic plasticity at the hippocampal CA1 synapse by differentially regulating NMDAREPSCs in a subunit-specific fashion (Figure 1). Specifically, PAC1R enhances the function of GluN2A-containing NMDARs by increasing Src phosphorylation of GluN2A subunit at Y1325, whereas D1R upregulates GluN2B-containing NMDARs through increased Fyn phosphorylation of GluN2B at Y1472. Moreover, by regulating the ratio of functional GluN2A- and GluN2B-containing NMDARs, PAC1R and D1R in turn modulate the direction of synaptic plasticity, favouring the production of LTP and LTD, respectively.While consistent with the recently proposed hypothesis that GluN2A and GluN2B may have preferential roles in the induction of hippocampal CA1 LTP and LTD (Collingridge et al, 2004; but see also Morishita et al, 2007), the current study further emphasizes the importance of GluN2A/GluN2B ratios in regulating LTP and LTD thresholds: increased ratio favours LTP, while reduced ratio promotes LTD. However, this seems to contradict some recent studies where the reduction and increase in the GluN2A/GluN2B ratio appeared to, respectively, favour LTP (Cho et al, 2009; Xu et al, 2009) and LTD (Xu et al, 2009). Therefore, the direction of plasticity change is likely modulated not only by the GluN2A/GluN2B ratio, but also by additional factors such as experimental conditions, developmental stages, and brain regions.Under many experimental conditions, LTP and LTD are usually induced by HFS and LFS stimulating protocols, respectively, but it remains essentially unknown how LTP and LTD are physiologically or pathologically generated in animals. To this end, the identification of different GPCRs as the endogenous upstream regulators of NMDA receptor subpopulations, and hence regulators of synaptic plasticity, is the major novelty of Yang and colleagues'' work. Future studies are needed to investigate if and how PAC1R and/or D1R are critically involved in the production of LTP or LTD in animals under physiological or pathological conditions. Given the fact that Src family kinases may be required for LTP induced by HFS in hippocampal slices (Salter and Kalia, 2004), an equally intriguing question would be whether these GPCRs are actually required for LTP/LTD induced by HFS/LFS experimental paradigms. In line with this conjecture, it would be interesting to determine if ligands for various GPCRs co-exist in the glutamatergic presynaptic terminals and, if so, can be differentially co-released with glutamate in a frequency-dependent manner, thereby contributing to either HFS-induced LTP or LFS-induced LTD.The findings by Yang and colleagues establish an exciting mechanistic model by which GPCRs can govern the direction of synaptic plasticity by determining the contributions of GluN2A- and GluN2B-NMDARs through differential tyrosine phosphorylation of respective NMDA receptor subtypes. Additional studies further validating this model under physiological and pathological conditions will greatly improve our understanding of the molecular mechanisms underlying synaptic plasticity and cognitive brain functions. In addition, NMDARs, depending on their subunit composition and/or subcellular localization, may also have complex roles in mediating neuronal survival and death (Lai et al, 2011). Considering that neurotoxicity produced by over-activation of NMDARs is widely accepted to be a common mechanism for neuronal loss in a number of acute brain injuries and chronic neurodegenerative diseases, Yang and colleagues'' finding of the differential regulation of NMDAR subunits by different GPCRs could have wider implications beyond synaptic plasticity.  相似文献   

15.
NMDA‐type glutamate receptors (NMDAR) are central actors in the plasticity of excitatory synapses. During adaptive processes, the number and composition of synaptic NMDAR can be rapidly modified, as in neonatal hippocampal synapses where a switch from predominant GluN2B‐ to GluN2A‐containing receptors is observed after the induction of long‐term potentiation (LTP). However, the cellular pathways by which surface NMDAR subtypes are dynamically regulated during activity‐dependent synaptic adaptations remain poorly understood. Using a combination of high‐resolution single nanoparticle imaging and electrophysiology, we show here that GluN2B‐NMDAR are dynamically redistributed away from glutamate synapses through increased lateral diffusion during LTP in immature neurons. Strikingly, preventing this activity‐dependent GluN2B‐NMDAR surface redistribution through cross‐linking, either with commercial or with autoimmune anti‐NMDA antibodies from patient with neuropsychiatric symptoms, affects the dynamics and spine accumulation of CaMKII and impairs LTP. Interestingly, the same impairments are observed when expressing a mutant of GluN2B‐NMDAR unable to bind CaMKII. We thus uncover a non‐canonical mechanism by which GluN2B‐NMDAR surface dynamics plays a critical role in the plasticity of maturing synapses through a direct interplay with CaMKII.  相似文献   

16.
GABA (gamma-aminobutyric-acid), the main inhibitory neurotransmitter in the adult brain, exerts depolarizing (excitatory) actions during development and this GABAergic depolarization cooperates with NMDARs (N-methyl-D-aspartate receptors) to drive spontaneous synchronous activity (SSA) that is fundamentally important for developing neuronal networks. Although GABAergic depolarization is known to assist in the activation of NMDARs during development, the subcellular localization of NMDARs relative to GABAergic synapses is still unknown. Here, we investigated the subcellular distribution of NMDARs in association with GABAergic synapses at the developmental stage when SSA is most prominent in mice. Using multiple immunofluorescent labeling and confocal laser-scanning microscopy in the developing mouse hippocampus, we found that NMDARs were associated with both glutamatergic and GABAergic synapses at postnatal day 6-7 and we observed a direct colocalization of GABA(A)- and NMDA-receptor labeling in GABAergic synapses. Electron microscopy of pre-embedding immunogold-immunoperoxidase reactions confirmed that GluN1, GluN2A and GluN2B NMDAR subunits were all expressed in glutamatergic and GABAergic synapses postsynaptically. Finally, quantitative post-embedding immunogold labeling revealed that the density of NMDARs was 3 times higher in glutamatergic than in GABAergic synapses. Since GABAergic synapses were larger, there was little difference in the total number of NMDA receptors in the two types of synapses. In addition, receptor density in synapses was substantially higher than extrasynaptically. These data can provide the neuroanatomical basis of a new interpretation of previous physiological data regarding the GABA(A)R-NMDAR cooperation during early development. We suggest that during SSA, synaptic GABA(A)R-mediated depolarization assists NMDAR activation right inside GABAergic synapses and this effective spatial cooperation of receptors and local change of membrane potential will reach developing glutamatergic synapses with a higher probability and efficiency even further away on the dendrites. This additional level of cooperation that operates within the depolarizing GABAergic synapse, may also allow its own modification triggered by Ca(2+)-influx through the NMDA receptors.  相似文献   

17.
18.
Abnormally high concentrations of extracellular glutamate in the brain may cause neuronal damage via excitotoxicity. Thus, tight regulation of glutamate release is critical to neuronal function and survival. Excitotoxicity is caused mainly by overactivation of the extrasynaptic NMDA receptor (NMDAR) and results in specific cellular changes, including calcium-induced activation of calpain proteases. Here, we report that presenilin-1 (PS1) null mouse cortical neuronal cultures have increased amounts of calpain-dependent spectrin breakdown products (SBDPs) compared with WT cultures. NMDAR antagonists blocked accumulation of SBDPs, suggesting abnormal activation of this receptor in PS1 null cultures. Importantly, an increase in SBDPs was detected in cultures of at least 7 days in vitro but not in younger cultures. Conditioned medium from PS1 null neuronal cultures at 8 days in vitro contained higher levels of glutamate than medium from WT cultures and stimulated production of SBDPs when added to WT cultures. Use of glutamate reuptake inhibitors indicated that accumulation of this neurotransmitter in the media of PS1 null cultures was due to increased rates of release. PS1 null neurons showed decreased cell surface expression and phosphorylation of the GluN2B subunit of NMDAR, indicating decreased amounts of extrasynaptic NMDAR in the absence of PS1. Inhibition of γ-secretase activity in WT neurons caused changes similar to those observed in PS1 null neurons. Together, these data indicate that the PS1/γ-secretase system regulates release of glutamate, tyrosine phosphorylation, and surface expression of GluN2B-containing NMDARs.  相似文献   

19.
N-Methyl-D-aspartate receptors (NMDARs), one of three main classes of ionotropic glutamate receptors, play major roles in synaptic plasticity, synaptogenesis, and excitotoxicity. Unlike non-NMDA receptors, NMDARs are thought to comprise obligatory heterotetrameric complexes mainly composed of GluN1 and GluN2 subunits. When expressed alone in heterogenous cells, such as HEK293 cells, most of the NMDAR subunits can neither leave the endoplasmic reticulum (ER) nor be expressed in the cell membrane because of the ER retention signals. Only when NMDARs are heteromerically assembled can the ER retention signals be masked and NMDARs be expressed in the surface membrane. However, the mechanisms underlying NMDAR assembly remain poorly understood. To identify regions in subunits that mediate this assembly, we made a series of truncated or chimeric cDNA constructs. Using FRET measurement in living cells combined with immunostaining and coimmunoprecipitation analysis, we examined the assembly-determining domains of NMDAR subunits. Our results indicate that the transmembrane region of subunits is necessary for the assembly of NMDAR subunits, both for the homodimer and the heteromer.  相似文献   

20.
N-Methyl-d-aspartate receptors (NMDARs) are known to be involved in a range of neurological and neurodegenerative disorders and consequently the development of compounds that modulate the function of these receptors has been the subject of intense interest. We have recently reported that 6-bromocoumarin-3-carboxylic acid (UBP608) is a negative allosteric modulator with weak selectivity for GluN2A-containing NMDARs. In the present study, a series of commercially available and newly synthesized coumarin derivatives have been evaluated in a structure-activity relationship (SAR) study as modulators of recombinant NMDAR activity. The main conclusions from this SAR study were that substituents as large as iodo were accommodated at the 6-position and that 6,8-dibromo or 6,8-diiodo substitution of the coumarin ring enhanced the inhibitory activity at NMDARs. These coumarin derivatives are therefore excellent starting points for the development of more potent and GluN2 subunit selective inhibitors, which may have application in the treatment of a range of neurological disorders such as neuropathic pain, epilepsy and depression. Surprisingly, 4-methyl substitution of UBP608 to give UBP714, led to conversion of the inhibitory activity of UBP608 into potentiating activity at recombinant GluN1/GluN2 receptors. UBP714 also enhanced NMDAR mediated field EPSPs in the CA1 region of the hippocampus. UBP714 is therefore a novel template for the development of potent and subunit selective NMDAR potentiators that may have therapeutic applicability in the treatment of patients with cognitive deficits or schizophrenia.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号