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 31 毫秒
1.
Kim MJ  Dunah AW  Wang YT  Sheng M 《Neuron》2005,46(5):745-760
NMDA receptors (NMDARs) control bidirectional synaptic plasticity by regulating postsynaptic AMPA receptors (AMPARs). Here we show that NMDAR activation can have differential effects on AMPAR trafficking, depending on the subunit composition of NMDARs. In mature cultured neurons, NR2A-NMDARs promote, whereas NR2B-NMDARs inhibit, the surface expression of GluR1, primarily by regulating its surface insertion. In mature neurons, NR2B is coupled to inhibition rather than activation of the Ras-ERK pathway, which drives surface delivery of GluR1. Moreover, the synaptic Ras GTPase activating protein (GAP) SynGAP is selectively associated with NR2B-NMDARs in brain and is required for inhibition of NMDAR-dependent ERK activation. Preferential coupling of NR2B to SynGAP could explain the subtype-specific function of NR2B-NMDARs in inhibition of Ras-ERK, removal of synaptic AMPARs, and weakening of synaptic transmission.  相似文献   

2.
The N-methyl-D-aspartate receptor (NMDAR), a major excitatory ligand-gated ion channel in the central nervous system (CNS), is a principal mediator of synaptic plasticity. Here we report that neuropilin tolloid-like 1 (Neto1), a complement C1r/C1s, Uegf, Bmp1 (CUB) domain-containing transmembrane protein, is a novel component of the NMDAR complex critical for maintaining the abundance of NR2A-containing NMDARs in the postsynaptic density. Neto1-null mice have depressed long-term potentiation (LTP) at Schaffer collateral-CA1 synapses, with the subunit dependency of LTP induction switching from the normal predominance of NR2A- to NR2B-NMDARs. NMDAR-dependent spatial learning and memory is depressed in Neto1-null mice, indicating that Neto1 regulates NMDA receptor-dependent synaptic plasticity and cognition. Remarkably, we also found that the deficits in LTP, learning, and memory in Neto1-null mice were rescued by the ampakine CX546 at doses without effect in wild-type. Together, our results establish the principle that auxiliary proteins are required for the normal abundance of NMDAR subunits at synapses, and demonstrate that an inherited learning defect can be rescued pharmacologically, a finding with therapeutic implications for humans.  相似文献   

3.

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 hippocamus 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.  相似文献   

4.
Regulation of neuronal NMDA receptor (NMDAR) is critical in synaptic transmission and plasticity. Protein kinase C (PKC) promotes NMDAR trafficking to the cell surface via interaction with NMDAR-associated proteins (NAPs). Little is known, however, about the NAPs that are critical to PKC-induced NMDAR trafficking. Here, we showed that calcium/calmodulin-dependent protein kinase II (CaMKII) could be a NAP that mediates the potentiation of NMDAR trafficking by PKC. PKC activation promoted the level of autophosphorylated CaMKII and increased association with NMDARs, accompanied by functional NMDAR insertion, at postsynaptic sites. This potentiation, along with PKC-induced long term potentiation of the AMPA receptor-mediated response, was abolished by CaMKII antagonist or by disturbing the interaction between CaMKII and NR2A or NR2B. Further mutual occlusion experiments demonstrated that PKC and CaMKII share a common signaling pathway in the potentiation of NMDAR trafficking and long-term potentiation (LTP) induction. Our results revealed that PKC promotes NMDA receptor trafficking and induces synaptic plasticity through indirectly triggering CaMKII autophosphorylation and subsequent increased association with NMDARs.  相似文献   

5.
NMDA receptors (NMDARs) play a pivotal role in the regulation of neuronal communication and synaptic function in the central nervous system. The subunit composition and compartmental localization of NMDARs in neurons affect channel activity and downstream signaling. This review discusses the distinct NMDAR subtypes and their function at synaptic, perisynaptic, and extrasynaptic sites of excitatory and inhibitory neurons. Many neurons express more than one of the modulatory NR2 subunits that participate in the formation of di- and/or triheteromeric channel assemblies (e.g., NR1/NR2A, NR1/NR2B, and/or NR1/NR2A/NR2B). Depending on the subunit composition and presence or absence of intracellular binding partners along the postsynaptic membrane, these NMDAR subtypes are allocated to distinct synaptic inputs converging onto a neuron or are distributed differentially among synaptic or extrasynaptic sites. These sites can carry NR2A and NR2B subunits, supporting the hypothesis that the spatial distribution of scaffolding and signaling complexes critically determines the full spectrum of NMDAR signaling.The author thanks the Deutsche Forschungsgemeinschaft for financial support (Ko 1064/5).  相似文献   

6.
Excitatory synaptic transmission and plasticity are critically modulated by N-methyl-D-aspartate receptors (NMDARs). Activation of NMDARs elevates intracellular Ca(2+) affecting several downstream signaling pathways that involve Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). Importantly, NMDAR activation triggers CaMKII translocation to synaptic sites. NMDAR activation failed to induce Ca(2+) responses in hippocampal neurons lacking the mandatory NMDAR subunit NR1, and no EGFP-CaMKIIalpha translocation was observed. In cells solely expressing Ca(2+)-impermeable NMDARs containing NR1(N598R)-mutant subunits, prolonged NMDA application elevated internal Ca(2+) to the same degree as in wild-type controls, yet failed to translocate CaMKIIalpha. Brief local NMDA application evoked smaller Ca(2+) transients in dendritic spines of mutant compared to wild-type cells. CaMKIIalpha mutants that increase binding to synaptic sites, namely CaMKII-T286D and CaMKII-TT305/306VA, rescued the translocation in NR1(N598R) cells in a glutamate receptor-subtype-specific manner. We conclude that CaMKII translocation requires Ca(2+) entry directly through NMDARs, rather than other Ca(2+) sources activated by NMDARs. Together with the requirement for activated, possibly ligand-bound, NMDARs as CaMKII binding partners, this suggests that synaptic CaMKII accumulation is an input-specific signaling event.  相似文献   

7.
NMDA receptors (NMDARs) activation in the hippocampus and insular cortex is necessary for spatial memory formation. Recent studies suggest that localization of NMDARs to lipid rafts enhance their signalization, since the kinases that phosphorylate its subunits are present in larger proportion in lipid raft membrane microdomains. We sought to determine the possibility that NMDAR translocation to synaptic lipid rafts occurs during plasticity processes such as memory formation. Our results show that water maze training induces a rapid recruitment of NMDAR subunits (NR1, NR2A, NR2B) and PSD-95 to synaptic lipid rafts and decrease in the post-synaptic density plus an increase of NR2B phosphorylation at tyrosine 1472 in the rat insular cortex. In the hippocampus, spatial training induces selective translocation of NR1 and NR2A subunits to lipid rafts. These results suggest that NMDARs translocate from the soluble fraction of post-synaptic membrane (non-raft PSD) to synaptic lipid raft during spatial memory formation. The recruitment of NMDA receptors and other proteins to lipid rafts could be an important mechanism for increasing the efficiency of synaptic transmission during synaptic plasticity process.  相似文献   

8.
We recently found that ethanol-induced long-term facilitation (LTF) of NMDAR activity is mediated by NR2B-NMDARs and is observed in the dorsomedial striatum (DMS) but not in the dorsolateral striatum (DLS). We also showed that repeated administration of ethanol causes a long-lasting increase in NMDAR activity in the DMS, resulting from ethanol-mediated Fyn phosphorylation of NR2B subunits. In this addendum, we report that the different sensitivity of NMDARs to ethanol between the DMS and DLS is not attributed to the abundance of synaptic NR2B-NMDARs or differences in Fyn levels. We further show that LTF is specific for NR2B-, but not NR2A-NMDARs, and that the duration of the in vivo ethanol-mediated increase in NMDAR activity is associated with the period of ethanol exposure, but not with alteration in NR1 or NR2A protein levels. Together, these results suggest that upregulation of NR2B-NMDAR activity by ethanol is selective and that ethanol's effect on NMDAR activity is gradual and cumulative.  相似文献   

9.
The N-methyl-D-aspartate receptor (NMDAR) plays a critical role in synaptic plasticity and is one of the main targets for alcohol (ethanol) in the brain. Trafficking of the NMDAR is emerging as a key regulatory mechanism that underlies channel activity and synaptic plasticity. Here we show that exposure of hippocampal neurons to ethanol increases the internalization of the NR2A but not NR2B subunit of the NMDAR via the endocytic pathway. We further observed that ethanol exposure results in NR2A endocytosis through the activation of H-Ras and the inhibition of the tyrosine kinase Src. Importantly, ethanol treatment alters functional subunit composition from NR2A/NR2B- to mainly NR2B-containing NMDARs. Our results suggest that addictive drugs such as ethanol alter NMDAR trafficking and subunit composition. This may be an important mechanism by which ethanol exerts its effects on NMDARs to produce alcohol-induced aberrant plasticity.  相似文献   

10.
Muscarinic receptor activation facilitates the induction of synaptic plasticity and enhances cognitive function. However, the specific muscarinic receptor subtype involved and the critical intracellular signaling pathways engaged have remained controversial. Here, we show that the recently discovered highly selective allosteric M(1) receptor agonist 77-LH-28-1 facilitates long-term potentiation (LTP) induced by theta burst stimulation at Schaffer collateral synapses in the hippocampus. Similarly, release of acetylcholine by stimulation of cholinergic fibers facilitates LTP via activation of M(1) receptors. N-methyl-D-aspartate receptor (NMDAR) opening during theta burst stimulation was enhanced by M(1) receptor activation, indicating this is the mechanism for LTP facilitation. M(1) receptors were found to enhance NMDAR activation by inhibiting SK channels that otherwise act to hyperpolarize postsynaptic spines and inhibit NMDAR opening. Thus, we describe a mechanism where M(1) receptor activation inhibits SK channels, allowing enhanced NMDAR activity and leading to a facilitation of LTP induction in the hippocampus.  相似文献   

11.
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   

12.
Tse YC  Bagot RC  Hutter JA  Wong AS  Wong TP 《PloS one》2011,6(11):e27215
Stress exerts a profound impact on learning and memory, in part, through the actions of adrenal corticosterone (CORT) on synaptic plasticity, a cellular model of learning and memory. Increasing findings suggest that CORT exerts its impact on synaptic plasticity by altering the functional properties of glutamate receptors, which include changes in the motility and function of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid subtype of glutamate receptor (AMPAR) that are responsible for the expression of synaptic plasticity. Here we provide evidence that CORT could also regulate synaptic plasticity by modulating the function of synaptic N-methyl-D-aspartate receptors (NMDARs), which mediate the induction of synaptic plasticity. We found that stress level CORT applied to adult rat hippocampal slices potentiated evoked NMDAR-mediated synaptic responses within 30 min. Surprisingly, following this fast-onset change, we observed a slow-onset (>1 hour after termination of CORT exposure) increase in synaptic expression of GluN2A-containing NMDARs. To investigate the consequences of the distinct fast- and slow-onset modulation of NMDARs for synaptic plasticity, we examined the formation of long-term potentiation (LTP) and long-term depression (LTD) within relevant time windows. Paralleling the increased NMDAR function, both LTP and LTD were facilitated during CORT treatment. However, 1-2 hours after CORT treatment when synaptic expression of GluN2A-containing NMDARs is increased, bidirectional plasticity was no longer facilitated. Our findings reveal the remarkable plasticity of NMDARs in the adult hippocampus in response to CORT. CORT-mediated slow-onset increase in GluN2A in hippocampal synapses could be a homeostatic mechanism to normalize synaptic plasticity following fast-onset stress-induced facilitation.  相似文献   

13.
Previous studies have shown that Csk plays critical roles in the regulation of neural development, differentiation and glutamate-mediated synaptic plasticity. It has been found that Csk associates with the NR2A and 2B subunits of N-methyl-D-aspartate receptors (NMDARs) in a Src activity-dependent manner and serves as an intrinsic mechanism to provide a “brake” on the induction of long-term synaptic potentiation (LTP) mediated by NMDARs. In contrast to the NR2A and 2B subunits, no apparent tyrosine phosphorylation is found in the NR1 subunit of NMDARs. Here, we report that Csk can also associate with the NR1 subunit in a Src activity-dependent manner. The truncation of the NR1 subunit C-tail which contains only one tyrosine (Y837) significantly reduced the Csk association with the NR1-1a/NR2A receptor complex. Furthermore, we found that either the truncation of NR2A C-tail at aa 857 or the mutation of Y837 in the NR1-1a subunit to phenylalanine blocked the inhibition of NR1-1a/NR2A receptors induced by intracellular application of Csk. Thus, both the NR1 and NR2 subunits are required for the regulation of NMDAR activity by Csk.  相似文献   

14.
N-methyl-D-aspartate receptors (NMDARs) are critical for the maturation and plasticity of glutamatergic synapses. In the hippocampus, NMDARs mainly contain GluN2A and/or GluN2B regulatory subunits. The amyloid precursor protein (APP) has emerged as a putative regulator of NMDARs, but the impact of this interaction to their function is largely unknown. By combining patch-clamp electrophysiology and molecular approaches, we unravel a dual mechanism by which APP controls GluN2B-NMDARs, depending on the life stage. We show that APP is highly abundant specifically at the postnatal postsynapse. It interacts with GluN2B-NMDARs, controlling its synaptic content and mediated currents, both in infant mice and primary neuronal cultures. Upon aging, the APP amyloidogenic-derived C-terminal fragments, rather than APP full-length, contribute to aberrant GluN2B-NMDAR currents. Accordingly, we found that the APP processing is increased upon aging, both in mice and human brain. Interfering with stability or production of the APP intracellular domain normalized the GluN2B-NMDARs currents. While the first mechanism might be essential for synaptic maturation during development, the latter could contribute to age-related synaptic impairments.  相似文献   

15.
In cerebral cortex there is a developmental switch from NR2B- to NR2A-containing NMDA receptors (NMDARs) driven by activity and sensory experience. This subunit switch alters NMDAR function, influences synaptic plasticity, and its dysregulation is associated with neurological disorders. However, the mechanisms driving the subunit switch are not known. Here, we show in hippocampal CA1 pyramidal neurons that the NR2B to NR2A switch driven acutely by activity requires activation of NMDARs and mGluR5, involves PLC, Ca(2+) release from IP(3)R-dependent stores, and PKC activity. In mGluR5 knockout mice the developmental NR2B-NR2A switch in CA1 is deficient. Moreover, in visual cortex of mGluR5 knockout mice, the NR2B-NR2A switch evoked in?vivo by visual experience is absent. Thus, we establish that mGluR5 and NMDARs are required for the activity-dependent NR2B-NR2A switch and play a critical role in experience-dependent regulation of NMDAR subunit composition in?vivo.  相似文献   

16.
The induction of long-term potentiation at CA3-CA1 synapses is caused by an N-methyl-d-aspartate (NMDA) receptordependent accumulation of intracellular Ca(2+), followed by Src family kinase activation and a positive feedback enhancement of NMDA receptors (NMDARs). Nevertheless, the amplitude of baseline transmission remains remarkably constant even though low frequency stimulation is also associated with an NMDAR-dependent influx of Ca(2+) into dendritic spines. We show here that an interaction between C-terminal Src kinase (Csk) and NMDARs controls the Src-dependent regulation of NMDAR activity. Csk associates with the NMDAR signaling complex in the adult brain, inhibiting the Src-dependent potentiation of NMDARs in CA1 neurons and attenuating the Src-dependent induction of long-term potentiation. Csk associates directly with Src-phosphorylated NR2 subunits in vitro. An inhibitory antibody for Csk disrupts this physical association, potentiates NMDAR mediated excitatory postsynaptic currents, and induces long-term potentiation at CA3-CA1 synapses. Thus, Csk serves to maintain the constancy of baseline excitatory synaptic transmission by inhibiting Src kinase-dependent synaptic plasticity in the hippocampus.  相似文献   

17.
Activation of N-methyl-D-aspartate receptors (NMDARs) has been implicated in various forms of synaptic plasticity depending on the receptor subtypes involved. However, the contribution of NR2A and NR2B subunits in the induction of long-term depression (LTD) of excitatory postsynaptic currents (EPSCs) in layer II/III pyramidal neurons of the young rat visual cortex remains unclear. The present study used whole-cell patch-clamp recordings in vitro to investigate the role of NR2A- and NR2B-containing NMDARs in the induction of LTD in visual cortical slices from 12- to 15-day old rats. We found that LTD was readily induced in layer II/III pyramidal neurons of the rat visual cortex with 10-min 1-Hz stimulation paired with postsynaptic depolarization. D-APV, a selective NMDAR antagonist, blocked the induction of LTD. Moreover, the selective NR2B-containing NMDAR antagonists (Ro 25-6981 and ifenprodil) also prevented the induction of LTD. However, Zn2+, a voltage-independent NR2A-containing NMDAR antagonist, displayed no influence on the induction of LTD. These results suggest that the induction of LTD in layer II/III pyramidal neurons of the young rat visual cortex is NMDAR-dependent and requires NR2B-containing NMDARs, not NR2A-containing NMDARs.  相似文献   

18.
NMDA receptor (NMDAR)-mediated excitatory synaptic transmission plays a critical role in synaptic plasticity and memory formation, whereas its dysfunction may underlie neuropsychiatric and neurodegenerative diseases. The neuroactive steroid pregnenolone sulfate (PS) acts as a cognitive enhancer in impaired animals, augments LTP in hippocampal slices by enhancing NMDAR activity, and may participate in the reduction of schizophrenia's negative symptoms by systemic pregnenolone. We report that the effects of PS on NMDAR function are diverse, varying with subunit composition and NR1 splice variant. While PS potentiates NR1-1a/NR2B receptors through a critical steroid modulatory domain in NR2B that also modulates tonic proton inhibition, potentiation of the NMDA response is not dependent upon relief of such inhibition, a finding that distinguishes it from spermine. In contrast, the presence of an NR2A subunit confers enhanced PS-potentiation at reduced pH, suggesting that it may indeed act like spermine does at NR2B-containing receptors. Additional tuning of the NMDAR response by PS comes via the N-terminal exon-5 splicing insert of NR1-1b, which regulates the magnitude of proton-dependent PS potentiation. For NR2C- and NR2D-containing receptors, negative modulation at NR2C receptors is pH-independent (like NR2B) while negative modulation at NR2D receptors is pH-dependent (like NR2A). Taken together, PS displays a rich modulatory repertoire that takes advantage of the structural diversity of NMDARs in the CNS. The differential pH sensitivity of NMDAR isoforms to PS modulation may be especially important given the emerging role of proton sensors to both learning and memory, as well as brain injury.  相似文献   

19.
NMDA receptors (NMDARs) are involved in excitatory synaptic transmission and plasticity associated with a variety of brain functions, from memory formation to chronic pain. Subunit-selective antagonists for NMDARs provide powerful tools to dissect NMDAR functions in neuronal activities. Recently developed antagonist for NR2A-containing receptors, NVP-AAM007, triggered debates on its selectivity and involvement of the NMDAR subunits in bi-directional synaptic plasticity. Here, we re-examined the pharmacological properties of NMDARs in the anterior cingulate cortex (ACC) using NVP-AAM007 as well as ifenprodil, a selective antagonist for NR2B-containing NMDARs. By alternating sequence of drug application and examining different concentrations of NVP-AAM007, we found that the presence of NVP-AAM007 did not significantly affect the effect of ifenprodil on NMDAR-mediated EPSCs. These results suggest that NVP-AAM007 shows great preference for NR2A subunit and could be used as a selective antagonist for NR2A-containing NMDARs in the ACC.  相似文献   

20.
N-methyl- D -aspartate receptors (NMDARs) play a pivotal role in excitatory neurotransmission, synaptic plasticity and brain development. Clinical and experimental evidence suggests a dysregulation of NMDAR function and glutamatergic pathways in the pathophysiology of schizophrenia. We evaluated electrophysiological and behavioral properties of NMDAR deficiency utilizing mice that express only 5–10% of the normal level of NMDAR NR1 subunit. Auditory and visual event related potentials yielded significantly increased amplitudes for the P20 and N40 components in NMDAR deficient (NR1neo−/−) mice suggesting decreased inhibitory tone. Compared to wild types, NR1neo−/− mice spent less time in social interactions and showed reduced nest building. NR1neo−/− mice displayed a preference for open arms of a zero maze and central zone of an open field, possibly reflecting decreased anxiety-related behavioral inhibition. However, locomotor activity did not differ between groups in either home cage environment or during behavioral testing. NR1neo−/− mice displayed hyperactivity only when placed in a large unfamiliar environment, suggesting that neither increased anxiety nor non-specific motor activation accounts for differential behavioral patterns. Data suggest that NMDAR NR1 deficiency causes disinhibition in sensory processing as well as reduced behavioral inhibition and impaired social interactions. The behavioral signature in NR1neo−/− mice supports the impact of impaired NMDAR function in a mouse model with possible relevance to negative symptoms in 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