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 796 毫秒
1.
Postsynaptic density protein‐95 (PSD‐95) is a central element of the postsynaptic architecture of glutamatergic synapses. PSD‐95 mediates postsynaptic localization of AMPA receptors and NMDA receptors and plays an important role in synaptic plasticity. PSD‐95 is released from postsynaptic membranes in response to Ca2+ influx via NMDA receptors. Here, we show that Ca2+/calmodulin (CaM) binds at the N‐terminus of PSD‐95. Our NMR structure reveals that both lobes of CaM collapse onto a helical structure of PSD‐95 formed at its N‐terminus (residues 1–16). This N‐terminal capping of PSD‐95 by CaM blocks palmitoylation of C3 and C5, which is required for postsynaptic PSD‐95 targeting and the binding of CDKL5, a kinase important for synapse stability. CaM forms extensive hydrophobic contacts with Y12 of PSD‐95. The PSD‐95 mutant Y12E strongly impairs binding to CaM and Ca2+‐induced release of PSD‐95 from the postsynaptic membrane in dendritic spines. Our data indicate that CaM binding to PSD‐95 serves to block palmitoylation of PSD‐95, which in turn promotes Ca2+‐induced dissociation of PSD‐95 from the postsynaptic membrane.  相似文献   

2.
Homeostatic synaptic plasticity is a negative-feedback mechanism for compensating excessive excitation or inhibition of neuronal activity. When neuronal activity is chronically suppressed, neurons increase synaptic strength across all affected synapses via synaptic scaling. One mechanism for this change is alteration of synaptic AMPA receptor (AMPAR) accumulation. Although decreased intracellular Ca2+ levels caused by chronic inhibition of neuronal activity are believed to be an important trigger of synaptic scaling, the mechanism of Ca2+-mediated AMPAR-dependent synaptic scaling is not yet understood. Here, we use dissociated mouse cortical neurons and employ Ca2+ imaging, electrophysiological, cell biological, and biochemical approaches to describe a novel mechanism in which homeostasis of Ca2+ signaling modulates activity deprivation-induced synaptic scaling by three steps: (1) suppression of neuronal activity decreases somatic Ca2+ signals; (2) reduced activity of calcineurin, a Ca2+-dependent serine/threonine phosphatase, increases synaptic expression of Ca2+-permeable AMPARs (CPARs) by stabilizing GluA1 phosphorylation; and (3) Ca2+ influx via CPARs restores CREB phosphorylation as a homeostatic response by Ca2+-induced Ca2+ release from the ER. Therefore, we suggest that synaptic scaling not only maintains neuronal stability by increasing postsynaptic strength but also maintains nuclear Ca2+ signaling by synaptic expression of CPARs and ER Ca2+ propagation.  相似文献   

3.
Calmodulin‐dependent kinase II (CaMKII) is key for long‐term potentiation of synaptic AMPA receptors. Whether CaMKII is involved in activity‐dependent plasticity of other ionotropic glutamate receptors is unknown. We show that repeated pairing of pre‐ and postsynaptic stimulation at hippocampal mossy fibre synapses induces long‐term depression of kainate receptor (KAR)‐mediated responses, which depends on Ca2+ influx, activation of CaMKII, and on the GluK5 subunit of KARs. CaMKII phosphorylation of three residues in the C‐terminal domain of GluK5 subunit markedly increases lateral mobility of KARs, possibly by decreasing the binding of GluK5 to PSD‐95. CaMKII activation also promotes surface expression of KARs at extrasynaptic sites, but concomitantly decreases its synaptic content. Using a molecular replacement strategy, we demonstrate that the direct phosphorylation of GluK5 by CaMKII is necessary for KAR‐LTD. We propose that CaMKII‐dependent phosphorylation of GluK5 is responsible for synaptic depression by untrapping of KARs from the PSD and increased diffusion away from synaptic sites.  相似文献   

4.
Ca2+/calmodulin-dependent kinase IIα (CaMKIIα) is essential for synaptic plasticity and learning by decoding synaptic Ca2+ oscillations. Despite decades of extensive research, new mechanisms underlying CaMKIIα’s function in synapses are still being discovered. Here, we discover that Shank3 is a specific binding partner for autoinhibited CaMKIIα. We demonstrate that Shank3 and GluN2B, via combined actions of Ca2+ and phosphatases, reciprocally bind to CaMKIIα. Under basal condition, CaMKIIα is recruited to the Shank3 subcompartment of postsynaptic density (PSD) via phase separation. Rise of Ca2+ concentration induces GluN2B-mediated recruitment of active CaMKIIα and formation of the CaMKIIα/GluN2B/PSD-95 condensates, which are autonomously dispersed upon Ca2+ removal. Protein phosphatases control the Ca2+-dependent shuttling of CaMKIIα between the two PSD subcompartments and PSD condensate formation. Activation of CaMKIIα further enlarges the PSD assembly and induces structural LTP. Thus, Ca2+-induced and phosphatase-checked shuttling of CaMKIIα between distinct PSD nano-domains can regulate phase separation-mediated PSD assembly and synaptic plasticity.Subject terms: Cell biology, Molecular biology  相似文献   

5.
Changes in synaptic function require both qualitative and quantitative reorganization of the synaptic components. Ca2+ plays a central role in this process, but the mechanism has not been fully elucidated. Zhang et al report a novel mechanism whereby Ca2+/calmodulin (CaM) regulates the stability of the postsynaptic scaffold. Ca2+/CaM interacts with PSD-95, a core protein in the postsynaptic density (PSD) that supports synaptic signaling and structural components. Ca2+/CaM interferes with the palmitoylation of PSD-95, resulting in the dissociation of PSD-95 from the postsynaptic membrane. This process may explain the reduction of surface glutamate receptor observed during synaptic depression and homeostatic regulation of the synaptic response after prolonged neuronal activity.The adaptation of synaptic strength is a fundamental process for learning and memory. This form of synaptic plasticity is triggered by influx of Ca2+ from NMDA-type glutamate receptor (NMDAR), leading to the synaptic insertion or removal of AMPA-type glutamate receptor (AMPAR) and determines the strength of synaptic transmission. This process is mediated by the gross reorganization of the postsynaptic composition in a qualitative and quantitative fashion (Bosch et al, 2014). Importantly, the number of synaptic AMPAR is regulated by the number and affinity of postsynaptic ‘slots’, a hypothetical receptor binding site within a synapse, which is regulated during synaptic plasticity processes.PSD-95, a scaffolding protein at excitatory synapses, has been considered as a major candidate for the slot. It interacts with AMPAR through the TARP/stargazin protein family and modulates the synaptic localization of the receptor as well as the strength of synaptic transmission (El-Husseini et al, 2000). In turn, the localization of PSD-95 at the synapse is regulated by a constant cycle of palmitoylation by protein palmitoyl acyltransferases (PAT) at cysteines 3 and 5, which is required for efficient synaptic targeting of the protein, and depalmitoylation by palmitoyl protein thioesterases (PPT) (El-Husseini et al, 2002; Noritake et al, 2009). Activation of glutamate receptors increases depalmitoylated PSD-95 and releases it from the postsynaptic site (El-Husseini et al, 2002; Sturgill et al, 2009), whereas blockage of neuronal activity by TTX increases palmitoylated PSD-95 and targets it to the synapse (Noritake et al, 2009). However, it remains unclear how neuronal activity controls the palmitoylation/depalmitoylation cycle and the subsequent trafficking of PSD-95 to and from the synapse.In this issue of The EMBO Journal, using a combination of structural biological, biochemical, and cell biological approaches, Zhang et al (2014) revealed a novel mechanism by which Ca2+ regulates the synaptic localization of PSD-95. The authors found that Ca2+ complexed to CaM can bind PSD-95 within the first 13 residues—the exact location where palmitoylation takes place. Ca2+/CaM binds preferentially to unmodified PSD-95, thereby blocking the accessibility of the PAT to the palmitoylation sites. In contrast, Ca2+/CaM does not bind to palmitoylated PSD-95, and therefore, palmitoylated PSD-95 is subject to depalymitoylation by PPT. Overall, the net effect of Ca2+/CaM is a reduction of PSD-95 bound to the synaptic membrane (Fig(Fig1).1). Consistent with this, a PSD-95 mutant that was unable to bind Ca2+/CaM does not leave the synapse following glutamate/glycine treatment. Furthermore, the mutant PSD-95 showed an increased in synaptic accumulation, indicating that the treatment also increases palmitoylation activity but it is normally dominated by the depalmitoylating action of Ca2+/CaM binding (Noritake et al, 2009).Open in a separate windowFigure 1Calcium influx-induced release of PSD-95 and glutamate receptor from the synapseAt a synapse, cysteine residues of PSD-95 (C3 and C5) are under a continuous cycle of palmitoylation by protein palmitoyl acyltransferase (PAT), and depalmitoylation by palmitoyl protein thioesterase (PPT). A. Palmitoylated PSD-95 associates with the synaptic membrane and CDKL5 and serves as a ‘slot’ for AMPAR at the synapse through the interaction with TARP/stargazin. B. Upon glutamate stimulation, Ca2+ influx through NMDARs induces binding of Ca2+/CaM to PSD-95. Ca2+/CaM blocks the accessibility of PAT, thereby facilitating the depalmitoylation of PSD-95, which subsequently allows PSD-95 to dissociate from the synaptic membrane and CDKL5. C. The dissociation of PSD-95 from the synaptic membrane reduces the number of available ‘slots’ for AMPAR on the postsynaptic membrane, leading to a reduction of AMPAR-mediated synaptic transmission.The beauty of the work by Zhang et al (2014) is that the structure fully explains the biology. However, several important questions remain. Above all, it is still unclear when this mechanism would operate. Generally, the palmitoylation/depalmitoylation cycle is considered to be in the order of minutes. Therefore, for Ca2+/CaM to effectively reduce the palmitoylation of PSD-95, a prolonged influx of Ca2+ is required. In contrast, the rise in intracellular Ca2+ induced by a single excitatory postsynaptic current (EPSC) or dendritic action potential is in the order of milliseconds to seconds. A slow repetitive stimulation protocol, such as that used to induce long-term depression (LTD) (1 Hz, 15 min), may be effective in increasing Ca2+/CaM for a sufficiently long period of time. Homeostatic scaling induced by a prolonged increase in network activity (for example, via the pharmacological blockade of inhibitory synaptic transmission) may also be a possible mechanism for the Ca2+/CaM-mediated removal of synaptic PSD-95. In contrast, stimulation used to induce long-term potentiation (LTP) (a brief high-frequency stimulation such as 100 Hz, 1 s) may not be effective. Indeed, Bosch et al (2014) found that the induction of LTP at single dendritic spines in hippocampal CA1 pyramidal neurons does not decrease or increase PSD-95 during the first hour after induction even if the dendritic spine enlarges during this period.In this context, it is important to understand what impact Ca2+/CaM-mediated removal of synaptic PSD-95 has on synaptic transmission. If PSD-95 is indeed the slot for AMPAR, the Ca2+/CaM-mediated removal of synaptic PSD-95 is expected to reduce the synaptic transmission. The stimulation protocol used here by Zhang et al (bath application of glutamate/glycine) is similar to previously reported approaches to induce ‘chemical’ LTD and hence can provide an explanation for the decrease in PSD-95 from the synapse for 10–15 min after stimulation. The PSD-95 mutant that is unable to bind Ca2+/CaM will be useful to further analyze the link between the observed PSD-95-Ca2+/CaM interaction and synaptic plasticity.PSD-95 is also known to interact with the cyclin-dependent protein kinase-like kinase 5 (CDKL5) at the first 19 residues in a palmitoylation-dependent manner (Zhu et al, 2013). As expected, Ca2+/CaM binding also regulates CDKL5 association with PSD-95. The treatment of neurons with NMDA reduces the palmitoylation of PSD-95 and, concomitantly, the association with CDKL5. Mutations of CDKL5 and netrin-G1 gene have been reported in patients with an atypical form of Rett syndrome. Netrin-G1 ligand (NGL1) has been identified as an interaction partner and substrate of CDKL5 (Ricciardi et al, 2012). CDKL5 phosphorylates NGL1, and this phosphorylation stabilizes the interaction of NGL1 with PSD-95. Given that the Ca2+ signal only lasts a few milliseconds to seconds, it is important to investigate the spatiotemporal interaction between CaM, PSD-95, CDKL5, and NGL1 in dendritic spines during physiological and pathological conditions. In addition, CDKL5 can function as an upstream modulator of Rac signaling during development (Chen et al, 2010). Together with the fact that Rac also plays an important role in long-term potentiation, the PSD-95/CDKL5 complex might regulate Rac activity in the vicinity of the PSD.Other neuronal proteins including AMPAR subunit GluR1/2, glutamate receptor interacting protein (GRIP), G-protein-coupled receptors, δ-catenin, and small and trimeric G-proteins can also undergo palmitoylation, suggesting that this process plays an essential role in subcellular targeting and that these proteins can be regulated by activity (Kang et al, 2008). The question of whether Ca2+/CaM can regulate the palmitoylation of these proteins remains. Interestingly, the Ca2+/CaM interaction site of PSD-95 does not conform to a canonical IQ-motif, a CaM binding motif found in many other proteins (Zhang et al, 2014). Therefore, a bioinformatic approach is not possible at this point in time. This opens further directions of research.  相似文献   

6.
7.
Dopamine and glutamate systems are both involved in cognitive, behavioral, and motor processes. Dysfunction of dopamine–glutamate interplay has been suggested in several psychotic diseases, above all in schizophrenia, for which there exists a need for novel medications. Intracellular calcium-dependent transduction pathways are key determinants of dopamine–glutamate interactions, which take place mainly, albeit not exclusively, in the postsynaptic density (PSD), a highly specialized postsynaptic ultrastructure. Stimulation of dopamine and glutamate receptors modulates the gene expression and the function of specific PSD proteins, the “scaffolding” proteins (Homer, Shank, and PSD95), belonging to a complex Ca2+-regulated network that integrates and converges dopamine and glutamate signaling to appropriate nuclear targets. Dysfunction of scaffolding proteins leads to severe impairment of Ca2+-dependent signaling, which may underlie the dopamine–glutamate aberrations putatively implicated in the pathogenesis of psychotic disorders. Antipsychotic therapy has been demonstrated to directly and indirectly affect the neuronal Ca2+-dependent pathways through the modulation of PSD scaffolding proteins, such as Homer, therefore influencing both dopaminergic and glutamatergic functions and enforcing Ca2+-mediated long-term synaptic changes. In this review, we will discuss the role of PSD scaffolding proteins in routing Ca2+-dependent signals to the nucleus. In particular, we will address the implication of PSD scaffolding proteins in the intracellular connections between dopamine and glutamate pathways, which involve both Ca2+-dependent and Ca2+-independent mechanisms. Finally, we will discuss how new strategies for the treatment of psychosis aim at developing antipsychotics that may impact both glutamate and dopamine signaling, and what should be the possible role of PSD scaffolding proteins.  相似文献   

8.
Cochlear inner hair cells (IHCs) develop from pre‐sensory pacemaker to sound transducer. Here, we report that this involves changes in structure and function of the ribbon synapses between IHCs and spiral ganglion neurons (SGNs) around hearing onset in mice. As synapses matured they changed from holding several small presynaptic active zones (AZs) and apposed postsynaptic densities (PSDs) to one large AZ/PSD complex per SGN bouton. After the onset of hearing (i) IHCs had fewer and larger ribbons; (ii) CaV1.3 channels formed stripe‐like clusters rather than the smaller and round clusters at immature AZs; (iii) extrasynaptic CaV1.3‐channels were selectively reduced, (iv) the intrinsic Ca2+ dependence of fast exocytosis probed by Ca2+ uncaging remained unchanged but (v) the apparent Ca2+ dependence of exocytosis linearized, when assessed by progressive dihydropyridine block of Ca2+ influx. Biophysical modeling of exocytosis at mature and immature AZ topographies suggests that Ca2+ influx through an individual channel dominates the [Ca2+] driving exocytosis at each mature release site. We conclude that IHC synapses undergo major developmental refinements, resulting in tighter spatial coupling between Ca2+ influx and exocytosis.  相似文献   

9.
The effect of synaptic junction (SJ) on microtubule assembly was examined. After preincubation with ATP at 37°C, rat SJ decreased the initial velocity and the extent of the porcine brain microtubule assembly (initiated by the addition of GTP) in a Ca2+/calmodulin (CaM)-dependent manner. The degree of the inhibition reached 35% of the control assembly (0-min preincubation) after 20-min preincubation with ATP. There was no inhibition either with heat-treated SJ, at 0°C, or in the presence of EGTA or W-7 (CaM antagonist). The inhibition was due neither to protease(s) nor CaM contaminating the preparations. Free Ca2+ concentration level required for the inhibition of microtubule assembly was 10–6 M. Phosphorylation of microtubule proteins was inhibited by SJ in a Ca2+/CaM-dependent manner, and the inhibition occurred in a physiological increase range of intracellular Ca2+ concentration (10–6M) The heat-treated SJ caused no inhibition. The result suggested that the microtubule assembly in the postsynaptic region was regulated by a Ca2+/CaM-dependent protein kinase associated with SJ; i. e., major postsynaptic density protein.Abbreviations used CaM calmodulin - DTT dithiothreitol - MAPs microtubule-associated proteins - MES 2-(N-morphorino)ethanesulfonic acid - mPSDp major postsynaptic density protein - PSD postsynaptic density - SDS PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis - W-7 N-(6-aminohexyl)-5-chloro-1-naphthalenesulfonamide hydrochloride  相似文献   

10.
Ca2+ signalling in neurons through calmodulin (CaM) has a prominent function in regulating synaptic vesicle trafficking, transport, and fusion. Importantly, Ca2+–CaM binds a conserved region in the priming proteins Munc13‐1 and ubMunc13‐2 and thus regulates synaptic neurotransmitter release in neurons in response to residual Ca2+ signals. We solved the structure of Ca2+4–CaM in complex with the CaM‐binding domain of Munc13‐1, which features a novel 1‐5‐8‐26 CaM‐binding motif with two separated mobile structural modules, each involving a CaM domain. Photoaffinity labelling data reveal the same modular architecture in the complex with the ubMunc13‐2 isoform. The N‐module can be dissociated with EGTA to form the half‐loaded Munc13/Ca2+2–CaM complex. The Ca2+ regulation of these Munc13 isoforms can therefore be explained by the modular nature of the Munc13/Ca2+–CaM interactions, where the C‐module provides a high‐affinity interaction activated at nanomolar [Ca2+]i, whereas the N‐module acts as a sensor at micromolar [Ca2+]i. This Ca2+/CaM‐binding mode of Munc13 likely constitutes a key molecular correlate of the characteristic Ca2+‐dependent modulation of short‐term synaptic plasticity.  相似文献   

11.
Calmodulin (CaM) binds to the FERM domain of 80 kDa erythrocyte protein 4.1R (R30) independently of Ca2+ but, paradoxically, regulates R30 binding to transmembrane proteins in a Ca2+-dependent manner. We have previously mapped a Ca2+-independent CaM-binding site, pep11 (A264KKLWKVCVEHHTFFR), in 4.1R FERM domain and demonstrated that CaM, when saturated by Ca2+ (Ca2+/CaM), interacts simultaneously with pep11 and with Ser185 in A181KKLSMYGVDLHKAKD (pep9), the binding affinity of Ca2+/CaM for pep9 increasing dramatically in the presence of pep11. Based on these findings, we hypothesized that pep11 induced key conformational changes in the Ca2+/CaM complex. By differential scanning calorimetry analysis, we established that the C-lobe of CaM was more stable when bound to pep11 either in the presence or absence of Ca2+. Using nuclear magnetic resonance spectroscopy, we identified 8 residues in the N-lobe and 14 residues in the C-lobe of pep11 involved in interaction with CaM in both of presence and absence of Ca2+. Lastly, Kratky plots, generated by small-angle X-ray scattering analysis, indicated that the pep11/Ca2+/CaM complex adopted a relaxed globular shape. We propose that these unique properties may account in part for the previously described Ca2+/CaM-dependent regulation of R30 binding to membrane proteins.  相似文献   

12.
Ca2+ influx into synaptic compartments during activity is a key mediator of neuronal plasticity. Although the role of presynaptic Ca2+ in triggering vesicle fusion though the Ca2+ sensor synaptotagmin 1 (Syt 1) is established, molecular mechanisms that underlie responses to postsynaptic Ca2+ influx remain unclear. In this study, we demonstrate that fusion-competent Syt 4 vesicles localize postsynaptically at both neuromuscular junctions (NMJs) and central nervous system synapses in Drosophila melanogaster. Syt 4 messenger RNA and protein expression are strongly regulated by neuronal activity, whereas altered levels of postsynaptic Syt 4 modify synaptic growth and presynaptic release properties. Syt 4 is required for known forms of activity-dependent structural plasticity at NMJs. Synaptic proliferation and retrograde signaling mediated by Syt 4 requires functional C2A and C2B Ca2+–binding sites, as well as serine 284, an evolutionarily conserved substitution for a key Ca2+-binding aspartic acid found in other synaptotagmins. These data suggest that Syt 4 regulates activity-dependent release of postsynaptic retrograde signals that promote synaptic plasticity, similar to the role of Syt 1 as a Ca2+ sensor for presynaptic vesicle fusion.  相似文献   

13.
Membrane skeletal protein 4.1R80 plays a key role in regulation of erythrocyte plasticity. Protein 4.1R80 interactions with transmembrane proteins, such as glycophorin C (GPC), are regulated by Ca2+-saturated calmodulin (Ca2+/CaM) through simultaneous binding to a short peptide (pep11; A264KKLWKVCVEHHTFFRL) and a serine residue (Ser185), both located in the N-terminal 30 kDa FERM domain of 4.1R80 (H·R30). We have previously demonstrated that CaM binding to H·R30 is Ca2+-independent and that CaM binding to H·R30 is responsible for the maintenance of H·R30 β-sheet structure. However, the mechanisms responsible for the regulation of CaM binding to H·R30 are still unknown. To investigate this, we took advantage of similarities and differences in the structure of Coracle, the Drosophila sp. homologue of human 4.1R80, i.e. conservation of the pep11 sequence but substitution of the Ser185 residue with an alanine residue. We show that the H·R30 homologue domain of Coracle, Cor30, also binds to CaM in a Ca2+-independent manner and that the Ca2+/CaM complex does not affect Cor30 binding to the transmembrane protein GPC. We also document that both H·R30 and Cor30 bind to phosphatidylinositol-4,5 bisphosphate (PIP2) and other phospholipid species and that that PIP2 inhibits Ca2+-free CaM but not Ca2+-saturated CaM binding to Cor30. We conclude that PIP2 may play an important role as a modulator of apo-CaM binding to 4.1R80 throughout evolution.  相似文献   

14.
The medial habenula (mHb) is an understudied small brain nucleus linking forebrain and midbrain structures controlling anxiety and fear behaviors. The mechanisms that maintain the structural and functional integrity of mHb neurons and their synapses remain unknown. Using spatiotemporally controlled Cre-mediated recombination in adult mice, we found that the glial cell–derived neurotrophic factor receptor alpha 1 (GFRα1) is required in adult mHb neurons for synaptic stability and function. mHb neurons express some of the highest levels of GFRα1 in the mouse brain, and acute ablation of GFRα1 results in loss of septohabenular and habenulointerpeduncular glutamatergic synapses, with the remaining synapses displaying reduced numbers of presynaptic vesicles. Chemo- and optogenetic studies in mice lacking GFRα1 revealed impaired circuit connectivity, reduced AMPA receptor postsynaptic currents, and abnormally low rectification index (R.I.) of AMPARs, suggesting reduced Ca2+ permeability. Further biochemical and proximity ligation assay (PLA) studies defined the presence of GluA1/GluA2 (Ca2+ impermeable) as well as GluA1/GluA4 (Ca2+ permeable) AMPAR complexes in mHb neurons, as well as clear differences in the levels and association of AMPAR subunits with mHb neurons lacking GFRα1. Finally, acute loss of GFRα1 in adult mHb neurons reduced anxiety-like behavior and potentiated context-based fear responses, phenocopying the effects of lesions to septal projections to the mHb. These results uncover an unexpected function for GFRα1 in the maintenance and function of adult glutamatergic synapses and reveal a potential new mechanism for regulating synaptic plasticity in the septohabenulointerpeduncular pathway and attuning of anxiety and fear behaviors.

The GPI-anchored protein GFRα1 is highly expressed in the habenula, but its role in the adult nervous system remains unknown. This study explores the role of GFRα1 in habenular neuron function, revealing that it regulates the maintenance of adult synapses in the septohabenular pathway, AMPA-mediated glutamatergic signaling, and fear and anxiety behaviors.  相似文献   

15.
The intracellular Ca2+ sensor calmodulin (CaM) regulates the cardiac Ca2+ release channel/ryanodine receptor 2 (RyR2), and mutations in CaM cause arrhythmias such as catecholaminergic polymorphic ventricular tachycardia (CPVT) and long QT syndrome. Here, we investigated the effect of CaM mutations causing CPVT (N53I), long QT syndrome (D95V and D129G), or both (CaM N97S) on RyR2-mediated Ca2+ release. All mutations increased Ca2+ release and rendered RyR2 more susceptible to store overload-induced Ca2+ release (SOICR) by lowering the threshold of store Ca2+ content at which SOICR occurred and the threshold at which SOICR terminated. To obtain mechanistic insights, we investigated the Ca2+ binding of the N- and C-terminal domains (N- and C-domain) of CaM in the presence of a peptide corresponding to the CaM-binding domain of RyR2. The N53I mutation decreased the affinity of Ca2+ binding to the N-domain of CaM, relative to CaM WT, but did not affect the C-domain. Conversely, mutations N97S, D95V, and D129G had little or no effect on Ca2+ binding to the N-domain but markedly decreased the affinity of the C-domain for Ca2+. These results suggest that mutations D95V, N97S, and D129G alter the interaction between CaM and the CaMBD and thus RyR2 regulation. Because the N53I mutation minimally affected Ca2+ binding to the C-domain, it must cause aberrant regulation via a different mechanism. These results support aberrant RyR2 regulation as the disease mechanism for CPVT associated with CaM mutations and shows that CaM mutations not associated with CPVT can also affect RyR2. A model for the CaM-RyR2 interaction, where the Ca2+-saturated C-domain is constitutively bound to RyR2 and the N-domain senses increases in Ca2+ concentration, is proposed.  相似文献   

16.
Oscillations in cytosolic free calcium determine the polarity of tip‐growing root hairs. The Ca2+ channel cyclic nucleotide gated channel 14 (CNGC14) contributes to the dynamic changes in Ca2+ concentration gradient at the root hair tip. However, the mechanisms that regulate CNGC14 are unknown. In this study, we detected a direct interaction between calmodulin 7 (CaM7) and CNGC14 through yeast two‐hybrid and bimolecular fluorescence complementation assays. We demonstrated that the third EF‐hand domain of CaM7 specifically interacts with the cytosolic C‐terminal domain of CNGC14. A two‐electrode voltage clamp assay showed that CaM7 completely inhibits CNGC14‐mediated Ca2+ influx, suggesting that CaM7 negatively regulates CNGC14‐mediated calcium signaling. Furthermore, CaM7 overexpressing lines phenocopy the short root hair phenotype of a cngc14 mutant and this phenotype is insensitive to changes in external Ca2+ concentrations. We, thus, identified CaM7‐CNGC14 as a novel interacting module that regulates polar growth in root hairs by controlling the tip‐focused Ca2+ signal.  相似文献   

17.
Bats C  Groc L  Choquet D 《Neuron》2007,53(5):719-734
Accumulation of AMPA receptors at synapses is a fundamental feature of glutamatergic synaptic transmission. Stargazin, a member of the TARP family, is an AMPAR auxiliary subunit allowing interaction of the receptor with scaffold proteins of the postsynaptic density, such as PSD-95. How PSD-95 and Stargazin regulate AMPAR number in synaptic membranes remains elusive. We show, using single quantum dot and FRAP imaging in live hippocampal neurons, that exchange of AMPAR by lateral diffusion between extrasynaptic and synaptic sites mostly depends on the interaction of Stargazin with PSD-95 and not upon the GluR2 AMPAR subunit C terminus. Disruption of interactions between Stargazin and PSD-95 strongly increases AMPAR surface diffusion, preventing AMPAR accumulation at postsynaptic sites. Furthermore, AMPARs and Stargazin diffuse as complexes in and out synapses. These results propose a model in which the Stargazin-PSD-95 interaction plays a key role to trap and transiently stabilize diffusing AMPARs in the postsynaptic density.  相似文献   

18.
Ca2+ influx through voltage‐activated Ca2+ channels and its feedback regulation by Ca2+‐activated K+ (BK) channels is critical in Ca2+‐dependent cellular processes, including synaptic transmission, growth and homeostasis. Here we report differential roles of cacophony (CaV2) and Dmca1D (CaV1) Ca2+ channels in synaptic transmission and in synaptic homeostatic regulations induced by slowpoke (slo) BK channel mutations. At Drosophila larval neuromuscular junctions (NMJs), a well‐established homeostatic mechanism of transmitter release enhancement is triggered by experimentally suppressing postsynaptic receptor response. In contrast, a distinct homeostatic adjustment is induced by slo mutations. To compensate for the loss of BK channel control presynaptic Sh K+ current is upregulated to suppress transmitter release, coupled with a reduction in quantal size. We demonstrate contrasting effects of cac and Dmca1D channels in decreasing transmitter release and muscle excitability, respectively, consistent with their predominant pre‐ vs. postsynaptic localization. Antibody staining indicated reduced postsynaptic GluRII receptor subunit density and altered ratio of GluRII A and B subunits in slo NMJs, leading to quantal size reduction. Such slo‐triggered modifications were suppressed in cac;;slo larvae, correlated with a quantal size reversion to normal in double mutants, indicating a role of cac Ca2+ channels in slo‐triggered homeostatic processes. In Dmca1D;slo double mutants, the quantal size and quantal content were not drastically different from those of slo, although Dmca1D suppressed the slo‐induced satellite bouton overgrowth. Taken together, cac and Dmca1D Ca2+ channels differentially contribute to functional and structural aspects of slo‐induced synaptic modifications. © 2013 Wiley Periodicals, Inc. Develop Neurobiol 74: 1–15, 2014  相似文献   

19.
20.
Neurogranin (Ng) is a postsynaptic IQ-motif containing protein that accelerates Ca2+ dissociation from calmodulin (CaM), a key regulator of long-term potentiation and long-term depression in CA1 pyramidal neurons. The exact physiological role of Ng, however, remains controversial. Two genetic knockout studies of Ng showed opposite outcomes in terms of the induction of synaptic plasticity. To understand its function, we test the hypothesis that Ng could regulate the spatial range of action of Ca2+/CaM based on its ability to accelerate the dissociation of Ca2+ from CaM. Using a mathematical model constructed on the known biochemistry of Ng, we calculate the cycle time that CaM molecules alternate between the fully Ca2+ saturated state and the Ca2+ unbound state. We then use these results and include diffusion of CaM to illustrate the impact that Ng has on modulating the spatial profile of Ca2+-saturated CaM within a model spine compartment. Finally, the first-passage time of CaM to transition from the Ca2+-free state to the Ca2+-saturated state was calculated with or without Ng present. These analyses suggest that Ng regulates the encounter rate between Ca2+ saturated CaM and its downstream targets during postsynaptic Ca2+ transients.  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