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 46 毫秒
1.
Prolonged medically induced coma (pMIC) is carried out routinely in intensive care medicine. pMIC leads to cognitive impairment, yet the underlying neuromorphological correlates are still unknown, as no direct studies of MIC exceeding ∼6 h on neural circuits exist. Here, we establish pMIC (up to 24 h) in adolescent and mature mice, and combine longitudinal two-photon imaging of cortical synapses with repeated behavioral object recognition assessments. We find that pMIC affects object recognition, and that it is associated with enhanced synaptic turnover, generated by enhanced synapse formation during pMIC, while the postanesthetic period is dominated by synaptic loss. Our results demonstrate major side effects of prolonged anesthesia on neural circuit structure.

Cognitive impairment due to prolonged medically induced coma (pMIC) represents an enormous clinical and socioeconomic burden affecting millions of patients worldwide (13). In adulthood, clinical trials have proven that intensive care unit (ICU) survivors frequently suffer from lasting cognitive impairment (2, 4), yet the neuromorphopathological underpinnings of this impairment have remained elusive. In early life, when the brain is highly plastic, MIC (maximum of 6 h tested in rodents to date) has been shown to result in synaptic changes and long-term cognitive impairment (57). However, basic animal research suggests that, during later adolescence and adulthood, dendrites and dendritic spines are stable under physiological conditions (810) and short-term MIC (11). Whether this notion holds true for pMIC is unknown.  相似文献   

2.
Neurotransmitter release during synaptic transmission comprises a tightly orchestrated sequence of molecular events, and Munc13-1 is a cornerstone of the fusion machinery. A forward genetic screen for defects in neurotransmitter release in Caenorhabditis elegans identified a mutation in the Munc13-1 ortholog UNC-13 that eliminated its unique and deeply conserved C-terminal module (referred to as HC2M) containing a Ca2+-insensitive C2 domain flanked by membrane-binding helices. The HC2M module could be functionally replaced in vivo by protein domains that localize to synaptic vesicles but not to the plasma membrane. HC2M is broadly conserved in other Unc13 family members and is required for efficient synaptic vesicle priming. We propose that the HC2M domain evolved as a vesicle/endosome adaptor and acquired synaptic vesicle specificity in the Unc13ABC protein family.

Chemical synaptic transmission is the primary mode of cellular communication within the nervous system. The presynaptic piece of this process encompasses a remarkable set of sequential and highly regulated interactions between a host of proteins, synaptic vesicles (SV), the plasma membrane, and calcium ions (Ca2+). Fusion of neurotransmitter-containing vesicles with the presynaptic plasma membrane is driven by the assembly of the neuronal SNAREs SNAP-25 and Syntaxin 1 on the plasma membrane and Synaptobrevin-2/VAMP2 on the SV. The assembly process and its coupling to intracellular Ca2+ are choreographed by a deeply conserved group of proteins including Munc13, Munc18, Synaptotagmin 1, and Complexin (14). Together with the SNAREs, these proteins form the core of the fusion apparatus across all metazoan nervous systems (57).First identified in a landmark genetic screen for nervous system mutants in the nematode Caenorhabditis elegans, UNC-13 is the founding member of the highly conserved metazoan Unc13 secretory protein family that includes Unc13ABC in humans (Munc13-1/2/3 in mice) (810). Munc13-1/UNC-13 localizes to the presynaptic active zone and is implicated in numerous presynaptic functions including initiation of release site assembly, SV docking and priming, Ca2+- and lipid-dependent forms of short-term synaptic plasticity, opening and positioning Syntaxin 1 for SNARE assembly, and protecting SNARE complexes from disassembly by NSF/alpha-SNAP (3, 1113). Loss of Munc13-1 orthologs in the nervous system almost entirely eliminates all forms of chemical synaptic transmission, establishing the Unc13 family as essential to this process (1416). All UNC-13 orthologs contain a large Syntaxin-binding MUN domain flanked by a Ca2+- and lipid-binding C1-C2 module and an additional C2 domain on its C terminus referred to as C2C (5, 10, 17).The C-terminal end of UNC-13 is the least understood domain within the Unc13 protein family in terms of both structure and mechanism (18, 19). Recent work on the MUN and C2C domains of Munc13-1 both in vitro and in cultured hippocampal synapses supports the notion that the MUN-C2C region attaches Munc13-1 to SVs as a means of preparing SVs for fusion (20, 21), but several questions remain unresolved. Is the SV interaction mediated by direct membrane binding? Does the C2C domain itself bind to SVs or does the MUN domain serve this role? Does either domain provide cargo specificity as part of the priming process? Interestingly, the C-terminal end of the MUN domain of CAPS, another Unc13 family member, can bind dense-core vesicles (DCVs) although it lacks a C-terminal C2 domain (22). Moreover, the MUN domain without the C2C domain has also been demonstrated to bind liposomes through an interaction with Synaptobrevin 2 (23). These observations bring up several possibilities for interactions with the C terminus of Munc13 including direct MUN–membrane interactions, C2C–membrane interactions, or protein–protein interactions involving either or both domains. Other Unc13 family members possessing a MUN domain with a C-terminal C2 domain such as Unc13D/Munc13-4 and BAIAP3 have been proposed to tether specific cargo such as endosomes, secretory granules, and large DCVs (24, 25). How Unc13 proteins select among different cargos remains largely unanswered (24, 26, 27).Through behavioral, electrophysiological, biochemical, and genetic approaches, we uncover a deeply conserved C-terminal membrane-binding domain within Munc13-1/UNC-13 termed the Munc13 C-terminal (MCT) domain. This region, together with C2C and a neighboring N-terminal helix fold together into a stable membrane-binding protein domain in vitro, and loss of any part of this module in vivo impairs SV priming and nervous system function. Moreover, the C-terminal domain can be replaced by foreign domains that bind SVs but not the plasma membrane, demonstrating a role in SV interactions at the synapse. Phylogenetic protein sequence comparisons suggest that the ancestral Unc13/BAIAP3 homolog possessed a similar C-terminal domain prior to the emergence of metazoa, and subsequently, the UNC-13ABC subfamily domain evolved as an SV adaptor that plays a critical role in neurotransmission in all animals.  相似文献   

3.
Little is known about the cellular signals that organize synapse formation. To explore what signaling pathways may be involved, we employed heterologous synapse formation assays in which a synaptic adhesion molecule expressed in a nonneuronal cell induces pre- or postsynaptic specializations in cocultured neurons. We found that interfering pharmacologically with microtubules or actin filaments impaired heterologous synapse formation, whereas blocking protein synthesis had no effect. Unexpectedly, pharmacological inhibition of c-jun N-terminal kinases (JNKs), protein kinase-A (PKA), or AKT kinases also suppressed heterologous synapse formation, while inhibition of other tested signaling pathways—such as MAP kinases or protein kinase C—did not alter heterologous synapse formation. JNK and PKA inhibitors suppressed formation of both pre- and postsynaptic specializations, whereas AKT inhibitors impaired formation of post- but not presynaptic specializations. To independently test whether heterologous synapse formation depends on AKT signaling, we targeted PTEN, an enzyme that hydrolyzes phosphatidylinositol 3-phosphate and thereby prevents AKT kinase activation, to postsynaptic sites by fusing PTEN to Homer1. Targeting PTEN to postsynaptic specializations impaired heterologous postsynaptic synapse formation induced by presynaptic adhesion molecules, such as neurexins and additionally decreased excitatory synapse function in cultured neurons. Taken together, our results suggest that heterologous synapse formation is driven via a multifaceted and multistage kinase network, with diverse signals organizing pre- and postsynaptic specializations.

Synapse formation is the universal process that underlies construction of all of the brain’s circuits, but little is known about its mechanisms. Unknown signaling pathways presumably organize synapses, but what pathways are involved remains unclear. Synapse formation likely requires interactions between pre- and postsynaptic neurons via adhesion molecules that transmit bidirectional signals to pre- and postsynaptic neurons and organize pre- and postsynaptic specializations (reviewed in refs. 13). Synapses exhibit canonical features that include a presynaptic side that releases neurotransmitters rapidly and transiently and a postsynaptic side that recognizes these neurotransmitters. Interestingly, only the presynaptic side of a synapse harbors canonical features that are shared by all synapses, such as synaptic vesicles and active zones with the same components in excitatory and inhibitory synapses. In contrast, the postsynaptic sides differ dramatically between excitatory and inhibitory synapses. Even excitatory and inhibitory neurotransmitter receptors exhibit no homology, and few if any molecular components are shared among excitatory and inhibitory postsynaptic specializations.At present, it is unknown what intracellular signaling pathways are involved in the assembly of pre- and postsynaptic specializations, whether different types of signaling pathways exist for pre- vs. postsynaptic specializations, and how excitatory vs. inhibitory synapses are organized. In the present study, we chose the heterologous synapse formation assay as an approach in order to begin to address these fundamental questions (4). In the heterologous synapse formation assay, nonneuronal cells, such as HEK293T cells, express a synaptic adhesion molecule that then induces pre- or postsynaptic specializations when these nonneuronal cells are cocultured with neurons (59). For example, if a postsynaptic adhesion molecule, such as neuroligin-1 (Nlgn1) or latrophilin-3, is expressed in HEK293T cells, and the HEK293T cells are cocultured with neurons, these neurons form presynaptic specializations on the HEK293T cells (5, 10). If, conversely, a presynaptic adhesion molecule, such as a neurexin or teneurin, is expressed in HEK293T cells, postsynaptic specializations are induced in cocultured neurons (8, 9, 11).Many adhesion molecules have been shown to induce heterologous synapse formation, including neurexins, neuroligins, latrophilins, teneurins, SynCAMs, neuronal pentraxin receptors, SALMs, LAR-type PTPRs, and others (5, 6, 815), suggesting that there are common “synapse signaling” pathways and that the heterologous synapse formation assay nonspecifically transduces different adhesion molecules signals into a response that organizes pre- or postsynaptic specializations. Even engagement of neuronal AMPA-type glutamate receptors by the neuronal pentraxin receptor, when expressed in HEK293T cells, causes organization of postsynaptic specializations in the heterologous synapse formation assay, testifying to the broad nature of the signals that mediate heterologous synapse formation (12). Strikingly, any given adhesion molecule triggers only either pre- or postsynaptic specializations, but not both, indicating signaling specificity. Most adhesion molecules—with the exception of teneurin splice variants (11)—induce both excitatory and inhibitory synaptic specializations at the same time. Heterologous synapses resemble real synapses and are functional (6, 7). Overall, these observations suggest that specific signaling pathways regulate synapse formation and that the heterologous synapse formation assay provides a plausible and practical paradigm to dissect such signaling pathways, even though it represents an artificial system that lacks much of the specificity of physiological synapse formation.In the present study, we have employed pharmacological inhibitors and molecular interventions to probe the nature of the signals mediating heterologous synapse formation. Our data reveal that multiple parallel protein kinase signaling pathways are required for heterologous synapse formation. We identified a role for both JNK and PKA signaling in the formation of pre- and postsynaptic specializations and found that the PI3 kinase pathway is specifically required for the formation of post- but not presynaptic specializations. Thus, our data provide initial insight into the signaling mechanisms underlying heterologous synapse formation that may be relevant for synapse formation in general.  相似文献   

4.
Hippocampal synaptic plasticity is important for learning and memory formation. Homeostatic synaptic plasticity is a specific form of synaptic plasticity that is induced upon prolonged changes in neuronal activity to maintain network homeostasis. While astrocytes are important regulators of synaptic transmission and plasticity, it is largely unclear how they interact with neurons to regulate synaptic plasticity at the circuit level. Here, we show that neuronal activity blockade selectively increases the expression and secretion of IL-33 (interleukin-33) by astrocytes in the hippocampal cornu ammonis 1 (CA1) subregion. This IL-33 stimulates an increase in excitatory synapses and neurotransmission through the activation of neuronal IL-33 receptor complex and synaptic recruitment of the scaffold protein PSD-95. We found that acute administration of tetrodotoxin in hippocampal slices or inhibition of hippocampal CA1 excitatory neurons by optogenetic manipulation increases IL-33 expression in CA1 astrocytes. Furthermore, IL-33 administration in vivo promotes the formation of functional excitatory synapses in hippocampal CA1 neurons, whereas conditional knockout of IL-33 in CA1 astrocytes decreases the number of excitatory synapses therein. Importantly, blockade of IL-33 and its receptor signaling in vivo by intracerebroventricular administration of its decoy receptor inhibits homeostatic synaptic plasticity in CA1 pyramidal neurons and impairs spatial memory formation in mice. These results collectively reveal an important role of astrocytic IL-33 in mediating the negative-feedback signaling mechanism in homeostatic synaptic plasticity, providing insights into how astrocytes maintain hippocampal network homeostasis.

Synaptic plasticity, the ability of neurons to alter the structure and strength of synapses, is important for the refinement of neuronal circuits in response to sensory experience during development (1) as well as learning and memory formation in adults (2, 3). To maintain the stability of neuronal network activity, the synaptic strength of neurons is modified through a negative-feedback mechanism termed homeostatic synaptic plasticity (46). Specifically, inhibiting neuronal activity in cultured neuronal cells or hippocampal slices by pharmacological administration of the sodium channel blocker tetrodotoxin (TTX) increases the strength of excitatory synapses to rebalance network activity (57).The hippocampus, which comprises the cornu ammonis 1 (CA1), CA2, CA3, and dentate gyrus subregions, is important for memory storage and retrieval. In particular, the CA1 subregion constitutes the primary output of the hippocampus, which is thought to be essential for most hippocampus-dependent memories (8, 9). Moreover, experience-driven synaptic changes in the CA1 microcircuitry impact how information is integrated (10, 11). Accordingly, the induction and expression of synaptic plasticity at hippocampal CA1 excitatory synapses are critically dependent on the structural remodeling and composition of synapses as well as functional modifications of pre- and postsynaptic proteins and neurotransmitter receptors (46, 12). As such, structural plasticity is a major regulatory mechanism of homeostatic synaptic plasticity in the hippocampal CA1 region. While most excitatory synapses are located at dendritic spines, morphological changes of dendritic spines likely participate in compensatory adaptations of hippocampal network activity and are therefore involved in learning, memory formation (13), and memory extinction (14).The efficacy of synaptic transmission and the wiring of neuronal circuitry are regulated not only by bidirectional communication between pre- and postsynaptic neurons, but also through the interactions between neurons and their associated glial cells (1517). Astrocytes, as the most abundant type of glia in the central nervous system, actively regulate synapse formation, function, and maintenance during development and in the adult brain (1820). However, the molecular basis of astrocyte–neuron communication in synaptic plasticity is largely unknown. Nevertheless, one of the mechanisms by which astrocytes regulate synapses is by secreting factors (2125); the most well-characterized one is TNFα. Notably, pharmacologically induced deprivation of neuronal activity increases TNFα release from astrocytes, which modulates homeostatic plasticity in both excitatory and inhibitory neurons through regulation of neuronal glutamate and GABA receptor trafficking (24, 26). Further in vivo studies on germline knockout mice support the roles of astrocyte-secreted TNFα in homeostatic adaptations of cortical circuitry during sensory deprivation (27, 28). Another cytokine interleukin-33 (IL-33) is secreted by astrocytes to regulate synapse development in spinal cord and thalamus (29). Nevertheless, it remains largely unknown how astrocytes respond to changes in neuronal activity to regulate homeostatic synaptic plasticity in the hippocampus as well as learning and memory formation.In this study, we identified IL-33 as an astrocyte-secreted factor which mediates homeostatic synaptic plasticity in the CA1 subregion of adult hippocampus. Pharmacological blockade of neuronal activity or in vivo optogenetic inhibition of CA1 pyramidal neurons stimulates a local increase in the expression and release of IL-33 from the astrocytes. In turn, this astrocyte-secreted IL-33 and its ST2/IL-1RAcP receptor complex mediate the increase of excitatory synapses and neurotransmission in homeostatic synaptic plasticity. Two-photon imaging of CA1 pyramidal neurons in vivo reveals that IL-33 promotes dendritic spine formation through the synaptic recruitment of postsynaptic scaffolding protein PSD-95. Importantly, conditional knockout of IL-33 in astrocytes decreases excitatory synapses in the CA1 subregion, and inhibition of IL-33/ST2 signaling in adult mice abolishes the homeostatic synaptic plasticity in CA1 pyramidal neurons, resulting in impaired spatial memory formation. Hence, our findings collectively show that astrocyte-secreted IL-33 plays an important role in homeostatic synaptic plasticity in the adult hippocampus and spatial memory formation.  相似文献   

5.
Behaviors that rely on the hippocampus are particularly susceptible to chronological aging, with many aged animals (including humans) maintaining cognition at a young adult-like level, but many others the same age showing marked impairments. It is unclear whether the ability to maintain cognition over time is attributable to brain maintenance, sufficient cognitive reserve, compensatory changes in network function, or some combination thereof. While network dysfunction within the hippocampal circuit of aged, learning-impaired animals is well-documented, its neurobiological substrates remain elusive. Here we show that the synaptic architecture of hippocampal regions CA1 and CA3 is maintained in a young adult-like state in aged rats that performed comparably to their young adult counterparts in both trace eyeblink conditioning and Morris water maze learning. In contrast, among learning-impaired, but equally aged rats, we found that a redistribution of synaptic weights amplifies the influence of autoassociational connections among CA3 pyramidal neurons, yet reduces the synaptic input onto these same neurons from the dentate gyrus. Notably, synapses within hippocampal region CA1 showed no group differences regardless of cognitive ability. Taking the data together, we find the imbalanced synaptic weights within hippocampal CA3 provide a substrate that can explain the abnormal firing characteristics of both CA3 and CA1 pyramidal neurons in aged, learning-impaired rats. Furthermore, our work provides some clarity with regard to how some animals cognitively age successfully, while others’ lifespans outlast their “mindspans.”

Aging is the biggest risk factor for Alzheimer’s disease, but many aged individuals nevertheless retain the ability to perform cognitive tasks with young adult (YA)-like competency, and are thus resilient to age-related cognitive decline and dementias (1, 2). The mechanisms of such resilience are unknown, but are thought to involve neural or cognitive reserve, brain network adaptations, or simply the ability to maintain cognitive brain circuits in a YA-like state (35). Much of the cellular and functional insight into the concept or risk of/resilience against age-related cognitive impairments has come from animal models of normal/nonpathological aging (610). Many of these studies have shown that circuit function abnormalities are associated with behavioral impairments. The cellular and structural bases for such functional aberrations, however, remain largely unknown.Two of the most well-studied cognitive domains that show susceptibility to chronological aging in both rodents and nonhuman primates are working memory and spatial/temporal memory (610). Importantly, these cognitive domains engage anatomically distinct neurocognitive systems, with the former relying on prefrontal/orbitofrontal cortical circuits and the latter relying on hippocampal circuitry. Interestingly, although behavioral deficits in these two domains (in the case of rat models of cognitive aging) begin to emerge, worsen, and become increasingly prevalent between 12 and 18 mo of age in most strains (reviewed in refs. 9 and 11), cognitive aging within hippocampus-dependent forms of learning and memory are relatively independent of those that engage the prefrontal/orbitofrontal cortical neural systems (69, 1215).Neither the mechanisms underlying the conservation of memory function across chronological aging nor those contributing to the age-related emergence and exacerbation of memory impairments are clearly understood for either neurocognitive system. It is clear, however, that neither frank neuronal loss (16, 17) nor overall synapse loss (18) contributes to cognitive aging within the medial temporal lobe/hippocampal memory system. Rather, the intriguing idea that has emerged from work in both the hippocampal and the prefrontal/orbitofrontal cortical memory systems is that there are functional alterations in the synaptic connections in individual microcircuits embedded within these larger neuroanatomical systems (610, 1931).Axospinous synapses (including those in hippocampal and cortical circuits) are characterized on the basis of the three-dimensional morphology of their postsynaptic densities (PSDs) (20, 3234). The most-abundant axospinous synaptic subtype has a continuous, macular, disk-shaped PSD, as compared to the less-abundant perforated synaptic subtype, which has at least one discontinuity in its PSD (34). In addition to differing substantially with regard to relative frequency, perforated and nonperforated synapses also harbor major differences in size and synaptic AMPA-type and NMDA-type receptor expression levels (AMPAR and NMDAR, respectively) (3438). There is also evidence that perforated and nonperforated synapses are differentially involved in synaptic plasticity (3944) and in preservation of—or reductions in—memory function during chronological aging (6, 20, 45). Layered onto these general distinctions between perforated and nonperforated synapses are more specific differences in their characteristics when considered within neural circuits. For example, perforated synapses have a stronger and more consistent influence on neuronal computation within hippocampal region CA1 than their nonperforated counterparts, which nevertheless outnumber the former by a roughly 9-to-1 ratio (34, 46, 47).These and other circuit-specific differences necessitate a circuit-based approach to understanding the synaptic bases underlying the retention or loss of YA-like memory function in the aging brain. In many ways, the hippocampal system is particularly convenient for such circuit-based approaches (48, 49). Information about the internal and external world is funneled to the parahippocampal system and then relayed via the entorhinal cortex to the dentate gyrus, the first component of the so-called trisynaptic circuit in the hippocampus proper. Granule cells in the dentate gyrus then transmit their computations to hippocampal region CA3 via the mossy fibers, which form very large and anatomically distinct synapses called mossy fiber bouton–thorny excrescence synaptic complexes in the stratum lucidum (SL). CA3 pyramidal neurons then integrate information from their autoassociational connections in the stratum radiatum (SR) and stratum oriens (SO), with both direct entorhinal inputs in stratum lacunosum-moleculare (SLM) and the dentate gyrus inputs in the SL, and convey this information to hippocampal CA1 pyramidal neurons. Neurons in hippocampal CA1 then integrate this information in their basal and apical SR dendrites with direct entorhinal cortical inputs in their most distal, tufted dendrites in the SLM, and represent the first and largest extrahippocampal output from the hippocampus proper. Thus, the computations performed both within individual hippocampal subregions and between them as an interconnected neurocognitive system are complex, and involve a combination of intrinsic (i.e., membrane-bound ion channels that regulate membrane excitability) and synaptic (i.e., ligand-gated ion channels expressed at both excitatory and inhibitory synapses) influences. Additionally, age-related changes at any level of these complex circuits will have downstream consequences on the accuracy/reliability of the information being relayed to extrahippocampal regions via CA1 pyramidal neurons.Given the amount of evidence supporting a possible synaptic explanation for age-related learning and memory impairments in hippocampus-dependent forms of cognition (610), we combined patch-clamp physiology, serial section conventional and immunogold electron microscopy (EM), quantitative Western blot analyses, and behavioral characterization using two hippocampus-dependent forms of learning in YA (6- to 8-mo old) and aged rats (28- to 29-mo old) to examine two interconnected hippocampal regions implicated in cognitive aging: Regions CA1 and CA3. We focused on CA1 and CA3 because of their central location in the hippocampal circuit (4850), their similar laminar dendritic structure (4850), and their well-documented age-related changes in place field specificity and reliability (5156).We find that the synaptic architecture and balance of synaptic weights in YA and aged, learning-unimpaired (AU) rats is remarkably similar, but that both are different in aged, learning-impaired (AI) rats. Moreover, this restructuring among “unsuccessful” cognitive agers has an intriguing specificity: It involves only AMPARs, only perforated axospinous synapses, and only hippocampal region CA3, which together shift the balance of synaptic weights that drive action potential output in CA3 pyramidal neurons maladaptively toward an overemphasis of the autoassociational synapses that interconnect CA3 pyramidal neurons.  相似文献   

6.
Ketamine is a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist that produces rapid antidepressant action in some patients with treatment-resistant depression. However, recent data suggest that ∼50% of patients with treatment-resistant depression do not respond to ketamine. The factors that contribute to the nonresponsiveness to ketamine’s antidepressant action remain unclear. Recent studies have reported a role for secreted glycoprotein Reelin in regulating pre- and postsynaptic function, which suggests that Reelin may be involved in ketamine’s antidepressant action, although the premise has not been tested. Here, we investigated whether the disruption of Reelin-mediated synaptic signaling alters ketamine-triggered synaptic plasticity and behavioral effects. To this end, we used mouse models with genetic deletion of Reelin or apolipoprotein E receptor 2 (Apoer2), as well as pharmacological inhibition of their downstream effectors, Src family kinases (SFKs) or phosphoinositide 3-kinase. We found that disruption of Reelin, Apoer2, or SFKs blocks ketamine-driven behavioral changes and synaptic plasticity in the hippocampal CA1 region. Although ketamine administration did not affect tyrosine phosphorylation of DAB1, an adaptor protein linked to downstream signaling of Reelin, disruption of Apoer2 or SFKs impaired baseline NMDA receptor–mediated neurotransmission. These results suggest that maintenance of baseline NMDA receptor function by Reelin signaling may be a key permissive factor required for ketamine’s antidepressant effects. Taken together, our results suggest that impairments in Reelin-Apoer2-SFK pathway components may in part underlie nonresponsiveness to ketamine’s antidepressant action.

Major depressive disorder (MDD) is a serious disorder that affects ∼20.6% of the US population and is a leading cause of suicide (1). One crucial problem in treating patients with MDD is an incomplete response rate to medications, where a large fraction of patients do not show a response to primary antidepressant medications (2, 3). Recent clinical findings demonstrate that a subanesthetic dose of ketamine, a noncompetitive N-methyl-d-aspartate receptor (NMDAR) antagonist, produces rapid antidepressant effects within hours in some patients with treatment-resistant depression or MDD (46). However, ∼50% of patients with treatment-resistant depression do not respond to ketamine (7), and factors involved in the nonresponsiveness to ketamine remain unclear.The hippocampus is a brain region that has been linked to the pathophysiological changes in MDD. Patients with MDD show a decrease in hippocampal volume and function (812). In contrast, MDD patients treated with classic antidepressants have a reversal in hippocampal volume changes along with an improvement in hippocampus-dependent cognitive function (1315). Previous preclinical studies have shown animal models of depression also exhibit a decrease in hippocampal volume and function (13), and hippocampal synaptic functional enhancement is required to mediate antidepressant responses (1618). This enhancement of hippocampal function has been suggested to be a key requirement to exert an antidepressant response.Ketamine induces rapid molecular changes that elicit synaptic plasticity in the hippocampus (16, 1922). Specifically, ketamine rapidly generates synaptic potentiation of field excitatory postsynaptic potentials (fEPSPs) in CA3–CA1 synapses in the hippocampus (ketamine potentiation) by inducing the rapid translation of brain-derived neurotrophic factor (BDNF) and trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) onto the postsynaptic surface (16, 19, 23, 24). Recent studies have shown that if key factors for the antidepressant effects of ketamine, such as BDNF (16, 25, 26) or AMPA receptors (16, 27), are deleted or blocked, the synaptic potentiation in the hippocampus concurrently disappears, suggesting that the synaptic potentiation underlies ketamine’s antidepressant effects (16, 19).Ketamine-mediated potentiation of fEPSPs in CA3–CA1 synapses has been shown to require a block of NMDAR activation by spontaneous glutamate release. Ketamine produces synaptic potentiation in the presence of tetrodotoxin, which blocks sodium channels, and thereby the generation of action potentials, suggesting that blocking NMDARs activated by the spontaneous presynaptic release is key to producing the synaptic potentiation (19, 21, 28, 29). In agreement with this premise, deletion of Vps10p-tail-interactor-1a (Vti1a) and vesicle-associated membrane protein 7 (VAMP7), which are soluble N-ethylmaleimide–sensitive factor attachment protein receptor proteins selectively involved in spontaneous neurotransmitter release (30, 31) in the CA3 hippocampal region, occluded the ketamine potentiation (32). Collectively, these lines of evidence suggest spontaneous glutamate release, and NMDARs are important factors for ketamine potentiation. Thus, it is possible that if these pre- or postsynaptic components are impaired, ketamine may not produce the synaptic potentiation and antidepressant effects.Reelin is a secreted glycoprotein and acts as a neuromodulator in the adult brain by regulating pre- and postsynaptic machinery. Reelin binds to its receptors, apolipoprotein E receptor 2 (Apoer2) and very-low-density lipoprotein receptor (VLDLR) and increases tyrosine phosphorylation in Disabled-1 (DAB1) (3335). The Reelin pathway regulates pre- or postsynaptic function through its downstream signaling pathways in the adult brain. In presynaptic terminals, the Reelin-Apoer2 pathway activates phosphoinositide 3-kinase (PI3K) and increases Ca2+ release from intracellular stores, which in turn mobilizes VAMP7-containing synaptic vesicles and augments spontaneous release (31). At the postsynaptic sites, Reelin’s binding to Apoer2 reciprocally activates DAB1 and Src family kinases (SFKs). Subsequently, the activated SFKs increase tyrosine phosphorylation in NMDAR subunits, GluN2A and GluN2B (3437), and increase NMDAR open probability (3739). Since pre- and postsynaptic components regulated by Reelin have been suggested to be important for ketamine potentiation (16, 1921, 32), it is conceivable that disrupted Reelin signaling may abrogate the antidepressant action and synaptic plasticity of ketamine.To examine this premise, we used genetically modified mice with a deletion of either Reelin or Apoer2 and investigated changes in antidepressant-like behaviors and synaptic potentiation in the CA1 hippocampal region following ketamine treatment. We also used pharmacological inhibitors to examine the effects of signaling molecules downstream of Reelin-Apoer2, specifically SFKs and PI3K, on ketamine-induced behavioral changes and synaptic plasticity. Lastly, we investigated whether the disruption of ketamine’s effects is due to a requirement for the activation of Reelin-dependent signaling or the impairment of NMDAR function by the disruption of Reelin-dependent signaling. Our results suggest that disruption of the Reelin-Apoer2-SFKs pathway depresses NMDAR function and diminishes ketamine’s use-dependent NMDAR antagonism, thereby rendering synapses nonresponsive to ketamine’s action as well as subsequent antidepressant responses. Taken together, these results provide insight into understanding the cellular and molecular mechanisms underlying ketamine’s antidepressant effects.  相似文献   

7.
8.
Physiological functioning and homeostasis of the brain rely on finely tuned synaptic transmission, which involves nanoscale alignment between presynaptic neurotransmitter-release machinery and postsynaptic receptors. However, the molecular identity and physiological significance of transsynaptic nanoalignment remain incompletely understood. Here, we report that epilepsy gene products, a secreted protein LGI1 and its receptor ADAM22, govern transsynaptic nanoalignment to prevent epilepsy. We found that LGI1–ADAM22 instructs PSD-95 family membrane-associated guanylate kinases (MAGUKs) to organize transsynaptic protein networks, including NMDA/AMPA receptors, Kv1 channels, and LRRTM4–Neurexin adhesion molecules. Adam22ΔC5/ΔC5 knock-in mice devoid of the ADAM22–MAGUK interaction display lethal epilepsy of hippocampal origin, representing the mouse model for ADAM22-related epileptic encephalopathy. This model shows less-condensed PSD-95 nanodomains, disordered transsynaptic nanoalignment, and decreased excitatory synaptic transmission in the hippocampus. Strikingly, without ADAM22 binding, PSD-95 cannot potentiate AMPA receptor-mediated synaptic transmission. Furthermore, forced coexpression of ADAM22 and PSD-95 reconstitutes nano-condensates in nonneuronal cells. Collectively, this study reveals LGI1–ADAM22–MAGUK as an essential component of transsynaptic nanoarchitecture for precise synaptic transmission and epilepsy prevention.

Epilepsy, characterized by unprovoked, recurrent seizures, affects 1 to 2% of the population worldwide. Many genes that cause inherited epilepsy when mutated encode ion channels, and dysregulated synaptic transmission often causes epilepsy (1, 2). Although antiepileptic drugs have mainly targeted ion channels, they are not always effective and have adverse effects. It is therefore important to clarify the detailed processes for synaptic transmission and how they are affected in epilepsy.Recent superresolution imaging of the synapse reveals previously overlooked subsynaptic nano-organizations and pre- and postsynaptic nanodomains (36), and mathematical simulation suggests their nanometer-scale coordination in individual synapses for efficient synaptic transmission: presynaptic neurotransmitter release machinery and postsynaptic receptors precisely align across the synaptic cleft to make “transsynaptic nanocolumns” (7, 8).So far, numerous transsynaptic cell-adhesion molecules have been identified (912), including presynaptic Neurexins and type IIa receptor protein tyrosine phosphatases (PTPδ, PTPσ, and LAR) and postsynaptic Neuroligins, LRRTMs, NGL-3, IL1RAPL1, Slitrks, and SALMs. Neurexins–Neuroligins have attracted particular attention because of their synaptogenic activities when overexpressed and their genetic association with neuropsychiatric disorders (e.g., autism). Another type of transsynaptic adhesion complex mediated by synaptically secreted Cblns (e.g., Neurexin–Cbln1–GluD2) promotes synapse formation and maintenance (1315). Genetic studies in Caenorhabditis elegans show that secreted Ce-Punctin, the ortholog of the mammalian ADAMTS-like family, specifies cholinergic versus GABAergic identity of postsynaptic domains and functions as an extracellular synaptic organizer (16). However, the molecular identity and in vivo physiological significance of transsynaptic nanocolumns remain incompletely understood.LGI1, a neuronal secreted protein, and its receptor ADAM22 have recently emerged as major determinants of brain excitability (17) as 1) mutations in the LGI1 gene cause autosomal dominant lateral temporal lobe epilepsy (18); 2) mutations in the ADAM22 gene cause infantile epileptic encephalopathy with intractable seizures and intellectual disability (19, 20); 3) Lgi1 or Adam22 knockout mice display lethal epilepsy (2124); and 4) autoantibodies against LGI1 cause limbic encephalitis characterized by seizures and amnesia (2528). Functionally, LGI1–ADAM22 regulates AMPA receptor (AMPAR) and NMDA receptor (NMDAR)-mediated synaptic transmission (17, 22, 29) and Kv1 channel-mediated neuronal excitability (30, 31). Recent structural analysis shows that LGI1 and ADAM22 form a 2:2 heterotetrameric assembly (ADAM22–LGI1–LGI1–ADAM22) (32), suggesting the transsynaptic configuration.In this study, we identify ADAM22-mediated synaptic protein networks in the brain, including pre- and postsynaptic MAGUKs and their functional bindings to transmembrane proteins (NMDA/AMPA glutamate receptors, voltage-dependent ion channels, cell-adhesion molecules, and vesicle-fusion machinery). ADAM22 knock-in mice lacking the MAGUK-binding motif show lethal epilepsy of hippocampal origin. In this mouse, postsynaptic PSD-95 nano-assembly as well as nano-scale alignment between pre- and postsynaptic proteins are significantly impaired. Importantly, PSD-95 is no longer able to modulate AMPAR-mediated synaptic transmission without binding to ADAM22. These findings establish that LGI1–ADAM22 instructs MAGUKs to organize transsynaptic nanocolumns and guarantee the stable brain activity.  相似文献   

9.
Long-term potentiation (LTP) is a cellular mechanism of learning and memory that results in a sustained increase in the probability of vesicular release of neurotransmitter. However, previous work in hippocampal area CA1 of the adult rat revealed that the total number of vesicles per synapse decreases following LTP, seemingly inconsistent with the elevated release probability. Here, electron-microscopic tomography (EMT) was used to assess whether changes in vesicle density or structure of vesicle tethering filaments at the active zone might explain the enhanced release probability following LTP. The spatial relationship of vesicles to the active zone varies with functional status. Tightly docked vesicles contact the presynaptic membrane, have partially formed SNARE complexes, and are primed for release of neurotransmitter upon the next action potential. Loosely docked vesicles are located within 8 nm of the presynaptic membrane where SNARE complexes begin to form. Nondocked vesicles comprise recycling and reserve pools. Vesicles are tethered to the active zone via filaments composed of molecules engaged in docking and release processes. The density of tightly docked vesicles was increased 2 h following LTP compared to control stimulation, whereas the densities of loosely docked or nondocked vesicles congregating within 45 nm above the active zones were unchanged. The tethering filaments on all vesicles were shorter and their attachment sites shifted closer to the active zone. These findings suggest that tethering filaments stabilize more vesicles in the primed state. Such changes would facilitate the long-lasting increase in release probability following LTP.

Long-term potentiation (LTP) is the persistent strengthening of synapses after a brief high-frequency stimulation and is widely accepted as a cellular correlate of learning and memory (1, 2). Within minutes after the induction of LTP, new receptors are inserted into the postsynaptic membrane. The resulting increase in the excitatory postsynaptic potential is immediate and can persist for hours in vitro or days to months in vivo (1, 38). Quantal content is also increased soon after LTP induction and reflects an increase in the number of presynaptic vesicles that release neurotransmitter (913). This increase in release probability is sustained several hours following LTP (14), concurrent with postsynaptic growth and spine enlargement (15). One might expect that the enhanced probability of release would involve increasing the number of vesicles docked and primed for neurotransmitter release. However, 2 h after induction of LTP, the total number of both docked and nondocked vesicles per presynaptic bouton are markedly decreased relative to control stimulation (16). These findings raise the question of whether an altered structure of docking and priming molecules leads to local clustering of vesicles that would elevate the probability of release following LTP.The proteins that connect synaptic vesicles to the plasma membrane can be visualized as filaments with electron-microscopic tomography (EMT) connecting vesicles to the presynaptic active zone (1724). Studies suggest that the SNARE complex begins to form when a vesicle and presynaptic membrane are within 8 nm of each other rendering them loosely docked (2527). Vesicles are then drawn toward the active zone, and the SNARE complex bundle is fully formed when the vesicle is within 2 nm of the presynaptic membrane (26, 28). Tightly docked vesicles are defined as being in contact with the presynaptic membrane and correspond to primed vesicles that comprise the readily releasable pool (27). Recent studies have suggested that docked vesicles can oscillate between loosely and tightly docked states (27, 29), providing a target mechanism for synaptic plasticity.To address the question of how changes in vesicle proximity and tethering might enhance the probability of release, we used EMT, which enabled us to acquire high-resolution structural data from small volumes of presynaptic boutons that were enriched in synaptic vesicles. We targeted active zones of hippocampal synapses, comparing their structure 2 h after LTP induction to control stimulation. The vesicle density and tethering filament dimensions were unchanged for the loosely docked and nondocked vesicles. In contrast, the density of tightly docked vesicles was increased, their tethering filaments were shorter, and the filament attachment sites on the vesicles were positioned closer to the side of the vesicle membrane facing the presynaptic membrane. Such alterations could contribute to the sustained increase in the probability of neurotransmitter release following LTP.  相似文献   

10.
Analysis of the presynaptic action potential’s (APsyn) role in synaptic facilitation in hippocampal pyramidal neurons has been difficult due to size limitations of axons. We overcame these size barriers by combining high-resolution optical recordings of membrane potential, exocytosis, and Ca2+ in cultured hippocampal neurons. These recordings revealed a critical and selective role for Kv1 channel inactivation in synaptic facilitation of excitatory hippocampal neurons. Presynaptic Kv1 channel inactivation was mediated by the Kvβ1 subunit and had a surprisingly rapid onset that was readily apparent even in brief physiological stimulation paradigms including paired-pulse stimulation. Genetic depletion of Kvβ1 blocked all broadening of the APsyn during high-frequency stimulation and eliminated synaptic facilitation without altering the initial probability of vesicle release. Thus, using all quantitative optical measurements of presynaptic physiology, we reveal a critical role for presynaptic Kv channels in synaptic facilitation at presynaptic terminals of the hippocampus upstream of the exocytic machinery.

The action potential (AP) firing pattern or “spike code” as typically measured from the soma is a gold standard for neural excitability within circuits. However, at presynaptic terminals the quantitative relationship between the input AP spike code and the magnitude of exocytosis, or vesicle fusion events per AP, can change dynamically as a result of stimulation frequency or firing pattern. Increased firing frequency can significantly increase the number of vesicles that fuse from an identical number of APs. This phenomenon is known as short-term synaptic facilitation, which can significantly enhance information transfer at synapses influencing several aspects of learning and memory (1). Thus, it is important to completely understand the underlying molecular and cellular mechanisms of synaptic facilitation.A critical initial step in exocytosis is the arrival of APsyn at boutons, whose waveform can exhibit plasticity based on firing frequency. Repetitive firing may cause inactivation of axonal voltage-gated sodium (Nav) channels and voltage-gated potassium (Kv) channels that control the depolarization and hyperpolarization of the waveform, respectively. Kv inactivation primarily leads to an increase in AP width or broadening (29). The width of the APsyn controls the fraction of time that Ca2+ channels open and the driving force of Ca2+ entry (10). These changes in voltage kinetics during the APsyn will also impact the shape or profile of the Ca2+ microdomain envelope that builds locally around open Ca2+ channels in the terminal (11, 12). The highly nonlinear influence of Ca2+ on exocytosis (13, 14) thus dictates that modest APsyn broadening has the potential to critically impact synaptic facilitation (1517). Indeed, APsyn broadening during repetitive firing has been demonstrated to cause the facilitation of exocytosis in the pituitary nerve (3), dorsal root ganglion (18), and mossy fiber bouton (2), all due to Kv channel inactivation. However, the APsyn waveform in Purkinje cells has also been shown to undergo frequency-dependent decreases in amplitude that substantially contribute to synaptic depression (19). Thus, it is best to consider the APsyn as a plastic signal that can powerfully modulate exocytosis bidirectionally, rather than as a digital spike acting solely as an initiation signal. We therefore reason that the somatic AP has proven to be a poor predictor of exocytosis magnitude as a result of a failure to resolve the APsyn waveform and its molecular regulators in the majority of brain regions.As opposed to the majority of larger synapses, en passant terminals are most commonly involved in brain regions associated with synaptic plasticity. Investigating the molecular regulation of APsyn in the common en passant nerve terminals of the cortex and hippocampus remains elusive due to the small size of these structures (<1 µm), which makes them inaccessible for whole-cell patch clamp recording. An innovative initial strategy to overcome these size restrictions was the use of voltage dyes, which failed to detect use-dependent changes in the APsyn in hippocampal slices (20). However, these dyes were limited by very low voltage sensitivity (<0.5% change in fluorescence for an AP) requiring population averaging, and these dyes were unable to report a stable resting voltage during stimulation (20). Moreover, it was found only later that this class of voltage dyes were phototoxic and altered membrane physiology, limiting their usefulness in small axons (21). As a result of these complications, the question of APsyn plasticity as a modulator of synaptic facilitation remains unanswered for hippocampal neurons. Our group has overcome the size barrier of hippocampal axons, while also avoiding cell population averaging and dye toxicity by pioneering the use of genetically encoded rhodopsin-based voltage indicators. Here, we measure the APsyn of individual en passant terminals from both inhibitory and excitatory hippocampal neurons. These measurements demonstrate a striking contrast between facilitating excitatory and depressing inhibitory nerve terminals in the hippocampus. We discovered that excitatory axons and terminals are uniquely enriched with a combination of Kv1.1/1.2 heteromers and Kvβ1 subunits. This combination of Kv subunits causes rapid APsyn broadening during brief periods of high-frequency firing. This broadening was essential for enabling synaptic facilitation without altering initial exocytosis. We also found that simply overexpressing this Kvβ1 subunit made inhibitory neurons switch from depressing during high-frequency stimulation to facilitation. Taken together, these results suggest that the molecular control of presynaptic Kv channel inactivation is an important modulator of synaptic facilitation upstream of vesicle release machinery.  相似文献   

11.
The cyclic adenosine monophosphate (cAMP)-dependent potentiation of neurotransmitter release is important for higher brain functions such as learning and memory. To reveal the underlying mechanisms, we applied paired pre- and postsynaptic recordings from hippocampal mossy fiber-CA3 synapses. Ca2+ uncaging experiments did not reveal changes in the intracellular Ca2+ sensitivity for transmitter release by cAMP, but suggested an increase in the local Ca2+ concentration at the release site, which was much lower than that of other synapses before potentiation. Total internal reflection fluorescence (TIRF) microscopy indicated a clear increase in the local Ca2+ concentration at the release site within 5 to 10 min, suggesting that the increase in local Ca2+ is explained by the simple mechanism of rapid Ca2+ channel accumulation. Consistently, two-dimensional time-gated stimulated emission depletion microscopy (gSTED) microscopy showed an increase in the P/Q-type Ca2+ channel cluster size near the release sites. Taken together, this study suggests a potential mechanism for the cAMP-dependent increase in transmission at hippocampal mossy fiber-CA3 synapses, namely an accumulation of active zone Ca2+ channels.

Communication between neurons is largely mediated by chemical synapses. Synaptic strengths are not fixed, but change dynamically in the short and longer term in an activity-dependent manner (short- and long-term plasticity, 13). Moreover, neuromodulators act on presynaptic terminals to modulate synaptic strength. Such activity-dependent or modulatory changes are often mediated by the activation of second messengers, such as protein kinase A and C (2). Second messenger systems, particularly the cyclic adenosine monophosphate (cAMP)/PKA-dependent system, are important for higher brain functions, including learning and memory in Aplysia (3), flies (4, 5), and the mammalian brain (6). Despite its functional importance, the cellular and molecular mechanisms of cAMP-dependent modulation are still poorly understood regardless of whether Aplysia synapses and Drosophila neuromuscular junctions have been investigated (2, 7). Mammalian central synapses are no exception here, also reflecting technical difficulties due to the generally small size of the presynaptic terminals in the mammalian brain.Hippocampal mossy fiber-CA3 (MF-CA3) synapses are characterized by exceptionally large presynaptic terminals (hippocampal mossy fiber bouton, hMFB), which allow for the direct analysis of the cellular mechanisms of synaptic transmission and plasticity by using patch-clamp recordings (810). Thus, hMFBs provide a suitable model of cortical synapses in the mammalian brain. Moreover, these synapses are functionally important for brain function such as pattern separation (11). Mossy fiber synapses are known to exhibit unique presynaptic forms of short- and long-term synaptic potentiation and depression, which share the cAMP/PKA-dependent induction mechanism (1215). In addition, the cAMP-dependent plasticity pathway is important for presynaptic modulation by dopamine and noradrenaline (1618), which modulates hippocampal network activity and behavior. However, its underlying cellular mechanisms remain largely unclear. Enhancement of the molecular priming and docking of synaptic vesicles at mossy fiber synapses has been suggested by previous studies using genetics and electron microscopy (1921). In particular, RIM1, an active zone scaffold protein, is crucial for cAMP-dependent long-term potentiation (LTP) (19) and is phosphorylated by PKA, although a corresponding phosphorylation mutant of RIM1 was found to have no effect on long-term potentiation (22, but see ref. 23). Other studies on hMFBs have implicated a role in positional priming, i.e., changes in the spatial coupling between Ca2+ channels and the release machinery (24). However, there is a lack of the direct visualization or manipulation of this regulation.In order to measure the intracellular Ca2+ sensitivity of transmitter release directly and examine the mechanisms of cAMP-dependent modulation quantitatively, we here carried out Ca2+ uncaging experiments at hippocampal mossy fiber synapses. Unexpectedly, our results failed to show changes in Ca2+ sensitivity, but instead uncovered an increase in local Ca2+ concentrations at the release sites. Furthermore, by live imaging of local Ca2+ using total internal reflection fluorescence (TIRF) microscopy as well as superresolution time gated STED (gSTED) microscopy, we provided evidence that rather rapid Ca2+ channel accumulation may underlie cAMP-induced potentiation instead of release machinery modulations. This study thus provides a potential mechanism of presynaptic modulation at central synapses.  相似文献   

12.
Neurotransmitter release occurs by regulated exocytosis from synaptic vesicles (SVs). Evolutionarily conserved proteins mediate the essential aspects of this process, including the membrane fusion step and priming steps that make SVs release-competent. Unlike the proteins constituting the core fusion machinery, the SV protein Mover does not occur in all species and all synapses. Its restricted expression suggests that Mover may modulate basic aspects of transmitter release and short-term plasticity. To test this hypothesis, we analyzed synaptic transmission electrophysiologically at the mouse calyx of Held synapse in slices obtained from wild-type mice and mice lacking Mover. Spontaneous transmission was unaffected, indicating that the basic release machinery works in the absence of Mover. Evoked release and vesicular release probability were slightly reduced, and the paired pulse ratio was increased in Mover knockout mice. To explore whether Mover’s role is restricted to certain subpools of SVs, we analyzed our data in terms of two models of priming. A model assuming two SV pools in parallel showed a reduced release probability of so-called “superprimed vesicles” while “normally primed” ones were unaffected. For the second model, which holds that vesicles transit sequentially from a loosely docked state to a tightly docked state before exocytosis, we found that knocking out Mover selectively decreased the release probability of tight state vesicles. These results indicate that Mover regulates a subclass of primed SVs in the mouse calyx of Held.

Synaptic transmission is initiated by exocytosis of neurotransmitters from presynaptic nerve terminals. Exocytosis involves tethering of synaptic vesicles (SVs) at release sites, priming to make SVs release-competent, and membrane fusion mediated by Synaptotagmins and SNAREs (1, 2). SVs in the release-competent state constitute the readily releasable pool (RRP) (3). Presynaptic proteins, such as Munc13s, Munc18s, and CAPS, are essential for generating the RRP and for regulating replenishment of this pool during short-term plasticity (4).Historically, the RRP had been regarded as a uniform set of primed SVs. But more recent evidence suggested that a fraction of SVs in the RRP might be more prone to undergoing exocytosis than others, and these SVs were dubbed “superprimed.” In this view, both primed and superprimed SVs are release-competent, albeit with different kinetics (57). More recently, evidence emerged that the primed states may be dynamic, reversible, and activity-dependent (8, 9) and they may correlate with morphological features of the synapse (10, 11). Furthermore, molecular evidence indicates that priming proceeds in distinct, reversible steps (12). These reports gave rise to a model which holds that primed SVs fluctuate between a loosely docked state (LS), in which SNARE complexes are partially zippered, and a tightly docked state (TS), in which SNARE zippering has progressed further. In this model, release predominantly occurs from TS vesicles; the transition from LS to TS is slow at rest but can occur rapidly in the presence of Ca2+, on a millisecond time scale (13).Exocytosis is abolished when components of the core machinery are perturbed, such as Munc13s, Munc18s, or Synaptotagmin-1. However, it is unknown whether there are proteins specifically fine-tuning parts of the priming process. Unlike most presynaptic proteins, the SV protein Mover is not expressed in all species and all synapses (14, 15). Mover is thus a prime candidate for modulating the ubiquitous release machinery. Here, we have tested the role of Mover for modulating synaptic transmission at the calyx of Held. Our data indicate that Mover regulates the release probability (pr) of a subset of primed SVs.  相似文献   

13.
14.
Current proteomic studies clarified canonical synaptic proteins that are common to many types of synapses. However, proteins of diversified functions in a subset of synapses are largely hidden because of their low abundance or structural similarities to abundant proteins. To overcome this limitation, we have developed an “ultra-definition” (UD) subcellular proteomic workflow. Using purified synaptic vesicle (SV) fraction from rat brain, we identified 1,466 proteins, three times more than reported previously. This refined proteome includes all canonical SV proteins, as well as numerous proteins of low abundance, many of which were hitherto undetected. Comparison of UD quantifications between SV and synaptosomal fractions has enabled us to distinguish SV-resident proteins from potential SV-visitor proteins. We found 134 SV residents, of which 86 are present in an average copy number per SV of less than one, including vesicular transporters of nonubiquitous neurotransmitters in the brain. We provide a fully annotated resource of all categorized SV-resident and potential SV-visitor proteins, which can be utilized to drive novel functional studies, as we characterized here Aak1 as a regulator of synaptic transmission. Moreover, proteins in the SV fraction are associated with more than 200 distinct brain diseases. Remarkably, a majority of these proteins was found in the low-abundance proteome range, highlighting its pathological significance. Our deep SV proteome will provide a fundamental resource for a variety of future investigations on the function of synapses in health and disease.

The functions of eukaryotic cells, in all their complexity, depend upon highly specific compartmentalization into subcellular domains, including organelles. These compartments represent functional units characterized by specific supramolecular protein complexes. A major goal of modern biology is to establish an exhaustive, quantitative inventory of the protein components of each intracellular compartment. Such inventories are points of departure, not only for functional understanding and reconstruction of biological systems, but also for a multitude of investigations, such as evolutionary diversification and derivation of general principles of biological regulation and homeostasis.Essential to communication within the nervous system, chemical synapses constitute highly specific compartments that are connected by axons to frequently distant neuronal cell bodies. Common to all chemical synapses are protein machineries that orchestrate exocytosis of synaptic vesicles (SVs) filled with neurotransmitters in response to presynaptic action potentials (APs), resulting in activation of postsynaptic receptors. Moreover, synapses are composed of structurally and functionally distinct subcompartments, such as free and docked SVs, endosomes, active zones (AZs) at the presynaptic side, and receptor-containing membranes with associated scaffold proteins on the postsynaptic side. Thus, it is not surprising that mass spectrometry (MS)-based proteomics, combined with subcellular fractionation, yields protein inventories of high complexity. For instance, >2,000 protein species were identified in synaptosomes (1), ∼400 in the SV fraction (2), ∼1,500 in postsynaptic densities (3), and ∼100 in an AZ-enriched preparation (4).While these studies provide insights into the protein composition of synaptic structures, they are still inherently limited for two reasons. First, synapses are functionally diverse with respect to the chemical nature of their neurotransmitters, as well as their synaptic strength, kinetics, and plasticity properties (5). Therefore, analyzed subcellular fractions represent “averages” of a great diversity of synapses (6) or SVs (2). The second limitation is that proteins known to be present in specific subsets were not found in these studies, despite the unprecedented sensitivity of modern mass spectrometers. In fact, many functionally critical synaptic proteins have remained undetected. For example, the synaptotagmin (Syt) family, major Ca2+ sensors of SV exocytosis, comprises >15 members, of which only 5 had been identified in previous SV proteomics (2, 4, 7). Missing isoforms included Syt7, involved in asynchronous transmitter release (8), synaptic plasticity (9), and SV recycling (10). Likewise, the vesicular transporters for monoamines (VMATs) and acetylcholine (VAChT) neurotransmitters were missing in these studies. Clearly, known components of the diversified synaptic proteome have been missing, and it is not possible to predict how many more such proteins remain hidden.What are the reasons for the continuing incompleteness of the synaptic protein inventory? Proteome identification and quantification rely heavily on MS detectability of peptides generated by digestion of extracted proteins with sequence-specific enzymes, such as trypsin. However, in MS analysis of complex biological samples, peptide signals from a few abundant proteins often mask those that are less abundant. Additionally, the probability of obtaining peptides with similar masses, but different amino acid sequences, increases with increasing sample complexity (11, 12). To overcome these limitations, we have elaborated a workflow with dual-enzymatic protein digestion in sequence combined with an extensive peptide separation prior to MS analysis. As proof of concept, we have utilized purified SV fractions from rat whole brain, which serve as a benchmark for quantitative organellar proteomics (2). As a result, we detected ∼1,500 proteins in the SV fraction, three times more than reported previously. This proteome not only covers all known canonical SV proteins but also contains proteins previously overlooked, such as the low-abundance Syts and SV transporters. Moreover, peptide quantification allowed for differentiating “SV-resident” from “SV-visitor” proteins. In fact, most “SV-resident” proteins revealed in our SV proteomics are of low abundance, with an average copy number of less than 1 per SV, suggesting a larger molecular and functional diversity of SVs than previously thought. Remarkably, more than 200 proteins detected in the SV fraction are genetically associated with brain disorders, 76% of which were previously hidden.  相似文献   

15.
In nerve cells the genes encoding for α2δ subunits of voltage-gated calcium channels have been linked to synaptic functions and neurological disease. Here we show that α2δ subunits are essential for the formation and organization of glutamatergic synapses. Using a cellular α2δ subunit triple-knockout/knockdown model, we demonstrate a failure in presynaptic differentiation evidenced by defective presynaptic calcium channel clustering and calcium influx, smaller presynaptic active zones, and a strongly reduced accumulation of presynaptic vesicle-associated proteins (synapsin and vGLUT). The presynaptic defect is associated with the downscaling of postsynaptic AMPA receptors and the postsynaptic density. The role of α2δ isoforms as synaptic organizers is highly redundant, as each individual α2δ isoform can rescue presynaptic calcium channel trafficking and expression of synaptic proteins. Moreover, α2δ-2 and α2δ-3 with mutated metal ion-dependent adhesion sites can fully rescue presynaptic synapsin expression but only partially calcium channel trafficking, suggesting that the regulatory role of α2δ subunits is independent from its role as a calcium channel subunit. Our findings influence the current view on excitatory synapse formation. First, our study suggests that postsynaptic differentiation is secondary to presynaptic differentiation. Second, the dependence of presynaptic differentiation on α2δ implicates α2δ subunits as potential nucleation points for the organization of synapses. Finally, our results suggest that α2δ subunits act as transsynaptic organizers of glutamatergic synapses, thereby aligning the synaptic active zone with the postsynaptic density.

In synapses neurotransmitter release is triggered by the entry of calcium through voltage-gated calcium channels (VGCCs). Neuronal VGCCs consist of an ion-conducting α1 subunit and the auxiliary β and α2δ subunits. α2δ subunits, the targets of the widely prescribed antiepileptic and antiallodynic drugs gabapentin and pregabalin, are membrane-anchored extracellular glycoproteins, which modulate VGCC trafficking and calcium currents (15). In nerve cells α2δ subunits have been linked to neuropathic pain and epilepsy (4) and they interact with mutant prion proteins (6) and regulate synaptic release probability (7). Importantly, all α2δ isoforms are implicated in synaptic functions. Presynaptic effects of α2δ-1, for example, may be mediated by an interaction with α-neurexins (8) or N-methyl-D-aspartate receptors (e.g., refs. 9 and 10). In contrast, postsynaptic α2δ-1 acts as a receptor for thrombospondins (11) and promotes spinogenesis via postsynaptic Rac1 (12). α2δ-2 is necessary for normal structure and function of auditory hair cell synapses (13); it has been identified as a regulator of axon growth and hence a suppressor of axonal regeneration (14) and was recently shown to control structure and function of cerebellar climbing fiber synapses (15). A splice variant of α2δ-2 regulates postsynaptic GABAA receptor (GABAAR) abundance and axonal wiring (16). In invertebrates, α2δ loss of function was associated with abnormal presynaptic development in motoneurons (17, 18) and in mice the loss of α2δ-3 results in aberrant synapse formation of auditory nerve fibers (19). Finally, α2δ-4 is required for the organization of rod and cone photoreceptor synapses (20, 21).Despite these important functions, knockout mice for α2δ-1 and α2δ-3 show only mild neurological phenotypes (5, 10, 2225). In contrast, mutant mice for α2δ-2 (ducky) display impaired gait, ataxia, and epileptic seizures (26), all phenotypes consistent with a cerebellar dysfunction due to the predominant expression of α2δ-2 in the cerebellum (e.g., ref. 15). Hence, in contrast to the specific functions of α2δ isoforms (discussed above) the phenotypes of the available knockout or mutant mouse models suggest a partial functional redundancy in central neurons. Moreover, detailed mechanistic insights into the putative synaptic functions of α2δ subunits are complicated by the simultaneous and strong expression of three isoforms (α2δ-1 to -3) in neurons of the central nervous system (27).In this study, by transfecting cultured hippocampal neurons from α2δ-2/-3 double-knockout mice with short hairpin RNA (shRNA) against α2δ-1, we developed a cellular α2δ subunit triple-knockout/knockdown model. Excitatory synapses from these cultures show a severe failure of synaptic vesicle recycling associated with severely reduced presynaptic calcium transients, loss of presynaptic calcium channels and presynaptic vesicle-associated proteins, and a reduced size of the presynaptic active zone (AZ). Lack of presynaptic α2δ subunits also induces a failure of postsynaptic PSD-95 and AMPA receptor (AMPAR) localization and a thinning of the postsynaptic density (PSD). Each individual α2δ isoform (α2δ-1 to -3) could rescue the severe phenotype, revealing the highly redundant role of presynaptic α2δ isoforms in glutamatergic synapse formation and differentiation. Together our results show that α2δ subunits regulate presynaptic differentiation as well as the transsynaptic alignment of postsynaptic receptors and are thus critical for the function of glutamatergic synapses.  相似文献   

16.
Heterozygous NRXN1 deletions constitute the most prevalent currently known single-gene mutation associated with schizophrenia, and additionally predispose to multiple other neurodevelopmental disorders. Engineered heterozygous NRXN1 deletions impaired neurotransmitter release in human neurons, suggesting a synaptic pathophysiological mechanism. Utilizing this observation for drug discovery, however, requires confidence in its robustness and validity. Here, we describe a multicenter effort to test the generality of this pivotal observation, using independent analyses at two laboratories of patient-derived and newly engineered human neurons with heterozygous NRXN1 deletions. Using neurons transdifferentiated from induced pluripotent stem cells that were derived from schizophrenia patients carrying heterozygous NRXN1 deletions, we observed the same synaptic impairment as in engineered NRXN1-deficient neurons. This impairment manifested as a large decrease in spontaneous synaptic events, in evoked synaptic responses, and in synaptic paired-pulse depression. Nrxn1-deficient mouse neurons generated from embryonic stem cells by the same method as human neurons did not exhibit impaired neurotransmitter release, suggesting a human-specific phenotype. Human NRXN1 deletions produced a reproducible increase in the levels of CASK, an intracellular NRXN1-binding protein, and were associated with characteristic gene-expression changes. Thus, heterozygous NRXN1 deletions robustly impair synaptic function in human neurons regardless of genetic background, enabling future drug discovery efforts.

Schizophrenia is a devastating brain disorder that affects millions of people worldwide and exhibits a strong genetic component. In a key discovery, deletions or duplications of larger stretches of chromosomal DNA that lead to copy number variations (CNVs) were identified two decades ago (1, 2). CNVs occur unexpectedly frequently, are often de novo, and usually affect multiple genes depending on the size of the deleted or duplicated stretch of DNA. Strikingly, the biggest genetic risk for schizophrenia was identified in three unrelated CNVs: a duplication of region 16p11.2 and deletions of 22q11.2 and of 2p16.3 (39). Of these CNVs, 16p11.2 and 22q11.2 CNVs affect more than 20 genes, whereas 2p16.3 CNVs impact only one or more exons of a single gene, NRXN1, which encodes the presynaptic cell-adhesion molecule neurexin-1 (4, 7, 912). NRXN1 CNVs confer an approximately 10-fold increase in risk of schizophrenia, and additionally strongly predispose to other neuropsychiatric disorders, especially autism and Tourette syndrome (13, 14). Moreover, genome-wide association studies using DNA microarrays identified common changes in many other genes that predispose to schizophrenia with smaller effect sizes (1521). Viewed together, these studies indicate that variations in a large number of genes are linked to schizophrenia. Among these genetic variations, heterozygous exonic CNVs of NRXN1 are rare events, but nevertheless constitute the most prevalent high-risk single-gene association at present.Neurexins are central regulators of neural circuits that control diverse synapse properties, such as the presynaptic release probability, the postsynaptic receptor composition, and synaptic plasticity (2228). To test whether heterozygous NRXN1 mutations might cause functional impairments in human neurons, we previously generated conditionally mutant human embryonic stem (ES) cells that enabled induction of heterozygous NRXN1 deletions using Cre-recombinase (29). We then analyzed the effects of the deletion on the properties of neurons induced from the conditionally mutant ES cells using forced expression of Ngn2, a method that generates a relatively homogeneous population of excitatory neurons that are also referred to as induced neuronal (iN) cells (30). These experiments thus examined isogenic neurons without or with a heterozygous NRXN1 loss-of-function mutation that mimicked the schizophrenia-associated 2p16.3 CNVs, enabling precise control of the genetic background. The heterozygous NRXN1 deletion produced a robust but discrete impairment in neurotransmitter release without major changes in neuronal development or morphology (29). These results were exciting because they suggested that a discrete impairment in neurotransmitter release could underlie the predisposition to schizophrenia conferred by the 2p16.3 CNV, but these experiments did not reveal whether the NRXN1 mutation induces the same synaptic impairment in schizophrenia patients (31).The present project was initiated to achieve multiple overlapping aims emerging from the initial study on human NRXN1 mutations (29). First, we aimed to validate or refute the results obtained with neurons generated from engineered conditionally mutant ES cells with neurons generated from patient-derived induced pluripotent stem (iPS) cells containing NRXN1 mutations (Fig. 1A). This goal was pursued in order to gain confidence in the disease-relevance of the observed phenotypes. Second, we wanted to test whether the observed phenotype is independent of the laboratory of analysis (i.e., whether it is sufficiently robust to be replicated at multiple sites) (Fig. 1A). This goal was motivated by the observation of limited reproducibility in some studies of the phenotypes of patient-derived neurons. We hypothesized that this lack of reproducibility is due to variations in experimental conditions rather than an experimental failure, and designed our studies to demonstrate robustness of the findings through replication. Third, we aimed to generate reagents that could be broadly used by the scientific community for investigating the cellular basis of neuropsychiatric disorders (32). This goal was prompted by the challenges posed by the finding that many different genes appear to be linked to schizophrenia. Fourth, we aimed to definitively establish or exclude the possibility that human neurons are uniquely sensitive to a heterozygous loss of NRXN1 compared with mouse neurons (Fig. 1B). The goal here was to test whether at least as regards to NRXN1, mouse and human neurons exhibit fundamental differences. Fifth and finally, we hoped to gain further insights into the mechanisms by which NRXN1 mutations predispose to schizophrenia, an obviously needed objective given our lack of understanding of this severe disorder. As described in detail below, our data provide advances toward meeting these goals, establishing unequivocally that heterozygous NRXN1 deletions in human but not in mouse neurons cause a robust impairment in neurotransmitter release that is replicable in multiple laboratories.Open in a separate windowFig. 1.Overall study design illustrating the experimental approach to analyze human heterozygous NRXN1 loss-of-function mutations, to achieve cross-laboratory and cross-platform validation of observed phenotypes, and to perform cross-paradigm evaluations of these phenotypes in human and mouse neurons. (A) Experimental strategy for analyzing the functional effects of heterozygous NRXN1 loss-of-function mutations in human patient-derived neurons and for validating the observed phenotypes in a cross-laboratory and cross-platform comparison. PBMCs from schizophrenia patients with NRXN1 deletions and from control individuals were reprogrammed into iPS cells by Rutgers University (RUCDR Infinite Biologics). iPS cells that passed QC were shipped to Stanford and to FCDI for expansion, banking, and transdifferentiation into induced neurons. The indicated subsequent analyses were carried out at Stanford University and at Rutgers University. FCDI manufactured industry-scale human induced neurons that were shipped to Rutgers for analysis, whereas Stanford generated induced neurons at an academic single-laboratory scale for analysis. (B) Experimental strategy to evaluate the conservation of NRXN1-deletion phenotypes observed in human neurons in mouse neurons (cross-paradigm evaluation). Human and mouse stem cells that carried heterozygous engineered conditional NRXN1/Nrxn1 deletions were transdifferentiated into neurons by Ngn2 expression and analyzed using similar approaches to ensure comparability. In this approach, isogenic human and mouse neurons without or with NRXN1/Nrxn1 deletions were compared to test whether side-by-side analysis of human and mouse neurons prepared by indistinguishable approaches yields similar phenotypes.  相似文献   

17.
As biological invasions continue to increase globally, eradication programs have been undertaken at significant cost, often without consideration of relevant ecological theory. Theoretical fisheries models have shown that harvest can actually increase the equilibrium size of a population, and uncontrolled studies and anecdotal reports have documented population increases in response to invasive species removal (akin to fisheries harvest). Both findings may be driven by high levels of juvenile survival associated with low adult abundance, often referred to as overcompensation. Here we show that in a coastal marine ecosystem, an eradication program resulted in stage-specific overcompensation and a 30-fold, single-year increase in the population of an introduced predator. Data collected concurrently from four adjacent regional bays without eradication efforts showed no similar population increase, indicating a local and not a regional increase. Specifically, the eradication program had inadvertently reduced the control of recruitment by adults via cannibalism, thereby facilitating the population explosion. Mesocosm experiments confirmed that adult cannibalism of recruits was size-dependent and could control recruitment. Genomic data show substantial isolation of this population and implicate internal population dynamics for the increase, rather than recruitment from other locations. More broadly, this controlled experimental demonstration of stage-specific overcompensation in an aquatic system provides an important cautionary message for eradication efforts of species with limited connectivity and similar life histories.

Theoretical population models can produce counterintuitive predictions regarding the consequences of harvest or removal of predatory species. These models show that for simple predator-prey systems, there can be positive population responses to predator mortality resulting from harvest for fisheries or population management, which can create an increased equilibrium level of that predator species (15). Among these mortality processes is the “hydra effect,” named after the mythical multi-headed serpent that grew two new heads for each one that was removed (6, 7). This counterintuitive outcome can be driven by a density-dependent process known as overcompensation. The hydra effect typically refers to higher equilibrium or time-averaged densities in response to increased mortality, typically involving consumer populations undergoing population cycles. Population increases in response to mortality can be the result of stage-specific overcompensation, which involves an increase in a specific life history stage or a size class following increased mortality. The first analysis of overcompensatory responses to mortality did not depend on stage specificity and was applied initially to fisheries harvests (1). Subsequent models have included stage specificity and have been applied to a broad range of systems in which species have been harvested for consumption or removed for population control of non-native species (4, 5, 815).Theory suggests that overcompensation in response to harvest or removal can occur for a variety of reasons, including 1) reduced competition for resources and increased adult reproduction rates, 2) faster rates of juvenile maturation or greater success in reaching the adult stage, and 3) increased juvenile or adult survival rates (17). An increase in reproductive output in response to reduced adult density can be the result of a reduction in resource competition (SI Appendix, Fig. S1).While there is substantial evidence that conditions that could produce density-dependent overcompensation occur frequently, evidence for overcompensation in natural populations is rare. For only a few populations do we have the long-term demographic data collected over a sufficiently long duration and for population densities over a wide enough range to detect this effect. Unfortunately, recent reviews of population increases in response to increased mortality do not include field studies with explicit controls for removals (1317).There are examples of density-dependent overcompensation from field populations (4, 1315), as well as a larger number of studies from the laboratory and greenhouse typically involving plant and insect populations (1822). Among the field examples is a population control program for smallmouth bass in a lake in upstate New York, which paradoxically resulted in greater bass abundance, primarily of juveniles, after 7 y of removal efforts (23, 24). Another field study in the United Kingdom showed that perch populations responded similarly when an unidentified pathogen decimated adults (25). Other programs that attempted to remove invasive fishes, including pikeperch in England (26), brook trout in Idaho (27), and Tilapia in Australia (28), showed similar results. However, although many of these examples involved well-executed studies with substantial field data, none had explicit controls for removal, such as comparable populations without harvest (or disease). Thus, despite the support of current theory in these studies, the contribution of external factors to observed population responses to harvest remains uncertain. To date, we are unaware of any experimental studies with comparable controls in a field population that demonstrates overcompensation in a single species (1315).  相似文献   

18.
Long-term potentiation (LTP) has long been considered as an important cellular mechanism for learning and memory. LTP expression involves NMDA receptor-dependent synaptic insertion of AMPA receptors (AMPARs). However, how AMPARs are recruited and anchored at the postsynaptic membrane during LTP remains largely unknown. In this study, using CRISPR/Cas9 to delete the endogenous AMPARs and replace them with the mutant forms in single neurons, we have found that the amino-terminal domain (ATD) of GluA1 is required for LTP maintenance. Moreover, we show that GluA1 ATD directly interacts with the cell adhesion molecule neuroplastin-65 (Np65). Neurons lacking Np65 exhibit severely impaired LTP maintenance, and Np65 deletion prevents GluA1 from rescuing LTP in AMPARs-deleted neurons. Thus, our study reveals an essential role for GluA1/Np65 binding in anchoring AMPARs at the postsynaptic membrane during LTP.

In 1973, Bliss and Lomo published the first observation of long-term potentiation (LTP) in which a tetanic stimulus caused a prolonged enhancement of synaptic transmission in rabbit hippocampus (1). Numerous studies have since demonstrated that LTP contributes to the neuronal mechanisms underlying learning and memory (24). The classic NMDA receptor (NMDAR)-dependent LTP is found in many brain regions and is studied mostly in hippocampal CA1 synapses (5, 6). Mechanistically, LTP can be divided into two sequential phases: initiation and maintenance. During LTP initiation, tetanic stimulation activates NMDARs that mediate rapid Ca2+ influx into dendritic spines, resulting in CaMKII activation, which subsequently recruits more AMPA-type glutamate receptors (AMPARs) into synapses, thus strengthening AMPAR-mediated excitatory postsynaptic currents (AMPAR-EPSCs) (7, 8). LTP maintenance is thought to require the newly recruited AMPARs to remain at the postsynaptic membrane for an extended period of time, a process called synaptic trapping (9).AMPAR cellular trafficking, synapse anchoring, and synaptic function are dependent on the subunit composition of the core functional ion channel, which consists of a tetramer of subunits GluA1-GluA4. Each subunit consists of an amino-terminal domain (ATD, also known as N-terminal domain), a ligand-binding domain, four membrane-spanning segments, and an intracellular C-terminal domain (CTD). In mouse hippocampal CA1 pyramidal neurons, the most common types of AMPAR subunits are GluA1, GluA2, and GluA3 (10). Early studies using virus-based overexpression of the green fluorecent protein (GFP)-tagged AMPAR subunits suggest that GluA1 and GluA2 have differential trafficking capabilities in hippocampal neurons (11, 12); GluA2/A3 heteromers are constitutively trafficked to dendritic spines, while the synaptic cooperation of GluA1-containing AMPARs is dependent on neuronal activity. Thus, a subunit-specific model for AMPAR trafficking has been proposed (13), and LTP is thought to require certain sequences or domains of GluA1 (14). The emergent roles of the GluA1 CTD in synaptic plasticity have been extensively documented (13, 1517). However, the observation that the CTD-lacking GluA1 is still present at the postsynaptic membrane and mediates LTP (18, 19) challenged the absolute requirement for GluA1 CTD in synaptic transmission and plasticity, indicating that other domains, such as the ATD, might have a previously uncovered role in LTP.Indeed, recent studies have revealed that the GluA1 ATD is required for synaptic transmission and LTP (20, 21). The ATD, which accounts for nearly half of the AMPAR coding sequence, projects nearly midway into the synaptic cleft where it may dynamically interact with proteins; such interactions might contribute to synaptic plasticity (22, 23). N-cadherin (24), a cell adhesion molecule, and neuronal pentraxins (25), secretory proteins, have been reported to associate with the ATDs of GluA2 and GluA4, respectively. However, whether GluA1 ATD has binding partners in the synaptic cleft remains unclear.In this study, we aimed to further understand the role of the GluA1 ATD in synaptic transmission and LTP and to investigate the molecular mechanism underlying its function. We found that the ATD is required for GluA1 synaptic function, both under basal conditions and during LTP. Furthermore, we have identified that GluA1 ATD directly interacts with neuroplastin-65 (referred to throughout as Np65), a single-transmembrane protein belonging to the immunoglobulin superfamily of cell adhesion molecules. Interaction of Np65 with the ATD of GluA1 is required for prolonged enhancement in synaptic transmission during LTP. Interestingly, it has been reported that as early as 20 y ago, Np65 antibody treatment causes impairment in LTP maintenance in hippocampal slices (26). Therefore, our results provide a molecular mechanism for GluA1- and Np65-mediated LTP maintenance.  相似文献   

19.
20.
Glutamate uptake into synaptic vesicles (SVs) depends on cation/H+ exchange activity, which converts the chemical gradient (ΔpH) into membrane potential (Δψ) across the SV membrane at the presynaptic terminals. Thus, the proper recruitment of cation/H+ exchanger to SVs is important in determining glutamate quantal size, yet little is known about its localization mechanism. Here, we found that secretory carrier membrane protein 5 (SCAMP5) interacted with the cation/H+ exchanger NHE6, and this interaction regulated NHE6 recruitment to glutamatergic presynaptic terminals. Protein–protein interaction analysis with truncated constructs revealed that the 2/3 loop domain of SCAMP5 is directly associated with the C-terminal region of NHE6. The use of optical imaging and electrophysiological recording showed that small hairpin RNA–mediated knockdown (KD) of SCAMP5 or perturbation of SCAMP5/NHE6 interaction markedly inhibited axonal trafficking and the presynaptic localization of NHE6, leading to hyperacidification of SVs and a reduction in the quantal size of glutamate release. Knockout of NHE6 occluded the effect of SCAMP5 KD without causing additional defects. Together, our results reveal that as a key regulator of axonal trafficking and synaptic localization of NHE6, SCAMP5 could adjust presynaptic strength by regulating quantal size at glutamatergic synapses. Since both proteins are autism candidate genes, the reduced quantal size by interrupting their interaction may underscore synaptic dysfunction observed in autism.

The uptake of classical transmitters into synaptic vesicles (SVs) depends on a proton electrochemical gradient (ΔμH+), consisting of the chemical gradient (ΔpH) and membrane potential (Δψ) across the SV membrane (1). ΔμH+ is generated by vacuolar-type H+-ATPases and ion channels/transporters on SVs (2). There are five classes of vesicular neurotransmitter transporters on SVs, namely, vesicular glutamate transporters (VGLUT 1 to 3), vesicular GABA/glycine transporter (VGAT, VIAAT), vesicular monoamine transporters (VMAT 1, 2), vesicular acetylcholine transporter (VAChT), and vesicular nucleotide transporter (VNUT); these all use ΔpH and Δψ, but to different extents, to fill SVs with neurotransmitters (3). For example, transporters for the anionic neurotransmitter, glutamate, mainly utilize Δψ, whereas the transporters for the cationic neurotransmitter, acetylcholine, and zwitterionic neurotransmitters, GABA and glycine, depend on ΔpH and both ΔpH and Δψ, respectively (4). Therefore, homeostatic regulation of ΔμH+ is important for controlling the quantal size of neurotransmitter release.Cation/H+ exchange activity across the membrane is mostly attributed to monovalent Na+(K+)/H+ exchangers (NHEs) that present at the plasma membrane or intracellular organelles (5). In humans, the NHE superfamily comprises nine isoforms consisting of NHE1 to NHE5 on the plasma membrane, NHE6 and NHE9 on intracellular vesicles, and NHE7 and NHE8 on the Golgi apparatus (6). Plasma membrane isoforms recognize Na+ but not K+ and have important roles in the regulation of cytoplasmic pH, while the intracellular isoforms recognize K+ as well as Na+ (7), but their physiological roles remain poorly understood. Loss-of-function mutations of NHE6 and NHE9, the two endosomal subtypes (eNHEs), are implicated in multiple neurodevelopmental and neuropsychiatric disorders, including autism, Christianson syndrome, X-linked intellectual disability, and Angelman syndrome (813). NHE6 and NHE9 are highly expressed in the brain, including the hippocampus and cortex (14). Previous studies have found that SVs show an NHE activity that plays an important role in glutamate uptake into SVs by dissipating ΔpH and increasing Δψ (15, 16), and thus, eNHEs were considered to reside in SVs to perform this function (14). However, it was found that there was no defect in vesicular filling with glutamate or GABA in NHE9 knockout (KO) neurons, and NHE9 regulated the luminal pH of axonal endosomes rather than recycling SVs (17). In contrast, NHE6 was identified on SVs by using quantitative proteomics (18), and its presynaptic localization was shown by immunofluorescent analysis (19). In addition, more severe synaptic dysfunction was observed in NHE6 KO mice (19) than in NHE9 KO mice (17). These results suggest that NHE6 and NHE9 are not functionally redundant, and NHE6 is responsible for NHE activity in SVs that regulates glutamate uptake at presynaptic terminals. Evidently, the proper localization of NHE6 to SVs is of utmost importance in determining the glutamate quantal size, but the mechanism underlying NHE6 recruitment to SVs is mostly unknown.Secretory carrier membrane proteins (SCAMPs) are known to regulate vesicular trafficking and vesicle recycling processes. Of the five known SCAMPs, SCAMP1 and SCAMP5 are highly expressed in the brain and enriched in SVs (20). The study of SCAMP1 KO mice showed that it was not necessary for brain function and synaptic physiology (21), suggesting a critical role of SCAMP5 in synaptic function. Indeed, a recent genetic analysis showed that SCAMP5 was silenced on a derivative chromosome and was reduced in expression to ∼40% of normal in a patient with idiopathic, sporadic autism (22), and several SCAMP5 mutations reported in humans have been implicated in neurodevelopmental and neurodegenerative disorders such as intellectual disability, seizure, and Parkinson’s disease (23, 24). We also have provided evidence that SCAMP5 plays a critical role in SV endocytosis during strong neuronal activity (25) and the protein clearance at the presynaptic SV release site (26). These results suggest that SCAMP5 plays an important role in regulating the function and trafficking of synaptic proteins at presynaptic terminals.In this study, we showed that SCAMP5 directly interacted with NHE6. Optical imaging and electrophysiological recordings proved that when perturbing their interaction, axonal trafficking and synaptic localization of NHE6 were severely impaired, which subsequently lowered the resting luminal pH of SVs and reduced the amounts of glutamate release. Knockout of NHE6 occluded the effect of SCAMP5 knockdown (KD) without causing additional defects. Since NHE6 and SCAMP5 are candidate genes for autism (9, 22), the reduced quantal size following impairment of their interaction may relate to the synaptic dysfunction observed in autism.  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