Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 174(6): 1450-1464.e23, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100184

ABSTRACT

Synapses are fundamental units of communication in the brain. The prototypical synapse-organizing complex neurexin-neuroligin mediates synapse development and function and is central to a shared genetic risk pathway in autism and schizophrenia. Neurexin's role in synapse development is thought to be mediated purely by its protein domains, but we reveal a requirement for a rare glycan modification. Mice lacking heparan sulfate (HS) on neurexin-1 show reduced survival, as well as structural and functional deficits at central synapses. HS directly binds postsynaptic partners neuroligins and LRRTMs, revealing a dual binding mode involving intrinsic glycan and protein domains for canonical synapse-organizing complexes. Neurexin HS chains also bind novel ligands, potentially expanding the neurexin interactome to hundreds of HS-binding proteins. Because HS structure is heterogeneous, our findings indicate an additional dimension to neurexin diversity, provide a molecular basis for fine-tuning synaptic function, and open therapeutic directions targeting glycan-binding motifs critical for brain development.


Subject(s)
Heparitin Sulfate/metabolism , Neural Cell Adhesion Molecules/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Glycopeptides/analysis , Heparitin Sulfate/chemistry , Humans , Membrane Proteins , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/antagonists & inhibitors , Neural Cell Adhesion Molecules/genetics , Neurons/cytology , Neurons/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Rats , Sequence Alignment
2.
J Neurosci ; 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942471

ABSTRACT

The mechanisms utilized by neurons to regulate the efficacy of phasic and tonic inhibition and their impacts on synaptic plasticity and behavior are incompletely understood. Cleft lip and palate transmembrane protein 1 (Clptm1) is a membrane-spanning protein that interacts with multiple γ-aminobutyric acid type A receptor (GABAAR) subunits, trapping them in the endoplasmic reticulum and Golgi network. Overexpression and knockdown studies suggest that Clptm1 modulates GABAAR-mediated phasic inhibition and tonic inhibition as well as activity-induced inhibitory synaptic homeostasis in cultured hippocampal neurons. To investigate the role of Clptm1 in the modulation of GABAARs in vivo, we generated Clptm1 knockout mice. Here, we show that genetic knockout of Clptm1 elevated phasic and tonic inhibitory transmission in both male and female heterozygous mice. Although basal excitatory synaptic transmission was not affected, Clptm1 haploinsufficiency significantly blocked high-frequency stimulation induced long-term potentiation in hippocampal CA3-CA1 synapses. In the hippocampus-dependent contextual fear conditioning behavior task, both male and female Clptm1 heterozygous knockout mice exhibited impairment in contextual fear memory. In addition, LTP and contextual fear memory were rescued by application of L-655,708, a negative allosteric modulator of the extrasynaptic GABAAR α5 subunit. These results suggest that haploinsufficiency of Clptm1 contributes to cognitive deficits through altered synaptic transmission and plasticity by elevation of inhibitory neurotransmission, with tonic inhibition playing a major role.Significance Statement The CLPTM1 gene was originally identified as disrupted in a family with cleft lip and palate. At the molecular level, Clptm1 interacts with multiple GABAA receptor subunits to limit their surface expression. Here, we generated Clptm1 knockout mice to uncover its functions in vivo. Clptm1 not only limited hippocampal inhibitory phasic and tonic transmission, it was required for excitatory synaptic plasticity and hippocampus-dependent cognitive function. A modulator of extrasynaptic GABAA receptors rescued the deficits in plasticity and behavior in Clptm1 heterozygous knockout mice, indicating the importance of tonic inhibition. These findings reveal a role for Clptm1 in balancing inhibitory strength and raise the possibility that disruptions of Clptm1 function may contribute to synaptic and cognitive deficits in neurological diseases.

3.
J Neurosci ; 44(14)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38360747

ABSTRACT

Growing evidence suggests a remarkable diversity and complexity in the molecular composition of synapses, forming the basis for the brain to execute complex behaviors. Hence, there is considerable interest in visualizing the spatial distribution of such molecular diversity at individual synapses within intact brain circuits. Yet this task presents significant technical challenges. Expansion microscopy approaches have revolutionized our view of molecular anatomy. However, their use to study synapse-related questions outside of the labs developing them has been limited. Here we independently adapted a version of Magnified Analysis of the Proteome (MAP) and present a step-by-step protocol for visualizing over 40 synaptic proteins in brain circuits. Surprisingly, our findings show that the advantage of MAP over conventional immunolabeling was primarily due to improved antigen recognition and secondarily physical expansion. Furthermore, we demonstrated the versatile use of MAP in brains perfused with paraformaldehyde or fresh-fixed with formalin and in formalin-fixed paraffin-embedded tissue. These tests expand the potential applications of MAP to combinations with slice electrophysiology or clinical pathology specimens. Using male and female mice expressing YFP-ChR2 exclusively in interneurons, we revealed a distinct composition of AMPA and NMDA receptors and Shank family members at synapses on hippocampal interneurons versus on pyramidal neurons. Quantitative single synapse analyses yielded comprehensive cell type distributions of synaptic proteins and their relationships. These findings exemplify the value of the versatile adapted MAP procedure presented here as an accessible tool for the broad neuroscience community to unravel the complexity of the "synaptome" across brain circuits and disease states.


Subject(s)
Proteome , Synapses , Mice , Male , Animals , Female , Proteome/metabolism , Synapses/physiology , Pyramidal Cells/physiology , Brain/metabolism , Formaldehyde , Hippocampus/metabolism
4.
J Neurosci ; 42(48): 8936-8947, 2022 11 30.
Article in English | MEDLINE | ID: mdl-36261284

ABSTRACT

In addition to its role in Alzheimer's disease, amyloid precursor protein (APP) has physiological roles in synapse development and function. APP induces presynaptic differentiation when presented to axons, but the mechanism is unknown. Here we show that APP binds neurexin to mediate this synaptogenic activity. APP specifically binds ß not α neurexins modulated by splice site 4. Binding to neurexin heparan sulfate glycan and LNS protein domains is required for high-affinity interaction and for full-length APP to recruit axonal neurexin. The synaptogenic activity of APP is abolished by triple knockdown of neurexins in hippocampal neurons pooled from male and female rats. Based on these and previous results, our model is that a dendritic-axonal trans dimer of full-length APP binds to axonal neurexin-ß to promote synaptic differentiation and function. Furthermore, soluble sAPPs also bind neurexin-ß and inhibit its interaction with neuroligin-1, raising the possibility that disruption of neurexin function by altered levels of full-length APP and its cleavage products may contribute to early synaptic deficits in Alzheimer's disease.SIGNIFICANCE STATEMENT The prevailing model for the basis of Alzheimer's disease is the amyloid cascade triggered by altered cleavage of amyloid precursor protein (APP). APP also has physiological roles at the synapse, but the molecular basis for its synaptic functions is not well understood. Here, we show that APP binds the presynaptic organizing protein neurexin-ß and that neurexin is essential for the synaptogenic activity of APP. Furthermore, soluble APP forms generated by APP cleavage also bind neurexin-ß and can block interaction with transmembrane synaptogenic ligands of neurexin. These findings reveal a role for neurexin-APP interaction in synapse development and raise the possibility that disruptions of neurexin function may contribute to synaptic and cognitive deficits in the critical early stage of Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Male , Female , Rats , Animals , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/metabolism , Nerve Tissue Proteins/metabolism , Synapses/physiology , Neurons/physiology
5.
Proc Natl Acad Sci U S A ; 115(23): E5382-E5389, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29784826

ABSTRACT

Leucine-rich repeat transmembrane (LRRTM) proteins are synaptic cell adhesion molecules that influence synapse formation and function. They are genetically associated with neuropsychiatric disorders, and via their synaptic actions likely regulate the establishment and function of neural circuits in the mammalian brain. Here, we take advantage of the generation of a LRRTM1 and LRRTM2 double conditional knockout mouse (LRRTM1,2 cKO) to examine the role of LRRTM1,2 at mature excitatory synapses in hippocampal CA1 pyramidal neurons. Genetic deletion of LRRTM1,2 in vivo in CA1 neurons using Cre recombinase-expressing lentiviruses dramatically impaired long-term potentiation (LTP), an impairment that was rescued by simultaneous expression of LRRTM2, but not LRRTM4. Mutation or deletion of the intracellular tail of LRRTM2 did not affect its ability to rescue LTP, while point mutations designed to impair its binding to presynaptic neurexins prevented rescue of LTP. In contrast to previous work using shRNA-mediated knockdown of LRRTM1,2, KO of these proteins at mature synapses also caused a decrease in AMPA receptor-mediated, but not NMDA receptor-mediated, synaptic transmission and had no detectable effect on presynaptic function. Imaging of recombinant photoactivatable AMPA receptor subunit GluA1 in the dendritic spines of cultured neurons revealed that it was less stable in the absence of LRRTM1,2. These results illustrate the advantages of conditional genetic deletion experiments for elucidating the function of endogenous synaptic proteins and suggest that LRRTM1,2 proteins help stabilize synaptic AMPA receptors at mature spines during basal synaptic transmission and LTP.


Subject(s)
CA1 Region, Hippocampal/physiology , Long-Term Potentiation/physiology , Neural Cell Adhesion Molecules/deficiency , Pyramidal Cells/physiology , Receptors, AMPA/metabolism , Animals , CA1 Region, Hippocampal/metabolism , Dendritic Spines/metabolism , Excitatory Postsynaptic Potentials/physiology , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synaptic Transmission/physiology
6.
PLoS Comput Biol ; 15(6): e1007113, 2019 06.
Article in English | MEDLINE | ID: mdl-31211786

ABSTRACT

In order to further our understanding of how gene expression contributes to key functional properties of neurons, we combined publicly accessible gene expression, electrophysiology, and morphology measurements to identify cross-cell type correlations between these data modalities. Building on our previous work using a similar approach, we distinguished between correlations which were "class-driven," meaning those that could be explained by differences between excitatory and inhibitory cell classes, and those that reflected graded phenotypic differences within classes. Taking cell class identity into account increased the degree to which our results replicated in an independent dataset as well as their correspondence with known modes of ion channel function based on the literature. We also found a smaller set of genes whose relationships to electrophysiological or morphological properties appear to be specific to either excitatory or inhibitory cell types. Next, using data from PatchSeq experiments, allowing simultaneous single-cell characterization of gene expression and electrophysiology, we found that some of the gene-property correlations observed across cell types were further predictive of within-cell type heterogeneity. In summary, we have identified a number of relationships between gene expression, electrophysiology, and morphology that provide testable hypotheses for future studies.


Subject(s)
Electrophysiological Phenomena/physiology , Neurons , Transcriptome/physiology , Animals , Computational Biology , Gene Expression Profiling , Mice , Models, Biological , Neurons/classification , Neurons/metabolism , Neurons/physiology , Single-Cell Analysis , Visual Cortex/cytology
7.
J Neurosci ; 35(36): 12425-31, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26354911

ABSTRACT

Neurotrophin-3 (NT-3) and its high-affinity receptor TrkC play crucial trophic roles in neuronal differentiation, axon outgrowth, and synapse development and plasticity in the nervous system. We demonstrated previously that postsynaptic TrkC functions as a glutamatergic synapse-inducing (synaptogenic) cell adhesion molecule trans-interacting with presynaptic protein tyrosine phosphatase σ (PTPσ). Given that NT-3 and PTPσ bind distinct domains of the TrkC extracellular region, here we tested the hypothesis that NT-3 modulates TrkC/PTPσ binding and synaptogenic activity. NT-3 enhanced PTPσ binding to cell surface-expressed TrkC and facilitated the presynapse-inducing activity of TrkC in rat hippocampal neurons. Imaging of recycling presynaptic vesicles combined with TrkC knockdown and rescue approaches demonstrated that NT-3 rapidly potentiates presynaptic function via binding endogenous postsynaptic TrkC in a tyrosine kinase-independent manner. Thus, NT-3 positively modulates the TrkC-PTPσ complex for glutamatergic presynaptic assembly and function independently from TrkC kinase activation. Our findings provide new insight into synaptic roles of neurotrophin signaling and mechanisms controlling synaptic organizing complexes. Significance statement: Although many synaptogenic adhesion complexes have been identified in recent years, little is known about modulatory mechanisms. Here, we demonstrate a novel role of neurotrophin-3 in synaptic assembly and function as a positive modulator of the TrkC-protein tyrosine phosphatase σ complex. This study provides new insight into the involvement of neurotrophin signaling in synapse development and plasticity, presenting a molecular mechanism that may underlie previous observations of short- and long-term enhancement of presynaptic function by neurotrophin. Given the links of synaptogenic adhesion molecules to autism and schizophrenia, this study might also contribute to a better understanding of the pathogenesis of these disorders and provide a new direction for ameliorating imbalances in synaptic signaling networks.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Neurotrophin 3/metabolism , Receptor, trkC/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Synapses/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Hippocampus/cytology , Male , Neurons/physiology , Protein Binding , Rats , Receptor, trkC/genetics , Synapses/physiology
8.
J Neurosci ; 35(22): 8462-79, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26041915

ABSTRACT

NMDA receptors play a central role in shaping the strength of synaptic connections throughout development and in mediating synaptic plasticity mechanisms that underlie some forms of learning and memory formation in the CNS. In the hippocampus and the neocortex, GluN1 is combined primarily with GluN2A and GluN2B, which are differentially expressed during development and confer distinct molecular and physiological properties to NMDA receptors. The contribution of each subunit to the synaptic traffic of NMDA receptors and therefore to their role during development and in synaptic plasticity is still controversial. We report a critical role for the GluN2B subunit in regulating NMDA receptor synaptic targeting. In the absence of GluN2B, the synaptic levels of AMPA receptors are increased and accompanied by decreased constitutive endocytosis of GluA1-AMPA receptor. We used quantitative proteomic analysis to identify changes in the composition of postsynaptic densities from GluN2B(-/-) mouse primary neuronal cultures and found altered levels of several ubiquitin proteasome system components, in particular decreased levels of proteasome subunits. Enhancing the proteasome activity with a novel proteasome activator restored the synaptic levels of AMPA receptors in GluN2B(-/-) neurons and their endocytosis, revealing that GluN2B-mediated anchoring of the synaptic proteasome is responsible for fine tuning AMPA receptor synaptic levels under basal conditions.


Subject(s)
Brain/cytology , Neurons/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Endocytosis/physiology , Excitatory Amino Acid Agents/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hydrazones/pharmacology , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Subcellular Fractions/metabolism , Subcellular Fractions/ultrastructure , Synapses/drug effects , Tetrodotoxin/pharmacology , Time Factors , ras GTPase-Activating Proteins/metabolism
9.
Neurobiol Dis ; 89: 223-34, 2016 May.
Article in English | MEDLINE | ID: mdl-26581639

ABSTRACT

Global cerebral ischemia induces selective degeneration of specific subsets of neurons throughout the brain, particularly in the hippocampus and cortex. One of the major hallmarks of cerebral ischemia is excitotoxicity, characterized by overactivation of glutamate receptors leading to intracellular Ca(2+) overload and ultimately neuronal demise. N-methyl-d-aspartate receptors (NMDARs) are considered to be largely responsible for excitotoxic injury due to their high Ca(2+) permeability. In the hippocampus and cortex, these receptors are most prominently composed of combinations of two GluN1 subunits and two GluN2A and/or GluN2B subunits. Due to the controversy regarding the differential role of GluN2A and GluN2B subunits in excitotoxic cell death, we investigated the role of GluN2B in the activation of pro-death signaling following an in vitro model of global ischemia, oxygen and glucose deprivation (OGD). For this purpose, we used GluN2B(-/-) mouse cortical cultures and observed that OGD-induced damage was reduced in these neurons, and partially prevented in wild-type rat neurons by a selective GluN2B antagonist. Notably, we found a crucial role of the C-terminal domain of the GluN2B subunit in triggering excitotoxic signaling. Indeed, expression of YFP-GluN2B C-terminus mutants for the binding sites to post-synaptic density protein 95 (PSD95), Ca(2+)-calmodulin kinase IIα (CaMKIIα) or clathrin adaptor protein 2 (AP2) failed to mediate neuronal death in OGD conditions. We focused on the GluN2B-CaMKIIα interaction and found a determinant role of this interaction in OGD-induced death. Inhibition or knock-down of CaMKIIα exerted a neuroprotective effect against OGD-induced death, whereas overexpression of this kinase had a detrimental effect. Importantly, in comparison with neurons overexpressing wild-type CaMKIIα, neurons overexpressing a mutant form of the kinase (CaMKII-I205K), unable to interact with GluN2B, were partially protected against OGD-induced damage. Taken together, our results identify crucial determinants in the C-terminal domain of GluN2B subunits in promoting neuronal death in ischemic conditions. These mechanisms underlie the divergent roles of the GluN2A- and GluN2B-NMDARs in determining neuronal fate in cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Cell Death , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Hippocampus/metabolism , In Vitro Techniques , Mice , Mice, Knockout , Protein Subunits/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics
10.
J Biol Chem ; 289(50): 34530-42, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25352602

ABSTRACT

Calsyntenin 3 (Cstn3 or Clstn3), a recently identified synaptic organizer, promotes the development of synapses. Cstn3 localizes to the postsynaptic membrane and triggers presynaptic differentiation. Calsyntenin members play an evolutionarily conserved role in memory and learning. Cstn3 was recently shown in cell-based assays to interact with neurexin 1α (n1α), a synaptic organizer that is implicated in neuropsychiatric disease. Interaction would permit Cstn3 and n1α to form a trans-synaptic complex and promote synaptic differentiation. However, it is contentious whether Cstn3 binds n1α directly. To understand the structure and function of Cstn3, we determined its architecture by electron microscopy and delineated the interaction between Cstn3 and n1α biochemically and biophysically. We show that Cstn3 ectodomains form monomers as well as tetramers that are stabilized by disulfide bonds and Ca(2+), and both are probably flexible in solution. We show further that the extracellular domains of Cstn3 and n1α interact directly and that both Cstn3 monomers and tetramers bind n1α with nanomolar affinity. The interaction is promoted by Ca(2+) and requires minimally the LNS domain of Cstn3. Furthermore, Cstn3 uses a fundamentally different mechanism to bind n1α compared with other neurexin partners, such as the synaptic organizer neuroligin 2, because Cstn3 does not strictly require the sixth LNS domain of n1α. Our structural data suggest how Cstn3 as a synaptic organizer on the postsynaptic membrane, particularly in tetrameric form, may assemble radially symmetric trans-synaptic bridges with the presynaptic synaptic organizer n1α to recruit and spatially organize proteins into networks essential for synaptic function.


Subject(s)
Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Cattle , Extracellular Space/metabolism , HEK293 Cells , Humans , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Rats , Receptors, Cell Surface/chemistry , Synapses/metabolism
11.
J Biol Chem ; 289(42): 29350-64, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25190809

ABSTRACT

Synapses, the basic units of communication in the brain, require complex molecular machinery for neurotransmitter release and reception. Whereas numerous components of excitatory postsynaptic sites have been identified, relatively few proteins are known that function at inhibitory postsynaptic sites. One such component is neuroligin-2 (NL2), an inhibitory synapse-specific cell surface protein that functions in cell adhesion and synaptic organization via binding to neurexins. In this study, we used a transgenic tandem affinity purification and mass spectrometry strategy to isolate and characterize NL2-associated complexes. Complexes purified from brains of transgenic His6-FLAG-YFP-NL2 mice showed enrichment in the Gene Ontology terms cell-cell signaling and synaptic transmission relative to complexes purified from wild type mice as a negative control. In addition to expected components including GABA receptor subunits and gephyrin, several novel proteins were isolated in association with NL2. Based on the presence of multiple components involved in trafficking and endocytosis, we showed that NL2 undergoes dynamin-dependent endocytosis in response to soluble ligand and colocalizes with VPS35 retromer in endosomes. Inhibitory synapses in brain also present a particular challenge for imaging. Whereas excitatory synapses on spines can be imaged with a fluorescent cell fill, inhibitory synapses require a molecular tag. We find the His6-FLAG-YFP-NL2 to be a suitable tag, with the unamplified YFP signal localizing appropriately to inhibitory synapses in multiple brain regions including cortex, hippocampus, thalamus, and basal ganglia. Altogether, we characterize NL2-associated complexes, demonstrate regulated trafficking of NL2, and provide tools for further proteomic and imaging studies of inhibitory synapses.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Nerve Tissue Proteins/metabolism , Proteomics/methods , Synapses/metabolism , Animals , Brain/metabolism , COS Cells , Chlorocebus aethiops , Endocytosis , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/physiology , Neurons/metabolism , Protein Transport/genetics , Proteome , Synaptic Transmission/physiology , Transgenes
12.
J Neurosci ; 33(13): 5867-5877, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23536098

ABSTRACT

Although the contribution of postsynaptic mechanisms to long-term synaptic plasticity has been studied extensively, understanding the contribution of presynaptic modifications to this process lags behind, primarily because of a lack of techniques with which to directly and quantifiably measure neurotransmitter release from synaptic terminals. Here, we developed a method to measure presynaptic activity through the biotinylation of vesicular transporters in vesicles fused with presynaptic membranes during neurotransmitter release. This method allowed us for the first time to selectively quantify the spontaneous or evoked release of glutamate or GABA at their respective synapses. Using this method to investigate presynaptic changes during the expression of group I metabotropic glutamate receptor (mGluR1/5)-mediated long-term depression (LTD) in cultured rat hippocampal neurons, we discovered that this form of LTD was associated with increased presynaptic release of glutamate, despite reduced miniature EPSCs measured with whole-cell recording. Moreover, we found that specific blockade of AMPA receptor (AMPAR) endocytosis with a membrane-permeable GluR2-derived peptide not only prevented the expression of LTD but also eliminated LTD-associated increase in presynaptic release. Thus, our work not only demonstrates that mGluR1/5-mediated LTD is associated with increased endocytosis of postsynaptic AMPARs but also reveals an unexpected homeostatic/compensatory increase in presynaptic release. In addition, this study indicates that biotinylation of vesicular transporters in live cultured neurons is a valuable tool for studying presynaptic function.


Subject(s)
Long-Term Synaptic Depression/physiology , Neurons/cytology , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Receptors, Metabotropic Glutamate/metabolism , Analysis of Variance , Animals , Bicuculline/analogs & derivatives , Bicuculline/pharmacology , Biophysics , Biotin/analogs & derivatives , Biotin/pharmacology , Biotinylation , Electric Stimulation , Endocytosis/drug effects , Endocytosis/physiology , Excitatory Amino Acid Antagonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , Hippocampus/cytology , Long-Term Synaptic Depression/drug effects , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Microtubule-Associated Proteins/metabolism , Neurons/physiology , Organ Culture Techniques , Patch-Clamp Techniques , Peptides/pharmacology , Potassium/pharmacology , Protein Transport/drug effects , Protein Transport/physiology , Quinoxalines/pharmacology , Rats , Receptors, AMPA/chemistry , Receptors, Transferrin/metabolism , Sodium Channel Blockers/pharmacology , Succinimides/pharmacology , Synaptotagmin I/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tetrodotoxin/pharmacology , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
13.
Front Mol Neurosci ; 17: 1381534, 2024.
Article in English | MEDLINE | ID: mdl-38783902

ABSTRACT

Functions of the cerebellar cortex, from motor learning to emotion and cognition, depend on the appropriate molecular composition at diverse synapse types. Glutamate receptor distributions have been partially mapped using immunogold electron microscopy. However, information is lacking on the distribution of many other components, such as Shank2, a postsynaptic scaffolding protein whose cerebellar dysfunction is associated with autism spectrum disorders. Here, we used an adapted Magnified Analysis of the Proteome, an expansion microscopy approach, to map multiple glutamate receptors, scaffolding and signaling proteins at single synapse resolution in the cerebellar cortex. Multiple distinct synapse-selective distribution patterns were observed. For example, AMPA receptors were most concentrated at synapses on molecular layer interneurons and at climbing fiber synapses, Shank1 was most concentrated at parallel fiber synapses on Purkinje cells, and Shank2 at both climbing fiber and parallel fiber synapses on Purkinje cells but little on molecular layer interneurons. Our results are consistent with gene expression data but also reveal input-selective targeting within Purkinje cells. In specialized glomerular structures of the granule cell layer, AMPA receptors as well as most other synaptic components preferentially targeted to synapses. However, NMDA receptors and the synaptic GTPase activating protein SynGAP preferentially targeted to extrasynaptic sites. Thus, glomeruli may be considered integrative signaling units through which mossy fibers differentially activate synaptic AMPA and extrasynaptic NMDA receptor complexes. Furthermore, we observed NMDA receptors and SynGAP at adherens junctions, suggesting a role in structural plasticity of glomeruli. Altogether, these data contribute to mapping the cerebellar 'synaptome'.

14.
J Neurosci ; 32(12): 3992-4003, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22442066

ABSTRACT

The NMDAR plays a unique and vital role in subcellular signaling. Calcium influx initiates signaling cascades important for both synaptic plasticity and survival; however, overactivation of the receptor leads to toxicity and cell death. This dichotomy is partially explained by the subcellular location of the receptor. NMDARs located at the synapse stimulate cell survival pathways, while extrasynaptic receptors signal for cell death. Thus far, this interplay between synaptic and extrasynaptic NMDARs has been studied exclusively in cortical (CTX) and hippocampal neurons. It was unknown whether other cell types, such as GABAergic medium-sized spiny projection neurons of the striatum (MSNs), which bear the brunt of neurodegeneration in Huntington's disease, follow the same pattern. Here we report synaptic versus extrasynaptic NMDAR signaling in striatal MSNs and resultant activation of cAMP response element binding protein (CREB), in rat primary corticostriatal cocultures. Similarly to CTX, we found in striatal MSNs that synaptic NMDARs activate CREB, whereas extrasynaptic NMDARs dominantly oppose CREB activation. However, MSNs are much less susceptible to NMDA-mediated toxicity than CTX cells and show differences in subcellular GluN2B distribution. Blocking NMDARs with memantine (30 µm) or GluN2B-containing receptors with ifenprodil (3 µm) prevents CREB shutoff effectively in CTX and MSNs, and also rescues both neuronal types from NMDA-mediated toxicity. This work may provide cell and NMDAR subtype-specific targets for treatment of diseases with putative NMDAR involvement, including neurodegenerative disorders and ischemia.


Subject(s)
Cerebral Cortex/cytology , Corpus Striatum/cytology , Neurons/cytology , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Synapses/physiology , 4-Aminopyridine/pharmacology , Analysis of Variance , Animals , Bicuculline/pharmacology , CREB-Binding Protein/metabolism , Calcium Channel Blockers/pharmacology , Cells, Cultured , Coculture Techniques , Electric Stimulation , Embryo, Mammalian , Excitatory Amino Acid Agents/pharmacology , Female , GABA-A Receptor Antagonists/pharmacology , Glutamate Decarboxylase/metabolism , Glycine Agents/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microfluidic Analytical Techniques/methods , N-Methylaspartate/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Nifedipine/pharmacology , Patch-Clamp Techniques , Potassium Channel Blockers/pharmacology , Pregnancy , Rats , Rats, Wistar , Sodium Channel Blockers/pharmacology , Strychnine/pharmacology , Tetrodotoxin/pharmacology , Transfection/methods , Vesicular Glutamate Transport Protein 1/metabolism
15.
J Biol Chem ; 287(33): 27432-45, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22740692

ABSTRACT

Trafficking of NMDA receptors to the surface of neurons and to synapses is critical for proper brain function and activity-dependent plasticity. Recent evidence suggests that surface trafficking of other ionotropic glutamate receptors requires ligand binding for exit from the endoplasmic reticulum. Here, we show that glutamate binding to GluN2 is required for trafficking of NMDA receptors to the cell surface. We expressed a panel of GluN2B ligand binding mutants in heterologous cells with GluN1 or in rat cultured neurons and found that surface expression correlates with glutamate efficacy. Such a correlation was found even in the presence of dominant negative dynamin to inhibit endocytosis and surface expression correlated with Golgi localization, indicating differences in forward trafficking. Co-expression of wild type GluN2B did not enhance surface expression of the mutants, suggesting that glutamate must bind to both GluN2 subunits in a tetramer and that surface expression is limited by the least avid of the two glutamate binding sites. Surface trafficking of a constitutively closed cleft GluN2B was indistinguishable from that of wild type, suggesting that glutamate concentrations are typically not limiting for forward trafficking. YFP-GluN2B expressed in hippocampal neurons from GluN2B(-/-) mice rescued synaptic accumulation at similar levels to wild type. Under these conditions, surface synaptic accumulation of YFP-GluN2B mutants also correlated with apparent glutamate affinity. Altogether, these results indicate that glutamate controls forward trafficking of NMDA receptors to the cell surface and to synapses and raise the intriguing idea that NMDA receptors may be functional at intracellular sites.


Subject(s)
Endocytosis/physiology , Glutamic Acid/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , COS Cells , Chlorocebus aethiops , Glutamic Acid/genetics , Mice , Mice, Knockout , Mutation , Neurons/cytology , Protein Binding/physiology , Protein Subunits , Protein Transport/physiology , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Synapses/genetics
16.
Mol Cell Neurosci ; 51(3-4): 68-78, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22902837

ABSTRACT

The calcium-calmodulin activated kinase CaMKII mediates many forms of learning and memory. Activity-regulated translocation of CaMKII to synapses is important for its functions in synaptic plasticity. Here, we tested the role of the NMDA receptor subunit GluN2B in recruiting CaMKIIα to synapses with different paradigms: global bath stimulation of NMDA receptors, a chemical long term potentiation (cLTP) protocol that selectively activates synaptic NMDA receptors, or local stimulation of NMDA receptors at a contiguous set of ~10-30 synapses that triggers a propagating synaptic accumulation of CaMKII. Global or cLTP-induced synaptic accumulation of CaMKIIα occurred in wild-type but not sister GluN2B -/- cultured mouse hippocampal neurons. Expression of YFP-GluN2B, but not a similar level of YFP-GluN2A, rescued global and cLTP-induced CaMKIIα translocation. Using chimeric constructs, the pore-forming extracellular and membrane domains of GluN2A combined with the cytoplasmic tail of GluN2B were sufficient to rescue CaMKIIα translocation, whereas the reverse chimera was ineffective. Furthermore, the dual point mutation R1300Q,S1303D in GluN2B that blocks interaction of this high affinity site with CaMKII abolished rescue. Thus, CaMKII binding to GluN2B is required for global and cLTP-induced synaptic accumulation of CaMKIIα. However, surprisingly, locally induced propagating synaptic accumulation of CaMKIIα occurred normally in GluN2B -/- neurons, indistinguishably from wild-type. Thus, synaptic trapping of CaMKII during locally induced propagating translocation occurs by different mechanisms and molecular partners compared with global stimulation and cLTP paradigms. These findings underscore the complex regulatory properties and molecular interactions of CaMKIIα, a key player in synaptic plasticity.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Long-Term Potentiation , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Mice , Neurons/metabolism , Neurons/physiology , Point Mutation , Protein Structure, Tertiary , Protein Transport , Rats , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synapses/physiology
17.
Cell Rep ; 42(7): 112714, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37384525

ABSTRACT

Neurexin synaptic organizing proteins are central to a genetic risk pathway in neuropsychiatric disorders. Neurexins also exemplify molecular diversity in the brain, with over a thousand alternatively spliced forms and further structural heterogeneity contributed by heparan sulfate glycan modification. Yet, interactions between these modes of post-transcriptional and post-translational modification have not been studied. We reveal that these regulatory modes converge on neurexin-1 splice site 5 (S5): the S5 insert increases the number of heparan sulfate chains. This is associated with reduced neurexin-1 protein level and reduced glutamatergic neurotransmitter release. Exclusion of neurexin-1 S5 in mice boosts neurotransmission without altering the AMPA/NMDA ratio and shifts communication and repetitive behavior away from phenotypes associated with autism spectrum disorders. Thus, neurexin-1 S5 acts as a synaptic rheostat to impact behavior through the intersection of RNA processing and glycobiology. These findings position NRXN1 S5 as a potential therapeutic target to restore function in neuropsychiatric disorders.


Subject(s)
Alternative Splicing , Autistic Disorder , Animals , Mice , Alternative Splicing/genetics , Autistic Disorder/genetics , Autistic Disorder/metabolism , Brain/metabolism , Heparitin Sulfate/metabolism , Neural Cell Adhesion Molecules/genetics , Synapses/metabolism , Synaptic Transmission
18.
J Neurosci ; 31(29): 10481-93, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21775594

ABSTRACT

Dynamics of GABAergic synaptic components have been studied previously over milliseconds to minutes, revealing mobility of postsynaptic scaffolds and receptors. Here we image inhibitory synapses containing fluorescently tagged postsynaptic scaffold Gephyrin, together with presynaptic vesicular GABA transporter (VGAT) or postsynaptic GABA(A) receptor γ2 subunit (GABA(A)Rγ2), over seconds to days in cultured rat hippocampal neurons, revealing modes of inhibitory synapse formation and remodeling. Entire synapses were mobile, translocating rapidly within a confined region and exhibiting greater nonstochastic motion over multihour periods. Presynaptic and postsynaptic components moved in unison, maintaining close apposition while translocating distances of several micrometers. An observed flux in the density of synaptic puncta partially resulted from the apparent merging and splitting of preexisting clusters. De novo formation of inhibitory synapses was observed, marked by the appearance of stably apposed Gephyrin and VGAT clusters at sites previously lacking either component. Coclustering of GABA(A)Rγ2 supports the identification of such new clusters as synapses. Nascent synapse formation occurred by gradual accumulation of components over several hours, with VGAT clustering preceding that of Gephyrin and GABA(A)Rγ2. Comparing VGAT labeling by active uptake of a luminal domain antibody with post hoc immunocytochemistry indicated that recycling vesicles from preexisting boutons significantly contribute to vesicle pools at the majority of new inhibitory synapses. Although new synapses formed primarily on dendrite shafts, some also formed on dendritic protrusions, without apparent interconversion. Altogether, the long-term imaging of GABAergic presynaptic and postsynaptic components reveals complex dynamics and perpetual remodeling with implications for mechanisms of assembly and synaptic integration.


Subject(s)
Dendrites/physiology , Neurons/cytology , Neurons/physiology , Presynaptic Terminals/physiology , Synapses/physiology , Synaptic Vesicles/physiology , Animals , Carrier Proteins/genetics , Cells, Cultured , Dendrites/ultrastructure , Embryo, Mammalian , Female , Hippocampus/cytology , Humans , Luminescent Proteins/genetics , Male , Membrane Proteins/genetics , Microscopy, Confocal/methods , Microtubule-Associated Proteins/metabolism , Mutagenesis, Site-Directed/methods , Neural Inhibition/physiology , Neurons/metabolism , Presynaptic Terminals/metabolism , Protein Transport/physiology , Rats , Receptors, GABA-A/genetics , Synapses/ultrastructure , Synaptic Vesicles/metabolism , Time Factors , Transfection/methods , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
19.
J Biol Chem ; 286(32): 28331-42, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21676872

ABSTRACT

NMDA receptors are calcium-permeable ionotropic receptors that detect coincident glutamate binding and membrane depolarization and are essential for many forms of synaptic plasticity in the mammalian brain. The obligatory GluN1 subunit of NMDA receptors is alternatively spliced at multiple sites, generating forms that vary in N-terminal N1 and C-terminal C1, C2, and C2' cassettes. Based on expression of GluN1 constructs in heterologous cells and in wild type neurons, the prevalent view is that the C-terminal cassettes regulate synaptic accumulation and its modulation by homeostatic activity blockade and by protein kinase C (PKC). Here, we tested the role of GluN1 splicing in regulated synaptic accumulation of NMDA receptors by lentiviral expression of individual GluN1 splice variants in hippocampal neurons cultured from GluN1 (-/-) mice. High efficiency transduction of GluN1 at levels similar to endogenous was achieved. Under control conditions, the C2' cassette mediated enhanced synaptic accumulation relative to the alternate C2 cassette, whereas the presence or absence of N1 or C1 had no effect. Surprisingly all GluN1 splice variants showed >2-fold increased synaptic accumulation with chronic blockade of NMDA receptor activity. Furthermore, in this neuronal rescue system, all GluN1 splice variants were equally rapidly dispersed upon activation of PKC. These results indicate that the major mechanisms mediating homeostatic synaptic accumulation and PKC dispersal of NMDA receptors occur independently of GluN1 splice isoform.


Subject(s)
Alternative Splicing/physiology , Protein Kinase C/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Synaptic Membranes/metabolism , Animals , Enzyme Activation/physiology , Mice , Mice, Knockout , Protein Kinase C/genetics , Protein Structure, Tertiary , Receptors, N-Methyl-D-Aspartate/genetics , Synaptic Membranes/genetics
20.
Elife ; 112022 06 06.
Article in English | MEDLINE | ID: mdl-35662394

ABSTRACT

LRRTMs are postsynaptic cell adhesion proteins that have region-restricted expression in the brain. To determine their role in the molecular organization of synapses in vivo, we studied synapse development and plasticity in hippocampal neuronal circuits in mice lacking both Lrrtm1 and Lrrtm2. We found that LRRTM1 and LRRTM2 regulate the density and morphological integrity of excitatory synapses on CA1 pyramidal neurons in the developing brain but are not essential for these roles in the mature circuit. Further, they are required for long-term-potentiation in the CA3-CA1 pathway and the dentate gyrus, and for enduring fear memory in both the developing and mature brain. Our data show that LRRTM1 and LRRTM2 regulate synapse development and function in a cell-type and developmental-stage-specific manner, and thereby contribute to the fine-tuning of hippocampal circuit connectivity and plasticity.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules , Animals , Hippocampus/physiology , Long-Term Potentiation/physiology , Mice , Neural Cell Adhesion Molecules/metabolism , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL