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

2.
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'.

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.
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
5.
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
6.
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
7.
Neuron ; 109(4): 566-568, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33600752

ABSTRACT

The relationship between synaptogenesis and dendritogenesis is poorly understood, particularly in mammals. In this issue of Neuron, Takeo et al. (2021) manipulate synaptic organizers GluD2 and cerebellin-1 to show that Purkinje cells regulate how their dendrites branch by competing with neighboring cells for synaptic real estate.


Subject(s)
Neurons , Purkinje Cells , Animals , Dendrites , Neurogenesis
9.
Elife ; 92020 09 11.
Article in English | MEDLINE | ID: mdl-32915137

ABSTRACT

Post-transcriptional mechanisms regulating cell surface synaptic organizing complexes that control the properties of connections in brain circuits are poorly understood. Alternative splicing regulates the prototypical synaptic organizing complex, neuroligin-neurexin. In contrast to the well-studied neuroligin splice site B, little is known about splice site A. We discovered that inclusion of the positively charged A1 insert in mouse neuroligin-1 increases its binding to heparan sulphate, a modification on neurexin. The A1 insert increases neurexin recruitment, presynaptic differentiation, and synaptic transmission mediated by neuroligin-1. We propose that the A1 insert could be a target for alleviating the consequences of deleterious NLGN1/3 mutations, supported by assays with the autism-linked neuroligin-1-P89L mutant. An enrichment of neuroligin-1 A1 in GABAergic neuron types suggests a role in synchrony of cortical circuits. Altogether, these data reveal an unusual mode by which neuroligin splicing controls synapse development through protein-glycan interaction and identify it as a potential therapeutic target.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules, Neuronal/metabolism , Polysaccharides/metabolism , Synapses/physiology , Animals , Female , Male , Mice , Mice, Knockout , Rats
10.
Neuron ; 105(6): 1007-1017.e5, 2020 03 18.
Article in English | MEDLINE | ID: mdl-31974009

ABSTRACT

LRRTM4 is a transsynaptic adhesion protein regulating glutamatergic synapse assembly on dendrites of central neurons. In the mouse retina, we find that LRRTM4 is enriched at GABAergic synapses on axon terminals of rod bipolar cells (RBCs). Knockout of LRRTM4 reduces RBC axonal GABAA and GABAC receptor clustering and disrupts presynaptic inhibition onto RBC terminals. LRRTM4 removal also perturbs the stereotyped output synapse arrangement at RBC terminals. Synaptic ribbons are normally apposed to two distinct postsynaptic "dyad" partners, but in the absence of LRRTM4, "monad" and "triad" arrangements are also formed. RBCs from retinas deficient in GABA release also demonstrate dyad mis-arrangements but maintain LRRTM4 expression, suggesting that defects in dyad organization in the LRRTM4 knockout could originate from reduced GABA receptor function. LRRTM4 is thus a key synapse organizing molecule at RBC terminals, where it regulates function of GABAergic synapses and assembly of RBC synaptic dyads.


Subject(s)
Membrane Proteins/physiology , Nerve Tissue Proteins/physiology , Neural Inhibition/physiology , Presynaptic Terminals/physiology , Retinal Bipolar Cells/physiology , Animals , Female , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Receptors, GABA/metabolism , Receptors, GABA/physiology , Retina/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , gamma-Aminobutyric Acid/metabolism
11.
Article in English | MEDLINE | ID: mdl-31191292

ABSTRACT

Formation of synapses between neurons depends in part on binding between axonal and dendritic cell surface synaptic organizing proteins, which recruit components of the developing presynaptic and postsynaptic specializations. One of these presynaptic organizing molecules is protein tyrosine phosphatase σ (PTPσ). Although the protein domains involved in adhesion between PTPσ and its postsynaptic binding partners are known, the mechanisms by which it signals into the presynaptic neuron to recruit synaptic vesicles and other necessary components for regulated transmitter release are not well understood. One attractive candidate to mediate this function is liprin-α, a scaffolding protein with well-established roles at the synapse. We systematically mutated residues of the PTPσ intracellular region (ICR) and used the yeast dihydrofolate reductase (DHFR) protein complementation assay to screen for disrupted interactions between these mutant forms of PTPσ and its various binding partners. Using a molecular replacement strategy, we show that disrupting the interaction between PTPσ and liprin-α, but not between PTPσ and itself or another binding partner, caskin, abolishes presynaptic differentiation. Furthermore, phosphatase activity of PTPσ and binding to extracellular heparan sulfate (HS) proteoglycans are dispensable for presynaptic induction. Previous reports have suggested that binding between PTPσ and liprin-α is mediated by the PTPσ membrane-distal phosphatase-like domain. However, we provide evidence here that both of the PTPσ phosphatase-like domains mediate binding to liprin-α and are required for PTPσ-mediated presynaptic differentiation. These findings further our understanding of the mechanistic basis by which PTPσ acts as a presynaptic organizer.

12.
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
13.
Curr Opin Neurobiol ; 57: 71-80, 2019 08.
Article in English | MEDLINE | ID: mdl-30771697

ABSTRACT

Synapse development depends on a dynamic balance between synapse promoters and suppressors. MDGAs, immunoglobulin superfamily proteins, negatively regulate synapse development through blocking neuroligin-neurexin interactions. Recent analyses of MDGA-neuroligin complexes revealed the structural basis of this activity and indicate that MDGAs interact with all neuroligins with differential affinities. Surprisingly, analyses of mouse mutants revealed a functional divergence, with targeted mutation of Mdga1 and Mdga2 elevating inhibitory and excitatory synapses, respectively, on hippocampal pyramidal neurons. Further research is needed to determine the synapse-specific organizing properties of MDGAs in neural circuits, which may depend on relative levels and subcellular distributions of each MDGA, neuroligin and neurexin. Behavioral deficits in Mdga mutant mice support genetic links to schizophrenia and autism spectrum disorders and raise the possibility of harnessing these interactions for therapeutic purposes.


Subject(s)
Synapses , Animals , Cell Adhesion Molecules, Neuronal , Dansyl Compounds , Galactosamine/analogs & derivatives , Mice , Nerve Tissue Proteins
14.
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
15.
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
16.
Neuron ; 97(3): 596-610.e8, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29395912

ABSTRACT

In contrast with numerous studies of glutamate receptor-associated proteins and their involvement in the modulation of excitatory synapses, much less is known about mechanisms controlling postsynaptic GABAA receptor (GABAAR) numbers. Using tandem affinity purification from tagged GABAAR γ2 subunit transgenic mice and proteomic analysis, we isolated several GABAAR-associated proteins, including Cleft lip and palate transmembrane protein 1 (Clptm1). Clptm1 interacted with all GABAAR subunits tested and promoted GABAAR trapping in the endoplasmic reticulum. Overexpression of Clptm1 reduced GABAAR-mediated currents in a recombinant system, in cultured hippocampal neurons, and in brain, with no effect on glycine or AMPA receptor-mediated currents. Conversely, knockdown of Clptm1 increased phasic and tonic inhibitory transmission with no effect on excitatory synaptic transmission. Furthermore, altering the expression level of Clptm1 mimicked activity-induced inhibitory synaptic scaling. Thus, in complement to other GABAAR-associated proteins that promote receptor surface expression, Clptm1 limits GABAAR forward trafficking and regulates inhibitory homeostatic plasticity.


Subject(s)
Inhibitory Postsynaptic Potentials , Membrane Proteins/metabolism , Neurons/metabolism , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , COS Cells , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Female , HEK293 Cells , Hippocampus/metabolism , Homeostasis , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Protein Subunits/metabolism , Protein Transport , Proteomics , Rats
17.
Cell Rep ; 21(13): 3637-3645, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29281813

ABSTRACT

Synaptopathies contributing to neurodevelopmental disorders are linked to mutations in synaptic organizing molecules, including postsynaptic neuroligins, presynaptic neurexins, and MDGAs, which regulate their interaction. The role of MDGA1 in suppressing inhibitory versus excitatory synapses is controversial based on in vitro studies. We show that genetic deletion of MDGA1 in vivo elevates hippocampal CA1 inhibitory, but not excitatory, synapse density and transmission. Furthermore, MDGA1 is selectively expressed by pyramidal neurons and regulates perisomatic, but not distal dendritic, inhibitory synapses. Mdga1-/- hippocampal networks demonstrate muted responses to neural excitation, and Mdga1-/- mice are resistant to induced seizures. Mdga1-/- mice further demonstrate compromised hippocampal long-term potentiation, consistent with observed deficits in spatial and context-dependent learning and memory. These results suggest that mutations in MDGA1 may contribute to cognitive deficits through altered synaptic transmission and plasticity by loss of suppression of inhibitory synapse development in a subcellular domain- and cell-type-selective manner.


Subject(s)
Cognition , Nerve Net/metabolism , Neural Cell Adhesion Molecules/metabolism , Neural Inhibition , Synapses/metabolism , Animals , CA1 Region, Hippocampal/pathology , Gene Deletion , Long-Term Potentiation , Mice, Inbred C57BL , Mice, Knockout , Neural Cell Adhesion Molecules/deficiency , Synapses/ultrastructure , Synaptic Transmission
19.
Neuron ; 95(4): 896-913.e10, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28817804

ABSTRACT

Neuroligin-neurexin (NL-NRX) complexes are fundamental synaptic organizers in the central nervous system. An accurate spatial and temporal control of NL-NRX signaling is crucial to balance excitatory and inhibitory neurotransmission, and perturbations are linked with neurodevelopmental and psychiatric disorders. MDGA proteins bind NLs and control their function and interaction with NRXs via unknown mechanisms. Here, we report crystal structures of MDGA1, the NL1-MDGA1 complex, and a spliced NL1 isoform. Two large, multi-domain MDGA molecules fold into rigid triangular structures, cradling a dimeric NL to prevent NRX binding. Structural analyses guided the discovery of a broad, splicing-modulated interaction network between MDGA and NL family members and helped rationalize the impact of autism-linked mutations. We demonstrate that expression levels largely determine whether MDGAs act selectively or suppress the synapse organizing function of multiple NLs. These results illustrate a potentially brain-wide regulatory mechanism for NL-NRX signaling modulation.


Subject(s)
Dansyl Compounds/metabolism , Galactosamine/analogs & derivatives , Neurturin/metabolism , Signal Transduction/physiology , Synapses/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , COS Cells , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Chickens , Coculture Techniques , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Galactosamine/genetics , Galactosamine/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Models, Molecular , Mutagenesis, Site-Directed , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurturin/genetics , Protein Interaction Maps , Receptors, N-Methyl-D-Aspartate/metabolism , Sequence Alignment
20.
Neuron ; 91(5): 1052-1068, 2016 Sep 07.
Article in English | MEDLINE | ID: mdl-27608760

ABSTRACT

Mutations in a synaptic organizing pathway contribute to autism. Autism-associated mutations in MDGA2 (MAM domain containing glycosylphosphatidylinositol anchor 2) are thought to reduce excitatory/inhibitory transmission. However, we show that mutation of Mdga2 elevates excitatory transmission, and that MDGA2 blocks neuroligin-1 interaction with neurexins and suppresses excitatory synapse development. Mdga2(+/-) mice, modeling autism mutations, demonstrated increased asymmetric synapse density, mEPSC frequency and amplitude, and altered LTP, with no change in measures of inhibitory synapses. Behavioral assays revealed an autism-like phenotype including stereotypy, aberrant social interactions, and impaired memory. In vivo voltage-sensitive dye imaging, facilitating comparison with fMRI studies in autism, revealed widespread increases in cortical spontaneous activity and intracortical functional connectivity. These results suggest that mutations in MDGA2 contribute to altered cortical processing through the dual disadvantages of elevated excitation and hyperconnectivity, and indicate that perturbations of the NRXN-NLGN pathway in either direction from the norm increase risk for autism.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Cerebral Cortex/physiology , Cognition/physiology , GPI-Linked Proteins/physiology , Haploinsufficiency/physiology , Neural Cell Adhesion Molecules/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Disks Large Homolog 4 Protein , Excitatory Postsynaptic Potentials/physiology , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Guanylate Kinases/metabolism , Hippocampus/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/physiology , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , Receptors, AMPA/metabolism , Receptors, AMPA/physiology , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL