Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Neurosci ; 44(23)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38684366

ABSTRACT

Latrophilin-1 (Lphn1, aka CIRL1 and CL1; gene symbol Adgrl1) is an adhesion GPCR that has been implicated in excitatory synaptic transmission as a candidate receptor for α-latrotoxin. Here we analyzed conditional knock-in/knock-out mice for Lphn1 that contain an extracellular myc epitope tag. Mice of both sexes were used in all experiments. Surprisingly, we found that Lphn1 is localized in cultured neurons to synaptic nanoclusters that are present in both excitatory and inhibitory synapses. Conditional deletion of Lphn1 in cultured neurons failed to elicit a detectable impairment in excitatory synapses but produced a decrease in inhibitory synapse numbers and synaptic transmission that was most pronounced for synapses close to the neuronal soma. No changes in axonal or dendritic outgrowth or branching were observed. Our data indicate that Lphn1 is among the few postsynaptic adhesion molecules that are present in both excitatory and inhibitory synapses and that Lphn1 by itself is not essential for excitatory synaptic transmission but is required for some inhibitory synaptic connections.


Subject(s)
Mice, Knockout , Receptors, Peptide , Synapses , Animals , Female , Male , Mice , Cells, Cultured , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Hippocampus/cytology , Inhibitory Postsynaptic Potentials/physiology , Mice, Inbred C57BL , Neural Inhibition/physiology , Neurons/metabolism , Neurons/physiology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Synapses/metabolism , Synapses/physiology , Synaptic Transmission/physiology
2.
bioRxiv ; 2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38328199

ABSTRACT

Cadherin EGF Laminin G seven-pass G-type receptors (CELSRs or ADGRCs) are conserved adhesion G protein-coupled receptors which are essential for animal development. CELSRs have extracellular regions (ECRs) containing 23 adhesion domains which couple adhesion to intracellular signaling. However, molecular-level insight into CELSR function is sparsely available. We report the 4.3 Å cryo-EM reconstruction of the mCELSR1 ECR with 13 domains resolved in the structure. These domains form a compact module mediated by interdomain interactions with contact between the N- and C-terminal domains. We show the mCELSR1 ECR forms an extended species in the presence of Ca 2+ , which we propose represents the antiparallel cadherin repeat dimer. Using assays for adhesion and G protein-coupling, we assign the N-terminal CADH1-8 module as necessary for cell adhesion and we show the C-terminal CAHD9-GAIN module regulates signaling. Our work provides important molecular context to the literature on CELSR function and opens the door towards further mechanistic studies.

3.
Cell Rep ; 42(6): 112552, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37224017

ABSTRACT

Adhesion G protein-coupled receptors (aGPCRs) are a large GPCR class that direct diverse fundamental biological processes. One prominent mechanism for aGPCR agonism involves autoproteolytic cleavage, which generates an activating, membrane-proximal tethered agonist (TA). How universal this mechanism is for all aGPCRs is unclear. Here, we investigate G protein induction principles of aGPCRs using mammalian latrophilin 3 (LPHN3) and cadherin EGF LAG-repeat 7-transmembrane receptors 1-3 (CELSR1-3), members of two aGPCR families conserved from invertebrates to vertebrates. LPHNs and CELSRs mediate fundamental aspects of brain development, yet CELSR signaling mechanisms are unknown. We find that CELSR1 and CELSR3 are cleavage deficient, while CELSR2 is efficiently cleaved. Despite differential autoproteolysis, CELSR1-3 all engage GαS, and CELSR1 or CELSR3 TA point mutants retain GαS coupling activity. CELSR2 autoproteolysis enhances GαS coupling, yet acute TA exposure alone is insufficient. These studies support that aGPCRs signal via multiple paradigms and provide insights into CELSR biological function.


Subject(s)
Cadherins , Receptors, G-Protein-Coupled , Animals , Humans , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion , GTP-Binding Proteins/metabolism , Mammals/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Structure-Activity Relationship , HEK293 Cells
4.
bioRxiv ; 2023 Apr 03.
Article in English | MEDLINE | ID: mdl-37066404

ABSTRACT

Adhesion GPCRs (aGPCRs) are a large GPCR class that direct diverse fundamental biological processes. One prominent mechanism for aGPCR agonism involves autoproteolytic cleavage, which generates an activating, membrane-proximal tethered agonist (TA). How universal this mechanism is for all aGPCRs is unclear. Here, we investigate G protein induction principles of aGPCRs using mammalian LPHN3 and CELSR1-3, members of two aGPCR families conserved from invertebrates to vertebrates. LPHNs and CELSRs mediate fundamental aspects of brain development, yet CELSR signaling mechanisms are unknown. We found that CELSR1 and CELSR3 are cleavage-deficient, while CELSR2 is efficiently cleaved. Despite differential autoproteolysis, CELSR1-3 all engage GαS, and CELSR1 or CELSR3 TA point mutants retain GαS coupling activity. CELSR2 autoproteolysis enhances GαS coupling, yet acute TA exposure alone is insufficient. These studies support that aGPCRs signal via multiple paradigms and provide insights into CELSR biological function.

6.
J Cell Biol ; 221(2)2022 02 07.
Article in English | MEDLINE | ID: mdl-34913963

ABSTRACT

The physiological mechanisms driving synapse formation are elusive. Although numerous signals are known to regulate synapses, it remains unclear which signaling mechanisms organize initial synapse assembly. Here, we describe new tools, referred to as "SynTAMs" for synaptic targeting molecules, that enable localized perturbations of cAMP signaling in developing postsynaptic specializations. We show that locally restricted suppression of postsynaptic cAMP levels or of cAMP-dependent protein-kinase activity severely impairs excitatory synapse formation without affecting neuronal maturation, dendritic arborization, or inhibitory synapse formation. In vivo, suppression of postsynaptic cAMP signaling in CA1 neurons prevented formation of both Schaffer-collateral and entorhinal-CA1/temporoammonic-path synapses, suggesting a general principle. Retrograde trans-synaptic rabies virus tracing revealed that postsynaptic cAMP signaling is required for continuous replacement of synapses throughout life. Given that postsynaptic latrophilin adhesion-GPCRs drive synapse formation and produce cAMP, we suggest that spatially restricted postsynaptic cAMP signals organize assembly of postsynaptic specializations during synapse formation.


Subject(s)
Cyclic AMP/metabolism , Genetic Engineering , Signal Transduction , Synapses/metabolism , Animals , Hippocampus/metabolism , Mice, Inbred C57BL , Single-Domain Antibodies/metabolism , Synapses/ultrastructure
7.
Elife ; 102021 03 01.
Article in English | MEDLINE | ID: mdl-33646123

ABSTRACT

Neural circuit assembly in the brain requires precise establishment of synaptic connections, but the mechanisms of synapse assembly remain incompletely understood. Latrophilins are postsynaptic adhesion-GPCRs that engage in trans-synaptic complexes with presynaptic teneurins and FLRTs. In mouse CA1-region neurons, Latrophilin-2 and Latrophilin-3 are essential for formation of entorhinal-cortex-derived and Schaffer-collateral-derived synapses, respectively. However, it is unknown whether latrophilins function as GPCRs in synapse formation. Here, we show that Latrophilin-2 and Latrophilin-3 exhibit constitutive GPCR activity that increases cAMP levels, which was blocked by a mutation interfering with G-protein and arrestin interactions of GPCRs. The same mutation impaired the ability of Latrophilin-2 and Latrophilin-3 to rescue the synapse-loss phenotype in Latrophilin-2 and Latrophilin-3 knockout neurons in vivo. Our results suggest that Latrophilin-2 and Latrophilin-3 require GPCR signaling in synapse formation, indicating that latrophilins promote synapse formation in the hippocampus by activating a classical GPCR-signaling pathway.


Subject(s)
Receptors, G-Protein-Coupled/physiology , Receptors, Peptide/physiology , Synapses/physiology , Animals , HEK293 Cells , Hippocampus/physiology , Humans , Mice , Mice, Knockout , Mutation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Synapses/genetics , Synapses/metabolism
8.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Article in English | MEDLINE | ID: mdl-33431662

ABSTRACT

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.


Subject(s)
Homer Scaffolding Proteins/genetics , Neurons/metabolism , PTEN Phosphohydrolase/genetics , Synapses/genetics , Actin Cytoskeleton/genetics , Calcium-Binding Proteins/genetics , Cell Adhesion Molecules/genetics , Cyclic AMP-Dependent Protein Kinases/genetics , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Microtubules/genetics , Neural Cell Adhesion Molecules/genetics , Phosphatidylinositol Phosphates , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , Synapses/physiology
9.
Nat Commun ; 11(1): 2140, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358586

ABSTRACT

The trans-synaptic interaction of the cell-adhesion molecules teneurins (TENs) with latrophilins (LPHNs/ADGRLs) promotes excitatory synapse formation when LPHNs simultaneously interact with FLRTs. Insertion of a short alternatively-spliced region within TENs abolishes the TEN-LPHN interaction and switches TEN function to specify inhibitory synapses. How alternative-splicing regulates TEN-LPHN interaction remains unclear. Here, we report the 2.9 Å resolution cryo-EM structure of the TEN2-LPHN3 complex, and describe the trimeric TEN2-LPHN3-FLRT3 complex. The structure reveals that the N-terminal lectin domain of LPHN3 binds to the TEN2 barrel at a site far away from the alternatively spliced region. Alternative-splicing regulates the TEN2-LPHN3 interaction by hindering access to the LPHN-binding surface rather than altering it. Strikingly, mutagenesis of the LPHN-binding surface of TEN2 abolishes the LPHN3 interaction and impairs excitatory but not inhibitory synapse formation. These results suggest that a multi-level coincident binding mechanism mediated by a cryptic adhesion complex between TENs and LPHNs regulates synapse specificity.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/metabolism , Alternative Splicing/genetics , Alternative Splicing/physiology , Binding Sites/genetics , HEK293 Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mutation/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Binding/genetics , Protein Structure, Secondary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/chemistry , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Synapses/physiology
10.
Elife ; 82019 09 30.
Article in English | MEDLINE | ID: mdl-31566565

ABSTRACT

Nanobodies (nAbs) are small, minimal antibodies that have distinct attributes that make them uniquely suited for certain biomedical research, diagnostic and therapeutic applications. Prominent uses include as intracellular antibodies or intrabodies to bind and deliver cargo to specific proteins and/or subcellular sites within cells, and as nanoscale immunolabels for enhanced tissue penetration and improved spatial imaging resolution. Here, we report the generation and validation of nAbs against a set of proteins prominently expressed at specific subcellular sites in mammalian brain neurons. We describe a novel hierarchical validation pipeline to systematically evaluate nAbs isolated by phage display for effective and specific use as intrabodies and immunolabels in mammalian cells including brain neurons. These nAbs form part of a robust toolbox for targeting proteins with distinct and highly spatially-restricted subcellular localization in mammalian brain neurons, allowing for visualization and/or modulation of structure and function at those sites.


Subject(s)
Brain/cytology , Neurons/metabolism , Protein Transport , Single-Domain Antibodies/metabolism , Staining and Labeling/methods , Animals , Cells, Cultured , Protein Binding , Rats , Single-Domain Antibodies/isolation & purification
11.
Science ; 363(6429)2019 02 22.
Article in English | MEDLINE | ID: mdl-30792275

ABSTRACT

Bidirectional signaling by cell adhesion molecules is thought to mediate synapse formation, but the mechanisms involved remain elusive. We found that the adhesion G protein-coupled receptors latrophilin-2 and latrophilin-3 selectively direct formation of perforant-path and Schaffer-collateral synapses, respectively, to hippocampal CA1-region neurons. Latrophilin-3 binds to two transcellular ligands: fibronectin leucine-rich repeat transmembrane proteins (FLRTs) and teneurins. In transgenic mice in vivo, both binding activities were required for input-specific synapse formation, which suggests that coincident binding of both ligands is necessary for synapse formation. In cultured neurons in vitro, teneurin or FLRT alone did not induce excitatory synapse formation, whereas together they potently did so. Thus, postsynaptic latrophilins promote excitatory synapse formation by simultaneous binding of two unrelated presynaptic ligands, which is required for formation of synaptic inputs at specific dendritic localizations.


Subject(s)
CA1 Region, Hippocampal/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Peptide/metabolism , Synapses/physiology , Animals , CA1 Region, Hippocampal/cytology , HEK293 Cells , Humans , Membrane Glycoproteins , Mice , Mice, Knockout , Protein Binding , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Synapses/genetics
13.
Cell ; 173(3): 735-748.e15, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29677516

ABSTRACT

Teneurins (TENs) are cell-surface adhesion proteins with critical roles in tissue development and axon guidance. Here, we report the 3.1-Å cryoelectron microscopy structure of the human TEN2 extracellular region (ECR), revealing a striking similarity to bacterial Tc-toxins. The ECR includes a large ß barrel that partially encapsulates a C-terminal domain, which emerges to the solvent through an opening in the mid-barrel region. An immunoglobulin (Ig)-like domain seals the bottom of the barrel while a ß propeller is attached in a perpendicular orientation. We further show that an alternatively spliced region within the ß propeller acts as a switch to regulate trans-cellular adhesion of TEN2 to latrophilin (LPHN), a transmembrane receptor known to mediate critical functions in the central nervous system. One splice variant activates trans-cellular signaling in a LPHN-dependent manner, whereas the other induces inhibitory postsynaptic differentiation. These results highlight the unusual structural organization of TENs giving rise to their multifarious functions.


Subject(s)
Bacterial Toxins/chemistry , Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Synapses/metabolism , Alternative Splicing , Amino Acid Motifs , Animals , Axons , Cell Adhesion , Cell Line , Cyclic AMP/metabolism , Female , HEK293 Cells , Hormones/chemistry , Humans , Insecta , Membrane Proteins/metabolism , Mice , Molecular Conformation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neuropeptides/chemistry , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/chemistry , Signal Transduction
14.
J Cell Biol ; 216(11): 3831-3846, 2017 11 06.
Article in English | MEDLINE | ID: mdl-28972101

ABSTRACT

Synapse assembly likely requires postsynaptic target recognition by incoming presynaptic afferents. Using newly generated conditional knock-in and knockout mice, we show in this study that latrophilin-2 (Lphn2), a cell-adhesion G protein-coupled receptor and presumptive α-latrotoxin receptor, controls the numbers of a specific subset of synapses in CA1-region hippocampal neurons, suggesting that Lphn2 acts as a synaptic target-recognition molecule. In cultured hippocampal neurons, Lphn2 maintained synapse numbers via a postsynaptic instead of a presynaptic mechanism, which was surprising given its presumptive role as an α-latrotoxin receptor. In CA1-region neurons in vivo, Lphn2 was specifically targeted to dendritic spines in the stratum lacunosum-moleculare, which form synapses with presynaptic entorhinal cortex afferents. In this study, postsynaptic deletion of Lphn2 selectively decreased spine numbers and impaired synaptic inputs from entorhinal but not Schaffer-collateral afferents. Behaviorally, loss of Lphn2 from the CA1 region increased spatial memory retention but decreased learning of sequential spatial memory tasks. Thus, Lphn2 appears to control synapse numbers in the entorhinal cortex/CA1 region circuit by acting as a domain-specific postsynaptic target-recognition molecule.


Subject(s)
CA1 Region, Hippocampal/metabolism , Entorhinal Cortex/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Synaptic Membranes/metabolism , Animals , Behavior, Animal , CA1 Region, Hippocampal/pathology , CA1 Region, Hippocampal/physiopathology , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/pathology , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Fear , Genotype , Maze Learning , Memory , Mice, Mutant Strains , Motor Activity , Neurons/pathology , Phenotype , Presynaptic Terminals/pathology , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Rotarod Performance Test , Smell , Synaptic Membranes/pathology , Synaptic Potentials , Time Factors , Transfection
15.
Neuron ; 94(2): 312-321.e3, 2017 Apr 19.
Article in English | MEDLINE | ID: mdl-28426966

ABSTRACT

Synaptic excitation mediates a broad spectrum of structural changes in neural circuits across the brain. Here, we examine the morphologies, wiring, and architectures of single synapses of projection neurons in the murine hippocampus that developed in virtually complete absence of vesicular glutamate release. While these neurons had smaller dendritic trees and/or formed fewer contacts in specific hippocampal subfields, their stereotyped connectivity was largely preserved. Furthermore, loss of release did not disrupt the morphogenesis of presynaptic terminals and dendritic spines, suggesting that glutamatergic neurotransmission is unnecessary for synapse assembly and maintenance. These results underscore the instructive role of intrinsic mechanisms in synapse formation.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , Animals , Dendrites/physiology , Dendritic Spines/physiology , Glutamic Acid/metabolism , Mice , Neurogenesis/physiology , Synapses/physiology , Synaptic Transmission/physiology
16.
PLoS Biol ; 14(7): e1002516, 2016 07.
Article in English | MEDLINE | ID: mdl-27429220

ABSTRACT

Individual synapses vary significantly in their neurotransmitter release properties, which underlie complex information processing in neural circuits. Presynaptic Ca2+ homeostasis plays a critical role in specifying neurotransmitter release properties, but the mechanisms regulating synapse-specific Ca2+ homeostasis in the mammalian brain are still poorly understood. Using electrophysiology and genetically encoded Ca2+ sensors targeted to the mitochondrial matrix or to presynaptic boutons of cortical pyramidal neurons, we demonstrate that the presence or absence of mitochondria at presynaptic boutons dictates neurotransmitter release properties through Mitochondrial Calcium Uniporter (MCU)-dependent Ca2+ clearance. We demonstrate that the serine/threonine kinase LKB1 regulates MCU expression, mitochondria-dependent Ca2+ clearance, and thereby, presynaptic release properties. Re-establishment of MCU-dependent mitochondrial Ca2+ uptake at glutamatergic synapses rescues the altered neurotransmitter release properties characterizing LKB1-null cortical axons. Our results provide novel insights into the cellular and molecular mechanisms whereby mitochondria control neurotransmitter release properties in a bouton-specific way through presynaptic Ca2+ clearance.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyramidal Cells/metabolism , Synapses/metabolism , AMP-Activated Protein Kinases , Action Potentials/physiology , Animals , Axons/metabolism , Axons/physiology , Blotting, Western , COS Cells , Calcium Channels/genetics , Calcium Channels/metabolism , Cells, Cultured , Chlorocebus aethiops , Coculture Techniques , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Patch-Clamp Techniques , Presynaptic Terminals/physiology , Protein Serine-Threonine Kinases/genetics , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Time-Lapse Imaging/methods
17.
Structure ; 23(9): 1678-1691, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26235030

ABSTRACT

Fibronectin leucine-rich repeat transmembrane proteins (FLRTs) are cell-adhesion molecules with emerging functions in cortical development and synapse formation. Their extracellular regions interact with latrophilins (LPHNs) to mediate synapse development, and with Uncoordinated-5 (UNC5)/netrin receptors to control the migration of neurons in the developing cortex. Here, we present the crystal structures of FLRT3 in isolation and in complex with LPHN3. The LPHN3/FLRT3 structure reveals that LPHN3 binds to FLRT3 at a site distinct from UNC5. Structure-based mutations specifically disrupt LPHN3/FLRT3 binding, but do not disturb their interactions with other proteins or their cell-membrane localization. Thus, they can be used as molecular tools to dissect the functions of FLRTs and LPHNs in vivo. Our results suggest that UNC5 and LPHN3 can simultaneously bind to FLRT3, forming a trimeric complex, and that FLRT3 may form transsynaptic complexes with both LPHN3 and UNC5. These findings provide molecular insights for understanding the role of cell-adhesion proteins in synapse function.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/chemistry , Receptors, Peptide/metabolism , Binding Sites , Cell Adhesion , Crystallography, X-Ray , HEK293 Cells , Humans , Membrane Glycoproteins , Membrane Proteins/genetics , Models, Molecular , Mutation , Netrin Receptors , Protein Multimerization , Receptors, Cell Surface/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Synapses/metabolism
18.
Cell Rep ; 11(7): 1054-66, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25959820

ABSTRACT

At presynaptic active zones, exocytosis of neurotransmitter vesicles (SVs) is driven by SNARE complexes that recruit Syb2 and SNAP25. However, it remains unknown which SNAREs promote the secretion of neuronal proteins, including those essential for circuit development and experience-dependent plasticity. Here we demonstrate that Syb2 and SNAP25 mediate the vesicular release of BDNF in axons and dendrites of cortical neurons, suggesting these SNAREs act in multiple spatially segregated secretory pathways. Remarkably, axonal secretion of BDNF is also strongly regulated by SNAP47, which interacts with SNAP25 but appears to be dispensable for exocytosis of SVs. Cell-autonomous ablation of SNAP47 disrupts the layer-specific branching of callosal axons of projection cortical neurons in vivo, and this phenotype is recapitulated by ablation of BDNF or its receptor, TrkB. Our results provide insights into the molecular mechanisms of protein secretion, and they define the functions of SNAREs in BDNF signaling and regulation of neuronal connectivity.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Exocytosis/physiology , Neurons/metabolism , Synaptosomal-Associated Protein 25/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Animals , Cells, Cultured , Immunohistochemistry , Immunoprecipitation , Mice , Patch-Clamp Techniques , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Transfection
19.
Neuron ; 84(1): 107-122, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25277456

ABSTRACT

The structural organization of neural circuits is strongly influenced by experience, but the underlying mechanisms are incompletely understood. We found that, in the developing dentate gyrus (DG), excitatory drive promotes the somatic innervation of principal granule cells (GCs) by parvalbumin (PV)-positive basket cells. In contrast, presynaptic differentiation of GCs and interneuron subtypes that inhibit GC dendrites is largely resistant to loss of glutamatergic neurotransmission. The networks of PV basket cells in the DG are regulated by vesicular release from projection entorhinal cortical neurons and, at least in part, by NMDA receptors in interneurons. Finally, we present evidence that glutamatergic inputs and NMDA receptors regulate these networks through a presynaptic mechanism that appears to control the branching of interneuron axons. Our results provide insights into how cortical activity tunes the inhibition in a subcortical circuit and reveal new principles of interneuron plasticity.


Subject(s)
Dentate Gyrus/physiology , Dentate Gyrus/ultrastructure , Nerve Net/physiology , Nerve Net/ultrastructure , Neuronal Plasticity/physiology , Animals , Cell Differentiation/physiology , Interneurons/physiology , Interneurons/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Culture Techniques
20.
Nat Methods ; 10(11): 1085-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056874

ABSTRACT

Acute manipulation of gene and protein function in the brain is essential for understanding the mechanisms of nervous system development, plasticity and information processing. Here we describe a technique based on a destabilized Cre recombinase (DD-Cre) whose activity is controlled by the antibiotic trimethoprim (TMP). We show that DD-Cre triggers rapid TMP-dependent recombination of loxP-flanked ('floxed') alleles in mouse neurons in vivo and validate the use of this system for neurobehavioral research.


Subject(s)
Gene Expression Regulation , Integrases/metabolism , Animals , Gene Expression Regulation/drug effects , Humans , Mice , Recombination, Genetic/drug effects , Trimethoprim/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...