Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nature ; 595(7866): 261-265, 2021 07.
Article in English | MEDLINE | ID: mdl-34135511

ABSTRACT

Ionotropic glutamate delta receptors 1 (GluD1) and 2 (GluD2) exhibit the molecular architecture of postsynaptic ionotropic glutamate receptors, but assemble into trans-synaptic adhesion complexes by binding to secreted cerebellins that in turn interact with presynaptic neurexins1-4. It is unclear whether neurexin-cerebellin-GluD1/2 assemblies serve an adhesive synapse-formation function or mediate trans-synaptic signalling. Here we show in hippocampal synapses, that binding of presynaptic neurexin-cerebellin complexes to postsynaptic GluD1 controls glutamate receptor activity without affecting synapse numbers. Specifically, neurexin-1-cerebellin-2 and neurexin-3-cerebellin-2 complexes differentially regulate NMDA (N-methyl-D-aspartate) receptors and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid) receptors by activating distinct postsynaptic GluD1 effector signals. Of note, minimal GluD1 and GluD2 constructs containing only their N-terminal cerebellin-binding and C-terminal cytoplasmic domains, joined by an unrelated transmembrane region, fully control the levels of NMDA and AMPA receptors. The distinct signalling specificity of presynaptic neurexin-1 and neurexin-35,6 is encoded by their alternatively spliced splice site 4 sequences, whereas the regulatory functions of postsynaptic GluD1 are mediated by conserved cytoplasmic sequence motifs spanning 5-13 residues. Thus, GluDs are signalling molecules that regulate NMDA and AMPA receptors by an unexpected transduction mechanism that bypasses their ionotropic receptor architecture and directly converts extracellular neurexin-cerebellin signals into postsynaptic receptor responses.


Subject(s)
Glutamate Dehydrogenase/metabolism , Receptors, Ionotropic Glutamate/metabolism , Signal Transduction , Amino Acid Motifs , Animals , Calcium-Binding Proteins/metabolism , Cell Membrane/metabolism , Excitatory Postsynaptic Potentials , Female , Male , Mice , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Protein Precursors/metabolism , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
2.
Mol Psychiatry ; 26(11): 6253-6268, 2021 11.
Article in English | MEDLINE | ID: mdl-33931733

ABSTRACT

Cannabis and cannabinoid drugs are central agents that are used widely recreationally and are employed broadly for treating psychiatric conditions. Cannabinoids primarily act by stimulating presynaptic CB1 receptors (CB1Rs), the most abundant G-protein-coupled receptors in brain. CB1R activation decreases neurotransmitter release by inhibiting presynaptic Ca2+ channels and induces long-term plasticity by decreasing cellular cAMP levels. Here we identified an unanticipated additional mechanism of acute cannabinoid signaling in presynaptic terminals that regulates the size of synaptic vesicle pools available for neurotransmitter release. Specifically, we show that activation of CB1Rs in human and mouse neurons rapidly recruits vesicles to nerve terminals by suppressing the cAMP-dependent phosphorylation of synapsins. We confirmed this unanticipated mechanism using conditional deletion of synapsin-1, the predominant synapsin isoform in human neurons, demonstrating that synapsin-1 significantly contributes to the CB1R-dependent regulation of neurotransmission. Interestingly, acute activation of the Gi-DREADD hM4D mimics the effect of CB1R activation in a synapsin-1-dependent manner, suggesting that the control of synaptic vesicle numbers by synapsin-1 phosphorylation is a general presynaptic mechanism of neuromodulation. Thus, we uncovered a CB1R-dependent presynaptic mechanism that rapidly regulates the organization and neurotransmitter release properties of synapses.


Subject(s)
Cannabinoids , Synapsins , Animals , Cannabinoids/pharmacology , Humans , Mice , Receptors, Cannabinoid , Synapses/physiology , Synaptic Transmission/physiology , Synaptic Vesicles
3.
J Clin Invest ; 131(7)2021 04 01.
Article in English | MEDLINE | ID: mdl-33539324

ABSTRACT

Dystonia is a debilitating hyperkinetic movement disorder, which can be transmitted as a monogenic trait. Here, we describe homozygous frameshift, nonsense, and missense variants in TSPOAP1, which encodes the active-zone RIM-binding protein 1 (RIMBP1), as a genetic cause of autosomal recessive dystonia in 7 subjects from 3 unrelated families. Subjects carrying loss-of-function variants presented with juvenile-onset progressive generalized dystonia, associated with intellectual disability and cerebellar atrophy. Conversely, subjects carrying a pathogenic missense variant (p.Gly1808Ser) presented with isolated adult-onset focal dystonia. In mice, complete loss of RIMBP1, known to reduce neurotransmission, led to motor abnormalities reminiscent of dystonia, decreased Purkinje cell dendritic arborization, and reduced numbers of cerebellar synapses. In vitro analysis of the p.Gly1808Ser variant showed larger spike-evoked calcium transients and enhanced neurotransmission, suggesting that RIMBP1-linked dystonia can be caused by either reduced or enhanced rates of spike-evoked release in relevant neural networks. Our findings establish a direct link between dysfunction of the presynaptic active zone and dystonia and highlight the critical role played by well-balanced neurotransmission in motor control and disease pathogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Alleles , Calcium Signaling , Dendrites/metabolism , Dystonic Disorders , Mutation, Missense , Purkinje Cells/metabolism , Synaptic Transmission , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Substitution , Animals , Dendrites/genetics , Dystonic Disorders/genetics , Dystonic Disorders/metabolism , Female , Humans , Male , Mice , Mice, Knockout
4.
Cell ; 179(2): 498-513.e22, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31585084

ABSTRACT

Neuromodulators bind to pre- and postsynaptic G protein-coupled receptors (GPCRs), are able to quickly change intracellular cyclic AMP (cAMP) and Ca2+ levels, and are thought to play important roles in neuropsychiatric and neurodegenerative diseases. Here, we discovered in human neurons an unanticipated presynaptic mechanism that acutely changes synaptic ultrastructure and regulates synaptic communication. Activation of neuromodulator receptors bidirectionally controlled synaptic vesicle numbers within nerve terminals. This control correlated with changes in the levels of cAMP-dependent protein kinase A-mediated phosphorylation of synapsin-1. Using a conditional deletion approach, we reveal that the neuromodulator-induced control of synaptic vesicle numbers was largely dependent on synapsin-1. We propose a mechanism whereby non-phosphorylated synapsin-1 "latches" synaptic vesicles to presynaptic clusters at the active zone. cAMP-dependent phosphorylation of synapsin-1 then removes the vesicles. cAMP-independent dephosphorylation of synapsin-1 in turn recruits vesicles. Synapsin-1 thereby bidirectionally regulates synaptic vesicle numbers and modifies presynaptic neurotransmitter release as an effector of neuromodulator signaling in human neurons.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Presynaptic Terminals/metabolism , Synapsins/metabolism , Synaptic Transmission , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neurotransmitter Agents/metabolism , Receptors, Neurotransmitter/metabolism , Signal Transduction
5.
Sci Transl Med ; 10(452)2018 08 01.
Article in English | MEDLINE | ID: mdl-30068571

ABSTRACT

Fragile X syndrome (FXS) is an X chromosome-linked disease leading to severe intellectual disabilities. FXS is caused by inactivation of the fragile X mental retardation 1 (FMR1) gene, but how FMR1 inactivation induces FXS remains unclear. Using human neurons generated from control and FXS patient-derived induced pluripotent stem (iPS) cells or from embryonic stem cells carrying conditional FMR1 mutations, we show here that loss of FMR1 function specifically abolished homeostatic synaptic plasticity without affecting basal synaptic transmission. We demonstrated that, in human neurons, homeostatic plasticity induced by synaptic silencing was mediated by retinoic acid, which regulated both excitatory and inhibitory synaptic strength. FMR1 inactivation impaired homeostatic plasticity by blocking retinoic acid-mediated regulation of synaptic strength. Repairing the genetic mutation in the FMR1 gene in an FXS patient cell line restored fragile X mental retardation protein (FMRP) expression and fully rescued synaptic retinoic acid signaling. Thus, our study reveals a robust functional impairment caused by FMR1 mutations that might contribute to neuronal dysfunction in FXS. In addition, our results suggest that FXS patient iPS cell-derived neurons might be useful for studying the mechanisms mediating functional abnormalities in FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Homeostasis , Mutation/genetics , Neuronal Plasticity , Neurons/metabolism , Signal Transduction , Synapses/metabolism , Tretinoin/metabolism , Alleles , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Excitatory Postsynaptic Potentials/drug effects , Fragile X Syndrome/genetics , Fragile X Syndrome/physiopathology , Homeostasis/drug effects , Humans , Mice , Neuronal Plasticity/drug effects , Neurons/drug effects , Signal Transduction/drug effects , Synapses/drug effects , Tretinoin/pharmacology , Trinucleotide Repeats/genetics , Up-Regulation/drug effects
6.
Rare Dis ; 4(1): e1131884, 2016.
Article in English | MEDLINE | ID: mdl-27141410

ABSTRACT

The use of human pluripotent stem cells to model human diseases has become a new standard in biomedical sciences. To this end, patient-derived somatic cells are studied in vitro to mimic human pathological conditions. Here, we describe an alternative experimental strategy, the 'conditional KO approach', which allows engineering disease-relevant mutations in pluripotent stem cells from healthy donors. In combination with the Cre/Lox technology, this strategy enables us to study the molecular causes of human diseases independent of the genetic background or of genetic alterations induced by clonal selection. As a proof-of-principle, we generated pluripotent stem cells with conditional loss-of-function mutations in the human STXBP1 gene that encodes Munc18-1. Using neurons derived from these cells, we show that heterozygous disruption of STXBP1 produces a specific and selective impairment in synaptic transmission that may account for the severe neurological disease caused by such mutations in human patients.

7.
Science ; 352(6286): aaf2669, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26966193

ABSTRACT

Heterozygous SHANK3 mutations are associated with idiopathic autism and Phelan-McDermid syndrome. SHANK3 is a ubiquitously expressed scaffolding protein that is enriched in postsynaptic excitatory synapses. Here, we used engineered conditional mutations in human neurons and found that heterozygous and homozygous SHANK3 mutations severely and specifically impaired hyperpolarization-activated cation (Ih) channels. SHANK3 mutations caused alterations in neuronal morphology and synaptic connectivity; chronic pharmacological blockage of Ih channels reproduced these phenotypes, suggesting that they may be secondary to Ih-channel impairment. Moreover, mouse Shank3-deficient neurons also exhibited severe decreases in Ih currents. SHANK3 protein interacted with hyperpolarization-activated cyclic nucleotide-gated channel proteins (HCN proteins) that form Ih channels, indicating that SHANK3 functions to organize HCN channels. Our data suggest that SHANK3 mutations predispose to autism, at least partially, by inducing an Ih channelopathy that may be amenable to pharmacological intervention.


Subject(s)
Autism Spectrum Disorder/genetics , Channelopathies/genetics , Genetic Predisposition to Disease , Haploinsufficiency/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Action Potentials , Animals , Cells, Cultured , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 22/genetics , Embryonic Stem Cells/metabolism , Gene Deletion , Genetic Engineering , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Mice , Mice, Knockout , Microfilament Proteins , Mutagenesis , Nerve Tissue Proteins/metabolism , Synapses/physiology , Synaptic Transmission
8.
J Exp Med ; 213(4): 499-515, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27001749

ABSTRACT

Hundreds of L1CAM gene mutations have been shown to be associated with congenital hydrocephalus, severe intellectual disability, aphasia, and motor symptoms. How such mutations impair neuronal function, however, remains unclear. Here, we generated human embryonic stem (ES) cells carrying a conditional L1CAM loss-of-function mutation and produced precisely matching control and L1CAM-deficient neurons from these ES cells. In analyzing two independent conditionally mutant ES cell clones, we found that deletion of L1CAM dramatically impaired axonal elongation and, to a lesser extent, dendritic arborization. Unexpectedly, we also detected an ∼20-50% and ∼20-30% decrease, respectively, in the levels of ankyrinG and ankyrinB protein, and observed that the size and intensity of ankyrinG staining in the axon initial segment was significantly reduced. Overexpression of wild-type L1CAM, but not of the L1CAM point mutants R1166X and S1224L, rescued the decrease in ankyrin levels. Importantly, we found that the L1CAM mutation selectively decreased activity-dependent Na(+)-currents, altered neuronal excitability, and caused impairments in action potential (AP) generation. Thus, our results suggest that the clinical presentations of L1CAM mutations in human patients could be accounted for, at least in part, by cell-autonomous changes in the functional development of neurons, such that neurons are unable to develop normal axons and dendrites and to generate normal APs.


Subject(s)
Action Potentials , Axons/metabolism , Dendrites/metabolism , Gene Deletion , Hydrocephalus , Neural Cell Adhesion Molecule L1/genetics , Axons/pathology , Dendrites/pathology , Human Embryonic Stem Cells , Humans , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/pathology , Hydrocephalus/physiopathology
9.
J Clin Invest ; 125(9): 3560-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26280581

ABSTRACT

Heterozygous mutations in the syntaxin-binding protein 1 (STXBP1) gene, which encodes Munc18-1, a core component of the presynaptic membrane-fusion machinery, cause infantile early epileptic encephalopathy (Ohtahara syndrome), but it is unclear how a partial loss of Munc18-1 produces this severe clinical presentation. Here, we generated human ES cells designed to conditionally express heterozygous and homozygous STXBP1 loss-of-function mutations and studied isogenic WT and STXBP1-mutant human neurons derived from these conditionally mutant ES cells. We demonstrated that heterozygous STXBP1 mutations lower the levels of Munc18-1 protein and its binding partner, the t-SNARE-protein Syntaxin-1, by approximately 30% and decrease spontaneous and evoked neurotransmitter release by nearly 50%. Thus, our results confirm that using engineered human embryonic stem (ES) cells is a viable approach to studying disease-associated mutations in human neurons on a controlled genetic background, demonstrate that partial STXBP1 loss of function robustly impairs neurotransmitter release in human neurons, and suggest that heterozygous STXBP1 mutations cause early epileptic encephalopathy specifically through a presynaptic impairment.


Subject(s)
Munc18 Proteins , Mutation , Neurons , Spasms, Infantile , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Evoked Potentials/genetics , HEK293 Cells , Humans , Infant , Infant, Newborn , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Spasms, Infantile/genetics , Spasms, Infantile/metabolism , Spasms, Infantile/pathology , Syntaxin 1/genetics , Syntaxin 1/metabolism
10.
Neuron ; 78(5): 785-98, 2013 Jun 05.
Article in English | MEDLINE | ID: mdl-23764284

ABSTRACT

Available methods for differentiating human embryonic stem cells (ESCs) and induced pluripotent cells (iPSCs) into neurons are often cumbersome, slow, and variable. Alternatively, human fibroblasts can be directly converted into induced neuronal (iN) cells. However, with present techniques conversion is inefficient, synapse formation is limited, and only small amounts of neurons can be generated. Here, we show that human ESCs and iPSCs can be converted into functional iN cells with nearly 100% yield and purity in less than 2 weeks by forced expression of a single transcription factor. The resulting ES-iN or iPS-iN cells exhibit quantitatively reproducible properties independent of the cell line of origin, form mature pre- and postsynaptic specializations, and integrate into existing synaptic networks when transplanted into mouse brain. As illustrated by selected examples, our approach enables large-scale studies of human neurons for questions such as analyses of human diseases, examination of human-specific genes, and drug screening.


Subject(s)
Biophysical Phenomena/physiology , Gene Expression Regulation/physiology , Nerve Tissue Proteins/metabolism , Neurons/physiology , Pluripotent Stem Cells/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Biophysical Phenomena/genetics , Biophysics , Brain/cytology , Calcium/metabolism , Cells, Cultured , Collagen Type VII/genetics , Electric Stimulation , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/pathology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fibroblasts , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Humans , Mice , Microscopy, Confocal , Munc18 Proteins/genetics , Munc18 Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , RNA, Small Interfering/physiology , Rhodopsin/genetics , Sodium Channel Blockers/pharmacology , Synapses/physiology , Tetrodotoxin/pharmacology , Time Factors , Transfection
11.
J Neurosci ; 30(8): 2897-910, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181587

ABSTRACT

The coxsackievirus-adenovirus receptor (CAR) is a member of the Ig superfamily strongly expressed in the developing nervous system. Our histological investigations during development reveal an initial uniform distribution of CAR on all neural cells with a concentration on membranes that face the margins of the nervous system (e.g., the basal laminae and the ventricular side). At more advanced stages, CAR becomes downregulated and restricted to specific regions including areas rich in axonal and dendritic surfaces. To study the function of CAR on neural cells, we used the fiber knob of the adenovirus, extracellular CAR domains, blocking antibodies to CAR, as well as CAR-deficient neural cells. Blocking antibodies were found to inhibit neurite extension in retina organ and retinal explant cultures, whereas the application of the recombinant fiber knob of the adenovirus subtype Ad2 or extracellular CAR domains promoted neurite extension and adhesion to extracellular matrices. We observed a promiscuous interaction of CAR with extracellular matrix glycoproteins, which was deduced from analytical ultracentrifugation experiments, affinity chromatography, and adhesion assays. The membrane proximal Ig domain of CAR, termed D2, was found to bind to a fibronectin fragment, including the heparin-binding domain 2, which promotes neurite extension of wild type, but not of CAR-deficient neural cells. In contrast to heterophilic interactions, homophilic association of CAR involves both Ig domains, as was revealed by ultracentrifugation, chemical cross-linking, and adhesion studies. The results of these functional and binding studies are correlated to a U-shaped homodimer of the complete extracellular domains of CAR detected by x-ray crystallography.


Subject(s)
Cell Differentiation/genetics , Central Nervous System/embryology , Central Nervous System/metabolism , Gene Expression Regulation, Developmental/genetics , Neurogenesis/genetics , Neurons/metabolism , Receptors, Virus/metabolism , Animals , Antibodies, Blocking/pharmacology , CHO Cells , Cell Adhesion/genetics , Cells, Cultured , Central Nervous System/cytology , Chick Embryo , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Cricetinae , Cricetulus , Crystallography, X-Ray , Dimerization , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Neurites/metabolism , Neurites/ultrastructure , Neurons/cytology , Organ Culture Techniques , Protein Structure, Tertiary/physiology , Receptors, Virus/chemistry , Receptors, Virus/genetics , Retina/cytology , Retina/embryology , Retina/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...