Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Nat Commun ; 13(1): 3690, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35760976

ABSTRACT

It is unclear how the 22q11.2 deletion predisposes to psychiatric disease. To study this, we generated induced pluripotent stem cells from deletion carriers and controls and utilized CRISPR/Cas9 to introduce the heterozygous deletion into a control cell line. Here, we show that upon differentiation into neural progenitor cells, the deletion acted in trans to alter the abundance of transcripts associated with risk for neurodevelopmental disorders including autism. In excitatory neurons, altered transcripts encoded presynaptic factors and were associated with genetic risk for schizophrenia, including common and rare variants. To understand how the deletion contributed to these changes, we defined the minimal protein-protein interaction network that best explains gene expression alterations. We found that many genes in 22q11.2 interact in presynaptic, proteasome, and JUN/FOS transcriptional pathways. Our findings suggest that the 22q11.2 deletion impacts genes that may converge with psychiatric risk loci to influence disease manifestation in each deletion carrier.


Subject(s)
DiGeorge Syndrome , Induced Pluripotent Stem Cells , Schizophrenia , Cell Line , DiGeorge Syndrome/genetics , Humans , Neurons , RNA , Schizophrenia/genetics
2.
iScience ; 24(1): 101935, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33409479

ABSTRACT

Genetic variation of the 16p11.2 deletion locus containing the KCTD13 gene and of CUL3 is linked with autism. This genetic connection suggested that substrates of a CUL3-KCTD13 ubiquitin ligase may be involved in disease pathogenesis. Comparison of Kctd13 mutant (Kctd13 -/- ) and wild-type neuronal ubiquitylomes identified adenylosuccinate synthetase (ADSS), an enzyme that catalyzes the first step in adenosine monophosphate (AMP) synthesis, as a KCTD13 ligase substrate. In Kctd13 -/- neurons, there were increased levels of succinyl-adenosine (S-Ado), a metabolite downstream of ADSS. Notably, S-Ado levels are elevated in adenylosuccinate lyase deficiency, a metabolic disorder with autism and epilepsy phenotypes. The increased S-Ado levels in Kctd13 -/- neurons were decreased by treatment with an ADSS inhibitor. Lastly, functional analysis of human KCTD13 variants suggests that KCTD13 variation may alter ubiquitination of ADSS. These data suggest that succinyl-AMP metabolites accumulate in Kctd13 -/- neurons, and this observation may have implications for our understanding of 16p11.2 deletion syndrome.

3.
Sci Transl Med ; 12(544)2020 05 20.
Article in English | MEDLINE | ID: mdl-32434848

ABSTRACT

Fragile X syndrome is caused by FMR1 gene silencing and loss of the encoded fragile X mental retardation protein (FMRP), which binds to mRNA and regulates translation. Studies in the Fmr1-/y mouse model of fragile X syndrome indicate that aberrant cerebral protein synthesis downstream of metabotropic glutamate receptor 5 (mGluR5) signaling contributes to disease pathogenesis, but clinical trials using mGluR5 inhibitors were not successful. Animal studies suggested that treatment with lithium might be an alternative approach. Targets of lithium include paralogs of glycogen synthase kinase 3 (GSK3), and nonselective small-molecule inhibitors of these enzymes improved disease phenotypes in a fragile X syndrome mouse model. However, the potential therapeutic use of GSK3 inhibitors has been hampered by toxicity arising from inhibition of both α and ß paralogs. Recently, we developed GSK3 inhibitors with sufficient paralog selectivity to avoid a known toxic consequence of dual inhibition, that is, increased ß-catenin stabilization. We show here that inhibition of GSK3α, but not GSK3ß, corrected aberrant protein synthesis, audiogenic seizures, and sensory cortex hyperexcitability in Fmr1-/y mice. Although inhibiting either paralog prevented induction of NMDA receptor-dependent long-term depression (LTD) in the hippocampus, only inhibition of GSK3α impaired mGluR5-dependent and protein synthesis-dependent LTD. Inhibition of GSK3α additionally corrected deficits in learning and memory in Fmr1-/y mice; unlike mGluR5 inhibitors, there was no evidence of tachyphylaxis or enhanced psychotomimetic-induced hyperlocomotion. GSK3α selective inhibitors may have potential as a therapeutic approach for treating fragile X syndrome.


Subject(s)
Fragile X Syndrome , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/drug therapy , Glycogen Synthase Kinase 3 , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Cell Rep ; 23(8): 2509-2523, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29791859

ABSTRACT

Transcription factor programming of pluripotent stem cells (PSCs) has emerged as an approach to generate human neurons for disease modeling. However, programming schemes produce a variety of cell types, and those neurons that are made often retain an immature phenotype, which limits their utility in modeling neuronal processes, including synaptic transmission. We report that combining NGN2 programming with SMAD and WNT inhibition generates human patterned induced neurons (hpiNs). Single-cell analyses showed that hpiN cultures contained cells along a developmental continuum, ranging from poorly differentiated neuronal progenitors to well-differentiated, excitatory glutamatergic neurons. The most differentiated neurons could be identified using a CAMK2A::GFP reporter gene and exhibited greater functionality, including NMDAR-mediated synaptic transmission. We conclude that utilizing single-cell and reporter gene approaches for selecting successfully programmed cells for study will greatly enhance the utility of hpiNs and other programmed neuronal populations in the modeling of nervous system disorders.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , Adult , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Differentiation , Cells, Cultured , Fetus/cytology , Gene Expression Regulation , Humans , Neurons/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Receptors, AMPA/metabolism , Receptors, Glutamate/metabolism , Smad Proteins/metabolism , Synapses/metabolism , Time Factors , Transcription, Genetic , Wnt Proteins/metabolism
5.
Biol Psychiatry ; 73(7): 683-90, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23237312

ABSTRACT

BACKGROUND: Ankyrin 3 (ANK3) has been strongly implicated as a risk gene for bipolar disorder (BD) by recent genome-wide association studies of patient populations. However, the genetic variants of ANK3 contributing to BD risk and their pathological function are unknown. METHODS: To gain insight into the potential disease relevance of ANK3, we examined the function of mouse Ank3 in the regulation of psychiatric-related behaviors using genetic, neurobiological, pharmacological, and gene-environment interaction (G×E) approaches. Ank3 expression was reduced in mouse brain either by viral-mediated RNA interference or through disruption of brain-specific Ank3 in a heterozygous knockout mouse. RESULTS: RNA interference of Ank3 in hippocampus dentate gyrus induced a highly specific and consistent phenotype marked by decreased anxiety-related behaviors and increased activity during the light phase, which were attenuated by chronic treatment with the mood stabilizer lithium. Similar behavioral alterations of reduced anxiety and increased motivation for reward were also exhibited by Ank3+/- heterozygous mice compared with wild-type Ank3+/+ mice. Remarkably, the behavioral traits of Ank3+/- mice transitioned to depression-related features after chronic stress, a trigger of mood episodes in BD. Ank3+/- mice also exhibited elevated serum corticosterone, suggesting that reduced Ank3 expression is associated with elevated stress reactivity. CONCLUSIONS: This study defines a new role for Ank3 in the regulation of psychiatric-related behaviors and stress reactivity that lends support for its involvement in BD and establishes a general framework for determining the disease relevance of genes implicated by patient genome-wide association studies.


Subject(s)
Ankyrins/genetics , Anxiety Disorders/genetics , Anxiety Disorders/physiopathology , Bipolar Disorder/genetics , Lithium Chloride/pharmacology , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Animals , Ankyrins/physiology , Anxiety Disorders/blood , Anxiety Disorders/drug therapy , Corticosterone/blood , Dentate Gyrus/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Stress, Psychological/blood , Stress, Psychological/drug therapy
6.
Biol Mood Anxiety Disord ; 2: 18, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23025490

ABSTRACT

Bipolar disorder (BD) is a multi-factorial disorder caused by genetic and environmental influences. It has a large genetic component, with heritability estimated between 59-93%. Recent genome-wide association studies (GWAS) using large BD patient populations have identified a number of genes with strong statistical evidence for association with susceptibility for BD. Among the most significant and replicated genes is ankyrin 3 (ANK3), a large gene that encodes multiple isoforms of the ankyrin G protein. This article reviews the current evidence for genetic association of ANK3 with BD, followed by a comprehensive overview of the known biology of the ankyrin G protein, focusing on its neural functions and their potential relevance to BD. Ankyrin G is a scaffold protein that is known to have many essential functions in the brain, although the mechanism by which it contributes to BD is unknown. These functions include organizational roles for subcellular domains in neurons including the axon initial segment and nodes of Ranvier, through which ankyrin G orchestrates the localization of key ion channels and GABAergic presynaptic terminals, as well as creating a diffusion barrier that limits transport into the axon and helps define axo-dendritic polarity. Ankyrin G is postulated to have similar structural and organizational roles at synaptic terminals. Finally, ankyrin G is implicated in both neurogenesis and neuroprotection. ANK3 and other BD risk genes participate in some of the same biological pathways and neural processes that highlight several mechanisms by which they may contribute to BD pathophysiology. Biological investigation in cellular and animal model systems will be critical for elucidating the mechanism through which ANK3 confers risk of BD. This knowledge is expected to lead to a better understanding of the brain abnormalities contributing to BD symptoms, and to potentially identify new targets for treatment and intervention approaches.

7.
Neuron ; 72(4): 545-58, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-22099458

ABSTRACT

Disrupted in Schizophrenia-1 (DISC1) is a candidate gene for psychiatric disorders and has many roles during brain development. Common DISC1 polymorphisms (variants) are associated with neuropsychiatric phenotypes including altered cognition, brain structure, and function; however, it is unknown how this occurs. Here, we demonstrate using mouse, zebrafish, and human model systems that DISC1 variants are loss of function in Wnt/GSK3ß signaling and disrupt brain development. The DISC1 variants A83V, R264Q, and L607F, but not S704C, do not activate Wnt signaling compared with wild-type DISC1 resulting in decreased neural progenitor proliferation. In zebrafish, R264Q and L607F could not rescue DISC1 knockdown-mediated aberrant brain development. Furthermore, human lymphoblast cell lines endogenously expressing R264Q displayed impaired Wnt signaling. Interestingly, S704C inhibited the migration of neurons in the developing neocortex. Our data demonstrate DISC1 variants impair Wnt signaling and brain development and elucidate a possible mechanism for their role in neuropsychiatric phenotypes.


Subject(s)
Brain Chemistry/genetics , Brain/growth & development , Glycogen Synthase Kinase 3/genetics , Nerve Tissue Proteins/genetics , Polymorphism, Genetic/genetics , Signal Transduction/genetics , Wnt3A Protein/genetics , Animals , Cell Line, Tumor , Female , Genetic Variation/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Mice , Phenotype , Pregnancy , Zebrafish
8.
PLoS One ; 6(10): e26203, 2011.
Article in English | MEDLINE | ID: mdl-22022567

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited cause of intellectual disability. In addition to cognitive deficits, FXS patients exhibit hyperactivity, attention deficits, social difficulties, anxiety, and other autistic-like behaviors. FXS is caused by an expanded CGG trinucleotide repeat in the 5' untranslated region of the Fragile X Mental Retardation (FMR1) gene leading to epigenetic silencing and loss of expression of the Fragile X Mental Retardation protein (FMRP). Despite the known relationship between FMR1 CGG repeat expansion and FMR1 silencing, the epigenetic modifications observed at the FMR1 locus, and the consequences of the loss of FMRP on human neurodevelopment and neuronal function remain poorly understood. To address these limitations, we report on the generation of induced pluripotent stem cell (iPSC) lines from multiple patients with FXS and the characterization of their differentiation into post-mitotic neurons and glia. We show that clones from reprogrammed FXS patient fibroblast lines exhibit variation with respect to the predominant CGG-repeat length in the FMR1 gene. In two cases, iPSC clones contained predominant CGG-repeat lengths shorter than measured in corresponding input population of fibroblasts. In another instance, reprogramming a mosaic patient having both normal and pre-mutation length CGG repeats resulted in genetically matched iPSC clonal lines differing in FMR1 promoter CpG methylation and FMRP expression. Using this panel of patient-specific, FXS iPSC models, we demonstrate aberrant neuronal differentiation from FXS iPSCs that is directly correlated with epigenetic modification of the FMR1 gene and a loss of FMRP expression. Overall, these findings provide evidence for a key role for FMRP early in human neurodevelopment prior to synaptogenesis and have implications for modeling of FXS using iPSC technology. By revealing disease-associated cellular phenotypes in human neurons, these iPSC models will aid in the discovery of novel therapeutics for FXS and other autism-spectrum disorders sharing common pathophysiology.


Subject(s)
Epigenesis, Genetic , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Models, Biological , Nervous System/growth & development , Adult , Case-Control Studies , Cell Differentiation/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Child, Preschool , CpG Islands/genetics , DNA Methylation/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fragile X Mental Retardation Protein/metabolism , Humans , Infant, Newborn , Male , Middle Aged , Mosaicism , Mutation/genetics , Nervous System/metabolism , Nervous System/pathology , Neurons/metabolism , Neurons/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Donors , Trinucleotide Repeat Expansion/genetics
9.
Chem Biol ; 18(7): 891-906, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21802010

ABSTRACT

Target identification remains challenging for the field of chemical biology. We describe an integrative chemical genomic and proteomic approach combining the use of differentially active analogs of small molecule probes with stable isotope labeling by amino acids in cell culture-mediated affinity enrichment, followed by subsequent testing of candidate targets using RNA interference-mediated gene silencing. We applied this approach to characterizing the natural product K252a and its ability to potentiate neuregulin-1 (Nrg1)/ErbB4 (v-erb-a erythroblastic leukemia viral oncogene homolog 4)-dependent neurotrophic factor signaling and neuritogenesis. We show that AAK1 (adaptor-associated kinase 1) is a relevant target of K252a, and that the loss of AAK1 alters ErbB4 trafficking and expression levels, providing evidence for a previously unrecognized role for AAK1 in Nrg1-mediated neurotrophic factor signaling. Similar strategies should lead to the discovery of novel targets for therapeutic development.


Subject(s)
ErbB Receptors/metabolism , Nerve Growth Factors/metabolism , Neuregulin-1/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Carbazoles/metabolism , ErbB Receptors/genetics , Gene Knockdown Techniques , Genomics/methods , Humans , Indole Alkaloids/metabolism , Models, Molecular , Nerve Growth Factors/genetics , Neuregulin-1/genetics , Neurites/metabolism , PC12 Cells , Protein Serine-Threonine Kinases/genetics , Proteomics/methods , Rats , Receptor, ErbB-4 , Signal Transduction
10.
ACS Chem Neurosci ; 1(4): 325-342, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-20495671

ABSTRACT

Genetic findings have suggested that neuregulin-1 (Nrg1) and its receptor v-erb-a erythroblastic leukemia viral oncogene homolog 4 (ErbB4) may play a role in neuropsychiatric diseases. However, the downstream signaling events and relevant phenotypic consequences of altered Nrg1 signaling in the nervous system remain poorly understood. To identify small molecules for probing Nrg1-ErbB4 signaling, a PC12-cell model was developed and used to perform a live-cell, image-based screen of the effects of small molecules on Nrg1-induced neuritogenesis. By comparing the resulting phenotypic data to that of a similar screening performed with nerve growth factor (NGF), this multidimensional screen identified compounds that directly inhibit Nrg1-ErbB4 signaling, such as the 4-anilino-quinazoline Iressa (gefitinib), as well as compounds that potentiate Nrg1-ErbB4 signaling, such as the indolocarbazole K-252a. These findings provide new insights into the regulation of Nrg1-ErbB4 signaling events and demonstrate the feasibility of using such a multidimensional, chemical-genetic approach for discovering probes of pathways implicated in neuropsychiatric diseases.

11.
Cell ; 136(6): 1017-31, 2009 Mar 20.
Article in English | MEDLINE | ID: mdl-19303846

ABSTRACT

The Disrupted in Schizophrenia 1 (DISC1) gene is disrupted by a balanced chromosomal translocation (1; 11) (q42; q14.3) in a Scottish family with a high incidence of major depression, schizophrenia, and bipolar disorder. Subsequent studies provided indications that DISC1 plays a role in brain development. Here, we demonstrate that suppression of DISC1 expression reduces neural progenitor proliferation, leading to premature cell cycle exit and differentiation. Several lines of evidence suggest that DISC1 mediates this function by regulating GSK3beta. First, DISC1 inhibits GSK3beta activity through direct physical interaction, which reduces beta-catenin phosphorylation and stabilizes beta-catenin. Importantly, expression of stabilized beta-catenin overrides the impairment of progenitor proliferation caused by DISC1 loss of function. Furthermore, GSK3 inhibitors normalize progenitor proliferation and behavioral defects caused by DISC1 loss of function. Together, these results implicate DISC1 in GSK3beta/beta-catenin signaling pathways and provide a framework for understanding how alterations in this pathway may contribute to the etiology of psychiatric disorders.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Signal Transduction , beta Catenin/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Brain/cytology , Brain/embryology , Embryo, Mammalian/metabolism , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism
12.
Cell ; 133(5): 903-15, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18510933

ABSTRACT

We show that miR-1, a conserved muscle-specific microRNA, regulates aspects of both pre- and postsynaptic function at C. elegans neuromuscular junctions. miR-1 regulates the expression level of two nicotinic acetylcholine receptor (nAChR) subunits (UNC-29 and UNC-63), thereby altering muscle sensitivity to acetylcholine (ACh). miR-1 also regulates the muscle transcription factor MEF-2, which results in altered presynaptic ACh secretion, suggesting that MEF-2 activity in muscles controls a retrograde signal. The effect of the MEF-2-dependent retrograde signal on secretion is mediated by the synaptic vesicle protein RAB-3. Finally, acute activation of levamisole-sensitive nAChRs stimulates MEF-2-dependent transcriptional responses and induces the MEF-2-dependent retrograde signal. We propose that miR-1 refines synaptic function by coupling changes in muscle activity to changes in presynaptic function.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , MicroRNAs/metabolism , Neuromuscular Junction/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Levamisole/pharmacology , MicroRNAs/genetics , Mutation , Nicotinic Agonists/metabolism , Receptors, Nicotinic/metabolism , Transcription, Genetic , rab3 GTP-Binding Proteins/metabolism
13.
Neuron ; 58(3): 346-61, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18466746

ABSTRACT

GABA synapses play a critical role in many aspects of circuit development and function. For example, conditions that perturb GABA transmission have been implicated in epilepsy. To identify genes that regulate GABA transmission, we performed an RNAi screen for genes whose inactivation increases the activity of C. elegans body muscles, which receive direct input from GABAergic motor neurons. We identified 90 genes, 21 of which were previously implicated in seizure syndromes, suggesting that this screen has effectively identified candidate genes for epilepsy. Electrophysiological recordings and imaging of excitatory and inhibitory synapses indicate that several genes alter muscle activity by selectively regulating GABA transmission. In particular, we identify two humoral pathways and several protein kinases that modulate GABA transmission but have little effect on excitatory transmission at cholinergic neuromuscular junctions. Our data suggest these conserved genes are components of signaling pathways that regulate GABA transmission and consequently may play a role in epilepsy and other cognitive or psychiatric disorders.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Genetic Testing/methods , RNA Interference , Synapses/physiology , gamma-Aminobutyric Acid/physiology , Acetylcholine/physiology , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Endocrine System/physiology , Epilepsy/genetics , Epilepsy/physiopathology , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , Inhibitory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/physiology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Motor Neurons/physiology , Movement/physiology , Muscles/physiology , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Receptors, GABA/genetics , Receptors, GABA/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Synaptic Transmission/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
14.
Nat Neurosci ; 10(1): 49-57, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17128266

ABSTRACT

The secretion of neurotransmitters and neuropeptides is mediated by distinct organelles-synaptic vesicles (SVs) and dense-core vesicles (DCVs), respectively. Relatively little is known about the factors that differentially regulate SV and DCV secretion. Here we show that protein kinase C-1 (PKC-1), which is most similar to the vertebrate PKC eta and epsilon isoforms, regulates exocytosis of DCVs in Caenorhabditis elegans motor neurons. Mutants lacking PCK-1 activity had delayed paralysis induced by the acetylcholinesterase inhibitor aldicarb, whereas mutants with increased PKC-1 activity had more rapid aldicarb-induced paralysis. Imaging and electrophysiological assays indicated that SV release occurred normally in pkc-1 mutants. By contrast, genetic analysis of aldicarb responses and imaging of fluorescently tagged neuropeptides indicated that mutants lacking PKC-1 had reduced neuropeptide secretion. Similar neuropeptide secretion defects were found in mutants lacking unc-31 (encoding the protein CAPS) or unc-13 (encoding Munc13). These results suggest that PKC-1 selectively regulates DCV release from neurons.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Motor Neurons/metabolism , Neuropeptides/metabolism , Protein Kinase C/physiology , Aldicarb/pharmacology , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Carrier Proteins , Cholinesterase Inhibitors/pharmacology , Cloning, Molecular/methods , Diagnostic Imaging/methods , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Exocytosis/drug effects , Exocytosis/genetics , Gene Expression/genetics , Green Fluorescent Proteins/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Motor Neurons/drug effects , Muscles/drug effects , Muscles/metabolism , Mutant Proteins/physiology , Patch-Clamp Techniques/methods , Secretory Vesicles/drug effects , Secretory Vesicles/physiology
15.
Neuron ; 51(3): 303-15, 2006 Aug 03.
Article in English | MEDLINE | ID: mdl-16880125

ABSTRACT

Priming of synaptic vesicles (SVs) is essential for synaptic transmission. UNC-13 proteins are required for priming. Current models propose that UNC-13 stabilizes the open conformation of Syntaxin, in which the SNARE helix is available for interactions with Synaptobrevin and SNAP-25. Here we show that Tomosyn inhibits SV priming. Tomosyn contains a SNARE motif, which forms an inhibitory SNARE complex with Syntaxin and SNAP-25. Mutants lacking Tomosyn have increased synaptic transmission, an increased pool of primed vesicles, and increased abundance of UNC-13 at synapses. Behavioral, imaging, and electrophysiological studies suggest that SV priming was reconstituted in unc-13 mutants by expressing a constitutively open mutant Syntaxin, or by mutations eliminating Tomosyn. Thus, priming is modulated by the balance between Tomosyn and UNC-13, perhaps by regulating the availability of open-Syntaxin. Even when priming was restored, synaptic transmission remained defective in unc-13 mutants, suggesting that UNC-13 is also required for other aspects of secretion.


Subject(s)
Caenorhabditis elegans Proteins/physiology , SNARE Proteins/physiology , Synaptic Vesicles/physiology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Carrier Proteins , Molecular Sequence Data , Mutation , Qa-SNARE Proteins/physiology , SNARE Proteins/genetics , Synaptic Vesicles/genetics
16.
Curr Biol ; 15(24): 2236-42, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16271476

ABSTRACT

Neurotransmitter secretion at synapses is controlled by several processes-morphological docking of vesicles at release sites, priming of docked vesicles to make them fusion competent, and calcium-dependent fusion of vesicles with the plasma membrane . In worms, flies, and mice, mutants lacking UNC-13 have defects in vesicle priming . Current models propose that UNC-13 primes vesicles by stabilizing Syntaxin's "open" conformation by directly interacting with its amino-terminal regulatory domain . However, the functional significance of the UNC-13/Syntaxin interaction has not been tested directly. A truncated protein containing the Munc homology domains (MHD1 and MHD2) and the carboxy-terminal C2 domain partially rescued both the behavioral and secretion defects of unc-13 mutants in C. elegans. A double mutation in MHD2 (F1000A/K1002A) disrupts the UNC-13/Syntaxin interaction. The rate of endogenous synaptic events and the amplitude of nerve-evoked excitatory post-synaptic currents (EPSCs) were both significantly reduced in UNC-13S(F1000A/K1002A). However, the pool of primed (i.e., fusion-competent) vesicles was normal. These results suggest that the UNC-13/Syntaxin interaction is conserved in C. elegans and that, contrary to current models, the UNC-13/Syntaxin interaction is required for nerve-evoked vesicle fusion rather than synaptic-vesicle priming. Thus, UNC-13 may regulate multiple steps of the synaptic-vesicle cycle.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , Qa-SNARE Proteins/metabolism , Synaptic Transmission/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Carrier Proteins , Electrophysiology , Molecular Sequence Data , Mutation/genetics , Qa-SNARE Proteins/genetics , Sequence Alignment , Sucrose , Synaptic Transmission/genetics , Synaptic Vesicles/metabolism , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...