Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 267
Filter
1.
Mol Psychiatry ; 22(3): 375-383, 2017 03.
Article in English | MEDLINE | ID: mdl-27217145

ABSTRACT

Neuroligins are postsynaptic cell-adhesion molecules implicated in autism and other neuropsychiatric disorders. Despite extensive work, the role of neuroligins in synapse function and plasticity, especially N-methyl-d-aspartate (NMDA) receptor (NMDAR)-dependent long-term potentiation (LTP), remains unclear. To establish which synaptic functions unequivocally require neuroligins, we analyzed single and triple conditional knockout (cKO) mice for all three major neuroligin isoforms (NL1-NL3). We inactivated neuroligins by stereotactic viral expression of Cre-recombinase in hippocampal CA1 region pyramidal neurons at postnatal day 0 (P0) or day 21 (P21) and measured synaptic function, synaptic plasticity and spine numbers in acute hippocampal slices 2-3 weeks later. Surprisingly, we find that ablation of neuroligins in newborn or juvenile mice only modestly impaired basal synaptic function in hippocampus and caused no alteration in postsynaptic spine numbers. However, triple cKO of NL1-NL3 or single cKO of NL1 impaired NMDAR-mediated excitatory postsynaptic currents and abolished NMDAR-dependent LTP. Strikingly, the NL1 cKO also abolished LTP elicited by activation of L-type Ca2+-channels during blockade of NMDARs. These findings demonstrate that neuroligins are generally not essential for synapse formation in CA1 pyramidal neurons but shape synaptic properties and that NL1 specifically is required for LTP induced by postsynaptic Ca2+-elevations, a function which may contribute to the pathophysiological role of neuroligins in brain disorders.


Subject(s)
CA1 Region, Hippocampal/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Electric Stimulation , Excitatory Postsynaptic Potentials/physiology , Hippocampus/metabolism , Long-Term Potentiation/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synaptic Transmission/physiology
2.
Mol Psychiatry ; 22(10): 1483-1491, 2017 10.
Article in English | MEDLINE | ID: mdl-27725662

ABSTRACT

Neuroligins are postsynaptic cell-adhesion molecules that bind to presynaptic neurexins. Mutations in neuroligin-3 predispose to autism, but how such mutations affect synaptic function remains incompletely understood. Here we systematically examined the effect of three autism-associated mutations, the neuroligin-3 knockout, the R451C knockin, and the R704C knockin, on synaptic transmission in the calyx of Held, a central synapse ideally suited for high-resolution analyses of synaptic transmission. Surprisingly, germline knockout of neuroligin-3 did not alter synaptic transmission, whereas the neuroligin-3 R451C and R704C knockins decreased and increased, respectively, synaptic transmission. These puzzling results prompted us to ask whether neuroligin-3 mutant phenotypes may be reshaped by developmental plasticity. Indeed, conditional knockout of neuroligin-3 during late development produced a marked synaptic phenotype, whereas conditional knockout of neuroligin-3 during early development caused no detectable effect, mimicking the germline knockout. In canvassing potentially redundant candidate genes, we identified developmentally early expression of another synaptic neurexin ligand, cerebellin-1. Strikingly, developmentally early conditional knockout of cerebellin-1 only modestly impaired synaptic transmission, whereas in contrast to the individual single knockouts, developmentally early conditional double knockout of both cerebellin-1 and neuroligin-3 severely decreased synaptic transmission. Our data suggest an unanticipated mechanism of developmental compensation whereby cerebellin-1 and neuroligin-3 functionally occlude each other during development of calyx synapses. Thus, although acute manipulations more likely reveal basic gene functions, developmental plasticity can be a major factor in shaping the overall phenotypes of genetic neuropsychiatric disorders.


Subject(s)
Autistic Disorder/genetics , Autistic Disorder/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Synapses/physiology , Animals , Disease Models, Animal , Gene Knock-In Techniques , Hippocampus/metabolism , Mice , Mice, Knockout , Mutation , Neuronal Plasticity/genetics , Neurons/metabolism , Synapses/metabolism , Synaptic Transmission , Trapezoid Body/metabolism , Trapezoid Body/physiology
3.
Mol Psychiatry ; 21(2): 169-77, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25778475

ABSTRACT

Neuroligins are postsynaptic cell-adhesion molecules that bind to presynaptic neurexins. Although the general synaptic role of neuroligins is undisputed, their specific functions at a synapse remain unclear, even controversial. Moreover, many neuroligin gene mutations were associated with autism, but the pathophysiological relevance of these mutations is often unknown, and their mechanisms of action uninvestigated. Here, we examine the synaptic effects of an autism-associated neuroligin-4 substitution (called R704C), which mutates a cytoplasmic arginine residue that is conserved in all neuroligins. We show that the R704C mutation, when introduced into neuroligin-3, enhances the interaction between neuroligin-3 and AMPA receptors, increases AMPA-receptor internalization and decreases postsynaptic AMPA-receptor levels. When introduced into neuroligin-4, conversely, the R704C mutation unexpectedly elevated AMPA-receptor-mediated synaptic responses. These results suggest a general functional link between neuroligins and AMPA receptors, indicate that both neuroligin-3 and -4 act at excitatory synapses but perform surprisingly distinct functions, and demonstrate that the R704C mutation significantly impairs the normal function of neuroligin-4, thereby validating its pathogenicity.


Subject(s)
Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Synaptic Transmission/physiology , Animals , Cell Culture Techniques , Hippocampus/metabolism , Mice, Inbred Strains , Mutation , Neurons/metabolism , Olfactory Bulb/metabolism , Receptors, AMPA/metabolism , Synapses/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid
5.
Mol Psychiatry ; 20(7): 850-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25824299

ABSTRACT

Abnormal activity in the medial prefrontal cortex (mPFC) is consistently observed in neuropsychiatric disorders, but the mechanisms involved remain unclear. Chronic aberrant excitation and/or inhibition of mPFC neurons were proposed to cause cognitive impairments. However, direct evidence for this hypothesis is lacking because it is technically challenging to control synaptic properties in a chronic and locally restricted, yet specific, manner. Here, we generated conditional knockout (cKO) mice of neuroligin-2 (Nlgn2), a postsynaptic cell-adhesion molecule of inhibitory synapses linked to neuropsychiatric disorders. cKO of Nlgn2 in adult mPFC rendered Nlgn2 protein undetectable after already 2-3 weeks, but induced major reductions in synaptic inhibition after only 6-7 weeks, and caused parallel impairments in anxiety, fear memory and social interaction behaviors. Moreover, cKO of Nlgn2 severely impaired behavioral stimulation of immediate-early gene expression in the mPFC, suggesting that chronic reduction in synaptic inhibition uncoupled the mPFC from experience-dependent inputs. Our results indicate that Nlgn2 is required for continuous maintenance of inhibitory synapses in the adult mPFC, and that chronic impairment of local inhibition disengages the mPFC from its cognitive functions by partially uncoupling the mPFC from experience-induced inputs.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/physiology , Cognition Disorders/physiopathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neural Inhibition/physiology , Prefrontal Cortex/physiopathology , Synapses/physiology , Animals , Anxiety/pathology , Anxiety/physiopathology , Cell Adhesion Molecules, Neuronal/deficiency , Cognition Disorders/pathology , Conditioning, Psychological/physiology , Fear/physiology , Membrane Potentials/physiology , Memory/physiology , Mice, Knockout , Nerve Tissue Proteins/deficiency , Patch-Clamp Techniques , Prefrontal Cortex/pathology , Social Behavior , Synapses/pathology , Synaptic Transmission/physiology , Tissue Culture Techniques
6.
Genes Brain Behav ; 11(2): 201-13, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22103334

ABSTRACT

The presynaptic protein RIM1α mediates multiple forms of presynaptic plasticity at both excitatory and inhibitory synapses. Previous studies of mice lacking RIM1α (RIM1α(-/-) throughout the brain showed that deletion of RIM1α results in multiple behavioral abnormalities. In an effort to begin to delineate the brain regions in which RIM1 deletion mediates these abnormal behaviors, we used conditional (floxed) RIM1 knockout mice (fRIM1). By crossing these fRIM1 mice to previously characterized transgenic cre lines, we aimed to delete RIM1 selectively in the dentate gyrus (DG), using a specific preproopiomelanocortin promoter driving cre recombinase (POMC-cre) line , and in pyramidal neurons of the CA3 region of hippocampus, using the kainate receptor subunit 1 promoter driving cre recombinase (KA-cre). Neither of these cre driver lines was uniquely selective to the targeted regions. In spite of this, we were able to reproduce a subset of the global RIM1α(-/-) behavioral abnormalities, thereby narrowing the brain regions in which loss of RIM1 is sufficient to produce these behavioral differences. Most interestingly, hypersensitivity to the pyschotomimetic MK-801 was shown in mice lacking RIM1 selectively in the DG, arcuate nucleus of the hypothalamus and select cerebellar neurons, implicating novel brain regions and neuronal subtypes in this behavior.


Subject(s)
Behavior, Animal/physiology , Brain/metabolism , GTP-Binding Proteins/genetics , Animals , Association Learning/drug effects , Association Learning/physiology , Behavior, Animal/drug effects , Brain/drug effects , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dizocilpine Maleate/pharmacology , GTP-Binding Proteins/metabolism , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Neurons/drug effects , Neurons/metabolism , Phenotype , Promoter Regions, Genetic , Reflex, Startle/drug effects , Reflex, Startle/genetics , Rotarod Performance Test , Sensory Gating/drug effects , Sensory Gating/genetics , Social Behavior , Synapses/drug effects , Synapses/genetics , Synapses/metabolism
7.
Diabetologia ; 54(2): 415-22, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20978738

ABSTRACT

AIMS/HYPOTHESIS: Type 2 diabetes is caused by relative deficiency of insulin secretion and is associated with dysregulation of glucagon secretion during the late stage of diabetes development. Like insulin secretion from beta cells, glucagon secretion is dependent on calcium signals and a calcium sensing protein, synaptotagmin-7. In this study, we tested the relative contribution of dysregulated glucagon secretion and reduced insulin release in the development of hyperglycaemia and type 2 diabetes by using synaptotagmin-7 knockout (KO) mice, which exhibit glucose intolerance, reduced insulin secretion and nearly abolished Ca(2+)-stimulated glucagon secretion. METHODS: We fed the synaptotagmin-7 KO and control mice with a high-fat diet (HFD) for 14 weeks, and compared their body weight, glucose levels, glucose and insulin tolerance, and insulin and glucagon secretion. RESULTS: On the HFD, synaptotagmin-7 KO mice showed progressive impairment of glucose tolerance and insulin secretion, along with continued maintenance of a low glucagon level. The control mice were less affected in terms of glucose intolerance, and showed enhanced insulin secretion with a concurrent increase in glucagon levels. Unexpectedly, after 14 weeks of HFD feeding, only the control mice displayed resting hyperglycaemia, whereas in synaptotagmin-7 KO mice defective insulin secretion and reduced insulin sensitivity were not sufficient to cause hyperglycaemia in the absence of enhanced glucagon secretion. CONCLUSIONS/INTERPRETATION: Our data uncover a previously overlooked role of dysregulated glucagon secretion in promoting hyperglycaemia and the ensuing diabetes, and strongly suggest maintenance of adequate regulation of glucagon secretion as an important therapeutic target in addition to the preservation of beta cell function and mass in the prevention and treatment of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon/metabolism , Hyperglycemia/metabolism , Synaptotagmins/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/genetics , Dietary Fats , Glycogen/metabolism , Hyperglycemia/blood , Hyperglycemia/genetics , Male , Mice , Mice, Knockout , Synaptotagmins/genetics
8.
Genes Brain Behav ; 8(1): 114-26, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19016888

ABSTRACT

Neuroligins (NL) are postsynaptic cell adhesion molecules that are thought to specify synapse properties. Previous studies showed that mutant mice carrying an autism-associated point mutation in NL3 exhibit social interaction deficits, enhanced inhibitory synaptic function and increased staining of inhibitory synaptic puncta without changes in overall inhibitory synapse numbers. In contrast, mutant mice lacking NL2 displayed decreased inhibitory synaptic function. These studies raised two relevant questions. First, does NL2 deletion impair inhibitory synaptic function by altering the number of inhibitory synapses, or by changing their efficacy? Second, does this effect of NL2 deletion on inhibition produce behavioral changes? We now show that although NL2-deficient mice exhibit an apparent decrease in number of inhibitory synaptic puncta, the number of symmetric synapses as determined by electron microscopy is unaltered, suggesting that NL2 deletion impairs the function of inhibitory synapses without decreasing their numbers. This decrease in inhibitory synaptic function in NL2-deficient mice correlates with a discrete behavioral phenotype that includes a marked increase in anxiety-like behavior, a decrease in pain sensitivity and a slight decrease in motor co-ordination. This work confirms that NL2 modulates inhibitory synaptic function and is the first demonstration that global deletion of NL2 can lead to a selective behavioral phenotype.


Subject(s)
Anxiety/genetics , Anxiety/psychology , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Animals , Behavior, Animal/physiology , Blotting, Western , Cell Adhesion Molecules, Neuronal , Electroshock , Gene Deletion , Interpersonal Relations , Learning/physiology , Male , Mice , Mice, Knockout , Microscopy, Electron , Motor Activity/physiology , Pain/genetics , Pain/psychology , Pain Measurement/psychology , Postural Balance/physiology , Social Behavior , Synapses/metabolism , Synapses/ultrastructure
9.
Eur J Neurosci ; 27(12): 3118-31, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18598257

ABSTRACT

Cysteine string protein (CSPalpha) is a synaptic vesicle protein present in most central and peripheral nervous system synapses. Previous studies demonstrated that the deletion of CSPalpha results in postnatal sensorial and motor impairment and premature lethality. To understand the participation of CSPalpha in neural function in vertebrates, we have studied the properties of synaptic transmission of motor terminals in wild-type and CSPalpha knockout mice. Our results demonstrate that, in the absence of CSPalpha, fast Ca2+-triggered release was not affected at postnatal day (P)14 but was dramatically reduced at P18 and P30 without a change in release kinetics. Although mutant terminals also exhibited a reduction in functional vesicle pool size by P30, further analysis showed that neurotransmission could be 'rescued' by high extracellular [Ca2+] or by the presence of a phorbol ester, suggesting that an impairment in the fusion machinery, or in vesicle recycling, was not the primary cause of the dysfunction of this synapse. The specific shift to the right of the Ca2+ dependence of synchronous release, and the lineal dependence of secretion on extracellular [Ca2+] in mutant terminals after P18, suggests that CSPalpha is indispensable for a normal Ca2+ sensitivity of exocytosis in vertebrate mature synapses.


Subject(s)
Calcium/metabolism , Exocytosis/physiology , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Synapses/physiology , Animals , Calcium Channels/physiology , Carcinogens/pharmacology , Evoked Potentials/drug effects , Evoked Potentials/physiology , Exocytosis/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Models, Neurological , Neurons/physiology , Phorbol Esters/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
10.
Proc Natl Acad Sci U S A ; 102(51): 18664-9, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16352718

ABSTRACT

Synaptotagmin 1 likely acts as a Ca2+ sensor in neurotransmitter release by Ca2+-binding to its two C2 domains. This notion was strongly supported by the observation that a mutation in the C2A domain causes parallel decreases in the apparent Ca2+ affinity of synaptotagmin 1 and in the Ca2+ sensitivity of release. However, this study was based on a single loss-of-function mutation. We now show that tryptophan substitutions in the synaptotagmin 1 C2 domains act as gain-of-function mutations to increase the apparent Ca2+ affinity of synaptotagmin 1. The same substitutions, when introduced into synaptotagmin 1 expressed in neurons, enhance the Ca2+ sensitivity of release. Mutations in the two C2 domains lead to comparable and additive effects in release. Our results thus show that the apparent Ca2+ sensitivity of release is dictated by the apparent Ca2+ affinity of synaptotagmin 1 in both directions, and that Ca2+ binding to both C2 domains contributes to Ca2+ triggering of release.


Subject(s)
Calcium/metabolism , Glutamic Acid/metabolism , Synaptotagmin I/metabolism , Animals , Calcium/pharmacology , Cations, Divalent/metabolism , Cations, Divalent/pharmacology , Cells, Cultured , Mice , Mice, Knockout , Models, Molecular , Mutation/genetics , Protein Structure, Tertiary , Synaptotagmin I/chemistry , Synaptotagmin I/deficiency , Synaptotagmin I/genetics , Time Factors , Tryptophan/genetics , Tryptophan/metabolism
11.
Biochem Soc Trans ; 33(Pt 6): 1345-9, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16246115

ABSTRACT

RIM1alpha (Rab3-interacting molecule 1alpha) is a large multidomain protein that is localized to presynaptic active zones [Wang, Okamoto, Schmitz, Hofmann and Südhof (1997) Nature (London) 388, 593-598] and is the founding member of the RIM protein family that also includes RIM2alpha, 2beta, 2gamma, 3gamma and 4gamma [Wang and Südhof (2003) Genomics 81, 126-137]. In presynaptic nerve termini, RIM1alpha interacts with a series of presynaptic proteins, including the synaptic vesicle GTPase Rab3 and the active zone proteins Munc13, liprins and ELKS (a protein rich in glutamate, leucine, lysine and serine). Mouse KOs (knockouts) revealed that, in different types of synapses, RIM1alpha is essential for different forms of synaptic plasticity. In CA1-region Schaffer-collateral excitatory synapses and in GABAergic synapses (where GABA is gamma-aminobutyric acid), RIM1alpha is required for maintaining normal neurotransmitter release and short-term synaptic plasticity. In contrast, in excitatory CA3-region mossy fibre synapses and cerebellar parallel fibre synapses, RIM1alpha is necessary for presynaptic long-term, but not short-term, synaptic plasticity. In these synapses, the function of RIM1alpha in presynaptic long-term plasticity depends, at least in part, on phosphorylation of RIM1alpha at a single site, suggesting that RIM1alpha constitutes a 'phosphoswitch' that determines synaptic strength. However, in spite of the progress in understanding RIM1alpha function, the mechanisms by which RIM1alpha acts remain unknown. For example, how does phosphorylation regulate RIM1alpha, what is the relationship of the function of RIM1alpha in basic release to synaptic plasticity and what is the physiological significance of different forms of RIM-dependent plasticity? Moreover, the roles of other RIM isoforms are unclear. Addressing these important questions will contribute to our view of how neurotransmitter release is regulated at the presynaptic active zone.


Subject(s)
GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Protein Isoforms/metabolism , Synapses/metabolism , Animals , Protein Structure, Tertiary , rab3 GTP-Binding Proteins/metabolism
12.
Neuroscience ; 118(4): 985-1002, 2003.
Article in English | MEDLINE | ID: mdl-12732244

ABSTRACT

In humans, mutations in the alpha-synuclein gene or exposure to the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) produce Parkinson's disease with loss of dopaminergic neurons and depletion of nigrostriatal dopamine. alpha-Synuclein is a vertebrate-specific component of presynaptic nerve terminals that may function in modulating synaptic transmission. To test whether MPTP toxicity involves alpha-synuclein, we generated alpha-synuclein-deficient mice by homologous recombination, and analyzed the effect of deleting alpha-synuclein on MPTP toxicity using these knockout mice. In addition, we examined commercially available mice that contain a spontaneous loss of the alpha-synuclein gene. As described previously, deletion of alpha-synuclein had no significant effects on brain structure or composition. In particular, the levels of synaptic proteins were not altered, and the concentrations of dopamine, dopamine metabolites, and dopaminergic proteins were unchanged. Upon acute MPTP challenge, alpha-synuclein knockout mice were partly protected from chronic depletion of nigrostriatal dopamine when compared with littermates of the same genetic background, whereas mice carrying the spontaneous deletion of the alpha-synuclein gene exhibited no protection. Furthermore, alpha-synuclein knockout mice but not the mice with the alpha-synuclein gene deletion were slightly more sensitive to methamphetamine than littermate control mice. These results demonstrate that alpha-synuclein is not obligatorily coupled to MPTP sensitivity, but can influence MPTP toxicity on some genetic backgrounds, and illustrate the need for extensive controls in studies aimed at describing the effects of mouse knockouts on MPTP sensitivity.


Subject(s)
Nerve Tissue Proteins/metabolism , Parkinsonian Disorders/metabolism , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Antibodies/metabolism , Blastomeres/metabolism , Blotting, Southern/methods , Corpus Striatum/metabolism , DNA Primers/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Glutamic Acid/metabolism , Hippocampus/metabolism , Homovanillic Acid/metabolism , Humans , Immunoblotting/methods , Immunohistochemistry/methods , MPTP Poisoning , Methamphetamine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Mice, Knockout/metabolism , Mice, Transgenic , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurons/metabolism , Parkinsonian Disorders/chemically induced , Piperazines/pharmacology , Rats , Reserpine/pharmacology , Serotonin/metabolism , Stem Cells/metabolism , Subcellular Fractions/metabolism , Substantia Nigra/metabolism , Synucleins , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein
13.
Neuroscience ; 112(1): 51-63, 2002.
Article in English | MEDLINE | ID: mdl-12044471

ABSTRACT

Ca(2+)-signalling plays a major role in regulating all aspects of neuronal function. Different types of neurons exhibit characteristic differences in the responses to Ca(2+)-signals. Correlating with differences in Ca(2+)-response are expression patterns of Ca(2+)-binding proteins that often serve as markers for various types of neurons. For example, in the cerebral cortex the EF-hand Ca(2+)-binding proteins parvalbumin and calbindin are primarily expressed in inhibitory interneurons where they influence Ca(2+)-dependent responses. We have now identified a new family of proteins called NECABs (neuronal Ca(2+)-binding proteins). NECABs contain an N-terminal EF-hand domain that binds Ca(2+), but different from many other neuronal EF-hand Ca(2+)-binding proteins, only a single EF-hand domain is present. At the C-terminus, NECABs include a DUF176 motif, a bacterial domain of unknown function that was previously not observed in eukaryotes. In rat at least three closely related NECAB genes are expressed either primarily in brain (NECABs 1 and 2) or in brain and muscle (NECAB 3). Immunocytochemistry revealed that NECAB 1 is restricted to subsets of neurons. In cerebral cortex, NECAB 1 is highly and uniformly expressed only in layer 4 pyramidal neurons, whereas in hippocampus only inhibitory interneurons and CA2 pyramidal cells contain NECAB 1. In these neurons, NECAB 1 fills the entire cytoplasm similar to other EF-hand Ca(2+)-binding proteins, and is not concentrated in any particular subcellular compartment. We suggest that NECABs represent a novel family of regulatory Ca(2+)-binding proteins with an unusual domain structure and a limited expression in a subclass of neurons.


Subject(s)
Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Amino Acid Sequence/genetics , Animals , Brain/metabolism , COS Cells , Calcium-Binding Proteins/isolation & purification , Cloning, Molecular , Eye Proteins/isolation & purification , Immunohistochemistry , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/isolation & purification , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , RNA, Messenger/metabolism , Rats , Tissue Distribution
14.
Science ; 294(5544): 1117-22, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11691998

ABSTRACT

SNAREs (soluble NSF-attachment protein receptors) are generally acknowledged as central components of membrane fusion reactions, but their precise function has remained enigmatic. Competing hypotheses suggest roles for SNAREs in mediating the specificity of fusion, catalyzing fusion, or actually executing fusion. We generated knockout mice lacking synaptobrevin/VAMP 2, the vesicular SNARE protein responsible for synaptic vesicle fusion in forebrain synapses, to make use of the exquisite temporal resolution of electrophysiology in measuring fusion. In the absence of synaptobrevin 2, spontaneous synaptic vesicle fusion and fusion induced by hypertonic sucrose were decreased approximately 10-fold, but fast Ca2+-triggered fusion was decreased more than 100-fold. Thus, synaptobrevin 2 may function in catalyzing fusion reactions and stabilizing fusion intermediates but is not absolutely required for synaptic fusion.


Subject(s)
Membrane Fusion , Membrane Proteins/physiology , Synapses/physiology , Synaptic Vesicles/physiology , Vesicular Transport Proteins , Action Potentials , Animals , Calcium/metabolism , Calcium/pharmacology , Cells, Cultured , Hypertonic Solutions , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutation , Patch-Clamp Techniques , Potassium/pharmacology , Presynaptic Terminals/physiology , Prosencephalon/physiology , R-SNARE Proteins , SNARE Proteins , Sucrose/pharmacology , Synaptic Transmission
15.
J Biol Chem ; 276(51): 47869-76, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11604393

ABSTRACT

Rearrangements of the actin cytoskeleton are involved in a variety of cellular processes from locomotion of cells to morphological alterations of the cell surface. One important question is how local interactions of cells with the extracellular space are translated into alterations of their membrane organization. To address this problem, we studied CASK, a member of the membrane-associated guanylate kinase homologues family of adaptor proteins. CASK has been shown to bind the erythrocyte isoform of protein 4.1, a class of proteins that promote formation of actin/spectrin microfilaments. In neurons, CASK also interacts via its PDZ domain with the cytosolic C termini of neurexins, neuron-specific cell-surface proteins. We now show that CASK binds a brain-enriched isoform of protein 4.1, and nucleates local assembly of actin/spectrin filaments. These interactions can be reconstituted on the cytosolic tail of neurexins. Furthermore, CASK can be recovered with actin filaments prepared from rat brain extracts, and neurexins are recruited together with CASK and protein 4.1 into these actin filaments. Thus, analogous to the PDZ-domain protein p55 and glycophorin C at the erythrocyte membrane, a similar complex comprising CASK and neurexins exists in neurons. Our data suggest that intercellular junctions formed by neurexins, such as junctions initiated by beta-neurexins with neuroligins, are at least partially coupled to the actin cytoskeleton via an interaction with CASK and protein 4.1.


Subject(s)
Actins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases , Cytoskeletal Proteins , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides , Nucleoside-Phosphate Kinase/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cytoplasm/metabolism , Glycophorins/metabolism , Guanylate Kinases , Molecular Sequence Data , Nucleoside-Phosphate Kinase/chemistry , Protein Binding , Rats , Sequence Homology, Amino Acid , Spectrin/metabolism
16.
Neuron ; 31(6): 987-99, 2001 Sep 27.
Article in English | MEDLINE | ID: mdl-11580898

ABSTRACT

We identify a chaperone complex composed of (1) the synaptic vesicle cysteine string protein (CSP), thought to function in neurotransmitter release, (2) the ubiquitous heat-shock protein cognate Hsc70, and (3) the SGT protein containing three tandem tetratricopeptide repeats. These three proteins interact with each other to form a stable trimeric complex that is located on the synaptic vesicle surface, and is disrupted in CSP knockout mice. The CSP/SGT/Hsc70 complex functions as an ATP-dependent chaperone that reactivates a denatured substrate. SGT overexpression in cultured neurons inhibits neurotransmitter release, suggesting that the CSP/SGT/Hsc70 complex is important for maintenance of a normal synapse. Taken together, our results identify a novel trimeric complex that functions as a synapse-specific chaperone machine.


Subject(s)
Exocytosis/physiology , HSP70 Heat-Shock Proteins/physiology , Membrane Proteins/physiology , Molecular Chaperones/physiology , Nerve Tissue Proteins/physiology , Neurotransmitter Agents/metabolism , Proteins/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Adenosine Triphosphate/physiology , Animals , Brain Chemistry , Carrier Proteins , Cells, Cultured , HSC70 Heat-Shock Proteins , HSP40 Heat-Shock Proteins , HSP70 Heat-Shock Proteins/chemistry , Hippocampus/cytology , Macromolecular Substances , Male , Membrane Proteins/chemistry , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Models, Biological , Molecular Chaperones/chemistry , Nerve Tissue Proteins/chemistry , Protein Binding , Protein Folding , Proteins/chemistry , Rats , Rats, Wistar , Synaptic Vesicles/chemistry , Two-Hybrid System Techniques
17.
Neuropharmacology ; 41(6): 643-9, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11640918

ABSTRACT

Rab3A is a GTP-binding protein of synaptic vesicles that regulates neurotransmitter release and cycles on and off synaptic vesicles as a function of exocytosis. Rab3A presumably functions via GTP-dependent interactions with effectors. Two putative rab3A effectors have been described in neurons, rabphilin which is a soluble protein that moves onto and off synaptic vesicles in concert with rab3A, and RIM which is an active zone protein that only binds to rab3A on docked vesicles. Rabphilin is an abundant, evolutionarily conserved protein whose function has remained enigmatic since a knockout of rabphilin does not display the functional deficiencies observed in the rab3A knockout. However, previous studies have shown that rabphilin is phosphorylated by protein kinase A and CaM Kinase II, suggesting that it may have a regulatory role. In the present study, we have examined the site and regulation of rabphilin phosphorylation in living nerve terminals using phospho-specific antibodies raised against phospho-serine234 of rabphilin. With these antibodies, we demonstrate that rabphilin is physiologically phosphorylated on serine234, and that soluble rabphilin which is not bound to rab3A on synaptic vesicles is the primary target. However, different from synapsins which are induced to dissociate from synaptic vesicles by PKA phosphorylation, phosphorylation of rabphilin is not instrumental for dissociating rabphilin from synaptic vesicles. Our data support the notion that dissociated rabphilin is a synaptic phosphoprotein in vivo that may play a role in the regulation of nerve terminal protein-protein interactions.


Subject(s)
Antibody Specificity , Immune Sera/metabolism , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , rab GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cattle , Hippocampus/metabolism , Humans , Immune Sera/biosynthesis , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/immunology , Phosphoproteins/immunology , Phosphorylation , Rats , Serine/metabolism , Synaptosomes/metabolism , Vesicular Transport Proteins , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/immunology , Rabphilin-3A
18.
Neuron ; 31(4): 581-91, 2001 Aug 30.
Article in English | MEDLINE | ID: mdl-11545717

ABSTRACT

Secretory vesicles dock at the plasma membrane before Ca(2+) triggers their exocytosis. Exocytosis requires the assembly of SNARE complexes formed by the vesicle protein Synaptobrevin and the membrane proteins Syntaxin-1 and SNAP-25. We analyzed the role of Munc18-1, a cytosolic binding partner of Syntaxin-1, in large dense-core vesicle (LDCV) secretion. Calcium-dependent LDCV exocytosis was reduced 10-fold in mouse chromaffin cells lacking Munc18-1, but the kinetic properties of the remaining release, including single fusion events, were not different from controls. Concomitantly, mutant cells displayed a 10-fold reduction in morphologically docked LDCVs. Moreover, acute overexpression of Munc18-1 in bovine chromaffin cells increased the amount of releasable vesicles and accelerated vesicle supply. We conclude that Munc18-1 functions upstream of SNARE complex formation and promotes LDCV docking.


Subject(s)
Chromaffin Cells/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Vesicles/metabolism , Vesicular Transport Proteins , Animals , Antigens, Surface/metabolism , Cattle , Cell Membrane/metabolism , Chromaffin Cells/ultrastructure , Exocytosis/physiology , Female , Fetus/cytology , Gene Deletion , Gene Expression/physiology , Membrane Potentials/physiology , Mice , Mice, Mutant Strains , Microscopy, Electron , Munc18 Proteins , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Pregnancy , Syntaxin 1
19.
Neuroscience ; 106(2): 263-74, 2001.
Article in English | MEDLINE | ID: mdl-11566499

ABSTRACT

Apoptotic protease-activating factor-1 (Apaf-1), dATP, and procaspase-9 form a multimeric complex that triggers programmed cell death through the activation of caspases upon release of cytochrome c from the mitochondria into the cytosol. Although cell death pathways exist that can bypass the requirement for cytochrome c release and caspase activation, several gene knockout studies have shown that the cytochrome c-mediated apoptotic pathway is critical for neural development. Specifically, the number of neuronal progenitor cells is abnormally increased in Apaf-1-, caspase-9-, caspase-3-deficient mice. However, the role of the cytochrome c cell death pathway for apoptosis of postmitotic, differentiated neurons in the developing brain has not been investigated in vivo. In this study we investigated embryonic neuronal cell death caused by trophic factor deprivation or lack of neurotransmitter release by analyzing Apaf-1/tyrosine kinase receptor A (TrkA) and Apaf-1/Munc-18 double mutant mice. Histological analysis of the double mutants' brains (including cell counting and terminal (TdT)-mediated dUTP-biotin nick end labeling (TUNEL) staining) reveals that neuronal cell death caused by these stimuli can proceed independent of Apaf-1. We propose that a switch between apoptotic programs (and their respective proteins) characterizes the transition of a neuronal precursor cell from the progenitor pool to the postmitotic population of differentiated neurons.


Subject(s)
Apoptosis/genetics , Nerve Growth Factors/metabolism , Nerve Tissue Proteins , Nervous System/embryology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Proteins/metabolism , Stem Cells/metabolism , Vesicular Transport Proteins , Animals , Apoptotic Protease-Activating Factor 1 , Caspases/metabolism , Cell Cycle/genetics , Cell Differentiation/genetics , Cytochrome c Group/metabolism , Ganglia, Sensory/cytology , Ganglia, Sensory/embryology , Ganglia, Sensory/metabolism , Mice , Mice, Knockout , Munc18 Proteins , Nervous System/cytology , Nervous System/metabolism , Neurons/cytology , Proteins/genetics , Receptor, trkA/deficiency , Receptor, trkA/genetics , Signal Transduction/genetics , Stem Cells/cytology
20.
Proc Natl Acad Sci U S A ; 98(20): 11680-5, 2001 Sep 25.
Article in English | MEDLINE | ID: mdl-11562488

ABSTRACT

Synaptotagmin I is a synaptic vesicle-associated protein essential for synchronous neurotransmission. We investigated its impact on the intracellular Ca(2+)-dependence of large dense-core vesicle (LDCV) exocytosis by combining Ca(2+)-uncaging and membrane capacitance measurements in adrenal slices from mouse synaptotagmin I null mutants. Synaptotagmin I-deficient chromaffin cells displayed prolonged exocytic delays and slow, yet Ca(2+)-dependent fusion rates, resulting in strongly reduced LDCV release in response to short depolarizations. Vesicle recruitment, the shape of individual amperometric events, and endocytosis appeared unaffected. These findings demonstrate that synaptotagmin I is required for rapid, highly Ca(2+)-sensitive LDCV exocytosis and indicate that it regulates the equilibrium between a slowly releasable and a readily releasable state of the fusion machinery. Alternatively, synaptotagmin I could function as calcium sensor for the readily releasable pool, leading to the destabilization of the pool in its absence.


Subject(s)
Adrenal Glands/physiology , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Chromaffin Cells/physiology , Exocytosis/physiology , Intracellular Fluid/physiology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Calcium-Binding Proteins/deficiency , In Vitro Techniques , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Synaptotagmin I , Synaptotagmins
SELECTION OF CITATIONS
SEARCH DETAIL
...