Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 24(9): 1210-1215, 2021 09.
Article in English | MEDLINE | ID: mdl-34341585

ABSTRACT

Cortical and subcortical circuitry are thought to play distinct roles in the generation of sleep oscillations and global state control, respectively. Here we silenced a subset of neocortical layer 5 pyramidal and archicortical dentate gyrus granule cells in male mice by ablating SNAP25. This markedly increased wakefulness and reduced rebound of electroencephalographic slow-wave activity after sleep deprivation, suggesting a role for the cortex in both vigilance state control and sleep homeostasis.


Subject(s)
Dentate Gyrus/physiology , Neocortex/physiology , Neurons/physiology , Sleep/physiology , Wakefulness/physiology , Animals , Male , Mice , Mice, Transgenic , Synaptosomal-Associated Protein 25/deficiency
2.
J Cell Sci ; 132(3)2019 02 12.
Article in English | MEDLINE | ID: mdl-30659110

ABSTRACT

In mammals, fertilization initiates Ca2+ oscillations in metaphase II oocytes, which are required for the activation of embryo development. Germinal vesicle (GV) oocytes also display Ca2+ oscillations, although these unfold spontaneously in the absence of any known agonist(s) and their function remains unclear. We found that the main intracellular store of Ca2+ in GV oocytes, the endoplasmic reticulum ([Ca2+]ER), constitutively 'leaks' Ca2+ through the type 1 inositol 1,4,5-trisphosphate receptor. The [Ca2+]ER leak ceases around the resumption of meiosis, the GV breakdown (GVBD) stage, which coincides with the first noticeable accumulation of Ca2+ in the stores. It also concurs with downregulation of the Ca2+ influx and termination of the oscillations, which seemed underpinned by the inactivation of the putative plasma membrane Ca2+ channels. Lastly, we demonstrate that mitochondria take up Ca2+ during the Ca2+ oscillations, mounting their own oscillations that stimulate the mitochondrial redox state and increase the ATP levels of GV oocytes. These distinct features of Ca2+ homeostasis in GV oocytes are likely to underpin the acquisition of both maturation and developmental competence, as well as fulfill stage-specific cellular functions during oocyte maturation.


Subject(s)
Calcium Signaling/genetics , Calcium/metabolism , Endoplasmic Reticulum/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mitochondria/metabolism , Oocytes/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Cell Membrane/metabolism , Cell Nucleus/metabolism , Female , Gene Expression Regulation , Homeostasis/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Metaphase , Mice , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Oocytes/cytology , Oogenesis/genetics , Primary Cell Culture , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Synaptosomal-Associated Protein 25/deficiency , Synaptosomal-Associated Protein 25/genetics
3.
Hippocampus ; 28(10): 735-744, 2018 10.
Article in English | MEDLINE | ID: mdl-29995325

ABSTRACT

Adult neurogenesis is necessary for proper cognition and behavior, however, the mechanisms that underlie the integration and maturation of newborn neurons into the pre-existing hippocampal circuit are not entirely known. In this study, we sought to determine the role of action potential (AP)-dependent synaptic transmission by adult-generated dentate granule cells (DGCs) in their survival and function within the existing circuitry. We used a triple transgenic mouse (NestinCreERT2 :Snap25fl/fl : tdTomato) to inducibly inactivate AP-dependent synaptic transmission within adult hippocampal progenitors and their progeny. Behavioral testing in a hippocampal-dependent A/B contextual fear-discrimination task revealed impaired discrimination learning in mice harboring SNAP-25-deficient adult-generated dentate granule cells (DGCs). Despite poor performance on this neurogenesis-dependent task, the production and survival of newborn DGCs was quantitatively unaltered in tamoxifen-treated NestinCreERT2 :Snap25fl/fl : tdTomato SNAP compared to tamoxifen-treated NestinCreERT2 :Snap25wt/wt : tdTomato control mice. Although SNAP-25-deficient adult DGCs displayed a small but statistically significant enhancement in proximal dendritic branching, their overall dendritic length and distal branching complexity was unchanged. SNAP-25-deficient newborn DGCs also displayed robust efferent mossy fiber output to CA3, with normal linear density of large mossy fiber terminals (LMTs). These studies suggest that AP-dependent neurotransmitter release by newborn DGCs is not essential for their survival or rudimentary structural maturation within the adult hippocampus.


Subject(s)
Hippocampus/cytology , Hippocampus/growth & development , Learning Disabilities/genetics , Neurogenesis/physiology , Neurons/physiology , Synaptosomal-Associated Protein 25/deficiency , Animals , Animals, Newborn , Cells, Cultured , Discrimination Learning/drug effects , Discrimination Learning/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Fear/physiology , Glutamic Acid/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Learning Disabilities/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Neurons/drug effects , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Synaptosomal-Associated Protein 25/genetics , Transfection
4.
J Cell Sci ; 130(11): 1877-1889, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28404788

ABSTRACT

Neuronal dense-core vesicles (DCVs) transport and secrete neuropeptides necessary for development, plasticity and survival, but little is known about their fusion mechanism. We show that Snap-25-null mutant (SNAP-25 KO) neurons, previously shown to degenerate after 4 days in vitro (DIV), contain fewer DCVs and have reduced DCV fusion probability in surviving neurons at DIV14. At DIV3, before degeneration, SNAP-25 KO neurons show normal DCV fusion, but one day later fusion is significantly reduced. To test if other SNAP homologs support DCV fusion, we expressed SNAP-23, SNAP-29 or SNAP-47 in SNAP-25 KO neurons. SNAP-23 and SNAP-29 rescued viability and supported DCV fusion in SNAP-25 KO neurons, but SNAP-23 did so more efficiently. SNAP-23 also rescued synaptic vesicle (SV) fusion while SNAP-29 did not. SNAP-47 failed to rescue viability and did not support DCV or SV fusion. These data demonstrate a developmental switch, in hippocampal neurons between DIV3 and DIV4, where DCV fusion becomes SNAP-25 dependent. Furthermore, SNAP-25 homologs support DCV and SV fusion and neuronal viability to variable extents - SNAP-23 most effectively, SNAP-29 less so and SNAP-47 ineffectively.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Secretory Vesicles/metabolism , Synaptosomal-Associated Protein 25/genetics , Animals , Biological Transport , Cell Death/genetics , Embryo, Nonmammalian , Exocytosis , Gene Expression Regulation , Genetic Complementation Test , Hippocampus/pathology , Membrane Fusion , Mice , Mice, Knockout , Neurons/pathology , Presynaptic Terminals/pathology , Primary Cell Culture , Protein Isoforms/genetics , Protein Isoforms/metabolism , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , Secretory Vesicles/pathology , Synaptic Transmission , Synaptosomal-Associated Protein 25/deficiency
5.
Proc Natl Acad Sci U S A ; 112(31): E4326-35, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195742

ABSTRACT

Synaptosomal-associated protein of 25 kDa (SNAP-25) is a key molecule in the soluble N-ethylmaleimide-sensitive factor attachment protein (SNARE) complex mediating fast Ca(2+)-triggered release of hormones and neurotransmitters, and both splice variants, SNAP-25a and SNAP-25b, can participate in this process. Here we explore the hypothesis that minor alterations in the machinery mediating regulated membrane fusion can increase the susceptibility for metabolic disease and precede obesity and type 2 diabetes. Thus, we used a mouse mutant engineered to express normal levels of SNAP-25 but only SNAP-25a. These SNAP-25b-deficient mice were exposed to either a control or a high-fat/high-sucrose diet. Monitoring of food intake, body weight, hypothalamic function, and lipid and glucose homeostases showed that SNAP-25b-deficient mice fed with control diet developed hyperglycemia, liver steatosis, and adipocyte hypertrophy, conditions dramatically exacerbated when combined with the high-fat/high-sucrose diet. Thus, modified SNARE function regulating stimulus-dependent exocytosis can increase the vulnerability to and even provoke metabolic disease. When combined with a high-fat/high-sucrose diet, this vulnerability resulted in diabesity. Our SNAP-25b-deficient mouse may represent a diabesity model.


Subject(s)
Metabolic Diseases/metabolism , Synaptosomal-Associated Protein 25/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Animals , Blood Glucose/metabolism , Body Weight , Dyslipidemias/pathology , Energy Intake , Energy Metabolism , Feeding Behavior , Female , Homeostasis , Hypertrophy , Hypothalamus/metabolism , Insulin/metabolism , Insulin Secretion , Leptin/blood , Liver/metabolism , Liver/pathology , Male , Metabolic Diseases/blood , Mice, Obese , Phenotype , Receptors, Leptin/metabolism , Synaptosomal-Associated Protein 25/deficiency
6.
Lancet Neurol ; 14(4): 420-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25792100

ABSTRACT

The congenital myasthenic syndromes (CMS) are a diverse group of genetic disorders caused by abnormal signal transmission at the motor endplate, a special synaptic contact between motor axons and each skeletal muscle fibre. Most CMS stem from molecular defects in the muscle nicotinic acetylcholine receptor, but they can also be caused by mutations in presynaptic proteins, mutations in proteins associated with the synaptic basal lamina, defects in endplate development and maintenance, or defects in protein glycosylation. The specific diagnosis of some CMS can sometimes be reached by phenotypic clues pointing to the mutated gene. In the absence of such clues, exome sequencing is a useful technique for finding the disease gene. Greater understanding of the mechanisms of CMS have been obtained from structural and electrophysiological studies of the endplate, and from biochemical studies. Present therapies for the CMS include cholinergic agonists, long-lived open-channel blockers of the acetylcholine receptor ion channel, and adrenergic agonists. Although most CMS are treatable, caution should be exercised as some drugs that are beneficial in one syndrome can be detrimental in another.


Subject(s)
Adrenergic Agonists/therapeutic use , Cholinergic Agonists/therapeutic use , Mutation , Myasthenic Syndromes, Congenital , Neuromuscular Junction/metabolism , Receptors, Cholinergic/genetics , Acetylcholinesterase/genetics , Choline O-Acetyltransferase/deficiency , Collagen/genetics , Exome/genetics , Humans , Laminin/deficiency , Laminin/genetics , Muscle Proteins/genetics , Myasthenic Syndromes, Congenital/diagnosis , Myasthenic Syndromes, Congenital/drug therapy , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism , Myasthenic Syndromes, Congenital/physiopathology , Neuromuscular Junction/genetics , Neuromuscular Junction/physiopathology , Receptors, Cholinergic/metabolism , Sequence Analysis, DNA , Synaptosomal-Associated Protein 25/deficiency , Synaptotagmin II/deficiency
7.
Eur J Neurosci ; 35(10): 1586-94, 2012 May.
Article in English | MEDLINE | ID: mdl-22607004

ABSTRACT

We are interested in the role of neural activity mediated through regulated vesicular release in the stopping and early branching of the thalamic projections in the cortex. Axon outgrowth, arrival at the cortical subplate, side-branch formation during the waiting period and cortical plate innervation of embryonic thalamocortical projections occurs without major abnormalities in the absence of regulated release in Snap25 (-/-) null mutant mice [Washbourne et al. (2002) Nat. Neurosci. 5:19-26; Molnár et al. (2002) J. Neurosci. 22:10313-10323]. The fact that Snap25 (-/-) null mutant mice die at birth limited our previous experiments to the prenatal period. We therefore investigated the behaviour of thalamic projections in co-culture paradigms by using heterochronic thalamic [embryonic day (E)16-E18] and cortical [postnatal day (P)0-P3] explants, in which the stopping and branching behaviour has been previously documented. Our current co-culture experiments established that thalamic projections from E16-E18 Snap25(+/+) or Snap25 (-/-) explants behaved in an identical fashion in P0-P3 Snap25 (+/+) cortical explants after 7 days in vitro. Thalamic projections from Snap25 (-/-) explants developed similar patterns of fibre ingrowth to the cortex, and stopped and formed branches at a similar depth in the Snap25(+/+) cortical slice as in control cultures. These results imply that thalamic projections can reach their ultimate target cells in layer 4, stop, and start to develop branches in the absence of regulated vesicular transmitter release from their own terminals.


Subject(s)
Axons/physiology , Cerebral Cortex , Neural Pathways , Neurons/cytology , Synaptosomal-Associated Protein 25/deficiency , Thalamus , Amino Acids/metabolism , Animals , Animals, Newborn , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Coculture Techniques , Embryo, Mammalian , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/cytology , Neural Pathways/embryology , Neural Pathways/growth & development , Organ Culture Techniques , Statistics, Nonparametric , Thalamus/cytology , Thalamus/embryology , Thalamus/growth & development
8.
Nat Neurosci ; 15(5): 738-45, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22406549

ABSTRACT

Synaptic vesicles in the brain harbor several soluble N-ethylmaleimide-sensitive-factor attachment protein receptor (SNARE) proteins. With the exception of synaptobrevin2, or VAMP2 (syb2), which is directly involved in vesicle fusion, the role of these SNAREs in neurotransmission is unclear. Here we show that in mice syb2 drives rapid Ca(2+)-dependent synchronous neurotransmission, whereas the structurally homologous SNARE protein VAMP4 selectively maintains bulk Ca(2+)-dependent asynchronous release. At inhibitory nerve terminals, up- or downregulation of VAMP4 causes a correlated change in asynchronous release. Biochemically, VAMP4 forms a stable complex with SNAREs syntaxin-1 and SNAP-25 that does not interact with complexins or synaptotagmin-1, proteins essential for synchronous neurotransmission. Optical imaging of individual synapses indicates that trafficking of VAMP4 and syb2 show minimal overlap. Taken together, these findings suggest that VAMP4 and syb2 diverge functionally, traffic independently and support distinct forms of neurotransmission. These results provide molecular insight into how synapses diversify their release properties by taking advantage of distinct synaptic vesicle-associated SNAREs.


Subject(s)
Neurons/cytology , R-SNARE Proteins/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/physiology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Ammonium Chloride/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Cells, Cultured , Cholecystokinin/metabolism , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Gene Knockdown Techniques , Hippocampus/cytology , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Mice , Mice, Knockout , Microscopy, Immunoelectron , Mutation/physiology , Neurons/drug effects , Neurons/ultrastructure , Patch-Clamp Techniques , Protein Binding/drug effects , Protein Binding/genetics , Protein Transport/drug effects , Protein Transport/genetics , R-SNARE Proteins/genetics , RNA Interference/physiology , Rats , Rats, Sprague-Dawley , SNARE Proteins/metabolism , Synapses/genetics , Synapses/ultrastructure , Synaptic Transmission/genetics , Synaptic Vesicles/drug effects , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/deficiency , Syntaxin 1/metabolism , Transfection , Valine/analogs & derivatives , Valine/pharmacology , Vesicle-Associated Membrane Protein 2/deficiency
9.
Brain Res ; 1431: 1-12, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22119397

ABSTRACT

Paired pulse facilitation (PPF) is a form of short-term synaptic plasticity that results from an interaction of residual presynaptic Ca(2+) ([Ca(2+)](res)), number of release-competent vesicles, and the sensitivity of the vesicle release mechanisms to Ca(2+). While PPF is predominant at hippocampal Schaffer collateral-CA1 (SC-CA1) synapses, facilitation is greater in adult mice (designated Tkneo) that over express an isoform of the plasma membrane-targeted SNARE protein, SNAP-25a, which is normally predominantly expressed in juvenile animals. SNAP-25 is essential for action potential-dependent neuroexocytosis, yet the significance of the shift between the alternatively spliced variants SNAP-25a and SNAP-25b is not fully understood. This alteration of a key component of the protein machinery required for neurotransmitter release in Tkneo mice, therefore, provides a useful tool to further investigate presynaptic mechanisms that influence short-term plasticity. To explore this link between SNAP-25 and PPF, we simultaneously measured postsynaptic potentials and presynaptic [Ca(2+)](res) during paired-pulses in adult Tkneo, heterozygote null (HET), and wild type (WT) mice. We demonstrate that enhanced PPF is maintained at mature hippocampal synapses of Tkneo mice that predominantly express SNAP-25a, and that [Ca(2+)](res) kinetics are altered at synapses of Tkneo and HET mice, both of which exhibit reduced levels of total SNAP-25 expression. To evaluate the role of SNAP-25 in short-term plasticity and [Ca(2+)](res) regulation, we applied a vesicular release probability model for neurotransmission. Our results suggest that the isoform expression and total level of SNAP-25 affect both [Ca(2+)](res) dynamics and the ability of releasable vesicles to enter into a facilitated state.


Subject(s)
Calcium/metabolism , Hippocampus/cytology , Presynaptic Terminals/metabolism , Synapses/genetics , Synaptosomal-Associated Protein 25/deficiency , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Action Potentials/genetics , Animals , Biophysics , Electric Stimulation , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Presynaptic Terminals/drug effects , Sodium Channel Blockers/pharmacology , Synapses/drug effects , Synapses/physiology , Tetrodotoxin/pharmacology , Time Factors
11.
EMBO J ; 29(15): 2477-90, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20562829

ABSTRACT

The SNARE-complex consisting of synaptobrevin-2/VAMP-2, SNAP-25 and syntaxin-1 is essential for evoked neurotransmission and also involved in spontaneous release. Here, we used cultured autaptic hippocampal neurons from Snap-25 null mice rescued with mutants challenging the C-terminal, N-terminal and middle domains of the SNARE-bundle to dissect out the involvement of these domains in neurotransmission. We report that the stabilities of two different sub-domains of the SNARE-bundle have opposing functions in setting the probability for both spontaneous and evoked neurotransmission. Destabilizing the C-terminal end of the SNARE-bundle abolishes spontaneous neurotransmitter release and reduces evoked release probability, indicating that the C-terminal end promotes both modes of release. In contrast, destabilizing the middle or deleting the N-terminal end of the SNARE-bundle increases both spontaneous and evoked release probabilities. In both cases, spontaneous release was affected more than evoked neurotransmission. In addition, the N-terminal deletion delays vesicle priming after a high-frequency train. We propose that the stability of N-terminal two-thirds of the SNARE-bundle has a function for vesicle priming and limiting spontaneous release.


Subject(s)
Synaptic Transmission , Synaptosomal-Associated Protein 25/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Gene Deletion , Hippocampus/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Mutation , Sequence Alignment , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/deficiency , Synaptosomal-Associated Protein 25/genetics
12.
Ann N Y Acad Sci ; 1152: 145-53, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19161385

ABSTRACT

The evolutionary conserved soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) fusion machinery is the operational unit in the release of neurotransmitters and hormones from excitable cells. The SNARE core complex consists of three proteins named SNAP-25 (synaptosomal-associated protein of 25 kD), syntaxin 1, and VAMP (vesicle-associated membrane protein)/synaptobrevin. Syntaxin 1 is, together with SNAP-25, localized to the plasma membrane, whereas VAMP/synaptobrevin is a component of secretory vesicles. In concert with the SNAREs, accessory factors govern the docking and priming of secretory vesicles prior to trans-SNARE complex formation and ultimately Ca(2+)-triggered fusion pore opening at the plasma membrane. The synaptic SNAP-25 protein exists as two closely related protein variants, named SNAP-25a and SNAP-25b. SNAP-25a and SNAP-25b are both encoded from a single copy gene and generated by obligate alternative splicing between two similar exon 5 sequences. Exon 5 spans a region of SNAP-25 that is subject to posttranslational palmitoylation and implicated in membrane anchoring of this cytosolic protein. The alternative splicing is strictly developmentally and neuroanatomically regulated, but the biological relevance of the distinct expression of these two similar protein variants is still a question of debate. However, recent findings in gene-targeted mouse mutants have started to unravel the importance that physiological levels of total SNAP-25 protein are present and, importantly, that this is accompanied by a balanced expression of SNAP-25a and SNAP-25b.


Subject(s)
Synaptosomal-Associated Protein 25/metabolism , Animals , Gene Expression Regulation/genetics , Humans , Mice , Mutation/genetics , Synaptosomal-Associated Protein 25/deficiency , Synaptosomal-Associated Protein 25/genetics , Transgenes/genetics
13.
BMC Neurosci ; 9: 105, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18959796

ABSTRACT

BACKGROUND: The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, comprised of SNAP-25, syntaxin 1A, and VAMP-2, has been shown to be responsible for action potential (AP)-dependent, calcium-triggered release of several neurotransmitters. However, this basic fusogenic protein complex may be further specialized to suit the requirements for different neurotransmitter systems, as exemplified by neurons and neuroendocrine cells. In this study, we investigate the effects of SNAP-25 ablation on spontaneous neuronal activity and the expression of functionally distinct isoforms of this t-SNARE in GABAergic and glutamatergic neurons of the adult brain. RESULTS: We found that neurons cultured from Snap25 homozygous null mutant (Snap25-/-) mice failed to develop synchronous network activity seen as spontaneous AP-dependent calcium oscillations and were unable to trigger glial transients following depolarization. Voltage-gated calcium channel (VGCC) mediated calcium transients evoked by depolarization, nevertheless, did not differ between soma of SNAP-25 deficient and control neurons. Furthermore, we observed that although the expression of SNAP-25 RNA transcripts varied among neuronal populations in adult brain, the relative ratio of the transcripts encoding alternatively spliced SNAP-25 variant isoforms was not different in GABAergic and glutamatergic neurons. CONCLUSION: We propose that the SNAP-25b isoform is predominantly expressed by both mature glutamatergic and GABAergic neurons and serves as a fundamental component of SNARE complex used for fast synaptic communication in excitatory and inhibitory circuits required for brain function. Moreover, SNAP-25 is required for neurons to establish AP-evoked synchronous network activity, as measured by calcium transients, whereas the loss of this t-SNARE does not affect voltage-dependent calcium entry.


Subject(s)
Calcium Signaling/physiology , Glutamic Acid/metabolism , Neurons/metabolism , Synaptosomal-Associated Protein 25/metabolism , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Analysis of Variance , Animals , Astrocytes/cytology , Astrocytes/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Calcium Channels/physiology , Calcium Signaling/drug effects , Cells, Cultured , Fluorescence , Glutamate Decarboxylase/metabolism , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SNARE Proteins/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Synaptosomal-Associated Protein 25/deficiency , Synaptosomal-Associated Protein 25/genetics
14.
J Neurochem ; 103(1): 115-23, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877635

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a CAG-expansion in the gene encoding the protein huntingtin. The disease is characterized by progressive motor disturbances, cognitive defects, dementia, and weight loss. Using western blotting and immunohistochemistry we have assessed the expression levels and patterns of a number of proteins involved in neurotransmitter release in post-mortem frontal cortex samples from 10 HD cases with different disease grades. We report a loss of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein, synaptosome-associated protein 25 (SNAP 25) in HD brains of grades I-IV. Moreover, in brains of grade III and IV we found a reduction in rabphilin 3a, a protein involved in vesicle docking and recycling. These losses appear to be specific and not due to a general loss of synapses in the HD cortex. Thus, levels of synaptobrevin II, syntaxin 1, rab3a or synaptophysin are unaltered in the same patient samples. SNAP 25 and rabphilin 3a are crucial for neurotransmitter release. Therefore, we suggest that a deficient pre-synaptic transmitter release may underlie some of the symptoms of HD.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Frontal Lobe/chemistry , Huntington Disease/pathology , Nerve Tissue Proteins/deficiency , Somatosensory Cortex/chemistry , Synaptosomal-Associated Protein 25/deficiency , Vesicular Transport Proteins/deficiency , Adaptor Proteins, Signal Transducing/analysis , Adult , Aged , Aged, 80 and over , Exocytosis/genetics , Female , Frontal Lobe/pathology , Humans , Male , Middle Aged , Nerve Tissue Proteins/analysis , Severity of Illness Index , Somatosensory Cortex/pathology , Synapses/pathology , Synaptophysin/analysis , Synaptosomal-Associated Protein 25/analysis , Syntaxin 1/analysis , Vesicle-Associated Membrane Protein 2/analysis , Vesicular Transport Proteins/analysis , rab3A GTP-Binding Protein/analysis , Rabphilin-3A
15.
J Neurosci ; 27(35): 9380-91, 2007 Aug 29.
Article in English | MEDLINE | ID: mdl-17728451

ABSTRACT

The SNAP receptor (SNARE) complex, consisting of synaptosome-associated protein of 25 kDa (SNAP-25), synaptobrevin-2, and syntaxin-1, is involved in synaptic vesicles exocytosis. In addition, SNAP-25 has been implicated in constitutive exocytosis processes required for neurite outgrowth. However, at least three isoforms of SNAP-25 have been reported from neurons: SNAP-23, which is also present in non-neuronal cells, and the two alternative splice variants SNAP-25a and SNAP-25b. Here, we studied the differential ability of these isoforms to support the functions previously broadly ascribed to "SNAP-25." We studied the rescue of snap-25 null neurons in culture with different SNAP-25 homologs. We find that deletion of SNAP-25 leads to strongly reduced neuron survival, and, in the few surviving cells, impaired arborization, reduced spontaneous release, and complete arrest of evoked release. Lentiviral expression of SNAP-25a, SNAP-25b, or SNAP-23 rescued neuronal survival, arborization, amplitude, and frequency of spontaneous events. Also evoked release was rescued by all isoforms, but synchronous release required SNAP-25a/b in both glutamatergic and GABAergic neurons. SNAP-23 supported asynchronous release only, reminiscent of synaptotagmin-1 null neurons. SNAP-25b was superior to SNAP-25a in vesicle priming, resembling the shift to larger releasable vesicle pools that accompanies synaptic maturation. These data demonstrate a differential ability of SNAP-25b, SNAP-25a, and SNAP-23 to support neuronal function.


Subject(s)
Neurons/physiology , Synaptosomal-Associated Protein 25/physiology , Alternative Splicing/physiology , Analysis of Variance , Animals , Cell Survival/genetics , Cells, Cultured , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Embryo, Mammalian , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Gene Expression Regulation/physiology , Genetic Vectors/physiology , Glutamic Acid/metabolism , Hippocampus/cytology , Lentivirus/physiology , Mice , Mice, Knockout , Patch-Clamp Techniques/methods , Pyridinium Compounds/metabolism , Synaptosomal-Associated Protein 25/deficiency , gamma-Aminobutyric Acid/metabolism
16.
J Neurophysiol ; 98(2): 794-806, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17553942

ABSTRACT

At the synapse, SNAP-25, along with syntaxin/HPC-1 and synaptobrevin/VAMP, forms SNARE N-ethylmaleimide-sensitive factor [soluble (NSF) attachment protein receptor] complexes that are thought to catalyze membrane fusion. Results from neuronal cultures of synaptobrevin-2 knockout (KO) mice showed that loss of synaptobrevin has a more severe effect on calcium-evoked release than on spontaneous release or on release evoked by hypertonicity. In this study, we recorded neurotransmitter release from neuronal cultures of SNAP-25 KO mice to determine whether they share this property. In neurons lacking SNAP-25, as those deficient in synaptobrevin-2, we found that approximately 10-12% of calcium-independent excitatory and inhibitory neurotransmitter release persisted. However, in contrast to synaptobrevin-2 knockouts, this remaining readily releasable pool in SNAP-25-deficient synapses was virtually insensitive to calcium-dependent-evoked stimulation. Although field stimulation reliably evoked neurotransmitter release in synaptobrevin-2 KO neurons, responses were rare in neurons lacking SNAP-25, and unlike synaptobrevin-2-deficient synapses, SNAP-25-deficient synapses did not exhibit facilitation of release during high-frequency stimulation. This severe loss of evoked exocytosis was matched by a reduction, but not a complete loss, of endocytosis during evoked stimulation. Moreover, synaptic vesicle turnover probed by FM-dye uptake and release during hypertonic stimulation was relatively unaffected by the absence of SNAP-25. This last difference indicates that in contrast to synaptobrevin, SNAP-25 does not directly function in endocytosis. Together, these results suggest that SNAP-25 has a more significant role in calcium-secretion coupling than synaptobrevin-2.


Subject(s)
Calcium/metabolism , Pyramidal Cells/physiology , Synapses/physiology , Synaptic Transmission/genetics , Synaptosomal-Associated Protein 25/deficiency , Animals , Calcium/pharmacology , Cells, Cultured , Dose-Response Relationship, Radiation , Electric Stimulation , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/cytology , Ionomycin/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Knockout , Microscopy, Immunoelectron/methods , Patch-Clamp Techniques , Potassium/pharmacology , Pyramidal Cells/drug effects , Pyridinium Compounds/pharmacokinetics , Quaternary Ammonium Compounds/pharmacokinetics , Synapses/drug effects , Synapses/ultrastructure , Synapsins/metabolism , Synaptic Transmission/drug effects , Time Factors
17.
Mol Biol Cell ; 16(12): 5675-85, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16195346

ABSTRACT

The essential membrane fusion apparatus in mammalian cells, the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, consists of four alpha-helices formed by three proteins: SNAP-25, syntaxin 1, and synaptobrevin 2. SNAP-25 contributes two helices to the complex and is targeted to the plasma membrane by palmitoylation of four cysteines in the linker region. It is alternatively spliced into two forms, SNAP-25a and SNAP-25b, differing by nine amino acids substitutions. When expressed in chromaffin cells from SNAP-25 null mice, the isoforms support different levels of secretion. Here, we investigated the basis of that different secretory phenotype. We found that two nonconservative substitutions in the N-terminal SNARE domain and not the different localization of one palmitoylated cysteine cause the functional difference between the isoforms. Biochemical and molecular dynamic simulation experiments revealed that the two substitutions do not regulate secretion by affecting the property of SNARE complex itself, but rather make the SNAP-25b-containing SNARE complex more available for the interaction with accessory factor(s).


Subject(s)
Alternative Splicing , SNARE Proteins/genetics , Synaptosomal-Associated Protein 25/genetics , Amino Acid Sequence , Amino Acid Substitution , Animals , Chromaffin Cells/physiology , Circular Dichroism , Computer Simulation , Exons/genetics , Mice , Mice, Knockout , Molecular Sequence Data , SNARE Proteins/chemistry , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...