Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Biochim Biophys Acta Mol Basis Dis ; 1868(2): 166304, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34826585

ABSTRACT

OBJECTIVE: SNAP-25 is one of the key proteins involved in formation of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complexes that are at the core of hormonal secretion and synaptic transmission. Altered expression or function of SNAP-25 can contribute to the development of neuropsychiatric and metabolic disease. A dominant negative (DN) I67T missense mutation in the b-isoform of SNAP-25 (DN-SNAP25mut) mice leads to abnormal interactions within the SNARE complex and impaired exocytotic vesicle recycling, yet the significance of this mutation to any association between the central nervous system and metabolic homeostasis is unknown. METHODS: Here we explored aspects of metabolism, steroid hormone production and neurobehavior of DN-SNAP25mut mice. RESULTS: DN-SNAP25mut mice displayed enhanced insulin function through increased Akt phosphorylation, alongside increased adrenal and gonadal hormone production. In addition, increased anxiety behavior and beigeing of white adipose tissue with increased energy expenditure were observed in mutants. CONCLUSIONS: Our results show that SNAP25 plays an important role in bridging central neurological systems with peripheral metabolic homeostasis, and provide potential insights between metabolic disease and neuropsychiatric disorders in humans.


Subject(s)
Behavior, Animal , Gonadal Steroid Hormones/metabolism , Homeostasis , Insulin Resistance , Metabolic Diseases/pathology , Mutation , Synaptosomal-Associated Protein 25/genetics , Animals , Female , Male , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Mice , Mice, Inbred C3H , Synaptic Transmission , Synaptosomal-Associated Protein 25/physiology
2.
Neurosci Lett ; 715: 134616, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31705923

ABSTRACT

INTRODUCTION: Synaptosomal associated proteins of 25 kDa (SNAP-25), as a member of stable soluble N-ethylmaleimide-sensitive factor attachment protein receptor complex, is critical for membrane fusion and required for the release of neurotransmitters. The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor is implicated in pathologic pain. This study aimed to investigate whether and how SNAP-25 regulated AMPA receptors in neuropathic pain. METHODS: Male Sprague-Dawley rats underwent L4 spinal nerve ligation (SNL) or the sham procedure. After assessing mechanical allodynia and thermal sensitivity, the ipsilateral portion of the L4-5 spinal cord was harvested. The expression level of SNAP-25 was analyzed by Western blot analysis and real-time quantitative polymerase chain reaction. SNAP-25 phosphorylation and AMPA receptor membrane trafficking levels were evaluated with Western blot analysis. An association between SNAP-25 and AMPA membrane trafficking was confirmed by SNAP-25 expression or phosphorylation inhibition. RESULTS: The SNL procedure induced and maintained mechanical allodynia and thermal hyperalgesia. SNL increased the expression and phosphorylation of SNAP-25 and the membrane trafficking of AMPA receptors in the spinal cord. SNAP-25 expression or phosphorylation inhibition alleviated neuropathic pain and downregulated membrane trafficking of AMPA receptors after SNL. GluA1-containing AMPA receptor inhibition relieved mechanical allodynia and thermal hyperalgesia after SNL. CONCLUSIONS: The upregulation of SNAP-25-dependent membrane trafficking of AMPA receptors via SNAP-25 phosphorylation at Ser187 contributed to SNL-induced neuropathic pain. Thus, the inhibition of SNAP-25 expression or phosphorylation might serve as a treatment for neuropathic pain. However, the mechanism of GluA1-containing AMPA receptor membrane trafficking mediated by SNAP-25 phosphorylation in neuropathic pain deserves further exploration.


Subject(s)
Neuralgia/metabolism , Receptors, AMPA/metabolism , Spinal Injuries/metabolism , Spinal Injuries/physiopathology , Synaptosomal-Associated Protein 25/physiology , Animals , Male , Neuralgia/prevention & control , Pain Measurement , Phosphorylation/drug effects , RNA, Small Interfering/pharmacology , Rats , Receptors, AMPA/antagonists & inhibitors , Synaptosomal-Associated Protein 25/antagonists & inhibitors , Synaptosomal-Associated Protein 25/biosynthesis
3.
Neuroscience ; 423: 86-97, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31705888

ABSTRACT

Synaptosomal-associated protein 25 (SNAP-25) plays an important role in neuropathic pain. However, the underlying mechanism is largely unknown. Vesicular glutamate transporter 2 (VGluT2) is an isoform of vesicular glutamate transporters that controls the storage and release of glutamate. In the present study, we found the expression levels of VGluT2 correlated with the upregulation of SNAP-25 in the spinal cord of rats following chronic constriction injury (CCI)-induced neuropathic pain. Cleavage of SNAP-25 by Botulinum toxin A (BoNT/A) attenuated mechanical allodynia, downregulated the expression of VGluT2 and reduced glutamate release. Overexpression of VGluT2 abolished the antinociceptive effect of BoNT/A. Upregulation of SNAP-25 in naive rats increased VGluT2 expression and induced pain-responsive behaviors. In pheochromocytoma (PC12) cells, the expression of VGluT2 was also depended on SNAP-25 dysregulation. Moreover, we found VGluT2 was involved in SNAP-25-mediated regulation of astrocyte expression and activation of the PKA/p-CREB pathway mediated the upregulation of SNAP-25 in neuropathic pain. The findings of our study indicate that VGluT2 contributes to the effect of SNAP-25 in maintaining the development of neuropathic pain and suggests a novel mechanism underlying SNAP-25 regulation of neuropathic pain.


Subject(s)
Hyperalgesia/prevention & control , Neuralgia/physiopathology , Synaptosomal-Associated Protein 25/physiology , Vesicular Glutamate Transport Protein 2/biosynthesis , Animals , Astrocytes/metabolism , Botulinum Toxins, Type A/pharmacology , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation/drug effects , Glutamic Acid/metabolism , Hyperalgesia/physiopathology , Male , Protein Kinases/metabolism , Rats , Signal Transduction/physiology , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism , Synaptosomal-Associated Protein 25/metabolism , Up-Regulation
4.
Sci Rep ; 9(1): 1211, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30718891

ABSTRACT

Homozygous mutations in SNAP29, encoding a SNARE protein mainly involved in membrane fusion, cause CEDNIK (Cerebral Dysgenesis, Neuropathy, Ichthyosis and Keratoderma), a rare congenital neurocutaneous syndrome associated with short life expectancy, whose pathogenesis is unclear. Here, we report the analysis of the first genetic model of CEDNIK in zebrafish. Strikingly, homozygous snap29 mutant larvae display CEDNIK-like features, such as microcephaly and skin defects. Consistent with Snap29 role in membrane fusion during autophagy, we observe accumulation of the autophagy markers p62 and LC3, and formation of aberrant multilamellar organelles and mitochondria. Importantly, we find high levels of apoptotic cell death during early development that might play a yet uncharacterized role in CEDNIK pathogenesis. Mutant larvae also display mouth opening problems, feeding impairment and swimming difficulties. These alterations correlate with defective trigeminal nerve formation and excess axonal branching. Since the paralog Snap25 is known to promote axonal branching, Snap29 might act in opposition with, or modulate Snap25 activity during neurodevelopment. Our vertebrate genetic model of CEDNIK extends the description in vivo of the multisystem defects due to loss of Snap29 and could provide the base to test compounds that might ameliorate traits of the disease.


Subject(s)
Keratoderma, Palmoplantar/metabolism , Neurocutaneous Syndromes/metabolism , SNARE Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Autophagy , Humans , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/physiopathology , Membrane Fusion , Models, Genetic , Mutation , Nervous System Malformations/metabolism , Neurocutaneous Syndromes/genetics , Neurocutaneous Syndromes/physiopathology , Phenotype , Protein Binding , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/metabolism , SNARE Proteins/physiology , Synaptosomal-Associated Protein 25/metabolism , Synaptosomal-Associated Protein 25/physiology , Zebrafish/metabolism , Zebrafish Proteins/physiology
5.
Cereb Cortex ; 29(5): 2148-2159, 2019 05 01.
Article in English | MEDLINE | ID: mdl-29850799

ABSTRACT

Synaptosomal associated protein 25 kDa (SNAP25) is an essential component of the SNARE complex regulating synaptic vesicle fusion. SNAP25 deficiency has been implicated in a variety of cognitive disorders. We ablated SNAP25 from selected neuronal populations by generating a transgenic mouse (B6-Snap25tm3mcw (Snap25-flox)) with LoxP sites flanking exon5a/5b. In the presence of Cre-recombinase, Snap25-flox is recombined to a truncated transcript. Evoked synaptic vesicle release is severely reduced in Snap25 conditional knockout (cKO) neurons as shown by live cell imaging of synaptic vesicle fusion and whole cell patch clamp recordings in cultured hippocampal neurons. We studied Snap25 cKO in subsets of cortical projection neurons in vivo (L5-Rbp4-Cre; L6-Ntsr1-Cre; L6b-Drd1a-Cre). cKO neurons develop normal axonal projections, but axons are not maintained appropriately, showing signs of swelling, fragmentation and eventually complete absence. Onset and progression of degeneration are dependent on the neuron type, with L5 cells showing the earliest and most severe axonal loss. Ultrastructural examination revealed that cKO neurites contain autophagosome/lysosome-like structures. Markers of inflammation such as Iba1 and lipofuscin are increased only in adult cKO cortex. Snap25 cKO can provide a model to study genetic interactions with environmental influences in several disorders.


Subject(s)
Brain/growth & development , Brain/pathology , Neurons/pathology , Neurons/physiology , Synaptosomal-Associated Protein 25/physiology , Animals , Axons/pathology , Axons/physiology , Axons/ultrastructure , Brain/ultrastructure , Female , Male , Mice, Knockout , Neurons/ultrastructure , Synaptic Transmission , Synaptic Vesicles
6.
Biol Psychol ; 138: 35-40, 2018 10.
Article in English | MEDLINE | ID: mdl-30092259

ABSTRACT

Attention deficit/hyperactivity disorder (ADHD) is a highly heritable neurodevelopment disorder. The deficit in working memory is a central cognitive impairment in ADHD. The SNAP-25 is a neurotransmitter vesicular docking protein whose MnlI polymorphism (rs3746544) is located in the 3'-untranslated region (3'-UTR) and known to be linked to ADHD, but the underlying mechanism of this polymorphism remains unclear. Using a functional connectivity density (FCD) mapping method based on resting-state functional magnetic resonance imaging in a sample of male children diagnosed with ADHD, we first investigated the correlation between SNAP-25 rs3746544 and FCD hubs. Compared with rs3746544 G-allele carriers, TT homozygous, which confers a high risk for ADHD, exhibited significantly decreased local and long-range FCD in anterior cingulate cortex, and decreased local FCD in the dorsal lateral prefrontal cortex. Moreover, both higher local and long-range FCD could predict better WM capacity. The current findings provide new insights into the underlying neural mechanisms linking SNAP-25 rs3746544 with the risk for ADHD via the endophenotype of brain functional connectivity.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Magnetic Resonance Imaging , Memory, Short-Term/physiology , Synaptosomal-Associated Protein 25/physiology , Attention Deficit Disorder with Hyperactivity/diagnostic imaging , Attention Deficit Disorder with Hyperactivity/physiopathology , Brain/diagnostic imaging , Brain/physiopathology , Child , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/physiopathology , Homozygote , Humans , Male , Polymorphism, Genetic , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/physiopathology
7.
Drug Des Devel Ther ; 12: 1905-1915, 2018.
Article in English | MEDLINE | ID: mdl-29983545

ABSTRACT

BACKGROUND: Botulinum neurotoxin type A (BoNT/A) is emerging as a treatment modality for Raynaud's phenomenon (RP). However, the mechanism of the role of BoNT/A in antagonizing the constriction of arteriola in RP remains unclear. MATERIALS AND METHODS: We tested the constriction of arteriole diameter and the distribution of adrenergic receptors on the rat cremaster modle. Moreover, we measured the secretion of norepinephrine (NE), protein level changes and related receptors on cultured rat superior cervical ganglia neurons(SCGNs), a model of sympathetic neuron. RESULTS: Based on our results, the inhibition of arteriole vasoconstriction was increased with increasing doses of BoNT/A. BoNT/A, prazosin, and BQ123 treatment can result in significant inhibition of arteriole vasoconstriction with the same electrical stimulation. The inhibition effect of prazosin was equivalent to BoNT/A, while BQ123 has a synergistic effect with BoNT/A. After treating SCGNs using BoNT/A for 30 min, the decrease in fluorescence intensity of FM1-43 slowed down which was correlated with the doses of BoNT/A. Furthermore, release of NE in the supernatant was significantly decreased as measured by enzyme-linked immunosorbent assay, 24 h after a high dose of BoNT/A (25 µ/mL). Cleaved-SNAP-25 was detected by Western blotting 24 h following BoNT/A (50 µ/mL) treatment. Moreover, receptor SV2C, GM1, and FGFR3 were detected on sympathetic neurons, similarly to cholinergic neurons. CONCLUSION: Our study showed that BoNT/A could significantly inhibit electrical stimulation-induced arteriole vasoconstriction through the sympathetic pathway. The mechanism was similar to the cholinergic one, in which the vesicle release of sympathetic neurons could be inhibited by cleavage of SNAP-25. The end result was blocked vesicle fusion with the presynaptic membrane after BoNT/A treatment, inhibiting the release of the NE.


Subject(s)
Botulinum Toxins, Type A/therapeutic use , Raynaud Disease/drug therapy , Animals , Arterioles/drug effects , Arterioles/physiology , Botulinum Toxins, Type A/pharmacology , Dose-Response Relationship, Drug , Male , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/analysis , Synaptosomal-Associated Protein 25/physiology
8.
Article in English | MEDLINE | ID: mdl-29248569

ABSTRACT

It is generally accepted that information about some of the odorants in the natal streams of anadromous Pacific salmon (Genus Oncorhynchus) is imprinted during their seaward migration, and that anadromous Pacific salmon use olfaction to identify their natal streams during the homeward migration. However, little is known about the molecular mechanisms of the various pre-synaptic functions that are important for olfactory imprinting and memory retrieval in the salmon brain. Synaptosome-associated protein-25 (SNAP-25) mediates pre-synaptic vesicle exocytosis and regulates synaptic transmission and neuronal plasticity. Despite the importance of synaptic plasticity for memorization, the expression of SNAP-25 in the salmon brain is not well understood. In this study, snap25 expression was detected in chum salmon (O. keta) brains using molecular biological techniques. Two cDNAs encoding salmon SNAP-25 were isolated and sequenced (SNAP-25a and SNAP-25b). These cDNAs encoded proteins with 204 amino acid residues, which showed marked homology with each other (97%). The protein and nucleotide sequences demonstrated a high level of homology between salmon SNAP-25s and those of other teleost species. By quantitative PCR, the expression of snap25a and snap25b was detected in all regions of the salmon brain, especially in the telencephalon. The expression levels of snap25a in the olfactory blub were higher during seaward migration than in upriver and post-upriver migrations, reflecting synaptogenesis in the olfactory nervous system, and snap25b in the telencephalon was increased during upriver period. Our results indicated that snap25s gene is involved in synaptic plasticity for olfactory imprinting and/or olfactory memory retrieval in Pacific salmon.


Subject(s)
Animal Migration , Brain/metabolism , Salmon/physiology , Synaptosomal-Associated Protein 25/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Exocytosis/physiology , Female , Imprinting, Psychological , Male , Memory , Neuronal Plasticity , Olfactory Bulb/metabolism , Phylogeny , Real-Time Polymerase Chain Reaction , Sequence Homology, Amino Acid , Smell , Synaptic Transmission/physiology , Synaptosomal-Associated Protein 25/physiology
9.
PLoS One ; 12(6): e0178518, 2017.
Article in English | MEDLINE | ID: mdl-28575017

ABSTRACT

The neurodegenerative disease glaucoma is one of the leading causes of blindness in the world. Glaucoma is characterized by progressive visual field loss caused by retinal ganglion cell (RGC) death. Both surgical glaucoma treatments and medications are available, however, they only halt glaucoma progression and are unable to reverse damage. Furthermore, many patients do not respond well to treatments. It is therefore important to better understand the mechanisms involved in glaucoma pathogenesis. Patients with Axenfeld-Rieger syndrome (ARS) offer important insight into glaucoma progression. ARS patients are at 50% risk of developing early onset glaucoma and respond poorly to treatments, even when surgical treatments are combined with medications. Mutations in the transcription factor FOXC1 cause ARS. Alterations in FOXC1 levels cause ocular malformations and disrupt stress response in ocular tissues, thereby contributing to glaucoma progression. In this study, using biochemical and molecular techniques, we show that FOXC1 regulates the expression of RAB3GAP1, RAB3GAP2 and SNAP25, three genes with central roles in both exocytosis and endocytosis, responsible for extracellular trafficking. FOXC1 positively regulates RAB3GAP1 and RAB3GAP2, while either increase or decrease in FOXC1 levels beyond its normal range results in decreased SNAP25. In addition, we found that FOXC1 regulation of RAB3GAP1, RAB3GAP2 and SNAP25 affects secretion of Myocilin (MYOC), a protein associated with juvenile onset glaucoma and steroid-induced glaucoma. The present work reveals that FOXC1 is an important regulator of exocytosis and establishes a new link between FOXC1 and MYOC-associated glaucoma.


Subject(s)
Cytoskeletal Proteins/metabolism , Exocytosis , Eye Proteins/metabolism , Forkhead Transcription Factors/physiology , Glycoproteins/metabolism , Synaptosomal-Associated Protein 25/physiology , rab3 GTP-Binding Proteins/physiology , Forkhead Transcription Factors/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Luciferases/genetics , RNA, Messenger/genetics , Synaptosomal-Associated Protein 25/genetics , Transcriptional Activation , rab3 GTP-Binding Proteins/genetics
10.
Proc Natl Acad Sci U S A ; 113(50): E8031-E8040, 2016 12 13.
Article in English | MEDLINE | ID: mdl-27911771

ABSTRACT

Synaptic soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) couple their stepwise folding to fusion of synaptic vesicles with plasma membranes. In this process, three SNAREs assemble into a stable four-helix bundle. Arguably, the first and rate-limiting step of SNARE assembly is the formation of an activated binary target (t)-SNARE complex on the target plasma membrane, which then zippers with the vesicle (v)-SNARE on the vesicle to drive membrane fusion. However, the t-SNARE complex readily misfolds, and its structure, stability, and dynamics are elusive. Using single-molecule force spectroscopy, we modeled the synaptic t-SNARE complex as a parallel three-helix bundle with a small frayed C terminus. The helical bundle sequentially folded in an N-terminal domain (NTD) and a C-terminal domain (CTD) separated by a central ionic layer, with total unfolding energy of ∼17 kBT, where kB is the Boltzmann constant and T is 300 K. Peptide binding to the CTD activated the t-SNARE complex to initiate NTD zippering with the v-SNARE, a mechanism likely shared by the mammalian uncoordinated-18-1 protein (Munc18-1). The NTD zippering then dramatically stabilized the CTD, facilitating further SNARE zippering. The subtle bidirectional t-SNARE conformational switch was mediated by the ionic layer. Thus, the t-SNARE complex acted as a switch to enable fast and controlled SNARE zippering required for synaptic vesicle fusion and neurotransmission.


Subject(s)
SNARE Proteins/chemistry , Amino Acid Sequence , Animals , Membrane Fusion , Mice , Microscopy, Atomic Force , Molecular Dynamics Simulation , Munc18 Proteins/chemistry , Munc18 Proteins/physiology , Optical Tweezers , Protein Conformation , Protein Domains , Protein Folding , Protein Stability , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/physiology , SNARE Proteins/genetics , SNARE Proteins/physiology , Synaptic Transmission/physiology , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/physiology , Vesicle-Associated Membrane Protein 2/chemistry , Vesicle-Associated Membrane Protein 2/physiology
11.
Curr Psychiatry Rep ; 18(8): 77, 2016 08.
Article in English | MEDLINE | ID: mdl-27371030

ABSTRACT

Schizophrenia is a serious psychiatric illness which is experienced by about 1 % of individuals worldwide and has a debilitating impact on perception, cognition, and social function. Over the years, several models/hypotheses have been developed which link schizophrenia to dysregulations of the dopamine, glutamate, and serotonin receptor pathways. An important segment of these pathways that have been extensively studied for the pathophysiology of schizophrenia is the presynaptic neurotransmitter release mechanism. This set of molecular events is an evolutionarily well-conserved process that involves vesicle recruitment, docking, membrane fusion, and recycling, leading to efficient neurotransmitter delivery at the synapse. Accumulated evidence indicate dysregulation of this mechanism impacting postsynaptic signal transduction via different neurotransmitters in key brain regions implicated in schizophrenia. In recent years, after ground-breaking work that elucidated the operations of this mechanism, research efforts have focused on the alterations in the messenger RNA (mRNA) and protein expression of presynaptic neurotransmitter release molecules in schizophrenia and other neuropsychiatric conditions. In this review article, we present recent evidence from schizophrenia human postmortem studies that key proteins involved in the presynaptic release mechanism are dysregulated in the disorder. We also discuss the potential impact of dysfunctional presynaptic neurotransmitter release on the various neurotransmitter systems implicated in schizophrenia.


Subject(s)
Brain/physiopathology , Schizophrenia/physiopathology , Schizophrenic Psychology , Synaptic Vesicles/physiology , Animals , Brain/pathology , Humans , Munc18 Proteins/physiology , Neurotransmitter Agents/metabolism , Qa-SNARE Proteins/physiology , R-SNARE Proteins/physiology , RNA, Messenger/genetics , Receptors, Presynaptic/physiology , SNARE Proteins/physiology , Schizophrenia/pathology , Signal Transduction/physiology , Synapsins/physiology , Synaptic Vesicles/genetics , Synaptophysin/physiology , Synaptosomal-Associated Protein 25/physiology
12.
Behav Brain Res ; 296: 393-400, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26300453

ABSTRACT

Autism spectrum disorder (ASD), attention-deficit hyperactivity disorder (ADHD), schizophrenia, Alzheimer's and Parkinson's disease are characterized by attentional deficits. In the present study we first applied the virtual object recognition test (VORT), where 3D objects were replaced with highly discriminated geometrical shapes and presented on two 3.5-inch widescreen displays, in different inbred mice strains (C57BL/6N, DBA/2J, BALB/cJ), in comparison with the standard object recognition test (NOR). In both NOR and VORT, there was a progressive decay of performance in terms of reduced discrimination index from 5 min to 72 h of inter-trial delay in all strains. However, BALB/cJ inbred mice showed a better long lasting performance than C57BL/6N and DBA/2J, when tested in NOR. In VORT, BALB/cJ showed the best performance. Total exploration time was always higher in BALB/cJ than C57BL/6N and DBA/2J mice. C57BL/6N were less explorative strain than DBA/2J and BALB/cJ mice. When VORT was applied to SNAP-25(+/-) mice, an impairment in both NOR and VORT was shown. However, when moving shapes were applied, these heterozygous mice improved their performance, suggesting that the introduction of motion is a strong cue that makes the task more valuable to study attention deficits. Taken together, these data indicate that VORT provides a useful and rapid tool to identify the attentional deficit in different inbred strains and genetically modified mice, enhancing the value of psychiatric mouse models.


Subject(s)
Attention/physiology , Behavior, Animal/physiology , Pattern Recognition, Visual/physiology , Synaptosomal-Associated Protein 25/physiology , Animals , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Models, Animal , Synaptosomal-Associated Protein 25/genetics
13.
BMC Musculoskelet Disord ; 15: 191, 2014 May 31.
Article in English | MEDLINE | ID: mdl-24885975

ABSTRACT

BACKGROUND: SNAP-25 protein is contributory to plasma membrane and synaptic vesicle fusions that are critical points in neurotransmission. SNAP-25 gene is associated with behavioral symptoms, personality and psychological disorders. In addition, SNAP-25 protein can be related to different neurotransmitter functions due to its association with vesicle membrane transition and fusion. This is important because neurologic, cognitive, and psychologic disorders in fibromyalgia syndrome (FMS) can be related to this function. This relationship may be enlightening for etiopathogenesis of FMS and treatment approaches. We aimed to study a SNAP-25 gene polymorphism, which is related to many psychiatric diseases, and FMS association in this prospective study. METHODS: We included 71 patients who were diagnosed according to new criteria and 57 matched healthy women in this study. Both groups were evaluated regarding age, height, weight, BMI, education level, marital and occupational status. A new diagnosis of FMS was made from criteria scoring, SF-36, Beck depression scale, and VAS that were applied to the patient group. SNAP-25 gene polymorphism and disease activity score correlations were compared. RESULTS: Mean age was 38±5,196 and 38.12±4.939 in patient and control groups, respectively (p=0.542). No significant difference was found between groups regarding age, height, weight, BMI, education level, marital or occupational status (p > 0.05). Ddel T/C genotype was significantly higher in the patient group (p = 0.009). MnlI gene polymorphism did not show a correlation with any score whereas a significant correlation was found between Ddel T/C genotype and Beck depression scale and VAS score (p < 0.05). CONCLUSION: FMS etiopathogenesis is not clearly known. Numerous neurologic, cognitive and psychological disorders were found during studies looking at cause. Our study showed increased SNAP-25 Ddel T/C genotype in FMS patients compared to the control group, which is related to behavioral symptoms, personality and psychological disorders in FMS patients.


Subject(s)
Central Nervous System Sensitization/genetics , Depressive Disorder/genetics , Fibromyalgia/genetics , Polymorphism, Single Nucleotide , Synaptosomal-Associated Protein 25/genetics , Adult , Case-Control Studies , Female , Fibromyalgia/psychology , Humans , Neural Conduction/physiology , Pain Measurement , Symptom Assessment , Synaptosomal-Associated Protein 25/physiology , Syndrome
14.
FEBS J ; 281(14): 3243-60, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24863955

ABSTRACT

Sympathetic neurons ramify to innervate multiple cells in target tissues. In compartmentalized cultures of rat superior cervical ganglion neurons, cleavage of synaptosomal-associated protein of Mr  = 25 000 (SNAP-25) in neurites exposed to botulinum neurotoxin type A (BoNT/A) arrested their growth and collapsed interstitial branches, but this required large, nonclinical doses. A protease-inactive mutant proved ineffective, confirming involvement of SNAP-25 in neurite extension and arborization. BoNT/C1 acted like BoNT/A, but BoNT/E caused only mild inhibition, likely due to transient SNAP-25 proteolysis. Near-total lack of susceptibility to BoNT/B or BoNT/D revealed that vesicle-associated membrane protein (VAMPs) isoforms 1-3 are not essential. Neurite length was not reduced when either BoNT/A or BoNT/C1 was applied to the somata, with no detrimental effect on neuron viability being observed. Treatments that protect cells from deprivation of nerve growth factor failed to prevent the toxin-induced loss of neurites. Inactivation of SNAP-25 caused the accumulation at neurite branch sites of Golgi-derived organelles labelled with N-(4,4-difluoro-5,7-dimethyl-4-bora-3a,4a-diaza-s-indacene-3-pentanoyl)-sphingosine conjugated to bovine serum albumin, prior to the collapse of arbors. Notably, neurite growth was ~ 1000-fold less susceptible to BoNT/A than cholinergic transmission in these neurons. Accordingly, a BoNT/A acceptor synaptic vesicle protein 2 (SV2) was found to be colocalized with VAMP 1-3, but not with VAMP 7, which is implicated in the growth of neurites. In conclusion, neurites depend on SNAP-25 for extension but this is quite resistant to BoNT/A, possibly, because of a low density of SV2 at growth sites that are distant from the highly susceptible regions of neurotransmitter release.


Subject(s)
Golgi Apparatus/physiology , Neurites/physiology , Neurons/physiology , Animals , Botulinum Toxins/metabolism , Botulinum Toxins, Type A , Cells, Cultured , Golgi Apparatus/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurons/cytology , Rats , Superior Cervical Ganglion/cytology , Synaptic Transmission , Synaptosomal-Associated Protein 25/physiology
15.
Neuron ; 80(4): 934-46, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24210904

ABSTRACT

Reelin is a glycoprotein that is critical for proper layering of neocortex during development as well as dynamic regulation of glutamatergic postsynaptic signaling in mature synapses. Here, we show that Reelin also acts presynaptically, resulting in robust rapid enhancement of spontaneous neurotransmitter release without affecting properties of evoked neurotransmission. This effect of Reelin requires a modest but significant increase in presynaptic Ca(2+) initiated via ApoER2 signaling. The specificity of Reelin action on spontaneous neurotransmitter release is encoded at the level of vesicular SNARE machinery as it requires VAMP7 and SNAP-25 but not synaptobrevin2, VAMP4, or vti1a. These results uncover a presynaptic regulatory pathway that utilizes the heterogeneity of synaptic vesicle-associated SNAREs and selectively augments action potential-independent neurotransmission.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Extracellular Matrix Proteins/physiology , Nerve Tissue Proteins/physiology , R-SNARE Proteins/physiology , Serine Endopeptidases/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/physiology , Animals , Apolipoproteins E/metabolism , Calcium Signaling/physiology , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Electrophysiological Phenomena , Excitatory Postsynaptic Potentials/physiology , Extracellular Matrix Proteins/genetics , Female , Lentivirus/genetics , Male , Mice , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Phosphatidylinositol 3-Kinases/metabolism , R-SNARE Proteins/genetics , Rats , Rats, Sprague-Dawley , Reelin Protein , SNARE Proteins/metabolism , Serine Endopeptidases/genetics , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/physiology , Tetrodotoxin/pharmacology
16.
J Physiol ; 591(16): 3935-47, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23774277

ABSTRACT

The group II metabotropic glutamate receptors (group II mGluRs) have emerged as the new drug targets for the treatment of mental disorders like schizophrenia. To understand the potential mechanisms underlying the antipsychotic effects of group II mGluRs, we examined their impact on NMDA receptors (NMDARs), since NMDAR hypofunction has been implicated in schizophrenia. The activation of group II mGluRs caused a significant enhancement of NMDAR currents in cortical pyramidal neurons, which was associated with increased NMDAR surface expression and synaptic localization. We further examined whether these effects of group II mGluRs are through the regulation of NMDAR exocytosis via SNARE proteins, a family of proteins involved in vesicle fusion. We found that the enhancing effect of APDC, a selective agonist of group II mGluRs, on NMDAR currents was abolished when botulinum toxin was delivered into the recorded neurons to disrupt the SNARE complex. Inhibiting the function of two key SNARE proteins, SNAP-25 and syntaxin 4, also eliminated the effect of APDC on NMDAR currents. Moreover, the application of APDC increased the activity of Rab4, a small Rab GTPase mediating fast recycling from early endosomes to the plasma membrane, and enhanced the interaction between syntaxin 4 and Rab4. Knockdown of Rab4 or expression of dominant-negative Rab4 attenuated the effect of APDC on NMDAR currents. Taken together, these results have identified key molecules involved in the group II mGluR-induced potentiation of NMDAR exocytosis and function.


Subject(s)
Frontal Lobe/physiology , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/physiology , SNARE Proteins/physiology , Animals , Frontal Lobe/embryology , In Vitro Techniques , Rats , Synaptosomal-Associated Protein 25/physiology
17.
PLoS One ; 8(2): e57080, 2013.
Article in English | MEDLINE | ID: mdl-23451149

ABSTRACT

SNAP-25 is a core component of the trimeric SNARE complex mediating vesicle exocytosis during membrane addition for neuronal growth, neuropeptide/growth factor secretion, and neurotransmitter release during synaptic transmission. Here, we report a novel microRNA mechanism of SNAP-25 regulation controlling motor neuron development, neurosecretion, synaptic activity, and movement in zebrafish. Loss of miR-153 causes overexpression of SNAP-25 and consequent hyperactive movement in early zebrafish embryos. Conversely, overexpression of miR-153 causes SNAP-25 down regulation resulting in near complete paralysis, mimicking the effects of treatment with Botulinum neurotoxin. miR-153-dependent changes in synaptic activity at the neuromuscular junction are consistent with the observed movement defects. Underlying the movement defects, perturbation of miR-153 function causes dramatic developmental changes in motor neuron patterning and branching. Together, our results indicate that precise control of SNAP-25 expression by miR-153 is critically important for proper neuronal patterning as well as neurotransmission.


Subject(s)
MicroRNAs/physiology , Motor Neurons/cytology , Synaptic Transmission/physiology , Synaptosomal-Associated Protein 25/physiology , Animals , Base Sequence , Exocytosis/physiology , Green Fluorescent Proteins/genetics , MicroRNAs/genetics , Sequence Homology, Amino Acid , Signal Transduction/physiology , Zebrafish/embryology
18.
Neuropharmacology ; 66: 31-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22626985

ABSTRACT

While postsynaptic ionotropic and metabotropic glutamate receptors have received the lions share of attention in studies of long-term activity-dependent synaptic plasticity, it is becoming clear that presynaptic metabotropic glutamate receptors play critical roles in both short-term and long-term plasticity of vesicular transmitter release, and that they act both at the level of voltage-dependent calcium channels and directly on proteins of the vesicular release machinery. Activation of G protein-coupled receptors can transiently inhibit vesicular release through the release of Gßγ which binds to both voltage-dependent calcium channels to reduce calcium influx, and directly to the C-terminus region of the SNARE protein SNAP-25. Our recent work has revealed that the binding of Gßγ to SNAP-25 is necessary, but not sufficient, to elicit long-term depression (LTD) of vesicular glutamate release, and that the concomitant release of Gα(i) and the second messenger nitric oxide are also necessary steps in the presynaptic LTD cascade. Here, we review the current state of knowledge of the molecular steps mediating short-term and long-term plasticity of vesicular release at glutamatergic synapses, and the many gaps that remain to be addressed. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.


Subject(s)
Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Receptors, Metabotropic Glutamate/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Animals , Brain/growth & development , Brain/metabolism , Calcium Channels/physiology , Neural Networks, Computer , Receptors, G-Protein-Coupled/physiology , Synaptosomal-Associated Protein 25/metabolism , Synaptosomal-Associated Protein 25/physiology
19.
Cell Mol Life Sci ; 70(1): 153-65, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22890573

ABSTRACT

Changes in the structure and number of synapses modulate learning, memory and cognitive disorders. Ubiquitin-mediated protein modification is a key mechanism for regulating synaptic activity, though the precise control of this process remains poorly understood. RING finger protein 13 (RNF13) is a recently identified E3 ubiquitin ligase, and its in vivo function remains completely unknown. We show here that genetic deletion of RNF13 in mice leads to a significant deficit in spatial learning as determined by the Morris water maze test and Y-maze learning test. At the ultrastructral level, the synaptic vesicle density was decreased and the area of the active zone was increased at hippocampal synapses of RNF13-null mice compared with those of wild-type littermates. We found no change in the levels of SNARE (soluble N-ethylmaleimide-sensitive factor-attachment protein receptor) complex proteins in the hippocampus of RNF13-null mice, but impaired SNARE complex assembly. RNF13 directly interacted with snapin, a SNAP-25-interacting protein. Interestingly, snapin was ubiquitinated by RNF13 via the lysine-29 conjugated polyubiquitin chain, which in turn promoted the association of snapin with SNAP-25. Consistently, we found an attenuated interaction between snapin and SNAP-25 in the RNF13-null mice. Therefore, these results suggest that RNF13 is involved in the regulation of the SNARE complex, which thereby controls synaptic function.


Subject(s)
SNARE Proteins/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Hippocampus/metabolism , Hippocampus/ultrastructure , Maze Learning/physiology , Mice , Mice, Knockout , Synapses/genetics , Synapses/ultrastructure , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/metabolism , Synaptosomal-Associated Protein 25/physiology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Vesicular Transport Proteins/metabolism
20.
Int J Biochem Cell Biol ; 44(12): 2288-98, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23010346

ABSTRACT

SNARE complex mediates cellular membrane fusion events essential for neurotransmitter release and synaptogenesis. SNAP25, a member of the SNARE proteins, plays critical roles during the development of the central nervous system via regulation by alternative splicing and protein kinase phosphorylation. To date, little information is available regarding the protein in the spinal cord regeneration, especially for the postnatal highly expressed isoform SNAP25b. In the present study, we characterized gecko SNAP25b, which shared high identity with those of other vertebrates. Expression of gecko SNAP25b was temporally upregulated in both neurons of spinal cord and forming ependymal tube following tail amputation, coinciding with the occurrence of regenerate re-innervation. Overexpression of gecko wild type SNAP25b in the SH-SY5Y and undifferentiated PC12 cells promoted the elongation and outgrowth of neurites, while mutant constructs at Serine(187) resulted in differential effects for which S187A had a promoting role. Knockdown of endogenous SNAP25b affected the formation of neurites, which could be rescued by overexpression of SNAP25b. FM1-43 staining revealed that transfection of S187E mutant construct reduced the recruitment of vesicles. In addition, transfection of gecko SNAP25b in the astrocyte, which is absent from neuronal specific VAMP2, was capable of enhancing process elongation, indicating a potential for various alternative protein combinations. Taken together, our data suggest that gecko SNAP25b is involved in spinal cord regeneration by promoting outgrowth and elongation of neurites in a more extensive protein binding manner.


Subject(s)
Nerve Regeneration , Neurites/physiology , Reptilian Proteins/physiology , Spinal Cord/physiology , Synaptosomal-Associated Protein 25/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Shape , Cloning, Molecular , Conserved Sequence , Gene Expression , Gene Knockdown Techniques , Humans , Lizards , Molecular Sequence Data , Neurites/metabolism , Organ Specificity , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , RNA, Small Interfering/genetics , Reptilian Proteins/genetics , Reptilian Proteins/metabolism , SNARE Proteins/metabolism , Spinal Cord/metabolism , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL