Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Biochem Biophys Res Commun ; 514(1): 105-111, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31027731

ABSTRACT

Assembly of neuronal SNARE protein complexes is essential for fusion of synaptic vesicles with the presynaptic plasma membrane, which releases neurotransmitters into the synaptic cleft and mediates neurotransmission. However, despite the potential of pharmacological regulation of this process for the treatment of various neurological disorders, only a few reagents, including botulinum neurotoxins, are currently available. Here, we report that buforin-1, an antimicrobial peptide from the Asian toad Bufo gargarizans, inhibits neuronal SNARE complex assembly, resulting in neuronal SNARE-mediated membrane fusion in vitro via its direct association with neuronal t-SNAREs syntaxin-1 and SNAP-25. Consistently, buforin-1 significantly inhibited neuronal-SNARE-mediated exocytosis in PC-12 cells. Thus, buforin-1 has potential for the treatment of neurological disorders caused by dysregulated neurotransmission.


Subject(s)
Membrane Fusion/drug effects , Neurons/drug effects , Proteins/pharmacology , SNARE Proteins/antagonists & inhibitors , Animals , Bufonidae , Cell Line , Exocytosis/drug effects , Male , Mice, Inbred C57BL , Neurons/metabolism , Rats , SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism
2.
Mol Cell Biochem ; 442(1-2): 97-109, 2018 May.
Article in English | MEDLINE | ID: mdl-29019108

ABSTRACT

Synaptotagmin 1 (Syt1) is the Ca2+ sensor protein with an essential role in neurotransmitter release. Since the wrinkle formation is due to the excessive muscle fiber stimulation in the face, a helpful stratagem to diminish the wrinkle line intenseness is to weaken the innervating neuron activity through Syt1 inhibition which is one of the possible therapeutic strategies against wrinkles. Recently, experimental evidence showed that botox-like peptides, which are typically used as SNARE modulators, may inhibit Syt1. In this work, we applied molecular modeling to (1) characterize the structural framework and (2) define the atomistic information of the factors for the inhibition mechanism. The modeling identified the plausible binding cleft able to efficiently bind all botox-like peptides. The MD simulations revealed that all peptides induced significant Syt1 rigidity by binding in the cleft of the C2A-C2B interface. The consequence of this binding event is the suppression of the protein motion associated with conformational change of Syt1 from the closed form to the open form. On this basis, this finding may therefore be of subservience for the advancement of novel botox-like molecules for the therapeutic treatment of wrinkle, targeting and modulating the function of Syt1.


Subject(s)
Molecular Docking Simulation , Peptides/chemistry , SNARE Proteins , Synaptotagmin I/chemistry , Humans , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/chemistry
3.
Proc Natl Acad Sci U S A ; 110(41): E3955-64, 2013 Oct 08.
Article in English | MEDLINE | ID: mdl-24065831

ABSTRACT

Atrial myocytes are continuously exposed to mechanical forces including shear stress. However, in atrial myocytes, the effects of shear stress are poorly understood, particularly with respect to its effect on ion channel function. Here, we report that shear stress activated a large outward current from rat atrial myocytes, with a parallel decrease in action potential duration. The main ion channel underlying the increase in current was found to be Kv1.5, the recruitment of which could be directly observed by total internal reflection fluorescence microscopy, in response to shear stress. The effect was primarily attributable to recruitment of intracellular pools of Kv1.5 to the sarcolemma, as the response was prevented by the SNARE protein inhibitor N-ethylmaleimide and the calcium chelator BAPTA. The process required integrin signaling through focal adhesion kinase and relied on an intact microtubule system. Furthermore, in a rat model of chronic hemodynamic overload, myocytes showed an increase in basal current despite a decrease in Kv1.5 protein expression, with a reduced response to shear stress. Additionally, integrin beta1d expression and focal adhesion kinase activation were increased in this model. This data suggests that, under conditions of chronically increased mechanical stress, the integrin signaling pathway is overactivated, leading to increased functional Kv1.5 at the membrane and reducing the capacity of cells to further respond to mechanical challenge. Thus, pools of Kv1.5 may comprise an inducible reservoir that can facilitate the repolarization of the atrium under conditions of excessive mechanical stress.


Subject(s)
Heart Atria/cytology , Kv1.5 Potassium Channel/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction/physiology , Stress, Physiological/physiology , Analysis of Variance , Animals , Biomechanical Phenomena , Blotting, Western , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Ethylmaleimide/pharmacology , Fluorescent Antibody Technique , Integrin beta1/metabolism , Male , Microscopy, Fluorescence , Models, Biological , Patch-Clamp Techniques , Rats , Rats, Wistar , SNARE Proteins/antagonists & inhibitors , Sarcolemma/metabolism , Shear Strength
4.
Biochemistry ; 54(9): 1831-40, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25714795

ABSTRACT

Alzheimer's disease (AD) and Parkinson's disease (PD) are caused by ß-amyloid (Aß) and α-synuclein (αS), respectively. Ample evidence suggests that these two pathogenic proteins are closely linked and have a synergistic effect on eliciting neurodegenerative disorders. However, the pathophysiological consequences of Aß and αS coexistence are still elusive. Here, we show that large-sized αS oligomers, which are normally difficult to form, are readily generated by Aß42-seeding and that these oligomers efficiently hamper neuronal SNARE-mediated vesicle fusion. The direct binding of the Aß-seeded αS oligomers to the N-terminal domain of synaptobrevin-2, a vesicular SNARE protein, is responsible for the inhibition of fusion. In contrast, large-sized Aß42 oligomers (or aggregates) or the products of αS incubated without Aß42 have no effect on vesicle fusion. These results are confirmed by examining PC12 cell exocytosis. Our results suggest that Aß and αS cooperate to escalate the production of toxic oligomers, whose main toxicity is the inhibition of vesicle fusion and consequently prompts synaptic dysfunction.


Subject(s)
Amyloid beta-Peptides/physiology , Cytoplasmic Vesicles/physiology , Membrane Fusion , SNARE Proteins/antagonists & inhibitors , alpha-Synuclein/physiology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Exocytosis/genetics , Humans , Membrane Fusion/genetics , PC12 Cells , Protein Binding/genetics , Protein Multimerization/physiology , Rats , SNARE Proteins/metabolism , Synapses/genetics , Synapses/metabolism , Transfection , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
5.
Blood ; 117(4): 1425-35, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21063020

ABSTRACT

Vascular endothelial growth factor receptor 2 (VEGFR2) plays a key role in physiologic and pathologic angiogenesis. Plasma membrane (PM) levels of VEGFR2 are regulated by endocytosis and secretory transport through the Golgi apparatus. To date, the mechanism whereby the VEGFR2 traffics through the Golgi apparatus remains incompletely characterized. We show in human endothelial cells that binding of VEGF to the cell surface localized VEGFR2 stimulates exit of intracellular VEGFR2 from the Golgi apparatus. Brefeldin A treatment reduced the level of surface VEGFR2, confirming that VEGFR2 traffics through the Golgi apparatus en route to the PM. Mechanistically, we show that inhibition of syntaxin 6, a Golgi-localized target membrane-soluble N-ethylmaleimide attachment protein receptor (t-SNARE) protein, interferes with VEGFR2 trafficking to the PM and facilitates lysosomal degradation of the VEGFR2. In cell culture, inhibition of syntaxin 6 also reduced VEGF-induced cell proliferation, cell migration, and vascular tube formation. Furthermore, in a mouse ear model of angiogenesis, an inhibitory form of syntaxin 6 reduced VEGF-induced neovascularization and permeability. Our data demonstrate the importance of syntaxin 6 in the maintenance of cellular VEGFR2 levels, and suggest that the inhibitory form of syntaxin 6 has good potential as an antiangiogenic agent.


Subject(s)
Golgi Apparatus/metabolism , Neovascularization, Physiologic/physiology , Qa-SNARE Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Nude , Neovascularization, Physiologic/drug effects , Protein Transport/drug effects , Qa-SNARE Proteins/antagonists & inhibitors , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/genetics , SNARE Proteins/metabolism , SNARE Proteins/physiology , Transfection , Vascular Endothelial Growth Factor A/pharmacology
6.
J Immunol ; 187(1): 391-400, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21642540

ABSTRACT

The role of exocytosis in the human neutrophil respiratory burst was determined using a fusion protein (TAT-SNAP-23) containing the HIV transactivator of transcription (TAT) cell-penetrating sequence and the N-terminal SNARE domain of synaptosome-associated protein-23 (SNAP-23). This agent inhibited stimulated exocytosis of secretory vesicles and gelatinase and specific granules but not azurophil granules. GST pulldown showed that TAT-SNAP-23 bound to the combination of vesicle-associated membrane protein-2 and syntaxin-4 but not to either individually. TAT-SNAP-23 reduced phagocytosis-stimulated hydrogen peroxide production by 60% without affecting phagocytosis or generation of HOCl within phagosomes. TAT-SNAP-23 had no effect on fMLF-stimulated superoxide release but significantly inhibited priming of this response by TNF-α and platelet-activating factor. Pretreatment with TAT-SNAP-23 inhibited the increase in plasma membrane expression of gp91(phox) in TNF-α-primed neutrophils, whereas TNF-α activation of ERK1/2 and p38 MAPK was not affected. The data demonstrate that neutrophil granule exocytosis contributes to phagocytosis-induced respiratory burst activity and plays a critical role in priming of the respiratory burst by increasing expression of membrane components of the NADPH oxidase.


Subject(s)
Cytoplasmic Granules/immunology , Exocytosis/immunology , Neutrophil Activation/immunology , Respiratory Burst/immunology , Apoptosis/genetics , Apoptosis/immunology , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Exocytosis/genetics , Gene Products, tat/antagonists & inhibitors , Gene Products, tat/genetics , Gene Products, tat/metabolism , HIV-1/immunology , Humans , Neutrophil Activation/genetics , Phagocytosis/genetics , Phagocytosis/immunology , Platelet Activating Factor/physiology , Protein Structure, Tertiary/genetics , Qb-SNARE Proteins/antagonists & inhibitors , Qb-SNARE Proteins/genetics , Qb-SNARE Proteins/metabolism , Qc-SNARE Proteins/antagonists & inhibitors , Qc-SNARE Proteins/genetics , Qc-SNARE Proteins/metabolism , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Respiratory Burst/genetics , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/genetics , SNARE Proteins/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology
7.
Planta Med ; 78(3): 233-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22109835

ABSTRACT

Most cosmetic and therapeutic applications of Clostridium botulinum neurotoxin (BoNT) are related to muscle paralysis caused by the blocking of neurotransmitter release at the neuromuscular junction. BoNT specifically cleaves SNARE proteins at the nerve terminal and impairs neuroexocytosis. Recently, we have shown that several polyphenols inhibit neurotransmitter release from neuronal PC12 cells by interfering with SNARE complex formation. Based on our previous result, we report here that myricetin, delphinidin, and cyanidin indeed paralyze muscle by inhibiting acetylcholine release at the neuromuscular junction. While the effect of myricetin on muscle paralysis was modest compared to BoNT/A, myricetin exhibited a shorter response time than BoNT/A. Intraperitoneally-injected myricetin at an extreme dose of 1000 mg/kg did not induce death of mice, alleviating the safety issue. Thus, these polyphenols might be useful in treating various human hypersecretion diseases for which BoNT/A has been the only option of choice.


Subject(s)
Botulinum Toxins, Type A/pharmacology , Cosmetics/pharmacology , Neuromuscular Blocking Agents/pharmacology , Polyphenols/pharmacology , SNARE Proteins/antagonists & inhibitors , Animals , Anthocyanins/pharmacology , Female , Flavonoids/pharmacology , Humans , Mice , Phytotherapy , Plant Extracts/pharmacology , SNARE Proteins/metabolism
8.
Pharm Biol ; 50(9): 1157-67, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22881141

ABSTRACT

CONTEXT: Botulinum neurotoxins (BoNTs) are popularly used to treat various diseases and for cosmetic purposes. They act by blocking neurotransmission through specific cleavage of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins. Recently, several polyphenols were shown to interfere with SNARE complex formation by wedging into the hydrophobic core interface, thereby leading to reduced neuroexocytosis. OBJECTIVE: In order to find industrially-viable plant extract that functions like BoNT, 71 methanol extracts of flowers were screened and BoNT-like activity of selected extract was evaluated. MATERIALS AND METHODS: After evaluating the inhibitory effect of 71 flower methanol extracts on SNARE complex formation, seven candidates were selected and they were subjected to SNARE-driven membrane fusion assay. Neurotransmitter release from neuronal PC12 cells and SNARE complex formation inside the cell was also evaluated. Finally, the effect of one selected extract on muscle contraction and digit abduction score was determined. RESULTS: The extract of Potentilla chinensis Ser. (Rosaceae)(Chinese cinquefoil) flower inhibited neurotransmitter release from neuronal PC12 cells by approximately 90% at a concentration of 10 µg/mL. The extract inhibited neuroexocytosis by interfering with SNARE complex formation inside cells. It reduced muscle contraction of phrenic nerve-hemidiaphragm by approximately 70% in 60 min, which is comparable to the action of the Ca²âº-channel blocker verapamil and BoNT type A. DISCUSSION AND CONCLUSION: While BoNT blocks neuroexocytosis by cleaving SNARE proteins, the Potentilla chinensis extract exhibited the same activity by inhibiting SNARE complex formation. The extract paralyzed muscle as efficiently as BoNT, suggesting the potential versatility in cosmetics and therapeutics.


Subject(s)
Membrane Fusion/drug effects , Muscle Contraction/drug effects , Neuromuscular Agents/pharmacology , Neurons/drug effects , Plant Extracts/pharmacology , Potentilla/chemistry , SNARE Proteins/antagonists & inhibitors , Animals , Botulinum Toxins/adverse effects , Botulinum Toxins/pharmacology , Drug Discovery , Exocytosis/drug effects , Female , Flowers/chemistry , Lower Extremity , Mice , Mice, Inbred ICR , Muscle, Skeletal/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neuromuscular Agents/adverse effects , Neurons/metabolism , Norepinephrine/metabolism , PC12 Cells , Plant Extracts/adverse effects , Rats , SNARE Proteins/metabolism , Synaptic Transmission/drug effects
9.
J Neurosci ; 30(10): 3624-33, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20219996

ABSTRACT

Glycogen synthase kinase-3 (GSK-3), a Ser/Thr protein kinase abundantly expressed in neurons, plays diverse functions in physiological and neurodegenerative conditions. Our recent study shows that upregulation of GSK-3 suppresses long-term potentiation and presynaptic release of glutamate; however, the underlying mechanism is elusive. Here, we show that activation of GSK-3beta retards the synaptic vesicle exocytosis in response to membrane depolarization. Using calcium imaging, whole-cell patch-clamp, as well as specific Ca(2+) channel inhibitors, we demonstrate that GSK-3beta phosphorylates the intracellular loop-connecting domains II and III (L(II-III)) of P/Q-type Ca(2+) channels, which leads to a decrease of intracellular Ca(2+) rise through the P/Q-type voltage-dependent calcium channel. To further illustrate the mechanisms of GSK-3beta's action, we show that activation of GSK-3beta interferes with the formation of the soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) complex through: (1) weakening the association of synaptobrevin with SNAP25 and syntaxin; (2) reducing the interactions among the phosphorylated L(II-III) and synaptotagmin, SNAP25, and syntaxin; and (3) inhibiting dissociation of synaptobrevin from synaptophysin I. These results indicate that GSK-3beta negatively regulates synaptic vesicle fusion events via interfering with Ca(2+)-dependent SNARE complex formation.


Subject(s)
Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/metabolism , Glycogen Synthase Kinase 3/physiology , Presynaptic Terminals/metabolism , SNARE Proteins/antagonists & inhibitors , Synaptic Vesicles/metabolism , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , Exocytosis/physiology , Glycogen Synthase Kinase 3/biosynthesis , Glycogen Synthase Kinase 3 beta , Molecular Sequence Data , Neural Inhibition/physiology , Phosphorylation , Rats , SNARE Proteins/biosynthesis
10.
Biochem J ; 429(2): 391-401, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20450495

ABSTRACT

VLDLs (very-low-density lipoproteins) are synthesized in the liver and play an important role in the pathogenesis of atherosclerosis. Following their biogenesis in hepatic ER (endoplasmic reticulum), nascent VLDLs are exported to the Golgi which is a physiologically regulatable event. We have previously shown that a unique ER-derived vesicle, the VTV (VLDL-transport vesicle), mediates the targeted delivery of VLDL to the Golgi lumen. Because VTVs are different from other ER-derived transport vesicles in their morphology and biochemical composition, we speculated that a distinct set of SNARE (soluble N-ethylmaleimide-sensitive factor-attachment protein receptor) proteins would form a SNARE complex which would eventually facilitate the docking/fusion of VTVs with Golgi. Our results show that Sec22b is concentrated in VTVs as compared with the ER. Electron microscopic results show that Sec22b co-localizes with p58 and Sar1 on the VTV surface. Pre-treatment of VTV with antibodies against Sec22b inhibited VTV-Golgi fusion, indicating its role as a v-SNARE (vesicle SNARE). To isolate the SNARE complex, we developed an in vitro docking assay in which VTVs were allowed to dock with the Golgi, but fusion was prevented to stabilize the SNARE complex. After the docking reaction, VTV-Golgi complexes were collected, solubilized in 2% Triton X-100 and the SNARE complex was co-immunoprecipitated using anti-Sec22b or GOS28 antibodies. A approximately 110 kDa complex was identified in non-boiled samples that was dissociated upon boiling. The components of the complex were identified as Sec22b, syntaxin 5, rBet1 and GOS28. Antibodies against each SNARE component significantly inhibited VTV-Golgi fusion. We conclude that the SNARE complex required for VTV-Golgi fusion is composed of Sec22b, syntaxin 5, rBet1 and GOS28.


Subject(s)
Golgi Apparatus/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , SNARE Proteins/metabolism , Animals , Biological Transport, Active , Endoplasmic Reticulum/metabolism , In Vitro Techniques , Membrane Fusion/physiology , Qa-SNARE Proteins/metabolism , Qb-SNARE Proteins/metabolism , R-SNARE Proteins/metabolism , Rats , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/immunology , Transport Vesicles/metabolism
11.
Rev Neurol (Paris) ; 167(1): 46-50, 2011 Jan.
Article in French | MEDLINE | ID: mdl-21194720

ABSTRACT

Botulinum toxin type A (BTX-A) is a potent neurotoxin that blocks acetylcholine release from presynaptic nerve terminals by cleaving the SNARE complex. BTX-A has been reported to have analgesic effects independent of its action on muscle tone. The most robust results have been observed in patients with neuropathic pain. Neuropathic pain due to peripheral lesions has been the most widely studied. BTX-A has shown its efficacy on pain and allodynia in various animal models of inflammatory neuropathic pain. The only randomized, double-blind, placebo-controlled trial in patients with focal painful neuropathies due to nerve trauma or postherpetic neuralgia demonstrated significant effects on average pain intensity from 2 weeks after the injections to 14 weeks. Most patients reported pain during the injections, but there were no further local or systemic side effects. The efficacy of BTX-A in painful peripheral neuropathies has been more recently studied. Results were positive in the only study in an animal model of peripheral neuropathy. One study in patients with diabetic painful peripheral neuropathy demonstrated a significant decrease in Visual Analog Scale. In conclusion, one session of multiple intradermal injection of BTX-A produces long-lasting analgesic effects in patients with focal painful neuropathies and diabetic neuropathic pain, and is particularly well tolerated. The findings are consistent with a reduction of peripheral sensitisation, the place of a possible central effect remaining to define. Further studies are needed to assess some important issues, i.e. BTX-A efficacy in patients with small fiber neuropathies and the relevance of early and repeated injections. Future studies could also provide valuable insights into pathophysiology of neuropathic pain.


Subject(s)
Botulinum Toxins, Type A/therapeutic use , Neuralgia/drug therapy , Animals , Botulinum Toxins, Type A/pharmacology , Controlled Clinical Trials as Topic , Diabetic Neuropathies/drug therapy , Drug Evaluation, Preclinical , Humans , Injections, Intradermal , Neuritis/drug therapy , Rats , SNARE Proteins/antagonists & inhibitors
12.
Sci Rep ; 11(1): 10955, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040104

ABSTRACT

The primary hallmark of Parkinson's disease (PD) is the generation of Lewy bodies of which major component is α-synuclein (α-Syn). Because of increasing evidence of the fundamental roles of α-Syn oligomers in disease progression, α-Syn oligomers have become potential targets for therapeutic interventions for PD. One of the potential toxicities of α-Syn oligomers is their inhibition of SNARE-mediated vesicle fusion by specifically interacting with vesicle-SNARE protein synaptobrevin-2 (Syb2), which hampers dopamine release. Here, we show that α-Syn monomers and oligomers cooperatively inhibit neuronal SNARE-mediated vesicle fusion. α-Syn monomers at submicromolar concentrations increase the fusion inhibition by α-Syn oligomers. This cooperative pathological effect stems from the synergically enhanced vesicle clustering. Based on this cooperative inhibition mechanism, we reverse the fusion inhibitory effect of α-Syn oligomers using small peptide fragments. The small peptide fragments, derivatives of α-Syn, block the binding of α-Syn oligomers to Syb2 and dramatically reverse the toxicity of α-Syn oligomers in vesicle fusion. Our findings demonstrate a new strategy for therapeutic intervention in PD and related diseases based on this specific interaction of α-Syn.


Subject(s)
Membrane Fusion/drug effects , SNARE Proteins/antagonists & inhibitors , alpha-Synuclein/pharmacology , Amino Acid Sequence , Amino Acid Substitution , Dopamine/metabolism , Dopamine/pharmacology , Drug Evaluation, Preclinical , Liposomes , Membrane Lipids/metabolism , Models, Molecular , Mutation, Missense , Peptide Fragments/pharmacology , Point Mutation , Protein Binding , Protein Multimerization , Proteolipids/chemistry , Recombinant Fusion Proteins/pharmacology , SNARE Proteins/physiology , Vesicle-Associated Membrane Protein 2/antagonists & inhibitors , Vesicle-Associated Membrane Protein 2/physiology , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/toxicity
13.
J Neurosci ; 29(39): 12292-301, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19793988

ABSTRACT

The soluble N-ethylmaleimide-sensitive fusion (NSF) attachment protein (SNAP) receptor (SNARE) protein syntaxin 1A forms nano-sized clusters (membrane rafts) on the plasma membrane (PM) that are in equilibrium with freely diffusing syntaxin molecules. SNARE-complex formation between syntaxin 1A and SNAP-25 (synaptosome-associated protein of 25 kDa) on the PM and synaptobrevin 2 on the vesicles (trans-SNAREs) is crucial for vesicle priming and fusion. This process might be impeded by the spontaneous accumulation of non-fusogenic cis-SNARE complexes formed when all three SNARE proteins reside on the PM. We investigated the kinetics of cis-SNARE complex assembly and disassembly and both exhibited biphasic behavior. The experimental measurements were analyzed through integration of differential rate equations pertinent to the reaction mechanism and through the application of a heuristic search for time constants and concentrations using a genetic algorithm. Reconstruction of the measurements necessitated the partitioning of syntaxin into two phases that might represent the syntaxin clusters and free syntaxin outside the clusters. The analysis suggests that most of the syntaxin in the clusters is concentrated in a nonreactive form. Consequently, cis-SNARE complex assembly in the clusters is substantially slower than outside the rafts. Interestingly, the clusters also mediate efficient disassembly of cis-SNARE complexes possibly attributable to the high local concentration of complexes in the clusters area that allows efficient disassembly by the enzymatic reaction of NSF.


Subject(s)
Membrane Microdomains/metabolism , Qa-SNARE Proteins/metabolism , Animals , Energy Metabolism/genetics , Energy Metabolism/physiology , Intracellular Membranes/metabolism , Intracellular Membranes/physiology , Kinetics , Membrane Microdomains/physiology , Multigene Family/physiology , PC12 Cells , Protein Binding/physiology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/physiology , Rats , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/biosynthesis , SNARE Proteins/metabolism
14.
Biol Reprod ; 81(2): 396-405, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19369646

ABSTRACT

Acrosomal exocytosis in mammalian sperm is a regulated secretion with unusual characteristics. One of its most striking features is the loss of the outer acrosomal membrane and the overlying plasma membrane as hybrid vesicles. We have reported previously in human sperm that by preventing the release of calcium from the acrosome, the exocytic process can be arrested at a stage where SNARE proteins are assembled in loose trans complexes. Transmission electron micrographs of sperm at this stage showed that the acrosomes were profusely swollen, with deep invaginations of the outer acrosomal membrane. The protruding edges of these invaginations were tightly apposed (i.e., docked) to the plasma membrane. Docking was prevented when streptolysin O-permeabilized sperm were stimulated in the presence of tetanus toxin or botulinum neurotoxin C, two SNARE-specific proteases. We propose that SNAREs present in the plasma membrane interact with SNAREs in the protruding edge of cup-shaped invaginations of the outer acrosomal membrane to form trans complexes. Fusion pore opening and expansion in this ring of apposed membranes would generate the hybrid vesicles that are released during the acrosome reaction.


Subject(s)
Acrosome Reaction/physiology , Acrosome/ultrastructure , Cell Membrane/metabolism , SNARE Proteins/metabolism , Spermatozoa/physiology , Acrosome/physiology , Adult , Calcium Channel Blockers/pharmacology , Cell Membrane/ultrastructure , Chelating Agents/pharmacology , Enzyme Inhibitors/pharmacology , Exocytosis/drug effects , Exocytosis/physiology , Humans , Ionophores/pharmacology , Male , Membrane Fusion/drug effects , Membrane Fusion/physiology , Microscopy, Electron, Transmission , Models, Biological , Nuclear Envelope/metabolism , Nuclear Envelope/ultrastructure , Permeability , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , SNARE Proteins/antagonists & inhibitors , Spermatozoa/metabolism , Spermatozoa/ultrastructure , Surface Properties , Transport Vesicles/metabolism , Transport Vesicles/ultrastructure
15.
Biotechnol Lett ; 31(3): 361-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19023663

ABSTRACT

Neuronal soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) proteins mediate membrane fusion between synaptic vesicle and presynaptic membrane, resulting in neurotransmitter release. SNARE proteins are specific substrates of botulinum neurotoxins (BoNT) which are now widely used for therapeutic and cosmetic purposes. While BoNT blocks neuroexocytosis by cleaving SNAREs, inhibiting SNARE assembly process might exert the same effect on neurotransmission. In the present study, some extracts of 100 plants reduced neurotransmitter release by inhibiting SNARE complex formation in neuronal cells. The extracts effectively paralyzed muscle of rat phrenic nerve-hemidiaphragm preparation. Our results raise the possibility that SNARE folding inhibitors from natural resources might replace some special BoNT application fields.


Subject(s)
Exocytosis/drug effects , Neuromuscular Blocking Agents/pharmacology , Neurons/drug effects , Plant Extracts/pharmacology , SNARE Proteins/antagonists & inhibitors , Synaptic Transmission/drug effects , Animals , Diaphragm/drug effects , Paralysis , Phrenic Nerve/drug effects , Rats
16.
Nat Genet ; 51(10): 1475-1485, 2019 10.
Article in English | MEDLINE | ID: mdl-31548722

ABSTRACT

The mechanisms by which common risk variants of small effect interact to contribute to complex genetic disorders are unclear. Here, we apply a genetic approach, using isogenic human induced pluripotent stem cells, to evaluate the effects of schizophrenia (SZ)-associated common variants predicted to function as SZ expression quantitative trait loci (eQTLs). By integrating CRISPR-mediated gene editing, activation and repression technologies to study one putative SZ eQTL (FURIN rs4702) and four top-ranked SZ eQTL genes (FURIN, SNAP91, TSNARE1 and CLCN3), our platform resolves pre- and postsynaptic neuronal deficits, recapitulates genotype-dependent gene expression differences and identifies convergence downstream of SZ eQTL gene perturbations. Our observations highlight the cell-type-specific effects of common variants and demonstrate a synergistic effect between SZ eQTL genes that converges on synaptic function. We propose that the links between rare and common variants implicated in psychiatric disease risk constitute a potentially generalizable phenomenon occurring more widely in complex genetic disorders.


Subject(s)
Gene Expression Regulation , Genetic Predisposition to Disease , Induced Pluripotent Stem Cells/pathology , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Schizophrenia/genetics , Schizophrenia/pathology , CRISPR-Cas Systems , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Chloride Channels/metabolism , Female , Furin/antagonists & inhibitors , Furin/genetics , Furin/metabolism , Gene Editing , Genome-Wide Association Study , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Monomeric Clathrin Assembly Proteins/antagonists & inhibitors , Monomeric Clathrin Assembly Proteins/genetics , Monomeric Clathrin Assembly Proteins/metabolism , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/genetics , SNARE Proteins/metabolism
17.
FEBS J ; 275(12): 3051-63, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18459979

ABSTRACT

Soluble N-ethylmaleimide sensitive-factor attachment receptor (SNARE) proteins have crucial roles in driving exocytic membrane fusion. Molecular recognition between vesicle-associated (v)-SNARE and target membrane (t)-SNARE leads to the formation of a four-helix bundle, which facilitates the merging of two apposing membranes. Synthetic peptides patterned after the SNARE motifs are predicted to block SNARE complex formation by competing with the parental SNAREs, inhibiting neuronal exocytosis. As an initial attempt to identify the peptide sequences that block SNARE assembly and membrane fusion, we created thirteen 17-residue synthetic peptides derived from the SNARE motifs of v- and t-SNAREs. The effects of these peptides on SNARE-mediated membrane fusion were investigated using an in vitro lipid-mixing assay, in vivo neurotransmitter release and SNARE complex formation assays in PC12 cells. Peptides derived from the N-terminal region of SNARE motifs had significant inhibitory effects on neuroexocytosis, whereas middle- and C-terminal-mimicking peptides did not exhibit much inhibitory function. N-terminal mimicking peptides blocked N-terminal zippering of SNAREs, a rate-limiting step in SNARE-driven membrane fusion. Therefore, the results suggest that the N-terminal regions of SNARE motifs are excellent targets for the development of drugs to block SNARE-mediated membrane fusion and neurotransmitter release.


Subject(s)
Exocytosis/drug effects , Neurons/drug effects , Peptides/pharmacology , SNARE Proteins/antagonists & inhibitors , Amino Acid Motifs , Amino Acid Sequence , Animals , Kinetics , Membrane Fusion/drug effects , Molecular Sequence Data , Neurons/metabolism , Norepinephrine/metabolism , PC12 Cells , Peptides/chemistry , Rats , SNARE Proteins/chemistry
18.
Trends Cell Biol ; 28(8): 631-645, 2018 08.
Article in English | MEDLINE | ID: mdl-29706534

ABSTRACT

Recent structural and functional studies of the synaptic vesicle fusion machinery suggest an inhibited tripartite complex consisting of neuronal soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs), synaptotagmin, and complexin prior to Ca2+-triggered synaptic vesicle fusion. We speculate that Ca2+-triggered fusion commences with the release of inhibition by Ca2+ binding to synaptotagmin C2 domains. Subsequently, fusion is assisted by SNARE complex zippering and by active membrane remodeling properties of synaptotagmin. This additional, inhibitory role of synaptotagmin may be a general principle since other recent studies suggest that Ca2+ binding to extended synaptotagmin C2 domains enables lipid transport by releasing an inhibited state of the system, and that Munc13 may nominally be in an inhibited state, which is released upon Ca2+ binding to one of its C2 domains.


Subject(s)
Calcium/pharmacology , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Animals , Binding Sites/drug effects , C2 Domains/drug effects , Calcium/metabolism , Humans , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/metabolism , Synaptotagmin I/antagonists & inhibitors , Synaptotagmin I/metabolism
19.
Nat Commun ; 9(1): 370, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371650

ABSTRACT

Astrocytes are implicated in modulation of neuronal excitability and synaptic function, but it remains unknown if these glial cells can directly control activities of motor circuits to influence complex behaviors in vivo. This study focused on the vital respiratory rhythm-generating circuits of the preBötzinger complex (preBötC) and determined how compromised function of local astrocytes affects breathing in conscious experimental animals (rats). Vesicular release mechanisms in astrocytes were disrupted by virally driven expression of either the dominant-negative SNARE protein or light chain of tetanus toxin. We show that blockade of vesicular release in preBötC astrocytes reduces the resting breathing rate and frequency of periodic sighs, decreases rhythm variability, impairs respiratory responses to hypoxia and hypercapnia, and dramatically reduces the exercise capacity. These findings indicate that astrocytes modulate the activity of CNS circuits generating the respiratory rhythm, critically contribute to adaptive respiratory responses in conditions of increased metabolic demand and determine the exercise capacity.


Subject(s)
Astrocytes/physiology , Brain Stem/physiology , Periodicity , Physical Conditioning, Animal/physiology , Respiration , Action Potentials/physiology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Animals, Newborn , Astrocytes/cytology , Brain Stem/cytology , Calcium/metabolism , Female , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hypercapnia/metabolism , Hypercapnia/physiopathology , Hypoxia/metabolism , Hypoxia/physiopathology , Male , Medulla Oblongata/cytology , Medulla Oblongata/physiology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , SNARE Proteins/antagonists & inhibitors , SNARE Proteins/genetics , SNARE Proteins/metabolism
20.
Cell Calcium ; 66: 71-77, 2017 09.
Article in English | MEDLINE | ID: mdl-28807151

ABSTRACT

Regulated exocytosis can be split into a sequence of steps ending with the formation and the dilation of a fusion pore, a neck-like connection between the vesicle and the plasma membrane. Each of these steps is precisely controlled to achieve the optimal spatial and temporal profile of the release of signalling molecules. At the level of the fusion pore, tuning of the exocytosis can be achieved by preventing its formation, by stabilizing the unproductive narrow fusion pore, by altering the speed of fusion pore expansion and by completely closing the fusion pore. The molecular structure and dynamics of fusion pores have become a major focus of cell research, especially as a promising target for therapeutic strategies. Electrophysiological, optical and electrochemical methods have been used extensively to illuminate how cells regulate secretion at the level of a single fusion pore. Here, we describe recent advances in the structure and mechanisms of the initial fusion pore formation and the progress in therapeutic strategies with the focus on exocytosis.


Subject(s)
Exocytosis/physiology , Membrane Fusion/physiology , SNARE Proteins/metabolism , Animals , Clostridium botulinum/metabolism , Exocytosis/drug effects , Ketamine/pharmacology , Membrane Fusion/drug effects , Neurotoxins/metabolism , Neurotoxins/pharmacology , SNARE Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL