Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Protoc ; 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956381

RÉSUMÉ

Here, we present a protocol for isolating functionally intact glutamatergic synaptic vesicles from whole-mouse brain tissue and using them in a single-vesicle assay to examine their association and fusion with plasma membrane mimic vesicles. This is a Protocol Extension, building on our previous protocol, which used a purely synthetic system comprised of reconstituted proteins in liposomes. We also describe the generation of a peptide based on the vesicular glutamate transporter, which is essential in the isolation process of glutamatergic synaptic vesicles. This method uses easily accessible reagents to generate fusion-competent glutamatergic synaptic vesicles through immunoisolation. The generation of the vGlut peptide can be accomplished in 6 d, while the isolation of the synaptic vesicles by using the peptide can be accomplished in 2 d, with an additional day to fluorescently label the synaptic vesicles for use in a single-vesicle hybrid fusion assay. The single-vesicle fusion assay can be accomplished in 1 d and can unambiguously delineate synaptic vesicle association, dissociation, Ca2+-independent and Ca2+-dependent fusion modalities. This assay grants control of the synaptic vesicle environment while retaining the complexity of the synaptic vesicles themselves. This protocol can be adapted to studies of other types of synaptic vesicles or, more generally, different secretory or transport vesicles. The workflow described here requires expertise in biochemistry techniques, in particular, protein purification and fluorescence imaging. We assume that the laboratory has protein-purification equipment, including chromatography systems.

2.
Cell Rep ; 43(5): 114026, 2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38809756

RÉSUMÉ

Synaptic vesicle docking and priming are dynamic processes. At the molecular level, SNAREs (soluble NSF attachment protein receptors), synaptotagmins, and other factors are critical for Ca2+-triggered vesicle exocytosis, while disassembly factors, including NSF (N-ethylmaleimide-sensitive factor) and α-SNAP (soluble NSF attachment protein), disassemble and recycle SNAREs and antagonize fusion under some conditions. Here, we introduce a hybrid fusion assay that uses synaptic vesicles isolated from mouse brains and synthetic plasma membrane mimics. We included Munc18, Munc13, complexin, NSF, α-SNAP, and an ATP-regeneration system and maintained them continuously-as in the neuron-to investigate how these opposing processes yield fusogenic synaptic vesicles. In this setting, synaptic vesicle association is reversible, and the ATP-regeneration system produces the most synchronous Ca2+-triggered fusion, suggesting that disassembly factors perform quality control at the early stages of synaptic vesicle association to establish a highly fusogenic state. We uncovered a functional role for Munc13 ancillary to the MUN domain that alleviates an α-SNAP-dependent inhibition of Ca2+-triggered fusion.

3.
Curr Res Physiol ; 7: 100121, 2024.
Article de Anglais | MEDLINE | ID: mdl-38572021

RÉSUMÉ

Synaptic vesicles specific to inhibitory GABA-releasing neurons are critical for regulating neuronal excitability. To study the specific molecular composition, architecture, and function of inhibitory synaptic vesicles, we have developed a new method to isolate and purify GABA synaptic vesicles from mouse brains. GABA synaptic vesicles were immunoisolated from mouse brain tissue using an engineered fragment antigen-binding region (Fab) against the vesicular GABA transporter (vGAT) and purified. Western blot analysis confirmed that the GABA synaptic vesicles were specifically enriched for vGAT and largely depleted of contaminants from other synaptic vesicle types, such as vesicular glutamate transporter (vGLUT1), and other cellular organelles. This degree of purity was achieved despite the relatively low abundance of vGAT vesicles compared to the total synaptic vesicle pool in mammalian brains. Cryo-electron microscopy images of these isolated GABA synaptic vesicles revealed intact morphology with circular shape and protruding proteinaceous densities. The GABA synaptic vesicles are functional, as assessed by a hybrid (ex vivo/in vitro) vesicle fusion assay, and they undergo synchronized fusion with synthetic plasma membrane mimic vesicles in response to Ca2+-triggering, but, as a negative control, not to Mg2+-triggering. Our immunoisolation method could also be applied to other types of vesicles.

4.
Proc Natl Acad Sci U S A ; 120(44): e2310174120, 2023 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-37883437

RÉSUMÉ

α-synuclein (α-Syn) is a presynaptic protein that is involved in Parkinson's and other neurodegenerative diseases and binds to negatively charged phospholipids. Previously, we reported that α-Syn clusters synthetic proteoliposomes that mimic synaptic vesicles. This vesicle-clustering activity depends on a specific interaction of α-Syn with anionic phospholipids. Here, we report that α-Syn surprisingly also interacts with the neutral phospholipid lysophosphatidylcholine (lysoPC). Even in the absence of anionic lipids, lysoPC facilitates α-Syn-induced vesicle clustering but has no effect on Ca2+-triggered fusion in a single vesicle-vesicle fusion assay. The A30P mutant of α-Syn that causes familial Parkinson disease has a reduced affinity to lysoPC and does not induce vesicle clustering. Taken together, the α-Syn-lysoPC interaction may play a role in α-Syn function.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Humains , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme , Vésicules synaptiques/métabolisme , Lysolécithine/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Phospholipides/métabolisme
5.
Cell Rep ; 42(4): 112345, 2023 04 25.
Article de Anglais | MEDLINE | ID: mdl-37027300

RÉSUMÉ

The AAA+ NSF complex is responsible for SNARE complex disassembly both before and after membrane fusion. Loss of NSF function results in pronounced developmental and degenerative defects. In a genetic screen for sensory deficits in zebrafish, we identified a mutation in nsf, I209N, that impairs hearing and balance in a dosage-dependent manner without accompanying defects in motility, myelination, and innervation. In vitro experiments demonstrate that while the I209N NSF protein recognizes SNARE complexes, the effects on disassembly are dependent upon the type of SNARE complex and I209N concentration. Higher levels of I209N protein produce a modest decrease in binary (syntaxin-SNAP-25) SNARE complex disassembly and residual ternary (syntaxin-1A-SNAP-25-synaptobrevin-2) disassembly, whereas at lower concentrations binary disassembly activity is strongly reduced and ternary disassembly activity is absent. Our study suggests that the differential effect on disassembly of SNARE complexes leads to selective effects on NSF-mediated membrane trafficking and auditory/vestibular function.


Sujet(s)
Fusion membranaire , Protéines SNARE , Animaux , Protéines SNARE/génétique , Protéines SNARE/métabolisme , Danio zébré/génétique , Danio zébré/métabolisme , Protéines SNAP/génétique , Protéines SNAP/métabolisme , N-Ethylmaleimide-sensitive factors/métabolisme , Mutation/génétique , Contrôle de qualité
6.
Proc Natl Acad Sci U S A ; 120(13): e2300360120, 2023 03 28.
Article de Anglais | MEDLINE | ID: mdl-36940324

RÉSUMÉ

The Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) introduced a relatively large number of mutations, including three mutations in the highly conserved heptad repeat 1 (HR1) region of the spike glycoprotein (S) critical for its membrane fusion activity. We show that one of these mutations, N969K induces a substantial displacement in the structure of the heptad repeat 2 (HR2) backbone in the HR1HR2 postfusion bundle. Due to this mutation, fusion-entry peptide inhibitors based on the Wuhan strain sequence are less efficacious. Here, we report an Omicron-specific peptide inhibitor designed based on the structure of the Omicron HR1HR2 postfusion bundle. Specifically, we inserted an additional residue in HR2 near the Omicron HR1 K969 residue to better accommodate the N969K mutation and relieve the distortion in the structure of the HR1HR2 postfusion bundle it introduced. The designed inhibitor recovers the loss of inhibition activity of the original longHR2_42 peptide with the Wuhan strain sequence against the Omicron variant in both a cell-cell fusion assay and a vesicular stomatitis virus (VSV)-SARS-CoV-2 chimera infection assay, suggesting that a similar approach could be used to combat future variants. From a mechanistic perspective, our work suggests the interactions in the extended region of HR2 may mediate the initial landing of HR2 onto HR1 during the transition of the S protein from the prehairpin intermediate to the postfusion state.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , SARS-CoV-2/génétique , SARS-CoV-2/métabolisme , Protéines de l'enveloppe virale/génétique , Séquence d'acides aminés , Structure secondaire des protéines , Glycoprotéine de spicule des coronavirus/métabolisme , Peptides/génétique , Peptides/pharmacologie , Peptides/composition chimique , Antirétroviraux
7.
FEBS Open Bio ; 13(1): 26-50, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36305864

RÉSUMÉ

Characterizing interactions of Synaptotagmin-1 with the SNARE complex is crucial to understand the mechanism of neurotransmitter release. X-ray crystallography revealed how the Synaptotagmin-1 C2 B domain binds to the SNARE complex through a so-called primary interface and to a complexin-1-SNARE complex through a so-called tripartite interface. Mutagenesis and electrophysiology supported the functional relevance of both interfaces, and extensive additional data validated the primary interface. However, ITC evidence suggesting that binding via the tripartite interface occurs in solution was called into question by subsequent NMR data. Here, we describe joint efforts to address this apparent contradiction. Using the same ITC approach with the same C2 B domain mutant used previously (C2 BKA-Q ) but including ion exchange chromatography to purify it, which is crucial to remove polyacidic contaminants, we were unable to observe the substantial endothermic ITC signal that was previously attributed to binding of this mutant to the complexin-1-SNARE complex through the tripartite interface. We were also unable to detect substantial populations of the tripartite interface in NMR analyses of the ITC samples or in measurements of paramagnetic relaxation effects, despite the high sensitivity of this method to detect weak protein complexes. However, these experiments do not rule out the possibility of very low affinity (KD > 1 mm) binding through this interface. These results emphasize the need to develop methods to characterize the structure of synaptotagmin-1-SNARE complexes between two membranes and to perform further structure-function analyses to establish the physiological relevance of the tripartite interface.


Sujet(s)
Protéines de tissu nerveux , Protéines SNARE , Protéines SNARE/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines adaptatrices du transport vésiculaire/génétique , Protéines adaptatrices du transport vésiculaire/métabolisme , Cytoplasme/métabolisme , Transmission synaptique/physiologie
8.
Proc Natl Acad Sci U S A ; 119(40): e2210990119, 2022 10 04.
Article de Anglais | MEDLINE | ID: mdl-36122200

RÉSUMÉ

Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge currently available coronavirus disease 2019 vaccines and monoclonal antibody therapies through epitope change on the receptor binding domain of the viral spike glycoprotein. Hence, there is a specific urgent need for alternative antivirals that target processes less likely to be affected by mutation, such as the membrane fusion step of viral entry into the host cell. One such antiviral class includes peptide inhibitors, which block formation of the so-called heptad repeat 1 and 2 (HR1HR2) six-helix bundle of the SARS-CoV-2 spike (S) protein and thus interfere with viral membrane fusion. We performed structural studies of the HR1HR2 bundle, revealing an extended, well-folded N-terminal region of HR2 that interacts with the HR1 triple helix. Based on this structure, we designed an extended HR2 peptide that achieves single-digit nanomolar inhibition of SARS-CoV-2 in cell-based and virus-based assays without the need for modifications such as lipidation or chemical stapling. The peptide also strongly inhibits all major SARS-CoV-2 variants to date. This extended peptide is ∼100-fold more potent than all previously published short, unmodified HR2 peptides, and it has a very long inhibition lifetime after washout in virus infection assays, suggesting that it targets a prehairpin intermediate of the SARS-CoV-2 S protein. Together, these results suggest that regions outside the HR2 helical region may offer new opportunities for potent peptide-derived therapeutics for SARS-CoV-2 and its variants, and even more distantly related viruses, and provide further support for the prehairpin intermediate of the S protein.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Glycoprotéine de spicule des coronavirus , Antiviraux/composition chimique , Antiviraux/pharmacologie , Humains , Peptides/composition chimique , Peptides/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques
9.
bioRxiv ; 2022 Aug 11.
Article de Anglais | MEDLINE | ID: mdl-35982670

RÉSUMÉ

Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge currently available COVID-19 vaccines and monoclonal antibody therapies through epitope change on the receptor binding domain of the viral spike glycoprotein. Hence, there is a specific urgent need for alternative antivirals that target processes less likely to be affected by mutation, such as the membrane fusion step of viral entry into the host cell. One such antiviral class includes peptide inhibitors which block formation of the so-called HR1HR2 six-helix bundle of the SARS-CoV-2 spike (S) protein and thus interfere with viral membrane fusion. Here we performed structural studies of the HR1HR2 bundle, revealing an extended, well-folded N-terminal region of HR2 that interacts with the HR1 triple helix. Based on this structure, we designed an extended HR2 peptide that achieves single-digit nanomolar inhibition of SARS-CoV-2 in cell-based fusion, VSV-SARS-CoV-2 chimera, and authentic SARS-CoV-2 infection assays without the need for modifications such as lipidation or chemical stapling. The peptide also strongly inhibits all major SARS-CoV-2 variants to date. This extended peptide is ~100-fold more potent than all previously published short, unmodified HR2 peptides, and it has a very long inhibition lifetime after washout in virus infection assays, suggesting that it targets a pre-hairpin intermediate of the SARS-CoV-2 S protein. Together, these results suggest that regions outside the HR2 helical region may offer new opportunities for potent peptide-derived therapeutics for SARS-CoV-2 and its variants, and even more distantly related viruses, and provide further support for the pre-hairpin intermediate of the S protein. Significance Statement: SARS-CoV-2 infection requires fusion of viral and host membranes, mediated by the viral spike glycoprotein (S). Due to the importance of viral membrane fusion, S has been a popular target for developing vaccines and therapeutics. We discovered a simple peptide that inhibits infection by all major variants of SARS-CoV-2 with nanomolar efficacies. In marked contrast, widely used shorter peptides that lack a key N-terminal extension are about 100 x less potent than this peptide. Our results suggest that a simple peptide with a suitable sequence can be a potent and cost-effective therapeutic against COVID-19 and they provide new insights at the virus entry mechanism.

10.
Front Pharmacol ; 13: 891041, 2022.
Article de Anglais | MEDLINE | ID: mdl-35814209

RÉSUMÉ

The so-called primary interface between the SNARE complex and synaptotagmin-1 (Syt1) is essential for Ca2+-triggered neurotransmitter release in neuronal synapses. The interacting residues of the primary interface are conserved across different species for synaptotagmins (Syt1, Syt2, Syt9), SNAP-25, and syntaxin-1A homologs involved in fast synchronous release. This Ca2+-independent interface forms prior to Ca2+-triggering and plays a role in synaptic vesicle priming. This primary interface is also conserved in the fusion machinery that is responsible for mucin granule membrane fusion. Ca2+-stimulated mucin secretion is mediated by the SNAREs syntaxin-3, SNAP-23, VAMP8, Syt2, and other proteins. Here, we designed and screened a series of hydrocarbon-stapled peptides consisting of SNAP-25 fragments that included some of the key residues involved in the primary interface as observed in high-resolution crystal structures. We selected a subset of four stapled peptides that were highly α-helical as assessed by circular dichroism and that inhibited both Ca2+-independent and Ca2+-triggered ensemble lipid-mixing with neuronal SNAREs and Syt1. In a single-vesicle content-mixing assay with reconstituted neuronal SNAREs and Syt1 or with reconstituted airway SNAREs and Syt2, the selected peptides also suppressed Ca2+-triggered fusion. Taken together, hydrocarbon-stapled peptides that interfere with the primary interface consequently inhibit Ca2+-triggered exocytosis. Our inhibitor screen suggests that these compounds may be useful to combat mucus hypersecretion, which is a major cause of airway obstruction in the pathophysiology of COPD, asthma, and cystic fibrosis.

11.
Proc Natl Acad Sci U S A ; 119(16): e2119467119, 2022 04 19.
Article de Anglais | MEDLINE | ID: mdl-35363556

RÉSUMÉ

Variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge currently available COVID-19 vaccines and monoclonal antibody therapies due to structural and dynamic changes of the viral spike glycoprotein (S). The heptad repeat 1 (HR1) and heptad repeat 2 (HR2) domains of S drive virus­host membrane fusion by assembly into a six-helix bundle, resulting in delivery of viral RNA into the host cell. We surveyed mutations of currently reported SARS-CoV-2 variants and selected eight mutations, including Q954H, N969K, and L981F from the Omicron variant, in the postfusion HR1HR2 bundle for functional and structural studies. We designed a molecular scaffold to determine cryogenic electron microscopy (cryo-EM) structures of HR1HR2 at 2.2­3.8 Å resolution by linking the trimeric N termini of four HR1 fragments to four trimeric C termini of the Dps4 dodecamer from Nostoc punctiforme. This molecular scaffold enables efficient sample preparation and structure determination of the HR1HR2 bundle and its mutants by single-particle cryo-EM. Our structure of the wild-type HR1HR2 bundle resolves uncertainties in previously determined structures. The mutant structures reveal side-chain positions of the mutations and their primarily local effects on the interactions between HR1 and HR2. These mutations do not alter the global architecture of the postfusion HR1HR2 bundle, suggesting that the interfaces between HR1 and HR2 are good targets for developing antiviral inhibitors that should be efficacious against all known variants of SARS-CoV-2 to date. We also note that this work paves the way for similar studies in more distantly related viruses.


Sujet(s)
COVID-19 , SARS-CoV-2 , Glycoprotéine de spicule des coronavirus , Séquence conservée , Humains , Domaines protéiques , SARS-CoV-2/génétique , Glycoprotéine de spicule des coronavirus/composition chimique , Glycoprotéine de spicule des coronavirus/génétique , Pénétration virale
12.
Nature ; 603(7903): 949-956, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35322233

RÉSUMÉ

Membrane fusion triggered by Ca2+ is orchestrated by a conserved set of proteins to mediate synaptic neurotransmitter release, mucin secretion and other regulated exocytic processes1-4. For neurotransmitter release, the Ca2+ sensitivity is introduced by interactions between the Ca2+ sensor synaptotagmin and the SNARE complex5, and sequence conservation and functional studies suggest that this mechanism is also conserved for mucin secretion6. Disruption of Ca2+-triggered membrane fusion by a pharmacological agent would have therapeutic value for mucus hypersecretion as it is the major cause of airway obstruction in the pathophysiology of respiratory viral infection, asthma, chronic obstructive pulmonary disease and cystic fibrosis7-11. Here we designed a hydrocarbon-stapled peptide that specifically disrupts Ca2+-triggered membrane fusion by interfering with the so-called primary interface between the neuronal SNARE complex and the Ca2+-binding C2B domain of synaptotagmin-1. In reconstituted systems with these neuronal synaptic proteins or with their airway homologues syntaxin-3, SNAP-23, VAMP8, synaptotagmin-2, along with Munc13-2 and Munc18-2, the stapled peptide strongly suppressed Ca2+-triggered fusion at physiological Ca2+ concentrations. Conjugation of cell-penetrating peptides to the stapled peptide resulted in efficient delivery into cultured human airway epithelial cells and mouse airway epithelium, where it markedly and specifically reduced stimulated mucin secretion in both systems, and substantially attenuated mucus occlusion of mouse airways. Taken together, peptides that disrupt Ca2+-triggered membrane fusion may enable the therapeutic modulation of mucin secretory pathways.


Sujet(s)
Calcium , Hydrocarbures , Fusion membranaire , Mucines , Protéines SNARE , Animaux , Calcium/métabolisme , Hydrocarbures/composition chimique , Fusion membranaire/physiologie , Souris , Mucines/métabolisme , Agents neuromédiateurs/métabolisme , Peptides/pharmacologie , Muqueuse respiratoire , Protéines SNARE/métabolisme
13.
Cell Death Dis ; 12(10): 939, 2021 10 13.
Article de Anglais | MEDLINE | ID: mdl-34645799

RÉSUMÉ

Lysosome-autophagosome fusion is critical to autophagosome maturation. Although several proteins that regulate this fusion process have been identified, the prefusion architecture and its regulation remain unclear. Herein, we show that upon stimulation, multiple lysosomes form clusters around individual autophagosomes, setting the stage for membrane fusion. The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein on lysosomes-vesicle-associated membrane protein 8 (VAMP8)-plays an important role in forming this prefusion state of lysosomal clusters. To study the potential role of phosphorylation on spontaneous fusion, we investigated the effect of phosphorylation of C-terminal residues of VAMP8. Using a phosphorylation mimic, we observed a decrease of fusion in an ensemble lipid mixing assay and an increase of unfused lysosomes associated with autophagosomes. These results suggest that phosphorylation not only reduces spontaneous fusion for minimizing autophagic flux under normal conditions, but also preassembles multiple lysosomes to increase the fusion probability for resuming autophagy upon stimulation. VAMP8 phosphorylation may thus play an important role in chemotherapy drug resistance by influencing autophagosome maturation.


Sujet(s)
Autophagosomes/métabolisme , Lysosomes/métabolisme , Fusion membranaire , Protéines R-SNARE/métabolisme , Autophagosomes/effets des médicaments et des substances chimiques , Autophagosomes/ultrastructure , [(3-Chlorophényl)hydrazono]malononitrile/pharmacologie , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Cellules HeLa , Humains , Lysosomes/effets des médicaments et des substances chimiques , Lysosomes/ultrastructure , Fusion membranaire/effets des médicaments et des substances chimiques , Phosphorylation/effets des médicaments et des substances chimiques , Protéines R-SNARE/composition chimique , Protéines SNARE/métabolisme , Témozolomide/pharmacologie
14.
EMBO J ; 38(22): e101603, 2019 11 15.
Article de Anglais | MEDLINE | ID: mdl-31566781

RÉSUMÉ

Neurexins are presynaptic, cell-adhesion molecules that specify the functional properties of synapses via interactions with trans-synaptic ligands. Neurexins are extensively alternatively spliced at six canonical sites that regulate multifarious ligand interactions, but the structural mechanisms underlying alternative splicing-dependent neurexin regulation are largely unknown. Here, we determined high-resolution structures of the complex of neurexophilin-1 and the second laminin/neurexin/sex-hormone-binding globulin domain (LNS2) of neurexin-1 and examined how alternative splicing at splice site #2 (SS2) regulates the complex. Our data reveal a unique, extensive, neurexophilin-neurexin binding interface that extends the jelly-roll ß-sandwich of LNS2 of neurexin-1 into neurexophilin-1. The SS2A insert of LNS2 augments this interface, increasing the binding affinity of LNS2 for neurexophilin-1. Taken together, our data reveal an unexpected architecture of neurexophilin-neurexin complexes that accounts for the modulation of binding by alternative splicing, which in turn regulates the competition of neurexophilin for neurexin binding with other ligands.


Sujet(s)
Épissage alternatif , Protéines de liaison au calcium/composition chimique , Protéines de liaison au calcium/métabolisme , Glycoprotéines/composition chimique , Glycoprotéines/métabolisme , Laminine/métabolisme , Molécules d'adhérence cellulaire neurales/composition chimique , Molécules d'adhérence cellulaire neurales/métabolisme , Neuropeptides/composition chimique , Neuropeptides/métabolisme , Séquence d'acides aminés , Animaux , Protéines de liaison au calcium/génétique , Cristallographie aux rayons X , Glycoprotéines/génétique , Ligands , Souris , Modèles moléculaires , Molécules d'adhérence cellulaire neurales/génétique , Neuropeptides/génétique , Liaison aux protéines , Conformation des protéines , Domaines protéiques , Rats , Similitude de séquences
15.
Elife ; 72018 09 10.
Article de Anglais | MEDLINE | ID: mdl-30198481

RÉSUMÉ

The recycling of SNARE proteins following complex formation and membrane fusion is an essential process in eukaryotic trafficking. A highly conserved AAA+ protein, NSF (N-ethylmaleimide sensitive factor) and an adaptor protein, SNAP (soluble NSF attachment protein), disassemble the SNARE complex. We report electron-cryomicroscopy structures of the complex of NSF, αSNAP, and the full-length soluble neuronal SNARE complex (composed of syntaxin-1A, synaptobrevin-2, SNAP-25A) in the presence of ATP under non-hydrolyzing conditions at ~3.9 Å resolution. These structures reveal electrostatic interactions by which two αSNAP molecules interface with a specific surface of the SNARE complex. This interaction positions the SNAREs such that the 15 N-terminal residues of SNAP-25A are loaded into the D1 ring pore of NSF via a spiral pattern of interactions between a conserved tyrosine NSF residue and SNAP-25A backbone atoms. This loading process likely precedes ATP hydrolysis. Subsequent ATP hydrolysis then drives complete disassembly.


Sujet(s)
N-Ethylmaleimide-sensitive factors/métabolisme , Protéines SNAP/composition chimique , Protéines SNAP/métabolisme , Adénosine triphosphate/métabolisme , Animaux , Cricetulus , Cinétique , Modèles moléculaires , N-Ethylmaleimide-sensitive factors/composition chimique , N-Ethylmaleimide-sensitive factors/ultrastructure , Conformation des protéines , Sous-unités de protéines/composition chimique , Sous-unités de protéines/métabolisme , Protéines SNAP/ultrastructure , Spécificité du substrat
16.
Elife ; 72018 07 09.
Article de Anglais | MEDLINE | ID: mdl-29985126

RÉSUMÉ

SNARE complex disassembly by the ATPase NSF is essential for neurotransmitter release and other membrane trafficking processes. We developed a single-molecule FRET assay to monitor repeated rounds of NSF-mediated disassembly and reassembly of individual SNARE complexes. For ternary neuronal SNARE complexes, disassembly proceeds in a single step within 100 msec. We observed short- (<0.32 s) and long-lived (≥0.32 s) disassembled states. The long-lived states represent fully disassembled SNARE complex, while the short-lived states correspond to failed disassembly or immediate reassembly. Either high ionic strength or decreased αSNAP concentration reduces the disassembly rate while increasing the frequency of short-lived states. NSF is also capable of disassembling anti-parallel ternary SNARE complexes, implicating it in quality control. Finally, complexin-1 competes with αSNAP binding to the SNARE complex; addition of complexin-1 has an effect similar to that of decreasing the αSNAP concentration, possibly differentially regulating cis and trans SNARE complexes disassembly.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/métabolisme , N-Ethylmaleimide-sensitive factors/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines SNARE/métabolisme , Animaux , Cricetulus , Transfert d'énergie par résonance de fluorescence , Cinétique , Souris , Protéines mutantes/métabolisme , Mutation/génétique , N-Ethylmaleimide-sensitive factors/ultrastructure , Concentration osmolaire , Liaison aux protéines , Domaines protéiques , Protéines Qa-SNARE/composition chimique , Protéines Qa-SNARE/métabolisme , Rats , Imagerie de molécules uniques , Protéines SNAP/métabolisme , Protéines SNAP/ultrastructure
17.
Neuron ; 95(3): 591-607.e10, 2017 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-28772123

RÉSUMÉ

Munc13 catalyzes the transit of syntaxin from a closed complex with Munc18 into the ternary SNARE complex. Here we report a new function of Munc13, independent of Munc18: it promotes the proper syntaxin/synaptobrevin subconfiguration during assembly of the ternary SNARE complex. In cooperation with Munc18, Munc13 additionally ensures the proper syntaxin/SNAP-25 subconfiguration. In a reconstituted fusion assay with SNAREs, complexin, and synaptotagmin, inclusion of both Munc13 and Munc18 quadruples the Ca2+-triggered amplitude and achieves Ca2+ sensitivity at near-physiological concentrations. In Munc13-1/2 double-knockout neurons, expression of a constitutively open mutant of syntaxin could only minimally restore neurotransmitter release relative to Munc13-1 rescue. Together, the physiological functions of Munc13 may be related to regulation of proper SNARE complex assembly.


Sujet(s)
Exocytose/physiologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines Munc18/métabolisme , Protéines de tissu nerveux/métabolisme , Agents neuromédiateurs/métabolisme , Protéines SNARE/métabolisme , Vésicules synaptiques/métabolisme , Animaux , Cellules cultivées , Protéines et peptides de signalisation intracellulaire/génétique , Souris , Protéines de tissu nerveux/génétique , Neurones/physiologie , Transmission synaptique/physiologie
18.
Proc Natl Acad Sci U S A ; 113(47): E7590-E7599, 2016 11 22.
Article de Anglais | MEDLINE | ID: mdl-27821736

RÉSUMÉ

In presynaptic nerve terminals, complexin regulates spontaneous "mini" neurotransmitter release and activates Ca2+-triggered synchronized neurotransmitter release. We studied the role of the C-terminal domain of mammalian complexin in these processes using single-particle optical imaging and electrophysiology. The C-terminal domain is important for regulating spontaneous release in neuronal cultures and suppressing Ca2+-independent fusion in vitro, but it is not essential for evoked release in neuronal cultures and in vitro. This domain interacts with membranes in a curvature-dependent fashion similar to a previous study with worm complexin [Snead D, Wragg RT, Dittman JS, Eliezer D (2014) Membrane curvature sensing by the C-terminal domain of complexin. Nat Commun 5:4955]. The curvature-sensing value of the C-terminal domain is comparable to that of α-synuclein. Upon replacement of the C-terminal domain with membrane-localizing elements, preferential localization to the synaptic vesicle membrane, but not to the plasma membrane, results in suppression of spontaneous release in neurons. Membrane localization had no measurable effect on evoked postsynaptic currents of AMPA-type glutamate receptors, but mislocalization to the plasma membrane increases both the variability and the mean of the synchronous decay time constant of NMDA-type glutamate receptor evoked postsynaptic currents.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/composition chimique , Protéines adaptatrices du transport vésiculaire/métabolisme , Membrane cellulaire/métabolisme , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/métabolisme , Neurones/métabolisme , Animaux , Calcium/métabolisme , Cellules cultivées , Souris , Neurones/cytologie , Rats , Protéines SNARE/métabolisme , Vésicules synaptiques/métabolisme , Synaptotagmines/métabolisme , alpha-Synucléine/composition chimique , alpha-Synucléine/métabolisme
19.
Proc Natl Acad Sci U S A ; 113(32): E4698-707, 2016 08 09.
Article de Anglais | MEDLINE | ID: mdl-27444020

RÉSUMÉ

Complexin activates Ca(2+)-triggered neurotransmitter release and regulates spontaneous release in the presynaptic terminal by cooperating with the neuronal soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and the Ca(2+)-sensor synaptotagmin. The N-terminal domain of complexin is important for activation, but its molecular mechanism is still poorly understood. Here, we observed that a split pair of N-terminal and central domain fragments of complexin is sufficient to activate Ca(2+)-triggered release using a reconstituted single-vesicle fusion assay, suggesting that the N-terminal domain acts as an independent module within the synaptic fusion machinery. The N-terminal domain can also interact independently with membranes, which is enhanced by a cooperative interaction with the neuronal SNARE complex. We show by mutagenesis that membrane binding of the N-terminal domain is essential for activation of Ca(2+)-triggered fusion. Consistent with the membrane-binding property, the N-terminal domain can be substituted by the influenza virus hemagglutinin fusion peptide, and this chimera also activates Ca(2+)-triggered fusion. Membrane binding of the N-terminal domain of complexin therefore cooperates with the other fusogenic elements of the synaptic fusion machinery during Ca(2+)-triggered release.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/physiologie , Calcium/physiologie , Fusion membranaire , Protéines adaptatrices du transport vésiculaire/composition chimique , Humains , Domaines protéiques , Protéines SNARE/physiologie , Vésicules synaptiques/physiologie , Synaptotagmine I/physiologie
20.
EMBO J ; 35(16): 1810-21, 2016 08 15.
Article de Anglais | MEDLINE | ID: mdl-27402227

RÉSUMÉ

Membrane fusion is essential for eukaryotic life, requiring SNARE proteins to zipper up in an α-helical bundle to pull two membranes together. Here, we show that vesicle fusion can be suppressed by phosphorylation of core conserved residues inside the SNARE domain. We took a proteomics approach using a PKCB knockout mast cell model and found that the key mast cell secretory protein VAMP8 becomes phosphorylated by PKC at multiple residues in the SNARE domain. Our data suggest that VAMP8 phosphorylation reduces vesicle fusion in vitro and suppresses secretion in living cells, allowing vesicles to dock but preventing fusion with the plasma membrane. Markedly, we show that the phosphorylation motif is absent in all eukaryotic neuronal VAMPs, but present in all other VAMPs. Thus, phosphorylation of SNARE domains is a general mechanism to restrict how much cells secrete, opening the door for new therapeutic strategies for suppression of secretion.


Sujet(s)
Protéine kinase C/métabolisme , Maturation post-traductionnelle des protéines , Protéines R-SNARE/métabolisme , Vésicules de sécrétion/métabolisme , Animaux , Lignée cellulaire , Mastocytes/physiologie , Phosphorylation , Protéomique , Rats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...