Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 9261, 2022 06 03.
Article in English | MEDLINE | ID: mdl-35661757

ABSTRACT

Neurotransmitter release of synaptic vesicles relies on the assembly of the soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex, consisting of syntaxin and SNAP-25 on the plasma membrane and synaptobrevin on the synaptic vesicle. The formation of the SNARE complex progressively zippers towards the membranes, which drives membrane fusion between the plasma membrane and the synaptic vesicle. However, the underlying molecular mechanism of SNARE complex regulation is unclear. In this study, we investigated the syntaxin-3b isoform found in the retinal ribbon synapses using single-molecule fluorescence resonance energy transfer (smFRET) to monitor the conformational changes of syntaxin-3b that modulate the SNARE complex formation. We found that syntaxin-3b is predominantly in a self-inhibiting closed conformation, inefficiently forming the ternary SNARE complex. Conversely, a phosphomimetic mutation (T14E) at the N-terminal region of syntaxin-3b promoted the open conformation, similar to the constitutively open form of syntaxin LE mutant. When syntaxin-3b is bound to Munc18-1, SNARE complex formation is almost completely blocked. Surprisingly, the T14E mutation of syntaxin-3b partially abolishes Munc18-1 regulation, acting as a conformational switch to trigger SNARE complex assembly. Thus, we suggest a model where the conformational change of syntaxin-3b induced by phosphorylation initiates the release of neurotransmitters in the ribbon synapses.


Subject(s)
Membrane Fusion , SNARE Proteins , Membrane Fusion/physiology , Munc18 Proteins/metabolism , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Synapses/metabolism , Synaptic Transmission , Synaptic Vesicles/metabolism , Syntaxin 1/genetics , Syntaxin 1/metabolism
2.
Biomolecules ; 13(1)2022 12 20.
Article in English | MEDLINE | ID: mdl-36671389

ABSTRACT

The N-methyl-D-aspartate (NMDA)-sensitive glutamate receptor (NMDAR) helps assemble downstream signaling pathways through protein interactions within the postsynaptic density (PSD), which are mediated by its intracellular C-terminal domain (CTD). The most abundant NMDAR subunits in the brain are GluN2A and GluN2B, which are associated with a developmental switch in NMDAR composition. Previously, we used single molecule fluorescence resonance energy transfer (smFRET) to show that the GluN2B CTD contained an intrinsically disordered region with slow, hop-like conformational dynamics. The CTD from GluN2B also undergoes liquid-liquid phase separation (LLPS) with synaptic proteins. Here, we extend these observations to the GluN2A CTD. Sequence analysis showed that both subunits contain a form of intrinsic disorder classified as weak polyampholytes. However, only GluN2B contained matched patterning of arginine and aromatic residues, which are linked to LLPS. To examine the conformational distribution, we used discrete molecular dynamics (DMD), which revealed that GluN2A favors extended disordered states containing secondary structures while GluN2B favors disordered globular states. In contrast to GluN2B, smFRET measurements found that GluN2A lacked slow conformational dynamics. Thus, simulation and experiments found differences in the form of disorder. To understand how this affects protein interactions, we compared the ability of these two NMDAR isoforms to undergo LLPS. We found that GluN2B readily formed condensates with PSD-95 and SynGAP, while GluN2A failed to support LLPS and instead showed a propensity for colloidal aggregation. That GluN2A fails to support this same condensate formation suggests a developmental switch in LLPS propensity.


Subject(s)
Glutamic Acid , N-Methylaspartate , Glutamic Acid/metabolism , N-Methylaspartate/metabolism , Brain/metabolism , Signal Transduction , Receptors, N-Methyl-D-Aspartate/metabolism
3.
Elife ; 102021 11 03.
Article in English | MEDLINE | ID: mdl-34730514

ABSTRACT

The dimeric ER Ca2+ sensor STIM1 controls store-operated Ca2+ entry (SOCE) through the regulated binding of its CRAC activation domain (CAD) to Orai channels in the plasma membrane. In resting cells, the STIM1 CC1 domain interacts with CAD to suppress SOCE, but the structural basis of this interaction is unclear. Using single-molecule Förster resonance energy transfer (smFRET) and protein crosslinking approaches, we show that CC1 interacts dynamically with CAD in a domain-swapped configuration with an orientation predicted to sequester its Orai-binding region adjacent to the ER membrane. Following ER Ca2+ depletion and release from CAD, cysteine crosslinking indicates that the two CC1 domains become closely paired along their entire length in the active Orai-bound state. These findings provide a structural basis for the dual roles of CC1: sequestering CAD to suppress SOCE in resting cells and propelling it toward the plasma membrane to activate Orai and SOCE after store depletion.


Subject(s)
Calcium Signaling , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , Stromal Interaction Molecule 1/genetics , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Fluorescence Resonance Energy Transfer , Neoplasm Proteins/metabolism , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism
4.
EMBO J ; 38(22): e101603, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31566781

ABSTRACT

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.


Subject(s)
Alternative Splicing , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Laminin/metabolism , Neural Cell Adhesion Molecules/chemistry , Neural Cell Adhesion Molecules/metabolism , Neuropeptides/chemistry , Neuropeptides/metabolism , Amino Acid Sequence , Animals , Calcium-Binding Proteins/genetics , Crystallography, X-Ray , Glycoproteins/genetics , Ligands , Mice , Models, Molecular , Neural Cell Adhesion Molecules/genetics , Neuropeptides/genetics , Protein Binding , Protein Conformation , Protein Domains , Rats , Sequence Homology
5.
Curr Opin Struct Biol ; 54: 179-188, 2019 02.
Article in English | MEDLINE | ID: mdl-30986753

ABSTRACT

Here, we review recent insights into the neuronal presynaptic fusion machinery that releases neurotransmitter molecules into the synaptic cleft upon stimulation. The structure of the pre-fusion state of the SNARE/complexin-1/synaptotagmin-1 synaptic protein complex suggests a new model for the initiation of fast Ca2+-triggered membrane fusion. Functional studies have revealed roles of the essential factors Munc18 and Munc13, demonstrating that a part of their function involves the proper assembly of synaptic protein complexes. Near-atomic resolution structures of the NSF/αSNAP/SNARE complex provide first glimpses of the molecular machinery that disassembles the SNARE complex during the synaptic vesicle cycle. These structures show how this machinery captures the SNARE substrate and provide clues as to a possible processing mechanism.


Subject(s)
Synapses/metabolism , Animals , Calcium/metabolism , Humans , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism
6.
Biomolecules ; 9(3)2019 03 22.
Article in English | MEDLINE | ID: mdl-30909517

ABSTRACT

The common conception of intrinsically disordered proteins (IDPs) is that they stochastically sample all possible configurations driven by thermal fluctuations. This is certainly true for many IDPs, which behave as swollen random coils that can be described using polymer models developed for homopolymers. However, the variability in interaction energy between different amino acid sequences provides the possibility that some configurations may be strongly preferred while others are forbidden. In compact globular IDPs, core hydration and packing density can vary between segments of the polypeptide chain leading to complex conformational dynamics. Here, we describe a growing number of proteins that appear intrinsically disordered by biochemical and bioinformatic characterization but switch between restricted regions of conformational space. In some cases, spontaneous switching between conformational ensembles was directly observed, but few methods can identify when an IDP is acting as a restricted chain. Such switching between disparate corners of conformational space could bias ligand binding and regulate the volume of IDPs acting as structural or entropic elements. Thus, mapping the accessible energy landscape and capturing dynamics across a wide range of timescales are essential to recognize when an IDP is acting as such a switch.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Humans , Protein Conformation
7.
Elife ; 72018 09 10.
Article in English | MEDLINE | ID: mdl-30198481

ABSTRACT

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.


Subject(s)
N-Ethylmaleimide-Sensitive Proteins/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/chemistry , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Cricetulus , Kinetics , Models, Molecular , N-Ethylmaleimide-Sensitive Proteins/chemistry , N-Ethylmaleimide-Sensitive Proteins/ultrastructure , Protein Conformation , Protein Subunits/chemistry , Protein Subunits/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/ultrastructure , Substrate Specificity
8.
Elife ; 72018 07 09.
Article in English | MEDLINE | ID: mdl-29985126

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , N-Ethylmaleimide-Sensitive Proteins/metabolism , Nerve Tissue Proteins/metabolism , SNARE Proteins/metabolism , Animals , Cricetulus , Fluorescence Resonance Energy Transfer , Kinetics , Mice , Mutant Proteins/metabolism , Mutation/genetics , N-Ethylmaleimide-Sensitive Proteins/ultrastructure , Osmolar Concentration , Protein Binding , Protein Domains , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/metabolism , Rats , Single Molecule Imaging , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/metabolism , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/ultrastructure
9.
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
10.
Annu Rev Biophys ; 47: 469-497, 2018 05 20.
Article in English | MEDLINE | ID: mdl-29792815

ABSTRACT

This review summarizes current knowledge of synaptic proteins that are central to synaptic vesicle fusion in presynaptic active zones, including SNAREs (soluble N-ethylmaleimide sensitive factor attachment protein receptors), synaptotagmin, complexin, Munc18 (mammalian uncoordinated-18), and Munc13 (mammalian uncoordinated-13), and highlights recent insights in the cooperation of these proteins for neurotransmitter release. Structural and functional studies of the synaptic fusion machinery suggest new molecular models of synaptic vesicle priming and Ca2+-triggered fusion. These studies will be a stepping-stone toward answering the question of how the synaptic vesicle fusion machinery achieves such high speed and sensitivity.


Subject(s)
Neurotransmitter Agents/therapeutic use , Synaptic Transmission/genetics , Biological Transport , Humans , Neurotransmitter Agents/pharmacology
11.
Neuron ; 95(3): 591-607.e10, 2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28772123

ABSTRACT

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.


Subject(s)
Exocytosis/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Munc18 Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Intracellular Signaling Peptides and Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neurons/physiology , Synaptic Transmission/physiology
12.
EMBO J ; 36(6): 816-829, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28137749

ABSTRACT

The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein syntaxin-1 adopts a closed conformation when bound to Munc18-1, preventing binding to synaptobrevin-2 and SNAP-25 to form the ternary SNARE complex. Although it is known that the MUN domain of Munc13-1 catalyzes the transition from the Munc18-1/syntaxin-1 complex to the SNARE complex, the molecular mechanism is unclear. Here, we identified two conserved residues (R151, I155) in the syntaxin-1 linker region as key sites for the MUN domain interaction. This interaction is essential for SNARE complex formation in vitro and synaptic vesicle priming in neuronal cultures. Moreover, this interaction is important for a tripartite Munc18-1/syntaxin-1/MUN complex, in which syntaxin-1 still adopts a closed conformation tightly bound to Munc18-1, whereas the syntaxin-1 linker region changes its conformation, similar to that of the LE mutant of syntaxin-1 when bound to Munc18-1. We suggest that the conformational change of the syntaxin-1 linker region induced by Munc13-1 initiates ternary SNARE complex formation in the neuronal system.


Subject(s)
Exocytosis , Munc18 Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Qa-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Synapses/physiology , Animals , Cells, Cultured , Humans , Models, Biological , Protein Conformation , Qa-SNARE Proteins/chemistry , Rats
13.
Proc Natl Acad Sci U S A ; 113(47): E7590-E7599, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821736

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Cell Membrane/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Mice , Neurons/cytology , Rats , SNARE Proteins/metabolism , Synaptic Vesicles/metabolism , Synaptotagmins/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism
14.
Proc Natl Acad Sci U S A ; 113(32): E4698-707, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27444020

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Calcium/physiology , Membrane Fusion , Adaptor Proteins, Vesicular Transport/chemistry , Humans , Protein Domains , SNARE Proteins/physiology , Synaptic Vesicles/physiology , Synaptotagmin I/physiology
15.
Elife ; 52016 06 02.
Article in English | MEDLINE | ID: mdl-27253060

ABSTRACT

Complexin regulates spontaneous and activates Ca(2+)-triggered neurotransmitter release, yet the molecular mechanisms are still unclear. Here we performed single molecule fluorescence resonance energy transfer experiments and uncovered two conformations of complexin-1 bound to the ternary SNARE complex. In the cis conformation, complexin-1 induces a conformational change at the membrane-proximal C-terminal end of the ternary SNARE complex that specifically depends on the N-terminal, accessory, and central domains of complexin-1. The complexin-1 induced conformation of the ternary SNARE complex may be related to a conformation that is juxtaposing the synaptic vesicle and plasma membranes. In the trans conformation, complexin-1 can simultaneously interact with a ternary SNARE complex via the central domain and a binary SNARE complex consisting of syntaxin-1A and SNAP-25A via the accessory domain. The cis conformation may be involved in activation of synchronous neurotransmitter release, whereas both conformations may be involved in regulating spontaneous release.


Subject(s)
SNARE Proteins/chemistry , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer/methods , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Conformation , Rats , SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/chemistry , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/chemistry , Syntaxin 1/metabolism
16.
Nature ; 525(7567): 62-7, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26280336

ABSTRACT

Synaptotagmin-1 and neuronal SNARE proteins have central roles in evoked synchronous neurotransmitter release; however, it is unknown how they cooperate to trigger synaptic vesicle fusion. Here we report atomic-resolution crystal structures of Ca(2+)- and Mg(2+)-bound complexes between synaptotagmin-1 and the neuronal SNARE complex, one of which was determined with diffraction data from an X-ray free-electron laser, leading to an atomic-resolution structure with accurate rotamer assignments for many side chains. The structures reveal several interfaces, including a large, specific, Ca(2+)-independent and conserved interface. Tests of this interface by mutagenesis suggest that it is essential for Ca(2+)-triggered neurotransmitter release in mouse hippocampal neuronal synapses and for Ca(2+)-triggered vesicle fusion in a reconstituted system. We propose that this interface forms before Ca(2+) triggering, moves en bloc as Ca(2+) influx promotes the interactions between synaptotagmin-1 and the plasma membrane, and consequently remodels the membrane to promote fusion, possibly in conjunction with other interfaces.


Subject(s)
Exocytosis , Neurons/metabolism , SNARE Proteins/chemistry , SNARE Proteins/metabolism , Synaptotagmins/chemistry , Synaptotagmins/metabolism , Animals , Binding Sites/genetics , Calcium/chemistry , Calcium/metabolism , Cell Membrane/metabolism , Crystallography, X-Ray , Electrons , Hippocampus/cytology , Lasers , Magnesium/chemistry , Magnesium/metabolism , Membrane Fusion , Mice , Models, Biological , Models, Molecular , Mutation/genetics , Neurons/chemistry , Neurons/cytology , SNARE Proteins/genetics , Synaptic Transmission , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Synaptotagmins/genetics
17.
J Biol Chem ; 290(16): 10518-34, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25716318

ABSTRACT

Sec1/Munc18 (SM) proteins are essential for membrane trafficking, but their molecular mechanism remains unclear. Using a single vesicle-vesicle content-mixing assay with reconstituted neuronal SNAREs, synaptotagmin-1, and complexin-1, we show that the neuronal SM protein Munc18a/nSec1 has no effect on the intrinsic kinetics of both spontaneous fusion and Ca(2+)-triggered fusion between vesicles that mimic synaptic vesicles and the plasma membrane. However, wild type Munc18a reduced vesicle association ∼50% when the vesicles bearing the t-SNAREs syntaxin-1A and SNAP-25 were preincubated with Munc18 for 30 min. Single molecule experiments with labeled SNAP-25 indicate that the reduction of vesicle association is a consequence of sequestration of syntaxin-1A by Munc18a and subsequent release of SNAP-25 (i.e. Munc18a captures syntaxin-1A via its high affinity interaction). Moreover, a phosphorylation mimic mutant of Munc18a with reduced affinity to syntaxin-1A results in less reduction of vesicle association. In summary, Munc18a does not directly affect fusion, although it has an effect on the t-SNARE complex, depending on the presence of other factors and experimental conditions. Our results suggest that Munc18a primarily acts at the prefusion stage.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Calcium/metabolism , Membrane Fusion , Munc18 Proteins/genetics , Nerve Tissue Proteins/genetics , Synaptic Vesicles/metabolism , Synaptotagmin I/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Munc18 Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Binding , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission , Synaptic Vesicles/chemistry , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Thermodynamics , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
18.
Structure ; 22(10): 1458-66, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25220472

ABSTRACT

Multidomain scaffold proteins serve as hubs in the signal transduction network. By physically colocalizing sequential steps in a transduction pathway, scaffolds catalyze and direct incoming signals. Much is known about binary interactions with individual domains, but it is unknown whether "scaffolding activity" is predictable from pairwise affinities. Here, we characterized multivalent binding to PSD-95, a scaffold protein containing three PDZ domains connected in series by disordered linkers. We used single molecule fluorescence to watch soluble PSD-95 recruit diffusing proteins to a surface-attached receptor cytoplasmic domain. Different ternary complexes showed unique concentration dependence for scaffolding despite similar pairwise affinity. The concentration dependence of scaffolding activity was not predictable based on binary interactions. PSD-95 did not stabilize specific complexes, but rather increased the frequency of transient binding events. Our results suggest that PSD-95 maintains a loosely connected pleomorphic ensemble rather than forming a stereospecific complex containing all components.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Multiprotein Complexes/chemistry , PDZ Domains/physiology , Disks Large Homolog 4 Protein , Fluorescence Polarization , Inhibitory Concentration 50 , Models, Molecular , Multiprotein Complexes/metabolism , Nitric Oxide Synthase Type I/chemistry , Nitric Oxide Synthase Type I/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Stochastic Processes
19.
J Biol Chem ; 288(31): 22506-15, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23782697

ABSTRACT

The NMDA-sensitive glutamate receptor is a ligand-gated ion channel that mediates excitatory synaptic transmission in the nervous system. Extracellular zinc allosterically regulates the NMDA receptor by binding to the extracellular N-terminal domain, which inhibits channel gating. Phosphorylation of the intrinsically disordered intracellular C-terminal domain alleviates inhibition by extracellular zinc. The mechanism for this functional effect is largely unknown. Proline is a hallmark of intrinsic disorder, so we used proline mutagenesis to modulate disorder in the cytoplasmic domain. Proline depletion selectively uncoupled zinc inhibition with little effect on receptor biogenesis, surface trafficking, or ligand-activated gating. Proline depletion also reduced the affinity for a PDZ domain involved in synaptic trafficking and affected small molecule binding. To understand the origin of these phenomena, we used single molecule fluorescence and ensemble biophysical methods to characterize the structural effects of proline mutagenesis. Proline depletion did not eliminate intrinsic disorder, but the underlying conformational dynamics were changed. Thus, we altered the form of intrinsic disorder, which appears sufficient to affect the biological activity. These findings suggest that conformational dynamics within the intrinsically disordered cytoplasmic domain are important for the allosteric regulation of NMDA receptor gating.


Subject(s)
Cytoplasm/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Allosteric Regulation , Electrophoresis, Polyacrylamide Gel , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Protein Binding , Receptors, N-Methyl-D-Aspartate/metabolism , Solubility
20.
Methods Mol Biol ; 896: 3-20, 2012.
Article in English | MEDLINE | ID: mdl-22821514

ABSTRACT

Fluorescence resonance energy transfer provides information about protein structure and dynamics. Single-molecule analysis can capture the information normally lost through ensemble averaging of heterogeneous and dynamic samples. Immobilization of single molecules, under conditions that retain their biological activity, allows for extended observation of the same molecule for tens of seconds. This can capture slow conformational transitions or protein binding and unbinding cycles. Using an open geometry for immobilization allows for direct observation of the response to changing solution conditions or adding ligands. Here we provide detailed methods for immobilization and observation of fluorescently labeled single proteins using total internal reflection microscopy that are widely applicable to the study of intrinsically disordered proteins.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Immobilized Proteins/chemistry , Immobilized Proteins/metabolism , Animals , Biotinylation , Cattle , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Microscopy , Polyethylene Glycols/chemistry , Protein Conformation , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Streptavidin/metabolism , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...