Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 432
1.
Alzheimers Res Ther ; 16(1): 82, 2024 Apr 13.
Article En | MEDLINE | ID: mdl-38615037

BACKGROUND: Previous studies have demonstrated that early intervention was the best plan to inhibit the progression of Alzheimer's disease (AD), which relied on the discovery of early diagnostic biomarkers. In this study, synaptic vesicle glycoprotein 2 A (SV2A) was examined to improve the early diagnostic efficiency in AD. METHODS: In this study, biomarker testing was performed through the single-molecule array (Simoa). A total of 121 subjects including cognitively unimpaired controls, amnestic mild cognitive impairment (aMCI), AD and other types of dementia underwent cerebrospinal fluid (CSF) SV2A testing; 430 subjects including health controls, aMCI, AD and other types of dementia underwent serum SV2A, glial fibrillary acidic protein (GFAP), neurofilament light chain (NfL) and p-tau217 testing; 92 subjects including aMCI and AD underwent both CSF SV2A and serum SV2A testing; 115 cognitively unimpaired subjects including APOE ε4 carriers and APOE ε4 non-carriers were tested for serum SV2A, GFAP, NfL and p-tau217. Then, the efficacy of SV2A for the early diagnosis of AD and its ability to identify those at high risk of AD from a cognitively unimpaired population were further analyzed. RESULTS: Both CSF and serum SV2A significantly and positively correlated with cognitive performance in patients with AD, and their levels gradually decreased with the progression of AD. Serum SV2A demonstrated excellent diagnostic efficacy for aMCI, with a sensitivity of 97.8%, which was significantly higher than those of NfL, GFAP, and p-tau217. The SV2A-positive rates ranged from 92.86 to 100% in aMCI cases that were negative for the above three biomarkers. Importantly, of all the biomarkers tested, serum SV2A had the highest positivity rate (81.82%) in individuals at risk for AD. CONCLUSIONS: Serum SV2A was demonstrated to be a novel and ideal biomarker for the early diagnosis of AD, which can effectively distinguish those at high risk of AD in cognitively unimpaired populations.


Alzheimer Disease , Membrane Glycoproteins , Nerve Tissue Proteins , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Apolipoprotein E4 , Biomarkers , Early Diagnosis , Glycoproteins , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Membrane Glycoproteins/cerebrospinal fluid , Membrane Glycoproteins/chemistry , Nerve Tissue Proteins/cerebrospinal fluid , Nerve Tissue Proteins/chemistry
2.
J Mol Biol ; 435(1): 167710, 2023 01 15.
Article En | MEDLINE | ID: mdl-35777466

Complexins play a critical role in regulating SNARE-mediated exocytosis of synaptic vesicles. Evolutionary divergences in complexin function have complicated our understanding of the role these proteins play in inhibiting the spontaneous fusion of vesicles. Previous structural and functional characterizations of worm and mouse complexins have indicated the membrane curvature-sensing C-terminal domain of these proteins is responsible for differences in inhibitory function. We have characterized the structure and dynamics of the mCpx1 CTD in the absence and presence of membranes and membrane mimetics using NMR, ESR, and optical spectroscopies. In the absence of lipids, the mCpx1 CTD features a short helix near its N-terminus and is otherwise disordered. In the presence of micelles and small unilamellar vesicles, the mCpx1 CTD forms a discontinuous helical structure in its C-terminal 20 amino acids, with no preference for specific lipid compositions. In contrast, the mCpx1 CTD shows distinct compositional preferences in its interactions with large unilamellar vesicles. These studies identify structural divergences in the mCpx1 CTD relative to the wCpx1 CTD in regions that are known to be critical to the wCpx1 CTD's role in inhibiting spontaneous fusion of synaptic vesicles, suggesting a potential structural basis for evolutionary divergences in complexin function.1.


Adaptor Proteins, Vesicular Transport , Nerve Tissue Proteins , Unilamellar Liposomes , Animals , Mice , Adaptor Proteins, Vesicular Transport/chemistry , Calcium/chemistry , Exocytosis , Membrane Fusion , Nerve Tissue Proteins/chemistry , Protein Binding , SNARE Proteins/metabolism , Synaptic Vesicles/chemistry , Unilamellar Liposomes/chemistry , Protein Domains
3.
Eur Biophys J ; 51(6): 465-482, 2022 Sep.
Article En | MEDLINE | ID: mdl-35904588

The size, polydispersity, and electron density profile of synaptic vesicles (SVs) can be studied by small-angle X-ray scattering (SAXS), i.e. by X-ray diffraction from purified SV suspensions in solution. Here we show that size and shape transformations, as they appear in the functional context of these important synaptic organelles, can also be monitored by SAXS. In particular, we have investigated the active uptake of neurotransmitters, and find a mean vesicle radius increase of about 12% after the uptake of glutamate, which indicates an unusually large extensibility of the vesicle surface, likely to be accompanied by conformational changes of membrane proteins and rearrangements of the bilayer. Changes in the electron density profile (EDP) give first indications for such a rearrangement. Details of the protein structure are screened, however, by SVs polydispersity. To overcome the limitations of large ensemble averages and heterogeneous structures, we therefore propose serial X-ray diffraction by single free electron laser pulses. Using simulated data for realistic parameters, we show that this is in principle feasible, and that even spatial distances between vesicle proteins could be assessed by this approach.


Glutamic Acid , Synaptic Vesicles , Biological Transport , Proteins/metabolism , Scattering, Small Angle , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , X-Ray Diffraction
4.
Cell Rep Methods ; 2(4): 100199, 2022 04 25.
Article En | MEDLINE | ID: mdl-35497490

A complete understanding of synaptic-vesicle recycling requires the use of multiple microscopy methods to obtain complementary information. However, many currently available probes are limited to a specific microscopy modality, which necessitates the use of multiple probes and labeling paradigms. Given the complexity of vesicle populations and recycling pathways, having new single-vesicle probes that could be used for multiple microscopy techniques would complement existing sets of tools for studying vesicle function. Here, we present a probe based on the membrane-binding C2 domain of cytosolic phospholipase A2 (cPLA2) that fulfills this need. By conjugating the C2 domain with different detectable tags, we demonstrate that a single, modular probe can allow synaptic vesicles to be imaged at multiple levels of spatial and temporal resolution. Moreover, as a general endocytic marker, the C2 domain may also be used to study membrane recycling in many cell types.


Multimodal Imaging , Synaptic Vesicles , Synaptic Vesicles/chemistry
5.
Nat Struct Mol Biol ; 29(2): 97-107, 2022 02.
Article En | MEDLINE | ID: mdl-35132256

Neurotransmitter release is mediated by proteins that drive synaptic vesicle fusion with the presynaptic plasma membrane. While soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs) form the core of the fusion apparatus, additional proteins play key roles in the fusion pathway. Here, we report that the C-terminal amphipathic helix of the mammalian accessory protein, complexin (Cpx), exerts profound effects on membranes, including the formation of pores and the efficient budding and fission of vesicles. Using nanodisc-black lipid membrane electrophysiology, we demonstrate that the membrane remodeling activity of Cpx modulates the structure and stability of recombinant exocytic fusion pores. Cpx had particularly strong effects on pores formed by small numbers of SNAREs. Under these conditions, Cpx increased the current through individual pores 3.5-fold, and increased the open time fraction from roughly 0.1 to 1.0. We propose that the membrane sculpting activity of Cpx contributes to the phospholipid rearrangements that underlie fusion by stabilizing highly curved membrane fusion intermediates.


Adaptor Proteins, Vesicular Transport/chemistry , Nerve Tissue Proteins/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , HEK293 Cells , Humans , Lipid Bilayers/chemistry , Membrane Fusion/physiology , Molecular Dynamics Simulation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Pore/chemistry , Nuclear Pore/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation, alpha-Helical , Protein Stability , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism
6.
Biochim Biophys Acta Biomembr ; 1864(1): 183817, 2022 02 01.
Article En | MEDLINE | ID: mdl-34767780

Here, carbon nanodots synthesized from ß-alanine (Ala-CDs) and detonation nanodiamonds (NDs) were assessed using (1) radiolabeled excitatory neurotransmitters L-[14C]glutamate, D-[2,33H]aspartate, and inhibitory ones [3H]GABA, [3H]glycine for registration of their extracellular concentrations in rat cortex nerve terminals; (2) the fluorescent ratiometric probe NR12S and pH-sensitive probe acridine orange for registration of the membrane lipid order and synaptic vesicle acidification, respectively; (3) suspended bilayer lipid membrane (BLM) to monitor changes in transmembrane current. In nerve terminals, Ala-CDs and NDs increased the extracellular concentrations of neurotransmitters and decreased acidification of synaptic vesicles, whereas have not changed sufficiently the lipid order of membrane. Both nanoparticles, Ala-CDs and NDs, were capable of increasing the conductance of the BLM by inducing stable potential-dependent cation-selective pores. Introduction of divalent cations, Zn2+ or Cd2+ on the particles` application side (cis-side) increased the rate of Ala-CDs pore-formation in the BLM. The application of positive potential (+100 mV) to the cis-chamber with Ala-CDs or NDs also activated the insertion as compared with the negative potential (-100 mV). The Ala-CD pores exhibited a wide-range distribution of conductances between 10 and 60 pS and consecutive increase in conductance of each major peak by ~10 pS, which suggest the clustering of the same basic ion-conductive structure. NDs also formed ion-conductive pores ranging from 6 pS to 60 pS with the major peak of conductance at ~12 pS in cholesterol-containing membrane. Observed Ala-CDs and NDs-induced increase in transmembrane current coincides with disturbance of excitatory and inhibitory neurotransmitter transport in nerve terminals.


Cerebral Cortex/metabolism , Nanoparticles/chemistry , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Vesicles/chemistry , Alanine/chemical synthesis , Alanine/chemistry , Animals , Aspartic Acid/chemical synthesis , Aspartic Acid/chemistry , Aspartic Acid/pharmacology , Carbon/chemistry , Carbon/pharmacology , Carbon Radioisotopes/chemistry , Carbon Radioisotopes/pharmacology , Cations/pharmacology , Cerebral Cortex/radiation effects , Cholesterol/chemistry , Glutamic Acid/chemical synthesis , Glutamic Acid/chemistry , Glutamic Acid/pharmacology , Lipid Bilayers/chemistry , Nanodiamonds/chemistry , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/pharmacology , Rats , Synapses/chemistry , Synaptic Transmission/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , gamma-Aminobutyric Acid/chemical synthesis , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/pharmacology
7.
Arch Biochem Biophys ; 709: 108966, 2021 09 30.
Article En | MEDLINE | ID: mdl-34139199

Chemical neurotransmission is the major mechanism of neuronal communication. Neurotransmitters are released from secretory organelles, the synaptic vesicles (SVs) via exocytosis into the synaptic cleft. Fusion of SVs with the presynaptic plasma membrane is balanced by endocytosis, thus maintaining the presynaptic membrane at steady-state levels. The protein machineries responsible for exo- and endocytosis have been extensively investigated. In contrast, less is known about the role of lipids in synaptic transmission and how the lipid composition of SVs is affected by dynamic exo-endocytotic cycling. Here we summarize the current knowledge about the composition, organization, and function of SV membrane lipids. We also cover lipid biogenesis and maintenance during the synaptic vesicle cycle.


Synaptic Membranes/chemistry , Synaptic Vesicles/chemistry , Animals , Endocytosis/physiology , Exocytosis/physiology , Humans , Membrane Lipids/chemistry , Membrane Lipids/metabolism , Synaptic Membranes/metabolism , Synaptic Vesicles/metabolism
8.
Neuroimage ; 238: 118236, 2021 09.
Article En | MEDLINE | ID: mdl-34091034

The mismatch in the spatial resolution of Arterial Spin Labeling (ASL) MRI perfusion images and the anatomy of functionally distinct tissues in the brain leads to a partial volume effect (PVE), which in turn confounds the estimation of perfusion into a specific tissue of interest such as gray or white matter. This confound occurs because the image voxels contain a mixture of tissues with disparate perfusion properties, leading to estimated perfusion values that reflect primarily the volume proportions of tissues in the voxel rather than the perfusion of any particular tissue of interest within that volume. It is already recognized that PVE influences studies of brain perfusion, and that its effect might be even more evident in studies where changes in perfusion are co-incident with alterations in brain structure, such as studies involving a comparison between an atrophic patient population vs control subjects, or studies comparing subjects over a wide range of ages. However, the application of PVE correction (PVEc) is currently limited and the employed methodologies remain inconsistent. In this article, we outline the influence of PVE in ASL measurements of perfusion, explain the main principles of PVEc, and provide a critique of the current state of the art for the use of such methods. Furthermore, we examine the current use of PVEc in perfusion studies and whether there is evidence to support its wider adoption. We conclude that there is sound theoretical motivation for the use of PVEc alongside conventional, 'uncorrected', images, and encourage such combined reporting. Methods for PVEc are now available within standard neuroimaging toolboxes, which makes our recommendation straightforward to implement. However, there is still more work to be done to establish the value of PVEc as well as the efficacy and robustness of existing PVEc methods.


Algorithms , Alzheimer Disease/diagnostic imaging , Brain/diagnostic imaging , Magnetic Resonance Imaging/methods , Neuroimaging/methods , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/analysis , Aniline Compounds , Brain/pathology , Brain/physiopathology , Carbon Radioisotopes , Cerebral Arteries , Cognitive Dysfunction/diagnostic imaging , Cognitive Dysfunction/pathology , Cognitive Dysfunction/physiopathology , Entorhinal Cortex/diagnostic imaging , Entorhinal Cortex/pathology , Entorhinal Cortex/physiopathology , Hippocampus/diagnostic imaging , Hippocampus/pathology , Hippocampus/physiopathology , Image Processing, Computer-Assisted/methods , Membrane Glycoproteins/analysis , Nerve Tissue Proteins/analysis , Organ Size , Perfusion , Positron-Emission Tomography , Pyridines , Pyrrolidinones , Radiopharmaceuticals , Spin Labels , Synaptic Vesicles/chemistry , Thiazoles
9.
Nat Commun ; 12(1): 927, 2021 02 10.
Article En | MEDLINE | ID: mdl-33568632

α-Synuclein (αS) is a presynaptic disordered protein whose aberrant aggregation is associated with Parkinson's disease. The functional role of αS is still debated, although it has been involved in the regulation of neurotransmitter release via the interaction with synaptic vesicles (SVs). We report here a detailed characterisation of the conformational properties of αS bound to the inner and outer leaflets of the presynaptic plasma membrane (PM), using small unilamellar vesicles. Our results suggest that αS preferentially binds the inner PM leaflet. On the basis of these studies we characterise in vitro a mechanism by which αS stabilises, in a concentration-dependent manner, the docking of SVs on the PM by establishing a dynamic link between the two membranes. The study then provides evidence that changes in the lipid composition of the PM, typically associated with neurodegenerative diseases, alter the modes of binding of αS, specifically in a segment of the sequence overlapping with the non-amyloid component region. Taken together, these results reveal how lipid composition modulates the interaction of αS with the PM and underlie its functional and pathological behaviours in vitro.


Lipids/chemistry , Synaptic Membranes/metabolism , Synaptic Vesicles/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Humans , Lipid Metabolism , Protein Conformation , Synaptic Membranes/chemistry , Synaptic Membranes/genetics , Synaptic Vesicles/chemistry , Synaptic Vesicles/genetics , alpha-Synuclein/genetics
10.
Phys Chem Chem Phys ; 23(3): 2117-2125, 2021 Jan 28.
Article En | MEDLINE | ID: mdl-33437978

Complexin-1 (Cpx) and α-synuclein (α-Syn) are involved in neurotransmitter release through an interaction with synaptic vesicles (SVs). Recent studies demonstrated that Cpx and α-Syn preferentially associate with highly curved membranes, like SVs, to correctly position them for fusion. Here, based on recent experimental results, to further propose a possible explanation for this mechanism, we performed in silico simulations probing interactions between Cpx or α-Syn and membranes of varying curvature. We found that the preferential association is attributed to smaller, curved membranes containing more packing defects that expose hydrophobic acyl tails, which may favorably interact with hydrophobic residues of Cpx and α-Syn. The number of membrane defects is proportional to the curvature and the size can be regulated by cholesterol.


Adaptor Proteins, Vesicular Transport/metabolism , Lipid Bilayers/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Vesicles/metabolism , alpha-Synuclein/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Cholesterol/chemistry , Hydrogen Bonding , Lipid Bilayers/chemistry , Molecular Dynamics Simulation , Nerve Tissue Proteins/chemistry , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Phosphatidylserines/chemistry , Protein Binding , Synaptic Vesicles/chemistry , alpha-Synuclein/chemistry
11.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Article En | MEDLINE | ID: mdl-33468652

Neurotransmitter release is governed by eight central proteins among other factors: the neuronal SNAREs syntaxin-1, synaptobrevin, and SNAP-25, which form a tight SNARE complex that brings the synaptic vesicle and plasma membranes together; NSF and SNAPs, which disassemble SNARE complexes; Munc18-1 and Munc13-1, which organize SNARE complex assembly; and the Ca2+ sensor synaptotagmin-1. Reconstitution experiments revealed that Munc18-1, Munc13-1, NSF, and α-SNAP can mediate Ca2+-dependent liposome fusion between synaptobrevin liposomes and syntaxin-1-SNAP-25 liposomes, but high fusion efficiency due to uncontrolled SNARE complex assembly did not allow investigation of the role of synaptotagmin-1 on fusion. Here, we show that decreasing the synaptobrevin-to-lipid ratio in the corresponding liposomes to very low levels leads to inefficient fusion and that synaptotagmin-1 strongly stimulates fusion under these conditions. Such stimulation depends on Ca2+ binding to the two C2 domains of synaptotagmin-1. We also show that anchoring SNAP-25 on the syntaxin-1 liposomes dramatically enhances fusion. Moreover, we uncover a synergy between synaptotagmin-1 and membrane anchoring of SNAP-25, which allows efficient Ca2+-dependent fusion between liposomes bearing very low synaptobrevin densities and liposomes containing very low syntaxin-1 densities. Thus, liposome fusion in our assays is achieved with a few SNARE complexes in a manner that requires Munc18-1 and Munc13-1 and that depends on Ca2+ binding to synaptotagmin-1, all of which are fundamental features of neurotransmitter release in neurons.


Munc18 Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/metabolism , Synaptotagmin I/metabolism , Animals , Calcium/metabolism , Gene Expression Regulation , Liposomes/chemistry , Liposomes/metabolism , Membrane Fusion , Munc18 Proteins/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurotransmitter Agents/genetics , Neurotransmitter Agents/metabolism , Phospholipids/chemistry , Phospholipids/metabolism , Rats , Synaptic Transmission , Synaptic Vesicles/chemistry , Synaptosomal-Associated Protein 25/genetics , Synaptotagmin I/genetics , Syntaxin 1/genetics , Syntaxin 1/metabolism , Vesicle-Associated Membrane Protein 2/genetics , Vesicle-Associated Membrane Protein 2/metabolism
12.
Proc Natl Acad Sci U S A ; 117(38): 23527-23538, 2020 09 22.
Article En | MEDLINE | ID: mdl-32907943

Clathrin light chain (CLC) subunits in vertebrates are encoded by paralogous genes CLTA and CLTB, and both gene products are alternatively spliced in neurons. To understand how this CLC diversity influences neuronal clathrin function, we characterized the biophysical properties of clathrin comprising individual CLC variants for correlation with neuronal phenotypes of mice lacking either CLC-encoding gene. CLC splice variants differentially influenced clathrin knee conformation within assemblies, and clathrin with neuronal CLC mixtures was more effective in membrane deformation than clathrin with single neuronal isoforms nCLCa or nCLCb. Correspondingly, electrophysiological recordings revealed that neurons from mice lacking nCLCa or nCLCb were both defective in synaptic vesicle replenishment. Mice with only nCLCb had a reduced synaptic vesicle pool and impaired neurotransmission compared to WT mice, while nCLCa-only mice had increased synaptic vesicle numbers, restoring normal neurotransmission. These findings highlight differences between the CLC isoforms and show that isoform mixing influences tissue-specific clathrin activity in neurons, which requires their functional balance.


Clathrin Light Chains , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , Cells, Cultured , Clathrin Light Chains/chemistry , Clathrin Light Chains/genetics , Clathrin Light Chains/metabolism , Mice , Mice, Knockout , Neurons/cytology , Neurons/metabolism , Protein Isoforms/chemistry , Protein Isoforms/metabolism
13.
Elife ; 92020 08 24.
Article En | MEDLINE | ID: mdl-32831174

Previously, we showed that modulation of the energy barrier for synaptic vesicle fusion boosts release rates supralinearly (Schotten, 2015). Here we show that mouse hippocampal synapses employ this principle to trigger Ca2+-dependent vesicle release and post-tetanic potentiation (PTP). We assess energy barrier changes by fitting release kinetics in response to hypertonic sucrose. Mimicking activation of the C2A domain of the Ca2+-sensor Synaptotagmin-1 (Syt1), by adding a positive charge (Syt1D232N) or increasing its hydrophobicity (Syt14W), lowers the energy barrier. Removing Syt1 or impairing its release inhibitory function (Syt19Pro) increases spontaneous release without affecting the fusion barrier. Both phorbol esters and tetanic stimulation potentiate synaptic strength, and lower the energy barrier equally well in the presence and absence of Syt1. We propose a model where tetanic stimulation activates Syt1-independent mechanisms that lower the energy barrier and act additively with Syt1-dependent mechanisms to produce PTP by exerting multiplicative effects on release rates.


Neuronal Plasticity/physiology , Synaptic Vesicles , Synaptotagmin I/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Female , Hippocampus/cytology , Hippocampus/metabolism , Male , Membrane Fusion/physiology , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism
14.
FEBS Lett ; 594(21): 3450-3463, 2020 11.
Article En | MEDLINE | ID: mdl-32860428

Synaptic vesicle proteins, including N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), Synaptotagmin-1 and Complexin, are responsible for controlling the synchronised fusion of synaptic vesicles with the presynaptic plasma membrane in response to elevated cytosolic calcium levels. A range of structures of SNAREs and their regulatory proteins have been elucidated, but the exact organisation of these proteins at synaptic junction membranes remains elusive. Here, we have used cryoelectron tomography to investigate the arrangement of synaptic proteins in an in vitro reconstituted fusion system. We found that the separation between vesicle and target membranes strongly correlates with the organisation of protein complexes at junctions. At larger membrane separations, protein complexes assume a 'clustered' distribution at the docking site, inducing a protrusion in the target membrane. As the membrane separation decreases, protein complexes become displaced radially outwards and assume a 'ring-like' arrangement. Our findings indicate that docked vesicles can possess a wide range of protein complex numbers and be heterogeneous in their protein arrangements.


Cell Membrane/metabolism , Membrane Proteins/metabolism , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Cell Membrane/ultrastructure , Cryoelectron Microscopy , In Vitro Techniques , Membrane Proteins/ultrastructure , Synaptic Vesicles/ultrastructure , Tomography
15.
Int J Mol Sci ; 21(9)2020 May 07.
Article En | MEDLINE | ID: mdl-32392751

Parkinson's disease (PD) is the second most common neurodegenerative disease; it is characterized by the loss of dopaminergic neurons in the midbrain and the accumulation of neuronal inclusions, mainly consisting of α-synuclein (α-syn) fibrils in the affected regions. The prion-like property of the pathological forms of α-syn transmitted via neuronal circuits has been considered inherent in the nature of PD. Thus, one of the potential targets in terms of PD prevention is the suppression of α-syn conversion from the functional form to pathological forms. Recent studies suggested that α-syn interacts with synaptic vesicle membranes and modulate the synaptic functions. A series of studies suggest that transient interaction of α-syn as multimers with synaptic vesicle membranes composed of phospholipids and other lipids is required for its physiological function, while an α-syn-lipid interaction imbalance is believed to cause α-syn aggregation and the resultant pathological α-syn conversion. Altered lipid metabolisms have also been implicated in the modulation of PD pathogenesis. This review focuses on the current literature reporting the role of lipids, especially phospholipids, and lipid metabolism in α-syn dynamics and aggregation processes.


Lipid Metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Humans , Phospholipids/metabolism , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , alpha-Synuclein/chemistry
16.
Mol Brain ; 13(1): 9, 2020 01 20.
Article En | MEDLINE | ID: mdl-31959215

Synaptic vesicles (SV) contain high concentrations of specific proteins. How these proteins are transported from soma to synapses, and how they become concentrated at SV clusters at presynaptic terminals were examined by immunogold electron microscopy in dissociated rat hippocampal neurons at 3-6 days in culture, a developmental stage when axonal transport of SV proteins is robust. In neuronal somas, labels for the SV integral membrane proteins (synaptophysin, SV2, VAMP/synaptobrevin, and synaptotagmin) were localized at Golgi complexes and other membranous structures that were dispersed in the cytoplasm as individual vesicle/vacuoles. These vesicles/vacuoles became aggregated in axons, with the size of aggregates ranging from 0.2 to 2 µm in length. Pleomorphic vesicle/vacuoles within the aggregate were typically larger (50-300 nm) than SVs, which were uniform in size at ~ 40 nm. These pleomorphic vesicles/vacuoles are probably transport cargos carrying SV integral membrane proteins from the soma, and then are preferentially sorted into axons at early developmental stages. Serial thin sections of young axons indicated that many labeled aggregates were not synaptic, and in fact, some of these axons were without dendritic contacts. In contrast, labels for two SV-associated proteins, synapsin I and α-synuclein, were not localized at the Golgi complexes or associated with membranous structures in the soma, but were dispersed in the cytoplasm. However, these SV-associated proteins became highly concentrated on clusters of SV-like vesicles in axons, and such clusters were already distinctive in axons as early as 3 days in culture. These clusters consisted of ~ 4-30 vesicles in single thin sections, and the vesicles were of a uniform size (~ 40 nm). Serial sectioning analysis showed that these clusters could be part of nascent synapses or exist in axons without any dendritic contact. Importantly, the vesicles were intensely labeled for SV integral membrane proteins as well as SV-associated proteins. Thus, these EM observations reveal that the two groups of proteins, SV integral membrane and SV-associated, proceed through different routes of biosynthesis and axon transport, and are only sorted into the same final compartment, SV clusters, when they are in the axons.


Hippocampus/cytology , Immunohistochemistry , Nerve Tissue Proteins/analysis , Neurons/chemistry , Synaptic Vesicles/chemistry , Animals , Axonal Transport , Axons/chemistry , Axons/ultrastructure , Cells, Cultured , Golgi Apparatus/chemistry , Golgi Apparatus/ultrastructure , Hippocampus/embryology , Membrane Proteins/analysis , Microscopy, Electron , Neurons/ultrastructure , Protein Transport , Rats , Secretory Vesicles/chemistry , Secretory Vesicles/ultrastructure , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/analysis , Vacuoles/chemistry , Vacuoles/ultrastructure
17.
IUBMB Life ; 72(4): 568-576, 2020 04.
Article En | MEDLINE | ID: mdl-31981303

The vacuolar H+ -adenosine triphosphatases (vATPases) acidify multiple intracellular organelles, including synaptic vesicles (SVs) and secretory granules. Acidification of SVs represents a critical point during the SV cycle: without acidification, neurotransmitters cannot be loaded into SVs. Despite the obvious importance of the vesicle acidification process for neurotrasmission and the life of complex organisms, little is known about the regulation of vATPase at the neuronal synapse. In addition, the composition of the vATPase complex on the SVs is unclear. Here, we summarize the key features of vATPase found on SVs, and propose a model of how vATPase activity is regulated during the SV cycle. It is anticipated that the information from the SV lumen is communicated to SV surface in order to signal successful acidification and neurotransmitter loading: we postulate here that the regulators of the vATPase activity exist (e.g., Rabconnectin-3) that promote the recruitment of SV peripheral proteins and, consequently, SV fusion.


Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Humans , Neurons/metabolism , Synapses/physiology
18.
Neuropharmacology ; 169: 107554, 2020 06 01.
Article En | MEDLINE | ID: mdl-30826343

Communication between cells relies on regulated exocytosis, a multi-step process that involves the docking, priming and fusion of vesicles with the plasma membrane, culminating in the release of neurotransmitters and hormones. Key proteins and lipids involved in exocytosis are subjected to Brownian movement and constantly switch between distinct motion states which are governed by short-lived molecular interactions. Critical biochemical reactions between exocytic proteins that occur in the confinement of nanodomains underpin the precise sequence of priming steps which leads to the fusion of vesicles. The advent of super-resolution microscopy techniques has provided the means to visualize individual molecules on the plasma membrane with high spatiotemporal resolution in live cells. These techniques are revealing a highly dynamic nature of the nanoscale organization of the exocytic machinery. In this review, we focus on soluble N-ethylmaleimide-sensitive factor attachment receptor (SNARE) syntaxin-1, which mediates vesicular fusion. Syntaxin-1 is highly mobile at the plasma membrane, and its inherent speed allows fast assembly and disassembly of syntaxin-1 nanoclusters which are associated with exocytosis. We reflect on recent studies which have revealed the mechanisms regulating syntaxin-1 nanoclustering on the plasma membrane and draw inferences on the effect of synaptic activity, phosphoinositides, N-ethylmaleimide-sensitive factor (NSF), α-soluble NSF attachment protein (α-SNAP) and SNARE complex assembly on the dynamic nanoscale organization of syntaxin-1. This article is part of the special issue entitled 'Mobility and trafficking of neuronal membrane proteins'.


Cell Membrane/metabolism , Exocytosis/physiology , Membrane Fusion/physiology , Nanoparticles/metabolism , Synapses/metabolism , Syntaxin 1/metabolism , Animals , Cell Membrane/chemistry , Humans , Markov Chains , Nanoparticles/analysis , Protein Binding/physiology , Protein Transport/physiology , Synapses/chemistry , Synaptic Vesicles/chemistry , Synaptic Vesicles/metabolism , Syntaxin 1/analysis
19.
J Am Chem Soc ; 141(44): 17507-17511, 2019 11 06.
Article En | MEDLINE | ID: mdl-31644274

Analytical tools for quantitative measurements of glutamate, the principal excitatory neurotransmitter in the brain, are lacking. Here, we introduce a new enzyme-based amperometric sensor technique for the counting of glutamate molecules stored inside single synaptic vesicles. In this method, an ultra-fast enzyme-based glutamate sensor is placed into a solution of isolated synaptic vesicles, which stochastically rupture at the sensor surface in a potential-dependent manner at a constant negative potential. The continuous amperometric signals are sampled at high speed (10 kHz) to record sub-millisecond spikes, which represent glutamate release from single vesicles that burst open. Glutamate quantification is achieved by a calibration curve that is based on measurements of glutamate release from vesicles pre-filled with various glutamate concentrations. Our measurements show that an isolated single synaptic vesicle encapsulates about 8000 glutamate molecules and is comparable to the measured exocytotic quantal glutamate release in amperometric glutamate sensing in the nucleus accumbens of mouse brain tissue. Hence, this new methodology introduces the means to quantify ultra-small amounts of glutamate and to study synaptic vesicle physiology, pathogenesis, and drug treatments for neuronal disorders where glutamate is involved.


Amino Acid Oxidoreductases/chemistry , Electrochemical Techniques/methods , Glutamic Acid/analysis , Neurotransmitter Agents/analysis , Synaptic Vesicles/chemistry , Animals , Brain Chemistry , Carbon/chemistry , Electrochemical Techniques/instrumentation , Electrodes , Glutamic Acid/chemistry , Gold/chemistry , Male , Metal Nanoparticles/chemistry , Mice, Inbred C57BL , Neurotransmitter Agents/chemistry , Rats, Sprague-Dawley , Unilamellar Liposomes/chemistry
20.
ACS Chem Neurosci ; 10(9): 3927-3938, 2019 09 18.
Article En | MEDLINE | ID: mdl-31394034

The synaptic vesicle glycoprotein 2 (SV2) family is comprised of three paralogues: SV2A, SV2B, and SV2C. In vertebrates, SV2s are 12-transmembrane proteins present on every secretory vesicle, including synaptic vesicles, and are critical to neurotransmission. Structural and functional studies suggest that SV2 proteins may play several roles to promote proper vesicular function. Among these roles are their potential to stabilize the transmitter content of vesicles, to maintain and orient the releasable pool of vesicles, and to regulate vesicular calcium sensitivity to ensure efficient, coordinated release of the transmitter. The SV2 family is highly relevant to human health in a number of ways. First, SV2A plays a role in neuronal excitability and as such is the specific target for the antiepileptic drug levetiracetam. SV2 proteins also act as the target by which potent neurotoxins, particularly botulinum, gain access to neurons and exert their toxicity. Both SV2B and SV2C are increasingly implicated in diseases such as Alzheimer's disease and Parkinson's disease. Interestingly, despite decades of intensive research, their exact function remains elusive. Thus, SV2 proteins are intriguing in their potentially diverse roles within the presynaptic terminal, and several recent developments have enhanced our understanding and appreciation of the protein family. Here, we review the structure and function of SV2 proteins as well as their relevance to disease and therapeutic development.


Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Nervous System Diseases/metabolism , Synaptic Vesicles/metabolism , Animals , Humans , Nervous System Diseases/pathology , Protein Transport/physiology , Synaptic Vesicles/chemistry , Synaptic Vesicles/pathology
...