Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.368
1.
J Physiol ; 602(12): 2873-2898, 2024 Jun.
Article En | MEDLINE | ID: mdl-38723211

Neurons in the central nervous system communicate with each other by activating billions of tiny synaptic boutons distributed along their fine axons. These presynaptic varicosities are very crowded environments, comprising hundreds of synaptic vesicles. Only a fraction of these vesicles can be recruited in a single release episode, either spontaneous or evoked by action potentials. Since the seminal work by Fatt and Katz, spontaneous release has been modelled as a memoryless process. Nevertheless, at central synapses, experimental evidence indicates more complex features, including non-exponential distributions of release intervals and power-law behaviour in their rate. To describe these features, we developed a probabilistic model of spontaneous release based on Brownian motion of synaptic vesicles in the presynaptic environment. To account for different diffusion regimes, we based our simulations on fractional Brownian motion. We show that this model can predict both deviation from the Poisson hypothesis and power-law features in experimental quantal release series, thus suggesting that the vesicular motion by diffusion could per se explain the emergence of these properties. We demonstrate the efficacy of our modelling approach using electrophysiological recordings at single synaptic boutons and ultrastructural data. When this approach was used to simulate evoked responses, we found that the replenishment of the readily releasable pool driven by Brownian motion of vesicles can reproduce the characteristic binomial release distributions seen experimentally. We believe that our modelling approach supports the idea that vesicle diffusion and readily releasable pool dynamics are crucial factors for the physiological functioning of neuronal communication. KEY POINTS: We developed a new probabilistic model of spontaneous and evoked vesicle fusion based on simple biophysical assumptions, including the motion of vesicles before they dock to the release site. We provide closed-form equations for the interval distribution of spontaneous releases in the special case of Brownian diffusion of vesicles, showing that a power-law heavy tail is generated. Fractional Brownian motion (fBm) was exploited to simulate anomalous vesicle diffusion, including directed and non-directed motion, by varying the Hurst exponent. We show that our model predicts non-linear features observed in experimental spontaneous quantal release series as well as ultrastructural data of synaptic vesicles spatial distribution. Evoked exocytosis based on a diffusion-replenished readily releasable pool might explain the emergence of power-law behaviour in neuronal activity.


Synaptic Transmission , Synaptic Vesicles , Synaptic Vesicles/physiology , Synaptic Vesicles/ultrastructure , Animals , Synaptic Transmission/physiology , Models, Neurological , Presynaptic Terminals/physiology , Presynaptic Terminals/ultrastructure , Rats , Diffusion
2.
Microsc Res Tech ; 87(7): 1647-1653, 2024 Jul.
Article En | MEDLINE | ID: mdl-38461470

The synaptic basal lamina of the electrocytes was disclosed to be electron-translucent to some extent when viewed in an en-face direction in embedment-free section transmission electron microscopy (EFS-TEM), and synaptic vesicles located close to the presynaptic membrane were seen through the synaptic basal lamina together with the presynaptic and postsynaptic membranes. This feature of translucency has the potential to analyze possible spatial interrelations in situ between bioactive molecules in the synaptic basal lamina and the synaptic vesicles in further studies. The synaptic basal lamina, appearing as an electron-dense line sandwiched by two parallel lines representing the presynaptic and postsynaptic membranes in ultrathin sections cut right to the synaptic junctional plane in conventional TEM, was not fully continuous but randomly intermittent along its trajectory. Compatible with the intermittent line appearance, the en-face 3D view in embedment-free section TEM revealed for the first time partial irregular defects of the synaptic basal lamina. Considering the known functional significance of several molecules contained in the synaptic basal lamina in the maintenance and exertion of the synapse, its partial defects may not represent its rigid structural features, but its immature structure under remodeling or its dynamic changes in consistency such as the sol/gel transition, whose validity needs further examination. RESEARCH HIGHLIGHTS: In embedment-free section TEM, a 3D en-face view of synaptic basal lamina in situ is reliably possible. The basal lamina en-face is electron-translucent, which makes it possible to analyze spatial interrelation between pre- and post-synaptic components. Partial irregular defects in the basal lamina are revealed in Torpedo electrocytes, suggesting its remodeling or dynamic changes in consistency.


Microscopy, Electron, Transmission , Animals , Microscopy, Electron, Transmission/methods , Synaptic Vesicles/ultrastructure , Electrical Synapses/ultrastructure , Electrical Synapses/physiology , Synapses/ultrastructure , Synaptic Membranes/ultrastructure , Imaging, Three-Dimensional/methods
3.
J Biol Chem ; 299(11): 105282, 2023 11.
Article En | MEDLINE | ID: mdl-37742923

The intracellular domains of connexins are essential for the assembly of gap junctions. For connexin 36 (Cx36), the major neuronal connexin, it has been shown that a dysfunctional PDZ-binding motif interferes with electrical synapse formation. However, it is still unknown how this motif coordinates the transport of Cx36. In the present study, we characterize a phenotype of Cx36 mutants that lack a functional PDZ-binding motif using HEK293T cells as an expression system. We provide evidence that an intact PDZ-binding motif is critical for proper endoplasmic reticulum (ER) export of Cx36. Removing the PDZ-binding motif of Cx36 results in ER retention and the formation of multimembrane vesicles containing gap junction-like connexin aggregates. Using a combination of site-directed mutagenesis and electron micrographs, we reveal that these vesicles consist of Cx36 channels that docked prematurely in the ER. Our data suggest a model in which ER-retained Cx36 channels reshape the ER membrane into concentric whorls that are released into the cytoplasm.


Connexins , Endoplasmic Reticulum , Gap Junctions , Humans , Connexins/genetics , Connexins/metabolism , Endoplasmic Reticulum/metabolism , Gap Junctions/metabolism , HEK293 Cells , Protein Domains , Amino Acid Motifs , Electrical Synapses/physiology , Mutation , Protein Transport/genetics , Synaptic Vesicles/pathology , Synaptic Vesicles/ultrastructure , Microscopy, Electron, Scanning , Gap Junction delta-2 Protein
4.
Elife ; 112022 12 23.
Article En | MEDLINE | ID: mdl-36562477

Ribbon synapses of cochlear inner hair cells (IHCs) are specialized to indefatigably transmit sound information at high rates. To understand the underlying mechanisms, structure-function analysis of the active zone (AZ) of these synapses is essential. Previous electron microscopy studies of synaptic vesicle (SV) dynamics at the IHC AZ used potassium stimulation, which limited the temporal resolution to minutes. Here, we established optogenetic IHC stimulation followed by quick freezing within milliseconds and electron tomography to study the ultrastructure of functional synapse states with good temporal resolution in mice. We characterized optogenetic IHC stimulation by patch-clamp recordings from IHCs and postsynaptic boutons revealing robust IHC depolarization and neurotransmitter release. Ultrastructurally, the number of docked SVs increased upon short (17-25 ms) and long (48-76 ms) light stimulation paradigms. We did not observe enlarged SVs or other morphological correlates of homotypic fusion events. Our results indicate a rapid recruitment of SVs to the docked state upon stimulation and suggest that univesicular release prevails as the quantal mechanism of exocytosis at IHC ribbon synapses.


Electron Microscope Tomography , Optogenetics , Mice , Animals , Synapses/physiology , Synaptic Vesicles/ultrastructure , Hair Cells, Auditory, Inner/physiology , Exocytosis/physiology
5.
Cell Rep ; 40(12): 111382, 2022 09 20.
Article En | MEDLINE | ID: mdl-36130490

Rab3A-interacting molecule (RIM) is crucial for fast Ca2+-triggered synaptic vesicle (SV) release in presynaptic active zones (AZs). We investigated hippocampal giant mossy fiber bouton (MFB) AZ architecture in 3D using electron tomography of rapid cryo-immobilized acute brain slices in RIM1α-/- and wild-type mice. In RIM1α-/-, AZs are larger with increased synaptic cleft widths and a 3-fold reduced number of tightly docked SVs (0-2 nm). The distance of tightly docked SVs to the AZ center is increased from 110 to 195 nm, and the width of their electron-dense material between outer SV membrane and AZ membrane is reduced. Furthermore, the SV pool in RIM1α-/- is more heterogeneous. Thus, RIM1α, besides its role in tight SV docking, is crucial for synaptic architecture and vesicle pool organization in MFBs.


Synapses , Synaptic Vesicles , Animals , Mice , Mossy Fibers, Hippocampal/ultrastructure , Presynaptic Terminals/ultrastructure , Synapses/ultrastructure , Synaptic Transmission , Synaptic Vesicles/ultrastructure
6.
Sci Rep ; 12(1): 1410, 2022 01 26.
Article En | MEDLINE | ID: mdl-35082358

Dp40 is ubiquitously expressed including the central nervous system. In addition to being present in the nucleus, membrane, and cytoplasm, Dp40 is detected in neurites and postsynaptic spines in hippocampal neurons. Although Dp40 is expressed from the same promoter as Dp71, its role in the cognitive impairment present in Duchenne muscular dystrophy patients is still unknown. Here, we studied the effects of overexpression of Dp40 and Dp40L170P during the neuronal differentiation of PC12 Tet-On cells. We found that Dp40 overexpression increased the percentage of PC12 cells with neurites and neurite length, while Dp40L170P overexpression decreased them compared to Dp40 overexpression. Two-dimensional gel electrophoresis analysis showed that the protein expression profile was modified in nerve growth factor-differentiated PC12-Dp40L170P cells compared to that of the control cells (PC12 Tet-On). The proteins α-internexin and S100a6, involved in cytoskeletal structure, were upregulated. The expression of vesicle-associated membrane proteins increased in differentiated PC12-Dp40 cells, in contrast to PC12-Dp40L170P cells, while neurofilament light-chain was decreased in both differentiated cells. These results suggest that Dp40 has an important role in the neuronal differentiation of PC12 cells through the regulation of proteins involved in neurofilaments and exocytosis of synaptic vesicles, functions that might be affected in PC12-Dp40L170P.


Amino Acid Substitution , Dystrophin/genetics , Intermediate Filaments/metabolism , Neuronal Outgrowth/genetics , Neurons/metabolism , Synaptic Vesicles/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Dystrophin/metabolism , Exocytosis , Gene Expression Regulation , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Intermediate Filaments/ultrastructure , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/cytology , PC12 Cells , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , S100 Calcium Binding Protein A6/genetics , S100 Calcium Binding Protein A6/metabolism , Signal Transduction , Synaptic Vesicles/ultrastructure
7.
Cereb Cortex ; 32(9): 1840-1865, 2022 04 20.
Article En | MEDLINE | ID: mdl-34530440

Synapses "govern" the computational properties of any given network in the brain. However, their detailed quantitative morphology is still rather unknown, particularly in humans. Quantitative 3D-models of synaptic boutons (SBs) in layer (L)6a and L6b of the temporal lobe neocortex (TLN) were generated from biopsy samples after epilepsy surgery using fine-scale transmission electron microscopy, 3D-volume reconstructions and electron microscopic tomography. Beside the overall geometry of SBs, the size of active zones (AZs) and that of the three pools of synaptic vesicles (SVs) were quantified. SBs in L6 of the TLN were middle-sized (~5 µm2), the majority contained only a single but comparatively large AZ (~0.20 µm2). SBs had a total pool of ~1100 SVs with comparatively large readily releasable (RRP, ~10 SVs L6a), (RRP, ~15 SVs L6b), recycling (RP, ~150 SVs), and resting (~900 SVs) pools. All pools showed a remarkably large variability suggesting a strong modulation of short-term synaptic plasticity. In conclusion, L6 SBs are highly reliable in synaptic transmission within the L6 network in the TLN and may act as "amplifiers," "integrators" but also as "discriminators" for columnar specific, long-range extracortical and cortico-thalamic signals from the sensory periphery.


Neocortex , Presynaptic Terminals , Adult , Humans , Neocortex/ultrastructure , Presynaptic Terminals/ultrastructure , Synapses/ultrastructure , Synaptic Transmission , Synaptic Vesicles/ultrastructure , Temporal Lobe/ultrastructure
8.
Neurosci Lett ; 771: 136390, 2022 02 06.
Article En | MEDLINE | ID: mdl-34896437

Detrimental consequences following exposure to severe stress, either acute or chronic are well recognized. Chronic mild stress (CMS) is also a leading cause of emotional distress and neuropsychiatric conditions such as anxiety disorders. However, the neurobiological substrates of the latter, particularly at the ultrastructural levels have not been adequately investigated. In this study, adult male Wistar rats were subjected to 4 h daily mild restraint for 20 days and their behavior in open field and elevated plus maze (EPM) were evaluated 24 h after the last restraint. Anxiety-like behavior was evident in CMS exposed rats by increases in rearing and grooming in the open field and the avoidance of open arms in the EPM. Concomitant ultrastructural alterations such as chromatolysis, agglutination of synaptic vesicles or mitochondrial damage were also observed in the central nucleus of amygdala (CNA), an area intimately involved in emotional and fear response, in CMS exposed rats. These results while confirming detrimental consequences of CMS, also suggest that ultrastructural alterations in CNA may be a basis for CMS-induced anxiety.


Amygdala/ultrastructure , Anxiety/pathology , Stress, Psychological/pathology , Amygdala/physiopathology , Animals , Anxiety/etiology , Anxiety/physiopathology , Male , Maze Learning , Mitochondria/ultrastructure , Rats , Rats, Wistar , Stress, Psychological/complications , Stress, Psychological/physiopathology , Synaptic Vesicles/ultrastructure
9.
Nature ; 599(7883): 147-151, 2021 11.
Article En | MEDLINE | ID: mdl-34616045

Understanding cellular architecture is essential for understanding biology. Electron microscopy (EM) uniquely visualizes cellular structures with nanometre resolution. However, traditional methods, such as thin-section EM or EM tomography, have limitations in that they visualize only a single slice or a relatively small volume of the cell, respectively. Focused ion beam-scanning electron microscopy (FIB-SEM) has demonstrated the ability to image small volumes of cellular samples with 4-nm isotropic voxels1. Owing to advances in the precision and stability of FIB milling, together with enhanced signal detection and faster SEM scanning, we have increased the volume that can be imaged with 4-nm voxels by two orders of magnitude. Here we present a volume EM atlas at such resolution comprising ten three-dimensional datasets for whole cells and tissues, including cancer cells, immune cells, mouse pancreatic islets and Drosophila neural tissues. These open access data (via OpenOrganelle2) represent the foundation of a field of high-resolution whole-cell volume EM and subsequent analyses, and we invite researchers to explore this atlas and pose questions.


Datasets as Topic , Information Dissemination , Microscopy, Electron, Scanning , Organelles/ultrastructure , Animals , Cell Line , Cells, Cultured , Drosophila melanogaster/cytology , Drosophila melanogaster/ultrastructure , Female , Golgi Apparatus/ultrastructure , Humans , Interphase , Islets of Langerhans/cytology , Male , Mice , Microscopy, Electron, Scanning/methods , Microscopy, Electron, Scanning/standards , Microtubules/ultrastructure , Neuroglia/ultrastructure , Neurons/ultrastructure , Open Access Publishing , Ovarian Neoplasms/immunology , Ovarian Neoplasms/ultrastructure , Ribosomes/ultrastructure , Synaptic Vesicles/ultrastructure , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure
10.
Elife ; 102021 09 20.
Article En | MEDLINE | ID: mdl-34543184

Synaptotagmin 7 (SYT7) has emerged as a key regulator of presynaptic function, but its localization and precise role in the synaptic vesicle cycle remain the subject of debate. Here, we used iGluSnFR to optically interrogate glutamate release, at the single-bouton level, in SYT7KO-dissociated mouse hippocampal neurons. We analyzed asynchronous release, paired-pulse facilitation, and synaptic vesicle replenishment and found that SYT7 contributes to each of these processes to different degrees. 'Zap-and-freeze' electron microscopy revealed that a loss of SYT7 diminishes docking of synaptic vesicles after a stimulus and inhibits the recovery of depleted synaptic vesicles after a stimulus train. SYT7 supports these functions from the axonal plasma membrane, where its localization and stability require both γ-secretase-mediated cleavage and palmitoylation. In summary, SYT7 is a peripheral membrane protein that controls multiple modes of synaptic vesicle (SV) exocytosis and plasticity, in part, through enhancing activity-dependent docking of SVs.


Amyloid Precursor Protein Secretases/metabolism , Axons/enzymology , Cell Membrane/enzymology , Hippocampus/enzymology , Synaptic Vesicles/enzymology , Synaptotagmins/metabolism , Animals , Axons/ultrastructure , Cell Membrane/ultrastructure , Cells, Cultured , Exocytosis , Hippocampus/ultrastructure , Lipoylation , Mice, Knockout , Molecular Docking Simulation , Neuronal Plasticity , Protein Processing, Post-Translational , Protein Transport , Proteolysis , Rats, Sprague-Dawley , Synaptic Transmission , Synaptic Vesicles/ultrastructure , Synaptotagmins/genetics , Time Factors
11.
Cell Rep ; 36(1): 109333, 2021 07 06.
Article En | MEDLINE | ID: mdl-34233191

While misfolding of alpha-synuclein (αSyn) is central to the pathogenesis of Parkinson's disease (PD), fundamental questions about its structure and function at the synapse remain unanswered. We examine synaptosomes from non-transgenic and transgenic mice expressing wild-type human αSyn, the E46K fPD-causing mutation, or an amplified form of E46K ("3K"). Synaptosomes from mice expressing the 3K mutant show reduced Ca2+-dependent vesicle exocytosis, altered synaptic vesicle ultrastructure, decreased SNARE complexes, and abnormal levels of certain synaptic proteins. With our intra-synaptosomal nuclear magnetic resonance (NMR) method, we reveal that WT αSyn participates in heterogeneous interactions with synaptic components dependent on endogenous αSyn and synaptosomal integrity. The 3K mutation markedly alters these interactions. The synaptic microenvironment is necessary for αSyn to reach its native conformations and establish a physiological interaction network. Its inability to populate diverse conformational ensembles likely represents an early step in αSyn dysfunction that contributes to the synaptotoxicity observed in synucleinopathies.


Parkinson Disease/metabolism , Parkinson Disease/pathology , Synaptic Vesicles/pathology , Synaptosomes/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Animals , Brain/pathology , Calcium/metabolism , Disease Models, Animal , Exocytosis , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Spectroscopy , Models, Biological , Protein Conformation , Protein Folding , Protein Multimerization , Recombinant Proteins/metabolism , SNARE Proteins/metabolism , Solubility , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomes/ultrastructure
12.
Neuroimage ; 239: 118302, 2021 10 01.
Article En | MEDLINE | ID: mdl-34174391

The positron emission tomography (PET) radioligand [11C]UCB-J binds to synaptic vesicle protein 2A (SV2A) and is used to investigate synaptic density in the living brain. Clinical studies have indicated reduced [11C]UCB-J binding in Alzheimer's disease (AD) and Parkinson's disease (PD) brains compared to healthy controls. Still, it is unknown whether [11C]UCB-J PET can visualise synaptic loss in mouse models of these disorders. Such models are essential for understanding disease pathology and for evaluating the effects of novel disease-modifying drug candidates. In the present study, synaptic density in transgenic models of AD (ArcSwe) and PD (L61) was studied using [11C]UCB-J PET. Data were acquired during 60 min after injection, and time-activity curves (TACs) in different brain regions and the left ventricle of the heart were generated based on the dynamic PET images. The [11C]UCB-J brain concentrations were expressed as standardised uptake value (SUV) over time. The area under the SUV curve (AUC), the ratio of AUC in the brain to that in the heart (AUCbrain/blood), and the volume of distribution (VT) obtained by kinetic modelling using the heart TAC as input were compared between transgenic and age-matched wild type (WT) mice. The L61 mice displayed 11-13% lower AUCbrain/blood ratio and brain VT generated by kinetic modeling compared to the control WT mice. In general, also transgenic ArcSwe mice tended to show lower [11C]UCB-J brain exposure than age-matched WT controls, but variation within the different animal groups was high. Older WT mice (18-20 months) showed lower [11C]UCB-J brain exposure than younger WT mice (8-9 months). Together, these data imply that [11C]UCB-J PET reflects synaptic density in mouse models of neurodegeneration and that inter-subject variation is large. In addition, the study suggested that model-independent AUCbrain/blood ratio can be used to evaluate [11C]UCB-J binding as an alternative to full pharmacokinetic modelling.


Amyloid beta-Peptides/analysis , Brain/diagnostic imaging , Carbon Radioisotopes/pharmacokinetics , Disease Models, Animal , Membrane Glycoproteins/analysis , Nerve Tissue Proteins/analysis , Peptide Fragments/analysis , Positron-Emission Tomography/methods , Pyridines/pharmacokinetics , Pyrrolidinones/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Synaptic Vesicles/ultrastructure , Synucleinopathies/diagnostic imaging , Aging , Alzheimer Disease , Amyloid beta-Peptides/genetics , Animals , Area Under Curve , Brain/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity , Parkinson Disease , Peptide Fragments/genetics
13.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Article En | MEDLINE | ID: mdl-33875591

Long-term potentiation (LTP) is a cellular mechanism of learning and memory that results in a sustained increase in the probability of vesicular release of neurotransmitter. However, previous work in hippocampal area CA1 of the adult rat revealed that the total number of vesicles per synapse decreases following LTP, seemingly inconsistent with the elevated release probability. Here, electron-microscopic tomography (EMT) was used to assess whether changes in vesicle density or structure of vesicle tethering filaments at the active zone might explain the enhanced release probability following LTP. The spatial relationship of vesicles to the active zone varies with functional status. Tightly docked vesicles contact the presynaptic membrane, have partially formed SNARE complexes, and are primed for release of neurotransmitter upon the next action potential. Loosely docked vesicles are located within 8 nm of the presynaptic membrane where SNARE complexes begin to form. Nondocked vesicles comprise recycling and reserve pools. Vesicles are tethered to the active zone via filaments composed of molecules engaged in docking and release processes. The density of tightly docked vesicles was increased 2 h following LTP compared to control stimulation, whereas the densities of loosely docked or nondocked vesicles congregating within 45 nm above the active zones were unchanged. The tethering filaments on all vesicles were shorter and their attachment sites shifted closer to the active zone. These findings suggest that tethering filaments stabilize more vesicles in the primed state. Such changes would facilitate the long-lasting increase in release probability following LTP.


Hippocampus/physiology , Long-Term Potentiation/physiology , Synaptic Vesicles/ultrastructure , Animals , Brain/metabolism , Brain/physiology , Cytoskeleton , Electron Microscope Tomography/methods , Hippocampus/metabolism , Long-Term Potentiation/genetics , Male , Neurotransmitter Agents , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Rats , Rats, Long-Evans , Synapses/physiology , Synaptic Membranes/physiology , Synaptic Membranes/ultrastructure , Synaptic Vesicles/physiology
14.
Cell Rep ; 34(11): 108842, 2021 03 16.
Article En | MEDLINE | ID: mdl-33730593

Synaptic vesicle (SV) docking is a dynamic multi-stage process that is required for efficient neurotransmitter release in response to nerve impulses. Although the steady-state SV docking likely involves the cooperation of Synaptotagmin-1 (Syt1) and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), where and how the docking process initiates remains unknown. Phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) can interact with Syt1 and SNAREs to contribute to vesicle exocytosis. In the present study, using the CRISPRi-mediated multiplex gene knockdown and 3D electron tomography approaches, we show that in mouse hippocampal synapses, SV docking initiates at ∼12 nm to the active zone (AZ) by Syt1. Furthermore, we demonstrate that PI(4,5)P2 is the membrane partner of Syt1 to initiate SV docking, and disrupting their interaction could abolish the docking initiation. In contrast, the SNARE complex contributes only to the tight SV docking within 0-2 nm. Therefore, Syt1 interacts with PI(4,5)P2 to loosely dock SVs within 2-12 nm to the AZ in hippocampal neurons.


Hippocampus/cytology , Neurons/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Synaptic Vesicles/metabolism , Synaptotagmin I/metabolism , Animals , HEK293 Cells , Humans , Mice, Inbred C57BL , Neurons/ultrastructure , Protein Binding , SNARE Proteins/metabolism , Synaptic Vesicles/ultrastructure , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Vesicle-Associated Membrane Protein 2/metabolism
15.
Hum Mol Genet ; 30(13): 1175-1187, 2021 06 17.
Article En | MEDLINE | ID: mdl-33601422

Synaptic dysfunction and cognitive decline in Huntington's disease (HD) involve hyperactive A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10). To identify the molecular mechanisms through which ADAM10 is associated with synaptic dysfunction in HD, we performed an immunoaffinity purification-mass spectrometry (IP-MS) study of endogenous ADAM10 in the brains of wild-type and HD mice. We found that proteins implicated in synapse organization, synaptic plasticity, and vesicle and organelles trafficking interact with ADAM10, suggesting that it may act as hub protein at the excitatory synapse. Importantly, the ADAM10 interactome is enriched in presynaptic proteins and ADAM10 co-immunoprecipitates with piccolo (PCLO), a key player in the recycling and maintenance of synaptic vesicles. In contrast, reduced ADAM10/PCLO immunoprecipitation occurs in the HD brain, with decreased density of synaptic vesicles in the reserve and docked pools at the HD presynaptic terminal. Conditional heterozygous deletion of ADAM10 in the forebrain of HD mice reduces active ADAM10 to wild-type level and normalizes ADAM10/PCLO complex formation and synaptic vesicle density and distribution. The results indicate that presynaptic ADAM10 and PCLO are a relevant component of HD pathogenesis.


ADAM10 Protein/metabolism , Cytoskeletal Proteins/metabolism , Huntington Disease/metabolism , Neuropeptides/metabolism , Synaptic Vesicles/metabolism , ADAM10 Protein/genetics , Animals , Blotting, Western , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Humans , Huntington Disease/genetics , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Presynaptic Terminals/metabolism , Protein Binding , Protein Interaction Maps/genetics , Proteomics/methods , Synaptic Vesicles/ultrastructure , Synaptosomes/metabolism , Tandem Mass Spectrometry/methods
16.
Nat Commun ; 12(1): 858, 2021 02 08.
Article En | MEDLINE | ID: mdl-33558502

Synaptic vesicles are storage organelles for neurotransmitters. They pass through a trafficking cycle and fuse with the pre-synaptic membrane when an action potential arrives at the nerve terminal. While molecular components and biophysical parameters of synaptic vesicles have been determined, our knowledge on the protein interactions in their membranes is limited. Here, we apply cross-linking mass spectrometry to study interactions of synaptic vesicle proteins in an unbiased approach without the need for specific antibodies or detergent-solubilisation. Our large-scale analysis delivers a protein network of vesicle sub-populations and functional assemblies including an active and an inactive conformation of the vesicular ATPase complex as well as non-conventional arrangements of the luminal loops of SV2A, Synaptophysin and structurally related proteins. Based on this network, we specifically target Synaptobrevin-2, which connects with many proteins, in different approaches. Our results allow distinction of interactions caused by 'crowding' in the vesicle membrane from stable interaction modules.


Cross-Linking Reagents/chemistry , Mass Spectrometry , Synaptic Membranes/metabolism , Synaptic Vesicles/metabolism , Animals , Brain/metabolism , Membrane Fusion , Protein Binding , Protein Interaction Maps , Proteolipids , Proteome/metabolism , Rats , Synaptic Membranes/ultrastructure , Synaptic Vesicles/ultrastructure , Synaptophysin/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vesicle-Associated Membrane Protein 2/metabolism
17.
Nat Commun ; 12(1): 263, 2021 01 11.
Article En | MEDLINE | ID: mdl-33431828

Clusters of tightly packed synaptic vesicles (SVs) are a defining feature of nerve terminals. While SVs are mobile within the clusters, the clusters have no boundaries consistent with a liquid phase. We previously found that purified synapsin, a peripheral SV protein, can assemble into liquid condensates and trap liposomes into them. How this finding relates to the physiological formation of SV clusters in living cells remains unclear. Here, we report that synapsin alone, when expressed in fibroblasts, has a diffuse cytosolic distribution. However, when expressed together with synaptophysin, an integral SV membrane protein previously shown to be localized on small synaptic-like microvesicles when expressed in non-neuronal cells, is sufficient to organize such vesicles in clusters highly reminiscent of SV clusters and with liquid-like properties. This minimal reconstitution system can be a powerful model to gain mechanistic insight into the assembly of structures which are of fundamental importance in synaptic transmission.


Neurons/metabolism , Synapsins/metabolism , Synaptic Vesicles/metabolism , Synaptophysin/metabolism , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Chlorocebus aethiops , Cytosol/metabolism , Endocytosis , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Mice , Static Electricity , Synaptic Vesicles/ultrastructure
18.
Proc Natl Acad Sci U S A ; 118(5)2021 02 02.
Article En | MEDLINE | ID: mdl-33468631

Controlled release of neurotransmitters stored in synaptic vesicles (SVs) is a fundamental process that is central to all information processing in the brain. This relies on tight coupling of the SV fusion to action potential-evoked presynaptic Ca2+ influx. This Ca2+-evoked release occurs from a readily releasable pool (RRP) of SVs docked to the plasma membrane (PM). The protein components involved in initial SV docking/tethering and the subsequent priming reactions which make the SV release ready are known. Yet, the supramolecular architecture and sequence of molecular events underlying SV release are unclear. Here, we use cryoelectron tomography analysis in cultured hippocampal neurons to delineate the arrangement of the exocytosis machinery under docked SVs. Under native conditions, we find that vesicles are initially "tethered" to the PM by a variable number of protein densities (∼10 to 20 nm long) with no discernible organization. In contrast, we observe exactly six protein masses, each likely consisting of a single SNAREpin with its bound Synaptotagmins and Complexin, arranged symmetrically connecting the "primed" vesicles to the PM. Our data indicate that the fusion machinery is likely organized into a highly cooperative framework during the priming process which enables rapid SV fusion and neurotransmitter release following Ca2+ influx.


Nerve Tissue Proteins/metabolism , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Cryoelectron Microscopy , Hippocampus/cytology , Imaging, Three-Dimensional , Mice, Inbred C57BL , Neurons/metabolism , Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Synaptic Vesicles/ultrastructure
19.
J Neurosci ; 41(6): 1174-1190, 2021 02 10.
Article En | MEDLINE | ID: mdl-33303681

The BAD-BAX-caspase-3 cascade is a canonical apoptosis pathway. Macroautophagy ("autophagy" hereinafter) is a process by which organelles and aggregated proteins are delivered to lysosomes for degradation. Here, we report a new function of the BAD-BAX-caspase-3 cascade and autophagy in the control of synaptic vesicle pools. We found that, in hippocampal neurons of male mice, the BAD-BAX-caspase-3 pathway regulates autophagy, which in turn limits the size of synaptic vesicle pools and influences the kinetics of activity-induced depletion and recovery of synaptic vesicle pools. Moreover, the caspase-autophagy pathway is engaged by fear conditioning to facilitate associative fear learning and memory. This work identifies a new mechanism for controlling synaptic vesicle pools, and a novel, nonapoptotic, presynaptic function of the BAD-BAX-caspase-3 cascade.SIGNIFICANCE STATEMENT Despite the importance of synaptic vesicles for neurons, little is known about how the size of synaptic vesicle pools is maintained under basal conditions and regulated by neural activity. This study identifies a new mechanism for the control of synaptic vesicle pools, and a new, nonapoptotic function of the BAD-BAX-caspase-3 pathway in presynaptic terminals. Additionally, it indicates that autophagy is not only a homeostatic mechanism to maintain the integrity of cells and tissues, but also a process engaged by neural activity to regulate synaptic vesicle pools for optimal synaptic responses, learning, and memory.


Autophagy/physiology , Caspase 3/deficiency , Signal Transduction/physiology , Synaptic Vesicles/metabolism , bcl-2-Associated X Protein/deficiency , bcl-Associated Death Protein/deficiency , Animals , Caspase 3/genetics , Cells, Cultured , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Molecular Imaging/methods , Organ Culture Techniques , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure , bcl-2-Associated X Protein/genetics , bcl-Associated Death Protein/genetics
20.
Methods Mol Biol ; 2233: 101-111, 2021.
Article En | MEDLINE | ID: mdl-33222130

The efficient recycling of synaptic vesicles (SVs) during neuronal activity is central for sustaining brain function. During intense neuronal activity, the dominant mechanism of SV retrieval is activity-dependent bulk endocytosis (ADBE). Here, we describe a method to monitor ADBE in isolation from other SV endocytosis modes, via the uptake of large fluorescent fluid-phase markers in primary neuronal culture. Furthermore, we outline how to monitor ADBE using this approach across a field of neurons or in individual neurons.


Endocytosis/genetics , Neurons/ultrastructure , Primary Cell Culture/methods , Synaptic Vesicles/ultrastructure , Animals , Dextrans/pharmacology , Endosomes/drug effects , Endosomes/ultrastructure , Fluorescent Dyes/pharmacology , Humans , Mice , Neurons/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Synaptic Vesicles/drug effects
...