Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO J ; 37(20)2018 10 15.
Article in English | MEDLINE | ID: mdl-30185408

ABSTRACT

Neuropeptides are essential signaling molecules transported and secreted by dense-core vesicles (DCVs), but the number of DCVs available for secretion, their subcellular distribution, and release probability are unknown. Here, we quantified DCV pool sizes in three types of mammalian CNS neurons in vitro and in vivo Super-resolution and electron microscopy reveal a total pool of 1,400-18,000 DCVs, correlating with neurite length. Excitatory hippocampal and inhibitory striatal neurons in vitro have a similar DCV density, and thalamo-cortical axons in vivo have a slightly higher density. Synapses contain on average two to three DCVs, at the periphery of synaptic vesicle clusters. DCVs distribute equally in axons and dendrites, but the vast majority (80%) of DCV fusion events occur at axons. The release probability of DCVs is 1-6%, depending on the stimulation. Thus, mammalian CNS neurons contain a large pool of DCVs of which only a small fraction can fuse, preferentially at axons.


Subject(s)
Axons , Corpus Striatum , Hippocampus , Neurites , Secretory Vesicles , Synapses , Animals , Axons/metabolism , Axons/ultrastructure , Corpus Striatum/metabolism , Corpus Striatum/ultrastructure , Hippocampus/metabolism , Hippocampus/ultrastructure , Mice , Neurites/metabolism , Neurites/ultrastructure , Secretory Vesicles/metabolism , Secretory Vesicles/ultrastructure , Synapses/metabolism , Synapses/ultrastructure
2.
Nat Commun ; 8: 15915, 2017 06 21.
Article in English | MEDLINE | ID: mdl-28635948

ABSTRACT

Synaptic transmission requires a stable pool of release-ready (primed) vesicles. Here we show that two molecules involved in SNARE-complex assembly, Munc13-1 and Munc18-1, together stabilize release-ready vesicles by preventing de-priming. Replacing neuronal Munc18-1 by a non-neuronal isoform Munc18-2 (Munc18-1/2SWAP) supports activity-dependent priming, but primed vesicles fall back into a non-releasable state (de-prime) within seconds. Munc13-1 deficiency produces a similar defect. Inhibitors of N-ethylmaleimide sensitive factor (NSF), N-ethylmaleimide (NEM) or interfering peptides, prevent de-priming in munc18-1/2SWAP or munc13-1 null synapses, but not in CAPS-1/2 null, another priming-deficient mutant. NEM rescues synaptic transmission in munc13-1 null and munc18-1/2SWAP synapses, in acute munc13-1 null slices and even partially in munc13-1/2 double null synapses. Together these data indicate that Munc13-1 and Munc18-1, but not CAPS-1/2, stabilize primed synaptic vesicles by preventing NSF-dependent de-priming.


Subject(s)
Munc18 Proteins/metabolism , N-Ethylmaleimide-Sensitive Proteins/metabolism , Nerve Tissue Proteins/metabolism , Synaptic Vesicles/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Female , Mice , Mice, Inbred C57BL , Munc18 Proteins/genetics , N-Ethylmaleimide-Sensitive Proteins/genetics , Nerve Tissue Proteins/genetics , SNARE Proteins/genetics , SNARE Proteins/metabolism , Synapses/metabolism , Synaptic Transmission , Synaptic Vesicles/genetics
3.
J Neurosci ; 37(17): 4525-4539, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28348137

ABSTRACT

The loss of presynaptic proteins Munc18-1, syntaxin-1, or SNAP-25 is known to produce cell death, but the underlying features have not been compared experimentally. Here, we investigated these features in cultured mouse CNS and DRG neurons. Side-by-side comparisons confirmed massive cell death, before synaptogenesis, within 1-4 DIV upon loss of t-SNAREs (syntaxin-1, SNAP-25) or Munc18-1, but not v-SNAREs (synaptobrevins/VAMP1/2/3 using tetanus neurotoxin (TeNT), also in TI-VAMP/VAMP7 knock-out (KO) neurons). A condensed cis-Golgi was the first abnormality observed upon Munc18-1 or SNAP-25 loss within 3 DIV. This phenotype was distinct from the Golgi fragmentation observed in apoptosis. Cell death was too rapid after syntaxin-1 loss to study Golgi abnormalities. Syntaxin-1 and Munc18-1 depend on each other for normal cellular levels. We observed that endogenous syntaxin-1 accumulates at the Golgi of Munc18-1 KO neurons. However, expression of a non-neuronal Munc18 isoform that does not bind syntaxin-1, Munc18-3, in Munc18-1 KO neurons prevented cell death and restored normal cis-Golgi morphology, but not synaptic transmission or syntaxin-1 targeting. Finally, we observed that DRG neurons are the only Munc18-1 KO neurons that do not degenerate in vivo or in vitro In these neurons, cis-Golgi abnormalities were less severe, with no changes in Golgi shape. Together, these data demonstrate that cell death upon Munc18-1, syntaxin-1, or SNAP-25 loss occurs via a degenerative pathway unrelated to the known synapse function of these proteins and involving early cis-Golgi abnormalities, distinct from apoptosis.SIGNIFICANCE STATEMENT This study provides new insights in a neurodegeneration pathway triggered by the absence of specific proteins involved in synaptic transmission (syntaxin-1, Munc18-1, SNAP-25), whereas other proteins involved in the same molecular process (synaptobrevins, Munc13-1/2) do not cause degeneration. Massive cell death occurs in cultured neurons upon depleting syntaxin-1, Munc18-1, and/or SNAP-25, well before synapse formation. This study characterizes several relevant cellular phenotypes, especially early cis-Golgi abnormalities, distinct from abnormalities observed during apoptosis, and rules out several other phenotypes as causal (defects in syntaxin-1 targeting and synaptic transmission). As proteins, such as syntaxin-1, Munc18-1, or SNAP-25, modulate α-synuclein neuropathy and/or are dysregulated in Alzheimer's disease, understanding this type of neurodegeneration may provide new links between synaptic defects and neurodegeneration in humans.


Subject(s)
Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Munc18 Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Synaptosomal-Associated Protein 25/genetics , Syntaxin 1/genetics , Animals , Apoptosis , Cell Death/genetics , Cells, Cultured , Exocytosis/genetics , Golgi Apparatus/pathology , Mice , Mice, Knockout , Neurodegenerative Diseases/pathology , Synapses/pathology
5.
Elife ; 42015 Feb 26.
Article in English | MEDLINE | ID: mdl-25719439

ABSTRACT

Neuropeptides released from dense-core vesicles (DCVs) modulate neuronal activity, but the molecules driving DCV secretion in mammalian neurons are largely unknown. We studied the role of calcium-activator protein for secretion (CAPS) proteins in neuronal DCV secretion at single vesicle resolution. Endogenous CAPS-1 co-localized with synaptic markers but was not enriched at every synapse. Deletion of CAPS-1 and CAPS-2 did not affect DCV biogenesis, loading, transport or docking, but DCV secretion was reduced by 70% in CAPS-1/CAPS-2 double null mutant (DKO) neurons and remaining fusion events required prolonged stimulation. CAPS deletion specifically reduced secretion of stationary DCVs. CAPS-1-EYFP expression in DKO neurons restored DCV secretion, but CAPS-1-EYFP and DCVs rarely traveled together. Synaptic localization of CAPS-1-EYFP in DKO neurons was calcium dependent and DCV fusion probability correlated with synaptic CAPS-1-EYFP expression. These data indicate that CAPS-1 promotes fusion competence of immobile (tethered) DCVs in presynaptic terminals and that CAPS-1 localization to DCVs is probably not essential for this role.


Subject(s)
Calcium-Binding Proteins/physiology , Nerve Tissue Proteins/physiology , Neurons/physiology , Presynaptic Terminals/physiology , Animals , Hippocampus/cytology , Mice , Neurons/cytology , Synaptic Transmission
6.
J Neurosci Methods ; 236: 1-10, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25109903

ABSTRACT

BACKGROUND: Adrenal chromaffin cells are a widely used model system to study regulated exocytosis and other membrane-associated processes. Alterations in the amount and localization of the proteins involved in these processes can be visualized with fluorescent probes that report the effect of different stimuli or genetic modifications. However, the quantitative analysis of such images remains difficult, especially when focused on specific locations, such as the plasma membrane. NEW METHOD: We developed an image analysis algorithm, named plasma membrane analysis in chromaffin cells (PlasMACC). PlasMACC enables automatic detection of the plasma membrane region and quantitative analysis of multi-fluorescent signals from spherical cells. PlasMACC runs in the image analysis software ImageJ environment, it is user-friendly and freely available. RESULTS: PlasMACC delivers detailed information about intensity, thickness and density of fluorescent signals at the plasma membrane of both living and fixed cells. Individual signals can be compared between cells and different signals within one cell can be correlated. PlasMACC can process conventional laser-scanning confocal images as well as data obtained by super-resolution methods such as structured illumination microscopy. COMPARISON WITH EXISTING METHOD(S): By comparing PlasMACC to methods currently used in the field, we show more consistent quantitative data due to the fully automated algorithm. PlasMACC also provides an expanded set of novel analysis parameters. CONCLUSION: PlasMACC enables a detailed quantification of fluorescent signals at the plasma membrane of spherical cells in an unbiased and reliable fashion.


Subject(s)
Actins/metabolism , Cell Membrane/metabolism , Chromaffin Cells/metabolism , Image Processing, Computer-Assisted/methods , Microscopy, Confocal/methods , Pattern Recognition, Automated/methods , Actins/genetics , Adrenal Glands/metabolism , Algorithms , Animals , CD146 Antigen/genetics , CD146 Antigen/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Cytosol/metabolism , Fluorescence , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Mice, Knockout , Mice, Transgenic , Potassium/metabolism , Software Validation
7.
J Cell Biol ; 204(5): 759-75, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24590174

ABSTRACT

Munc18-1 is a soluble protein essential for synaptic transmission. To investigate the dynamics of endogenous Munc18-1 in neurons, we created a mouse model expressing fluorescently tagged Munc18-1 from the endogenous munc18-1 locus. We show using fluorescence recovery after photobleaching in hippocampal neurons that the majority of Munc18-1 trafficked through axons and targeted to synapses via lateral diffusion together with syntaxin-1. Munc18-1 was strongly expressed at presynaptic terminals, with individual synapses showing a large variation in expression. Axon-synapse exchange rates of Munc18-1 were high: during stimulation, Munc18-1 rapidly dispersed from synapses and reclustered within minutes. Munc18-1 reclustering was independent of syntaxin-1, but required calcium influx and protein kinase C (PKC) activity. Importantly, a PKC-insensitive Munc18-1 mutant did not recluster. We show that synaptic Munc18-1 levels correlate with synaptic strength, and that synapses that recruit more Munc18-1 after stimulation have a larger releasable vesicle pool. Hence, PKC-dependent dynamic control of Munc18-1 levels enables individual synapses to tune their output during periods of activity.


Subject(s)
Munc18 Proteins/analysis , Presynaptic Terminals/metabolism , Protein Kinase C/metabolism , Animals , Axons/metabolism , Electrophysiology , Gene Knock-In Techniques , Mice , Munc18 Proteins/metabolism , Protein Transport , Synapses/metabolism , Syntaxin 1/metabolism
8.
Biol Cell ; 102(8): 479-88, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20497124

ABSTRACT

BACKGROUND INFORMATION: During development, growth cones of outgrowing neurons express proteins involved in vesicular secretion, such as SNARE (soluble N-ethylmaleimide-sensitive fusion protein-attachment protein receptor) proteins, Munc13 and Munc18. Vesicles are known to fuse in growth cones prior to synapse formation, which may contribute to outgrowth. RESULTS: We tested this possibility in dissociated cell cultures and organotypic slice cultures of two release-deficient mice (Munc18-1 null and Munc13-1/2 double null). Both types of release-deficient neurons have a decreased outgrowth speed and therefore have a smaller total neurite length during early development [DIV1-4 (day in vitro 1-4)]. In addition, more filopodia per growth cone were observed in Munc18-1 null, but not WT (wild-type) or Munc13-1/2 double null neurons. The smaller total neurite length during early development was no longer observed after synaptogenesis (DIV14-23). CONCLUSION: These data suggest that the inability of vesicle fusion in the growth cone affects outgrowth during the initial phases when outgrowth speed is high, but not during/after synaptogenesis. Overall, the outgrowth speed is probably not rate-limiting during neuronal network formation, at least in vitro. In addition, Munc18, but not Munc13, regulates growth cone filopodia, potentially via its previously observed effect on filamentous actin.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Munc18 Proteins/physiology , Nerve Tissue Proteins/physiology , Neurites/physiology , Animals , Growth Cones/physiology , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Transgenic , Munc18 Proteins/deficiency , Nerve Tissue Proteins/deficiency , Pseudopodia/metabolism
9.
J Neurosci Methods ; 178(2): 378-84, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19146878

ABSTRACT

Cellular traffic is a central aspect of cell function in health and disease. It is highly dynamic, and can be investigated at increasingly finer temporal and spatial resolution due to new imaging techniques and probes. Manual tracking of these data is labor-intensive and observer-biased and existing automation is only semi-automatic and requires near-perfect object detection and high-contrast images. Here, we describe a novel automated technique for quantifying cellular traffic. Using local intrinsic information from adjacent images in a sequence and a model for object characteristics, our approach detects and tracks multiple objects in living cells via Multiple Hypothesis Tracking and handles several confounds (merge/split, birth/death, and clutters), as reliable as expert observers. By replacing the related component (e.g. using a different appearance model) the method can be easily adapted for quantitative analysis of other biological samples.


Subject(s)
Astrocytes/metabolism , Neurons/metabolism , Algorithms , Animals , Automation , Bayes Theorem , Biological Transport , Brain/metabolism , Cell Line , Cells, Cultured , Cytoplasmic Vesicles/metabolism , Humans , Mice , Neuropeptide Y/metabolism , Organelles/metabolism , Recombinant Fusion Proteins/metabolism , Software , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...