Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Sensors (Basel) ; 22(20)2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36298299

ABSTRACT

In this paper, we present a new LoRa transceiver scheme to ensure discrete communications secure from potential eavesdroppers by leveraging a simple and elegant spread spectrum philosophy. The scheme modifies both preamble and payload waveforms by adapting a current state-of-the-art LoRa synchronization front-end. This scheme can also be seen as a self-jamming approach. Furthermore, we introduce a new payload demodulation method that avoids the adverse effects of the traditional cross-correlation solution that would otherwise be used. Our simulation results show that the self-jamming scheme exhibits very good symbol error rate (SER) performance with a loss of just 0.5 dB for a frequency spread factor of up to 10.

2.
Elife ; 102021 12 08.
Article in English | MEDLINE | ID: mdl-34878402

ABSTRACT

Precise quantitative information about the molecular architecture of synapses is essential to understanding the functional specificity and downstream signaling processes at specific populations of synapses. Glycine receptors (GlyRs) are the primary fast inhibitory neurotransmitter receptors in the spinal cord and brainstem. These inhibitory glycinergic networks crucially regulate motor and sensory processes. Thus far, the nanoscale organization of GlyRs underlying the different network specificities has not been defined. Here, we have quantitatively characterized the molecular arrangement and ultra-structure of glycinergic synapses in spinal cord tissue using quantitative super-resolution correlative light and electron microscopy. We show that endogenous GlyRs exhibit equal receptor-scaffold occupancy and constant packing densities of about 2000 GlyRs µm-2 at synapses across the spinal cord and throughout adulthood, even though ventral horn synapses have twice the total copy numbers, larger postsynaptic domains, and more convoluted morphologies than dorsal horn synapses. We demonstrate that this stereotypic molecular arrangement is maintained at glycinergic synapses in the oscillator mouse model of the neuromotor disease hyperekplexia despite a decrease in synapse size, indicating that the molecular organization of GlyRs is preserved in this hypomorph. We thus conclude that the morphology and size of inhibitory postsynaptic specializations rather than differences in GlyR packing determine the postsynaptic strength of glycinergic neurotransmission in motor and sensory spinal cord networks.


Subject(s)
Receptors, Glycine/physiology , Receptors, Glycine/ultrastructure , Spinal Cord/physiology , Spinal Cord/ultrastructure , Synapses/physiology , Synapses/ultrastructure , Animals , Mice , Molecular Structure
3.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507990

ABSTRACT

Long-term synaptic plasticity is believed to be the cellular substrate of learning and memory. Synaptic plasticity rules are defined by the specific complement of receptors at the synapse and the associated downstream signaling mechanisms. In young rodents, at the cerebellar synapse between granule cells (GC) and Purkinje cells (PC), bidirectional plasticity is shaped by the balance between transcellular nitric oxide (NO) driven by presynaptic N-methyl-D-aspartate receptor (NMDAR) activation and postsynaptic calcium dynamics. However, the role and the location of NMDAR activation in these pathways is still debated in mature animals. Here, we show in adult rodents that NMDARs are present and functional in presynaptic terminals where their activation triggers NO signaling. In addition, we find that selective genetic deletion of presynaptic, but not postsynaptic, NMDARs prevents synaptic plasticity at parallel fiber-PC (PF-PC) synapses. Consistent with this finding, the selective deletion of GC NMDARs affects adaptation of the vestibulo-ocular reflex. Thus, NMDARs presynaptic to PCs are required for bidirectional synaptic plasticity and cerebellar motor learning.


Subject(s)
Learning/physiology , Neuronal Plasticity/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Brain/physiology , Cerebellum/physiology , Excitatory Postsynaptic Potentials/physiology , Female , Humans , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/physiology , Neurons/metabolism , Presynaptic Terminals/physiology , Purkinje Cells/metabolism , Synapses/metabolism
4.
PLoS Biol ; 19(8): e3001375, 2021 08.
Article in English | MEDLINE | ID: mdl-34428203

ABSTRACT

Pyramidal neurons (PNs) are covered by thousands of dendritic spines receiving excitatory synaptic inputs. The ultrastructure of dendritic spines shapes signal compartmentalization, but ultrastructural diversity is rarely taken into account in computational models of synaptic integration. Here, we developed a 3D correlative light-electron microscopy (3D-CLEM) approach allowing the analysis of specific populations of synapses in genetically defined neuronal types in intact brain circuits. We used it to reconstruct segments of basal dendrites of layer 2/3 PNs of adult mouse somatosensory cortex and quantify spine ultrastructural diversity. We found that 10% of spines were dually innervated and 38% of inhibitory synapses localized to spines. Using our morphometric data to constrain a model of synaptic signal compartmentalization, we assessed the impact of spinous versus dendritic shaft inhibition. Our results indicate that spinous inhibition is locally more efficient than shaft inhibition and that it can decouple voltage and calcium signaling, potentially impacting synaptic plasticity.


Subject(s)
Dendritic Spines/ultrastructure , Excitatory Postsynaptic Potentials , Inhibitory Postsynaptic Potentials , Models, Neurological , Pyramidal Cells/ultrastructure , Animals , Calcium Signaling , Dendritic Spines/physiology , Female , Mice , Microscopy, Electron, Scanning/methods , Neuronal Plasticity , Pregnancy , Somatosensory Cortex/physiology , Somatosensory Cortex/ultrastructure
5.
Nat Cell Biol ; 21(12): 1544-1552, 2019 12.
Article in English | MEDLINE | ID: mdl-31792378

ABSTRACT

Multiciliated cells (MCCs) amplify large numbers of centrioles that convert into basal bodies, which are required for producing multiple motile cilia. Most centrioles amplified by MCCs grow on the surface of organelles called deuterosomes, whereas a smaller number grow through the centriolar pathway in association with the two parent centrioles. Here, we show that MCCs lacking deuterosomes amplify the correct number of centrioles with normal step-wise kinetics. This is achieved through a massive production of centrioles on the surface and in the vicinity of parent centrioles. Therefore, deuterosomes may have evolved to relieve, rather than supplement, the centriolar pathway during multiciliogenesis. Remarkably, MCCs lacking parent centrioles and deuterosomes also amplify the appropriate number of centrioles inside a cloud of pericentriolar and fibrogranular material. These data show that the centriole number is set independently of their nucleation platforms and suggest that massive centriole production in MCCs is a robust process that can self-organize.


Subject(s)
Centrioles/physiology , Cilia/physiology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Xenopus laevis
6.
Sci Rep ; 9(1): 13060, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31506528

ABSTRACT

Reproductive and respiratory organs, along with brain ventricles, are lined by multiciliated epithelial cells (MCC) that generate cilia-powered fluid flows. MCC hijack the centrosome duplication pathway to form hundreds of centrioles and nucleate motile cilia. In these cells, the large majority of procentrioles are formed associated with partially characterized organelles called deuterosomes. We recently challenged the paradigm that deuterosomes and procentrioles are formed de novo by providing data, in brain MCC, suggesting that they are nucleated from the pre-existing centrosomal younger centriole. However, the origin of deuterosomes and procentrioles is still under debate. Here, we further question centrosome importance for deuterosome and centriole amplification. First, we provide additional data confirming that centriole amplification occurs sequentially from the centrosomal region, and that the first procentriole-loaded deuterosomes are associated with the daughter centriole or in the centrosomal centriole vicinity. Then, to further test the requirement of the centrosome in deuterosome and centriole formation, we depleted centrosomal centrioles using a Plk4 inhibitor. We reveal unexpected limited consequences in deuterosome/centriole number in absence of centrosomal centrioles. Notably, in absence of the daughter centriole only, deuterosomes are not seen associated with the mother centriole. In absence of both centrosomal centrioles, procentrioles are still amplified sequentially and with no apparent structural defects. They seem to arise from a focal region, characterized by microtubule convergence and pericentriolar material (PCM) assembly. The relevance of deuterosome association with the daughter centriole as well as the role of the PCM in the focal and sequential genesis of centrioles in absence of centrosomal centrioles are discussed.


Subject(s)
Brain/physiology , Centrioles/metabolism , Centrosome/metabolism , Cilia/metabolism , Epithelial Cells/metabolism , Biomarkers , Cell Cycle , Fluorescent Antibody Technique , Humans , Molecular Imaging , Organelles/metabolism
7.
Protist ; 166(5): 506-21, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26386358

ABSTRACT

The marine pennate diatom Phaeodactylum tricornutum has become a model for diatom biology, due to its ease of culture and accessibility to reverse genetics approaches. While several features underlying the molecular mechanisms of cell division have been described, morphological analyses are less advanced than they are in other diatoms. We therefore examined cell ultrastructure changes prior to and during cytokinesis. Following chloroplast division, cleavage furrows are formed at both longitudinal ends of the cell and are accompanied by significant vesicle transport. Although neither spindle nor microtubules were observed, the nucleus appeared to be split by the furrow after duplication of the Golgi apparatus. Finally, centripetal cytokinesis was completed by fusion of the furrows. Additionally, F-actin formed a ring structure and its diameter became smaller, accompanying the ingrowing furrows. To further analyse vesicular transport during cytokinesis, we generated transgenic cells expressing yellow fluorescent protein (YFP) fusions with putative diatom orthologs of small GTPase Sec4 and t-SNARE protein SyntaxinA. Time-lapse observations revealed that SyntaxinA-YFP localization expands from both cell tips toward the center, whereas Sec4-YFP was found in the Golgi and subsequently relocalizes to the future division plane. This work provides fundamental new information about cell replication processes in P. tricornutum.


Subject(s)
Algal Proteins/metabolism , Cytokinesis , Diatoms/physiology , Diatoms/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Diatoms/cytology , Diatoms/metabolism , Microscopy, Electron, Transmission , Organisms, Genetically Modified/physiology , Protein Transport
8.
Cell Rep ; 8(5): 1271-9, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25159150

ABSTRACT

Dysfunction of microglia, the tissue macrophages of the brain, has been associated with the etiology of several neuropsychiatric disorders. Consistently, microglia have been shown to regulate neurogenesis and synaptic maturation at perinatal and postnatal stages. However, microglia invade the brain during mid-embryogenesis and thus could play an earlier prenatal role. Here, we show that embryonic microglia, which display a transiently uneven distribution, regulate the wiring of forebrain circuits. Using multiple mouse models, including cell-depletion approaches and cx3cr1(-/-), CR3(-/-), and DAP12(-/-) mutants, we find that perturbing microglial activity affects the outgrowth of dopaminergic axons in the forebrain and the laminar positioning of subsets of neocortical interneurons. Since defects in both dopamine innervation and cortical networks have been linked to neuropsychiatric diseases, our study provides insights into how microglial dysfunction can impact forebrain connectivity and reveals roles for immune cells during normal assembly of brain circuits.


Subject(s)
Microglia/metabolism , Neurogenesis , Prosencephalon/cytology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Interneurons/cytology , Interneurons/metabolism , Mice , Mice, Inbred C57BL , Prosencephalon/embryology , Prosencephalon/physiology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism
9.
Nano Lett ; 14(6): 2994-3001, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24754795

ABSTRACT

Small-molecule chemical calcium (Ca(2+)) indicators are invaluable tools for studying intracellular signaling pathways but have severe shortcomings for detecting local Ca(2+) entry. Nanobiosensors incorporating functionalized quantum dots (QDs) have emerged as promising alternatives but their intracellular use remains a major challenge. We designed cell-penetrating FRET-based Ca(2+) nanobiosensors for the detection of local Ca(2+) concentration transients, using commercially available CANdot565QD as a donor and CaRuby, a custom red-emitting Ca(2+) indicator, as an acceptor. With Ca(2+)-binding affinities covering the range of 3-20 µM, our CaRubies allow building sensors with a scalable affinity for detecting intracellular Ca(2+) transients at various concentrations. To facilitate their cytoplasmic delivery, QDs were further functionalized with a small cell-penetrating peptide (CPP) derived from hadrucalcin (HadUF1-11: H11), a ryanodine receptor-directed scorpion toxin identified within the venom of Hadrurus gertschi. Efficient internalization of QDs doubly functionalized with PEG5-CaRuby and H11 (in a molar ratio of 1:10:10, respectively) is demonstrated. In BHK cells expressing a N-methyl-d-aspartate receptor (NMDAR) construct, these nanobiosensors report rapid intracellular near-membrane Ca(2+) transients following agonist application when imaged by TIRF microscopy. Our work presents the elaboration of cell-penetrating FRET-based nanobiosensors and validates their function for detection of intracellular Ca(2+) transients.


Subject(s)
Biosensing Techniques/methods , Calcium Signaling/physiology , Calcium/metabolism , Cell-Penetrating Peptides/chemistry , Fluorescence Resonance Energy Transfer , Quantum Dots/chemistry , Animals , Cricetinae , HEK293 Cells , Humans , Scorpion Venoms/chemistry
10.
Neuron ; 79(2): 308-21, 2013 Jul 24.
Article in English | MEDLINE | ID: mdl-23889935

ABSTRACT

The strength of synaptic transmission is controlled by the number and activity of neurotransmitter receptors. However, little is known about absolute numbers and densities of receptor and scaffold proteins and the stoichiometry of molecular interactions at synapses. Here, we conducted three-dimensional and quantitative nanoscopic imaging based on single-molecule detections to characterize the ultrastructure of inhibitory synapses and to count scaffold proteins and receptor binding sites. We observed a close correspondence between the spatial organization of gephyrin scaffolds and glycine receptors at spinal cord synapses. Endogenous gephyrin was clustered at densities of 5,000-10,000 molecules/µm(2). The stoichiometry between gephyrin molecules and receptor binding sites was approximately 1:1, consistent with a two-dimensional scaffold in which all gephyrin molecules can contribute to receptor binding. The competition of glycine and GABAA receptor complexes for synaptic binding sites highlights the potential of single-molecule imaging to quantify synaptic plasticity on the nanoscopic scale.


Subject(s)
Carrier Proteins/ultrastructure , Membrane Proteins/ultrastructure , Nanostructures/chemistry , Nanostructures/ultrastructure , Neural Inhibition/physiology , Synapses/ultrastructure , Animals , Binding Sites/physiology , Carrier Proteins/chemistry , Cells, Cultured , Membrane Proteins/chemistry , Molecular Imaging/methods , Protein Binding/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/chemistry , Receptors, GABA-A/metabolism , Receptors, GABA-A/ultrastructure , Receptors, Glycine/chemistry , Receptors, Glycine/metabolism , Receptors, Glycine/ultrastructure , Synapses/chemistry , Synapses/metabolism
11.
PLoS One ; 7(4): e35488, 2012.
Article in English | MEDLINE | ID: mdl-22523599

ABSTRACT

The BCH (BNIP2 and Cdc42GAP Homology) domain-containing protein Bmcc1/Prune2 is highly enriched in the brain and is involved in the regulation of cytoskeleton dynamics and cell survival. However, the molecular mechanisms accounting for these functions are poorly defined. Here, we have identified Bmcc1s, a novel isoform of Bmcc1 predominantly expressed in the mouse brain. In primary cultures of astrocytes and neurons, Bmcc1s localized on intermediate filaments and microtubules and interacted directly with MAP6/STOP, a microtubule-binding protein responsible for microtubule cold stability. Bmcc1s overexpression inhibited MAP6-induced microtubule cold stability by displacing MAP6 away from microtubules. It also resulted in the formation of membrane protrusions for which MAP6 was a necessary cofactor of Bmcc1s. This study identifies Bmcc1s as a new MAP6 interacting protein able to modulate MAP6-induced microtubule cold stability. Moreover, it illustrates a novel mechanism by which Bmcc1 regulates cell morphology.


Subject(s)
Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Nerve Tissue Proteins/metabolism , Protein Isoforms/metabolism , Amino Acid Sequence , Animals , Astrocytes/metabolism , Brain/metabolism , Cold Temperature , HeLa Cells , Humans , Mice , Microtubule-Associated Proteins/drug effects , Microtubules/physiology , Molecular Sequence Data , Neoplasm Proteins , Neurons/metabolism
12.
Proc Natl Acad Sci U S A ; 109(4): E197-205, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22167804

ABSTRACT

Fine control of neuronal activity is crucial to rapidly adjust to subtle changes of the environment. This fine tuning was thought to be purely neuronal until the discovery that astrocytes are active players of synaptic transmission. In the adult hippocampus, microglia are the other major glial cell type. Microglia are highly dynamic and closely associated with neurons and astrocytes. They react rapidly to modifications of their environment and are able to release molecules known to control neuronal function and synaptic transmission. Therefore, microglia display functional features of synaptic partners, but their involvement in the regulation of synaptic transmission has not yet been addressed. We have used a combination of pharmacological approaches with electrophysiological analysis on acute hippocampal slices and ATP assays in purified cell cultures to show that activation of microglia induces a rapid increase of spontaneous excitatory postsynaptic currents. We found that this modulation is mediated by binding of ATP to P2Y1R located on astrocytes and is independent of TNFα or NOS2. Our data indicate that, on activation, microglia cells rapidly release small amounts of ATP, and astrocytes, in turn, amplified this release. Finally, P2Y1 stimulation of astrocytes increased excitatory postsynaptic current frequency through a metabotropic glutamate receptor 5-dependent mechanism. These results indicate that microglia are genuine regulators of neurotransmission and place microglia as upstream partners of astrocytes. Because pathological activation of microglia and alteration of neurotransmission are two early symptoms of most brain diseases, our work also provides a basis for understanding synaptic dysfunction in neuronal diseases.


Subject(s)
Astrocytes/physiology , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , Microglia/physiology , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Astrocytes/metabolism , Astrocytes/ultrastructure , Blotting, Western , DNA Primers/genetics , Electrophysiology , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Microglia/ultrastructure , Microscopy, Confocal , Microscopy, Electron , Real-Time Polymerase Chain Reaction , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/metabolism , Receptors, Purinergic P2Y1/metabolism
13.
J Neurosci ; 31(1): 3-14, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209184

ABSTRACT

Formation and stabilization of postsynaptic glycine receptor (GlyR) clusters result from their association with the polymerized scaffold protein gephyrin. At the cell surface, lateral diffusion and local trapping of GlyR by synaptic gephyrin clusters is one of the main factors controlling their number. However, the mechanisms regulating gephyrin/GlyR cluster sizes are not fully understood. To identify molecular binding partners able to control gephyrin cluster stability, we performed pull-down assays with full-length or truncated gephyrin forms incubated in a rat spinal cord extract, combined with mass spectrometric analysis. We found that heat shock cognate protein 70 (Hsc70), a constitutive member of the heat shock protein 70 (Hsp70) family, selectively binds to the gephyrin G-domain. Immunoelectron microscopy of mouse spinal cord sections showed that Hsc70 could be colocalized with gephyrin at inhibitory synapses. Furthermore, ternary Hsc70-gephyrin-GlyR coclusters were formed following transfection of COS-7 cells. Upon overexpression of Hsc70 in mouse spinal cord neurons, synaptic accumulation of gephyrin was significantly decreased, but GlyR amounts were unaffected. In the same way, Hsc70 inhibition increased gephyrin accumulation at inhibitory synapses without modifying GlyR clustering. Single particle tracking experiments revealed that the increase of gephyrin molecules reduced GlyR diffusion rates without altering GlyR residency at synapses. Our findings demonstrate that Hsc70 regulates gephyrin polymerization independently of its interaction with GlyR. Therefore, gephyrin polymerization and synaptic clustering of GlyR are uncoupled events.


Subject(s)
Carrier Proteins/metabolism , HSC70 Heat-Shock Proteins/physiology , Membrane Proteins/metabolism , Adenosine Diphosphate/pharmacology , Animals , COS Cells , Carrier Proteins/genetics , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/metabolism , Chlorocebus aethiops , Dendrites/metabolism , Dendrites/ultrastructure , Dose-Response Relationship, Drug , Green Fluorescent Proteins/genetics , HSC70 Heat-Shock Proteins/chemistry , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/ultrastructure , Immunoprecipitation/methods , Luminescent Proteins/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Neurons/metabolism , Neurons/ultrastructure , Polymerization/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/genetics , Protein Transport/drug effects , Protein Transport/genetics , Proteomics/methods , Rats , Receptors, Glycine/genetics , Receptors, Glycine/ultrastructure , Spinal Cord/cytology , Spinal Cord/metabolism , Synapses/metabolism , Synapses/ultrastructure , Transfection/methods , Red Fluorescent Protein
14.
Neuron ; 66(2): 235-47, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20435000

ABSTRACT

Miniature synaptic currents have long been known to represent random transmitter release under resting conditions, but much remains to be learned about their nature and function in central synapses. In this work, we describe a new class of miniature currents ("preminis") that arise by the autocrine activation of axonal receptors following random vesicular release. Preminis are prominent in gabaergic synapses made by cerebellar interneurons during the development of the molecular layer. Unlike ordinary miniature postsynaptic currents in the same cells, premini frequencies are strongly enhanced by subthreshold depolarization, suggesting that the membrane depolarization they produce belongs to a feedback loop regulating neurotransmitter release. Thus, preminis could guide the formation of the interneuron network by enhancing neurotransmitter release at recently formed synaptic contacts.


Subject(s)
Interneurons/physiology , Neural Inhibition/physiology , Presynaptic Terminals/physiology , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Cerebellum/metabolism , Cerebellum/physiology , Electrophysiology , Inhibitory Postsynaptic Potentials/physiology , Interneurons/metabolism , Microscopy, Electron , Miniature Postsynaptic Potentials/physiology , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Synapses/metabolism , Synapses/physiology
15.
Neuron ; 65(1): 53-65, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-20152113

ABSTRACT

The density of GABA(A) receptors (GABA(A)Rs) at synapses regulates brain excitability, and altered inhibition may contribute to Huntington's disease, which is caused by a polyglutamine repeat in the protein huntingtin. However, the machinery that delivers GABA(A)Rs to synapses is unknown. We demonstrate that GABA(A)Rs are trafficked to synapses by the kinesin family motor protein 5 (KIF5). We identify the adaptor linking the receptors to KIF5 as the huntingtin-associated protein 1 (HAP1). Disrupting the HAP1-KIF5 complex decreases synaptic GABA(A)R number and reduces the amplitude of inhibitory postsynaptic currents. When huntingtin is mutated, as in Huntington's disease, GABA(A)R transport and inhibitory synaptic currents are reduced. Thus, HAP1-KIF5-dependent GABA(A)R trafficking is a fundamental mechanism controlling the strength of synaptic inhibition in the brain. Its disruption by mutant huntingtin may explain some of the defects in brain information processing occurring in Huntington's disease and provides a molecular target for therapeutic approaches.


Subject(s)
Kinesins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Protein Isoforms/metabolism , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Humans , Huntingtin Protein , Huntington Disease/metabolism , Kinesins/genetics , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/genetics , Patch-Clamp Techniques , Peptides/genetics , Peptides/metabolism , Protein Isoforms/genetics , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Synapses/ultrastructure , Synaptic Transmission/physiology
16.
J Neurosci ; 27(34): 9022-31, 2007 Aug 22.
Article in English | MEDLINE | ID: mdl-17715339

ABSTRACT

The parallel fibers relay information coming into the cerebellar cortex from the mossy fibers, and they form synapses with molecular layer interneurons (MLIs) and Purkinje cells. Here we show that activation of ionotropic GABA receptors (GABA(A)Rs) induces glutamate release from parallel fibers onto both MLIs and Purkinje cells. These GABA-induced EPSCs have kinetics and amplitudes identical to random spontaneous currents (sEPSCs), but, unlike sEPSCs, they occur in bursts of between one and five successive events. The variation in amplitude of events within bursts is significantly less than the variation of all sEPSC amplitudes, suggesting that the bursts result from repetitive activation of single presynaptic fibers. Electron microscopy of immunogold-labeled alpha-1 subunits revealed GABA(A)Rs on parallel fiber terminals. We suggest that the activation of these receptors underlies the increased amplitude of parallel fiber-evoked Purkinje cell EPSCs seen with application of exogenous GABA or after the release of GABA from local interneurons. These results occur only when molecular layer GABA(A)Rs are activated, and the effects are abolished when the receptors are blocked by the GABA(A)R antagonist gabazine (5 microM). From these data, we conclude that GABA(A)Rs located on parallel fibers depolarize parallel fiber terminals beyond the threshold for Na+ channel activation and thereby induce glutamate release onto MLIs and Purkinje cells.


Subject(s)
Cerebellum/cytology , Glutamic Acid/metabolism , Presynaptic Terminals/metabolism , Receptors, GABA-A/physiology , Synapses/metabolism , Animals , Animals, Newborn , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/radiation effects , GABA Agonists/pharmacology , GABA Antagonists/pharmacology , In Vitro Techniques , Interneurons/cytology , Microscopy, Electron, Transmission/methods , Muscimol/pharmacology , Patch-Clamp Techniques/methods , Phosphinic Acids/pharmacology , Presynaptic Terminals/radiation effects , Presynaptic Terminals/ultrastructure , Propanolamines/pharmacology , Purkinje Cells/cytology , Rats , Synapses/ultrastructure , Time Factors , gamma-Aminobutyric Acid/pharmacology
17.
J Neurosci ; 27(26): 6868-77, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17596435

ABSTRACT

Presynaptic terminals are specialized for mediating rapid fusion of synaptic vesicles (SVs) after calcium influx. The regulated trafficking of SVs likely results from a highly organized cytomatrix. How this cytomatrix links SVs, maintains them near the active zones (AZs) of release, and organizes docked SVs at the release sites is not fully understood. To analyze the three-dimensional (3D) architecture of the presynaptic cytomatrix, electron tomography of presynaptic terminals contacting spines was performed in the stratum radiatum of the rat hippocampal CA1 area. To preserve the cytomatrix, hippocampal slices were immobilized using high-pressure freezing, followed by cryosubstitution and embedding. SVs are surrounded by a dense network of filaments. A given vesicle is connected to approximately 1.5 neighboring ones. SVs at the periphery of this network are also linked to the plasma membrane, by longer filaments. More of these filaments are found at the AZ. At the AZ, docked SVs are grouped around presynaptic densities. Filaments with adjacent SVs emerge from these densities. Immunogold localizations revealed that synapsin is located in the presynaptic bouton, whereas Bassoon and CAST (ERC2) are at focal points next to the AZ. In synapsin triple knock-out mice, the number of SVs is reduced by 63%, but the size of the boutons is reduced by only 18%, and the mean distance of SVs to the AZ is unchanged. This 3D analysis reveals the morphological constraints exerted by the presynaptic molecular scaffold. SVs are tightly interconnected in the axonal bouton, and this network is preferentially connected to the AZ.


Subject(s)
Dendritic Spines/ultrastructure , Extracellular Matrix/ultrastructure , Hippocampus/ultrastructure , Presynaptic Terminals/ultrastructure , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dendritic Spines/metabolism , Extracellular Matrix/metabolism , Hippocampus/metabolism , Image Cytometry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Synapsins/genetics , Synapsins/metabolism , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Tomography, X-Ray Computed
18.
Cell ; 127(2): 277-89, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-17055430

ABSTRACT

The auditory inner hair cell (IHC) ribbon synapse operates with an exceptional temporal precision and maintains a high level of neurotransmitter release. However, the molecular mechanisms underlying IHC synaptic exocytosis are largely unknown. We studied otoferlin, a predicted C2-domain transmembrane protein, which is defective in a recessive form of human deafness. We show that otoferlin expression in the hair cells correlates with afferent synaptogenesis and find that otoferlin localizes to ribbon-associated synaptic vesicles. Otoferlin binds Ca(2+) and displays Ca(2+)-dependent interactions with the SNARE proteins syntaxin1 and SNAP25. Otoferlin deficient mice (Otof(-/-)) are profoundly deaf. Exocytosis in Otof(-/-) IHCs is almost completely abolished, despite normal ribbon synapse morphogenesis and Ca(2+) current. Thus, otoferlin is essential for a late step of synaptic vesicle exocytosis and may act as the major Ca(2+) sensor triggering membrane fusion at the IHC ribbon synapse.


Subject(s)
Cochlea/metabolism , Deafness/metabolism , Exocytosis , Hair Cells, Auditory, Inner/metabolism , Membrane Proteins/metabolism , Synapses/metabolism , Animals , Auditory Pathways/metabolism , Calcium/metabolism , Cochlea/growth & development , Deafness/genetics , Deafness/physiopathology , Evoked Potentials, Auditory, Brain Stem , Hair Cells, Auditory, Inner/ultrastructure , Humans , Membrane Fusion , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Synaptic Transmission , Synaptic Vesicles/metabolism , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/metabolism , Time Factors
19.
Eur J Neurosci ; 24(12): 3463-74, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17229095

ABSTRACT

Electron microscopy allows the analysis of synaptic ultrastructure and its modifications during learning or in pathological conditions. However, conventional electron microscopy uses aldehyde fixatives that alter the morphology of the synapse by changing osmolarity and collapsing its molecular components. We have used high-pressure freezing (HPF) to capture within a few milliseconds structural features without aldehyde fixative, and thus to provide a snapshot of living synapses. CA1 hippocampal area slices from P21 rats were frozen at -173 degrees C under high pressure to reduce crystal formation, and synapses on dendritic spines were analysed after cryosubstitution and embedding. Synaptic terminals were larger than after aldehyde fixation, and synaptic vesicles in these terminals were less densely packed. Small filaments linked the vesicles in subgroups. The postsynaptic densities (PSDs) exhibited filamentous projections extending into the spine cytoplasm. Tomographic analysis showed that these projections were connected with the spine cytoskeletal meshwork. Using immunocytochemistry, we found as expected GluR1 at the synaptic cleft and CaMKII in the PSD. Actin immunoreactivity (IR) labelled the cytoskeletal meshwork beneath the filamentous projections, but was very scarce within the PSD itself. ProSAP2/Shank3, cortactin and Ena/VASP-IRs were concentrated on the cytoplasmic face of the PSD, at the level of the PSD projections. Synaptic ultrastructure after HPF was different from that observed after aldehyde fixative. The boutons were larger, and filamentous components were preserved. Particularly, filamentous projections were observed linking the PSD to the actin cytoskeleton. Thus, synaptic ultrastructure can be analysed under more realistic conditions following HPF.


Subject(s)
Cryopreservation/methods , Synapses/ultrastructure , Tomography , Aldehydes/pharmacology , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cortactin/metabolism , Freezing , Hippocampus/ultrastructure , Microscopy, Electron, Transmission/methods , Microscopy, Immunoelectron/methods , Rats , Receptors, Glutamate/metabolism , Synapses/drug effects , Synapses/metabolism , Tissue Fixation/methods
20.
Mol Cell Neurosci ; 27(4): 394-403, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15555918

ABSTRACT

Inhibitory transmission in the hippocampus is predominantly GABAergic, but electrophysiological data evidenced strychnine-sensitive glycine-induced currents. However, synaptic currents have not been reported. Here, we describe, for the first time, the presence of GlyR clusters in several areas of the hippocampus as well as in cultured hippocampal neurons. In contrast with spinal cord, hippocampal GlyRs contain alpha2 but no alpha1 subunit. Optical and electron microscopy indicates that GlyRs can be synaptic as well as extrasynaptic. Synaptic GlyRs were apposed to glycinergic boutons characterized by the expression of the vesicular and the plasma membrane transporters of glycine (VIAAT and GlyT2, respectively). Double labeling with calcium-binding proteins showed that GlyT2 could be detected in boutons innervating both excitatory cells (soma and dendrites) and interneurons. Finally, GlyR clusters could be detected at synaptic sites with the GABAA receptor gamma2 subunit and gephyrin, suggesting that mixed GABA/glycine synapses might exist in the hippocampus.


Subject(s)
Glycine/metabolism , Hippocampus/metabolism , Neural Inhibition/physiology , Synapses/metabolism , Amino Acid Transport Systems/metabolism , Amino Acid Transport Systems, Neutral/metabolism , Animals , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Cells, Cultured , Dendrites/metabolism , Dendrites/ultrastructure , Glycine Plasma Membrane Transport Proteins , Hippocampus/ultrastructure , Immunohistochemistry , Interneurons/metabolism , Interneurons/ultrastructure , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley , Receptor Aggregation/physiology , Receptors, GABA-A/metabolism , Synapses/ultrastructure , Synaptic Membranes/metabolism , Synaptic Membranes/ultrastructure , Synaptic Transmission/physiology , Vesicular Inhibitory Amino Acid Transport Proteins , Vesicular Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...