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











Publication year range
1.
bioRxiv ; 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38659918

ABSTRACT

Neurotransmitters are released from synaptic vesicles with remarkable precision in response to presynaptic Ca2+ influx but exhibit significant heterogeneity in exocytosis timing and efficacy based on the recent history of activity. This heterogeneity is critical for information transfer in the brain, yet its molecular basis remains poorly understood. Here, we employ a biochemically-defined fusion assay under physiologically-relevant conditions to delineate the minimal protein machinery sufficient to account for different modes of Ca2+-triggered vesicle fusion and short-term facilitation. We find that Synaptotagmin-1, Synaptotagmin-7, and Complexin, synergistically restrain SNARE complex assembly, thus preserving vesicles in a stably docked state at rest. Upon Ca2+ activation, Synaptotagmin-1 induces rapid vesicle fusion, while Synaptotagmin-7 mediates delayed fusion. Competitive binding of Synaptotagmin-1 and Synaptotagmin-7 to the same SNAREs, coupled with differential rates of Ca2+-triggered fusion clamp reversal, govern the kinetics of vesicular fusion. Under conditions mimicking sustained neuronal activity, the Synaptotagmin-7 fusion clamp is destabilized by the elevated basal Ca2+ concentration, thereby enhancing the synchronous component of fusion. These findings provide a direct demonstration that a small set of proteins is sufficient to account for how nerve terminals adapt and regulate the Ca2+-evoked neurotransmitter exocytosis process to support their specialized functions in the nervous system.

2.
Commun Biol ; 6(1): 1091, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37891212

ABSTRACT

Calcium-evoked release of neurotransmitters from synaptic vesicles (SVs) is catalysed by SNARE proteins. The predominant view is that, at rest, complete assembly of SNARE complexes is inhibited ('clamped') by synaptotagmin and complexin molecules. Calcium binding by synaptotagmins releases this fusion clamp and triggers fast SV exocytosis. However, this model has not been quantitatively tested over physiological timescales. Here we describe an experimentally constrained computational modelling framework to quantitatively assess how the molecular architecture of the fusion clamp affects SV exocytosis. Our results argue that the 'release-of-inhibition' model can indeed account for fast calcium-activated SV fusion, and that dual binding of synaptotagmin-1 and synaptotagmin-7 to the same SNARE complex enables synergistic regulation of the kinetics and plasticity of neurotransmitter release. The developed framework provides a powerful and adaptable tool to link the molecular biochemistry of presynaptic proteins to physiological data and efficiently test the plausibility of calcium-activated neurotransmitter release models.


Subject(s)
Calcium , Synapses , Calcium/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , SNARE Proteins/metabolism , Neurotransmitter Agents/metabolism
3.
Res Sq ; 2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37163032

ABSTRACT

Calcium-evoked release of neurotransmitters from synaptic vesicles (SVs) is catalysed by SNARE proteins. The predominant view is that, at rest, complete assembly of SNARE complexes is inhibited ('clamped') by synaptotagmin and complexin molecules. Calcium binding by synaptotagmins releases this fusion clamp and triggers fast SV exocytosis. However, this model has not been quantitatively tested over physiological timescales. Here we describe an experimentally constrained computational modelling framework to quantitatively assess how the molecular architecture of the fusion clamp affects SV exocytosis. Our results argue that the "release-of-inhibition" model can indeed account for fast calcium-activated SV fusion, and that dual binding of synaptotagmin-1 and synaptotagmin-7 to the same SNARE complex enables synergistic regulation of the kinetics and plasticity of neurotransmitter release. The developed framework provides a powerful and adaptable tool to link the molecular biochemistry of presynaptic proteins to physiological data and efficiently test the plausibility of calcium-activated neurotransmitter release models.

4.
Nat Commun ; 13(1): 3497, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715404

ABSTRACT

The balance between fast synchronous and delayed asynchronous release of neurotransmitters has a major role in defining computational properties of neuronal synapses and regulation of neuronal network activity. However, how it is tuned at the single synapse level remains poorly understood. Here, using the fluorescent glutamate sensor SF-iGluSnFR, we image quantal vesicular release in tens to hundreds of individual synaptic outputs from single pyramidal cells with 4 millisecond temporal and 75 nm spatial resolution. We find that the ratio between synchronous and asynchronous synaptic vesicle exocytosis varies extensively among synapses supplied by the same axon, and that the synchronicity of release is reduced at low release probability synapses. We further demonstrate that asynchronous exocytosis sites are more widely distributed within the release area than synchronous sites. Together, our results reveal a universal relationship between the two major functional properties of synapses - the timing and the overall efficacy of neurotransmitter release.


Subject(s)
Glutamic Acid , Synapses , Exocytosis/physiology , Neurotransmitter Agents , Synapses/physiology , Synaptic Transmission/physiology
5.
J Neurochem ; 156(1): 48-58, 2021 01.
Article in English | MEDLINE | ID: mdl-32418206

ABSTRACT

Brain function relies on vesicular release of neurotransmitters at chemical synapses. The release probability depends on action potential-evoked presynaptic Ca2+ entry, but also on the resting Ca2+ level. Whether these basic aspects of presynaptic calcium homeostasis show any consistent trend along the axonal path, and how they are controlled by local network activity, remains poorly understood. Here, we take advantage of the recently advanced FLIM-based method to monitor presynaptic Ca2+ with nanomolar sensitivity. We find that, in cortical pyramidal neurons, action potential-evoked calcium entry (range 10-300 nM), but not the resting Ca2+ level (range 10-100 nM), tends to increase with higher order of axonal branches. Blocking astroglial glutamate uptake reduces evoked Ca2+ entry but has little effect on resting Ca2+ whereas both appear boosted by the constitutive activation of group 1/2 metabotropic glutamate receptors. We find no consistent effect of transient somatic depolarization or hyperpolarization on presynaptic Ca2+ entry or its basal level. The results unveil some key aspects of presynaptic machinery in cortical circuits, shedding light on basic principles of synaptic connectivity in the brain.


Subject(s)
Calcium/metabolism , Glutamic Acid/metabolism , Neurons/metabolism , Optical Imaging/methods , Synaptic Transmission/physiology , Animals , Cerebral Cortex/metabolism , Mice , Mice, Inbred C57BL , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Receptors, Metabotropic Glutamate/metabolism , Synapses/metabolism
6.
Proc Natl Acad Sci U S A ; 117(7): 3819-3827, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32015138

ABSTRACT

Synaptotagmin 1 (Syt1) synchronizes neurotransmitter release to action potentials (APs) acting as the fast Ca2+ release sensor and as the inhibitor (clamp) of spontaneous and delayed asynchronous release. While the Syt1 Ca2+ activation mechanism has been well-characterized, how Syt1 clamps transmitter release remains enigmatic. Here we show that C2B domain-dependent oligomerization provides the molecular basis for the Syt1 clamping function. This follows from the investigation of a designed mutation (F349A), which selectively destabilizes Syt1 oligomerization. Using a combination of fluorescence imaging and electrophysiology in neocortical synapses, we show that Syt1F349A is more efficient than wild-type Syt1 (Syt1WT) in triggering synchronous transmitter release but fails to clamp spontaneous and synaptotagmin 7 (Syt7)-mediated asynchronous release components both in rescue (Syt1-/- knockout background) and dominant-interference (Syt1+/+ background) conditions. Thus, we conclude that Ca2+-sensitive Syt1 oligomers, acting as an exocytosis clamp, are critical for maintaining the balance among the different modes of neurotransmitter release.


Subject(s)
Neurotransmitter Agents/metabolism , Synaptotagmin I/metabolism , Animals , Exocytosis , Mice , Mice, Knockout , Mutation, Missense , Synapses/metabolism , Synaptic Transmission , Synaptotagmin I/genetics
7.
Proc Natl Acad Sci U S A ; 115(32): E7624-E7631, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30038018

ABSTRACT

Regulated exocytosis, which underlies many intercellular signaling events, is a tightly controlled process often triggered by calcium ion(s) (Ca2+). Despite considerable insight into the central components involved, namely, the core fusion machinery [soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)] and the principal Ca2+ sensor [C2-domain proteins like synaptotagmin (Syt)], the molecular mechanism of Ca2+-dependent release has been unclear. Here, we report that the Ca2+-sensitive oligomers of Syt1, a conserved structural feature among several C2-domain proteins, play a critical role in orchestrating Ca2+-coupled vesicular release. This follows from pHluorin-based imaging of single-vesicle exocytosis in pheochromocytoma (PC12) cells showing that selective disruption of Syt1 oligomerization using a structure-directed mutation (F349A) dramatically increases the normally low levels of constitutive exocytosis to effectively occlude Ca2+-stimulated release. We propose a parsimonious model whereby Ca2+-sensitive oligomers of Syt (or a similar C2-domain protein) assembled at the site of docking physically block spontaneous fusion until disrupted by Ca2+ Our data further suggest Ca2+-coupled vesicular release is triggered by removal of the inhibition, rather than by direct activation of the fusion machinery.


Subject(s)
Calcium/metabolism , Exocytosis , Membrane Fusion/physiology , Protein Multimerization/physiology , Synaptotagmin I/metabolism , Animals , Cations, Divalent/metabolism , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/ultrastructure , Fluorescent Antibody Technique , Green Fluorescent Proteins/chemistry , Microscopy, Electron , Mutation , PC12 Cells , Protein Binding/physiology , Rats , Recombinant Proteins/metabolism , Synaptotagmin I/genetics , Vesicle-Associated Membrane Protein 2/metabolism
8.
BMC Biol ; 16(1): 70, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29925374

ABSTRACT

BACKGROUND: Recent advances in clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) genome editing have led to the use of long single-stranded DNA (lssDNA) molecules for generating conditional mutations. However, there is still limited available data on the efficiency and reliability of this method. RESULTS: We generated conditional mouse alleles using lssDNA donor templates and performed extensive characterization of the resulting mutations. We observed that the use of lssDNA molecules as donors efficiently yielded founders bearing the conditional allele, with seven out of nine projects giving rise to modified alleles. However, rearranged alleles including nucleotide changes, indels, local rearrangements and additional integrations were also frequently generated by this method. Specifically, we found that alleles containing unexpected point mutations were found in three of the nine projects analyzed. Alleles originating from illegitimate repairs or partial integration of the donor were detected in eight projects. Furthermore, additional integrations of donor molecules were identified in four out of the seven projects analyzed by copy counting. This highlighted the requirement for a thorough allele validation by polymerase chain reaction, sequencing and copy counting of the mice generated through this method. We also demonstrated the feasibility of using lssDNA donors to generate thus far problematic point mutations distant from active CRISPR cutting sites by targeting two distinct genes (Gckr and Rims1). We propose a strategy to perform extensive quality control and validation of both types of mouse models generated using lssDNA donors. CONCLUSION: lssDNA donors reproducibly generate conditional alleles and can be used to introduce point mutations away from CRISPR/Cas9 cutting sites in mice. However, our work demonstrates that thorough quality control of new models is essential prior to reliably experimenting with mice generated by this method. These advances in genome editing techniques shift the challenge of mutagenesis from generation to the validation of new mutant models.


Subject(s)
DNA, Single-Stranded , Gene Editing/methods , Gene Targeting , Mice/genetics , Alleles , Animals , CRISPR-Cas Systems , Mutation , Reproducibility of Results
9.
Curr Opin Neurobiol ; 51: 154-162, 2018 08.
Article in English | MEDLINE | ID: mdl-29886350

ABSTRACT

Quantal neurotransmitter release at nerve terminals is tightly regulated by the presynaptic Ca2+ concentration. Here, we summarise current advances in understanding how the interplay between presynaptic Ca2+ dynamics and different Ca2+ release sensors shapes action potential-evoked release on a timescale from hundreds of microseconds to hundreds of milliseconds. In particular, we review recent studies that reveal the synergistic roles of the low Ca2+ affinity/fast release sensors synaptotagmins 1, 2 and 9 and the high affinity/slow release sensor synaptotagmin 7 in the regulation of synchronous and asynchronous release and of short-term synaptic plasticity. We also examine new biochemical and structural data and outline a working model that could potentially explain the cooperative roles of different synaptotagmins in molecular terms.


Subject(s)
Cell Membrane/metabolism , Neurons/cytology , Neurotransmitter Agents/metabolism , Synaptotagmins/metabolism , Animals , Calcium/metabolism , Neurons/physiology
10.
Cell Rep ; 20(2): 333-343, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28700936

ABSTRACT

Alternative splicing of pre-mRNAs is prominent in the mammalian brain, where it is thought to expand proteome diversity. For example, alternative splicing of voltage-gated Ca2+ channel (VGCC) α1 subunits can generate thousands of isoforms with differential properties and expression patterns. However, the impact of this molecular diversity on brain function, particularly on synaptic transmission, which crucially depends on VGCCs, is unclear. Here, we investigate how two major splice isoforms of P/Q-type VGCCs (Cav2.1[EFa/b]) regulate presynaptic plasticity in hippocampal neurons. We find that the efficacy of P/Q-type VGCC isoforms in supporting synaptic transmission is markedly different, with Cav2.1[EFa] promoting synaptic depression and Cav2.1[EFb] synaptic facilitation. Following a reduction in network activity, hippocampal neurons upregulate selectively Cav2.1[EFa], the isoform exhibiting the higher synaptic efficacy, thus effectively supporting presynaptic homeostatic plasticity. Therefore, the balance between VGCC splice variants at the synapse is a key factor in controlling neurotransmitter release and presynaptic plasticity.


Subject(s)
Alternative Splicing/physiology , Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/metabolism , Alternative Splicing/genetics , Animals , Cells, Cultured , Electrophysiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , GABA Antagonists/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Picrotoxin/pharmacology , RNA Interference , Rats , Synapses/drug effects , Synapses/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
11.
Proc Natl Acad Sci U S A ; 114(9): 2395-2400, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193892

ABSTRACT

Although action potentials propagate along axons in an all-or-none manner, subthreshold membrane potential fluctuations at the soma affect neurotransmitter release from synaptic boutons. An important mechanism underlying analog-digital modulation is depolarization-mediated inactivation of presynaptic Kv1-family potassium channels, leading to action potential broadening and increased calcium influx. Previous studies have relied heavily on recordings from blebs formed after axon transection, which may exaggerate the passive propagation of somatic depolarization. We recorded instead from small boutons supplied by intact axons identified with scanning ion conductance microscopy in primary hippocampal cultures and asked how distinct potassium channels interact in determining the basal spike width and its modulation by subthreshold somatic depolarization. Pharmacological or genetic deletion of Kv1.1 broadened presynaptic spikes without preventing further prolongation by brief depolarizing somatic prepulses. A heterozygous mouse model of episodic ataxia type 1 harboring a dominant Kv1.1 mutation had a similar broadening effect on basal spike shape as deletion of Kv1.1; however, spike modulation by somatic prepulses was abolished. These results argue that the Kv1.1 subunit is not necessary for subthreshold modulation of spike width. However, a disease-associated mutant subunit prevents the interplay of analog and digital transmission, possibly by disrupting the normal stoichiometry of presynaptic potassium channels.


Subject(s)
Action Potentials , Ataxia/metabolism , Hippocampus/metabolism , Kv1.1 Potassium Channel/genetics , Myokymia/metabolism , Neurons/metabolism , Protein Subunits/genetics , Animals , Ataxia/genetics , Ataxia/pathology , Disease Models, Animal , Gene Expression , Hippocampus/pathology , Kv1.1 Potassium Channel/deficiency , Mice , Mice, Knockout , Myokymia/genetics , Myokymia/pathology , Neurons/pathology , Patch-Clamp Techniques , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Primary Cell Culture , Protein Subunits/deficiency , Synaptic Transmission
12.
Nat Commun ; 7: 12102, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27381274

ABSTRACT

Brain development and interictal function are unaffected in many paroxysmal neurological channelopathies, possibly explained by homoeostatic plasticity of synaptic transmission. Episodic ataxia type 1 is caused by missense mutations of the potassium channel Kv1.1, which is abundantly expressed in the terminals of cerebellar basket cells. Presynaptic action potentials of small inhibitory terminals have not been characterized, and it is not known whether developmental plasticity compensates for the effects of Kv1.1 dysfunction. Here we use visually targeted patch-clamp recordings from basket cell terminals of mice harbouring an ataxia-associated mutation and their wild-type littermates. Presynaptic spikes are followed by a pronounced afterdepolarization, and are broadened by pharmacological blockade of Kv1.1 or by a dominant ataxia-associated mutation. Somatic recordings fail to detect such changes. Spike broadening leads to increased Ca(2+) influx and GABA release, and decreased spontaneous Purkinje cell firing. We find no evidence for developmental compensation for inherited Kv1.1 dysfunction.


Subject(s)
Action Potentials/drug effects , Ataxia/physiopathology , Channelopathies/physiopathology , Kv1.1 Potassium Channel/metabolism , Myokymia/physiopathology , Purkinje Cells/metabolism , Animals , Ataxia/genetics , Ataxia/metabolism , Calcium/metabolism , Channelopathies/genetics , Channelopathies/metabolism , Disease Models, Animal , Elapid Venoms/pharmacology , Female , Gene Expression , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.1 Potassium Channel/genetics , Mice , Mice, Transgenic , Microtomy , Mutation , Myokymia/genetics , Myokymia/metabolism , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Purkinje Cells/drug effects , Purkinje Cells/pathology , Synaptic Transmission/drug effects , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
13.
Front Cell Neurosci ; 9: 239, 2015.
Article in English | MEDLINE | ID: mdl-26190970

ABSTRACT

Action potential-dependent release of synaptic vesicles and short-term synaptic plasticity are dynamically regulated by the endogenous Ca(2+) buffers that shape [Ca(2+)] profiles within a presynaptic bouton. Calmodulin is one of the most abundant presynaptic proteins and it binds Ca(2+) faster than any other characterized endogenous neuronal Ca(2+) buffer. Direct effects of calmodulin on fast presynaptic Ca(2+) dynamics and vesicular release however have not been studied in detail. Using experimentally constrained three-dimensional diffusion modeling of Ca(2+) influx-exocytosis coupling at small excitatory synapses we show that, at physiologically relevant concentrations, Ca(2+) buffering by calmodulin plays a dominant role in inhibiting vesicular release and in modulating short-term synaptic plasticity. We also propose a novel and potentially powerful mechanism for short-term facilitation based on Ca(2+)-dependent dynamic dislocation of calmodulin molecules from the plasma membrane within the active zone.

14.
Neurology ; 84(6): 575-9, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25589670

ABSTRACT

OBJECTIVE: To determine whether immunoglobulin G (IgG) from patients with Lambert-Eaton myasthenic syndrome (LEMS) decreases action potential­evoked synaptic vesicle exocytosis,and whether the effect is mediated by P/Q-type voltage-gated calcium channels (VGCCs). METHODS: IgG was obtained from 4 patients with LEMS (3 males, 1 female), including 2 patients with lung malignancy. Antibodies against P/Q-type VGCCs were detected in all 4 patients, and against N-type VGCCs in 2. We incubated neuronal cultures with LEMS IgG and determined the size of the total recycling pool of synaptic vesicles and the rate of action potential­evoked exocytosis using fluorescence imaging of the amphiphilic dye SynaptoRed C1. Pooled IgG from healthy volunteers was used as a control. We repeated the experiments on synapses lacking P/Q-type calcium channels from a Cacna1a knockout mouse to determine whether these channels account for the pathogenic effect of LEMS IgG. RESULTS: LEMS IgG had no effect on the total recycling pool size but significantly reduced the rate of action potential­evoked synaptic exocytosis in wild-type neurons when compared with neurons treated with control IgG. In contrast, LEMS IgG had no effect on the rate of synaptic vesicle exocytosis in neurons lacking P/Q-type channels. CONCLUSIONS: These data provide direct evidence that LEMS IgG inhibits neurotransmitter release by acting on P/Q-type VGCCs.


Subject(s)
Action Potentials/drug effects , Calcium Channel Blockers/therapeutic use , Calcium Channels/metabolism , Immunoglobulin G/therapeutic use , Lambert-Eaton Myasthenic Syndrome/metabolism , Neurons/metabolism , Synaptic Transmission/drug effects , Adult , Aged , Animals , Autoantibodies/blood , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Exocytosis , Female , Fluorescent Dyes , Humans , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Lambert-Eaton Myasthenic Syndrome/immunology , Male , Mice , Mice, Knockout , Middle Aged
15.
Nat Neurosci ; 16(12): 1754-1763, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185424

ABSTRACT

The role of voltage-gated Ca2+ channels (VGCCs) in spontaneous miniature neurotransmitter release is incompletely understood. We found that stochastic opening of P/Q-, N- and R-type VGCCs accounts for ∼50% of all spontaneous glutamate release at rat cultured hippocampal synapses, and that R-type channels have a far greater role in spontaneous than in action potential-evoked exocytosis. VGCC-dependent miniature neurotransmitter release (minis) showed similar sensitivity to presynaptic Ca2+ chelation as evoked release, arguing for direct triggering of spontaneous release by transient spatially localized Ca(2+) domains. Experimentally constrained three-dimensional diffusion modeling of Ca2+ influx-exocytosis coupling was consistent with clustered distribution of VGCCs in the active zone of small hippocampal synapses and revealed that spontaneous VGCCs openings can account for the experimentally observed VGCC-dependent minis, although single channel openings triggered release with low probability. Uncorrelated stochastic VGCC opening is therefore a major trigger for spontaneous glutamate release, with differential roles for distinct channel subtypes.


Subject(s)
Calcium Channels/classification , Calcium Channels/physiology , Glutamic Acid/metabolism , Ion Channel Gating/physiology , Neurons/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers , Cells, Cultured , Chelating Agents/pharmacology , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Hippocampus/cytology , Ion Channel Gating/drug effects , Models, Biological , Neurons/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Stochastic Processes
16.
Neuron ; 79(6): 1067-77, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24050398

ABSTRACT

Direct electrical access to presynaptic ion channels has hitherto been limited to large specialized terminals such as the calyx of Held or hippocampal mossy fiber bouton. The electrophysiology and ion-channel complement of far more abundant small synaptic terminals (≤ 1 µm) remain poorly understood. Here we report a method based on superresolution scanning ion conductance imaging of small synapses in culture at approximately 100-150 nm 3D resolution, which allows presynaptic patch-clamp recordings in all four configurations (cell-attached, inside-out, outside-out, and whole-cell). Using this technique, we report presynaptic recordings of K(+), Na(+), Cl(-), and Ca(2+) channels. This semiautomated approach allows direct investigation of the distribution and properties of presynaptic ion channels at small central synapses.


Subject(s)
Ion Channel Gating/physiology , Ion Channels/physiology , Neurons/cytology , Presynaptic Terminals/physiology , Animals , Animals, Newborn , Biophysical Phenomena/physiology , Calcium/metabolism , Cells, Cultured , Dendritic Spines/physiology , Dendritic Spines/ultrastructure , Electric Stimulation , Electrodes , Fluorescent Dyes/metabolism , Hippocampus/cytology , Imaging, Three-Dimensional , Ion Channels/ultrastructure , Membrane Potentials/physiology , Microscopy, Scanning Tunneling , Patch-Clamp Techniques , Presynaptic Terminals/ultrastructure , Rats
17.
PLoS Biol ; 10(9): e1001396, 2012.
Article in English | MEDLINE | ID: mdl-23049481

ABSTRACT

The efficacy of action potential evoked neurotransmitter release varies widely even among synapses supplied by the same axon, and the number of release-ready vesicles at each synapse is a major determinant of this heterogeneity. Here we identify a second, equally important, mechanism for release heterogeneity at small hippocampal synapses, the inter-synaptic variation of the exocytosis probability of release-ready vesicles. Using concurrent measurements of vesicular pool sizes, vesicular exocytosis rates, and presynaptic Ca²âº dynamics, in the same small hippocampal boutons, we show that the average fusion probability of release-ready vesicles varies among synapses supplied by the same axon with the size of the spike-evoked Ca²âº concentration transient. We further show that synapses with a high vesicular release probability exhibit a lower Ca²âº cooperativity, arguing that this is a direct consequence of increased Ca²âº influx at the active zone. We conclude that variability of neurotransmitter release under basal conditions at small central synapses is accounted for not only by the number of release-ready vesicles, but also by their fusion probabilities, which are set independently of bouton size by variable spike-evoked presynaptic Ca²âº influx.


Subject(s)
Calcium Signaling , Calcium/metabolism , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , Action Potentials/physiology , Animals , Axons/metabolism , Buffers , Exocytosis , Fluorescence , Rats , Synaptic Vesicles/metabolism
18.
Proc Natl Acad Sci U S A ; 108(29): 12113-8, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21724987

ABSTRACT

Latrophilin 1 (LPH1), a neuronal receptor of α-latrotoxin, is implicated in neurotransmitter release and control of presynaptic Ca(2+). As an "adhesion G-protein-coupled receptor," LPH1 can convert cell surface interactions into intracellular signaling. To examine the physiological functions of LPH1, we used LPH1's extracellular domain to purify its endogenous ligand. A single protein of ∼275 kDa was isolated from rat brain and termed Lasso. Peptide sequencing and molecular cloning have shown that Lasso is a splice variant of teneurin-2, a brain-specific orphan cell surface receptor with a function in neuronal pathfinding and synaptogenesis. We show that LPH1 and Lasso interact strongly and specifically. They are always copurified from rat brain extracts. Coculturing cells expressing LPH1 with cells expressing Lasso leads to their mutual attraction and formation of multiple junctions to which both proteins are recruited. Cells expressing LPH1 form chimerical synapses with hippocampal neurons in cocultures; LPH1 and postsynaptic neuronal protein PSD-95 accumulate on opposite sides of these structures. Immunoblotting and immunoelectron microscopy of purified synapses and immunostaining of cultured hippocampal neurons show that LPH1 and Lasso are enriched in synapses; in both systems, LPH1 is presynaptic, whereas Lasso is postsynaptic. A C-terminal fragment of Lasso interacts with LPH1 and induces Ca(2+) signals in presynaptic boutons of hippocampal neurons and in neuroblastoma cells expressing LPH1. Thus, LPH1 and Lasso can form transsynaptic complexes capable of inducing presynaptic Ca(2+) signals, which might affect synaptic functions.


Subject(s)
Calcium Signaling/physiology , Hippocampus/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Peptide/metabolism , Synapses/metabolism , Synaptic Transmission/physiology , Animals , Base Sequence , Cloning, Molecular , Hippocampus/physiology , Immunoblotting , Microscopy, Immunoelectron , Molecular Sequence Data , Rats , Sequence Analysis, DNA
19.
J Biol Chem ; 284(10): 6495-506, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19124473

ABSTRACT

The unusual adhesion G-protein-coupled receptors (aGPCRs) contain large extracellular N-terminal domains, which resemble cell-adhesion receptors, and C-terminal heptahelical domains, which may couple to G-proteins. These receptors are cleaved post-translationally between these domains into two fragments (NTF and CTF). Using the aGPCR latrophilin 1, we previously demonstrated that the fragments behave as independent cell-surface proteins. Upon binding the agonist, alpha-latrotoxin (LTX), latrophilin fragments reassemble and induce intracellular signaling. Our observations raised important questions: is the aGPCR signaling mediated by reassembled fragments or by any non-cleaved receptors? Also, can the fragments originating from distinct aGPCRs form hybrid complexes? To answer these questions, we created two types of chimerical constructs. One contained the CTF of latrophilin joined to the NTF of another aGPCR, EMR2; the resulting protein did not bind LTX but, similar to latrophilin, could couple to G-proteins. In another construct, the NTF of latrophilin was fused with the C terminus of neurexin; this chimera bound LTX but could not signal via G-proteins. Both constructs were efficiently cleaved in cells. When the two constructs were co-expressed, their fragments could cross-interact, as shown by immunoprecipitation. Furthermore, LTX(N4C) induced intracellular Ca2+ signaling only in cells expressing both constructs but not each individual construct. Finally, we demonstrated that fragments of unrelated aGPCRs can be cross-immunoprecipitated from live tissues. Thus, (i) aGPCR fragments behave as independent proteins, (ii) the complementary fragments from distinct aGPCRs can cross-interact, and (iii) these cross-complexes are functionally active. This unusual cross-assembly of aGPCR fragments could couple cell-surface interactions to multiple signaling pathways.


Subject(s)
Calcium Signaling/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Humans , Protein Structure, Tertiary/physiology , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
Ther Adv Neurol Disord ; 1(1): 13-24, 2008 Jul.
Article in English | MEDLINE | ID: mdl-21180561

ABSTRACT

Levetiracetam (LEV) is a new antiepileptic drug that is clinically effective in generalized and partial epilepsy syndromes as sole or add-on medication. Nevertheless, its underlying mechanism of action is poorly understood. It has a unique preclinical profile; unlike other antiepileptic drugs (AEDs), it modulates seizure-activity in animal models of chronic epilepsy with no effect in most animal models of acute seizures. Yet it is effective in acute in-vitro 'seizure' models. A possible explanation for these dichotomous findings is that LEV has different mechanisms of actions, whether given acutely or chronically and in 'epileptic' and control tissue. Here we review the general mechanism of action of AEDs, give an updated and critical overview about the experimental findings of LEV's cellular targets (in particular the synaptic vesicular protein SV2A) and ask whether LEV represents a new class of AED.

SELECTION OF CITATIONS
SEARCH DETAIL