Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 288
Filter
1.
Science ; 385(6705): 168-174, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38900912

ABSTRACT

Intercellular communication in the nervous system occurs through the release of neurotransmitters into the synaptic cleft between neurons. In the presynaptic neuron, the proton pumping vesicular- or vacuolar-type ATPase (V-ATPase) powers neurotransmitter loading into synaptic vesicles (SVs), with the V1 complex dissociating from the membrane region of the enzyme before exocytosis. We isolated SVs from rat brain using SidK, a V-ATPase-binding bacterial effector protein. Single-particle electron cryomicroscopy allowed high-resolution structure determination of V-ATPase within the native SV membrane. In the structure, regularly spaced cholesterol molecules decorate the enzyme's rotor and the abundant SV protein synaptophysin binds the complex stoichiometrically. ATP hydrolysis during vesicle loading results in a loss of the V1 region of V-ATPase from the SV membrane, suggesting that loading is sufficient to induce dissociation of the enzyme.


Subject(s)
Cholesterol , Cryoelectron Microscopy , Synaptic Vesicles , Vacuolar Proton-Translocating ATPases , Animals , Synaptic Vesicles/ultrastructure , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/ultrastructure , Vacuolar Proton-Translocating ATPases/chemistry , Rats , Cholesterol/metabolism , Synaptophysin/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/ultrastructure , Brain/ultrastructure , Brain/enzymology , Hydrolysis
2.
Nature ; 611(7937): 827-834, 2022 11.
Article in English | MEDLINE | ID: mdl-36418452

ABSTRACT

Vacuolar-type adenosine triphosphatases (V-ATPases)1-3 are electrogenic rotary mechanoenzymes structurally related to F-type ATP synthases4,5. They hydrolyse ATP to establish electrochemical proton gradients for a plethora of cellular processes1,3. In neurons, the loading of all neurotransmitters into synaptic vesicles is energized by about one V-ATPase molecule per synaptic vesicle6,7. To shed light on this bona fide single-molecule biological process, we investigated electrogenic proton-pumping by single mammalian-brain V-ATPases in single synaptic vesicles. Here we show that V-ATPases do not pump continuously in time, as suggested by observing the rotation of bacterial homologues8 and assuming strict ATP-proton coupling. Instead, they stochastically switch between three ultralong-lived modes: proton-pumping, inactive and proton-leaky. Notably, direct observation of pumping revealed that physiologically relevant concentrations of ATP do not regulate the intrinsic pumping rate. ATP regulates V-ATPase activity through the switching probability of the proton-pumping mode. By contrast, electrochemical proton gradients regulate the pumping rate and the switching of the pumping and inactive modes. A direct consequence of mode-switching is all-or-none stochastic fluctuations in the electrochemical gradient of synaptic vesicles that would be expected to introduce stochasticity in proton-driven secondary active loading of neurotransmitters and may thus have important implications for neurotransmission. This work reveals and emphasizes the mechanistic and biological importance of ultraslow mode-switching.


Subject(s)
Brain , Mammals , Vacuolar Proton-Translocating ATPases , Animals , Adenosine Triphosphate/metabolism , Brain/enzymology , Brain/metabolism , Mammals/metabolism , Protons , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Neurotransmitter Agents/metabolism , Synaptic Transmission , Time Factors , Kinetics
3.
Elife ; 102021 09 20.
Article in English | MEDLINE | ID: mdl-34543184

ABSTRACT

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.


Subject(s)
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
4.
Elife ; 92020 05 04.
Article in English | MEDLINE | ID: mdl-32364493

ABSTRACT

Mechanisms regulating the turnover of synaptic vesicle (SV) proteins are not well understood. They are thought to require poly-ubiquitination and degradation through proteasome, endo-lysosomal or autophagy-related pathways. Bassoon was shown to negatively regulate presynaptic autophagy in part by scaffolding Atg5. Here, we show that increased autophagy in Bassoon knockout neurons depends on poly-ubiquitination and that the loss of Bassoon leads to elevated levels of ubiquitinated synaptic proteins per se. Our data show that Bassoon knockout neurons have a smaller SV pool size and a higher turnover rate as indicated by a younger pool of SV2. The E3 ligase Parkin is required for increased autophagy in Bassoon-deficient neurons as the knockdown of Parkin normalized autophagy and SV protein levels and rescued impaired SV recycling. These data indicate that Bassoon is a key regulator of SV proteostasis and that Parkin is a key E3 ligase in the autophagy-mediated clearance of SV proteins.


Subject(s)
Autophagy , Hippocampus/enzymology , Nerve Tissue Proteins/deficiency , Presynaptic Terminals/enzymology , Synaptic Vesicles/enzymology , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Female , Hippocampus/ultrastructure , Male , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Presynaptic Terminals/ultrastructure , Proteolysis , Proteostasis , Signal Transduction , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Vesicle-Associated Membrane Protein 2/metabolism
5.
Adv Biol Regul ; 75: 100688, 2020 01.
Article in English | MEDLINE | ID: mdl-31836314

ABSTRACT

Diacylglycerol kinases (DGKs) contribute to an important part of intracellular signaling because, in addition to reducing diacylglycerol levels, they generate phosphatidic acid (PtdOH) Recent research has led to the discovery of ten mammalian DGK isoforms, all of which are found in the mammalian brain. Many of these isoforms have studied functions within the brain, while others lack such understanding in regards to neuronal roles, regulation, and structural dynamics. However, while previously a neuronal function for DGKθ was unknown, it was recently found that DGKθ is required for the regulation of synaptic vesicle endocytosis and work is currently being conducted to elucidate the mechanism behind this regulation. Here we will review some of the roles of all mammalian DGKs and hypothesize additional roles. We will address the topic of redundancy among the ten DGK isoforms and discuss the possibility that DGKθ, among other DGKs, may have unstudied postsynaptic functions. We also hypothesize that in addition to DGKθ's presynaptic endocytic role, DGKθ might also regulate the endocytosis of AMPA receptors and other postsynaptic membrane proteins.


Subject(s)
Diacylglycerol Kinase/metabolism , Endocytosis , Neurons/enzymology , Synaptic Membranes/enzymology , Synaptic Vesicles/enzymology , Animals , Diacylglycerol Kinase/genetics , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Phosphatidic Acids/genetics , Phosphatidic Acids/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synaptic Membranes/genetics , Synaptic Vesicles/genetics
6.
Elife ; 72018 04 13.
Article in English | MEDLINE | ID: mdl-29652249

ABSTRACT

Newly-formed synaptic vesicles (SVs) are rapidly acidified by vacuolar adenosine triphosphatases (vATPases), generating a proton electrochemical gradient that drives neurotransmitter loading. Clathrin-mediated endocytosis is needed for the formation of new SVs, yet it is unclear when endocytosed vesicles acidify and refill at the synapse. Here, we isolated clathrin-coated vesicles (CCVs) from mouse brain to measure their acidification directly at the single vesicle level. We observed that the ATP-induced acidification of CCVs was strikingly reduced in comparison to SVs. Remarkably, when the coat was removed from CCVs, uncoated vesicles regained ATP-dependent acidification, demonstrating that CCVs contain the functional vATPase, yet its function is inhibited by the clathrin coat. Considering the known structures of the vATPase and clathrin coat, we propose a model in which the formation of the coat surrounds the vATPase and blocks its activity. Such inhibition is likely fundamental for the proper timing of SV refilling.


Subject(s)
Adenosine Triphosphate/metabolism , Clathrin-Coated Vesicles/enzymology , Clathrin-Coated Vesicles/metabolism , Clathrin/metabolism , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Animals , Brain/metabolism , Hydrolysis , Mice
7.
Mol Cell Biochem ; 444(1-2): 1-13, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29159770

ABSTRACT

The JNK-interacting protein 3 (JIP3) is a molecular scaffold, expressed predominantly in neurons, that serves to coordinate the activation of the c-Jun N-terminal kinase (JNK) by binding to JNK and the upstream kinases involved in its activation. The JNK pathway is involved in the regulation of many cellular processes including the control of cell survival, cell death and differentiation. JIP3 also associates with microtubule motor proteins such as kinesin and dynein and is likely an adapter protein involved in the tethering of vesicular cargoes to the motors involved in axonal transport in neurons. We have used immunofluorescence microscopy and biochemical fractionation to investigate the subcellular distribution of JIP3 in relation to JNK and to vesicular and organelle markers in rat pheochromocytoma cells (PC12) differentiating in response to nerve growth factor. In differentiated PC12 cells, JIP3 was seen to accumulate in growth cones at the tips of developing neurites where it co-localised with both JNK and the JNK substrate paxillin. Cellular fractionation of PC12 cells showed that JIP3 was associated with a subpopulation of vesicles in the microsomal fraction, distinct from synaptic vesicles, likely to be an anterograde-directed exocytic vesicle pool. In differentiated PC12 cells, JIP3 did not appear to associate with retrograde endosomal vesicles thought to be involved in signalling axonal injury. Together, these observations indicate that JIP3 may be involved in transporting vesicular cargoes to the growth cones of PC12 cells, possibly targeting JNK to its substrate paxillin, and thus facilitating neurite outgrowth.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation , Growth Cones/enzymology , Nerve Tissue Proteins/metabolism , Neurites/enzymology , Secretory Vesicles/enzymology , Synaptic Vesicles/enzymology , Animals , PC12 Cells , Rats
8.
Adv Biol Regul ; 67: 141-147, 2018 01.
Article in English | MEDLINE | ID: mdl-28986032

ABSTRACT

In cortical and hippocampal neurons of the mammalian brain, the synaptic vesicle cycle is a series of steps that tightly regulate exo- and endocytosis of vesicles. Many proteins contribute to this regulation, but lipids have recently emerged as critical regulators as well. Of all the many lipid signaling molecules, phosphatidic acid is important to the physical processes of membrane fusion. Therefore, the lipid-metabolizing enzymes that produce phosphatidic acid are vital to the regulation of the cycle. Our lab is particularly interested in the potential regulatory mechanisms and neuronal roles of two phosphatidic acid-producing enzymes: diacylglycerol kinase theta (DGKθ) and phospholipase D (PLD). We recently discovered a regulatory role of DGKθ on evoked endocytosis (Goldschmidt et al., 2016). In addition to this enzyme, studies implicate PLD1 in neurotransmission, although its precise role is of some debate. Altogether, the production of phosphatidic acid by these enzymes offer an interesting and novel pathway for the regulation of the synaptic vesicle cycle.


Subject(s)
Lipid Metabolism/physiology , Neurons/metabolism , Phosphatidic Acids/metabolism , Phospholipase D/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/enzymology , Animals , Endocytosis/physiology , Humans , Phosphatidic Acids/genetics , Phospholipase D/genetics , Synaptic Vesicles/genetics
9.
J Biol Chem ; 292(34): 14092-14107, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28637871

ABSTRACT

Tyrosine hydroxylase (TH) catalyzes the conversion of l-tyrosine into l-DOPA, which is the rate-limiting step in the synthesis of catecholamines, such as dopamine, in dopaminergergic neurons. Low dopamine levels and death of the dopaminergic neurons are hallmarks of Parkinson's disease (PD), where α-synuclein is also a key player. TH is highly regulated, notably by phosphorylation of several Ser/Thr residues in the N-terminal tail. However, the functional role of TH phosphorylation at the Ser-31 site (THSer(P)-31) remains unclear. Here, we report that THSer(P)-31 co-distributes with the Golgi complex and synaptic-like vesicles in rat and human dopaminergic cells. We also found that the TH microsomal fraction content decreases after inhibition of cyclin-dependent kinase 5 (Cdk5) and ERK1/2. The cellular distribution of an overexpressed phospho-null mutant, TH1-S31A, was restricted to the soma of neuroblastoma cells, with decreased association with the microsomal fraction, whereas a phospho-mimic mutant, TH1-S31E, was distributed throughout the soma and neurites. TH1-S31E associated with vesicular monoamine transporter 2 (VMAT2) and α-synuclein in neuroblastoma cells, and endogenous THSer(P)-31 was detected in VMAT2- and α-synuclein-immunoprecipitated mouse brain samples. Microtubule disruption or co-transfection with α-synuclein A53T, a PD-associated mutation, caused TH1-S31E accumulation in the cell soma. Our results indicate that Ser-31 phosphorylation may regulate TH subcellular localization by enabling its transport along microtubules, notably toward the projection terminals. These findings disclose a new mechanism of TH regulation by phosphorylation and reveal its interaction with key players in PD, opening up new research avenues for better understanding dopamine synthesis in physiological and pathological states.


Subject(s)
Dopaminergic Neurons/enzymology , Golgi Apparatus/enzymology , Microtubules/enzymology , Protein Processing, Post-Translational , Serine/metabolism , Synaptic Vesicles/enzymology , Tyrosine 3-Monooxygenase/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Golgi Apparatus/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/metabolism , Mutagenesis, Site-Directed , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Protein Transport , Rats , Recombinant Fusion Proteins/metabolism , Synaptic Vesicles/metabolism , Tyrosine 3-Monooxygenase/genetics
10.
Neuron ; 90(2): 278-91, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27068791

ABSTRACT

Changes in neuronal activity create local and transient changes in energy demands at synapses. Here we discover a metabolic compartment that forms in vivo near synapses to meet local energy demands and support synaptic function in Caenorhabditis elegans neurons. Under conditions of energy stress, glycolytic enzymes redistribute from a diffuse localization in the cytoplasm to a punctate localization adjacent to synapses. Glycolytic enzymes colocalize, suggesting the ad hoc formation of a glycolysis compartment, or a "glycolytic metabolon," that can maintain local levels of ATP. Local formation of the glycolytic metabolon is dependent on presynaptic scaffolding proteins, and disruption of the glycolytic metabolon blocks the synaptic vesicle cycle, impairs synaptic recovery, and affects locomotion. Our studies indicate that under energy stress conditions, energy demands in C. elegans synapses are met locally through the assembly of a glycolytic metabolon to sustain synaptic function and behavior. VIDEO ABSTRACT.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/enzymology , Phosphofructokinase-1/metabolism , Presynaptic Terminals/enzymology , Presynaptic Terminals/physiology , Stress, Physiological , Animals , Caenorhabditis elegans/metabolism , Endocytosis , Hypoxia , Metabolomics , Mutation , Presynaptic Terminals/metabolism , Synaptic Vesicles/enzymology , Synaptic Vesicles/metabolism
11.
J Cell Biol ; 212(7): 789-801, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-27022091

ABSTRACT

Differentiation of the presynaptic terminal is a complex and rapid event that normally occurs in spatially specific axonal regions distant from the soma; thus, it is believed to be dependent on intra-axonal mechanisms. However, the full nature of the local events governing presynaptic assembly remains unknown. Herein, we investigated the involvement of the ubiquitin-proteasome system (UPS), the major degradative pathway, in the local modulation of presynaptic differentiation. We found that proteasome inhibition has a synaptogenic effect on isolated axons. In addition, formation of a stable cluster of synaptic vesicles onto a postsynaptic partner occurs in parallel to an on-site decrease in proteasome degradation. Accumulation of ubiquitinated proteins at nascent sites is a local trigger for presynaptic clustering. Finally, proteasome-related ubiquitin chains (K11 and K48) function as signals for the assembly of presynaptic terminals. Collectively, we propose a new axon-intrinsic mechanism for presynaptic assembly through local UPS inhibition. Subsequent on-site accumulation of proteins in their polyubiquitinated state triggers formation of presynapses.


Subject(s)
Cell Differentiation , Hippocampus/enzymology , Polyubiquitin/metabolism , Presynaptic Terminals/enzymology , Proteasome Endopeptidase Complex/metabolism , Ubiquitinated Proteins/metabolism , Animals , Axons/enzymology , Cell Differentiation/drug effects , Cells, Cultured , Hippocampus/drug effects , Hippocampus/embryology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Presynaptic Terminals/drug effects , Proteasome Inhibitors/pharmacology , Proteolysis , Rats, Wistar , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , Synaptic Vesicles/enzymology , Time Factors , Time-Lapse Imaging , Transfection , Ubiquitination
12.
J Biol Chem ; 291(4): 1948-1956, 2016 Jan 22.
Article in English | MEDLINE | ID: mdl-26627835

ABSTRACT

Variation in PPP3CC, the gene that encodes the γ isoform of the calcineurin catalytic subunit, has been reported to be associated with schizophrenia. Because of its low expression level in most tissues, there has been little research devoted to the specific function of the calcineurin Aγ (CNAγ) versus the calcineurin Aα (CNAα) and calcineurin Aß (CNAß) catalytic isoforms. Consequently, we have a limited understanding of the role of altered CNAγ function in psychiatric disease. In this study, we demonstrate that CNAγ is present in the rodent and human brain and dephosphorylates a presynaptic substrate of calcineurin. Through a combination of immunocytochemistry and immuno-EM, we further show that CNAγ is localized to presynaptic terminals in hippocampal neurons. Critically, we demonstrate that RNAi-mediated knockdown of CNAγ leads to a disruption of synaptic vesicle cycling in cultured rat hippocampal neurons. These data indicate that CNAγ regulates a critical aspect of synaptic vesicle cycling and suggest that variation in PPP3CC may contribute to psychiatric disease by altering presynaptic function.


Subject(s)
Calcineurin/metabolism , Endocytosis , Synaptic Vesicles/enzymology , Animals , Calcineurin/genetics , Cells, Cultured , Hippocampus/cytology , Hippocampus/enzymology , Humans , Male , Mice , Mice, Inbred BALB C , Neurons/enzymology , Rats , Synaptic Vesicles/genetics
13.
Toxicon ; 107(Pt A): 32-6, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26130523

ABSTRACT

Botulinum neurotoxins (BoNTs) are Janus toxins, as they are at the same time the most deadly substances known and one of the safest drugs used in human therapy. They specifically block neurotransmission at peripheral nerves through the proteolysis of SNARE proteins, i.e. the essential proteins which are the core of the neuroexocytosis machinery. Even if BoNTs are traditionally known as seven main serotypes, their actual number is much higher as each serotype exists in many different subtypes, with individual biological properties and little antigenic relations. Since BoNTs can be used as biological weapons, and the only currently available therapy is based on immunological approaches, the existence of so many different subtypes is a major safety problem. Nevertheless, all BoNT isoforms are structurally similar and intoxicate peripheral nerve endings via a conserved mechanism. They consist of two chains linked by a unique disulphide bond which must be reduced to enable their toxicity. We found that thioredoxin 1 and its reductase compose the cell redox system responsible for this reduction, and its inhibition via specific chemicals significantly reduces BoNTs activity, in vitro as well as in vivo. Such molecules can be considered as lead compounds for the development of pan-inhibitors.


Subject(s)
Botulinum Toxins/metabolism , Synaptic Vesicles/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Botulinum Antitoxin/metabolism , Humans , Oxidation-Reduction , Peripheral Nerves/enzymology , Peripheral Nerves/metabolism , Protein Isoforms/metabolism , Synaptic Vesicles/enzymology , Thioredoxin-Disulfide Reductase/antagonists & inhibitors
14.
Biochem Biophys Res Commun ; 456(1): 145-50, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25446113

ABSTRACT

Previous studies demonstrated that depletion of membrane cholesterol by 10mM methyl-beta-cyclodextrin (MCD) results in increased spontaneous exocytosis at both peripheral and central synapses. Here, we investigated the role of protein kinase C in the enhancement of spontaneous exocytosis at frog motor nerve terminals after cholesterol depletion using electrophysiological and optical methods. Inhibition of the protein kinase C by myristoylated peptide and chelerythrine chloride prevented MCD-induced increases in FM1-43 unloading, whereas the frequency of spontaneous postsynaptic events remained enhanced. The increase in FM1-43 unloading still could be observed if sulforhodamine 101 (the water soluble FM1-43 quencher that can pass through the fusion pore) was added to the extracellular solution. This suggests a possibility that exocytosis of synaptic vesicles under these conditions could occur through the kiss-and-run mechanism with the formation of a transient fusion pore. Inhibition of phospholipase C did not lead to similar change in MCD-induced exocytosis.


Subject(s)
Cholesterol/metabolism , Enzyme Inhibitors/chemistry , Exocytosis/drug effects , Protein Kinase C/antagonists & inhibitors , Synaptic Vesicles/enzymology , Animals , Benzophenanthridines/chemistry , Electrophysiology , Microscopy, Fluorescence , Myristic Acid/chemistry , Neuromuscular Junction/drug effects , Protein Kinase C/metabolism , Ranidae , Rhodamines/chemistry , Synaptic Transmission/drug effects , Synaptic Vesicles/drug effects , Type C Phospholipases/metabolism , beta-Cyclodextrins/chemistry
15.
Ross Fiziol Zh Im I M Sechenova ; 100(4): 385-93, 2014 Apr.
Article in Russian | MEDLINE | ID: mdl-25272450

ABSTRACT

Calpains are calcium-activated neutral cysteine proteases, involved in the regulation of a number of physiological functions. Substrates of calpains include receptors, kinases, phosphatases, cytoskeleton and synaptosomal proteins. Some of them undergo complete degradation, though most of the substrates are subjected to limited proteolysis, which results in proteins having new properties. In the following review, we discuss involvement of calpains in the regulation of synapse structure and function. Namely, calpains participate in the regulation of synthesis, release and reuptake of neurotransmitters, modulation of receptors, stabilization or destabilization of the neuronal cytoskeleton. However, uncontrolled hyperactivation of calpains leads to dysregulation of these processes causing neuronal death.


Subject(s)
Calpain/metabolism , Neurotransmitter Agents/metabolism , Proteolysis , Synaptic Transmission/physiology , Synaptic Vesicles/enzymology , Animals , Humans
16.
Cell Rep ; 8(6): 1870-1878, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25220457

ABSTRACT

Botulinum neurotoxins consist of a metalloprotease linked via a conserved interchain disulfide bond to a heavy chain responsible for neurospecific binding and translocation of the enzymatic domain in the nerve terminal cytosol. The metalloprotease activity is enabled upon disulfide reduction and causes neuroparalysis by cleaving the SNARE proteins. Here, we show that the thioredoxin reductase-thioredoxin protein disulfide-reducing system is present on synaptic vesicles and that it is functional and responsible for the reduction of the interchain disulfide of botulinum neurotoxin serotypes A, C, and E. Specific inhibitors of thioredoxin reductase or thioredoxin prevent intoxication of cultured neurons in a dose-dependent manner and are also very effective inhibitors of the paralysis of the neuromuscular junction. We found that this group of inhibitors of botulinum neurotoxins is very effective in vivo. Most of them are nontoxic and are good candidates as preventive and therapeutic drugs for human botulism.


Subject(s)
Botulinum Toxins/toxicity , Paralysis/prevention & control , Synaptic Vesicles/drug effects , Synaptic Vesicles/enzymology , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Cytoplasm/metabolism , Disulfides/pharmacology , Disulfides/therapeutic use , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Imidazoles/pharmacology , Imidazoles/therapeutic use , Male , Mice , Neurons/drug effects , Neurons/metabolism , Paralysis/etiology , Serotyping , Synaptosomal-Associated Protein 25/metabolism , Thioredoxin-Disulfide Reductase/antagonists & inhibitors , Thioredoxins/antagonists & inhibitors
17.
Neuroscience ; 280: 275-81, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25194787

ABSTRACT

Lipoprotein lipase (LPL) is expressed at high levels in hippocampal neurons, although its function is unclear. We previously reported that LPL-deficient mice have learning and memory impairment and fewer synaptic vesicles in hippocampal neurons, but properties of synaptic activity in LPL-deficient neurons remain unexplored. In this study, we found reduced frequency of miniature excitatory postsynaptic currents (mEPSCs) and readily releasable pool (RRP) size in LPL-deficient neurons, which led to presynaptic dysfunction and plasticity impairment without altering postsynaptic activity. We demonstrated that synaptic vesicle recycling, which is known to play an important role in maintaining the RRP size in active synapses, is impaired in LPL-deficient neurons. Moreover, lipid assay revealed deficient docosahexaenoic acid (DHA) and arachidonic acid (AA) in the hippocampus of LPL-deficient mice; exogenous DHA or AA supplement partially restored synaptic vesicle recycling capability. These results suggest that impaired synaptic vesicle recycling results from deficient DHA and AA and contributes to the presynaptic dysfunction and plasticity impairment in LPL-deficient neurons.


Subject(s)
Hippocampus/physiopathology , Lipoprotein Lipase/deficiency , Presynaptic Terminals/enzymology , Synaptic Vesicles/enzymology , Animals , Arachidonic Acid/administration & dosage , Arachidonic Acid/metabolism , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/metabolism , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gas Chromatography-Mass Spectrometry , Hippocampus/drug effects , Hyperlipoproteinemia Type I/drug therapy , Hyperlipoproteinemia Type I/physiopathology , Lipoprotein Lipase/genetics , Male , Mice, Inbred C57BL , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Patch-Clamp Techniques , Presynaptic Terminals/drug effects , Pyramidal Cells/drug effects , Pyramidal Cells/physiopathology , Synaptic Vesicles/drug effects , Tissue Culture Techniques
18.
Nat Commun ; 5: 4246, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24977345

ABSTRACT

Phosphorylation has emerged as a crucial regulatory mechanism in the nervous system to integrate the dynamic signalling required for proper synaptic development, function and plasticity, particularly during changes in neuronal activity. Here we present evidence that Minibrain (Mnb; also known as Dyrk1A), a serine/threonine kinase implicated in autism spectrum disorder and Down syndrome, is required presynaptically for normal synaptic growth and rapid synaptic vesicle endocytosis at the Drosophila neuromuscular junction (NMJ). We find that Mnb-dependent phosphorylation of Synaptojanin (Synj) is required, in vivo, for complex endocytic protein interactions and to enhance Synj activity. Neuronal stimulation drives Mnb mobilization to endocytic zones and triggers Mnb-dependent phosphorylation of Synj. Our data identify Mnb as a synaptic kinase that promotes efficient synaptic vesicle recycling by dynamically calibrating Synj function at the Drosophila NMJ, and in turn endocytic capacity, to adapt to conditions of high synaptic activity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/enzymology , Nerve Tissue Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Serine-Threonine Kinases/metabolism , Synaptic Vesicles/enzymology , Animals , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Endocytosis , Nerve Tissue Proteins/genetics , Neuromuscular Junction/enzymology , Neuromuscular Junction/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Synapses/enzymology , Synaptic Vesicles/genetics
19.
J Cell Biol ; 205(1): 7-9, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24733582

ABSTRACT

Neurons fire by releasing neurotransmitters via fusion of synaptic vesicles with the plasma membrane. Fusion can be evoked by an incoming signal from a preceding neuron or can occur spontaneously. Synaptic vesicle fusion requires the formation of trans complexes between SNAREs as well as Ca(2+) ions. Wang et al. (2014. J. Cell Biol. http://dx.doi.org/jcb.201312109) now find that the Ca(2+)-binding protein Calmodulin promotes spontaneous release and SNARE complex formation via its interaction with the V0 sector of the V-ATPase.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Neuromuscular Junction/enzymology , Qa-SNARE Proteins/metabolism , Synaptic Transmission , Synaptic Vesicles/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Animals
20.
J Cell Biol ; 205(1): 21-31, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24733584

ABSTRACT

Most chemical neurotransmission occurs through Ca(2+)-dependent evoked or spontaneous vesicle exocytosis. In both cases, Ca(2+) sensing is thought to occur shortly before exocytosis. In this paper, we provide evidence that the Ca(2+) dependence of spontaneous vesicle release may partly result from an earlier requirement of Ca(2+) for the assembly of soluble N-ethylmaleimide-sensitive fusion attachment protein receptor (SNARE) complexes. We show that the neuronal vacuolar-type H(+)-adenosine triphosphatase V0 subunit a1 (V100) can regulate the formation of SNARE complexes in a Ca(2+)-Calmodulin (CaM)-dependent manner. Ca(2+)-CaM regulation of V100 is not required for vesicle acidification. Specific disruption of the Ca(2+)-dependent regulation of V100 by CaM led to a >90% loss of spontaneous release but only had a mild effect on evoked release at Drosophila melanogaster embryo neuromuscular junctions. Our data suggest that Ca(2+)-CaM regulation of V100 may control SNARE complex assembly for a subset of synaptic vesicles that sustain spontaneous release.


Subject(s)
Calcium/metabolism , Calmodulin/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Neuromuscular Junction/enzymology , Qa-SNARE Proteins/metabolism , Synaptic Transmission , Synaptic Vesicles/enzymology , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Electric Stimulation , Hydrogen-Ion Concentration , Lysosomes/enzymology , Multiprotein Complexes , Protein Binding , Protein Subunits , Qa-SNARE Proteins/genetics , Time Factors , Vacuolar Proton-Translocating ATPases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...