Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4120, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37433762

ABSTRACT

In Parkinson's disease (PD), motor dysfunctions only become apparent after extensive loss of DA innervation. This resilience has been hypothesized to be due to the ability of many motor behaviors to be sustained through a diffuse basal tone of DA; but experimental evidence for this is limited. Here we show that conditional deletion of the calcium sensor synaptotagmin-1 (Syt1) in DA neurons (Syt1 cKODA mice) abrogates most activity-dependent axonal DA release in the striatum and mesencephalon, leaving somatodendritic (STD) DA release intact. Strikingly, Syt1 cKODA mice showed intact performance in multiple unconditioned DA-dependent motor tasks and even in a task evaluating conditioned motivation for food. Considering that basal extracellular DA levels in the striatum were unchanged, our findings suggest that activity-dependent DA release is dispensable for such tasks and that they can be sustained by a basal tone of extracellular DA. Taken together, our findings reveal the striking resilience of DA-dependent motor functions in the context of a near-abolition of phasic DA release, shedding new light on why extensive loss of DA innervation is required to reveal motor dysfunctions in PD.


Subject(s)
Dopamine , Parkinson Disease , Synaptotagmin I , Animals , Mice , Calcium , Corpus Striatum , Neostriatum , Niacinamide , Synaptotagmin I/physiology
2.
Mol Cell Neurosci ; 112: 103613, 2021 04.
Article in English | MEDLINE | ID: mdl-33753311

ABSTRACT

Presynaptic neurotransmitter release is strictly regulated by SNARE proteins, Ca2+ and a number of Ca2+ sensors including synaptotagmins (Syts) and Double C2 domain proteins (Doc2s). More than seventy years after the original description of spontaneous release, the mechanism that regulates this process is still poorly understood. Syt-1, Syt7 and Doc2 proteins contribute predominantly, but not exclusively, to synchronous, asynchronous and spontaneous phases of release. The proteins share a conserved tandem C2 domain architecture, but are functionally diverse in their subcellular location, Ca2+-binding properties and protein interactions. In absence of Syt-1, Doc2a and -b, neurons still exhibit spontaneous vesicle fusion which remains Ca2+-sensitive, suggesting the existence of additional sensors. Here, we selected Doc2c, rabphilin-3a and Syt-7 as three potential Ca2+ sensors for their sequence homology with Syt-1 and Doc2b. We genetically ablated each candidate gene in absence of Doc2a and -b and investigated spontaneous and evoked release in glutamatergic hippocampal neurons, cultured either in networks or on microglial islands (autapses). The removal of Doc2c had no effect on spontaneous or evoked release. Syt-7 removal also did not affect spontaneous release, although it altered short-term plasticity by accentuating short-term depression. The removal of rabphilin caused an increased spontaneous release frequency in network cultures, an effect that was not observed in autapses. Taken together, we conclude that Doc2c and Syt-7 do not affect spontaneous release of glutamate in hippocampal neurons, while our results suggest a possible regulatory role of rabphilin-3a in neuronal networks. These findings importantly narrow down the repertoire of synaptic Ca2+ sensors that may be implicated in the spontaneous release of glutamate.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Calcium-Binding Proteins/physiology , Calcium/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Synaptotagmin I/physiology , Vesicular Transport Proteins/physiology , Action Potentials , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cells, Cultured , Conserved Sequence , Glutamic Acid/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Protein Domains , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Synaptotagmin I/chemistry , Synaptotagmin I/deficiency , Synaptotagmin I/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/deficiency , Vesicular Transport Proteins/genetics , Rabphilin-3A
3.
J Neurochem ; 154(6): 598-617, 2020 09.
Article in English | MEDLINE | ID: mdl-32058590

ABSTRACT

Synaptotagmin-7 (Syt-7) is one of two major calcium sensors for exocytosis in adrenal chromaffin cells, the other being synaptotagmin-1 (Syt-1). Despite a broad appreciation for the importance of Syt-7, questions remain as to its localization, function in mediating discharge of dense core granule cargos, and role in triggering release in response to physiological stimulation. These questions were addressed using two distinct experimental preparations-mouse chromaffin cells lacking endogenous Syt-7 (KO cells) and a reconstituted system employing cell-derived granules expressing either Syt-7 or Syt-1. First, using immunofluorescence imaging and subcellular fractionation, it is shown that Syt-7 is widely distributed in organelles, including dense core granules. Total internal reflection fluorescence (TIRF) imaging demonstrates that the kinetics and probability of granule fusion in Syt-7 KO cells stimulated by a native secretagogue, acetylcholine, are markedly lower than in WT cells. When fusion is observed, fluorescent cargo proteins are discharged more rapidly when only Syt-1 is available to facilitate release. To determine the extent to which the aforementioned results are attributable purely to Syt-7, granules expressing only Syt-7 or Syt-1 were triggered to fuse on planar supported bilayers bearing plasma membrane SNARE proteins. Here, as in cells, Syt-7 confers substantially greater calcium sensitivity to granule fusion than Syt-1 and slows the rate at which cargos are released. Overall, this study demonstrates that by virtue of its high affinity for calcium and effects on fusion pore expansion, Syt-7 plays a central role in regulating secretory output from adrenal chromaffin cells.


Subject(s)
Chromaffin Granules/physiology , Receptors, Calcium-Sensing/physiology , Synaptotagmins/genetics , Synaptotagmins/physiology , Acetylcholine/pharmacology , Animals , Calcium Signaling/genetics , Calcium Signaling/physiology , Cell Movement/genetics , Cell Movement/physiology , Electrophysiological Phenomena , Exocytosis , Female , Kinetics , Male , Membrane Fusion , Mice , Mice, Inbred C57BL , Mice, Knockout , PC12 Cells , Rats , SNARE Proteins/metabolism , Subcellular Fractions/metabolism , Synaptotagmin I/physiology
4.
J Neurochem ; 152(1): 48-60, 2020 01.
Article in English | MEDLINE | ID: mdl-31587282

ABSTRACT

Phosphatidylserine (PS), a negatively charged phospholipid present predominantly at the inner leaflet of the plasma membrane, has been widely implicated in many cellular processes including membrane trafficking. Along this line, PS has been demonstrated to be important for endocytosis, however, the involved mechanisms remain uncertain. By monitoring clathrin-mediated endocytosis (CME) of single vesicles in mouse chromaffin cells using cell-attached capacitance measurements that offer millisecond time resolution, we demonstrate in the present study that the fission-pore duration is reduced by PS addition, indicating a stimulatory role of PS in regulating the dynamics of vesicle fission during CME. Furthermore, our results show that the PS-mediated effect on the fission-pore duration is Ca2+ -dependent and abolished in the absence of synaptotagmin 1 (Syt1), implying that Syt1 is necessary for the stimulatory role of PS in vesicle fission during CME. Consistently, a Syt1 mutant with a defective PS-Syt1 interaction increases the fission-pore duration. Taken together, our study suggests that PS-Syt1 interaction may be critical in regulating fission dynamics during CME.


Subject(s)
Chromaffin Cells/physiology , Clathrin-Coated Vesicles/physiology , Clathrin/physiology , Phosphatidylserines/physiology , Animals , Cells, Cultured , Endocytosis/physiology , Exocytosis/physiology , Female , Gene Knockout Techniques , Male , Mice , Mice, Inbred C57BL , Synaptotagmin I/genetics , Synaptotagmin I/physiology
5.
J Biol Chem ; 294(28): 10942-10953, 2019 07 12.
Article in English | MEDLINE | ID: mdl-31147445

ABSTRACT

Exocytosis mediates the release of neurotransmitters and hormones from neurons and neuroendocrine cells. Tandem C2 domain proteins in the synaptotagmin (syt) and double C2 domain (Doc2) families regulate exocytotic membrane fusion via direct interactions with Ca2+ and phospholipid bilayers. Syt1 is a fast-acting, low-affinity Ca2+ sensor that penetrates membranes upon binding Ca2+ to trigger synchronous vesicle fusion. The closely related Doc2ß is a slow-acting, high-affinity Ca2+ sensor that triggers spontaneous and asynchronous vesicle fusion, but whether it also penetrates membranes is unknown. Both syt1 and Doc2ß bind the dynamically regulated plasma membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2), but it is unclear whether PIP2 serves only as a membrane contact or enables specialized membrane-binding modes by these Ca2+ sensors. Furthermore, it has been shown that PIP2 uncaging can trigger rapid, syt1-dependent exocytosis in the absence of Ca2+ influx, suggesting that current models for the action of these Ca2+ sensors are incomplete. Here, using a series of steady-state and time-resolved fluorescence measurements, we show that Doc2ß, like syt1, penetrates membranes in a Ca2+-dependent manner. Unexpectedly, we observed that PIP2 can drive membrane penetration by both syt1 and Doc2ß in the absence of Ca2+, providing a plausible mechanism for Ca2+-independent, PIP2-dependent exocytosis. Quantitative measurements of penetration depth revealed that, in the presence of Ca2+, PIP2 drives Doc2ß, but not syt1, substantially deeper into the membrane, defining a biophysical regulatory mechanism specific to this high-affinity Ca2+ sensor. Our results provide evidence of a novel role for PIP2 in regulating, and under some circumstances triggering, exocytosis.


Subject(s)
Calcium-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Synaptotagmin I/metabolism , Animals , C2 Domains , Calcium/metabolism , Calcium-Binding Proteins/physiology , Cell Membrane/metabolism , Exocytosis/physiology , Membrane Fusion , Membrane Lipids/metabolism , Nerve Tissue Proteins/physiology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Phosphatidylinositol 4,5-Diphosphate/physiology , Phosphatidylinositols/metabolism , Protein Binding , Synapses/physiology , Synaptic Transmission , Synaptic Vesicles/metabolism , Synaptotagmin I/physiology
6.
Science ; 363(6430): 948-955, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30819957

ABSTRACT

We investigated the roles of components of neuronal synapses for development of the Drosophila air sac primordium (ASP). The ASP, an epithelial tube, extends specialized signaling filopodia called cytonemes that take up signals such as Dpp (Decapentaplegic, a homolog of the vertebrate bone morphogenetic protein) from the wing imaginal disc. Dpp signaling in the ASP was compromised if disc cells lacked Synaptobrevin and Synaptotagmin-1 (which function in vesicle transport at neuronal synapses), the glutamate transporter, and a voltage-gated calcium channel, or if ASP cells lacked Synaptotagmin-4 or the glutamate receptor GluRII. Transient elevations of intracellular calcium in ASP cytonemes correlate with signaling activity. Calcium transients in ASP cells depend on GluRII, are activated by l-glutamate and by stimulation of an optogenetic ion channel expressed in the wing disc, and are inhibited by EGTA and by the GluR inhibitor NASPM (1-naphthylacetyl spermine trihydrochloride). Activation of GluRII is essential but not sufficient for signaling. Cytoneme-mediated signaling is glutamatergic.


Subject(s)
Calcium Signaling , Drosophila Proteins/physiology , Glutamates/physiology , Imaginal Discs/physiology , Receptors, Ionotropic Glutamate/physiology , Synapses/physiology , Animals , Animals, Genetically Modified , Calcium Channels/physiology , Drosophila melanogaster/physiology , Optical Imaging , Pseudopodia/physiology , R-SNARE Proteins/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/physiology , Synaptotagmin I/physiology , Tissue Culture Techniques
7.
Cell Rep ; 26(9): 2340-2352.e5, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30811985

ABSTRACT

Information transfer across CNS synapses depends on the very low basal vesicle fusion rate and the ability to rapidly upregulate that rate upon Ca2+ influx. We show that local electrostatic repulsion participates in creating an energy barrier, which limits spontaneous synaptic transmission. The barrier amplitude is increased by negative charges and decreased by positive charges on the SNARE-complex surface. Strikingly, the effect of charges on the barrier is additive and this extends to evoked transmission, but with a shallower charge dependence. Action potential-driven synaptic release is equivalent to the abrupt addition of ∼35 positive charges to the fusion machine. Within an electrostatic model for triggering, the Ca2+ sensor synaptotagmin-1 contributes ∼18 charges by binding Ca2+, while also modulating the fusion barrier at rest. Thus, the energy barrier for synaptic vesicle fusion has a large electrostatic component, allowing synaptotagmin-1 to act as an electrostatic switch and modulator to trigger vesicle fusion.


Subject(s)
SNARE Proteins/chemistry , Synaptic Transmission , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Excitatory Postsynaptic Potentials , Female , Male , Membrane Fusion , Mice , Mice, Knockout , Models, Neurological , Static Electricity , Synaptotagmin I/physiology
8.
Biochim Biophys Acta Biomembr ; 1861(3): 677-684, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30615859

ABSTRACT

Abundant attention has focused on synaptotagmin's C2 domains, but less is known about the structure and function of its other regions. Here, we synthesized the N-acetylated, C-end amidated and Cys-palmitated peptide (VLTCCFCICK KCLFKKKNKK K) which includes the fatty acylated cysteine residues in the membrane-affiliated domain of synaptotagmin-1. Fourier-transform infrared spectrometry indicated that this peptide's conformation is influenced by environmental polarity. In artificial bilayer membranes, this peptide exhibited abundant ß-structure. Electron microscopy revealed that this peptide also promoted the stacking of liposome membranes. Together these results suggest that the fatty acylated region of synaptotagmin-1 is likely to adopt ß-structure in biological membranes. This preference for ß-structure versus α-helix has functional implications for the role of synaptotagmin-1 in synaptic vesicle exocytosis.


Subject(s)
Fatty Acids/chemistry , Fatty Acids/metabolism , Synaptotagmin I/chemistry , Synaptotagmin I/physiology , Acylation , Exocytosis/physiology , Humans , Liposomes/chemistry , Liposomes/metabolism , Mass Spectrometry , Membrane Fusion , Protein Domains , Protein Structure, Secondary/physiology , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship , Synaptic Transmission , Synaptotagmin I/metabolism
9.
Brain ; 141(9): 2576-2591, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30107533

ABSTRACT

Synaptotagmin 1 (SYT1) is a critical mediator of fast, synchronous, calcium-dependent neurotransmitter release and also modulates synaptic vesicle endocytosis. This paper describes 11 patients with de novo heterozygous missense mutations in SYT1. All mutations alter highly conserved residues, and cluster in two regions of the SYT1 C2B domain at positions Met303 (M303K), Asp304 (D304G), Asp366 (D366E), Ile368 (I368T) and Asn371 (N371K). Phenotypic features include infantile hypotonia, congenital ophthalmic abnormalities, childhood-onset hyperkinetic movement disorders, motor stereotypies, and developmental delay varying in severity from moderate to profound. Behavioural characteristics include sleep disturbance and episodic agitation. Absence of epileptic seizures and normal orbitofrontal head circumference are important negative features. Structural MRI is unremarkable but EEG disturbance is universal, characterized by intermittent low frequency high amplitude oscillations. The functional impact of these five de novo SYT1 mutations has been assessed by expressing rat SYT1 protein containing the equivalent human variants in wild-type mouse primary hippocampal cultures. All mutant forms of SYT1 were expressed at levels approximately equal to endogenous wild-type protein, and correctly localized to nerve terminals at rest, except for SYT1M303K, which was expressed at a lower level and failed to localize at nerve terminals. Following stimulation, SYT1I368T and SYT1N371K relocalized to nerve terminals at least as efficiently as wild-type SYT1. However, SYT1D304G and SYT1D366E failed to relocalize to nerve terminals following stimulation, indicative of impairments in endocytic retrieval and trafficking of SYT1. In addition, the presence of SYT1 variants at nerve terminals induced a slowing of exocytic rate following sustained action potential stimulation. The extent of disturbance to synaptic vesicle kinetics is mirrored by the severity of the affected individuals' phenotypes, suggesting that the efficiency of SYT1-mediated neurotransmitter release is critical to cognitive development. In summary, de novo dominant SYT1 missense mutations are associated with a recognizable neurodevelopmental syndrome, and further cases can now be diagnosed based on clinical features, electrophysiological signature and mutation characteristics. Variation in phenotype severity may reflect mutation-specific impact on the diverse physiological functions of SYT1.


Subject(s)
Synaptotagmin I/genetics , Synaptotagmin I/physiology , Action Potentials , Adolescent , Animals , Calcium/metabolism , Child , Child, Preschool , Electrophysiological Phenomena , Endocytosis , Female , Humans , Intellectual Disability/genetics , Male , Mice , Mice, Inbred C57BL , Movement Disorders/genetics , Mutation, Missense/genetics , Neurodevelopmental Disorders/metabolism , Neurons/metabolism , Rats , Synaptic Transmission , Synaptic Vesicles/genetics , Synaptic Vesicles/metabolism , Synaptic Vesicles/physiology , Young Adult
10.
J Neurosci ; 37(17): 4604-4617, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28363983

ABSTRACT

Parvalbumin-expressing inhibitory neurons in the mammalian CNS are specialized for fast transmitter release at their output synapses. However, the Ca2+ sensor(s) used by identified inhibitory synapses, including the output synapses of parvalbumin-expressing inhibitory neurons, have only recently started to be addressed. Here, we investigated the roles of Syt1 and Syt2 at two types of fast-releasing inhibitory connections in the mammalian CNS: the medial nucleus of the trapezoid body to lateral superior olive glycinergic synapse, and the basket/stellate cell-Purkinje GABAergic synapse in the cerebellum. We used conditional and conventional knock-out (KO) mouse lines, with viral expression of Cre-recombinase and a light-activated ion channel for optical stimulation of the transduced fibers, to produce Syt1-Syt2 double KO synapses in vivo Surprisingly, we found that KO of Syt2 alone had only minor effects on evoked transmitter release, despite the clear presence of the protein in inhibitory nerve terminals revealed by immunohistochemistry. We show that Syt1 is weakly coexpressed at these inhibitory synapses and must be genetically inactivated together with Syt2 to achieve a significant reduction and desynchronization of fast release. Thus, our work identifies the functionally relevant Ca2+ sensor(s) at fast-releasing inhibitory synapses and shows that two major Syt isoforms can cooperate to mediate release at a given synaptic connection.SIGNIFICANCE STATEMENT During synaptic transmission, the influx of Ca2+ into the presynaptic nerve terminal activates a Ca2+ sensor for vesicle fusion, a crucial step in the activity-dependent release of neurotransmitter. Synaptotagmin (Syt) proteins, and especially Syt1 and Syt2, have been identified as the Ca2+ sensor at excitatory synapses, but the Ca2+ sensor(s) at inhibitory synapses in native brain tissue are not well known. We found that both Syt1 and Syt2 need to be genetically inactivated to cause a significant reduction of activity-evoked release at two types of fast inhibitory synapses in mouse brain. Thus, we identify Syt2 as a functionally important Ca2+ sensor at fast-releasing inhibitory synapses, and show that Syt1 and Syt2 can redundantly control transmitter release at specific brain synapses.


Subject(s)
Neurons/physiology , Parvalbumins/metabolism , Synaptic Transmission/physiology , Synaptotagmin II/physiology , Synaptotagmin I/physiology , Animals , Cerebellum/metabolism , Glycine/metabolism , Mice , Mice, Knockout , Nerve Fibers/physiology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Photic Stimulation , gamma-Aminobutyric Acid/physiology
11.
J Exp Bot ; 67(21): 6161-6171, 2016 11.
Article in English | MEDLINE | ID: mdl-27811083

ABSTRACT

Arabidopsis synaptotagmin 1 (SYT1) is localized on the endoplasmic reticulum-plasma membrane (ER-PM) contact sites in leaf and root cells. The ER-PM localization of Arabidopsis SYT1 resembles that of the extended synaptotagmins (E-SYTs) in animal cells. In mammals, E-SYTs have been shown to regulate calcium signaling, lipid transfer, and endocytosis. Arabidopsis SYT1 was reported to be essential for maintaining cell integrity and virus movement. This study provides detailed insight into the subcellular localization of SYT1 and VAP27-1, another ER-PM-tethering protein. SYT1 and VAP27-1 were shown to be localized on distinct ER-PM contact sites. The VAP27-1-enriched ER-PM contact sites (V-EPCSs) were always in contact with the SYT1-enriched ER-PM contact sites (S-EPCSs). The V-EPCSs still existed in the leaf epidermal cells of the SYT1 null mutant; however, they were less stable than those in the wild type. The polygonal networks of cortical ER disassembled and the mobility of VAP27-1 protein on the ER-PM contact sites increased in leaf cells of the SYT1 null mutant. These results suggest that SYT1 is responsible for stabilizing the ER network and V-EPCSs.


Subject(s)
Arabidopsis Proteins/physiology , Cell Membrane/physiology , Endoplasmic Reticulum/physiology , Synaptotagmin I/physiology , Arabidopsis/metabolism , Blotting, Western , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , Microscopy, Confocal , Plant Leaves/cytology , Plant Leaves/metabolism , Plant Leaves/physiology , R-SNARE Proteins/physiology
12.
Proc Natl Acad Sci U S A ; 113(32): E4698-707, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27444020

ABSTRACT

Complexin activates Ca(2+)-triggered neurotransmitter release and regulates spontaneous release in the presynaptic terminal by cooperating with the neuronal soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs) and the Ca(2+)-sensor synaptotagmin. The N-terminal domain of complexin is important for activation, but its molecular mechanism is still poorly understood. Here, we observed that a split pair of N-terminal and central domain fragments of complexin is sufficient to activate Ca(2+)-triggered release using a reconstituted single-vesicle fusion assay, suggesting that the N-terminal domain acts as an independent module within the synaptic fusion machinery. The N-terminal domain can also interact independently with membranes, which is enhanced by a cooperative interaction with the neuronal SNARE complex. We show by mutagenesis that membrane binding of the N-terminal domain is essential for activation of Ca(2+)-triggered fusion. Consistent with the membrane-binding property, the N-terminal domain can be substituted by the influenza virus hemagglutinin fusion peptide, and this chimera also activates Ca(2+)-triggered fusion. Membrane binding of the N-terminal domain of complexin therefore cooperates with the other fusogenic elements of the synaptic fusion machinery during Ca(2+)-triggered release.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Calcium/physiology , Membrane Fusion , Adaptor Proteins, Vesicular Transport/chemistry , Humans , Protein Domains , SNARE Proteins/physiology , Synaptic Vesicles/physiology , Synaptotagmin I/physiology
13.
Proc Natl Acad Sci U S A ; 113(29): 8314-9, 2016 07 19.
Article in English | MEDLINE | ID: mdl-27364007

ABSTRACT

Inositol pyrophosphates such as 5-diphosphoinositol pentakisphosphate (5-IP7) are highly energetic inositol metabolites containing phosphoanhydride bonds. Although inositol pyrophosphates are known to regulate various biological events, including growth, survival, and metabolism, the molecular sites of 5-IP7 action in vesicle trafficking have remained largely elusive. We report here that elevated 5-IP7 levels, caused by overexpression of inositol hexakisphosphate (IP6) kinase 1 (IP6K1), suppressed depolarization-induced neurotransmitter release from PC12 cells. Conversely, IP6K1 depletion decreased intracellular 5-IP7 concentrations, leading to increased neurotransmitter release. Consistently, knockdown of IP6K1 in cultured hippocampal neurons augmented action potential-driven synaptic vesicle exocytosis at synapses. Using a FRET-based in vitro vesicle fusion assay, we found that 5-IP7, but not 1-IP7, exhibited significantly higher inhibitory activity toward synaptic vesicle exocytosis than IP6 Synaptotagmin 1 (Syt1), a Ca(2+) sensor essential for synaptic membrane fusion, was identified as a molecular target of 5-IP7 Notably, 5-IP7 showed a 45-fold higher binding affinity for Syt1 compared with IP6 In addition, 5-IP7-dependent inhibition of synaptic vesicle fusion was abolished by increasing Ca(2+) levels. Thus, 5-IP7 appears to act through Syt1 binding to interfere with the fusogenic activity of Ca(2+) These findings reveal a role of 5-IP7 as a potent inhibitor of Syt1 in controlling the synaptic exocytotic pathway and expand our understanding of the signaling mechanisms of inositol pyrophosphates.


Subject(s)
Exocytosis/drug effects , Inositol Phosphates/pharmacology , Synaptotagmin I/physiology , Animals , Hippocampus/cytology , Neurons/physiology , PC12 Cells , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Rats , Rats, Sprague-Dawley
14.
Mol Neurodegener ; 10: 31, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26202512

ABSTRACT

BACKGROUND: Accumulation of the ß-amyloid peptide (Aß) is a major pathological hallmark of Alzheimer's disease (AD). Recent studies have shown that synaptic Aß toxicity may directly impair synaptic function. However, proteins regulating Aß generation at the synapse have not been characterized. Here, we sought to identify synaptic proteins that interact with the extracellular domain of APP and regulate Aß generation. RESULTS: Affinity purification-coupled mass spectrometry identified members of the Synaptotagmin (Syt) family as novel interacting proteins with the APP ectodomain in mouse brains. Syt-1, -2 and -9 interacted with APP in cells and in mouse brains in vivo. Using a GST pull-down approach, we have further demonstrated that the Syt interaction site lies in the 108 amino acids linker region between the E1 and KPI domains of APP. Stable overexpression of Syt-1 or Syt-9 with APP in CHO and rat pheochromocytoma cells (PC12) significantly increased APP-CTF and sAPP levels, with a 2 to 3 fold increase in secreted Aß levels in PC12 cells. Moreover, using a stable knockdown approach to reduce the expression of endogenous Syt-1 in PC12 cells, we have observed a ~ 50% reduction in secreted Aß generation. APP processing also decreased in these cells, shown by lower CTF levels. Lentiviral-mediated knock down of endogenous Syt-1 in mouse primary neurons also led to a significant reduction in both Aß40 and Aß42 generation. As secreted sAPPß levels were significantly reduced in PC12 cells lacking Syt-1 expression, our results suggest that Syt-1 regulates Aß generation by modulating BACE1-mediated cleavage of APP. CONCLUSION: Altogether, our data identify the synaptic vesicle proteins Syt-1 and 9 as novel APP-interacting proteins that promote Aß generation and thus may play an important role in the pathogenesis of AD.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Synaptotagmins/physiology , Amyloid Precursor Protein Secretases/physiology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/chemistry , Animals , Aspartic Acid Endopeptidases/physiology , CHO Cells , Cricetinae , Cricetulus , Mice , Neurons/metabolism , PC12 Cells , Peptide Fragments/biosynthesis , Peptide Fragments/metabolism , Protein Interaction Mapping , Protein Processing, Post-Translational , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/genetics , Rats , Recombinant Fusion Proteins/metabolism , Species Specificity , Synaptotagmin I/deficiency , Synaptotagmin I/genetics , Synaptotagmin I/physiology , Synaptotagmin II/physiology
15.
J Cell Sci ; 128(12): 2229-35, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25964652

ABSTRACT

Synaptotagmin 1 (Syt1) is a synaptic vesicle protein that is important for the kinetics of both exocytosis and endocytosis, and is thus a candidate molecule to link these two processes. Although the tandem Ca(2+)-binding C2 domains of Syt1 have important roles in exocytosis and endocytosis, the function of the conserved juxtamembrane (jxm) linker region has yet to be determined. We now demonstrate that the jxm region of Syt1 interacts directly with the pleckstrin homology (PH) domain of the endocytic protein dynamin 1. By using cell-attached capacitance recordings with millisecond time resolution to monitor clathrin-mediated endocytosis of single vesicles in neuroendocrine chromaffin cells, we find that loss of this interaction prolongs the lifetime of the fission pore leading to defects in the dynamics of vesicle fission. These results indicate a previously undescribed interaction between two major regulatory proteins in the secretory vesicle cycle and that this interaction regulates endocytosis.


Subject(s)
Brain/metabolism , Chromaffin Cells/metabolism , Dynamin I/metabolism , Synaptic Vesicles/physiology , Synaptotagmin I/physiology , Amino Acid Sequence , Animals , Blotting, Western , Brain/cytology , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Chromaffin Cells/cytology , Clathrin/metabolism , Endocytosis/physiology , Exocytosis/physiology , Female , Humans , Immunoprecipitation , Male , Mice , Mice, Knockout , Molecular Sequence Data , Protein Interaction Domains and Motifs , Rats , Sequence Homology, Amino Acid , Synapses/physiology
16.
Sci Rep ; 5: 9267, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25791821

ABSTRACT

Synaptotagmin 1 (Syt1), a major Ca(2+) sensor in neuroexocytosis, utilizes SNARE- and membrane-binding to regulate vesicle fusion, a required process for neurotransmitter release at the synapse. However, the mechanism by which Syt1 orchestrates SNARE- and membrane- binding to control individual vesicle fusion steps is still unclear. In this study, we used a number of single vesicle assays that can differentiate intermediates of neuroexocytosis, to focus on Syt1 mutants that might impair Syt1-SNARE/PIP2 interaction, Ca(2+)-binding, or membrane penetration. Our results show that, although putative Syt1-SNARE/PIP2 coupling through the polybasic region of the C2B domain is critical for vesicle docking, its disruption does not affect content release. In contrast, Ca(2+)-binding and membrane-penetration mutants significantly reduce content release. Our results thus delineate multiple functions of Syt1 along the pathway of Ca(2+)-triggered exocytosis in unprecedented detail.


Subject(s)
Synaptotagmin I/physiology , Calcium/metabolism , Mutation , Protein Binding , SNARE Proteins/metabolism , Synaptotagmin I/genetics , Synaptotagmin I/metabolism
17.
Proc Natl Acad Sci U S A ; 112(12): 3793-8, 2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25775572

ABSTRACT

Synaptotagmin 1 (Syt1) is a synaptic vesicle integral membrane protein that regulates neurotransmitter release by activating fast synchronous fusion and suppressing slower asynchronous release. The cytoplasmic C2 domains of Syt1 interact with SNAREs and plasma membrane phospholipids in a Ca(2+)-dependent manner and can substitute for full-length Syt1 in in vitro membrane fusion assays. To determine whether synaptic vesicle tethering of Syt1 is required for normal fusion in vivo, we performed a structure-function study with tethering mutants at the Drosophila larval neuromuscular junction. Transgenic animals expressing only the cytoplasmic C2 domains or full-length Syt1 tethered to the plasma membrane failed to restore synchronous synaptic vesicle fusion, and also failed to clamp spontaneous vesicle release. In addition, transgenic animals with shorter, but not those with longer, linker regions separating the C2 domains from the transmembrane segment abolished Syt1's ability to activate synchronous vesicle fusion. Similar defects were observed when C2 domain alignment was altered to C2B-C2A from the normal C2A-C2B orientation, leaving the tether itself intact. Although cytoplasmic and plasma membrane-tethered Syt1 variants could not restore synchronous release in syt1 null mutants, they were very effective in promoting fusion through the slower asynchronous pathway. As such, the subcellular localization of Syt1 within synaptic terminals is important for the temporal dynamics that underlie synchronous and asynchronous neurotransmitter release.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster , Neurotransmitter Agents/metabolism , Synaptic Vesicles/pathology , Synaptotagmin I/physiology , Animals , Animals, Genetically Modified , Astacoidea , Calcium/metabolism , Cytoplasm/metabolism , Electrophysiological Phenomena , Exocytosis , Immunohistochemistry , Male , Membrane Fusion , Mutation , Neuromuscular Junction/metabolism , Protein Structure, Tertiary , Transgenes
18.
J Clin Invest ; 125(4): 1670-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25705886

ABSTRACT

Synaptotagmin-1 (SYT1) is a calcium-binding synaptic vesicle protein that is required for both exocytosis and endocytosis. Here, we describe a human condition associated with a rare variant in SYT1. The individual harboring this variant presented with an early onset dyskinetic movement disorder, severe motor delay, and profound cognitive impairment. Structural MRI was normal, but EEG showed extensive neurophysiological disturbances that included the unusual features of low-frequency oscillatory bursts and enhanced paired-pulse depression of visual evoked potentials. Trio analysis of whole-exome sequence identified a de novo SYT1 missense variant (I368T). Expression of rat SYT1 containing the equivalent human variant in WT mouse primary hippocampal cultures revealed that the mutant form of SYT1 correctly localizes to nerve terminals and is expressed at levels that are approximately equal to levels of endogenous WT protein. The presence of the mutant SYT1 slowed synaptic vesicle fusion kinetics, a finding that agrees with the previously demonstrated role for I368 in calcium-dependent membrane penetration. Expression of the I368T variant also altered the kinetics of synaptic vesicle endocytosis. Together, the clinical features, electrophysiological phenotype, and in vitro neuronal phenotype associated with this dominant negative SYT1 mutation highlight presynaptic mechanisms that mediate human motor control and cognitive development.


Subject(s)
Intellectual Disability/genetics , Motor Skills Disorders/genetics , Movement Disorders/genetics , Mutation, Missense , Point Mutation , Presynaptic Terminals/physiology , Synaptic Vesicles/physiology , Synaptotagmin I/physiology , Amino Acid Sequence , Animals , Calcium/metabolism , Cells, Cultured , Child , Endocytosis/genetics , Endocytosis/physiology , Evoked Potentials, Visual , Exocytosis/genetics , Exocytosis/physiology , Genes, Dominant , Hippocampus/cytology , Humans , Kinetics , Male , Membrane Fusion , Mice , Molecular Sequence Data , Rats , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Synaptotagmin I/genetics
19.
Neuron ; 80(4): 947-59, 2013 Nov 20.
Article in English | MEDLINE | ID: mdl-24267651

ABSTRACT

In forebrain neurons, knockout of synaptotagmin-1 blocks fast Ca(2+)-triggered synchronous neurotransmitter release but enables manifestation of slow Ca(2+)-triggered asynchronous release. Here, we show using single-cell PCR that individual hippocampal neurons abundantly coexpress two Ca(2+)-binding synaptotagmin isoforms, synaptotagmin-1 and synaptotagmin-7. In synaptotagmin-1-deficient synapses of excitatory and inhibitory neurons, loss of function of synaptotagmin-7 suppressed asynchronous release. This phenotype was rescued by wild-type but not mutant synaptotagmin-7 lacking functional Ca(2+)-binding sites. Even in synaptotagmin-1-containing neurons, synaptotagmin-7 ablation partly impaired asynchronous release induced by extended high-frequency stimulus trains. Synaptotagmins bind Ca(2+) via two C2 domains, the C2A and C2B domains. Surprisingly, synaptotagmin-7 function selectively required its C2A domain Ca(2+)-binding sites, whereas synaptotagmin-1 function required its C2B domain Ca(2+)-binding sites. Our data show that nearly all Ca(2+)-triggered release at a synapse is due to synaptotagmins, with synaptotagmin-7 mediating a slower form of Ca(2+)-triggered release that is normally occluded by faster synaptotagmin-1-induced release but becomes manifest upon synaptotagmin-1 deletion.


Subject(s)
Neurotransmitter Agents/metabolism , Synaptotagmin I/physiology , Synaptotagmins/physiology , Animals , Calcium/physiology , Cells, Cultured , Dependovirus/genetics , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/physiology , Lentivirus/genetics , Mice , Mice, Knockout , Neurons/metabolism , Patch-Clamp Techniques , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Calcium-Sensing/physiology , Synapses/metabolism , Synaptotagmin I/genetics , Synaptotagmins/genetics
20.
Insect Mol Biol ; 22(5): 457-72, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23782271

ABSTRACT

Gap junctions (GJs) provide a common form of intercellular communication in most animal cells and tissues, from Hydra to human, including electrical synaptic signalling. Cell coupling via GJs has an important role in development in general, and in neural network development in particular. However, quantitative studies monitoring GJ proteins throughout nervous system development are few. Direct investigations demonstrating a role for GJ proteins by way of experimental manipulation of their expression are also rare. In the current work we focused on the role of invertebrate GJ proteins (innexins) in the in vitro development of neural network functional topology, using two-dimensional neural culture preparations derived from the frontal ganglion of the desert locust, Schistocerca gregaria. Immunocytochemistry and quantitative real-time PCR revealed a dynamic expression pattern of the innexins during development of the cultured networks. Changes were observed both in the levels and in the localization of expression. Down-regulating the expression of innexins, by using double-strand RNA for the first time in locust neural cultures, induced clear changes in network morphology, as well as inhibition of synaptogenesis, thus suggesting a role for GJs during the development of the functional topology of neuronal networks.


Subject(s)
Connexins/physiology , Insect Proteins/physiology , Nerve Net/growth & development , Animals , Connexins/genetics , Gene Knockdown Techniques , Grasshoppers , Insect Proteins/genetics , Nerve Net/physiology , RNA, Double-Stranded/genetics , Synaptotagmin I/physiology , Tissue Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...