Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Neurosci ; 37(2): 383-396, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28077717

ABSTRACT

Synaptic vesicles fuse at morphological specializations in the presynaptic terminal termed active zones (AZs). Vesicle fusion can occur spontaneously or in response to an action potential. Following fusion, vesicles are retrieved and recycled within nerve terminals. It is still unclear whether vesicles that fuse spontaneously or following evoked release share similar recycling mechanisms. Genetic deletion of the SNARE-binding protein complexin dramatically increases spontaneous fusion, with the protein serving as the synaptic vesicle fusion clamp at Drosophila synapses. We examined synaptic vesicle recycling pathways at complexin null neuromuscular junctions, where spontaneous release is dramatically enhanced. We combined loading of the lipophilic dye FM1-43 with photoconversion, electron microscopy, and electrophysiology to monitor evoked and spontaneous recycling vesicle pools. We found that the total number of recycling vesicles was equal to those retrieved through spontaneous and evoked pools, suggesting that retrieval following fusion is partially segregated for spontaneous and evoked release. In addition, the kinetics of FM1-43 destaining and synaptic depression measured in the presence of the vesicle-refilling blocker bafilomycin indicated that spontaneous and evoked recycling pools partially intermix during the release process. Finally, FM1-43 photoconversion combined with electron microscopy analysis indicated that spontaneous recycling preferentially involves synaptic vesicles in the vicinity of AZs, whereas vesicles recycled following evoked release involve a larger intraterminal pool. Together, these results suggest that spontaneous and evoked vesicles use separable recycling pathways and then partially intermix during subsequent rounds of fusion. SIGNIFICANCE STATEMENT: Neurotransmitter release involves fusion of synaptic vesicles with the plasma membrane in response to an action potential, or spontaneously in the absence of stimulation. Upon fusion, vesicles are retrieved and recycled, and it is unclear whether recycling pathways for evoked and spontaneous vesicles are segregated after fusion. We addressed this question by taking advantage of preparations lacking the synaptic protein complexin, which have elevated spontaneous release that enables reliable tracking of the spontaneous recycling pool. Our results suggest that spontaneous and evoked recycling pathways are segregated during the retrieval process but can partially intermix during stimulation.


Subject(s)
Adaptor Proteins, Vesicular Transport/deficiency , Drosophila Proteins/deficiency , Exocytosis/physiology , Mutation/physiology , Nerve Tissue Proteins/deficiency , Signal Transduction/physiology , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Male , Nerve Tissue Proteins/genetics , Synaptic Vesicles/genetics , Synaptic Vesicles/ultrastructure
2.
Hum Mol Genet ; 25(15): 3164-3177, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288455

ABSTRACT

Huntington disease-like 2 (HDL2) and Huntington disease (HD) are adult-onset neurodegenerative diseases characterized by movement disorders, psychiatric disturbances and cognitive decline. Brain tissue from HD and HDL2 patients shows degeneration of the striatum and ubiquitinated inclusions immunoreactive for polyglutamine (polyQ) antibodies. Despite these similarities, the diseases result from different genetic mutations. HD is caused by a CAG repeat expansion in the huntingtin (HTT) gene, while HDL2 results from an expansion at the junctophilin 3 (JPH3) locus. Recent evidence indicates that the HDL2 expansion may give rise to a toxic polyQ protein translated from an antisense mRNA derived from the JPH3 locus. To investigate this hypothesis, we generated and characterized a Drosophila HDL2 model and compared it with a previously established HD model. We find that neuronal expression of HDL2-Q15 is not toxic, while the expression of an expanded HDL2-Q138 protein is lethal. HDL2-Q138 forms large nuclear aggregates, with only smaller puncta observed in the cytoplasm. This is in contrast to what is observed in a Drosophila model of HD, where polyQ aggregates localize exclusively to the cytoplasm. Altering localization of HLD2 with the addition of a nuclear localization or nuclear export sequence demonstrates that nuclear accumulation is required for toxicity in the Drosophila HDL2 model. Directing HDL2-Q138 to the nucleus exacerbates toxicity in multiple tissue types, while confining HDL2-Q138 to the cytoplasm restores viability to control levels. We conclude that while HD and HDL2 have similar clinical profiles, distinct pathogenic mechanisms are likely to drive toxicity in Drosophila models of these disorders.


Subject(s)
Cell Nucleus , Disease Models, Animal , Drosophila Proteins , Huntington Disease , Membrane Proteins , Trinucleotide Repeat Expansion , Adult , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Peptides/genetics , Peptides/metabolism
3.
J Neurosci ; 36(21): 5820-32, 2016 05 25.
Article in English | MEDLINE | ID: mdl-27225771

ABSTRACT

UNLABELLED: Prosap/Shank scaffolding proteins regulate the formation, organization, and plasticity of excitatory synapses. Mutations in SHANK family genes are implicated in autism spectrum disorder and other neuropsychiatric conditions. However, the molecular mechanisms underlying Shank function are not fully understood, and no study to date has examined the consequences of complete loss of all Shank proteins in vivo Here we characterize the single Drosophila Prosap/Shank family homolog. Shank is enriched at the postsynaptic membrane of glutamatergic neuromuscular junctions and controls multiple parameters of synapse biology in a dose-dependent manner. Both loss and overexpression of Shank result in defects in synaptic bouton number and maturation. We find that Shank regulates a noncanonical Wnt signaling pathway in the postsynaptic cell by modulating the internalization of the Wnt receptor Fz2. This study identifies Shank as a key component of synaptic Wnt signaling, defining a novel mechanism for how Shank contributes to synapse maturation during neuronal development. SIGNIFICANCE STATEMENT: Haploinsufficiency for SHANK3 is one of the most prevalent monogenic causes of autism spectrum disorder, making it imperative to understand how the Shank family regulates neurodevelopment and synapse function. We created the first animal model lacking all Shank proteins and used the Drosophila neuromuscular junction, a model glutamatergic synapse, to characterize the role of Shank at synapses. We identified a novel function of Shank in synapse maturation via regulation of Wnt signaling in the postsynaptic cell.


Subject(s)
Axon Guidance/physiology , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Presynaptic Terminals/physiology , Wnt Signaling Pathway/physiology , Animals , Animals, Genetically Modified , Drosophila/genetics , Humans , Nerve Tissue Proteins/genetics , Neuromuscular Junction/physiology
4.
Neuron ; 88(4): 749-61, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26590346

ABSTRACT

Synaptic plasticity is a fundamental feature of the nervous system that allows adaptation to changing behavioral environments. Most studies of synaptic plasticity have examined the regulated trafficking of postsynaptic glutamate receptors that generates alterations in synaptic transmission. Whether and how changes in the presynaptic release machinery contribute to neuronal plasticity is less clear. The SNARE complex mediates neurotransmitter release in response to presynaptic Ca(2+) entry. Here we show that the SNARE fusion clamp Complexin undergoes activity-dependent phosphorylation that alters the basic properties of neurotransmission in Drosophila. Retrograde signaling following stimulation activates PKA-dependent phosphorylation of the Complexin C terminus that selectively and transiently enhances spontaneous release. Enhanced spontaneous release is required for activity-dependent synaptic growth. These data indicate that SNARE-dependent fusion mechanisms can be regulated in an activity-dependent manner and highlight the key role of spontaneous neurotransmitter release as a mediator of functional and structural plasticity.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Drosophila Proteins/genetics , Nerve Tissue Proteins/genetics , Neuromuscular Junction/metabolism , Neuronal Plasticity/genetics , Synaptic Transmission/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Base Sequence , Calcium/metabolism , Drosophila , Drosophila Proteins/metabolism , Exocytosis/genetics , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Phosphorylation , SNARE Proteins/metabolism
5.
Mol Cell Neurosci ; 61: 241-54, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25066865

ABSTRACT

Synaptic communication requires precise alignment of presynaptic active zones with postsynaptic receptors to enable rapid and efficient neurotransmitter release. How transsynaptic signaling between connected partners organizes this synaptic apparatus is poorly understood. To further define the mechanisms that mediate synapse assembly, we carried out a chemical mutagenesis screen in Drosophila to identify mutants defective in the alignment of active zones with postsynaptic glutamate receptor fields at the larval neuromuscular junction. From this screen we identified a mutation in Actin 57B that disrupted synaptic morphology and presynaptic active zone organization. Actin 57B, one of six actin genes in Drosophila, is expressed within the postsynaptic bodywall musculature. The isolated allele, act(E84K), harbors a point mutation in a highly conserved glutamate residue in subdomain 1 that binds members of the Calponin Homology protein family, including spectrin. Homozygous act(E84K) mutants show impaired alignment and spacing of presynaptic active zones, as well as defects in apposition of active zones to postsynaptic glutamate receptor fields. act(E84K) mutants have disrupted postsynaptic actin networks surrounding presynaptic boutons, with the formation of aberrant actin swirls previously observed following disruption of postsynaptic spectrin. Consistent with a disruption of the postsynaptic actin cytoskeleton, spectrin, adducin and the PSD-95 homolog Discs-Large are all mislocalized in act(E84K) mutants. Genetic interactions between act(E84K) and neurexin mutants suggest that the postsynaptic actin cytoskeleton may function together with the Neurexin-Neuroligin transsynaptic signaling complex to mediate normal synapse development and presynaptic active zone organization.


Subject(s)
Actins/metabolism , Drosophila Proteins/metabolism , Neuromuscular Junction/genetics , Neuromuscular Junction/metabolism , Receptors, Glutamate/metabolism , Actins/genetics , Age Factors , Animals , Animals, Genetically Modified , Calcium-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Gene Expression Regulation/genetics , Glutamic Acid/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Horseradish Peroxidase/metabolism , Larva , Microfilament Proteins/metabolism , Microscopy, Electron, Transmission , Models, Molecular , Mutation/drug effects , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/ultrastructure , Patch-Clamp Techniques , Receptors, Glutamate/genetics , Synaptic Potentials/genetics , Calponins
6.
Proc Natl Acad Sci U S A ; 111(28): 10317-22, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982161

ABSTRACT

Complexin (Cpx) is a SNARE-binding protein that regulates neurotransmission by clamping spontaneous synaptic vesicle fusion in the absence of Ca(2+) influx while promoting evoked release in response to an action potential. Previous studies indicated Cpx may cross-link multiple SNARE complexes via a trans interaction to function as a fusion clamp. During Ca(2+) influx, Cpx is predicted to undergo a conformational switch and collapse onto a single SNARE complex in a cis-binding mode to activate vesicle release. To test this model in vivo, we performed structure-function studies of the Cpx protein in Drosophila. Using genetic rescue approaches with cpx mutants that disrupt SNARE cross-linking, we find that manipulations that are predicted to block formation of the trans SNARE array disrupt the clamping function of Cpx. Unexpectedly, these same mutants rescue action potential-triggered release, indicating trans-SNARE cross-linking by Cpx is not a prerequisite for triggering evoked fusion. In contrast, mutations that impair Cpx-mediated cis-SNARE interactions that are necessary for transition from an open to closed conformation fail to rescue evoked release defects in cpx mutants, although they clamp spontaneous release normally. Our in vivo genetic manipulations support several predictions made by the Cpx cross-linking model, but unexpected results suggest additional mechanisms are likely to exist that regulate Cpx's effects on SNARE-mediated fusion. Our findings also indicate that the inhibitory and activating functions of Cpx are genetically separable, and can be mapped to distinct molecular mechanisms that differentially regulate the SNARE fusion machinery.


Subject(s)
Calcium/metabolism , Drosophila Proteins/metabolism , Mutation , Nerve Tissue Proteins/metabolism , SNARE Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Nerve Tissue Proteins/genetics , SNARE Proteins/genetics
7.
J Neurosci ; 32(50): 18234-45, 2012 Dec 12.
Article in English | MEDLINE | ID: mdl-23238737

ABSTRACT

Neurotransmitter release following synaptic vesicle (SV) fusion is the fundamental mechanism for neuronal communication. Synaptic exocytosis is a specialized form of intercellular communication that shares a common SNARE-mediated fusion mechanism with other membrane trafficking pathways. The regulation of synaptic vesicle fusion kinetics and short-term plasticity is critical for rapid encoding and transmission of signals across synapses. Several families of SNARE-binding proteins have evolved to regulate synaptic exocytosis, including Synaptotagmin (SYT) and Complexin (CPX). Here, we demonstrate that Drosophila CPX controls evoked fusion occurring via the synchronous and asynchronous pathways. cpx(-/-) mutants show increased asynchronous release, while CPX overexpression largely eliminates the asynchronous component of fusion. We also find that SYT and CPX coregulate the kinetics and Ca(2+) co-operativity of neurotransmitter release. CPX functions as a positive regulator of release in part by coupling the Ca(2+) sensor SYT to the fusion machinery and synchronizing its activity to speed fusion. In contrast, syt(-/-); cpx(-/-) double mutants completely abolish the enhanced spontaneous release observe in cpx(-/-) mutants alone, indicating CPX acts as a fusion clamp to block premature exocytosis in part by preventing inappropriate activation of the SNARE machinery by SYT. CPX levels also control the size of synaptic vesicle pools, including the immediate releasable pool and the ready releasable pool-key elements of short-term plasticity that define the ability of synapses to sustain responses during burst firing. These observations indicate CPX regulates both spontaneous and evoked fusion by modulating the timing and properties of SYT activation during the synaptic vesicle cycle.


Subject(s)
Neurotransmitter Agents/metabolism , SNARE Proteins/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Synaptotagmins/metabolism , Animals , Blotting, Western , Drosophila , Drosophila Proteins/metabolism , Excitatory Postsynaptic Potentials/physiology , Exocytosis/physiology , Gene Knockout Techniques , Immunohistochemistry , Microscopy, Electron, Transmission , Neuromuscular Junction/metabolism , Patch-Clamp Techniques
8.
Mol Cell Neurosci ; 45(4): 389-97, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20678575

ABSTRACT

The SNARE-binding protein complexin (Cpx) has been demonstrated to regulate synaptic vesicle fusion. Previous studies are consistent with Cpx functioning either as a synaptic vesicle fusion clamp to prevent premature exocytosis, or as a facilitator to directly stimulate release. Here we examined conserved roles of invertebrate and mammalian Cpx isoforms in the regulation of neurotransmitter release using the Drosophila neuromuscular junction as a model synapse. We find that SNARE binding by Cpx is required for its role as a fusion clamp. All four mammalian Cpx proteins (mCpx), which have been demonstrated to facilitate release, also function as fusion clamps when expressed in Drosophila cpx null mutants, though their clamping abilities vary between isoforms. Moreover, expression of mCpx I, II or III isoforms dramatically enhance evoked release compared to mCpx IV or Drosophila Cpx. Differences in the clamping and facilitating properties of complexin isoforms can be partially attributed to differences in the C-terminal membrane tethering domain. Our findings indicate that the function of complexins as fusion clamps and facilitators of fusion are conserved across evolution, and that these roles are genetically separable within an isoform and across different isoforms.


Subject(s)
Exocytosis/physiology , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Synaptic Vesicles/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Drosophila , Immunohistochemistry , Mammals , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neuromuscular Junction , Phylogeny , Protein Isoforms/metabolism , SNARE Proteins/metabolism
9.
Neuron ; 57(6): 858-71, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18367087

ABSTRACT

Matrix metalloproteases (MMPs) play a role in remodeling the extracellular matrix during brain development and have been implicated in synaptic plasticity. Here, we report that a member of the neuronal pentraxin (NP) family, neuronal pentraxin receptor (NPR), undergoes regulated cleavage by the MMP tumor necrosis factor-alpha converting enzyme (TACE). NPR is enriched at excitatory synapses where it associates with AMPA-type glutamate receptors (AMPAR) and enhances synaptogenesis. However, in response to activation of group 1 mGluRs (mGluR1/5), TACE cleaves NPR and releases the pentraxin domain from its N-terminal transmembrane domain. Cleaved NPR rapidly accumulates in endosomes where it colocalizes with AMPAR. This process is necessary for mGluR1/5-dependent LTD in hippocampal and cerebellar synapses. These observations suggest that cleaved NPR functions to "capture" AMPAR for endocytosis and reveal a bifunctional role of NPs in both synapse strengthening and weakening.


Subject(s)
ADAM Proteins/pharmacology , Inhibitory Postsynaptic Potentials/physiology , Neurons/drug effects , Receptors, Cell Surface/metabolism , Receptors, Metabotropic Glutamate/physiology , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Animals, Newborn , C-Reactive Protein/deficiency , Cells, Cultured , Cerebellum/cytology , Embryo, Mammalian , Excitatory Amino Acid Agents/pharmacology , Hippocampus/cytology , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/radiation effects , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neuronal Plasticity/radiation effects , Neurons/physiology , Patch-Clamp Techniques/methods , Protein Structure, Tertiary/physiology , RNA, Small Interfering/pharmacology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/deficiency , Transfection
10.
Neuron ; 39(3): 513-28, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12895424

ABSTRACT

Narp is a neuronal immediate early gene that plays a role in excitatory synaptogenesis. Here, we report that native Narp in brain is part of a pentraxin complex that includes NP1. These proteins are covalently linked by disulfide bonds into highly organized complexes, and their relative ratio in the complex is dynamically dependent upon the neuron's activity history and developmental stage. Complex formation is dependent on their distinct N-terminal coiled-coil domains, while their closely homologous C-terminal pentraxin domains mediate association with AMPA-type glutamate receptors. Narp is substantially more effective in assays of cell surface cluster formation, coclustering of AMPA receptors, and excitatory synaptogenesis, yet their combined expression results in supraadditive effects. These studies support a model in which Narp can regulate the latent synaptogenic activity of NP1 by forming mixed pentraxin assemblies. This mechanism appears to contribute to both activity-independent and activity-dependent excitatory synaptogenesis.


Subject(s)
C-Reactive Protein/chemistry , C-Reactive Protein/physiology , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/physiology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/physiology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Synapses/metabolism , Amino Acid Sequence , Animals , C-Reactive Protein/biosynthesis , C-Reactive Protein/ultrastructure , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/ultrastructure , Escherichia coli Proteins/ultrastructure , Humans , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/ultrastructure , Rats , Rats, Sprague-Dawley , Sequence Homology, Amino Acid , Synapses/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...