Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Front Cell Neurosci ; 17: 1182493, 2023.
Article En | MEDLINE | ID: mdl-38045729

Synapse formation is critical for the wiring of neural circuits in the developing brain. The synaptic scaffolding protein S-SCAM/MAGI-2 has important roles in the assembly of signaling complexes at post-synaptic densities. However, the role of S-SCAM in establishing the entire synapse is not known. Here, we report significant effects of RNAi-induced S-SCAM knockdown on the number of synapses in early stages of network development in vitro. In vivo knockdown during the first three postnatal weeks reduced the number of dendritic spines in the rat brain neocortex. Knockdown of S-SCAM in cultured hippocampal neurons severely reduced the clustering of both pre- and post-synaptic components. This included synaptic vesicle proteins, pre- and post-synaptic scaffolding proteins, and cell adhesion molecules, suggesting that entire synapses fail to form. Correspondingly, functional and morphological characteristics of developing neurons were affected by reducing S-SCAM protein levels; neurons displayed severely impaired synaptic transmission and reduced dendritic arborization. A next-generation sequencing approach showed normal expression of housekeeping genes but changes in expression levels in 39 synaptic signaling molecules in cultured neurons. These results indicate that S-SCAM mediates the recruitment of all key classes of synaptic molecules during synapse assembly and is critical for the development of neural circuits in the developing brain.

2.
Methods Mol Biol ; 1148: 77-87, 2014.
Article En | MEDLINE | ID: mdl-24718796

Being able to use versatile light microscopy on live or fixed samples followed by electron microscopy imaging for high resolution analyses is a challenging goal. The advantage is of course that tracing and localizing fluorescently labeled molecules yields great information about dynamic cellular processes, while electron microscopy of the same sample provides exquisite information about subcellular structures. Here, I describe the straightforward combination of both methods by photoconversion of diaminobenzidine (DAB) through cyan fluorescent protein (CFP) tagged proteins localized to the Golgi apparatus in primary hippocampal neurons.


Green Fluorescent Proteins/chemistry , Neurons/ultrastructure , 3,3'-Diaminobenzidine/chemistry , Animals , Cells, Cultured , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Green Fluorescent Proteins/biosynthesis , Hippocampus/cytology , Mice , Microscopy, Electron, Transmission , Neurons/metabolism , Photochemical Processes , Primary Cell Culture , Protein Transport , Rats, Wistar , Staining and Labeling
3.
PLoS One ; 8(5): e63474, 2013.
Article En | MEDLINE | ID: mdl-23723986

With remarkably few exceptions, the molecules mediating synaptic vesicle exocytosis at active zones are structurally and functionally conserved between vertebrates and invertebrates. Mover was found in a yeast-2-hybrid assay using the vertebrate-specific active zone scaffolding protein bassoon as a bait. Peptides of Mover have been reported in proteomics screens for self-interacting proteins, phosphorylated proteins, and synaptic vesicle proteins, respectively. Here, we tested the predictions arising from these screens. Using flotation assays, carbonate stripping of peripheral membrane proteins, mass spectrometry, immunogold labelling of purified synaptic vesicles, and immuno-organelle isolation, we found that Mover is indeed a peripheral synaptic vesicle membrane protein. In addition, by generating an antibody against phosphorylated Mover and Western blot analysis of fractionated rat brain, we found that Mover is a bona fide phospho-protein. The localization of Mover to synaptic vesicles is phosphorylation dependent; treatment with a phosphatase caused Mover to dissociate from synaptic vesicles. A yeast-2-hybrid screen, co-immunoprecipitation and cell-based optical assays of homomerization revealed that Mover undergoes homophilic interaction, and regions within both the N- and C- terminus of the protein are required for this interaction. Deleting a region required for homomeric interaction abolished presynaptic targeting of recombinant Mover in cultured neurons. Together, these data prove that Mover is associated with synaptic vesicles, and implicate phosphorylation and multimerization in targeting of Mover to synaptic vesicles and presynaptic sites.


Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Synaptic Vesicles/metabolism , Animals , Chlorocebus aethiops , Membrane Potentials , Membrane Proteins/metabolism , Mice , Mutant Proteins/metabolism , Phosphorylation , Protein Binding , Protein Transport , Rats , Rats, Sprague-Dawley , Sequence Deletion , Subcellular Fractions/metabolism , Synaptic Vesicles/ultrastructure , Vero Cells
4.
PLoS One ; 7(3): e34167, 2012.
Article En | MEDLINE | ID: mdl-22470532

Profilins are prominent regulators of actin dynamics. While most mammalian cells express only one profilin, two isoforms, PFN1 and PFN2a are present in the CNS. To challenge the hypothesis that the expression of two profilin isoforms is linked to the complex shape of neurons and to the activity-dependent structural plasticity, we analysed how PFN1 and PFN2a respond to changes of neuronal activity. Simultaneous labelling of rodent embryonic neurons with isoform-specific monoclonal antibodies revealed both isoforms in the same synapse. Immunoelectron microscopy on brain sections demonstrated both profilins in synapses of the mature rodent cortex, hippocampus and cerebellum. Both isoforms were significantly more abundant in postsynaptic than in presynaptic structures. Immunofluorescence showed PFN2a associated with gephyrin clusters of the postsynaptic active zone in inhibitory synapses of embryonic neurons. When cultures were stimulated in order to change their activity level, active synapses that were identified by the uptake of synaptotagmin antibodies, displayed significantly higher amounts of both isoforms than non-stimulated controls. Specific inhibition of NMDA receptors by the antagonist APV in cultured rat hippocampal neurons resulted in a decrease of PFN2a but left PFN1 unaffected. Stimulation by the brain derived neurotrophic factor (BDNF), on the other hand, led to a significant increase in both synaptic PFN1 and PFN2a. Analogous results were obtained for neuronal nuclei: both isoforms were localized in the same nucleus, and their levels rose significantly in response to KCl stimulation, whereas BDNF caused here a higher increase in PFN1 than in PFN2a. Our results strongly support the notion of an isoform specific role for profilins as regulators of actin dynamics in different signalling pathways, in excitatory as well as in inhibitory synapses. Furthermore, they suggest a functional role for both profilins in neuronal nuclei.


Neurons/metabolism , Profilins/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/immunology , Brain-Derived Neurotrophic Factor/pharmacology , Carrier Proteins/metabolism , Cell Line , Cell Nucleus/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Mice , Profilins/analysis , Profilins/genetics , Protein Isoforms , Rats , Synapses/drug effects , Synapses/metabolism
5.
Opt Express ; 19(9): 8066-72, 2011 Apr 25.
Article En | MEDLINE | ID: mdl-21643055

We show that far-field fluorescence nanoscopy by stimulated emission depletion (STED) can be realized with compact off-the-shelf laser diodes, such as those used in laser pointers and DVDs. A spatial resolution of 40-50 nm is attained by pulsing a 660 nm DVD-diode. The efficacy of these low-cost STED microscopes in biological imaging is demonstrated by differentiating between clusters of the synaptic protein bassoon and transport vesicles in hippocampal neurons, based on the feature diameter. Our results facilitate the implementation of this all-molecular-transition based superresolution method in many applications ranging from nanoscale fluorescence imaging to nanoscale fluorescence sensing.


Image Enhancement/instrumentation , Lasers, Semiconductor , Microscopy, Fluorescence/instrumentation , Nanotechnology/instrumentation , Equipment Design , Equipment Failure Analysis
6.
Proc Natl Acad Sci U S A ; 106(32): 13564-9, 2009 Aug 11.
Article En | MEDLINE | ID: mdl-19628693

Presynaptic nerve terminals pass through distinct stages of maturation after their initial assembly. Here we show that the postsynaptic cell adhesion molecule Neuroligin1 regulates key steps of presynaptic maturation. Presynaptic terminals from Neuroligin1-knockout mice remain structurally and functionally immature with respect to active zone stability and synaptic vesicle pool size, as analyzed in cultured hippocampal neurons. Conversely, overexpression of Neuroligin1 in immature neurons, that is within the first 5 days after plating, induced the formation of presynaptic boutons that had hallmarks of mature boutons. In particular, Neuroligin1 enhanced the size of the pool of recycling synaptic vesicles, the rate of synaptic vesicle exocytosis, the fraction of boutons responding to depolarization, as well as the responsiveness of the presynaptic release machinery to phorbol ester stimulation. Moreover, Neuroligin1 induced the formation of active zones that remained stable in the absence of F-actin, another hallmark of advanced maturation. Acquisition of F-actin independence of the active zone marker Bassoon during culture development or induced via overexpression of Neuroligin1 was activity-dependent. The extracellular domain of Neuroligin1 was sufficient to induce assembly of functional presynaptic terminals, while the intracellular domain was required for terminal maturation. These data show that induction of presynaptic terminal assembly and maturation involve mechanistically distinct actions of Neuroligins, and that Neuroligin1 is essential for presynaptic terminal maturation.


Neural Cell Adhesion Molecules/metabolism , Presynaptic Terminals/metabolism , Synaptic Potentials/physiology , Actins/metabolism , Animals , Cell Adhesion Molecules, Neuronal , Green Fluorescent Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/chemistry , Protein Structure, Tertiary , Rats , Rats, Wistar , Recombinant Fusion Proteins/metabolism
7.
J Neurosci ; 29(2): 351-8, 2009 Jan 14.
Article En | MEDLINE | ID: mdl-19144835

Presynaptic sites typically appear as varicosities (boutons) distributed along axons. Ultrastructurally, presynaptic boutons lack obvious physical barriers that separate them from the axon proper, yet activity-related and constitutive dynamics continuously promote the "reshuffling" of presynaptic components and even their dispersal into flanking axonal segments. How presynaptic sites manage to maintain their organization and individual characteristics over long durations is thus unclear. Conceivably, presynaptic tenacity might depend on the active zone (AZ), an electron-dense specialization of the presynaptic membrane, and particularly on the cytoskeletal matrix associated with the AZ (CAZ) that could act as a relatively stable "core scaffold" that conserves and dictates presynaptic organization. At present, however, little is known on the molecular dynamics of CAZ molecules, and thus, the factual basis for this hypothesis remains unclear. To examine the stability of the CAZ, we studied the molecular dynamics of the major CAZ molecule Bassoon in cultured hippocampal neurons. Fluorescence recovery after photobleaching and photoactivation experiments revealed that exchange rates of green fluorescent protein and photoactivatable green fluorescent protein-tagged Bassoon at individual presynaptic sites are very low (tau > 8 h). Exchange rates varied between boutons and were only slightly accelerated by stimulation. Interestingly, photoactivation experiments revealed that Bassoon lost from one synapse was occasionally assimilated into neighboring presynaptic sites. Our findings indicate that Bassoon is engaged in relatively stable associations within the CAZ and thus support the notion that the CAZ or some of its components might constitute a relatively stable presynaptic core scaffold.


Cytoskeleton/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Nonlinear Dynamics , Presynaptic Terminals/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Cells, Cultured , Electric Stimulation/methods , Excitatory Amino Acid Antagonists/pharmacology , Fluorescent Dyes/pharmacology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Nerve Tissue Proteins/genetics , Neurons/drug effects , Photic Stimulation/methods , Presynaptic Terminals/drug effects , Pyridinium Compounds/pharmacology , Quaternary Ammonium Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Transfection/methods
8.
FEBS Lett ; 581(24): 4727-33, 2007 Oct 02.
Article En | MEDLINE | ID: mdl-17869247

Presynaptic nerve terminals contain scaffolding proteins that orchestrate neurotransmitter release at active zones. Here we describe mover, a yet unknown non-transmembrane protein that is targeted to presynaptic terminals when overexpressed in cultured neurons. Confocal immunomicroscopy revealed that mover colocalizes with presynaptic markers in the calyx of Held. In the hippocampus, mover localizes to mossy fibre terminals, but is absent from inhibitory nerve terminals. By contrast, mover localizes to inhibitory terminals throughout the cerebellar cortex. Our results suggest that mover may act in concert with generally expressed scaffolding proteins in distinct sets of presynaptic terminals.


Central Nervous System/metabolism , Nerve Tissue Proteins/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Conserved Sequence , Humans , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Organ Specificity , Rats , Sequence Alignment , Tissue Culture Techniques
9.
J Biol Chem ; 281(9): 6038-47, 2006 Mar 03.
Article En | MEDLINE | ID: mdl-16373352

Neurotransmitter release from presynaptic nerve terminals is restricted to specialized areas of the plasma membrane, so-called active zones. Active zones are characterized by a network of cytoplasmic scaffolding proteins involved in active zone generation and synaptic transmission. To analyze the modes of biogenesis of this cytomatrix, we asked how Bassoon and Piccolo, two prototypic active zone cytomatrix molecules, are delivered to nascent synapses. Although these proteins may be transported via vesicles, little is known about the importance of a vesicular pathway and about molecular determinants of cytomatrix molecule trafficking. We found that Bassoon and Piccolo co-localize with markers of the trans-Golgi network in cultured neurons. Impairing vesicle exit from the Golgi complex, either using brefeldin A, recombinant proteins, or a low temperature block, prevented transport of Bassoon out of the soma. Deleting a newly identified Golgi-binding region of Bassoon impaired subcellular targeting of recombinant Bassoon. Overexpressing this region to specifically block Golgi binding of the endogenous protein reduced the concentration of Bassoon at synapses. These results suggest that, during the period of bulk synaptogenesis, a primordial cytomatrix assembles in a trans-Golgi compartment. They further indicate that transport via Golgi-derived vesicles is essential for delivery of cytomatrix proteins to the synapse. Paradigmatically this establishes Golgi transit as an obligatory step for subcellular trafficking of distinct cytoplasmic scaffolding proteins.


Cytoskeletal Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , trans-Golgi Network/physiology , Animals , Biomarkers/metabolism , Brefeldin A/pharmacology , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuropeptides/genetics , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , Protein Transport/physiology , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synapses/ultrastructure , trans-Golgi Network/ultrastructure
10.
FEBS Lett ; 579(20): 4254-8, 2005 Aug 15.
Article En | MEDLINE | ID: mdl-16051233

Raver2 was identified as a novel member of the hnRNP family based on sequence homology within three RNA recognition motifs and its general domain organization reminiscent of the previously described raver1 protein. Like raver1, raver2 contains two putative nuclear localization signals and a potential nuclear export sequence, and also displays nucleo-cytoplasmic shuttling in a heterokaryon assay. In glia cells and neurons, raver2 localizes to the nucleus. Moreover, the protein interacts with polypyrimidine tract binding protein (PTB) suggesting that it may participate in PTB-mediated nuclear functions. In contrast to ubiquitously expressed raver1, raver2 exerts a distinct spatio-temporal expression pattern during embryogenesis and is essentially restricted to brain, lung, and kidney in the adult mouse.


Heterogeneous-Nuclear Ribonucleoproteins/chemistry , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cytoplasm/chemistry , Cytoplasm/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Heterogeneous-Nuclear Ribonucleoproteins/classification , Humans , Mice , Molecular Sequence Data , Neuroglia/cytology , Neurons/chemistry , Neurons/metabolism , Nuclear Localization Signals , Nuclear Proteins/metabolism , Polypyrimidine Tract-Binding Protein/analysis , Polypyrimidine Tract-Binding Protein/metabolism , Protein Transport , RNA-Binding Proteins , Ribonucleoproteins , Tissue Distribution
11.
Mol Biol Cell ; 15(4): 1600-8, 2004 Apr.
Article En | MEDLINE | ID: mdl-14767055

Profilin 1 (PFN1) is a regulator of the microfilament system and is involved in various signaling pathways. It interacts with many cytoplasmic and nuclear ligands. The importance of PFN1 for human tissue differentiation has been demonstrated by the findings that human cancer cells, expressing conspicuously low PFN1 levels, adopt a nontumorigenic phenotype upon raising their PFN1 level. In the present study, we characterize the ligand binding site crucial for profilin's tumor suppressor activity. Starting with CAL51, a human breast cancer cell line highly tumorigenic in nude mice, we established stable clones that express PFN1 mutants differentially defective in ligand binding. Clones expressing PFN1 mutants with reduced binding to either poly-proline-stretch ligands or phosphatidyl-inositol-4,5-bisphosphate, but with a functional actin binding site, were normal in growth, adhesion, and anchorage dependence, with only a weak tendency to elicit tumors in nude mice, similar to controls expressing wild-type PFN1. In contrast, clones expressing a mutant with severely reduced capacity to bind actin still behaved like the parental CAL51 and were highly tumorigenic. We conclude that the actin binding site on profilin is instrumental for normal differentiation of human epithelia and the tumor suppressor function of PFN1.


Actins/chemistry , Contractile Proteins/physiology , Genes, Tumor Suppressor , Microfilament Proteins/physiology , Neoplasms/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Binding Sites , Cell Adhesion , Cell Division , Cell Line, Tumor , Cell Movement , Collagen/pharmacology , Cytoplasm/metabolism , Drug Combinations , Epithelium/metabolism , Female , Humans , Immunoblotting , Laminin/pharmacology , Ligands , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Phenotype , Phosphatidylinositol 4,5-Diphosphate/chemistry , Point Mutation , Profilins , Proteoglycans/pharmacology , Recombinant Proteins/chemistry , Signal Transduction , Time Factors , Transfection
...