Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Cells ; 12(18)2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37759474

ABSTRACT

The faithful formation and, consequently, function of a synapse requires continuous and tightly controlled delivery of synaptic material. At the presynapse, a variety of proteins with unequal molecular properties are indispensable to compose and control the molecular machinery concerting neurotransmitter release through synaptic vesicle fusion with the presynaptic membrane. As presynaptic proteins are produced mainly in the neuronal soma, they are obliged to traffic along microtubules through the axon to reach the consuming presynapse. This anterograde transport is performed by highly specialised and diverse presynaptic precursor vesicles, membranous organelles able to transport as different proteins such as synaptic vesicle membrane and membrane-associated proteins, cytosolic active zone proteins, ion-channels, and presynaptic membrane proteins, coordinating synaptic vesicle exo- and endocytosis. This review aims to summarise and categorise the diverse and numerous findings describing presynaptic precursor cargo, mode of trafficking, kinesin-based axonal transport and the molecular mechanisms of presynaptic precursor vesicles biogenesis in both vertebrate and invertebrate model systems.

2.
J Cell Biol ; 220(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33822845

ABSTRACT

Reliable delivery of presynaptic material, including active zone and synaptic vesicle proteins from neuronal somata to synaptic terminals, is prerequisite for successful synaptogenesis and neurotransmission. However, molecular mechanisms controlling the somatic assembly of presynaptic precursors remain insufficiently understood. We show here that in mutants of the small GTPase Rab2, both active zone and synaptic vesicle proteins accumulated in the neuronal cell body at the trans-Golgi and were, consequently, depleted at synaptic terminals, provoking neurotransmission deficits. Ectopic presynaptic material accumulations consisted of heterogeneous vesicles and short tubules of 40 × 60 nm, segregating in subfractions either positive for active zone or synaptic vesicle proteins and LAMP1, a lysosomal membrane protein. Genetically, Rab2 acts upstream of Arl8, a lysosomal adaptor controlling axonal export of precursors. Collectively, we identified a Golgi-associated assembly sequence of presynaptic precursor biogenesis dependent on a Rab2-regulated protein export and sorting step at the trans-Golgi.


Subject(s)
Golgi Apparatus/metabolism , Synaptic Vesicles/metabolism , rab2 GTP-Binding Protein/metabolism , Animals , Axons/metabolism , Drosophila melanogaster/metabolism , Female , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/metabolism , Male , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Protein Transport/physiology , Synaptic Transmission/physiology
3.
Curr Biol ; 31(8): 1711-1725.e5, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33651992

ABSTRACT

As a result of developmental synapse formation, the presynaptic neurotransmitter release machinery becomes accurately matched with postsynaptic neurotransmitter receptors. Trans-synaptic signaling is executed through cell adhesion proteins such as Neurexin::Neuroligin pairs but also through diffusible and cytoplasmic signals. How exactly pre-post coordination is ensured in vivo remains largely enigmatic. Here, we identified a "molecular choreography" coordinating pre- with postsynaptic assembly during the developmental formation of Drosophila neuromuscular synapses. Two presynaptic Neurexin-binding scaffold proteins, Syd-1 and Spinophilin (Spn), spatio-temporally coordinated pre-post assembly in conjunction with two postsynaptically operating, antagonistic Neuroligin species: Nlg1 and Nlg2. The Spn/Nlg2 module promoted active zone (AZ) maturation by driving the accumulation of AZ scaffold proteins critical for synaptic vesicle release. Simultaneously, these regulators restricted postsynaptic glutamate receptor incorporation. Both functions of the Spn/Nlg2 module were directly antagonized by Syd-1/Nlg1. Nlg1 and Nlg2 also had divergent effects on Nrx-1 in vivo motility. Concerning diffusible signals, Spn and Syd-1 antagonistically controlled the levels of Munc13-family protein Unc13B at nascent AZs, whose release function facilitated glutamate receptor incorporation at assembling postsynaptic specializations. As a result, we here provide direct in vivo evidence illustrating how a highly regulative and interleaved communication between cell adhesion protein signaling complexes and diffusible signals allows for a precise coordination of pre- with postsynaptic assembly. It will be interesting to analyze whether this logic also transfers to plasticity processes.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Animals , Cell Adhesion Molecules , Drosophila , Drosophila Proteins/genetics , Receptors, Glutamate , Synapses
4.
J Cell Biol ; 219(7)2020 07 06.
Article in English | MEDLINE | ID: mdl-32369542

ABSTRACT

At presynaptic active zones, arrays of large conserved scaffold proteins mediate fast and temporally precise release of synaptic vesicles (SVs). SV release sites could be identified by clusters of Munc13, which allow SVs to dock in defined nanoscale relation to Ca2+ channels. We here show in Drosophila that RIM-binding protein (RIM-BP) connects release sites physically and functionally to the ELKS family Bruchpilot (BRP)-based scaffold engaged in SV recruitment. The RIM-BP N-terminal domain, while dispensable for SV release site organization, was crucial for proper nanoscale patterning of the BRP scaffold and needed for SV recruitment of SVs under strong stimulation. Structural analysis further showed that the RIM-BP fibronectin domains form a "hinge" in the protein center, while the C-terminal SH3 domain tandem binds RIM, Munc13, and Ca2+ channels release machinery collectively. RIM-BPs' conserved domain architecture seemingly provides a relay to guide SVs from membrane far scaffolds into membrane close release sites.


Subject(s)
Carrier Proteins/chemistry , Central Nervous System/metabolism , Cytoskeletal Proteins/chemistry , Drosophila Proteins/chemistry , Drosophila melanogaster/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism , rab3 GTP-Binding Proteins/chemistry , Animals , Animals, Genetically Modified , Binding Sites , Calcium Channels/genetics , Calcium Channels/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Central Nervous System/ultrastructure , Cloning, Molecular , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/ultrastructure , Escherichia coli/genetics , Escherichia coli/metabolism , Female , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Larva/genetics , Larva/metabolism , Larva/ultrastructure , Male , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synapses/ultrastructure , Synaptic Transmission , Synaptic Vesicles/ultrastructure , rab3 GTP-Binding Proteins/genetics , rab3 GTP-Binding Proteins/metabolism
5.
Nat Commun ; 10(1): 1085, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30842428

ABSTRACT

Neuronal communication across synapses relies on neurotransmitter release from presynaptic active zones (AZs) followed by postsynaptic transmitter detection. Synaptic plasticity homeostatically maintains functionality during perturbations and enables memory formation. Postsynaptic plasticity targets neurotransmitter receptors, but presynaptic mechanisms regulating the neurotransmitter release apparatus remain largely enigmatic. By studying Drosophila neuromuscular junctions (NMJs) we show that AZs consist of nano-modular release sites and identify a molecular sequence that adds modules within minutes of inducing homeostatic plasticity. This requires cognate transport machinery and specific AZ-scaffolding proteins. Structural remodeling is not required for immediate potentiation of neurotransmitter release, but necessary to sustain potentiation over longer timescales. Finally, mutations in Unc13 disrupting homeostatic plasticity at the NMJ also impair short-term memory when central neurons are targeted, suggesting that both plasticity mechanisms utilize Unc13. Together, while immediate synaptic potentiation capitalizes on available material, it triggers the coincident incorporation of modular release sites to consolidate synaptic potentiation.


Subject(s)
Drosophila Proteins/metabolism , Long-Term Potentiation/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuromuscular Junction/metabolism , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Animals , Animals, Genetically Modified , Behavior, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Male , Membrane Proteins/genetics , Memory, Short-Term/physiology , Models, Animal , Mushroom Bodies/cytology , Mushroom Bodies/metabolism , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism
6.
J Cell Sci ; 132(6)2019 03 18.
Article in English | MEDLINE | ID: mdl-30745339

ABSTRACT

Protein scaffolds at presynaptic active zone membranes control information transfer at synapses. For scaffold biogenesis and maintenance, scaffold components must be safely transported along axons. A spectrum of kinases has been suggested to control transport of scaffold components, but direct kinase-substrate relationships and operational principles steering phosphorylation-dependent active zone protein transport are presently unknown. Here, we show that extensive phosphorylation of a 150-residue unstructured region at the N-terminus of the highly elongated Bruchpilot (BRP) active zone protein is crucial for ordered active zone precursor transport in Drosophila Point mutations that block SRPK79D kinase-mediated phosphorylation of the BRP N-terminus interfered with axonal transport, leading to BRP-positive axonal aggregates that also contain additional active zone scaffold proteins. Axonal aggregates formed only in the presence of non-phosphorylatable BRP isoforms containing the SRPK79D-targeted N-terminal stretch. We assume that specific active zone proteins are pre-assembled in transport packages and are thus co-transported as functional scaffold building blocks. Our results suggest that transient post-translational modification of a discrete unstructured domain of the master scaffold component BRP blocks oligomerization of these building blocks during their long-range transport.


Subject(s)
Axonal Transport/physiology , Drosophila Proteins/metabolism , Drosophila/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Phosphorylation , Presynaptic Terminals/metabolism , Synapses/metabolism
7.
Neuron ; 99(6): 1216-1232.e7, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30174114

ABSTRACT

Nervous system function relies on the polarized architecture of neurons, established by directional transport of pre- and postsynaptic cargoes. While delivery of postsynaptic components depends on the secretory pathway, the identity of the membrane compartment(s) supplying presynaptic active zone (AZ) and synaptic vesicle (SV) proteins is unclear. Live imaging in Drosophila larvae and mouse hippocampal neurons provides evidence that presynaptic biogenesis depends on axonal co-transport of SV and AZ proteins in presynaptic lysosome-related vesicles (PLVs). Loss of the lysosomal kinesin adaptor Arl8 results in the accumulation of SV- and AZ-protein-containing vesicles in neuronal cell bodies and a corresponding depletion of SV and AZ components from presynaptic sites, leading to impaired neurotransmission. Conversely, presynaptic function is facilitated upon overexpression of Arl8. Our data reveal an unexpected function for a lysosome-related organelle as an important building block for presynaptic biogenesis.


Subject(s)
Axonal Transport/physiology , Lysosomes/metabolism , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Animals , Drosophila/metabolism , Hippocampus/metabolism , Mice , Neurons/metabolism , Protein Transport/physiology , Synaptic Transmission/physiology
8.
Nature ; 536(7615): 151-2, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27462816
9.
Curr Opin Neurobiol ; 39: 69-76, 2016 08.
Article in English | MEDLINE | ID: mdl-27131423

ABSTRACT

Cognitive processes including memory formation and learning rely on a precise, local and dynamic control of synapse functionality executed by molecular changes within both presynaptic and postsynaptic compartments. Recently, the size of the presynaptic active zone scaffold, a cluster of large multi-domain proteins decorating the presynaptic plasma membrane, was found to directly scale with the action potential evoked release of synaptic vesicles. The challenge now is to constitute an integrated picture of how long-range axonal transport, local exchange and localization mechanisms at the scaffold and degradation processes are integrated to allow for dynamic and controlled scaffold rearrangements. Here we discuss findings from multiple model systems emphasizing both short-term and long-term regulations of active zone composition and function.


Subject(s)
Memory/physiology , Nerve Tissue Proteins/metabolism , Synapses/physiology , Humans , Synaptic Vesicles/metabolism
10.
Curr Biol ; 24(22): R1076-80, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25458214

ABSTRACT

Synapses are specialized asymmetric cell-cell connections permitting the controlled transfer of an electrical or chemical signal between a presynaptic neuronal cell and a postsynaptic target cell (e.g. neuron or muscle). Adequate synapse function is an essential prerequisite of all neuronal processing, including higher cognitive functions, such as learning and memory. At synapses, neurotransmitters (e.g. amino acids, amines, peptides, and acetylcholine) are released from synaptic vesicles into the synaptic cleft in response to action potentials. The Nobel Prize for Physiology and Medicine in 2013 was awarded to James E. Rothman, Randy W. Schekman and Thomas C. Südhof "for their discoveries of the machinery regulating vesicle traffic, a major transport system in our cells". This included crucial revelations, such as the identification of the core machinery of synaptic vesicle fusion. However, in contrast to the advances concerning the organization of the core functions of the synapse, our current understanding of the processes of synapse formation and maintenance--i.e. 'synaptogenesis'--is still somewhat fragmentary. Here, we will outline the current status and future directions of the field of synaptogenesis, primarily from the perspective of the presynaptic release site.


Subject(s)
Models, Biological , Synapses/metabolism , Synaptic Vesicles/metabolism , Biological Transport , Synaptic Transmission , Synaptic Vesicles/physiology , Synaptic Vesicles/ultrastructure
11.
Curr Biol ; 24(17): 2059-65, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25131677

ABSTRACT

Glutamate-releasing synapses dominate excitatory release in the brain. Mechanisms governing their assembly are of major importance for circuit development and long-term plasticity underlying learning and memory. AMPA/Kainate-type glutamate receptors (GluRs) are tetrameric ligand-gated ion channels that open their ion-conducting pores in response to binding of the neurotransmitter. Changes in subunit composition of postsynaptic GluRs are highly relevant for plasticity and development of glutamatergic synapses [1-4]. To date, posttranslational modifications, mostly operating via the intracellular C-terminal domains (CTDs) of GluRs, are presumed to be the major regulator of trafficking [5]. In recent years, structural and electrophysiological analyses have improved our understanding of GluR gating mechanism [6-11]. However, whether conformational changes subsequent to glutamate binding may per se be able to influence GluR trafficking has remained an unaddressed question. Using a Drosophila system allowing for extended visualization of GluR trafficking in vivo, we here provide evidence that mutations changing the gating behavior alter GluR distribution and trafficking. GluR mutants associated with reduced charge transfer segregated from coexpressed wild-type GluRs on the level of individual postsynaptic densities. Segregation was lost upon blocking of evoked glutamate release. Photobleaching experiments suggested increased mobility of mutants with reduced charge transfer, which accumulated prematurely during early steps of synapse assembly, but failed to further increase their level in accordance with assembly of the presynaptic scaffold. In summary, gating characteristics seem to be a new variable for the understanding of GluR trafficking relevant to both development and plasticity.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Receptors, Glutamate/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Glutamic Acid/metabolism , Ion Channel Gating , Mutation , Protein Transport , Receptors, Glutamate/metabolism
12.
Dis Model Mech ; 6(3): 689-700, 2013 May.
Article in English | MEDLINE | ID: mdl-23335205

ABSTRACT

The C subunit of the vacuolar H(+)-ATPase or V-ATPase regulates the activity and assembly of the proton pump at cellular membranes. It has been shown to be strongly upregulated in oral squamous cell carcinoma, a highly metastatic epithelial cancer. In addition, increased V-ATPase activity appears to correlate with invasiveness of cancer cells, but the underlying mechanism is largely unknown. Using the Drosophila wing imaginal epithelium as an in vivo model system, we demonstrate that overexpression of Vha44, the Drosophila orthologue of the C subunit, causes a tumor-like tissue transformation in cells of the wing epithelium. Overexpressing cells are excluded from the epithelium and acquire invasive properties while displaying high apoptotic rates. Blocking apoptosis in these cells unmasks a strong proliferation stimulus, leading to overgrowth. Furthermore, we show that excess Vha44 greatly increases acidification of endocytic compartments and interferes with endosomal trafficking. As a result, cargoes such as GFP-Lamp1 and Notch accumulate in highly acidified enlarged endolysosomal compartments. Consistent with previous reports on the endocytic activation of Eiger/JNK signaling, we find that V-ATPase stimulation by Vha44 causes JNK signaling activation whereas downmodulation of JNK signaling rescues the invasive phenotypes. In summary, our in vivo-findings demonstrate that increased levels of V-ATPase C subunit induce a Eiger/JNK-dependent cell transformation within an epithelial organ that recapitulates early carcinoma stages.


Subject(s)
Cell Movement , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Drosophila melanogaster/growth & development , Epithelium/growth & development , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Apoptosis , Body Patterning , Cell Compartmentation , Drosophila melanogaster/cytology , Endosomes/metabolism , Epithelium/enzymology , Hydrogen-Ion Concentration , Lysosomes/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Models, Biological , Mutant Proteins/metabolism , Oncogenes , Proteolysis , Receptors, Notch/metabolism , Wings, Animal/cytology , Wings, Animal/growth & development , ras Proteins/metabolism
13.
Development ; 139(10): 1874-84, 2012 May.
Article in English | MEDLINE | ID: mdl-22491943

ABSTRACT

In bilateria, positioning and looping of visceral organs requires proper left-right (L/R) asymmetry establishment. Recent work in Drosophila has identified a novel situs inversus gene encoding the unconventional type ID myosin (MyoID). In myoID mutant flies, the L/R axis is inverted, causing reversed looping of organs, such as the gut, spermiduct and genitalia. We have previously shown that MyoID interacts physically with ß-Catenin, suggesting a role of the adherens junction in Drosophila L/R asymmetry. Here, we show that DE-Cadherin co-immunoprecipitates with MyoID and is required for MyoID L/R activity. We further demonstrate that MyoIC, a closely related unconventional type I myosin, can antagonize MyoID L/R activity by preventing its binding to adherens junction components, both in vitro and in vivo. Interestingly, DE-Cadherin inhibits MyoIC, providing a protective mechanism to MyoID function. Conditional genetic experiments indicate that DE-Cadherin, MyoIC and MyoID show temporal synchronicity for their function in L/R asymmetry. These data suggest that following MyoID recruitment by ß-Catenin at the adherens junction, DE-Cadherin has a twofold effect on Drosophila L/R asymmetry by promoting MyoID activity and repressing that of MyoIC. Interestingly, the product of the vertebrate situs inversus gene inversin also physically interacts with ß-Catenin, suggesting that the adherens junction might serve as a conserved platform for determinants to establish L/R asymmetry both in vertebrates and invertebrates.


Subject(s)
Body Patterning/physiology , Cadherins/metabolism , Drosophila Proteins/metabolism , Myosins/metabolism , Animals , Body Patterning/genetics , Cadherins/genetics , Drosophila , Drosophila Proteins/genetics , Immunoprecipitation , Myosin Type I/genetics , Myosin Type I/metabolism , Myosins/genetics , Protein Binding , beta Catenin/genetics , beta Catenin/metabolism
14.
Pediatr Nephrol ; 26(9): 1523-7, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21380625

ABSTRACT

The planar cell polarity (PCP) pathway polarizes epithelia in the plane of a tissue. It regulates form and function of tissues and manifests itself by the polarized formation of cellular appendages such as epidermal hairs and cilia. Defects in the pathway are often associated with organ malformation and disease. In the kidney, the molecular events leading to cyst formation in polycystic kidney disease involve the PCP pathway. PCP is, however, best understood in Drosophila where genetic screens have identified a group of PCP core proteins including the Wnt receptor Frizzled (Fz), Dishevelled (Dsh), and Flamingo (Fmi). These proteins can localize to opposite parts of the plasma membrane in response to a poorly understood symmetry breaking event. Recent evidence suggests that proton transporters may play a role in regulating the asymmetric localization of PCP core proteins. Several papers have reported that the (pro)renin receptor, which is an associated subunit of the proton pumping V-ATPase, is required for PCP, but also for canonical Wnt signaling. Here, we discuss the implications of these findings for diverse developmental settings.


Subject(s)
Epithelial Cells/metabolism , Kidney/metabolism , Proton Pumps/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Cell Polarity , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Kidney/embryology , Morphogenesis , Proton Pumps/genetics , Signal Transduction/genetics , Wnt Proteins/genetics
15.
Curr Biol ; 20(19): 1773-8, 2010 Oct 12.
Article in English | MEDLINE | ID: mdl-20832313

ABSTRACT

Handed asymmetry in organ shape and positioning is a common feature among bilateria, yet little is known about the morphogenetic mechanisms underlying left-right (LR) organogenesis. We utilize the directional 360° clockwise rotation of genitalia in Drosophila to study LR-dependent organ looping. Using time-lapse imaging, we show that rotation of genitalia by 360° results from an additive process involving two ring-shaped domains, each undergoing 180° rotation. Our results show that the direction of rotation for each ring is autonomous and strictly depends on the LR determinant myosin ID (MyoID). Specific inactivation of MyoID in one domain causes rings to rotate in opposite directions and thereby cancels out the overall movement. We further reveal a specific pattern of apoptosis at the ring boundaries and show that local cell death is required for the movement of each domain, acting as a brake-releaser. These data indicate that organ looping can proceed through an incremental mechanism coupling LR determination and apoptosis. Furthermore, they suggest a model for the stepwise evolution of genitalia posture in Diptera, through the emergence and duplication of a 180° LR module.


Subject(s)
Apoptosis/physiology , Body Patterning/physiology , Drosophila melanogaster , Organogenesis/physiology , Animals , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Genitalia/anatomy & histology , Genitalia/embryology , Genitalia/growth & development , Time-Lapse Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...