Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Proc Natl Acad Sci U S A ; 117(21): 11760-11769, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32393629

ABSTRACT

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two related neurodegenerative diseases that present with similar TDP-43 pathology in patient tissue. TDP-43 is an RNA-binding protein which forms aggregates in neurons of ALS and FTD patients as well as in a subset of patients diagnosed with other neurodegenerative diseases. Despite our understanding that TDP-43 is essential for many aspects of RNA metabolism, it remains obscure how TDP-43 dysfunction contributes to neurodegeneration. Interestingly, altered neuronal dendritic morphology is a common theme among several neurological disorders and is thought to precede neurodegeneration. We previously found that both TDP-43 overexpression (OE) and knockdown (KD) result in reduced dendritic branching of cortical neurons. In this study, we used TRIBE (targets of RNA-binding proteins identified by editing) as an approach to identify signaling pathways that regulate dendritic branching downstream of TDP-43. We found that TDP-43 RNA targets are enriched for pathways that signal to the CREB transcription factor. We further found that TDP-43 dysfunction inhibits CREB activation and CREB transcriptional output, and restoring CREB signaling rescues defects in dendritic branching. Finally, we demonstrate, using RNA sequencing, that TDP-43 OE and KD cause similar changes in the abundance of specific messenger RNAs, consistent with their ability to produce similar morphological defects. Our data therefore provide a mechanism by which TDP-43 dysfunction interferes with dendritic branching, and may define pathways for therapeutic intervention in neurodegenerative diseases.


Subject(s)
Cyclic AMP Response Element-Binding Protein , DNA-Binding Proteins , Dendrites , Gene Expression Regulation/genetics , Signal Transduction , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendrites/metabolism , Dendrites/pathology , HEK293 Cells , Humans , RNA, Messenger/metabolism , Rats , Signal Transduction/genetics , Signal Transduction/physiology , TDP-43 Proteinopathies
2.
PLoS Genet ; 13(10): e1007052, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29028801

ABSTRACT

Lowe Syndrome is a developmental disorder characterized by eye, kidney, and neurological pathologies, and is caused by mutations in the phosphatidylinositol-5-phosphatase OCRL. OCRL plays diverse roles in endocytic and endolysosomal trafficking, cytokinesis, and ciliogenesis, but it is unclear which of these cellular functions underlie specific patient symptoms. Here, we show that mutation of Drosophila OCRL causes cell-autonomous activation of hemocytes, which are macrophage-like cells of the innate immune system. Among many cell biological defects that we identified in docrl mutant hemocytes, we pinpointed the cause of innate immune cell activation to reduced Rab11-dependent recycling traffic and concomitantly increased Rab7-dependent late endosome traffic. Loss of docrl amplifies multiple immune-relevant signals, including Toll, Jun kinase, and STAT, and leads to Rab11-sensitive mis-sorting and excessive secretion of the Toll ligand Spåtzle. Thus, docrl regulation of endosomal traffic maintains hemocytes in a poised, but quiescent state, suggesting mechanisms by which endosomal misregulation of signaling may contribute to symptoms of Lowe syndrome.


Subject(s)
Cytokinesis/genetics , Immunity, Innate/genetics , Oculocerebrorenal Syndrome/genetics , Phosphoric Monoester Hydrolases/genetics , Animals , Drosophila , Endosomes/genetics , Endosomes/pathology , Hemocytes/metabolism , Hemocytes/pathology , Humans , Mutation , Oculocerebrorenal Syndrome/pathology , Protein Binding
3.
Mol Ther ; 26(2): 648-658, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29396265

ABSTRACT

Most of the peptides used for promoting cellular uptake bear positive charges. In our previous study, we reported an example of taurine (bearing negative charges in physiological conditions) promoting cellular uptake of D-peptides. Taurine, conjugated to a small D-peptide via an ester bond, promotes the cellular uptake of this D-peptide. Particularly, intracellular carboxylesterase (CES) instructs the D-peptide to self-assemble and to form nanofibers, which largely disfavors efflux and further enhances the intracellular accumulation of the D-peptide, as supported by that the addition of CES inhibitors partially impaired cellular uptake of this molecule in mammalian cell lines. Using dynamin 1, 2, and 3 triple knockout (TKO) mouse fibroblasts, we demonstrated that cells took up this molecule via macropinocytosis and dynamin-dependent endocytosis. Imaging of Drosophila larval blood cells derived from endocytic mutants confirmed the involvement of multiple endocytosis pathways. Electron microscopy (EM) indicated that the precursors can form aggregates on the cell surface to facilitate the cellular uptake via macropinocytosis. EM also revealed significantly increased numbers of vesicles in the cytosol. This work provides new insights into the cellular uptake of taurine derivative for intracellular delivery and self-assembly of D-peptides.


Subject(s)
Dynamins/metabolism , Endocytosis/drug effects , Peptides/pharmacology , Pinocytosis/drug effects , Taurine , Animals , Biological Transport , Cell Line , Fluorescent Antibody Technique , HeLa Cells , Humans , Molecular Structure , Peptides/chemistry , Signal Transduction/drug effects , Taurine/chemistry
4.
Proc Natl Acad Sci U S A ; 113(38): E5552-61, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27601635

ABSTRACT

Membrane remodeling by Fes/Cip4 homology-Bin/Amphiphysin/Rvs167 (F-BAR) proteins is regulated by autoinhibitory interactions between their SRC homology 3 (SH3) and F-BAR domains. The structural basis of autoregulation, and whether it affects interactions of SH3 domains with other cellular ligands, remain unclear. Here we used single-particle electron microscopy to determine the structure of the F-BAR protein Nervous Wreck (Nwk) in both soluble and membrane-bound states. On membrane binding, Nwk SH3 domains do not completely dissociate from the F-BAR dimer, but instead shift from its concave surface to positions on either side of the dimer. Unexpectedly, along with controlling membrane binding, these autoregulatory interactions inhibit the ability of Nwk-SH3a to activate Wiskott-Aldrich syndrome protein (WASp)/actin related protein (Arp) 2/3-dependent actin filament assembly. In Drosophila neurons, Nwk autoregulation restricts SH3a domain-dependent synaptopod formation, synaptic growth, and actin organization. Our results define structural rearrangements in Nwk that control F-BAR-membrane interactions as well as SH3 domain activities, and suggest that these two functions are tightly coordinated in vitro and in vivo.


Subject(s)
Drosophila Proteins/chemistry , Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Neurons/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/chemistry , Amino Acid Sequence/genetics , Animals , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Humans , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Binding , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics , src Homology Domains/genetics
5.
Traffic ; 17(2): 87-101, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26538429

ABSTRACT

Neurons require target-derived autocrine and paracrine growth factors to maintain proper identity, innervation, homeostasis and survival. Neuronal growth factor signaling is highly dependent on membrane traffic, both for the packaging and release of the growth factors themselves, and for regulation of intracellular signaling by their transmembrane receptors. Here, we review recent findings from the Drosophila larval neuromuscular junction (NMJ) that illustrate how specific steps of intracellular traffic and inter-organelle interactions impinge on signaling, particularly in the bone morphogenic protein, Wingless and c-Jun-activated kinase pathways, regulating elaboration and stability of NMJ arbors, construction of synapses and synaptic transmission and homeostasis. These membrane trafficking and signaling pathways have been implicated in human motor neuron diseases including amyotrophic lateral sclerosis and hereditary spastic paraplegia, highlighting their importance for neuronal health and survival.


Subject(s)
Drosophila/physiology , Nervous System Diseases/physiopathology , Protein Transport/physiology , Signal Transduction/physiology , Synapses/physiology , Animals , Humans , Motor Neurons/pathology , Neuromuscular Junction/physiology , Synaptic Transmission/physiology
6.
J Am Chem Soc ; 140(10): 3505-3509, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29481071

ABSTRACT

Despite the advancement of molecular imaging techniques, there is an unmet need for probes for direct imaging of membrane dynamics of live cells. Here we report a novel type of active (or enzyme responsive) probes to directly image membrane dynamics of live cells with high spatial and temporal resolution over extended time scales and areas. Because lipid rafts enrich cholesterols and GPI-anchored enzymes (e.g., ectophosphatases), we design probes that consist of an enzymatic trigger, a fluorophore, and a cholesterol that are affinitive to the cell membrane. Being water-soluble and as the substrate of ectophosphatase, these cell compatible probes preferentially and rapidly assemble in plasma membrane, exhibit strong fluorescence, work at micromolar concentrations, and easily achieve high resolution monitoring of nanoscale heterogeneity in membranes of live cells, the release of exosomes, and the membrane dynamics of live cells. This work provides a facile means to link membrane dynamics and heterogeneity to cellular processes for understanding the interactions between membranes and proteins.


Subject(s)
Cell Membrane/metabolism , Fluorescent Dyes/chemistry , Molecular Imaging , Cell Line , Cell Membrane/chemistry , Cell Survival , Humans , Molecular Structure
7.
J Am Chem Soc ; 140(30): 9566-9573, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29995402

ABSTRACT

The endoplasmic reticulum (ER) is responsible for the synthesis and folding of a large number of proteins, as well as intracellular calcium regulation, lipid synthesis, and lipid transfer to other organelles, and is emerging as a target for cancer therapy. However, strategies for selectively targeting the ER of cancer cells are limited. Here we show that enzymatically generated crescent-shaped supramolecular assemblies of short peptides disrupt cell membranes and target ER for selective cancer cell death. As revealed by sedimentation assay, the assemblies interact with synthetic lipid membranes. Live cell imaging confirms that the assemblies impair membrane integrity, which is further supported by lactate dehydrogenase (LDH) assays. According to transmission electron microscopy (TEM), static light scattering (SLS), and critical micelle concentration (CMC), attaching an l-amino acid at the C-terminal of a d-tripeptide results in the crescent-shaped supramolecular assemblies. Structure-activity relationship suggests that the crescent-shaped morphology is critical for interacting with membranes and for controlling cell fate. Moreover, fluorescent imaging indicates that the assemblies accumulate on the ER. Time-dependent Western blot and ELISA indicate that the accumulation causes ER stress and subsequently activates the caspase signaling cascade for cell death. As an approach for in situ generating membrane binding scaffolds (i.e., the crescent-shaped supramolecular assemblies), this work promises a new way to disrupt the membrane and to target the ER for developing anticancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Oligopeptides/pharmacology , Phosphopeptides/pharmacology , Alkaline Phosphatase/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Humans , Liposomes/metabolism , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/metabolism , Phosphopeptides/chemical synthesis , Phosphopeptides/chemistry , Phosphopeptides/metabolism , Protein Multimerization , Structure-Activity Relationship
8.
Angew Chem Int Ed Engl ; 56(51): 16297-16301, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29125896

ABSTRACT

Higher-order assemblies of proteins, with a structural and dynamic continuum, is an important concept in biology, but these insights have yet to be applied in designing biomaterials. Dynamic assemblies of supramolecular phosphoglycopeptides (sPGPs) transform a 2D cell sheet into 3D cell spheroids. A ligand-receptor interaction between a glycopeptide and a phosphopeptide produces sPGPs that form nanoparticles, which transform into nanofibrils upon partial enzymatic dephosphorylation. The assemblies form dynamically and hierarchically in situ on the cell surface, and interact with the extracellular matrix molecules and effectively abolish contact inhibition of locomotion (CIL) of the cells. Integrating molecular recognition, catalysis, and assembly, these active assemblies act as a dynamic continuum to disrupt CIL, thus illustrating a new kind of biomaterial for regulating cell behavior.


Subject(s)
Glycopeptides/metabolism , Spheroids, Cellular/chemistry , Cell Line , Fluorescence , Glycopeptides/chemistry , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Molecular Structure , Spheroids, Cellular/metabolism
9.
PLoS Genet ; 9(4): e1003450, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23593037

ABSTRACT

The formation of synapses and the proper construction of neural circuits depend on signaling pathways that regulate cytoskeletal structure and dynamics. After the mutual recognition of a growing axon and its target, multiple signaling pathways are activated that regulate cytoskeletal dynamics to determine the morphology and strength of the connection. By analyzing Drosophila mutations in the cytoplasmic FMRP interacting protein Cyfip, we demonstrate that this component of the WAVE complex inhibits the assembly of filamentous actin (F-actin) and thereby regulates key aspects of synaptogenesis. Cyfip regulates the distribution of F-actin filaments in presynaptic neuromuscular junction (NMJ) terminals. At cyfip mutant NMJs, F-actin assembly was accelerated, resulting in shorter NMJs, more numerous satellite boutons, and reduced quantal content. Increased synaptic vesicle size and failure to maintain excitatory junctional potential amplitudes under high-frequency stimulation in cyfip mutants indicated an endocytic defect. cyfip mutants exhibited upregulated bone morphogenetic protein (BMP) signaling, a major growth-promoting pathway known to be attenuated by endocytosis at the Drosophila NMJ. We propose that Cyfip regulates synapse development and endocytosis by inhibiting actin assembly.


Subject(s)
Adaptor Proteins, Signal Transducing , Cytoskeleton , Drosophila Proteins , Drosophila melanogaster , Synapses , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Mutation , Neuromuscular Junction/metabolism , Signal Transduction , Synapses/genetics , Synapses/metabolism , Synapses/physiology
10.
J Neurosci ; 33(45): 17560-8, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24198346

ABSTRACT

The fruit fly Drosophila melanogaster has been established as a premier experimental model system for neuroscience research. These organisms are genetically tractable, yet their nervous systems are sufficiently complex to study diverse processes that are conserved across metazoans, including neural cell fate determination and migration, axon guidance, synaptogenesis and function, behavioral neurogenetics, and responses to neuronal injury. For several decades, Drosophila neuroscientists have taken advantage of a vast toolkit of genetic and molecular techniques to reveal fundamental principles of neuroscience illuminating to all systems, including the first behavioral mutants from Seymour Benzer's pioneering work in the 1960s and 1970s, the cloning of the first potassium channel in the 1980s, and the identification of the core genes that orchestrate axon guidance and circadian rhythms in the 1990s. Over the past decade, new tools and innovations in genetic, imaging, and electrophysiological technologies have enabled the visualization, in vivo, of dynamic processes in synapses with unprecedented resolution. We will review some of the fresh insights into synaptic development, function, and plasticity that have recently emerged in Drosophila with an emphasis on the unique advantages of this model system.


Subject(s)
Drosophila/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/physiology , Animals , Behavior, Animal/physiology , Neurogenesis/physiology
11.
Proc Natl Acad Sci U S A ; 108(29): 11884-9, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730168

ABSTRACT

Evidence for cooperation between actin nucleators is growing. The WH2-containing nucleator Spire and the formin Cappuccino interact directly, and both are essential for assembly of an actin mesh during Drosophila oogenesis. Their interaction requires the kinase noncatalytic C-lobe domain (KIND) domain of Spire and the C-terminal tail of the formin. Here we describe the crystal structure of the KIND domain of human Spir1 alone and in complex with the tail of Fmn2, a mammalian ortholog of Cappuccino. The KIND domain is structurally similar to the C-lobe of protein kinases. The Fmn2 tail is coordinated in an acidic cleft at the base of the domain that appears to have evolved via deletion of a helix from the canonical kinase fold. Our functional analysis of Cappuccino reveals an unexpected requirement for its tail in actin assembly. In addition, we find that the KIND/tail interaction blocks nucleation by Cappuccino and promotes its displacement from filament barbed ends providing insight into possible modes of cooperation between Spire and Cappuccino.


Subject(s)
Actins/metabolism , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Models, Molecular , Nerve Tissue Proteins/chemistry , Oogenesis/physiology , Protein Conformation , Protein Structure, Tertiary/genetics , Animals , Crystallization , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , Fluorescence Polarization , Humans , Microfilament Proteins/chemistry , Microfilament Proteins/genetics
12.
SIAM J Appl Math ; 84(3): S476-S492, 2024.
Article in English | MEDLINE | ID: mdl-38912397

ABSTRACT

The transport of particles in cells is influenced by the properties of intracellular networks they traverse while searching for localized target regions or reaction partners. Moreover, given the rapid turnover in many intracellular structures, it is crucial to understand how temporal changes in the network structure affect diffusive transport. In this work, we use network theory to characterize complex intracellular biological environments across scales. We develop an efficient computational method to compute the mean first passage times for simulating a particle diffusing along two-dimensional planar networks extracted from fluorescence microscopy imaging. We first benchmark this methodology in the context of synthetic networks, and subsequently apply it to live-cell data from endoplasmic reticulum tubular networks.

13.
bioRxiv ; 2024 May 05.
Article in English | MEDLINE | ID: mdl-38746182

ABSTRACT

Extracellular vesicles (EVs) are released by many cell types including neurons, carrying cargoes involved in signaling and disease. It is unclear whether EVs promote intercellular signaling or serve primarily to dispose of unwanted materials. We show that loss of multivesicular endosome-generating ESCRT (endosomal sorting complex required for transport) machinery disrupts release of EV cargoes from Drosophila motor neurons. Surprisingly, ESCRT depletion does not affect the signaling activities of the EV cargo Synaptotagmin-4 (Syt4) and disrupts only some signaling activities of the EV cargo Evenness Interrupted (Evi). Thus, these cargoes may not require intercellular transfer via EVs, and instead may be conventionally secreted or function cell autonomously in the neuron. We find that EVs are phagocytosed by glia and muscles, and that ESCRT disruption causes compensatory autophagy in presynaptic neurons, suggesting that EVs are one of several redundant mechanisms to remove cargoes from synapses. Our results suggest that synaptic EV release serves primarily as a proteostatic mechanism for certain cargoes.

14.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38842573

ABSTRACT

Extracellular vesicles (EVs) are released by many cell types, including neurons, carrying cargoes involved in signaling and disease. It is unclear whether EVs promote intercellular signaling or serve primarily to dispose of unwanted materials. We show that loss of multivesicular endosome-generating endosomal sorting complex required for transport (ESCRT) machinery disrupts release of EV cargoes from Drosophila motor neurons. Surprisingly, ESCRT depletion does not affect the signaling activities of the EV cargo Synaptotagmin-4 (Syt4) and disrupts only some signaling activities of the EV cargo evenness interrupted (Evi). Thus, these cargoes may not require intercellular transfer via EVs, and instead may be conventionally secreted or function cell-autonomously in the neuron. We find that EVs are phagocytosed by glia and muscles, and that ESCRT disruption causes compensatory autophagy in presynaptic neurons, suggesting that EVs are one of several redundant mechanisms to remove cargoes from synapses. Our results suggest that synaptic EV release serves primarily as a proteostatic mechanism for certain cargoes.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Endosomal Sorting Complexes Required for Transport , Extracellular Vesicles , Motor Neurons , Signal Transduction , Synapses , Animals , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , Extracellular Vesicles/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Synapses/metabolism , Motor Neurons/metabolism , Autophagy , Synaptotagmins/metabolism , Synaptotagmins/genetics , Neuroglia/metabolism
15.
bioRxiv ; 2023 Nov 13.
Article in English | MEDLINE | ID: mdl-38014140

ABSTRACT

Neuromuscular junctions (NMJs) are evolutionarily ancient, specialized contacts between neurons and muscles. Axons and NMJs must endure mechanical strain through a lifetime of muscle contraction, making them vulnerable to aging and neurodegenerative conditions. However, cellular strategies for mitigating this mechanical stress remain unknown. In this study, we used Drosophila larval NMJs to investigate the role of actin and myosin (actomyosin)-mediated contractility in generating and responding to cellular forces at the neuron-muscle interface. We identified a new long-lived, low-turnover presynaptic actin core traversing the NMJ, which partly co-localizes with non-muscle myosin II (NMII). Neuronal RNAi of NMII induced disorganization of this core, suggesting that this structure might have contractile properties. Interestingly, neuronal RNAi of NMII also decreased NMII levels in the postsynaptic muscle proximal to neurons, suggesting that neuronal actomyosin rearrangements propagate their effects transsynaptically. We also observed reduced Integrin levels upon NMII knockdown, indicating that neuronal actomyosin disruption triggers rearrangements of Integrin-mediated connections between neurons and surrounding muscle tissue. In summary, our study identifies a previously uncharacterized presynaptic actomyosin subpopulation that upholds the neuronal mechanical continuum, transmits signals to adjacent muscle tissue, and collaborates with Integrin receptors to govern the mechanobiology of the neuromuscular junction.

16.
Mol Biol Cell ; 34(6): ar51, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36542486

ABSTRACT

Following exocytosis at active zones, synaptic vesicle membranes and membrane-bound proteins must be recycled. The endocytic machinery that drives this recycling accumulates in the periactive zone (PAZ), a region of the synapse adjacent to active zones, but the organization of this machinery within the PAZ, and how PAZ composition relates to active zone release properties, remains unknown. The PAZ is also enriched for cell adhesion proteins, but their function at these sites is poorly understood. Here, using Airyscan and stimulated emission depletion imaging of Drosophila synapses, we develop a quantitative framework describing the organization and ultrastructure of the PAZ. Different endocytic proteins localize to distinct regions of the PAZ, suggesting that subdomains are specialized for distinct biochemical activities, stages of membrane remodeling, or synaptic functions. We find that the accumulation and distribution of endocytic but not adhesion PAZ proteins correlate with the abundance of the scaffolding protein Bruchpilot at active zones-a structural correlate of release probability. These data suggest that endocytic and exocytic activities are spatially correlated. Taken together, our results identify novel relationships between the exocytic and endocytic apparatus at the synapse and provide a new conceptual framework to quantify synaptic architecture.


Subject(s)
Drosophila Proteins , Synapses , Animals , Synapses/metabolism , Synaptic Vesicles/metabolism , Drosophila/metabolism , Membrane Proteins/metabolism , Drosophila Proteins/metabolism , Synaptic Transmission
17.
Nat Commun ; 14(1): 999, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36890170

ABSTRACT

Dominant mutations in tyrosyl-tRNA synthetase (YARS1) and six other tRNA ligases cause Charcot-Marie-Tooth peripheral neuropathy (CMT). Loss of aminoacylation is not required for their pathogenicity, suggesting a gain-of-function disease mechanism. By an unbiased genetic screen in Drosophila, we link YARS1 dysfunction to actin cytoskeleton organization. Biochemical studies uncover yet unknown actin-bundling property of YARS1 to be enhanced by a CMT mutation, leading to actin disorganization in the Drosophila nervous system, human SH-SY5Y neuroblastoma cells, and patient-derived fibroblasts. Genetic modulation of F-actin organization improves hallmark electrophysiological and morphological features in neurons of flies expressing CMT-causing YARS1 mutations. Similar beneficial effects are observed in flies expressing a neuropathy-causing glycyl-tRNA synthetase. Hence, in this work, we show that YARS1 is an evolutionary-conserved F-actin organizer which links the actin cytoskeleton to tRNA-synthetase-induced neurodegeneration.


Subject(s)
Actins , Tyrosine-tRNA Ligase , Animals , Humans , Actins/metabolism , Charcot-Marie-Tooth Disease/genetics , Drosophila/genetics , Glycine-tRNA Ligase/genetics , Mutation , RNA, Transfer , Tyrosine-tRNA Ligase/genetics , Tyrosine-tRNA Ligase/metabolism , Cell Line, Tumor
18.
Neuron ; 110(5): 735-737, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35240059

ABSTRACT

In this issue of Neuron, Yang et al. show that autophagy machinery is tightly coupled to neuronal activity via endocytic cycling of the transmembrane protein ATG-9 at presynaptic terminals.


Subject(s)
Autophagosomes , Endocytosis , Autophagosomes/metabolism , Autophagy/physiology , Autophagy-Related Proteins/metabolism , Endocytosis/physiology , Neurons/metabolism
19.
J Cell Biol ; 221(5)2022 05 02.
Article in English | MEDLINE | ID: mdl-35320349

ABSTRACT

Neuronal extracellular vesicles (EVs) are locally released from presynaptic terminals, carrying cargoes critical for intercellular signaling and disease. EVs are derived from endosomes, but it is unknown how these cargoes are directed to the EV pathway rather than for conventional endolysosomal degradation. Here, we find that endocytic machinery plays an unexpected role in maintaining a release-competent pool of EV cargoes at synapses. Endocytic mutants, including nervous wreck (nwk), shibire/dynamin, and AP-2, unexpectedly exhibit local presynaptic depletion specifically of EV cargoes. Accordingly, nwk mutants phenocopy synaptic plasticity defects associated with loss of the EV cargo synaptotagmin-4 (Syt4) and suppress lethality upon overexpression of the EV cargo amyloid precursor protein (APP). These EV defects are genetically separable from canonical endocytic functions in synaptic vesicle recycling and synaptic growth. Endocytic machinery opposes the endosomal retromer complex to regulate EV cargo levels and acts upstream of synaptic cargo removal by retrograde axonal transport. Our data suggest a novel molecular mechanism that locally promotes cargo loading into synaptic EVs.


Subject(s)
Extracellular Vesicles , Synaptic Vesicles , Endosomes , Extracellular Vesicles/metabolism , Presynaptic Terminals/metabolism , Synapses/metabolism , Synaptic Vesicles/metabolism
20.
Curr Biol ; 18(6): R259-61, 2008 Mar 25.
Article in English | MEDLINE | ID: mdl-18364232

ABSTRACT

Acute photo-inactivation of clathrin in Drosophila synapses sheds new light on a 35-year-old debate over mechanisms of synaptic-vesicle endocytosis: clathrin is essential for reformation of functional synaptic vesicles, but not for bulk membrane internalization.


Subject(s)
Clathrin/metabolism , Endocytosis/physiology , Synaptic Vesicles/metabolism , Animals , Drosophila , Dynamins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL