Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Neurosci ; 40(33): 6270-6288, 2020 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-32631939

RESUMEN

Structural and functional plasticity induced by neuronal competition is a common feature of developing nervous systems. However, the rules governing how postsynaptic cells differentiate between presynaptic inputs are unclear. In this study, we characterized synaptic interactions following manipulations of tonic Ib or phasic Is glutamatergic motoneurons that coinnervate postsynaptic muscles of male or female Drosophila melanogaster larvae. After identifying drivers for each neuronal subtype, we performed ablation or genetic manipulations to alter neuronal activity and examined the effects on synaptic innervation and function at neuromuscular junctions. Ablation of either Ib or Is resulted in decreased muscle response, with some functional compensation occurring in the Ib input when Is was missing. In contrast, the Is terminal failed to show functional or structural changes following loss of the coinnervating Ib input. Decreasing the activity of the Ib or Is neuron with tetanus toxin light chain resulted in structural changes in muscle innervation. Decreased Ib activity resulted in reduced active zone (AZ) number and decreased postsynaptic subsynaptic reticulum volume, with the emergence of filopodial-like protrusions from synaptic boutons of the Ib input. Decreased Is activity did not induce structural changes at its own synapses, but the coinnervating Ib motoneuron increased the number of synaptic boutons and AZs it formed. These findings indicate that tonic Ib and phasic Is motoneurons respond independently to changes in activity, with either functional or structural alterations in the Ib neuron occurring following ablation or reduced activity of the coinnervating Is input, respectively.SIGNIFICANCE STATEMENT Both invertebrate and vertebrate nervous systems display synaptic plasticity in response to behavioral experiences, indicating that underlying mechanisms emerged early in evolution. How specific neuronal classes innervating the same postsynaptic target display distinct types of plasticity is unclear. Here, we examined whether Drosophila tonic Ib and phasic Is motoneurons display competitive or cooperative interactions during innervation of the same muscle, or compensatory changes when the output of one motoneuron is altered. We established a system to differentially manipulate the motoneurons and examined the effects of cell type-specific changes to one of the inputs. Our findings indicate Ib and Is motoneurons respond differently to activity mismatch or loss of the coinnervating input, with the Ib subclass responding robustly compared with Is motoneurons.


Asunto(s)
Neuronas Motoras/citología , Neuronas Motoras/fisiología , Unión Neuromuscular/citología , Unión Neuromuscular/fisiología , Plasticidad Neuronal , Sinapsis/fisiología , Animales , Drosophila melanogaster , Femenino , Ácido Glutámico/fisiología , Masculino , Potenciales de la Membrana , Terminales Presinápticos/fisiología
2.
J Neurosci ; 37(12): 3425-3439, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28235896

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by expansion of a polyglutamine (polyQ) stretch within the Huntingtin (Htt) protein. Pathogenic Htt disrupts multiple neuronal processes, including gene expression, axonal trafficking, proteasome and mitochondrial activity, and intracellular vesicle trafficking. However, the primary pathogenic mechanism and subcellular site of action for mutant Htt are still unclear. Using a Drosophila HD model, we found that pathogenic Htt expression leads to a profound overgrowth of synaptic connections that correlates directly with the levels of Htt at nerve terminals. Branches of the same nerve containing different levels of Htt show distinct phenotypes, indicating that Htt acts locally to disrupt synaptic growth. The effects of pathogenic Htt on synaptic growth arise from defective synaptic endosomal trafficking, leading to expansion of a recycling endosomal signaling compartment containing Sorting Nexin 16 and a reduction in late endosomes containing Rab11. The disruption of endosomal compartments leads to elevated BMP signaling within nerve terminals, driving excessive synaptic growth. Blocking aberrant signaling from endosomes or reducing BMP activity ameliorates the severity of HD pathology and improves viability. Pathogenic Htt is present largely in a nonaggregated form at synapses, indicating that cytosolic forms of the protein are likely to be the toxic species that disrupt endosomal signaling. Our data indicate that pathogenic Htt acts locally at nerve terminals to alter trafficking between endosomal compartments, leading to defects in synaptic structure that correlate with pathogenesis and lethality in the Drosophila HD model.SIGNIFICANCE STATEMENT Huntington's disease (HD) is the most commonly inherited polyglutamine expansion disorder, but how mutant Huntingtin (Htt) disrupts neuronal function is unclear. In particular, it is unknown within what subcellular compartment pathogenic Htt acts and whether the pathogenesis is associated with aggregated or more soluble forms of the protein. Using a Drosophila HD model, we find that nonaggregated pathogenic Htt acts locally at synaptic terminals to disrupt endosomal compartments, leading to aberrant wiring defects. Genetic manipulations to increase endosomal trafficking of synaptic growth receptors from signaling endosomes or to reduce BMP signaling reduce pathology in this HD model. These data indicate that pathogenic Htt can act locally within nerve terminals to disrupt synaptic endosomal signaling and induce neuropathology.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Endosomas/patología , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Animales , Aumento de la Célula , Drosophila/metabolismo , Endosomas/metabolismo , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Transducción de Señal , Tasa de Supervivencia
3.
J Neurosci ; 36(21): 5820-32, 2016 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-27225771

RESUMEN

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.


Asunto(s)
Orientación del Axón/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis/fisiología , Terminales Presinápticos/fisiología , Vía de Señalización Wnt/fisiología , Animales , Animales Modificados Genéticamente , Drosophila/genética , Humanos , Proteínas del Tejido Nervioso/genética , Unión Neuromuscular/fisiología
4.
Mol Cell Neurosci ; 61: 241-54, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25066865

RESUMEN

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.


Asunto(s)
Actinas/metabolismo , Proteínas de Drosophila/metabolismo , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Receptores de Glutamato/metabolismo , Actinas/genética , Factores de Edad , Animales , Animales Modificados Genéticamente , Proteínas de Unión al Calcio/metabolismo , Drosophila , Proteínas de Drosophila/genética , Regulación de la Expresión Génica/genética , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Peroxidasa de Rábano Silvestre/metabolismo , Larva , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica de Transmisión , Modelos Moleculares , Mutación/efectos de los fármacos , Mutación/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Neuromuscular/ultraestructura , Técnicas de Placa-Clamp , Receptores de Glutamato/genética , Potenciales Sinápticos/genética , Calponinas
5.
J Neurosci ; 33(1): 187-200, 2013 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-23283333

RESUMEN

Synaptic vesicle fusion mediates communication between neurons and is triggered by rapid influx of Ca(2+). The Ca(2+)-triggering step for fusion is regulated by the synaptic vesicle transmembrane protein Synaptotagmin 1 (Syt1). Syt1 contains two cytoplasmic C2 domains, termed C2A and C2B, which coordinate Ca(2+) binding. Although C2A and C2B share similar topology, binding of Ca(2+) ions to the C2B domain has been suggested as the only critical trigger for evoked vesicle release. If and how C2A domain function is coordinated with C2B remain unclear. In this study, we generated a panel of Syt1 chimeric constructs in Drosophila to delineate the unique and shared functions of each C2 domain in regulation of synaptic vesicle fusion. Expression of Syt 1 transgenes containing only individual C2 domains, or dual C2A-C2A or C2B-C2B chimeras, failed to restore Syt1 function in a syt1(-/-) null mutant background, indicating both C2A and C2B are specifically required to support fast synchronous release. Mutations that disrupted Ca(2+) binding to both C2 domains failed to rescue evoked release, but supported synaptic vesicle docking and endocytosis, indicating that these functions of Syt1 are Ca(2+)-independent. The dual C2 domain Ca(2+)-binding mutant also enhanced spontaneous fusion while dramatically increasing evoked release when coexpressed with native Syt1. Together, these data indicate that synaptic transmission can be regulated by Syt1 multimerization and that both C2 domains of Syt1 are uniquely required for modulating Ca(2+)-independent spontaneous fusion and Ca(2+)-dependent synchronous release.


Asunto(s)
Calcio/metabolismo , Neuronas/metabolismo , Transmisión Sináptica/genética , Vesículas Sinápticas/genética , Sinaptotagmina I/genética , Animales , Drosophila , Mutación , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/metabolismo , Transgenes
6.
J Neurosci ; 33(44): 17253-63, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174659

RESUMEN

Neurotransmitter release from synaptic vesicle fusion is the fundamental mechanism for neuronal communication at synapses. Evoked release following an action potential has been well characterized for its function in activating the postsynaptic cell, but the significance of spontaneous release is less clear. Using transgenic tools to image single synaptic vesicle fusion events at individual release sites (active zones) in Drosophila, we characterized the spatial and temporal dynamics of exocytotic events that occur spontaneously or in response to an action potential. We also analyzed the relationship between these two modes of fusion at single release sites. A majority of active zones participate in both modes of fusion, although release probability is not correlated between the two modes of release and is highly variable across the population. A subset of active zones is specifically dedicated to spontaneous release, indicating a population of postsynaptic receptors is uniquely activated by this mode of vesicle fusion. Imaging synaptic transmission at individual release sites also revealed general rules for spontaneous and evoked release, and indicate that active zones with similar release probability can cluster spatially within individual synaptic boutons. These findings suggest neuronal connections contain two information channels that can be spatially segregated and independently regulated to transmit evoked or spontaneous fusion signals.


Asunto(s)
Potenciales de Acción/fisiología , Potenciales Evocados/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Drosophila , Femenino , Masculino , Vesículas Sinápticas/fisiología
7.
Mol Cell Neurosci ; 52: 161-72, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23159779

RESUMEN

Complexins are small α-helical proteins that modulate neurotransmitter release by binding to SNARE complexes during synaptic vesicle exocytosis. They have been found to function as fusion clamps to inhibit spontaneous synaptic vesicle fusion in the absence of Ca(2+), while also promoting evoked neurotransmitter release following an action potential. Complexins consist of an N-terminal domain and an accessory α-helix that regulates the activating and inhibitory properties of the protein, respectively, and a central α-helix that binds the SNARE complex and is essential for both functions. In addition, complexins contain a largely unstructured C-terminal domain whose role in synaptic vesicle cycling is poorly defined. Here, we demonstrate that the C-terminus of Drosophila complexin (DmCpx) regulates localization to synapses and that alternative splicing of the C-terminus can differentially regulate spontaneous and evoked neurotransmitter release. Characterization of the single DmCpx gene by mRNA analysis revealed expression of two alternatively expressed isoforms, DmCpx7A and DmCpx7B, which encode proteins with different C-termini that contain or lack a membrane tethering prenylation domain. The predominant isoform, DmCpx7A, is further modified by RNA editing within this C-terminal region. Functional analysis of the splice isoforms showed that both are similarly localized to synaptic boutons at larval neuromuscular junctions, but have differential effects on the regulation of evoked and spontaneous fusion. These data indicate that the C-terminus of Drosophila complexin regulates both spontaneous and evoked release through separate mechanisms and that alternative splicing generates isoforms with distinct effects on the two major modes of synaptic vesicle fusion at synapses.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Unión Neuromuscular/metabolismo , Neurotransmisores/metabolismo , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Western Blotting , Drosophila , Proteínas de Drosophila/genética , Exocitosis , Inmunohistoquímica , Hibridación in Situ , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo
8.
J Neurosci ; 32(50): 18054-67, 18067a, 2012 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23238721

RESUMEN

Sustained neuronal communication relies on the coordinated activity of multiple proteins that regulate synaptic vesicle biogenesis and cycling within the presynaptic terminal. Synaptogyrin and synaptophysin are conserved MARVEL domain-containing transmembrane proteins that are among the most abundant synaptic vesicle constituents, although their role in the synaptic vesicle cycle has remained elusive. To further investigate the function of these proteins, we generated and characterized a synaptogyrin (gyr)-null mutant in Drosophila, whose genome encodes a single synaptogyrin isoform and lacks a synaptophysin homolog. We demonstrate that Drosophila synaptogyrin plays a modulatory role in synaptic vesicle biogenesis at larval neuromuscular junctions. Drosophila lacking synaptogyrin are viable and fertile and have no overt deficits in motor function. However, ultrastructural analysis of gyr larvae revealed increased synaptic vesicle diameter and enhanced variability in the size of synaptic vesicles. In addition, the resolution of endocytic cisternae into synaptic vesicles in response to strong stimulation is defective in gyr mutants. Electrophysiological analysis demonstrated an increase in quantal size and a concomitant decrease in quantal content, suggesting functional consequences for transmission caused by the loss of synaptogyrin. Furthermore, high-frequency stimulation resulted in increased facilitation and a delay in recovery from synaptic depression, indicating that synaptic vesicle exo-endocytosis is abnormally regulated during intense stimulation conditions. These results suggest that synaptogyrin modulates the synaptic vesicle exo-endocytic cycle and is required for the proper biogenesis of synaptic vesicles at nerve terminals.


Asunto(s)
Vesículas Sinápticas/genética , Vesículas Sinápticas/metabolismo , Sinaptogirinas/genética , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Western Blotting , Drosophila , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Mutación , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/ultraestructura , Técnicas de Placa-Clamp , Vesículas Sinápticas/ultraestructura
9.
J Neurosci ; 32(50): 18234-45, 2012 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-23238737

RESUMEN

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.


Asunto(s)
Neurotransmisores/metabolismo , Proteínas SNARE/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Sinaptotagminas/metabolismo , Animales , Western Blotting , Drosophila , Proteínas de Drosophila/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Exocitosis/fisiología , Técnicas de Inactivación de Genes , Inmunohistoquímica , Microscopía Electrónica de Transmisión , Unión Neuromuscular/metabolismo , Técnicas de Placa-Clamp
10.
Elife ; 122023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37368474

RESUMEN

Heparan sulfate proteoglycans (HSPGs) form essential components of the extracellular matrix (ECM) and basement membrane (BM) and have both structural and signaling roles. Perlecan is a secreted ECM-localized HSPG that contributes to tissue integrity and cell-cell communication. Although a core component of the ECM, the role of Perlecan in neuronal structure and function is less understood. Here, we identify a role for Drosophila Perlecan in the maintenance of larval motoneuron axonal and synaptic stability. Loss of Perlecan causes alterations in the axonal cytoskeleton, followed by axonal breakage and synaptic retraction of neuromuscular junctions. These phenotypes are not prevented by blocking Wallerian degeneration and are independent of Perlecan's role in Wingless signaling. Expression of Perlecan solely in motoneurons cannot rescue synaptic retraction phenotypes. Similarly, removing Perlecan specifically from neurons, glia, or muscle does not cause synaptic retraction, indicating the protein is secreted from multiple cell types and functions non-cell autonomously. Within the peripheral nervous system, Perlecan predominantly localizes to the neural lamella, a specialized ECM surrounding nerve bundles. Indeed, the neural lamella is disrupted in the absence of Perlecan, with axons occasionally exiting their usual boundary in the nerve bundle. In addition, entire nerve bundles degenerate in a temporally coordinated manner across individual hemi-segments throughout larval development. These observations indicate disruption of neural lamella ECM function triggers axonal destabilization and synaptic retraction of motoneurons, revealing a role for Perlecan in axonal and synaptic integrity during nervous system development.


Asunto(s)
Proteínas de la Matriz Extracelular , Proteoglicanos de Heparán Sulfato , Animales , Axones/metabolismo , Drosophila/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo
11.
bioRxiv ; 2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36711745

RESUMEN

Although neuronal subtypes display unique synaptic organization and function, the underlying transcriptional differences that establish these features is poorly understood. To identify molecular pathways that contribute to synaptic diversity, single neuron PatchSeq RNA profiling was performed on Drosophila tonic and phasic glutamatergic motoneurons. Tonic motoneurons form weaker facilitating synapses onto single muscles, while phasic motoneurons form stronger depressing synapses onto multiple muscles. Super-resolution microscopy and in vivo imaging demonstrated synaptic active zones in phasic motoneurons are more compact and display enhanced Ca 2+ influx compared to their tonic counterparts. Genetic analysis identified unique synaptic properties that mapped onto gene expression differences for several cellular pathways, including distinct signaling ligands, post-translational modifications and intracellular Ca 2+ buffers. These findings provide insights into how unique transcriptomes drive functional and morphological differences between neuronal subtypes.

12.
Neuron ; 111(22): 3554-3569.e7, 2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37611584

RESUMEN

Although neuronal subtypes display unique synaptic organization and function, the underlying transcriptional differences that establish these features are poorly understood. To identify molecular pathways that contribute to synaptic diversity, single-neuron Patch-seq RNA profiling was performed on Drosophila tonic and phasic glutamatergic motoneurons. Tonic motoneurons form weaker facilitating synapses onto single muscles, while phasic motoneurons form stronger depressing synapses onto multiple muscles. Super-resolution microscopy and in vivo imaging demonstrated that synaptic active zones in phasic motoneurons are more compact and display enhanced Ca2+ influx compared with their tonic counterparts. Genetic analysis identified unique synaptic properties that mapped onto gene expression differences for several cellular pathways, including distinct signaling ligands, post-translational modifications, and intracellular Ca2+ buffers. These findings provide insights into how unique transcriptomes drive functional and morphological differences between neuronal subtypes.


Asunto(s)
Drosophila , Sinapsis , Animales , Sinapsis/fisiología , Neuronas Motoras/fisiología , Transducción de Señal
13.
Elife ; 102021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34713802

RESUMEN

Synaptic vesicle (SV) release probability (Pr) is a key presynaptic determinant of synaptic strength established by cell-intrinsic properties and further refined by plasticity. To characterize mechanisms that generate Pr heterogeneity between distinct neuronal populations, we examined glutamatergic tonic (Ib) and phasic (Is) motoneurons in Drosophila with stereotyped differences in Pr and synaptic plasticity. We found the decoy soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) Tomosyn is differentially expressed between these motoneuron subclasses and contributes to intrinsic differences in their synaptic output. Tomosyn expression enables tonic release in Ib motoneurons by reducing SNARE complex formation and suppressing Pr to generate decreased levels of SV fusion and enhanced resistance to synaptic fatigue. In contrast, phasic release dominates when Tomosyn expression is low, enabling high intrinsic Pr at Is terminals at the expense of sustained release and robust presynaptic potentiation. In addition, loss of Tomosyn disrupts the ability of tonic synapses to undergo presynaptic homeostatic potentiation.


Nerve cells transmit messages in the form of electrical and chemical signals. Electrical impulses travel along a neuron to the junction between two neighbouring cells, the synapse. There, chemical messengers called neurotransmitters are released from one cell and detected by the next, which can either excite or inhibit the recipient cell. Synapses differ in their ability to propagate signals and their signalling activity also fluctuates at times. Moreover, synaptic connections can be strengthened or weakened in a process called plasticity, which is a key part of learning new skills and recovering from a brain injury. It is thought that synaptic signalling might be amped up or dialled down to change the output of the connection between two cells, but exactly how this happens remains unclear. To investigate why synapses differ and how their signalling capabilities change, Sauvola et al. examined the connections between neurons and muscle cells in developing fruit flies. In fruit fly larvae, two types of neurons ­ called tonic Ib and phasic Is neurons ­ form synapses with muscle cells. But their synapses have different signalling properties: Ib synapses are weaker than Is synapses. Sauvola et al. hypothesised that a protein called Tomosyn ­ which is thought to restrict chemical signalling at the synapse ­ might be more active at weaker Ib synapses. Sauvola et al. found that Tomosyn was indeed more abundant at Ib synapses than at Is synapses, appearing to reflect their differences in signalling properties. In flies engineered to lack the Tomosyn protein, Ib synapses became four times stronger than usual, while Is synapses hardly changed. This supports the idea that Tomosyn restricts the release of neurotransmitters at typically weak Ib synapses. Further experiments showed Ib synapses in flies lacking Tomosyn also lost their malleability and ability to become strengthened during synaptic plasticity. Though the precise molecular interactions need further investigation, the findings suggest that Tomosyn is required for some forms of synaptic plasticity by controlling how much chemical signal neurons release. In summary, this work advances our understanding of synaptic signalling and brain plasticity, showing once again how the brain can change itself.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Homeostasis/genética , Plasticidad Neuronal , Proteínas SNARE/genética , Animales , Proteínas de Drosophila/metabolismo , Masculino , Proteínas SNARE/metabolismo
14.
Mol Cell Neurosci ; 40(2): 199-206, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19026748

RESUMEN

We combined recordings of spontaneous quantal events with electron microscopy analysis of synaptic ultrastructure to demonstrate that the size of a neurosecretory quantum increases following an activation of the endosomal endocytic pathway. We reversibly activated the endosomal endocytic pathway in Drosophila motor boutons by application of high K+ solution. This treatment produced the formation of numerous cisternae, vacuoles and enlarged vesicles. Spontaneous quantal events recorded immediately after the cessation of high K+ application were significantly enlarged, and this increase in quantal size was reversed after a 10 minute resting period. Actin depolymerization produced by latrunculin B pretreatment inhibited both the formation of endosome-like structures and the increase in quantal size. Loading the preparations with the dye FM1-43 followed by photoconversion of the dye combined with electron microscopy analysis revealed that the observed cisternae are likely to be the product of both bulk membrane retrieval and vesicle fusion.


Asunto(s)
Endocitosis/fisiología , Endosomas/metabolismo , Transmisión Sináptica/fisiología , Animales , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/fisiología , Endosomas/ultraestructura , Potasio/metabolismo , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Tiazolidinas/farmacología
15.
Elife ; 92020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32343229

RESUMEN

Synchronous neurotransmitter release is triggered by Ca2+ binding to the synaptic vesicle protein Synaptotagmin 1, while asynchronous fusion and short-term facilitation is hypothesized to be mediated by plasma membrane-localized Synaptotagmin 7 (SYT7). We generated mutations in Drosophila Syt7 to determine if it plays a conserved role as the Ca2+ sensor for these processes. Electrophysiology and quantal imaging revealed evoked release was elevated 2-fold. Syt7 mutants also had a larger pool of readily-releasable vesicles, faster recovery following stimulation, and intact facilitation. Syt1/Syt7 double mutants displayed more release than Syt1 mutants alone, indicating SYT7 does not mediate the residual asynchronous release remaining in the absence of SYT1. SYT7 localizes to an internal membrane tubular network within the peri-active zone, but does not enrich at active zones. These findings indicate the two Ca2+ sensor model of SYT1 and SYT7 mediating all phases of neurotransmitter release and facilitation is not applicable at Drosophila synapses.


Asunto(s)
Sinapsis/fisiología , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Sinaptotagminas/metabolismo , Animales , Calcio/metabolismo , Membrana Celular/metabolismo , Drosophila/metabolismo , Exocitosis/fisiología , Neurotransmisores/metabolismo
16.
Elife ; 72018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29989549

RESUMEN

Neurons communicate through neurotransmitter release at specialized synaptic regions known as active zones (AZs). Using biosensors to visualize single synaptic vesicle fusion events at Drosophila neuromuscular junctions, we analyzed the developmental and molecular determinants of release probability (Pr) for a defined connection with ~300 AZs. Pr was heterogeneous but represented a stable feature of each AZ. Pr remained stable during high frequency stimulation and retained heterogeneity in mutants lacking the Ca2+ sensor Synaptotagmin 1. Pr correlated with both presynaptic Ca2+ channel abundance and Ca2+ influx at individual release sites. Pr heterogeneity also correlated with glutamate receptor abundance, with high Pr connections developing receptor subtype segregation. Intravital imaging throughout development revealed that AZs acquire high Pr during a multi-day maturation period, with Pr heterogeneity largely reflecting AZ age. The rate of synapse maturation was activity-dependent, as both increases and decreases in neuronal activity modulated glutamate receptor field size and segregation.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Neurotransmisores/metabolismo , Receptores Ionotrópicos de Glutamato/metabolismo , Sinapsis/fisiología , Vesículas Sinápticas/metabolismo , Sinaptotagmina I/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/genética , Exocitosis , Femenino , Masculino , Mutación , Neuronas/citología , Neuronas/metabolismo , Terminales Presinápticos/fisiología , Receptores Ionotrópicos de Glutamato/genética , Transmisión Sináptica , Sinaptotagmina I/genética
17.
Brain Res ; 1178: 52-64, 2007 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-17904536

RESUMEN

We employed optical detection of the lipophylic dye FM1-43 and focal recordings of quantal release to investigate how synapsin affects vesicle cycling at the neuromuscular junction of synapsin knockout (Syn KO) Drosophila. Loading the dye employing high K+ stimulation, which presumably involves the recycling pool of vesicles in exo/endocytosis, stained the periphery of wild type (WT) boutons, while in Syn KO the dye was redistributed towards the center of the bouton. When endocytosis was promoted by cyclosporin A pretreatment, the dye uptake was significantly enhanced in WT boutons, and the entire boutons were stained, suggesting staining of the reserve vesicle pool. In Syn KO boutons, the same loading paradigm produced fainter staining and significantly faster destaining. When the axon was stimulated electrically, a distinct difference in dye loading patterns was observed in WT boutons at different stimulation frequencies: a low stimulation frequency (3 Hz) produced a ring-shaped staining pattern, while at a higher frequency (10 Hz) the dye was redistributed towards the center of the bouton and the fluorescence intensity was significantly increased. This difference in staining patterns was essentially disrupted in Syn KO boutons, although synapsin did not affect the rate of quantal release. Stimulation of the nerve in the presence of bafilomycin, the blocker of the transmitter uptake, produced significantly stronger depression in Syn KO boutons. These results, taken together, suggest that synapsin maintains the reserve pool of vesicles and segregation between the recycling and reserve pools, and that it mediates mobilization of the reserve pool during intense stimulation.


Asunto(s)
Drosophila/fisiología , Terminales Presinápticos/fisiología , Receptores Presinapticos/fisiología , Sinapsinas/fisiología , Vesículas Sinápticas/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Estimulación Eléctrica , Colorantes Fluorescentes , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Larva , Macrólidos/farmacología , Microscopía Confocal , Terminales Presinápticos/ultraestructura , Receptores Presinapticos/ultraestructura , Sinapsinas/genética , Vesículas Sinápticas/ultraestructura
18.
Elife ; 62017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28895532

RESUMEN

The synaptic vesicle Ca2+ sensor Synaptotagmin binds Ca2+ through its two C2 domains to trigger membrane interactions. Beyond membrane insertion by the C2 domains, other requirements for Synaptotagmin activity are still being elucidated. To identify key residues within Synaptotagmin required for vesicle cycling, we took advantage of observations that mutations in the C2B domain Ca2+-binding pocket dominantly disrupt release from invertebrates to humans. We performed an intragenic screen for suppressors of lethality induced by expression of Synaptotagmin C2B Ca2+-binding mutants in Drosophila. This screen uncovered essential residues within Synaptotagmin that suggest a structural basis for several activities required for fusion, including a C2B surface implicated in SNARE complex interaction that is required for rapid synchronization and Ca2+ cooperativity of vesicle release. Using electrophysiological, morphological and computational characterization of these mutants, we propose a sequence of molecular interactions mediated by Synaptotagmin that promote Ca2+ activation of the synaptic vesicle fusion machinery.


Asunto(s)
Calcio/metabolismo , Vesículas Citoplasmáticas/metabolismo , Fusión de Membrana , Proteínas SNARE/metabolismo , Sinaptotagminas/metabolismo , Animales , Drosophila , Sinaptotagminas/genética
19.
Neuron ; 88(4): 749-61, 2015 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-26590346

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Drosophila/genética , Proteínas del Tejido Nervioso/genética , Unión Neuromuscular/metabolismo , Plasticidad Neuronal/genética , Transmisión Sináptica/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Secuencia de Bases , Calcio/metabolismo , Drosophila , Proteínas de Drosophila/metabolismo , Exocitosis/genética , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/metabolismo , Neurotransmisores/metabolismo , Fosforilación , Proteínas SNARE/metabolismo
20.
J Cell Biol ; 193(1): 201-17, 2011 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-21464232

RESUMEN

Structural remodeling of synapses in response to growth signals leads to long-lasting alterations in neuronal function in many systems. Synaptic growth factor receptors alter their signaling properties during transit through the endocytic pathway, but the mechanisms controlling cargo traffic between endocytic compartments remain unclear. Nwk (Nervous Wreck) is a presynaptic F-BAR/SH3 protein that regulates synaptic growth signaling in Drosophila melanogaster. In this paper, we show that Nwk acts through a physical interaction with sorting nexin 16 (SNX16). SNX16 promotes synaptic growth signaling by activated bone morphogenic protein receptors, and live imaging in neurons reveals that SNX16-positive early endosomes undergo transient interactions with Nwk-containing recycling endosomes. We identify an alternative signal termination pathway in the absence of Snx16 that is controlled by endosomal sorting complex required for transport (ESCRT)-mediated internalization of receptors into the endosomal lumen. Our results define a presynaptic trafficking pathway mediated by SNX16, NWK, and the ESCRT complex that functions to control synaptic growth signaling at the interface between endosomal compartments.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/metabolismo , Endosomas/metabolismo , Transducción de Señal , Vesículas Sinápticas/metabolismo , Animales , Células Cultivadas , Proteínas de Drosophila/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nexinas de Clasificación/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA