Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.040
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Eur J Neurosci ; 59(10): 2483-2501, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38532289

RESUMO

Dopaminergic neurons of the substantia nigra exist in a persistent state of vulnerability resulting from high baseline oxidative stress, high-energy demand, and broad unmyelinated axonal arborisations. Impairments in the storage of dopamine compound this stress because of cytosolic reactions that transform the vital neurotransmitter into an endogenous neurotoxicant, and this toxicity is thought to contribute to the dopamine neuron degeneration that occurs Parkinson's disease. We have previously identified synaptic vesicle glycoprotein 2C (SV2C) as a modifier of vesicular dopamine function, demonstrating that genetic ablation of SV2C in mice results in decreased dopamine content and evoked dopamine release in the striatum. Here, we adapted a previously published in vitro assay utilising false fluorescent neurotransmitter 206 (FFN206) to visualise how SV2C regulates vesicular dopamine dynamics and determined that SV2C promotes the uptake and retention of FFN206 within vesicles. In addition, we present data indicating that SV2C enhances the retention of dopamine in the vesicular compartment with radiolabelled dopamine in vesicles isolated from immortalised cells and from mouse brain. Further, we demonstrate that SV2C enhances the ability of vesicles to store the neurotoxicant 1-methyl-4-phenylpyridinium (MPP+) and that genetic ablation of SV2C results in enhanced 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced vulnerability in mice. Together, these findings suggest that SV2C functions to enhance vesicular storage of dopamine and neurotoxicants and helps maintain the integrity of dopaminergic neurons.


Assuntos
Dopamina , Neurônios Dopaminérgicos , Glicoproteínas de Membrana , Proteínas do Tecido Nervoso , Vesículas Sinápticas , Animais , Humanos , Camundongos , Corpo Estriado/metabolismo , Corpo Estriado/efeitos dos fármacos , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/efeitos dos fármacos
2.
PLoS Biol ; 19(6): e3001149, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34153028

RESUMO

Synaptic plasticity is a cellular model for learning and memory. However, the expression mechanisms underlying presynaptic forms of plasticity are not well understood. Here, we investigate functional and structural correlates of presynaptic potentiation at large hippocampal mossy fiber boutons induced by the adenylyl cyclase activator forskolin. We performed 2-photon imaging of the genetically encoded glutamate sensor iGluu that revealed an increase in the surface area used for glutamate release at potentiated terminals. Time-gated stimulated emission depletion microscopy revealed no change in the coupling distance between P/Q-type calcium channels and release sites mapped by Munc13-1 cluster position. Finally, by high-pressure freezing and transmission electron microscopy analysis, we found a fast remodeling of synaptic ultrastructure at potentiated boutons: Synaptic vesicles dispersed in the terminal and accumulated at the active zones, while active zone density and synaptic complexity increased. We suggest that these rapid and early structural rearrangements might enable long-term increase in synaptic strength.


Assuntos
Fibras Musgosas Hipocampais/metabolismo , Terminações Pré-Sinápticas/metabolismo , Animais , Colforsina/farmacologia , Ácido Glutâmico/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência por Excitação Multifotônica , Fibras Musgosas Hipocampais/efeitos dos fármacos , Fibras Musgosas Hipocampais/ultraestrutura , Neurotransmissores/metabolismo , Terminações Pré-Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo
3.
Cell Mol Life Sci ; 78(11): 4973-4992, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33864480

RESUMO

Amyloid beta (Aß) is linked to the pathology of Alzheimer's disease (AD). At physiological concentrations, Aß was proposed to enhance neuroplasticity and memory formation by increasing the neurotransmitter release from presynapse. However, the exact mechanisms underlying this presynaptic effect as well as specific contribution of endogenously occurring Aß isoforms remain unclear. Here, we demonstrate that Aß1-42 and Aß1-16, but not Aß17-42, increased size of the recycling pool of synaptic vesicles (SV). This presynaptic effect was driven by enhancement of endogenous cholinergic signalling via α7 nicotinic acetylcholine receptors, which led to activation of calcineurin, dephosphorylation of synapsin 1 and consequently resulted in reorganization of functional pools of SV increasing their availability for sustained neurotransmission. Our results identify synapsin 1 as a molecular target of Aß and reveal an effect of physiological concentrations of Aß on cholinergic modulation of glutamatergic neurotransmission. These findings provide new mechanistic insights in cholinergic dysfunction observed in AD.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Fragmentos de Peptídeos/farmacologia , Sinapses/metabolismo , Sinapsinas/metabolismo , Vesículas Sinápticas/efeitos dos fármacos , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Cálcio/metabolismo , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurotransmissores/metabolismo , Nicotina/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Vesículas Sinápticas/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/deficiência , Receptor Nicotínico de Acetilcolina alfa7/genética
4.
J Neurochem ; 157(4): 1052-1068, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33341946

RESUMO

The Rho kinase (ROCK) signaling pathway is an attractive therapeutic target in neurodegeneration since it has been linked to the prevention of neuronal death and neurite regeneration. The isoquinoline derivative fasudil is a potent ROCK inhibitor, which is already approved for chronic clinical treatment in humans. However, the effects of chronic fasudil treatments on neuronal function are still unknown. We analyzed here chronic fasudil treatment in primary rat hippocampal cultures. Neurons were stimulated with 20 Hz field stimulation and we investigated pre-synaptic mechanisms and parameters regulating synaptic transmission after fasudil treatment by super resolution stimulated emission depletion (STED) microscopy, live-cell fluorescence imaging, and western blotting. Fasudil did not affect basic synaptic function or the amount of several synaptic proteins, but it altered the chronic dynamics of the synaptic vesicles. Fasudil reduced the proportion of the actively recycling vesicles, and shortened the vesicle lifetime, resulting overall in a reduction of the synaptic response upon stimulation. We conclude that fasudil does not alter synaptic structure, accelerates vesicle turnover, and decreases the number of released vesicles. This broadens the known spectrum of effects of this drug, and suggests new potential clinical uses.


Assuntos
1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/análogos & derivados , Neurônios/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Feminino , Hipocampo/efeitos dos fármacos , Masculino , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos , Ratos Wistar , Quinases Associadas a rho/antagonistas & inibidores
5.
Neurobiol Learn Mem ; 183: 107480, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34153453

RESUMO

Perturbations in the glutamate-glutamine cycle and glutamate release from presynaptic terminals have been involved in the development of cognitive deficits in Alzheimer's disease (AD) patients and mouse models. Glutamate transporter-1 (GLT-1) removes glutamate from the synaptic cleft and transports it into astrocytes, where it is used as substrate for the glutamate-glutamine cycle. Ceftriaxone has been reported to improve cognitive deficits in AD mice by increasing GLT-1 expression, glutamate transformation to glutamine, and glutamine efflux from astrocytes. However, the impact of ceftriaxone on glutamine metabolism in neurons is unknown. The present study aimed to investigate whether ceftriaxone regulated the production and vesicular assembly of glutamate in the presynaptic terminals of neurons and to determine GLT-1 involvement in this process. We used the amyloid precursor protein (APP)/presenilin-1 (PS1) AD mouse model and GLT-1 knockdown APP/PS1 (GLT-1+/-/APP/PS1) mice. The expression levels of sodium-coupled neutral amino-acid transporter 1 (SNAT1) and vesicular glutamate transporters 1 and 2 (VGLUT1/2) were analyzed by immunofluorescence and immunohistochemistry staining as well as by Western blotting. Glutaminase activity was assayed by fluorometry. Ceftriaxone treatment significantly increased SNAT1 expression and glutaminase activity in neurons in APP/PS1 mice. Similarly, VGLUT1/2 levels were increased in the presynaptic terminals of APP/PS1 mice treated with ceftriaxone. The deletion of one GLT-1 allele in APP/PS1 mice prevented the ceftriaxone-induced upregulation of SNAT1 and VGLUT1/2 expression, indicating that GLT-1 played an important role in ceftriaxone effect. Based on the role of SNAT1, glutaminase, and VGLUT1/2 in the glutamate-glutamine cycle in neurons, the present results suggested that ceftriaxone improved the production and vesicular assembly of glutamate as a neurotransmitter in presynaptic terminals by acting on GLT-1 in APP/PS1 mice.


Assuntos
Doença de Alzheimer/metabolismo , Antibacterianos/farmacologia , Ceftriaxona/farmacologia , Transportador 2 de Aminoácido Excitatório/efeitos dos fármacos , Terminações Pré-Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Doença de Alzheimer/genética , Sistema A de Transporte de Aminoácidos/efeitos dos fármacos , Sistema A de Transporte de Aminoácidos/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Modelos Animais de Doenças , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/metabolismo , Técnicas de Silenciamento de Genes , Ácido Glutâmico/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Glutaminase/efeitos dos fármacos , Glutaminase/metabolismo , Camundongos , Camundongos Transgênicos , Presenilina-1/genética , Terminações Pré-Sinápticas/metabolismo , Vesículas Sinápticas/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/efeitos dos fármacos , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/efeitos dos fármacos , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
6.
Cereb Cortex ; 30(3): 1422-1435, 2020 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-31504271

RESUMO

While a lot is known about classical, anterograde neurotransmission, less is known about the mechanisms and molecules involved in retrograde neurotransmission. Our hypothesis is that N-acetylaspartylglutamate (NAAG), the most abundant dipeptide in the brain, may act as a retrograde transmitter in the brain. NAAG was predominantly localized in dendritic compartments of glutamatergic synapses in the intact hippocampus, where it was present in close proximity to synaptic-like vesicles. In acute hippocampal slices, NAAG was depleted from postsynaptic dendritic elements during neuronal stimulation induced by depolarizing concentrations of potassium or by exposure to glutamate receptor (GluR) agonists. The depletion was completely blocked by botulinum toxin B and strictly dependent on extracellular calcium, indicating exocytotic release. In contrast, there were low levels of NAAG and no effect by depolarization or GluR agonists in presynaptic glutamatergic terminals or GABAergic pre- and postsynaptic elements. Together these data suggest a possible role for NAAG as a retrograde signaling molecule at glutamatergic synapses via exocytotic release.


Assuntos
Dipeptídeos/farmacologia , Exocitose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Agonistas de Aminoácidos Excitatórios/farmacologia , Exocitose/fisiologia , Hipocampo/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Terminações Pré-Sinápticas/efeitos dos fármacos , Ratos Wistar , Vesículas Sinápticas/efeitos dos fármacos
7.
Mol Cell Neurosci ; 107: 103528, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32650044

RESUMO

Multiple C2 and Transmembrane Domain Proteins (MCTPs) are putative calcium sensors. Proteins that contain C2 domains play essential roles in membrane trafficking and exocytosis; however, MCTPs functions in neurotransmitter release are not known. Here we report that in C. elegans mctp-1 is under the control of two promoters - one active in the nervous system and the second in the spermatheca. We generated and characterized a loss of function amt1 mutant and compared it to a previously published loss of function mutant (av112). Loss of mctp-1 function causes defects in egg-laying, crawling velocity, and thrashing rates. Both amt1 and av112 mutants are hyposensitive to the acetylcholinesterase blocker aldicarb, suggesting that MCTP-1 may play a role in synaptic vesicle release.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/análogos & derivados , Exocitose/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Cálcio/metabolismo , Exocitose/fisiologia , Proteínas de Membrana/metabolismo , Neurotransmissores/farmacologia , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/metabolismo
8.
Hum Mutat ; 41(11): 1999-2011, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32906212

RESUMO

Clinical and genetic features of five unrelated patients with de novo pathogenic variants in the synaptic vesicle-associated membrane protein 2 (VAMP2) reveal common features of global developmental delay, autistic tendencies, behavioral disturbances, and a higher propensity to develop epilepsy. For one patient, a cognitively impaired adolescent with a de novo stop-gain VAMP2 mutation, we tested a potential treatment strategy, enhancing neurotransmission by prolonging action potentials with the aminopyridine family of potassium channel blockers, 4-aminopyridine and 3,4-diaminopyridine, in vitro and in vivo. Synaptic vesicle recycling and neurotransmission were assayed in neurons expressing three VAMP2 variants by live-cell imaging and electrophysiology. In cellular models, two variants decrease both the rate of exocytosis and the number of synaptic vesicles released from the recycling pool, compared with wild-type. Aminopyridine treatment increases the rate and extent of exocytosis and total synaptic charge transfer and desynchronizes GABA release. The clinical response of the patient to 2 years of off-label aminopyridine treatment includes improved emotional and behavioral regulation by parental report, and objective improvement in standardized cognitive measures. Aminopyridine treatment may extend to patients with pathogenic variants in VAMP2 and other genes influencing presynaptic function or GABAergic tone, and tested in vitro before treatment.


Assuntos
4-Aminopiridina/farmacologia , Mutação/genética , Proteína 2 Associada à Membrana da Vesícula/genética , Adulto , Eletrofisiologia , Exocitose/efeitos dos fármacos , Feminino , Humanos , Masculino , Transmissão Sináptica/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo
9.
Neurobiol Dis ; 143: 104977, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32553709

RESUMO

Environmental toxicants have the potential to contribute to the pathophysiology of multiple complex diseases, but the underlying mechanisms remain obscure. One such toxicant is the widely used fungicide ziram, a dithiocarbamate known to have neurotoxic effects and to increase the risk of Parkinson's disease. We have used Drosophila melanogaster as an unbiased discovery tool to identify novel molecular pathways by which ziram may disrupt neuronal function. Consistent with previous results in mammalian cells, we find that ziram increases the probability of synaptic vesicle release by dysregulation of the ubiquitin signaling system. In addition, we find that ziram increases neuronal excitability. Using a combination of live imaging and electrophysiology, we find that ziram increases excitability in both aminergic and glutamatergic neurons. This increased excitability is phenocopied and occluded by null mutant animals of the ether a-go-go (eag) potassium channel. A pharmacological inhibitor of the temperature sensitive hERG (human ether-a-go-go related gene) phenocopies the excitability effects of ziram but only at elevated temperatures. seizure (sei), a fly ortholog of hERG, is thus another candidate target of ziram. Taken together, the eag family of potassium channels emerges as a candidate for mediating some of the toxic effects of ziram. We propose that ziram may contribute to the risk of complex human diseases by blockade of human eag and sei orthologs, such as hERG.


Assuntos
Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Fungicidas Industriais/toxicidade , Neurônios/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos , Ziram/toxicidade , Animais , Drosophila melanogaster , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Neurônios/metabolismo , Neurotransmissores/metabolismo , Vesículas Sinápticas/metabolismo
10.
Neurochem Res ; 45(1): 109-121, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30793220

RESUMO

The finding that ketamine, an anaesthetic, can elicit a rapid antidepressant effect at low doses that lasts for weeks in patients with depression is arguably a major achievement in psychiatry in the last decades. However, the mechanisms of action are unclear. The glutamatergic hypothesis of ketamine action posits that ketamine is a N-methyl-D-aspartate receptor (NMDAR) antagonist modulating downstream cytoplasmic events in neurons. In addition to targeting NMDARs in synaptic transmission, ketamine may modulate the function of astroglia, key homeostasis-providing cells in the central nervous system, also playing a role in many neurologic diseases including depression, which affects to 20% of the population globally. We first review studies on astroglia revealing that (sub)anaesthetic doses of ketamine attenuate stimulus-evoked calcium signalling, a process of astroglial cytoplasmic excitability, regulating the exocytotic release of gliosignalling molecules. Then we address how ketamine alters the fusion pore activity of secretory vesicles, and how ketamine affects extracellular glutamate and K+ homeostasis, both considered pivotal in depression. Finally, we also provide evidence indicating reduced cytoplasmic mobility of astroglial vesicles carrying the inward rectifying potassium channel (Kir4.1), which may regulate the density of Kir4.1 at the plasma membrane. These results indicate that the astroglial capacity to control extracellular K+ concentration may be altered by ketamine and thus indirectly affect the action potential firing of neurons, as is the case in lateral habenula in a rat disease model of depression. Hence, ketamine-altered functions of astroglia extend beyond neuronal NMDAR antagonism and provide a basis for its antidepressant action through glia.


Assuntos
Astrócitos/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Humanos , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Vesículas Sinápticas/efeitos dos fármacos
11.
Molecules ; 25(7)2020 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-32276517

RESUMO

Oxidative stress triggers a lethal cascade, leading to Parkinson's disease by causing degeneration of dopaminergic neurons. In this study, eight antioxidants were screened for their neuroprotective effect on PC12 cells (pheochromocytoma cell line) under oxidative stress induced by salsolinol (OSibS). Hydroxytyrosol was found to be the strongest neuroprotective agent; it improved viability of PC12 cells by up to 81.69% under OSibS. Afterward, two synaptic vesicle proteins, synapsin-1 and septin-5, were screened for their neuroprotective role; the overexpression of synapsin-1 and the downregulation of septin-5 separately improved the viability of PC12 cells by up to 71.17% and 67.00%, respectively, compared to PC12 cells only treated with salsolinol (PoTwS) under OSibS. Subsequently, the PC12+syn++sep- cell line was constructed and pretreated with 100 µM hydroxytyrosol, which improved its cell viability by up to 99.03% and led to 14.71- and 6.37-fold reductions in the levels of MDA and H2O2, respectively, and 6.8-, 12.97-, 10.57-, and 7.57-fold increases in the activity of catalase, glutathione reductase, superoxide dismutase, and glutathione peroxidase, respectively, compared to PoTwS under OSibS. Finally, alcohol dehydrogenase-6 from Saccharomyces cerevisiae was expressed in PC12+syn++sep- cells to convert 3,4-dihydroxyphenylacetaldehyde (an endogenous neurotoxin) into hydroxytyrosol. The PC12+syn++sep-+ADH6+ cell line also led to 22.38- and 12.33-fold decreases in the production of MDA and H2O2, respectively, and 7.15-, 13.93-, 12.08-, and 8.11-fold improvements in the activity of catalase, glutathione reductase, superoxide dismutase, and glutathione peroxidase, respectively, compared to PoTwS under OSibS. Herein, we report the endogenous production of a powerful antioxidant, hydroxytyrosol, from 3,4-dihydroxyphenylacetaldehyde, and evaluate its synergistic neuroprotective effect, along with synapsin-1 and septin-5, on PC12 cells under OSibS.


Assuntos
Neoplasias das Glândulas Suprarrenais/patologia , Isoquinolinas/toxicidade , Fármacos Neuroprotetores/farmacologia , Álcool Feniletílico/análogos & derivados , Feocromocitoma/patologia , Vesículas Sinápticas/metabolismo , Animais , Catalase/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Dopamina/metabolismo , Sinergismo Farmacológico , Flavonoides/farmacologia , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Malondialdeído/metabolismo , Metaboloma , Células PC12 , Álcool Feniletílico/farmacologia , Ratos , Superóxido Dismutase/metabolismo , Vesículas Sinápticas/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
12.
J Neurosci ; 38(7): 1725-1736, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29326174

RESUMO

Block of neurotransmitter receptors at the neuromuscular junction (NMJ) has been shown to trigger upregulation of the number of synaptic vesicles released (quantal content, QC), a response termed homeostatic synaptic plasticity. The mechanism underlying this plasticity is not known. Here, we used selective toxins to demonstrate that block of α1-containing nicotinic acetylcholine receptors (nAChRs) at the NMJ of male and female mice triggers the upregulation of QC. Reduction of current flow through nAChRs, induced by drugs with antagonist activity, demonstrated that reduction in synaptic current per se does not trigger upregulation of QC. These data led to the remarkable conclusion that disruption of synaptic transmission is not sensed to trigger upregulation of QC. During studies of the effect of partial block of nAChRs on QC, we observed a small but reproducible increase in the decay kinetics of miniature synaptic currents. The change in kinetics was correlated with the increase in QC and raises the possibility that a change in postsynaptic nAChR conformation may be associated with the presynaptic increase in QC. We propose that, in addition to functioning in synaptic transmission, ionotropic muscle nicotonic nAChRs may serve as signaling molecules that participate in synaptic plasticity. Because nAChRs have been implicated in a number of disease states, the finding that nAChRs may be involved in triggering synaptic plasticity could have wide-reaching implications.SIGNIFICANCE STATEMENT The signals that initiate synaptic plasticity of the nervous system are still incompletely understood. Using the mouse neuromuscular junction as a model synapse, we studied how block of neurotransmitter receptors is sensed to trigger synaptic plasticity. Our studies led to the surprising conclusion that neither changes in synaptic current nor spiking of the presynaptic or postsynaptic cell are sensed to initiate synaptic plasticity. Instead, postsynaptic nicotinic acetylcholine receptors (nAChRs), in addition to functioning in synaptic transmission, may serve as signaling molecules that trigger synaptic plasticity. Because nAChRs have been implicated in a number of disease states, the finding that they may mediate synaptic plasticity has broad implications.


Assuntos
Músculo Esquelético/efeitos dos fármacos , Junção Neuromuscular/efeitos dos fármacos , Neurotoxinas/farmacologia , Antagonistas Nicotínicos/farmacologia , Receptores Nicotínicos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Feminino , Cinética , Masculino , Camundongos , Potenciais Sinápticos/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Vesículas Sinápticas/efeitos dos fármacos
13.
Neurochem Res ; 44(3): 683-691, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29052089

RESUMO

Abnormal processing of amyloid precursor protein (APP) and aggregation of the Aß peptide are known to play a key role in the pathogenesis of Alzheimer disease, but the function of endogenous APP under normal physiological conditions remains poorly understood. In this study, we investigated presynaptic changes in APP knockout (KO) mice. We demonstrate that both sucrose-induced neurotransmission and synaptic depletion in response to high frequency stimulation are significantly enhanced in APP KO compared to wild type littermates. In addition, the level of phosphorylated forms of synapsins, but not total synapsins, is elevated in the KO mice. Furthermore, we show that the inhibition of L-type calcium channels normalizes phosphorylated synapsins and slows down the high frequency induced synaptic depletion in APP KO mice. These results suggest a new mechanism by which APP regulates synaptic vesicle dynamics through synapsin-dependent phosphorylation.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Fosforilação/fisiologia , Sinapsinas/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Camundongos Knockout , Neurotransmissores/farmacologia , Sinapsinas/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/efeitos dos fármacos
14.
Br J Anaesth ; 123(2): 219-227, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31056238

RESUMO

BACKGROUND: P/Q- and N-type voltage-gated calcium channels (VGCC) are the principal subtypes mediating synaptic vesicle (SV) exocytosis. Both the degree of isoflurane inhibition of SV exocytosis and VGCC subtype expression vary between brain regions and neurotransmitter phenotype. We hypothesised that differences in VGCC subtype expression contribute to synapse-selective presynaptic effects of isoflurane. METHODS: We used quantitative live-cell imaging to measure exocytosis in cultured rat hippocampal neurones after transfection of the fluorescent biosensor vGlut1-pHluorin. Selective inhibitors of P/Q- and N-type VGCCs were used to isolate subtype-specific effects of isoflurane. RESULTS: Inhibition of N-type channels by 1 µM ω-conotoxin GVIA reduced SV exocytosis to 81±5% of control (n=10). Residual exocytosis mediated by P/Q-type channels was further inhibited by isoflurane to 42±4% of control (n=10). The P/Q-type channel inhibitor ω-agatoxin IVA at 0.4 µM inhibited SV exocytosis to 29±3% of control (n=10). Residual exocytosis mediated by N-type channels was further inhibited by isoflurane to 17±3% of control (n=10). Analysis of isoflurane effects at the level of individual boutons revealed no difference in sensitivity to isoflurane between P/Q- or N-type channel-mediated SV exocytosis (P=0.35). There was no correlation between the effect of agatoxin (P=0.91) or conotoxin (P=0.15) and the effect of isoflurane on exocytosis. CONCLUSIONS: Sensitivity of SV exocytosis to isoflurane in rat hippocampal neurones is independent of the specific VGCC subtype coupled to exocytosis. The differential sensitivity of VGCC subtypes to isoflurane does not explain the observed neurotransmitter-selective effects of isoflurane in hippocampus.


Assuntos
Anestésicos Inalatórios/farmacologia , Canais de Cálcio/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Isoflurano/farmacologia , Vesículas Sinápticas/efeitos dos fármacos , Animais , Células Cultivadas , Técnicas In Vitro , Modelos Animais , Neurônios/efeitos dos fármacos , Ratos
15.
Cereb Cortex ; 28(4): 1087-1104, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28158493

RESUMO

Dendritic spines compartmentalize information in the brain, and their morphological characteristics are thought to underly synaptic plasticity. Here we identify copine-6 as a novel modulator of dendritic spine morphology. We found that brain-derived neurotrophic factor (BDNF) - a molecule essential for long-term potentiation of synaptic strength - upregulated and recruited copine-6 to dendritic spines in hippocampal neurons. Overexpression of copine-6 increased mushroom spine number and decreased filopodia number, while copine-6 knockdown had the opposite effect and dramatically increased the number of filopodia, which lacked PSD95. Functionally, manipulation of post-synaptic copine-6 levels affected miniature excitatory post-synaptic current (mEPSC) kinetics and evoked synaptic vesicle recycling in contacting boutons, and post-synaptic knockdown of copine-6 reduced hippocampal LTP and increased LTD. Mechanistically, copine-6 promotes BDNF-TrkB signaling and recycling of activated TrkB receptors back to the plasma membrane surface, and is necessary for BDNF-induced increases in mushroom spines in hippocampal neurons. Thus copine-6 regulates BDNF-dependent changes in dendritic spine morphology to promote synaptic plasticity.


Assuntos
Proteínas de Transporte/metabolismo , Espinhas Dendríticas/fisiologia , Hipocampo/citologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Neurônios/ultraestrutura , Vesículas Sinápticas/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Proteínas de Transporte/genética , Células Cultivadas , Espinhas Dendríticas/ultraestrutura , Proteína 4 Homóloga a Disks-Large/metabolismo , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Técnicas de Cultura de Órgãos , Ratos , Receptor trkB/genética , Receptor trkB/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Sinapses/ultraestrutura , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/genética , Vesículas Sinápticas/efeitos dos fármacos , Sinaptossomos/metabolismo , Sinaptossomos/ultraestrutura , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo
16.
Mol Cell Proteomics ; 16(8): 1394-1415, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28659490

RESUMO

The therapeutic use of RhoA inhibitors (RhoAi) has been experimentally tested in spinal cord injury (SCI). In order to decipher the underlying molecular mechanisms involved in such a process, an in vitro neuroproteomic-systems biology platform was developed in which the pan-proteomic profile of the dorsal root ganglia (DRG) cell line ND7/23 DRG was assessed in a large array of culture conditions using RhoAi and/or conditioned media obtained from SCI ex vivo derived spinal cord slices. A fine mapping of the spatio-temporal molecular events of the RhoAi treatment in SCI was performed. The data obtained allow a better understanding of regeneration/degeneration induced above and below the lesion site. Results notably showed a time-dependent alteration of the transcription factors profile along with the synthesis of growth cone-related factors (receptors, ligands, and signaling pathways) in RhoAi treated DRG cells. Furthermore, we assessed in a rat SCI model the in vivo impact of RhoAi treatment administered in situ via alginate scaffold that was combined with FK506 delivery. The improved recovery of locomotion was detected only at the early postinjury time points, whereas after overall survival a dramatic increase of synaptic contacts on outgrowing neurites in affected segments was observed. We validate these results by in vivo proteomic studies along the spinal cord segments from tissue and secreted media analyses, confirming the increase of the synaptogenesis expression factors under RhoAi treatment. Taken together, we demonstrate that RhoAi treatment seems to be useful to stimulate neurite outgrowth in both in vitro as well in vivo environments. However, for in vivo experiments there is a need for sustained delivery regiment to facilitate axon regeneration and promote synaptic reconnections with appropriate target neurons also at chronic phase, which in turn may lead to higher assumption for functional improvement.


Assuntos
Axônios/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Crescimento Neuronal/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Vesículas Sinápticas/efeitos dos fármacos , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Análise de Variância , Animais , Axônios/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/uso terapêutico , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiopatologia , Locomoção/efeitos dos fármacos , Crescimento Neuronal/fisiologia , Proteômica , Ratos , Regeneração/efeitos dos fármacos , Traumatismos da Medula Espinal/fisiopatologia , Vesículas Sinápticas/fisiologia , Tacrolimo , Fatores de Tempo , Fatores de Transcrição/metabolismo
17.
J Neurosci ; 37(25): 6043-6052, 2017 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-28576942

RESUMO

α-Synuclein is a presynaptic protein the function of which has yet to be identified, but its neuronal content increases in patients of synucleinopathies including Parkinson's disease. Chronic overexpression of α-synuclein reportedly expresses various phenotypes of synaptic dysfunction, but the primary target of its toxicity has not been determined. To investigate this, we acutely loaded human recombinant α-synuclein or its pathological mutants in their monomeric forms into the calyces of Held presynaptic terminals in slices from auditorily mature and immature rats of either sex. Membrane capacitance measurements revealed significant and specific inhibitory effects of WT monomeric α-synuclein on vesicle endocytosis throughout development. However, the α-synuclein A53T mutant affected vesicle endocytosis only at immature calyces, whereas the A30P mutant had no effect throughout. The endocytic impairment by WT α-synuclein was rescued by intraterminal coloading of the microtubule (MT) polymerization blocker nocodazole. Furthermore, it was reversibly rescued by presynaptically loaded photostatin-1, a photoswitcheable inhibitor of MT polymerization, in a light-wavelength-dependent manner. In contrast, endocytic inhibition by the A53T mutant at immature calyces was not rescued by nocodazole. Functionally, presynaptically loaded WT α-synuclein had no effect on basal synaptic transmission evoked at a low frequency, but significantly attenuated exocytosis and impaired the fidelity of neurotransmission during prolonged high-frequency stimulation. We conclude that monomeric WT α-synuclein primarily inhibits vesicle endocytosis via MT overassembly, thereby impairing high-frequency neurotransmission.SIGNIFICANCE STATEMENT Abnormal α-synuclein abundance is associated with synucleinopathies including Parkinson's disease, but neither the primary target of α-synuclein toxicity nor its mechanism is identified. Here, we loaded monomeric α-synuclein directly into mammalian glutamatergic nerve terminals and found that it primarily inhibits vesicle endocytosis and subsequently impairs exocytosis and neurotransmission fidelity during prolonged high-frequency stimulation. Such α-synuclein toxicity could be rescued by blocking microtubule polymerization, suggesting that microtubule overassembly underlies the toxicity of acutely elevated α-synuclein in the nerve terminal.


Assuntos
Vias Auditivas/efeitos dos fármacos , Vias Auditivas/metabolismo , Endocitose/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , alfa-Sinucleína/toxicidade , Animais , Exocitose/efeitos dos fármacos , Exocitose/genética , Feminino , Humanos , Masculino , Mutação/genética , Nocodazol/farmacologia , Polimerização , Ratos , Ratos Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/toxicidade , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/genética , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/genética , alfa-Sinucleína/genética
18.
J Neurosci ; 37(7): 1888-1899, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-28093477

RESUMO

The relative contributions of glycolysis and oxidative phosphorylation to neuronal presynaptic energy demands are unclear. In rat hippocampal neurons, ATP production by either glycolysis or oxidative phosphorylation alone sustained basal evoked synaptic transmission for up to 20 min. However, combined inhibition of both ATP sources abolished evoked transmission. Neither action potential propagation failure nor depressed Ca2+ influx explained loss of evoked synaptic transmission. Rather, inhibition of ATP synthesis caused massive spontaneous vesicle exocytosis, followed by arrested endocytosis, accounting for the disappearance of evoked postsynaptic currents. In contrast to its weak effects on basal transmission, inhibition of oxidative phosphorylation alone depressed recovery from vesicle depletion. Local astrocytic lactate shuttling was not required. Instead, either ambient monocarboxylates or neuronal glycolysis was sufficient to supply requisite substrate. In summary, basal transmission can be sustained by glycolysis, but strong presynaptic demands are met preferentially by oxidative phosphorylation, which can be maintained by bulk but not local monocarboxylates or by neuronal glycolysis.SIGNIFICANCE STATEMENT Neuronal energy levels are critical for proper CNS function, but the relative roles for the two main sources of ATP production, glycolysis and oxidative phosphorylation, in fueling presynaptic function in unclear. Either glycolysis or oxidative phosphorylation can fuel low-frequency synaptic function and inhibiting both underlies loss of synaptic transmission via massive vesicle release and subsequent failure to endocytose lost vesicles. Oxidative phosphorylation, fueled by either glycolysis or endogenously released monocarboxylates, can fuel more metabolically demanding tasks such as vesicle recovery after depletion. Our work demonstrates the flexible nature of fueling presynaptic function to maintain synaptic function.


Assuntos
Trifosfato de Adenosina/metabolismo , Neurônios/fisiologia , Terminações Pré-Sinápticas/metabolismo , Transmissão Sináptica/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Células Cultivadas , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Inibidores Enzimáticos/farmacologia , Glicólise/efeitos dos fármacos , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Neurotransmissores/farmacologia , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Potenciais Sinápticos/efeitos dos fármacos , Potenciais Sinápticos/fisiologia , Vesículas Sinápticas/efeitos dos fármacos
19.
J Neurochem ; 144(4): 466-482, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28960313

RESUMO

Proper neuronal function requires essential biological cargoes to be packaged within membranous vesicles and transported, intracellularly, through the extensive outgrowth of axonal and dendritic fibers. The precise spatiotemporal movement of these cargoes is vital for neuronal survival and, thus, is highly regulated. In this study we test how the axonal movement of a neuropeptide-containing dense-core vesicle (DCV) responds to alcohol stressors. We found that ethanol induces a strong anterograde bias in vesicle movement. Low doses of ethanol stimulate the anterograde movement of neuropeptide-DCV while high doses inhibit bi-directional movement. This process required the presence of functional kinesin-1 motors as reduction in kinesin prevented the ethanol-induced stimulation of the anterograde movement of neuropeptide-DCV. Furthermore, expression of inactive glycogen synthase kinase 3 (GSK-3ß) also prevented ethanol-induced stimulation of neuropeptide-DCV movement, similar to pharmacological inhibition of GSK-3ß with lithium. Conversely, inhibition of PI3K/AKT signaling with wortmannin led to a partial prevention of ethanol-stimulated transport of neuropeptide-DCV. Taken together, we conclude that GSK-3ß signaling mediates the stimulatory effects of ethanol. Therefore, our study provides new insight into the physiological response of the axonal movement of neuropeptide-DCV to exogenous stressors. Cover Image for this Issue: doi: 10.1111/jnc.14165.


Assuntos
Transporte Axonal/efeitos dos fármacos , Axônios/metabolismo , Depressores do Sistema Nervoso Central/farmacologia , Drosophila/fisiologia , Etanol/farmacologia , Neurônios Motores/metabolismo , Neuropeptídeos/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Axônios/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/metabolismo , Imuno-Histoquímica , Cinesinas/fisiologia , Larva , Lítio/farmacologia , Neurônios Motores/efeitos dos fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais/efeitos dos fármacos , Estimulação Química , Vesículas Sinápticas/efeitos dos fármacos , Wortmanina/farmacologia
20.
Brain Behav Immun ; 73: 222-234, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29751053

RESUMO

We previously reported that l-Cysteine, an H2S donor, significantly alleviated brain injury after hypoxia-ischemic (HI) injury in neonatal mice. However, the mechanisms underlying this neuroprotective effect of l-Cysteine against HI insult remain unknown. In the present study, we tested the hypothesis that the protective effects of l-Cysteine are associated with glial responses and autophagy, and l-Cysteine attenuates synaptic injury as well as behavioral deficits resulting from HI. Consistent with our previous findings, we found that treatment with l-Cysteine after HI reduced early brain injury, improved behavioral deficits and synaptic damage, effects which were associated with an up-regulation of synaptophysin and postsynaptic density protein 95 expression in the lesioned cortex. l-Cysteine attenuated the accumulation of CD11b+/CD45high cells, activation of microglia and astrocytes and diminished HI-induced increases in reactive oxygen species and malondialdehyde within the lesioned cortex. In addition, l-Cysteine increased microtubule associated protein 1 light chain 3-II and Beclin1 expression, decreased p62 expression and phosphor-mammalian target of rapamycin and phosphor-signal transducer and activator of transcription 3. Further support for a critical role of l-Cysteine was revealed from results demonstrating that treatment with an inhibitor of the H2S-producing enzyme, amino-oxyacetic acid, reversed the beneficial effects of l-Cysteine described above. These results demonstrate that l-Cysteine effectively alleviates HI injury and improves behavioral outcomes by inhibiting reactive glial responses and synaptic damage and an accompanying triggering of autophagic flux. Accordingly, l-Cysteine may provide a new a therapeutic approach for the treatment of HI via the formation of H2S.


Assuntos
Cisteína/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Ácido Amino-Oxiacético/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Autofagia/efeitos dos fármacos , Cisteína/metabolismo , Sulfeto de Hidrogênio , Hipóxia , Camundongos , Microglia/metabolismo , Neuroglia/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Vesículas Sinápticas/efeitos dos fármacos , Sinaptofisina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA