Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Toxicol Appl Pharmacol ; 482: 116776, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38043803

RESUMO

Bisphenol A (BPA) has been implicated in cognitive impairment. Icariin is the main active ingredient extracted from Epimedium Herb with protective function of nervous system. However, the potential therapeutic effects of Icariin on spatial memory deficits induced by developmental BPA exposure in Sprague-Dawley rats have not been investigated. This study investigated the therapeutic effect of Icariin (10 mg/kg/day, from postnatal day (PND) 21 to PND 60 by gavage) on spatial memory deficits in rat induced by developmental BPA exposure (1 mg/kg/day, from embryonic to PND 60), demonstrating that Icariin can markedly improve spatial memory in BPA-exposed rat. Furthermore, intra-gastric administration of Icariin could attenuate abnormal hippocampal cell dispersion and loss, improved the dendritic spine density and Nissl bodies. Moreover, Icariin reversed BPA induced reduction of frequency of miniature excitatory postsynaptic currents(mEPSC) and decrease of Vesicular glutamate transporter 1(VGlut1). Collectively, Icariin could effectively rescue BPA-induced spatial memory impairment in male rats by preventing cell loss and reduction of dendritic spines in the hippocampus. In addition, we also found that VGlut1 is a critical target in the repair of BPA-induced spatial memory by Icariin. Thus, Icariin may be a promising therapeutic agent to attenuate BPA-induced spatial memory deficits.


Assuntos
Flavonoides , Hipocampo , Fenóis , Memória Espacial , Ratos , Animais , Masculino , Ratos Sprague-Dawley , Compostos Benzidrílicos/toxicidade , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/prevenção & controle , Aprendizagem em Labirinto
2.
Cell Mol Neurobiol ; 43(6): 2785-2799, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36689065

RESUMO

Abelson non-receptor tyrosine kinases (Abl1 and Abl2) are established cellular signaling proteins, implicated in cytoskeletal reorganization essential for modulation of cell morphology and motility. During development of the central nervous system, Abl kinases play fundamental roles in neurulation and neurite outgrowth, relaying information from axon guidance cues and growth factor receptors to promote cytoskeletal rearrangements. In mature neurons, Abl kinases localize to pre- and postsynaptic compartments and are involved in regulation of synaptic stability and plasticity. Although emerging evidence indicates interchangeability of these isoforms in managing of cellular functions, in healthy adult neurons, Abl1 contribution is less elucidated, while Abl2 is required for optimal synaptic functioning. Our previous study demonstrated compartmentalization of Abl1 to the presynapse and Abl2 to the postsynapse and characterized their modulatory effect on spontaneous excitatory synaptic transmission. Here, we further delineate the role of Abl2 on regulation of the postsynaptic component of miniature excitatory postsynaptic current (mEPSC). Our findings show that both acute and prolonged activation of Abl2, in line with reduction of mEPSC amplitude, also decrease AMPA and NMDA current amplitudes. In contrast with the current-detrimental effect, prolonged Abl2 activity stabilizes spines, particularly contributing to maintenance of active synapses at distal (perhaps apical) segments of dendrites. Hence, we propose that attenuation of ion currents via ionotropic glutamatergic receptors by Abl2 kinase derives from either reduction of the receptor sensitivity for glutamate or is due to alteration of channel gating mechanisms. Abl2 and excitatory postsynapses: Abl2 expression level affects active excitatory synapse density on distal dendrites, while Abl2 activity impacts current density through AMPA and NMDA receptors.


Assuntos
Sinapses , Transmissão Sináptica , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo , Transmissão Sináptica/fisiologia , Sinapses/metabolismo , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
3.
Cereb Cortex ; 31(2): 1182-1200, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33063109

RESUMO

As axons from the raphe nuclei densely innervate the somatosensory cortex, we investigated how serotonin (5-HT) modulates transmitter release in layer II pyramidal cells of rat barrel cortex. In the presence of tetrodotoxin and gabazine, 10 µM 5-HT caused a waxing and waning in the frequency of miniature excitatory postsynaptic currents (mEPSC) with no effect on amplitude. Specifically, within 15 min of recording the mEPSC frequency initially increased by 28 ± 7%, then dropped to below control (-15 ± 3%), before resurging back to 27 ± 7% larger than control. These changes were seen in 47% of pyramidal cells (responders) and were mediated by 5-HT2C receptors (5-HT2CR). Waxing resulted from phospholipase C activation, IP3 production, and Ca2+ release from presynaptic stores. Waning was prevented if PKC was blocked. In contrast, in paired recordings, the unitary EPSC amplitude was reduced by 50 ± 3% after 5-HT exposure in almost all cases with no significant effect on paired-pulse ratio and synaptic dynamics. This sustained EPSC reduction was also caused by 5-HT2R, but was mediated by presynaptic Gßγ subunits likely limiting influx through CaV2 channels. EPSC reduction, together with enhanced spontaneous noise in a restricted subset of inputs, could temporarily diminish the signal-to-noise ratio and affect the computation in the neocortical microcircuit.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Células Piramidais/metabolismo , Agonistas do Receptor de Serotonina/farmacologia , Serotonina/farmacologia , Córtex Somatossensorial/metabolismo , Animais , Técnicas de Cultura de Órgãos , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Wistar , Receptor 5-HT2C de Serotonina/metabolismo , Córtex Somatossensorial/citologia , Córtex Somatossensorial/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 116(7): 2701-2706, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30692251

RESUMO

Glutamate is the most abundant excitatory neurotransmitter, present at the bulk of cortical synapses, and participating in many physiologic and pathologic processes ranging from learning and memory to stroke. The tripeptide, glutathione, is one-third glutamate and present at up to low millimolar intracellular concentrations in brain, mediating antioxidant defenses and drug detoxification. Because of the substantial amounts of brain glutathione and its rapid turnover under homeostatic control, we hypothesized that glutathione is a relevant reservoir of glutamate and could influence synaptic excitability. We find that drugs that inhibit generation of glutamate by the glutathione cycle elicit decreases in cytosolic glutamate and decreased miniature excitatory postsynaptic potential (mEPSC) frequency. In contrast, pharmacologically decreasing the biosynthesis of glutathione leads to increases in cytosolic glutamate and enhanced mEPSC frequency. The glutathione cycle can compensate for decreased excitatory neurotransmission when the glutamate-glutamine shuttle is inhibited. Glutathione may be a physiologic reservoir of glutamate neurotransmitter.


Assuntos
Glutationa/metabolismo , Sinapses/metabolismo , Animais , Células Cultivadas , Potenciais Pós-Sinápticos Excitadores/fisiologia , Ácido Glutâmico/metabolismo , Homeostase , Neurônios/fisiologia , Ratos Sprague-Dawley , Transmissão Sináptica/fisiologia
5.
Cell Mol Neurobiol ; 41(3): 431-448, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32399753

RESUMO

Amyloid beta (Aß) peptides represent one of the most studied etiological factors of Alzheimer's disease. Nevertheless, the effects elicited by different molecular forms of amyloid beta peptides widely vary between the studies, mostly depending on experimental conditions. Despite the enormous amount of accumulated evidences concerning the pathological effects of amyloid beta peptides, the exact identity of the amyloid beta species is still controversial, and even less is clear as regards to the downstream effectors that mediate the devastating impact of these peptides on synapses in the central nervous system. Recent publications indicate that some of the neurotoxic effects of amyloid beta peptides may be mediated via the activation of proteins belonging to the Abelson non-receptor tyrosine kinase (Abl) family, that are known to regulate actin cytoskeleton structure as well as phosphorylate microtubule-associated tau protein, a hallmark of Alzheimer's disease. By performing series of miniature excitatory postsynaptic currents (mEPSC) recordings in cultured hippocampal cells, we demonstrate that activation of Abl kinases by acute application of 42 amino acid-length monomeric amyloid beta (Aß1-42) peptides reduces spontaneous synaptic release, while this effect can be rescued by pharmacologic inhibition of Abl kinase activity, or by reduction of Abl expression with small interfering RNAs. Our electrophysiological data are further reinforced by a subsequent biochemical analysis, showing enhanced phosphorylation of Abl kinase substrate CT10 Regulator of Kinase-homolog-Like (Crkl) upon treatment of hippocampal neurons with Aß peptides. Thus, we conclude that Abl kinase activation may be involved in Aß-induced weakening of synaptic transmission.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Fragmentos de Peptídeos/toxicidade , Proteínas Proto-Oncogênicas c-abl/metabolismo , Sinapses/metabolismo , Animais , Ativação Enzimática/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hidantoínas/farmacologia , Mesilato de Imatinib/farmacologia , Neurotransmissores/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Pirimidinas/farmacologia , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
6.
J Neurophysiol ; 123(6): 2426-2436, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32401126

RESUMO

In this study, the effect of extracellular pH on glutamatergic synaptic transmission was examined in mechanically dissociated rat hippocampal CA3 pyramidal neurons using a whole-cell patch-clamp technique under voltage-clamp conditions. Native synaptic boutons were isolated without using any enzymes, using a so-called "synapse bouton preparation," and preserved for the electrical stimulation of single boutons. Both the frequency and amplitude of spontaneous excitatory postsynaptic currents (sEPSCs) were found to decrease and increase in response to modest acidic (~pH 6.5) and basic (~pH 8.5) solutions, respectively. These changes in sEPSC frequency were not affected by the addition of TTX but completely disappeared by successive addition of Cd2+. However, changes in sEPSC amplitude induced by acidic and basic extracellular solutions were not affected by the addition of neither TTX nor Cd2+. The glutamate-induced whole-cell currents were decreased and increased by acidic and basic solutions, respectively. Acidic pH also decreased the amplitude and increased the failure rate (Rf) and paired-pulse rate (PPR) of glutamatergic electrically evoked excitatory postsynaptic currents (eEPSCs), while a basic pH increased the amplitude and decreased both the Rf and PPR of eEPSCs. The kinetics of the currents were not affected by changes in pH. Acidic and basic solutions decreased and increased voltage-gated Ca2+ but not Na+ channel currents in the dentate gyrus granule cell bodies. Our results indicate that extracellular pH modulates excitatory transmission via both pre- and postsynaptic sites, with the presynaptic modulation correlated to changes in voltage-gated Ca2+ channel currents.NEW & NOTEWORTHY The effects of external pH changes on spontaneous, miniature, and evoked excitatory synaptic transmission in CA3 hippocampal synapses were examined using the isolated nerve bouton preparation, which allowed for the accurate regulation of extracellular pH at the synapses. Acidification generally reduced transmission, partly via effects on presynaptic Ca2+ channel currents, while alkalization generally enhanced transmission. Both pre- and postsynaptic sites contributed to these effects.


Assuntos
Região CA3 Hipocampal/fisiologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Concentração de Íons de Hidrogênio , Terminações Pré-Sinápticas/fisiologia , Células Piramidais/fisiologia , Animais , Região CA3 Hipocampal/química , Feminino , Ácido Glutâmico/metabolismo , Masculino , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/química , Células Piramidais/química , Ratos , Ratos Wistar
7.
Int J Mol Sci ; 20(13)2019 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-31324059

RESUMO

Anti-inflammatory cytokines are known to exert neuroprotective action ameliorating aberrant neuronal network activity associated with inflammatory responses. Yet, it is still not fully understood if anti-inflammatory cytokines play a significant role in the regulation of synaptic activity under normal conditions. Thus, the aim of our study was to investigate the effect of Interleukin-10 (IL-10) on neuronal synaptic transmission and plasticity. For this we tested the effect of IL-10 on miniature excitatory postsynaptic currents (mEPSC) and intracellular Ca2+ responses using whole-cell patch clamp and fluorescence microscopy in 13-15 DIV primary hippocampal neuroglial culture. We found that IL-10 significantly potentiated basal glutamatergic excitatory synaptic transmission within 15 min after application. Obtained results revealed a presynaptic nature of the effect, as IL-10 in a dose-dependent manner significantly increased the frequency but not the amplitude of mEPSC. Further, we tested the effect of IL-10 on mEPSC in a model of homeostatic synaptic plasticity (HSP) induced by treatment of primary hippocampal culture with 1 µM of tetrodotoxin (TTX) for a 24 h. It was found that 15 min application of IL-10 at established HSP resulted in enhanced mEPSC frequency, thus partially compensating for a decrease in the mEPSC frequency associated with TTX-induced HSP. Next, we studied if IL-10 can influence induction of HSP. We found that co-incubation of IL-10 with 1 µM of TTX for 24 h induced synaptic scaling, significantly increasing the amplitude of mEPSC and Ca2+ responses to application of the AMPA agonist, 5-Fluorowillardiine, thus facilitating a compensatory postsynaptic mechanism at HSP condition. Our results indicate that IL-10 potentiates synaptic activity in a dose- and time-dependent manner exerting both presynaptic (short-term exposure) and postsynaptic (long-term exposure) action. Obtained results demonstrate involvement of IL-10 in the regulation of basal glutamatergic synaptic transmission and plasticity at normal conditions.


Assuntos
Hipocampo/citologia , Interleucina-10/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Alanina/análogos & derivados , Alanina/farmacologia , Animais , Células Cultivadas , Microscopia de Fluorescência , Plasticidade Neuronal/efeitos dos fármacos , Técnicas de Patch-Clamp , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Tetrodotoxina/farmacologia
8.
J Neurosci ; 37(16): 4301-4310, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28320843

RESUMO

At chemical synapses, voltage-activated calcium channels (VACCs) mediate Ca2+ influx to trigger action potential-evoked neurotransmitter release. However, the mechanisms by which Ca2+ regulates spontaneous transmission have not been fully determined. We have shown that VACCs are a major trigger of spontaneous release at neocortical inhibitory synapses but not at excitatory synapses, suggesting fundamental differences in spontaneous neurotransmission at GABAergic and glutamatergic synapses. Recently, VACC blockers were reported to reduce spontaneous release of glutamate and it was proposed that there was conservation of underlying mechanisms of neurotransmission at excitatory and inhibitory synapses. Furthermore, it was hypothesized that the different effects on excitatory and inhibitory synapses may have resulted from off-target actions of Cd2+, a nonselective VACC blocker, or other variations in experimental conditions. Here we report that in mouse neocortical neurons, selective and nonselective VACC blockers inhibit spontaneous release at inhibitory but not at excitatory terminals, and that this pattern is observed in culture and slice preparations as well as in synapses from acute slices of the auditory brainstem. The voltage dependence of Cd2+ block of VACCs accounts for the apparent lower potency of Cd2+ on spontaneous release of GABA than on VACC current amplitudes. Our findings indicate fundamental differences in the regulation of spontaneous release at inhibitory and excitatory synapses by stochastic VACC activity that extend beyond the cortex to the brainstem.SIGNIFICANCE STATEMENT Presynaptic Ca2+ entry via voltage-activated calcium channels (VACCs) is the major trigger of action potential-evoked synaptic release. However, the role of VACCs in the regulation of spontaneous neurotransmitter release (in the absence of a synchronizing action potential) remains controversial. We show that spontaneous release is affected differently by VACCs at excitatory and inhibitory synapses. At inhibitory synapses, stochastic openings of VACCs trigger the majority of spontaneous release, whereas they do not affect spontaneous release at excitatory synapses. We find this pattern to be wide ranging, holding for large and small synapses in the neocortex and brainstem. These findings indicate fundamental differences of the Ca2+ dependence of spontaneous release at excitatory and inhibitory synapses and heterogeneity of the mechanisms of release across the CNS.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/metabolismo , Potenciais Pós-Sinápticos Excitadores , Potenciais Pós-Sinápticos Inibidores , Sinapses/metabolismo , Animais , Tronco Encefálico/citologia , Cádmio/farmacologia , Células Cultivadas , Feminino , Masculino , Camundongos , Potenciais Pós-Sinápticos em Miniatura , Neocórtex/citologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
9.
J Neurosci ; 35(2): 776-85, 2015 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-25589770

RESUMO

Severe autonomic dysfunction, including the loss of control of the cardiovascular, respiratory, and gastrointestinal systems, is a common comorbidity of stroke and other bleeding head injuries. Previous studies suggest that this collapse of autonomic control may be caused by thrombin acting on astrocytic protease-activated receptors (PAR1) in the hindbrain. Using calcium imaging and electrophysiological techniques, we evaluated the mechanisms by which astrocytic PAR1s modulate the activity of presynaptic vagal afferent terminals and postsynaptic neurons in the rat nucleus of the solitary tract (NST). Our calcium-imaging data show that astrocytic and neuronal calcium levels increase after brain slices are treated with the PAR1 agonist SFLLRN-NH2. This increase in activity is blocked by pretreating the slices with the glial metabolic blocker fluorocitrate. In addition, PAR1-activated astrocytes communicate directly with NST neurons by releasing glutamate. Calcium responses to SFLLRN-NH2 in the astrocytes and neurons significantly increase after bath application of the excitatory amino acid transporter blocker DL-threo-ß-benzyloxyaspartic acid (TBOA) and significantly decrease after bath application of the NMDA receptor antagonist DL-2-amino-5-phosphonopentanoic acid (DL-AP5). Furthermore, astrocytic glutamate activates neuronal GluN2B-containing NMDA receptors. Voltage-clamp recordings of miniature EPSCs (mEPSCs) from NST neurons show that astrocytes control presynaptic vagal afferent excitability directly under resting and activated conditions. Fluorocitrate significantly decreases mEPSC frequency and SFLLRN-NH2 significantly increases mEPSC frequency. These data show that astrocytes act within a tripartite synapse in the NST, controlling the excitability of both postsynaptic NST neurons and presynaptic vagal afferent terminals.


Assuntos
Astrócitos/metabolismo , Neurônios Aferentes/fisiologia , Receptor PAR-1/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleo Solitário/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , Animais , Ácido Aspártico/farmacologia , Astrócitos/efeitos dos fármacos , Cálcio/metabolismo , Citratos/farmacologia , Antagonistas de Aminoácidos Excitatórios , Potenciais Pós-Sinápticos Excitadores , Feminino , Ácido Glutâmico/metabolismo , Masculino , Potenciais Pós-Sinápticos em Miniatura , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Fragmentos de Peptídeos/farmacologia , Ratos , Ratos Long-Evans , Receptor PAR-1/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Núcleo Solitário/citologia , Núcleo Solitário/metabolismo , Sinapses/metabolismo , Sinapses/fisiologia , Nervo Vago/citologia , Nervo Vago/metabolismo , Nervo Vago/fisiologia
10.
J Biol Chem ; 290(42): 25548-60, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26338711

RESUMO

The regulation of AMPA-type receptor (AMPAR) abundance in the postsynaptic membrane is an important mechanism involved in learning and memory formation. Recent data suggest that one of the constituents of the AMPAR complex is carnitine palmitoyltransferase 1C (CPT1C), a brain-specific isoform located in the endoplasmic reticulum of neurons. Previous results had demonstrated that CPT1C deficiency disrupted spine maturation in hippocampal neurons and impaired spatial learning, but the role of CPT1C in AMPAR physiology had remained mostly unknown. In the present study, we show that CPT1C binds GluA1 and GluA2 and that the three proteins have the same expression profile during neuronal maturation. Moreover, in hippocampal neurons of CPT1C KO mice, AMPAR-mediated miniature excitatory postsynaptic currents and synaptic levels of AMPAR subunits GluA1 and GluA2 are significantly reduced. We show that AMPAR expression is dependent on CPT1C levels because total protein levels of GluA1 and GluA2 are decreased in CPT1C KO neurons and are increased in CPT1C-overexpressing neurons, whereas other synaptic proteins remain unaltered. Notably, mRNA levels of AMPARs remained unchanged in those cultures, indicating that CPT1C is post-transcriptionally involved. We demonstrate that CPT1C is directly involved in the de novo synthesis of GluA1 and not in protein degradation. Moreover, in CPT1C KO cultured neurons, GluA1 synthesis after chemical long term depression was clearly diminished, and brain-derived neurotrophic factor treatment was unable to phosphorylate the mammalian target of rapamycin (mTOR) and stimulate GluA1 protein synthesis. These data newly identify CPT1C as a regulator of AMPAR translation efficiency and therefore also synaptic function in the hippocampus.


Assuntos
Carnitina O-Palmitoiltransferase/metabolismo , Hipocampo/metabolismo , Receptores de AMPA/biossíntese , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Carnitina O-Palmitoiltransferase/genética , Hipocampo/citologia , Hipocampo/enzimologia , Camundongos , Camundongos Knockout , Neurônios/enzimologia , Neurônios/metabolismo , Receptores de AMPA/química , Receptores de AMPA/metabolismo , Transmissão Sináptica
11.
J Neurophysiol ; 113(3): 720-9, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25376786

RESUMO

Sex differences in neuron electrophysiological properties were traditionally associated with brain regions directly involved in reproduction in adult, postpubertal animals. There is growing acknowledgement that sex differences can exist in other developmental periods and brain regions as well. This includes the dorsal striatum (caudate/putamen), which shows robust sex differences in gene expression, neuromodulator action (including dopamine and 17ß-estradiol), and relevant sensorimotor behaviors and pathologies such as the responsiveness to drugs of abuse. Here we examine whether these sex differences extend to striatal neuron electrophysiology. We test the hypothesis that passive and active medium spiny neuron (MSN) electrophysiological properties in prepubertal rat dorsal striatum differ by sex. We made whole cell recordings from male and females MSNs from acute brain slices. The slope of the evoked firing rate to current injection curve was increased in MSNs recorded from females compared with males. The initial action potential firing rate was increased in MSNs recorded from females compared with males. Action potential after-hyperpolarization peak was decreased, and threshold was hyperpolarized in MSNs recorded from females compared with males. No sex differences in passive electrophysiological properties or miniature excitatory synaptic currents were detected. These findings indicate that MSN excitability is increased in prepubertal females compared with males, providing a new mechanism that potentially contributes to generating sex differences in striatal-mediated processes. Broadly, these findings demonstrate that sex differences in neuron electrophysiological properties can exist prepuberty in brain regions not directly related to reproduction.


Assuntos
Potenciais de Ação , Corpo Estriado/fisiologia , Neurônios/fisiologia , Animais , Corpo Estriado/citologia , Corpo Estriado/crescimento & desenvolvimento , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Fatores Sexuais
12.
J Neurophysiol ; 111(3): 648-58, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24225540

RESUMO

PSD-95-like, disc-large (DLG) family membrane-associated guanylate kinase proteins (PSD/DLG-MAGUKs) are essential for regulating synaptic AMPA receptor (AMPAR) function and activity-dependent trafficking of AMPARs. Using a molecular replacement strategy to replace endogenous PSD-95 with SAP97ß, we show that the prototypic ß-isoform of the PSD-MAGUKs, SAP97ß, has distinct NMDA receptor (NMDAR)-dependent roles in regulating basic properties of AMPAR-containing synapses. SAP97ß enhances the number of AMPAR-containing synapses in an NMDAR-dependent manner, whereas its effect on the size of unitary synaptic response is not fully dependent on NMDAR activity. These effects contrast with those of PSD-95α, which increases both the number of AMPAR-containing synapses and the size of unitary synaptic responses, with or without NMDAR activity. Our results suggest that SAP97ß regulates synaptic AMPAR content by increasing surface expression of GluA1-containing AMPARs, whereas PSD-95α enhances synaptic AMPAR content presumably by increasing the synaptic scaffold capacity for synaptic AMPARs. Our approach delineates discrete effects of different PSD-MAGUKs on principal properties of glutamatergic synaptic transmission. Our results suggest that the molecular diversity of PSD-MAGUKs can provide rich molecular substrates for differential regulation of glutamatergic synapses in the brain.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Membrana/metabolismo , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteína 4 Homóloga a Disks-Large , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Potenciais da Membrana , Proteínas de Membrana/genética , Neurônios/metabolismo , Neurônios/fisiologia , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/genética , Receptores de N-Metil-D-Aspartato/genética , Sinapses/fisiologia , Membranas Sinápticas/metabolismo
13.
J Neurochem ; 128(4): 476-90, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24117520

RESUMO

Hippocampal neurons are affected by chronic stress and have a high density of cytoplasmic mineralocorticoid and glucocorticoid receptors (MR and GR). Detailed studies on the genomic effects of the stress hormone corticosterone at physiologically relevant concentrations on different steps in synaptic transmission are limited. In this study, we tried to delineate how activation of MR and GR by different concentrations of corticosterone affects synaptic transmission at various levels. The effect of 3-h corticosterone (25, 50, and 100 nM) treatment on depolarization-mediated calcium influx, vesicular release and properties of miniature excitatory post-synaptic currents (mEPSCs) were studied in cultured hippocampal neurons. Activation of MR with 25 nM corticosterone treatment resulted in enhanced depolarization-mediated calcium influx via a transcription-dependent process and increased frequency of mEPSCs with larger amplitude. On the other hand, activation of GR upon 100 nM corticosterone treatment resulted in increase in the rate of vesicular release via the genomic actions of GR. Furthermore, GR activation led to significant increase in the frequency of mEPSCs with larger amplitude and faster decay. Our studies indicate that differential activation of the dual receptor system of MR and GR by corticosterone targets the steps in synaptic transmission differently.


Assuntos
Corticosterona/farmacologia , Receptores de Glucocorticoides/efeitos dos fármacos , Receptores de Mineralocorticoides/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Cálcio/metabolismo , Cálcio/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Relação Dose-Resposta a Droga , Imunofluorescência , Ácido Glutâmico/fisiologia , Hipocampo/citologia , Hipocampo/metabolismo , Rede Nervosa/citologia , Rede Nervosa/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Cultura Primária de Células , RNA/biossíntese , RNA/isolamento & purificação , Ratos , Ratos Wistar
14.
Eur J Neurosci ; 39(7): 1214-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24713000

RESUMO

In our previous studies, phosphorylation-dependent tau mislocalization to dendritic spines resulted in early cognitive and synaptic deficits. It is well known that amyloid beta (Aß) oligomers cause synaptic dysfunction by inducing calcineurin-dependent AMPA receptor (AMPAR) internalization. However, it is unknown whether Aß-induced synaptic deficits depend upon tau phosphorylation. It is also unknown whether changes in tau can cause calcineurin-dependent loss of AMPARs in synapses. Here, we show that tau mislocalizes to dendritic spines in cultured hippocampal neurons from APPSwe Alzheimer's disease (AD)-transgenic mice and in cultured rat hippocampal neurons treated with soluble Aß oligomers. Interestingly, Aß treatment also impairs synaptic function by decreasing the amplitude of miniature excitatory postsynaptic currents (mEPSCs). The above tau mislocalization and Aß-induced synaptic impairment are both diminished by the expression of AP tau, indicating that these events require tau phosphorylation. The phosphatase activity of calcineurin is important for AMPAR internalization via dephosphorylation of GluA1 residue S845. The effects of Aß oligomers on mEPSCs are blocked by the calcineurin inhibitor FK506. Aß-induced loss of AMPARs is diminished in neurons from knock-in mice expressing S845A mutant GluA1 AMPA glutamate receptor subunits. This finding suggests that changes in phosphorylation state at S845 are involved in this pathogenic cascade. Furthermore, FK506 rescues deficits in surface AMPAR clustering on dendritic spines in neurons cultured from transgenic mice expressing P301L tau proteins. Together, our results support the role of tau and calcineurin as two intermediate signaling molecules between Aß initiation and eventual synaptic dysfunction early in AD pathogenesis.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Espinhas Dendríticas/metabolismo , Potenciais Pós-Sinápticos em Miniatura , Fragmentos de Peptídeos/toxicidade , Receptores de AMPA/metabolismo , Proteínas tau/metabolismo , Animais , Calcineurina/metabolismo , Inibidores de Calcineurina/farmacologia , Células Cultivadas , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , Potenciais Pós-Sinápticos Excitadores , Camundongos , Mutação , Fosforilação , Transporte Proteico , Receptores de AMPA/genética , Tacrolimo/farmacologia , Proteínas tau/genética
15.
Mol Cell Neurosci ; 56: 244-54, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23769723

RESUMO

The COMPLEXIN (CPX) proteins play a critical role in synaptic vesicle fusion and neurotransmitter release. Previous studies demonstrated that CPX functions in both activation of evoked neurotransmitter release and inhibition/clamping of spontaneous synaptic vesicle fusion. Here we report a new cpx mutant in Drosophila melanogaster, cpx(1257), revealing spatially defined and separable pools of CPX which make distinct contributions to the activation and clamping functions. In cpx(1257), lack of only the last C-terminal amino acid of CPX is predicted to disrupt prenylation and membrane targeting of CPX. Immunocytochemical analysis established localization of wild-type CPX to active zone (AZ) regions containing neurotransmitter release sites as well as broader presynaptic membrane compartments including synaptic vesicles. Parallel biochemical studies confirmed CPX membrane association and demonstrated robust binding interactions of CPX with all three SNAREs. This is in contrast to the cpx(1257) mutant, in which AZ localization of CPX persists but general membrane localization and, surprisingly, the bulk of CPX-SNARE protein interactions are abolished. Furthermore, electrophysiological analysis of neuromuscular synapses revealed interesting differences between cpx(1257) and a cpx null mutant. The cpx null exhibited a marked decrease in the EPSC amplitude, slowed EPSC rise and decay times and an increased mEPSC frequency with respect to wild-type. In contrast, cpx(1257) exhibited a wild-type EPSC with an increased mEPSC frequency and thus a selective failure to clamp spontaneous release. These results indicate that spatially distinct and separable interactions of CPX with presynaptic membranes and SNARE proteins mediate separable activation and clamping functions of CPX in neurotransmitter release.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Drosophila melanogaster/metabolismo , Exocitose , Mutação , Junção Neuromuscular/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Sítios de Ligação , Drosophila melanogaster/genética , Potenciais Pós-Sinápticos Excitadores , Junção Neuromuscular/fisiologia , Ligação Proteica , Transporte Proteico , Proteínas SNARE/metabolismo , Vesículas Sinápticas/metabolismo
16.
Behav Brain Res ; 476: 115233, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39233145

RESUMO

The prefrontal cortex (PFC) plays an important role in social behavior and is sensitive to stressful circumstances. Challenging life conditions might change PFC function and put individuals at risk for maladaptive social behavior. The excitation-inhibition (EI) balance of prefrontal neurons appears to play a crucial role in this process. Here, we examined how a challenging life condition in C57BL/6JolaHsd mice, i.e. group-housing 6 mice in a complex environment for 10 days in adulthood, changes the EI-balance of infralimbic prefrontal neurons in layer 2/3, compared to standard pair-housing. Slices were prepared from "undisturbed" mice, i.e. the first mouse taken from the cage, or mice taken ∼15 min later, who were mildly aroused after removal of the first mouse. We observed a housing-condition by arousal-state interaction, with in the complex housing group an elevated EI-balance in undisturbed and reduced EI-balance in mildly aroused animals, while no differences were observed in standard housed animals. The change was explained by a shift in mIPSC and mEPSC frequency, while amplitudes remained unaffected. Female mice showed no housing-by-state interaction, but a main effect of housing was found for mIPSCs, with a higher frequency in complex- versus standard-housed females. No effects were observed in males who were complex-housed from a young age onwards. Explorative investigations support a potential mediating role of corticosterone in housing effects on the EI-balance of males. We argue that taking the arousal state of individuals into account is necessary to better understand the consequences of exposure to challenging life conditions for prefrontal function.

17.
Methods Mol Biol ; 2799: 257-267, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38727912

RESUMO

The NMDAR is a heterotetramer composed of two GluN1 subunits and two GluN2 and/or GluN3 subunits, with the GluN2 subunits exhibiting significant diversity in their structure and function. Recent studies have highlighted the importance of characterizing the specific roles of each GluN2 subunit across central nervous system regions and developmental stages, as well as their unique contributions to NMDAR-mediated signaling and plasticity. Understanding the distinct functions of GluN2 subunits is critical for the development of targeted therapeutic strategies for NMDAR-related disorders. However, measuring the functional contribution of individual GluN2 subtypes in ex vivo slices is challenging. Conventionally, pharmacological or genetic approaches are used, but, in many cases, this is not possible or is restricted to population-level NMDAR responses. Here, we describe a technique for using biophysical properties of miniature synaptic NMDAR responses as a proxy to measure the functional contribution of specific GluN2-NMDAR subunits to individual synapses within a neuron.


Assuntos
Subunidades Proteicas , Receptores de N-Metil-D-Aspartato , Sinapses , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Animais , Sinapses/metabolismo , Subunidades Proteicas/metabolismo , Camundongos , Neurônios/metabolismo , Ratos , Técnicas de Patch-Clamp/métodos , Transmissão Sináptica
18.
BJA Open ; 6: 100143, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37588178

RESUMO

Background: Inflammation and general anaesthesia likely contribute to perioperative neurocognitive disorders, possibly by causing a neuronal imbalance of excitation and inhibition. We showed previously that treatment with lipopolysaccharide (LPS) and sevoflurane causes a sustained increase in a tonic inhibitory conductance in the hippocampus; however, whether excitatory neurotransmission is also altered remains unknown. The goal of this study was to examine excitatory synaptic currents in the hippocampus after treatment with LPS and sevoflurane. Synaptic plasticity in the hippocampus, a cellular correlate of learning and memory, was also studied. Methods: Mice were injected with vehicle or LPS (1 mg kg-1 i.p.), and after 24 h they were then exposed to vehicle or sevoflurane (2.3%; 2 h). Hippocampal slices were prepared 48 h later. Excitatory synaptic currents were recorded from pyramidal neurones. Long-term potentiation (LTP) and long-term depression (LTD) were studied in the Schaffer collateral-cornu ammonis 1 pathway. Results: The amplitude of miniature excitatory postsynaptic currents (EPSCs) was reduced after LPS+sevoflurane (P<0.001), whereas that of spontaneous EPSCs was unaltered, as evidenced by cumulative distribution plots. The frequency, area, and kinetics of both miniature and spontaneous EPSCs were unchanged, as were LTP and LTD. Conclusions: The reduced amplitude of miniature EPSCs, coupled with the previously reported increase in tonic inhibition, indicates that the combination of LPS and sevoflurane markedly disrupts the balance of excitation and inhibition. Restoring this balance by pharmacologically enhancing excitatory neurotransmission and inhibiting the tonic current may represent an effective therapeutic option for perioperative neurocognitive disorders.

19.
Neuroscience ; 525: 38-46, 2023 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-37295597

RESUMO

Astrocytes have been increasingly acknowledged to play active roles in regulating synaptic transmission and plasticity. Through a variety of metabotropic and ionotropic receptors expressed on their surface, astrocytes detect extracellular neurotransmitters, and in turn, release gliotransmitters to modify synaptic strength, while they can also alter neuronal membrane excitability by modulating extracellular ionic milieu. Given the seemingly large repertoire of synaptic modulation, when, where and how astrocytes interact with synapses remain to be fully understood. Previously, we have identified a role for astrocyte NMDA receptor and L-VGCCs signaling in heterosynaptic presynaptic plasticity and promoting the heterogeneity of presynaptic strengths at hippocampal synapses. Here, we have sought to further clarify the mode by which astrocytes regulate presynaptic plasticity by exploiting a reduced culture system to globally evoke NMDA receptor-dependent presynaptic plasticity. Recording from a postsynaptic neuron intracellularly loaded with BAPTA, briefly bath applying NMDA and glycine induces a stable decrease in the rate of spontaneous glutamate release, which requires the presence of astrocytes and the activation of A1 adenosine receptors. Upon preventing astrocyte calcium signaling or blocking L-VGCCs, NMDA + glycine application triggers an increase, rather than a decrease, in the rate of spontaneous glutamate release, thereby shifting the presynaptic plasticity to promote an increase in strength. Our findings point to a crucial and surprising role of astrocytes in controlling the polarity of NMDA receptor and adenosine-dependent presynaptic plasticity. Such a pivotal mechanism unveils the power of astrocytes in regulating computations performed by neural circuits and is expected to profoundly impact cognitive processes.


Assuntos
Astrócitos , Sinalização do Cálcio , Astrócitos/metabolismo , Sinalização do Cálcio/fisiologia , Receptores de N-Metil-D-Aspartato/metabolismo , N-Metilaspartato/farmacologia , N-Metilaspartato/metabolismo , Transmissão Sináptica/fisiologia , Sinapses/metabolismo , Glutamatos/metabolismo , Glicina/metabolismo , Cálcio/metabolismo , Plasticidade Neuronal
20.
Artigo em Inglês | MEDLINE | ID: mdl-36812097

RESUMO

Aim: Activation of microglial NLRP3 inflammasome is an essential contributor to neuroinflammation underlying HIV-associated neurological disorders (HAND). Under pathological conditions, microglia-derived-EVs (MDEVs) can affect neuronal functions by delivering neurotoxic mediators to recipient cells. However, the role of microglial NLRP3 in mediating neuronal synaptodendritic injury has remained unexplored to date. In the present study, we sought to assess the regulatory role of HIV-1 Tat induced microglial NLRP3 in neuronal synaptodendritic injury. We hypothesized that HIV-1 Tat mediated microglia EVs carrying significant levels of NLRP3 contribute to the synaptodendritic injury, thereby affecting the maturation of neurons. Methods: To understand the cross-talk between microglia and neuron, we isolated EVs from BV2 and human primary microglia (HPM) cells with or without NLRP3 depletion using siNLRP3 RNA. EVs were isolated by differential centrifugation, characterized by ZetaView nanoparticle tracking analysis, electron microscopy, and western blot analysis for exosome markers. Purified EVs were exposed to primary rat neurons isolated from E18 rats. Along with green fluorescent protein (GFP) plasmid transfection, immunocytochemistry was performed to visualize neuronal synaptodendritic injury. Western blotting was employed to measure siRNA transfection efficiency and the extent of neuronal synaptodegeneration. Images were captured in confocal microscopy, and subsequently, Sholl analysis was performed for analyzing dendritic spines using neuronal reconstruction software Neurolucida 360. Electrophysiology was performed on hippocampal neurons for functional assessment. Results: Our findings demonstrated that HIV-1 Tat induced expression of microglial NLRP3 and IL1ß, and further that these were packaged in microglial exosomes (MDEV) and were also taken up by the neurons. Exposure of rat primary neurons to microglial Tat-MDEVs resulted in downregulation of synaptic proteins- PSD95, synaptophysin, excitatory vGLUT1, as well as upregulation of inhibitory proteins- Gephyrin, GAD65, thereby implicating impaired neuronal transmissibility. Our findings also showed that Tat-MDEVs not only caused loss of dendritic spines but also affected numbers of spine sub-types- mushroom and stubby. Synaptodendritic injury further affected functional impairment as evidenced by the decrease in miniature excitatory postsynaptic currents (mEPSCs). To assess the regulatory role of NLRP3 in this process, neurons were also exposed to Tat-MDEVs from NLRP3 silenced microglia. Tat-MDEVs from NLRP3 silenced microglia exerted a protective role on neuronal synaptic proteins, spine density as well as mEPSCs. Conclusion: In summary, our study underscores the role of microglial NLRP3 as an important contributor to Tat-MDEV mediated synaptodendritic injury. While the role of NLRP3 in inflammation is well-described, its role in EV-mediated neuronal damage is an interesting finding, implicating it as a target for therapeutics in HAND.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA