Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 51(3): 1300-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25056275

RESUMO

Sphingosine kinase-1 (Sphk1-1, EC 2.7.1.91) is a regulator of pro-survival signalling, and its alterations have been observed in Alzheimer's disease, brain ischemia and other neurological disorders. In this study we addressed the question whether Sphk1 and its product, sphingosine-1-phosphate (S1P), play a significant role in glucose deprivation (GD)/glucose reload (GR) stress in hippocampal neuronal cells (HT22). It was found that GD (6 h) followed by 24 h of GR evoked enhancement of the free radical level and neuronal HT22 cell death. Moreover, the significantly stronger gene expression for the pro-apoptotic Bax protein and down-regulation of the anti-apoptotic Bcl-2 and Bcl-XL proteins were observed. Concomitantly, this stress up-regulated: gene expression, protein level and activity of Sphk1. Exogenous S1P at 1 µM concentration and the other agonists of the S1P1 receptor (SEW 2871 and P-FTY720) enhanced HT22 cell viability affected by GD/GR stress. This mechanism is mediated by S1P receptor(s) signalling and by the activation of gene expression for Bcl-2 and Bcl-XL. Summarising, our data suggest that sphingolipid metabolism may play an important role in the early events that take place in neuronal cell survival/death under GD/GR stress. Our data demonstrate that exogenous S1P, through the activation of specific receptors S1P1 and S1P3 signalling pathways, regulates the gene expression for anti-apoptotic proteins and enhances neuronal cell survival affected by GD/GR stress.


Assuntos
Glucose/metabolismo , Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Estresse Fisiológico , Doença de Alzheimer/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Morte Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Camundongos , Organofosfatos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Esfingosina/metabolismo , Esfingosina/farmacologia
2.
Folia Neuropathol ; 52(3): 260-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25310737

RESUMO

Sphingosine kinases (Sphk1/2 EC 2.7.1.91) are responsible for synthesis of sphingosine-1-phosphate (S1P) and for regulation of the bioactive sphingolipids homeostasis. Sphingosine-1-phosphate can act as a potent messenger in an autocrine/paracrine manner through five specific G protein-coupled receptors (GPCR) S1P1-5. This sphingolipid is involved in the mechanism of transcription, mitochondrial function, neuronal viability and degeneration. Until now the involvement of Sphk1/2 and sphingolipid alterations in Parkinson's disease (PD) remains unknown. Recent studies have indicated the role of sphingolipids in the regulation of alpha-synuclein (ASN) in the PD brain. Our latest data demonstrated significant inhibition of Sphk1 gene expression and activity in an in vitro PD model, induced by 1-methyl-4-phenylpyridinium (MPP+). The aim of this study was to investigate the role of Sphks inhibition in ASN secretion and in the molecular mechanism of neuronal death in the PD model. Our study was carried out using neuronal dopaminergic SH-SY5Y control cells, transfected with the human gene for ASN or with an empty vector. These cells were treated with MPP+ (1-3 mM), which represents an experimental PD model, or with the Sphks inhibitor (1-5 µM SKI II) for 3-24 h. Our data indicated that MPP+ (3 mM) induced significant alterations of Sphks and S1P lyase (SPL) gene expression. Reduced activity of Sphk1 and Sphk2 in the cytosolic fraction and in the crude nuclear fraction, respectively, was observed. Sphks inhibition evoked enhancement of ASN secretion, suppression of PI3K/Akt phosphorylation and activation of gene expression for the pro-apoptotic Bcl-2 proteins Bax and BH3-only protein Harakiri. Moreover, a lower level of cytochrome c in the mitochondrial fraction and caspase-dependent degradation of DNA-bound enzyme poly(ADP-ribose) polymerase (PARP-1) were observed. The caspase inhibitor (20 µM Z-VAD-FMK) significantly enhanced neuronal cell viability in MPP+ oxidative stress. However, exogenous S1P (1 µM) exerted a more efficient neuroprotective effect as compared to Z-VAD-FMK. In summary, these data indicated that Sphk1 inhibition plays an important role in caspase-dependent apoptotic neuronal death in an experimental PD model.


Assuntos
Apoptose/fisiologia , Neurônios/enzimologia , Doença de Parkinson/enzimologia , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Humanos , Imuno-Histoquímica , Estresse Oxidativo/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Transcriptoma , Transfecção , alfa-Sinucleína/metabolismo
3.
Mol Neurobiol ; 50(1): 38-48, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24399507

RESUMO

Sphingosine kinases (Sphk1/2) are crucial enzymes in regulation of the biostat between sphingosine-1-phosphate (S1P) and ceramide and play an important role in the pathogenesis/pathomechanism of Alzheimer's disease (AD). These enzymes synthesise S1P, which regulates neurotransmission, synaptic function and neuron cell proliferation, by activating five G protein-coupled receptors (S1P1-5). However, S1P synthesised by Sphk2 could be involved in amyloid ß (Aß) release by stimulation of Aß precursor protein degradation. The significance of this bioactive sphingolipid in the pathogenesis of Parkinson's disease (PD) is unknown. The aim of our study was to investigate the expression level of Sphk1 and its role in human dopaminergic neuronal cell (SH-SY5Y) viability under oxidative stress, evoked by 1-methyl-4-phenylpyridinium (MPP+). Moreover, the mechanism of S1P action on the death signalling pathway in these experimental conditions was evaluated. Our study indicated marked downregulation of Sphk1 expression in this cellular PD model. Inhibition of Sphk1 decreased SH-SY5Y cell viability and concomitantly enhanced the reactive oxygen species (ROS) level. It was found that exogenous S1P (1 µM) exerted the neuroprotective effect by activation of Sphk1 and S1P1 receptor gene expression. Moreover, S1P downregulated Bax and harakiri, death protein 5 (Hrk/DP5) expression and enhanced cell viability in MPP+-treated cells. The neuroprotective mechanism of S1P is mainly dependent on S1P1 receptor signalling, which was indicated by using specific agonists and antagonists of S1P1 receptor. The results show that S1P and S1P1 receptor agonists protected a significant population of neuronal cells against death.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Neurônios Dopaminérgicos/metabolismo , Lisofosfolipídeos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Esfingosina/análogos & derivados , Linhagem Celular Tumoral , Ceramidas/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Humanos , Doença de Parkinson/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo
4.
Neurochem Int ; 62(5): 626-36, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23439385

RESUMO

The human genome is exposed to oxidative/genotoxic stress by several endogenous and exogenous compounds. These events evoke DNA damage and activate poly(ADP-ribose) polymerase-1 (PARP-1), the key enzyme involved in DNA repair. The massive stress and over-activation of this DNA-bound enzyme can be responsible for an energy crisis and neuronal death. The last data indicated that product of PARP-1, i.e. poly(ADP-ribose) (PAR), acts as a signalling molecule and plays a significant role in nucleus-mitochondria cross-talk. PAR translocated to the mitochondria can be involved in mitochondrial permeability, the release of an apoptosis-inducing factor (AIF). Its translocation into the nucleus leads to chromatin condensation, fragmentation and cell death. The exact mechanism of this novel death pathway has not yet fully been understood. In this study the relationship between AIF and PARP/PAR in death signalling in the neuronal cell line (HT22) subjected to oxidative/genotoxic stress evoked by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) was explored. The neuroprotective influence of docosahexaenoic acid (DHA), major dietary ω-3 long-chain polyunsaturated fatty acids as well as the action of tetracyclines, the novel suppressors of PARP-1, were examined. The effect of these all compounds was compared with specific PARP-1 inhibitors. The oxidative/genotoxic stress evoked by MNNG enhanced the level of PAR in a time-dependent manner with a concomitant significant decrease in the mitochondrial AIF protein level. Moreover, the down-regulation of the anti-apoptotic proteins (Bcl-2 and Bcl-xL) and the up-regulation of the Bax pro-apoptotic protein were presented. In these conditions massive HT22 cell death was observed. Both PARP-1 inhibitors: 3-aminobenzamide (3-AB) and PJ 34, tetracycline: doxocycline and minocycline, as well as DHA protected the cells against PAR formation and AIF translocation. Moreover, all of these compounds enhanced Bcl-xL gene expression and protected the cells against MNNG-induced death. Our data show that both DHA and tetracyclines offer a novel neuroprotective strategy for oxidative/genotoxic stress treatment.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Mutagênicos/toxicidade , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Poli(ADP-Ribose) Polimerases , Tetraciclinas/farmacologia , Animais , Linhagem Celular Transformada , Hipocampo/efeitos dos fármacos , Hipocampo/enzimologia , Hipocampo/metabolismo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA