Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Immunopathol Pharmacol ; 24(3): 735-47, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21978706

RESUMO

The present study aims to assess the protective role of the antioxidant enzyme catalase (CAT) with relation to hydrogen peroxide (H(2)O(2)) degradation in oxygen plus water on electrophysiological and fluorescence changes induced by in vitro ischemia and on brain damage produced by transient in vivo ischemia. Neuroprotective effects of CAT were determined by means of electrophysiological recordings and confocal fluorescence microscopy in the hippocampal slice preparation. Ischemia was simulated in vitro by oxygen/glucose deprivation (OGD). In vivo ischemia was produced by transient middle cerebral artery occlusion (MCAo). A protection of the rat CA1 field excitatory postsynaptic potential (fEPSP) loss caused by a prolonged OGD (40 min) was observed after exogenous CAT (500 U/mL) bath-applied before a combined exposure to OGD and H(2)O(2) (3 mM). Of note, neither H(2)O(2) nor exogenous CAT alone had a protective action when OGD lasted for 40 min. The CAT-induced neuroprotection was confirmed in a transgenic mouse model over-expressing human CAT [Tg(CAT)]. In the presence of H(2)O(2), the hippocampus of Tg(CAT) showed an increased resistance against OGD compared to that of wild-type (WT) animals. Moreover, CAT treatment reduced for about 50 min fEPSP depression evoked by repeated applications of H(2)O(2) in normoxia. A lower sensitivity to H(2)O(2)-induced depression of fEPSPs was also indicated by the rightward shift of concentration-response curve in Tg(CAT) compared to WT mice. Noteworthy, Tg(CAT) mice had a reduced infarct size after MCAo. Our data suggest new strategies to reduce neuronal damage produced by transient brain ischemia through the manipulation of CAT enzyme.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/enzimologia , Catalase/fisiologia , Catalase/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Animais , Encéfalo/enzimologia , Encéfalo/patologia , Isquemia Encefálica/patologia , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/metabolismo , Catalase/biossíntese , Infarto Cerebral/patologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Glucose/deficiência , Peróxido de Hidrogênio/farmacologia , Hipóxia Encefálica/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Oxidantes/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética , Superóxidos/metabolismo , Transmissão Sináptica/efeitos dos fármacos
2.
Br J Pharmacol ; 153(5): 1022-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18223675

RESUMO

BACKGROUND AND PURPOSE: Reactive oxygen species (ROS) have been postulated to play a crucial role in the pathogenesis of ischaemia-reperfusion injury. Among these, hydrogen peroxide (H(2)O(2)) is known to be a toxic compound responsible for free-radical-dependent neuronal damage. In recent years, however, the 'bad reputation' of H(2)O(2) and other ROS molecules has changed. The aim of this study was to assess the protective role of H(2)O(2) and modification in its endogenous production on the electrophysiological and morphological changes induced by oxygen/glucose deprivation (OGD) on CA1 hippocampal neurons. EXPERIMENTAL APPROACH: Neuroprotective effects of exogenous and endogenous H(2)O(2) were determined using extracellular electrophysiological recordings of field excitatory post synaptic potentials (fEPSPs) and morphological studies in a hippocampal slice preparation. In vitro OGD was delivered by switching to an artificial cerebrospinal fluid solution with no glucose and with oxygen replaced by nitrogen. KEY RESULTS: Neuroprotection against in vitro OGD was observed in slices treated with H(2)O(2) (3 mM). The rescuing action of H(2)O(2) was mediated by catalase as pre-treatment with the catalase inhibitor 3-amino-1,2,4-triazole blocked this effect. More interestingly, we showed that an increase of the endogenous levels of H(2)O(2), due to a combination of an inhibitor of the glutathione peroxidase enzyme and addition of Cu,Zn-superoxide dismutase in the tissue bath, prevented the OGD-induced irreversible depression of fEPSPs. CONCLUSIONS AND IMPLICATIONS: Taken together, our results suggest new possible strategies to lessen the damage produced by a transient brain ischaemia by increasing the endogenous tissue level of H(2)O(2).


Assuntos
Isquemia Encefálica/tratamento farmacológico , Peróxido de Hidrogênio/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Isquemia Encefálica/fisiopatologia , Catalase/efeitos dos fármacos , Catalase/metabolismo , Modelos Animais de Doenças , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Peróxido de Hidrogênio/metabolismo , Técnicas In Vitro , Masculino , Fármacos Neuroprotetores/metabolismo , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Ratos , Ratos Wistar
3.
Nat Cell Biol ; 3(11): 1030-3, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11715026

RESUMO

Genetic and biochemical evidence have led to the suggestion that presenilins could be the long-searched-for gamma-secretase, the proteolytic activity that generates the carboxy terminus of amyloid beta-peptides. This activity is also thought to be responsible for the release of the Notch intracellular domain (NICD) from Notch. Here, we report the production of endogenous secreted and intracellular 40- and 42-amino-acid Abeta peptides in mouse fibroblasts deficient in presenilin 1, presenilin 2 or both. We show that the endogenous production of Abeta40 and Abeta42 was not altered by presenilin deficiency. By contrast, inactivating presenilin genes fully abolished NICD production. These data indicate that Abeta and NICD production are distinct catabolic events. Also, even though NICD formation is indeed presenilin dependent, endogenous secreted and intracellular beta-amyloid peptides are still generated in absence of presenilins, indicating that there is a gamma-secretase activity distinct from presenilins, at least in murine fibroblasts.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Proteínas de Membrana/biossíntese , Proteínas de Membrana/fisiologia , Fragmentos de Peptídeos/biossíntese , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Presenilina-1 , Presenilina-2 , Receptores Notch
5.
Eur J Pharmacol ; 397(2-3): 291-6, 2000 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-10844126

RESUMO

An N-terminal dopamine D(2s) receptor clone was constructed and coexpressed in COS-7 cells together with a separate gene fragment coding for the C-terminal sequence of the dopamine D(2s) receptor. The truncated receptor (referred to as D(2trunc)) contained transmembrane domains I-V and the N-terminal portion of the third cytoplasmic loop, whereas the C-terminal receptor fragment (referred to as D(2tail)) contained transmembrane domains VI and VII and the adjacent intra- and extracellular sequences of the dopamine D(2s) receptor. Expression in COS-7 cells of either of these two polypeptides alone did not result in any detectable [3H]methylspiperone binding activity. However, specific [3H]methylspiperone binding could be observed after coexpression of the D(2trunc) and D(2tail) gene constructs; the number of receptors present on the plasma membrane was about 10% with respect to that of the wild type. The binding properties of the coexpressed fragments were similar to those of the wild-type dopamine D(2s) receptor for agonists and antagonists. Functional stimulation of the cotransfected D(2trunc) and D(2tail) fragments with quinpirole resulted in the inhibition of adenylate cyclase activity. Maximal inhibition corresponds to a 28% decrease in forskolin-stimulated adenylate cyclase. The apparent IC(50) of quinpirole was 5.1+/-0.3 mcM. These findings confirm and extend analogous data for other G protein-coupled receptors and indicate that this phenomenon is of general importance for the entire family of these proteins.


Assuntos
Fragmentos de Peptídeos/fisiologia , Receptores de Dopamina D2/fisiologia , Animais , Apomorfina/farmacologia , Ligação Competitiva/efeitos dos fármacos , Células COS , Clozapina/farmacologia , Colforsina/farmacologia , AMP Cíclico/metabolismo , Dopamina/farmacologia , Agonistas de Dopamina/metabolismo , Agonistas de Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Expressão Gênica , Haloperidol/farmacologia , Membranas/metabolismo , Fragmentos de Peptídeos/efeitos dos fármacos , Fragmentos de Peptídeos/genética , Pergolida/farmacologia , Quimpirol/farmacologia , Ensaio Radioligante , Ratos , Receptores de Dopamina D2/efeitos dos fármacos , Receptores de Dopamina D2/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/fisiologia , Espiperona/análogos & derivados , Espiperona/metabolismo , Espiperona/farmacologia , Transfecção , Trítio
7.
Neurotox Res ; 1(4): 285-97, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12835096

RESUMO

Epidemiological studies have shown a reduced incidence of cancer in Parkinson's disease. Since nearly all parkinsonian patients with clinical impairment are treated with L-beta-3,4-dihydroxyphenylalanine (L-DOPA) and dopamine (DA)ergic agonists, a possibility exists that these therapeutic agents can influence the risk of cancer. We studied the antiproliferative effect of these therapeutic agents (and substances structurally correlated) on Chinese hamster ovary (CHO)-K1 cell growth. Among the compounds tested, apomorphine proved to be the most potent inhibitor of CHO-K1 cell growth, with an EC(50) of 3.35 +/- 0.12 micro M. The apomorphine analogues, apocodeine and hydroxyethylnorapomorphine, were less active as inhibitors of CHO-K1 cell growth. The activity of DA, 6-hydroxydopamine (6-OHDA), phenylethylamine (PEA), L-DOPA and bromocriptine as antiproliferative was one order of magnitude lower than that of apomorphine while pergolide was ineffective. To test whether or not the oxidative potential of these compounds was important for their antiproliferative effect, several antioxidants were assayed. Among them glutathione (GSH) and dithiothreitol (DTT) were effective in reversing the anti-proliferative effect of apomorphine, DA, 6-OHDA and PEA, conversely they did not work with bromocriptine. GSH and DTT are sulphydryl-reducing agents; while their effect could explain the efficacy against apomorphine, DA and 6-OHDA, it is difficult to understand why they should have any effect on PEA as this substance does not react with sulphydryl groups. The oxidative potential as a mechanism of action was also questioned by the results obtained with dihydrorhodamine 123, a probe that changes its fluorescent emission wave when oxidized. None of the compounds, with the exception of 6-OHDA, had any effect on the fluorescent emission wave of the probe at the maximal concentrations used to inhibit CHO-K1 cell growth. At concentrations five times higher, apomorphine and DA generated reactive oxygen species but PEA and bromocriptine did not. These data demonstrate that the antiproliferative effect of these compounds is not due to their oxidative potential, but another mechanism must be postulated.

8.
J Neural Transm Suppl ; 55: 47-55, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10335492

RESUMO

Apomorphine is a potent non selective agonist at the D1 and D2 dopamine receptors acting both pre- and post-synaptically. In this report we describe a novel function of apomorphine, independent from its dopaminergic activity. Apomorphine inhibits Chinese hamster ovary (CHO)-K1 cell proliferation in a dose-dependent manner. The EC50 of apomorphine-induced inhibition of CHO-K1 cell proliferation determined by cell counting was 3.24 +/- 0.07 microM. Remarkably, the dose-response curve obtained by measuring the incorporation of [3H]thymidine was practically identical to the previous one giving an EC50 of 3.52 +/- 0.04 microM. The dopaminergic antagonists SCH23390 and spiperone at a concentration of 10 microM (well beyond their Kd values for the dopamine D1- and D2-like receptors respectively) were not able to antagonize the effect of apomorphine on CHO-K1 cell proliferation. Apomorphine exerts its effect early during incubation; CHO-K1 cells exposed to apomorphine for a period as short as 1 h and then allowed to grow for three days were significantly reduced in number with respect to untreated control cells. After four hours of exposition to apomorphine (10 microM) the antiproliferative effect was similar to that seen when this compound was present in the bath for all three days. Concentrations of apomorphine higher than 10 microM induced cell death, and the colony was completely destroyed at 50 microM. Cytometric analyses showed a significant accumulation of CHO-K1 cells in the G2/M phase.


Assuntos
Apomorfina/farmacologia , Animais , Células CHO , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Cricetinae
9.
J Neurochem ; 71(6): 2439-46, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9832142

RESUMO

The repeated finding of an apparent protective effect of cigarette smoking on the risk of Parkinson's disease is one of the few consistent results in the epidemiology of this disorder. Among the numerous substances that originate from tobacco smoke, nicotine is by far the most widely studied. Nicotine is a natural alkaloid that has considerable stimulatory effects on the CNS. Its effects on the CNS are mediated by the activation of neuronal heteromeric acetylcholine-gated ion channel receptors (nAChRs, also termed nicotinic acetylcholine receptors). In the present study, we describe the neuroprotective effects of (-)-nicotine in two animal models of parkinsonism: diethyldithiocarbamate-induced enhancement of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity in mice and methamphetamine-induced neurotoxicity in rats and mice. The neuroprotective effect of (-)-nicotine was very similar to that of the noncompetitive NMDA receptor antagonist (+)-MK-801. In parallel experiments, we found that (-)-nicotine induces the basic fibroblast growth factor-2 (FGF-2) and the brain-derived neurotrophic factor in rat striatum. The effect of (-)-nicotine on the induction of FGF-2 was prevented by the nAChR antagonist mecamylamine. We also found that (+)-MK-801 was able to induce FGF-2 in the striatum. As trophic factors have been reported to be neuroprotective for dopaminergic cells, our data suggest that the increase in neurotrophic factors is a possible mechanism by which (-)-nicotine protects from experimental parkinsonisms.


Assuntos
Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Fatores de Crescimento Neural/metabolismo , Nicotina/farmacologia , Doença de Parkinson/prevenção & controle , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Células Cultivadas , Dopamina/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Imuno-Histoquímica , Masculino , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Tirosina 3-Mono-Oxigenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...