Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Pineal Res ; 58(1): 86-106, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25407782

RESUMEN

We have demonstrated that mitochondrial oxidative damage and PKCδ overexpression contribute to methamphetamine-induced dopaminergic degeneration. Although it is recognized that antioxidant melatonin is effective in preventing neurotoxicity induced by methamphetamine, its precise mechanism remains elusive. C57BL/6J wild-type mice exhibited a similar degree of dopaminergic deficit when methamphetamine was administered during light and dark phases. Furthermore, dopaminergic neuroprotection by genetic inhibition of PKCδ during the light phase was comparable to that during the dark phase. Thus, we have focused on the light phase to examine whether melatonin modulates PKCδ-mediated neurotoxic signaling after multiple high doses of methamphetamine. To enhance the bioavailability of melatonin, we applied liposomal melatonin. Treatment with methamphetamine resulted in hyperthermia, mitochondrial translocation of PKCδ, oxidative damage (mitochondria > cytosol), mitochondrial dysfunction, pro-apoptotic changes, ultrastructural mitochondrial degeneration, dopaminergic degeneration, and behavioral impairment in wild-type mice. Treatment with liposomal melatonin resulted in a dose-dependent attenuation against degenerative changes induced by methamphetamine in wild-type mice. Attenuation by liposomal melatonin might be comparable to that by genetic inhibition (using PKCδ((-/-)) mice or PKCδ antisense oligonucleotide). However, liposomal melatonin did not show any additional protective effects on the attenuation by genetic inhibition of PKCδ. Our results suggest that the circadian cycle cannot be a key factor in modulating methamphetamine toxicity under the current experimental condition and that PKCδ is one of the critical target genes for melatonin-mediated protective effects against mitochondrial burdens (dysfunction), oxidative stress, pro-apoptosis, and dopaminergic degeneration induced by methamphetamine.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de Captación de Dopamina/efectos adversos , Melatonina/farmacología , Metanfetamina/efectos adversos , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Proteína Quinasa C-delta/antagonistas & inhibidores , Animales , Dopamina/genética , Dopamina/metabolismo , Inhibidores de Captación de Dopamina/farmacología , Liposomas , Metanfetamina/farmacología , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Neurodegenerativas/inducido químicamente , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/genética , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo
2.
J Appl Toxicol ; 35(8): 927-44, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25523949

RESUMEN

Recently, we have demonstrated that ginsenoside Re protects methamphetamine (MA)-induced dopaminergic toxicity in mice via genetic inhibition of PKCδ and attenuation of mitochondrial stress. In addition, we have reported that induction of mitochondrial glutathione peroxidase (GPx) is also important for neuroprotection mediated by ginsenoside Re. To extend our knowledge, we examined the effects of ginsenoside Re against MA toxicity in vitro condition using SH-SY5Y neuroblastoma cells. Treatment with ginsenoside Re resulted in significant attenuations against a decrease in the activity of GPx and an increase in the activity of superoxide dismutase (SOD) in the cytosolic and mitochondrial fraction. The changes in glutathione (GSH) paralleled those in GPx in the same experimental condition. Consistently, ginsenoside Re treatment exhibited significant protections against cytosolic and mitochondrial oxidative damage (i.e. lipid peroxidation and protein oxidation), mitochondrial translocation of PKCδ, mitochondrial dysfunction (mitochondrial transmembrane potential and intra-mitochondrial Ca(2+)), apoptotic events [i.e., cytochrome c release from mitochondria, cleavage of caspase-3 and poly(ADP-ribose)polymerase-1, nuclear condensation, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive apoptotic cells], and a reduction in the tyrosine hydroxylase (TH) expression and TH activity induced by MA in SH-SY5Y neuroblastoma cells. These protective effects of ginsenoside Re were comparable to those of PKCδ antisense oligonucleotide (ASO). However, ginsenoside Re did not significantly provide additional protective effects mediated by genetic inhibition of PKCδ. Our results suggest that PKCδ is a specific target for ginsenoside Re-mediated protective activity against MA toxicity in SH-SY5Y neuroblastoma cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/toxicidad , Ginsenósidos/farmacología , Metanfetamina/antagonistas & inhibidores , Metanfetamina/toxicidad , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteína Quinasa C-delta/genética , Antioxidantes/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Citosol/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Tirosina 3-Monooxigenasa/metabolismo
3.
J Pharmacol Sci ; 113(4): 404-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20644336

RESUMEN

Parishin C, a major component of Gastrodia elata BLUME (GE), was purified from GE. Because GE modulates the serotonergic system and the 5-HT(1A) receptor is an important therapeutic target of schizophrenia, we examined whether parishin C affects phencyclidine-induced abnormal behaviors in mice. Phencyclidine-induced abnormal behaviors were significantly ameliorated by parishin C. These effects were reversed by WAY 100635, a 5HT(1A)-receptor antagonist. Consistently, parishin C showed high affinity at 5-HT(1A) receptor as well as a 5-HT(1A)-agonist activity in a 8-OH-DPAT-stimulated [(35)S]GTP-gammaS binding assay. Our results suggest that the antipsychotic effects of parishin C require activation of 5-HT(1A) receptors.


Asunto(s)
Citratos/farmacología , Glucósidos/farmacología , Fenciclidina/toxicidad , Trastornos Psicóticos/prevención & control , Receptor de Serotonina 5-HT1A/fisiología , Esquizofrenia/prevención & control , Animales , Ratones
4.
Free Radic Res ; 50(4): 467-83, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26930476

RESUMEN

We investigated the possible roles of angiotensin II type 1 receptor (AT1R) and oxidative stress responsive nuclear factor κB (NFκB) in renal damage caused by multiple doses of cocaine in glutathione peroxidase (GPx)-1 gene-depleted mice. Treatment with cocaine resulted in significant increases in malondialdehyde, protein carbonyl, and pro-apoptotic Bax expression and decreases in the ratio of glutathione (GSH) and its oxidized form (GSSG), GSH-dependent enzymes, and anti-apoptotic factors in the kidney. These alterations were more pronounced in GPx-1 knockout (-/-) mice than in wild type (WT) mice. Notably, the AT1R antagonist losartan protected against the renal toxicity induced by cocaine, whereas the NFκB inhibitor pyrrolidine dithiocarbamate was not protective. The toxicity was more pronounced in GPx-1 (-/-) mice than in WT mice. The protective effect afforded by losartan against cocaine toxicity appeared to be more sensitive in GPx-1 (-/-) mice than that in WT mice. These losartan-mediated protective effects were inhibited by the phosphatidyl-inositol-3-kinase (PI3K) inhibitor LY294002, indicating that losartan provides significant protection from cocaine-induced renal toxicity through PI3K/Akt signaling. Our results suggest that genetic inhibition of GPx-1 potentiates cocaine-induced renal damage via activation of AT1R by inhibition of PI3K-Akt signaling, and that AT1R can be a therapeutic target against renal toxicity induced by cocaine.


Asunto(s)
Cocaína/toxicidad , Glutatión Peroxidasa/genética , Riñón/efectos de los fármacos , Losartán/farmacología , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Receptor de Angiotensina Tipo 1/genética , Animales , Cromonas/farmacología , Cocaína/antagonistas & inhibidores , Femenino , Regulación de la Expresión Génica , Glutatión/metabolismo , Glutatión Peroxidasa/deficiencia , Riñón/metabolismo , Riñón/patología , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Noqueados , Morfolinas/farmacología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Carbonilación Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirrolidinas/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Tiocarbamatos/farmacología , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , Glutatión Peroxidasa GPX1
5.
Mol Neurobiol ; 49(3): 1400-21, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24430743

RESUMEN

Ginsenoside Re, one of the main constituents of Panax ginseng, possesses novel antioxidant and anti-inflammatory properties. However, the pharmacological mechanism of ginsenoside Re in dopaminergic degeneration remains elusive. We suggested that protein kinase C (PKC) δ mediates methamphetamine (MA)-induced dopaminergic toxicity. Treatment with ginsenoside Re significantly attenuated methamphetamine-induced dopaminergic degeneration in vivo by inhibiting impaired enzymatic antioxidant systems, mitochondrial oxidative stress, mitochondrial translocation of protein kinase Cδ, mitochondrial dysfunction, pro-inflammatory microglial activation, and apoptosis. These protective effects were comparable to those observed with genetic inhibition of PKCδ in PKCδ knockout (-/-) mice and with PKCδ antisense oligonucleotides, and ginsenoside Re did not provide any additional protective effects in the presence of PKCδ inhibition. Our results suggest that PKCδ is a critical target for ginsenoside Re-mediated protective activity in response to dopaminergic degeneration induced by MA.


Asunto(s)
Neuronas Dopaminérgicas/efectos de los fármacos , Ginsenósidos/farmacología , Metanfetamina/toxicidad , Microglía/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteína Quinasa C-delta/antagonistas & inhibidores , Animales , Neuronas Dopaminérgicas/metabolismo , Ginsenósidos/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Mitocondrias/metabolismo , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Estrés Oxidativo/fisiología , Proteína Quinasa C-delta/metabolismo , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología
6.
Behav Brain Res ; 250: 351-60, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23714077

RESUMEN

Senescence-accelerated mouse prone 8 (SAMP8) is a model of aging characterized by the early onset of learning and memory impairment and various pathological features of Alzheimer's disease (AD). Our recent studies have demonstrated that nobiletin, a polymethoxylated flavone from citrus peels, ameliorates learning and memory impairment in olfactory-bulbectomized mice, amyloid precursor protein transgenic mice, and NMDA receptor antagonist-treated mice. Here, we present evidence that this natural compound improves age-related cognitive impairment and reduces oxidative stress and tau phosphorylation in SAMP8 mice. Treatment with nobiletin (10 or 50mg/kg) reversed the impairment of recognition memory and context-dependent fear memory in SAMP8 mice. Treatment with nobiletin also restored the decrease in the GSH/GSSG ratio in the brain of SAMP8 mice. In addition, increases in glutathione peroxidase and manganese-superoxide dismutase activities, as well as a decrease in protein carbonyl level, were observed in the brain of nobiletin-treated SAMP8 mice. Furthermore, nobiletin reduced tau phosphorylation in the hippocampus of SAMP8 mice. Together, the markedly beneficial effects of nobiletin represent a potentially useful treatment for ameliorating the learning and memory deficits, oxidative stress, and hyperphosphorylation of tau in aging as well as age-related neurodegenerative diseases such as AD.


Asunto(s)
Envejecimiento/efectos de los fármacos , Antioxidantes/uso terapéutico , Trastornos del Conocimiento/tratamiento farmacológico , Flavonas/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Proteínas tau/metabolismo , Envejecimiento/genética , Envejecimiento/fisiología , Análisis de Varianza , Animales , Antioxidantes/química , Antioxidantes/farmacología , Peso Corporal/efectos de los fármacos , Peso Corporal/genética , Estudios de Casos y Controles , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Conducta Exploratoria/efectos de los fármacos , Miedo/efectos de los fármacos , Miedo/fisiología , Flavonas/química , Flavonas/farmacología , Glutatión/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Carbonilación Proteica/efectos de los fármacos , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo
7.
Exp Neurobiol ; 20(3): 130-6, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22110371

RESUMEN

We previously demonstrated that repeated exposure to extremely low frequency magnetic fields (ELF-MF) increases locomotor activity via stimulation of dopaminergic D1 receptor (J. Pharmacol. Sci., 2007;105:367-371). Since it has been demonstrated that activator protein-1 (AP-1) transcription factors, especially 35-kDa fos-related antigen (FRA), play a key role in the neuronal and behavioral adaptation in response to various stimuli, we examined whether repeated ELF-MF exposure induces FRA-immunoreactivity (FRA-IR) in the striatum and nucleus accumbens (striatal complex) of the mice. Repeated exposure to ELF-MF (0.3 or 2.4 mT, 1 h/day, for consecutive fourteen days) significantly induced hyperlocomotor activity and FRA-IR in the striatal complex in a field intensity-dependent manner. ELF-MF-induced FRA-IR lasted for at least 1 year, while locomotor activity returned near control level 3 months after the final exposure to ELF-MF. Pretreatment with SCH23390, a dopaminergic D1 receptor antagonist, but not with sulpiride, a dopaminergic D2 receptor antagonist, significantly attenuated hyperlocomotor activity and FRA-IR induced by ELF-MF. Our results suggest that repeated exposure to ELF-MF leads to prolonged locomotor stimulation and long-term expression of FRA in the striatal complex of the mice via stimulation of dopaminergic D1 receptor.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA