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











Base de dados
Intervalo de ano de publicação
1.
Eur Neuropsychopharmacol ; 24(10): 1698-708, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25108314

RESUMO

We recently identified ASP5736, (N-(diaminomethylene)-1-(3,5-difluoropyridin-4-yl)-4-fluoroisoquinoline-7-carboxamide (2E)-but-2-enedioate), a novel antagonist of 5-HT5A receptor, and here describe the in vitro and in vivo characterization of this compound. ASP5736 exhibited a high affinity for the human 5-HT5A receptor (Ki = 3.6 ± 0.66 nM) and antagonized 5-carboxamidotryptamine (5-CT)-induced Ca(2+) influx in human cells stably expressing the 5-HT5A receptor with approximately 200-fold selectivity over other receptors, including other 5-HT receptor subtypes, enzymes, and channels except human 5-HT2c receptor (Ki = 286.8 nM) and 5-HT7 receptor (Ki = 122.9 nM). Further, ASP5736 dose-dependently antagonized the 5-CT-induced decrease in cAMP levels in HEK293 cells stably expressing the 5-HT5A receptor. We then evaluated the effects of ASP5736 on cognitive impairments in several animal models of schizophrenia. Working memory deficit in MK-801-treated mice and visual learning deficit in neonatally phencyclidine (PCP)-treated mice were both ameliorated by ASP5736. In addition, ASP5736 also attenuated MK-801- and methamphetamine (MAP)-induced hyperactivity in mice without causing sedation, catalepsy, or plasma prolactin increase. The addition of olanzapine did not affect ASP5736-induced cognitive enhancement, and neither the sedative nor cataleptogenic effects of olanzapine were worsened by ASP5736. These results collectively suggest that ASP5736 is a novel and potent 5-HT5A receptor antagonist that not only ameliorates positive-like symptoms but also cognitive impairments in animal models of schizophrenia, without adverse effects. Present studies also indicate that ASP5736 holds potential to satisfy currently unmet medical needs for the treatment of schizophrenia by either mono-therapy or co-administered with commercially available antipsychotics.


Assuntos
Antipsicóticos/farmacologia , Guanidinas/farmacologia , Isoquinolinas/farmacologia , Esquizofrenia/tratamento farmacológico , Antagonistas da Serotonina/farmacologia , Animais , Antipsicóticos/química , Antipsicóticos/farmacocinética , Cálcio/metabolismo , Catalepsia/tratamento farmacológico , Catalepsia/fisiopatologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/fisiopatologia , AMP Cíclico/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Guanidinas/química , Guanidinas/farmacocinética , Células HEK293 , Humanos , Isoquinolinas/química , Isoquinolinas/farmacocinética , Masculino , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos ICR , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Receptor 5-HT2C de Serotonina/metabolismo , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Esquizofrenia/fisiopatologia , Antagonistas da Serotonina/química , Antagonistas da Serotonina/farmacocinética
2.
Neuropharmacology ; 79: 412-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24373902

RESUMO

γ-Secretase is the enzyme responsible for the intramembranous proteolysis of various substrates, such as amyloid precursor protein (APP) and Notch. Amyloid-ß peptide 42 (Aß42) is produced through the sequential proteolytic cleavage of APP by ß- and γ-secretase and causes the synaptic dysfunction associated with memory impairment in Alzheimer's disease. Here, we identified a novel cyclohexylamine-derived γ-secretase modulator, {(1R*,2S*,3R*)-3-[(cyclohexylmethyl)(3,3-dimethylbutyl)amino]-2-[4-(trifluoromethyl)phenyl]cyclohexyl}acetic acid (AS2715348), that may inhibit this pathological response. AS2715348 was seen to reduce both cell-free and cellular production of Aß42 without increasing levels of APP ß-carboxyl terminal fragment or inhibiting Notch signaling. Additionally, the compound increased Aß38 production, suggesting a shift of the cleavage site in APP. The inhibitory potency of AS2715348 on endogenous Aß42 production was similar across human, mouse, and rat cells. Oral administration with AS2715348 at 1 mg/kg and greater significantly reduced brain Aß42 levels in rats, and no Notch-related toxicity was observed after 28-day treatment at 100 mg/kg. Further, AS2715348 significantly ameliorated cognitive deficits in APP-transgenic Tg2576 mice. Finally, AS2715348 significantly reduced brain Aß42 levels in cynomolgus monkeys. These findings collectively show the promise for AS2715348 as a potential disease-modifying drug for Alzheimer's disease.


Assuntos
Acetatos/farmacologia , Doença de Alzheimer/tratamento farmacológico , Secretases da Proteína Precursora do Amiloide/metabolismo , Encéfalo/efeitos dos fármacos , Cicloexilaminas/farmacologia , Fármacos Neuroprotetores/farmacologia , Acetatos/efeitos adversos , Acetatos/farmacocinética , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Linhagem Celular Tumoral , Cognição/efeitos dos fármacos , Cicloexilaminas/efeitos adversos , Cicloexilaminas/farmacocinética , Modelos Animais de Doenças , Feminino , Humanos , Macaca fascicularis , Masculino , Camundongos , Camundongos Transgênicos , Estrutura Molecular , Fármacos Neuroprotetores/efeitos adversos , Fármacos Neuroprotetores/farmacocinética , Nootrópicos/efeitos adversos , Nootrópicos/química , Nootrópicos/farmacologia , Fragmentos de Peptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Notch/metabolismo
3.
J Neurosci ; 32(6): 2037-50, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22323718

RESUMO

γ-Secretase inhibitors (GSIs) reduce amyloid-ß (Aß) peptides but inevitably increase the ß-C-terminal fragment (ß-CTF) of amyloid precursor protein (APP), potentially having undesirable effects on synapses. In contrast, γ-secretase modulators (GSMs) reduce Aß42 without increasing ß-CTF. Although the Aß-lowering effects of these compounds have been extensively studied, little effort has been made to investigate their effects on cognition. Here, we compared the effects of two GSIs--(2S)-2-hydroxy-3-methyl-N-[(2S)-1-{[(1S)-3-methyl-2-oxo-2,3,4,5-tetrahydro-1H-3-benzazepin-1-yl]amino}-1-oxopropan-2-yl]butanamide (LY450139, semagacestat) and (2R)-2-[[(4-chlorophenyl)sulfonyl][[2-fluoro-4-(1,2,4-oxazol-3-yl)phenyl]methyl]amino-5,5,5-trifluoropentanamide (BMS-708163)--and a second-generation GSM [{(2S,4R)-1-[(4R)-1,1,1-trifluoro-7-methyloctan-4-yl]-2-[4-(trifluoromethyl)phenyl]piperidin-4-yl}acetic acid (GSM-2)] on spatial working memory in APP-transgenic (Tg2576) and nontransgenic mice using the Y-maze task. While acute dosing with either GSI ameliorated memory deficits in 5.5-month-old Tg2576 mice, these effects disappeared after 8 d subchronic dosing. Subchronic dosing with either GSI rather impaired normal cognition in 3-month-old Tg2576 mice, with no inhibition on the processing of other γ-secretase substrates, such as Notch, N-cadherin, or EphA4, in the brain. LY450139 also impaired normal cognition in wild-type mice; however, the potency was 10-fold lower than that in Tg2576 mice, indicating an APP-dependent mechanism likely with ß-CTF accumulation. Immunofluorescence studies revealed that the ß-CTF accumulation was localized in the presynaptic terminals of the hippocampal stratum lucidum and dentate hilus, implying an effect on presynaptic function in the mossy fibers. In contrast, both acute and subchronic dosing with GSM-2 significantly ameliorated memory deficits in Tg2576 mice and did not affect normal cognition in wild-type mice. We demonstrated a clear difference between GSI and GSM in effects on functional consequences, providing new insights into strategies for developing these drugs against Alzheimer's disease.


Assuntos
Alanina/análogos & derivados , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/fisiologia , Precursor de Proteína beta-Amiloide/fisiologia , Azepinas/farmacologia , Cognição/efeitos dos fármacos , Cognição/fisiologia , Inibidores de Proteases/farmacologia , Alanina/farmacologia , Precursor de Proteína beta-Amiloide/genética , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos
4.
Behav Brain Res ; 216(2): 561-8, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-20816897

RESUMO

Body weight gain is one of the most serious side effects associated with clinical use of antipsychotics. However, the mechanisms by which antipsychotics induce body weight gain are unknown, and no reliable animal models of antipsychotics-induced weight gain have been established. The present studies were designed to establish male rat models of weight gain induced by chronic and acute treatment with antipsychotics. Six-week chronic treatment with olanzapine (5, 7.5, and 10mg/kg/day) in male Sprague-Dawley rats fed a daily diet resembling a human macronutrient diet, significantly increased body weight gain and weight of fatty tissues. In contrast, ziprasidone (1.25, 2.5, and 5mg/kg/day) administration caused no observable adverse effects. We then investigated feeding behavior with acute antipsychotic treatment in male rats using an automated food measurement apparatus. Rats were allowed restricted access to normal laboratory chow (4h/day). With acute olanzapine (0.5, 1, and 2mg/kg, i.p.) treatment in the light phase, food intake volume and duration were significantly increased, while treatment with ziprasidone (0.3, 1, and 3mg/kg, i.p.) did not increase food intake volume or meal time duration. Findings from the present studies showed that chronic treatment with olanzapine in male rats induced body weight gain, and acute injection induced hyperphagia, suggesting that hyperphagia may be involved in the weight gain and obesity-inducing properties of chronically administered olanzapine. These animal models may provide useful experimental platforms for analysis of the mechanism of hyperphagia and evaluating the potential risk of novel antipsychotics to induce weight gain in humans.


Assuntos
Antipsicóticos/efeitos adversos , Benzodiazepinas/efeitos adversos , Modelos Animais de Doenças , Hiperfagia/induzido quimicamente , Piperazinas/efeitos adversos , Tiazóis/efeitos adversos , Aumento de Peso/efeitos dos fármacos , Animais , Antipsicóticos/administração & dosagem , Benzodiazepinas/administração & dosagem , Relação Dose-Resposta a Droga , Comportamento Alimentar/efeitos dos fármacos , Masculino , Olanzapina , Piperazinas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Tiazóis/administração & dosagem
5.
Pain ; 146(1-2): 26-33, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19646816

RESUMO

Fibromyalgia is a prevalent and burdensome disorder characterized by chronic widespread pain and complex comorbid symptoms. To develop better treatments for pain-centered fibromyalgia symptoms, there is still a need for animal models which mimic the features of fibromyalgia patients. In the present study, we have established a fibromyalgia animal model by utilizing a never-before-published pharmacological effect of reserpine. Repeated administration of reserpine (1mg/kg s.c., once daily, for three consecutive days) causes a significant decrease in the muscle pressure threshold and tactile allodynia, which are sustained for 1week or more in both male and female rats. This treatment regimen decreases the amount of biogenic amines (dopamine, norepinephrine, and 5-hydroxytryptamine) in the spinal cord, thalamus, and prefrontal cortex, which are deeply involved in pain signal processing. It also significantly increases immobility time in the forced swim test, which is indicative of depression, a common comorbid symptom of fibromyalgia. Pregabalin, duloxetine, and pramipexole significantly attenuated the reserpine-induced decrease in muscle pressure threshold, but diclofenac did not. The validity of the use of this reserpinized animal as a fibromyalgia model is demonstrated from three different aspects, i.e., face validity (manifestation of chronic pain and comorbid symptoms), construct validity (dysfunction of biogenic amine-mediated central nervous system pain control is involved), and predictive validity (similar responses to treatments used in fibromyalgia patients). This animal model is expected to contribute to the better understanding of fibromyalgia pathophysiology and the evaluation of drugs, especially those which would activate biogenic amine system.


Assuntos
Aminas Biogênicas/fisiologia , Transtorno Depressivo/induzido quimicamente , Transtorno Depressivo/psicologia , Fibromialgia/induzido quimicamente , Fibromialgia/psicologia , Hiperalgesia/induzido quimicamente , Doenças Musculares/induzido quimicamente , Dor/induzido quimicamente , Inibidores da Captação Adrenérgica/farmacologia , Animais , Aminas Biogênicas/metabolismo , Sistema Nervoso Central/metabolismo , Transtorno Depressivo/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Fibromialgia/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Masculino , Músculo Esquelético/fisiologia , Doenças Musculares/tratamento farmacológico , Dor/tratamento farmacológico , Limiar da Dor/efeitos dos fármacos , Estimulação Física , Pressão , Ratos , Ratos Sprague-Dawley , Reserpina/farmacologia , Natação/psicologia , Proteínas Vesiculares de Transporte de Monoamina/antagonistas & inibidores
6.
Behav Brain Res ; 194(2): 152-61, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18657577

RESUMO

Recent evidence indicates that adenosine A(2A) receptor antagonists hold therapeutic potential for the treatment of Parkinson's disease (PD). A study on the novel adenosine A(1) and A(2A) receptor dual antagonist 5-[5-amino-3-(4-fluorophenyl)pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854) showed it to be effective in various rodents models of PD and cognition. In the present study, we further investigated the potential of ASP5854 as an anti-PD drug using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated common marmosets, which is a highly predictive model of clinical efficacy in PD, and compared its effect with those of existing anti-PD drugs. ASP5854 significantly and dose-dependently improved the total motor disability score for 7h at doses higher than 1mg/kg, and significantly increased total locomotor activity at doses higher than 0.1mg/kg without adverse effects. l-3,4-Dihydroxyphenylalanine+benserazide and bromocriptine also significantly improved the motor disability score and the hypolocomotion caused by MPTP treatment in a dose-dependent fashion. This amelioration was significant at 32+8 and 10-32 mg/kg, respectively, although bromocriptine induced severe emesis. Trihexiphenidyl also significantly improved the total motor disability score at doses of 10-32 mg/kg; however, while a significant increase in the total locomotor activity was observed at 10mg/kg, the drug induced ataxia-like behavior at 32 mg/kg. On the other hand, neither selegiline nor amantadine improved the total motor disability and hypolocomotion. These data substantiate the evidence that the novel adenosine antagonist ASP5854 exerts comparable anti-PD activity with existing anti-PD drugs, which indicates that ASP5854 might have potential to ameliorate motor deficits in PD.


Assuntos
Aminopiridinas/uso terapêutico , Antiparkinsonianos/uso terapêutico , Intoxicação por MPTP/complicações , Transtornos dos Movimentos/tratamento farmacológico , Transtornos dos Movimentos/etiologia , Piperazinas/uso terapêutico , Antagonistas Purinérgicos , Análise de Variância , Animais , Callithrix , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Intoxicação por MPTP/induzido quimicamente , Masculino , Atividade Motora/efeitos dos fármacos , Fatores de Tempo
7.
J Nucl Med ; 49(7): 1183-8, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18552135

RESUMO

UNLABELLED: The purpose of the present study was to measure adenosine A(2A) receptor (A(2A)R) occupancy in the brain by a novel adenosine A(1)/A(2A) antagonist, 5-[5-amino-3-(4fluorophenyl)pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854), and to determine the degree of receptor occupancy necessary to inhibit haloperidol-induced catalepsy in rhesus monkeys. METHODS: A(2A)R occupancy by ASP5854 (0.001-0.1 mg/kg) was examined in the striatum using an A(2A)R-specific radiotracer, (11)C-SCH442416, and PET in conscious rhesus monkeys. A(2A)R occupancy was monitored after a single intravenous administration of ASP5854 in 3 animals, and a dynamic PET scan was performed at 1, 4, and 8 h after an intravenous bolus injection of the tracer for approximately 740 MBq. Catalepsy was induced by haloperidol (0.03 mg/kg, intramuscularly) and examined for incidence and duration. RESULTS: ASP5854 dose-dependently increased A(2A)R occupancy in the striatum and showed long-lasting occupancy even after the reduction of plasma concentration. Haloperidol induced severe catalepsy at 40 min after intramuscular injection. The incidence and duration of cataleptic posture were dose-dependently reduced by ASP5854 at 1 h after oral administration, and the minimum ED(50) value was 0.1 mg/kg. Administration of a dose of 0.1 mg/kg yielded a plasma concentration of 97 +/- 16.3 ng/mL, which corresponded to 85%-90% of A(2A)R occupancy. CONCLUSION: These results showed that ASP5854 antagonized A(2A)R in the striatum, and the dissociation from A(2A)R was relatively slow. In addition, more than 85% A(2A)R occupancy by ASP5854 resulted in an inhibition of haloperidol-induced catalepsy. Thus, such a pharmacodynamic study directly demonstrates both the kinetics of a drug in the brain and the relationship between dose-dependent receptor occupancy and plasma level.


Assuntos
Antagonistas do Receptor A1 de Adenosina , Aminopiridinas/metabolismo , Corpo Estriado/metabolismo , Piperazinas/metabolismo , Receptor A2A de Adenosina/metabolismo , Antagonistas do Receptor A2 de Adenosina , Aminopiridinas/uso terapêutico , Animais , Radioisótopos de Carbono , Catalepsia/induzido quimicamente , Catalepsia/tratamento farmacológico , Corpo Estriado/diagnóstico por imagem , Haloperidol , Macaca mulatta , Masculino , Piperazinas/uso terapêutico , Tomografia por Emissão de Pósitrons , Pirazóis/metabolismo , Pirimidinas/metabolismo
8.
J Pharmacol Exp Ther ; 323(2): 708-19, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17684118

RESUMO

Central adenosine A(2A) receptor is a promising target for drugs to treat Parkinson's disease (PD), and the central blockade of adenosine A(1) receptor improves cognitive function. In the present study, we investigated the effect of a novel adenosine A(1) and A(2A) dual antagonist, 5-[5-amino-3-(4-fluorophenyl) pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854), in animal models of PD and cognition. The binding affinities of ASP5854 for human A(1) and A(2A) receptors were 9.03 and 1.76 nM, respectively, with higher specificity and no species differences. ASP5854 also showed antagonistic action on A(1) and A(2A) agonist-induced increases of intracellular Ca(2+) concentration. ASP5854 ameliorated A(2A) agonist 2-[p-(2-carboxyethyl) phenethylamino]-5'-N-ethylcarboxamidoadenosine (CGS21680)- and haloperidol-induced catalepsy in mice, with the minimum effective doses of 0.32 and 0.1 mg/kg, respectively, and it also improved haloperidol-induced catalepsy in rats at doses higher than 0.1 mg/kg. In unilateral 6-hydroxydopamine-lesioned rats, ASP5854 significantly potentiated l-dihydroxyphenylalanine (L-DOPA)-induced rotational behavior at doses higher than 0.032 mg/kg. ASP5854 also significantly restored the striatal dopamine content reduced by 1-metyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment in mice at doses higher than 0.1 mg/kg. Furthermore, in the rat passive avoidance test, ASP5854 significantly reversed the scopolamine-induced memory deficits, whereas the specific adenosine A(2A) antagonist 8-((E)-2-(3,4-dimethoxyphenyl)ethenyl)-1,3-diethyl-7-methyl-3,7-dihydro-1H-purine-2,6-dione (KW-6002; istradefylline) did not. Scopolamine- or 5H-dibenzo[a,d]cyclohepten-5,10-imine (dizocilpine maleate) (MK-801)-induced impairment of spontaneous alternation in the mouse Y-maze test was ameliorated by ASP5854, whereas KW-6002 did not exert improvement at therapeutically relevant dosages. These results demonstrate that the novel, selective, and orally active dual adenosine A(1) and A(2A) receptors antagonist ASP5854 improves motor impairments, is neuroprotective via A(2A) antagonism, and also enhances cognitive function through A(1) antagonism.


Assuntos
Antagonistas do Receptor A1 de Adenosina , Antagonistas do Receptor A2 de Adenosina , Aminopiridinas/uso terapêutico , Transtornos Cognitivos/tratamento farmacológico , Intoxicação por MPTP/tratamento farmacológico , Piperazinas/uso terapêutico , Adenosina/análogos & derivados , Adenosina/farmacologia , Aminopiridinas/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Dopamina/análise , Dopamina/metabolismo , Haloperidol/farmacologia , Humanos , Masculino , Transtornos da Memória/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Fenetilaminas/farmacologia , Piperazinas/farmacologia , Ratos , Ratos Endogâmicos F344 , Ratos Wistar , Receptor A1 de Adenosina/fisiologia , Receptor A2A de Adenosina/fisiologia , Escopolamina/farmacologia
9.
Eur J Pharmacol ; 563(1-3): 40-8, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17343843

RESUMO

Mitochondrial membrane potential plays an important role in cell survival. Transitions in mitochondrial permeability, which indicate the imminent destruction of the organelles, have been observed in damaged neuronal cells both in vitro and in vivo. In this study, C57/BL6n mouse thymocytes were put under stress using thapsigargin, a Ca2+ ATP-ase inhibitor, after which the change in mitochondrial membrane potential was monitored with a JC-1 dual-emission probe. This was done in an attempt to identify a novel compound that can suppress mitochondrial membrane potential reduction and cell death. In this assay system, the novel compound SCH-20148 [2,3-dihydroxypropyl-5-bromo-N-(2-methyl-3-trifluoromethylphenyl)anthranilate] was found to protect mouse thymocytes against thapsigargin (3 nM)-induced mitochondrial membrane potential reduction (IC50=42 nM). SCH-20148 also prevented A23187- or ionomycin-induced shifts in mitochondrial membrane potential but it did not have any effect on the changes induced by tunicamycin, staurosporine, or dexamethasone. The potent immunosuppressants tacrolimus and cyclosporine A prevented the effect of thapsigargin, but did not prevent the A23187- or ionomycin-induced changes. Calcium-modulating agents, an anti-oxidant, a protein kinase C inhibitor, and anti-inflammatory agents were not effective against thapsigargin-induced mitochondrial permeability transition which implies that SCH-20148 exerts a protective effect via its specific mechanism. In addition, SH-20148 demonstrated a neuroprotective effect against thapsigargin-induced neuronal cell death in neuroblastoma SH-SY5Y cells. Taken together, these results suggest the potential of SCH-20148 as novel neuroprotective drug.


Assuntos
Inibidores Enzimáticos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Tapsigargina/farmacologia , Timo/efeitos dos fármacos , ortoaminobenzoatos/farmacologia , Animais , Calcimicina/farmacologia , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Relação Dose-Resposta a Droga , Humanos , Imunossupressores/farmacologia , Ionomicina/farmacologia , Ionóforos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Permeabilidade/efeitos dos fármacos , Tacrolimo/farmacologia , Timo/metabolismo , Timo/patologia
10.
Brain Res ; 1149: 181-90, 2007 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-17391653

RESUMO

The immunosuppressant cyclosporin A (CsA) has been shown to exert potent neuroprotective effects, possibly via the inhibition of calcineurin and mitochondrial permeability transition pore formation. Here, we investigated the neuroprotective profile of a novel derivative of CsA, FR901459, by evaluating its effects against in vitro mitochondrial damage and in vivo brain damage in transient global or focal cerebral ischemia models, in comparison with those of CsA. Efficacy of calcineurin inhibition was estimated from its immunosuppressive effect on the mixed lymphocyte reaction. Results showed that the immunosuppressive effect of FR901459 was approximately 7-fold less potent than that of CsA. In contrast, FR901459 suppressed Ca(2+)-induced mitochondrial swelling measured in isolated liver mitochondria with greater potency than CsA. Further, FR901459 showed approximately 30-fold greater neuroprotective potency than CsA against neuronal cell damage induced by thapsigargin in SH-SY5Y cells. In a transient global cerebral ischemia model in gerbils, FR901459 showed the dose-dependent suppression of neuronal cell death, while FR901459 was less efficacious than CsA. In a rat transient focal ischemia model, FR901459 tended to reduce brain damage on both intravenous injection as well as intracerebroventricular infusion, but with less efficacy than CsA which significantly reduced the damage. These findings suggest that FR901459 exerts a potent neuroprotective effect by inhibiting mitochondrial damage in vitro, but that in in vivo transient cerebral ischemia, its immunosuppressive component which possibly acts via the inhibition of calcineurin may play a more important role in attenuating brain damage than its inhibitory effect against mitochondrial damage.


Assuntos
Ciclosporina/farmacologia , Hipóxia Encefálica/prevenção & controle , Ataque Isquêmico Transitório/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Linhagem Celular Tumoral , Gerbillinae , Humanos , Hipóxia Encefálica/etiologia , Técnicas In Vitro , Ataque Isquêmico Transitório/complicações , Teste de Cultura Mista de Linfócitos , Mitocôndrias/patologia , Ratos
11.
J Pharmacol Exp Ther ; 320(3): 1087-96, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17167170

RESUMO

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily and function as ligand-modulated transcription factors that regulate gene expression in many important biological processes. The PPARdelta subtype has the highest expression in the brain and is postulated to play a major role in neuronal cell function; however, the precise physiological roles of this receptor remain to be elucidated. Herein, we show that the high-affinity PPARdelta agonists L-165041 [4-[3-(4-acetyl-3-hydroxy-2-propylphenoxy)-propoxyl]phenoxy]-acetic acid] and GW501516 [2-methyl4-((4-methyl-2-(4-trifluoromethylphenyl)-1,3-triazol-5-yl)-methylsulfanyl)phenoxy acetic acid] protect against cytotoxin-induced SH-SY5Y cell injury in vitro and both ischemic brain injury and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity in vivo. In the SH-SY5Y studies, treatment with L-165041 or GW501516 significantly and concentration-dependently attenuated cell death following thapsigargin, 1-methyl-4-phenylpyridinium, or staurosporine exposure, with the extent of damage correlated with the level of caspase-3 inhibition. In the transient (90 min) middle cerebral artery occlusion model of ischemic brain injury in rats, i.c.v. infusion of L-165041 or GW501516 significantly attenuated the ischemic brain damage measured 24 h after reperfusion. Moreover, the PPARdelta agonists also significantly attenuated MPTP-induced depletion of striatal dopamine and related metabolite contents in mouse brain. These results demonstrate that subtype-selective PPARdelta agonists possess antiapoptotic properties in vitro, which may underlie their potential neuroprotective potential in in vivo experimental models of cerebral ischemia and Parkinson's disease (PD). These findings suggest that PPARdelta agonists could be useful tools for understanding the role of PPARdelta in other neurodegenerative disorders, as well as attractive therapeutic candidates for stroke and neurodegenerative diseases such as PD.


Assuntos
Isquemia Encefálica/prevenção & controle , Encéfalo/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , PPAR delta/agonistas , Doença de Parkinson/prevenção & controle , Acetatos/farmacocinética , Acetatos/farmacologia , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Dopamina/metabolismo , Genes Reporter , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacocinética , PPAR delta/genética , Doença de Parkinson/metabolismo , Fenóis/farmacocinética , Fenóis/farmacologia , Fenoxiacetatos , Ratos , Ratos Wistar , Especificidade por Substrato , Tiazóis/farmacocinética , Tiazóis/farmacologia
12.
J Biomol Screen ; 11(2): 155-64, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16361696

RESUMO

Measurement of neurite outgrowth is a common assay of neurotrophic activity. However, currently available techniques for measuring neurite outgrowth are either time or resource intensive. The authors established a system in which chronic treatment of a subcloned SH-SY5Y cell line with aphidicolin and various concentrations of nerve growth factor (NGF) induced discernable alterations in proliferation and differentiation. Cells were fixed, labeled with a nonfluorescent dye, and evaluated both manually and with an automated analysis system. NGF increased multiple parameters of differentiation, including neurite length, the proportion of cells extending neurites, and branching, as well as promoting cellular survival/proliferation. Interestingly, although NGF treatment increased the total number of branches, it actually decreased the proportion of branches per neurite length. The authors observed no differences in results obtained using the manual and automated systems, but the automated system was orders of magnitude faster. To demonstrate the flexibility of the system, the authors also show that they could measure changes in differentiation induced by a small-molecule Rho kinase inhibitor, as well as by retinoic acid cotreatment with brain-derived neurotrophic factor. In addition to this flexibility, this system does not require specialized equipment or fluorescent antibodies for analysis and therefore provides a less resource-intensive alternative to fluorescence-based systems.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Fator de Crescimento Neural/farmacologia , Neuritos/química , Tretinoína/farmacologia , Proteínas rho de Ligação ao GTP/antagonistas & inibidores , Afidicolina/farmacologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Clonais , Sinergismo Farmacológico , Humanos , Neuritos/ultraestrutura
13.
Eur J Pharmacol ; 509(1): 11-9, 2005 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-15713424

RESUMO

Immunophilin ligands are neuroregenerative agents, characterized by binding to FK506 binding proteins (FKBPs), which stimulate recovery of neurons in a variety of injury paradigms. Here we report the discovery of a novel, non-immunosuppressive immunophilin ligand, FK1706. FK1706, a derivative of FK506, showed similarly high affinity for two FKBP subtypes, FKBP-12 and FKBP-52, but inhibited T-cell proliferation and interleukin-2 cytokine production with much lower potency and efficacy than FK506. FK1706 (0.1 to 10 nM) significantly potentiated nerve growth factor (NGF)-induced neurite outgrowth in SH-SY5Y cells, as did FK506. This neurite potentiation could be blocked by an anti-FKBP-52 antibody, as well as by specific pharmacological inhibitors of phospholipase C (PLC), phosphatidylinositol 3-kinase (PI3K), and the Ras/Raf/Mitogen-Activated Protein Kinase (MAPK) signaling pathway. FK1706 also potentiated NGF-induced MAPK activation, with a similar dose-dependency to that necessary for potentiating neurite outgrowth. Taken together, these data suggest that FK1706 is a non-immunosuppressive immunophilin ligand with significant neurotrophic effects, putatively mediated via FKBP-52 and the Ras/Raf/MAPK signaling pathway, and therefore that FK1706 may have therapeutic potential in a variety of neurological disorders.


Assuntos
Imunofilinas/farmacologia , Fatores de Crescimento Neural/farmacologia , Fatores de Crescimento Neural/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Imunofilinas/química , Imunofilinas/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Crescimento Neural/antagonistas & inibidores , Receptor trkB/metabolismo , Transdução de Sinais/fisiologia , Tacrolimo/análogos & derivados , Tacrolimo/química , Tacrolimo/imunologia , Tacrolimo/metabolismo , Tacrolimo/farmacologia , Proteína 1A de Ligação a Tacrolimo/química , Proteína 1A de Ligação a Tacrolimo/metabolismo , Proteína 1A de Ligação a Tacrolimo/farmacologia , Trítio
14.
J Pharmacol Sci ; 96(1): 42-52, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15351792

RESUMO

Adenosine A1 receptors in the brain are believed to play an important role in brain functioning. We have discovered a novel adenosine A1 receptor antagonist, FR194921 (2-(1-methyl-4-piperidinyl)-6-(2-phenylpyrazolo[1,5-a]pyridin-3-yl)-3(2H)-pyridazinone), and characterized the pharmacological activity in the present study. FR194921 showed potent and selective affinity for the adenosine A1 receptor without affinity for A2A and A3 receptors and did not show any species differences in binding affinity profile among human, rat, and mouse. Pharmacokinetic study in rats revealed that FR194921 was orally active and highly brain penetrable. Oral administration of FR194921 dose-dependently ameliorated the hypolocomotion induced by the A1 receptor agonist N6-cyclopentyladenosine in rats, indicating this compound exerts A1-antagonistic action in vivo. In the passive avoidance test, scopolamine (1 mg/kg)-induced memory deficits were significantly ameliorated by FR194921 (0.32, 1 mg/kg). In two animal models of anxiety, the social interaction test and elevated plus maze, FR194921 showed specific anxiolytic activity without significantly influencing general behavior. In contrast, FR194921 did not show antidepressant activity even at a dose of 32 mg/kg in the rat forced swimming test. These results indicate that the novel, potent, and selective adenosine A1 receptor antagonist FR194921 exerts both cognitive-enhancing and anxiolytic activity, suggesting the therapeutic potential of this compound for dementia and anxiety disorders.


Assuntos
Antagonistas do Receptor A1 de Adenosina , Adenosina/análogos & derivados , Piperidinas/farmacologia , Piridazinas/farmacologia , Piridinas/farmacologia , Receptor A1 de Adenosina/metabolismo , Adenosina/farmacologia , Administração Oral , Animais , Células CHO , Cricetinae , Relação Dose-Resposta a Droga , Humanos , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Camundongos , Piperidinas/administração & dosagem , Piperidinas/química , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Piridazinas/administração & dosagem , Piridazinas/química , Piridinas/administração & dosagem , Piridinas/química , Ratos , Ratos Sprague-Dawley , Ratos Wistar
15.
J Med Chem ; 47(17): 4151-4, 2004 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15293985

RESUMO

A novel class of quinazolinone derivatives as potent poly(ADP-ribose)polymerase-1 (PARP-1) inhibitors has been discovered. Key to success was application of a rational discovery strategy involving structure-based design, combinatorial chemistry, and classical SAR for improvement of potency and bioavailability. The new inhibitors were shown to bind to the nicotinamide-ribose binding site (NI site) and the adenosine-ribose binding site (AD site) of NAD+.


Assuntos
Barreira Hematoencefálica/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases , Quinazolinas/síntese química , Administração Oral , Animais , Disponibilidade Biológica , Encéfalo/metabolismo , Domínio Catalítico , Técnicas de Química Combinatória , Cães , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/patologia , Poli(ADP-Ribose) Polimerases/química , Quinazolinas/química , Quinazolinas/farmacocinética , Ratos , Relação Estrutura-Atividade
16.
J Pharmacol Exp Ther ; 310(2): 425-36, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15075382

RESUMO

The activation of poly(ADP-ribose) polymerase-1 (PARP-1) after exposure to nitric oxide or oxygen-free radicals can lead to cell injury via severe, irreversible depletion of NAD. Genetic deletion or pharmacological inhibition of PARP-1 attenuates brain injury after focal ischemia and neurotoxicity in several neurodegenerative models in animals. FR247304 (5-chloro-2-[3-(4-phenyl-3,6-dihydro-1(2H)-pyridinyl)propyl]-4(3H)-quinazolinone) is a novel PARP-1 inhibitor that has recently been identified through structure-based drug design. In an enzyme kinetic analysis, FR247304 exhibits potent and competitive inhibition of PARP-1 activity, with a K(i) value of 35 nM. Here, we show that prevention of PARP activation by FR247304 treatment protects against both reactive oxygen species-induced PC12 cell injury in vitro and ischemic brain injury in vivo. In cell death model, treatment with FR247304 (10(-8)-10(-5) M) significantly reduced NAD depletion by PARP-1 inhibition and attenuated cell death after hydrogen peroxide (100 microM) exposure. After 90 min of middle cerebral artery occlusion in rats, poly(ADP-ribosy)lation and NAD depletion were markedly increased in the cortex and striatum from 1 h after reperfusion. The increased poly(ADP-ribose) immunoreactivity and NAD depletion were attenuated by FR247304 (32 mg/kg i.p.) treatment, and FR247304 significantly decreased ischemic brain damage measured at 24 h after reperfusion. Whereas other PARP inhibitors such as 3-aminobenzamide and PJ34 [N-(6-oxo-5,6-dihydro-phenanthridin-2-yl)-N,N-dimethylactamide] showed similar neuroprotective actions, they were less potent in in vitro assays and less efficacious in an in vivo model compared with FR247304. These results indicate that the novel PARP-1 inhibitor FR247304 exerts its neuroprotective efficacy in in vitro and in vivo experimental models of cerebral ischemia via potent PARP-1 inhibition and also suggest that FR247304 or its derivatives could be attractive therapeutic candidates for stroke and neurodegenerative disease.


Assuntos
Isquemia Encefálica/enzimologia , Inibidores Enzimáticos/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Inibidores de Poli(ADP-Ribose) Polimerases , Piridinas/farmacologia , Quinazolinas/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Isquemia Encefálica/prevenção & controle , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Inibidores Enzimáticos/uso terapêutico , Humanos , Peróxido de Hidrogênio/toxicidade , Masculino , Camundongos , Células PC12 , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Piridinas/química , Piridinas/uso terapêutico , Quinazolinas/química , Quinazolinas/uso terapêutico , Quinazolinonas , Ratos , Ratos Wistar
17.
J Pharmacol Exp Ther ; 310(3): 1114-24, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15113847

RESUMO

Methamphetamine (METH) administration in mice, results in a chronic dopamine (DA) depletion associated with nerve terminal damage, with DA oxidation and generation of reactive oxygen species (ROS) primarily mediating this neurotoxicity. The oxidative stress induced by METH putatively activates nuclear enzyme poly(ADP-ribose) polymerase (PARP), with excessive PARP activation eventually leading to cell death. In this study, we show that prevention of PARP activation by treatment with FR261529 [2-(4-chlorophenyl)-5-quinoxalinecarboxamide], the compound that was recently identified as a novel PARP inhibitor (IC50 for PARP-1 = 33 nM, IC50 for PARP-2 = 7 nM), protects against both ROS-induced cells injury in vitro and METH-induced dopaminergic neuronal damage in an in vivo Parkinson's disease (PD) model. In PC12 cells, exposure of hydrogen peroxide or METH markedly induced PARP activation, and treatment with FR261529 (1 microM) significantly reduced PARP activation and attenuated cell death. In the mouse METH model, METH (15 mg/kg x 2 i.p., 2 h apart) intoxication accelerated DA metabolism and oxidation in the striatum, with subsequent cell damage in nigrostriatal dopaminergic neurons after 4 days. Oral administration of FR261529 (10 or 32 mg/kg) attenuated the damage of dopaminergic neurons via marked reduction of PARP activity and not via changes in dopamine metabolism or body temperature. These findings indicate that the neuroprotective effects of a novel PARP inhibitor, FR261529, were accompanied by inhibition of METH-induced PARP activation, suggesting that METH induces nigrostriatal dopaminergic neurodegeneration involving PARP activation and also orally active and brain-penetrable PARP inhibitor FR261529 could be a novel attractive therapeutic candidate for neurodegenerative disorders such as PD.


Assuntos
Inibidores Enzimáticos/farmacologia , Metanfetamina/toxicidade , Inibidores de Poli(ADP-Ribose) Polimerases , Quinoxalinas/farmacologia , Animais , Benzamidas/farmacologia , Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Inibidores Enzimáticos/farmacocinética , Peroxidação de Lipídeos/efeitos dos fármacos , Metanfetamina/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacocinética , Fármacos Neuroprotetores/farmacologia , Células PC12 , Ratos , Substância Negra/efeitos dos fármacos , Substância Negra/metabolismo
18.
J Pharmacol Exp Ther ; 309(3): 1067-78, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-14985416

RESUMO

The massive activation of poly(ADP-ribose) polymerase-1 (PARP-1) by DNA-damaging stimuli, such as exposure to reactive oxygen species (ROS), can lead to cell injury via severe, irreversible depletion of the NAD and ATP pool, and PARP-1 inhibitors have been expected to rescue neurons from degeneration in a number of disease models. We have recently identified 2-[3-[4-(4-chlorophenyl)-1-piperazinyl] propyl]-4(3H)-quinazolinone (FR255595) as a novel and potent PARP-1 inhibitor through structure-based drug design and high-throughput screening. This compound potently inhibited PARP activity with an IC(50) value of 11 nM and was orally active and highly brain penetrable. Here, we show that prevention of PARP activation by FR255595 protects against both ROS-induced cells injury in vitro and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal dopaminergic damage in an in vivo Parkinson's disease (PD) model. In cell death models in vitro, exposure of hydrogen peroxide induced cell death with PARP overactivation in PC12 cells and SH-SY5Y cells, and pre- and post-treatment with FR255595 (10(-9)-10(-5) M) significantly reduced PARP activation and cell death. In mouse MPTP model, MPTP (20 mg/kg i.p.) intoxication lead to PARP activation and cell damage in the nigrostriatal dopaminergic pathway, which was significantly ameliorated by oral administration of FR255595 (10-32 mg/kg), both in the substantia nigra and in the striatum via marked reduction of PARP activation, even with delayed treatment. These findings clearly indicate that the novel PARP-1 inhibitor FR255595 exerts neuroprotective effect through its potent PARP-1 inhibitory actions in PD model, suggesting that the drug could be an attractive candidate for several neurodegenerative disorders, including PD.


Assuntos
Morte Celular/efeitos dos fármacos , Intoxicação por MPTP/prevenção & controle , Piperazinas/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases , Quinazolinas/uso terapêutico , Animais , Células Cultivadas , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Piperazinas/farmacocinética , Piperazinas/farmacologia , Poli(ADP-Ribose) Polimerases , Quinazolinas/farmacocinética , Quinazolinas/farmacologia , Quinazolinonas , Ratos
19.
Br J Pharmacol ; 140(5): 825-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14559856

RESUMO

Nerve growth factor (NGF) and other members of the neurotrophin family are critical for the survival and differentiation of neurons within the peripheral and central nervous systems. Neurophilin ligands, including FK506, potentiate NGF-induced neurite outgrowth in several experimental models, although the mechanism of this potentiation is unclear. Therefore, we tested which signaling pathways were involved in FK506-potentiated neurite outgrowth in SH-SY5Y neuroblastoma cells using specific pharmacological inhibitors of various signaling molecules. Inhibitors of Ras (lovastatin), Raf (GW5074), or MAP kinase (PD98059 and U0126) blocked FK506 activity, as did inhibitors of phospholipase C (U73122) and phosphatidylinositol 3' kinase (LY294002). Protein kinase C inhibitors (Go6983 and Ro31-8220) slightly but significantly inhibited neurite outgrowth, whereas inhibitors of p38 MAPK (SB203580) or c-Jun N-terminal kinase (SP600125) had no effect. These data suggest that FK506 potentiates neurite outgrowth through the Ras/Raf/MAP kinase signaling pathway downstream of phospholipase C and phosphatidylinositol 3' kinase.


Assuntos
Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Proteínas Proto-Oncogênicas c-raf/fisiologia , Tacrolimo/farmacologia , Proteínas ras/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia
20.
J Biol Chem ; 277(1): 623-9, 2002 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-11679586

RESUMO

Emerging evidence has shown that tumor suppressor p53 expression is enhanced in response to brain ischemia/hypoxia and that p53 plays a critical role in the cell death pathway in such an acute neurological insult. However the mechanism remains unclear. Recently it was reported that Peg3/Pw1, originally identified as a paternally expressed gene, plays a pivotal role in the p53-mediated cell death pathway in mouse fibroblast cell lines. In this study, we found that Peg3/Pw1 expression is enhanced in peri-ischemic neurons in rat stroke model by in situ hybridization analysis, where p53 expression was also induced by immunohistochemical analysis. Moreover, we found that p53 was co-localized with Peg3/Pw1 in brain ischemia/hypoxia by double staining analysis. In human neuroblastoma-derived SK-N-SH cells, Peg3/Pw1 mRNA expression is enhanced remarkably at 24 h post-hypoxia, when p53 protein expression was also enhanced at high levels. Subcellular localization of Peg3/Pw1 was observed in the nucleus. Adenovirus-mediated high dose p53 overexpression induced Peg3/Pw1 mRNA expression. Overexpression of Peg3/Pw1 reduced cell viability under hypoxic conditions, whereas that of the C-terminal-deleted mutant and anti-sense Peg3/Pw1 inhibited hypoxia-induced cell death. These results suggest that Peg3/Pw1 is involved in the p53-mediated cell death pathway as a downstream effector of p53 in brain ischemia/hypoxia.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Proteínas Quinases , Proteínas/fisiologia , Fatores de Transcrição , Proteína Supressora de Tumor p53/fisiologia , Animais , Morte Celular , Sobrevivência Celular , Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Hipóxia-Isquemia Encefálica/patologia , Fatores de Transcrição Kruppel-Like , Proteínas/análise , Proteínas/genética , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA