Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 193: 108619, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34023336

RESUMO

The newfound antidepressant efficacy of ketamine has provided opportunities for the development of new-generation, rapid-acting, glutamate-based antidepressants. We previously identified that methoxetamine (MXE), a ketamine analog, and an N-Methyl-d-aspartate (NMDA) receptor antagonist, produced rapid and sustained antidepressant effects in mice. MXE (R, S (±)-MXE) is a racemic mixture containing equal parts of S (+)-MXE and R (-)-MXE. However, studies have yet to investigate the antidepressant effects of its enantiomers. Here, we examined the potential antidepressant properties and behavioral side effects of S- and R-MXE in mice. Both S- and R-MXE showed significant NMDA receptor affinity and appreciable inhibitory activity on serotonin transporter. Also, S- and R-MXE (10 mg kg-1) exerted antidepressant effects and increased gamma waves (electroencephalography) but were inhibited by NBQX (an AMPA receptor antagonist). Subsequently, they increased mammalian target of rapamycin phosphorylation and AMPA receptor subunits GluA1 and GluA2 protein levels in the hippocampus or prefrontal cortex. Furthermore, they increased 5HT2a and 5HT2c receptor mRNA levels in the prefrontal cortex, with their antidepressant effects inhibited by ketanserin (a 5HT2a/c receptor antagonist). Taken together, S-MXE and R-MXE elicit antidepressant effects that are probably mediated via glutamatergic and serotonergic mechanisms. Unlike S-MXE, R-MXE did not induce prepulse inhibition deficits, hyperlocomotion, conditioned place preference, and locomotor sensitization, although it acutely altered motor coordination. This suggests that R-MXE induces fewer behavioral side effects and is a safer antidepressant than S-MXE. Overall, this study provides significant implications for future research on the next generation of rapid-acting, glutamate-based antidepressant drugs.


Assuntos
Antidepressivos/efeitos adversos , Antidepressivos/farmacologia , Cicloexanonas/farmacologia , Cicloexilaminas/farmacologia , Depressão/tratamento farmacológico , Depressão/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cicloexanonas/efeitos adversos , Cicloexilaminas/efeitos adversos , Teste de Labirinto em Cruz Elevado , Células HEK293 , Elevação dos Membros Posteriores , Humanos , Ketamina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de Serotonina/metabolismo , Homólogo LST8 da Proteína Associada a mTOR/metabolismo
2.
Birth Defects Res ; 109(1): 49-54, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-27801979

RESUMO

BACKGROUND: While pharmacological activation of the Hedgehog (HH) signaling pathway may have therapeutic benefits for developmental and adult diseases, its teratogenic potential is of concern. The membrane molecule Smoothened (SMO) transduces HH signaling and can be acutely modulated by antagonists and agonists. The objective of the current experiments was to determine how maternal treatment with the Smo agonist, SAG, affects the developing limb. METHODS: Pregnant C57BL/6J mice received a single injection of SAG (15, 17, or 20 mg/kg, i.p.) or its vehicle on gestational day (GD) 9.25, the time of limb bud induction. Embryos were examined on GD 15 for gross dysmorphology and skeletal staining was performed to visualize the number and type of digits on the fore- and hindlimbs. Additionally, in situ hybridization was performed 4 hr after GD 9.25 SAG administration to determine SAG's effects on Gli1 and Gli2 mRNA expression. RESULTS: The most prevalent effect of SAG was the dose-dependent induction of pre-axial polydactyly; defects ranged from a broad thumb to the duplication of two finger-like digits on the preaxial side of the thumb. The highest SAG dose was effective in ca. 80% of the embryos and increased Gli1 and Gli2 mRNA expression in the limb bud, with Gli1 mRNA being the most upregulated. CONCLUSION: Preaxial polydactyly can be caused in the developing embryo by acute maternal administration of a Smo agonist that activates HH signaling. These results are consistent with the preaxial polydactyly induced in developmental disorders associated with mutations in HH signaling genes.Birth Defects Research 109:49-54, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Cicloexilaminas/efeitos adversos , Cicloexilaminas/metabolismo , Polidactilia/fisiopatologia , Tiofenos/efeitos adversos , Tiofenos/metabolismo , Animais , Extremidades , Feminino , Deformidades da Mão/genética , Deformidades da Mão/metabolismo , Proteínas Hedgehog/genética , Botões de Extremidades/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Polidactilia/genética , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Transdução de Sinais/genética , Receptor Smoothened/agonistas , Receptor Smoothened/metabolismo , Polegar/anormalidades , Polegar/fisiopatologia , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco/efeitos dos fármacos , Proteína GLI1 em Dedos de Zinco/genética , Proteína Gli2 com Dedos de Zinco/efeitos dos fármacos , Proteína Gli2 com Dedos de Zinco/genética
3.
Br J Clin Pharmacol ; 81(5): 971-9, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26620151

RESUMO

AIMS: Endothelial-derived epoxyeicosatrienoic acids may regulate vascular tone and are metabolized by soluble epoxide hydrolase enzymes (sEH). GSK2256294 is a potent and selective sEH inhibitor that was tested in two phase I studies. METHODS: Single escalating doses of GSK2256294 2-20 mg or placebo were administered in a randomized crossover design to healthy male subjects or obese smokers. Once daily doses of 6 or 18 mg or placebo were administered for 14 days to obese smokers. Data were collected on safety, pharmacokinetics, sEH enzyme inhibition and blood biomarkers. Single doses of GSK2256294 10 mg were also administered to healthy younger males or healthy elderly males and females with and without food. Data on safety, pharmacokinetics and biliary metabolites were collected. RESULTS: GSK2256294 was well-tolerated with no serious adverse events (AEs) attributable to the drug. The most frequent AEs were headache and contact dermatitis. Plasma concentrations of GSK2256294 increased with single doses, with a half-life averaging 25-43 h. There was no significant effect of age, food or gender on pharmacokinetic parameters. Inhibition of sEH enzyme activity was dose-dependent, from an average of 41.9% on 2 mg (95% confidence interval [CI] -51.8, 77.7) to 99.8% on 20 mg (95% CI 99.3, 100.0) and sustained for up to 24 h. There were no significant changes in serum VEGF or plasma fibrinogen. CONCLUSIONS: GSK2256294 was well-tolerated and demonstrated sustained inhibition of sEH enzyme activity. These data support further investigation in patients with endothelial dysfunction or abnormal tissue repair, such as diabetes, wound healing or COPD.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Cicloexilaminas/farmacologia , Inibidores Enzimáticos/farmacologia , Epóxido Hidrolases/antagonistas & inibidores , Obesidade/tratamento farmacológico , Triazinas/farmacologia , Ácido 8,11,14-Eicosatrienoico/metabolismo , Adolescente , Adulto , Idoso , Estudos de Coortes , Estudos Cross-Over , Cicloexilaminas/efeitos adversos , Cicloexilaminas/farmacocinética , Dermatite de Contato/etiologia , Método Duplo-Cego , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Epóxido Hidrolases/metabolismo , Feminino , Meia-Vida , Cefaleia/induzido quimicamente , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Triazinas/efeitos adversos , Triazinas/farmacocinética , Adulto Jovem
4.
Exp Neurol ; 261: 404-11, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24842488

RESUMO

BACKGROUND AND PURPOSE: Activation of mitogen-activated protein kinases (MAPKs), particularly c-jun-N-terminal kinases (JNK) and p38 exacerbates stroke injury by provoking pro-apoptotic and pro-inflammatory cellular signaling. MAPK phosphatase-1 (MKP-1) restrains the over-activation of MAPKs via rapid de-phosphorylation of the MAPKs. We therefore examined the role of MKP-1 in stroke and studied its inhibitory effects on MAPKs after experimental stroke. METHODS: Male mice were subjected to transient middle cerebral artery occlusion (MCAO). MKP-1 knockout (KO) mice and a MKP-1 pharmacological inhibitor were utilized. We utilized flow cytometry, immunohistochemistry (IHC), and Western blots analysis to explore MKP-1 signaling and its effects on apoptosis/inflammation in the brain and specifically in microglia after stroke. RESULTS: MKP-1 was highly expressed in the nuclei of both neurons and microglia after stroke. MKP-1 genetic deletion exacerbated stroke outcome by increasing infarct, neurological deficits and hemorrhagic transformation. Additionally, delayed treatment of the MKP-1 pharmacological inhibitor worsened stroke outcome in wild type (WT) mice but had no effect in MKP-1 KO mice. Furthermore, MKP-1 deletion led to increased c-jun-N-terminal kinase (JNK) activation and microglial p38 activation after stroke. Finally, MKP-1 deletion or inhibition increased inflammatory and apoptotic response as evidenced by the increased levels of interleukin-6 (IL-6), tumor necrosis factor α (TNFα), ratio of p-c-jun/c-jun and cleaved caspase-3 following ischemia. CONCLUSIONS: We have demonstrated that MKP-1 signaling is an endogenous protective mechanism in stroke. Our data imply that MKP-1 possesses anti-apoptotic and anti-inflammatory properties by simultaneously controlling the activities of JNK and microglial p38.


Assuntos
Fosfatase 1 de Especificidade Dupla/deficiência , Encefalite/etiologia , Regulação Enzimológica da Expressão Gênica/genética , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/etiologia , Animais , Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Cicloexilaminas/efeitos adversos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fosfatase 1 de Especificidade Dupla/genética , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Indenos/efeitos adversos , Infarto da Artéria Cerebral Média/genética , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos Knockout , Exame Neurológico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
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
6.
Curr Med Res Opin ; 3(4): 218-24, 1975.
Artigo em Inglês | MEDLINE | ID: mdl-1097197
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA