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










Base de dados
Intervalo de ano de publicação
1.
Neuropharmacology ; 77: 131-44, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24071566

RESUMO

Kappa opioid receptors and their endogenous neuropeptide ligand, dynorphin A, are densely localized in limbic and cortical areas comprising the brain reward system, and appear to play a key role in modulating stress and mood. Growing literature indicates that kappa receptor antagonists may be beneficial in the treatment of mood and addictive disorders. However, existing literature on kappa receptor antagonists has used extensively JDTic and nor-BNI which exhibit long-lasting pharmacokinetic properties that complicate experimental design and interpretation of results. Herein, we report for the first time the in vitro and in vivo pharmacological profile of a novel, potent kappa opioid receptor antagonist with excellent selectivity over other receptors and markedly improved drug-like properties over existing research tools. LY2456302 exhibits canonical pharmacokinetic properties that are favorable for clinical development, with rapid absorption (t(max): 1-2 h) and good oral bioavailability (F = 25%). Oral LY2456302 administration selectively and potently occupied central kappa opioid receptors in vivo (ED50 = 0.33 mg/kg), without evidence of mu or delta receptor occupancy at doses up to 30 mg/kg. LY2456302 potently blocked kappa-agonist-mediated analgesia and disruption of prepulse inhibition, without affecting mu-agonist-mediated effects at doses >30-fold higher. Importantly, LY2456302 did not block kappa-agonist-induced analgesia one week after administration, indicating lack of long-lasting pharmacodynamic effects. In contrast to the nonselective opioid antagonist naltrexone, LY2456302 produced antidepressant-like effects in the mouse forced swim test and enhanced the effects of imipramine and citalopram. LY2456302 reduced ethanol self-administration in alcohol-preferring (P) rats and, unlike naltrexone, did not exhibit significant tolerance upon 4 days of repeated dosing. LY2456302 is a centrally-penetrant, potent, kappa-selective antagonist with pharmacokinetic properties favorable for clinical development and activity in animal models predictive of efficacy in mood and addictive disorders.


Assuntos
Benzamidas/farmacologia , Depressão/tratamento farmacológico , Antagonistas de Entorpecentes/farmacologia , Pirrolidinas/farmacologia , Receptores Opioides kappa/antagonistas & inibidores , Reflexo de Sobressalto/efeitos dos fármacos , Filtro Sensorial/efeitos dos fármacos , Analgesia , Animais , Antidepressivos/farmacocinética , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Benzamidas/farmacocinética , Benzamidas/uso terapêutico , Modelos Animais de Doenças , Etanol/administração & dosagem , Masculino , Camundongos , Antagonistas de Entorpecentes/farmacocinética , Antagonistas de Entorpecentes/uso terapêutico , Pirrolidinas/farmacocinética , Pirrolidinas/uso terapêutico , Ratos , Autoadministração
2.
Am J Physiol Endocrinol Metab ; 305(2): E282-92, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23715724

RESUMO

Obesity continues to be a global health problem, and thus it is imperative that new pathways regulating energy balance be identified. Recently, it was reported: (Hayashi K, Cao T, Passmore H, Jourdan-Le Saux C, Fogelgren B, Khan S, Hornstra I, Kim Y, Hayashi M, Csiszar K. J Invest Dermatol 123: 864-871, 2004) that mice carrying a missense mutation in myelin protein zero-like 3 (Mpzl3rc) have reduced body weight. To determine how Mpzl3 controls energy balance in vivo, we generated mice deficient in myelin protein zero-like 3 (Mpzl3-KO). Interestingly, KO mice were hyperphagic yet had reduced body weight and fat mass. Moreover, KO mice were highly resistant to body weight and fat mass gain after exposure to a high-fat, energy-dense diet. These effects on body weight and adiposity were driven, in part, by a pronounced increase in whole body energy expenditure levels in KO mice. KO mice also had reduced blood glucose levels during an intraperitoneal glucose challenge and significant reductions in circulating insulin levels suggesting an increase in insulin sensitivity. In addition, there was an overall increase in oxidative capacity and contractile force in skeletal muscle isolated from KO mice. Hepatic triglyceride levels were reduced by 92% in livers of KO mice, in part due to a reduction in de novo lipid synthesis. Interestingly, Mpzl3 mRNA expression in liver was increased in diet-induced obese mice. Moreover, KO mice exhibited an increase in insulin-stimulated Akt signaling in the liver, further demonstrating that Mpzl3 can regulate insulin sensitivity in this tissue. We have determined that Mpzl3 has a novel physiological role in controlling body weight regulation, energy expenditure, glycemic control, and hepatic triglyceride synthesis in mice.


Assuntos
Glicemia/fisiologia , Metabolismo Energético/fisiologia , Lipídeos/biossíntese , Fígado/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Adiposidade/genética , Adiposidade/fisiologia , Animais , Análise Química do Sangue , Western Blotting , Temperatura Corporal/fisiologia , Dieta , Dislipidemias/genética , Dislipidemias/metabolismo , Teste de Tolerância a Glucose , Hiperglicemia/genética , Hiperglicemia/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Contração Muscular/fisiologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Reação em Cadeia da Polimerase em Tempo Real , Triglicerídeos/metabolismo , Aumento de Peso/fisiologia
3.
Obesity (Silver Spring) ; 18(9): 1710-7, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20300082

RESUMO

Binge eating disorder (BED) is characterized by excessive food intake during a short period of time and is often associated with obesity. Mouse models of binge-like eating behavior are lacking making it difficult to employ genetic models in the identification of mechanisms regulating excessive eating. We report a rapid and simple model to induce binge-like eating behavior in mice that does not require food deprivation or exogenous stressors. Weekly 24 h access to a nutritionally complete high energy diet (HED), along with continuous access to standard chow, resulted in a significant increase in HED intake following its presentation compared to mice that had continuous access to both diets. Mice exhibiting binge-like eating consumed one-third of their normal total daily caloric intake within 2.5 h of HED presentation. Moreover, total 24-h caloric intakes were increased by 50% in mice exhibiting binge-like eating. Following repeated cycles, binge-like eating of the HED was maintained over several weeks with no evidence of habituation or significant alterations in body weight and adiposity. Pharmacological evaluation of binge-like eating behavior was performed using clinically employed compounds. Interestingly, binge-like eating was dose-dependently decreased by fluoxetine, but not baclofen or topiramate. These data support clinical validation of this mouse model of binge-like eating behavior, as fluoxetine has been shown to reduce binge frequency in human subjects with BED. The availability of transgenic and knockout mice will allow for the determination of genes that are involved in the initiation and maintenance of binge-like eating behavior.


Assuntos
Comportamento Animal , Transtorno da Compulsão Alimentar , Dieta , Modelos Animais de Doenças , Ingestão de Energia/fisiologia , Camundongos Endogâmicos C57BL , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Adiposidade/fisiologia , Animais , Fármacos Antiobesidade/farmacologia , Baclofeno/farmacologia , Comportamento Animal/efeitos dos fármacos , Transtorno da Compulsão Alimentar/tratamento farmacológico , Transtorno da Compulsão Alimentar/fisiopatologia , Peso Corporal/fisiologia , Relação Dose-Resposta a Droga , Ingestão de Energia/efeitos dos fármacos , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Frutose/análogos & derivados , Frutose/farmacologia , Agonistas dos Receptores de GABA-B/farmacologia , Habituação Psicofisiológica , Masculino , Camundongos , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Topiramato
4.
Am J Physiol Regul Integr Comp Physiol ; 295(2): R463-71, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18525013

RESUMO

An analog of the trans-3,4-dimethyl-4-(3-hydroxyphenyl)piperidine series (LY255582) exhibits high in vitro binding affinity and antagonist potency for the mu-, delta-, and kappa-opioid receptors. In vivo, LY255582 exhibits potent effects in reducing food intake and body weight in several rodent models of obesity. In the present study, we evaluated the effects of LY255582 to prevent the consumption of a highly palatable (HP) diet (a high-fat/high-carbohydrate diet) both when the food was novel and following daily limited access to the HP diet. Additionally, we examined the effects of consumption of the HP diet and of LY255582 treatment on mesolimbic dopamine (DA) signaling by in vivo microdialysis. Consumption of the HP diet increased extracellular DA levels within the nucleus accumbens (NAc) shell. Increased DA in the NAc shell was not related to the quantity of the HP diet consumed, and the DA response did not habituate following daily scheduled access to the HP diet. Interestingly, treatment with LY255582 inhibited consumption of the HP diet and the HP diet-associated increase in NAc shell DA levels. Moreover, the increased HP diet consumption observed following daily limited access to the HP diet was completely prevented by LY255582 treatment. LY255582 may be a useful tool in understanding the neural mechanisms involved in the reinforcement mechanisms regulating food intake.


Assuntos
Regulação do Apetite/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Cicloexanos/farmacologia , Dopamina/metabolismo , Antagonistas de Entorpecentes/farmacologia , Neurônios/efeitos dos fármacos , Núcleo Accumbens/efeitos dos fármacos , Piperidinas/farmacologia , Animais , Ingestão de Alimentos/efeitos dos fármacos , Preferências Alimentares , Masculino , Microdiálise , Neurônios/metabolismo , Núcleo Accumbens/citologia , Núcleo Accumbens/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Opioides/metabolismo , Reforço Psicológico , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...