Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biomol NMR ; 18(1): 65-8, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11061229

RESUMO

A powerful screening by NMR methodology (WaterLOGSY), based on transfer of magnetization from bulk water, for the identification of compounds that interact with target biomolecules (proteins, RNA and DNA fragments) is described. The method exploits efficiently the large reservoir of H2O magnetization. The high sensitivity of the technique reduces the amount of biomolecule and ligands needed for the screening, which constitutes an important requirement for high throughput screening by NMR of large libraries of compounds. Application of the method to a compound mixture against the cyclin-dependent kinase 2 (cdk2) protein is presented.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/química , Ligantes , Ressonância Magnética Nuclear Biomolecular/métodos , Proteínas Serina-Treonina Quinases/química , Água/química , Quinase 2 Dependente de Ciclina
2.
Epilepsia ; 40(11): 1523-8, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10565578

RESUMO

PURPOSE: PNU-151774E [(S)-(+)-2-(4-(3-fluorobenzyloxy) benzylamino) propanamide, methanesulfonate] is a novel antiepileptic drug (AED) with a broad spectrum of activity in a variety of chemically and mechanically induced seizures. The objective of this study was to evaluate the activity of PNU-151774E in the amygdala fully kindled rat model of complex partial seizures, and to compare its effects with those of carbamazepine (CBZ), phenytoin (PHT), lamotrigine (LTG), and gabapentin (GBP), drugs used to treat this disease state. METHODS: Male Wistar rats were stimulated daily through electrodes implanted in the amygdala with a threshold current until fully generalized seizures developed. The rats were then treated with various doses of a single compound. Control values for each rat and drug dose were determined after vehicle administration followed by electrical stimulation 1 day before drug treatment. RESULTS: PNU-151774E (1, 10, 30 mg/kg; i.p.) reduced the duration of behavioral seizures significantly and dose-dependently at doses starting from 1 mg/kg. Higher doses significantly reduced seizure severity and afterdischarge duration. In contrast, no dose-related effects were noted after administration of PHT, whereas after CBZ treatment, a plateau of activity was noted from the intermediate to higher doses. The effects of PNU-151774E were comparable to those of LTG and GBP. CONCLUSIONS: The activity shown by PNU-151774E at doses similar to those that are active in models of generalized seizures indicates that PNU-151774E would also have potential efficacy in the treatment of complex partial seizures.


Assuntos
Alanina/análogos & derivados , Aminas , Tonsila do Cerebelo/fisiopatologia , Anticonvulsivantes/farmacologia , Benzilaminas/farmacologia , Ácidos Cicloexanocarboxílicos , Epilepsia Parcial Complexa/prevenção & controle , Excitação Neurológica/fisiologia , Ácido gama-Aminobutírico , Acetatos/farmacologia , Acetatos/uso terapêutico , Alanina/farmacologia , Alanina/uso terapêutico , Animais , Anticonvulsivantes/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Benzilaminas/uso terapêutico , Carbamazepina/farmacologia , Carbamazepina/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia Parcial Complexa/etiologia , Epilepsia Parcial Complexa/fisiopatologia , Gabapentina , Lamotrigina , Masculino , Fenitoína/farmacologia , Fenitoína/uso terapêutico , Ratos , Ratos Wistar , Índice de Gravidade de Doença , Triazinas/farmacologia , Triazinas/uso terapêutico
3.
Bioorg Med Chem Lett ; 9(17): 2521-4, 1999 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-10498200

RESUMO

Sodium channel blocking, anticonvulsant activity, and sigma (sigma) binding of selected leads in a series of alpha-amino amide anticonvulsants were examined. While anticonvulsant compounds were always endowed with low micromolar sodium (Na+) channel site-2 binding, compounds with low site-2 Na+ channel affinity failed to control seizures. No correlation could be drawn with sigma1 binding. Both anticonvulsant and Na+ channel blocking activities were independent of stereochemistry, while sigma1 binding seems to be favoured by an S-configuration on the aminoamide moiety.


Assuntos
Amidas/farmacologia , Anticonvulsivantes/farmacologia , Receptores sigma/metabolismo , Bloqueadores dos Canais de Sódio , Amidas/metabolismo , Animais , Anticonvulsivantes/metabolismo , Ratos
4.
Bioorg Med Chem Lett ; 9(13): 1783-8, 1999 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-10406642

RESUMO

Dextromethorphan 1 is an effective neuroprotectant in animal models of epilepsy and ischemia but showed side-effects during clinical trials limiting its potential use in a clinical setting. Here we describe the enantioselective and enantiospecific syntheses and the initial in vitro and in vivo biological evaluation of new hybrid structures between 1 and a previously disclosed alpha-amino amide anticonvulsant (3).


Assuntos
Alanina/análogos & derivados , Anticonvulsivantes/síntese química , Anticonvulsivantes/farmacologia , Benzilaminas/síntese química , Benzilaminas/farmacologia , Dextrometorfano/química , Alanina/síntese química , Alanina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Dextrometorfano/análogos & derivados , Concentração Inibidora 50 , Cinética , Camundongos , Ratos
5.
Farmaco ; 54(3): 152-60, 1999 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-10371028

RESUMO

A series of pyrrolo[3,2-c]quinoline derivatives were synthesised and evaluated as inhibitors of selected enzymes of the kynurenine pathway. 7-Chloro-3-methyl-1H-pyrrolo[3,2-c]quinoline-4-carboxylic acid (7a) was found to be a relatively potent and selective inhibitor of kynurenine-3-hydroxylase (KYN-3-OHase). A molecular modelling study showed a good superimposition of 7a with PNU-156561 and kynurenine the natural substrate of KYN-3-OHase.


Assuntos
Inibidores Enzimáticos/síntese química , Oxigenases de Função Mista/antagonistas & inibidores , Pirróis/síntese química , Quinolinas/síntese química , Animais , Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase , Fígado/enzimologia , Estrutura Molecular , Pirróis/farmacologia , Quinolinas/farmacologia , Ratos
6.
J Pharmacol Exp Ther ; 288(3): 1151-9, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10027853

RESUMO

PNU-151774E [(S)-(+)-2-(4-(3-fluorobenzyloxy)benzylamino)propanamide methanesulfonate], a new anticonvulsant that displays a wide therapeutic window, has a potency comparable or superior to that of most classic anticonvulsants. PNU-151774E is chemically unrelated to current antiepileptics. In animal seizure models it possesses a broad spectrum of action. In the present study, the action mechanism of PNU-151774E has been investigated using electrophysiological and biochemical assays. Binding studies performed with rat brain membranes show that PNU-151774E has high affinity for binding site 2 of the sodium channel receptor, which is greater than that of phenytoin or lamotrigine (IC50, 8 microM versus 47 and 185 microM, respectively). PNU-151774E reduces sustained repetitive firing in a use-dependent manner without modifying the first action potential in hippocampal cultured neurons. In the same preparation PNU-151774E inhibits tetrodotoxin-sensitive fast sodium currents and high voltage-activated calcium currents under voltage-clamp conditions. These electrophysiological activities of PNU-151774E correlate with its ability to inhibit veratrine and KCl-induced glutamate release in rat hippocampal slices (IC50, 56.4 and 185.5 microM, respectively) and calcium inward currents in mouse cortical neurons. On the other hand, PNU-151774E does not affect whole-cell gamma-aminobutryic acid- and glutamate-induced currents in cultured mouse cortical neurons. These results suggest that PNU-151774E exerts its anticonvulsant activity, at least in part, through inhibition of sodium and calcium channels, stabilizing neuronal membrane excitability and inhibiting transmitter release. The possible relevance of these pharmacological properties to its antiepileptic potential is discussed.


Assuntos
Alanina/análogos & derivados , Anticonvulsivantes/farmacologia , Benzilaminas/farmacologia , Encéfalo/efeitos dos fármacos , Alanina/farmacologia , Animais , Encéfalo/metabolismo , Canais de Cálcio/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Masculino , Membranas/efeitos dos fármacos , Camundongos , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Canais de Potássio/efeitos dos fármacos , Cloreto de Potássio/farmacologia , Ratos , Ratos Wistar , Veratrina/farmacologia
7.
Eur J Pharmacol ; 359(1): 27-32, 1998 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-9831289

RESUMO

Kainic acid-induced multifocal status epilepticus in the rat is a model of medically intractable complex partial seizures and neurotoxicity. The exact mechanisms of kainic acid epileptogenic and neurotoxic effects are unknown, but enhanced glutamate release seems to be an important factor. PNU-151774E ((S)-(+)-2-(4-(3-fluorobenzyloxy) benzylamino) propanamide, methanesulfonate) is a broad-spectrum new anticonvulsant with Na+ channel-blocking and glutamate release inhibiting properties. We have examined the effect of pretreatment with this compound on both seizure activity and hippocampal neuronal damage induced by systemic injection of kainic acid in rats. Lamotrigine, a recently developed anticonvulsant with similar glutamate release inhibitory properties, was tested for comparison, together with diazepam as reference standard, on the basis of its anticonvulsant and neuroprotectant properties in this animal model. PNU-151774E, lamotrigine (10, 30 mg/kg; i.p.) and diazepam (20 mg/kg; i.p.) were administered 15 min before kainic acid (10 mg/kg; i.p.). In the vehicle-treated group, kainic acid injection caused status epilepticus in 86% of animals. Hippocampal neuronal cell loss was 66% in the CA4 hippocampal area at 7 days after kainic acid administration. Diazepam inhibited both seizures and neurotoxicity. Lamotrigine reduced hippocampal neuronal cell loss at both doses, even when it did not protect from seizures, although it showed a trend toward protection. On the other hand PNU-151774E protected from both hippocampal neurodegeneration and status epilepticus. Thus, these data support the concept that seizure prevention and neuroprotection might not be tightly coupled. Glutamate release inhibition may play a major role in neuroprotection, but an additional mechanism(s) of action might be relevant for the anticonvulsant activity of PNU-151774E in this model.


Assuntos
Alanina/análogos & derivados , Anticonvulsivantes/uso terapêutico , Benzilaminas/uso terapêutico , Epilepsia/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Alanina/uso terapêutico , Animais , Diazepam/uso terapêutico , Epilepsia/induzido quimicamente , Hipocampo/fisiologia , Ácido Caínico , Lamotrigina , Masculino , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Wistar , Triazinas/uso terapêutico
8.
J Pharmacol Exp Ther ; 285(2): 397-403, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9580576

RESUMO

PNU-151774E [(S)-(+)-2-(4-(3-fluorobenzyloxy) benzylamino) propanamide, methanesulfonate] is a structurally novel anticonvulsant having Na+ channel-blocking and glutamate release-inhibiting properties, as well as being a MAOB inhibitor. Its anticonvulsant activity was evaluated in the maximal electroshock (MES) test and in chemically induced seizures (bicuculline, BIC; picrotoxin, PIC; 3-mercaptopropionic acid, 3-MPA; pentylenetetrazole, PTZ; strychnine, STRYC). Behavioral toxicity was evaluated in the rotorod test with measurements of spontaneous locomotor activity and passive avoidance responding. The anti-MES activity of PNU-151774E in both mice and rats, respectively, produced ED50 values of 4.1 mg/kg and 6.9 mg/kg after i.p. administration or 8.0 mg/kg and 11.8 mg/kg after p.o. administration. Oral anti-MES activity in rats peaked between 1 and 2 h after administration and was evident up to 4 h. This activity was related to brain levels of unchanged drug which peaked at 37 mM within 1 h. Oral ED50 values (mg/kg) effective in blocking tonic extension seizures by chemical convulsants in mice were: BIC (26.9), PIC (60.6), 3-MPA (21.5), STRYC (104.1) and PTZ (26.8). This potency was associated with high therapeutic indices relative to: MES (78.2), BIC (23.3), PIC (10.3), 3-MPA (29.1) and STRYC (6.0). No evidence of tolerance to anti-MES activity after repeated dosing was observed. PNU-151774E did not show anti-absence seizure activity as assessed by i.v. infusion of PTZ. PNU-151774E impaired spontaneous activity in rats only at the oral rotorod ED50 dose of 700 mg/kg p.o. PNU-151774E did not impair passive avoidance responding at doses up to 40 times the oral MES ED50 dose in rats. These results indicate that PNU-151774E is an anticonvulsant effective in various seizure models with a wide therapeutic window, and with a low potential to induce tolerance and locomotor or cognitive side effects.


Assuntos
Alanina/análogos & derivados , Anticonvulsivantes/farmacologia , Benzilaminas/farmacologia , Alanina/efeitos adversos , Alanina/farmacocinética , Alanina/farmacologia , Animais , Anticonvulsivantes/efeitos adversos , Anticonvulsivantes/farmacocinética , Aprendizagem da Esquiva/efeitos dos fármacos , Benzilaminas/efeitos adversos , Benzilaminas/farmacocinética , Tolerância a Medicamentos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Ratos Wistar
9.
J Med Chem ; 41(4): 579-90, 1998 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-9484507

RESUMO

Although most epilepsies are adequately treated by conventional antiepileptic therapy, there remains an unfulfilled need for safer and more effective anticonvulsant agents. Starting from milacemide, a weak anticonvulsant, and trying to elucidate its mechanism of action, we discovered a structurally novel class of potent and preclinically safe anticonvulsants. Here we report the structure-activity relationship (SAR) study within this series of compounds. Different parts of the structural lead 2-[[4-(3-chlorobenzoxy)benzyl]amino]acetamide (6) were thus varied (Figure 1), and many potent anticonvulsants were found. As an outcome of this study, 57 ((S)-2-[[4-(3-fluorobenzoxy)benzyl]amino]propanamide methanesulfonate, PNU-151774E) emerged as a promising candidate for further development for its potent anticonvulsant activity and outstanding therapeutic indexes (TIs) in different animal tests.


Assuntos
Alanina/síntese química , Anticonvulsivantes/síntese química , Benzilaminas/síntese química , Convulsões/prevenção & controle , Alanina/análogos & derivados , Alanina/química , Alanina/farmacologia , Animais , Anticonvulsivantes/química , Anticonvulsivantes/farmacologia , Benzilaminas/química , Benzilaminas/farmacologia , Bicuculina , Desenho de Fármacos , Eletrochoque , Indicadores e Reagentes , Masculino , Camundongos , Camundongos Endogâmicos ICR , Modelos Moleculares , Estrutura Molecular , Atividade Motora/efeitos dos fármacos , Picrotoxina , Equilíbrio Postural/efeitos dos fármacos , Equilíbrio Postural/fisiologia , Ratos , Ratos Endogâmicos , Convulsões/induzido quimicamente , Convulsões/etiologia , Relação Estrutura-Atividade , Estricnina
10.
Bioorg Med Chem Lett ; 8(20): 2907-12, 1998 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-9873646

RESUMO

Kynurenine 3-hydroxylase (KYN 3-OHase) is a key enzyme in the kynurenine pathway of tryptophan degradation and its inhibition may be an effective mechanism for counteracting neuronal excitotoxic damage. We present here a new class of inhibitors derived from a structure-activity relationship (SAR) study of the benzoylalanine side-chain of 1. 2-hydroxy-4-(3,4-dichlorophenyl)-4-oxobutanoic acid (8) and 2-benzyl-4-(3,4-dichlorophenyl)-4-oxo-butanoic acid (10) emerged as the most interesting derivatives. Enantiospecific synthesis for both enantiomers of 8 and diastereomeric salt resolution for those of 10 were successfully applied.


Assuntos
Inibidores Enzimáticos/química , Oxigenases de Função Mista/antagonistas & inibidores , Fenilbutiratos/química , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Inibidores Enzimáticos/farmacologia , Humanos , Concentração Inibidora 50 , Quinurenina 3-Mono-Oxigenase , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fenilbutiratos/farmacologia , Ratos , Relação Estrutura-Atividade , Triptofano/metabolismo
11.
Biochem Pharmacol ; 48(5): 905-14, 1994 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-8093103

RESUMO

A series of analogues of the anticonvulsant drug milacemide (2-(n-pentylamino)-acetamide; Compound I) has been synthesized: 2-(benzylamino)acetamide (Compound II), 2-(phenethylamino)acetamide (Compound III), 2-(2-indol-3-yl)-ethylamino acetamide (Compound IV), 2-(2-(5-methoxyindol-3-yl)ethylamino)-acetamide (Compound V), 2-(2(4-chlorobenzamido)-ethylamino)acetamide (Compound VI), 2-(2-benzamidoethylamino)-acetamide (Compound VII) and 2-(4-(3-chlorobenzyloxy)phenethylamino)acetamide (Compound VIII). These compounds involve retention of the aminoacetamide portion of milacemide but replacement of the pentyl moiety with aromatic residues present in the structures of substrates and inhibitors of the monoamine oxidases. All the compounds tested were substrates for ox liver monoamine oxidase-B (MAO-B), producing an aldehyde that could act as a substrate for ox liver aldehyde dehydrogenase and H2O2 as a result of oxidative cleavage which also released glycinamide, although their Michaelis-Menten parameters differed markedly. None showed detectable activity as substrates for rat liver monoamine oxidase-A (MAO-A). Inhibition of the MAO-B by all the compounds except Compounds VIII and IV showed marked time dependence and was at least partly irreversible. There was no apparent change in the inhibition of MAO-A during enzyme-inhibitor preincubation at 37 degrees for 60 min. Compound VIII was a potent reversible inhibitor of both MAO-A and MAO-B (Ki = 2.8 +/- 0.1 and 4.1 +/- 0.8 microM), respectively. Comparison of the inhibitory potencies and the specificity constants of the series of compounds as substrates for MAO-B revealed no simple correlations with their anticonvulsant activities, as measured by their ability to prevent bicuculline-induced convulsions and death in the mouse. These results suggest that neither inhibition of MAO nor oxidative cleavage by this enzyme to yield glycinamide plays the major role in the anticonvulsant action of these compounds.


Assuntos
Acetamidas/farmacologia , Anticonvulsivantes/farmacologia , Inibidores da Monoaminoxidase/farmacologia , Acetamidas/química , Acetamidas/metabolismo , Animais , Anticonvulsivantes/metabolismo , Sítios de Ligação , Bovinos , Mitocôndrias Hepáticas/enzimologia , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/metabolismo , Ratos
12.
Xenobiotica ; 22(2): 191-7, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1632108

RESUMO

1. It has previously been established that monoamine oxidase (MAO)-B participates in the metabolism of milacemide [2-(pentylamino)acetamide]. Furthermore, in rats, inhibition of FAD-dependent polyamine oxidase (PAO) was found to decrease the urinary excretion of two milacemide metabolites, termed UK1 and UK2. 2. Using gas chromatography-mass spectrometry, UK1 was identified as oxamic acid and UK2 as 2-hydroxyacetamide, confirming that PAO is involved in the metabolism of milacemide. 3. Thus, two FAD-dependent amine-oxidizing enzymes, MAO and PAO, contribute to the metabolism of milacemide. Milacemide appears to be the first non-polyamine xenobiotic in the metabolism of which PAO participates.


Assuntos
Acetamidas/urina , Anticonvulsivantes/urina , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Animais , Flavina-Adenina Dinucleotídeo/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Masculino , Ácido Oxâmico/urina , Ratos , Ratos Endogâmicos , Poliamina Oxidase
13.
Eur J Drug Metab Pharmacokinet ; 16(3): 231-9, 1991.
Artigo em Inglês | MEDLINE | ID: mdl-1814741

RESUMO

The purpose of this study was to compare the disposition and the metabolic pattern of Reboxetine in several species, including man. [14C]-Reboxetine was given orally to the rat, the dog, the monkey (5 mg/kg) and man (2 and 4 mg/kg). Radioactivity was eliminated both by the renal and faecal route in the rat and the dog, mainly in urine in the monkey and man. Reboxetine was extensively metabolized. A number of urinary metabolites were quantified by radio-HPLC and tentatively identified by comparison with the retention times of reference compounds. Suggested routes of metabolic transformation are: 2-O-dealkylation; hydroxylation of the ethoxyphenoxy ring; oxidation of the morpholine ring; morpholine ring-opening; and combinations of these. Metabolites were partially or completely conjugated with glucuronic acid and/or sulphuric acid.


Assuntos
Antidepressivos/farmacocinética , Morfolinas/farmacocinética , Administração Oral , Animais , Antidepressivos/administração & dosagem , Antidepressivos/metabolismo , Cromatografia Líquida de Alta Pressão , Remoção de Radical Alquila , Cães/metabolismo , Humanos , Hidroxilação , Macaca fascicularis/metabolismo , Masculino , Morfolinas/administração & dosagem , Morfolinas/metabolismo , Ratos , Ratos Endogâmicos/metabolismo , Reboxetina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA