Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Neuroscience ; 97(2): 243-51, 2000.
Article de Anglais | MEDLINE | ID: mdl-10799756

RÉSUMÉ

Competing enzymatic mechanisms degrade the tryptophan metabolite L-kynurenine to kynurenate, an inhibitory and neuroprotective compound, and to the neurotoxins 3-hydroxykynurenine and quinolinate. Kynurenine 3-hydroxylase inhibitors such as PNU 156561 shift metabolism towards enhanced kynurenate production, and this effect may underlie the recently discovered anticonvulsant and neuroprotective efficacy of these drugs. Using electrophysiological and neurotoxicological endpoints, we now used PNU 156561 as a tool to examine the functional interplay of kynurenate, 3-hydroxykynurenine and quinolinate in the rat hippocampus in vivo. First, population spike amplitude in area CA1 and the extent of quinolinate-induced excitotoxic neurodegeneration were studied in animals receiving acute or prolonged intravenous infusions of L-kynurenine, PNU 156561, (L-kynurenine+PNU 156561) or kynurenate. Only the latter two treatments, but not L-kynurenine or PNU 156561 alone, caused substantial inhibition of evoked responses in area CA1, and only prolonged (3h) infusion of (L-kynurenine+PNU 156561) or kynurenate was neuroprotective. Biochemical analyses in separate animals revealed that the levels of kynurenate attained in both blood and brain (hippocampus) were essentially identical in rats receiving extended infusions of L-kynurenine alone or (L-kynurenine+PNU 156561) (4 and 7microM, respectively, after an infusion of 90 or 180min). However, addition of the kynurenine 3-hydroxylase inhibitor resulted in a significant decrement in the formation of 3-hydroxykynurenine and quinolinate in both blood and brain. These data suggest that the ratio between kynurenate and 3-hydroxykynurenine and/or quinolinate in the brain is a critical determinant of neuronal excitability and viability. The anticonvulsant and neuroprotective potency of kynurenine 3-hydroxylase inhibitors may therefore be due to the drugs' dual action on both branches of the kynurenine pathway of tryptophan degradation.


Sujet(s)
Butyrates/pharmacologie , Hippocampe/physiologie , Acide kynurénique/métabolisme , Cynurénine/métabolisme , Neuroprotecteurs/pharmacologie , Acide 3-hydroxy-anthranilique/métabolisme , Animaux , Barrière hémato-encéphalique , Antienzymes/pharmacologie , Hippocampe/effets des médicaments et des substances chimiques , Acide kynurénique/pharmacologie , Cynurénine/analogues et dérivés , Cynurénine/pharmacologie , Kynurenine 3-monooxygenase , Mâle , Mixed function oxygenases/antagonistes et inhibiteurs , Neurotoxines/pharmacologie , Cellules pyramidales/effets des médicaments et des substances chimiques , Cellules pyramidales/physiologie , Acide quinolinique/pharmacologie , Rats , Rat Sprague-Dawley
2.
Epilepsia ; 40(11): 1523-8, 1999 Nov.
Article de Anglais | MEDLINE | ID: mdl-10565578

RÉSUMÉ

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.


Sujet(s)
Alanine/analogues et dérivés , Amines , Amygdale (système limbique)/physiopathologie , Anticonvulsivants/pharmacologie , Benzylamines/pharmacologie , Acides cyclohexanecarboxyliques , Épilepsie partielle complexe/prévention et contrôle , Embrasement/physiologie , Acide gamma-amino-butyrique , Acétates/pharmacologie , Acétates/usage thérapeutique , Alanine/pharmacologie , Alanine/usage thérapeutique , Animaux , Anticonvulsivants/usage thérapeutique , Comportement animal/effets des médicaments et des substances chimiques , Comportement animal/physiologie , Benzylamines/usage thérapeutique , Carbamazépine/pharmacologie , Carbamazépine/usage thérapeutique , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Épilepsie partielle complexe/étiologie , Épilepsie partielle complexe/physiopathologie , Gabapentine , Lamotrigine , Mâle , Phénytoïne/pharmacologie , Phénytoïne/usage thérapeutique , Rats , Rat Wistar , Indice de gravité de la maladie , Triazines/pharmacologie , Triazines/usage thérapeutique
3.
Bioorg Med Chem Lett ; 9(17): 2521-4, 1999 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-10498200

RÉSUMÉ

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.


Sujet(s)
Amides/pharmacologie , Anticonvulsivants/pharmacologie , Récepteur sigma/métabolisme , Bloqueurs de canaux sodiques , Amides/métabolisme , Animaux , Anticonvulsivants/métabolisme , Rats
4.
Farmaco ; 54(5): 288-96, 1999 May 30.
Article de Anglais | MEDLINE | ID: mdl-10418123

RÉSUMÉ

Ergot comprises a group of indole alkaloids which are predominantly found in various species of the ascomycete Claviceps. In pharmacopoeias, the sclerotia of Claviceps purpurea (Fr.) Tulasne parasitizing on rye, Secale cereale L., are designed as ergot or Secale cornutum. Now, the term ergot is used in a broader sense to describe the sclerotia of various Claviceps species growing on different host plants or their saprophytic mycelia. Due to their many fascinating features, there is a continuing and extensive interest in these secondary metabolites. Thus, the chemistry of ergot alkaloids and derivatives has presented many challenges to organic chemists. The ergot alkaloids and derivatives have attracted great interest for their broad spectrum of pharmacological action that includes central, neurohumoral and peripheral effects. These are mainly responses mediated by noradrenaline, serotonin, or dopamine receptors. No other group of natural products exhibits such a wide spectrum of biological action. For this reason, ergot has been termed a veritable treasure house of pharmacological constituents'. Moreover, ergot alkaloids have been an important stimulus in the development of new drugs by providing structural prototypes of molecules with pronounced pharmacological activities. This concise review, moving from the experience of our group in Pharmacia & Upjohn, will briefly mention the most representative ergoline derivatives featured in the literature. Our work in this field originated compounds with quite different pharmacological activities. In fact, by continuous modification of the same main template structure, the ergoline skeleton, it ultimately led to the development of new dopaminergic agents and to the identification of new series of serotonergic agents.


Sujet(s)
Ergolines/pharmacologie , Récepteurs dopaminergiques/effets des médicaments et des substances chimiques , Récepteurs sérotoninergiques/effets des médicaments et des substances chimiques , Animaux , Cabergoline , Agents dopaminergiques/pharmacologie , Humains , Prolactine/antagonistes et inhibiteurs , Récepteurs dopaminergiques/métabolisme , Récepteurs sérotoninergiques/métabolisme , Antisérotonines/pharmacologie , Agonistes des récepteurs de la sérotonine/pharmacologie
5.
Bioorg Med Chem Lett ; 9(13): 1783-8, 1999 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-10406642

RÉSUMÉ

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).


Sujet(s)
Alanine/analogues et dérivés , Anticonvulsivants/synthèse chimique , Anticonvulsivants/pharmacologie , Benzylamines/synthèse chimique , Benzylamines/pharmacologie , Dextrométhorphane/composition chimique , Alanine/synthèse chimique , Alanine/pharmacologie , Animaux , Encéphale/effets des médicaments et des substances chimiques , Dextrométhorphane/analogues et dérivés , Concentration inhibitrice 50 , Cinétique , Souris , Rats
6.
Farmaco ; 54(3): 152-60, 1999 Mar 31.
Article de Anglais | MEDLINE | ID: mdl-10371028

RÉSUMÉ

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.


Sujet(s)
Antienzymes/synthèse chimique , Mixed function oxygenases/antagonistes et inhibiteurs , Pyrroles/synthèse chimique , Quinoléines/synthèse chimique , Animaux , Antienzymes/pharmacologie , Kynurenine 3-monooxygenase , Foie/enzymologie , Structure moléculaire , Pyrroles/pharmacologie , Quinoléines/pharmacologie , Rats
7.
J Pharmacol Exp Ther ; 288(3): 1151-9, 1999 Mar.
Article de Anglais | MEDLINE | ID: mdl-10027853

RÉSUMÉ

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.


Sujet(s)
Alanine/analogues et dérivés , Anticonvulsivants/pharmacologie , Benzylamines/pharmacologie , Encéphale/effets des médicaments et des substances chimiques , Alanine/pharmacologie , Animaux , Encéphale/métabolisme , Canaux calciques/effets des médicaments et des substances chimiques , Acide glutamique/métabolisme , Mâle , Membranes/effets des médicaments et des substances chimiques , Souris , Neurones/effets des médicaments et des substances chimiques , Techniques de patch-clamp , Canaux potassiques/effets des médicaments et des substances chimiques , Chlorure de potassium/pharmacologie , Rats , Rat Wistar , Vératrine/pharmacologie
8.
Eur J Pharmacol ; 359(1): 27-32, 1998 Oct 16.
Article de Anglais | MEDLINE | ID: mdl-9831289

RÉSUMÉ

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.


Sujet(s)
Alanine/analogues et dérivés , Anticonvulsivants/usage thérapeutique , Benzylamines/usage thérapeutique , Épilepsie/traitement médicamenteux , Hippocampe/effets des médicaments et des substances chimiques , Alanine/usage thérapeutique , Animaux , Diazépam/usage thérapeutique , Épilepsie/induit chimiquement , Hippocampe/physiologie , Acide kaïnique , Lamotrigine , Mâle , Neuroprotecteurs/usage thérapeutique , Rats , Rat Wistar , Triazines/usage thérapeutique
9.
J Pharmacol Exp Ther ; 285(2): 397-403, 1998 May.
Article de Anglais | MEDLINE | ID: mdl-9580576

RÉSUMÉ

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.


Sujet(s)
Alanine/analogues et dérivés , Anticonvulsivants/pharmacologie , Benzylamines/pharmacologie , Alanine/effets indésirables , Alanine/pharmacocinétique , Alanine/pharmacologie , Animaux , Anticonvulsivants/effets indésirables , Anticonvulsivants/pharmacocinétique , Apprentissage par évitement/effets des médicaments et des substances chimiques , Benzylamines/effets indésirables , Benzylamines/pharmacocinétique , Tolérance aux médicaments , Mâle , Souris , Souris de lignée ICR , Activité motrice/effets des médicaments et des substances chimiques , Rats , Rat Sprague-Dawley , Rat Wistar
10.
Farmaco ; 53(1): 65-72, 1998 Jan.
Article de Anglais | MEDLINE | ID: mdl-9543728

RÉSUMÉ

A series of ergoline-amides was synthesised in the discovery of new dopaminomimetic agents. Several compounds exhibited in vivo high prolactin lowering activity (indirectly measured by the nidation test) in rats. For the most active, the potential anti-Parkinson activity was evaluated by observation of the contralateral turning behaviour in 6-OH-DA lesioned rats. The acute toxicity by oral route in mice was also studied.


Sujet(s)
Antiparkinsoniens/synthèse chimique , Ergolines/synthèse chimique , Animaux , Antiparkinsoniens/pharmacologie , Comportement animal/effets des médicaments et des substances chimiques , Ergolines/pharmacologie , Femelle , Mâle , Souris , Rats , Rat Sprague-Dawley , Relation structure-activité
11.
J Med Chem ; 41(4): 579-90, 1998 Feb 12.
Article de Anglais | MEDLINE | ID: mdl-9484507

RÉSUMÉ

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.


Sujet(s)
Alanine/synthèse chimique , Anticonvulsivants/synthèse chimique , Benzylamines/synthèse chimique , Crises épileptiques/prévention et contrôle , Alanine/analogues et dérivés , Alanine/composition chimique , Alanine/pharmacologie , Animaux , Anticonvulsivants/composition chimique , Anticonvulsivants/pharmacologie , Benzylamines/composition chimique , Benzylamines/pharmacologie , Bicuculline , Conception de médicament , Électrochoc , Indicateurs et réactifs , Mâle , Souris , Souris de lignée ICR , Modèles moléculaires , Structure moléculaire , Activité motrice/effets des médicaments et des substances chimiques , Picrotoxine , Équilibre postural/effets des médicaments et des substances chimiques , Équilibre postural/physiologie , Rats , Lignées consanguines de rats , Crises épileptiques/induit chimiquement , Crises épileptiques/étiologie , Relation structure-activité , Strychnine
12.
Bioorg Med Chem Lett ; 8(9): 1117-22, 1998 May 05.
Article de Anglais | MEDLINE | ID: mdl-9871719

RÉSUMÉ

Novel classes of 13- and 14-tertbutyl-ergoline derivatives were prepared, and characterised in vitro for their affinity for adrenergic, dopaminergic and serotonergic binding sites. This study particularly examines the importance of the presence and the position of the tert-butyl group in conferring either significant 5-HT1A or 5-HT2 affinity and selectivity respectively.


Sujet(s)
Ergolines/synthèse chimique , Récepteurs alpha-adrénergiques/métabolisme , Récepteurs dopaminergiques/métabolisme , Récepteurs sérotoninergiques/métabolisme , Agonistes des récepteurs de la sérotonine/synthèse chimique , Animaux , Fixation compétitive , Conception de médicament , Ergolines/composition chimique , Ergolines/pharmacologie , Hippocampe/métabolisme , Indicateurs et réactifs , Cortex préfrontal/métabolisme , Dosage par compétition , Rats , Récepteurs de la sérotonine de type 5-HT1 , Agonistes des récepteurs de la sérotonine/composition chimique , Agonistes des récepteurs de la sérotonine/pharmacologie , Relation structure-activité
13.
Bioorg Med Chem Lett ; 8(5): 447-52, 1998 Mar 03.
Article de Anglais | MEDLINE | ID: mdl-9871596

RÉSUMÉ

Syntheses of both the alpha-methyl and benzyl analogs of quisqualic acid are described. Testing of these compounds for their activity at excitatory amino acid receptors revealed a striking change in activity in comparison to quisqualic acid. This structural modification results in the loss of quisqualate's potent agonist action at both non-NMDA ionotropic glutamate receptors as well as at group I mGluRs, while allowing these analogs to acquire antagonist properties with relative selectivity for group II metabotropic glutamate receptors.


Sujet(s)
Acide quisqualique/analogues et dérivés , Récepteurs métabotropes au glutamate/antagonistes et inhibiteurs , Animaux , Calcium/métabolisme , Lignée cellulaire , Cervelet/cytologie , Cervelet/effets des médicaments et des substances chimiques , Cervelet/métabolisme , Cricetinae , Antagonistes des acides aminés excitateurs/composition chimique , Antagonistes des acides aminés excitateurs/pharmacologie , Transport des ions , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Acide quisqualique/synthèse chimique , Acide quisqualique/pharmacologie , Rats
14.
Bioorg Med Chem Lett ; 8(20): 2907-12, 1998 Oct 20.
Article de Anglais | MEDLINE | ID: mdl-9873646

RÉSUMÉ

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.


Sujet(s)
Antienzymes/composition chimique , Mixed function oxygenases/antagonistes et inhibiteurs , Phénylbutyrates/composition chimique , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/enzymologie , Antienzymes/pharmacologie , Humains , Concentration inhibitrice 50 , Kynurenine 3-monooxygenase , Foie/effets des médicaments et des substances chimiques , Foie/enzymologie , Phénylbutyrates/pharmacologie , Rats , Relation structure-activité , Tryptophane/métabolisme
15.
J Med Chem ; 40(21): 3369-80, 1997 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-9341912

RÉSUMÉ

A possible bioisosterism between the benzamido and the phenylimidazolidin-2-one moieties has been suggested on the basis of the similarity between the molecular electrostatic potential (MEP) of metoclopramide, a D2 receptor antagonist with weak 5-HT3 receptor antagonist properties, and zetidoline, a D2 receptor antagonist. Starting from this premise, a series of phenylimidazolidin-2-one derivatives bearing a basic azabicycloalkyl or an imidazolylalkyl moiety were synthesized and evaluated for 5-HT3 receptor radioligand binding affinity ([3H]-GR 43,694). In vitro 5-HT3 receptor antagonist activity was tested in the guinea pig ileum assay (GPI). A number of high-affinity ligands were shown to be potent 5-HT3 receptor antagonists in vivo as determined by inhibition of the Bezold--Jarisch reflex in the anesthetized rat. In general, the imidazolylalkyl derivatives were found to be more active than azabicycloalkyls. 1-(3,5-Dichlorophenyl)-3-[(5-methyl-1H-imidazol-4-yl)methyl]imidazoli din-2-one (58), in particular, displayed very high affinity for the 5-HT3 receptor (Ki of 0.038 nM) with a Kb of 5.62 nM in the GPI assay, being more potent than the reference compounds (ondansetron, tropisetron, granisetron, and BRL 46,470) tested. 58 showed an ID50 comparable to that of ondansetron (2.2 micrograms/kg i.v.) in the Bezold--Jarisch reflex. A molecular modeling study based on this structurally novel series of compounds allowed the refinement of previously reported 5-HT3 receptor antagonist pharmacophore models.


Sujet(s)
Imidazoles/pharmacologie , Récepteurs sérotoninergiques/métabolisme , Antisérotonines/pharmacologie , Animaux , Fixation compétitive , Encéphale/effets des médicaments et des substances chimiques , Cochons d'Inde , Rythme cardiaque/effets des médicaments et des substances chimiques , Iléum/effets des médicaments et des substances chimiques , Imidazoles/synthèse chimique , Imidazoles/composition chimique , Imidazoles/métabolisme , Modèles moléculaires , Structure moléculaire , Muscles/effets des médicaments et des substances chimiques , Rats , Récepteurs sérotoninergiques 5-HT3 , Antisérotonines/synthèse chimique , Antisérotonines/composition chimique , Antisérotonines/métabolisme
16.
Science ; 276(5315): 1119-22, 1997 May 16.
Article de Anglais | MEDLINE | ID: mdl-9148807

RÉSUMÉ

Prion diseases are transmissible neurodegenerative conditions characterized by the accumulation of protease-resistant forms of the prion protein (PrP), termed PrPres, in the brain. Insoluble PrPres tends to aggregate into amyloid fibrils. The anthracycline 4'-iodo-4'-deoxy-doxorubicin (IDX) binds to amyloid fibrils and induces amyloid resorption in patients with systemic amyloidosis. To test IDX in an experimental model of prion disease, Syrian hamsters were inoculated intracerebrally either with scrapie-infected brain homogenate or with infected homogenate coincubated with IDX. In IDX-treated hamsters, clinical signs of disease were delayed and survival time was prolonged. Neuropathological examination showed a parallel delay in the appearance of brain changes and in the accumulation of PrPres and PrP amyloid.


Sujet(s)
Doxorubicine/analogues et dérivés , Prions/métabolisme , Tremblante/traitement médicamenteux , Amyloïde/métabolisme , Animaux , Comportement animal , Encéphale/métabolisme , Encéphale/anatomopathologie , Maladie de Creutzfeldt-Jakob/métabolisme , Cricetinae , Doxorubicine/métabolisme , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Femelle , Humains , Mesocricetus , ARN messager/génétique , ARN messager/métabolisme , Tremblante/métabolisme , Tremblante/anatomopathologie , Tubuline/analyse
17.
Eur J Pharmacol ; 315(3): 263-7, 1996 Nov 21.
Article de Anglais | MEDLINE | ID: mdl-8982663

RÉSUMÉ

Kynurenic acid is an endogenous excitatory amino-acid receptor antagonist with neuroprotective and anticonvulsant properties. We demonstrate here that systemic administration of the new and potent kynurenine 3-hydroxylase inhibitor (R,S)-3,4-dichlorobenzoylalanine (FCE 28833A) causes a dose-dependent elevation in endogenous kynurenine and kynurenic acid levels in rat brain tissue. In hippocampal microdialysates, peak increases of 10- and 80-fold above basal kynurenic acid concentrations, respectively, were obtained after a single oral or intraperitoneal administration of 400 mg/kg FCE 28833A. After intraperitoneal treatment with FCE 28833A, extracellular brain kynurenic acid levels remained significantly elevated for at least 22 h, rendering this compound a far more effective enhancer of kynurenic acid levels than the previously described kynurenine 3-hydroxylase blocker m-nitrobenzoylalanine. FCE 28833A and similar molecules may have therapeutic value in diseases which are linked to a hyperfunction of excitatory amino-acid receptors.


Sujet(s)
Encéphale/effets des médicaments et des substances chimiques , Butyrates/pharmacologie , Acide kynurénique/métabolisme , Neuroprotecteurs/pharmacologie , Animaux , Encéphale/métabolisme , Mâle , Rats , Rat Wistar
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE