Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Aujourdhui ; 205(1): 29-31, 2011.
Artigo em Francês | MEDLINE | ID: mdl-21501572

RESUMO

Novel strategies are needed to treat epilepsy, in order to ensure efficiency, security and prevention. The search for innovating anti-epileptics is based on finding appropriate target molecules, among which the most pertinent appear to be chlore and potassium channels. Transcriptomics and proteomics are also prone to detect genes or proteins implicated in the disease, in particular when biopsies from healthy and epileptic brains are compared. Animal genetic models provide information about epilepsies with a unique origin. Finally some targets are identified through fortuitous findings from research in other fields, notably that of pro-inflammatory cytokines.


Assuntos
Epilepsia/genética , Epilepsia/fisiopatologia , Animais , Canais de Cloreto , Citocinas , Modelos Animais de Doenças , Epilepsia/tratamento farmacológico , Perfilação da Expressão Gênica , Humanos , Canais de Potássio , Proteômica
2.
J Soc Biol ; 203(3): 249-69, 2009.
Artigo em Francês | MEDLINE | ID: mdl-19833071

RESUMO

This paper reviews the process used in the pharmaceutical industry to discover new innovative drugs originating from medicinal chemistry or biotherapeutics. After a rapid description of the global Research and Development process (preclinical and clinical studies), the different steps involved specifically in the Discovery Research phase (from the initiation of the research program to the proposal of a candidate for development) are analysed in detail. As far as compounds originating from medicinal chemistry are concerned, we discuss the choice of the biological target (molecular or cellular) and its functional validation, the process used for the screening of chemical libraries and the generation of chemical hits, the hit-to-lead process and finally the chemical optimisation of the leads into drug candidates which will be proposed for preclinical and clinical development. As far as biotherapeutics are concerned, the process used for the generation and manufacturing of humanized naked or conjugated monoclonal antibodies and recombinant therapeutic proteins is discussed in detail.


Assuntos
Descoberta de Drogas/métodos , Indústria Farmacêutica/métodos , Experimentação Animal , Animais , Terapia Biológica , Células Cultivadas/efeitos dos fármacos , Ensaios Clínicos como Assunto/métodos , Sistemas de Liberação de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/uso terapêutico , Humanos , Projetos de Pesquisa , Bibliotecas de Moléculas Pequenas
3.
Pharmacol Biochem Behav ; 91(1): 47-58, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18621075

RESUMO

On native human, rat and mouse glycine transporter-1(GlyT1), SSR130800 behaves as a selective inhibitor with IC50 values of 1.9, 5.3 and 6.8 nM, respectively. It reversibly blocked glycine uptake in mouse brain cortical homogenates, increased extracellular levels of glycine in the rat prefrontal cortex, and potentiated NMDA-mediated excitatory postsynaptic currents in rat hippocampal slices. SSR103800 (30 mg/kg, p.o.) decreased MK-801- and PCP-induced locomotor hyperactivity in rodents. SSR103800 (1 and 10 mg/kg, p.o.) attenuated social recognition deficit in adult rats induced by neonatal injections of PCP (10 mg/kg, s.c., on post-natal day 7, 9 and 11). SSR103800 (3 mg/kg, p.o.) counteracted the deficit in short-term visual episodic-like memory induced by a low challenge dose of PCP (1 mg/kg, i.p.), in PCP-sensitized rats (10 mg/kg, i.p.). SSR103800 (30 mg/kg, i.p.) increased the prepulse inhibition of the startle reflex in DBA/1J mice. SSR103800 decreased defensive- and despair-related behaviors in the tonic immobility test in gerbils (10 and 30 mg/kg, p.o.) and in the forced-swimming procedure in rats (1 and 3 mg/kg, p.o.), respectively. These findings suggest that SSR103800 may have a therapeutic potential in the management of the core symptoms of schizophrenia and comorbid depression states.


Assuntos
Antipsicóticos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Esquizofrenia/tratamento farmacológico , Animais , Antidepressivos/farmacologia , Discriminação Psicológica/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Gerbillinae , Glicina/metabolismo , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , N-Metilaspartato/fisiologia , Fenciclidina/farmacologia , Reconhecimento Psicológico/efeitos dos fármacos , Reflexo de Sobressalto/efeitos dos fármacos , Estereoisomerismo , Natação/psicologia
4.
Neuropsychopharmacology ; 33(3): 574-87, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17460614

RESUMO

The characterization of the first selective orally active and brain-penetrant beta3-adrenoceptor agonist, SR58611A (amibegron), has opened new possibilities for exploring the involvement of this receptor in stress-related disorders. By using a battery of tests measuring a wide range of anxiety-related behaviors in rodents, including the mouse defense test battery, the elevated plus-maze, social interaction, stress-induced hyperthermia, four-plate, and punished drinking tests, we demonstrated for the first time that the stimulation of the beta3 receptor by SR58611A resulted in robust anxiolytic-like effects, with minimal active doses ranging from 0.3 to 10 mg/kg p.o., depending on the procedure. These effects paralleled those obtained with the prototypical benzodiazepine anxiolytic diazepam or chlordiazepoxide. Moreover, when SR58611A was tested in acute or chronic models of depression in rodents, such as the forced-swimming and the chronic mild stress tests, it produced antidepressant-like effects, which were comparable in terms of the magnitude of the effects to those of the antidepressant fluoxetine or imipramine. Supporting these behavioral data, SR58611A modified spontaneous sleep parameters in a manner comparable to that observed with fluoxetine. Importantly, SR58611A was devoid of side effects related to cognition (as shown in the Morris water maze and object recognition tasks), motor activity (in the rotarod), alcohol interaction, or physical dependence. Antagonism studies using pharmacological tools targeting a variety of neurotransmitters involved in anxiety and depression and the use of mice lacking the beta3 adrenoceptor suggested that these effects of SR58611A are mediated by beta3 adrenoceptors. Taken as a whole, these findings indicate that the pharmacological stimulation of beta3 adrenoceptors may represent an innovative approach for the treatment of anxiety and depressive disorders.


Assuntos
Agonistas de Receptores Adrenérgicos beta 3 , Agonistas Adrenérgicos beta/uso terapêutico , Transtornos de Ansiedade/tratamento farmacológico , Transtorno Depressivo/tratamento farmacológico , Tetra-Hidronaftalenos/uso terapêutico , Agonistas Adrenérgicos beta/administração & dosagem , Agressão/efeitos dos fármacos , Animais , Ansiolíticos/farmacologia , Antidepressivos de Segunda Geração/farmacologia , Antidepressivos Tricíclicos/farmacologia , Transtornos de Ansiedade/psicologia , Comportamento Animal/efeitos dos fármacos , Cognição/efeitos dos fármacos , Transtorno Depressivo/psicologia , Diazepam/farmacologia , Etanol/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Fluoxetina/farmacologia , Gerbillinae , Imipramina/farmacologia , Relações Interpessoais , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Ratos Wistar , Receptores Adrenérgicos beta 3/genética , Sono/efeitos dos fármacos , Transtornos Relacionados ao Uso de Substâncias/psicologia , Natação/psicologia , Tetra-Hidronaftalenos/administração & dosagem
5.
Eur J Neurosci ; 26(12): 3458-64, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18052990

RESUMO

In the present study, we examined the occurrence and potential regulation of endocannabinoid release by cannabinoid CB1 receptors in the rat brain. To this end, we developed a highly sensitive (limit of sensitivity 30-300 amol) new analytical method, combining online brain microdialysis with solid-phase extraction-liquid chromatography-tandem mass spectrometry, which allowed the detection in real time of trace amounts of endocannabinoids in the extracellular fluid. In the hypothalamus, anandamide and 2-arachidonoyl-glycerol release was stimulated following depolarization via local administration of K(+), with or without addition of Ca(2+), or glutamate application. Inhibition of fatty acid amide hydrolase by systemic administration of intraperitoneal (i.p.) URB597 (0.5 mg/kg) induced an increase of anandamide, but not 2-arachidonoyl-glycerol, outflow. The CB1 receptor antagonist rimonabant (10 mg/kg i.p.) increased, whereas the CB1 agonist WIN55,212-2 (2.5 mg/kg i.p.) decreased, anandamide release. Interestingly, the same treatments induced opposite changes in 2-arachidonoyl-glycerol release. At a dose of 3 mg/kg i.p., which by itself did not affect endocannabinoid release, rimonabant fully antagonized the effect of WIN55,212-2 (2.5 mg/kg i.p.). Taken together, these results suggest that CB1 receptors are able to control the local release of endocannabinoids in the hypothalamus via a feedback mechanism and strengthen the view that anandamide and 2-arachidonoyl-glycerol have distinct physiological roles.


Assuntos
Moduladores de Receptores de Canabinoides/metabolismo , Cromatografia Líquida , Endocanabinoides , Hipotálamo/metabolismo , Microdiálise , Receptor CB1 de Canabinoide/fisiologia , Espectrometria de Massas em Tandem , Amidoidrolases/antagonistas & inibidores , Animais , Ácidos Araquidônicos/metabolismo , Benzamidas/farmacologia , Benzoxazinas/farmacologia , Canabinoides/antagonistas & inibidores , Canabinoides/farmacologia , Carbamatos/farmacologia , Líquido Extracelular/metabolismo , Glicerídeos/metabolismo , Hipotálamo/efeitos dos fármacos , Masculino , Morfolinas/farmacologia , Naftalenos/farmacologia , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/metabolismo , Potássio/farmacologia , Pirazóis/farmacologia , Ratos , Ratos Wistar , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Rimonabanto
6.
Hepatology ; 46(1): 122-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17526015

RESUMO

This study investigated the effects of rimonabant (SR141716), an antagonist of the cannabinoid receptor type 1 (CB1), on obesity-associated hepatic steatosis and related features of metabolic syndrome: inflammation (elevated plasma levels of tumor necrosis factor alpha [TNFalpha]), dyslipidemia, and reduced plasma levels of adiponectin. We report that oral treatment of obese (fa/fa) rats with rimonabant (30 mg/kg) daily for 8 weeks abolished hepatic steatosis. This treatment reduced hepatomegaly, reduced elevation of plasma levels of enzyme markers of hepatic damage (alanine aminotransferase, gamma glutamyltransferase, and alkaline phosphatase) and decreased the high level of local hepatic TNFalpha currently associated with steatohepatitis. In parallel, treatment of obese (fa/fa) rats with rimonabant reduced the high plasma level of the proinflammatory cytokine TNFalpha and increased the reduced plasma level of the anti-inflammatory hormone adiponectin. Finally, rimonabant treatment also improved dyslipidemia by both decreasing plasma levels of triglycerides, free fatty acids, and total cholesterol and increasing the HDLc/LDLc ratio. All the effects of rimonabant found in this study were not or only slightly observed in pair-fed obese animals, highlighting the additional beneficial effects of treatment with rimonabant compared to diet. These results demonstrate that rimonabant plays a hepatoprotective role and suggest that this CB1 receptor antagonist potentially has clinical applications in the treatment of obesity-associated liver diseases and related features of metabolic syndrome.


Assuntos
Fígado Gorduroso/prevenção & controle , Síndrome Metabólica/prevenção & controle , Obesidade/complicações , Piperidinas/uso terapêutico , Pirazóis/uso terapêutico , Animais , Antagonistas de Receptores de Canabinoides , Fígado Gorduroso/etiologia , Inflamação/prevenção & controle , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Síndrome Metabólica/etiologia , RNA/genética , RNA/isolamento & purificação , Ratos , Ratos Zucker , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rimonabanto , Fator de Necrose Tumoral alfa/sangue
7.
Neuropsychopharmacology ; 32(1): 1-16, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17019409

RESUMO

In this paper, we report on the pharmacological and functional profile of SSR180711 (1,4-Diazabicyclo[3.2.2]nonane-4-carboxylic acid, 4-bromophenyl ester), a new selective alpha7 acetylcholine nicotinic receptor (n-AChRs) partial agonist. SSR180711 displays high affinity for rat and human alpha7 n-AChRs (K(i) of 22+/-4 and 14+/-1 nM, respectively). Ex vivo (3)[H]alpha-bungarotoxin binding experiments demonstrate that SSR180711 rapidly penetrates into the brain (ID(50)=8 mg/kg p.o.). In functional studies performed with human alpha7 n-AChRs expressed in Xenopus oocytes or GH4C1 cells, the compound shows partial agonist effects (intrinsic activity=51 and 36%, EC(50)=4.4 and 0.9 microM, respectively). In rat cultured hippocampal neurons, SSR180711 induced large GABA-mediated inhibitory postsynaptic currents and small alpha-bungarotoxin sensitive currents through the activation of presynaptic and somato-dendritic alpha7 n-AChRs, respectively. In mouse hippocampal slices, the compound increased the amplitude of both glutamatergic (EPSCs) and GABAergic (IPSCs) postsynaptic currents evoked in CA1 pyramidal cells. In rat and mouse hippocampal slices, a concentration of 0.3 muM of SSR180711 increased long-term potentiation (LTP) in the CA1 field. Null mutation of the alpha7 n-AChR gene totally abolished SSR180711-induced modulation of EPSCs, IPSCs and LTP in mice. Intravenous administration of SSR180711 strongly increased the firing rate of single ventral pallidum neurons, extracellularly recorded in anesthetized rats. In microdialysis experiments, administration of the compound (3-10 mg/kg i.p.) dose-dependently increased extracellular acetylcholine (ACh) levels in the hippocampus and prefrontal cortex of freely moving rats. Together, these results demonstrate that SSR180711 is a selective and partial agonist at human, rat and mouse alpha7 n-AChRs, increasing glutamatergic neurotransmission, ACh release and LTP in the hippocampus.


Assuntos
Agonistas Nicotínicos/farmacologia , Agonistas Nicotínicos/farmacocinética , Receptores Nicotínicos/fisiologia , Animais , Animais Recém-Nascidos , Sítios de Ligação/efeitos dos fármacos , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Interações Medicamentosas , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Hipocampo/citologia , Humanos , Técnicas In Vitro , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Agonistas Nicotínicos/química , Antagonistas Nicotínicos/farmacologia , Oócitos/fisiologia , Técnicas de Patch-Clamp/métodos , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/fisiologia , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/deficiência , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Receptor Nicotínico de Acetilcolina alfa7 , Ácido gama-Aminobutírico/farmacologia
8.
Neuropsychopharmacology ; 32(1): 17-34, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16936709

RESUMO

SSR180711 (4-bromophenyl 1,4diazabicyclo(3.2.2) nonane-4-carboxylate, monohydrochloride) is a selective alpha7 nicotinic receptor (n-AChR) partial agonist. Based on the purported implication of this receptor in cognitive deficits associated with schizophrenia, the present study assessed efficacy of SSR180711 (i.p. and p.o.) in different types of learning and memory involved in this pathology. SSR180711 enhanced episodic memory in the object recognition task in rats and mice (MED: 0.3 mg/kg), an effect mediated by the alpha7 n-AChR, as it was no longer seen in mice lacking this receptor. Efficacy was retained after repeated treatment (eight administrations over 5 days, 1 mg/kg), indicating lack of tachyphylaxia. SSR180711 also reversed (MED: 0.3 mg/kg) MK-801-induced deficits in retention of episodic memory in rats (object recognition). The drug reversed (MED: 0.3 mg/kg) selective attention impaired by neonatal phencyclidine (PCP) treatment and restored MK-801- or PCP-induced memory deficits in the Morris or linear maze (MED: 1-3 mg/kg). In neurochemical and electrophysiological correlates of antipsychotic drug action, SSR180711 increased extracellular levels of dopamine in the prefrontal cortex (MED: 1 mg/kg) and enhanced (3 mg/kg) spontaneous firing of retrosplenial cortex neurons in rats. Selectivity of SSR180711 was confirmed as these effects were abolished by methyllycaconitine (3 mg/kg, i.p. and 1 mg/kg, i.v., respectively), a selective alpha7 n-AChR antagonist. Additional antidepressant-like properties of SSR180711 were demonstrated in the forced-swimming test in rats (MED: 1 mg/kg), the maternal separation-induced ultrasonic vocalization paradigm in rat pups (MED: 3 mg/kg) and the chronic mild stress procedure in mice (10 mg/kg o.d. for 3 weeks). Taken together, these findings characterize SSR180711 as a promising new agent for the treatment of cognitive symptoms of schizophrenia. The antidepressant-like properties of SSR180711 are of added interest, considering the high prevalence of depressive symptoms in schizophrenic patients.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Agonistas Nicotínicos/uso terapêutico , Receptores Nicotínicos/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Maleato de Dizocilpina/farmacologia , Dopamina/metabolismo , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Feminino , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenciclidina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Nicotínicos/deficiência , Reconhecimento Psicológico/efeitos dos fármacos , Esquizofrenia/complicações , Receptor Nicotínico de Acetilcolina alfa7
9.
Mol Pharmacol ; 69(2): 471-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16282221

RESUMO

Adipocyte cell proliferation is an important process in body fat mass development in obesity. Adiponectin or Acrp30 is an adipocytokine exclusively expressed and secreted by adipose tissue that regulates lipid and glucose metabolism and plays a key role in body weight regulation and homeostasis. Adiponectin mRNA expression in adipose tissue and plasma level of adiponectin are decreased in obesity and type 2 diabetes. In obese rodents, the selective CB(1) receptor antagonist rimonabant reduces food intake and body weight and improves lipid and glucose parameters. We have reported previously that rimonabant stimulated adiponectin mRNA expression in adipose tissue of obese fa/fa rats, by a direct effect on adipocytes. We report here that rimonabant (10-400 nM) inhibits cell proliferation of cultured mouse 3T3 F442A preadipocytes in a concentration-dependent manner. In parallel to this inhibitory effect on preadipocyte cell proliferation, rimonabant (25-100 nM) stimulates mRNA expression and protein levels of two late markers of adipocyte differentiation (adiponectin and glyceraldehyde-3-phosphate dehydrogenase) with a maximal effect at 100 nM, without inducing the accumulation of lipid droplets. Furthermore, treatment of mouse 3T3 F442A preadipocytes with rimonabant (100 nM) inhibits basal and serum-induced p42/44 mitogen-activated protein (MAP) kinase activity. These results suggest that inhibition of MAP kinase activity by rimonabant may be one of mechanisms involved in the inhibition of 3T3 F442A preadipocyte cell proliferation and stimulation of adiponectin and GAPDH expression. The inhibition of preadipocyte cell proliferation and the induction of adipocyte late "maturation" may participate in rimonabant-induced antiobesity effects, particularly the reduction of body fat mass.


Assuntos
Adipócitos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Adipócitos/citologia , Adiponectina/genética , Adiponectina/metabolismo , Animais , Biomarcadores/análise , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/genética , Gliceraldeído-3-Fosfato Desidrogenase (Fosforiladora)/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , RNA Mensageiro/análise , RNA Mensageiro/metabolismo , Rimonabanto
10.
Neuropsychopharmacology ; 30(11): 1963-85, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15956994

RESUMO

Noncompetitive N-methyl-D-aspartate (NMDA) blockers induce schizophrenic-like symptoms in humans, presumably by impairing glutamatergic transmission. Therefore, a compound potentiating this neurotransmission, by increasing extracellular levels of glycine (a requisite co-agonist of glutamate), could possess antipsychotic activity. Blocking the glycine transporter-1 (GlyT1) should, by increasing extracellular glycine levels, potentiate glutamatergic neurotransmission. SSR504734, a selective and reversible inhibitor of human, rat, and mouse GlyT1 (IC50=18, 15, and 38 nM, respectively), blocked reversibly the ex vivo uptake of glycine (mouse cortical homogenates: ID50: 5 mg/kg i.p.), rapidly and for a long duration. In vivo, it increased (minimal efficacious dose (MED): 3 mg/kg i.p.) extracellular levels of glycine in the rat prefrontal cortex (PFC). This resulted in an enhanced glutamatergic neurotransmission, as SSR504734 potentiated NMDA-mediated excitatory postsynaptic currents (EPSCs) in rat hippocampal slices (minimal efficacious concentration (MEC): 0.5 microM) and intrastriatal glycine-induced rotations in mice (MED: 1 mg/kg i.p.). It normalized activity in rat models of hippocampal and PFC hypofunctioning (through activation of presynaptic CB1 receptors): it reversed the decrease in electrically evoked [3H]acetylcholine release in hippocampal slices (MEC: 10 nM) and the reduction of PFC neurons firing (MED: 0.3 mg/kg i.v.). SSR504734 prevented ketamine-induced metabolic activation in mice limbic areas and reversed MK-801-induced hyperactivity and increase in EEG spectral energy in mice and rats, respectively (MED: 10-30 mg/kg i.p.). In schizophrenia models, it normalized a spontaneous prepulse inhibition deficit in DBA/2 mice (MED: 15 mg/kg i.p.), and reversed hypersensitivity to locomotor effects of d-amphetamine and selective attention deficits (MED: 1-3 mg/kg i.p.) in adult rats treated neonatally with phencyclidine. Finally, it increased extracellular dopamine in rat PFC (MED: 10 mg/kg i.p.). The compound showed additional activity in depression/anxiety models, such as the chronic mild stress in mice (10 mg/kg i.p.), ultrasonic distress calls in rat pups separated from their mother (MED: 1 mg/kg s.c.), and the increased latency of paradoxical sleep in rats (MED: 30 mg/kg i.p.). In conclusion, SSR504734 is a potent and selective GlyT1 inhibitor, exhibiting activity in schizophrenia, anxiety and depression models. By targeting one of the primary causes of schizophrenia (hypoglutamatergy), it is expected to be efficacious not only against positive but also negative symptoms, cognitive deficits, and comorbid depression/anxiety states.


Assuntos
Benzamidas/farmacologia , Química Encefálica/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Neurônios/efeitos dos fármacos , Piperidinas/farmacologia , Acetilcolina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Anfetamina/farmacologia , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Isótopos de Carbono/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Ritmo Circadiano/efeitos dos fármacos , Inibidores da Captação de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/química , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Feminino , Glicina/metabolismo , Hipocampo/citologia , Humanos , Técnicas In Vitro , Concentração Inibidora 50 , Masculino , Camundongos , Atividade Motora/efeitos dos fármacos , Inibição Neural/efeitos dos fármacos , Neurônios/fisiologia , Técnicas de Patch-Clamp/métodos , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/efeitos dos fármacos
11.
Biol Psychiatry ; 57(3): 261-7, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15691527

RESUMO

BACKGROUND: The endocannabinoid system has been implicated in the modulation of emotional processes. METHODS: These experiments aimed to investigate the effects of the cannabinoid CB1 receptor antagonist rimonabant (SR141716) in animal models measuring aspects of emotional reactivity and depression. RESULTS: Rimonabant had weak anxiolytic-like activity in the elevated plus-maze and failed to affect flight and risk assessment activities in the mouse defense test battery (MDTB). It produced clear anxiolytic-like effects in the Vogel conflict test (.3-3 mg/kg intraperitoneal [i.p.]) and on defensive aggression in the MDTB (1 and 10 mg/kg, i.p.). The effects of rimonabant in the MDTB paralleled those observed with CB1 receptor knockout mice in this procedure. In the forced-swimming test in rats and the tonic immobility paradigm in gerbils, rimonabant (3 and 10 mg/kg per os [p.o.]) produced antidepressant-like effects that were comparable to those observed with the reference antidepressant, fluoxetine. In the chronic mild stress model in mice, repeated administration of rimonabant (10 mg/kg, p.o.) for 5 weeks improved the deleterious effects produced by stress. CONCLUSIONS: These findings point further to a role for the endocannabinoid system in the modulation of emotional processes and suggest that it may be primarily involved in the adaptive responses to unavoidable stressful stimuli.


Assuntos
Comportamento Animal/efeitos dos fármacos , Emoções/efeitos dos fármacos , Modelos Animais , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Agressão/efeitos dos fármacos , Análise de Variância , Animais , Ansiolíticos/farmacologia , Antidepressivos de Segunda Geração/uso terapêutico , Depressão/tratamento farmacológico , Diazepam/farmacologia , Relação Dose-Resposta a Droga , Eletrochoque/efeitos adversos , Fluoxetina/uso terapêutico , Gerbillinae , Resposta de Imobilidade Tônica/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Testes Neuropsicológicos , Piperidinas/uso terapêutico , Pirazóis/uso terapêutico , Ratos , Ratos Wistar , Rimonabanto
12.
J Pharmacol Exp Ther ; 309(1): 414-24, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14722330

RESUMO

4-chloro-3-[(3R)-(+)-5-chloro-1-(2,4-dimethoxybenzyl)-3-methyl-2-oxo-2,3-dihydro-1H-indol-3-yl]-N-ethyl-N-(3-pyridylmethyl)benzamide, hydrochloride (SSR126768A), a new potent and selective, orally active oxytocin (OT) receptor antagonist was characterized in several biochemical and pharmacological models. In binding studies, SSR126768A showed nanomolar affinity for rat and human recombinant and native OT receptors (K(i) = 0.44 nM) and exhibited much lower affinity for V(1a), V(1b), and V(2) receptors. In addition, it did not interact with a large number of other receptors, enzymes, and ion channels (1 microM). In autoradiographic experiments performed on at-term human pregnant uterus sections, SSR126768A dose dependently displaced [I(125)]d(CH(2))(5)[Tyr(Me)(2), Thr(4), Orn(8) (125)I-Tyr-NH(2)(9)]VT in situ labeling to OT receptors highly expressed in these tissues. In functional studies, SSR126768A behaved as a full antagonist and potently antagonized OT-induced intracellular Ca(2+) increase (K(i) = 0.50 nM) and prostaglandin release (K(i) = 0.45 nM) in human uterine smooth muscle cells. In rat isolated myometrium, OT-induced uterine contractions were competitively antagonized by SSR126768A (pA(2) = 8.47). Similarly, in human pregnant myometrial strips, SSR126768A inhibited the contractile uterine response to OT. In conscious telemetrated rats, oral administration of SSR126768A (1-10 mg/kg) produced a competitive inhibition of the dose response to OT on uterine contractions up to 24 h at 3 mg/kg p.o.; no tachyphylaxis was observed after 4-day repeated treatment. Finally, SSR126768A (30 mg/kg p.o.) significantly delayed parturition in pregnant rats in labor similar to ritodrine (10 mg/kg p.o.). Thus, SSR126768A is a potent, highly selective, orally active OT receptor antagonist with a long duration of action. This molecule could find therapeutic application as a tocolytic agent for acute and chronic oral management of preterm labor.


Assuntos
Benzamidas/uso terapêutico , Indóis/uso terapêutico , Trabalho de Parto Prematuro/prevenção & controle , Receptores de Ocitocina/antagonistas & inibidores , Tocolíticos/uso terapêutico , Animais , Autorradiografia , Células CHO , Cálcio/metabolismo , Cricetinae , Feminino , Humanos , Parto/efeitos dos fármacos , Gravidez , Prenhez , Prostaglandinas/metabolismo , Ratos , Ratos Sprague-Dawley , Telemetria , Contração Uterina/efeitos dos fármacos
13.
Neuropsychopharmacology ; 28(11): 1889-902, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12902993

RESUMO

SSR181507 ((3-exo)-8-benzoyl-N-(((2S)7-chloro-2,3-dihydro-1,4-benzodioxin-1-yl)methyl)-8-azabicyclo(3.2.1)octane-3-methanamine monohydrochloride) is a novel tropanemethanamine benzodioxane that displays antagonist activity at dopamine D(2) receptors and agonist activity at 5-HT(1A) receptors. SSR181507 antagonized apomorphine-induced climbing in mice and stereotypies in rats (ED(50) of 2 and 3.4 mg/kg i.p., respectively) and blocked D-amphetamine-induced hyperlocomotion in rats at lower doses (0.3-1 mg/kg i.p.). At 1-10 mg/kg, it was found to disrupt active avoidance in mice. SSR181507 did not induce catalepsy in rats (MED>60 mg/kg i.p.) and antagonized (3-10 mg/kg i.p.) haloperidol-induced catalepsy. SSR181507 was also active in two models sensitive to antidepressant/anxiolytic drugs: in a guinea-pig pup/mother separation test, it decreased (1-3 mg/kg i.p.) the time spent vocalizing during the separation episode, and in a lithium-induced taste aversion procedure in rats, it partially reversed (3 mg/kg i.p.) the decrease of intake of a saccharin solution. Furthermore, SSR181507 increased (3 mg/kg i.p.) the latency time to paradoxical sleep in rats, an effect commonly observed with antidepressants. Coadministration of the selective 5-HT(1A) blocker SL88.0338 produced catalepsy and antagonized the effects of SSR181507 in the depression/anxiety tests, confirming the view that activation of 5-HT(1A) receptors confers an atypical profile on SSR181507, and is responsible for its antidepressant/anxiolytic properties. Finally, SSR181507 (1-3 mg/kg) did not affect memory performance in a Morris water maze task in rats. The pharmacological profile of SSR181507 suggests that it should control the symptoms of schizophrenia, in the absence of extrapyramidal signs and cognitive deficits, with the additional benefit of antidepressant/anxiolytic activities.


Assuntos
Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Dioxanos/farmacologia , Antagonistas dos Receptores de Dopamina D2 , Agonistas do Receptor 5-HT1 de Serotonina , Antagonistas da Serotonina/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Tropanos/farmacologia , Animais , Feminino , Cobaias , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Valor Preditivo dos Testes , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptor 5-HT1A de Serotonina/fisiologia , Receptores de Dopamina D2/fisiologia
14.
Neuropsychopharmacology ; 28(12): 2064-76, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12902994

RESUMO

SSR181507 ((3-exo)-8-benzoyl-N-[[(2S)7-chloro-2,3-dihydro-1,4-benzodioxin-1-yl]methyl]-8-azabicyclo[3.2.1]octane-3-methanamine monohydrochloride) is a novel tropanemethanamine benzodioxane derivative that possesses high and selective affinities for D2-like and 5-HT(1A) receptors (K(I)=0.8, 0.2, and 0.2 nM for human D(2), D(3), and 5-HT(1A), respectively). In vivo, SSR181507 inhibited [(3)H]raclopride binding to D(2) receptors in the rat (ID(50)=0.9 and 1 mg/kg, i.p. in limbic system and striatum, respectively). It displayed D(2) antagonist and 5-HT(1A) agonist properties in the same concentration range in vitro (IC(50)=5.3 nM and EC(50)=2.3 nM, respectively, in the GTPgammaS model) and in the same dose range in vivo (ED(50)=1.6 and 0.7 mg/kg, i.p. on striatal DA and 5-HT synthesis, respectively, and 0.03-0.3 mg/kg, i.v. on dorsal raphe nucleus firing rate). It selectively enhanced Fos immunoreactivity in mesocorticolimbic areas as compared to the striatum. This regional selectivity was confirmed in electrophysiological studies where SSR181507, given acutely (0.1-3 mg/kg, i.p.) or chronically (3 mg/kg, i.p., o.d., 22 days), increased or decreased, respectively, the number of spontaneous active DA cells in the ventral tegmental area, but not in the substantia nigra. Moreover, SSR181507 increased both basal and phasic DA efflux (as assessed by microdialysis and electrochemistry) in the medial prefrontal cortex and nucleus accumbens, but not in the striatum. This study shows that the combination of D(2) receptor antagonism and 5-HT(1A) agonism, in the same dose range, confers on SSR181507 a unique neurochemical and electrophysiological profile and suggests the potential of this compound for the treatment of the main dimensions of schizophrenia.


Assuntos
Encéfalo/efeitos dos fármacos , Antagonistas dos Receptores de Dopamina D2 , Antagonistas do Receptor 5-HT1 de Serotonina , 5-Hidroxitriptofano/metabolismo , Potenciais de Ação/efeitos dos fármacos , Animais , Antipsicóticos/farmacologia , Comportamento Animal/efeitos dos fármacos , Sítios de Ligação , Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Contagem de Células , Dioxanos/farmacologia , Dopamina/metabolismo , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Interações Medicamentosas , Estimulação Elétrica , Eletroquímica/métodos , Guanosina 5'-O-(3-Tiotrifosfato)/farmacocinética , Haloperidol/farmacologia , Humanos , Imuno-Histoquímica/métodos , Técnicas In Vitro , Levodopa/metabolismo , Masculino , Microdiálise/métodos , Neurônios/efeitos dos fármacos , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Piridinas/farmacologia , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptor 5-HT1A de Serotonina/química , Receptores de Dopamina D2/química , Antagonistas da Serotonina/farmacologia , Isótopos de Enxofre/farmacocinética , Fatores de Tempo , Tropanos/farmacologia
15.
CNS Drug Rev ; 9(1): 3-20, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12595909

RESUMO

SL651498 (6-fluoro-9-methyl-2-phenyl-4-(pyrrolidin-1-yl-carbonyl)-2,9-dihydro-1H-pyrido[3,4-b]indol-1-one) was identified as a drug development candidate from a research program designed to discover subtype-selective GABA(A) receptor agonists for the treatment of generalized anxiety disorder and muscle spasms. The drug displays high affinity for rat native GABA(A) receptors containing alpha(1) (K(i) = 6.8 nM) and alpha(2) (K(i) = 12.3 nM) subunits, and weaker affinity for alpha5-containing GABA(A) receptors (K(i) = 117 nM). Studies on recombinant rat GABA(A) receptors confirm these findings and indicate intermediate affinity for the alpha(3)beta(2)gamma(2) subtype. SL651498 behaves as a full agonist at recombinant rat GABA(A) receptors containing alpha(2) and alpha(3) subunits, and as a partial agonist at recombinant GABA(A) receptors expressing alpha(1) and alpha(5) subunits. SL651498 produced anxiolytic-like and skeletal muscle relaxant effects qualitatively similar to those of benzodiazepines (BZs) [minimal effective dose (MED): 1 to 10 mg/kg, i.p. and 3 to 10 mg/kg, p.o.]. However, unlike these latter drugs, SL651498 induced muscle weakness, ataxia or sedation at doses much higher than those having anxiolytic-like activity (MED: 30 to 100 mg/kg, i.p. or p.o.). Moreover, in contrast to BZs, SL651498 did not produce tolerance to its anticonvulsant activity or physical dependence. It was much less active than BZs in potentiating the depressant effects of ethanol or impairing cognitive processes in rodents. The differential profile of SL651498 as compared to BZs may be related to its selective efficacy at the alpha(2)- and alpha(3)-containing GABA(A) receptors. This suggests that selectively targeting GABA(A) receptor subtypes can lead to drugs with increased clinical specificity. SL651498 represents a promising alternative to agents currently used for the treatment of anxiety disorders and muscle spasms without the major side effects seen with classical BZs.


Assuntos
Transtornos de Ansiedade/tratamento farmacológico , Agonistas de Receptores de GABA-A , Indóis/farmacologia , Indóis/uso terapêutico , Pirróis/farmacologia , Pirróis/uso terapêutico , Espasmo/tratamento farmacológico , Amnésia Anterógrada/induzido quimicamente , Animais , Ataxia/induzido quimicamente , Sistema Nervoso Central/efeitos dos fármacos , Aprendizagem por Discriminação/efeitos dos fármacos , Discriminação Psicológica/efeitos dos fármacos , Interações Medicamentosas , Tolerância a Medicamentos , Etanol/farmacologia , Agonistas GABAérgicos/administração & dosagem , Agonistas GABAérgicos/efeitos adversos , Agonistas GABAérgicos/química , Agonistas GABAérgicos/farmacologia , Agonistas GABAérgicos/uso terapêutico , Indóis/efeitos adversos , Indóis/química , Camundongos , Pirróis/efeitos adversos , Pirróis/química , Ratos , Transtornos Relacionados ao Uso de Substâncias/etiologia
16.
J Pharmacol Exp Ther ; 302(2): 731-41, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12130738

RESUMO

SL65.0155 [5-(8-amino-7-chloro-2,3-dihydro-1,4-benzodioxin-5-yl)-3-[1-(2-phenyl ethyl)-4-piperidinyl]-1,3,4-oxadiazol-2(3H)-one monohydrochloride] is a novel benzodioxanoxadiazolone compound with high affinity for human 5-hydroxytryptamine (5-HT)(4) receptors (K(i) of 0.6 nM) and good selectivity (greater than 100-fold for all other receptors tested). In cells expressing the 5-HT(4(b)) and 5-HT(4(e)) splice variants, SL65.0155 acted as a partial agonist, stimulating cAMP production with a maximal effect of 40 to 50% of serotonin. However, in the rat esophagus preparation, SL65.0155 acted as a 5-HT(4) antagonist with a pK(b) of 8.81. In addition, SL65.0155 potently improved performance in several tests of learning and memory. In the object recognition task, it improved retention at 24 h when administered i.p. or p.o. (0.001-0.1 mg/kg). This effect was antagonized by the 5-HT(4) antagonist SDZ 205,557, itself without effect, demonstrating that the promnesic effects of SL65.0155 are mediated by 5-HT(4) agonism. SL65.0155 also reversed the cognitive deficits of aged rats in the linear maze task and the scopolamine-induced deficit of mice in the water maze task. Furthermore, the combined administration of an inactive dose of SL65.0155 with the cholinesterase inhibitor rivastigmine resulted in a significant promnesic effect, suggesting a synergistic interaction. SL65.0155 was devoid of unwanted cardiovascular, gastrointestinal, or central nervous system effects with doses up to more than 100-fold higher than those active in the cognitive tests. These results characterize SL65.0155 as a novel promnesic agent acting via 5-HT(4) receptors, with an excellent preclinical profile. Its broad range of activity in cognitive tests and synergism with cholinesterase inhibitors suggest that SL65.0155 represents a promising new agent for the treatment of dementia.


Assuntos
Cognição/fisiologia , AMP Cíclico/metabolismo , Dioxanos/farmacologia , Aprendizagem em Labirinto/fisiologia , Oxidiazóis/farmacologia , Receptores de Serotonina/fisiologia , Agonistas do Receptor de Serotonina/farmacologia , Processamento Alternativo , Animais , Pressão Sanguínea/efeitos dos fármacos , Células CHO , Células COS , Chlorocebus aethiops , Cognição/efeitos dos fármacos , Cricetinae , Esôfago/efeitos dos fármacos , Esôfago/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Cobaias , Frequência Cardíaca/efeitos dos fármacos , Íleo/efeitos dos fármacos , Íleo/fisiologia , Aprendizagem em Labirinto/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/fisiologia , Ensaio Radioligante , Ratos , Receptores de Serotonina/efeitos dos fármacos , Receptores de Serotonina/genética , Receptores 5-HT4 de Serotonina , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Transfecção
17.
J Pharmacol Exp Ther ; 301(3): 1067-78, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12023539

RESUMO

In the present study, we have investigated the potential neuroprotective effects of a novel peripheral benzodiazepine binding site (PBR) ligand, 7-chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (SSR180575), in models of central and peripheral neurodegeneration in vivo and its effect on steroid concentrations in plasma and nervous tissue. SSR180575 shows high affinity (IC(50), 2.5-3.5 nM) and selectivity for the rat and human PBR and potently inhibits the in vivo binding of [(3)H]alpidem to PBR in the rat brain and spleen after oral or i.p. administration (ID(50), 0.1-0.3 mg/kg). In an experimental model of motoneuron degeneration induced by facial nerve axotomy in the immature rat, SSR180575 given i.p. or orally for 8 days rescued facial motoneurons, increasing their survival by 40 to 72% at 6 and 10 mg/kg p.o. b.i.d. Moreover, in this model, SSR180575 (10 mg/kg p.o. b.i.d.) increased by 87% the number of motoneurons immunoreactive to peripherin, a type III intermediate filament, whose expression is up-regulated during nerve regeneration. SSR180575 also improved functional recovery in acrylamide-induced neuropathy in the rat when given therapeutically at 2.5 to 10 mg/kg/day p.o. Furthermore, SSR180575 (3 mg/kg i.p. b.i.d.) accelerated functional recovery of the blink reflex after local injury of the facial nerve in the rat. SSR180575 increased pregnenolone accumulation in the brain and sciatic nerve (+100% at 3 mg/kg i.p.), suggesting that its neuroprotective effects are steroid-mediated. These results indicate that PBR ligands (e.g., SSR180575) promote neuronal survival and repair in axotomy and neuropathy models and have potential for the treatment of neurodegenerative diseases (e.g., peripheral neuropathies or amyotrophic lateral sclerosis).


Assuntos
Acetamidas/metabolismo , Acetamidas/farmacologia , Agonistas de Receptores de GABA-A , Indóis/metabolismo , Indóis/farmacologia , Neurônios Motores/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Acetamidas/química , Envelhecimento/efeitos dos fármacos , Envelhecimento/fisiologia , Animais , Ansiolíticos/metabolismo , Axotomia , Ligação Competitiva , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Nervo Facial/efeitos dos fármacos , Nervo Facial/metabolismo , Antagonistas de Receptores de GABA-A , Humanos , Imidazóis/metabolismo , Indóis/química , Ligantes , Masculino , Neurônios Motores/metabolismo , Fármacos Neuroprotetores/química , Fármacos Neuroprotetores/metabolismo , Piridazinas/química , Piridazinas/metabolismo , Piridazinas/farmacologia , Piridinas/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/metabolismo
18.
Proc Natl Acad Sci U S A ; 99(9): 6370-5, 2002 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-11959912

RESUMO

The limbic localization of the arginine vasopressin V(1b) receptor has prompted speculation as to a potential role of this receptor in the control of emotional processes. To investigate this possibility, we have studied the behavioral effects of SSR149415, the first selective and orally active non-peptide antagonist of vasopressin V(1b) receptors, in a variety of classical (punished drinking, elevated plus-maze, and light/dark tests) and atypical (fear/anxiety defense test battery and social defeat-induced anxiety) rodent models of anxiety, and in two models of depression [forced swimming and chronic mild stress (CMS)]. When tested in classical tests of anxiety, SSR149415 produced anxiolytic-like activity at doses that ranged from 1 to 30 mg/kg (i.p. or p.o.), but the magnitude of these effects was overall less than that of the benzodiazepine anxiolytic diazepam, which was used as a positive control. In contrast, SSR149415 produced clear-cut anxiolytic-like activity in models involving traumatic stress exposure, such as the social defeat paradigm and the defense test battery (1-30 mg/kg, p.o.). In the forced swimming test, SSR149415 (10-30 mg/kg, p.o.) produced antidepressant-like effects in both normal and hypophysectomized rats. Moreover, in the CMS model in mice, repeated administration of SSR149415 (10 and 30 mg/kg, i.p.) for 39 days improved the degradation of the physical state, anxiety, despair, and the loss of coping behavior produced by stress. These findings point to a role for vasopressin in the modulation of emotional processes via the V(1b) receptor, and suggest that its blockade may represent a novel avenue for the treatment of affective disorders.


Assuntos
Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Antagonistas dos Receptores de Hormônios Antidiuréticos , Ansiedade/tratamento farmacológico , Depressão/tratamento farmacológico , Indóis/farmacologia , Pirrolidinas/farmacologia , Estresse Fisiológico , Animais , Antidepressivos de Segunda Geração/farmacologia , Antidepressivos Tricíclicos/farmacologia , Comportamento Animal/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Fluoxetina/farmacologia , Imipramina/farmacologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Medição de Risco , Estresse Psicológico , Fatores de Tempo
19.
J Pharmacol Exp Ther ; 301(1): 322-32, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11907190

RESUMO

4-(2-Chloro-4-methoxy-5-methylphenyl)-N-[(1S)-2-cyclopropyl-1- (3-fluoro-4-methylphenyl)ethyl]5-methyl-N-(2-propynyl)-1,3-thiazol-2-amine hydrochloride (SSR125543A), a new 2-aminothiazole derivative, shows nanomolar affinity for human cloned or native corticotrophin-releasing factor (CRF)(1) receptors (pK(i) values of 8.73 and 9.08, respectively), and a 1000-fold selectivity for CRF(1) versus CRF(2 alpha) receptor and CRF binding protein. SSR125543A antagonizes CRF-induced stimulation of cAMP synthesis in human retinoblastoma Y 79 cells (IC(50) = 3.0 +/- 0.4 nM) and adrenocorticotropin hormone (ACTH) secretion in mouse pituitary tumor AtT-20 cells. SSR125543A is devoid of agonist activity in these models. Its brain penetration was demonstrated in rats by using an ex vivo [(125)I-Tyr(0)] ovine CRF binding assay. SSR125543A displaced radioligand binding to the CRF(1) receptor in the brain with an ID(50) of 6.5 mg/kg p.o. (duration of action >24 h). SSR125543A also inhibited the increase in plasma ACTH levels elicited in rats by i.v. CRF (4 microg/kg) injection (ID(50) = 1, 5, or 5 mg/kg i.v., i.p., and p.o., respectively); this effect lasted for more than 6 h when the drug was given orally at a dose of 30 mg/kg. SSR125543A (10 mg/kg p.o.) reduced by 73% the increase in plasma ACTH levels elicited by a 15-min restraint stress in rats. Moreover, SSR125543A (20 mg/kg i.p.) also antagonized the increase of hippocampal acetylcholine release induced by i.c.v. injection of 1 microg of CRF in rats. Finally, SSR125543A reduced forepaw treading induced by i.c.v. injection of 1 microg of CRF in gerbils (ID(50) = approximately 10 mg/kg p.o.). Altogether, these data indicate that SSR125543A is a potent, selective, and orally active CRF(1) receptor antagonist.


Assuntos
Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Tiazóis/farmacologia , Acetilcolina/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Animais , Química Encefálica/efeitos dos fármacos , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetinae , AMP Cíclico/biossíntese , Feminino , Gerbillinae , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Microdiálise , Ratos , Receptores de Hormônio Liberador da Corticotropina/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/metabolismo
20.
J Pharmacol Exp Ther ; 301(1): 333-45, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11907191

RESUMO

The present study investigated the effects of the novel corticotrophin-releasing factor (CRF)(1) receptor antagonist 4-(2-chloro-4-methoxy-5-methylphenyl)-N-[(1S)-2-cyclopropyl-1-(3-fluoro-4-methylphenyl)ethyl]5-methyl-N-(2-propynyl)-1,3-thiazol-2-amine hydrochloride (SSR125543A) in a variety of rodent models of anxiety, including conflict procedures (punished drinking and four-plate), exploration models (elevated plus-maze and light/dark), a fear/anxiety defense test battery, and several procedures based on stress-induced changes in physiological (isolation-induced hyperthermia and tail pinch-induced cortical norepinephrine release) or behavioral (social defeat-induced anxiety, maternal separation-induced vocalization) parameters. Moreover, the effects of SSR125543A were investigated in acute (forced swimming) and chronic (chronic mild stress; CMS) models of depression. SSR125543A and the CRF(1) receptor antagonist antalarmin displayed limited efficacy in exploration-based anxiety models. In contrast, both compounds produced clear-cut anxiolytic-like activity in models involving inescapable stress, including the conflict procedures, the social defeat-induced anxiety paradigm and the defense test battery (3-30 mg/kg i.p. or p.o.). These effects paralleled those of the anxiolytic diazepam. In addition, SSR125543A and antalarmin antagonized stress-induced hyperthermia, distress vocalization, and cortical norepinephrine release. In the forced swimming test, 30 mg/kg p.o. SSR125543A and 3 to 30 mg/kg p.o. antalarmin produced clear antidepressant-like effects. These latter results were strengthened by the findings from the CMS, which showed that repeated administration of 10 mg/kg i.p. SSR125543A for 30 days improved the degradation of the physical state, the reduction of body weight gain, and anxiety produced by stress. Together, these data indicate that SSR125543A shows good activity in acute and chronic tests of unavoidable stress exposure, suggesting that it may have a potential in the treatment of depression and some forms of anxiety disorders.


Assuntos
Comportamento Animal/efeitos dos fármacos , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Estresse Psicológico/tratamento farmacológico , Tiazóis/farmacologia , Animais , Antidepressivos/uso terapêutico , Ansiedade/tratamento farmacológico , Ansiedade/psicologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Conflito Psicológico , Relação Dose-Resposta a Droga , Comportamento Exploratório/efeitos dos fármacos , Medo/efeitos dos fármacos , Hipotermia/prevenção & controle , Masculino , Memória/efeitos dos fármacos , Camundongos , Atividade Motora/efeitos dos fármacos , Norepinefrina/metabolismo , Punição , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Isolamento Social/psicologia , Estresse Psicológico/fisiopatologia , Natação/psicologia , Vocalização Animal/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...