Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Mol Pharmacol ; 92(2): 151-161, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28588066

RESUMEN

N-methyl-d-aspartate (NMDA)-type ionotropic glutamate receptors mediate excitatory neurotransmission in the central nervous system and are critically involved in brain function. NMDA receptors are also implicated in psychiatric and neurological disorders and have received considerable attention as therapeutic targets. In this regard, administration of d-cycloserine (DCS), which is a glycine site NMDA receptor agonist, can enhance extinction of conditioned fear responses. The intriguing behavioral effects of DCS have been linked to its unique pharmacological profile among NMDA receptor subtypes (GluN1/2A-D), in which DCS is a superagonist at GluN2C-containing receptors compared with glycine and a partial agonist at GluN2B-containing receptors. Here, we identify (R)-2-amino-3-(4-(2-ethylphenyl)-1H-indole-2-carboxamido)propanoic acid (AICP) as a glycine site agonist with unique GluN2-dependent differences in agonist efficacy at recombinant NMDA receptor subtypes. AICP is a full agonist at GluN1/2A (100% response compared with glycine), a partial agonist at GluN1/2B and GluN1/2D (10% and 27%, respectively), and a highly efficacious superagonist at GluN1/2C receptors (353%). Furthermore, AICP potencies are enhanced compared with DCS with EC50 values in the low nanomolar range (1.7 nM at GluN1/2C). We show that GluN1/2C superagonism of AICP and DCS is mediated by overlapping but distinct mechanisms and that AICP selectively enhances responses from recombinant GluN1/2C receptors in the presence of physiological glycine concentrations. This functional selectivity of AICP for GluN2C-containing NMDA receptors is more pronounced compared with DCS, suggesting that AICP can be a useful tool compound for uncovering the roles of GluN2C subunits in neuronal circuit function and in the development of new therapeutic strategies.


Asunto(s)
Agonistas de Aminoácidos Excitadores/metabolismo , Glicina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/farmacología , Glicina/agonistas , Células HEK293 , Humanos , Proteínas del Tejido Nervioso/agonistas , Ratas , Receptores de N-Metil-D-Aspartato/agonistas , Xenopus laevis
2.
Eur J Neurosci ; 46(3): 1887-1896, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28635024

RESUMEN

Nav 1.1 (SCN1A) channels primarily located in gamma-aminobutyric acid (GABA)ergic fast-spiking interneurons are pivotal for action potential generation and propagation in these neurons. Inappropriate function of fast-spiking interneurons, leading to disinhibition of pyramidal cells and network desynchronization, correlates with decreased cognitive capability. Further, reduced functionality of Nav 1.1 channels is linked to various diseases in the central nervous system. There is, at present, however no subtype selective pharmacological activators of Nav 1.1 channels available for studying pharmacological modulation of interneuron function. In the current study, we identified a small molecule Nav 1.1 activator, 3-amino-5-(4-methoxyphenyl)thiophene-2-carboxamide, named AA43279, and provided an in vitro to in vivo characterization of the compound. In HEK-293 cells expressing human Nav 1.1 channels, AA43279 increased the Nav 1.1-mediated current in a concentration-dependent manner mainly by impairing the fast inactivation kinetics of the channels. In rat hippocampal brain slices, AA43279 increased the firing activity of parvalbumin-expressing, fast-spiking GABAergic interneurons and increased the spontaneous inhibitory post-synaptic currents (sIPSCs) recorded from pyramidal neurons. When tested in vivo, AA43279 had anti-convulsive properties in the maximal electroshock seizure threshold test. AA43279 was tested for off-target effects on 72 different proteins, including Nav 1.2, Nav 1.4, Nav 1.5, Nav 1.6 and Nav 1.7 and exhibited reasonable selectivity. Taken together, AA43279 might constitute a valuable tool compound for revealing biological functions of Nav 1.1 channels.


Asunto(s)
Anticonvulsivantes/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Convulsiones/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/farmacología , Tiofenos/farmacología , Potenciales de Acción , Animales , Anticonvulsivantes/síntesis química , Anticonvulsivantes/uso terapéutico , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Células HEK293 , Humanos , Interneuronas/metabolismo , Interneuronas/fisiología , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/uso terapéutico
3.
Bioorg Med Chem Lett ; 24(1): 288-93, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24291041

RESUMEN

In this Letter, we describe a chemical lead optimization campaign starting from a novel, weak α7 nicotinic acetylcholine receptor positive allosteric modulator (PAM) hit from a HTS screen. Exploration of the structure-activity relationships for α7 PAM potency, intrinsic hepatic clearance, the structure-property relationships for lipophilicity, and thermodynamic solubility, led to the identification of Lu AF58801: a potent, orally available, brain penetrant PAM of the α7 nicotinic acetylcholine receptor, showing efficacy in a novel object recognition task in rats treated subchronically with phencyclidine (PCP).


Asunto(s)
Encéfalo/efectos de los fármacos , Trastornos del Conocimiento/tratamiento farmacológico , Ciclopropanos/farmacología , Descubrimiento de Drogas , Alcohol Feniletílico/análogos & derivados , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Administración Oral , Regulación Alostérica/efectos de los fármacos , Animales , Encéfalo/metabolismo , Trastornos del Conocimiento/inducido químicamente , Ciclopropanos/administración & dosificación , Ciclopropanos/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Fenciclidina/administración & dosificación , Alcohol Feniletílico/administración & dosificación , Alcohol Feniletílico/química , Alcohol Feniletílico/farmacología , Ratas , Ratas Endogámicas , Relación Estructura-Actividad
4.
Bioorg Med Chem Lett ; 22(15): 5134-40, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22677319

RESUMEN

We describe the discovery of a series of compounds based on 1-{3-[4-(2-oxo-2,3-dihydro-benzoimidazol-1-yl)-piperidin-1-yl]-propyl}-3,4-dihydro-1H-quinolin-2-one (3), showing combined D(2) receptor affinity and M(1) receptor agonism. Based on a strategy of controlling logP, we herein describe a hit-to-lead investigation with the aim of retaining the combined D(2)/M(1) profile, while removing the propensity of the compounds to inhibit the hERG channel, as well as at obtaining acceptable pharmacokinetic properties. Although a SAR was evident for all four parameters in question, it was not possible to separate hERG channel inhibition and D(2) receptor affinity by this effort; whilst it was feasible to obtain compounds with M(1) receptor agonism, acceptable clearance, and weak hERG inhibition.


Asunto(s)
Antipsicóticos/química , Ligandos , Quinolonas/química , Receptor Muscarínico M1/agonistas , Receptores de Dopamina D2/agonistas , Antipsicóticos/síntesis química , Antipsicóticos/farmacocinética , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Semivida , Humanos , Microsomas Hepáticos/metabolismo , Quinolonas/síntesis química , Quinolonas/farmacocinética , Receptor Muscarínico M1/metabolismo , Receptores de Dopamina D2/metabolismo , Relación Estructura-Actividad
5.
Anal Biochem ; 418(1): 66-72, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21782781

RESUMEN

To develop a real-time thallium flux assay for high-throughput screening (HTS) of human KCNQ4 (Kv7.4) potassium channel openers, we used CHO-K1 cells stably expressing human KCNQ4 channel protein and a thallium-sensitive dye based on the permeability of thallium through potassium channels. The electrophysiological and pharmacological properties of the cell line expressing the KCNQ4 protein were found to be in agreement with that reported elsewhere. The EC(50) values of the positive control compound (retigabine) determined by the thallium and (86)rubidium flux assays were comparable to and consistent with those documented in the literature. Signal-to-background (S/B) ratio and Z factor of the thallium influx assay system were assessed to be 8.82 and 0.63, respectively. In a large-scale screening of 98,960 synthetic and natural compounds using the thallium influx assay, 76 compounds displayed consistent KCNQ4 activation, and of these 6 compounds demonstrated EC(50) values of less than 20 µmol/L and 2 demonstrated EC(50) values of less than 1 µmol/L. Taken together, the fluorescence-based thallium flux assay is a highly efficient, automatable, and robust tool to screen potential KCNQ4 openers. This approach may also be expanded to identify and evaluate potential modulators of other potassium channels.


Asunto(s)
Canales de Potasio KCNQ/química , Talio/química , Animales , Células CHO , Cricetinae , Evaluación Preclínica de Medicamentos/métodos , Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Humanos , Activación del Canal Iónico , Canales de Potasio KCNQ/metabolismo , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Talio/metabolismo
6.
Eur J Pharmacol ; 887: 173440, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32745603

RESUMEN

Activation of the voltage-gated Kv7 channels holds therapeutic promise in several neurological and psychiatric disorders, including epilepsy, schizophrenia, and depression. Here, we present a pharmacological characterization of Lu AA41178, a novel, pan-selective Kv7.2-7.5 opener, using both in vitro assays and a broad range of in vivo assays with relevance to epilepsy, schizophrenia, and depression. Electrophysiological characterization in Xenopus oocytes expressing human Kv7.2-Kv7.5 confirmed Lu AA41178 as a pan-selective opener of Kv7 channels by significantly left-shifting the activation threshold. Additionally, Lu AA41178 was tested in vitro for off-target effects, demonstrating a clean Kv7-selective profile, with no impact on common cardiac ion channels, and no potentiating activity on GABAA channels. Lu AA41178 was evaluated across preclinical in vivo assays with relevance to neurological and psychiatric disorders. In the maximum electroshock seizure threshold test and PTZ seizure threshold test, Lu AA41178 significantly increased the seizure thresholds in mice, demonstrating anticonvulsant efficacy. Lu AA41178 demonstrated antipsychotic-like activity by reducing amphetamine-induced hyperlocomotion in mice as well as lowering conditioned avoidance responses in rats. In the mouse forced swim test, a model with antidepressant predictivity, Lu AA41178 significantly reduced immobility. Additionally, behavioral effects typically observed with Kv7 openers was also characterized. In vivo assays were accompanied by plasma and brain exposures, revealing minimum effective plasma levels <1000 ng/ml. Lu AA41178, a potent opener of neuronal Kv7 channels demonstrate efficacy in assays of epilepsy, schizophrenia and depression and might serve as a valuable tool for exploring the role of Kv7 channels in both neurological and psychiatric disorders.


Asunto(s)
Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Canal de Potasio KCNQ2/agonistas , Trastornos Mentales/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Animales , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Canal de Potasio KCNQ2/metabolismo , Masculino , Trastornos Mentales/metabolismo , Trastornos Mentales/psicología , Ratones , Ratones Endogámicos C57BL , Psicotrópicos/farmacología , Psicotrópicos/uso terapéutico , Ratas , Ratas Wistar , Convulsiones/metabolismo , Convulsiones/psicología , Resultado del Tratamiento , Xenopus laevis
7.
Neurosci Lett ; 662: 29-35, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-28987817

RESUMEN

Mental disorders such as schizophrenia are associated with impaired firing properties of fast spiking inhibitory interneurons (FSINs) causing reduced task-evoked gamma-oscillation in prefrontal cortex. The voltage-gated sodium channel NaV1.1 is highly expressed in PV-positive interneurons, but only at low levels in principal cells. Positive modulators of Nav1.1 channels are for this reason considered potential candidates for the treatment of cognitive disorders. Here we examined the effect of the novel positive modulator of voltage-gated sodium channels Lu AE98134. We found that Lu AE98134 facilitated the sodium current mediated by NaV1.1 expressed in HEK cells by shifting its activation to more negative values, decreasing its inactivation kinetics and promoting a persistent inward current. In a slice preparation from the brain of adult mice, Lu AE98134 promoted the excitability of fast spiking interneurons by decreasing the threshold for action potentials. We then tested if Lu AE98134 could normalize the altered firing properties of FSINs in Dlx5/6+/- mutant mice. FSINs of this model for schizophrenia are characterized by broader action potentials and higher spike threshold. We found that in the presence of Lu AE98134, the firing frequency was increased while the spike duration and the threshold were decreased. Compounds with similar mode of action appear as promising candidates for restoring cognitive deficits present in schizophrenia.


Asunto(s)
Compuestos Heterocíclicos con 3 Anillos/farmacología , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Sulfonamidas/farmacología , Animales , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Ratones Mutantes
8.
Brain Res ; 1689: 1-11, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29274875

RESUMEN

The multimodal antidepressant vortioxetine is thought to mediate its pharmacological effects via 5-HT1A receptor agonism, 5-HT1B receptor partial agonism, 5-HT1D, 5-HT3, 5-HT7 receptor antagonism and 5-HT transporter inhibition. Here we studied vortioxetine's functional effects across species (canine, mouse, rat, guinea pig and human) in cellular assays with heterologous expression of 5-HT3A receptors (in Xenopus oocytes and HEK-293 cells) and in mouse neuroblastoma N1E-115 cells with endogenous expression of 5-HT3A receptors. Furthermore, we studied the effects of vortioxetine on activity of CA1 Stratum Radiatum interneurons in rat hippocampus slices using current- and voltage-clamping methods. The patched neurons were subsequently filled with biocytin for confirmation of 5-HT3 receptor mRNA expression by in situ hybridization. Whereas, both vortioxetine and the 5-HT3 receptor antagonist ondansetron potently antagonized 5-HT-induced currents in the cellular assays, vortioxetine had a slower off-rate than ondansetron in oocytes expressing 5-HT3A receptors. Furthermore, vortioxetine's but not ondansetron's 5-HT3 receptor antagonistic potency varied considerably across species. Vortioxetine had the highest potency at rat and the lowest potency at guinea pig 5-HT3A receptors. Finally, in 5-HT3 receptor-expressing GABAergic interneurons from the CA1 stratum radiatum, vortioxetine and ondansetron blocked depolarizations induced by superfusion of either 5-HT or the 5-HT3 receptor agonist mCPBG. Taken together, these data add to a growing literature supporting the idea that vortioxetine may inhibit GABAergic neurotransmission in some brain regions via a 5-HT3 receptor antagonism-dependent mechanism and thereby disinhibit pyramidal neurons and enhance glutamatergic signaling.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Antidepresivos/farmacología , Interneuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Vortioxetina/farmacología , Potenciales de Acción/fisiología , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/metabolismo , Perros , Ácido Glutámico/metabolismo , Cobayas , Células HEK293 , Humanos , Interneuronas/metabolismo , Ratones , Ondansetrón/farmacología , Oocitos , Células Piramidales/metabolismo , Ratas , Receptores de Serotonina 5-HT3/metabolismo , Serotonina/metabolismo , Técnicas de Cultivo de Tejidos , Xenopus laevis , Ácido gamma-Aminobutírico/metabolismo
9.
Eur J Pharmacol ; 538(1-3): 23-31, 2006 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-16647053

RESUMEN

In order to establish if the canine 5-hydroxytryptamine type 3A (5-HT(3A)) receptors share the pharmacological profile with human 5-HT(3A) receptors, we cloned and performed a molecular pharmacological characterization of the canine 5-HT(3A) receptor. The 5-HT(3A) cDNA was cloned from canine brain by polymerase chain reaction amplification. It encodes a 483 amino acid peptide that exhibits from 80% (mice) to 90% (ferrets) identity to other sequenced mammalian 5-HT(3A) receptors. The receptor agonists 5-hydroxytryptamine (5-HT) and meta-chlorophenylbiguanide (mCPBG) showed little differences between the two species, whereas 2-methyl-5-hydroxytryptamine (2-Me-5-HT) was ten times weaker at canine receptors than at human receptors. The potencies at the canine 5-HT(3) receptors were 9.9 microM (5-HT), 79 microM (2-Me-5-HT) and 0.8 microM (mCPBG). The selective, competitive receptor antagonist ondansetron was ten times more potent at human receptors compared to canine receptors (K(b)=0.9 nM), while (+)-tubocurarine was 1000-fold more potent at canine receptors (K(b)=3.0 nM) than at human receptors. Examination of the presumed ligand binding extracellular domain revealed one residue, where the canine receptor differs from all previously characterized 5-HT(3A) receptors, i.e. other species contain a conserved Trp(195), whereas the canine orthologue contains a Leu(195). To address the differences in potencies at the human and canine 5-HT(3A) receptors seen in this study, we introduced a L195W point mutation in the canine orthologue. Data showed that the 195 residue can affect receptor agonist potency and efficacy as well as antagonist potency, but did produce a pharmacological profile identical to the human orthologue. We therefore conclude that position 195 is strongly involved in the receptor-ligand interaction, but additional residues must contribute to the overall pharmacological profile.


Asunto(s)
Perros/genética , Receptores de Serotonina 5-HT3/genética , Secuencia de Aminoácidos , Animales , Biguanidas/farmacología , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Relación Dosis-Respuesta a Droga , Femenino , Potenciales de la Membrana/efectos de los fármacos , Datos de Secuencia Molecular , Mutación , Ondansetrón/farmacología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Receptores de Serotonina 5-HT3/fisiología , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Serotonina/farmacología , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Xenopus
10.
Biochem Pharmacol ; 110-111: 92-108, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27086281

RESUMEN

We have previously identified a novel class of 5-hydroxytryptamine type 3 receptor (5-HT3R) agonists sharing little structural similarity with orthosteric 5-HT3R ligands (Jørgensen et al., 2011). In the present study we have elucidated the functional characteristics and the mechanism of action of one of these compounds, trans-3-(4-methoxyphenyl)-N-(pentan-3-yl)acrylamide (TMPPAA). In electrophysiological recordings TMPPAA was found to be a highly-efficacious partial agonist equipotent with 5-HT at the 5-HT3A receptor (5-HT3AR) expressed in COS-7 cells and somewhat less potent at the receptor expressed in Xenopus oocytes. The desensitization kinetics of TMPPAA-evoked currents were very different from those mediated by 5-HT. Moreover, repeated TMPPAA applications resulted in progressive current run-down and persistent non-responsiveness of the receptor to TMPPAA, but not to 5-HT. In addition to its direct activation, TMPPAA potentiated 5-HT-mediated 5-HT3AR signalling, and the allosteric link between the two binding sites was corroborated by the analogous ability of 5-HT to potentiate TMPPAA-evoked responses. The agonism and potentiation exerted by TMPPAA at a chimeric α7-nACh/5-HT3A receptor suggested that the ligand acts through the transmembrane domain of 5-HT3AR, a notion further substantiated by its functional properties at chimeric and mutant human/murine 5-HT3ARs. A residue in the transmembrane helix 4 of 5-HT3A was identified as an important molecular determinant for the different agonist potencies exhibited by TMPPAA at human and murine 5-HT3ARs. In conclusion, TMPPAA is a novel allosteric agonist and positive allosteric modulator of 5-HT3Rs, and its aberrant signalling characteristics compared to 5-HT at the 5-HT3AR underline the potential in Cys-loop receptor modulation and activation through allosteric sites.


Asunto(s)
Acrilamidas/farmacología , Proteínas Mutantes Quiméricas/agonistas , Éteres Fenílicos/farmacología , Receptores de Serotonina 5-HT3/genética , Agonistas del Receptor de Serotonina 5-HT3/farmacología , Acrilamidas/síntesis química , Regulación Alostérica , Sitio Alostérico , Animales , Células COS , Chlorocebus aethiops , Potenciales Evocados/efectos de los fármacos , Potenciales Evocados/fisiología , Expresión Génica , Cinética , Ratones , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/metabolismo , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Técnicas de Placa-Clamp , Éteres Fenílicos/síntesis química , Unión Proteica , Estructura Secundaria de Proteína , Receptores de Serotonina 5-HT3/metabolismo , Serotonina/farmacología , Agonistas del Receptor de Serotonina 5-HT3/síntesis química , Xenopus laevis
11.
Eur J Pharmacol ; 748: 10-7, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25530269

RESUMEN

Torsades de Pointes (TdP) is a potentially lethal cardiac arrhythmia and a known adverse effect of many drugs secondary to block of the rapidly activating delayed rectifier potassium current (IKr). In animal models antipsychotic drugs have shown reduced pro-arrhythmic potential compared to drugs with comparable IKr-blocking characteristics. The reduced pro-arrhythmic properties of antipsychotic drugs has been attributed to a variety of different causes e.g., effects on α1-adrenergic receptors, ß-adrenergic receptors, muscarinic receptors or cardiac ion channels like Ca(2+)- and Na(+)-channels. Since only limited experimental information exists about the effects of α1-adrenergic receptor activity of antipsychotic drugs in pro-arrhythmic models, we have decided to investigate this. In this study we show that four antipsychotic drugs all have high affinity for α1-adrenergic receptor (sertindole>risperidone>haloperidol>olanzapine) and all block IKr (sertindole>haloperidol>risperidone>olanzapine). In canine Purkinje fibres, α1-adrenergic stimulation prolonged action potential duration; however, the stimulation does not cause afterdepolarizations, even in the presence of dofetilide-induced delayed repolarization. We showed for the first time in an in vivo pro-arrhythmic rabbit model that several antipsychotic drugs in accordance with their known α1-adrenergic receptor blocking properties reduced the incidence of drug-induced TdP and that the overall ability of the antipsychotic drugs to prevent TdP was associated with prevention of methoxamine induced increase in blood pressure. Further investigations are required to clarify the relative importance of α1-adrenergic receptor antagonism in conjunction with the additional effects of antipsychotic drugs on various receptors and ion channels.


Asunto(s)
Antiarrítmicos/farmacología , Antipsicóticos/farmacología , Receptores Adrenérgicos alfa 1/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Presión Sanguínea/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Perros , Femenino , Masculino , Modelos Animales , Potasio/metabolismo , Ramos Subendocárdicos/efectos de los fármacos , Ramos Subendocárdicos/fisiología , Conejos
12.
ACS Chem Neurosci ; 6(8): 1302-8, 2015 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-26114759

RESUMEN

Voltage-gated sodium channels (Nav) are crucial to the initiation and propagation of action potentials (APs) in electrically excitable cells, and during the past decades they have received considerable attention due to their therapeutic potential. Here, we report for the first time the synthesis and the electrophysiological evaluation of 16 ligands based on a 2-methylbenzamide scaffold that have been identified as Nav1.1 modulators. Among these compounds, N,N'-(1,3-phenylene)bis(2-methylbenzamide) (3a) has been selected and evaluated in ex-vivo experiments in order to estimate the activation impact of such a compound profile. It appears that 3a increases the Nav1.1 channel activity although its overall impact remains moderate. Altogether, our preliminary results provide new insights into the development of small molecule activators targeting specifically Nav1.1 channels to design potential drugs for treating CNS diseases.


Asunto(s)
Benzamidas/química , Moduladores del Transporte de Membrana/farmacología , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Animales , Región CA1 Hipocampal/efectos de los fármacos , Región CA1 Hipocampal/fisiología , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Interneuronas/efectos de los fármacos , Interneuronas/fisiología , Potenciales de la Membrana/efectos de los fármacos , Moduladores del Transporte de Membrana/síntesis química , Moduladores del Transporte de Membrana/química , Estructura Molecular , Ratas , Técnicas de Cultivo de Tejidos
13.
Psychopharmacology (Berl) ; 221(3): 451-68, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22124672

RESUMEN

RATIONALE: A growing body of evidence suggests that negative modulation of γ-aminobutyric acid (GABA) GABA(A) α5 receptors may be a promising strategy for the treatment of certain facets of cognitive impairment; however, selective modulators of GABA(A) α5 receptors have not yet been tested in "schizophrenia-relevant" cognitive assay/model systems in animals. OBJECTIVES: The objectives of this study were to investigate the potential of RO4938581, a negative modulator of GABA(A) α5 receptors, and to attenuate cognitive impairments induced following sub-chronic (sub-PCP) and early postnatal PCP (neo-PCP) administration in the novel object recognition (NOR) and intra-extradimensional shift (ID/ED) paradigms in rats. Complementary in vitro, ex vivo and in vivo studies were performed to confirm negative modulatory activity of RO4938581 and to investigate animal model validity, concept validity and potential side effect issues, respectively. RESULTS: In vitro studies confirmed the reported negative modulatory activity of RO4938581, whilst immunohistochemical analyses revealed significantly reduced parvalbumin-positive cells in the prefrontal cortex of sub-PCP- and neo-PCP-treated rats. RO4938581 (1 mg/kg) ameliorated both sub-PCP- and neo-PCP-induced cognitive deficits in NOR and ID/ED performance, respectively. In contrast, QH-II-066 (1 and 3 mg/kg), a GABA(A) α5 receptor positive modulator, impaired cognitive performance in the NOR task when administered to vehicle-treated animals. Additional studies revealed that both RO4938581 (1 mg/kg) and QH-II-066 (1 and 3 mg/kg) attenuated amphetamine-induced hyperactivity in rats. CONCLUSIONS: Taken together, these novel findings suggest that negative modulation of GABA(A) α5 receptors may represent an attractive treatment option for the cognitive impairments, and potentially positive symptoms, associated with schizophrenia.


Asunto(s)
Benzodiazepinas/farmacología , Trastornos del Conocimiento/tratamiento farmacológico , Imidazoles/farmacología , Fenciclidina/toxicidad , Receptores de GABA-A/efectos de los fármacos , Anfetamina/farmacología , Animales , Células CHO , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos del Conocimiento/inducido químicamente , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Hipercinesia/inducido químicamente , Masculino , Oocitos , Parvalbúminas/metabolismo , Fenciclidina/administración & dosificación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/fisiopatología , Xenopus laevis
14.
J Med Chem ; 54(9): 3206-21, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21486038

RESUMEN

The synthesis and structure-activity relationship of a novel series of compounds with combined effects on 5-HT(3A) and 5-HT(1A) receptors and on the serotonin (5-HT) transporter (SERT) are described. Compound 5m (Lu AA21004) was the lead compound, displaying high affinity for recombinant human 5-HT(1A) (K(i) = 15 nM), 5-HT(1B) (K(i) = 33 nM), 5-HT(3A) (K(i) = 3.7 nM), 5-HT(7) (K(i) = 19 nM), and noradrenergic ß(1) (K(i) = 46 nM) receptors, and SERT (K(i) = 1.6 nM). Compound 5m displayed antagonistic properties at 5-HT(3A) and 5-HT(7) receptors, partial agonist properties at 5-HT(1B) receptors, agonistic properties at 5-HT(1A) receptors, and potent inhibition of SERT. In conscious rats, 5m significantly increased extracellular 5-HT levels in the brain after acute and 3 days of treatment. Following the 3-day treatment (5 or 10 (mg/kg)/day) SERT occupancies were only 43% and 57%, respectively. These characteristics indicate that 5m is a novel multimodal serotonergic compound, and 5m is currently in clinical development for major depressive disorder.


Asunto(s)
Antidepresivos/síntesis química , Trastorno Depresivo Mayor/tratamiento farmacológico , Piperazinas/síntesis química , Sulfuros/síntesis química , Animales , Antidepresivos/química , Antidepresivos/farmacología , Línea Celular , Agonismo Parcial de Drogas , Estabilidad de Medicamentos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Oocitos/efectos de los fármacos , Oocitos/fisiología , Piperazinas/química , Piperazinas/farmacología , Ensayo de Unión Radioligante , Ratas , Receptor de Serotonina 5-HT1A/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Receptores de Serotonina/metabolismo , Receptores de Serotonina 5-HT1/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/antagonistas & inhibidores , Serotonina/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/síntesis química , Agonistas del Receptor de Serotonina 5-HT1/química , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Antagonistas del Receptor de Serotonina 5-HT3/síntesis química , Antagonistas del Receptor de Serotonina 5-HT3/química , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/síntesis química , Inhibidores Selectivos de la Recaptación de Serotonina/química , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Relación Estructura-Actividad , Sulfuros/química , Sulfuros/farmacología , Vortioxetina , Xenopus
15.
J Pharmacol Exp Ther ; 307(2): 776-84, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12966159

RESUMEN

Inhibition of the potassium current IKr and QT prolongation are associated with drug-induced torsades de pointes arrhythmias (TdP) and sudden cardiac death. We investigated the cardiac electrophysiological effects of sertindole, an antipsychotic drug reported to prolong the QT interval in schizophrenic patients. In cell cultures, sertindole seemed to be a selective blocker of IHERG over other ion currents. For IHERG, the IC50 value was 64 +/- 7 nM, whereas ISCN5A, ICa,L, ICa,T, IKCNQ1/KCNE1, and IKv4.3 were blocked in the micromolar range. In canine ventricular myocytes, the IC50 value for IKr inhibition by sertindole was 107 +/- 21 nM. Action potentials in these cells prolonged in a reverse rate- and concentration-dependent manner at 10 to 300 nM sertindole. In vivo, sertindole was administered to anesthetized dogs at clinically relevant (0.05-0.20 mg/kg) and high doses (1.0-2.0 mg/kg) i.v. At 0.05 to 0.20 mg/kg sertindole (plasma concentrations 30-157 nM), QTc was prolonged by 1 to 5% in normal dogs and by 9 to 20% in dogs with remodeled hearts due to chronic atrioventricular block (CAVB). TdP was not induced at these doses in normal dogs or in CAVB dogs with reproducible induction of TdP by dofetilide in previous experiments. At 1.0 to 2.0 mg/kg sertindole (plasma concentrations 0.5-3.1 microM), QTc prolonged by 6 to 11% in normal dogs and by 22% in dofetilide-sensitive CAVB dogs. TdP occurred in three of five animals in the latter group. Thus, at high i.v. doses sertindole can pose a serious proarrhythmic risk when electrical remodeling of the ventricles is present. At clinically relevant doses, however, sertindole does not cause TdP in anesthetized dogs with normal or remodeled hearts.


Asunto(s)
Antipsicóticos/farmacología , Corazón/efectos de los fármacos , Imidazoles/farmacología , Indoles/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Antipsicóticos/efectos adversos , Arritmias Cardíacas/inducido químicamente , Perros , Electrofisiología , Corazón/fisiología , Imidazoles/efectos adversos , Indoles/efectos adversos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Fenetilaminas/farmacología , Sulfonamidas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA