Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Pharmacol Exp Ther ; 372(3): 339-353, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31818916

RESUMEN

The serine hydrolase monoacylglycerol lipase (MAGL) is the rate-limiting enzyme responsible for the degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG) into arachidonic acid and glycerol. Inhibition of 2-AG degradation leads to elevation of 2-AG, the most abundant endogenous agonist of the cannabinoid receptors (CBs) CB1 and CB2. Activation of these receptors has demonstrated beneficial effects on mood, appetite, pain, and inflammation. Therefore, MAGL inhibitors have the potential to produce therapeutic effects in a vast array of complex human diseases. The present report describes the pharmacologic characterization of [1-(4-fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone (JNJ-42226314), a reversible and highly selective MAGL inhibitor. JNJ-42226314 inhibits MAGL in a competitive mode with respect to the 2-AG substrate. In rodent brain, the compound time- and dose-dependently bound to MAGL, indirectly led to CB1 occupancy by raising 2-AG levels, and raised norepinephrine levels in cortex. In vivo, the compound exhibited antinociceptive efficacy in both the rat complete Freund's adjuvant-induced radiant heat hypersensitivity and chronic constriction injury-induced cold hypersensitivity models of inflammatory and neuropathic pain, respectively. Though 30 mg/kg induced hippocampal synaptic depression, altered sleep onset, and decreased electroencephalogram gamma power, 3 mg/kg still provided approximately 80% enzyme occupancy, significantly increased 2-AG and norepinephrine levels, and produced neuropathic antinociception without synaptic depression or decreased gamma power. Thus, it is anticipated that the profile exhibited by this compound will allow for precise modulation of 2-AG levels in vivo, supporting potential therapeutic application in several central nervous system disorders. SIGNIFICANCE STATEMENT: Potentiation of endocannabinoid signaling activity via inhibition of the serine hydrolase monoacylglycerol lipase (MAGL) is an appealing strategy in the development of treatments for several disorders, including ones related to mood, pain, and inflammation. [1-(4-Fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone is presented in this report to be a novel, potent, selective, and reversible noncovalent MAGL inhibitor that demonstrates dose-dependent enhancement of the major endocannabinoid 2-arachidonoylglycerol as well as efficacy in models of neuropathic and inflammatory pain.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Monoacilglicerol Lipasas/antagonistas & inhibidores , Piperazinas/farmacología , Animales , Unión Competitiva , Encéfalo/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/sangre , Escherichia coli/enzimología , Escherichia coli/genética , Células HeLa , Humanos , Cinética , Leucocitos Mononucleares/enzimología , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Monoacilglicerol Lipasas/genética , Dolor/tratamiento farmacológico , Piperazinas/sangre , Unión Proteica , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB2/agonistas , Sueño REM/efectos de los fármacos , Especificidad por Sustrato
2.
J Pharmacol Exp Ther ; 357(2): 394-414, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26989142

RESUMEN

Members of the α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid (AMPA) subtype of ionotropic glutamate receptors mediate the majority of fast synaptic transmission within the mammalian brain and spinal cord, representing attractive targets for therapeutic intervention. Here, we describe novel AMPA receptor modulators that require the presence of the accessory protein CACNG8, also known as transmembrane AMPA receptor regulatory protein γ8 (TARP-γ8). Using calcium flux, radioligand binding, and electrophysiological assays of wild-type and mutant forms of TARP-γ8, we demonstrate that these compounds possess a novel mechanism of action consistent with a partial disruption of the interaction between the TARP and the pore-forming subunit of the channel. One of the molecules, 5-[2-chloro-6-(trifluoromethoxy)phenyl]-1,3-dihydrobenzimidazol-2-one (JNJ-55511118), had excellent pharmacokinetic properties and achieved high receptor occupancy following oral administration. This molecule showed strong, dose-dependent inhibition of neurotransmission within the hippocampus, and a strong anticonvulsant effect. At high levels of receptor occupancy in rodent in vivo models, JNJ-55511118 showed a strong reduction in certain bands on electroencephalogram, transient hyperlocomotion, no motor impairment on rotarod, and a mild impairment in learning and memory. JNJ-55511118 is a novel tool for reversible AMPA receptor inhibition, particularly within the hippocampus, with potential therapeutic utility as an anticonvulsant or neuroprotectant. The existence of a molecule with this mechanism of action demonstrates the possibility of pharmacological targeting of accessory proteins, increasing the potential number of druggable targets.


Asunto(s)
Bencimidazoles/uso terapéutico , Canales de Calcio/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Receptores AMPA/efectos de los fármacos , Animales , Canales de Calcio/genética , Señalización del Calcio/efectos de los fármacos , Diseño de Fármacos , Electroencefalografía/efectos de los fármacos , Células HEK293 , Humanos , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Mutación/genética , Neuronas/efectos de los fármacos , Equilibrio Postural/efectos de los fármacos , Ratas Sprague-Dawley , Receptores AMPA/genética
3.
Mol Pharmacol ; 88(5): 911-25, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26349500

RESUMEN

GPR139 is an orphan G-protein-coupled receptor expressed in the central nervous system. To identify its physiologic ligand, we measured GPR139 receptor activity from recombinant cells after treatment with amino acids, orphan ligands, serum, and tissue extracts. GPR139 activity was measured using guanosine 5'-O-(3-[(35)S]thio)-triphosphate binding, calcium mobilization, and extracellular signal-regulated kinases phosphorylation assays. Amino acids L-tryptophan (L-Trp) and L-phenylalanine (L-Phe) activated GPR139, with EC50 values in the 30- to 300-µM range, consistent with the physiologic concentrations of L-Trp and L-Phe in tissues. Chromatography of rat brain, rat serum, and human serum extracts revealed two peaks of GPR139 activity, which corresponded to the elution peaks of L-Trp and L-Phe. With the purpose of identifying novel tools to study GPR139 function, a high-throughput screening campaign led to the identification of a selective small-molecule agonist [JNJ-63533054, (S)-3-chloro-N-(2-oxo-2-((1-phenylethyl)amino)ethyl) benzamide]. The tritium-labeled JNJ-63533054 bound to cell membranes expressing GPR139 and could be specifically displaced by L-Trp and L-Phe. Sequence alignment revealed that GPR139 is highly conserved across species, and RNA sequencing studies of rat and human tissues indicated its exclusive expression in the brain and pituitary gland. Immunohistochemical analysis showed specific expression of the receptor in circumventricular regions of the habenula and septum in mice. Together, these findings suggest that L-Trp and L-Phe are candidate physiologic ligands for GPR139, and we hypothesize that this receptor may act as a sensor to detect dynamic changes of L-Trp and L-Phe in the brain.


Asunto(s)
Habénula/química , Proteínas del Tejido Nervioso/fisiología , Fenilalanina/fisiología , Receptores Acoplados a Proteínas G/fisiología , Tabique del Cerebro/química , Triptófano/fisiología , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/efectos de los fármacos , Fenilalanina/sangre , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/análisis , Receptores Acoplados a Proteínas G/efectos de los fármacos , Triptófano/sangre
4.
J Pharmacol Exp Ther ; 352(3): 590-601, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25583879

RESUMEN

Orexins (OXs) are peptides produced by perifornical (PeF) and lateral hypothalamic neurons that exert a prominent role in arousal-related processes, including stress. A critical role for the orexin-1 receptor (OX1R) in complex emotional behavior is emerging, such as overactivation of the OX1R pathway being associated with panic or anxiety states. Here we characterize a brain-penetrant, selective, and high-affinity OX1R antagonist, compound 56 [N-({3-[(3-ethoxy-6-methylpyridin-2-yl)carbonyl]-3-azabicyclo[4.1.0]hept-4-yl}methyl)-5-(trifluoromethyl)pyrimidin-2-amine]. Ex vivo receptor binding studies demonstrated that, after subcutaneous administration, compound 56 crossed the blood-brain barrier and occupied OX1Rs in the rat brain at lower doses than standard OX1R antagonists GSK-1059865 [5-bromo-N-({1-[(3-fluoro-2-methoxyphenyl)carbonyl]-5-methylpiperidin-2-yl}methyl)pyridin-2-amine], SB-334867 [1-(2-methyl-1,3-benzoxazol-6-yl)-3-(1,5-naphthyridin-4-yl)urea], and SB-408124 [1-(6,8-difluoro-2-methylquinolin-4-yl)-3-[4-(dimethylamino)phenyl]urea]. Although compound 56 did not alter spontaneous sleep in rats and in wild-type mice, its administration in orexin-2 receptor knockout mice selectively promoted rapid eye movement sleep, demonstrating target engagement and specific OX1R blockade. In a rat model of psychological stress induced by cage exchange, the OX1R antagonist prevented the prolongation of sleep onset without affecting sleep duration. In a rat model of panic vulnerability (involving disinhibition of the PeF OX region) to threatening internal state changes (i.e., intravenous sodium lactate infusion), compound 56 attenuated sodium lactate-induced panic-like behaviors and cardiovascular responses without altering baseline locomotor or autonomic activity. In conclusion, OX1R antagonism represents a novel therapeutic strategy for the treatment of various psychiatric disorders associated with stress or hyperarousal states.


Asunto(s)
Aminopiridinas/uso terapéutico , Nivel de Alerta/fisiología , Antagonistas de los Receptores de Orexina , Receptores de Orexina/metabolismo , Piperidinas/uso terapéutico , Estrés Psicológico/metabolismo , Estrés Psicológico/prevención & control , Aminopiridinas/metabolismo , Aminopiridinas/farmacología , Animales , Nivel de Alerta/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Hipnóticos y Sedantes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piperidinas/metabolismo , Piperidinas/farmacología , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley
5.
J Pharmacol Exp Ther ; 354(3): 471-82, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26177655

RESUMEN

Dual orexin receptor antagonists have been shown to promote sleep in various species, including humans. Emerging research indicates that selective orexin-2 receptor (OX2R) antagonists may offer specificity and a more adequate sleep profile by preserving normal sleep architecture. Here, we characterized JNJ-42847922 ([5-(4,6-dimethyl-pyrimidin-2-yl)-hexahydro-pyrrolo[3,4-c]pyrrol-2-yl]-(2-fluoro-6-[1,2,3]triazol-2-yl-phenyl)-methanone), a high-affinity/potent OX2R antagonist. JNJ-42847922 had an approximate 2-log selectivity ratio versus the human orexin-1 receptor. Ex vivo receptor binding studies demonstrated that JNJ-42847922 quickly occupied OX2R binding sites in the rat brain after oral administration and rapidly cleared from the brain. In rats, single oral administration of JNJ-42847922 (3-30 mg/kg) during the light phase dose dependently reduced the latency to non-rapid eye movement (NREM) sleep and prolonged NREM sleep time in the first 2 hours, whereas REM sleep was minimally affected. The reduced sleep onset and increased sleep duration were maintained upon 7-day repeated dosing (30 mg/kg) with JNJ-42847922, then all sleep parameters returned to baseline levels following discontinuation. Although the compound promoted sleep in wild-type mice, it had no effect in OX2R knockout mice, consistent with a specific OX2R-mediated sleep response. JNJ-42847922 did not increase dopamine release in rat nucleus accumbens or produce place preference in mice after subchronic conditioning, indicating that the compound lacks intrinsic motivational properties in contrast to zolpidem. In a single ascending dose study conducted in healthy subjects, JNJ-42847922 increased somnolence and displayed a favorable pharmacokinetic and safety profile for a sedative/hypnotic, thus emerging as a promising candidate for further clinical development for the treatment of insomnia.


Asunto(s)
Antagonistas de los Receptores de Orexina , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Sueño/efectos de los fármacos , Animales , Sitios de Unión/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células CHO , Línea Celular , Cricetulus , Dopamina/metabolismo , Células HEK293 , Humanos , Hipnóticos y Sedantes/farmacología , Masculino , Ratones , Ratones Noqueados , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Trastornos del Inicio y del Mantenimiento del Sueño/metabolismo , Fases del Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Zolpidem
6.
J Pharmacol Exp Ther ; 342(2): 429-40, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22570363

RESUMEN

In rodents 5-hydroxytryptamine type 7 (5-HT(7)) receptor blockade has been shown to be effective in models of depression and to increase the latency to rapid eye movement (REM) sleep and decrease REM duration. In the clinic, the REM sleep reduction observed with many antidepressants may serve as a biomarker. We report here the preclinical and clinical evaluation of a 5-HT(7) receptor antagonist, (3-(4-chlorophenyl)-1,4,5,6,7,8-hexahydro-1-(phenylmethyl)pyrazolo[3,4-d]azepine 2-hydroxy-1,2,3-propanetricarboxylate) (JNJ-18038683). In rodents, JNJ-18038683 increased the latency to REM sleep and decreased REM duration, and this effect was maintained after repeated administration for 7 days. The compound was effective in the mouse tail suspension test. JNJ-18038683 enhanced serotonin transmission, antidepressant-like behavior, and REM sleep suppression induced by citalopram in rodents. In healthy human volunteers JNJ-18038683 prolonged REM latency and reduced REM sleep duration, demonstrating that the effect of 5-HT(7) blockade on REM sleep translated from rodents to humans. Like in rats, JNJ-18038683 enhanced REM sleep suppression induced by citalopram in humans, although a drug-drug interaction could not be ruled out. In a double-blind, active, and placebo-controlled clinical trial in 225 patients suffering from major depressive disorder, neither treatment with pharmacologically active doses of JNJ-18038683 or escitalopram separated from placebo, indicating a failed study lacking assay sensitivity. Post hoc analyses using an enrichment window strategy, where all the efficacy data from sites with an implausible high placebo response [placebo group Montgomery-Åsberg Depression Rating Scale (MADRS) < = 12] and from sites with no placebo response (MADRS > = 28) are removed, there was a clinically meaningful difference between JNJ-18038683 and placebo. Further clinical studies are required to characterize the potential antidepressant efficacy of JNJ-18038683.


Asunto(s)
Antidepresivos/farmacología , Azepinas/farmacología , Trastorno Depresivo Mayor/tratamiento farmacológico , Receptores de Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Sueño REM/efectos de los fármacos , Ácidos Tricarboxílicos/farmacología , Adolescente , Adulto , Animales , Antidepresivos/uso terapéutico , Azepinas/uso terapéutico , Línea Celular Transformada , Citalopram/farmacología , Estudios de Cohortes , Estudios Cruzados , Trastorno Depresivo Mayor/metabolismo , Método Doble Ciego , Femenino , Células HEK293 , Suspensión Trasera/métodos , Humanos , Hipotermia/tratamiento farmacológico , Hipotermia/metabolismo , Hipotermia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Antagonistas de la Serotonina/uso terapéutico , Ácidos Tricarboxílicos/uso terapéutico , Adulto Joven
7.
Bioorg Med Chem Lett ; 20(14): 4210-4, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20561786

RESUMEN

The pre-clinical characterization of novel aryloxypyridine amides that are histamine H(3) receptor antagonists is described. These compounds are high affinity histamine H(3) ligands that penetrate the CNS and occupy the histamine H(3) receptor in rat brain. Several compounds were extensively profiled pre-clinically leading to the identification of two compounds suitable for nomination as development candidates.


Asunto(s)
Azepinas/farmacología , Antagonistas de los Receptores Histamínicos H3/farmacología , Piridinas/farmacología , Amidas/química , Animales , Azepinas/química , Evaluación Preclínica de Medicamentos , Piridinas/química , Ratas
8.
Bioorg Med Chem Lett ; 20(9): 2755-60, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20382018

RESUMEN

Pre-clinical characterization of novel substituted pyrrolidines that are high affinity histamine H(3) receptor antagonists is described. These compounds efficiently penetrate the CNS and occupy the histamine H(3) receptor in rat brain following oral administration. One compound, (2S,4R)-1-[2-(4-cyclobutyl-[1,4]diazepane-1-carbonyl)-4-(3-fluoro-phenoxy)-pyrrolidin-1-yl]-ethanone, was extensively profiled and shows promise as a potential clinical candidate.


Asunto(s)
Azepinas/química , Antagonistas de los Receptores Histamínicos H3/química , Pirrolidinas/química , Receptores Histamínicos H3/química , Administración Oral , Animales , Azepinas/síntesis química , Azepinas/farmacocinética , Encéfalo/metabolismo , Perros , Evaluación Preclínica de Medicamentos , Antagonistas de los Receptores Histamínicos H3/síntesis química , Antagonistas de los Receptores Histamínicos H3/farmacocinética , Humanos , Ratones , Pirrolidinas/síntesis química , Pirrolidinas/farmacocinética , Ratas , Receptores Histamínicos H3/metabolismo , Relación Estructura-Actividad
9.
Eur J Pharmacol ; 882: 173256, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32531213

RESUMEN

GPR139 is a G-protein coupled receptor expressed in circumventricular regions of the habenula and septum. Amino acids L-tryptophan and L-phenylalanine have been shown to activate GPR139 at physiologically relevant concentrations. The aim of the present study was to investigate the role of GPR139 on sleep modulation using pharmacological and genetic (GPR139 knockout mice, KO) rodent models. To evaluate the effects of GPR139 pharmacological activation on sleep, rats were orally dosed with the selective GPR139 agonist JNJ-63533054 (3-30 mg/kg). When acutely administered at the beginning of the light phase, the GPR139 agonist dose-dependently reduced non-rapid eye movement (NREM) latency and increased NREM sleep duration without altering rapid eye movement (REM) sleep. This effect progressively dissipated upon 7-day repeated dosing, suggesting functional desensitization. Under baseline conditions, GPR139 KO mice spent less time in REM sleep compared to their wild type littermates during the dark phase, whereas NREM sleep was not altered. Under conditions of pharmacologically enhanced monoamine endogenous tone, GPR139 KO mice showed a blunted response to citalopram or fluoxetine induced REM sleep suppression and an attenuated response to the wake promoting effect of amphetamine. These findings indicate an emerging role of GPR139 in the modulation of sleep states.


Asunto(s)
Proteínas del Tejido Nervioso , Receptores Acoplados a Proteínas G , Sueño , Animales , Citalopram/farmacología , Dextroanfetamina/farmacología , Dopamina/farmacología , Fluoxetina/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Proteínas del Tejido Nervioso/agonistas , Proteínas del Tejido Nervioso/genética , Norepinefrina/farmacología , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Serotonina/farmacología , Sueño/efectos de los fármacos , Sueño/genética
10.
ACS Med Chem Lett ; 11(10): 2002-2009, 2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33062185

RESUMEN

The orexin system consists of two neuropeptides (orexin-A and orexin-B) that exert their mode of action on two receptors (orexin-1 and orexin-2). While the role of the orexin-2 receptor is established as an important modulator of sleep wake states, the role of the orexin-1 receptor is believed to play a role in addiction, panic, or anxiety. In this manuscript, we describe the optimization of a nonselective substituted azabicyclo[2.2.1]heptane dual orexin receptor antagonist (DORA) into orally bioavailable, brain penetrating, selective orexin-1 receptor (OX1R) antagonists. This resulted in the discovery of our first candidate for clinical development, JNJ-54717793.

11.
Transl Psychiatry ; 10(1): 308, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32895369

RESUMEN

Orexin neurons originating in the perifornical and lateral hypothalamic area project to anxiety- and panic-associated neural circuitry, and are highly reactive to anxiogenic stimuli. Preclinical evidence suggests that the orexin system, and particularly the orexin-1 receptor (OX1R), may be involved in the pathophysiology of panic and anxiety. Selective OX1R antagonists thus may constitute a potential new treatment strategy for panic- and anxiety-related disorders. Here, we characterized a novel selective OX1R antagonist, JNJ-61393215, and determined its affinity and potency for human and rat OX1R in vitro. We also evaluated the safety, pharmacokinetic, and pharmacodynamic properties of JNJ-61393215 in first-in-human single- and multiple-ascending dose studies conducted. Finally, the potential anxiolytic effects of JNJ-61393215 were evaluated both in rats and in healthy men using 35% CO2 inhalation challenge to induce panic symptoms. In the rat CO2 model of panic anxiety, JNJ-61393215 demonstrated dose-dependent attenuation of CO2-induced panic-like behavior without altering baseline locomotor or autonomic activity, and had minimal effect on spontaneous sleep. In phase-1 human studies, JNJ-61393215 at 90 mg demonstrated significant reduction (P < 0.02) in CO2-induced fear and anxiety symptoms that were comparable to those obtained using alprazolam. The most frequently reported adverse events were somnolence and headache, and all events were mild in severity. These results support the safety, tolerability, and anxiolytic effects of JNJ-61393215, and validate CO2 exposure as a translational cross-species experimental model to evaluate the therapeutic potential of novel anxiolytic drugs.


Asunto(s)
Antagonistas de los Receptores de Orexina , Pánico , Roedores , Animales , Humanos , Modelos Teóricos , Receptores de Orexina , Ratas
12.
J Pharmacol Exp Ther ; 330(1): 142-51, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19363060

RESUMEN

Orexins are peptides produced by lateral hypothalamic neurons that exert a prominent role in the maintenance of wakefulness by activating orexin-1 (OX1R) and orexin-2 (OX2R) receptor located in wake-active structures. Pharmacological blockade of both receptors by the dual OX1/2R antagonist (2R)-2-[(1S)-6,7-dimethoxy-1-{2-[4-(trifluoromethyl)phenyl]ethyl}-3,4-dihydroisoquinolin-2(1H)-yl]-N-methyl-2-phenylethanamide (almorexant) has been shown to promote sleep in animals and humans during their active period. However, the selective distribution of OX1R and OX2R in distinct neuronal circuits may result in a differential impact of these receptors in sleep-wake modulation. The respective role of OX1R and OX2R on sleep in correlation with monoamine release was evaluated in rats treated with selective antagonists alone or in combination. When administered in either phase of the light/dark cycle, the OX2R antagonist 1-(2,4-dibromophenyl)-3-[(4S,5S)-2,2-dimethyl-4-phenyl-1,3-dioxan-5-yl]urea (JNJ-10397049) decreased the latency for persistent sleep and increased nonrapid eye movement and rapid eye movement sleep time. Almorexant produced less hypnotic activity, whereas the OX1R antagonist 1-(6,8-difluoro-2-methylquinolin-4-yl)-3-[4-(dimethylamino)phenyl]urea (SB-408124) had no effect. Microdialysis studies showed that either OX2R or OX1/2R antagonism decreased extracellular histamine concentration in the lateral hypothalamus, whereas both OX1R and OX1/2R antagonists increased dopamine release in the prefrontal cortex. Finally, coadministration of the OX1R with the OX2R antagonist greatly attenuated the sleep-promoting effects of the OX2R antagonist. These results indicate that blockade of OX2R is sufficient to initiate and prolong sleep, consistent with the hypothesis of a deactivation of the histaminergic system. In addition, it is suggested that simultaneous inhibition of OX1R attenuates the sleep-promoting effects mediated by selective OX2R blockade, possibly correlated with dopaminergic neurotransmission.


Asunto(s)
Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/fisiología , Receptores de Neuropéptido/antagonistas & inhibidores , Receptores de Neuropéptido/fisiología , Sueño/efectos de los fármacos , Sueño/fisiología , Acetamidas/farmacología , Animales , Isoquinolinas/farmacología , Masculino , Receptores de Orexina , Ratas , Ratas Sprague-Dawley
13.
Neuropharmacology ; 56(2): 448-54, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18948124

RESUMEN

5-HT(7) receptors are involved in REM sleep and possibly in mood disorders. REM sleep suppression and antidepressant-like behavior is observed in 5-HT(7)(-/-) mice and in rats treated with 5-HT(7) receptor antagonists. We recently demonstrated that pharmacological blockade of 5-HT(7) receptors enhances REM sleep suppression and antidepressant-like behavior induced by citalopram in rodents. It has been hypothesized that the effect of citalopram on sleep is essentially mediated by the activation of 5-HT(1A) receptors. The present study investigates the impact of 5-HT(7) receptor gene deletion on the effect of various reuptake inhibitors on REM sleep and probes the role of 5-HT(1A) receptors in this response. Three SSRIs (citalopram, fluoxetine and paroxetine) but not the tricyclic antidepressant desipramine had a significantly stronger REM sleep suppressive effect in 5-HT(7)(-/-) mice compared to 5-HT(7)(+/+) mice. In contrast, REM sleep was similarly reduced in 5-HT(7)(+/+) mice and 5-HT(7)(-/-) mice after treatment with the 5-HT(1A) receptor agonist ipsapirone. Furthermore, both 5-HT(7)(+/+) and 5-HT(7)(-/-) mice displayed the same increase in REM sleep duration produced by the 5-HT(1A) receptor antagonist WAY-100635. These findings indicate that 5-HT(7) receptor deletion augments the effect of various SSRIs on REM sleep suppression and that this effect is distinct from those mediated via 5-HT(1A) receptors.


Asunto(s)
Receptor de Serotonina 5-HT1A/fisiología , Receptores de Serotonina/deficiencia , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Sueño REM/efectos de los fármacos , Análisis de Varianza , Animales , Electroencefalografía/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pirimidinas/farmacología , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Receptor de Serotonina 5-HT1A/deficiencia , Receptores de Serotonina/genética , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Sueño REM/genética , Factores de Tiempo
14.
Eur J Pharmacol ; 576(1-3): 43-54, 2007 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-17765221

RESUMEN

Wake-promoting agents such as modafinil are used in the clinic as adjuncts to antidepressant therapy in order to alleviate lethargy. The wake-promoting action of histamine H(3) receptor antagonists has been evidenced in numerous animal studies. They may therefore be a viable strategy for use as an antidepressant therapy in conjunction with selective serotonin reuptake inhibitors. JNJ-28583867 (2-Methyl-4-(4-methylsulfanyl-phenyl)-7-(3-morpholin-4-yl-propoxy)-1,2,3,4-tetrahydro-isoquinoline) is a selective and potent histamine H(3) receptor antagonist (K(i)=10.6 nM) and inhibitor of the serotonin transporter (SERT) (K(i)=3.7 nM), with 30-fold selectivity for SERT over the dopamine and norepinephrine transporters. After subcutaneous administration, JNJ-28583867 occupied both the histamine H(3) receptor and the SERT in rat brain at low doses (<1 mg/kg). JNJ-28583867 blocked imetit-induced drinking (3-10 mg/kg i.p.), confirming in vivo functional activity at the histamine H(3) receptor and also significantly increased cortical extracellular levels of serotonin at doses of 0.3 mg/kg (s.c.) and higher. Smaller increases in cortical extracellular levels of norepinephrine and dopamine were also observed. JNJ-28583867 (3-30 mg/kg p.o.) showed antidepressant-like activity in the mouse tail suspension test. JNJ-28583867 (1-3 mg/kg s.c.) caused a dose-dependent increase in the time spent awake mirrored by a decrease in NREM. Concomitantly, JNJ-28583867 produced a potent suppression of REM sleep from the dose of 1 mg/kg onwards. JNJ-28583867 has good oral bioavailability in the rat (32%), a half-life of 6.9 h and a C(max) of 260 ng/ml after 10 mg/kg p.o. In summary, JNJ-28583867 is a combined histamine H(3) receptor antagonist-SERT inhibitor with in vivo efficacy in biochemical and behavioral models of depression and wakefulness.


Asunto(s)
Antagonistas de los Receptores Histamínicos/farmacología , Receptores Histamínicos H3/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Tetrahidroisoquinolinas/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células CHO , Línea Celular , Cricetinae , Cricetulus , Perros , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Antagonistas de los Receptores Histamínicos/farmacocinética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacocinética , Tetrahidroisoquinolinas/farmacocinética
15.
Front Behav Neurosci ; 11: 83, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28533747

RESUMEN

Orexins peptides exert a prominent role in arousal-related processes including stress responding, by activating orexin-1 (OX1R) and orexin-2 (OX2R) receptors located widely throughout the brain. Stress or orexin administration stimulates hyperarousal, adrenocorticotropic hormone (ACTH) and corticosterone release, and selective OX1R blockade can attenuate several stress-induced behavioral and cardiovascular responses but not the hypothalamic-pituitary-adrenal (HPA) axis activation. As opposed to OX1R, OX2R are preferentially expressed in the paraventricular hypothalamic nucleus which is involved in the HPA axis regulation. In the present study, we investigated the effects of a psychological stress elicited by cage exchange (CE) on ACTH release in two murine models (genetic and pharmacological) of selective OX2R inhibition. CE-induced stress produced a significant increase in ACTH serum levels. Mice lacking the OX2R exhibited a blunted stress response. Stress-induced ACTH release was absent in mice pre-treated with the selective OX2R antagonist JNJ-42847922 (30 mg/kg po), whereas pre-treatment with the dual OX1/2R antagonist SB-649868 (30 mg/kg po) only partially attenuated the increase of ACTH. To assess whether the intrinsic and distinct sleep-promoting properties of each antagonist could account for the differential stress response, a separate group of mice implanted with electrodes for standard sleep recording were orally dosed with JNJ-42847922 or SB-649868 during the light phase. While both compounds reduced the latency to non-rapid eye movement (NREM) sleep without affecting its duration, a prevalent REM-sleep promoting effect was observed only in mice treated with the dual OX1/2R antagonist. These data indicate that in a psychological stress model, genetic or pharmacological inhibition of OX2R markedly attenuated stress-induced ACTH secretion, as a separately mediated effect from the NREM sleep induction of OX2R antagonism.

16.
Front Pharmacol ; 8: 357, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28649201

RESUMEN

Orexin neurons originating in the perifornical and lateral hypothalamic area are highly reactive to anxiogenic stimuli and have strong projections to anxiety and panic-associated circuitry. Recent studies support a role for the orexin system and in particular the orexin 1 receptor (OX1R) in coordinating an integrative stress response. However, no selective OX1R antagonist has been systematically tested in two preclinical models of using panicogenic stimuli that induce panic attack in the majority of people with panic disorder, namely an acute hypercapnia-panic provocation model and a model involving chronic inhibition of GABA synthesis in the perifornical hypothalamic area followed by intravenous sodium lactate infusion. Here we report on a novel brain penetrant, selective and high affinity OX1R antagonist JNJ-54717793 (1S,2R,4R)-7-([(3-fluoro-2-pyrimidin-2-ylphenyl)carbonyl]-N-[5-(trifluoromethyl)pyrazin-2-yl]-7-azabicyclo[2.2.1]heptan-2-amine). JNJ-54717793 is a high affinity/potent OX1R antagonist and has an excellent selectivity profile including 50 fold versus the OX2R. Ex vivo receptor binding studies demonstrated that after oral administration JNJ-54717793 crossed the blood brain barrier and occupied OX1Rs in the rat brain. While JNJ-54717793 had minimal effect on spontaneous sleep in rats and in wild-type mice, its administration in OX2R knockout mice, selectively promoted rapid eye movement sleep, demonstrating target engagement and specific OX1R blockade. JNJ-54717793 attenuated CO2 and sodium lactate induced panic-like behaviors and cardiovascular responses without altering baseline locomotor or autonomic activity. These data confirm that selective OX1R antagonism may represent a novel approach of treating anxiety disorders, with no apparent sedative effects.

17.
Sleep ; 29(9): 1211-23, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17040009

RESUMEN

STUDY OBJECTIVES: Sex is an important determinant of the pathophysiology of several disorders that influence and/or impair sleep-wake regulation. To date, few studies have examined either the role of sex or the gonadal hormones on sleep and wakefulness. The difficulty in performing well-controlled clinical experiments on sex and sleep underscores the need for effective animal models to investigate the influence of the gonadal hormones on sleep-wake states. This study describes the influence of sex on sleep and wakefulness in mice, the primary mammalian genetic model for sleep analysis, and tests the hypothesis that gonadal function drives sex differences in sleep-wake states. DESIGN: Electroencephalogram/electromyogram sleep-wake patterns were recorded in intact and gonadectomized male and female C57BL/6J mice maintained on a 14-hour light:10-hour dark schedule. Following a 24-hour baseline recording, mice were sleep deprived during the light phase by gentle handling and given a 10-hour recovery opportunity during the immediate dark phase. MEASUREMENTS AND RESULTS: Intact female mice spent more time awake than intact males during 24 hours of baseline recording at the expense of non-rapid eye movement (NREM) sleep. Though the recovery response of NREM sleep was similar between males and females, when examined in reference to baseline levels, females exhibited a more robust recovery response. Gonadectomy in males and females reduced or eliminated the majority of sex differences in sleep architecture and homeostasis. CONCLUSIONS: These data demonstrate that the gonadal hormones influence the amount, distribution, and intensity of sleep but do not account for all sex differences in the sleep-wake cycle.


Asunto(s)
Ritmo Circadiano , Gónadas/fisiopatología , Trastornos del Sueño del Ritmo Circadiano/epidemiología , Trastornos del Sueño del Ritmo Circadiano/fisiopatología , Animales , Castración/estadística & datos numéricos , Modelos Animales de Enfermedad , Electroencefalografía , Electromiografía , Femenino , Gónadas/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Ovariectomía/estadística & datos numéricos , Polisomnografía , Factores Sexuales , Privación de Sueño/epidemiología , Trastornos del Sueño del Ritmo Circadiano/diagnóstico
18.
Front Neurosci ; 10: 53, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26973444

RESUMEN

Prolonged exposure to abnormally high calcium concentrations is thought to be a core mechanism underlying hippocampal damage in epileptic patients; however, no prior study has characterized calcium activity during seizures in the live, intact hippocampus. We have directly investigated this possibility by combining whole-brain electroencephalographic (EEG) measurements with microendoscopic calcium imaging of pyramidal cells in the CA1 hippocampal region of freely behaving mice treated with the pro-convulsant kainic acid (KA). We observed that KA administration led to systematic patterns of epileptiform calcium activity: a series of large-scale, intensifying flashes of increased calcium fluorescence concurrent with a cluster of low-amplitude EEG waveforms. This was accompanied by a steady increase in cellular calcium levels (>5 fold increase relative to the baseline), followed by an intense spreading calcium wave characterized by a 218% increase in global mean intensity of calcium fluorescence (n = 8, range [114-349%], p < 10(-4); t-test). The wave had no consistent EEG phenotype and occurred before the onset of motor convulsions. Similar changes in calcium activity were also observed in animals treated with 2 different proconvulsant agents, N-methyl-D-aspartate (NMDA) and pentylenetetrazol (PTZ), suggesting the measured changes in calcium dynamics are a signature of seizure activity rather than a KA-specific pathology. Additionally, despite reducing the behavioral severity of KA-induced seizures, the anticonvulsant drug valproate (VA, 300 mg/kg) did not modify the observed abnormalities in calcium dynamics. These results confirm the presence of pathological calcium activity preceding convulsive motor seizures and support calcium as a candidate signaling molecule in a pathway connecting seizures to subsequent cellular damage. Integrating in vivo calcium imaging with traditional assessment of seizures could potentially increase translatability of pharmacological intervention, leading to novel drug screening paradigms and therapeutics designed to target and abolish abnormal patterns of both electrical and calcium excitation.

19.
Sleep ; 28(4): 395-409, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16171284

RESUMEN

STUDY OBJECTIVES: The finding that deletion or mutation of core circadian clock genes in both mice and flies induce unexpected alterations in sleep amount, sleep architecture and the recovery response to sleep deprivation, has led to new insights into functions of the circadian system that extend beyond its role as a regulator of the timing of the sleep-wake cycle. A key transcription factor in the transcriptional/translational feedback loop of mammalian circadian genes is BMAL1/Mop3, a heterodimeric partner to CLOCK. It was previously shown that mice deficient in the BMAL1/Mop3 gene become immediately arrhythmic in constant darkness and have reduced locomotor activity levels under entrained and constant conditions. In this study, we tested the hypothesis that the mammalian BMAL1/Mop3 gene would have regulatory effects on sleep-wake patterns. DESIGN: In mice with targeted deletion of the BMAL1/Mop3 gene, EEG/EMG sleep-wake patterns were recorded under entrained and free-running conditions as well as following acute (6-hrs) sleep deprivation. MEASUREMENTS AND RESULTS: Mice homozygous for the BMAL1/Mop3 deletion showed an attenuated rhythm of sleep and wakefulness distribution across the 24-hr period. In addition, these mice showed increases in total sleep time, sleep fragmentation and EEG delta power under baseline conditions, and an attenuated compensatory response to acute sleep deprivation. CONCLUSIONS: These new data strengthen the hypothesis that molecular components of the circadian system play a central role in the generation of sleep and wakefulness beyond just the timing of these behavioral vigilance states.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ritmo Circadiano/fisiología , Privación de Sueño/genética , Factores de Transcripción ARNTL , Alelos , Animales , Temperatura Corporal/fisiología , Electroencefalografía , Electromiografía , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Polisomnografía , Fases del Sueño/fisiología , Sueño REM/fisiología
20.
J Med Chem ; 58(14): 5620-36, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26087021

RESUMEN

The preclinical characterization of novel octahydropyrrolo[3,4-c]pyrroles that are potent and selective orexin-2 antagonists is described. Optimization of physicochemical and DMPK properties led to the discovery of compounds with tissue distribution and duration of action suitable for evaluation in the treatment of primary insomnia. These selective orexin-2 antagonists are proven to promote sleep in rats, and this work ultimately led to the identification of a compound that progressed into human clinical trials for the treatment of primary insomnia. The synthesis, SAR, and optimization of the pharmacokinetic properties of this series of compounds as well as the identification of the clinical candidate, JNJ-42847922 (34), are described herein.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Neuropéptidos/antagonistas & inhibidores , Pirroles/química , Pirroles/farmacología , Animales , Ensayos Clínicos como Asunto , Perros , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Neuropéptidos/metabolismo , Receptores de Orexina/metabolismo , Orexinas , Pirroles/farmacocinética , Pirroles/uso terapéutico , Ratas , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Relación Estructura-Actividad , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA