Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Pharmacol Exp Ther ; 336(1): 165-77, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20947638

RESUMO

The normalization of excessive glutamatergic neurotransmission through the activation of metabotropic glutamate 2 (mGlu2) receptors may have therapeutic potential in a variety of psychiatric disorders, including anxiety/depression and schizophrenia. Here, we characterize the pharmacological properties of N-(4-((2-(trifluoromethyl)-3-hydroxy-4-(isobutyryl)phenoxy)methyl)benzyl)-1-methyl-1H-imidazole-4-carboxamide (THIIC), a structurally novel, potent, and selective allosteric potentiator of human and rat mGlu2 receptors (EC(50) = 23 and 13 nM, respectively). THIIC produced anxiolytic-like efficacy in the rat stress-induced hyperthermia assay and the mouse stress-induced elevation of cerebellar cGMP and marble-burying assays. THIIC also produced robust activity in three assays that detect antidepressant-like activity, including the mouse forced-swim test, the rat differential reinforcement of low rate 72-s assay, and the rat dominant-submissive test, with a maximal response similar to that of imipramine. Effects of THIIC in the forced-swim test and marble burying were deleted in mGlu2 receptor null mice. Analysis of sleep electroencephalogram (EEG) showed that THIIC had a sleep-promoting profile with increased non-rapid eye movement (REM) and decreased REM sleep. THIIC also decreased the dark phase increase in extracellular histamine in the medial prefrontal cortex and decreased levels of the histamine metabolite tele-methylhistamine (t-MeHA) in rat cerebrospinal fluid. Collectively, these results indicate that the novel mGlu2-positive allosteric modulator THIIC has robust activity in models used to predict anxiolytic/antidepressant efficacy, substantiating, at least with this molecule, differentiation in the biological impact of mGlu2 potentiation versus mGlu2/3 orthosteric agonism. In addition, we provide evidence that sleep EEG and CSF t-MeHA might function as viable biomarker approaches to facilitate the translational development of THIIC and other mGlu2 potentiators.


Assuntos
Ansiolíticos/farmacologia , Antidepressivos/farmacologia , Compostos de Benzil/farmacologia , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Agonistas de Aminoácidos Excitatórios/farmacologia , Imidazóis/farmacologia , Receptores de Glutamato Metabotrópico/agonistas , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Linhagem Celular , Sistema Nervoso Central/química , Cerebelo/química , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Sinergismo Farmacológico , Humanos , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Ratos Wistar , Receptores de Glutamato Metabotrópico/fisiologia
2.
Sleep ; 30(10): 1255-63, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17969459

RESUMO

STUDY OBJECTIVE: The mammalian circadian clock in the suprachiasmatic nuclei (SCN) of the hypothalamus conveys 24-h rhythmicity to sleep-wake cycles, locomotor activity, and other behavioral and physiological processes. The timing of rhythms relative to the light/dark (LD12:12) cycle is influenced in part by the endogenous circadian period and the time of day specific sensitivity of the clock to light. We now describe a novel circadian rhythm phenotype, and a locus influencing that phenotype, in a segregating population of mice. METHODS: By crossbreeding 2 genetically distinct nocturnal strains of mice (Cast/Ei and C57BL/6J) and backcrossing the resulting progeny to Cast/Ei, we have produced a novel circadian phenotype, called early runner mice. RESULTS: Early runner mice entrain to a light/dark cycle at an advanced phase, up to 9 hours before dark onset. This phenotype is not significantly correlated with circadian period in constant darkness and is not associated with disruption of molecular circadian rhythms in the SCN, as assessed by analysis of period gene expression. We have identified a genomic region that regulates this phenotype-a major quantitative trait locus on chromosome 18 (near D18Mit184) that we have named era1 for Early Runner Activity locus one. Phase delays caused by light exposure early in the subjective night were of smaller magnitude in backcross offspring that were homozygous Cast/Ei at D18Mit184 than in those that were heterozygous at this locus. CONCLUSION: Genetic variability in the circadian response to light may, in part, explain the variance in phase angle of entrainment in this segregating mouse population.


Assuntos
Relógios Biológicos/genética , Cromossomos de Mamíferos , Ritmo Circadiano/genética , Locos de Características Quantitativas/genética , Animais , Relógios Biológicos/fisiologia , Mapeamento Cromossômico , Ritmo Circadiano/fisiologia , Escuridão , Feminino , Luz , Masculino , Camundongos , Camundongos Transgênicos , Estimulação Luminosa
3.
Neuropharmacology ; 108: 415-25, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27150557

RESUMO

Most antidepressants suppress rapid eye movement (REM) sleep, which is thought to be important to brain function, yet the resulting REM sleep restriction is well tolerated. This study investigated the impact of antidepressants with different mechanisms of action, such as selective serotonin reuptake inhibitors (SSRIs) and tricyclic antidepressants (TCA), on the regulation of REM sleep in rats. REM sleep was first demonstrated to be homeostatically regulated using 5, 8 and 10 h of REM-sleep specific restriction through EEG-triggered arousals, with an average of 91 ± 10% of lost REM sleep recovered following a 26-29 -hour recovery period. Acute treatment with the antidepressants paroxetine, citalopram and imipramine inhibited REM sleep by 84 ± 8, 84 ± 8 and 69 ± 9% respectively relative to vehicle control. The pharmacologically-induced REM sleep deficits by paroxetine and citalopram were not fully recovered, whereas, after imipramine the REM sleep deficit was fully compensated. Given the marked difference between REM sleep recovery following the administration of paroxetine, citalopram, imipramine and REM sleep restriction, the homeostatic response was further examined by pairing REM sleep specific restriction with the three antidepressants. Surprisingly, the physiologically-induced REM sleep deficits incurred prior to suppression of REM sleep by all antidepressants was consistently recovered. The data indicate that REM sleep homeostasis remains operative following subsequent treatment with antidepressants and is unaffected by additional pharmacological inhibition of REM sleep.


Assuntos
Antidepressivos/farmacologia , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Sono REM/efeitos dos fármacos , Sono REM/fisiologia , Animais , Antidepressivos Tricíclicos/farmacologia , Masculino , Ratos , Ratos Wistar , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
4.
Psychopharmacology (Berl) ; 232(21-22): 3977-89, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25902875

RESUMO

While treatment options are available, excessive daytime sleepiness (EDS) remains a significant unmet medical need for many patients. Relatively little rodent behavioural pharmacology has been conducted in this context to assess potential pro-vigilant compounds for their ability to restore functional capacity following experimentally induced sleep loss. Male Wistar rats were prepared for electroencephalographic (EEG) recording and subject to 11 h of sleep restriction using a biofeedback-induced cage rotation protocol. A simple response latency task (SRLT) was used to behaviourally index sleep restriction and the effects of pro-vigilant compounds: modafinil, D-amphetamine, caffeine, and the mGlu5-positive allosteric modulator LSN2814617. Sleep restriction resulted in a consistent, quantified loss of non-rapid eye movement (NREM) and REM sleep that impaired SRLT performance in a manner suggestive of progressive task disengagement. In terms of EEG parameters, all compounds induced wakefulness. Amphetamine treatment further decreased SRLT performance capacity, whereas the other three compounds decreased omissions and allowed animals to re-engage in the task. Caffeine and modafinil also significantly increased premature responses during this period, an effect not observed for LSN2814617. While all compounds caused compensatory sleep responses, the magnitude of compensation observed for LSN2814617 was much smaller than would be predicted to result from the prolongation of wakefulness exhibited. Using simple response latencies to index performance, an mGlu5 PAM dramatically increased wakefulness and improved functional capacity of sleep-restricted animals, without eliciting a proportionate compensatory sleep response. This effect was qualitatively distinct from that of amphetamine, caffeine and modafinil.


Assuntos
Nível de Alerta/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Oxidiazóis/farmacologia , Piridinas/farmacologia , Receptor de Glutamato Metabotrópico 5/agonistas , Triazóis/farmacologia , Animais , Compostos Benzidrílicos/farmacologia , Biorretroalimentação Psicológica , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Condicionamento Operante/efeitos dos fármacos , Dextroanfetamina/farmacologia , Eletroencefalografia/efeitos dos fármacos , Masculino , Modafinila , Desempenho Psicomotor/efeitos dos fármacos , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Sono/efeitos dos fármacos , Privação do Sono , Sono REM/efeitos dos fármacos
5.
BMC Neurosci ; 3: 20, 2002 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-12495442

RESUMO

BACKGROUND: The cryptochrome 1 and 2 genes (cry1 and cry2) are necessary for the generation of circadian rhythms, as mice lacking both of these genes (cry1,2-/-) lack circadian rhythms. We studied sleep in cry1,2-/- mice under baseline conditions as well as under conditions of constant darkness and enforced wakefulness to determine whether cryptochromes influence sleep regulatory processes. RESULTS: Under all three conditions, cry1,2-/- mice exhibit the hallmarks of high non-REM sleep (NREMS) drive (i.e., increases in NREMS time, NREMS consolidation, and EEG delta power during NREMS). This unexpected phenotype was associated with elevated brain mRNA levels of period 1 and 2 (per1,2), and albumin d-binding protein (dbp), which are known to be transcriptionally inhibited by CRY1,2. To further examine the relationship between circadian genes and sleep homeostasis, we examined wild type mice and rats following sleep deprivation and found increased levels of per1,2 mRNA and decreased levels of dbp mRNA specifically in the cerebral cortex; these changes subsided with recovery sleep. The expression of per3, cry1,2, clock, npas2, bmal1, and casein-kinase-1epsilon did not change with sleep deprivation. CONCLUSIONS: These results indicate that mice lacking cryptochromes are not simply a genetic model of circadian arrhythmicity in rodents and functionally implicate cryptochromes in the homeostatic regulation of sleep.


Assuntos
Ritmo Circadiano/fisiologia , Proteínas de Ligação a DNA , Proteínas de Drosophila , Proteínas do Olho , Flavoproteínas/fisiologia , Células Fotorreceptoras de Invertebrados , Sono/fisiologia , Fatores de Transcrição ARNTL , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas CLOCK , Caseína Quinases , Proteínas de Ciclo Celular , Criptocromos , Escuridão , Ritmo Delta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Circadianas Period , Fenótipo , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G , Privação do Sono/metabolismo , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vigília
6.
Sleep ; 27(4): 619-27, 2004 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15282996

RESUMO

STUDY OBJECTIVES: Hypocretins (orexins) are involved in the sleep disorder narcolepsy. While hypocretin-1 has a daily oscillation, little is known regarding the relative contribution of circadian and homeostatic components on hypocretin release. The effect of lesions of the suprachiasmatic nucleus (SCN) on hypocretin-1 in the cerebrospinal fluid (CSF) was examined. DESIGN: SCN-ablated (SCNx) and sham-operated control rats were implanted with activity-temperature transmitters. Animals were housed individually under 1 of 3 lighting conditions: 12-hour:12-hour light:dark cycle (LD), constant light (LL), and constant darkness (DD). Lesions were verified histologically and shown not to affect hypocretin-containing cells. Hypocretin-1 concentrations in the CSF were determined every 4 hours using radioimmunoassays. MEASUREMENTS AND RESULTS: Control animals displayed robust circadian (LL, DD) and diurnal (LD) fluctuations in CSF hypocretin-1, locomotor activity, and temperature. Peak CSF hypocretin-1 was at the end of the active period. Activity, temperature, and CSF hypocretin-1 were arrhythmic in SCNx animals in LL and DD. In LD, a weak but significant fluctuation in activity and temperature but not CSF hypocretin-1 was observed in SCNx animals. We also explored correlations between CSF hypocretin-1, CSF corticosterone, and locomotor activity occurring prior to CSF sampling in arrhythmic SCNx rats under constant conditions. Significant correlations between hypocretin-1 and activity were observed both across and within animals, suggesting that interindividual and time-of-the-day differences in activity have significant effects on hypocretin release in arrhythmic animals. No correlation was found between CSF hypocretin-1 and corticosterone. CONCLUSIONS: Hypocretin-1 release is under SCN control. Locomotor activity influences the activity of the hypocretin neurons.


Assuntos
Proteínas de Transporte/metabolismo , Ritmo Circadiano/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos/metabolismo , Núcleo Supraquiasmático/patologia , Animais , Temperatura Corporal/fisiologia , Proteínas de Transporte/líquido cefalorraquidiano , Locomoção/fisiologia , Narcolepsia/líquido cefalorraquidiano , Narcolepsia/metabolismo , Narcolepsia/patologia , Neuropeptídeos/líquido cefalorraquidiano , Orexinas , Radioimunoensaio , Ratos
7.
Brain Res ; 955(1-2): 221-8, 2002 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-12419540

RESUMO

Mice lacking the GABA(A) receptor beta(3) subunit exhibit a profound disruption in thalamic circuitry. We have studied sleep in these mice under baseline conditions and following treatment with the benzodiazepine midazolam. Under baseline conditions, NREM sleep time did not differ between beta(3) subunit knockout mice and wild type mice, while REM sleep time was significantly lower in knockout mice than in wild type mice during the light portion of a 24-h light-dark cycle. In constant dark conditions, circadian rhythmicity remained intact in mutant mice for a period of at least 9 days. EEG delta power (1-4 Hz) was significantly greater in the knockout than in wild type mice during NREM sleep but not during other states. A transient increase in EEG power in the 12-16 Hz range that occurred in wild type mice just prior to the transition from NREM to REM sleep was present but significantly blunted in the knockout. Midazolam decreased NREM delta power and REM time in wild type mice. The former but not the latter response to midazolam was intact in the knockout. These results further support a role for GABAergic transmission in regulating REM sleep and EEG spectral phenomena associated with NREM sleep.


Assuntos
Eletroencefalografia , Receptores de GABA-A/deficiência , Receptores de GABA-A/genética , Fases do Sono/genética , Animais , Ritmo Circadiano/efeitos dos fármacos , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Relação Dose-Resposta a Droga , Eletroencefalografia/efeitos dos fármacos , Feminino , Masculino , Camundongos , Camundongos Knockout , Midazolam/farmacologia , Fases do Sono/efeitos dos fármacos , Fases do Sono/fisiologia
8.
Neuropharmacology ; 64: 224-39, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22884720

RESUMO

The demonstrated functional interaction of metabotropic glutamate 5 (mGlu5) receptors with N-methyl-d-aspartate (NMDA) receptors has prompted speculation that their activation may offer a potential treatment for aspects of schizophrenia. Development of selective mGlu5 agonists has been difficult, but several different positive allosteric modulator (PAM) molecules have now been identified. This study describes two novel mGlu5 PAMs, LSN2463359 (N-(1-methylethyl)-5-(pyridin-4-ylethynyl)pyridine-2-carboxamide) and LSN2814617 [(7S)-3-tert-butyl-7-[3-(4-fluorophenyl)-1,2,4-oxadiazol-5-yl]-5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-A]pyridine], which are useful tools for this field of research. Both compounds are potent and selective potentiators of human and rat mGlu5 receptors in vitro, displaying curve shift ratios of two to three fold in the concentration-response relationship to glutamate or the glutamate receptor agonist, DHPG, with no detectable intrinsic agonist properties. Both compounds displaced the mGlu5 receptor antagonist radioligand, [³H]MPEP in vitro and, following oral administration reached brain concentrations sufficient to occupy hippocampal mGlu5 receptors as measured in vivo by dose-dependent displacement from the hippocampus of intravenously administered MPEPy. In vivo EEG studies demonstrated that these mGlu5 PAMs have marked wake-promoting properties but little in the way of rebound hypersomnolence. In contrast, the previously described mGlu5 PAMs CDPPB and ADX47273 showed relatively poor evidence of in vivo target engagement in either receptor occupancy assays or EEG disturbance. Wake-promoting doses of LSN2463359 and LSN2814617 attenuated deficits in performance induced by the competitive NMDA receptor antagonist SDZ 220,581 in two tests of operant behaviour: the variable interval 30 s task and the DMTP task. These effects were lost if the dose of either compound extended into the range which disrupted performance in the baseline DMTP task. However, the improvements in response accuracy induced by the mGlu5 potentiators in SDZ 220,581-treated rats were not delay-dependent and, therefore, perhaps more likely reflected optimization of general arousal than specific beneficial effects on discrete cognitive processes. The systematic profiling of LSN2463359 and LSN2814617 alongside other previously described molecules will help determine more precisely how mGlu5 potentiator pharmacology might provide therapeutic benefit. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Assuntos
Córtex Cerebral/efeitos dos fármacos , Drogas em Investigação/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Neurônios/efeitos dos fármacos , Nootrópicos/farmacologia , Receptores de Glutamato Metabotrópico/agonistas , Esquizofrenia/tratamento farmacológico , Regulação Alostérica , Animais , Nível de Alerta/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Linhagem Celular , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Relação Dose-Resposta a Droga , Drogas em Investigação/efeitos adversos , Drogas em Investigação/metabolismo , Drogas em Investigação/uso terapêutico , Embrião de Mamíferos/citologia , Agonistas de Aminoácidos Excitatórios/efeitos adversos , Agonistas de Aminoácidos Excitatórios/metabolismo , Agonistas de Aminoácidos Excitatórios/uso terapêutico , Humanos , Masculino , Neurônios/citologia , Neurônios/metabolismo , Nootrópicos/efeitos adversos , Nootrópicos/metabolismo , Nootrópicos/uso terapêutico , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fases do Sono/efeitos dos fármacos , Distribuição Tecidual
9.
Neuron ; 76(3): 526-33, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-23141065

RESUMO

Rhythmic neural network activity patterns are defining features of sleep, but interdependencies between limbic and cortical oscillations at different frequencies and their functional roles have not been fully resolved. This is particularly important given evidence linking abnormal sleep architecture and memory consolidation in psychiatric diseases. Using EEG, local field potential (LFP), and unit recordings in rats, we show that anteroposterior propagation of neocortical slow-waves coordinates timing of hippocampal ripples and prefrontal cortical spindles during NREM sleep. This coordination is selectively disrupted in a rat neurodevelopmental model of schizophrenia: fragmented NREM sleep and impaired slow-wave propagation in the model culminate in deficient ripple-spindle coordination and disrupted spike timing, potentially as a consequence of interneuronal abnormalities reflected by reduced parvalbumin expression. These data further define the interrelationships among slow-wave, spindle, and ripple events, indicating that sleep disturbances may be associated with state-dependent decoupling of hippocampal and cortical circuits in psychiatric diseases.


Assuntos
Modelos Animais de Doenças , Hipocampo/crescimento & desenvolvimento , Córtex Pré-Frontal/crescimento & desenvolvimento , Esquizofrenia/fisiopatologia , Sono/fisiologia , Animais , Eletroencefalografia/métodos , Feminino , Vias Neurais/crescimento & desenvolvimento , Gravidez , Ratos , Ratos Sprague-Dawley
10.
Science ; 331(6019): 906-9, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21330544

RESUMO

Black bears hibernate for 5 to 7 months a year and, during this time, do not eat, drink, urinate, or defecate. We measured metabolic rate and body temperature in hibernating black bears and found that they suppress metabolism to 25% of basal rates while regulating body temperature from 30° to 36°C, in multiday cycles. Heart rates were reduced from 55 to as few as 9 beats per minute, with profound sinus arrhythmia. After returning to normal body temperature and emerging from dens, bears maintained a reduced metabolic rate for up to 3 weeks. The pronounced reduction and delayed recovery of metabolic rate in hibernating bears suggest that the majority of metabolic suppression during hibernation is independent of lowered body temperature.


Assuntos
Temperatura Corporal , Metabolismo Energético , Hibernação , Consumo de Oxigênio , Ursidae/fisiologia , Animais , Metabolismo Basal , Feminino , Frequência Cardíaca , Humanos , Masculino , Fatores de Tempo , Ursidae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA