Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Neuropsychopharmacology ; 38(12): 2401-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23722242

ABSTRACT

Dual orexin receptor antagonists (DORAs) induce sleep by blocking orexin 1 and orexin 2 receptor-mediated activities responsible for regulating wakefulness. DORAs represent a potential alternative mechanism to the current standard of care that includes the γ-aminobutyric acid (GABA)A receptor-positive allosteric modulators, eszopiclone and zolpidem. This work uses an innovative method to analyze electroencephalogram (EEG) spectral frequencies within sleep/wake states to differentiate the effects of GABAA modulators from DORA-22, an analog of the DORA MK-6096, in Sprague-Dawley rats. The effects of low, intermediate, and high doses of eszopiclone, zolpidem, and DORA-22 were examined after first defining each compound's ability to promote sleep during active-phase dosing. The EEG spectral frequency power within specific sleep stages was calculated in 1-Hz intervals from 1 to 100 Hz within each sleep/wake state for the first 4 h after the dose. Eszopiclone and zolpidem produced marked, dose-responsive disruptions in sleep stage-specific EEG spectral profiles compared with vehicle treatment. In marked contrast, DORA-22 exhibited marginal changes in the spectral profile, observed only during rapid eye movement sleep, and only at the highest dose tested. Moreover, while eszopiclone- and zolpidem-induced changes were evident in the inactive period, the EEG spectral responses to DORA-22 were absent during this phase. These results suggest that DORA-22 differs from eszopiclone and zolpidem whereby DORA-22 promotes somnolence without altering the neuronal network EEG activity observed during normal sleep.


Subject(s)
Azabicyclo Compounds/pharmacology , Brain/drug effects , GABA-A Receptor Agonists/pharmacology , Orexin Receptor Antagonists , Piperazines/pharmacology , Piperidines/pharmacology , Pyridines/pharmacology , Sleep Stages/drug effects , Triazoles/pharmacology , Animals , Azabicyclo Compounds/administration & dosage , Brain/physiology , Electroencephalography , Eszopiclone , GABA-A Receptor Agonists/administration & dosage , Male , Piperazines/administration & dosage , Piperidines/administration & dosage , Pyridines/administration & dosage , Rats , Rats, Sprague-Dawley , Sleep Stages/physiology , Triazoles/administration & dosage , Zolpidem
2.
J Neurogenet ; 25(4): 167-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22091728

ABSTRACT

Despite the substantial impact of sleep disturbances on human health and the many years of study dedicated to understanding sleep pathologies, the underlying genetic mechanisms that govern sleep and wake largely remain unknown. Recently, the authors completed large-scale genetic and gene expression analyses in a segregating inbred mouse cross and identified candidate causal genes that regulate the mammalian sleep-wake cycle, across multiple traits including total sleep time, amounts of rapid eye movement (REM), non-REM, sleep bout duration, and sleep fragmentation. Here the authors describe a novel approach toward validating candidate causal genes, while also identifying potential targets for sleep-related indications. Select small-molecule antagonists and agonists were used to interrogate candidate causal gene function in rodent sleep polysomnography assays to determine impact on overall sleep architecture and to evaluate alignment with associated sleep-wake traits. Significant effects on sleep architecture were observed in validation studies using compounds targeting the muscarinic acetylcholine receptor M3 subunit (Chrm3) (wake promotion), nicotinic acetylcholine receptor alpha4 subunit (Chrna4) (wake promotion), dopamine receptor D5 subunit (Drd5) (sleep induction), serotonin 1D receptor (Htr1d) (altered REM fragmentation), glucagon-like peptide-1 receptor (Glp1r) (light sleep promotion and reduction of deep sleep), and calcium channel, voltage-dependent, T type, alpha 1I subunit (Cacna1i) (increased bout duration of slow wave sleep). Taken together, these results show the complexity of genetic components that regulate sleep-wake traits and highlight the importance of evaluating this complex behavior at a systems level. Pharmacological validation of genetically identified putative targets provides a rapid alternative to generating knock out or transgenic animal models, and may ultimately lead towards new therapeutic opportunities.


Subject(s)
Crosses, Genetic , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/genetics , Sleep/drug effects , Sleep/genetics , Animals , Calcium Channels, N-Type , Calcium Channels, P-Type/genetics , Calcium Channels, Q-Type/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M3/genetics , Receptors, Dopamine D5/genetics , Receptors, Nicotinic/genetics , Sleep Wake Disorders/metabolism
3.
Brain Res ; 1416: 69-79, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21885038

ABSTRACT

Modulation of TASK-3 (Kcnk9) potassium channels affect neurotransmitter release in thalamocortical centers and other sleep-related nuclei having the capacity to regulate arousal cycles and REM sleep changes associated with mood disorders and antidepressant action. Circumstantial evidence from this and previous studies suggest the potential for TASK-3 to be a novel antidepressant therapeutic target; TASK-3 knock-out mice display augmented circadian amplitude and exhibit sleep architecture characterized by suppressed REM activity. Detailed analysis of locomotor activity indicates that the amplitudes of activity bout duration and bout number are augmented in TASK-3 mutants well beyond that seen in wildtypes, findings substantiated by amplitude increases in body temperature and EEG recordings of sleep stage bouts. Polysomnographic analysis of TASK-3 mutants reveals increases in nocturnal active wake and suppressed REM sleep time while increased slow wave sleep typifies the inactive phase, findings that have implications for the cognitive impact of reduced TASK-3 activity. In direct measures of their resistance to despair behavior, TASK-3 knock-outs displayed significant decreases in immobility relative to wildtype controls in both tail suspension and forced swim tests. Treatment of wildtype animals with the antidepressant Fluoxetine markedly reduced REM sleep, while leaving active wake and slow wave sleep relatively intact. Remarkably, these effects were absent in TASK-3 mutants indicating that TASK-3 is either directly involved in the mechanism of this drug's action, or participates in parallel pathways that achieve the same effect. Together, these results support the TASK-3 channel to act as a therapeutic target for antidepressant action.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Arousal/physiology , Circadian Rhythm/physiology , Fluoxetine/pharmacology , Potassium Channels/metabolism , Sleep, REM/physiology , Animals , Behavior, Animal/physiology , Depression/drug therapy , Depression/metabolism , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Male , Matched-Pair Analysis , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Neurologic Mutants , Phenotype , Potassium Channels/drug effects , Potassium Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL