Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cereb Cortex ; 29(3): 1090-1108, 2019 03 01.
Article in English | MEDLINE | ID: mdl-29462275

ABSTRACT

We have proposed that cortical nNOS/NK1R interneurons have a role in sleep homeostasis. The hypocretins (orexins) are wake-promoting neuropeptides and hypocretin/orexin (Hcrt) neurons project to the cortex. Hcrt peptides affect deep layer cortical neurons, and Hcrt receptor 1 (Hcrtr1; Ox1r) mRNA is expressed in cortical nNOS/NK1R cells. Therefore, we investigated whether Hcrt neuron stimulation affects cingulate cortex nNOS/NK1R neurons. Bath application of HCRT1/orexin-A evoked an inward current and membrane depolarization in most nNOS/NK1R cells which persisted in tetrodotoxin; optogenetic stimulation of Hcrt terminals expressing channelrhodopsin-2 confirmed these results, and pharmacological studies determined that HCRTR1 mediated these responses. Single-cell RT-PCR found Hcrtr1 mRNA in 31% of nNOS/NK1R cells without any Hcrtr2 mRNA expression; immunohistochemical studies of Hcrtr1-EGFP mice confirmed that a minority of nNOS/NK1R cells express HCRTR1. When Hcrt neurons degenerated in orexin-tTA;TetO DTA mice, the increased EEG delta power during NREM sleep produced in response to 4 h sleep deprivation and c-FOS expression in cortical nNOS/NK1R cells during recovery sleep were indistinguishable from that of controls. We conclude that Hcrt excitatory input to these deep layer cells is mediated through HCRTR1 but is unlikely to be involved in the putative role of cortical nNOS/NK1R neurons in sleep homeostasis.


Subject(s)
Gyrus Cinguli/physiology , Homeostasis , Neurons/physiology , Nitric Oxide Synthase Type I/physiology , Orexin Receptors/physiology , Receptors, Neurokinin-1/physiology , Sleep/physiology , Animals , Female , Gyrus Cinguli/drug effects , Hypothalamic Area, Lateral/physiology , Male , Mice, Inbred C57BL , Neurons/drug effects , Orexins/administration & dosage , Orexins/physiology
2.
Biol Psychiatry ; 82(9): 623-633, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-27919403

ABSTRACT

BACKGROUND: Narcolepsy, a disorder of rapid eye movement (REM) sleep, is characterized by excessive daytime sleepiness and cataplexy, a loss of muscle tone triggered by emotional stimulation. Current narcolepsy pharmacotherapeutics include controlled substances with abuse potential or drugs with undesirable side effects. As partial agonists at trace amine-associated receptor 1 (TAAR1) promote wakefulness in mice and rats, we evaluated whether TAAR1 agonism had beneficial effects in two mouse models of narcolepsy. METHODS: In the first experiment, male homozygous B6-Taar1tm1(NLSLacZ)Blt (Taar1 knockout) and wild-type mice were surgically implanted to record electroencephalogram, electromyogram, locomotor activity, and body temperature, and the efficacy of the TAAR1 agonist, RO5256390, on sleep/wake and physiological parameters was determined. In the second experiment, the effects of the TAAR1 full agonist RO5256390 and partial agonist RO5263397 on sleep/wake, locomotor activity, body temperature, and cataplexy were assessed in two mouse narcolepsy models. RESULTS: RO5256390 profoundly reduced rapid eye movement sleep in wild-type mice; these effects were eliminated in Taar1 knockout mice. The TAAR1 partial agonist RO5263397 also promoted wakefulness and suppressed nonrapid eye movement sleep. Both compounds reduced body temperature in the two narcolepsy models at the highest doses tested. Both TAAR1 compounds also mitigated cataplexy, the pathognomonic symptom of this disorder, in the narcolepsy models. The therapeutic benefit was mediated through a reduction in number of cataplexy episodes and time spent in cataplexy. CONCLUSIONS: These results suggest TAAR1 agonism as a new therapeutic pathway for treatment of this orphan disease. The common underlying mechanism may be the suppression of rapid eye movement sleep.


Subject(s)
Narcolepsy/drug therapy , Receptors, G-Protein-Coupled/agonists , Sleep, REM/drug effects , Wakefulness-Promoting Agents/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Electroencephalography , Electromyography , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Motor Activity/drug effects , Oxazoles/pharmacology , Rare Diseases
3.
Neuropsychopharmacology ; 42(6): 1305-1314, 2017 May.
Article in English | MEDLINE | ID: mdl-27658486

ABSTRACT

Trace amine-associated receptor 1 (TAAR1) agonists have been shown to have procognitive, antipsychotic-like, anxiolytic, weight-reducing, glucose-lowering, and wake-promoting activities. We used Taar1 knockout (KO) and overexpressing (OE) mice and TAAR1 agonists to elucidate the role of TAAR1 in sleep/wake. EEG, EMG, body temperature (Tb), and locomotor activity (LMA) were recorded in Taar1 KO, OE, and WT mice. Following a 24 h recording to characterize basal sleep/wake parameters, mice were sleep deprived (SD) for 6 h. In another experiment, mice were given three doses of the TAAR1 partial agonist RO5263397, caffeine, or vehicle p.o. Baseline wakefulness was modestly increased in OE compared with WT mice. Baseline theta (4.5-9 Hz) and low gamma (30-60 Hz) activity was elevated in KO compared with OE mice in NREM and REM sleep. Following SD, both KO and OE mice exhibited a homeostatic sleep rebound. In WT mice, RO5263397 increased waking and reduced NREM and REM sleep, decreased gamma power during wake and NREM, and decreased Tb without affecting LMA; these effects were absent in KO mice and potentiated in OE mice. In contrast, caffeine increased wake time, NREM gamma power, and LMA in all strains compared with vehicle; this effect was attenuated in KO and potentiated in OE mice. TAAR1 overexpression modestly increases wakefulness, whereas TAAR1 partial agonism increases wakefulness and also reduces NREM and also REM sleep. These results indicate a modulatory role for TAAR1 in sleep/wake and cortical activity and suggest TAAR1 as a novel target for wake-promoting therapeutics.


Subject(s)
Behavior, Animal/physiology , Brain Waves/physiology , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Oxazoles/pharmacology , Receptors, G-Protein-Coupled/physiology , Sleep Stages/physiology , Wakefulness/physiology , Animals , Behavior, Animal/drug effects , Brain Waves/drug effects , Caffeine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Male , Mice , Mice, Knockout , Oxazoles/administration & dosage , Receptors, G-Protein-Coupled/agonists , Sleep Stages/drug effects , Wakefulness/drug effects
4.
J Neurosci ; 34(19): 6495-509, 2014 May 07.
Article in English | MEDLINE | ID: mdl-24806676

ABSTRACT

The sleep disorder narcolepsy results from loss of hypothalamic orexin/hypocretin neurons. Although narcolepsy onset is usually postpubertal, current mouse models involve loss of either orexin peptides or orexin neurons from birth. To create a model of orexin/hypocretin deficiency with closer fidelity to human narcolepsy, diphtheria toxin A (DTA) was expressed in orexin neurons under control of the Tet-off system. Upon doxycycline removal from the diet of postpubertal orexin-tTA;TetO DTA mice, orexin neurodegeneration was rapid, with 80% cell loss within 7 d, and resulted in disrupted sleep architecture. Cataplexy, the pathognomic symptom of narcolepsy, occurred by 14 d when ∼5% of the orexin neurons remained. Cataplexy frequency increased for at least 11 weeks after doxycycline. Temporary doxycycline removal followed by reintroduction after several days enabled partial lesion of orexin neurons. DTA-induced orexin neurodegeneration caused a body weight increase without a change in food consumption, mimicking metabolic aspects of human narcolepsy. Because the orexin/hypocretin system has been implicated in the control of metabolism and addiction as well as sleep/wake regulation, orexin-tTA; TetO DTA mice are a novel model in which to study these functions, for pharmacological studies of cataplexy, and to study network reorganization as orexin input is lost.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Narcolepsy/drug therapy , Neurons/drug effects , Neuropeptides/antagonists & inhibitors , Animals , Body Weight/drug effects , Body Weight/physiology , Cataplexy/physiopathology , Diphtheria Toxin/genetics , Disease Models, Animal , Doxycycline/pharmacology , Drinking/drug effects , Drinking/physiology , Eating/drug effects , Eating/physiology , Electroencephalography , Electromyography , Female , Food , Male , Mice , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Narcolepsy/physiopathology , Orexins , Sleep/physiology , Wakefulness/physiology
5.
Brain ; 136(Pt 7): 2159-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23801738

ABSTRACT

Deficits in sleep and circadian organization have been identified as common early features in patients with Huntington's disease that correlate with symptom severity and may be instrumental in disease progression. Studies in Huntington's disease gene carriers suggest that alterations in the electroencephalogram may reflect underlying neuronal dysfunction that is present in the premanifest stage. We conducted a longitudinal characterization of sleep/wake and electroencephalographic activity in the R6/2 mouse model of Huntington's disease to determine whether analogous electroencephalographic 'signatures' could be identified early in disease progression. R6/2 and wild-type mice were implanted for electroencephalographic recordings along with telemetry for the continuous recording of activity and body temperature. Diurnal patterns of activity and core body temperature were progressively disrupted in R6/2 mice, with a large reduction in the amplitude of these rhythms apparent by 13 weeks of age. The diurnal variation in sleep/wake states was gradually attenuated as sleep became more fragmented and total sleep time was reduced relative to wild-type mice. These genotypic differences were augmented at 17 weeks and evident across the entire 24-h period. Quantitative electroencephalogram analysis revealed anomalous increases in high beta and gamma activity (25-60 Hz) in all sleep/wake states in R6/2 mice, along with increases in theta activity during both non-rapid eye movement and rapid eye movement sleep and a reduction of delta power in non-rapid eye movement sleep. These dramatic alterations in quantitative electroencephalographic measures were apparent from our earliest recording (9 weeks), before any major differences in diurnal physiology or sleep/wake behaviour occurred. In addition, the homeostatic response to sleep deprivation was greatly attenuated with disease progression. These findings demonstrate the sensitivity of quantitative electroencephalographic analysis to identify early pathophysiological alterations in the R6/2 model of Huntington's disease and suggest longitudinal studies in other preclinical Huntington's disease models are needed to determine the generality of these observations as a potential adjunct in therapeutic development.


Subject(s)
Brain Waves/physiology , Circadian Rhythm/physiology , Huntington Disease/complications , Sleep Stages/physiology , Sleep Wake Disorders/etiology , Analysis of Variance , Animals , Body Temperature/genetics , Brain Waves/genetics , Circadian Rhythm/genetics , Disease Models, Animal , Disease Progression , Electroencephalography , Electromyography , Humans , Huntingtin Protein , Huntington Disease/genetics , Longitudinal Studies , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Phenotype , Sleep Deprivation/physiopathology , Sleep Stages/genetics , Spectrum Analysis , Trinucleotide Repeats/genetics , Wakefulness/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...