Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
eNeuro ; 3(2)2016.
Article in English | MEDLINE | ID: mdl-27022631

ABSTRACT

Hypocretin 1 and 2 (Hcrts; also known as orexin A and B), excitatory neuropeptides synthesized in cells located in the tuberal hypothalamus, play a central role in the control of arousal. Hcrt inputs to the locus coeruleus norepinephrine (LC NE) system and the posterior hypothalamic histaminergic tuberomammillary nuclei (TMN HA) are important efferent pathways for Hcrt-induced wakefulness. The LC expresses Hcrt receptor 1 (HcrtR1), whereas HcrtR2 is found in the TMN. Although the dual Hcrt/orexin receptor antagonist almorexant (ALM) decreases wakefulness and increases NREM and REM sleep time, the neural circuitry that mediates these effects is currently unknown. To test the hypothesis that ALM induces sleep by selectively disfacilitating subcortical wake-promoting populations, we ablated LC NE neurons (LCx) or TMN HA neurons (TMNx) in rats using cell-type-specific saporin conjugates and evaluated sleep/wake following treatment with ALM and the GABAA receptor modulator zolpidem (ZOL). Both LCx and TMNx attenuated the promotion of REM sleep by ALM without affecting ALM-mediated increases in NREM sleep. Thus, eliminating either HcrtR1 signaling in the LC or HcrtR2 signaling in the TMN yields similar effects on ALM-induced REM sleep without affecting NREM sleep time. In contrast, neither lesion altered ZOL efficacy on any measure of sleep-wake regulation. These results contrast with those of a previous study in which ablation of basal forebrain cholinergic neurons attenuated ALM-induced increases in NREM sleep time without affecting REM sleep, indicating that Hcrt neurotransmission influences distinct aspects of NREM and REM sleep at different locations in the sleep-wake regulatory network.


Subject(s)
Acetamides/pharmacology , Hypothalamic Area, Lateral/physiology , Isoquinolines/pharmacology , Locus Coeruleus/physiology , Orexins/metabolism , Sleep, REM/drug effects , Analysis of Variance , Animals , Dose-Response Relationship, Drug , Electroencephalography , Electromyography , GABA-A Receptor Agonists/pharmacology , Histamine/metabolism , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/injuries , Locus Coeruleus/drug effects , Locus Coeruleus/injuries , Male , Norepinephrine/metabolism , Orexins/antagonists & inhibitors , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/toxicity , Saporins , Telemetry , Wakefulness/drug effects , Zolpidem
2.
Neuropsychopharmacology ; 41(4): 1144-55, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26289145

ABSTRACT

The dual hypocretin receptor (HcrtR) antagonist almorexant (ALM) may promote sleep through selective disfacilitation of wake-promoting systems, whereas benzodiazepine receptor agonists (BzRAs) such as zolpidem (ZOL) induce sleep through general inhibition of neural activity. Previous studies have indicated that HcrtR antagonists cause less-functional impairment than BzRAs. To gain insight into the mechanisms underlying these differential profiles, we compared the effects of ALM and ZOL on functional activation of wake-promoting systems at doses equipotent for sleep induction. Sprague-Dawley rats, implanted for EEG/EMG recording, were orally administered vehicle (VEH), 100 mg/kg ALM, or 100 mg/kg ZOL during their active phase and either left undisturbed or kept awake for 90 min after which their brains were collected. ZOL-treated rats required more stimulation to maintain wakefulness than VEH- or ALM-treated rats. We measured Fos co-expression with markers for wake-promoting cell groups in the lateral hypothalamus (Hcrt), tuberomammillary nuclei (histamine; HA), basal forebrain (acetylcholine; ACh), dorsal raphe (serotonin; 5HT), and singly labeled Fos(+) cells in the locus coeruleus (LC). Following SD, Fos co-expression in Hcrt, HA, and ACh neurons (but not in 5HT neurons) was consistently elevated in VEH- and ALM-treated rats, whereas Fos expression in these neuronal groups was unaffected by SD in ZOL-treated rats. Surprisingly, Fos expression in the LC was elevated in ZOL- but not in VEH- or ALM-treated SD animals. These results indicate that Hcrt signaling is unnecessary for the activation of Hcrt, HA, or ACh wake-active neurons, which may underlie the milder cognitive impairment produced by HcrtR antagonists compared to ZOL.


Subject(s)
Acetamides/administration & dosage , Brain/physiology , Isoquinolines/administration & dosage , Neurons/physiology , Orexin Receptor Antagonists/administration & dosage , Pyridines/administration & dosage , Sleep Stages/drug effects , Wakefulness/drug effects , Animals , Brain/drug effects , Brain/metabolism , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Electroencephalography , Electromyography , Histamine/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Orexins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/drug effects , Serotonergic Neurons/metabolism , Zolpidem
3.
Brain Struct Funct ; 221(2): 923-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25431268

ABSTRACT

Hypocretin/orexin (HCRT) neurons provide excitatory input to wake-promoting brain regions including the basal forebrain (BF). The dual HCRT receptor antagonist almorexant (ALM) decreases waking and increases sleep. We hypothesized that HCRT antagonists induce sleep, in part, through disfacilitation of BF neurons; consequently, ALM should have reduced efficacy in BF-lesioned (BFx) animals. To test this hypothesis, rats were given bilateral IgG-192-saporin injections, which predominantly targets cholinergic BF neurons. BFx and intact rats were then given oral ALM, the benzodiazepine agonist zolpidem (ZOL) or vehicle (VEH) at lights-out. ALM was less effective than ZOL at inducing sleep in BFx rats compared to controls. BF adenosine (ADO), γ-amino-butyric acid (GABA), and glutamate levels were then determined via microdialysis from intact, freely behaving rats following oral ALM, ZOL or VEH. ALM increased BF ADO and GABA levels during waking and mixed vigilance states, and preserved sleep-associated increases in GABA under low and high sleep pressure conditions. ALM infusion into the BF also enhanced cortical ADO release, demonstrating that HCRT input is critical for ADO signaling in the BF. In contrast, oral ZOL and BF-infused ZOL had no effect on ADO levels in either BF or cortex. ALM increased BF ADO (an endogenous sleep-promoting substance) and GABA (which is increased during normal sleep), and required an intact BF for maximal efficacy, whereas ZOL blocked sleep-associated BF GABA release, and required no functional contribution from the BF to induce sleep. ALM thus induces sleep by facilitating the neural mechanisms underlying the normal transition to sleep.


Subject(s)
Adenosine/metabolism , Basal Forebrain/physiology , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Orexins/antagonists & inhibitors , Sleep/physiology , gamma-Aminobutyric Acid/metabolism , Acetamides/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Basal Forebrain/drug effects , Basal Forebrain/metabolism , Butyric Acid , Cholinergic Neurons/drug effects , Cholinergic Neurons/metabolism , Glutamic Acid , Intracellular Signaling Peptides and Proteins , Isoquinolines/pharmacology , Male , Neuropeptides/metabolism , Orexins/metabolism , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins , Sleep/drug effects , Synaptic Transmission/drug effects , Wakefulness , Zolpidem
4.
Neuropsychopharmacology ; 40(3): 632-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25139062

ABSTRACT

Cortical interneurons, immunoreactive for neuronal nitric oxide synthase (nNOS) and the receptor NK1, express the functional activity marker Fos selectively during sleep. NREM sleep 'pressure' is hypothesized to accumulate during waking and to dissipate during sleep. We reported previously that the proportion of Fos(+) cortical nNOS/NK1 neurons is correlated with established electrophysiological markers of sleep pressure. As these markers covary with the amount of NREM sleep, it remained unclear whether cortical nNOS/NK1 neurons are activated to the same degree throughout NREM sleep or whether the extent of their activation is related to the sleep pressure that accrued during the prior waking period. To distinguish between these possibilities, we used hypnotic medications to control the amount of NREM sleep in rats while we varied prior wake duration and the resultant sleep pressure. Drug administration was preceded by 6 h of sleep deprivation (SD) ('high sleep pressure') or undisturbed conditions ('low sleep pressure'). We find that the proportion of Fos(+) cortical nNOS/NK1 neurons was minimal when sleep pressure was low, irrespective of the amount of time spent in NREM sleep. In contrast, a large proportion of cortical nNOS/NK1 neurons was Fos(+) when an equivalent amount of sleep was preceded by SD. We conclude that, although sleep is necessary for cortical nNOS/NK1 neuron activation, the proportion of cells activated is dependent upon prior wake duration.


Subject(s)
Cerebral Cortex/physiology , Homeostasis/physiology , Neurons/physiology , Nitric Oxide Synthase Type I/physiology , Receptors, Neurokinin-1/physiology , Sleep Stages/physiology , Acetamides/pharmacology , Animals , Cerebral Cortex/drug effects , Isoquinolines/pharmacology , Male , Neurons/immunology , Nitric Oxide Synthase Type I/immunology , Pyridines/pharmacology , Rats , Receptors, Neurokinin-1/immunology , Sleep Deprivation/physiopathology , Sleep Stages/drug effects , Zolpidem
5.
Sleep ; 36(3): 325-36, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23449602

ABSTRACT

STUDY OBJECTIVES: Humans with narcolepsy and orexin/ataxin-3 transgenic (TG) mice exhibit extensive, but incomplete, degeneration of hypo-cretin (Hcrt) neurons. Partial Hcrt cell loss also occurs in Parkinson disease and other neurologic conditions. Whether Hcrt antagonists such as almorexant (ALM) can exert an effect on the Hcrt that remains after Hcrt neurodegeneration has not yet been determined. The current study was designed to evaluate the hypnotic and cataplexy-inducing efficacy of a Hcrt antagonist in an animal model with low Hcrt tone and compare the ALM efficacy profile in the disease model to that produced in wild-type (WT) control animals. DESIGN: Counterbalanced crossover study. SETTING: Home cage. PATIENTS OR PARTICIPANTS: Nine TG mice and 10 WT mice. INTERVENTIONS: ALM (30, 100, 300 mg/kg), vehicle and positive control injections, dark/active phase onset. MEASUREMENTS AND RESULTS: During the 12-h dark period after dosing, ALM exacerbated cataplexy in TG mice and increased nonrapid eye movement sleep with heightened sleep/wake fragmentation in both genotypes. ALM showed greater hypnotic potency in WT mice than in TG mice. The 100 mg/kg dose conferred maximal promotion of cataplexy in TG mice and maximal promotion of REM sleep in WT mice. In TG mice, ALM (30 mg/ kg) paradoxically induced a transient increase in active wakefulness. Core body temperature (Tb) decreased after acute Hcrt receptor blockade, but the reduction in Tb that normally accompanies the wake-to-sleep transition was blunted in TG mice. CONCLUSIONS: These complex dose- and genotype-dependent interactions underscore the importance of effector mechanisms downstream from Hcrt receptors that regulate arousal state. Cataplexy promotion by ALM warrants cautious use of Hcrt antagonists in patient populations with Hcrt neurodegeneration, but may also facilitate the discovery of anticataplectic medications. CITATION: Black SW; Morairty SR; Fisher SP; Chen TM; Warrier DR; Kilduff TS. Almorexant promotes sleep and exacerbates cataplexy in a murine model of narcolepsy. SLEEP 2013;36(3):325-336.


Subject(s)
Acetamides/pharmacology , Cataplexy/chemically induced , Isoquinolines/pharmacology , Narcolepsy/drug therapy , Sleep/drug effects , Analysis of Variance , Animals , Cross-Over Studies , Disease Models, Animal , Electroencephalography/drug effects , Electromyography/drug effects , Intracellular Signaling Peptides and Proteins/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neuropeptides/drug effects , Orexins , Wakefulness/drug effects
6.
Article in English | MEDLINE | ID: mdl-22679419

ABSTRACT

We have previously demonstrated that Type I neuronal nitric oxide synthase (nNOS)-expressing neurons are sleep-active in the cortex of mice, rats, and hamsters. These neurons are known to be GABAergic, to express Neuropeptide Y (NPY) and, in rats, to co-express the Substance P (SP) receptor NK1, suggesting a possible role for SP in sleep/wake regulation. To evaluate the degree of co-expression of nNOS and NK1 in the cortex among mammals, we used double immunofluorescence for nNOS and NK1 and determined the anatomical distribution in mouse, rat, and squirrel monkey cortex. Type I nNOS neurons co-expressed NK1 in all three species although the anatomical distribution within the cortex was species-specific. We then performed in vitro patch clamp recordings in cortical neurons in mouse and rat slices using the SP conjugate tetramethylrhodamine-SP (TMR-SP) to identify NK1-expressing cells and evaluated the effects of SP on these neurons. Bath application of SP (0.03-1 µM) resulted in a sustained increase in firing rate of these neurons; depolarization persisted in the presence of tetrodotoxin. These results suggest a conserved role for SP in the regulation of cortical sleep-active neurons in mammals.

7.
J Neurosci ; 31(15): 5764-76, 2011 Apr 13.
Article in English | MEDLINE | ID: mdl-21490218

ABSTRACT

Down syndrome (DS) is a developmental disorder caused by a third chromosome 21 in humans (Trisomy 21), leading to neurological deficits and cognitive impairment. Studies in mouse models of DS suggest that cognitive deficits in the adult are associated with deficits in synaptic learning and memory mechanisms, but it is unclear whether alterations in the early wiring and refinement of neuronal circuits contribute to these deficits. Here, we show that early developmental refinement of visual circuits is perturbed in mouse models of Down syndrome. Specifically, we find excessive eye-specific segregation of retinal axons in the dorsal lateral geniculate nucleus. Indeed, the degree of refinement scales with defects in the "Down syndrome critical region" (DSCR) in a dose-dependent manner. We further identify Dscam (Down syndrome cell adhesion molecule), a gene within the DSCR, as a regulator of eye-specific segregation of retinogeniculate projections. Although Dscam is not the sole gene in the DSCR contributing to enhanced refinement in trisomy, Dscam dosage clearly regulates cell spacing and dendritic fasciculation in a specific class of retinal ganglion cells. Thus, altered developmental refinement of visual circuits that occurs before sensory experience is likely to contribute to visual impairment in individuals with Down syndrome.


Subject(s)
Down Syndrome/physiopathology , Geniculate Bodies/physiopathology , Retina/physiopathology , Algorithms , Animals , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cell Count , Dendrites/physiology , Dose-Response Relationship, Drug , Fasciculation/physiopathology , Gene Dosage , Immunohistochemistry , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microelectrodes , Neurons, Afferent/physiology , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Retinal Ganglion Cells/physiology , Trisomy/pathology , Visual Pathways/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...