Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.541
Filter
1.
Behav Pharmacol ; 35(4): 239-252, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38567447

ABSTRACT

Rapid-eye movement (REM) sleep deprivation (SD) can induce manic-like behaviors including hyperlocomotion. On the other hand, crocin (one of the main compounds of Crocus sativus L. or Saffron) may be beneficial in the improvement of mental and cognitive dysfunctions. Also, crocin can restore the deleterious effects of SD on mental and cognitive processes. In this study, we investigated the effect of REM SD on female rats' behaviors including depression- and anxiety-like behaviors, locomotion, pain perception, and obsessive-compulsive-like behavior, and also, the potential effect of crocin on REM SD effects. We used female rats because evidence on the role of REM SD in modulating psychological and behavioral functions of female (but not male) rats is limited. REM SD was induced for 14 days (6h/day), and crocin (25, 50, and 75 mg/kg) was injected intraperitoneally. Open field test, forced swim test, hot plate test, and marble burying test were used to assess rats' behaviors. The results showed REM SD-induced manic-like behavior (hyperlocomotion). Also, REM SD rats showed decreased anxiety- and depression-like behavior, pain subthreshold (the duration it takes for the rat to feel pain), and showed obsessive compulsive-like behavior. However, crocin at all doses partially or fully reversed REM SD-induced behavioral changes. In conclusion, our results suggested the possible comorbidity of OCD and REM SD-induced manic-like behavior in female rats or the potential role of REM SD in the etiology of OCD, although more studies are needed. In contrast, crocin can be a possible therapeutic choice for decreasing manic-like behaviors.


Subject(s)
Carotenoids , Crocus , Sleep Deprivation , Animals , Female , Rats , Sleep Deprivation/drug therapy , Sleep Deprivation/complications , Carotenoids/pharmacology , Obsessive-Compulsive Disorder/drug therapy , Anxiety/drug therapy , Behavior, Animal/drug effects , Mania/drug therapy , Depression/drug therapy , Rats, Wistar , Disease Models, Animal , Bipolar Disorder/drug therapy , Sleep, REM/drug effects , Dose-Response Relationship, Drug
2.
Psychopharmacology (Berl) ; 241(6): 1213-1225, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38427059

ABSTRACT

RATIONALE: Prepulse inhibition (PPI) impairment reflects sensorimotor gating problems, i.e. in schizophrenia. This study aims to enlighten the role of orexinergic regulation on PPI in a psychosis-like model. OBJECTIVES: In order to understand the impact of orexinergic innervation on PPI and how it is modulated by age and baseline PPI (bPPI), chronic orexin A (OXA) injections was carried on non-sleep-deprived and sleep-deprived rats that are grouped by their bPPI. METHODS: bPPI measurements were carried on male Wistar rats on P45 or P90 followed by grouping into low-PPI and high-PPI rats. The rats were injected with OXA twice per day for four consecutive days starting on P49 or P94, while the control groups received saline injections. 72 h REMSD was carried on via modified multiple platform technique on P94 and either OXA or saline was injected during REMSD. PPI tests were carried out 30 min. after the last injection. RESULTS: Our previous study with acute OXA injection after REMSD without bPPI grouping revealed that low OXA doses might improve REMSD-induced PPI impairment. Our current results present three important conclusions: (1) The effect of OXA on PPI is bPPI-dependent and age-dependent. (2) The effect of REMSD is bPPI-dependent. (3) The effect of OXA on PPI after REMSD also depends on bPPI. CONCLUSION: Orexinergic regulation of PPI response with and without REMSD can be predicted by bPPI levels. Our findings provide potential insights into the regulation of sensorimotor gating by sleep/wakefulness systems and present potential therapeutic targets for the disorders, where PPI is disturbed.


Subject(s)
Orexins , Prepulse Inhibition , Rats, Wistar , Sleep Deprivation , Animals , Orexins/pharmacology , Orexins/administration & dosage , Orexins/metabolism , Male , Sleep Deprivation/physiopathology , Rats , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Sleep, REM/drug effects , Sensory Gating/drug effects , Age Factors , Disease Models, Animal
3.
Food Funct ; 13(24): 12697-12706, 2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36408594

ABSTRACT

Turmeric (Curcuma longa) had been considered as a universal panacea in functional foods and traditional medicines. In recent, the sedative-hypnotic effect of turmeric extract (TE) was reported. However, sleep-promoting compounds in TE have been not yet demonstrated. Curcuminoids (curcumin, demethoxycurcumin, and bisdemethoxycurcumin) are the major constituents of turmeric being responsible for its various biological activities. Therefore, they can be first assumed to be sedative-hypnotic compounds of TE. In the present study, we aimed to investigate the effects and underlying mechanisms of curcuminoids and each constituent on the sleep-wake cycle of mice. Molecular docking studies, histamine H1 receptor (H1R) binding assays, and H1R knockout animal studies were used to investigate the molecular mechanisms underlying the sleep-promoting effects. Curcuminoids and their constituents reduced sleep latency and increased sleep duration in the pentobarbital-induced sleep test in mice. In addition, curcuminoids significantly increased the duration of NREMS and reduced sleep latency without altering the REMS and delta activity. Curcumin, demethoxycurcumin, and bisdemethoxycurcumin were predicted to interact with H1R in the molecular model. In the binding affinity assay, we found that curcuminoids, as well as their constituents, significantly bind to H1R with the Ki value of 1.49 µg mL-1. Furthermore, sleep latency was reduced and NREMS frequency was increased following curcuminoid administration in wild-type mice but not in H1R knockout mice. Therefore, we conclude that curcuminoids reduce sleep latency and enhance the quantity of NREMS by acting as modulators of H1R, indicating their usefulness in treating insomnia.


Subject(s)
Curcuma , Curcumin , Diarylheptanoids , Receptors, Histamine H1 , Sleep Aids, Pharmaceutical , Sleep Latency , Sleep, REM , Animals , Mice , Curcuma/chemistry , Curcumin/chemistry , Curcumin/pharmacology , Diarylheptanoids/pharmacology , Molecular Docking Simulation , Plant Extracts/chemistry , Plant Extracts/pharmacology , Receptors, Histamine H1/genetics , Receptors, Histamine H1/metabolism , Sleep Latency/drug effects , Sleep, REM/drug effects , Sleep Aids, Pharmaceutical/chemistry , Sleep Aids, Pharmaceutical/pharmacology
4.
Biochem Biophys Res Commun ; 632: 10-16, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36191372

ABSTRACT

Paradoxical sleep deprivation (PSD) is prevalent in modern society, and impaired memory is one of its serious consequences. The pathogenic mechanism is still unclear, and the therapeutic strategies for PSD are limited. Here, we found that quercetin treatment ameliorated memory impairments caused by PSD in a dose-dependent manner in an animal model. Quercetin could restore the dynamic changes of the gamma band while the animals performed novel object recognition (NOR) tasks as determined by electroencephalogram analysis. Morphological analysis showed that quercetin, by targeting the hippocampal CA1 region, strikingly ameliorated the overactivation of microglia induced by PSD. Mechanistically, quercetin inhibited the toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and nuclear factor kappa-b (NF-κB) cascade, which is critical for abnormal microglial activation following PSD stress. Our results provided experimental evidence for the therapeutic effects of quercetin on PSD-related memory impairments by suppressing TLR4/MyD88/NF-κB signaling that mediated abnormal microglia activation in the hippocampus.


Subject(s)
Memory Disorders , Microglia , Quercetin , Animals , Mice , Disease Models, Animal , Memory Disorders/drug therapy , Memory Disorders/etiology , Microglia/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Quercetin/pharmacology , Quercetin/therapeutic use , Sleep, REM/drug effects , Toll-Like Receptor 4/metabolism
5.
PLoS One ; 17(2): e0264386, 2022.
Article in English | MEDLINE | ID: mdl-35213655

ABSTRACT

Both chronic pain and sleep disorders are associated with a reduction in the quality of life. They can be both a cause and a consequence of each other, and should therefore be simultaneously treated. However, optimal treatments for chronic pain-related sleep disorders are not well established. Here, we aimed to investigate the effects of suvorexant, a novel sleep drug, and mirtazapine, a noradrenergic and specific serotonergic antidepressant, on pain-related changes in sleep parameters in a preclinical chronic pain mice model, by partial sciatic nerve ligation. We evaluated the quantity, duration, and depth of sleep by analyzing the electroencephalogram and voluntary activity by counting the number of wheel rotations to determine various symptoms of sleep disorders, including reduced total sleep time, fragmentation, low quality, and impaired activity in the daytime. Suvorexant and mirtazapine normalized the reduction in sleep time and fragmented sleep, further regaining the sleep depth at sleep onset in the chronic pain state in nerve-ligated mice. Mirtazapine also increased the percentage of rapid eye movement sleep in mice. Suvorexant decreased voluntary activity, which was prolonged after administration; however, mirtazapine did not decrease it. Although the effects of suvorexant and mirtazapine on sleep and activity are different, both suvorexant and mirtazapine could be potential therapeutic agents for chronic pain-related sleep disorders.


Subject(s)
Azepines/pharmacology , Mirtazapine/pharmacology , Sciatic Nerve , Sleep, REM/drug effects , Triazoles/pharmacology , Animals , Chronic Pain/drug therapy , Chronic Pain/physiopathology , Male , Mice , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology
6.
Sci Rep ; 12(1): 2117, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136089

ABSTRACT

Previous spectral analysis studies on insomnia have shown inconsistent results due to their heterogeneity and small sample sizes. We compared the difference of electroencephalogram (EEG) spectral power during sleep among participants without insomnia, insomniacs with no hypnotic use, hypnotic users with no insomnia complaints, and hypnotic users with insomnia complaints using the Sleep Heart Health Study data, which is large sample size and has good quality control. The fast Fourier transformation was used to calculate the EEG power spectrum for total sleep duration within contiguous 30-s epochs of sleep. For 1985 participants, EEG spectral power was compared among the groups while adjusting for potential confounding factors that could affect sleep EEG. The power spectra during total sleep differed significantly among the groups in all frequency bands (pcorr < 0.001). We found that quantitative EEG spectral power in the beta and sigma bands of total sleep differed (pcorr < 0.001) between participants without insomnia and hypnotic users with insomnia complaints after controlling for potential confounders. The higher beta and sigma power were found in the hypnotic users with insomnia complaints than in the non-insomnia participants. This study suggests differences in the microstructures of polysomnography-derived sleep EEG between the two groups.


Subject(s)
Brain Waves/drug effects , Polysomnography , Sleep Aids, Pharmaceutical/therapeutic use , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep, REM/drug effects , Adult , Aged , Case-Control Studies , Cohort Studies , Female , Humans , Male , Middle Aged , Sleep Aids, Pharmaceutical/pharmacology
7.
Neuropharmacology ; 206: 108940, 2022 03 15.
Article in English | MEDLINE | ID: mdl-34982973

ABSTRACT

The interaction among the acetylcholine (ACh)-ergic REM-ON neurons in the pedunculo-pontine area (PPT), noradrenergic REM-OFF neurons in locus coeruleus (LC) and GABA-ergic neurons in the regulation of rapid eye movement sleep (REMS) have been studied in relative details; however, many questions including the role of dopamine (DA) remain unanswered. The ventral tegmental area (VTA) is rich in DA-ergic neurons, which have been implicated with schizophrenia and depression, when REMS is significantly affected. Also, some of the symptoms of REMS and these diseases are common. As the ACh-ergic REM-ON neurons in the PPT project to VTA, we proposed that such inputs might affect REMS, dreams and hallucinations. We recorded sleep-wake-REMS in freely moving, chronically prepared rats under three controlled experimental conditions. In different sets of experiments, either the ACh-ergic inputs to the VTA were blocked by local microinjection of Scopolamine (Scop) alone, or, the PPT neurons were bilaterally stimulated by Glutamate (Glut), or, the PPT neurons were stimulated by Glut in presence of Scop into the VTA. It was observed that Glut into PPT and Scop into the VTA significantly increased and decreased REMS, respectively. Additionally, PPT stimulation induced increased REMS was prevented in the presence of Scop into the VTA. Based on these findings we propose that inputs from ACh-ergic REM-ON neurons to VTA increase REMS and it could be a possible circuitry for expressions of hallucinations and dreams.


Subject(s)
Cholinergic Neurons/physiology , Dopaminergic Neurons/physiology , Pontine Tegmentum/physiology , Sleep, REM/physiology , Ventral Tegmental Area/physiology , Animals , Cholinergic Antagonists/pharmacology , Rats , Scopolamine/pharmacology , Sleep, REM/drug effects , Ventral Tegmental Area/drug effects
8.
J Ethnopharmacol ; 285: 114866, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34822960

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Jiaotaiwan (JTW) is good at communicating the heart and kidney. That meets the main mechanism of insomnia in traditional Chinese medicine. But the mechanism of JTW in promoting sleep is not clear. AIM OF THE STUDY: To investigate the mechanism of JTW in promoting sleep and identify the main components. MATERIALS AND METHODS: In this study, we detected the levels of GABA in serum and brain via LC-MS/MS analysis and investigated the hypnotic effect of JTW and its role in promoting sleep via Sleep monitoring and vigilance state analysis. Further, the identification of the main components was carried out by using LC-MS/MS analysis. RESULTS: JTW could increase the GABA levels in serum, FC and BS of SDM rats. JTW reduced the amount of wakefulness, increased the time of NREM sleep and REM sleep. A total of 25 compounds were identified. CONCLUSIONS: The current work provides valuable information on the hypnotic effects of JTW and its regulatory mechanisms in promoting sleep.


Subject(s)
Brain/drug effects , Drugs, Chinese Herbal , Sleep, REM/drug effects , Sleep/drug effects , gamma-Aminobutyric Acid/blood , gamma-Aminobutyric Acid/metabolism , Animals , Brain/metabolism , Male , Mice , Rats , Rats, Sprague-Dawley , Sleep Initiation and Maintenance Disorders/drug therapy
9.
Neurosci Lett ; 765: 136247, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34530113

ABSTRACT

Accumulating evidence has shown that sleep disturbance is a common symptom in Alzheimer's disease (AD), which is regarded as a modifiable risk factor for AD. Orexin is a key modulator of the sleep-wake cycle and has been found to be dysregulated in AD patients. The increased orexin in cerebrospinal fluid (CSF) is associated with decreased sleep efficiency and REM sleep, as well as cognitive impairment in AD patients. The orexin system has profuse projections to brain regions that are implicated in arousal and cognition and has been found to participate in the progression of AD pathology. Conversely the orexin receptor antagonists are able to consolidate sleep and reduce AD pathology. Therefore, improved understanding of the mechanisms linking orexin system, sleep disturbance and AD could make orexin receptor antagonists a promising target for the prevention or treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Orexin Receptor Antagonists/therapeutic use , Orexins/metabolism , Sleep Wake Disorders/etiology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Cognition/drug effects , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Disease Models, Animal , Humans , Orexin Receptor Antagonists/pharmacology , Orexins/antagonists & inhibitors , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/pathology , Sleep, REM/drug effects
10.
Sci Rep ; 11(1): 16713, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34408180

ABSTRACT

The effects of chronic antidepressant (AD) treatment on sleep disturbances in rodent chronic stress models have not been thoroughly investigated. Here, we show that chronic social defeat stress (SDS) in rats induces prolonged social avoidance, alterations in sleep architecture (increased total rapid eye movement [REM] sleep duration, bout, and shortened REM latency), and contextual but not cued fear memory deficits, even 1 month after the last SDS. These abnormalities were associated with changes in electroencephalography (EEG) spectral powers, including reduced REM sleep theta power during the light phase. Chronic treatment with two different classes of antidepressants (ADs), imipramine and fluoxetine, significantly ameliorated these behavioral, sleep, and EEG abnormalities. Interestingly, REM theta power was normalized by chronic (1 month) but not 1 week AD administration and solely correlated with the ratio (an objective indicator) of social interaction 1 month after the last SDS. These data suggest that reductions in REM sleep theta power, an EEG parameter that has never been directly investigated in humans, is a core sleep symptom in socially defeated rats, and, potentially, also in patients with stress-related psychiatric disorders, including major depressive and posttraumatic stress disorders.


Subject(s)
Antidepressive Agents/adverse effects , Fluoxetine/adverse effects , Imipramine/adverse effects , Sleep, REM/drug effects , Stress, Psychological/physiopathology , Theta Rhythm/drug effects , Animals , Antidepressive Agents/pharmacology , Chronic Disease , Electroencephalography , Fluoxetine/pharmacology , Humans , Imipramine/pharmacology , Male , Rats
11.
Sci Rep ; 11(1): 16313, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381098

ABSTRACT

The first night effect (FNE) is a type of sleep disturbance caused by an unfamiliar environment, which leads to difficulty falling asleep and reduced sleep duration. Previously, we reported that Lactobacillus fermentum PS150 (PS150) improves sleep conditions in a pentobarbital-induced sleep mouse model. In this study, we aimed to evaluate the effect of PS150 on the FNE in mice. Briefly, mice were implanted with electrodes and orally administered PS150 for four weeks, and then the FNE was induced by cage changing. Analysis of polysomnographic signals revealed that intervention with PS150 restored non-rapid eye movement (NREM) sleep length under the FNE. Compared to diphenhydramine, a commonly used sleep aid, PS150 had no unwanted side effects, such as rapid eye movement (REM) sleep deprivation and fragmented sleep. Moreover, temporal analysis revealed that PS150 efficiently reduced both sleep latency and time spent restoring normal levels of REM sleep. Taken together, these results suggest that PS150 efficiently ameliorates sleep disturbance caused by the FNE. Additionally, V3-V4 16S rRNA sequencing revealed significant increases in Erysipelotrichia, Actinobacteria, and Coriobacteriia in fecal specimens of the PS150-treated group, indicating that PS150 induces gut microbiota remodeling.


Subject(s)
Limosilactobacillus fermentum/physiology , Sleep, REM/physiology , Animals , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Male , Mice , Mice, Inbred C57BL , Pentobarbital/pharmacology , Polysomnography/methods , RNA, Ribosomal, 16S/genetics , Sleep Deprivation/chemically induced , Sleep Deprivation/microbiology , Sleep Deprivation/physiopathology , Sleep Initiation and Maintenance Disorders/chemically induced , Sleep Initiation and Maintenance Disorders/microbiology , Sleep Initiation and Maintenance Disorders/physiopathology , Sleep Wake Disorders/chemically induced , Sleep Wake Disorders/microbiology , Sleep Wake Disorders/physiopathology , Sleep, REM/drug effects
12.
Biol Pharm Bull ; 44(6): 789-797, 2021.
Article in English | MEDLINE | ID: mdl-34078810

ABSTRACT

Sleep curtailment negatively affects cardiac activities and thus should be ameliorated by pharmacological methods. One of the therapeutic targets is melatonin receptors, which tune circadian rhythms. Ramelteon, a melatonin MT1/MT2 receptor agonist, has recently been developed to modulate sleep-wake rhythms. To date, the sleep-promoting effect of ramelteon has been widely delineated, but whether ramelteon treatment physiologically influences cardiac function is not well understood. To address this question, we recorded electrocardiograms, electromyograms, and electrocorticograms in the frontal cortex and the olfactory bulb of unrestrained rats treated with either ramelteon or vehicle. We detected vigilance states based on physiological measurements and analyzed cardiac and muscular activities. We found that during non-rapid eye movement (non-REM) sleep, heartrate variability was maintained by ramelteon treatment. Analysis of the electromyograms confirmed that neither microarousal during non-REM sleep nor the occupancy of phasic periods during REM sleep was altered by ramelteon. Our results indicate that ramelteon has a remedial effect on cardiac activity by keeping the heartrate variability and may reduce cardiac dysfunction during sleep.


Subject(s)
Heart Rate/drug effects , Indenes/pharmacology , Sleep, REM/drug effects , Animals , Electrocardiography , Electrocorticography , Electromyography , Frontal Lobe/drug effects , Frontal Lobe/physiology , Male , Olfactory Bulb/drug effects , Olfactory Bulb/physiology , Rats, Wistar , Sleep, REM/physiology
13.
Clin Pharmacol Ther ; 110(3): 808-815, 2021 09.
Article in English | MEDLINE | ID: mdl-33961287

ABSTRACT

In contrast to the dose-occupancy relationship in the treatment of schizophrenia, the minimal effective level of dopamine receptor 2 (D2R) blockade for antipsychotics in the treatment of bipolar depression is unknown. Lower doses aimed at reducing extrapyramidal side effects must be balanced against the need to retain the therapeutic benefit of D2R blockade on emergent cycling, mixed, manic, anxiety, and/or psychotic symptoms. Dose-reductions intended to lower D2R blockade, however, could also decrease concomitant serotonin receptor antagonism and its potential benefit on depressive symptoms. Here, we uncoupled the potential antidepressant activity in amisulpride, driven by 5-HT7 receptor (5-HT7R) antagonism, from the D2R-mediated antipsychotic activity by discovering that each enantiomer favors a different receptor. Aramisulpride was more potent at 5-HT7R relative to esamisulpride (Ki 47 vs. 1,900 nM, respectively), whereas esamisulpride was more potent at D2R (4.0 vs. 140 nM). We hypothesized that a nonracemic ratio might achieve greater 5-HT7R-mediated antidepressant effects at a lower level of D2R blockade. The dose-occupancy relationship of esamisulpride at D2R was determined by positron emission tomography (PET) imaging in human volunteers. Separately the dose-relationship of aramisulpride was established in humans using suppression of rapid eye movement (REM) sleep as a marker of 5-HT7R antagonism. These results led to the discovery of an 85:15 ratio of aramisulpride to esamisulpride (SEP-4199) that maximizes the potential for antidepressant benefit of aramisulpride via 5-HT7R and reduces esamisulpride to minimize D2R-related extrapyramidal side effects while still retaining D2R-mediated effects predicted to provide benefit in bipolar depression. The antidepressant efficacy of SEP-4199 was recently confirmed in a proof-of-concept trial for the treatment of bipolar depression (NCT03543410).


Subject(s)
Amisulpride/adverse effects , Amisulpride/therapeutic use , Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Dopamine D2 Receptor Antagonists/therapeutic use , Mood Disorders/drug therapy , Receptors, Serotonin/metabolism , Adult , Animals , Antidepressive Agents/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Female , Humans , Male , Mood Disorders/metabolism , Positron-Emission Tomography/methods , Rats , Rats, Wistar , Schizophrenia/drug therapy , Schizophrenia/metabolism , Sleep, REM/drug effects
14.
Neuropharmacology ; 193: 108607, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34023337

ABSTRACT

Dreams appear intermittently during phasic rapid eye movement sleep (REMS). Although reasonable progress has been made about neuro-physio-pharmacological mechanism of appearance of REMS, appearance of dreams is a mystery. Isolated studies have reported that substantia nigra (SN) withdraws inhibition from pedunculo-pontine tegmentum (PPT) acetylcholine (ACh)-ergic REM-ON neurons to trigger REMS; some REM-ON neurons become phasically active during REMS; amygdala (Amyg), a limbic structure associated with emotions, may be related with dreaming like state; Amyg receives projections from both SN-Dopamine (DA)-ergic and PPT-ACh-ergic neurons. Collating these isolated findings, we proposed that on the background of REMS, SN-DA-ergic and PPT-ACh-ergic inputs phasically activate Amyg-neurons to manifest dreams. In the absence of better criteria, we recorded electrophysiological characteristics of REMS as the closest objective read-out for dreams in surgically prepared, chronic, freely moving rats. Microinjection of either DA-ergic or ACh-ergic agonist [Quinpirole (Qnp) or Carbachol (Carb)] bilaterally into Amyg increased, while antagonists [Haloperidol (Hal) or Scopolamine (Scop)] reduced REMS. Electrical stimulation of either bilateral SN or PPT increased REMS, which however, was prevented when stimulated in presence of Hal or Scop, respectively into the Amyg. These findings confirm and support our contention that SN-DA-ergic and PPT-ACh-ergic inputs integrate in Amyg for REMS regulation. Further, subject to confirmation in humans, we propose that on the background of REMS, some phasic PPT-ACh-ergic-REM-ON neurons intermittently trigger some neurons in Amyg, the area known to be associated with memory and emotions, causing intermittent appearance of REMS-associated dreams and in REMS behavior disorder.


Subject(s)
Amygdala/physiology , Dopaminergic Neurons/physiology , Pontine Tegmentum/physiology , Sleep, REM/physiology , Substantia Nigra/physiology , Wakefulness/physiology , Amygdala/drug effects , Animals , Carbachol/pharmacology , Cholinergic Neurons , Electric Stimulation , Haloperidol/pharmacology , Male , Pontine Tegmentum/drug effects , Quinpirole/pharmacology , Rats , Rats, Wistar , Scopolamine/pharmacology , Sleep, REM/drug effects , Substantia Nigra/drug effects , Wakefulness/drug effects
15.
Life Sci ; 273: 119303, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33667518

ABSTRACT

AIM: The current study aims to investigate the impact of paradoxical (REM) sleep deprivation and/or epileptic seizures on rat's cortical brain tissues. MAIN METHODS: Animals were divided into four groups; control, epileptic, REM sleep deprived and epileptic subjected to REM sleep deprivation. Electrocorticogram (ECoG) signals were recorded and quantitatively analyzed for each group. Concentrations of amino acid neurotransmitters; proinflammatory cytokines; and oxidative stress parameters; and acetylcholinesterase activity were determined in the cortex of the animals in different groups. KEY FINDINGS: Results showed significant variations in the spectral distribution of ECoG waves in the epilepsy model, 24- and 48-hours of REM sleep deprivation and their combined effects indicating a state of cortical hyperexcitability. Significant increases in NO and taurine and significant decrement in glutamine, GABA and glycine were determined. In REM sleep deprived rats significant elevation in glutamate, aspartate, glycine and taurine and a significant lowering in GABA were obtained. This was accompanied by significant reduction in AchE and IL-ß. In the cortical tissue of epileptic rats deprived from REM sleep significant increases in lipid peroxidation, TNF-α, IL-1ß, IL-6 and aspartate and a significant reduction in AchE were observed. SIGNIFICANCE: The present data indicate that REM sleep deprivation induces an increase in lipid peroxidation and storming in proinflammatory cytokines in the cortex of rat model of epilepsy during SRS. These changes are associated with a decreased seizure threshold as inferred from the increase in alpha and Beta waves and a decrease in Delta waves of ECoG.


Subject(s)
Brain/pathology , Neurotransmitter Agents/toxicity , Oxidative Stress/drug effects , Seizures/complications , Sleep Deprivation/complications , Sleep, REM/drug effects , Animals , Brain/drug effects , Electrophysiology , Lipid Peroxidation , Male , Rats , Rats, Wistar
16.
Psychopharmacology (Berl) ; 238(6): 1437-1447, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33635384

ABSTRACT

RATIONALE: The medical uses of cannabidiol (CBD), a constituent of the Cannabis sativa, have accelerated the legal and social acceptance for CBD-based medications but has also given the momentum for questioning whether the long-term use of CBD during the early years of life may induce adverse neurobiological effects in adulthood, including sleep disturbances. Given the critical window for neuroplasticity and neuro-functional changes that occur during stages of adolescence, we hypothesized that CBD might influence the sleep-wake cycle in adult rats after their exposure to CBD during the adolescence. OBJECTIVES: Here, we investigated the effects upon behavior and neural activity in adulthood after long-term administrations of CBD in juvenile rats. METHODS: We pre-treated juvenile rats with CBD (5 or 30 mg/Kg, daily) from post-natal day (PND) 30 and during 2 weeks. Following the treatments, the sleep-wake cycle and NeuN expression was analyzed at PND 80. RESULTS: We found that systemic injections of CBD (5 or 30 mg/Kg, i.p.) given to adolescent rats (post-natal day 30) for 14 days increased in adulthood the wakefulness and decreased rapid eye movement sleep during the lights-on period whereas across the lights-off period, wakefulness was diminished and slow wave sleep was enhanced. In addition, we found that adult animals that received CBD during the adolescence displayed disruptions in sleep rebound period after total sleep deprivation. Finally, we determined how the chronic administrations of CBD during the adolescence affected in the adulthood the NeuN expression in the suprachiasmatic nucleus, a sleep-related brain region. CONCLUSIONS: Our findings are relevant for interpreting results of adult rats that were chronically exposed to CBD during the adolescence and provide new insights into how CBD may impact the sleep-wake cycle and neuronal activity during developmental stages.


Subject(s)
Cannabidiol/administration & dosage , Sleep Wake Disorders/chemically induced , Sleep/drug effects , Wakefulness/drug effects , Animals , Brain/drug effects , Cannabis/chemistry , Male , Neurons/drug effects , Rats , Rats, Wistar , Sleep Deprivation , Sleep, REM/drug effects
17.
Pharmacol Biochem Behav ; 203: 173153, 2021 04.
Article in English | MEDLINE | ID: mdl-33582097

ABSTRACT

The majority of the literature describing the relation of sleep/alertness disturbance and substance use disorders (SUD) has focused on the disruptive effects of substances with abuse liability on sleep and alertness. Rarely have studies or literature reviews assessed or discussed how sleep/alertness disturbance affects substance use. This paper focuses on the sleep/alertness disturbance side of the relation. We argue that the relation is bi-directional and review evidence showing that sleep/alertness disturbance affects all phases of the addiction cycle, including the initiation, maintenance and relapse of SUD. We review a variety of substances across all phases of the addiction cycle and conclude sleep/alertness disturbance is a critical factor in both understanding and treating SUD.


Subject(s)
Sleep Wake Disorders/complications , Sleep, REM/drug effects , Sleep, Slow-Wave/drug effects , Substance-Related Disorders/complications , Adolescent , Adult , Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Female , Humans , Male , Recurrence , Sleep Wake Disorders/metabolism , Substance-Related Disorders/metabolism , Young Adult
18.
Neuroimage ; 231: 117850, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33582277

ABSTRACT

Consciousness is a mental characteristic of the human mind, whose exact neural features remain unclear. We aimed to identify the critical nodes within the brain's global functional network that support consciousness. To that end, we collected a large fMRI resting state dataset with subjects in at least one of the following three consciousness states: preserved (including the healthy awake state, and patients with a brain injury history (BI) that is fully conscious), reduced (including the N1-sleep state, and minimally conscious state), and lost (including the N3-sleep state, anesthesia, and unresponsive wakefulness state). We also included a unique dataset of subjects in rapid eye movement sleep state (REM-sleep) to test for the presence of consciousness with minimum movements and sensory input. To identify critical nodes, i.e., hubs, within the brain's global functional network, we used a graph-theoretical measure of degree centrality conjoined with ROI-based functional connectivity. Using these methods, we identified various higher-order sensory and motor regions including the supplementary motor area, bilateral supramarginal gyrus (part of inferior parietal lobule), supragenual/dorsal anterior cingulate cortex, and left middle temporal gyrus, that could be important hubs whose degree centrality was significantly reduced when consciousness was reduced or absent. Additionally, we identified a sensorimotor circuit, in which the functional connectivity among these regions was significantly correlated with levels of consciousness across the different groups, and remained present in the REM-sleep group. Taken together, we demonstrated that regions forming a higher-order sensorimotor integration circuit are involved in supporting consciousness within the brain's global functional network. That offers novel and more mechanism-guided treatment targets for disorders of consciousness.


Subject(s)
Anesthesia/methods , Consciousness/physiology , Nerve Net/physiology , Sensorimotor Cortex/physiology , Sleep, REM/physiology , Wakefulness/physiology , Adult , Anesthetics, Intravenous/administration & dosage , Consciousness/drug effects , Electroencephalography/methods , Female , Humans , Magnetic Resonance Imaging/methods , Male , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Sensorimotor Cortex/diagnostic imaging , Sensorimotor Cortex/drug effects , Sleep, REM/drug effects , Wakefulness/drug effects , Young Adult
19.
Tijdschr Psychiatr ; 63(1): 70-73, 2021.
Article in Dutch | MEDLINE | ID: mdl-33537977

ABSTRACT

A 61-year-old woman with suspected schizophrenia has been attending an outpatient geriatrics service for some time, initially with memory complaints and panic attacks. During treatment, the diagnosis schizophrenia was rejected and psychopharmaceuticals were largely phased out, which improved cognitive functions. Eventually, flashbacks of incest experienced in childhood remained together with REM sleep pathology. The flashbacks, nightmares and the REM sleep pathology were responsive to rivastigmine. Rivastigmine use for the treatment of REM sleep pathology is known in the literature, but it has never been described previously that rivastigmine also impacts on flashbacks and nightmares..


Subject(s)
Dreams/drug effects , Hallucinations/drug therapy , Incest/psychology , Rivastigmine/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep, REM/drug effects , Female , Hallucinations/psychology , Humans , Middle Aged , Panic Disorder/etiology , Panic Disorder/psychology , Sleep, REM/physiology , Treatment Outcome
20.
Psychopharmacology (Berl) ; 238(3): 867-876, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33433644

ABSTRACT

RATIONALE: Previous research suggests that sleep polysomnography and EEG endpoints can be used to assess GABAergic activity; however, the impact of GABAB receptor positive allosteric modulators on sleep endpoints remains unclear. OBJECTIVES: This phase 1 study compared a single dose of ASP8062 (35 mg or 70 mg), a GABAB receptor positive allosteric modulator, with placebo and paroxetine (40 mg). METHODS: Healthy adult volunteers were randomized to four treatments (35 mg ASP8062, 70 mg ASP8062, paroxetine 40 mg, or matching placebo), each separated by a 14-day washout. Primary endpoints obtained by polysomnography were time in stage N3 or SWS and time in rapid eye movement (REM) sleep. Secondary endpoints included impact on sleep stages and electroencephalography parameters, pharmacokinetics, nighttime growth hormone (GH), and safety/tolerability. RESULTS: In 20 randomized volunteers, ASP8062 led to a significant and seemingly dose-dependent increase in SWS over the entire night; this increase was mainly observed during the first third of the night. ASP8062 did not impact time in REM sleep. Paroxetine had no effect on SWS but produced a significant reduction in time spent in REM sleep. A dose-dependent trend in increased GH release was also observed with ASP8062. Headache and nausea were the most commonly reported treatment-emergent adverse events (TEAEs) for ASP8062; most TEAEs were mild in severity. CONCLUSIONS: Single-dose ASP8062 (35 and 70 mg) appeared to result in CNS penetration and enhanced GABAergic activity as measured by increases in slow-wave sleep and growth hormone release.


Subject(s)
GABA Modulators/therapeutic use , Morpholines/therapeutic use , Polysomnography/drug effects , Pyrimidines/therapeutic use , Receptors, GABA-B/metabolism , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep, REM/drug effects , Sleep, Slow-Wave/drug effects , Adult , Electroencephalography/drug effects , Female , GABA Modulators/administration & dosage , GABA Modulators/adverse effects , GABA Modulators/pharmacokinetics , Humans , Male , Middle Aged , Morpholines/administration & dosage , Morpholines/adverse effects , Morpholines/pharmacokinetics , Paroxetine/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Sleep Initiation and Maintenance Disorders/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...