Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.617
Filter
Add more filters

Publication year range
1.
Int J Neuropsychopharmacol ; 27(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38875132

ABSTRACT

BACKGROUND: A compelling hypothesis about attention-deficit/hyperactivity disorder (ADHD) etiopathogenesis is that the ADHD phenotype reflects a delay in cortical maturation. Slow-wave activity (SWA) of non-rapid eye movement (NREM) sleep electroencephalogram (EEG) is an electrophysiological index of sleep intensity reflecting cortical maturation. Available data on ADHD and SWA are conflicting, and developmental differences, or the effect of pharmacological treatment, are relatively unknown. METHODS: We examined, in samples (Mage = 16.4, SD = 1.2), of ever-medicated adolescents at risk for ADHD (n = 18; 72% boys), medication-naïve adolescents at risk for ADHD (n = 15, 67% boys), and adolescents not at risk for ADHD (n = 31, 61% boys) matched for chronological age and controlling for non-ADHD pharmacotherapy, whether ADHD pharmacotherapy modulates the association between NREM SWA and ADHD risk in home sleep. RESULTS: Findings indicated medication-naïve adolescents at risk for ADHD exhibited greater first sleep cycle and entire night NREM SWA than both ever-medicated adolescents at risk for ADHD and adolescents not at risk for ADHD and no difference between ever-medicated, at-risk adolescents, and not at-risk adolescents. CONCLUSIONS: Results support atypical cortical maturation in medication-naïve adolescents at risk for ADHD that appears to be normalized by ADHD pharmacotherapy in ever-medicated adolescents at risk for ADHD. Greater NREM SWA may reflect a compensatory mechanism in middle-later adolescents at risk for ADHD that normalizes an earlier occurring developmental delay.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Electroencephalography , Humans , Attention Deficit Disorder with Hyperactivity/physiopathology , Attention Deficit Disorder with Hyperactivity/drug therapy , Adolescent , Male , Female , Sleep, Slow-Wave/physiology , Sleep, Slow-Wave/drug effects , Central Nervous System Stimulants/pharmacology , Sleep Stages/drug effects , Sleep Stages/physiology
2.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34903651

ABSTRACT

We provide evidence that human sleep is a competitive arena in which cognitive domains vie for limited resources. Using pharmacology and effective connectivity analysis, we demonstrate that long-term memory and working memory are served by distinct offline neural mechanisms that are mutually antagonistic. Specifically, we administered zolpidem to increase central sigma activity and demonstrated targeted suppression of autonomic vagal activity. With effective connectivity, we determined the central activity has greater causal influence over autonomic activity, and the magnitude of this influence during sleep produced a behavioral trade-off between offline long-term and working memory processing. These findings suggest a sleep switch mechanism that toggles between central sigma-dependent long-term memory and autonomic vagal-dependent working memory processing.


Subject(s)
Memory, Long-Term/physiology , Memory, Short-Term/physiology , Sleep/physiology , Adult , Autonomic Nervous System/drug effects , Autonomic Nervous System/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Female , Hippocampus/drug effects , Hippocampus/physiology , Humans , Male , Memory Consolidation/drug effects , Memory Consolidation/physiology , Memory, Long-Term/drug effects , Memory, Short-Term/drug effects , Models, Neurological , Neural Pathways , Sleep/drug effects , Sleep Stages/drug effects , Sleep Stages/physiology , Zolpidem/pharmacology
3.
J Neurophysiol ; 126(4): 1265-1275, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34469699

ABSTRACT

The electroencephalogram (EEG) provides an objective, neural correlate of consciousness. Opioid receptors modulate mammalian neuronal excitability, and this fact was used to characterize how opioids administered to mice alter EEG power and states of consciousness. The present study tested the hypothesis that antinociceptive doses of fentanyl, morphine, or buprenorphine differentially alter the EEG and states of sleep and wakefulness in adult, male C57BL/6J mice. Mice were anesthetized and implanted with telemeters that enabled wireless recordings of cortical EEG and electromyogram (EMG). After surgical recovery, EEG and EMG were used to objectively score states of consciousness as wakefulness, rapid eye movement (REM) sleep, or non-REM (NREM) sleep. Measures of EEG power (dB) were quantified as δ (0.5-4 Hz), θ (4-8 Hz), α (8-13 Hz), σ (12-15 Hz), ß (13-30 Hz), and γ (30-60 Hz). Compared with saline (control), fentanyl and morphine decreased NREM sleep, morphine eliminated REM sleep, and buprenorphine eliminated NREM sleep and REM sleep. Opioids significantly and differentially disrupted the temporal organization of sleep/wake states, altered specific EEG frequency bands, and caused dissociated states of consciousness. The results are discussed relative to the fact that opioids, pain, and sleep modulate interacting states of consciousness.NEW & NOTEWORTHY This study discovered that antinociceptive doses of fentanyl, morphine, and buprenorphine significantly and differentially disrupt EEG-defined states of consciousness in C57BL/6J mice. These data are noteworthy because: 1) buprenorphine is commonly used in medication-assisted therapy for opioid addiction, and 2) there is evidence that disordered sleep can promote addiction relapse. The results contribute to community phenotyping efforts by making publicly available all descriptive and inferential statistics from this study (Supplemental Tables S1-S8).


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics/pharmacology , Brain Waves/drug effects , Buprenorphine/pharmacology , Consciousness/drug effects , Dissociative Disorders/chemically induced , Electrocorticography/drug effects , Fentanyl/pharmacology , Morphine/pharmacology , Sleep Stages/drug effects , Wakefulness/drug effects , Analgesics/administration & dosage , Analgesics, Opioid/administration & dosage , Animals , Buprenorphine/administration & dosage , Disease Models, Animal , Electroencephalography , Electromyography , Fentanyl/administration & dosage , Male , Mice , Mice, Inbred C57BL , Morphine/administration & dosage
4.
Sleep Breath ; 25(2): 1029-1035, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32857320

ABSTRACT

PURPOSE: Drug induced sedation endoscopy (DISE) is performed to investigate patterns and sites of obstruction in patients with sleep-disordered breathing (SDB). During DISE the patients are sedated to obtain a muscular relaxation of the upper airway which mimics the relaxation during natural sleep. Different sleep stages are intended to be simulated by drug induced sedation, and it is helpful to measure the depth of sedation. The BiSpectral Index® (BIS) is often used for this procedure. Besides the BIS, other means of sedation depth monitoring exist in anaesthesiology but have not yet been investigated with respect to DISE. Monitoring of the Cerebral State Index® (CSI) is one of these methods. The aim of the study was to compare the BIS and CSI for sedation depth monitoring during DISE. METHODS: Sixty patients underwent DISE monitored by the BIS and CSI in parallel. The BIS and CSI values were compared using the Bland-Altman analysis. RESULTS: The BIS and CSI values differed during the course of sedation during DISE by a mean of - 6.07. At light sedation (BIS 60-80), lower values by 10 scale points of CSI compared with BIS were detectable. At deeper sedation levels (BIS 40-50), the CSI turned to present equal and even higher values compared with the BIS. CONCLUSION: Sedation depth measurement during DISE can be performed by the BIS or CSI, but the differences should be interpreted carefully as comparable data for sleep stages in natural sleep are available only for BIS.


Subject(s)
Deep Sedation , Endoscopy/methods , Hypnotics and Sedatives/pharmacology , Monitoring, Physiologic/methods , Sleep Stages/drug effects , Adult , Female , Humans , Male , Middle Aged , Prospective Studies
5.
Int J Neurosci ; 131(6): 580-590, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32228330

ABSTRACT

INTRODUCTION: There is evidence to suggest that melatonin diminishes non-rapid eye movement sleep (NREMS) latency in patients with Alzheimer´s disease (AD). However, melatonin's effects on cortical activity during NREMS in AD have not been studied. The objective of this research was to analyze the effects of melatonin on cortical activity during the stages of NREMS in 8 mild-to-moderate AD patients that received 5-mg of fast-release melatonin. METHODS: During a single-blind, placebo-controlled crossover study, polysomnographic recordings were obtained from C3-A1, C4-A2, F7-T3, F8-T4, F3-F4 and O1-O2. Also, the relative power (RP) and EEG coherences of the delta, theta, alpha1, alpha2, beta1, beta2 and gamma bands were calculated during NREMS-1, NREMS-2 and NREMS-3. These sleep latencies and all EEG data were then compared between the placebo and melatonin conditions. RESULTS: During NREMS-2, a significant RP increase was observed in the theta band of the left-central hemisphere. During NREMS-3, significant RP decreases in the beta bands were recorded in the right-central hemisphere, compared to the placebo group. After melatonin administration, significant decreases of EEG coherences in the beta2, beta1 and gamma bands were observed in the right hemisphere during NREMS-3. DISCUSSION: We conclude that short NREMS onset related to melatonin intake in AD patients is associated with a significant RP increase in the theta band and a decrease in RP and EEG coherences in the beta and gamma bands during NREMS-3. These results suggest that the GABAergic pathways are preserved in mild-to-moderate AD.


Subject(s)
Alzheimer Disease/complications , Brain Waves/drug effects , Central Nervous System Depressants/pharmacology , Electroencephalography Phase Synchronization/drug effects , Melatonin/pharmacology , Sleep Stages/drug effects , Sleep Wake Disorders/drug therapy , Aged , Alzheimer Disease/physiopathology , Brain Waves/physiology , Central Nervous System Depressants/administration & dosage , Cross-Over Studies , Humans , Male , Melatonin/administration & dosage , Middle Aged , Pilot Projects , Polysomnography , Severity of Illness Index , Single-Blind Method , Sleep Stages/physiology , Sleep Wake Disorders/etiology , Sleep Wake Disorders/physiopathology
6.
Glia ; 68(1): 44-59, 2020 01.
Article in English | MEDLINE | ID: mdl-31429116

ABSTRACT

Synaptic strength reduces during sleep, but the underlying mechanisms of this process are unclear. This study showed reduction of synaptic proteins in rat prefrontal cortex (PFC) at AM7 or Zeitgeber Time (ZT0), when the light phase or sleeping period for rats started. At this time point, microglia were weakly activated, displaying larger and more granular somata with increased CD11b expression compared with those at ZT12, as revealed by flow cytometry. Expression of opsonins, such as complements or MFG-E8, matrix metalloproteinases, and microglial markers at ZT0 were increased compared with that at ZT12. Microglia at ZT0 phagocytosed synapses, as revealed by immunohistochemical staining. Immunoblotting detected more synapsin I in the isolated microglia at ZT0 than at ZT12. Complement C3- or MFG-E8-bound synapses were the most abundant at ZT0, some of which were phagocytosed by microglia. Systemic administration of synthetic glucocorticoid dexamethasone reduced microglial size, granularity and CD11b expression at ZT0, resembling microglia at ZT12, and increased synaptic proteins and decreased the sleeping period. Noradrenaline (NA) suppressed glutamate-induced phagocytosis in primary cultured microglia. Systemic administration of the brain monoamine-depleting agent reserpine decreased NA content and synapsin I expression in PFC, and increased expression of microglia markers, C3 and MFG-E8, while increasing the sleeping period. A NA precursor l-threo-dihydroxyphenylserine abolished the reserpine-induced changes. These results suggest that microglia may eliminate presumably weak synapses during every sleep phase. The circadian changes in concentrations of circulating glucocorticoids and brain NA might be correlated with the circadian changes of microglial phenotypes and synaptic strength.


Subject(s)
Microglia/metabolism , Phagocytes/metabolism , Phagocytosis/physiology , Prefrontal Cortex/metabolism , Sleep Stages/physiology , Synapses/metabolism , Animals , Cells, Cultured , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Dexamethasone/pharmacology , Male , Microglia/drug effects , Phagocytes/drug effects , Phagocytosis/drug effects , Prefrontal Cortex/cytology , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Sleep Stages/drug effects , Synapses/drug effects
7.
Front Neuroendocrinol ; 52: 22-28, 2019 01.
Article in English | MEDLINE | ID: mdl-29908879

ABSTRACT

Melatonin is a physiological indoleamine involved in circadian rhythm regulation and it is currently used for secondary sleep disorders supported by empirical evidence. A small amount of evidence and some controversial results have been obtained in some randomized controlled trials (RCT). The objective of this meta-analysis is to determine the efficacy of exogenous melatonin versus placebo in managing secondary sleep disorders. Literature retrieval of eligible RCT was performed in 5 databases (PubMed, Embase, Cochrane Library, ClinicalTrials.gov, and Web of Science). In total, 7 studies of 205 patients were included. Pooled data demonstrate that exogenous melatonin lowers sleep onset latency and increases total sleep time, whereas it has little if any effect on sleep efficiency. Although, the efficacy of melatonin still requires further confirmation, this meta-analysis clearly supports the use of melatonin as a management for patients with secondary sleep disorders.


Subject(s)
Central Nervous System Depressants/pharmacology , Melatonin/pharmacology , Sleep Stages/drug effects , Sleep Wake Disorders/drug therapy , Humans
8.
Neurochem Res ; 45(8): 1791-1801, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32367385

ABSTRACT

GABA, the most abundant inhibitory neurotransmitter in the brain, is closely linked with sleep and wakefulness. As the largest area input to the ventral pallidum (VP), the nucleus accumbens (NAc) has been confirmed to play a pivotal role in promoting non-rapid eye movement (NREM) sleep through inhibitory projections from NAc adenosine A2A receptor-expressing neurons to VP GABAergic neurons which mostly express GABAA receptors. Although these studies demonstrate the possible role of VP GABAergic neurons in sleep-wake regulation, whether and how its modulate sleep-wake cycle is not completely clear. In our study, pharmacological manipulations were implemented in freely moving rats and then the EEG and the EMG were recorded to monitor the sleep-wake states. We found that microinjection of muscimol, a GABAA receptor agonist, into the VP increased NREM sleep in both light and dark period. Microinjection of bicuculline, a GABAA receptor antagonist, into the VP increased wakefulness in the light period. Collectively, our data identify the important role of VP GABAA receptor-expressing neurons in NREM sleep of rats which may help improve the understanding of the pathological sleep disorders.


Subject(s)
Basal Forebrain/drug effects , GABA-A Receptor Agonists/pharmacology , Muscimol/pharmacology , Receptors, GABA-A/metabolism , Sleep Stages/drug effects , Animals , Basal Forebrain/metabolism , Bicuculline/pharmacology , GABA-A Receptor Antagonists/pharmacology , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Male , Rats, Sprague-Dawley , Wakefulness/drug effects
9.
Anesthesiology ; 133(6): 1234-1243, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33001139

ABSTRACT

BACKGROUND: The administration of dexmedetomidine is limited to highly monitored care settings because it is only available for use in humans as intravenous medication. An oral formulation of dexmedetomidine may broaden its use to all care settings. The authors investigated the effect of a capsule-based solid oral dosage formulation of dexmedetomidine on sleep polysomnography. METHODS: The authors performed a single-site, placebo-controlled, randomized, crossover, double-blind phase II study of a solid oral dosage formulation of dexmedetomidine (700 mcg; n = 15). The primary outcome was polysomnography sleep quality. Secondary outcomes included performance on the motor sequence task and psychomotor vigilance task administered to each subject at night and in the morning to assess motor memory consolidation and psychomotor function, respectively. Sleep questionnaires were also administered. RESULTS: Oral dexmedetomidine increased the duration of non-rapid eye movement (non-REM) stage 2 sleep by 63 (95% CI, 19 to 107) min (P = 0.010) and decreased the duration of rapid eye movement (REM) sleep by 42 (5 to 78) min (P = 0.031). Overnight motor sequence task performance improved after placebo sleep (7.9%; P = 0.003) but not after oral dexmedetomidine-induced sleep (-0.8%; P = 0.900). In exploratory analyses, we found a positive correlation between spindle density during non-REM stage 2 sleep and improvement in the overnight test performance (Spearman rho = 0.57; P = 0.028; n = 15) for placebo but not oral dexmedetomidine (Spearman rho = 0.04; P = 0.899; n = 15). Group differences in overnight motor sequence task performance, psychomotor vigilance task metrics, and sleep questionnaires did not meet the threshold for statistical significance. CONCLUSIONS: These results demonstrate that the nighttime administration of a solid oral dosage formulation of dexmedetomidine is associated with increased non-REM 2 sleep and decreased REM sleep. Spindle density during dexmedetomidine sleep was not associated with overnight improvement in the motor sequence task.


Subject(s)
Dexmedetomidine/pharmacology , Hypnotics and Sedatives/pharmacology , Sleep Stages/drug effects , Administration, Oral , Adult , Cross-Over Studies , Dexmedetomidine/administration & dosage , Double-Blind Method , Female , Humans , Hypnotics and Sedatives/administration & dosage , Male , Polysomnography
10.
J Pineal Res ; 69(3): e12674, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32535982

ABSTRACT

Melatonin (MLT) is widely used to treat sleep disorders although the underlying mechanism is still elusive. In mice, using wheel-running detection, we found that exogenous MLT could completely recover the period length prolonged by N-methyl-D-aspartate receptor (NMDAR) impairment due to the injection of the NMDAR antagonist MK-801, a preclinical model of psychosis. The analysis of the possible underlying mechanisms indicated that MLT could regulate the homeostatic state in the ventrolateral preoptic nucleus (VLPO) instead of the circadian process in the suprachiasmatic nucleus (SCN). In addition, our data showed that MK-801 decreased Ca2+ -related CaMKII expression and CREB phosphorylation levels in the VLPO, and MLT could rescue these intracellular impairments but not NMDAR expression levels. Accordingly, Gcamp6 AAV virus was injected in-vivo to further monitor intracellular Ca2+ levels in the VLPO, and MLT demonstrated a unique ability to increase Ca2+ fluorescence compared with MK-801-injected mice. Additionally, using the selective melatonin MT2 receptor antagonist 4-phenyl-2-propionamidotetralin (4P-PDOT), we discovered that the pharmacological effects of MLT upon NMDAR impairments were mediated by melatonin MT2 receptors. Using electroencephalography/electromyography (EEG/EMG) recordings, we observed that the latency to the first nonrapid eye movement (NREM) sleep episode was delayed by MK-801, and MLT was able to recover this delay. In conclusion, exogenous MLT by acting upon melatonin MT2 receptors rescues sleep phase delayed by NMDAR impairment via increasing intracellular Ca2+ signaling in the VLPO, suggesting a regulatory role of the neurohormone on the homeostatic system.


Subject(s)
Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dizocilpine Maleate/pharmacology , Melatonin/pharmacology , Preoptic Area/metabolism , Receptor, Melatonin, MT2/metabolism , Sleep Stages/drug effects , Animals , Electroencephalography , Electromyography , Male , Melatonin/metabolism , Mice
11.
J Sleep Res ; 28(4): e12800, 2019 08.
Article in English | MEDLINE | ID: mdl-30565327

ABSTRACT

Studies have shown that both nicotine and sleep spindles are associated with enhanced memorisation. Further, a few recent studies have shown how cholinergic input through nicotinic and muscarinic receptors can trigger or modulate sleep processes in general, and sleep spindles in particular. To better understand the interaction between nicotine and sleep spindles, we compared in a single blind randomised study the characteristics of sleep spindles in 10 healthy participants recorded for 2 nights, one with a nicotine patch and one with a sham patch. We investigated differences in sleep spindle duration, amplitude, intra-spindle oscillation frequency and density (i.e. spindles per min). We found that under nicotine, spindles are more numerous (average increase: 0.057 spindles per min; 95% confidence interval: [0.025-0.089]; p = .0004), have higher amplitude (average amplification: 0.260 µV; confidence interval: [0.119-0.402]; p = .0032) and last longer (average lengthening: 0.025 s; confidence interval: [0.017-0.032]; p = 2.7e-11). These results suggest that nicotine can increase spindle activity by acting on nicotinic acetylcholine receptors, and offer an attractive hypothesis for common mechanisms that may support memorisation improvements previously reported to be associated with nicotine and sleep spindles.


Subject(s)
Electroencephalography/methods , Nicotine/adverse effects , Sleep Stages/drug effects , Sleep/drug effects , Adult , Female , Humans , Male , Single-Blind Method , Young Adult
12.
Eur J Neurosci ; 48(8): 2738-2758, 2018 10.
Article in English | MEDLINE | ID: mdl-29280209

ABSTRACT

Sleep abnormalities have recently gained renewed attention in patients diagnosed with schizophrenia. Disrupted thalamocortical brain oscillations hold promise as putative biomarkers or endophenotypes of the disorder. Despite an increase in studies related to sleep spindle and slow-wave activity, findings remain in part contradictory. Although sleep spindle deficits have been confirmed in several groups of patients with chronic, medicated schizophrenia, data on the early stages of the disorder and in unmedicated subjects are still insufficient. Findings on slow-wave abnormalities are largely inconclusive, possibly due to the different criteria employed to define the phenomenon and to the influence of atypical antipsychotics. In this review, we aim to address the methodological and practical issues that may have limited the consistency of findings across research groups and different patient populations. Given the neurobiological relevance of these oscillations, which reflect the integrity of thalamocortical and cortico-cortical function, research in this domain should be encouraged. To promote widespread consensus over the scientific and clinical implications of these sleep-related phenomena, we advocate uniform and sound methodological approaches. These should encompass electroencephalographic recording and analysis techniques but also selection criteria and characterization of clinical populations.


Subject(s)
Cerebral Cortex/physiopathology , Schizophrenia/physiopathology , Sleep Stages/physiology , Sleep Wake Disorders/physiopathology , Thalamus/physiopathology , Animals , Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Cerebral Cortex/drug effects , Forecasting , Humans , Schizophrenia/drug therapy , Sleep Stages/drug effects , Sleep Wake Disorders/chemically induced , Thalamus/drug effects
13.
Sleep Breath ; 22(4): 919-923, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30324545

ABSTRACT

PURPOSE: During drug-induced sleep endoscopy (DISE) in patients with obstructive sleep apnea, the increased depth of propofol anesthesia is related to the increased collapsibility of the upper airway with dose-dependent. We examined the effect of remifentanil on propofol concentration during DISE. METHODS: In a prospective randomized trial, 56 adult patients were divided into remifentanil-propofol (n = 28) and propofol alone (n = 28) groups. Anesthesia was administered using a target-controlled infusion system. In the remifentanil-propofol group, 0.5 ng/ml remifentanil was administered prior to propofol infusion and its concentration maintained; thereafter, in the propofol alone group, normal saline was injected instead of remifentanil. Propofol was infused at a concentration of 1.5 µg/ml after the target concentration of remifentanil was reached. In both groups, the concentration of propofol was increased by 0.5 µg/ml if the degree of sedation was not sufficient. The sedation level was targeted at observer's assessment of alertness/sedation (OAA/S) scale 3. RESULTS: The mean propofol concentration was 2.87 ± 0.60 µg/ml in the remifentanil-propofol group, which was lower than that in the propofol alone group (3.38 ± 0.72 µg/ml, P < 0.001). The time until sufficient sedation to perform DISE was shorter in the remifentanil-propofol group (P < 0.001). Apnea-hypopnea index and the lowest peripheral capillary oxygen saturation (SpO2) during polysomnography showed no statistical difference between groups (P > 0.05). The lowest SpO2 and VOTE classification during DISE were also not statistically different (P > 0.05). CONCLUSIONS: Use of remifentanil during DISE reduces the target concentration of propofol required for patient sedation to perform DISE without respiratory depression.


Subject(s)
Analgesics, Opioid/administration & dosage , Anesthesia/methods , Natural Orifice Endoscopic Surgery/methods , Remifentanil/administration & dosage , Sleep Apnea, Obstructive/therapy , Adult , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Humans , Hypnotics and Sedatives/administration & dosage , Male , Middle Aged , Polysomnography , Propofol/administration & dosage , Prospective Studies , Sleep/drug effects , Sleep Apnea, Obstructive/drug therapy , Sleep Stages/drug effects
14.
Mar Drugs ; 16(5)2018 Apr 24.
Article in English | MEDLINE | ID: mdl-29695101

ABSTRACT

In our previous studies, we have demonstrated that marine polyphenol phlorotannins promote sleep through the benzodiazepine site of the gamma-aminobutyric acid type A (GABAA) receptors. In this follow-up study, the sleep-promoting effects of triphlorethol A, one of the major phlorotannin constituents, were investigated. The effect of triphlorethol A on sleep-wake architecture and profiles was evaluated based on electroencephalogram and electromyogram data from C57BL/6N mice and compared with the well-known hypnotic drug zolpidem. Oral administration of triphlorethol A (5, 10, 25, and 50 mg/kg) dose-dependently decreased sleep latency and increased sleep duration during pentobarbital-induced sleep in imprinting control region mice. Triphlorethol A (50 mg/kg) significantly decreased sleep latency and increased the amount of non-rapid eye movement sleep (NREMS) in C57BL/6N mice, without affecting rapid eye movement sleep (REMS). There was no significant difference between the effects of triphlorethol A at 50 mg/kg and zolpidem at 10 mg/kg. Triphlorethol A had no effect on delta activity (0.5⁻4 Hz) of NREMS, whereas zolpidem significantly decreased it. These results not only support the sleep-promoting effects of marine polyphenol phlorotannins, but also suggest that the marine polyphenol compound triphlorethol A is a promising structure for developing novel sedative hypnotics.


Subject(s)
Phloroglucinol/analogs & derivatives , Seaweed/chemistry , Sleep Latency/drug effects , Sleep Stages/drug effects , Administration, Oral , Animals , Dose-Response Relationship, Drug , Hypnotics and Sedatives/chemistry , Hypnotics and Sedatives/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Molecular Structure , Phloroglucinol/administration & dosage , Phloroglucinol/chemistry , Phloroglucinol/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Zolpidem
15.
Pharmazie ; 73(1): 35-41, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29441949

ABSTRACT

Patients benefit from drug therapy not only through pharmacological mechanisms, but also through non-pharmacological action (placebo effect), which may be mediated in part by the prefrontal area of the brain. We consider that the difference between responders and non-responders to placebo might be related to polymorphisms in the serotonin transporter-linked polymorphic region (5-HTTLPR). To study this idea, we performed a randomized double-blind clinical trial using caffeine and lactose (placebo). Activity in the prefrontal area of the brain was measured in terms of blood flow by means of near-infrared spectroscopy (NIRS) as an objective indicator. Self-reported feelings of drowsiness on established scales were used as subjective indicators. Twenty-one subjects in block A took caffeine on the first day and placebo on the third day, and 21 in block B took placebo on the first day and placebo on the third day. After placebo administration, improvement of sleepiness was significantly enhanced, a similar extent to that after caffeine medication. Among the 42 subjects, 22 showed S/S type polymorphism in the serotonin transporter (52.4 %), 17 showed S/L type (40.5 %) and 3 showed L/L type (7.10 %). Statistical analysis of the results indicate that subjects with L/L genotype showed a significantly greater placebo response in terms of both self-reported feeling of drowsiness and blood flow in the prefrontal area of the brain associated with working memory (46 area). Our results indicate that the L/L genotype of 5-HTTLPR, which is rare in Japanese (3.2 %) but common in Americans (32.2 %), may be associated with a greater placebo effect.


Subject(s)
Caffeine/pharmacology , Prefrontal Cortex/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Sleep Stages/drug effects , Adult , Double-Blind Method , Female , Genotype , Humans , Male , Memory, Short-Term/physiology , Placebo Effect , Polymorphism, Genetic , Prefrontal Cortex/blood supply , Self Report , Sleep Stages/genetics , Spectroscopy, Near-Infrared , Young Adult
16.
JAAPA ; 31(12): 52-54, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30489392

ABSTRACT

The sleep architecture (or sleep kinetics) of schizophrenia is different from that of other mental illnesses, including major depressive disorder. However, clinicians rarely consider these parameters in clinical settings during treatment. This article discusses the use of polysomnography to characterize the sleeping patterns of patients diagnosed with schizophrenia and the positive influence of clozapine on sleep in patients with schizophrenia.


Subject(s)
Schizophrenia/therapy , Schizophrenic Psychology , Sleep/physiology , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Clozapine/pharmacology , Clozapine/therapeutic use , Humans , Polysomnography , Sleep/drug effects , Sleep Stages/drug effects , Sleep Stages/physiology
17.
J Neurochem ; 142(5): 710-720, 2017 09.
Article in English | MEDLINE | ID: mdl-28444769

ABSTRACT

Alcohol has a profound effect on sleep. However, neuronal substrates mediating sleep-promoting effects of alcohol are unknown. Since the basal forebrain (BF) cholinergic neurons are implicated in the homeostatic regulation of sleep, we hypothesized that the BF cholinergic neurons may have an important role in sleepiness observed after alcohol consumption. 192-IgG-saporin (bilateral BF infusions) was used to selectively lesion BF cholinergic neurons in adult male Sprague-Dawley rats. Standard surgical procedures were used to implant sleep recording electrodes or microdialysis guide cannulas. The first experiment used between-group design [lesion and sham (controls)] and examined effects of BF cholinergic neuronal lesions on alcohol (3 g/Kg; ig) induced sleep promotion. The second experiment used within-group design [lesion (ipsilateral BF) and sham (controls; contralateral BF) in same animal] and local reverse microdialysis infusion of alcohol (300 mM) to examine the effects of cholinergic neuronal lesions on extracellular adenosine in the BF. Alcohol had a robust sleep promoting effect in controls as evidenced by a significant reduction in sleep onset latency and wakefulness; non-rapid eye movement sleep was significantly increased. No such alcohol-induced sleep promotion was observed in lesioned rats with significantly fewer BF cholinergic neurons. Rapid eye movement sleep was minimally affected. Adenosine release was significantly reduced following local infusion of alcohol on the lesion side, with significantly fewer cholinergic neurons as compared with the control side. Based on these results, we suggest that alcohol promotes sleep by increasing extracellular adenosine via its action on cholinergic neurons of the BF. Read the Editorial Highlight for this article on page 620.


Subject(s)
Adenosine/metabolism , Alcohol Drinking/metabolism , Basal Forebrain/metabolism , Cholinergic Neurons/metabolism , Sleep Stages/physiology , Wakefulness/physiology , Adenosine/antagonists & inhibitors , Animals , Basal Forebrain/drug effects , Basal Forebrain/pathology , Cholinergic Neurons/pathology , Electroencephalography/drug effects , Electroencephalography/methods , Ethanol/administration & dosage , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Sleep Stages/drug effects , Wakefulness/drug effects
18.
Epilepsy Behav ; 76: 63-70, 2017 11.
Article in English | MEDLINE | ID: mdl-28927712

ABSTRACT

INTRODUCTION: Rufinamide is approved for the adjunctive treatment of seizures associated with Lennox-Gastaut syndrome (LGS) in patients aged ≥4years. The objective of this study was to provide real-world, long-term data on patients with LGS initiating rufinamide as add-on therapy and patients with LGS receiving other antiepileptic drugs (AEDs). METHODS: A Phase IV, noninterventional, multicenter registry study was conducted in patients with LGS aged ≥4years requiring modification to any AED treatment, including initiation of add-on rufinamide therapy. Safety/tolerability was assessed by evaluating treatment-emergent adverse events (TEAEs), and efficacy was assessed using a generic seizure frequency scale. RESULTS: A total of 111 patients from 64 sites in 8 European countries were included, of whom 64 initiated rufinamide ("rufinamide" group) and 21 did not receive rufinamide at any time during the study ("no-rufinamide" group). Mean ages were 16.1years (rufinamide) and 15.0years (no rufinamide). The median duration of follow-up was >2years (range: 1.3-46.4months). Antiepileptic drug-related TEAEs were reported for 40.6% (rufinamide) and 33.3% (no rufinamide) of patients and led to discontinuation of 7.8% and 4.8%, respectively. The most frequently reported rufinamide-related TEAEs (≥5% patients) were somnolence (7.8%) and decreased appetite (6.3%). There were no unexpected safety/tolerability findings. At month 12, the proportion of patients with improvement in all seizures ("much improved" or "very much improved") was 28.6% (12/42) for the rufinamide group and 14.3% (2/14) for the no-rufinamide group. CONCLUSION: The study provided valuable information on LGS and its management, and evidence that rufinamide has a consistent and generally favorable safety/tolerability profile when used in routine clinical practice. CLINICALTRIALS. GOV IDENTIFIER: NCT01991041.


Subject(s)
Anticonvulsants/therapeutic use , Lennox Gastaut Syndrome/drug therapy , Triazoles/therapeutic use , Adolescent , Child , Child, Preschool , Ethnicity , Europe , European Union , Female , Humans , Male , Registries , Seizures/drug therapy , Sleep Stages/drug effects , Treatment Outcome , Triazoles/adverse effects
19.
Epilepsy Behav ; 75: 176-182, 2017 10.
Article in English | MEDLINE | ID: mdl-28866338

ABSTRACT

OBJECTIVE: The objective of the study was to investigate the effects of lacosamide (LCM) on daytime sleepiness ascertained by the Epworth Sleepiness Scale (ESS) in adults with focal epilepsy in a randomized, controlled design. METHODS: Subjects taking ≤2 AEDs for ≥4weeks underwent polysomnography with EEG followed by the maintenance of wakefulness test (MWT) and completed the ESS and other patient-reported outcomes (PROs) at baseline, LCM 200mg/day, and LCM 400mg/day (Visit 4; V4). Primary endpoint was ESS change (V4 to baseline) between LCM and placebo. Noninferiority test on ESS used a one-sided t-test based on a hypothesized difference of 4-point change between groups. Superiority test used a two-sided t-test to investigate the difference in change in PROs and MWT mean sleep latency (MSL) between groups. Fifty-five subjects provided 80% power to show noninferiority of LCM assuming 10% dropout. RESULTS: Fifty-two subjects (mean age: 43.5±13.2years, 69% female, median monthly seizure frequency: 1 [0, 4.0]) participated. Baseline group characteristics including age, sex, ethnicity, standardized AED dose, seizure frequency, and ESS were similar. Abnormal baseline ESS scores were found in 35% of subjects. Noninferiority test found a ≤4-point increase in ESS (mean [95% CI]) in LCM subjects vs. placebo (-1.2 [-2.9, 0.53] vs. -1.1 [-5.2, 3.0], p=0.027) at V4. No significant difference in change in PROs, MSL, seizure frequency, or AED standardized dose was observed between groups. SIGNIFICANCE: Our interventional trial found that LCM is not a major contributor to daytime sleepiness based on subjective and objective measures. Inclusion of sleepiness measures in AED trials is warranted given the high prevalence of sleep-wake complaints in people with epilepsy.


Subject(s)
Acetamides/adverse effects , Anticonvulsants/adverse effects , Epilepsies, Partial/drug therapy , Sleep Stages/drug effects , Wakefulness/drug effects , Acetamides/therapeutic use , Adult , Anticonvulsants/therapeutic use , Epilepsies, Partial/physiopathology , Female , Humans , Lacosamide , Male , Middle Aged , Patient Reported Outcome Measures , Polysomnography , Treatment Outcome
20.
Sleep Breath ; 21(2): 319-325, 2017 May.
Article in English | MEDLINE | ID: mdl-27726069

ABSTRACT

PURPOSE: Sleep disturbance is common in Parkinson's disease (PD) and negatively impacts quality of life. There is little data on how dopamine agonists influence nocturnal sleep in PD, particularly in sleep laboratory data to measure sleep parameters and their changes objectively. The goal of this open-label study was to objectively evaluate the effect of rotigotine on sleep in PD patients by video-polysomnographic methods. METHODS: A total of 25 PD patients with complaints of nocturnal sleep impairment were enrolled. The sleep quality before and after stable rotigotine therapy was evaluated subjectively through questionnaire assessments and objectively measured by video-polysomnographic methods. The Parkinsonism, depression, anxiety, and quality of life of PD patients were also evaluated through questionnaire assessments. RESULTS: At the end of rotigotine treatment, the PD daytime functioning, motor performance, depression, subjective quality of sleep, and the quality of life improved. Video-polysomnographic analysis showed that the sleep efficiency and stage N1% were increased, while the sleep latency, wake after sleep onset, and the periodic leg movements in sleep index were decreased after rotigotine treatment. CONCLUSIONS: Video-polysomnographic analysis confirmed the subjective improvement of sleep after rotigotine treatment. This observation suggests that in PD rotigotine is a treatment option for patients complaining from sleep disturbances.


Subject(s)
Dopamine Agonists/therapeutic use , Parkinson Disease/drug therapy , Polysomnography/drug effects , Polysomnography/methods , Sleep Wake Disorders/drug therapy , Tetrahydronaphthalenes/therapeutic use , Thiophenes/therapeutic use , Video Recording/methods , Aged , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Parkinson Disease/diagnosis , Parkinson Disease/psychology , Quality of Life/psychology , Sleep Stages/drug effects , Sleep Wake Disorders/diagnosis , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL