Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
1.
Sleep Med ; 121: 144-150, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38972128

RESUMO

BACKGROUND: Although sleepwalking is one of the most prevalent and potentially injurious of the NREM parasomnias, it is still diagnosed primarily based on the patient's clinical history. Early pilot work suggested that sleep deprivation protocols could help obtain a polysomnographically-based (PSG) diagnosis of sleepwalking, but larger studies remain lacking. METHODS: We compared baseline PSG recordings with those obtained after 25hrs of sleep deprivation in a cohort of 124 consecutively assessed adult sleepwalkers. RESULTS: When compared to baseline recordings, post-sleep deprivation PSG assessments resulted in nearly twice as many somnambulistic episodes being recorded in the laboratory and significantly increased the proportion of patients (from 48 % to 63 %) experiencing at least one lab-based episode. Moreover, while 17 % of patients experienced a sleepwalking event exclusively during recovery sleep, only 2 % of patients did so solely at baseline. Sleep deprivation had similar facilitating effects on patents' somnambulistic events regardless of age of onset and positive versus negative family history for sleepwalking. Younger age and higher home episode frequency both predicted a positive response to sleep deprivation. A separate group of 17 patients with comorbid sleep disorders showed a similar increase in their proportion experiencing at least one episode during recovery sleep. CONCLUSION: The results from this large series of sleepwalkers provide strong support for the use of sleep deprivation in facilitating the occurrence of somnambulistic events in the sleep laboratory.

2.
bioRxiv ; 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-39005401

RESUMO

Decrease in cognitive performance after sleep deprivation followed by recovery after sleep suggests its key role, and especially non-rapid eye movement (NREM) sleep, in the maintenance of cognition. It remains unknown whether brain network reorganization in NREM sleep stages N2 and N3 can uniquely be mapped onto individual differences in cognitive performance after a recovery nap following sleep deprivation. Using resting state functional magnetic resonance imaging (fMRI), we quantified the integration and segregation of brain networks during NREM sleep stages N2 and N3 while participants took a 1-hour nap following 24-hour sleep deprivation, compared to well-rested wakefulness. Here, we advance a new analytic framework called the hierarchical segregation index (HSI) to quantify network segregation across spatial scales, from whole-brain to the voxel level, by identifying spatio-temporally overlapping large-scale networks and the corresponding voxel-to-region hierarchy. Our results show that network segregation increased in the default mode, dorsal attention and somatomotor networks during NREM sleep compared to wakefulness. Segregation within the visual, limbic, and executive control networks exhibited N2 versus N3 sleep-specific voxel-level patterns. More segregation during N3 was associated with worse recovery of working memory, executive attention, and psychomotor vigilance after the nap. The level of spatial resolution of network segregation varied among brain regions and was associated with the recovery of performance in distinct cognitive tasks. We demonstrated the sensitivity and reliability of voxel-level HSI to provide key insights into within-region variation, suggesting a mechanistic understanding of how NREM sleep replenishes cognition after sleep deprivation.

3.
Proc Natl Acad Sci U S A ; 121(31): e2400678121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39052838

RESUMO

Recollecting painful or traumatic experiences can be deeply troubling. Sleep may offer an opportunity to reduce such suffering. We developed a procedure to weaken older aversive memories by reactivating newer positive memories during sleep. Participants viewed 48 nonsense words each paired with a unique aversive image, followed by an overnight sleep. In the next evening, participants learned associations between half of the words and additional positive images, creating interference. During the following non-rapid-eye-movement sleep, auditory memory cues were unobtrusively delivered. Upon waking, presenting cues associated with both aversive and positive images during sleep, as opposed to not presenting cues, weakened aversive memory recall while increasing positive memory intrusions. Substantiating these memory benefits, computational modeling revealed that cueing facilitated evidence accumulation toward positive affect judgments. Moreover, cue-elicited theta brain rhythms during sleep predominantly predicted the recall of positive memories. A noninvasive sleep intervention can thus modify aversive recollection and affective responses.


Assuntos
Sinais (Psicologia) , Rememoração Mental , Sono , Humanos , Feminino , Sono/fisiologia , Masculino , Rememoração Mental/fisiologia , Adulto , Adulto Jovem , Memória/fisiologia
4.
Brain Sci ; 14(7)2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-39061403

RESUMO

A body of experimental research has aimed to investigate processes underlying dream formation by examining the effects of a range of pre-sleep stimuli and events on subsequent dream content. Given its ever-growing presence and salience in people's everyday lives, pre-sleep media consumption stands out as a key variable that could influence people's dreams. We conducted a scoping review to evaluate the experimental evidence of the effects of pre-sleep exposure to visual media on dream content. A systematic search on PubMed, PsycInfo, and Web of Science using terms related to moving visual media and dreams yielded 29 studies meeting the inclusion criteria. Overall, we found modest yet varied effects of pre-sleep exposure to visual media on dream content, with rates of stimulus-related incorporation ranging from 3% to 43% for REM dream reports, 4% to 30% for NREM sleep mentation reports, and between 11% and 35% for home dream reports. Our review highlights the large methodological heterogeneity and gaps across studies, the general difficulty in influencing dream content using pre-sleep exposure to visual media, and suggests promising venues for future research to advance our understanding of how and why digital media may impact people's dreams.

5.
Cell Rep ; 43(6): 114266, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38787724

RESUMO

Fragile X syndrome (FXS) is associated with disrupted cognition and sleep abnormalities. Sleep loss negatively impacts cognitive function, and one untested possibility is that disrupted cognition in FXS is exacerbated by abnormal sleep. We tested whether ML297, a hypnotic acting on G-protein-activated inward-rectifying potassium (GIRK) channels, could reverse sleep phenotypes and disrupted memory in Fmr1-/y mice. Fmr1-/y mice exhibit reduced non-rapid eye movement (NREM) sleep and fragmented NREM architecture, altered sleep electroencephalogram (EEG) oscillations, and reduced EEG coherence between cortical areas; these are partially reversed following ML297 administration. Treatment following contextual fear or spatial learning restores disrupted memory consolidation in Fmr1-/y mice. During memory recall, Fmr1-/y mice show an altered balance of activity among hippocampal principal neurons vs. parvalbumin-expressing interneurons; this is partially reversed by ML297. Because sleep disruption could impact neurophysiological phenotypes in FXS, augmenting sleep may improve disrupted cognition in this disorder.


Assuntos
Modelos Animais de Doenças , Eletroencefalografia , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Transtornos da Memória , Sono , Animais , Síndrome do Cromossomo X Frágil/fisiopatologia , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Síndrome do Cromossomo X Frágil/complicações , Transtornos da Memória/fisiopatologia , Transtornos da Memória/tratamento farmacológico , Camundongos , Sono/efeitos dos fármacos , Sono/fisiologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Masculino , Hipnóticos e Sedativos/farmacologia , Hipnóticos e Sedativos/uso terapêutico , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Camundongos Endogâmicos C57BL , Medo , Consolidação da Memória/efeitos dos fármacos
7.
J Neurosci ; 44(24)2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38604779

RESUMO

Memory reactivation during sleep is thought to facilitate memory consolidation. Most sleep reactivation research has examined how reactivation of specific facts, objects, and associations benefits their overall retention. However, our memories are not unitary, and not all features of a memory persist in tandem over time. Instead, our memories are transformed, with some features strengthened and others weakened. Does sleep reactivation drive memory transformation? We leveraged the Targeted Memory Reactivation technique in an object category learning paradigm to examine this question. Participants (20 female, 14 male) learned three categories of novel objects, where each object had unique, distinguishing features as well as features shared with other members of its category. We used a real-time EEG protocol to cue the reactivation of these objects during sleep at moments optimized to generate reactivation events. We found that reactivation improved memory for distinguishing features while worsening memory for shared features, suggesting a differentiation process. The results indicate that sleep reactivation does not act holistically on object memories, instead supporting a transformation where some features are enhanced over others.


Assuntos
Eletroencefalografia , Consolidação da Memória , Sono , Humanos , Feminino , Masculino , Sono/fisiologia , Adulto Jovem , Adulto , Consolidação da Memória/fisiologia , Eletroencefalografia/métodos , Memória/fisiologia , Adolescente
8.
Neurosci Biobehav Rev ; 158: 105567, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38309498

RESUMO

Prenatal alcohol exposure is the leading nongenetic cause of human intellectual impairment. The long-term impacts of prenatal alcohol exposure on health and well-being are diverse, including neuropathology leading to behavioral, cognitive, and emotional impairments. Additionally negative effects also occur on the physiological level, such as the endocrine, cardiovascular, and immune systems. Among these diverse impacts is sleep disruption. In this review, we describe how prenatal alcohol exposure affects sleep, and potential mechanisms of those effects. Furthermore, we outline the evidence that sleep disruption across the lifespan may be a mediator of some cognitive and behavioral impacts of developmental alcohol exposure, and thus may represent a promising target for treatment.


Assuntos
Transtornos do Espectro Alcoólico Fetal , Efeitos Tardios da Exposição Pré-Natal , Feminino , Humanos , Gravidez , Transtornos do Espectro Alcoólico Fetal/etiologia , Etanol/efeitos adversos , Sono
9.
Clin Neurophysiol ; 159: 56-65, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38335766

RESUMO

OBJECTIVE: Investigate sleep and temporal lobe epilepsy (TLE) effects on brain networks derived from electroencephalography (EEG). METHODS: High-density EEG was recorded during non-rapid eye movement (NREM) sleep stage 2 (N2) and wakefulness in 23 patients and healthy controls (HC). Epochs without epileptic discharges were source-reconstructed in 72 brain regions and connectivity was estimated. We calculated network integration and segregation at global (global efficiency, GE; average clustering coefficient, avgCC) and hemispheric level. These were compared between groups across frequency bands and correlated with the individual proportion of wakefulness- or sleep-related seizures. RESULTS: At the global level, patients had higher delta GE, delta avgCC and theta avgCC than controls, irrespective of the vigilance state. During wakefulness, theta GE of patients was higher than controls and, for patients, theta GE during wakefulness was higher than during N2. Wake-to-sleep differences in TLE were notable only in the ipsilateral hemisphere. Only measures from wakefulness recordings correlated with the proportion of wakefulness- or sleep-related seizures. CONCLUSIONS: TLE network alterations are more prominent during wakefulness and at lower frequencies. Increased integration and segregation suggest a pathological 'small world' configuration with a possible inhibitory role. SIGNIFICANCE: Network alterations in TLE occur and are easier to detect during wakefulness.


Assuntos
Epilepsia Reflexa , Epilepsia do Lobo Temporal , Humanos , Epilepsia do Lobo Temporal/diagnóstico , Movimentos Oculares , Vigília , Sono , Convulsões
10.
J Sleep Res ; : e14134, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38196146

RESUMO

The circuitry underlying the initiation, maintenance, and coordination of wakefulness, rapid eye movement sleep, and non-rapid eye movement sleep is not thoroughly understood. Sleep is thought to arise due to decreased activity in the ascending reticular arousal system, which originates in the brainstem and awakens the thalamus and cortex during wakefulness. Despite the conventional association of sleep-wake states with hippocampal rhythms, the mutual influence of the hippocampal formation in regulating vigilance states has been largely neglected. Here, we focus on the subiculum, the main output region of the hippocampal formation. The subiculum, particulary the ventral part, sends extensive monosynaptic projections to crucial regions implicated in sleep-wake regulation, including the thalamus, lateral hypothalamus, tuberomammillary nucleus, basal forebrain, ventrolateral preoptic nucleus, ventrolateral tegmental area, and suprachiasmatic nucleus. Additionally, second-order projections from the subiculum are received by the laterodorsal tegmental nucleus, locus coeruleus, and median raphe nucleus, suggesting the potential involvement of the subiculum in the regulation of the sleep-wake cycle. We also discuss alterations in the subiculum observed in individuals with sleep disorders and in sleep-deprived mice, underscoring the significance of investigating neuronal communication between the subiculum and pathways promoting both sleep and wakefulness.

11.
Brain Topogr ; 37(2): 312-328, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37253955

RESUMO

The majority of EEG microstate analyses concern wakefulness, and the existing sleep studies have focused on changes in spatial microstate properties and on microstate transitions between adjacent time points, the shortest available time scale. We present a more extensive time series analysis of unsmoothed EEG microstate sequences in wakefulness and non-REM sleep stages across many time scales. Very short time scales are assessed with Markov tests, intermediate time scales by the entropy rate and long time scales by a spectral analysis which identifies characteristic microstate frequencies. During the descent from wakefulness to sleep stage N3, we find that the increasing mean microstate duration is a gradual phenomenon explained by a continuous slowing of microstate dynamics as described by the relaxation time of the transition probability matrix. The finite entropy rate, which considers longer microstate histories, shows that microstate sequences become more predictable (less random) with decreasing vigilance level. Accordingly, the Markov property is absent in wakefulness but in sleep stage N3, 10/19 subjects have microstate sequences compatible with a second-order Markov process. A spectral microstate analysis is performed by comparing the time-lagged mutual information coefficients of microstate sequences with the autocorrelation function of the underlying EEG. We find periodic microstate behavior in all vigilance states, linked to alpha frequencies in wakefulness, theta activity in N1, sleep spindle frequencies in N2, and in the delta frequency band in N3. In summary, we show that EEG microstates are a dynamic phenomenon with oscillatory properties that slow down in sleep and are coupled to specific EEG frequencies across several sleep stages.


Assuntos
Eletroencefalografia , Vigília , Humanos , Sono , Fases do Sono , Cadeias de Markov , Encéfalo
12.
Curr Biol ; 34(2): 427-433.e5, 2024 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-38141616

RESUMO

Timing and quantity of sleep depend on a circadian (∼24-h) rhythm and a specific sleep requirement.1 Sleep curtailment results in a homeostatic rebound of more and deeper sleep, the latter reflected in increased electroencephalographic (EEG) slow-wave activity (SWA) during non-rapid eye movement (NREM) sleep.2 Circadian rhythms are synchronized by the light-dark cycle but persist under constant conditions.3,4,5 Strikingly, arctic reindeer behavior is arrhythmic during the solstices.6 Moreover, the Arctic's extreme seasonal environmental changes cause large variations in overall activity and food intake.7 We hypothesized that the maintenance of optimal functioning under these extremely fluctuating conditions would require adaptations not only in daily activity patterns but also in the homeostatic regulation of sleep. We studied sleep using non-invasive EEG in four Eurasian tundra reindeer (Rangifer tarandus tarandus) in Tromsø, Norway (69°N) during the fall equinox and both solstices. As expected, sleep-wake rhythms paralleled daily activity distribution, and sleep deprivation resulted in a homeostatic rebound in all seasons. Yet, these sleep rebounds were smaller in summer and fall than in winter. Surprisingly, SWA decreased not only during NREM sleep but also during rumination. Quantitative modeling revealed that sleep pressure decayed at similar rates during the two behavioral states. Finally, reindeer spent less time in NREM sleep the more they ruminated. These results suggest that they can sleep during rumination. The ability to reduce sleep need during rumination-undisturbed phases for both sleep recovery and digestion-might allow for near-constant feeding in the arctic summer.


Assuntos
Rena , Animais , Rena/fisiologia , Sono/fisiologia , Privação do Sono , Ritmo Circadiano/fisiologia , Eletroencefalografia , Regiões Árticas
13.
Curr Biol ; 34(1): 132-146.e5, 2024 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-38141615

RESUMO

Social interactions profoundly influence animal development, physiology, and behavior. Yet, how sleep-a central behavioral and neurophysiological process-is modulated by social interactions is poorly understood. Here, we characterized sleep behavior and neurophysiology in freely moving and co-living mice under different social conditions. We utilized wireless neurophysiological devices to simultaneously record multiple individuals within a group for 24 h, alongside video acquisition. We first demonstrated that mice seek physical contact before sleep initiation and sleep while in close proximity to each other (hereafter, "huddling"). To determine whether huddling during sleep is a motivated behavior, we devised a novel behavioral apparatus allowing mice to choose whether to sleep in close proximity to a conspecific or in solitude, under different environmental conditions. We also applied a deep-learning-based approach to classify huddling behavior. We demonstrate that mice are willing to forgo their preferred sleep location, even under thermoneutral conditions, to gain access to social contact during sleep. This strongly suggests that the motivation for prolonged physical contact-which we term somatolonging-drives huddling behavior. We then characterized sleep architecture under different social conditions and uncovered a social-dependent modulation of sleep. We also revealed coordination in multiple neurophysiological features among co-sleeping individuals, including in the timing of falling asleep and waking up and non-rapid eye movement sleep (NREMS) intensity. Notably, the timing of rapid eye movement sleep (REMS) was synchronized among co-sleeping male siblings but not co-sleeping female or unfamiliar mice. Our findings provide novel insights into the motivation for physical contact and the extent of social-dependent plasticity in sleep.


Assuntos
Sono REM , Sono , Masculino , Feminino , Camundongos , Animais , Sono/fisiologia , Sono REM/fisiologia , Eletroencefalografia , Vigília/fisiologia
14.
Mol Neurobiol ; 2023 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-37987958

RESUMO

Therapeutic options to contain seizures, a transitional stage of many neuropathologies, are limited due to the blood-brain barrier (BBB). Herbal nanoparticle formulations can be employed to enhance seizure prognosis. Bacoside A (BM3) and bacopaside I (BM4) were isolated from Bacopa monnieri and synthesized as nanoparticles (BM3NP and BM4NP, respectively) for an effective delivery system to alleviate seizures and associated conditions. After physicochemical characterization, cell viability was assessed on mouse neuronal stem cells (mNSC) and neuroblastoma cells (N2a). Thereafter, anti-seizure effects, mitochondrial membrane potential (MMP), apoptosis, immunostaining and epileptic marker mRNA expression were determined in vitro. The seizure-induced changes in the cortical electroencephalogram (EEG), electromyography (EMG), Non-Rapid Eye Movement (NREM) and Rapid Eye Movement (REM) sleep were monitored in vivo in a kainic acid (KA)-induced rat seizure model. The sizes of BM3NPs and BM4NPs were 165.5 nm and 689.6 nm, respectively. They were biocompatible and also aided in neuroplasticity in mNSC. BM3NPs and BM4NPs depicted more than 50% cell viability in N2a cells, with IC50 values of 1609 and 2962 µg/mL, respectively. Similarly, these nanoparticles reduced the cytotoxicity of N2a cells upon KA treatment. Nanoparticles decreased the expression of epileptic markers like fractalkine, HMGB1, FOXO3a and pro-inflammatory cytokines (P < 0.05). They protected neurons from apoptosis and restored MMP. After administration of BM3NPs and BM4NPs, KA-treated rats attained a significant reduction in the epileptic spikes, sleep latency and an increase in NREM sleep duration. Results indicate the potential of BM3NPs and BM4NPs in neutralizing the KA-induced excitotoxic seizures in neurons.

15.
J Sleep Res ; : e14078, 2023 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-37859564

RESUMO

Previous prospective studies examining associations of obstructive sleep apnea and sleep macroarchitecture with future cognitive function recruited older participants, many demonstrating baseline cognitive impairment. This study examined obstructive sleep apnea and sleep macroarchitecture predictors of visual attention, processing speed, and executive function after 8 years among younger community-dwelling men. Florey Adelaide Male Ageing Study participants (n = 477) underwent home-based polysomnography, with 157 completing Trail-Making Tests A and B and the Mini-Mental State Examination. Associations of obstructive sleep apnea (apnea-hypopnea index, oxygen desaturation index, and hypoxic burden index) and sleep macroarchitecture (sleep stage percentages and total sleep time) parameters with future cognitive function were examined using regression models adjusted for baseline demographic, biomedical, and behavioural factors, and cognitive task performance. The mean (standard deviation) age of the men at baseline was 58.9 (8.9) years, with severe obstructive sleep apnea (apnea-hypopnea index ≥30 events/h) in 9.6%. The median (interquartile range) follow-up was 8.3 (7.9-8.6) years. A minority of men (14.6%) were cognitively impaired at baseline (Mini-Mental State Examination score <28/30). A higher percentage of light sleep was associated with better Trail-Making Test A performance (B = -0.04, 95% confidence interval [CI] -0.06, -0.01; p = 0.003), whereas higher mean oxygen saturation was associated with worse performance (B = 0.11, 95% CI 0.02, 0.19; p = 0.012). While obstructive sleep apnea and sleep macroarchitecture might predict cognitive decline, future studies should consider arousal events and non-routine hypoxaemia measures, which may show associations with cognitive decline.

16.
Sleep Med ; 110: 44-53, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37536211

RESUMO

BACKGROUND: In clinical populations, the movement of cerebrospinal fluid (CSF) during sleep is a growing area of research with potential mechanistic connections in both neurodegenerative (e.g., Alzheimer's Disease) and neurodevelopmental disorders. However, we know relatively little about the processes that influence CSF movement. To inform clinical intervention targets this study assesses the coupling between (a) real-time CSF movement, (b) neuronal-driven movement, and (c) non-neuronal systemic physiology driven movement. METHODS: This study included eight young, healthy volunteers, with concurrently acquired neurofluid dynamics using functional Magnetic Resonance Imaging (MRI), neural activity using Electroencephalography (EEG), and non-neuronal systemic physiology with peripheral functional Near-Infrared Spectroscopy (fNIRS). Neuronal and non-neuronal drivers were assessed temporally; wherein, EEG measured slow wave activity that preceded CSF movement was considered neuronally driven. Similarly, slow wave oscillations (assessed via fNIRS) that coupled with CSF movement were considered non-neuronal systemic physiology driven. RESULTS AND CONCLUSIONS: Our results document neural contributions to CSF movement were only present during light NREM sleep but low-frequency non-neuronal oscillations were strongly coupled with CSF movement in all assessed states - awake, NREM-1, NREM-2. The clinical/research implications of these findings are two-fold. First, neuronal-driven oscillations contribute to CSF movement outside of deep sleep (NREM-3); therefore, interventions aimed at increasing CSF movement may yield meaningful increases with the promotion of NREM sleep more generally - a focus on NREM S3 may not be needed. Second, non-neuronal systemic oscillations contribute across wake and sleep stages; therefore, interventions may increase CSF movement by manipulating systemic physiology.


Assuntos
Eletroencefalografia , Sono , Humanos , Sono/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Neurônios
17.
Biomed Pharmacother ; 165: 115259, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37531785

RESUMO

Sleep disturbances are commonly non-motor symptoms in Parkinson's diseases (PD). However, standard dopamine replacement therapies for the treatment of motor symptoms often prove inadequate in combating sleep disturbances. Previous studies conducted by our research group have reported the neuroprotective effects of tenuigenin, a natural extract from Polygala tenuifolia root, which has been traditionally employed in treating insomnia. The objective of this study was to investigate the potential of tenuigenin in modulating sleep-wake behaviors and elucidate the underlying mechanisms. We employed EEG/EMG recordings to evaluate the impact of tenuigenin on sleep-wake profiles. Furthermore, we utilized c-Fos immunostaining, whole-cell patch clamping and local field potentials (LFP) recording to explore the mechanisms involved in sleep-promoting effects of tenuigenin. Additionally, we examined the effects of tenuigenin on sleep-promoting in MPTP PD mice. Here, we found tenuigenin demonstrated a significant increase in NREM sleep and a reduction in sleep latency in mice, without altering the EEG power density. Moreover, tenuigenin increased c-Fos expression in the ventrolateral preoptic area (VLPO) and stimulated sleep-promoting neurons in VLPO. The sleep-promoting effects of tenuigenin were abolished when mice were pretreated with flumazenil, an antagonist at the benzodiazepine site of the GABAA receptor. Furthermore, tenuigenin was found to ameliorate sleep disturbances in MPTP-induced mice. The results suggesting that tenuigenin facilitated a type of NREM sleep comparable to physiological NREM sleep through interaction with the GABAA receptor. Additionally, tenuigenin demonstrated improvements in sleep disturbances in MPTP-induced PD mice, suggesting its potential as a sleep-promoting substance, particularly for PD patients experiencing sleep disturbances.


Assuntos
Doença de Parkinson , Receptores de GABA-A , Camundongos , Animais , Doença de Parkinson/tratamento farmacológico , Movimentos Oculares , Sono , Eletroencefalografia
18.
Sleep ; 46(9)2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37478470

RESUMO

Ponto-geniculo-occipital or pontine (P) waves have long been recognized as an electrophysiological signature of rapid eye movement (REM) sleep. However, P-waves can be observed not just during REM sleep, but also during non-REM (NREM) sleep. Recent studies have uncovered that P-waves are functionally coupled with hippocampal sharp wave ripples (SWRs) during NREM sleep. However, it remains unclear to what extent P-waves during NREM sleep share their characteristics with P-waves during REM sleep and how the functional coupling to P-waves modulates SWRs. Here, we address these issues by performing multiple types of electrophysiological recordings and fiber photometry in both sexes of mice. P-waves during NREM sleep share their waveform shapes and local neural ensemble dynamics at a short (~100 milliseconds) timescale with their REM sleep counterparts. However, the dynamics of mesopontine cholinergic neurons are distinct at a longer (~10 seconds) timescale: although P-waves are accompanied by cholinergic transients, the cholinergic tone gradually reduces before P-wave genesis during NREM sleep. While P-waves are coupled to hippocampal theta rhythms during REM sleep, P-waves during NREM sleep are accompanied by a rapid reduction in hippocampal ripple power. SWRs coupled with P-waves are short-lived and hippocampal neural firing is also reduced after P-waves. These results demonstrate that P-waves are part of coordinated sleep-related activity by functionally coupling with hippocampal ensembles in a state-dependent manner.


Assuntos
Movimentos Oculares , Lobo Occipital , Masculino , Feminino , Animais , Camundongos , Lobo Occipital/fisiologia , Corpos Geniculados/fisiologia , Sono/fisiologia , Hipocampo/fisiologia , Ponte/fisiologia
19.
Cell Rep Med ; 4(7): 101100, 2023 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-37421946

RESUMO

Insufficient sleep impairs glucose regulation, increasing the risk of diabetes. However, what it is about the human sleeping brain that regulates blood sugar remains unknown. In an examination of over 600 humans, we demonstrate that the coupling of non-rapid eye movement (NREM) sleep spindles and slow oscillations the night before is associated with improved next-day peripheral glucose control. We further show that this sleep-associated glucose pathway may influence glycemic status through altered insulin sensitivity, rather than through altered pancreatic beta cell function. Moreover, we replicate these associations in an independent dataset of over 1,900 adults. Of therapeutic significance, the coupling between slow oscillations and spindles was the most significant sleep predictor of next-day fasting glucose, even more so than traditional sleep markers, relevant to the possibility of an electroencephalogram (EEG) index of hyperglycemia. Taken together, these findings describe a sleeping-brain-body framework of optimal human glucose homeostasis, offering a potential prognostic sleep signature of glycemic control.


Assuntos
Ondas Encefálicas , Sono , Adulto , Humanos , Sono/fisiologia , Eletroencefalografia , Glucose , Homeostase
20.
Neurobiol Stress ; 24: 100543, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37252645

RESUMO

Sleep is a vital and evolutionarily conserved process, critical to daily functioning and homeostatic balance. Losing sleep is inherently stressful and leads to numerous detrimental physiological outcomes. Despite sleep disturbances affecting everyone, women and female rodents are often excluded or underrepresented in clinical and pre-clinical studies. Advancing our understanding of the role of biological sex in the responses to sleep loss stands to greatly improve our ability to understand and treat health consequences of insufficient sleep. As such, this review discusses sex differences in response to sleep deprivation, with a focus on the sympathetic nervous system stress response and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We review sex differences in several stress-related consequences of sleep loss, including inflammation, learning and memory deficits, and mood related changes. Focusing on women's health, we discuss the effects of sleep deprivation during the peripartum period. In closing, we present neurobiological mechanisms, including the contribution of sex hormones, orexins, circadian timing systems, and astrocytic neuromodulation, that may underlie potential sex differences in sleep deprivation responses.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA