Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
Nat Commun ; 15(1): 3661, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38688901

ABSTRACT

Optochemistry, an emerging pharmacologic approach in which light is used to selectively activate or deactivate molecules, has the potential to alleviate symptoms, cure diseases, and improve quality of life while preventing uncontrolled drug effects. The development of in-vivo applications for optochemistry to render brain cells photoresponsive without relying on genetic engineering has been progressing slowly. The nucleus accumbens (NAc) is a region for the regulation of slow-wave sleep (SWS) through the integration of motivational stimuli. Adenosine emerges as a promising candidate molecule for activating indirect pathway neurons of the NAc expressing adenosine A2A receptors (A2ARs) to induce SWS. Here, we developed a brain-permeable positive allosteric modulator of A2ARs (A2AR PAM) that can be rapidly photoactivated with visible light (λ > 400 nm) and used it optoallosterically to induce SWS in the NAc of freely behaving male mice by increasing the activity of extracellular adenosine derived from astrocytic and neuronal activity.


Subject(s)
Adenosine , Nucleus Accumbens , Receptor, Adenosine A2A , Sleep, Slow-Wave , Animals , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiology , Male , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2A/genetics , Mice , Adenosine/metabolism , Adenosine/pharmacology , Allosteric Regulation , Sleep, Slow-Wave/physiology , Sleep, Slow-Wave/drug effects , Astrocytes/metabolism , Astrocytes/drug effects , Light , Neurons/metabolism , Neurons/drug effects , Mice, Inbred C57BL , Humans , Adenosine A2 Receptor Agonists/pharmacology
2.
Article in English | MEDLINE | ID: mdl-38683718

ABSTRACT

Sleep is vital to our daily activity. Lack of proper sleep can impair functionality and overall health. While stress is known for its detrimental impact on sleep quality, the precise effect of pre-sleep stress on subsequent sleep structure remains unknown. This study introduced a novel approach to study the pre-sleep stress effect on sleep structure, specifically slow-wave sleep (SWS) deficiency. To achieve this, we selected forehead resting EEG immediately before and upon sleep onset to extract stress-related neurological markers through power spectra and entropy analysis. These markers include beta/delta correlation, alpha asymmetry, fuzzy entropy (FuzzEn) and spectral entropy (SpEn). Fifteen subjects were included in this study. Our results showed that subjects lacking SWS often exhibited signs of stress in EEG, such as an increased beta/delta correlation, higher alpha asymmetry, and increased FuzzEn in frontal EEG. Conversely, individuals with ample SWS displayed a weak beta/delta correlation and reduced FuzzEn. Finally, we employed several supervised learning models and found that the selected neurological markers can predict subsequent SWS deficiency. Our investigation demonstrated that the classifiers could effectively predict varying levels of slow-wave sleep (SWS) from pre-sleep EEG segments, achieving a mean balanced accuracy surpassing 0.75. The SMOTE-Tomek resampling method could improve the performance to 0.77. This study suggests that stress-related neurological markers derived from pre-sleep EEG can effectively predict SWS deficiency. Such information can be integrated with existing sleep-improving techniques to provide a personalized sleep forecasting and improvement solution.


Subject(s)
Algorithms , Electroencephalography , Entropy , Sleep, Slow-Wave , Humans , Electroencephalography/methods , Male , Female , Sleep, Slow-Wave/physiology , Adult , Young Adult , Stress, Psychological/physiopathology , Alpha Rhythm/physiology , Forecasting , Beta Rhythm/physiology , Delta Rhythm , Sleep Deprivation/physiopathology , Reproducibility of Results
3.
Elife ; 122024 Apr 25.
Article in English | MEDLINE | ID: mdl-38661727

ABSTRACT

We are unresponsive during slow-wave sleep but continue monitoring external events for survival. Our brain wakens us when danger is imminent. If events are non-threatening, our brain might store them for later consideration to improve decision-making. To test this hypothesis, we examined whether novel vocabulary consisting of simultaneously played pseudowords and translation words are encoded/stored during sleep, and which neural-electrical events facilitate encoding/storage. An algorithm for brain-state-dependent stimulation selectively targeted word pairs to slow-wave peaks or troughs. Retrieval tests were given 12 and 36 hr later. These tests required decisions regarding the semantic category of previously sleep-played pseudowords. The sleep-played vocabulary influenced awake decision-making 36 hr later, if targeted to troughs. The words' linguistic processing raised neural complexity. The words' semantic-associative encoding was supported by increased theta power during the ensuing peak. Fast-spindle power ramped up during a second peak likely aiding consolidation. Hence, new vocabulary played during slow-wave sleep was stored and influenced decision-making days later.


Subject(s)
Memory, Long-Term , Sleep, Slow-Wave , Humans , Sleep, Slow-Wave/physiology , Male , Female , Memory, Long-Term/physiology , Adult , Young Adult , Brain/physiology , Decision Making/physiology , Vocabulary , Electroencephalography
4.
J Neurosci Res ; 102(4): e25325, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38562056

ABSTRACT

Brain states (wake, sleep, general anesthesia, etc.) are profoundly associated with the spatiotemporal dynamics of brain oscillations. Previous studies showed that the EEG alpha power shifted from the occipital cortex to the frontal cortex (alpha anteriorization) after being induced into a state of general anesthesia via propofol. The sleep research literature suggests that slow waves and sleep spindles are generated locally and propagated gradually to different brain regions. Since sleep and general anesthesia are conceptualized under the same framework of consciousness, the present study examines whether alpha anteriorization similarly occurs during sleep and how the EEG power in other frequency bands changes during different sleep stages. The results from the analysis of three polysomnography datasets of 234 participants show consistent alpha anteriorization during the sleep stages N2 and N3, beta anteriorization during stage REM, and theta posteriorization during stages N2 and N3. Although it is known that the neural circuits responsible for sleep are not exactly the same for general anesthesia, the findings of alpha anteriorization in this study suggest that, at macro level, the circuits for alpha oscillations are organized in the similar cortical areas. The spatial shifts of EEG power in different frequency bands during sleep may offer meaningful neurophysiological markers for the level of consciousness.


Subject(s)
Electroencephalography , Sleep, Slow-Wave , Humans , Electroencephalography/methods , Sleep, Slow-Wave/physiology , Sleep/physiology , Sleep Stages/physiology , Polysomnography
5.
Sleep ; 47(5)2024 May 10.
Article in English | MEDLINE | ID: mdl-38452190

ABSTRACT

STUDY OBJECTIVES: Sleep supports systems memory consolidation through the precise temporal coordination of specific oscillatory events during slow-wave sleep, i.e. the neocortical slow oscillations (SOs), thalamic spindles, and hippocampal ripples. Beneficial effects of sleep on memory are also observed in infants, although the contributing regions, especially hippocampus and frontal cortex, are immature. Here, we examined in rats the development of these oscillatory events and their coupling during early life. METHODS: EEG and hippocampal local field potentials were recorded during sleep in male rats at postnatal days (PD)26 and 32, roughly corresponding to early (1-2 years) and late (9-10 years) human childhood, and in a group of adult rats (14-18 weeks, corresponding to ~22-29 years in humans). RESULTS: SO and spindle amplitudes generally increased from PD26 to PD32. In parallel, frontocortical EEG spindles increased in density and frequency, while changes in hippocampal ripples remained nonsignificant. The proportion of SOs co-occurring with spindles also increased from PD26 to PD32. Whereas parietal cortical spindles were phase-locked to the depolarizing SO-upstate already at PD26, over frontal cortex SO-spindle phase-locking emerged not until PD32. Co-occurrence of hippocampal ripples with spindles was higher during childhood than in adult rats, but significant phase-locking of ripples to the excitable spindle troughs was observed only in adult rats. CONCLUSIONS: Results indicate a protracted development of synchronized thalamocortical processing specifically in frontocortical networks (i.e. frontal SO-spindle coupling). However, synchronization within thalamocortical networks generally precedes synchronization of thalamocortical with hippocampal processing as reflected by the delayed occurrence of spindle-ripple phase-coupling.


Subject(s)
Electroencephalography , Hippocampus , Animals , Rats , Male , Hippocampus/physiology , Thalamus/physiology , Neocortex/physiology , Sleep/physiology , Sleep, Slow-Wave/physiology , Brain Waves/physiology
6.
Commun Biol ; 7(1): 288, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38459227

ABSTRACT

Sleep boosts the integration of memories, and can thus facilitate relational learning. This benefit may be due to memory reactivation during non-REM sleep. We set out to test this by explicitly cueing reactivation using a technique called targeted memory reactivation (TMR), in which sounds are paired with learned material in wake and then softly played during subsequent sleep, triggering reactivation of the associated memories. We specifically tested whether TMR in slow wave sleep leads to enhancements in inferential thinking in a transitive inference task. Because the Up-phase of the slow oscillation is more responsive to cues than the Down-phase, we also asked whether Up-phase stimulation is more beneficial for such integration. Our data show that TMR during the Up-Phase boosts the ability to make inferences, but only for the most distant inferential leaps. Up-phase stimulation was also associated with detectable memory reinstatement, whereas Down-phase stimulation led to below-chance performance the next morning. Detection of memory reinstatement after Up-state stimulation was negatively correlated with performance on the most difficult inferences the next morning. These findings demonstrate that cueing memory reactivation at specific time points in sleep can benefit difficult relational learning problems.


Subject(s)
Sleep, Slow-Wave , Humans , Sleep, Slow-Wave/physiology , Learning/physiology , Sleep/physiology , Cues , Sound
7.
Eur J Neurosci ; 59(4): 584-594, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37038095

ABSTRACT

Besides regulating the amount of light that reaches the retina, fluctuations in pupil size also occur in isoluminant conditions during accommodation, during movement and in relation to cognitive workload, attention and emotion. Recent studies in mammals and birds revealed that the pupils are also highly dynamic in the dark during sleep. However, despite exhibiting similar sleep states (rapid eye movement [REM] and non-REM [NREM] sleep), wake and sleep state-dependent changes in pupil size are opposite between mammals and birds, due in part to differences in the type (striated vs. smooth) and control of the iris muscles. Given the link between pupil dynamics and cognitive processes occurring during wakefulness, sleep-related changes in pupil size might indicate when related processes are occurring during sleep. Moreover, the divergent pupillary behaviour observed between mammals and birds raises the possibility that changes in pupil size in birds are a readout of processes not reflected in the mammalian pupil.


Subject(s)
Sleep, Slow-Wave , Wakefulness , Animals , Wakefulness/physiology , Sleep/physiology , Sleep, REM/physiology , Sleep, Slow-Wave/physiology , Mammals , Electroencephalography
8.
Science ; 382(6669): 405-412, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37883555

ABSTRACT

Neural substrates of wakefulness, rapid eye movement sleep (REMS), and non-REMS (NREMS) in the mammalian hypothalamus overlap both anatomically and functionally with cellular networks that support physiological and behavioral homeostasis. Here, we review the roles of sleep neurons of the hypothalamus in the homeostatic control of thermoregulation or goal-oriented behaviors during wakefulness. We address how hypothalamic circuits involved in opposing behaviors such as core body temperature and sleep compute conflicting information and provide a coherent vigilance state. Finally, we highlight some of the key unresolved questions and challenges, and the promise of a more granular view of the cellular and molecular diversity underlying the integrative role of the hypothalamus in physiological and behavioral homeostasis.


Subject(s)
Hypothalamus , Neurons , Sleep, REM , Sleep, Slow-Wave , Wakefulness , Animals , Body Temperature Regulation , Electroencephalography , Hypothalamus/cytology , Hypothalamus/physiology , Sleep, REM/physiology , Wakefulness/physiology , Humans , Neurons/physiology , Sleep, Slow-Wave/physiology
9.
J Neural Eng ; 20(5)2023 10 05.
Article in English | MEDLINE | ID: mdl-37726002

ABSTRACT

Objective.Healthy sleep plays a critical role in general well-being. Enhancement of slow-wave sleep by targeting acoustic stimuli to particular phases of delta (0.5-2 Hz) waves has shown promise as a non-invasive approach to improve sleep quality. Closed-loop stimulation during other sleep phases targeting oscillations at higher frequencies such as theta (4-7 Hz) or alpha (8-12 Hz) could be another approach to realize additional health benefits. However, systems to track and deliver stimulation relative to the instantaneous phase of electroencephalogram (EEG) signals at these higher frequencies have yet to be demonstrated outside of controlled laboratory settings.Approach.Here we examine the feasibility of using an endpoint-corrected version of the Hilbert transform (ecHT) algorithm implemented on a headband wearable device to measure alpha phase and deliver phase-locked auditory stimulation during the transition from wakefulness to sleep, during which alpha power is greatest. First, the ecHT algorithm is implementedin silicoto evaluate the performance characteristics of this algorithm across a range of sleep-related oscillatory frequencies. Secondly, a pilot sleep study tests feasibility to use the wearable device by users in the home setting for measurement of EEG activity during sleep and delivery of real-time phase-locked stimulation.Main results.The ecHT is capable of computing the instantaneous phase of oscillating signals with high precision, allowing auditory stimulation to be delivered at the intended phases of neural oscillations with low phase error. The wearable system was capable of measuring sleep-related neural activity with sufficient fidelity for sleep stage scoring during the at-home study, and phase-tracking performance matched simulated results. Users were able to successfully operate the system independently using the companion smartphone app to collect data and administer stimulation, and presentation of auditory stimuli during sleep initiation did not negatively impact sleep onset.Significance.This study demonstrates the feasibility of closed-loop real-time tracking and neuromodulation of a range of sleep-related oscillations using a wearable EEG device. Preliminary results suggest that this approach could be used to deliver non-invasive neuromodulation across all phases of sleep.


Subject(s)
Electroencephalography , Sleep, Slow-Wave , Electroencephalography/methods , Sleep/physiology , Sleep, Slow-Wave/physiology , Sleep Stages/physiology , Acoustic Stimulation/methods
10.
Cell Rep ; 42(5): 112450, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37126447

ABSTRACT

Sleep consists of two basic stages: non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. NREM sleep is characterized by slow high-amplitude cortical electroencephalogram (EEG) signals, while REM sleep is characterized by desynchronized cortical rhythms. Despite this, recent electrophysiological studies have suggested the presence of slow waves (SWs) in local cortical areas during REM sleep. Electrophysiological techniques, however, have been unable to resolve the regional structure of these activities because of relatively sparse sampling. Here, we map functional gradients in cortical activity during REM sleep using mesoscale imaging in mice and show local SW patterns occurring mainly in somatomotor and auditory cortical regions with minimum presence within the default mode network. The role of the cholinergic system in local desynchronization during REM sleep is also explored by calcium imaging of cholinergic activity within the cortex and analyzing structural data. We demonstrate weaker cholinergic projections and terminal activity in regions exhibiting frequent SWs during REM sleep.


Subject(s)
Auditory Cortex , Sleep, Slow-Wave , Mice , Animals , Sleep, REM/physiology , Electroencephalography/methods , Sleep , Sleep, Slow-Wave/physiology
11.
J Clin Endocrinol Metab ; 108(10): e1044-e1055, 2023 09 18.
Article in English | MEDLINE | ID: mdl-37084404

ABSTRACT

CONTEXT: N3 sleep (i.e., slow-wave sleep), a marker of deep restorative sleep, is implicated in hormonal and blood pressure regulation and may impact cardiometabolic health. OBJECTIVE: We conducted cross-sectional and prospective analyses to test whether a higher proportion and longer duration of N3 sleep are associated with reduced type 2 diabetes risk. METHODS: A subsample of participants from the Multi-Ethnic Study of Atherosclerosis completed 1-night polysomnography at Exam 5 (2010-2013) and were prospectively followed until Exam 6 (2016-2018). We used modified Poisson regression to examine the cross-sectional associations of N3 proportion and duration with prevalent diabetes and Cox proportional hazards models to estimate risk of diabetes according to N3 measures. RESULTS: In cross-sectional analyses (n = 2026, mean age: 69 years), diabetes prevalence was 28% (n = 572). Compared with the first quartile (Q1) of the N3 proportion (<2.0%), participants in Q4 (≥15.4%) were 29% (95% CI 0.58, 0.87) less likely to have prevalent diabetes (P trend = .0016). The association attenuated after adjustment for demographics, lifestyles, and sleep-related factors (P trend = .3322). In prospective analyses of 1251 participants and 129 incident cases over 6346 person-years of follow-up, a curvilinear relationship was observed between N3 proportion and incident diabetes risk. In the fully adjusted model, the hazard ratio (95% CI) of developing diabetes vs Q1 was 0.47 (0.26, 0.87) for Q2, 0.34 (0.15, 0.77) for Q3, and 0.32 (0.10, 0.97) for Q4 (P nonlinearity = .0213). The results were similar for N3 duration. CONCLUSION: Higher N3 proportion and longer N3 duration were prospectively associated with lower type 2 diabetes risk in a nonlinear fashion among older American adults.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Type 2 , Sleep, Slow-Wave , Adult , Humans , Aged , Sleep, Slow-Wave/physiology , Prospective Studies , Cross-Sectional Studies , Sleep/physiology , Risk Factors
12.
Am J Geriatr Psychiatry ; 31(8): 643-652, 2023 08.
Article in English | MEDLINE | ID: mdl-37105885

ABSTRACT

Slow wave sleep (SWS), characterized by large electroencephalographic oscillations, facilitates crucial physiologic processes that maintain synaptic plasticity and overall brain health. Deficiency in older adults is associated with depression and cognitive dysfunction, such that enhancing sleep slow waves has emerged as a promising target for novel therapies. Enhancement of SWS has been noted after infusions of propofol, a commonly used anesthetic that induces electroencephalographic patterns resembling non-rapid eye movement sleep. This paper 1) reviews the scientific premise underlying the hypothesis that sleep slow waves are a novel therapeutic target for improving cognitive and psychiatric outcomes in older adults, and 2) presents a case series of two patients with late-life depression who each received two propofol infusions. One participant, a 71-year-old woman, had a mean of 2.8 minutes of evening SWS prior to infusions (0.7% of total sleep time). SWS increased on the night after each infusion, to 12.5 minutes (5.3% of total sleep time) and 24 minutes (10.6% of total sleep time), respectively. Her depression symptoms improved, reflected by a reduction in her Montgomery-Asberg Depression Rating Scale (MADRS) score from 26 to 7. In contrast, the other participant, a 77-year-old man, exhibited no SWS at baseline and only modest enhancement after the second infusion (3 minutes, 1.3% of total sleep time). His MADRS score increased from 13 to 19, indicating a lack of improvement in his depression. These cases provide proof-of-concept that propofol can enhance SWS and improve depression for some individuals, motivating an ongoing clinical trial (ClinicalTrials.gov NCT04680910).


Subject(s)
Propofol , Sleep, Slow-Wave , Humans , Male , Female , Aged , Sleep, Slow-Wave/physiology , Propofol/pharmacology , Propofol/therapeutic use , Depression/complications , Depression/drug therapy , Sleep/physiology , Brain , Electroencephalography
13.
Nature ; 616(7956): 312-318, 2023 04.
Article in English | MEDLINE | ID: mdl-36949193

ABSTRACT

Our understanding of the functions and mechanisms of sleep remains incomplete, reflecting their increasingly evident complexity1-3. Likewise, studies of interhemispheric coordination during sleep4-6 are often hard to connect precisely to known sleep circuits and mechanisms. Here, by recording from the claustra of sleeping bearded dragons (Pogona vitticeps), we show that, although the onsets and offsets of Pogona rapid-eye-movement (REMP) and slow-wave sleep are coordinated bilaterally, these two sleep states differ markedly in their inter-claustral coordination. During slow-wave sleep, the claustra produce sharp-wave ripples independently of one another, showing no coordination. By contrast, during REMP sleep, the potentials produced by the two claustra are precisely coordinated in amplitude and time. These signals, however, are not synchronous: one side leads the other by about 20 ms, with the leading side switching typically once per REMP episode or in between successive episodes. The leading claustrum expresses the stronger activity, suggesting bilateral competition. This competition does not occur directly between the two claustra or telencephalic hemispheres. Rather, it occurs in the midbrain and depends on the integrity of a GABAergic (γ-aminobutyric-acid-producing) nucleus of the isthmic complex, which exists in all vertebrates and is known in birds to underlie bottom-up attention and gaze control. These results reveal that a winner-take-all-type competition exists between the two sides of the brain of Pogona, which originates in the midbrain and has precise consequences for claustrum activity and coordination during REMP sleep.


Subject(s)
Brain , Functional Laterality , Lizards , Sleep , Animals , Brain/anatomy & histology , Brain/physiology , Lizards/anatomy & histology , Lizards/physiology , Mesencephalon/physiology , Sleep/physiology , Sleep, REM/physiology , Sleep, Slow-Wave/physiology , Functional Laterality/physiology , Time Factors , gamma-Aminobutyric Acid/metabolism , Fixation, Ocular , Attention , Birds/physiology
14.
J Sleep Res ; 32(4): e13846, 2023 08.
Article in English | MEDLINE | ID: mdl-36806335

ABSTRACT

Slow-wave sleep (SWS) is a fundamental physiological process, and its modulation is of interest for basic science and clinical applications. However, automatised protocols for the suppression of SWS are lacking. We describe the development of a novel protocol for the automated detection (based on the whole head topography of frontal slow waves) and suppression of SWS (through closed-loop modulated randomised pulsed noise), and assessed the feasibility, efficacy and functional relevance compared to sham stimulation in 15 healthy young adults in a repeated-measure sleep laboratory study. Auditory compared to sham stimulation resulted in a highly significant reduction of SWS by 30% without affecting total sleep time. The reduction of SWS was associated with an increase in lighter non-rapid eye movement sleep and a shift of slow-wave activity towards the end of the night, indicative of a homeostatic response and functional relevance. Still, cumulative slow-wave activity across the night was significantly reduced by 23%. Undisturbed sleep led to an evening to morning reduction of wake electroencephalographic theta activity, thought to reflect synaptic downscaling during SWS, while suppression of SWS inhibited this dissipation. We provide evidence for the feasibility, efficacy, and functional relevance of a novel fully automated protocol for SWS suppression based on auditory closed-loop stimulation. Future work is needed to further test for functional relevance and potential clinical applications.


Subject(s)
Sleep, Slow-Wave , Young Adult , Humans , Sleep, Slow-Wave/physiology , Feasibility Studies , Sleep/physiology , Polysomnography , Electroencephalography/methods , Acoustic Stimulation/methods
15.
Epilepsy Behav ; 140: 109105, 2023 03.
Article in English | MEDLINE | ID: mdl-36758358

ABSTRACT

Encephalopathy related to Status Epilepticus during slow Sleep (ESES) is a childhood epilepsy syndrome characterized by the appearance of cognitive, behavioral, and motor disturbances in conjunction with a striking activation of EEG epileptic abnormalities during non-REM sleep. After more than 50 years since the first description, the pathophysiological mechanisms underlying the appearance of encephalopathy in association with a sleep-related enhancement of epileptic discharges are incompletely elucidated. Recent experimental data support the hypothesis that the development of the ESES encephalopathic picture depends on a spike-induced impairment of the synaptic homeostasis processes occurring during normal sleep and that is particularly pronounced during the developmental age. During sleep, synaptic homeostasis is promoted by synaptic weakening/elimination after the increment of synaptic strength that occurs during wakefulness. The EEG can display modifications in synaptic strength by changes in sleep slow wave activity (SWA). Recent studies during active ESES have failed to show changes in sleep SWA, while these changes occurred again after recovery from ESES, thus supporting a spike-related interference on the normal homeostatic processes of sleep. This impairment, during the developmental period, can lead to disruption of cortical wiring and brain plastic remodeling, which lead to the, often irreversible, neuropsychological compromise typical of ESES. From the nosographic point of view, these pathophysiological data lend support to the maintenance of the term ESES, i.e., "encephalopathy related to status epilepticus during sleep". Indeed, this term conveys the concept that the extreme activation of epileptic discharges during sleep is directly responsible for the encephalopathy, hence the importance of defining this condition as an encephalopathy related to the exaggerated activation of epileptic activity during sleep. In this respect, ESES represents a genuine example of a "pure" epileptic encephalopathy in which sleep-related epileptic activity "per se" has a crucial role in determining the encephalopathic picture. This paper was presented at the 8th London-Innsbruck Colloquium on Status Epilepticus and Acute Seizures held in September 2022.


Subject(s)
Brain Diseases , Epilepsy , Sleep, Slow-Wave , Status Epilepticus , Humans , Child , Electroencephalography/methods , Sleep, Slow-Wave/physiology , Brain Diseases/complications , Epilepsy/complications , Sleep/physiology , Status Epilepticus/complications
16.
J Sleep Res ; 32(4): e13818, 2023 08.
Article in English | MEDLINE | ID: mdl-36631001

ABSTRACT

Dementia is the seventh leading cause of mortality, and a major source of disability and dependency in older individuals globally. Cognitive decline (and, to a lesser extent, normal ageing) are associated with sleep fragmentation and loss of slow-wave sleep. Evidence suggests a bidirectional causal link between these losses. Phase-locked auditory stimulation has emerged as a promising non-invasive tool to enhance slow-wave sleep, potentially ameliorating cognitive decline. In laboratory settings, auditory stimulation is usually supervised by trained experts. Different algorithms (simple amplitude thresholds, topographic correlation, sine-wave fitting, phase-locked loop, and phase vocoder) are used to precisely target auditory stimulation to a desired phase of the slow wave. While all algorithms work well in younger adults, the altered sleep physiology of older adults and particularly those with neurodegenerative disorders requires a tailored approach that can adapt to older adults' fragmented sleep and reduced amplitudes of slow waves. Moreover, older adults might require a continuous intervention that is not feasible in laboratory settings. Recently, several auditory stimulation-capable portable devices ('Dreem®', 'SmartSleep®' and 'SleepLoop®') have been developed. We discuss these three devices regarding their potential as tools for science, and as clinical remote-intervention tools to combat cognitive decline. Currently, SleepLoop® shows the most promise for scientific research in older adults due to high transparency and customizability but is not commercially available. Studies evaluating down-stream effects on cognitive abilities, especially in patient populations, are required before a portable auditory stimulation device can be recommended as a clinical preventative remote-intervention tool.


Subject(s)
Cognitive Dysfunction , Sleep, Slow-Wave , Humans , Aged , Sleep, Slow-Wave/physiology , Acoustic Stimulation , Electroencephalography , Sleep/physiology , Cognitive Dysfunction/prevention & control
17.
Neurosci Res ; 189: 37-43, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36584925

ABSTRACT

Evidence is accumulating that the brain actively consolidates long-term memory during sleep. Motor skill memory is a form of non-declarative procedural memory and can be coordinated with multi-sensory processing such as visual, tactile, and, auditory. Conversely, perception is affected by body movement signal from motor brain regions. Although both cortical and subcortical brain regions are involved in memory consolidation, cerebral cortex activity can be recorded and manipulated noninvasively or minimally invasively in humans and animals. NREM sleep, which is important for non-declarative memory consolidation, is characterized by slow and spindle waves representing thalamo-cortical population activity. In animals, electrophysiological recording, optical imaging, and manipulation approaches have revealed multi-scale cortical dynamics across learning and sleep. In the sleeping cortex, neural activity is affected by prior learning and neural circuits are continually reorganized. Here I outline how sensorimotor coordination is formed through awake learning and subsequent sleep.


Subject(s)
Memory Consolidation , Sleep, Slow-Wave , Humans , Animals , Memory Consolidation/physiology , Sleep/physiology , Brain/physiology , Learning/physiology , Sleep, Slow-Wave/physiology
18.
Cereb Cortex ; 33(9): 5238-5250, 2023 04 25.
Article in English | MEDLINE | ID: mdl-36376911

ABSTRACT

Previous studies have shown that modulating neural activity can affect rapid eye movement (REM) and non-rapid eye movement (NREM) sleep. Low-intensity transcranial ultrasound stimulation (TUS) can effectively modulate neural activity. However, the modulation effect of TUS on REM and NREM sleep is still unclear. In this study, we used ultrasound to stimulate motor cortex and hippocampus, respectively, and found the following: (i) In healthy mice, TUS increased the NREM sleep ratio and decreased the REM sleep ratio, and altered the relative power and sample entropy of the delta band and spindle in NREM sleep and that of the theta and gamma bands in REM sleep. (ii) In sleep-deprived mice, TUS decreased the ratio of REM sleep or the relative power of the theta band during REM sleep. (iii) In sleep-disordered Alzheimer's disease (AD) mice, TUS increased the total sleep time and the ratio of NREM sleep and modulated the relative power and the sample entropy of the delta and spindle bands during NREM and that of the theta band during REM sleep. These results demonstrated that TUS can effectively modulate REM and NREM sleep and that modulation effect depends on the sleep state of the samples, and can improve sleep in sleep-disordered AD mice.


Subject(s)
Sleep, REM , Sleep, Slow-Wave , Mice , Animals , Sleep, REM/physiology , Electroencephalography/methods , Sleep/physiology , Sleep, Slow-Wave/physiology , Hippocampus/physiology
19.
Nature ; 612(7940): 512-518, 2022 12.
Article in English | MEDLINE | ID: mdl-36477539

ABSTRACT

Progress has been made in the elucidation of sleep and wakefulness regulation at the neurocircuit level1,2. However, the intracellular signalling pathways that regulate sleep and the neuron groups in which these intracellular mechanisms work remain largely unknown. Here, using a forward genetics approach in mice, we identify histone deacetylase 4 (HDAC4) as a sleep-regulating molecule. Haploinsufficiency of Hdac4, a substrate of salt-inducible kinase 3 (SIK3)3, increased sleep. By contrast, mice that lacked SIK3 or its upstream kinase LKB1 in neurons or with a Hdac4S245A mutation that confers resistance to phosphorylation by SIK3 showed decreased sleep. These findings indicate that LKB1-SIK3-HDAC4 constitute a signalling cascade that regulates sleep and wakefulness. We also performed targeted manipulation of SIK3 and HDAC4 in specific neurons and brain regions. This showed that SIK3 signalling in excitatory neurons located in the cerebral cortex and the hypothalamus positively regulates EEG delta power during non-rapid eye movement sleep (NREMS) and NREMS amount, respectively. A subset of transcripts biased towards synaptic functions was commonly regulated in cortical glutamatergic neurons through the expression of a gain-of-function allele of Sik3 and through sleep deprivation. These findings suggest that NREMS quantity and depth are regulated by distinct groups of excitatory neurons through common intracellular signals. This study provides a basis for linking intracellular events and circuit-level mechanisms that control NREMS.


Subject(s)
Neurons , Sleep Duration , Sleep , Wakefulness , Animals , Mice , Electroencephalography , Neurons/metabolism , Neurons/physiology , Sleep/genetics , Sleep/physiology , Sleep Deprivation/genetics , Wakefulness/genetics , Wakefulness/physiology , Signal Transduction , Delta Rhythm , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Glutamic Acid/metabolism , Sleep, Slow-Wave/genetics , Sleep, Slow-Wave/physiology
20.
Neuroimage ; 264: 119682, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36240988

ABSTRACT

Slow-wave sleep is the deep non-rapid eye-movement (NREM) sleep stage that is most relevant for the recuperative function of sleep. Its defining property is the presence of slow oscillations (<2 Hz) in the scalp electroencephalogram (EEG). Slow oscillations are generated by a synchronous back and forth between highly active UP-states and silent DOWN-states in neocortical neurons. Growing evidence suggests that closed-loop sensory stimulation targeted at UP-states of EEG-defined slow oscillations can enhance the slow oscillatory activity, increase sleep depth, and boost sleep's recuperative functions. However, several studies failed to replicate such findings. Failed replications might be due to the use of conventional closed-loop stimulation algorithms that analyze the signal from one single electrode and thereby neglect the fact that slow oscillations vary with respect to their origins, distributions, and trajectories on the scalp. In particular, conventional algorithms nonspecifically target functionally heterogeneous UP-states of distinct origins. After all, slow oscillations at distinct sites of the scalp have been associated with distinct functions. Here we present a novel EEG-based closed-loop stimulation algorithm that allows targeting UP- and DOWN-states of distinct cerebral origins based on topographic analyses of the EEG: the topographic targeting of slow oscillations (TOPOSO) algorithm. We present evidence that the TOPOSO algorithm can detect and target local slow oscillations with specific, predefined voltage maps on the scalp in real-time. When compared to a more conventional, single-channel-based approach, TOPOSO leads to fewer but locally more specific stimulations in a simulation study. In a validation study with napping participants, TOPOSO targets auditory stimulation reliably at local UP-states over frontal, sensorimotor, and centro-parietal regions. Importantly, auditory stimulation temporarily enhanced the targeted local state. However, stimulation then elicited a standard frontal slow oscillation rather than local slow oscillations. The TOPOSO algorithm is suitable for the modulation and the study of the functions of local slow oscillations.


Subject(s)
Sleep, Slow-Wave , Humans , Sleep, Slow-Wave/physiology , Electroencephalography/methods , Sleep/physiology , Acoustic Stimulation , Neurons/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...