Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Physiol Rev ; 100(2): 805-868, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31804897

RESUMO

Sleep spindles are burstlike signals in the electroencephalogram (EEG) of the sleeping mammalian brain and electrical surface correlates of neuronal oscillations in thalamus. As one of the most inheritable sleep EEG signatures, sleep spindles probably reflect the strength and malleability of thalamocortical circuits that underlie individual cognitive profiles. We review the characteristics, organization, regulation, and origins of sleep spindles and their implication in non-rapid-eye-movement sleep (NREMS) and its functions, focusing on human and rodent. Spatially, sleep spindle-related neuronal activity appears on scales ranging from small thalamic circuits to functional cortical areas, and generates a cortical state favoring intracortical plasticity while limiting cortical output. Temporally, sleep spindles are discrete events, part of a continuous power band, and elements grouped on an infraslow time scale over which NREMS alternates between continuity and fragility. We synthesize diverse and seemingly unlinked functions of sleep spindles for sleep architecture, sensory processing, synaptic plasticity, memory formation, and cognitive abilities into a unifying sleep spindle concept, according to which sleep spindles 1) generate neural conditions of large-scale functional connectivity and plasticity that outlast their appearance as discrete EEG events, 2) appear preferentially in thalamic circuits engaged in learning and attention-based experience during wakefulness, and 3) enable a selective reactivation and routing of wake-instated neuronal traces between brain areas such as hippocampus and cortex. Their fine spatiotemporal organization reflects NREMS as a physiological state coordinated over brain and body and may indicate, if not anticipate and ultimately differentiate, pathologies in sleep and neurodevelopmental, -degenerative, and -psychiatric conditions.


Assuntos
Ondas Encefálicas , Encéfalo/fisiopatologia , Cognição , Doenças do Sistema Nervoso/fisiopatologia , Periodicidade , Fases do Sono , Transtornos do Sono-Vigília/fisiopatologia , Animais , Atenção , Encéfalo/metabolismo , Humanos , Inteligência , Memória , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/psicologia , Plasticidade Neuronal , Transtornos do Sono-Vigília/genética , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/psicologia , Fatores de Tempo
2.
Trends Genet ; 39(5): 338-339, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36858881

RESUMO

Distilling insomnia genome-wide association study (GWAS) variants, Palermo and colleagues identified several genes that participate in sleep regulation in two different model organisms. This workflow sets off an innovative strategy to extract biological relevance from large human genomic databases.


Assuntos
Estudo de Associação Genômica Ampla , Locos de Características Quantitativas , Humanos , Fenótipo , Sono/genética , Polimorfismo de Nucleotídeo Único/genética
3.
Biochem Biophys Res Commun ; 720: 150072, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38749187

RESUMO

The Eph receptor, a prototypically large receptor protein tyrosine kinase, interacts with ephrin ligands, forming a bidirectional signaling system that impacts diverse brain functions. Eph receptors and ephrins mediate forward and reverse signaling, affecting neurogenesis, axon guidance, and synaptic signaling. While mammalian studies have emphasized their roles in neurogenesis and synaptic plasticity, the Drosophila counterparts are less studied, especially in glial cells, despite structural similarities. Using RNAi to modulate Eph/ephrin expression in Drosophila neurons and glia, we studied their roles in brain development and sleep and circadian behavior. Knockdown of neuronal ephrin disrupted mushroom body development, while glial knockdown had minimal impact. Surprisingly, disrupting ephrin in neurons or glial cells altered sleep and circadian rhythms, indicating a direct involvement in these behaviors independent from developmental effects. Further analysis revealed distinct sleep phenotypes between neuronal and glial knockdowns, underscoring the intricate interplay within the neural circuits that govern behavior. Glia-specific knockdowns showed altered sleep patterns and reduced circadian rhythmicity, suggesting an intricate role of glia in sleep regulation. Our findings challenge simplistic models of Eph/ephrin signaling limited to neuron-glia communication and emphasize the complexity of the regulatory networks modulating behavior. Future investigations targeting specific glial subtypes will enhance our understanding of Eph/ephrin signaling's role in sleep regulation across species.


Assuntos
Ritmo Circadiano , Efrinas , Corpos Pedunculados , Neuroglia , Neurônios , Transdução de Sinais , Sono , Animais , Neuroglia/metabolismo , Sono/fisiologia , Sono/genética , Ritmo Circadiano/fisiologia , Neurônios/metabolismo , Efrinas/metabolismo , Efrinas/genética , Corpos Pedunculados/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Receptores da Família Eph/metabolismo , Receptores da Família Eph/genética , Drosophila melanogaster/metabolismo , Drosophila melanogaster/fisiologia , Drosophila melanogaster/genética , Drosophila/metabolismo
4.
J Sleep Res ; 33(2): e13936, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37217191

RESUMO

Adequate sleep is critical for development and facilitates the maturation of the neurophysiological circuitries at the basis of cognitive and behavioural function. Observational research has associated early life sleep problems with worse later cognitive, psychosocial, and somatic health outcomes. Yet, the extent to which day-to-day sleep behaviours (e.g., duration, regularity) in early life relate to non-rapid eye movement (NREM) neurophysiology-acutely and the long-term-remains to be studied. We measured sleep behaviours in 32 healthy 6-month-olds assessed with actimetry and neurophysiology with high-density electroencephalography (EEG) to investigate the association between NREM sleep and habitual sleep behaviours. Our study revealed four findings: first, daytime sleep behaviours are related to EEG slow-wave activity (SWA). Second, night-time movement and awakenings from sleep are connected with spindle density. Third, habitual sleep timing is linked to neurophysiological connectivity quantified as delta coherence. And lastly, delta coherence at 6 months predicts night-time sleep duration at 12 months. These novel findings widen our understanding that infants' sleep behaviours are closely intertwined with three particular levels of neurophysiology: sleep pressure (determined by SWA), the maturation of the thalamocortical system (spindles), and the maturation of cortical connectivity (coherence). The crucial next step is to extend this concept to clinical groups to objectively characterise infants' sleep behaviours 'at risk' that foster later neurodevelopmental problems.


Assuntos
Movimentos Oculares , Sono de Ondas Lentas , Lactente , Humanos , Eletroencefalografia , Sono/fisiologia , Encéfalo
5.
J Sleep Res ; 32(5): e13878, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-36934366

RESUMO

The orexin 2 receptor-selective agonist danavorexton (TAK-925) has been shown to produce wake-promoting effects in wild-type mice, narcolepsy-model mice, and individuals with narcolepsy type 1 and type 2. Here, we report wake-promoting effects of danavorexton in non-human primates and healthy men during their sleep phase. Electroencephalogram analyses revealed that subcutaneous administration of danavorexton significantly increased wakefulness in common marmosets (p < 0.05 at 0.1 mg kg-1 , and p < 0.001 at 1 mg kg-1 and 10 mg kg-1 ) and cynomolgus monkeys (p ≤ 0.05 at 1 mg kg-1 and 3 mg kg-1 ). In a phase 1b crossover, randomized, double-blind, placebo-controlled and active-controlled study in sleep-deprived healthy participants (ClinicalTrials.gov identifier: NCT03522506), modafinil 300 mg (used to demonstrate assay sensitivity) and continuous infusion of danavorexton 44 mg and danavorexton 112 mg showed statistically superior wake-promoting effects to placebo (n = 18). Measured using the Maintenance of Wakefulness Test, mean (standard deviation) sleep latencies during infusion of danavorexton 44 mg, danavorexton 112 mg and placebo were 21.4 (8.9), 31.8 (3.2) and 9.2 (6.4) min, respectively. Least-squares mean difference from placebo in average sleep latency was 16.8 min with danavorexton 44 mg and 30.2 min with danavorexton 112 mg (both p < 0.001). Karolinska Sleepiness Scale scores were statistically significantly lower (indicating decreased sleepiness) for participants receiving danavorexton than for those receiving placebo during infusion (danavorexton 44 mg, p = 0.010; danavorexton 112 mg, p < 0.001). Together, these results indicate that an orexin 2 receptor agonist increases wakefulness in non-human primates and healthy individuals during their sleep phase.


Assuntos
Narcolepsia , Orexinas , Vigília , Animais , Método Duplo-Cego , Narcolepsia/tratamento farmacológico , Orexinas/farmacologia , Primatas , Sonolência , Resultado do Tratamento , Vigília/efeitos dos fármacos , Humanos , Masculino
6.
J Sleep Res ; : e13929, 2023 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-37177872

RESUMO

Sleep modulates the immune response, and sleep loss can reduce vaccine immunogenicity; vice versa, immune responses impact sleep. We aimed to investigate the influence of mental health and sleep quality on the immunogenicity of COVID-19 vaccinations and, conversely, of COVID-19 vaccinations on sleep quality. The prospective CoVacSer study monitored mental health, sleep quality and Anti-SARS-CoV-2-Spike IgG titres in a cohort of 1082 healthcare workers from 29 September 2021 to 19 December 2022. Questionnaires and blood samples were collected before, 14 days, and 3 months after the third COVID-19 vaccination, as well as in 154 participants before and 14 days after the fourth COVID-19 vaccination. Healthcare workers with psychiatric disorders had slightly lower Anti-SARS-CoV-2-Spike IgG levels before the third COVID-19 vaccination. However, this effect was mediated by higher median age and body mass index in this subgroup. Antibody titres following the third and fourth COVID-19 vaccinations ("booster vaccinations") were not significantly different between subgroups with and without psychiatric disorders. Sleep quality did not affect the humoral immunogenicity of the COVID-19 vaccinations. Moreover, the COVID-19 vaccinations did not impact self-reported sleep quality. Our data suggest that in a working population neither mental health nor sleep quality relevantly impact the immunogenicity of COVID-19 vaccinations, and that COVID-19 vaccinations do not cause a sustained deterioration of sleep, suggesting that they are not a precipitating factor for insomnia. The findings from this large-scale real-life cohort study will inform clinical practice regarding the recommendation of COVID-19 booster vaccinations for individuals with mental health and sleep problems.

7.
J Sleep Res ; 31(4): e13598, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35502706

RESUMO

The two-process model serves as a major conceptual framework in sleep science. Although dating back more than four decades, it has not lost its relevance for research today. Retracing its origins, I describe how animal experiments aimed at exploring the oscillators driving the circadian sleep-wake rhythm led to the recognition of gradients of sleep states within the daily sleep period. Advances in signal analysis revealed that the level of slow-wave activity in non-rapid eye movement sleep electroencephalogram is high at the beginning of the 12-light period and then declines. After sleep deprivation, the level of slow-wave activity is enhanced. By scheduling recovery sleep to the animal's activity period, the conflict between the sleep-wake-dependent and the circadian influence resulted in a two-stage recovery pattern. These experiments provided the basis for the first version of the two-process model. Sleep deprivation experiments in humans showed that the decline of slow-wave activity during sleep is exponential. The two-process model posits that a sleep-wake-dependent homeostatic process (Process S) interacts with a process controlled by the circadian pacemaker (Process C). At present, homeostatic and circadian facets of sleep regulation are being investigated at the synaptic level as well as in the transcriptome and proteome domains. The notion of sleep has been extended from a global phenomenon to local representations, while the master circadian pacemaker has been supplemented by multiple peripheral oscillators. The original interpretation that the emergence of sleep may be viewed as an escape from the rigid control imposed by the circadian pacemaker is still upheld.


Assuntos
Privação do Sono , Sono , Animais , Ritmo Circadiano/fisiologia , Eletroencefalografia , Homeostase/fisiologia , Humanos , Sono/fisiologia , Vigília/fisiologia
8.
J Neurosci ; 40(1): 171-190, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31694962

RESUMO

Origin and functions of intermittent transitions among sleep stages, including brief awakenings and arousals, constitute a challenge to the current homeostatic framework for sleep regulation, focusing on factors modulating sleep over large time scales. Here we propose that the complex micro-architecture characterizing sleep on scales of seconds and minutes results from intrinsic non-equilibrium critical dynamics. We investigate θ- and δ-wave dynamics in control rats and in rats where the sleep-promoting ventrolateral preoptic nucleus (VLPO) is lesioned (male Sprague-Dawley rats). We demonstrate that bursts in θ and δ cortical rhythms exhibit complex temporal organization, with long-range correlations and robust duality of power-law (θ-bursts, active phase) and exponential-like (δ-bursts, quiescent phase) duration distributions, features typical of non-equilibrium systems self-organizing at criticality. We show that such non-equilibrium behavior relates to anti-correlated coupling between θ- and δ-bursts, persists across a range of time scales, and is independent of the dominant physiologic state; indications of a basic principle in sleep regulation. Further, we find that VLPO lesions lead to a modulation of cortical dynamics resulting in altered dynamical parameters of θ- and δ-bursts and significant reduction in θ-δ coupling. Our empirical findings and model simulations demonstrate that θ-δ coupling is essential for the emerging non-equilibrium critical dynamics observed across the sleep-wake cycle, and indicate that VLPO neurons may have dual role for both sleep and arousal/brief wake activation. The uncovered critical behavior in sleep- and wake-related cortical rhythms indicates a mechanism essential for the micro-architecture of spontaneous sleep-stage and arousal transitions within a novel, non-homeostatic paradigm of sleep regulation.SIGNIFICANCE STATEMENT We show that the complex micro-architecture of sleep-stage/arousal transitions arises from intrinsic non-equilibrium critical dynamics, connecting the temporal organization of dominant cortical rhythms with empirical observations across scales. We link such behavior to sleep-promoting neuronal population, and demonstrate that VLPO lesion (model of insomnia) alters dynamical features of θ and δ rhythms, and leads to significant reduction in θ-δ coupling. This indicates that VLPO neurons may have dual role for both sleep and arousal/brief wake control. The reported empirical findings and modeling simulations constitute first evidences of a neurophysiological fingerprint of self-organization and criticality in sleep- and wake-related cortical rhythms; a mechanism essential for spontaneous sleep-stage and arousal transitions that lays the bases for a novel, non-homeostatic paradigm of sleep regulation.


Assuntos
Sono/fisiologia , Vigília/fisiologia , Animais , Ritmo Delta , Eletroencefalografia , Masculino , Área Pré-Óptica/lesões , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Fases do Sono/fisiologia , Organismos Livres de Patógenos Específicos , Ritmo Teta
9.
BMC Neurosci ; 22(1): 13, 2021 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-33639837

RESUMO

BACKGROUND: Aside from the homeostatic and circadian components, light has itself an important, direct as well as indirect role in sleep regulation. Light exerts indirect sleep effect by modulating the circadian rhythms. Exposure to short light-dark cycle (LD 1:1, 1:1 h light - dark) eliminates the circadian sleep regulatory component but direct sleep effect of light could prevail. The aim of the present study was to examine the interaction between the light and the homeostatic influences regarding sleep regulation in a rat model. METHODS: Spontaneous sleep-wake and homeostatic sleep regulation by sleep deprivation (SD) and analysis of slow waves (SW) were examined in Wistar rats exposed to LD1:1 condition using LD12:12 regime as control. RESULTS: Slow wave sleep (SWS) and REM sleep were both enhanced, while wakefulness (W) was attenuated in LD1:1. SWS recovery after 6-h total SD was more intense in LD1:1 compared to LD12:12 and SWS compensation was augmented in the bright hours. Delta power increment during recovery was caused by the increase of SW number in both cases. More SW was seen during baseline in the second half of the day in LD1:1 and after SD compared to the LD12:12. Increase of SW number was greater in the bright hours compared to the dark ones after SD in LD1:1. Lights ON evoked immediate increase in W and decrease in both SWS and REM sleep during baseline LD1:1 condition, while these changes ceased after SD. Moreover, the initial decrease seen in SWS after lights ON, turned to an increase in the next 6-min bin and this increase was stronger after SD. These alterations were caused by the change of the epoch number in W, but not in case of SWS or REM sleep. Lights OFF did not alter sleep-wake times immediately, except W, which was increased by lights OFF after SD. CONCLUSIONS: Present results show the complex interaction between light and homeostatic sleep regulation in the absence of the circadian component and indicate the decoupling of SW from the homeostatic sleep drive in LD1:1 lighting condition.


Assuntos
Encéfalo/fisiologia , Homeostase/fisiologia , Fotoperíodo , Fases do Sono/fisiologia , Animais , Ritmo Circadiano , Eletrocorticografia , Masculino , Ratos , Ratos Wistar , Vigília/fisiologia
10.
J Sleep Res ; 30(5): e13314, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33601475

RESUMO

The COVID-19 confinement has dramatically altered daily routines, causing decreased sleep quality in adults. This necessitates careful observation, as sleep plays a crucial role in brain maturation and poor sleep increases the risk of psychopathology, particularly in the young population. Through an online survey with one baseline (April 2020) and two follow-up assessments (May and June 2020), we examined the effect of confinement on sleep quality in 452 babies (0-35 months) and 412 preschool children (36-71 months) from several, mainly European, countries. An acute decrease in sleep quality was found in both groups of children. However, at follow-up assessments, this effect rebounded to the level reported for the period before the confinement. Importantly, caregiver's stress level was identified as a substantial risk factor determining lower sleep quality in both groups of children across assessments. Protective factors conserving children's sleep quality included caregiver's engagement in mindfulness techniques or childcare, and the presence of siblings and pets. In the near future, we may repeatedly experience the circumstances of abruptly enforced confinement. Our findings reveal promising pathways of action to protect young children's sleep, with which to essentially mitigate the long-term consequences of the pandemic on brain development and mental health.


Assuntos
COVID-19 , Controle de Doenças Transmissíveis , Transtornos do Sono-Vigília , Sono , COVID-19/epidemiologia , COVID-19/prevenção & controle , Pré-Escolar , Europa (Continente)/epidemiologia , Feminino , Humanos , Lactente , Recém-Nascido , Estudos Longitudinais , Masculino , Fatores de Proteção , Fatores de Risco , Transtornos do Sono-Vigília/epidemiologia
11.
Proc Natl Acad Sci U S A ; 115(27): 7129-7134, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29915051

RESUMO

Sleep and metabolism are physiologically and behaviorally intertwined; however, the molecular basis for their interaction remains poorly understood. Here, we identified a serine metabolic pathway as a key mediator for starvation-induced sleep suppression. Transcriptome analyses revealed that enzymes involved in serine biosynthesis were induced upon starvation in Drosophila melanogaster brains. Genetic mutants of astray (aay), a fly homolog of the rate-limiting phosphoserine phosphatase in serine biosynthesis, displayed reduced starvation-induced sleep suppression. In contrast, a hypomorphic mutation in a serine/threonine-metabolizing enzyme, serine/threonine dehydratase (stdh), exaggerated starvation-induced sleep suppression. Analyses of double mutants indicated that aay and stdh act on the same genetic pathway to titrate serine levels in the head as well as to adjust starvation-induced sleep behaviors. RNA interference-mediated depletion of aay expression in neurons, using cholinergic Gal4 drivers, phenocopied aay mutants, while a nicotinic acetylcholine receptor antagonist selectively rescued the exaggerated starvation-induced sleep suppression in stdh mutants. Taken together, these data demonstrate that neural serine metabolism controls sleep during starvation, possibly via cholinergic signaling. We propose that animals have evolved a sleep-regulatory mechanism that reprograms amino acid metabolism for adaptive sleep behaviors in response to metabolic needs.


Assuntos
Encéfalo/metabolismo , Proteínas de Drosophila/metabolismo , L-Serina Desidratase/metabolismo , Mutação , Serina/metabolismo , Transdução de Sinais , Inanição/metabolismo , Animais , Comportamento Animal , Proteínas de Drosophila/genética , Drosophila melanogaster , L-Serina Desidratase/genética , Serina/genética , Inanição/genética
12.
Molecules ; 26(9)2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062829

RESUMO

Betulinic acid (BA) is a major constituent of Zizyphus seeds that have been long used as therapeutic agents for sleep-related issues in Asia. BA is a pentacyclic triterpenoid. It also possesses various anti-cancer and anti-inflammatory effects. Current commercially available sleep aids typically use GABAergic regulation, for which many studies are being actively conducted. However, few studies have focused on acetylcholine receptors that regulate wakefulness. In this study, we utilized BA as an antagonist of α3ß4 nicotinic acetylcholine receptors (α3ß4 nAChRs) known to regulate rapid-eye-movement (REM) sleep and wakefulness. Effects of BA on α3ß4 nAChRs were concentration-dependent, reversible, voltage-independent, and non-competitive. Site-directed mutagenesis and molecular-docking studies confirmed the binding of BA at the molecular level and showed that the α3 subunit L257 and the ß4 subunit I263 residues affected BA binding. These data demonstrate that BA can bind to a binding site different from the site for the receptor's ligand, acetylcholine (ACh). This suggests that BA may be an effective antagonist that is unaffected by large amounts of ACh released during wakefulness and REM sleep. Based on the above experimental results, BA is likely to be a therapeutically useful sleep aid and sedative.


Assuntos
Acetilcolina/metabolismo , Triterpenos Pentacíclicos/farmacologia , Receptores Nicotínicos/metabolismo , Animais , Sítios de Ligação , Bovinos , Eletrofisiologia , Ligantes , Simulação de Acoplamento Molecular , Mutagênese , Mutação , Oócitos/citologia , Oócitos/metabolismo , Ligação Proteica , Conformação Proteica , Subunidades Proteicas/química , Sementes , Sono , Distúrbios do Início e da Manutenção do Sono/metabolismo , Transcrição Gênica , Triterpenos/farmacologia , Xenopus laevis , Ziziphus , Ácido gama-Aminobutírico/metabolismo , Ácido Betulínico
13.
J Sleep Res ; 28(3): e12709, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-29869353

RESUMO

The photoperiod has been evidenced to influence sleep regulation in the rat. Nevertheless, lengthening of the photoperiod beyond 30 days seems to have little effect on the 24-hr baseline level of sleep and the response to total sleep deprivation. We studied the effects of 12:12 (habitual) and 16:8 (long) light-dark photoperiods on sleep, locomotor activity and body core temperature, before and after 24 hr of total sleep deprivation. Eight rats were submitted for 14 days to light-dark 12:12 (lights on: 08:00 hours-20:00 hours) followed by total sleep deprivation, and then for 14 days to light-dark 16:8 (light extended to 24:00 hours) followed by total sleep deprivation. Rats were simultaneously recorded for electroencephalogram, locomotor activity and body core temperature for 24 hr before and after total sleep deprivation. At baseline before total sleep deprivation, total sleep time and non-rapid eye movement sleep per 24 hr and during extended light hours (20:00 hours-24:00 hours) were higher (13% for total sleep time) after light-dark exposure compared with habitual photoperiod, while percentage delta power in non-rapid eye movements and rapid eye movements were unchanged. Locomotor activity and body core temperature were lower, particularly during extended light hours (20:00 hours-24:00 hours). Following total sleep deprivation, total sleep time and non-rapid eye movements were significantly lower after long photoperiod between 20:00 hours and 24:00 hours, and between 10:00 hours and 12:00 hours, and unchanged per 24 hr. The percentage delta power in non-rapid eye movements was lower between 08:00 hours and 11:00 hours. Total sleep deprivation decreased locomotor activity and body core temperature after habitual photoperiod exposure only. Fourteen days under long photoperiod (light-dark 16:8) increased non-rapid eye movements sleep, and decreased sleep rebound related to total sleep deprivation (lower non-rapid eye movements duration and delta power). This may create a model of sleep extension for the rat that has been found to favour anabolism in the brain and the periphery.


Assuntos
Fotoperíodo , Polissonografia/métodos , Privação do Sono/fisiopatologia , Sono/fisiologia , Animais , Masculino , Ratos , Ratos Wistar
14.
Eur J Neurosci ; 47(11): 1339-1352, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29737605

RESUMO

Obesity prevalence and sleep habit changes are commonplace nowadays, due to modern lifestyle. A bidirectional relationship likely exists between sleep quality and metabolic disruptions, which could impact quality of life. In our study, we investigated the effects of a chronic high-caloric diet on sleep architecture and sleep regulation in mice. We studied the effect of 3 months high-caloric diet (HCD, 45% fat) on sleep and the sleep electroencephalogram (EEG) in C57BL/6J mice during 24-hr baseline (BL) recordings, and after 6-hr sleep deprivation (SD). We examined the effect of HCD on sleep homeostasis, by performing parameter estimation analysis and simulations of the sleep homeostatic Process S, a measure of sleep pressure, which is reflected in the non-rapid-eye-movement (NREM) sleep slow-wave-activity (SWA, EEG power density between 0.5 and 4.0 Hz). Compared to controls (n = 11, 30.7 ± 0.8 g), mice fed with HCD (n = 9, 47.6 ± 0.8 g) showed an increased likelihood of consecutive NREM-REM sleep cycles, increased REM sleep and decreased NREM sleep EEG SWA. After SD, these effects were more pronounced. The simulation resulted in a close fit between the time course of SWA and Process S in both groups. HCD fed mice had a slower time constant (Ti  = 15.98 hr) for the increase in homeostatic sleep pressure compared with controls (5.95 hr) indicating a reduced effect of waking on the increase in sleep pressure. Our results suggest that chronic HCD consumption impacts sleep regulation.


Assuntos
Dieta/efeitos adversos , Ingestão de Energia , Homeostase/fisiologia , Privação do Sono/fisiopatologia , Sono REM/fisiologia , Sono de Ondas Lentas/fisiologia , Vigília/fisiologia , Animais , Eletroencefalografia , Eletromiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
Am J Phys Anthropol ; 166(3): 578-589, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29989159

RESUMO

OBJECTIVES: Primates spend almost half their lives asleep, yet we know little about how evolution has shaped variation in the duration or intensity of sleep (i.e., sleep regulation) across primate species. Our objective was to test hypotheses related to how sleeping site security influences sleep intensity in different lemur species. METHODS: We used actigraphy and infrared videography to generate sleep measures in 100 individuals (males = 51, females = 49) of seven lemur species (genera: Eulemur, Lemur, Propithecus, and Varecia) at the Duke Lemur Center in Durham, NC. We also generated experimental data using sleep deprivation for 16 individuals. This experiment used a pair-wise design for two sets of paired lemurs from each genus, where the experimental pair experienced a sleep deprivation protocol while the control experienced normal sleeping conditions. We calculated a sleep depth composite metric from weighted z scores of three sleep intensity variables. RESULTS: We found that, relative to cathemeral lemurs, diurnal Propithecus was characterized by the deepest sleep and exhibited the most disruptions to normal sleep-wake regulation when sleep deprived. In contrast, Eulemur mongoz was characterized by significantly lighter sleep than Propithecus, and E. mongoz showed the fewest disruptions to normal sleep-wake regulation when sleep deprived. Security of the sleeping site led to greater sleep depth, with access to outdoor housing linked to lighter sleep in all lemurs that were studied. CONCLUSIONS: We propose that sleeping site security was an essential component of sleep regulation throughout primate evolution. This work suggests that sleeping site security may have been an important factor associated with the evolution of sleep in early and later hominins.


Assuntos
Comportamento Animal/fisiologia , Lemur/fisiologia , Sono de Ondas Lentas/fisiologia , Animais , Antropologia Física , Ritmo Circadiano/fisiologia , Feminino , Masculino
16.
J Sleep Res ; 26(5): 539-546, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28488395

RESUMO

Slow waves occurring during non-rapid eye movement sleep have been associated with neurobehavioural performance and memory. In addition, the duration of previous wakefulness and sleep impacts characteristics of these slow waves. However, molecular mechanisms regulating the dynamics of slow-wave characteristics remain poorly understood. The EphA4 receptor regulates glutamatergic transmission and synaptic plasticity, which have both been linked to sleep slow waves. To investigate if EphA4 regulates slow-wave characteristics during non-rapid eye movement sleep, we compared individual parameters of slow waves between EphA4 knockout mice and wild-type littermates under baseline conditions and after a 6-h sleep deprivation. We observed that, compared with wild-type mice, knockout mice display a shorter duration of positive and negative phases of slow waves under baseline conditions and after sleep deprivation. However, the mutation did not change slow-wave density, amplitude and slope, and did not affect the sleep deprivation-dependent changes in slow-wave characteristics, suggesting that EphA4 is not involved in the response to elevated sleep pressure. Our present findings suggest a role for EphA4 in shaping cortical oscillations during sleep that is independent from sleep need.


Assuntos
Receptor EphA4/deficiência , Receptor EphA4/genética , Sono/fisiologia , Animais , Eletroencefalografia , Masculino , Camundongos , Camundongos Knockout , Receptor EphA4/metabolismo , Sono/genética , Privação do Sono/genética , Privação do Sono/fisiopatologia , Fatores de Tempo , Vigília/genética , Vigília/fisiologia
17.
J Comput Neurosci ; 41(1): 15-28, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27066796

RESUMO

In mammals, sleep is categorized by two main sleep stages, rapid eye movement (REM) and non-REM (NREM) sleep that are known to fulfill different functional roles, the most notable being the consolidation of memory. While REM sleep is characterized by brain activity similar to wakefulness, the EEG activity changes drastically with the emergence of K-complexes, sleep spindles and slow oscillations during NREM sleep. These changes are regulated by circadian and ultradian rhythms, which emerge from an intricate interplay between multiple neuronal populations in the brainstem, forebrain and hypothalamus and the resulting varying levels of neuromodulators. Recently, there has been progress in the understanding of those rhythms both from a physiological as well as theoretical perspective. However, how these neuromodulators affect the generation of the different EEG patterns and their temporal dynamics is poorly understood. Here, we build upon previous work on a neural mass model of the sleeping cortex and investigate the effect of those neuromodulators on the dynamics of the cortex and the corresponding transition between wakefulness and the different sleep stages. We show that our simplified model is sufficient to generate the essential features of human EEG over a full day. This approach builds a bridge between sleep regulatory networks and EEG generating neural mass models and provides a valuable tool for model validation.


Assuntos
Encéfalo/fisiologia , Modelos Neurológicos , Rede Nervosa/fisiologia , Sono/fisiologia , Animais , Mapeamento Encefálico , Eletroencefalografia , Humanos , Vigília
18.
J Sleep Res ; 23(6): 664-672, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25088756

RESUMO

Individuals with major depressive disorder typically exhibit sleep electroencephalograpy abnormalities which have been shown to vary by sex. Recent research has shown that depressed males display deficits in slow wave sleep and delta electroencephalograph (EEG) activity that are not apparent in depressed females. This may suggest that males and females with depression vary with respect to their homeostatic regulation of sleep. Utilizing archival data, the present study examined the effects of a 3-h sleep delay, which represents a mild sleep challenge, on slow wave activity in healthy controls and individuals with depression. All participants slept in the laboratory for three sequential nights. On the third night in the laboratory, the participants' bedtime was delayed by 3 h. Slow wave activity was calculated utilizing power spectral analysis and compared across groups. Following the sleep delay, males with depression exhibited the lowest slow wave activity compared to all other groups. These results may suggest that males with depression are at a greater risk for homeostatic dysregulation than females, and may require specialized intervention.


Assuntos
Transtorno Depressivo Maior/fisiopatologia , Caracteres Sexuais , Sono/fisiologia , Adulto , Estudos de Casos e Controles , Eletroencefalografia , Feminino , Homeostase/fisiologia , Humanos , Masculino , Fatores de Tempo , Adulto Jovem
19.
J Sleep Res ; 23(6): 619-627, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25109588

RESUMO

Cynomolgus monkeys are widely used as models of diseases and in pre-clinical studies to assess the impact of new pharmacotherapies on brain function and behaviour. However, the time course of electroencephalographic delta activity during sleep, which represents the main marker of sleep intensity associated with recovery during sleep, has never been described in this non-human primate. In this study, telemetry implants were used to record one spontaneous 24-h sleep-wake cycle in four freely-moving Cynomolgus monkeys, and to quantify the time course of electroencephalographic activity during sleep using spectral analysis. Animals presented a diurnal activity pattern interrupted by short naps. During the dark period, most of the time was spent in sleep with non-rapid eye movement sleep/rapid eye movement sleep alternations and sleep consolidation profiles intermediate between rodents and humans. Deep non-rapid eye movement sleep showed a typical predominance at the beginning of the night with decreased propensity in the course of the night, which was accompanied by a progressive increase in rapid eye movement sleep duration. Spectral profiles showed characteristic changes between vigilance states as reported in other mammalian species. Importantly, delta activity also followed the expected time course of variation, showing a build-up with wakefulness duration and dissipation across the night. Thus, Cynomolgus monkeys present typical characteristics of sleep architecture and spectral structure as those observed in other mammalian species including humans, validating the use of telemetry in this non-human primate model for translational sleep studies.


Assuntos
Macaca fascicularis/fisiologia , Sono/fisiologia , Telemetria , Animais , Atenção/fisiologia , Atenção/efeitos da radiação , Escuridão , Eletroencefalografia , Humanos , Luz , Masculino , Modelos Animais , Polissonografia , Sono/efeitos da radiação , Sono REM/fisiologia , Sono REM/efeitos da radiação , Fatores de Tempo , Vigília/fisiologia , Vigília/efeitos da radiação
20.
eNeuro ; 11(3)2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38212115

RESUMO

Sleep and muscle injury-related pain are in negative relationship, and sleep extension may be a favorable countermeasure. In response to muscle injury, an adaptive sleep response has been described in rats, characterized by an increase in total sleep time (TST) and nonrapid eye movement (NREM) sleep. This study examined the effects of photoperiod lengthening (a model of sleep prolongation in rats) on the sleep characteristics of muscle-injured rats and whether this lengthening could benefit injury-induced mechanical hyperalgesia using the Von Frey test. Switching from the conventional 12:12 light/dark (LD) photoperiod (light on: 08:00-20:00) to LD 16:8 (light extended to 24:00) gives rats an extra window of sleep. Our results show higher TST and NREM sleep times in LD 16:8 versus LD 12:12 injured rats during 4 h of light lengthening for 7 d postinjury, showing the efficiency of photoperiod lengthening to increase sleep time in injured rats. In addition, a cumulative effect with the adaptive sleep response to muscle injury occurred with higher TST and NREM sleep times in LD 16:8 injured versus noninjured rats during the dark period, reflecting the high need for sleep after the injury. Greater stability and higher relative delta power of NREM sleep during the extended light period were also observed in injured rats. Finally, the extended photoperiod limits the muscle injury-induced mechanical hyperalgesia for 13 d and allows faster recovery of the baseline mechanical threshold. This is associated with reduced pro-inflammatory cytokines levels in the hippocampus, a brain structure involved in pain processing.


Assuntos
Hiperalgesia , Fotoperíodo , Ratos , Animais , Sono/fisiologia , Hipocampo , Dor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA