Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pineal Res ; 70(3): e12714, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33378563

RESUMO

We examined whether dynamically changing light across a scheduled 16-h waking day influences sleepiness, cognitive performance, visual comfort, melatonin secretion, and sleep under controlled laboratory conditions in healthy men. Fourteen participants underwent a 49-h laboratory protocol in a repeated-measures study design. They spent the first 5 hours in the evening under standard lighting, followed by an 8-h nocturnal sleep episode at habitual bedtimes. Thereafter, volunteers either woke up to static light or to a dynamic light that changed spectrum and intensity across the scheduled 16-h waking day. Following an 8-h nocturnal sleep episode, the volunteers spent another 11 hours either under static or dynamic light. Static light attenuated the evening rise in melatonin levels more compared to dynamic light as indexed by a significant reduction in the melatonin AUC prior to bedtime during static light only. Participants felt less vigilant in the evening during dynamic light. After dynamic light, sleep latency was significantly shorter in both the baseline and treatment night while sleep structure, sleep quality, cognitive performance, and visual comfort did not significantly differ. The study shows that dynamic changes in spectrum and intensity of light promote melatonin secretion and sleep initiation in healthy men.


Assuntos
Ritmo Circadiano/efeitos da radiação , Luz , Iluminação , Melatonina/metabolismo , Glândula Pineal/efeitos da radiação , Sono/efeitos da radiação , Biomarcadores/metabolismo , Cognição/efeitos da radiação , Cor , Voluntários Saudáveis , Humanos , Masculino , Glândula Pineal/metabolismo , Saliva/metabolismo , Fatores de Tempo
2.
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
3.
Sleep ; 39(3): 613-24, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26612390

RESUMO

STUDY OBJECTIVES: Optimal sleep is ensured by the interaction of circadian and homeostatic processes. Although synaptic plasticity seems to contribute to both processes, the specific players involved are not well understood. The EphA4 tyrosine kinase receptor is a cell adhesion protein regulating synaptic plasticity. We investigated the role of EphA4 in sleep regulation using electrocorticography in mice lacking EphA4 and gene expression measurements. METHODS: EphA4 knockout (KO) mice, Clock(Δ19/Δ19) mutant mice and littermates, C57BL/6J and CD-1 mice, and Sprague-Dawley rats were studied under a 12 h light: 12 h dark cycle, under undisturbed conditions or 6 h sleep deprivation (SLD), and submitted to a 48 h electrophysiological recording and/or brain sampling at different time of day. RESULTS: EphA4 KO mice showed less rapid eye movement sleep (REMS), enhanced duration of individual bouts of wakefulness and nonrapid eye movement sleep (NREMS) during the light period, and a blunted daily rhythm of NREMS sigma activity. The NREMS delta activity response to SLD was unchanged in EphA4 KO mice. However, SLD increased EphA4 expression in the thalamic/hypothalamic region in C57BL/6J mice. We further show the presence of E-boxes in the promoter region of EphA4, a lower expression of EphA4 in Clock mutant mice, a rhythmic expression of EphA4 ligands in several brain areas, expression of EphA4 in the suprachiasmatic nuclei of the hypothalamus (SCN), and finally an unchanged number of cells expressing Vip, Grp and Avp in the SCN of EphA4 KO mice. CONCLUSIONS: Our results suggest that EphA4 is involved in circadian sleep regulation.


Assuntos
Ritmo Circadiano/fisiologia , Receptor EphA4/metabolismo , Privação do Sono/fisiopatologia , Sono/fisiologia , Animais , Proteínas CLOCK/genética , Ritmo Circadiano/genética , Escuridão , Eletrocorticografia , Fenômenos Eletrofisiológicos , Homeostase , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasticidade Neuronal , Regiões Promotoras Genéticas/genética , Ratos , Ratos Sprague-Dawley , Receptor EphA4/biossíntese , Receptor EphA4/deficiência , Receptor EphA4/genética , Sono/genética , Privação do Sono/genética , Sono REM/genética , Sono REM/fisiologia , Núcleo Supraquiasmático/metabolismo , Tálamo/metabolismo , Fatores de Tempo , Vigília/genética , Vigília/fisiologia
4.
Brain Behav Immun ; 47: 118-30, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25576803

RESUMO

Traumatic brain injury (TBI), including mild TBI (mTBI), is importantly associated with vigilance and sleep complaints. Because sleep is required for learning, plasticity and recovery, we here evaluated the bidirectional relationship between mTBI and sleep with two specific objectives: (1) Test that mTBI rapidly impairs sleep-wake architecture and the dynamics of the electrophysiological marker of sleep homeostasis (i.e., non-rapid eye movement sleep delta (1-4Hz) activity); (2) evaluate the impact of sleep loss following mTBI on the expression of plasticity markers that have been linked to sleep homeostasis and on genome-wide gene expression. A closed-head injury model was used to perform a 48h electrocorticographic (ECoG) recording in mice submitted to mTBI or Sham surgery. mTBI was found to immediately decrease the capacity to sustain long bouts of wakefulness as well as the amplitude of the time course of ECoG delta activity during wakefulness. Significant changes in ECoG spectral activity during wakefulness, non-rapid eye movement and rapid eye movement sleep were observed mainly on the second recorded day. A second experiment was performed to measure gene expression in the cerebral cortex and hippocampus after a mTBI followed either by two consecutive days of 6h sleep deprivation (SD) or of undisturbed behavior (quantitative PCR and next-generation sequencing). mTBI modified the expression of genes involved in immunity, inflammation and glial function (e.g., chemokines, glial markers) and SD changed that of genes linked to circadian rhythms, synaptic activity/neuronal plasticity, neuroprotection and cell death and survival. SD appeared to affect gene expression in the cerebral cortex more importantly after mTBI than Sham surgery including that of the astrocytic marker Gfap, which was proposed as a marker of clinical outcome after TBI. Interestingly, SD impacted the hippocampal expression of the plasticity elements Arc and EfnA3 only after mTBI. Overall, our findings reveal alterations in spectral signature across all vigilance states in the first days after mTBI, and show that sleep loss post-mTBI reprograms the transcriptome in a brain area-specific manner and in a way that could be deleterious to brain recovery.


Assuntos
Lesões Encefálicas/fisiopatologia , Encéfalo/fisiopatologia , Ritmo Circadiano/fisiologia , Sono/fisiologia , Transcriptoma , Animais , Encéfalo/metabolismo , Lesões Encefálicas/metabolismo , Eletrocorticografia , Masculino , Camundongos , Privação do Sono/metabolismo , Privação do Sono/fisiopatologia
5.
Ann Med ; 46(2): 62-72, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24428734

RESUMO

Sleep parallels brain functioning and mental health. Neuronal activity during wakefulness leads to a subsequent increase in sleep intensity as measured using electroencephalographic slow-wave activity (SWA; index of neuronal synchrony in the low-frequency range). Wakefulness, and particularly prolonged wakefulness, also drives important changes in brain gene expression and changes in protein regulation. The role of these two cellular mechanisms in sleep-wake regulation has typically been studied independently, and their exact contribution to SWA remains poorly defined. In this review, we highlight that many transcriptional pathways driven by sleep deprivation are associated to protein regulation. We first describe the relationship between cytokines, clock genes, and markers of sleep need with an emphasis on transcriptional processes. Observations regarding the role of protein metabolism in sleep-wake regulation are then depicted while presenting interconnections between transcriptional and translational responses driven by sleep loss. Lastly, a manner by which this integrated response can feed back on neuronal network activity to determine sleep intensity is proposed. Overall, the literature supports that a complex cross-talk between transcriptional and translational regulation during prolonged wakefulness drives the changes in sleep intensity as a function of the sleep/wake history.


Assuntos
Privação do Sono/metabolismo , Sono/fisiologia , Vigília/fisiologia , Animais , Encéfalo/fisiologia , Eletroencefalografia , Humanos , Processamento de Proteína Pós-Traducional/fisiologia , Transcrição Gênica/fisiologia
6.
Proc Natl Acad Sci U S A ; 110(24): 9974-9, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23716671

RESUMO

Maintaining wakefulness is associated with a progressive increase in the need for sleep. This phenomenon has been linked to changes in synaptic function. The synaptic adhesion molecule Neuroligin-1 (NLG1) controls the activity and synaptic localization of N-methyl-d-aspartate receptors, which activity is impaired by prolonged wakefulness. We here highlight that this pathway may underlie both the adverse effects of sleep loss on cognition and the subsequent changes in cortical synchrony. We found that the expression of specific Nlg1 transcript variants is changed by sleep deprivation in three mouse strains. These observations were associated with strain-specific changes in synaptic NLG1 protein content. Importantly, we showed that Nlg1 knockout mice are not able to sustain wakefulness and spend more time in nonrapid eye movement sleep than wild-type mice. These changes occurred with modifications in waking quality as exemplified by low theta/alpha activity during wakefulness and poor preference for social novelty, as well as altered delta synchrony during sleep. Finally, we identified a transcriptional pathway that could underlie the sleep/wake-dependent changes in Nlg1 expression and that involves clock transcription factors. We thus suggest that NLG1 is an element that contributes to the coupling of neuronal activity to sleep/wake regulation.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Neurônios/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Western Blotting , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Eletroencefalografia , Eletromiografia , Expressão Gênica , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos AKR , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Neurônios/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sono/genética , Privação do Sono/genética , Privação do Sono/fisiopatologia , Especificidade da Espécie , Fatores de Tempo , Vigília/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...