Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.346
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(25): 5500-5516.e21, 2023 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-38016470

RESUMEN

Most animals require sleep, and sleep loss induces serious pathophysiological consequences, including death. Previous experimental approaches for investigating sleep impacts in mice have been unable to persistently deprive animals of both rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Here, we report a "curling prevention by water" paradigm wherein mice remain awake 96% of the time. After 4 days of exposure, mice exhibit severe inflammation, and approximately 80% die. Sleep deprivation increases levels of prostaglandin D2 (PGD2) in the brain, and we found that elevated PGD2 efflux across the blood-brain-barrier-mediated by ATP-binding cassette subfamily C4 transporter-induces both accumulation of circulating neutrophils and a cytokine-storm-like syndrome. Experimental disruption of the PGD2/DP1 axis dramatically reduced sleep-deprivation-induced inflammation. Thus, our study reveals that sleep-related changes in PGD2 in the central nervous system drive profound pathological consequences in the peripheral immune system.


Asunto(s)
Privación de Sueño , Animales , Ratones , Citocinas/metabolismo , Inflamación , Prostaglandina D2 , Sueño/fisiología , Privación de Sueño/genética , Privación de Sueño/metabolismo , Síndrome , Humanos , Ratas , Línea Celular , Tormentas Ciclónicas , Neutrófilos/metabolismo
2.
Cell ; 181(6): 1307-1328.e15, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32502393

RESUMEN

The view that sleep is essential for survival is supported by the ubiquity of this behavior, the apparent existence of sleep-like states in the earliest animals, and the fact that severe sleep loss can be lethal. The cause of this lethality is unknown. Here we show, using flies and mice, that sleep deprivation leads to accumulation of reactive oxygen species (ROS) and consequent oxidative stress, specifically in the gut. ROS are not just correlates of sleep deprivation but drivers of death: their neutralization prevents oxidative stress and allows flies to have a normal lifespan with little to no sleep. The rescue can be achieved with oral antioxidant compounds or with gut-targeted transgenic expression of antioxidant enzymes. We conclude that death upon severe sleep restriction can be caused by oxidative stress, that the gut is central in this process, and that survival without sleep is possible when ROS accumulation is prevented. VIDEO ABSTRACT.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Antioxidantes/metabolismo , Drosophila , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Estrés Oxidativo/fisiología
3.
Proc Natl Acad Sci U S A ; 121(3): e2220532121, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38207077

RESUMEN

MicroRNAs (miRNAs) are key post-transcriptional regulators of gene expression that have been implicated in a plethora of neuronal processes. Nevertheless, their role in regulating brain activity in the context of sleep has so far received little attention. To test their involvement, we deleted mature miRNAs in post-mitotic neurons at two developmental ages, i.e., in early adulthood using conditional Dicer knockout (cKO) mice and in adult mice using an inducible conditional Dicer cKO (icKO) line. In both models, electroencephalographic (EEG) activity was affected and the response to sleep deprivation (SD) altered; while the rapid-eye-movement sleep (REMS) rebound was compromised in both, the increase in EEG delta (1 to 4 Hz) power during non-REMS (NREMS) was smaller in cKO mice and larger in icKO mice compared to controls. We subsequently investigated the effects of SD on the forebrain miRNA transcriptome and found that the expression of 48 miRNAs was affected, and in particular that of the activity-dependent miR-709. In vivo inhibition of miR-709 in the brain increased EEG power during NREMS in the slow-delta (0.75 to 1.75 Hz) range, particularly after periods of prolonged wakefulness. Transcriptome analysis of primary cortical neurons in vitro revealed that miR-709 regulates genes involved in glutamatergic neurotransmission. A subset of these genes was also affected in the cortices of sleep-deprived, miR-709-inhibited mice. Our data implicate miRNAs in the regulation of EEG activity and indicate that miR-709 links neuronal activity during wakefulness to brain synchrony during sleep through the regulation of glutamatergic signaling.


Asunto(s)
MicroARNs , Sueño , Ratones , Animales , Sueño/fisiología , Privación de Sueño/genética , Electroencefalografía , Vigilia/fisiología , Prosencéfalo , MicroARNs/genética , MicroARNs/farmacología
4.
Proc Natl Acad Sci U S A ; 120(26): e2214505120, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37339227

RESUMEN

Sleep loss robustly disrupts mood and emotion regulation in healthy individuals but can have a transient antidepressant effect in a subset of patients with depression. The neural mechanisms underlying this paradoxical effect remain unclear. Previous studies suggest that the amygdala and dorsal nexus (DN) play key roles in depressive mood regulation. Here, we used functional MRI to examine associations between amygdala- and DN-related resting-state connectivity alterations and mood changes after one night of total sleep deprivation (TSD) in both healthy adults and patients with major depressive disorder using strictly controlled in-laboratory studies. Behavioral data showed that TSD increased negative mood in healthy participants but reduced depressive symptoms in 43% of patients. Imaging data showed that TSD enhanced both amygdala- and DN-related connectivity in healthy participants. Moreover, enhanced amygdala connectivity to the anterior cingulate cortex (ACC) after TSD associated with better mood in healthy participants and antidepressant effects in depressed patients. These findings support the key role of the amygdala-cingulate circuit in mood regulation in both healthy and depressed populations and suggest that rapid antidepressant treatment may target the enhancement of amygdala-ACC connectivity.


Asunto(s)
Trastorno Depresivo Mayor , Adulto , Humanos , Trastorno Depresivo Mayor/diagnóstico por imagen , Trastorno Depresivo Mayor/tratamiento farmacológico , Privación de Sueño/diagnóstico por imagen , Amígdala del Cerebelo/diagnóstico por imagen , Giro del Cíngulo/diagnóstico por imagen , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Imagen por Resonancia Magnética/métodos
5.
J Neurosci ; 44(28)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38830763

RESUMEN

Chronic sleep disruption (CSD), from insufficient or fragmented sleep and is an important risk factor for Alzheimer's disease (AD). Underlying mechanisms are not understood. CSD in mice results in degeneration of locus ceruleus neurons (LCn) and CA1 hippocampal neurons and increases hippocampal amyloid-ß42 (Aß42), entorhinal cortex (EC) tau phosphorylation (p-tau), and glial reactivity. LCn injury is increasingly implicated in AD pathogenesis. CSD increases NE turnover in LCn, and LCn norepinephrine (NE) metabolism activates asparagine endopeptidase (AEP), an enzyme known to cleave amyloid precursor protein (APP) and tau into neurotoxic fragments. We hypothesized that CSD would activate LCn AEP in an NE-dependent manner to induce LCn and hippocampal injury. Here, we studied LCn, hippocampal, and EC responses to CSD in mice deficient in NE [dopamine ß-hydroxylase (Dbh)-/-] and control male and female mice, using a model of chronic fragmentation of sleep (CFS). Sleep was equally fragmented in Dbh -/- and control male and female mice, yet only Dbh -/- mice conferred resistance to CFS loss of LCn, LCn p-tau, and LCn AEP upregulation and activation as evidenced by an increase in AEP-cleaved APP and tau fragments. Absence of NE also prevented a CFS increase in hippocampal AEP-APP and Aß42 but did not prevent CFS-increased AEP-tau and p-tau in the EC. Collectively, this work demonstrates AEP activation by CFS, establishes key roles for NE in both CFS degeneration of LCn neurons and CFS promotion of forebrain Aß accumulation, and, thereby, identifies a key molecular link between CSD and specific AD neural injuries.


Asunto(s)
Péptidos beta-Amiloides , Cisteína Endopeptidasas , Hipocampo , Locus Coeruleus , Norepinefrina , Privación de Sueño , Animales , Péptidos beta-Amiloides/metabolismo , Norepinefrina/metabolismo , Ratones , Hipocampo/metabolismo , Hipocampo/patología , Privación de Sueño/metabolismo , Privación de Sueño/patología , Masculino , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Fragmentos de Péptidos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Dopamina beta-Hidroxilasa/metabolismo , Dopamina beta-Hidroxilasa/genética , Proteínas tau/metabolismo , Femenino , Degeneración Nerviosa/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/genética
6.
Proc Natl Acad Sci U S A ; 119(27): e2200047119, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35759656

RESUMEN

Adequate pain management is one of the biggest challenges of the modern healthcare system. Physician perception of patient subjective pain, which is crucial to pain management, is susceptible to a host of potential biases. Here we explore the timing of physicians' work as a previously unrecognized source of systematic bias in pain management. We hypothesized that during night shifts, sleep deprivation, fatigue, and stress would reduce physicians' empathy for others' pain, leading to underprescription of analgesics for patient pain relief. In study 1, 67 resident physicians, either following a night shift or not, performed empathy for pain assessment tasks and simulated patient scenarios in laboratory conditions. As predicted, following a night shift, physicians showed reduced empathy for pain. In study 2, we explored this phenomenon in medical decisions in the field. We analyzed three emergency department datasets from Israel and the United States that included discharge notes of patients arriving with pain complaints during 2013 to 2020 (n = 13,482). Across all datasets, physicians were less likely to prescribe an analgesic during night shifts (compared to daytime shifts) and prescribed fewer analgesics than generally recommended by the World Health Organization. This effect remained significant after adjusting for patient, physician, type of complaint, and emergency department characteristics. Underprescription for pain during night shifts was particularly prominent for opioids. We conclude that night shift work is an important and previously unrecognized source of bias in pain management, likely stemming from impaired perception of pain. We consider the implications for hospitals and other organizations employing night shifts.


Asunto(s)
Analgésicos , Prescripciones de Medicamentos , Empatía , Relaciones Médico-Paciente , Médicos , Horario de Trabajo por Turnos , Analgésicos/uso terapéutico , Conjuntos de Datos como Asunto , Humanos , Israel , Dolor/tratamiento farmacológico , Médicos/psicología , Horario de Trabajo por Turnos/psicología , Privación de Sueño , Estados Unidos
7.
J Neurosci ; 43(12): 2168-2177, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36804738

RESUMEN

Sleep loss pervasively affects the human brain at multiple levels. Age-related changes in several sleep characteristics indicate that reduced sleep quality is a frequent characteristic of aging. Conversely, sleep disruption may accelerate the aging process, yet it is not known what will happen to the age status of the brain if we can manipulate sleep conditions. To tackle this question, we used an approach of brain age to investigate whether sleep loss would cause age-related changes in the brain. We included MRI data of 134 healthy volunteers (mean chronological age of 25.3 between the age of 19 and 39 years, 42 females/92 males) from five datasets with different sleep conditions. Across three datasets with the condition of total sleep deprivation (>24 h of prolonged wakefulness), we consistently observed that total sleep deprivation increased brain age by 1-2 years regarding the group mean difference with the baseline. Interestingly, after one night of recovery sleep, brain age was not different from baseline. We also demonstrated the associations between the change in brain age after total sleep deprivation and the sleep variables measured during the recovery night. By contrast, brain age was not significantly changed by either acute (3 h time-in-bed for one night) or chronic partial sleep restriction (5 h time-in-bed for five continuous nights). Together, the convergent findings indicate that acute total sleep loss changes brain morphology in an aging-like direction in young participants and that these changes are reversible by recovery sleep.SIGNIFICANCE STATEMENT Sleep is fundamental for humans to maintain normal physical and psychological functions. Experimental sleep deprivation is a variable-controlling approach to engaging the brain among different sleep conditions for investigating the responses of the brain to sleep loss. Here, we quantified the response of the brain to sleep deprivation by using the change of brain age predictable with brain morphologic features. In three independent datasets, we consistently found increased brain age after total sleep deprivation, which was associated with the change in sleep variables. Moreover, no significant change in brain age was found after partial sleep deprivation in another two datasets. Our study provides new evidence to explain the brainwide effect of sleep loss in an aging-like direction.


Asunto(s)
Privación de Sueño , Sueño , Masculino , Femenino , Humanos , Adulto , Adulto Joven , Privación de Sueño/diagnóstico por imagen , Privación de Sueño/psicología , Sueño/fisiología , Encéfalo/diagnóstico por imagen , Vigilia/fisiología , Factores de Tiempo
8.
Neuroimage ; 299: 120837, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39241898

RESUMEN

Sleep deprivation has been demonstrated to exert widespread and intricate impacts on the brain network. The human brain network is a modular network composed of interconnected nodes. This network consists of provincial hubs and connector hubs, with provincial hubs having diverse connectivities within their own modules, while connector hubs distribute their connectivities across different modules. The latter is crucial for integrating information from various modules and ensuring the normal functioning of the modular brain. However, there has been a lack of systematic investigation into the impact of sleep deprivation on brain connector hubs. In this study, we utilized functional connectivity from resting-state functional magnetic resonance imaging, as well as structural connectivity from diffusion-weighted imaging, to systematically explore the variation of connector hub properties in the cerebral cortex after one night of sleep deprivation. The normalized participation coefficients (PCnorm) were utilized to identify connector hubs. In both the functional and structural networks, connector hubs exhibited a significant increase in average PCnorm, indicating the diversity enhancement of the connector hub following sleep deprivation. This enhancement is associated with increased network cost, reduced modularity, and decreased small-worldness, but enhanced global efficiency. This may potentially signify a compensatory mechanism within the brain following sleep deprivation. The significantly affected connector hubs were primarily observed in both the Control Network and Salience Network. We believe that the observed results reflect the increasing demand on the brain to invest more effort at preventing performance deterioration after sleep loss, in exchange for increased communication efficiency, especially involving systems responsible for neural resource allocation and cognitive control. These results have been replicated in an independent dataset. In conclusion, this study has enhanced our understanding of the compensatory mechanism in the brain response to sleep deprivation. This compensation is characterized by an enhancement in the connector hubs responsible for inter-modular communication, especially those related to neural resource and cognitive control. As a result, this compensation comes with a higher network cost but leads to an improvement in global communication efficiency, akin to a more random-like network manner.


Asunto(s)
Conectoma , Imagen de Difusión por Resonancia Magnética , Imagen por Resonancia Magnética , Red Nerviosa , Privación de Sueño , Humanos , Privación de Sueño/fisiopatología , Privación de Sueño/diagnóstico por imagen , Masculino , Adulto , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/fisiopatología , Red Nerviosa/fisiología , Conectoma/métodos , Adulto Joven , Femenino , Encéfalo/diagnóstico por imagen , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/fisiopatología , Corteza Cerebral/fisiología
9.
J Neurophysiol ; 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39412560

RESUMEN

Sleep deprivation (SD) has been shown to have a negative impact on alertness, as evidenced by behavioral and electroencephalographic studies. Nevertheless, conventional fixed-bandwidth spectral analysis confused the aperiodic and periodic components, and ignored some important periodic parameters (i.e., center frequency, bandwidth). Here, based on a large open access dataset of SD, we employed a standardized process for multiple electrode analysis and group inference. We found that, compared to SC, the aperiodic offset shifted overall after SD, primarily in the occipital region. And this shift was associated with a reduction in subjective alertness. For periodic component, we did not found any power change in alpha rhythm, but there is an increase in bandwidth of alpha. And the increased regions distributed in the occipital and temporal lobes. These findings highlight the potential significance and value of aperiodic parameters in behavioral and electrophysiological researches.

10.
J Neurochem ; 168(9): 2561-2576, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38676340

RESUMEN

Rapid eye movement sleep (REMS) maintains brain excitability at least by regulating Na-K ATPase activity. Although REMS deprivation (REMSD)-associated elevated noradrenaline (NA) increases Na-K ATPase protein expression, its mRNA transcription did not increase. We hypothesized and confirmed both in vivo as well as in vitro that elevated mRNA stability explains the apparent puzzle. The mRNA stability was measured in control and REMSD rat brain with or without in vivo treatment with α1-adrenoceptor (AR) antagonist, prazosin (PRZ). Upon REMSD, Na-K ATPase α1-, and α2-mRNA stability increased significantly, which was prevented by PRZ. To decipher the molecular mechanism of action, we estimated NA-induced Na-K ATPase mRNA stability in Neuro-2a cells under controlled conditions and by transcription blockage using Actinomycin D (Act-D). NA increased Na-K ATPase mRNA stability, which was prevented by PRZ and propranolol (PRP, ß-AR antagonist). The knockdown assay confirmed that the increased mRNA stabilization was induced by elevated cytoplasmic abundance of Human antigen R (HuR) and involving (Phospholipase C) PLC-mediated activation of Protein Kinase C (PKC). Additionally, using cell-impermeable Enz-link sulfo NHS-SS-Biotin, we observed that NA increased Na-K ATPase α1-subunits on the Neuro-2a cell surface. We conclude that REMSD-associated elevated NA, acting on α1- and ß-AR, increases nucleocytoplasmic translocation of HuR and increases Na-K ATPase mRNA stability, resulting in increased Na-K ATPase protein expression. The latter then gets translocated to the neuronal membrane surface involving both PKC and (Protein Kinase A) PKA-mediated pathways. These findings may be exploited for the amelioration of REMSD-associated chronic disorders and symptoms.


Asunto(s)
Proteína 1 Similar a ELAV , Norepinefrina , Proteína Quinasa C , Estabilidad del ARN , ATPasa Intercambiadora de Sodio-Potasio , Animales , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética , Ratas , Norepinefrina/metabolismo , Proteína Quinasa C/metabolismo , Proteína 1 Similar a ELAV/metabolismo , Proteína 1 Similar a ELAV/genética , Estabilidad del ARN/efectos de los fármacos , Masculino , Membrana Celular/metabolismo , Membrana Celular/efectos de los fármacos , ARN Mensajero/metabolismo , ARN Mensajero/biosíntesis , Ratones , Transducción de Señal/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Línea Celular Tumoral , Ratas Wistar , Humanos
11.
EMBO J ; 39(21): e103864, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32893934

RESUMEN

The fragile X autosomal homolog 1 (Fxr1) is regulated by lithium and has been GWAS-associated with schizophrenia and insomnia. Homeostatic regulation of synaptic strength is essential for the maintenance of brain functions and involves both cell-autonomous and system-level processes such as sleep. We examined the contribution of Fxr1 to cell-autonomous homeostatic synaptic scaling and neuronal responses to sleep loss, using a combination of gene overexpression and Crispr/Cas9-mediated somatic knockouts to modulate gene expression. Our findings indicate that Fxr1 is downregulated during both scaling and sleep deprivation via a glycogen synthase kinase 3 beta (GSK3ß)-dependent mechanism. In both conditions, downregulation of Fxr1 is essential for the homeostatic modulation of surface AMPA receptors and synaptic strength. Preventing the downregulation of Fxr1 during sleep deprivation results in altered EEG signatures. Furthermore, sequencing of neuronal translatomes revealed the contribution of Fxr1 to changes induced by sleep deprivation. These findings uncover a role of Fxr1 as a shared signaling hub between cell-autonomous homeostatic plasticity and system-level responses to sleep loss, with potential implications for neuropsychiatric illnesses and treatments.


Asunto(s)
Homeostasis/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Sueño/genética , Sueño/fisiología , Animales , Encéfalo/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasticidad Neuronal , Neuronas/metabolismo , Receptores AMPA/metabolismo , Transcriptoma
12.
Am J Physiol Heart Circ Physiol ; 327(4): H947-H955, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39178028

RESUMEN

Exaggerated blood pressure (BP) responses during exercise are independently associated with future development of hypertension. Partial sleep deprivation (PSD) can increase 24-h ambulatory BP, but the effects on exercise BP are unclear. We hypothesized that acute PSD would augment the BP response to constant load cycling exercise and a 20-min time trial. Twenty-two healthy adults (22 ± 3 yr old; 13 males; V̇o2peak, 43.6 ± 8.2 mL·kg-1·min-1) completed a randomized crossover trial in which they either slept normally (normal sleep-wake schedule for each participant) or sleep was partially deprived (early awakening, 40% of normal sleep duration). Each participant completed a 12-min warm-up consisting of two 6-min steps (step 1, 62 ± 25 W; step 2, 137 ± 60 W) followed by a 20-min time trial on a cycle ergometer. PSD did not alter power output during the 20-min time trial [(control vs. PSD) 170 ± 68 vs. 168 ± 68 W, P = 0.65]. Systolic BP did not differ during step 1 of the warm-up (141 ± 15 vs. 137 ± 12 mmHg, P = 0.39) but was lower following PSD during step 2 (165 ± 21 vs. 159 ± 22 mmHg, P = 0.004) and the 20-min time trial (171 ± 20 vs. 164 ± 23 mmHg, P < 0.001). These results were maintained when peak oxygen uptake (V̇o2peak) was included as a covariate. Systolic BP responses were modulated by sex (time × visit × sex interaction P = 0.03), with attenuated systolic BP during the warm-up and the 20-min time trial in males but not in females. In contrast to our hypothesis, acute PSD attenuates systolic BP responses during constant load and 20-min time trial cycling exercise; however, these observations appear to be primarily driven by changes in males.NEW & NOTEWORTHY A single night of partial sleep deprivation (PSD) can increase ambulatory blood pressure (BP) the following day. Despite this phenomenon, the present study found that acute PSD attenuates systolic BP responses to both constant load cycling and a 20-min cycling time trial in young healthy adults. Interestingly, the attenuated systolic BP responses following PSD appeared to be modulated by sex such that attenuations were observed in males but not in females.


Asunto(s)
Ciclismo , Presión Sanguínea , Estudios Cruzados , Ejercicio Físico , Privación de Sueño , Humanos , Masculino , Privación de Sueño/fisiopatología , Femenino , Adulto Joven , Adulto , Factores de Tiempo , Sueño , Consumo de Oxígeno
13.
Am J Physiol Heart Circ Physiol ; 326(1): H291-H301, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38038716

RESUMEN

Blood pressure (BP) follows a circadian rhythm intertwined with the sleep-wake cycle. Acute partial sleep deprivation (PSD; sleep ≤ 6 h) can increase BP, associated with increased cardiovascular risk. Acute exercise can reduce BP for up to 24 h, a phenomenon termed postexercise hypotension. The present study tested whether aerobic exercise could mitigate the augmented 24-h ambulatory BP caused by acute PSD. Twenty-four young otherwise healthy adults (22 ± 3 yr; 14 females; self-reported chronotypes: 6 early/10 intermediate/8 late; Pittsburgh sleep quality index: 17 good/7 poor sleepers) completed a randomized crossover trial in which, on different days, they slept normally (2300-0700), restricted sleep [0330-0700 (PSD)], and cycled for 50 min (70-80% predicted heart rate maximum) before PSD. Ambulatory BP was assessed every 30 min until 2100 the next day. Acute PSD increased 24-h systolic BP (control 117 ± 9 mmHg, PSD 122 ± 9 mmHg; P < 0.001) and prior exercise attenuated (exercise + PSD 120 ± 9 mmHg; P = 0.04 vs. PSD) but did not fully reverse this response (exercise + PSD, P = 0.02 vs. control). Subgroup analysis revealed that the 24-h systolic BP reduction following exercise was specific to late types (PSD 119 ± 7 vs. exercise + PSD 116 ± 6 mmHg; P < 0.05). Overall, habitual sleep quality was negatively correlated with the change in daytime systolic BP following PSD (r = -0.56, P < 0.01). These findings suggest that the ability of aerobic cycling exercise to counteract the hemodynamic effects of acute PSD in young adults may be dependent on chronotype and that habitual sleep quality can predict the daytime BP response to acute PSD.NEW & NOTEWORTHY We demonstrate that cycling exercise attenuates, but does not fully reverse, the augmented 24-h ambulatory blood pressure (BP) response caused by acute partial sleep deprivation (PSD). This response was primarily observed in late chronotypes. Furthermore, daytime BP after acute PSD is related to habitual sleep quality, with better sleepers being more prone to BP elevations. This suggests that habitual sleeping habits can influence BP responses to acute PSD and their interactions with prior cycling exercise.


Asunto(s)
Hipertensión , Privación de Sueño , Femenino , Humanos , Adulto Joven , Presión Sanguínea/fisiología , Monitoreo Ambulatorio de la Presión Arterial , Cronotipo , Ejercicio Físico/fisiología , Sueño/fisiología , Calidad del Sueño , Masculino , Adulto , Estudios Cruzados
14.
Hum Brain Mapp ; 45(13): e70013, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-39225144

RESUMEN

Insufficient sleep compromises cognitive performance, diminishes vigilance, and disrupts daily functioning in hundreds of millions of people worldwide. Despite extensive research revealing significant variability in vigilance vulnerability to sleep deprivation, the underlying mechanisms of these individual differences remain elusive. Locus coeruleus (LC) plays a crucial role in the regulation of sleep-wake cycles and has emerged as a potential marker for vigilance vulnerability to sleep deprivation. In this study, we investigate whether LC microstructural integrity, assessed by fractional anisotropy (FA) through diffusion tensor imaging (DTI) at baseline before sleep deprivation, can predict impaired psychomotor vigilance test (PVT) performance during sleep deprivation in a cohort of 60 healthy individuals subjected to a rigorously controlled in-laboratory sleep study. The findings indicate that individuals with high LC FA experience less vigilance impairment from sleep deprivation compared with those with low LC FA. LC FA accounts for 10.8% of the variance in sleep-deprived PVT lapses. Importantly, the relationship between LC FA and impaired PVT performance during sleep deprivation is anatomically specific, suggesting that LC microstructural integrity may serve as a biomarker for vigilance vulnerability to sleep loss.


Asunto(s)
Imagen de Difusión Tensora , Locus Coeruleus , Desempeño Psicomotor , Privación de Sueño , Humanos , Privación de Sueño/diagnóstico por imagen , Privación de Sueño/fisiopatología , Privación de Sueño/patología , Locus Coeruleus/diagnóstico por imagen , Locus Coeruleus/patología , Masculino , Femenino , Adulto , Adulto Joven , Desempeño Psicomotor/fisiología , Nivel de Alerta/fisiología , Anisotropía , Pruebas Neuropsicológicas
15.
Cogn Affect Behav Neurosci ; 24(3): 517-526, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38273105

RESUMEN

Sleep deprivation (SD) negatively affects many cognitive functions, such as language performance. However, what remains unclear is whether and how SD affects the language-related brain network based on gender and age differences. The current study of 86 healthy adults used resting-state functional magnetic resonance imaging (rs-fMRI) to measure language-related functional connectivity after full sleep or partial SD. Gender and age differences in functional connectivity were assessed across four linguistic aspects: phonetics, morphology, semantics, and syntax. The results showed that SD can affect the connectivity status of language-related brain networks, especially syntax-related networks. Furthermore, the influence of SD on the functional connectivity in language-related networks differed between male and female groups, and between younger and older groups. Specifically, there were gender differences in the temporal association cortex and age differences in the parietal association cortex, during full sleep versus partial SD. These findings highlight changes in the brain's functional connectivity in response to SD as a potential source of gender and age differences in brain function.


Asunto(s)
Mapeo Encefálico , Encéfalo , Lenguaje , Imagen por Resonancia Magnética , Caracteres Sexuales , Privación de Sueño , Humanos , Masculino , Femenino , Adulto , Privación de Sueño/fisiopatología , Privación de Sueño/diagnóstico por imagen , Adulto Joven , Encéfalo/fisiopatología , Encéfalo/diagnóstico por imagen , Encéfalo/fisiología , Vías Nerviosas/fisiopatología , Vías Nerviosas/fisiología , Vías Nerviosas/diagnóstico por imagen , Persona de Mediana Edad , Envejecimiento/fisiología , Adolescente
16.
J Pediatr ; 270: 114036, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38554747

RESUMEN

Findings from a recent survey of a community-based sample of Black youth ages 12 through 21 in Baltimore City, Maryland (n = 345) reveal that viewing fatal police violence videos is associated with significant increases in the odds of youth sleep disturbances, and about 30% of this association is attributable to emotional distress after viewing the videos.


Asunto(s)
Negro o Afroamericano , Policia , Trastornos del Sueño-Vigilia , Humanos , Adolescente , Trastornos del Sueño-Vigilia/epidemiología , Trastornos del Sueño-Vigilia/etiología , Masculino , Femenino , Niño , Adulto Joven , Baltimore/epidemiología , Violencia , Exposición a la Violencia/psicología
17.
Metabolomics ; 20(5): 97, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112673

RESUMEN

INTRODUCTION: The human salivary metabolome is a rich source of information for metabolomics studies. Among other influences, individual differences in sleep-wake history and time of day may affect the metabolome. OBJECTIVES: We aimed to characterize the influence of a single night of sleep deprivation compared to sufficient sleep on the metabolites present in oral fluid and to assess the implications of sampling time points for the design of metabolomics studies. METHODS: Oral fluid specimens of 13 healthy young males were obtained in Salivette® devices at regular intervals in both a control condition (repeated 8-hour sleep) and a sleep deprivation condition (total sleep deprivation of 8 h, recovery sleep of 8 h) and their metabolic contents compared in a semi-targeted metabolomics approach. RESULTS: Analysis of variance results showed factor 'time' (i.e., sampling time point) representing the major influencer (median 9.24%, range 3.02-42.91%), surpassing the intervention of sleep deprivation (median 1.81%, range 0.19-12.46%). In addition, we found about 10% of all metabolic features to have significantly changed in at least one time point after a night of sleep deprivation when compared to 8 h of sleep. CONCLUSION: The majority of significant alterations in metabolites' abundances were found when sampled in the morning hours, which can lead to subsequent misinterpretations of experimental effects in metabolomics studies. Beyond applying a within-subject design with identical sample collection times, we highly recommend monitoring participants' sleep-wake schedules prior to and during experiments, even if the study focus is not sleep-related (e.g., via actigraphy).


Asunto(s)
Metabolómica , Saliva , Sueño , Humanos , Masculino , Metabolómica/métodos , Saliva/metabolismo , Saliva/química , Sueño/fisiología , Adulto Joven , Adulto , Privación de Sueño/metabolismo , Metaboloma/fisiología , Factores de Tiempo
18.
Exp Eye Res ; 243: 109907, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38649019

RESUMEN

Sleep loss is common in modern society and is increasingly associated with eye diseases. However, the precise effects of sleep loss on retinal structure and function, particularly on the retinal circadian system, remain largely unexplored. This study investigates these effects using a chronic sleep deprivation (CSD) model in mice. Our investigation reveals that CSD significantly alters the retinal circadian transcriptome, leading to remarkable changes in the temporal patterns of enriched pathways. This perturbation extends to metabolic and immune-related transcriptomes, coupled with an accumulation of reactive oxygen species in the retina. Notably, CSD rhythmically affects the thickness of the ganglion cell complex, along with diurnal shifts in microglial migration and morphology within the retina. Most critically, we observe a marked decrease in both scotopic and photopic retinal function under CSD conditions. These findings underscore the broad impact of sleep deprivation on retinal health, highlighting its role in altering circadian gene expression, metabolism, immune response, and structural integrity. Our study provides new insights into the broader impact of sleep loss on retinal health.


Asunto(s)
Ritmo Circadiano , Ratones Endogámicos C57BL , Retina , Privación de Sueño , Transcriptoma , Animales , Privación de Sueño/fisiopatología , Privación de Sueño/metabolismo , Privación de Sueño/genética , Ratones , Ritmo Circadiano/fisiología , Masculino , Retina/metabolismo , Retina/fisiopatología , Modelos Animales de Enfermedad , Células Ganglionares de la Retina/patología , Células Ganglionares de la Retina/metabolismo , Electrorretinografía , Regulación de la Expresión Génica , Enfermedad Crónica
19.
FASEB J ; 37(5): e22899, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37002889

RESUMEN

Sleep is a fundamental medicine for cardiac homeostasis, and sleep-deprived individuals are prone to higher incidences of heart attack. The lipid-dense diet (obesogenic diet-OBD) is a cumulative risk factor for chronic inflammation in cardiovascular disease; thus, understanding how sleep fragmentation (SF) in an obesity setting impacts immune and cardiac health is an unmet medical need. We hypothesized whether the co-existence of SF with OBD dysregulates gut homeostasis and leukocyte-derived reparative/resolution mediators, thereby impairing cardiac repair. Two-month-old male C57BL/6J mice were randomized first into two groups, then four groups; Control, control + SF, OBD, and OBD + SF mice subjected to myocardial infarction (MI). OBD mice had higher levels of plasma linolenic acid with a decrease in eicosapentaenoic and docosahexaenoic acid. The OBD mice had lower Lactobacillus johnsonii indicating a loss of probiotic microbiota. SF in OBD mice increased Firmicutes/Bacteroidetes ratio indicative of a detrimental change in SF-directed microbiome. OBD + SF group increased in the neutrophil: lymphocyte ratio suggestive of suboptimal inflammation. As a result of SF, resolution mediators (RvD2, RvD3, RvD5, LXA4 , PD1, and MaR1) decreased and inflammatory mediators (PGD2 , PGE2 , PGF2a , 6k-PGF1a ) were increased in OBD mice post-MI. At the site of infarction, the proinflammatory cytokines Ccl2, IL1ß, and IL-6 were amplified in OBD + SF indicating a robust proinflammatory milieu post-MI. Also, brain circadian genes (Bmal1, Clock) were downregulated in SF-subjected control mice, but remained elevated in OBD mice post-MI. SF superimposed on obesity dysregulated physiological inflammation and disrupted resolving response thereby impaired cardiac repair and signs of pathological inflammation.


Asunto(s)
Insuficiencia Cardíaca , Microbiota , Infarto del Miocardio , Masculino , Ratones , Animales , Privación de Sueño/complicaciones , Lipidómica , Ratones Endogámicos C57BL , Inflamación/complicaciones , Insuficiencia Cardíaca/etiología , Infarto del Miocardio/patología , Citocinas/genética , Obesidad/complicaciones
20.
Brain Behav Immun ; 117: 12-19, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38157946

RESUMEN

Microglia, resident immune cells in the central nervous system, constantly monitor the state of the surrounding brain activity. The animal model induced by sleep deprivation (SD) is widely used to study the pathophysiological mechanisms of insomnia and bipolar disorder. However, it remains unclear whether SD affects behaviors in young and aged male mice and microglia in various brain regions. In this study, we confirmed brain region-specific changes in microglial density and morphology in the accumbens nucleus (Acb), amygdala (AMY), cerebellum (Cb), corpus callosum (cc), caudate putamen, hippocampus (HIP), hypothalamus (HYP), medial prefrontal cortex (mPFC), and thalamus (TH) of young mice. In addition, the density of microglia in old mice was higher than that in young mice. Compared with young mice, old mice showed a markedly increased microglial size, decreased total length of microglial processes, and decreased maximum length. Importantly, we found that 48-h SD decreased microglial density and morphology in old mice, whereas SD increased microglial density and morphology in most observed brain regions in young mice. SD-induced hyperactivity was observed only in young mice but not in old mice. Moreover, microglial density (HIP, AMY, mPFC, CPu) was significantly positively correlated with behaviors in SD- and vehicle-treated young mice. Contrarily, negative correlations were shown between the microglial density (cc, Cb, TH, HYP, Acb, AMY) and behaviors in vehicle-treated young and old mice. These results suggest that SD dysregulates the homeostatic state of microglia in a region- and age-dependent manner. Microglia may be involved in regulating age-related behavioral responses to SD.


Asunto(s)
Microglía , Privación de Sueño , Ratones , Masculino , Animales , Encéfalo , Hipocampo , Amígdala del Cerebelo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA