Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Gene ; 887: 147732, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-37625565

ABSTRACT

Sleep deprivation disrupt the circadian clock and exercise performance. Defective oxidative stress caused by sleep deprivation may affect the expression of genes involved in cell apoptosis. Since a number of studies have shown the anti-apoptotic effect of L-arginine, so the aim of this study was to evaluate the effect of eight weeks of L-arginine supplementation on the expression of brain and muscle ARNT-like protein 1 (BMAL1), cell cycle and apoptosis regulator 2 (CCAR2), and BAX and BCL2 genes during sleep deprivation and acute anaerobic exercise. Participants included 20 healthy men age 26-35 years, randomized into the L-arginine intervention group (n = 10) and a placebo control (n = 10). The running-based anaerobic sprint test (RAST) was used for anaerobic exercise. Intervention subjects took one 1000 mg L-arginine tablet daily for 8 weeks. The Real-Time PCR method was used to determine apoptosis gene expression in peripheral blood mononuclear cells (PBMCs). Acute anaerobic exercise and sleep deprivation both increased the expression of BAX and CCAR2 genes, and decreased the expression of BCL2 and BMAL1 genes (p < 0.05 for all). L-arginine supplementation increased the expression of BMAL1 and BCL2 genes and decreased the expression of BAX and CCAR2 genes relative to control (p < 0.05). L-Arginine controlled the increase in expression of BAX and CCAR2 genes and the decrease in expression of BCL2 and BMAL1 genes in response to sleep deprivation and acute anaerobic exercise (p < 0.05). Our results showed that 24-hour sleep deprivation and acute anaerobic exercise increased the expression of pro-apoptotic genes (BAX and CCAR2) and decreased the expression of anti-apoptotic genes (BCL2 and BMAL1), although the effect of sleep deprivation is greater. In this situation, L-arginine supplementation may balance the apoptotic state of peripheral blood mononuclear cells. However, any recommendation about this needs further research.


Subject(s)
ARNTL Transcription Factors , Sleep Deprivation , Adult , Humans , Male , Adaptor Proteins, Signal Transducing/metabolism , Anaerobiosis , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , Dietary Supplements , Leukocytes, Mononuclear/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Sleep Deprivation/genetics , Sleep Deprivation/metabolism
2.
Nature ; 612(7940): 512-518, 2022 12.
Article in English | MEDLINE | ID: mdl-36477539

ABSTRACT

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


Subject(s)
Neurons , Sleep Duration , Sleep , Wakefulness , Animals , Mice , Electroencephalography , Neurons/metabolism , Neurons/physiology , Sleep/genetics , Sleep/physiology , Sleep Deprivation/genetics , Wakefulness/genetics , Wakefulness/physiology , Signal Transduction , Delta Rhythm , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Hypothalamus/cytology , Hypothalamus/physiology , Glutamic Acid/metabolism , Sleep, Slow-Wave/genetics , Sleep, Slow-Wave/physiology
3.
Sheng Li Xue Bao ; 74(4): 534-540, 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-35993204

ABSTRACT

Sleep deprivation (SD) has many deleterious health effects and occurs in more than 70% of pregnant women. However, the changes in sex hormones and relevant mechanisms after SD have not been well clarified. The aim of the present study was to explore the effects of SD on the secretion of sex hormones and the underlying mechanisms. Twelve pregnant Wistar rats were divided into control (CON, n = 6) and SD (n = 6) groups. Pregnant rats in the SD group were deprived of sleep for 18 h, and allowed free rest for 6 h, and then the above procedures were repeated until delivery. The CON group lived in a 12 h light/dark light cycle environment. Estradiol (E2) and progesterone (P4) levels were detected by enzyme-linked immunosorbent assay (ELISA), and the expression of circadian clock genes, Bmal1, Clock and Per2, in hypothalamus and pituitary gland tissues were evaluated by immunohistochemistry (IHC) and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The PI3K and Akt phosphorylation levels in the hypothalamic and pituitary tissues were determined by Western blot. The results showed that, compared with the CON group, the SD group exhibited significantly reduced serum E2 and P4 levels, down-regulated Bmal1, Clock and Per2 expression, as well as decreased phosphorylation levels of PI3K and Akt. But there was no significant difference of the total PI3K and Akt protein expression levels between the two groups. These results suggest that SD might affect the expression of the circadian clock genes in the hypothalamus and pituitary via PI3K/Akt pathway, and subsequently regulate the secretion of sex hormones in the pregnant rats, which hints the important roles of SD-induced changes of serum sex hormone levels in the pregnant rats.


Subject(s)
Circadian Clocks , Gonadal Steroid Hormones , Hypothalamus , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Sleep Deprivation , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Circadian Clocks/genetics , Circadian Clocks/physiology , Circadian Rhythm/genetics , Female , Gene Expression Regulation/genetics , Gonadal Steroid Hormones/genetics , Gonadal Steroid Hormones/metabolism , Hypothalamus/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Pituitary Gland/metabolism , Pregnancy , Progesterone , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Signal Transduction , Sleep Deprivation/genetics , Sleep Deprivation/metabolism
4.
Article in English | MEDLINE | ID: mdl-33373678

ABSTRACT

Many people consume coffee to attenuate increased sleepiness and impaired vigilance and attention due to insufficient sleep. We investigated in genetically caffeine sensitive men and women whether 'real world' coffee consumption during a simulated busy work week counteracts disabling consequences of chronically restricted sleep. We subjected homozygous C-allele carriers of ADORA2A (gene encoding adenosine A2A receptors) to five nights of only 5 h time-in-bed. We administered regular coffee (n = 12; 200 mg caffeine at breakfast and 100 mg caffeine after lunch) and decaffeinated coffee (n = 14) in double-blind fashion on all days following sleep restriction. At regular intervals four times each day, participants rated their sleepiness and performed the psychomotor vigilance test, the visual search task, and the visuo-spatial and letter n-back tasks. At bedtime, we quantified caffeine and the major caffeine metabolites paraxanthine, theobromine and theophylline in saliva. The two groups did not differ in age, body-mass-index, sex-ratio, chronotype and mood states. Subjective sleepiness increased in both groups across consecutive sleep restriction days and did not differ. By contrast, regular coffee counteracted the impact of repeated sleep loss on sustained and selective attention, as well as executive control when compared to decaffeinated coffee. The coffee also induced initial or transient benefits on different aspects of baseline performance during insufficient sleep. All differences between the groups disappeared after the recovery night and the cessation of coffee administration. The data suggest that 'real world' coffee consumption can efficiently attenuate sleep restriction-induced impairments in vigilance and attention in genetically caffeine sensitive individuals. German Clinical Trial Registry: # DRSK00014379.


Subject(s)
Attention/drug effects , Caffeine/administration & dosage , Coffee , Purinergic P1 Receptor Antagonists/administration & dosage , Receptor, Adenosine A2A/genetics , Sleep Deprivation/psychology , Adult , Alleles , Double-Blind Method , Female , Humans , Male , Neuropsychological Tests , Psychomotor Performance/drug effects , Sleep Deprivation/genetics , Wakefulness/drug effects
5.
PLoS One ; 15(7): e0236318, 2020.
Article in English | MEDLINE | ID: mdl-32726319

ABSTRACT

Lately, Drosophila has been favored as a model in sleep and circadian rhythm research due to its conserved mechanism and easily manageable operation. These studies have revealed the sophisticated parameters in whole-day sleep profiles of Drosophila, drawing connections between Drosophila sleep and human sleep. In this study, we tested several sleep deprivation protocols (mechanical shakes and light interruptions) on Drosophila and delineated their influences on Drosophila sleep. We applied a daytime light-deprivation protocol (DD) mimicking jet-lag to screen drugs that alleviate sleep deprivation. Characteristically, classical sleep-aid compounds exhibited different forms of influence: phenobarbital and pentobarbital modified total sleep time, while melatonin only shortened the latency to sleep. Such results construct the basis for further research on sleep benefits in other treatments in Drosophila. We screened seven herb extracts, and found very diverse results regarding their effect on sleep regulation. For instance, Panax notoginseng and Withania somnifera extracts displayed potent influence on total sleep time, while Melissa officinalis increased the number of sleep episodes. By comparing these treatments, we were able to rank drug potency in different aspects of sleep regulation. Notably, we also confirmed the presence of sleep difficulties in a Drosophila Alzheimer's disease (AD) model with an overexpression of human Abeta, and recognized clear differences between the portfolios of drug screening effects in AD flies and in the control group. Overall, potential drug candidates and receipts for sleep problems can be identified separately for normal and AD Drosophila populations, outlining Drosophila's potential in drug screening tests in other populations if combined with the use of other genetic disease tools.


Subject(s)
Plant Extracts/pharmacology , Sleep Deprivation/drug therapy , Sleep Wake Disorders/drug therapy , Sleep/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/genetics , Animals , Circadian Rhythm/drug effects , Disease Models, Animal , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Expression Regulation/genetics , Humans , Melatonin/pharmacology , Mutation , Panax notoginseng/chemistry , Phenobarbital/pharmacology , Plant Extracts/chemistry , Sleep/drug effects , Sleep/genetics , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology , Sleep Wake Disorders/genetics , Sleep Wake Disorders/physiopathology , Withania/chemistry
6.
Proc Natl Acad Sci U S A ; 115(17): 4483-4488, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29632177

ABSTRACT

The effects of acute sleep deprivation on ß-amyloid (Aß) clearance in the human brain have not been documented. Here we used PET and 18F-florbetaben to measure brain Aß burden (ABB) in 20 healthy controls tested after a night of rested sleep (baseline) and after a night of sleep deprivation. We show that one night of sleep deprivation, relative to baseline, resulted in a significant increase in Aß burden in the right hippocampus and thalamus. These increases were associated with mood worsening following sleep deprivation, but were not related to the genetic risk (APOE genotype) for Alzheimer's disease. Additionally, baseline ABB in a range of subcortical regions and the precuneus was inversely associated with reported night sleep hours. APOE genotyping was also linked to subcortical ABB, suggesting that different Alzheimer's disease risk factors might independently affect ABB in nearby brain regions. In summary, our findings show adverse effects of one-night sleep deprivation on brain ABB and expand on prior findings of higher Aß accumulation with chronic less sleep.


Subject(s)
Amyloid beta-Peptides/metabolism , Hippocampus/metabolism , Sleep Deprivation/diagnostic imaging , Sleep Deprivation/metabolism , Thalamus/metabolism , Adult , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Apolipoproteins E/genetics , Female , Genotype , Hippocampus/diagnostic imaging , Humans , Male , Middle Aged , Risk Factors , Sleep Deprivation/genetics , Thalamus/diagnostic imaging
7.
Sleep ; 40(6)2017 06 01.
Article in English | MEDLINE | ID: mdl-28444394

ABSTRACT

Study objectives: Shortened or mistimed sleep affects metabolic homeostasis, which may in part be mediated by dysregulation of endogenous circadian clocks. In this study, we assessed the contribution of sleep disruption to metabolic dysregulation by analysing diurnal transcriptome regulation in metabolic tissues of mice subjected to a sleep restriction (SR) paradigm. Methods: Male mice were subjected to 2 × 5 days of SR with enforced waking during the first 6 hours of the light phase. SR and control mice were sacrificed at different time points of the day and RNA preparations from the mediobasal hypothalamus (MBH), liver, and epididymal white adipose tissue (eWAT) were subjected to whole-genome microarray hybridization. Transcriptional rhythms were associated with changes in behavioral and physiological parameters such as sleep, body temperature, and food intake. Rhythm detection was performed with CircWave and transcription profiles were compared by 2-way analysis of variance and t-tests with Benjamini-Hochberg corrections. Results: Clock gene rhythms were blunted in all tissues, while transcriptome regulation was associated with either clock gene expression, sleep patterns, or food intake in a tissue-specific manner. Clock gene expression was associated with apoptosis pathways in the MBH and with tumor necrosis factor alpha signalling in liver. Food intake-associated genes included cilium movement genes in the MBH and lipid metabolism-associated transcripts in liver. Conclusions: In mice, repeated SR profoundly alters behavioral and molecular diurnal rhythms, disrupting essential signalling pathways in MBH, liver, and eWAT, which may underlie the metabolic and cognitive disturbances observed in sleep-restricted humans such as shift workers.


Subject(s)
Circadian Rhythm/genetics , Organ Specificity/genetics , Sleep Deprivation/genetics , Transcriptome , Adipose Tissue, White/metabolism , Animals , Apoptosis/genetics , Body Temperature/genetics , Circadian Clocks/genetics , Eating/genetics , Gene Expression Profiling , Gene Expression Regulation , Hypothalamus/metabolism , Lipid Metabolism/genetics , Liver/metabolism , Male , Mice , Sleep/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
Peptides ; 88: 55-61, 2017 02.
Article in English | MEDLINE | ID: mdl-27988352

ABSTRACT

Orexins, also known as hypocretins, play a regulatory role in the sleep-wake cycle by activating orexin receptors. Previous animal studies have shown that sleep deprivation can elevate orexinergic peptide levels. However, the relationship between insomnia disorder and orexin-A levels in humans has not been explored. In the current study, we examined plasma orexin-A levels in patients with insomnia disorder and in normal sleepers. We also studied the possible mechanisms underlying changes in orexin-A levels between the study groups; this included investigations of prepro-orexin and orexin receptor gene polymorphisms as well as exploration of other variables. We measured plasma orexin-A levels in 228 patients with insomnia disorder and 282 normal sleepers. The results indicated that the patients with insomnia disorder had significantly higher orexin-A levels than normal sleepers (63.42±37.56 vs. 54.84±23.95pg/ml). A positive relationship was detected between orexin-A level and age in patients with insomnia disorder. Orexin-A levels were elevated in relation to course of insomnia, as well as in relation to increased Insomnia Severity Index score. None of the evaluated prepro-orexin gene single nucleotide polymorphisms were informative between the two study populations. After sequencing all orexin receptor exons, one variation (rs2271933) in the OX1R gene and one variation (rs2653349) in the OX2R gene were found. However, no significant differences were found in either genotypic or allelic frequency distributions between the two study groups. It is suggested that the increased plasma orexin-A levels in patients with insomnia disorder are associated with the course and severity of insomnia, but not with prepro-orexin and orexin receptor gene polymorphisms.


Subject(s)
Orexin Receptors/genetics , Orexins/blood , Sleep Initiation and Maintenance Disorders/genetics , Adult , Aged , Animals , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Hypothalamus/metabolism , Male , Middle Aged , Orexin Receptors/blood , Polymorphism, Single Nucleotide , Sleep/genetics , Sleep/physiology , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology , Sleep Initiation and Maintenance Disorders/blood , Sleep Initiation and Maintenance Disorders/physiopathology
9.
Eur Neuropsychopharmacol ; 26(4): 767-76, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26857197

ABSTRACT

The postpartum period is characterized by a post-withdrawal hormonal status, sleep deprivation, and susceptibility to affective disorders. Postpartum mothering involves automatic and attentional processes to screen out new external as well as internal stimuli. The present study investigated sensorimotor gating in relation to sleep duration, depression, as well as catecholaminergic and oxytocinergic genotypes in postpartum women. Prepulse inhibition (PPI) of the startle reflex and startle reactivity were assessed two months postpartum in 141 healthy and 29 depressed women. The catechol-O-methyltransferase (COMT) Val158Met, and oxytocin receptor (OXTR) rs237885 and rs53576 polymorphisms were genotyped, and data on sleep duration were collected. Short sleep duration (less than four hours in the preceding night) and postpartum depression were independently associated with lower PPI. Also, women with postpartum depression had higher startle reactivity in comparison with controls. The OXTR rs237885 genotype was related to PPI in an allele dose-dependent mode, with T/T healthy postpartum women carriers displaying the lowest PPI. Reduced sensorimotor gating was associated with sleep deprivation and depressive symptoms during the postpartum period. Individual neurophysiological vulnerability might be mediated by oxytocinergic genotype which relates to bonding and stress response. These findings implicate the putative relevance of lower PPI of the startle response as an objective physiological correlate of liability to postpartum depression.


Subject(s)
Depression, Postpartum/genetics , Postpartum Period/psychology , Prepulse Inhibition/genetics , Receptors, Oxytocin/genetics , Reflex, Startle/genetics , Sleep Deprivation/genetics , Sleep Deprivation/psychology , Acoustic Stimulation , Adult , Alleles , Case-Control Studies , Catechol O-Methyltransferase/genetics , Depression, Postpartum/physiopathology , Depression, Postpartum/psychology , Female , Genotype , Humans , Polymorphism, Genetic/genetics , Postpartum Period/genetics , Young Adult
10.
J Affect Disord ; 192: 64-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26707349

ABSTRACT

BACKGROUND: Combined Total sleep deprivation (TSD) and light therapy (LT) cause a rapid improvement in bipolar depression which has been hypothesized to be paralleled by changes in sleep homeostasis. Recent studies showed that bipolar patients had lower changes of EEG theta power after sleep and responders to antidepressant TSD+LT slept less and showed a lower increase of EEG theta power then non-responders. A polymorphism in PER3 gene has been associated with diurnal preference, sleep structure and homeostatic response to sleep deprivation in healthy subjects. We hypothesized that the individual variability in the homeostatic response to TSD could be a correlate of antidepressant response and be influenced by genetic factors. METHODS: We administered three TSD+LT cycles to bipolar depressed patients. Severity of depression was rated on Hamilton Depression Rating Scale. Actigraphic recordings were performed in a group of patients. RESULTS: PER3 polymorphism influenced changes in total sleep time (F=2.24; p=0.024): while PER3(4/4) and PER3(4/5) patients showed a reduction in it after treatment, PER3(5/5) subjects showed an increase of about 40min, suggesting a higher homeostatic pressure. The same polymorphism influenced the change of depressive symptomatology during treatment (F=3.72; p=0.028). LIMITATIONS: Sleep information was recorded till the day after the end of treatment: a longer period of observation could give more information about the possible maintenance of allostatic adaptation. CONCLUSIONS: A higher sleep homeostatic pressure reduced the antidepressant response to TSD+LT, while an allostatic adaptation to sleep loss was associated with better response. This process seems to be under genetic control.


Subject(s)
Bipolar Disorder/therapy , Depression/therapy , Period Circadian Proteins/genetics , Sleep Deprivation/genetics , Sleep/genetics , Actigraphy , Adult , Bipolar Disorder/genetics , Bipolar Disorder/psychology , Depression/genetics , Depression/psychology , Female , Homeostasis/genetics , Humans , Male , Middle Aged , Phototherapy , Polymorphism, Genetic , Pressure , Psychiatric Status Rating Scales , Sleep Deprivation/psychology , Treatment Outcome
11.
Sleep ; 39(3): 613-24, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26612390

ABSTRACT

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.


Subject(s)
Circadian Rhythm/physiology , Receptor, EphA4/metabolism , Sleep Deprivation/physiopathology , Sleep/physiology , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Darkness , Electrocorticography , Electrophysiological Phenomena , Homeostasis , Light , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity , Promoter Regions, Genetic/genetics , Rats , Rats, Sprague-Dawley , Receptor, EphA4/biosynthesis , Receptor, EphA4/deficiency , Receptor, EphA4/genetics , Sleep/genetics , Sleep Deprivation/genetics , Sleep, REM/genetics , Sleep, REM/physiology , Suprachiasmatic Nucleus/metabolism , Thalamus/metabolism , Time Factors , Wakefulness/genetics , Wakefulness/physiology
12.
Amino Acids ; 48(4): 949-957, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26645537

ABSTRACT

The purpose of this study was to identify sleep deprivation-induced atrophy and the muscle-specific fiber types affected and to determine the effects of leucine supplementation on atrophy and pertinent portions of the pathways of muscle protein synthesis and degradation in rats. A total of 46 Wistar rats were distributed in four groups: control (CTL), leucine supplementation (LEU), sleep deprivation (SD), and leucine supplementation + sleep deprivation (LEU + SD). Leucine supplementation was by gavage (1.35 g/kg/daily), and the animals were subjected to SD for 96 h. Testosterone and corticosterone concentrations, along with proteins involved in protein synthesis and degradation and proteasome activity levels, were measured in the gastrocnemius (GA) muscle. Myosin ATPase staining was used to evaluate the different muscle fibers. After sleep deprivation, GA muscle and body masses decreased in the SD group compared to the CTL, LEU, and LEU + SD groups. There was no difference between groups in type I fiber cross-sectional area (CSA). The CSAs for type IIa fibers were lower in the SD and LEU + SD groups vs. the CTL and LEU groups, while the IIb fiber CSA was lower in the SD group vs. the CSAs in all other groups. The phospho (p)-Akt levels were lower in the SD and LEU + SD groups vs. the CTL and LEU groups. The p-mTORC1 levels were higher in the LEU, SD, and LEU + SD groups vs. the CTL group. The p-p70S6k levels were higher in the LEU and LEU + SD groups; the 4E-BP1 levels were higher in the SD and LEU + SD groups compared to those in the CTL and LEU groups, and the p-4E-BP1 levels were higher in the LEU and SD groups compared to those in the CTL group and even higher in the LEU + SD group compared to those in the LEU and SD groups. Ubiquitinated proteins, LC3, and p62/SQSTM, and proteasome activity levels were higher in the SD and LEU + SD groups vs. the LEU and CTL groups. Sleep deprivation led to the atrophy of IIa and IIb muscle fibers; however, leucine supplementation prevented muscle loss and type IIb fiber atrophy.


Subject(s)
Leucine/administration & dosage , Muscle Fibers, Skeletal/drug effects , Muscular Atrophy/drug therapy , Sleep Deprivation/drug therapy , Administration, Oral , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Corticosterone/metabolism , Dietary Supplements , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins , Male , Mechanistic Target of Rapamycin Complex 1 , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Muscle Fibers, Skeletal/classification , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Atrophy/complications , Muscular Atrophy/genetics , Muscular Atrophy/physiopathology , Myosins/genetics , Myosins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Rats , Rats, Wistar , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Sleep Deprivation/complications , Sleep Deprivation/genetics , Sleep Deprivation/physiopathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Testosterone/metabolism
13.
Ann Neurol ; 78(2): 235-47, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25940842

ABSTRACT

OBJECTIVE: Even though wakefulness at night leads to profound performance deterioration and is regularly experienced by shift workers, its cerebral correlates remain virtually unexplored. METHODS: We assessed brain activity in young healthy adults during a vigilant attention task under high and low sleep pressure during night-time, coinciding with strongest circadian sleep drive. We examined sleep-loss-related attentional vulnerability by considering a PERIOD3 polymorphism presumably impacting on sleep homeostasis. RESULTS: Our results link higher sleep-loss-related attentional vulnerability to cortical and subcortical deactivation patterns during slow reaction times (i.e., suboptimal vigilant attention). Concomitantly, thalamic regions were progressively less recruited with time-on-task and functionally less connected to task-related and arousal-promoting brain regions in those volunteers showing higher attentional instability in their behavior. The data further suggest that the latter is linked to shifts into a task-inactive default-mode network in between task-relevant stimulus occurrence. INTERPRETATION: We provide a multifaceted view on cerebral correlates of sleep loss at night and propose that genetic predisposition entails differential cerebral coping mechanisms, potentially compromising adequate performance during night work.


Subject(s)
Arousal/genetics , Attention/physiology , Brain/physiopathology , Circadian Rhythm/genetics , Period Circadian Proteins/genetics , Reaction Time/genetics , Sleep Deprivation/genetics , Adult , Arousal/physiology , Brain Stem/physiopathology , Circadian Rhythm/physiology , Female , Functional Neuroimaging , Gyrus Cinguli/physiopathology , Humans , Magnetic Resonance Imaging , Male , Neural Pathways/physiopathology , Polymorphism, Genetic , Prefrontal Cortex/physiopathology , Psychomotor Performance/physiology , Reaction Time/physiology , Sleep Deprivation/physiopathology , Sleep Deprivation/psychology , Thalamus/physiopathology , Young Adult
14.
Sleep ; 33(1): 19-28, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20120617

ABSTRACT

STUDY OBJECTIVES: Genetic manipulation of cAMP-dependent protein kinase A (PKA) in Drosophila has implicated an important role for PKA in sleeplwake state regulation. Here, we characterize the role of this signaling pathway in the regulation of sleep using electroencephalographic (EEG) and electromyographic (EMG) recordings in R(AB) transgenic mice that express a dominant negative form of the regulatory subunit of PKA in neurons within cortex and hippocampus. Previous studies have revealed that these mutant mice have reduced PKA activity that results in the impairment of hippocampus-dependent long-term memory and long-lasting forms of hippocampal synaptic plasticity. DESIGN: PKA assays, in situ hybridization, immunoblots, and sleep studies were performed in R(AB) transgenic mice and wild-type control mice. MEASUREMENTS AND RESULTS: We have found that R(AB) transgenic mice have reduced PKA activity within cortex and reduced Ser845 phosphorylation of the glutamate receptor subunit GluR1. R(AB) transgenic mice exhibit non-rapid eye movement (NREM) sleep fragmentation and increased amounts of rapid eye movement (REM) sleep relative to wild-type mice. Further, R(AB) transgenic mice have more delta power but less sigma power during NREM sleep relative to wild-type mice. After sleep deprivation, the amounts of NREM and REM sleep were comparable between wild-type and R(AB) transgenic mice. However, the homeostatic rebound of sigma power in R(AB) transgenic mice was reduced. CONCLUSIONS: Alterations in cortical synaptic receptors, impairments in sleep continuity, and alterations in sleep oscillations in R(AB) mice imply that PKA is involved not only in synaptic plasticity and memory storage but also in the regulation of sleep/wake states.


Subject(s)
Biological Clocks/genetics , Cerebral Cortex/physiology , Cyclic AMP-Dependent Protein Kinases/genetics , Electroencephalography , Sleep/genetics , Thalamus/physiology , Animals , Circadian Rhythm/genetics , Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/genetics , Electromyography , Female , Gene Expression/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Pathways/physiology , Neuronal Plasticity/genetics , Neurons/physiology , Receptors, AMPA/genetics , Receptors, Neurotransmitter/genetics , Retention, Psychology/physiology , Sleep Deprivation/genetics , Sleep Stages/genetics , Wakefulness/genetics
15.
Am J Physiol Endocrinol Metab ; 298(3): E726-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20051529

ABSTRACT

Several pieces of evidence support that sleep duration plays a role in body weight control. Nevertheless, it has been assumed that, after the identification of orexins (hypocretins), the molecular basis of the interaction between sleep and energy homeostasis has been provided. However, no study has verified the relationship between neuropeptide Y (NPY) and orexin changes during hyperphagia induced by sleep deprivation. In the current study we aimed to establish the time course of changes in metabolite, endocrine, and hypothalamic neuropeptide expression of Wistar rats sleep deprived by the platform method for a distinct period (from 24 to 96 h) or sleep restricted for 21 days (SR-21d). Despite changes in the stress hormones, we found no changes in food intake and body weight in the SR-21d group. However, sleep-deprived rats had a 25-35% increase in their food intake from 72 h accompanied by slight weight loss. Such changes were associated with increased hypothalamus mRNA levels of prepro-orexin (PPO) at 24 h followed by NPY at 48 h of sleep deprivation. Conversely, sleep recovery reduced the expression of both PPO and NPY, which rapidly brought the animals to a hypophagic condition. Our data also support that sleep deprivation rapidly increases energy expenditure and therefore leads to a negative energy balance and a reduction in liver glycogen and serum triacylglycerol levels despite the hyperphagia. Interestingly, such changes were associated with increased serum levels of glucagon, corticosterone, and norepinephrine, but no effects on leptin, insulin, or ghrelin were observed. In conclusion, orexin activation accounts for the myriad changes induced by sleep deprivation, especially the hyperphagia induced under stress and a negative energy balance.


Subject(s)
Hyperphagia/metabolism , Hypothalamus/metabolism , Neuropeptide Y/metabolism , Sleep Deprivation/genetics , Animals , Body Weight , Eating , Gene Expression Regulation , Male , Rats , Rats, Wistar , Sleep Deprivation/metabolism
16.
Integr Cancer Ther ; 8(4): 329-36, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19926609

ABSTRACT

Circadian disruption has been linked with inflammation, an established cancer risk factor. Per3 clock gene polymorphisms have also been associated with circadian disruption and with increased cancer risk. Patients completed a questionnaire and provided a blood sample prior to undergoing a colonoscopy (n = 70). Adjusted mean serum cytokine concentrations (IL-6, TNF-alpha, gamma-INF, IL-1ra, IL-1-beta, VEGF) were compared among patients with high and low scores for fatigue (Multidimensional Fatigue Inventory), depressive symptoms (Beck Depression Inventory II), or sleep disruption (Pittsburgh Sleep Quality Index), or among patients with different Per3 clock gene variants. Poor sleep was associated with elevated VEGF, and fatigue-related reduced activity was associated with elevated TNF-alpha concentrations. Participants with the 4/5 or 5/5 Per3 variable tandem repeat sequence had elevated IL-6 concentrations compared to those with the 4/4 genotype. Biological processes linking circadian disruption with cancer remain to be elucidated. Increased inflammatory cytokine secretion may play a role.


Subject(s)
Chronobiology Disorders/genetics , Cytokines/blood , Period Circadian Proteins/genetics , Polymorphism, Genetic , Sleep Deprivation/genetics , Chronobiology Disorders/blood , Colonoscopy , Depression/blood , Depression/genetics , Fatigue/blood , Fatigue/genetics , Genetic Variation , Genotype , Humans , Inflammation/blood , Inflammation/genetics , Interleukin-6/blood , Male , Middle Aged , Sleep Deprivation/blood , Surveys and Questionnaires , Tandem Repeat Sequences , Tumor Necrosis Factor-alpha/blood , Vascular Endothelial Growth Factor A/blood
17.
Curr Pharm Des ; 15(22): 2637-49, 2009.
Article in English | MEDLINE | ID: mdl-19689334

ABSTRACT

Despite confirmed evidences about some neurochemical effects of antidepressant treatments, there is still a high level of uncertainty about which biological changes are needed to recover from a major depressive episode. Changes of monoaminergic neurotransmission are paralleled by profound changes in brain metabolism, neural responses to stimuli, sleep architecture, biological rhythms, and, at the intracellular level, neuronal signaling pathways regulating gene expression, neuroplasticity, and neurotrophic mechanisms. Sleep deprivation targets the biological mechanisms which are responsible for the possibility, unique to mood disorders, of rapid switching between depression, euthymia, and mania. The rapidity of action of sleep deprivation enables the study of the correlates of antidepressant response at close time points, providing a good model to study the biological basis of the antidepressant response and of the pathophysiology of affective illness. Current knowledge suggests that multiple neurobiological effects of sleep deprivation are responsible for the clinical mood amelioration, suggesting a multi-target mechanism of action. An impressive group of brain imaging studies using different brain imaging techniques (positron emission tomography, single photon emission tomography, functional magnetic resonance imaging, proton spectroscopy, arterial spin labeling) showed that clinical response is associated with changes in the functioning of specific brain areas. The combination of these new methodological acquisitions with the classical neurobiological and pharmacogenetic perspective provides an evolving knowledge about brain changes associated with antidepressant response, and will then help to identify the real targets of antidepressant treatment.


Subject(s)
Brain/diagnostic imaging , Chronotherapy/methods , Drug Discovery/methods , Mood Disorders/genetics , Mood Disorders/therapy , Serotonin Plasma Membrane Transport Proteins/genetics , Sleep Deprivation/diagnostic imaging , Sleep Deprivation/genetics , Animals , Biogenic Monoamines/metabolism , Brain/metabolism , Brain/pathology , Diagnostic Imaging , Humans , Magnetic Resonance Imaging , Mood Disorders/metabolism , Positron-Emission Tomography , Sleep Deprivation/metabolism , Thyroid Hormones/metabolism
18.
Sleep ; 31(7): 927-33, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18652088

ABSTRACT

STUDY OBJECTIVES: Chronic sleep deprivation of rats causes hyperphagia without body weight gain. Sleep deprivation hyperphagia is prompted by changes in pathways governing food intake; hyperphagia may be adaptive to sleep deprivation hypermetabolism. A recent paper suggested that sleep deprivation might inhibit ability of rats to increase food intake and that hyperphagia may be an artifact of uncorrected chow spillage. To resolve this, a palatable liquid diet (Ensure) was used where spillage is insignificant. DESIGN: Sleep deprivation of male Sprague Dawley rats was enforced for 10 days by the flowerpot/platform paradigm. Daily food intake and body weight were measured. On day 10, rats were transcardially perfused for analysis of hypothalamic mRNA expression of the orexigen, neuropeptide Y (NPY). SETTING: Morgan State University, sleep deprivation and transcardial perfusion; University of Maryland, NPY in situ hybridization and analysis. MEASUREMENTS AND RESULTS: Using a liquid diet for accurate daily measurements, there was no change in food intake in the first 5 days of sleep deprivation. Importantly, from days 6-10 it increased significantly, peaking at 29% above baseline. Control rats steadily gained weight but sleep-deprived rats did not. Hypothalamic NPY mRNA levels were positively correlated to stimulation of food intake and negatively correlated with changes in body weight. CONCLUSION: Sleep deprivation hyperphagia may not be apparent over the short term (i.e., < or = 5 days), but when extended beyond 6 days, it is readily observed. The timing of changes in body weight and food intake suggests that the negative energy balance induced by sleep deprivation prompts the neural changes that evoke hyperphagia.


Subject(s)
Hyperphagia/psychology , Sleep Deprivation/psychology , Animals , Body Weight/genetics , Energy Metabolism/genetics , Gene Expression Regulation/genetics , Hyperphagia/genetics , Hypothalamus/pathology , Male , Neuropeptide Y/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Sleep Deprivation/genetics
19.
Br Med Bull ; 86: 23-32, 2008.
Article in English | MEDLINE | ID: mdl-18487629

ABSTRACT

BACKGROUND: A long-standing challenge in the treatment of depression is the development of a rapidly acting antidepressant. Conventional antidepressants typically require 2-8 weeks for clinical remission. In contrast, chronobiological interventions such as sleep deprivation treatment dramatically reduce depressive symptoms within 24-48 h in 40-60% of depressed subjects. It is hypothesized that fast-acting treatments for depression may alter circadian rhythms through chronobiological mechanisms relevant to clock gene function. SOURCES OF DATA: A bibliographic review using Entrez PubMed with Boolean search terms 'circadian' and 'depressive' identified more than 1000 clinical papers published over a 40-year period (1966-present). AREAS OF AGREEMENT: A large body of clinical data reports that sleep, temperature, hormone and mood changes in depression are consistent with disturbances in circadian-related processes. AREAS OF CONTROVERSY: Consensus has not been achieved in terms of defining underlying chronobiological mechanisms for optimal methods to produce rapid and sustained antidepressant responses to circadian interventions. GROWING POINTS: Chronobiological augmentation using combinations of sleep deprivation with light therapy and/or sleep phase advance in medicated patients supports a clinical strategy for accelerating and sustaining antidepressant responses. AREAS TIMELY FOR DEVELOPING RESEARCH: Advances in technology including improved assays for clock gene expression will facilitate exploring the role of clock genes and may lead to new rapidly acting antidepressant strategies and potential novel drug targets.


Subject(s)
Antidepressive Agents/therapeutic use , Biological Clocks/genetics , Circadian Rhythm/genetics , Depressive Disorder/therapy , Sleep Deprivation/genetics , Biological Clocks/physiology , Circadian Rhythm/physiology , Depressive Disorder/genetics , Depressive Disorder/physiopathology , Humans , Sleep Deprivation/physiopathology
20.
Endocrinology ; 147(1): 421-31, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16210372

ABSTRACT

Chronic rapid eye movement (paradoxical) sleep deprivation (REM-SD) of rats leads to two conspicuous pathologies: hyperphagia coincident with body weight loss, prompted by elevated metabolism. Our goals were to test the hypotheses that 1) as a stressor, REM-SD would increase CRH gene expression in the hypothalamus and that 2) to account for hyperphagia, hypothalamic gene expression of the orexigen neuropeptide Y (NPY) would increase, but expression of the anorexigen proopiomelanocortin (POMC) would decrease. Enforcement of REM-SD of adult male rats for 20 d with the platform (flowerpot) method led to progressive hyperphagia, increasing to approximately 300% of baseline; body weight steadily declined by approximately 25%. Consistent with changes in food intake patterns, NPY expression rapidly increased in the hypothalamic arcuate nucleus by d 5 of REM-SD, peaking at d 20; by contrast, POMC expression decreased progressively during REM-SD. CRH expression was increased by d 5, both in mRNA and ability to detect neuronal perikaryal staining in paraventricular nucleus with immunocytochemistry, and it remained elevated thereafter with modest declines. Taken together, these data indicate that changes in hypothalamic neuropeptides regulating food intake are altered in a manner consistent with the hyperphagia seen with REM-SD. Changes in CRH, although indicative of REM-SD as a stressor, suggest that the anorexigenic actions of CRH are ineffective (or disabled). Furthermore, changes in NPY and POMC agree with current models of food intake behavior, but they are opposite to their acute effects on peripheral energy metabolism and thermogenesis.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Hypothalamus/physiopathology , Neuropeptide Y/genetics , Pro-Opiomelanocortin/genetics , Sleep Deprivation/physiopathology , Sleep, REM/physiology , Animals , Male , Rats , Rats, Sprague-Dawley , Sleep Deprivation/genetics , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL