Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 101
Filter
1.
Basic Clin Pharmacol Toxicol ; 128(2): 256-267, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32975883

ABSTRACT

Post-operative sleep disorders induce adverse effects on patients, especially the elderly, which may be associated with surgery and inhalational anaesthetics. Melatonin is a neuroendocrine regulator of the sleep-wake cycle. In this study, we analysed the alterations of post-operative sleep in aged melatonin-deficient (C57BL/6J) mice, and investigated if exogenous melatonin could facilitate entrainment of circadian rhythm after laparotomy under sevoflurane anaesthesia. The results showed that laparotomy under sevoflurane anaesthesia had a greater influence on post-operative sleep than sevoflurane alone. Laparotomy under anaesthesia led to circadian rhythm shifting forward, altered EEG power density and delta power of NREM sleep, and lengthened REM and NREM sleep latencies. In the light phase, the number of waking episodes tended to decline, and wake episode duration elevated. However, these indicators presented the opposite tendency during the dark phase. Melatonin showed significant efficacy for ameliorating the sleep disorder and restoring physiological sleep, and most of the beneficial effect of melatonin was antagonized by luzindole, a melatonin receptor antagonist.


Subject(s)
Anesthetics, Inhalation/toxicity , Circadian Rhythm/drug effects , Laparotomy/adverse effects , Melatonin/pharmacology , Postoperative Complications/prevention & control , Sevoflurane/toxicity , Sleep Aids, Pharmaceutical/pharmacology , Sleep Stages/drug effects , Sleep Wake Disorders/prevention & control , Activity Cycles/drug effects , Age Factors , Animals , Electroencephalography , Electromyography , Female , Melatonin/deficiency , Mice, Inbred C57BL , Photoperiod , Postoperative Complications/etiology , Postoperative Complications/metabolism , Postoperative Complications/physiopathology , Sleep Wake Disorders/etiology , Sleep Wake Disorders/metabolism , Sleep Wake Disorders/physiopathology , Sleep, REM/drug effects , Time Factors
2.
Nutrients ; 10(4)2018 Mar 23.
Article in English | MEDLINE | ID: mdl-29570610

ABSTRACT

Sialic acid (SA) is an integral component of gangliosides and signaling molecules in the brain and its dietary intake may support cognitive development. We previously reported that feeding sialyllactose, a milk oligosaccharide that contains SA, alters SA content and diffusivity in the pig brain. The present research sought to expand upon such results and describe the effects of feeding sialyllactose on recognition memory and sleep/wake activity using a translational pig model. Pigs were provided ad libitum access to a customized milk replacer containing 0 g/L or 380 g/L of sialyllactose from postnatal day (PND) 2-22. Beginning on PND 15, pigs were fitted with accelerometers to track home-cage activity and testing on the novel object recognition task began at PND 17. There were no significant effects of diet on average daily body weight gain, average daily milk intake, or the gain-to-feed ratio during the study (all p ≥ 0.11). Pigs on both diets were able to display recognition memory on the novel object recognition task (p < 0.01), but performance and exploratory behavior did not differ between groups (all p ≥ 0.11). Total activity and percent time spent sleeping were equivalent between groups during both day and night cycles (all p ≥ 0.56). Dietary sialyllactose did not alter growth performance of young pigs, and there was no evidence that providing SA via sialyllactose benefits the development of recognition memory or gross sleep-related behaviors.


Subject(s)
Activity Cycles/drug effects , Behavior, Animal/drug effects , Circadian Rhythm/drug effects , Diet , Lactose/analogs & derivatives , Recognition, Psychology/drug effects , Sialic Acids/administration & dosage , Age Factors , Animal Feed , Animals , Lactose/administration & dosage , Locomotion/drug effects , Male , Sus scrofa , Time Factors
3.
Anesth Analg ; 127(1): 255-262, 2018 07.
Article in English | MEDLINE | ID: mdl-29261550

ABSTRACT

BACKGROUND: Increasing evidence suggests that patients with eating disorders are more likely to develop chronic pain. A misaligned diet has been reported to disrupt the sleep-awake rhythms. Combined with our previous investigation on circadian pain, we aimed to investigate the role of misaligned diet in the pain sensitivity and the underlying mechanisms. METHODS: Two-month-old C57BL/6J male mice were administered chronic constriction injury (CCI) surgery to establish neuropathic pain models. CCI mice were randomized to scheduled food access throughout the whole day (CCI-free), during the daytime (CCI-misaligned), and at night (CCI-aligned), respectively. The paw withdrawal mechanical threshold, indicating pain behavior, was measured by Von Frey. The gross motor activity pattern indicating the sleep-awake rhythm was monitored by Mini-Mitter. Melatonin (Mel) was administered to ameliorate the sleep-awake rhythm (CCI-free + Mel and CCI-misaligned + Mel). The expressions of circadian pain-related proteins were detected by quantitative polymerase chain reaction and western blot. The primary outcome is the pain threshold and the secondary outcome is the sleep-awake rhythm. RESULTS: Misaligned diet during the peri-CCI surgery period significantly decreased the paw withdrawal mechanical threshold compared with the CCI-free mice (day 14: 0.40 ± 0.09 vs 0.64 ± 0.15; P = .03;) and altered the sleep-awake rhythm. Mel pretreatment alleviated the increased pain (day 14, CCI-misaligned + Mel versus CCI-misaligned: day 14: 0.60 ± 0.13 vs 0.35 ± 0.12; P = .022) and the disrupted sleep-awake rhythm caused by misaligned feeding. The mRNA levels of N-methyl-D-aspartate receptor subtype 2B (NR2B), Ca/calmodulin-dependent protein kinase II (CaMKII), and cyclic adenosine monophosphate-response element binding protein (CREB) in the spinal dorsal horn increased in CCI-misaligned mice compared with the CCI-free mice. The phosphor-NR2B, phosphor-CaMKII, and phosphor-CREB also increased in CCI-misaligned mice compared with the CCI-free mice. However, the expressions of NR2B, CaMKII, and CREB were decreased in CCI-misaligned + Mel mice compared to CCI-misaligned mice at both transcriptional and translational levels. CONCLUSIONS: Misaligned diet might aggravate pain sensitivity through the disruption of the sleep-awake cycle, which could be recovered by Mel. NR2B-CaMKII-CREB may participate in the disruption of sleep-awake rhythm-mediated pain aggravation.


Subject(s)
Activity Cycles , Behavior, Animal , Feeding Behavior , Meals , Motor Activity , Neuralgia/physiopathology , Sleep , Activity Cycles/drug effects , Animals , Behavior, Animal/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Male , Melatonin/pharmacology , Mice, Inbred C57BL , Motor Activity/drug effects , Neuralgia/metabolism , Neuralgia/prevention & control , Neuralgia/psychology , Pain Perception , Pain Threshold , Phosphorylation , Posterior Horn Cells/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Sleep/drug effects , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/physiopathology , Time Factors
4.
Chronobiol Int ; 34(9): 1273-1287, 2017.
Article in English | MEDLINE | ID: mdl-29039977

ABSTRACT

Exposure to environmental conditions that disturb the daily rhythms has been shown to enhance the proinflammatory responses of immunostimulant-challenged immune system. However, it is not known whether circadian disturbances may stimulate unchallenged immune responses and thus contribute per se to the development of inflammation-related diseases. Our aim was to ascertain an effect of various conditions threatening the behavioral activity/rest cycle regulation, namely aging with or without melatonin, 6 h advance/delay phase shifts in the light/dark cycle repeated with a 2-day frequency and constant light, on expression of immune markers in the rat colon. The impact of these conditions on parameters of behavioral activity and mRNA levels of selected immune markers in the colonic mucosa of Wistar rats, namely TNFα (Tnf), IL1a (Il1a), IL17RA (Il17ra), STAT3 (Stat3) and Rgs16 (Rsg16), were detected. Our results demonstrate that aging with or without melatonin as well as repeated 6 h advance/delay phase shifts in the light/dark cycle, which increased inactivity as a correlate of sleep during the dark phase of the light/dark cycle (i.e. during the active phase for nocturnal animals), had a minor effect on immune state in the colonic mucosa; all these conditions caused downregulation of gene Rgs16 which is involved in attenuation of the inflammatory response in the colon but did not affect expression of the other immune markers. Interestingly, a long-term absence of melatonin facilitated the aging-induced effect on immune state in the colon. In contrast, exposure to constant light, which perturbed the interval of inactivity (sleep) and led to the complete abolishment of activity/inactivity cycles, activated robustly proinflammatory state in the colon selectively via Stat3-dependent pathway. In spite all these experimental conditions (aging with or without melatonin, shifts in light/dark cycles, constant light) perturbed the activity/rest cycles, none of them induced sleep deprivation. These results provided the first evidence that disruptions in the behavioral activity/inactivity cycles may spontaneously (without immuno-stimulant) induce selective proinflammatory responses in the colonic mucosa. Such effects may take part in the mechanisms of modern lifestyle-induced inflammatory diseases of the gut. ABBREVIATIONS: B2M: ß2-microglobulin; DSS: dextran sodium sulfate; Gapdh: glyceraldehyde-3-phosphate dehydrogenase; Ifng: interferon g; Il1a: interleukin 1a; Il1b: interleukin 1b; Il2: interleukin 2; Il6: interleukin 6; Il17ra: interleukin 17 receptor a; LD: light/dark cycle; LL: constant light; LPS: lipopolysaccharide; Mntr1a: melatonin receptor 1a; PINX: pinealectomy; Rgs16: regulator of G protein signaling 16; RT qPCR: quantitative reverse transcription polymerase chain reaction; Stat3: signal transducer and activator of transcription 3; Th17: type 17 T helper cells; Tnfα: tumor necrosis factor α; Tnfrsf1b: tumor necrosis factor receptor superfamily member 1b.


Subject(s)
Circadian Rhythm/physiology , Colon/drug effects , Melatonin/pharmacology , Sleep Deprivation/drug therapy , Sleep/drug effects , Activity Cycles/drug effects , Animals , Colon/metabolism , Light , Motor Activity/drug effects , Motor Activity/physiology , Photoperiod , Rats, Wistar , Sleep/physiology , Time Factors
5.
Eur J Gastroenterol Hepatol ; 29(3): 302-308, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27977438

ABSTRACT

BACKGROUND AND AIM: Sleep disorders are frequently reported in patients with cirrhosis and hepatic encephalopathy (HE). This study assessed the effect of rifaximin on sleep architecture parameters in patients with recurrent HE. PATIENTS AND METHODS: This sequential, prospective, and exploratory study involved all patients with cirrhosis and recurrent HE admitted between June 2014 and September 2015. HE was assessed according to the West-Haven Classification. Patients underwent 24-h polysomnography (PSG) and 7-day actigraphy. Rapid eye movement (REM) sleep was considered to be an indicator of good sleep quality. Patients completed questionnaires assessing the quality of sleep and sleepiness. After a 28-day course of rifaximin, the same assessment was repeated. RESULTS: Fifteen patients were included (nine men, mean age: 57±11 years). Child-Pugh scores ranged from B7 to C15. Before rifaximin, the mean HE score was 2.7±0.7. Data from PSG analysis indicated long total sleep time (TST): 571±288 min, and limited REM sleep: 2.5% TST (0-19). Seven-day actigraphy showed an impaired number of steps: 1690/24 h (176-6945). Questionnaires indicated that patients experienced impaired sleep quality and excessive daytime sleepiness. After rifaximin, HE scores decreased to 1.7±0.6 (P<0.001). REM sleep increased to 8.5% TST (0-25) (P=0.003). No changes were observed for TST, number of steps, and on questionnaires. CONCLUSION: Patients with recurrent HE suffer from poor sleep quality and excessive daytime sleepiness. On 24-h PSG, rifaximin improves objective sleep architecture parameters with no changes in the subjective quality of sleep and sleepiness.


Subject(s)
Hepatic Encephalopathy/drug therapy , Liver Cirrhosis/complications , Rifamycins/therapeutic use , Sleep Aids, Pharmaceutical/therapeutic use , Sleep Wake Disorders/drug therapy , Sleep, REM/drug effects , Actigraphy , Activity Cycles/drug effects , Affect/drug effects , Aged , Belgium , Female , Hepatic Encephalopathy/diagnosis , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/physiopathology , Humans , Liver Cirrhosis/diagnosis , Male , Middle Aged , Polysomnography , Prospective Studies , Quality of Life , Recurrence , Rifamycins/adverse effects , Rifaximin , Sleep Aids, Pharmaceutical/adverse effects , Sleep Wake Disorders/diagnosis , Sleep Wake Disorders/etiology , Sleep Wake Disorders/physiopathology , Surveys and Questionnaires , Time Factors , Treatment Outcome
6.
Respir Res ; 17(1): 148, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27842553

ABSTRACT

BACKGROUND: Modafinil is a wake-promoting drug and has been widely used for daytime sleepiness in patients with narcolepsy and other sleep disorders. A recent case series reported that daily oral modafinil alleviated hypercapnic respiratory failure in patients with COPD. However, the precise action of modafinil on respiration such as hypercapnic and/or hypoxic ventilatory responses remains unclear. The aim of this study is to clarify the effect of modafinil on the ventilatory control. METHODS: We investigated the hypothesis that modafinil enhances resting ventilation as well as the stimulatory ventilatory responses to hypercapnia and hypoxia. We addressed the issue by examining minute ventilation, respiratory rate and volume components using plethysmography, combined with a concurrent EEG monitoring of the level of wakefulness before and after administration of modafinil in two doses of 100 mg/kg and 200 mg/kg in unanesthetized mice. In addition, we monitored the effect of the lower dose of modafinil on mice locomotor activity in a freely moving condition by video-recording. RESULTS: Wakefulness, locomotor activity and variability of the breathing pattern in tidal volume were promoted by both doses of modafinil. Neither dose of modafinil increased the absolute values of resting ventilation or promoted the ventilatory responses to hypercapnia and hypoxia. Rather, higher dose of modafinil slightly suppressed respiratory rate in room air condition. CONCLUSIONS: Modafinil is conducive to the state of wakefulness but does not augment resting ventilation or the hyperventilatory responses to chemical stimuli in unanesthetized rodents.


Subject(s)
Activity Cycles/drug effects , Benzhydryl Compounds/pharmacology , Lung/drug effects , Pulmonary Ventilation/drug effects , Respiration/drug effects , Wakefulness-Promoting Agents/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Hypercapnia/drug therapy , Hypercapnia/physiopathology , Hypoxia/drug therapy , Hypoxia/physiopathology , Locomotion/drug effects , Lung/physiopathology , Male , Mice, Inbred C57BL , Modafinil , Time Factors , Video Recording
7.
Chronobiol Int ; 32(9): 1311-4, 2015.
Article in English | MEDLINE | ID: mdl-26376345

ABSTRACT

A circadian rhythm is a cycle of approximately 24 h, responsible for many physiological adjustments, and ageing of the circadian clock contributes to cognitive decline. Rhythmicity is severely impaired in Alzheimer disease (AD) and few therapeutic attempts succeeded in improving sleep disorders in such context. This study evaluated sleep parameters by actigraphy in 30 AD patients before and after trazodone use for 2 weeks, and we show a significant improvement in relative rhythm amplitude (RRA), compatible with a more stable daytime behavioral pattern. So, trazodone appears to produce a stabilization of the circadian rhythms in individuals with AD.


Subject(s)
Activity Cycles/drug effects , Alzheimer Disease/drug therapy , Sleep Disorders, Circadian Rhythm/drug therapy , Sleep/drug effects , Trazodone/therapeutic use , Actigraphy , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/physiopathology , Female , Humans , Male , Sleep Disorders, Circadian Rhythm/diagnosis , Sleep Disorders, Circadian Rhythm/physiopathology , Time Factors , Trazodone/adverse effects , Treatment Outcome
8.
Drugs Today (Barc) ; 51(1): 29-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25685859

ABSTRACT

Tasimelteon (Hetlioz®), a melatonin receptor agonist, is the first, and, at the time of the publication, the only drug to be approved by the U.S. Food and Drug Administration (FDA) for the treatment of non-24-hour sleep-wake disorder (non-24). This circadian rhythm disorder occurs most commonly in blind individuals without light perception, and it results from their inability to entrain to the 24-hour photoperiod, although the indication does not specify a particular patient population. Non-24 is characterized by a persistent cycle of nighttime insomnia and daytime sleepiness, alternating with asymptomatic periods depending on an individual's degree of circadian rhythm synchronization with the photoperiod at any particular time. Phase II clinical trials in healthy individuals confirmed the circadian phase-shifting potential of tasimelteon. Phase III trials in totally blind subjects diagnosed with non-24 demonstrated the efficacy of tasimelteon in reducing both nighttime wakefulness and daytime napping. Physiologic monitoring revealed that tasimelteon resulted in a higher proportion of individuals becoming entrained to the 24-hour cycle compared with placebo. Safety assessments indicated that tasimelteon is well tolerated, with the most common adverse events being headache, alanine aminotransferase elevation, nightmares or unusual dreams, and upper respiratory or urinary tract infections. Tasimelteon is available as a capsule in a single 20-mg dose and it must be obtained through Vanda Pharmaceutical's HetliozSolutions program with dispensing through a specialty pharmacy. Safety studies in blind individuals diagnosed with non-24 are ongoing and a future clinical trial with Smith-Magenis syndrome patients is planned.


Subject(s)
Activity Cycles/drug effects , Benzofurans/therapeutic use , Blindness/complications , Cyclopropanes/therapeutic use , Receptors, Melatonin/agonists , Sleep Disorders, Circadian Rhythm/drug therapy , Animals , Benzofurans/adverse effects , Benzofurans/pharmacokinetics , Cyclopropanes/adverse effects , Cyclopropanes/pharmacokinetics , Drug Interactions , Humans , Receptors, Melatonin/metabolism , Signal Transduction/drug effects , Sleep/drug effects , Sleep Disorders, Circadian Rhythm/etiology , Sleep Disorders, Circadian Rhythm/metabolism , Sleep Disorders, Circadian Rhythm/physiopathology , Time Factors , Treatment Outcome , Wakefulness/drug effects
9.
Elife ; 32014 Dec 29.
Article in English | MEDLINE | ID: mdl-25546305

ABSTRACT

Ultradian (~4 hr) rhythms in locomotor activity that do not depend on the master circadian pacemaker in the suprachiasmatic nucleus have been observed across mammalian species, however, the underlying mechanisms driving these rhythms are unknown. We show that disruption of the dopamine transporter gene lengthens the period of ultradian locomotor rhythms in mice. Period lengthening also results from chemogenetic activation of midbrain dopamine neurons and psychostimulant treatment, while the antipsychotic haloperidol has the opposite effect. We further reveal that striatal dopamine levels fluctuate in synchrony with ultradian activity cycles and that dopaminergic tone strongly predicts ultradian period. Our data indicate that an arousal regulating, dopaminergic ultradian oscillator (DUO) operates in the mammalian brain, which normally cycles in harmony with the circadian clock, but can desynchronize when dopamine tone is elevated, thereby producing aberrant patterns of arousal which are strikingly similar to perturbed sleep-wake cycles comorbid with psychopathology.


Subject(s)
Activity Cycles/physiology , Arousal/physiology , Behavior, Animal , Dopamine/physiology , Activity Cycles/drug effects , Animals , Haloperidol/pharmacology , Locomotion , Methamphetamine/pharmacology , Mice , Mice, Knockout , Suprachiasmatic Nucleus/drug effects , Suprachiasmatic Nucleus/physiology
10.
Alcohol Clin Exp Res ; 38(3): 760-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24512529

ABSTRACT

BACKGROUND: Ethanol (EtOH) triggers cellular adaptations that induce tolerance in many brain areas, including the suprachiasmatic nucleus (SCN), the site of the master circadian clock. EtOH inhibits light-induced phase shifts in the SCN in vivo and glutamate-induced phase shifts in vitro. The in vitro phase shifts develop acute tolerance to EtOH, occurring within minutes of initial exposure, while the in vivo phase shifts exhibit no evidence of chronic tolerance. An intermediate form, rapid tolerance, is not well studied but may predict subsequent chronic tolerance. Here, we investigated rapid tolerance in the SCN clock. METHODS: Adult C57BL/6 mice were provided 15% EtOH or water for one 12-hour lights-off period. For in vitro experiments, SCN-containing brain slices were prepared in the morning and treated for 10 minutes with glutamate +/- EtOH the following night. Single-cell neuronal firing rates were recorded extracellularly during the subsequent day to determine SCN clock phase. For in vivo experiments, mice receiving EtOH 24 hours previously were exposed to a 30-minute light pulse immediately preceded by intraperitoneal saline or 2 g/kg EtOH injection. Mice were then placed in constant darkness and their phase-shifting responses measured. RESULTS: In vitro, the SCN clock from EtOH-exposed mice exhibited rapid tolerance, with a 10-fold increase in EtOH needed to inhibit glutamate-induced phase shifts. Co-application of brain-derived neurotrophic factor prevented EtOH inhibition, consistent with experiments using EtOH-naïve mice. Rapid tolerance lasts 48 to 96 hours, depending on whether assessing in vitro phase advances or phase delays. Similarly, in vivo, prior EtOH consumption prevented EtOH's acute blockade of photic phase delays. Finally, immunoblot experiments showed no changes in SCN glutamate receptor subunit (NR2B) expression or phosphorylation in response to rapid tolerance induction. CONCLUSIONS: The SCN circadian clock develops rapid tolerance to EtOH as assessed both in vivo and in vitro, and the tolerance lasts for several days. These data demonstrate the utility of the circadian system as a model for investigating cellular mechanisms through which EtOH acts in the brain.


Subject(s)
Central Nervous System Depressants/pharmacology , Circadian Clocks/drug effects , Drug Tolerance , Ethanol/pharmacology , Suprachiasmatic Nucleus/drug effects , Activity Cycles/drug effects , Animals , Brain-Derived Neurotrophic Factor , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Receptors, N-Methyl-D-Aspartate/metabolism
11.
Chronobiol Int ; 31(2): 283-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24128195

ABSTRACT

In a large, prospective, 8-week open study of 721 outpatients receiving agomelatine treatment for a current major depressive episode (MDE), morningness-eveningness (Composite Scale of Morningness) was assessed before and after treatment to investigate possible changes in morningness-eveningness after treatment and evaluate whether morningness-eveningness at baseline predicted treatment response. A change towards morningness was observed after treatment. This change was greater in responders than non-responders. Moreover, being a morning type at baseline was an independent predictor of response to treatment. Once thought to be a trait variable, morningness-eveningness is a potential treatment target that should be systematically assessed in MDE patients.


Subject(s)
Acetamides/therapeutic use , Antidepressive Agents/therapeutic use , Circadian Rhythm/drug effects , Depressive Disorder, Major/drug therapy , Activity Cycles/drug effects , Adult , Depressive Disorder, Major/diagnosis , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/psychology , Female , France , Humans , Male , Middle Aged , Prospective Studies , Social Behavior , Time Factors , Treatment Outcome
12.
J Neurosci ; 33(31): 12825-34, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23904617

ABSTRACT

Notch is a cell surface receptor that is known to regulate developmental processes by establishing physical contact between neighboring cells. Many recent studies show that it also plays an important role in the formation of long-term memory (LTM) in adults, implying that memory formation requires regulation at the level of cell-cell contacts among brain cells. Neither the target of Notch activity in LTM formation nor the underlying mechanism of regulation is known. We report here results of our studies in adult Drosophila melanogaster showing that Notch regulates dCrebB-17A, the CREB protein. CREB is a transcriptional factor that is pivotal for intrinsic and synaptic plasticity involved in LTM formation. Notch in conjunction with PKC activity upregulates the level of a hyperphosphorylated form of CREB (hyper-PO4 CREB) and triggers its ultradian oscillation, both of which are linked to LTM formation. One of the sites that is phosphorylated in hyper-PO4 CREB is serine 231, which is the functional equivalent of mammalian CREB serine 133, the phosphorylation of which is an important regulator of CREB functions. Our data suggest the model that Notch and PKC activities generate a cyclical accumulation of cytoplasmic hyper-PO4 CREB that is a precursor for generating the nuclear CREB isoforms. Cyclical accumulation of CREB might be important for repetitive aspects of LTM formation, such as memory consolidation. Because Notch, PKC, and CREB have been implicated in many neurodegenerative diseases (e.g., Alzheimer's disease), our data might also shed some light on memory loss and dementia.


Subject(s)
Activity Cycles/physiology , Brain/metabolism , Conditioning, Classical/physiology , Drosophila Proteins/metabolism , Memory, Long-Term/physiology , Receptors, Notch/metabolism , Activity Cycles/drug effects , Activity Cycles/genetics , Animals , Animals, Genetically Modified , Brain/cytology , CREB-Binding Protein/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Male , Mutation/genetics , Phorbol Esters/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Receptors, Notch/genetics , Temperature , Time Factors , Up-Regulation/drug effects , Up-Regulation/genetics
13.
Brain Res ; 1521: 51-8, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23701725

ABSTRACT

Studies of rats and voles suggest that distinct pathways emanating from the anterior hypothalamic-retrochiasmatic area and the mediobasal hypothalamic arcuate nucleus independently generate ultradian rhythms (URs) in hormone secretion and behavior. We evaluated the hypothesis that destruction of arcuate nucleus (ARC) neurons, in concert with dampening of suprachiasmatic nucleus (SCN) circadian rhythmicity, would compromize the generation of ultradian rhythms (URs) of locomotor activity. Siberian hamsters retain-->of both sexes treated neonatally with monosodium glutamate (MSG) that destroys ARC neurons were subjected in adulthood to a circadian disrupting phase-shift protocol (DPS) that produces SCN arrhythmia. MSG treatments induced hypogonadism and obesity, retain-->and markedly reduced the size of the optic chiasm and optic nerves. MSG-treated hamsters exhibited normal entrainment to the light-dark cycle, but MSG treatretain-->ment counteracted the circadian arrhythmicity induced by the DPS protocol: only 6% of retain-->MSG-treated hamsters exhibited circadian arrhythmia, whereas 50% of control hamsters were circadian disrupted. In MSG-treated hamsters that retained circadian rhythmicity after DPS treatment, quantitative parameters of URs appeared normal, but in the two MSG-treated hamsters that became circadian arrhythmic after DPS, both dark-phase and light-phase URs were abolished. Although preliminary, these data are consistent with reports in voles suggesting that the combined disruption of SCN and ARC function impairs the expression of behavioral URs. The data also suggest that light thresholds for entrainment of circadian rhythms may be lower than those required to disrupt circadian organization.


Subject(s)
Activity Cycles/physiology , Arcuate Nucleus of Hypothalamus/physiology , Sodium Glutamate/toxicity , Suprachiasmatic Nucleus/physiology , Activity Cycles/drug effects , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/drug effects , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Cricetinae , Female , Male , Motor Activity/drug effects , Motor Activity/physiology , Phodopus , Photoperiod , Suprachiasmatic Nucleus/drug effects
14.
J Neurosci ; 32(42): 14794-803, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-23077063

ABSTRACT

In mammals, the suprachiasmatic nucleus (SCN) of the hypothalamus generates a 24 h rhythm of sleep and arousal. While neuronal spiking activity in the SCN provides a functional circadian oscillator that propagates throughout the brain, the ultradian sleep-wake state is regulated by the basal forebrain/preoptic area (BF/POA). How this SCN circadian oscillation is integrated into the shorter sleep-wake cycles remains unclear. We examined the temporal patterns of neuronal activity in these key brain regions in freely behaving rats. Neuronal activity in various brain regions presented diurnal rhythmicity and/or sleep-wake state dependence. We identified a diurnal rhythm in the BF/POA that was selectively degraded when diurnal arousal patterns were disrupted by acute brain serotonin depletion despite robust circadian spiking activity in the SCN. Local blockade of serotonergic transmission in the BF/POA was sufficient to disrupt the diurnal sleep-wake rhythm of mice. These results suggest that the serotonergic system enables the BF/POA to couple the SCN circadian signal to ultradian sleep-wake cycles, thereby providing a potential link between circadian rhythms and psychiatric disorders.


Subject(s)
Activity Cycles/physiology , Circadian Clocks/physiology , Serotonin/physiology , Sleep Stages/physiology , Suprachiasmatic Nucleus/physiology , Wakefulness/physiology , Activity Cycles/drug effects , Animals , Circadian Clocks/drug effects , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Male , Mice , Mice, Inbred C57BL , Preoptic Area/drug effects , Preoptic Area/physiology , Prosencephalon/drug effects , Prosencephalon/physiology , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology , Sleep Stages/drug effects , Suprachiasmatic Nucleus/drug effects , Wakefulness/drug effects
15.
Proc Natl Acad Sci U S A ; 109(18): 7061-6, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22509009

ABSTRACT

Following general anesthesia, people are often confused about the time of day and experience sleep disruption and fatigue. It has been hypothesized that these symptoms may be caused by general anesthesia affecting the circadian clock. The circadian clock is fundamental to our well-being because it regulates almost all aspects of our daily biochemistry, physiology, and behavior. Here, we investigated the effects of the most common general anesthetic, isoflurane, on time perception and the circadian clock using the honeybee (Apis mellifera) as a model. A 6-h daytime anesthetic systematically altered the time-compensated sun compass orientation of the bees, with a mean anticlockwise shift in vanishing bearing of 87° in the Southern Hemisphere and a clockwise shift in flight direction of 58° in the Northern Hemisphere. Using the same 6-h anesthetic treatment, time-trained bees showed a delay in the start of foraging of 3.3 h, and whole-hive locomotor-activity rhythms were delayed by an average of 4.3 h. We show that these effects are all attributable to a phase delay in the core molecular clockwork. mRNA oscillations of the central clock genes cryptochrome-m and period were delayed by 4.9 and 4.3 h, respectively. However, this effect is dependent on the time of day of administration, as is common for clock effects, and nighttime anesthesia did not shift the clock. Taken together, our results suggest that general anesthesia during the day causes a persistent and marked shift of the clock effectively inducing "jet lag" and causing impaired time perception. Managing this effect in humans is likely to help expedite postoperative recovery.


Subject(s)
Anesthesia, General/adverse effects , Bees/physiology , Circadian Clocks/physiology , Time Perception/physiology , Activity Cycles/drug effects , Activity Cycles/physiology , Anesthetics, General/adverse effects , Animals , Base Sequence , Bees/drug effects , Bees/genetics , Circadian Clocks/drug effects , Circadian Clocks/genetics , Feeding Behavior/drug effects , Feeding Behavior/physiology , Flight, Animal/drug effects , Flight, Animal/physiology , Genes, Insect , Humans , Isoflurane/adverse effects , Models, Animal , Photoperiod , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Perception/drug effects
16.
Mol Endocrinol ; 25(6): 944-54, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21511880

ABSTRACT

Glucocorticoid (GC) hormones are secreted from the adrenal gland in a characteristic pulsatile pattern. This ultradian secretory activity exhibits remarkable plasticity, with distinct changes in response to both physiological and stressful stimuli in humans and experimental animals. It is therefore important to understand how the pattern of GC exposure regulates intracellular signaling through the GC receptor (GR). We have previously shown that each pulse of ligand initiates rapid, transient GR activation in several physiologically relevant and functionally diverse target cell types. Using chromatin immunoprecipitation assays, we detect cyclical shifts in the net equilibrium position of GR association with regulatory elements of GC-target genes and have investigated in detail the mechanism of pulsatile transcriptional regulation of the GC-induced Period 1 gene. Transient recruitment of the histone acetyl transferase complex cAMP response element-binding protein (CREB) binding protein (CBP)/p300 is found to precisely track the ultradian hormone rhythm, resulting in transient localized net changes in lysine acetylation at GC-regulatory regions after each pulse. Pulsatile changes in histone H4 acetylation and concomitant recruitment of RNA polymerase 2 precede ultradian bursts of Period 1 gene transcription. Finally, we report the crucial underlying role of the intranuclear heat shock protein 90 molecular chaperone complex in pulsatile GR regulation. Pharmacological interference of heat shock protein 90 (HSP90) with geldanamycin during the intranuclear chaperone cycle completely ablated GR's cyclical activity, cyclical cAMP response element-binding protein (CREB) binding protein (CBP)/p300 recruitment, and the associated cyclical acetylation at the promoter region. These data imply a key role for an intact nuclear chaperone cycle in cyclical transcriptional responses, regulated in time by the pattern of pulsatile hormone.


Subject(s)
Activity Cycles/drug effects , CREB-Binding Protein/genetics , Corticosterone/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Hydrocortisone/pharmacology , Receptors, Glucocorticoid/genetics , p300-CBP Transcription Factors/genetics , Acetylation , Animals , Benzoquinones/pharmacology , CREB-Binding Protein/metabolism , Cell Line , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Histones/metabolism , Humans , Lactams, Macrocyclic/pharmacology , Leupeptins/pharmacology , Ligands , Mice , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Protein Transport/drug effects , RNA Polymerase II/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Glucocorticoid/metabolism , Regulatory Elements, Transcriptional , Transcription, Genetic , p300-CBP Transcription Factors/metabolism
17.
Endocrinology ; 151(11): 5369-79, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20861228

ABSTRACT

Glucocorticoid hormones are released in rapid hourly hormone bursts by the adrenal gland. These ultradian oscillations are fundamental to hypothalamic-pituitary-adrenal activity and transcriptional regulation of glucocorticoid responsive genes. The physiological relevance of glucocorticoid pulsatility is however unknown. Using a novel automated infusion system, we artificially created different patterns (modulating pulse amplitude) of corticosterone (cort). Identical amounts of cort either in constant or in hourly pulses were infused into adrenalectomized rats. At the end of the infusion period, either during rising or falling concentrations of a cort pulse, animals were exposed to 99 dB noise stress (10 min). Pulsatile cort infusion led to a differential stress response, dependent on the phase of the pulse during which the stress was applied. Although constant administration of cort resulted in a blunted ACTH response to the stressor, a brisker response occurred during the rising phase of plasma cort than during the falling phase. This phase-dependent effect was also seen in the behavioral response to the stressor, which was again greater during the rising phase of each cort pulse. Within the brain itself, we found differential C-fos activation responses to noise stress in the pituitary, paraventricular nucleus, amygdala, and hippocampus. This effect was both glucocorticoid pulse amplitude and phase dependent, suggesting that different stress circuits are differentially responsive to the pattern of glucocorticoid exposure. Our data suggest that the oscillatory changes in plasma glucocorticoid levels are critical for the maintenance of normal physiological reactivity to a stressor and in addition modulate emotionality and exploratory behavior.


Subject(s)
Activity Cycles/physiology , Brain/physiology , Corticosterone/metabolism , Neurons/physiology , Stress, Physiological/physiology , Activity Cycles/drug effects , Adrenalectomy , Adrenocorticotropic Hormone/metabolism , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Corticosterone/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/physiology , Immunoradiometric Assay , In Situ Hybridization , Male , Neurons/drug effects , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/physiology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Stress, Physiological/drug effects
18.
Curr Med Res Opin ; 24(12): 3357-67, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19032118

ABSTRACT

UNLABELLED: OBJECTIVE AND SCOPE: This review article used data from an extensive literature search (including MEDLINE database searches) to explore the relationships between sleep, memory and Alzheimer's disease (AD). The importance of taking into account circadian rhythmicity and acetylcholine (ACh) levels when considering acetylcholinesterase inhibitors, galantamine in particular, in the treatment of patients with AD is discussed. REVIEW FINDINGS: Moderate changes of circadian rhythms may occur as part of the normal ageing process, but patients with AD exhibit circadian rhythm disturbances extending beyond those observed in non-demented elderly and this may lead to severe disruption of the sleep-wake cycle. Indeed, ACh plays an active role in maintaining a normal sleep pattern, which is important for memory consolidation. Low levels of ACh during slow-wave sleep compared with wakefulness have been shown to be critical for the consolidation of declarative memory. This suggests the existence of a circadian rhythm in central cholinergic transmission which modulates memory processes, with high ACh levels during wakefulness and reduced levels during slow-wave sleep. When using cholinesterase inhibitors to stimulate central cholinergic transmission in AD, respecting the natural circadian fluctuations of central cholinergic transmission may therefore be an important factor for patient improvement. Interfering with nocturnal cholinergic activity can add to memory problems and induce sleep disorders. Available data suggest that the type of cholinesterase inhibitor used and the time of administration may be critical with regard to the possible development of such disturbances. Plasma levels of galantamine, for example, are high during the waking day and lower at night, supporting a cholinergic stimulation that mirrors the physiological circadian rhythm of cholinergic activity. This may have beneficial implications with regard to sleep and memory. CONCLUSIONS: The pharmacokinetic properties of cholinesterase inhibitors may need to be taken into account to avoid interference with sleep architecture and to achieve optimum benefits from treatment on cognitive processes.


Subject(s)
Activity Cycles/drug effects , Alzheimer Disease/drug therapy , Cholinesterase Inhibitors/therapeutic use , Galantamine/therapeutic use , Memory/drug effects , Sleep Stages/drug effects , Acetylcholine/metabolism , Aging/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Cholinesterase Inhibitors/adverse effects , Female , Galantamine/adverse effects , Humans , Male
19.
Chronobiol Int ; 24(5): 921-37, 2007.
Article in English | MEDLINE | ID: mdl-17994346

ABSTRACT

The combination of total sleep deprivation (TSD) and light therapy (LT) in bipolar depression causes rapid antidepressant effects, and its mechanism of action has been hypothesized to involve the enhancement of all of the monoaminergic systems targeted by antidepressant drugs (serotonin, dopamine, norepinephrine). It is still unknown if the clinical effects are paralleled by changes in biological rhythms. In a before/after design of a study of biological correlates of response, 39 inpatients affected by Type I Bipolar Disorder whose current depressive episode was without psychotic features were treated for one week with repeated TSD combined with morning LT. Wrist actigraphy was recorded throughout the study. Two-thirds of the patients responded to treatment (50% reduction in Hamilton Depression score). Responders showed an increase in daytime activity, phase-advance of the activity-rest rhythm of 57 min compared to the pre-treatment baseline, and reduced nighttime sleep. Non-responders did not show significant changes in the parameters of their activity-rest rhythm. Phase advance of the activity-rest rhythm is an actimetric correlate of the antidepressant response to TSD and LT in bipolar depression. Results are consistent with the known effects of sleep-wake manipulations and neurotransmitter function on the suprachiasmatic nucleus.


Subject(s)
Antidepressive Agents/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/therapy , Chronotherapy , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Phototherapy , Sleep Deprivation/physiopathology , Activity Cycles/drug effects , Activity Cycles/physiology , Adult , Bipolar Disorder/physiopathology , Female , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...