Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Clin Exp Immunol ; 192(3): 284-291, 2018 06.
Article in English | MEDLINE | ID: mdl-29878323

ABSTRACT

This is the second report of the United Kingdom Primary Immunodeficiency (UKPID) registry. The registry will be a decade old in 2018 and, as of August 2017, had recruited 4758 patients encompassing 97% of immunology centres within the United Kingdom. This represents a doubling of recruitment into the registry since we reported on 2229 patients included in our first report of 2013. Minimum PID prevalence in the United Kingdom is currently 5·90/100 000 and an average incidence of PID between 1980 and 2000 of 7·6 cases per 100 000 UK live births. Data are presented on the frequency of diseases recorded, disease prevalence, diagnostic delay and treatment modality, including haematopoietic stem cell transplantation (HSCT) and gene therapy. The registry provides valuable information to clinicians, researchers, service commissioners and industry alike on PID within the United Kingdom, which may not otherwise be available without the existence of a well-established registry.


Subject(s)
Epidemiological Monitoring , Immunologic Deficiency Syndromes/epidemiology , Registries/statistics & numerical data , Female , Humans , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Male , United Kingdom/epidemiology
2.
Neuropharmacology ; 62(3): 1359-70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21521646

ABSTRACT

Neuroanatomical, electrophysiological and behavioural abnormalities following timed prenatal methylazoxymethanol acetate (MAM) treatment in rats model changes observed in schizophrenia. In particular, MAM treatment on gestational day 17 (E17) preferentially disrupts limbic-cortical circuits, and is a promising animal model of schizophrenia. The hypersensitivity of this model to the NMDA receptor antagonist-induced hyperactivity has been proposed to mimic the increase in sensitivity observed in schizophrenia patients following PCP and Ketamine administration. However, how this increase in sensitivity in both patients and animals translates to differences in EEG oscillatory activity is unknown. In this study we have shown that MAM-E17 treated animals have an increased response to the hyperlocomotor and wake promoting effects of Ketamine, PCP, and MK801 but not to the competitive antagonist SDZ 220,581. These behavioural changes were accompanied by altered EEG responses to the NMDAR antagonists, most evident in the gamma and high frequency (HFO) ranges; altered sensitivity of these neuronal network oscillations in MAM-exposed rats is regionally selective, and reflects altered interneuronal function in this neurodevelopmental model.


Subject(s)
Brain Waves/drug effects , Cerebral Cortex/embryology , Disease Models, Animal , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Schizophrenia/physiopathology , Animals , Biphenyl Compounds/pharmacology , Cerebral Cortex/drug effects , Cross-Over Studies , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Methylazoxymethanol Acetate/toxicity , Pregnancy , Propionates/pharmacology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/physiology , Schizophrenia/chemically induced
4.
Neuroscience ; 137(2): 593-605, 2006.
Article in English | MEDLINE | ID: mdl-16257491

ABSTRACT

Previous studies have demonstrated that macromolecular synthesis in the brain is modulated in association with the occurrence of sleep and wakefulness. Similarly, the spectral composition of electroencephalographic activity that occurs during sleep is dependent on the duration of prior wakefulness. Since this homeostatic relationship between wake and sleep is highly conserved across mammalian species, genes that are truly involved in the electroencephalographic response to sleep deprivation might be expected to be conserved across mammalian species. Therefore, in the rat cerebral cortex, we have studied the effects of sleep deprivation on the expression of immediate early gene and heat shock protein mRNAs previously shown to be upregulated in the mouse brain in sleep deprivation and in recovery sleep after sleep deprivation. We find that the molecular response to sleep deprivation and recovery sleep in the brain is highly conserved between these two mammalian species, at least in terms of expression of immediate early gene and heat shock protein family members. Using Affymetrix Neurobiology U34 GeneChips , we also screened the rat cerebral cortex, basal forebrain, and hypothalamus for other genes whose expression may be modulated by sleep deprivation or recovery sleep. We find that the response of the basal forebrain to sleep deprivation is more similar to that of the cerebral cortex than to the hypothalamus. Together, these results suggest that sleep-dependent changes in gene expression in the cerebral cortex are similar across rodent species and therefore may underlie sleep history-dependent changes in sleep electroencephalographic activity.


Subject(s)
Brain/metabolism , Gene Expression Regulation/physiology , Genes, Immediate-Early/genetics , Heat-Shock Proteins/genetics , Sleep Deprivation/genetics , Sleep/physiology , Action Potentials/genetics , Animals , Basal Nucleus of Meynert/anatomy & histology , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/physiopathology , Brain/anatomy & histology , Cerebral Cortex/anatomy & histology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Electroencephalography , Gene Expression Profiling , Heat-Shock Proteins/biosynthesis , Hypothalamus/anatomy & histology , Hypothalamus/metabolism , Hypothalamus/physiopathology , Male , Mice , Oligonucleotide Array Sequence Analysis , Rats , Rats, Wistar , Recovery of Function/genetics , Sleep Deprivation/metabolism , Species Specificity
5.
Neuroscience ; 131(2): 375-85, 2005.
Article in English | MEDLINE | ID: mdl-15708480

ABSTRACT

The Tg2576 mouse model of Alzheimer's disease (AD) exhibits age-dependent amyloid beta (Abeta) deposition in the brain. We studied electroencephalographically defined sleep and the circadian regulation of waking activities in Tg2576 mice to determine whether these animals exhibit sleep abnormalities akin to those in AD. In Tg2576 mice at all ages studied, the circadian period of wheel running rhythms in constant darkness was significantly longer than that of wild type mice. In addition, the increase in electroencephalographic delta (1-4 Hz) power that occurs during non-rapid eye movement sleep after sleep deprivation was blunted in Tg2576 mice relative to controls at all ages studied. Electroencephalographic power during non-rapid eye movement sleep was shifted to higher frequencies in plaque-bearing mice relative to controls. The wake-promoting efficacy of the acetylcholinesterase inhibitor donepezil was lower in plaque-bearing Tg2576 mice than in controls. Sleep abnormalities in Tg2576 mice may be due in part to a cholinergic deficit in these mice. At 22 months of age, two additional deficits emerged in female Tg2576 mice: time of day-dependent modulation of sleep was blunted relative to controls and rapid eye movement sleep as a percentage of time was lower in Tg2576 than in wild type controls. The rapid eye movement sleep deficit in 22 month-old female Tg2576 mice was abolished by brief passive immunization with an N-terminal antibody to Abeta. The Tg2576 model provides a uniquely powerful tool for studies on the pathophysiology of and treatments for sleep deficits and associated cholinergic abnormalities in AD.


Subject(s)
Alzheimer Disease/genetics , Cholinergic Fibers/physiology , Chronobiology Disorders/genetics , Sleep/genetics , Synaptic Transmission/genetics , Alzheimer Disease/physiopathology , Animals , Chronobiology Disorders/physiopathology , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
6.
Neuroreport ; 14(2): 233-8, 2003 Feb 10.
Article in English | MEDLINE | ID: mdl-12598736

ABSTRACT

The monoamine neurotransmitter serotonin has long been implicated in development and maintenance of sleep patterns, yet the role of the serotonin transporter (SERT) in these processes has not been evaluated in detail. We report that genetically engineered SERT knockout mice exhibit more REM sleep (REMS) than wild type littermates (11 vs 7% of recording time under baseline conditions) and display more frequent REMS bouts that last longer. This phenotype resembles the previously reported long-term effect of repeated treatment with SERT inhibitor compounds rather than the acute REMS suppressing effect of treatment with such compounds, and is thus likely to reflect neuroadaptations to the absence of SERT, rather than an acute effect of its absence in the adult. While electroencephalographic (EEG) spectra did not differ between SERT knockout and wild type mice during non-REM sleep (NREMS) or REMS, the dynamics of the EEG during the transition from NREMS to REMS differed between the genotypes. The surge in EEG power in both the 6-9 Hz and 10-16 Hz ranges that occurs just prior to the onset of REMS (pre-REMS power surge) is of greater magnitude in SERT knockout mice than in wild type littermate controls. This observation contrasts with the reduced magnitude pre-REMS power surge observed in rats subjected to REMS deprivation relative to yoked controls. These results indicate that the pre-REMS power surge is influenced by REMS history and by monoaminergic transmission. Genetic differences in serotonin systems and developmental exposure to SERT blockers are likely to exert effects on REMS.


Subject(s)
Membrane Glycoproteins/deficiency , Membrane Transport Proteins , Nerve Tissue Proteins , Sleep, REM/genetics , Animals , Carrier Proteins/genetics , Electroencephalography/methods , Female , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Serotonin Plasma Membrane Transport Proteins , Sleep, REM/physiology
8.
J Neurosci ; 21(5): 1787-94, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222668

ABSTRACT

The role of dopamine in sleep regulation and in mediating the effects of wake-promoting therapeutics is controversial. In this study, polygraphic recordings and caudate microdialysate dopamine measurements in narcoleptic dogs revealed that the wake-promoting antinarcoleptic compounds modafinil and amphetamine increase extracellular dopamine in a hypocretin receptor 2-independent manner. In mice, deletion of the dopamine transporter (DAT) gene reduced non-rapid eye movement sleep time and increased wakefulness consolidation independently from locomotor effects. DAT knock-out mice were also unresponsive to the normally robust wake-promoting action of modafinil, methamphetamine, and the selective DAT blocker GBR12909 but were hypersensitive to the wake-promoting effects of caffeine. Thus, dopamine transporters play an important role in sleep regulation and are necessary for the specific wake-promoting action of amphetamines and modafinil.


Subject(s)
Central Nervous System Stimulants/pharmacology , Dopamine/metabolism , Membrane Glycoproteins , Membrane Transport Proteins , Narcolepsy/metabolism , Nerve Tissue Proteins , Wakefulness/drug effects , Wakefulness/physiology , Amphetamine/administration & dosage , Animals , Benzhydryl Compounds/administration & dosage , Caffeine/administration & dosage , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caudate Nucleus/metabolism , Disease Models, Animal , Dogs , Dopamine/analysis , Dopamine Plasma Membrane Transport Proteins , Dopamine Uptake Inhibitors/pharmacology , Dose-Response Relationship, Drug , Electroencephalography , Electromyography , Methamphetamine/administration & dosage , Mice , Mice, Knockout , Microdialysis , Modafinil , Motor Activity/drug effects , Narcolepsy/drug therapy , Orexin Receptors , Receptors, G-Protein-Coupled , Receptors, Neuropeptide/metabolism
9.
J Biol Rhythms ; 16(1): 66-75, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11220781

ABSTRACT

Entrainment of the circadian pacemaker to nonphotic stimuli, such as scheduled wheel-running activity, is well characterized in nocturnal rodents, but little is known about activity-dependent entrainment in diurnal or crepuscular species. In the present study, effects of scheduled voluntary wheel-running activity on circadian timekeeping were investigated in Octodon degus, a hystricomorph rodent that exhibits robust crepuscular patterns of wakefulness. When housed in constant darkness, O. degus exhibited circadian rhythms in wheel-running activity and body temperature (Tb) with an average period length (tau) of 23.39 +/- 0.11 h. When wheel running was restricted to a fixed 2-h schedule every 24 h, tau increased on average 0.39 +/- 0.09 h but did not result in steady-state entrainment. Instead, relative coordination between the fixed running schedule and circadian timing was observed. Tau was greatest when scheduled wheel running occurred at CT 20.5 (0.4 h greater than DD baseline tau). Scheduled running activity also influenced Tb waveform symmetry, reflecting concomitant changes in the circadian activity-rest ratio (alpha:rho). Aftereffects of the scheduled wheel-running paradigm were also observed. In 2 animals, tau lengthened from 23.20 and 23.80 h to 24.14 and 24.15 h, respectively, and remained relatively stable for approximately 1 month during the wheel schedule. Although behavioral activity appears to be a weak zeitgeber in this species, these data suggest that nonphotic stimuli can phase delay the circadian pacemaker in O. degus at similar times of the day as in nocturnal hamsters and mice, and in humans.


Subject(s)
Biological Clocks/physiology , Body Temperature/physiology , Circadian Rhythm/physiology , Motor Activity/physiology , Animals , Humans , Light , Male , Periodicity , Rodentia
10.
Sleep ; 23(7): 867-74, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11083595

ABSTRACT

The hypocretin/orexin ligand-receptor system has recently been implicated in the sleep disorder narcolepsy. During the dark (active) period, null mutants of the prepro-orexin (prepro-hypocretin) gene have cataplectic attacks and increased levels of both rapid eye movement (REM) and non-REM (NREM) sleep. Intracerebroventricular injection of one of the encoded neuropeptides, orexin-A, early in the light period increases wakefulness and reduces REM sleep in the rat, suggesting that this system may be involved in the normal regulation of sleep and wakefulness. To further test this hypothesis, we measured hypocretin (hcrt) mRNA levels by both Northern hybridization and Taqman analysis in mouse and rat hypothalamus after short-term (6 h) sleep deprivation (SD) and 2-4 hours after recovery from SD. Although our SD procedures effectively induced a sleep debt and increased c-fos mRNA expression in the cortex and hypothalamus as described by other investigators, we found that hcrt mRNA levels were not significantly changed in either species either after SD or after recovery from SD. If the hcrt system is involved in normal regulation of sleep and wakefulness, longer periods of SD may be necessary to affect hcrt mRNA levels or changes may occur at the protein rather than mRNA level. Alternatively, this system may also be involved in another function that counterbalances any SD-induced changes in hcrt mRNA levels.


Subject(s)
Hypothalamus/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Sleep Deprivation/metabolism , Sleep, REM/physiology , Animals , Blotting, Northern , Electrodes, Implanted , Electroencephalography , Electromyography , Gene Expression , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Orexins , Polymerase Chain Reaction/methods , RNA, Messenger/genetics , Rats , Wakefulness/physiology
11.
J Neurosci ; 20(11): 4300-10, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10818165

ABSTRACT

The daily timing of rapid eye movement (REM) sleep reflects an interaction between the circadian pacemaker located in the suprachiasmatic nucleus of the hypothalamus (SCN) and a homeostatic process that induces compensatory REM sleep in response to REM sleep loss. Whether the circadian variation in REM sleep propensity is caused by active promotion, inhibition, or passive gating of REM sleep homeostasis by the SCN is unknown. To investigate these possibilities, compensatory responses to 24 hr REM sleep deprivation (RSD) were compared between SCN-lesioned (SCNx) and sham-lesioned rats at different times of day in constant dark. The attempts to enter REM sleep (REM tendency) increased during RSD in all rats and were modulated by circadian phase in sham-lesioned, but not SCNx rats. REM sleep homeostasis interacted with circadian time, such that REM tendency doubled during the rest phase in sham-lesioned rats relative to SCNx rats (F((6,93)) = 17.9; p = 0.0001). However, REM tendency was indistinguishable between SCNx and sham-lesioned rats during the activity phase, suggesting the SCN does not inhibit REM tendency at this time. By contrast, the amount of compensatory REM sleep examined 2, 6, 12, or 24 hr after RSD did not depend on circadian phase. Thus, transitions into REM sleep are facilitated by the SCN during the rest phase, but the amount of REM sleep, once initiated, is determined primarily by homeostatic mechanisms. This work supports a role for the SCN in the active promotion of REM sleep at specific times of day.


Subject(s)
Circadian Rhythm/physiology , Homeostasis/physiology , Sleep, REM/physiology , Suprachiasmatic Nucleus/physiology , Animals , Arousal/physiology , Body Temperature/physiology , Electroencephalography , Electromyography , Male , Rats , Rats, Wistar , Sleep Deprivation/physiopathology
12.
Am J Physiol Regul Integr Comp Physiol ; 278(5): R1385-9, 2000 May.
Article in English | MEDLINE | ID: mdl-10801311

ABSTRACT

Light exposure during the early and late subjective night generally phase delays and advances circadian rhythms, respectively. However, this generality was recently questioned in a photic entrainment study in Octodon degus. Because degus can invert their activity phase preference from diurnal to nocturnal as a function of activity level, assessment of phase preference is critical for computations of phase reference [circadian time (CT) 0] toward the development of a photic phase response curve. After determining activity phase preference in a 24-h light-dark cycle (LD 12:12), degus were released in constant darkness. In this study, diurnal (n = 5) and nocturnal (n = 7) degus were randomly subjected to 1-h light pulses (30-35 lx) at many circadian phases (CT 1-6: n = 7; CT 7-12: n = 8; CT 13-18: n = 8; and CT 19-24: n = 7). The circadian phase of body temperature (Tb) onset was defined as CT 12 in nocturnal animals. In diurnal animals, CT 0 was determined as Tb onset + 1 h. Light phase delayed and advanced circadian rhythms when delivered during the early (CT 13-16) and late (CT 20-23) subjective night, respectively. No significant phase shifts were observed during the middle of the subjective day (CT 3-10). Thus, regardless of activity phase preference, photic entrainment of the circadian pacemaker in Octodon degus is similar to most other diurnal and nocturnal species, suggesting that entrainment mechanisms do not determine overt diurnal and nocturnal behavior.


Subject(s)
Activity Cycles/physiology , Light , Rodentia/physiology , Adaptation, Physiological , Animals , Body Temperature , Male , Motor Activity , Photoperiod
13.
Pharmacol Biochem Behav ; 65(1): 155-62, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10638649

ABSTRACT

The adenosine antagonist caffeine disrupts sleep, but whether caffeine promotes wakefulness by interfering with the expression of sleep or by attenuating sleepiness is unknown. The ability of caffeine to reduce sleep tendency in rats was directly tested by quantifying the number of stimuli needed to maintain wakefulness during sleep deprivation for 6 h after systemic caffeine treatment. In addition, the influence of caffeine on the dynamics between nonrapid-eye-movement (NREM) and rapid-eye-movement (REM) sleep was investigated by comparing the magnitude and time course of the compensatory sleep responses for 42 h postsleep deprivation. Caffeine significantly reduced the attempts to sleep during sleep deprivation, F(1,9) 8.83, p = 0.0157; 44.9% of vehicle), but did not change compensatory slow-wave activity during recovery sleep. During the initial recovery phase, caffeine suppressed compensatory REM sleep and reduced, but did not block, compensatory NREM sleep duration and continuity. By 42 h postsleep deprivation, the amount of NREM recovered (70.0% of deficit) did not differ from vehicle. In contrast, the REM sleep deficit recovered after caffeine (100%) was more than after vehicle (43.9%). Thus, caffeine slowed the rate of compensatory sleep after sleep deprivation, as indexed by the duration of sleep states and sleep continuity.


Subject(s)
Caffeine/pharmacology , Sleep/drug effects , Animals , Circadian Rhythm , Male , Rats , Rats, Wistar , Sleep Deprivation , Sleep, REM/drug effects , Time Factors
14.
Sleep Res Online ; 3(3): 113-9, 2000.
Article in English | MEDLINE | ID: mdl-11382909

ABSTRACT

The squirrel monkey (Saimiri sciureus) exhibits a robust daily rhythm of sleep-wakefulness that is under circadian control, but the nature of homeostatic sleep regulation in this diurnal primate is poorly understood. Since delta frequency (0.5-2.0 Hz) activity in the electroencephalogram (EEG) during non-Rapid Eye Movement (NREM) sleep is thought to reflect homeostatic factors contributing to sleep tendency, we measured EEG delta power density and slow wave incidence and amplitude during NREM sleep during spontaneous sleep, occurring when monkeys were housed undisturbed in a 24-hour light-dark (LD) cycle and in constant light (LL). In LD and LL conditions, monkeys exhibited circadian rhythms in delta power density, wave incidence and wave amplitude that peaked in the middle of the subjective night, several hours after consolidated sleep onset. These results differ from predictions of a purely homeostatic model of sleep that would include maximal levels of delta activity at sleep onset.


Subject(s)
Delta Rhythm , Sleep/physiology , Animals , Circadian Rhythm/physiology , Darkness , Female , Light , Male , Ovariectomy , Saimiri , Sleep Stages/physiology , Sleep, REM/physiology , Statistics as Topic , Time Factors
15.
J Pharmacol Exp Ther ; 291(3): 1317-23, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10565857

ABSTRACT

Novel neuroactive steroids were evaluated for their effects on operant responding, rotorod motor performance, and electroencephalogram recording in rats. Co 134444, Co 177843, and Co 127501 were compared with the prototypical gamma-aminobutyric acid(A)-positive allosteric modulators triazolam, zolpidem, pentobarbital, pregnanolone, and CCD 3693. Each of the compounds produced a dose-related decrease in response rates under a variable-interval 2-min schedule of positive reinforcement in an operant paradigm. In addition, all compounds produced a dose-related increase in ataxia and significant increases in nonrapid eye movement sleep in this experiment or have been previously reported to do so. Co 134444, Co 177843, and Co 127501 increased nonrapid eye movement sleep at doses that had no effect on rapid eye movement sleep. All of the compounds were more potent at decreasing operant responding than they were at increasing ataxia. Furthermore, the potency of compounds to produce response-rate suppression in an operant paradigm appeared to be a better predictor of soporific potency than did potency in the rotorod assay. The screening for sedative-hypnotic activity resulted in the identification of the novel orally active neuroactive steroids Co 134444, Co 177843, and Co 127501.


Subject(s)
Conditioning, Operant/drug effects , Hypnotics and Sedatives/pharmacology , Steroids/pharmacology , Animals , Depression, Chemical , Dose-Response Relationship, Drug , Electroencephalography/drug effects , Male , Pentobarbital/pharmacology , Postural Balance/drug effects , Pregnanolone/analogs & derivatives , Pregnanolone/pharmacology , Pregnenolone/analogs & derivatives , Pregnenolone/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Triazolam/pharmacology , Zolpidem
16.
Sleep ; 22(1): 45-59, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9989365

ABSTRACT

A series of sleep deprivation (SD) experiments were performed to examine the relative influence of circadian and homeostatic factors on the timing of sleep in squirrel monkeys free-running in constant illumination. All SDs started at the beginning of subjective night and lasted 0, 1/4, 1/2, 1, 1 1/4, or 1 1/2 circadian cycles. These six lengths represented three pairs: (0.1), (1/4, 1 1/4), (1/2, 1 1/2). Within each pair, SD ended at the same circadian phase but differed by one circadian cycle in duration. Both before and after SD, consolidated sleep (CS) episodes occurred predominantly during subjective night, even after long SDs ending at the beginning of subjective day. CS duration was strongly influenced by circadian phase but had no overall correlation with prior wake duration. Sleep loss incurred during SDs longer than 1/4 cycle was only partially recovered over the next two circadian cycles, though total sleep duration was closer to baseline levels after the second circadian cycle after SD. There was a trend toward a positive correlation between prior wake duration and the amount of NREM and delta activity measures during subjective day. Delta activity was not increased in the first 2 hours of CS after the SD. Relatively high levels of delta activity occurred immediately after the SD ended and again at the time of baseline CS onset. These data indicate that the amount of sleep and delta activity after SD in squirrel monkeys is weakly dependent on prior wake duration. Circadian factors appear to dominate homeostatic processes in determining the timing, duration and content of sleep in these diurnal primates.


Subject(s)
Circadian Rhythm/physiology , Homeostasis/physiology , Sleep Deprivation/physiology , Sleep, REM/physiology , Animals , Electroencephalography , Female , Male , Saimiri/physiology , Time Factors , Wakefulness
17.
J Neurosci ; 19(1): 328-33, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9870962

ABSTRACT

Mechanisms differentiating diurnal from nocturnal species are thought to be innate components of the circadian timekeeping system and may be located downstream from the circadian pacemaker within the suprachiasmatic nucleus (SCN) of the hypothalamus. In the present study, we found that the dominant phase of behavioral activity and body temperature (Tb) is susceptible to modification by a specific modality of behavioral activity (wheel-running activity) in Octodon degus, a mammal that exhibits multiple chronotypes. Seven Octodon degus exhibited diurnal Tb and locomotor activity (LMA) circadian rhythms while entrained to a 24 h light/dark cycle (LD 12:12). When the diurnal animals were provided unrestricted access to a running wheel, the overt daily rhythms in these animals inverted to nocturnal. This nocturnal pattern was sustained in constant darkness and returned to diurnal after removal of the running wheel. Six additional animals exhibited nocturnal chronotypes in LD 12:12 regardless of access to running wheels. Wheel-running activity inverted the phase preference in the diurnal animals without changing the 24 hr mean LMA or Tb levels. Because wheel running did not increase the amplitude of the pre-existing diurnal pattern, simple masking effects on LMA and Tb cannot explain the rhythm inversion. The diurnal-nocturnal inversion occurred without reversing crepuscular-timed episodes of activity, suggesting that diurnal or nocturnal phase preference is controlled separately from the intrinsic timing mechanisms within the SCN and can be dependent on behavioral or environmental factors.


Subject(s)
Choice Behavior/physiology , Circadian Rhythm/physiology , Rodentia/physiology , Running/physiology , Animals , Body Temperature Regulation/physiology , Photic Stimulation
18.
Sleep ; 22(8): 1045-53, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10617165

ABSTRACT

The circadian timing system in mammals is thought to promote wakefulness and oppose sleep drive that accumulates across the activity phase in diurnal and nocturnal species. Whether the circadian system actively opposes compensatory sleep responses in mammals with episodes of alertness consolidated at dawn and dusk is unknown. In the present study, an interaction between circadian timed arousal at dawn and compensatory sleep responses after sleep deprivation (SD) was examined in Octodon degus, a hystricomorph rodent with crepuscular episodes of wakefulness. Recovery sleep was compared after 6 hours and 12 hours of SD ending at either CT 21 or 12, just before the dawn, and just after the dusk crepuscular episodes of consolidated wakefulness, respectively. Total sleep time and NREM sleep after SD increased proportionally to the amount of sleep loss; however, compensatory sleep responses after SD were attenuated at CT 23, a circadian time when a crepuscular event of wakefulness occurs in this species. EEG slow-wave activity (SWA) and body temperature levels in the first two hours after 6 and 12 hours of SD ending at CT 12 were similar. However, both were significantly higher than after 12 hours of SD ending at CT 21, suggesting factors other than the amount of prior wake duration can influence SWA levels. This study provides evidence that the circadian arousal system opposes compensatory sleep responses at dawn by actively promoting wakefulness in this species.


Subject(s)
Circadian Rhythm/physiology , Rodentia/physiology , Sleep Deprivation , Sleep, REM/physiology , Wakefulness/physiology , Animals , Arousal/physiology , Body Temperature/physiology , Electroencephalography , Homeostasis/physiology
19.
Am J Physiol ; 275(2): R555-65, 1998 08.
Article in English | MEDLINE | ID: mdl-9688693

ABSTRACT

Individual effects of corticotropin-releasing hormone (CRH) and glucocorticoids on sleep have been difficult to discern due to the feedback effects each hormone exerts on the other. In addition, it is not known whether hypothalamic-pituitary-adrenal axis hormones alter sleep homeostasis or circadian influences on sleep propensity. We therefore analyzed sleep architecture and electroencephalographic (EEG) power in freely moving rats before and after removal of corticosterone (thus elevating endogenous CRH) by surgical adrenalectomy. Adrenalectomy reduced the amplitude of the diurnal rhythms of maximal and average sleep bout lengths (P < 0.004). After adrenalectomy, power from 1 to 4 Hz decreased (P < 0.042), whereas power from 9 to 12 Hz increased in the power spectra of the EEG recording (P = 0.001). Administration of physiological corticosterone replacement reversed some of these effects. Supraphysiological corticosterone replacement in adrenalectomized rats reduced the amount of non-rapid-eye-movement sleep in the 24-h cycle (P = 0.001). During each endocrine condition, rats were sleep deprived for 6 h. Endocrine status did not alter the subsequent homeostatic response to sleep deprivation. Thus ADX and supraphysiological corticosteroid replacement each altered sleep architecture without a demonstrable effect on sleep homeostasis.


Subject(s)
Adrenalectomy , Corticosterone/physiology , Electroencephalography , Sleep/physiology , Analysis of Variance , Animals , Body Temperature/drug effects , Body Temperature/physiology , Corticosterone/pharmacology , Darkness , Electroencephalography/drug effects , Electromyography , Light , Male , Motor Activity , Multivariate Analysis , Rats , Rats, Wistar , Sleep/drug effects , Sleep Deprivation/physiology , Sleep, REM/drug effects , Sleep, REM/physiology
20.
Psychoneuroendocrinology ; 23(2): 161-73, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9621396

ABSTRACT

Considerable data support a role for cholinergic influences on the circadian system. The extent to which these influences are mediated by nicotinic acetylcholine receptors (nAChRs) has been controversial, as have the specific actions of nicotine and acetylcholine in the suprachiasmatic nucleus (SCN) of the hypothalamus. In this article we review the existing literature and present new data supporting an important role for nAChRs in both the developing and adult SCN. Specifically, we present data showing that nicotine is capable of causing phase shifts in the circadian rhythms of rats. Like light and carbachol, nicotine appears to cause phase delays in the early subjective night and phase advances in the late subjective night. In the isolated SCN slice, however, only phase advances are seen, and, surprisingly, nicotine appears to cause the inhibition rather than the excitation of neurons. Among nAChR subunit mRNAs, alpha 7 appears to be the most abundant subunit in the adult SCN, whereas in the perinatal period, the more typical nAChRs with higher affinity for nicotine predominate in the SCN. This developmental change in subunit expression may explain the dramatic sensitivity of the perinatal SCN to nicotine that we have previously observed. The effects of nicotine on the SCN may contribute to alterations caused by nicotine in other physiological systems. These effects might also contribute to the dependence properties of nicotine through influences on arousal.


Subject(s)
Circadian Rhythm/physiology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/physiology , Animals , Circadian Rhythm/drug effects , Humans , Rats , Receptors, Nicotinic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...