Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters










Publication year range
1.
Acta Physiol (Oxf) ; 222(4): e13017, 2018 04.
Article in English | MEDLINE | ID: mdl-29253320

ABSTRACT

Knowledge regarding the cellular mechanisms of sleep regulation is accumulating rapidly. In addition to neurones, also non-neuronal brain cells (astrocytes and microglia) are emerging as potential players. New techniques, particularly optogenetics and designed receptors activated by artificial ligands (DREADD), have provided also sleep research with important additional tools to study the effect of either silencing or activating specific neuronal groups/neuronal networks by opening or shutting ion channels on cells. The advantages of these strategies are the possibility to genetically target specific cell populations and the possibility to either activate or inhibit them with inducing light signal into the brain. Studies probing circuits of NREM and REM sleep regulation, as well as their role in memory consolidation, have been conducted recently. In addition, fundamentally new thoughts and potential mechanisms have been introduced to the field. The role of non-neuronal tissues in the regulation of many brain functions has become evident. These non-neuronal cells, particularly astrocytes, integrate large number of neurones, and it has been suggested that one of their functions is to integrate the (neural) activity in larger brain areas-a feature that is one of the prominent features of also the state of sleep.


Subject(s)
Astrocytes/physiology , Brain/physiology , Microglia/physiology , Neurons/physiology , Sleep/physiology , Animals , Humans
2.
Arch Ital Biol ; 152(2-3): 57-65, 2014.
Article in English | MEDLINE | ID: mdl-25828678

ABSTRACT

The state of sleep consists of different phases that proceed in successive, tightly regulated order through the night forming a physiological program, which for each individual is different but stabile from one night to another. Failure to accomplish this program results in feeling of unrefreshing sleep and tiredness in the morning. The pro- gram core is constructed by genetic factors but regulated by circadian rhythm and duration and intensity of day time brain activity. Many environmental factors modulate sleep, including stress, health status and ingestion of vigilance-affecting nutrients or medicines (e.g. caffeine). Knowledge of the factors that regulate the spontaneous sleep-wake cycle and factors that can affect this regulation forms the basis for diagnosis and treatment of the many common disorders of sleep.


Subject(s)
Brain/physiology , Circadian Rhythm , Sleep/physiology , Animals , Brain/drug effects , Brain/metabolism , Homeostasis , Humans , Stress, Physiological
3.
Acta Physiol (Oxf) ; 208(4): 311-28, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23746394

ABSTRACT

The state of sleep consists of different phases that proceed in successive, tightly regulated order through the night forming a physiological program, which for each individual is different but stabile from one night to another. Failure to accomplish this program results in feeling of unrefreshing sleep and tiredness in the morning. The program core is constructed by genetic factors but regulated by circadian rhythm and duration and intensity of day time brain activity. Many environmental factors modulate sleep, including stress, health status and ingestion of vigilance-affecting nutrients or medicines (e.g. caffeine). Acute sleep loss results in compromised cognitive performance, memory deficits, depressive mood and involuntary sleep episodes during the day. Moreover, prolonged sleep curtailment has many adverse health effects, as evidenced by both epidemiological and experimental studies. These effects include increased risk for depression, type II diabetes, obesity and cardiovascular diseases. In addition to voluntary restriction of sleep, shift work, irregular working hours, jet lag and stress are important factors that induce curtailed or bad quality sleep and/or insomnia. This review covers the current theories on the function of normal sleep and describes current knowledge on the physiologic effects of sleep loss. It provides insights into the basic mechanisms of the regulation of wakefulness and sleep creating a theoretical background for understanding different disturbances of sleep.


Subject(s)
Sleep Wake Disorders/physiopathology , Sleep/physiology , Aging , Brain/physiology , Energy Metabolism/physiology , Humans , Sleep/immunology
4.
Transl Psychiatry ; 2: e97, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22832862

ABSTRACT

Epidemiological studies show association between sleep duration and lipid metabolism. In addition, inactivation of circadian genes induces insulin resistance and hyperlipidemia. We hypothesized that sleep length and lipid metabolism are partially controlled by the same genes. We studied the association of total sleep time (TST) with 60 genetic variants that had previously been associated with lipids. The analyses were performed in a Finnish population-based sample (N = 6334) and replicated in 2189 twins. Finally, RNA expression from mononuclear leucocytes was measured in 10 healthy volunteers before and after sleep restriction. The genetic analysis identified two variants near TRIB1 gene that independently contributed to both blood lipid levels and to TST (rs17321515, P = 8.92(*)10(-5), Bonferroni corrected P = 0.0053, ß = 0.081 h per allele; rs2954029, P = 0.00025, corrected P = 0.015, ß = 0.076; P<0.001 for both variants after adjusting for blood lipid levels or body mass index). The finding was replicated in the twin sample (rs17321515, P = 0.022, ß = 0.063; meta-analysis of both samples P = 8.1(*)10(-6), ß = 0.073). After the experimentally induced sleep restriction period TRIB1 expression increased 1.6-fold and decreased in recovery phase (P = 0.006). In addition, a negative correlation between TRIB1 expression and slow wave sleep was observed in recovery from sleep restriction. These results show that allelic variants of TRIB1 are independently involved in regulation of lipid metabolism and sleep. The findings give evidence for the pleiotropic nature of TRIB1 and may reflect the shared roots of sleep and metabolism. The shared genetic background may at least partially explain the mechanism behind the well-established connection between diseases with disrupted metabolism and sleep.


Subject(s)
Alleles , Genetic Variation/genetics , Intracellular Signaling Peptides and Proteins/genetics , Lipid Metabolism/genetics , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Sleep/genetics , Adult , Aged , Cholesterol, HDL/blood , Cholesterol, LDL , Cohort Studies , Disorders of Excessive Somnolence/blood , Disorders of Excessive Somnolence/genetics , Female , Finland , Gene Expression/genetics , Gene Frequency/genetics , Genetic Association Studies , Genotype , Homeostasis/genetics , Humans , Male , Middle Aged , Protein Serine-Threonine Kinases/genetics , Sleep Deprivation/blood , Sleep Deprivation/genetics , Triglycerides/blood , Twins/genetics
5.
Neuroscience ; 212: 149-58, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22516018

ABSTRACT

Neonatal treatment of rat pups with clomipramine (CLI) has been shown to cause long-lasting and persistent depression-related behaviors and changes in sleep architecture and in brain-derived neurotrophic factor (BDNF) signaling in adult animals, producing an animal model of depression. However, the molecular mechanisms which mediate these effects of early-life CLI treatment on adult animals remain largely unknown. In order to characterize these further, we investigated in neonatally CLI-treated rats the sleep architecture as well as the extracellular and cellular levels of sleep regulators (nitric oxide, adenosine) and BDNF, respectively, in the basal forebrain (BF), i.e. the brain area which is implicated in sleep and depression. We found that CLI-treated rats exhibited a disturbed sleep architecture (REM sleep fragmentation was increased and NREM periods preceding REM were shorter) and reduced levels of BDNF and adenosine in the BF, whereas the levels of nitric oxide were elevated. Next, we examined sleep deprivation (SD)-induced homeostatic responses on sleep regulation and brain BDNF levels in CLI-treated rats. Compared to control rats, 3h of SD induced a smaller increase in the amount of NREM sleep during sleep recovery. At the molecular level, the normal homeostatic response was dissociated: the rise in the adenosine level was not accompanied by a rise in the nitric oxide concentration. Moreover, while BF BDNF levels decreased during SD in control rats, such a decline was not observed in CLI rats. Taken together, neonatal CLI treatment produces long-lasting functional changes in the sleep architecture and sleep regulation in adult rats, accompanied by dysregulated BDNF signaling in the BF.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Clomipramine/pharmacology , Depressive Disorder/chemically induced , Homeostasis/drug effects , Prosencephalon/drug effects , Sleep, REM/drug effects , Animals , Animals, Newborn , Behavior, Animal/drug effects , Behavior, Animal/physiology , Depressive Disorder/drug therapy , Depressive Disorder/physiopathology , Disease Models, Animal , Homeostasis/physiology , Male , Prosencephalon/growth & development , Prosencephalon/physiology , Rats , Rats, Wistar , Sleep, REM/physiology
6.
Brain Res ; 1399: 40-8, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21645878

ABSTRACT

The basal forebrain (BF) is an important mediator of cortical arousal, which is innervated by all ascending arousal systems. During sleep deprivation (SD) a site-specific accumulation of sleep factors in the BF results in increased sleep pressure (Kalinchuk et al., 2006; Porkka-Heiskanen et al., 1997; Porkka-Heiskanen et al., 2000). However, animals are able to stay awake and even increase their neuronal activity in the BF and cortex during SD, suggesting increased activity of the ascending arousal systems to counteract the effect of sleep pressure. This study used in vivo microdialysis to measure the effect of a 6h SD, by "gentle handling" in freely moving rats, on the extracellular levels of serotonin and dopamine metabolites (5-HIAA, and DOPAC and HVA respectively) in the BF. Additionally, because glucocorticoids can interact with monoaminergic neurotransmission, and SD could be stressful, corticosterone levels were measured. We found an increase in extracellular serotonin and dopamine metabolite levels (n=8, p≤0.05). No interaction between corticosterone and the monoaminergic systems was apparent. Extracellular corticosterone levels showed no increase during the first 3h of SD, and the subsequent increase (n=8, p≤0.05) did not result in values exceeding the normal diurnal maximum, indicating that no substantial stress was induced. The results demonstrate that SD increases extracellular dopamine and serotonin metabolites in the BF, suggesting increased activity of the ascending arousal systems. It remains to be investigated what the specific roles of the dopaminergic and serotonergic ascending arousal systems are in BF-mediated cortical arousal.


Subject(s)
Dopamine/metabolism , Extracellular Fluid/metabolism , Prosencephalon/metabolism , Serotonin/metabolism , Sleep Deprivation/pathology , Analysis of Variance , Animals , Chromatography, High Pressure Liquid/methods , Corticosterone/metabolism , Electroencephalography/methods , Male , Microdialysis/methods , Prosencephalon/cytology , Radioimmunoassay/methods , Rats , Rats, Wistar , Sleep Deprivation/physiopathology
7.
Acta Physiol (Oxf) ; 198(3): 237-49, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20003098

ABSTRACT

AIM: Orexin/hypocretin peptides are expressed in the lateral hypothalamus and involved in the regulation of autonomic functions, energy homeostasis and arousal states. The sleep disorder narcolepsy, which is characterized by excessive daytime sleepiness and occurrence of sudden rapid eye movement (REM) sleep, is associated with a loss of orexin neurones. Our study investigated the effects of orexins on sleep-wake patterns in a novel transgenic mouse line overexpressing the human prepro-orexin (hPPO) gene under the control of its endogenous promoter. METHODS: Orexin overexpression was investigated by PCR, Southern and Western blotting as well as immunohistochemistry. Polysomnographic recordings were performed for analyses of sleep-wake patterns and for electroencephalographic activity during 24 h baseline and during and after 6 h of sleep deprivation (SD). RESULTS: Transgenic hPPO mice had increased expression of human prepro-orexin (hPPO) and orexin-A in the hypothalamus. Transgene expression decreased endogenous orexin-2 receptors but not orexin-1 receptors in the hypothalamus without affecting orexin receptor levels in the basal forebrain, cortex or hippocampus. Transgenic mice compared with their wild type littermates showed small but significant differences in the amount of waking and slow wave sleep, particularly during the light-dark transition periods, in addition to a slight reduction in REM sleep during baseline and during recovery sleep after SD. CONCLUSION: The hPPO-overexpressing mice show a small reduction in REM sleep, in addition to differences in vigilance state amounts in the light/dark transition periods, but overall the sleep-wake patterns of hPPO-overexpressing mice do not significantly differ from their wild type littermates.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Sleep/physiology , Wakefulness/physiology , Animals , Arousal/physiology , Darkness , Electroencephalography , Humans , Hypothalamus/metabolism , Light , Mice , Mice, Transgenic , Orexin Receptors , Orexins , Polysomnography , Protein Isoforms/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Sleep Deprivation/physiopathology , Sleep, REM/physiology , Up-Regulation
8.
Neuroscience ; 157(1): 238-53, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-18805464

ABSTRACT

A topic of high current interest and controversy is the basis of the homeostatic sleep response, the increase in non-rapid-eye-movement (NREM) sleep and NREM-delta activity following sleep deprivation (SD). Adenosine, which accumulates in the cholinergic basal forebrain (BF) during SD, has been proposed as one of the important homeostatic sleep factors. It is suggested that sleep-inducing effects of adenosine are mediated by inhibiting the wake-active neurons of the BF, including cholinergic neurons. Here we examined the association between SD-induced adenosine release, the homeostatic sleep response and the survival of cholinergic neurons in the BF after injections of the immunotoxin 192 immunoglobulin G (IgG)-saporin (saporin) in rats. We correlated SD-induced adenosine level in the BF and the homeostatic sleep response with the cholinergic cell loss 2 weeks after local saporin injections into the BF, as well as 2 and 3 weeks after i.c.v. saporin injections. Two weeks after local saporin injection there was an 88% cholinergic cell loss, coupled with nearly complete abolition of the SD-induced adenosine increase in the BF, the homeostatic sleep response, and the sleep-inducing effects of BF adenosine infusion. Two weeks after i.c.v. saporin injection there was a 59% cholinergic cell loss, correlated with significant increase in SD-induced adenosine level in the BF and an intact sleep response. Three weeks after i.c.v. saporin injection there was an 87% cholinergic cell loss, nearly complete abolition of the SD-induced adenosine increase in the BF and the homeostatic response, implying that the time course of i.c.v. saporin lesions is a key variable in interpreting experimental results. Taken together, these results strongly suggest that cholinergic neurons in the BF are important for the SD-induced increase in adenosine as well as for its sleep-inducing effects and play a major, although not exclusive, role in sleep homeostasis.


Subject(s)
Adenosine/physiology , Antibodies, Monoclonal/pharmacology , Basal Ganglia/physiology , Cholinergic Agents/pharmacology , Homeostasis/physiology , Neurons/physiology , Parasympathetic Nervous System/physiology , Prosencephalon/physiology , Ribosome Inactivating Proteins, Type 1/pharmacology , Sleep/physiology , Acetylcholinesterase/metabolism , Adenosine/metabolism , Animals , Basal Ganglia/cytology , Basal Ganglia/metabolism , Choline O-Acetyltransferase/metabolism , Chromatography, High Pressure Liquid , Electroencephalography/drug effects , Electromyography/drug effects , Glutamate Decarboxylase/metabolism , Injections, Intraventricular , Male , Nerve Fibers/metabolism , Nerve Fibers/physiology , Parasympathetic Nervous System/cytology , Parasympathetic Nervous System/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , Rats , Rats, Wistar , Saporins , Sleep Stages/drug effects , Sleep Stages/physiology
9.
Neuroscience ; 147(3): 811-23, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17574765

ABSTRACT

A prolonged period of waking accumulates sleep pressure, increasing both the duration and the intensity of the subsequent sleep period. Delta power, which is calculated from the slow range electroencephalographic (EEG) oscillations (0.1-4 Hz), is regarded as the marker of sleep intensity. Recent findings indicate that not only the duration but also the quality of waking, determines the level of increase in the delta activity during the subsequent sleep period. Elevated levels of extracellular adenosine in the basal forebrain (BF) during prolonged waking have been proposed to act as the molecular signal of increased sleep pressure, but the role of BF neuronal activity in elevating adenosine has not been previously explored. We hypothesized that an increase in neuronal discharge in the BF would lead to increase in the extracellular adenosine and contribute to the increase in the subsequent sleep. To experimentally increase neuronal activity in the rat BF, we used 3 h in vivo microdialysis application of glutamate or its receptor agonists N-methyl-D-aspartate (NMDA) or AMPA. Samples for adenosine measurement were collected during the drug application and the EEG was recorded during and after the treatment, altogether for 24 h. All treatments increased the duration of the subsequent sleep following the application. In contrast, delta power was elevated only if both the waking EEG theta (5-9 Hz) power (which can be regarded as a marker of active waking) and the extracellular adenosine in the BF were increased during the application. These results indicate that increased neuronal activity in the BF, and particularly the type of neuronal activity coinciding with active waking, is one of the factors contributing to the buildup of the sleep pressure.


Subject(s)
Adenosine/metabolism , Extracellular Fluid/drug effects , Glutamic Acid/pharmacology , Prosencephalon/cytology , Sleep/drug effects , Animals , Chromatography, High Pressure Liquid , Dizocilpine Maleate/pharmacology , Electroencephalography/methods , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Fluid/metabolism , Male , Microdialysis/methods , Oncogene Proteins v-fos/metabolism , Polysomnography , Rats , Rats, Wistar
10.
J Neurochem ; 99(2): 483-98, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17029601

ABSTRACT

Sleep homeostasis is the process by which recovery sleep is generated by prolonged wakefulness. The molecular mechanisms underlying this important phenomenon are poorly understood. Here, we assessed the role of the intercellular gaseous signaling agent NO in sleep homeostasis. We measured the concentration of nitrite and nitrate, indicative of NO production, in the basal forebrain (BF) of rats during sleep deprivation (SD), and found the level increased by 100 +/- 51%. To test whether an increase in NO production might play a causal role in recovery sleep, we administered compounds into the BF that increase or decrease concentrations of NO. Infusion of either a NO scavenger, 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide, or a NO synthase inhibitor, N(omega)-nitro-L-arginine methyl ester (L-NAME), completely abolished non-rapid eye movement (NREM) recovery sleep. Infusion of a NO donor, (Z)-1-[N-(2-aminoethyl)-N-(2-ammonioethyl)amino]diazen-1-ium-1,2diolate (DETA/NO), produced an increase in NREM that closely resembled NREM recovery after prolonged wakefulness. The effects of inhibition of NO synthesis and the pharmacological induction of sleep were effective only in the BF area. Indicators of energy metabolism, adenosine, lactate and pyruvate increased during prolonged wakefulness and DETA/NO infusion, whereas L-NAME infusion during SD prevented the increases. We conclude that an increase in NO production in the BF is a causal event in the induction of recovery sleep.


Subject(s)
Diagonal Band of Broca/metabolism , Nitric Oxide/biosynthesis , Prosencephalon/metabolism , Recovery of Function/physiology , Sleep Deprivation/metabolism , Sleep/physiology , Adenosine/metabolism , Animals , Diagonal Band of Broca/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Lactic Acid/metabolism , Male , Microdialysis , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Prosencephalon/drug effects , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Signal Transduction/physiology , Sleep/drug effects , Sleep Deprivation/physiopathology , Up-Regulation/drug effects , Up-Regulation/physiology
11.
Eur J Neurosci ; 24(5): 1443-56, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16987226

ABSTRACT

Sleep homeostasis is the process by which recovery sleep is generated by prolonged wakefulness. The molecular mechanisms underlying this important phenomenon are poorly understood. We have previously shown that nitric oxide (NO) generation increases in the basal forebrain (BF) during sleep deprivation (SD). Moreover, both NO synthase (NOS) inhibition and a NO scavenger prevented recovery sleep induction, while administration of a NO donor during the spontaneous sleep-wake cycle increased sleep, indicating that NO is necessary and sufficient for the induction of recovery sleep. Next we wanted to know which NOS isoform is involved in the production of recovery sleep. Using in vivo microdialysis we infused specific inhibitors of NOS into the BF of rats during SD, and found that an inhibitor of inducible NOS (iNOS), 1400W, prevented non-rapid eye movement (NREM) recovery, while an inhibitor of neuronal NOS (nNOS), L-N-propyl-arginine, decreased REM recovery but did not affect NREM recovery. Using immunoblot analysis we found that iNOS was not expressed during the spontaneous sleep-wake cycle, but was induced by prolonged wakefulness (increased by 278%). A known iNOS inducer, lipopolysaccharide, evoked an increase in sleep that closely resembled recovery sleep, and its effects were abolished by 1400W. These results suggest that the elevation of NO produced by induction of iNOS in the BF during prolonged wakefulness is a specific mechanism for producing NREM recovery sleep and that the two NOS isoforms have a complementary role in NREM and REM recovery induction.


Subject(s)
Nitric Oxide Synthase Type II/metabolism , Nitric Oxide Synthase Type I/metabolism , Sleep Stages/physiology , Adenosine/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Electroencephalography/methods , Electromyography/methods , Imines/pharmacology , Lactic Acid/metabolism , Lipopolysaccharides/pharmacology , Male , Microinjections/methods , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Sleep Stages/drug effects , Wakefulness/physiology
12.
Eur J Endocrinol ; 150(5): 737-42, 2004 May.
Article in English | MEDLINE | ID: mdl-15132733

ABSTRACT

OBJECTIVE: Orexins have been implicated in the regulation of several physiological functions including reproduction, energy balance and vigilance state. For successful reproduction, the precisely timed hormonal secretions of the estrous cycle must be combined with appropriate nutritional and vigilance states. The steroid- and nutritional state-dependent modulation of LH release by orexins, as well as an increase of vigilance, suggest that orexins may co-ordinate these functions in the course of the estrous cycle. DESIGN: We studied the brain tissue levels of orexins in the course of the estrous cycle in young and middle-aged rats. Young cycling rats (3 months old) and irregularly/non-cycling (7-9 months old) female rats were inspected for vaginal smears and serum hormone levels. METHODS: Tissue concentrations of orexin A and B were measured in the hypothalamus and lateral hypothalamus on different days of the estrous cycle. RESULTS: Orexin A concentration in the hypothalamus of young cycling rats was higher on the day of proestrus 5-6 h after the lights were switched on than on the other days of the estrous cycle at the same circadian time. Orexin B concentration was higher on both the day of proestrus and the day of estrus as compared with the days of diestrus. The hypothalamic concentrations of both orexin A and B in the non-cycling middle-aged rats were lower than those in cycling rats on the days of proestrus and estrus. CONCLUSIONS: We have concluded that the high hypothalamic concentration of orexins on the day of proestrus may contribute to the LH and prolactin surges. High orexin A levels may also contribute to the decreased amount of sleep on the day of proestrus.


Subject(s)
Aging/metabolism , Carrier Proteins/metabolism , Estrus/metabolism , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins , Neuropeptides/metabolism , Animals , Estradiol/blood , Female , Hypothalamic Area, Lateral/metabolism , Orexins , Osmolar Concentration , Proestrus/metabolism , Rats , Rats, Wistar
13.
Int Clin Psychopharmacol ; 17(4): 177-84, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12131601

ABSTRACT

The effect of a single dose of 10 mg olanzapine on healthy volunteers of both sexes was examined using polysomnography and power spectral analysis. The structure and continuity of sleep were unaffected by olanzapine in both sexes. The increase in both actual sleep time and slow wave sleep in females correlated with the increase in theta power, while delta power was not significantly elevated, suggesting that theta power may be a sensitive indicator of changes in sleep. The changes in sleep had the same tendency in men, but they were not significant. The difference between the sexes could not be explained by differences in body mass index. Olanzapine affects sleep probably through 5-HT(2C) receptors. The receptor gene is located on the X-chromosome, inducing an allelic difference between the females and males. This difference may contribute to the different effects of olanzapine on sleep. Olanzapine seems to preserve the normal structure of sleep and increase the amount of slow-wave sleep, which might be of additional benefit in treatment of schizophrenia. The effective clinical dose may be lower for females than males.


Subject(s)
Antipsychotic Agents/pharmacology , Pirenzepine/analogs & derivatives , Pirenzepine/pharmacology , Serotonin Agents/pharmacology , Sleep/drug effects , Adult , Antipsychotic Agents/administration & dosage , Benzodiazepines , Electroencephalography , Female , Humans , Male , Olanzapine , Pirenzepine/administration & dosage , Polysomnography , Serotonin Agents/administration & dosage , Sex Factors , Sleep Stages/drug effects , Sleep Stages/genetics
14.
Neuroscience ; 104(3): 731-9, 2001.
Article in English | MEDLINE | ID: mdl-11440805

ABSTRACT

There is considerable evidence to suggest that adenosine is a modulator of behavioral state. Our previous reports showed that perfusion of adenosine into the basal forebrain decreased wakefulness. Furthermore, prolonged wakefulness resulted in increased levels of extracellular adenosine in the basal forebrain of cats and rats. However, the longer-term consequences of prolonged wakefulness and increased adenosine are largely unknown. We report here an increase in the DNA binding activity of the transcription factor, nuclear factor-kappa B (NF-kappaB) following 3 h of sustained wakefulness in the rat basal forebrain. Moreover, this treatment led to the appearance of the p65 subunit of NF-kappaB in the nucleus, as determined by western blot analysis of nuclear proteins. This contrasted with undetectable levels in the sleeping controls. A concomitant disappearance of I-kappaB in cytoplasm suggested the degradation of this inhibitor of NF-kappaB. In the acute in vitro basal forebrain slice preparation, perfusion of adenosine increased NF-kappaB DNA binding while pretreatment of the slices with the A1 adenosine receptor antagonist, cyclopentyl-1-3-dimethylxanthine, significantly reduced NF-kappaB DNA binding. These results are compatible with the hypothesis that increases in the levels of adenosine in the basal forebrain, that occur during prolonged wakefulness, act through an A1 adenosine receptor and a second messenger system to increase the activity of the transcription factor NF-kappaB. We further hypothesize that some of the long duration effects of prolonged wakefulness/sleep deprivation on performance and physiology, often termed 'sleep debt', might be mediated through adenosine and its activation of NF-kappaB, which is known to alter the expression of several behavioral state regulatory factors.


Subject(s)
Adenosine/metabolism , Basal Nucleus of Meynert/metabolism , Basal Nucleus of Meynert/physiology , NF-kappa B/metabolism , Neurons/metabolism , Receptors, Purinergic P1/metabolism , Wakefulness/physiology , Animals , Basal Nucleus of Meynert/cytology , Binding Sites/drug effects , Binding Sites/physiology , Cell Compartmentation/physiology , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cytoplasm/metabolism , Cytoplasm/ultrastructure , DNA/metabolism , Male , Neurons/cytology , Neurons/drug effects , Protein Transport/physiology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Rats , Rats, Long-Evans , Signal Transduction/physiology , Time Factors
15.
Neuroreport ; 12(8): 1577-80, 2001 Jun 13.
Article in English | MEDLINE | ID: mdl-11409719

ABSTRACT

Extracellular levels of adenosine increase in basal forebrain following prolonged wakefulness. Moreover, perfusion of adenosine into basal forebrain increases sleep. In this study we have examined the adenosine receptor subtypes, A1 and A2A, for changes in the levels of mRNA using RT-PCR and in situ hybridization and the receptor ligand binding efficiency using autoradiography following 3 and 6 h of sleep deprivation. We observed that A1 receptor mRNA levels increased in basal forebrain with no changes in other forebrain areas examined. A1 receptor binding was not affected. A2A receptor mRNA and ligand binding were undetectable in basal forebrain. However, in the olfactory tubercle, A2A mRNA and receptor binding decreased significantly. Based on the significant increase in the A1 but not in A2A receptor, we hypothesize that the effects of sleep deprivation-induced increased adenosine are mediated by A1 receptor in basal forebrain of rats.


Subject(s)
RNA, Messenger/metabolism , Receptors, Purinergic P1/genetics , Sleep Deprivation/metabolism , Animals , Autoradiography , In Situ Hybridization , Male , Rats , Rats, Long-Evans , Receptor, Adenosine A2A , Receptors, Purinergic P1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
16.
Biol Signals Recept ; 9(6): 319-27, 2000.
Article in English | MEDLINE | ID: mdl-11025338

ABSTRACT

Recent reports from our laboratory have shown that extracellular adenosine levels selectively increase in basal forebrain during prolonged wakefulness in cats and rats. Furthermore, microdialysis perfusion of adenosine into the basal forebrain (BF) increased sleepiness and decreased wakefulness in both the species, whereas perfusion of the A(1)-receptor-selective antagonist, cyclopentyl-1, 3-dimethylxanthine resulted in increased wakefulness, an observation similar to that found with caffeine or theophylline administration. The selective participation of the A(1) subtype of the adenosine receptor in mediating the effects of adenosine in the BF was further examined by the technique of single unit recording performed in conjunction with microdialysis perfusion of selective agonists and antagonists. Perfusion of the A(1) agonist cyclohexyladenosine, inhibited the activity of wake-active neurons in the basal forebrain. The effect of prolonged wakefulness-induced increases in adenosine levels were further investigated by determining the changes in the BF in the levels of A(1) receptor binding and the levels of its mRNA. We observed that A(1) receptor mRNA levels increase after 6 h of sleep deprivation. One of the transcription factors that showed increased DNA-binding activity was nuclear factor kappaB (NF-kappaB) and may regulate the expression of A(1) mRNA. We observed, using a gel shift assay, that the DNA-binding activity of NF-kappaB increased following 3 h of sleep deprivation. This was further supported by the increased appearance of NF-kappaB protein in the nuclear extracts and the consequent disappearance of cytoplasmic protein inhibitor kappaB (I-kappaB). Together our results reviewed in this report suggest that the somnogenic effects of adenosine in the BF area may be mediated by the A(1) subtype of adenosine receptor, and its expression might be regulated by induction in the NF-kappaB protein as its transcription factor. This positive feedback might mediate some of long-duration effects of sleep deprivation, including 'sleep debt'.


Subject(s)
Adenosine/metabolism , Sleep/physiology , Wakefulness/physiology , NF-kappa B/metabolism , Prosencephalon/physiology , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Sleep Deprivation/metabolism , Transcription Factors/metabolism
17.
Neuroscience ; 99(3): 507-17, 2000.
Article in English | MEDLINE | ID: mdl-11029542

ABSTRACT

Previous data suggested that increases in extracellular adenosine in the basal forebrain mediated the sleep-inducing effects of prolonged wakefulness. The present study sought to determine if the state-related changes found in basal forebrain adenosine levels occurred uniformly throughout the brain. In vivo microdialysis sample collection coupled to microbore high-performance liquid chromatography measured extracellular adenosine levels in six brain regions of the cat: basal forebrain, cerebral cortex, thalamus, preoptic area of hypothalamus, dorsal raphe nucleus and pedunculopontine tegmental nucleus. In all these brain regions extracellular adenosine levels showed a similar decline of 15 to 20% during episodes of spontaneous sleep relative to wakefulness. Adenosine levels during non-rapid eye movement sleep did not differ from rapid eye movement sleep. In the course of 6h of sleep deprivation, adenosine levels increased significantly in the cholinergic region of the basal forebrain (to 140% of baseline) and, to a lesser extent in the cortex, but not in the other regions. Following sleep deprivation, basal forebrain adenosine levels declined very slowly, remaining significantly elevated throughout a 3-h period of recovery sleep, but elsewhere levels were either similar to, or lower than, baseline. The site-specific accumulation of adenosine during sleep deprivation suggests a differential regulation of adenosine levels by as yet unidentified mechanisms. Moreover, the unique pattern of sleep-related changes in basal forebrain adenosine level lends strong support to the hypothesis that the sleep-promoting effects of adenosine, as well as the sleepiness associated with prolonged wakefulness, are both mediated by adenosinergic inhibition of a cortically projecting basal forebrain arousal system.


Subject(s)
Adenosine/metabolism , Brain Chemistry/physiology , Sleep Deprivation/metabolism , Sleep, REM/physiology , Animals , Arousal/physiology , Cats , Extracellular Space/metabolism , Male , Microdialysis , Preoptic Area/metabolism , Prosencephalon/metabolism , Raphe Nuclei/metabolism , Sleep Deprivation/physiopathology , Wakefulness/physiology
18.
Behav Brain Res ; 115(2): 183-204, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11000420

ABSTRACT

This review describes a series of animal experiments that investigate the role of endogenous adenosine (AD) in sleep. We propose that AD is a modulator of the sleepiness associated with prolonged wakefulness. More specifically, we suggest that, during prolonged wakefulness, extracellular AD accumulates selectively in the basal forebrain (BF) and cortex and promotes the transition from wakefulness to slow wave sleep (SWS) by inhibiting cholinergic and non-cholinergic wakefulness-promoting BF neurons at the AD A1 receptor. New in vitro data are also compatible with the hypothesis that, via presynaptic inhibition of GABAergic inhibitory input, AD may disinhibit neurons in the preoptic/anterior hypothalamus (POAH) that have SWS-selective activity and Fos expression. Our in vitro recordings initially showed that endogenous AD suppressed the discharge activity of neurons in the BF cholinergic zone via the AD A1 receptor. Moreover, in identified mesopontine cholinergic neurons, AD was shown to act post-synaptically by hyperpolarizng the membrane via an inwardly rectifying potassium current and inhibition of the hyperpolarization-activated current, I(h). In vivo microdialysis in the cat has shown that AD in the BF cholinergic zone accumulates during prolonged wakefulness, and declines slowly during subsequent sleep, findings confirmed in the rat. Moreover, increasing BF AD concentrations to approximately the level as during sleep deprivation by a nucleoside transport blocker mimicked the effect of sleep deprivation on both the EEG power spectrum and behavioral state distribution: wakefulness was decreased, and there were increases in SWS and REM sleep. As predicted, microdialyis application of the specific A1 receptor antagonist cyclopentyltheophylline (CPT) in the BF produced the opposite effects on behavioral state, increasing wakefulness and decreasing SWS and REM. Combined unit recording and microdialysis studies have shown neurons selectively active in wakefulness, compared with SWS, have discharge activity suppressed by both AD and the A1-specific agonist cyclohexyladenosine (CHA), while discharge activity is increased by the A1 receptor antagonist, CPT. We next addressed the question of whether AD exerts its effects locally or globally. Adenosine accumulation during prolonged wakefulness occurred in the BF and neocortex, although, unlike in the BF, cortical AD levels declined in the 6th h of sleep deprivation and declined further during subsequent recovery sleep. Somewhat to our surprise, AD concentrations did not increase during prolonged wakefulness (6 h) even in regions important in behavioral state control, such as the POAH, dorsal raphe nucleus, and pedunculopontine tegmental nucleus, nor did it increase in the ventrolateral/ventroanterior thalamic nucleii. These data suggest the presence of brain region-specific differences in AD transporters and/or degradation that become evident with prolonged wakefulness, even though AD concentrations are higher in all brain sites sampled during the naturally occurring (and shorter duration) episodes of wakefulness as compared to sleep episodes in the freely moving and behaving cat. Might AD also produce modulation of activity of neurons that have sleep selective transcriptional (Fos) and discharge activity in the preoptic/anterior hypothalamus zone? Whole cell patch clamp recordings in the in vitro horizontal slice showed fast and likely GABAergic inhibitory post-synaptic potentials and currents that were greatly decreased by bath application of AD. Adenosine may thus disinhibit and promote expression of sleep-related neuronal activity in the POAH. In summary, a growing body of evidence supports the role of AD as a mediator of the sleepiness following prolonged wakefulness, a role in which its inhibitory actions on the BF wakefulness-promoting neurons may be especially important.


Subject(s)
Adenosine/physiology , Anterior Hypothalamic Nucleus/physiology , Basal Ganglia/physiology , Behavior, Animal/physiology , Neurons/physiology , Preoptic Area/physiology , Prosencephalon/physiology , Animals , Anterior Hypothalamic Nucleus/anatomy & histology , Anterior Hypothalamic Nucleus/cytology , Basal Ganglia/anatomy & histology , Basal Ganglia/cytology , Cats , Electroencephalography , In Vitro Techniques , Microdialysis , Polysomnography , Preoptic Area/anatomy & histology , Preoptic Area/cytology , Prosencephalon/anatomy & histology , Prosencephalon/cytology , Rats , Rats, Long-Evans
19.
Pharmacol Biochem Behav ; 66(4): 721-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10973509

ABSTRACT

In order to study the role of endogenous somatostatin in the physiologic modulation of REM sleep (REMS), we measured the effect of intracerebroventricular (ICV) injection of somatostatin antagonist (SA) cyclo-(7-aminoheptanoyl-phe-d-trp-lys-thr(bzl)) on sleep in rats. The effect of ICV SA was also tested after 24-h REMS deprivation with the platform method. To study the role of locus coeruleus (LC) as a site of the sleep inducing action for somatostatin and galanin we microinjected SA, somatostatin, and galanin locally into LC. In all experiments, vigilance state was analyzed visually from 6 h post-injection EEG/EMG recording. Injection of 0.5 and 2 nmol of SA ICV reduced spontaneous REMS and 2 nmol dose reduced also rebound REMS after REMS deprivation when compared with controls (artificial cerebrospinal fluid vehicle). Microinjection of 0.25 nmol of SA into LC reduced REMS, whereas microinjection of somatostatin, galanin, and a combined injection of them were not effective to induce REMS. The results suggest that endogenous somatostatin may contribute to facilitation of REMS. Somatostatin receptors in the LC may be one possible mediator of this effect.


Subject(s)
Hormone Antagonists/pharmacology , Locus Coeruleus/physiology , Sleep, REM/drug effects , Somatostatin/analogs & derivatives , Somatostatin/antagonists & inhibitors , Animals , Electroencephalography/drug effects , Electromyography/drug effects , Galanin/administration & dosage , Galanin/pharmacology , Injections, Intraventricular , Male , Microinjections , Rats , Rats, Wistar , Somatostatin/administration & dosage , Somatostatin/pharmacology
20.
Brain Res Mol Brain Res ; 73(1-2): 1-10, 1999 Nov 10.
Article in English | MEDLINE | ID: mdl-10581392

ABSTRACT

In several brain areas, extracellular adenosine (AD) levels are higher during waking than sleep and during prolonged wakefulness AD levels in the basal forebrain increase progressively. Similarly, c-Fos levels in several brain areas are higher during waking than sleep and remain elevated during prolonged wakefulness. In the present study, we investigated the effect of extracellular AD levels on c-Fos protein and activator protein-1 (AP1) binding in the basal forebrain of rats. Increased levels of extracellular AD were induced either by keeping the animals awake, or by local perfusion of AD into the basal forebrain. During prolonged wakefulness extracellular AD concentration was monitored using in vivo microdialysis. The effect of AD perfusion on the behavioral states was recorded using polysomnography. At the end of the perfusion period the basal forebrain tissue was analyzed for the levels of c-Fos protein and AP1 binding. In vivo microdialysis measurements showed an increase in AD levels with prolonged wakefulness. Unilateral perfusion of AD (300 microM) increased non-REM sleep and delta power (0.5 to 4 Hz) when compared to rats perfused with artificial CSF. The levels of c-Fos protein and the AP1 DNA binding were high in the basal forebrain of both sleep-deprived animals and in animals perfused with AD. The results suggest that AD might mediate, at least in part, the long term effects of sleep deprivation by inducing c-Fos protein and subsequent AP1 binding.


Subject(s)
Adenosine/metabolism , Sleep/physiology , Wakefulness/physiology , Adenosine/pharmacology , Animals , Arousal/drug effects , Arousal/physiology , Choline O-Acetyltransferase/analysis , DNA/metabolism , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Extracellular Space/chemistry , Immunohistochemistry , Male , Prosencephalon/chemistry , Prosencephalon/drug effects , Prosencephalon/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Long-Evans , Sleep/drug effects , Sleep Deprivation , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/metabolism , Wakefulness/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...