Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 10730, 2018 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-30013200

RESUMO

The functions of purinergic P2 receptors (P2Rs) for extracellular adenosine triphosphate (ATP) are poorly understood. Here, for the first time, we show that activation of P2Rs in an important arousal region, the basal forebrain (BF), promotes wakefulness, whereas inhibition of P2Rs promotes sleep. Infusion of a non-hydrolysable P2R agonist, ATP-γ-S, into mouse BF increased wakefulness following sleep deprivation. ATP-γ-S depolarized BF cholinergic and cortically-projecting GABAergic neurons in vitro, an effect blocked by antagonists of ionotropic P2Rs (P2XRs) or glutamate receptors. In vivo, ATP-γ-S infusion increased BF glutamate release. Thus, activation of BF P2XRs promotes glutamate release and excitation of wake-active neurons. Conversely, pharmacological antagonism of BF P2XRs decreased spontaneous wakefulness during the dark (active) period. Together with previous findings, our results suggest sleep-wake regulation by BF extracellular ATP involves a balance between excitatory, wakefulness-promoting effects mediated by direct activation of P2XRs and inhibitory, sleep-promoting effects mediated by degradation to adenosine.


Assuntos
Prosencéfalo Basal/fisiologia , Receptores Purinérgicos P2/metabolismo , Vigília/fisiologia , Adenosina/metabolismo , Trifosfato de Adenosina/administração & dosagem , Trifosfato de Adenosina/análogos & derivados , Animais , Prosencéfalo Basal/citologia , Prosencéfalo Basal/efeitos dos fármacos , Eletrodos Implantados , Eletroencefalografia/instrumentação , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Neurônios GABAérgicos/efeitos dos fármacos , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Camundongos , Modelos Animais , Técnicas de Patch-Clamp , Agonistas do Receptor Purinérgico P2/administração & dosagem , Antagonistas do Receptor Purinérgico P2/administração & dosagem , Receptores de Glutamato/metabolismo , Receptores Purinérgicos P2/efeitos dos fármacos , Sono/efeitos dos fármacos , Sono/fisiologia , Vigília/efeitos dos fármacos
2.
J Sleep Res ; 24(5): 549-558, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25900125

RESUMO

Although chronic sleep restriction frequently produces long-lasting behavioural and physiological impairments in humans, the underlying neural mechanisms are unknown. Here we used a rat model of chronic sleep restriction to investigate the role of brain adenosine and noradrenaline systems, known to regulate sleep and wakefulness, respectively. The density of adenosine A1 and A2a receptors and ß-adrenergic receptors before, during and following 5 days of sleep restriction was assessed with autoradiography. Rats (n = 48) were sleep-deprived for 18 h day(-1) for 5 consecutive days (SR1-SR5), followed by 3 unrestricted recovery sleep days (R1-R3). Brains were collected at the beginning of the light period, which was immediately after the end of sleep deprivation on sleep restriction days. Chronic sleep restriction increased adenosine A1 receptor density significantly in nine of the 13 brain areas analysed with elevations also observed on R3 (+18 to +32%). In contrast, chronic sleep restriction reduced adenosine A2a receptor density significantly in one of the three brain areas analysed (olfactory tubercle which declined 26-31% from SR1 to R1). A decrease in ß-adrenergic receptors density was seen in substantia innominata and ventral pallidum which remained reduced on R3, but no changes were found in the anterior cingulate cortex. These data suggest that chronic sleep restriction can induce long-term changes in the brain adenosine and noradrenaline receptors, which may underlie the long-lasting neurocognitive impairments observed in chronic sleep restriction.


Assuntos
Encéfalo/metabolismo , Receptores Adrenérgicos/metabolismo , Receptores Purinérgicos P1/metabolismo , Privação do Sono/metabolismo , Animais , Autorradiografia , Prosencéfalo Basal/metabolismo , Doença Crônica , Giro do Cíngulo/metabolismo , Masculino , Transtornos Neurocognitivos/complicações , Transtornos Neurocognitivos/metabolismo , Tubérculo Olfatório/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor A2A de Adenosina/metabolismo , Sono/fisiologia , Privação do Sono/complicações , Substância Inominada/metabolismo , Fatores de Tempo , Vigília/fisiologia
3.
Neurosci Lett ; 580: 27-31, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25093703

RESUMO

Acute sleep loss increases pro-inflammatory and synaptic plasticity-related molecules in the brain, including interleukin-1 beta (IL-1ß), tumor necrosis factor-alpha (TNF-α), and brain-derived neurotrophic factor (BDNF). These molecules enhance non-rapid eye movement sleep slow wave activity (SWA), also known as electroencephalogram delta power, and modulate neurocognitive performance. Evidence suggests that chronic sleep restriction (CSR), a condition prevalent in today's society, does not elicit the enhanced SWA that is seen after acute sleep loss, although it cumulatively impairs neurocognitive functioning. Rats were continuously sleep deprived for 18h per day and allowed 6h of ad libitum sleep opportunity for 1 (SR1), 3 (SR3), or 5 (SR5) successive days (i.e., CSR). IL-1ß, TNF-α, and BDNF mRNA levels were determined in the somatosensory cortex, frontal cortex, hippocampus, and basal forebrain. Largely, brain IL-1ß and TNF-α expression were significantly enhanced throughout CSR. In contrast, BDNF mRNA levels were similar to baseline values in the cortex after 1 day of SR and significantly lower than baseline values in the hippocampus after 5 days of SR. In the basal forebrain, BDNF expression remained elevated throughout the 5 days of CSR, although IL-1ß expression was significantly reduced. The chronic elevations of IL-1ß and TNF-α and inhibition of BDNF might contribute to the reported lack of SWA responses reported after CSR. Further, the CSR-induced enhancements in brain inflammatory molecules and attenuations in hippocampal BDNF might contribute to neurocognitive and vigilance detriments that occur from CSR.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Interleucina-1beta/metabolismo , Privação do Sono/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Córtex Cerebral/metabolismo , Interleucina-1beta/genética , Masculino , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/genética
4.
Physiol Rev ; 92(3): 1087-187, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22811426

RESUMO

This review summarizes the brain mechanisms controlling sleep and wakefulness. Wakefulness promoting systems cause low-voltage, fast activity in the electroencephalogram (EEG). Multiple interacting neurotransmitter systems in the brain stem, hypothalamus, and basal forebrain converge onto common effector systems in the thalamus and cortex. Sleep results from the inhibition of wake-promoting systems by homeostatic sleep factors such as adenosine and nitric oxide and GABAergic neurons in the preoptic area of the hypothalamus, resulting in large-amplitude, slow EEG oscillations. Local, activity-dependent factors modulate the amplitude and frequency of cortical slow oscillations. Non-rapid-eye-movement (NREM) sleep results in conservation of brain energy and facilitates memory consolidation through the modulation of synaptic weights. Rapid-eye-movement (REM) sleep results from the interaction of brain stem cholinergic, aminergic, and GABAergic neurons which control the activity of glutamatergic reticular formation neurons leading to REM sleep phenomena such as muscle atonia, REMs, dreaming, and cortical activation. Strong activation of limbic regions during REM sleep suggests a role in regulation of emotion. Genetic studies suggest that brain mechanisms controlling waking and NREM sleep are strongly conserved throughout evolution, underscoring their enormous importance for brain function. Sleep disruption interferes with the normal restorative functions of NREM and REM sleep, resulting in disruptions of breathing and cardiovascular function, changes in emotional reactivity, and cognitive impairments in attention, memory, and decision making.


Assuntos
Encéfalo/fisiopatologia , Transtornos do Sono-Vigília/fisiopatologia , Sono , Vigília , Animais , Atenção , Encéfalo/metabolismo , Ondas Encefálicas , Cognição , Emoções , Predisposição Genética para Doença , Genômica , Humanos , Memória , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais/fisiopatologia , Fenótipo , Proteômica , Transdução de Sinais , Sono/genética , Transtornos do Sono-Vigília/genética , Transtornos do Sono-Vigília/metabolismo , Transtornos do Sono-Vigília/psicologia , Transtornos do Sono-Vigília/terapia , Sono REM , Vigília/genética
5.
Sleep ; 35(6): 861-9, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22654205

RESUMO

STUDY OBJECTIVE: Sleep responses to chronic sleep restriction (CSR) might be very different from those observed after short-term total sleep deprivation. For example, after sleep restriction continues for several consecutive days, animals no longer express compensatory increases in daily sleep time and sleep intensity. However, it is unknown if these allostatic, or adaptive, sleep responses to CSR are paralleled by behavioral and neurochemical measures of sleepiness. DESIGN: This study was designed to investigate CSR-induced changes in (1) sleep time and intensity as a measure of electrophysiological sleepiness, (2) sleep latency as a measure of behavioral sleepiness, and (3) brain adenosine A1 (A1R) and A2a receptor (A2aR) mRNA levels as a putative neurochemical correlate of sleepiness. SUBJECTS: Male Sprague-Dawley rats INTERVENTIONS: A 5-day sleep restriction (SR) protocol consisting of 18-h sleep deprivation and 6-h sleep opportunity each day. MEASUREMENT AND RESULTS: Unlike the first SR day, rats did not sleep longer or deeper on days 2 through 5, even though they exhibited significant elevations of behavioral sleepiness throughout all 5 SR days. For all SR days and recovery day 1, A1R mRNA in the basal forebrain was maintained at elevated levels, whereas A2aR mRNA in the frontal cortex was maintained at reduced levels. CONCLUSION: CSR LEADS TO A DECOUPLING OF SLEEPINESS FROM SLEEP TIME AND SLEEP INTENSITY, SUGGESTING THAT THERE ARE AT LEAST TWO DIFFERENT SLEEP REGULATORY SYSTEMS: one mediating sleepiness (homeostatic) and the other mediating sleep time/intensity (allostatic). The time course of changes observed in adenosine receptor mRNA levels suggests that the basal forebrain and cortical adenosine system might mediate sleepiness rather than sleep time or intensity.


Assuntos
Prosencéfalo/química , Receptores Purinérgicos P1/análise , Privação do Sono/fisiopatologia , Sono/fisiologia , Vigília/fisiologia , Animais , Eletroencefalografia , Masculino , Metiltransferases , Proteínas Nucleares , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptor A1 de Adenosina/análise , Receptor A1 de Adenosina/fisiologia , Receptores A2 de Adenosina/análise , Receptores A2 de Adenosina/fisiologia , Receptores Purinérgicos P1/fisiologia
6.
Neurosci Lett ; 469(1): 1-5, 2010 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-19914331

RESUMO

Sleep fragmentation (SF) impairs the restorative/cognitive benefits of sleep via as yet unidentified alterations in neural physiology. Previously, we found that hippocampal synaptic plasticity and spatial learning are impaired in a rat model of SF which utilizes a treadmill to awaken the animals every 2 min, mimicking the frequency of awakenings observed in human sleep apnea patients. Here, we investigated the cellular mechanisms responsible for these effects, using whole-cell patch-clamp recordings. 24h of SF decreased the excitability of hippocampal CA1 pyramidal neurons via decreased input resistance, without alterations in other intrinsic membrane or action potential properties (when compared to cage controls, or to exercise controls that experienced the same total amount of treadmill movement as SF rats). Contrary to our initial prediction, the hyperpolarizing response to bath applied adenosine (30 microM) was reduced in the CA1 neurons of SF treated rats. Our initial prediction was based on the evidence that sleep loss upregulates cortical adenosine A1 receptors; however, the present findings are consistent with a very recent report that hippocampal A1 receptors are not elevated by sleep loss. Thus, increased adenosinergic inhibition is unlikely to be responsible for reduced hippocampal long-term potentiation in SF rats. Instead, the reduced excitability of CA1 pyramidal neurons observed here may contribute to the loss of hippocampal long-term potentiation and hippocampus-dependent cognitive impairments associated with sleep disruption.


Assuntos
Adenosina/fisiologia , Região CA1 Hipocampal/fisiologia , Células Piramidais/fisiologia , Privação do Sono , Potenciais de Ação , Adenosina/farmacologia , Animais , Região CA1 Hipocampal/citologia , Masculino , Potenciais da Membrana , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/biossíntese , Membranas Sinápticas/fisiologia , Regulação para Cima
7.
Sleep ; 31(10): 1393-8, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18853936

RESUMO

STUDY OBJECTIVE: The inhibitory neuromodulator adenosine has been proposed as a homeostatic sleep factor that acts potently in the basal forebrain (BF) to increase sleepiness. Here 300 microM of adenosine was dialyzed in the BF of rats, and the effect on vigilance was determined in the rat Psychomotor Vigilance Task (rPVT). DESIGN: Rats experienced all experimental conditions in a repeated-measures, cross-over design. PATIENTS OR PARTICIPANTS: Twelve young adult male Fischer-Norway rats. INTERVENTIONS: Sustained attention performance in the rPVT was evaluated following 2 hours of bilateral microdialysis perfusion of vehicle, adenosine (300 microM), or codialysis of 300 microM of adenosine with the A1 receptor antagonist 8-cyclopentyltheophylline. MEASUREMENTS AND RESULTS: During rPVT performance, response latencies and performance lapses increased significantly after adenosine dialysis when compared with baseline (no dialysis) or vehicle dialysis sessions. The codialysis of 8-cyclopentyltheophylline with adenosine completely blocked the effects produced by adenosine alone, resulting in performance equivalent to that of the vehicle sessions. CONCLUSIONS: Pharmacologic elevation of BF adenosine in rats produced vigilance impairments resembling the effect of sleep deprivation on vigilance performance in both man and rats. This effect of exogenous adenosine was completely blocked by codialysis with an adenosine A1 receptor antagonist. The results are consistent with the hypothesis that sleep loss induces elevations of BF adenosine that, acting via A1 receptors, lead to increased sleepiness and impaired vigilance.


Assuntos
Adenosina/farmacologia , Nível de Alerta/efeitos dos fármacos , Atenção/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Sono/efeitos dos fármacos , Adenosina/antagonistas & inibidores , Animais , Comportamento Apetitivo/efeitos dos fármacos , Microdiálise , Ratos , Ratos Endogâmicos F344 , Tempo de Reação/efeitos dos fármacos , Teofilina/análogos & derivados
8.
J Physiol ; 586(21): 5215-29, 2008 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-18787037

RESUMO

Sleep fragmentation (SF), a primary feature of obstructive sleep apnoea (OSA), impairs hippocampal long-term potentiation and causes cognitive/attention deficits. However, its influence upon respiratory control has hardly been studied. This study examined the effect of SF on ventilatory long-term facilitation (LTF, a persistent augmentation of respiratory activity after episodic hypoxia) and the hypoxic ventilatory response (HVR), and investigated the role of adenosine A1 receptors in these SF effects in conscious adult male Sprague-Dawley rats. SF, confirmed by sleep architecture recordings, was achieved by periodic, forced locomotion in a rotating drum (30 s rotation/90 s stop for 24 h). LTF, elicited by five episodes of 5 min poikilocapnic hypoxia (10% O2) with 5 min intervals, was measured by plethysmography. Resting ventilation and metabolic rate were unchanged, HVR was reduced (150.6 +/- 3.5% versus 110.4 +/- 12.3%) and LTF was eliminated (22.6 +/- 0.5% versus -0.1 +/- 1.3%) shortly after 24 h SF. The SF-induced impairments were SF duration dependent, and completely reversible as HVR (< 24 h) and LTF (< 48 h) returned spontaneously to their pre-SF values. The SF-impaired HVR was improved (130.3 +/- 4.2%) and SF-eliminated LTF was restored (19.6 +/- 0.9%) by systemic injection of the adenosine A1 receptor antagonist 8-CPT (2.5 mg kg(-1)) approximately 30 min before LTF elicitation. Both HVR and LTF were also similarly impaired by 24 h total sleep deprivation or 24 h repeated cage tapping-induced SF, but not by a 24 h locomotion control protocol for SF. Collectively, these data suggest that: (1) 24 h SF impairs LTF and poikilocapnic HVR; (2) these impairments require A1 receptors; and (3) SF of OSA may exacerbate OSA via impaired ventilatory control mechanisms.


Assuntos
Receptor A1 de Adenosina/metabolismo , Mecânica Respiratória/fisiologia , Privação do Sono/metabolismo , Animais , Metabolismo Basal/fisiologia , Temperatura Corporal , Corticosterona/sangue , Frequência Cardíaca , Hipóxia , Masculino , Ratos , Ratos Sprague-Dawley
10.
Prog Neurobiol ; 73(6): 379-96, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15313333

RESUMO

This review addresses three principal questions about adenosine and sleep-wake regulation: (1) Is adenosine an endogenous sleep factor? (2) Are there specific brain regions/neuroanatomical targets and receptor subtypes through which adenosine mediates sleepiness? (3) What are the molecular mechanisms by which adenosine may mediate the long-term effects of sleep loss? Data suggest that adenosine is indeed an important endogenous, homeostatic sleep factor, likely mediating the sleepiness that follows prolonged wakefulness. The cholinergic basal forebrain is reviewed in detail as an essential area for mediating the sleep-inducing effects of adenosine by inhibition of wake-promoting neurons via the A1 receptor. The A2A receptor in the subarachnoid space below the rostral forebrain may play a role in the prostaglandin D2-mediated somnogenic effects of adenosine. Recent evidence indicates that a cascade of signal transduction induced by basal forebrain adenosine A1 receptor activation in cholinergic neurons leads to increased transcription of the A1 receptor; this may play a role in mediating the longer-term effects of sleep deprivation, often called sleep debt.


Assuntos
Adenosina/fisiologia , Sono/fisiologia , Vigília/fisiologia , Adenosina/metabolismo , Animais , Sistema Nervoso Central/fisiologia , Humanos , Receptores Purinérgicos P1/efeitos dos fármacos , Receptores Purinérgicos P1/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA