Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Curr Opin Neurobiol ; 44: 159-166, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28538168

RESUMO

The diverse cell-types of the basal forebrain control sleep-wake states, cortical activity and reward processing. Large, slow-firing, cholinergic neurons suppress cortical delta activity and promote cortical plasticity in response to reinforcers. Large, fast-firing, cortically-projecting GABAergic neurons promote wakefulness and fast cortical activity. In particular, parvalbumin/GABAergic neurons promote neocortical gamma band activity. Conversely, excitation of slower-firing somatostatin/GABAergic neurons promotes sleep through inhibition of cortically-projecting neurons. Activation of glutamatergic neurons promotes wakefulness, likely by exciting other cortically-projecting neurons. Similarly, cholinergic neurons indirectly promote wakefulness by excitation of wake-promoting, cortically-projecting GABAergic neurons and/or inhibition of sleep-promoting somatostatin/GABAergic neurons. Both glia and neurons increase the levels of adenosine during prolonged wakefulness. Adenosine presynaptically inhibits glutamatergic inputs to wake-promoting cholinergic and GABAergic/parvalbumin neurons, promoting sleep.


Assuntos
Prosencéfalo Basal/citologia , Prosencéfalo Basal/fisiologia , Neurônios/fisiologia , Animais , Humanos , Neurônios/citologia , Sono/fisiologia , Vigília/fisiologia
2.
J Sleep Res ; 26(3): 377-385, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28397310

RESUMO

Sleep has been postulated to promote brain energy restoration. It is as yet unknown if increasing the energy availability within the brain reduces sleep need. The guanidine amino acid creatine (Cr) is a well-known energy booster in cellular energy homeostasis. Oral Cr-monohydrate supplementation (CS) increases exercise performance and has been shown to have substantial effects on cognitive performance, neuroprotection and circadian rhythms. The effect of CS on cellular high-energy molecules and sleep-wake behaviour is unclear. Here, we examined the sleep-wake behaviour and brain energy metabolism before and after 4-week-long oral administration of CS in the rat. CS decreased total sleep time and non-rapid eye movement (NREM) sleep significantly during the light (inactive) but not during the dark (active) period. NREM sleep and NREM delta activity were decreased significantly in CS rats after 6 h of sleep deprivation. Biochemical analysis of brain energy metabolites showed a tendency to increase in phosphocreatine after CS, while cellular adenosine triphosphate (ATP) level decreased. Microdialysis analysis showed that the sleep deprivation-induced increase in extracellular adenosine was attenuated after CS. These results suggest that CS reduces sleep need and homeostatic sleep pressure in rats, thereby indicating its potential in the treatment of sleep-related disorders.


Assuntos
Creatina/farmacologia , Homeostase/efeitos dos fármacos , Sono/efeitos dos fármacos , Sono/fisiologia , Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Creatina/administração & dosagem , Eletroencefalografia , Metabolismo Energético/efeitos dos fármacos , Masculino , Microdiálise , Fosfocreatina/metabolismo , Ratos , Ratos Sprague-Dawley , Privação do Sono/tratamento farmacológico , Privação do Sono/metabolismo , Sono REM/efeitos dos fármacos , Sono REM/fisiologia
3.
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
4.
Eur J Neurosci ; 41(2): 182-95, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25369989

RESUMO

The tight coordination of biochemical and electrophysiological mechanisms underlies the homeostatic sleep pressure (HSP) produced by sleep deprivation (SD). We have reported that during SD the levels of inducible nitric oxide synthase (iNOS), extracellular nitric oxide (NO), adenosine [AD]ex , lactate [Lac]ex and pyruvate [Pyr]ex increase in the basal forebrain (BF). However, it is not clear whether all of them contribute to HSP leading to increased electroencephalogram (EEG) delta activity during non-rapid eye movement (NREM) recovery sleep (RS) following SD. Previously, we showed that NREM delta increase evident during RS depends on the presence of BF cholinergic (ChBF) neurons. Here, we investigated the role of ChBF cells in coordination of biochemical and EEG changes seen during SD and RS in the rat. Increases in low-theta power (5-7 Hz), but not high-theta (7-9 Hz), during SD correlated with the increase in NREM delta power during RS, and with the changes in nitrate/nitrite [NOx ]ex and [AD]ex . Lesions of ChBF cells using IgG 192-saporin prevented increases in [NOx ]ex , [AD]ex and low-theta activity, during SD, but did not prevent increases in [Lac]ex and [Pyr]ex . Infusion of NO donor DETA NONOate into the saporin-treated BF failed to increase NREM RS and delta power, suggesting ChBF cells are important for mediating NO homeostatic effects. Finally, SD-induced iNOS was mostly expressed in ChBF cells, and the intensity of iNOS induction correlated with the increase in low-theta activity. Together, our data indicate ChBF cells are important in regulating the biochemical and EEG mechanisms that contribute to HSP.


Assuntos
Prosencéfalo Basal/fisiologia , Neurônios Colinérgicos/fisiologia , Homeostase/fisiologia , Sono/fisiologia , Adenosina/metabolismo , Animais , Anticorpos Monoclonais , Prosencéfalo Basal/efeitos dos fármacos , Prosencéfalo Basal/fisiopatologia , Neurônios Colinérgicos/efeitos dos fármacos , Ritmo Delta/efeitos dos fármacos , Ritmo Delta/fisiologia , Homeostase/efeitos dos fármacos , Ácido Láctico/metabolismo , Masculino , Nitratos/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Nitritos/metabolismo , Compostos Nitrosos/farmacologia , Ácido Pirúvico/metabolismo , Ratos Wistar , Proteínas Inativadoras de Ribossomos Tipo 1 , Saporinas , Sono/efeitos dos fármacos , Privação do Sono/fisiopatologia , Ritmo Teta/efeitos dos fármacos , Ritmo Teta/fisiologia
5.
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
6.
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
7.
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
9.
Brain Imaging Behav ; 5(4): 252-61, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21728040

RESUMO

The current report used structural magnetic resonance imaging (MRI) to objectively measure olfactory bulb volume and olfactory sulcal depth in patients diagnosed with chronic schizophrenia and healthy controls. Additional measures were obtained to assess olfactory function. The olfactory bulb and sulcus were manually traced on structural 3T MRIs for 25 right-handed male patients diagnosed with chronic schizophrenia and 25 matched male healthy controls. A sub-set of subjects received the University of Pennsylvania Smell Identification Test (UPSIT). Olfactory bulb volume was significantly decreased in patients with schizophrenia compared to healthy controls, as was their performance on the UPSIT. Additionally, a positive correlation was seen in patients between right bulb volume and UPSIT scores. Overall, our findings support earlier research studies showing morphometric and functional changes in the olfactory system in patients with schizophrenia.


Assuntos
Bulbo Olfatório/patologia , Condutos Olfatórios/patologia , Esquizofrenia/patologia , Adulto , Antipsicóticos/uso terapêutico , Manual Diagnóstico e Estatístico de Transtornos Mentais , Lateralidade Funcional/fisiologia , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Memória/fisiologia , Pessoa de Meia-Idade , Testes Neuropsicológicos , Transtornos do Olfato/diagnóstico , Transtornos do Olfato/fisiopatologia , Córtex Pré-Frontal/patologia , Desempenho Psicomotor/fisiologia , Esquizofrenia/tratamento farmacológico , Psicologia do Esquizofrênico , Olfato/fisiologia , Fumar/psicologia , Fatores Socioeconômicos , Lobo Temporal/patologia , Teste de Sequência Alfanumérica , Adulto Jovem
10.
J Neurochem ; 116(2): 260-72, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21062286

RESUMO

Both adenosine and nitric oxide (NO) are known for their role in sleep homeostasis, with the basal forebrain (BF) wakefulness center as an important site of action. Previously, we reported a cascade of homeostatic events, wherein sleep deprivation (SD) induces the production of inducible nitric oxide synthase (iNOS)-dependent NO in BF, leading to enhanced release of extracellular adenosine. In turn, increased BF adenosine leads to enhanced sleep intensity, as measured by increased non-rapid eye movement sleep EEG delta activity. However, the presence and time course of similar events in cortex has not been studied, although a frontal cortical role for the increase in non-rapid eye movement recovery sleep EEG delta power is known. Accordingly, we performed simultaneous hourly microdialysis sample collection from BF and frontal cortex (FC) during 11 h SD. We observed that both areas showed sequential increases in iNOS and NO, followed by increases in adenosine. BF increases began at 1 h SD, whereas FC increases began at 5 h SD. iNOS and Fos-double labeling indicated that iNOS induction occurred in BF and FC wake-active neurons. These data support the role of BF adenosine and NO in sleep homeostasis and indicate the temporal and spatial sequence of sleep homeostatic cascade for NO and adenosine.


Assuntos
Adenosina/metabolismo , Córtex Cerebral/metabolismo , Homeostase/fisiologia , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico/fisiologia , Prosencéfalo/metabolismo , Privação do Sono/metabolismo , Fases do Sono/fisiologia , Adenosina/fisiologia , Animais , Córtex Cerebral/fisiologia , Eletroencefalografia/métodos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/fisiologia , Prosencéfalo/fisiologia , Ratos , Ratos Wistar , Fatores de Tempo
11.
J Neurosci ; 30(26): 9007-16, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20592221

RESUMO

Sleep is one of the most pervasive biological phenomena, but one whose function remains elusive. Although many theories of function, indirect evidence, and even common sense suggest sleep is needed for an increase in brain energy, brain energy levels have not been directly measured with modern technology. We here report that ATP levels, the energy currency of brain cells, show a surge in the initial hours of spontaneous sleep in wake-active but not in sleep-active brain regions of rat. The surge is dependent on sleep but not time of day, since preventing sleep by gentle handling of rats for 3 or 6 h also prevents the surge in ATP. A significant positive correlation was observed between the surge in ATP and EEG non-rapid eye movement delta activity (0.5-4.5 Hz) during spontaneous sleep. Inducing sleep and delta activity by adenosine infusion into basal forebrain during the normally active dark period also increases ATP. Together, these observations suggest that the surge in ATP occurs when the neuronal activity is reduced, as occurs during sleep. The levels of phosphorylated AMP-activated protein kinase (P-AMPK), well known for its role in cellular energy sensing and regulation, and ATP show reciprocal changes. P-AMPK levels are lower during the sleep-induced ATP surge than during wake or sleep deprivation. Together, these results suggest that sleep-induced surge in ATP and the decrease in P-AMPK levels set the stage for increased anabolic processes during sleep and provide insight into the molecular events leading to the restorative biosynthetic processes occurring during sleep.


Assuntos
Trifosfato de Adenosina/metabolismo , Encéfalo/fisiologia , Sono/fisiologia , Proteínas Quinases Ativadas por AMP/metabolismo , Adenosina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Creatina/metabolismo , Ritmo Delta , Eletroencefalografia , Masculino , Fosfocreatina/metabolismo , Fosforilação , Fotoperíodo , Ratos , Ratos Sprague-Dawley , Privação do Sono/metabolismo , Fases do Sono/efeitos dos fármacos , Fases do Sono/fisiologia , Fatores de Tempo , Vigília/fisiologia
12.
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
13.
Brain Res ; 1258: 53-8, 2009 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-19146833

RESUMO

Adenosine, increasing after sleep deprivation and acting via the A(1) adenosine receptor (A(1)AR), is likely a key factor in the homeostatic control of sleep. This study examines the impact of sleep deprivation on A(1)AR density in different parts of the rat brain with [(3)H]CPFPX autoradiography. Binding of [(3)H]CPFPX was significantly increased in parietal cortex (PAR) (7%), thalamus (11%) and caudate-putamen (9%) after 24 h of sleep deprivation compared to a control group with an undisturbed circadian sleep-wake rhythm. Sleep deprivation of 12 h changed receptor density regionally between -5% and +9% (motor cortex (M1), statistically significant) compared to the circadian control group. These results suggest cerebral A(1)ARs are involved in effects of sleep deprivation and the regulation of sleep. The increase of A(1)AR density could serve the purpose of not only maintaining the responsiveness to increased adenosine levels but also amplifying the effect of sleep deprivation and is in line with a sleep-induced homoeostatic reorganization at the synaptic level.


Assuntos
Encéfalo/metabolismo , Receptor A1 de Adenosina/metabolismo , Privação do Sono/metabolismo , Sono/fisiologia , Antagonistas do Receptor A1 de Adenosina , Análise de Variância , Animais , Autorradiografia , Ritmo Circadiano , Masculino , Ratos , Ratos Sprague-Dawley , Trítio , Xantinas/metabolismo
14.
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
15.
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
16.
Schizophr Res ; 100(1-3): 261-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17884348

RESUMO

As cigarette smoking prevalence rates approach 90% in schizophrenia, an important emerging question is the role of nicotine in the disease-related disturbance in cognition. We therefore tested a total of 38 male cigarette smokers (22 schizophrenia, 16 normal control), matched on nicotine dependence, on the Attention Network Test (ANT) at three nicotine conditions (baseline, 8 h overnight withdrawal, 3 h 21 mg nicotine patch). The results indicated that the groups did not differ in performance on either of three ANT measures (alertness, orienting, and executive) across baseline, patch, and withdrawal conditions. However, in comparison to the controls, the participants with schizophrenia showed faster ANT reaction time (RT) for the nicotine patch in relation to the baseline condition. In comparison to controls, the participants with schizophrenia also showed reduced ANT accuracy at withdrawal but not at patch condition. These results suggest that overall processing speed and accuracy are affected differently by nicotine levels in participants with schizophrenia, with evidence supporting greater impairment from withdrawal and greater improvement from nicotine administration.


Assuntos
Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/diagnóstico , Nicotina/efeitos adversos , Nicotina/uso terapêutico , Esquizofrenia/diagnóstico , Fumar/efeitos adversos , Síndrome de Abstinência a Substâncias/etiologia , Síndrome de Abstinência a Substâncias/psicologia , Administração Cutânea , Adolescente , Adulto , Atenção , Transtornos Cognitivos/psicologia , Comorbidade , Grupos Controle , Humanos , Pessoa de Meia-Idade , Testes Neuropsicológicos/estatística & dados numéricos , Nicotina/administração & dosagem , Desempenho Psicomotor/fisiologia , Tempo de Reação/fisiologia , Esquizofrenia/epidemiologia , Psicologia do Esquizofrênico , Absorção Cutânea , Fumar/epidemiologia , Fumar/psicologia , Inquéritos e Questionários , Tabagismo/complicações , Tabagismo/psicologia
17.
Neuroreport ; 18(18): 1895-9, 2007 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-18007182

RESUMO

Sleep deprivation increases the levels of extracellular adenosine and A1 receptor (A1R)mRNA in the cholinergic zone of the basal forebrain, a region involved in sleep homeostasis. To evaluate homeostatic control mechanisms, we examined the sleep deprivation-induced changes in the A1R density in rodent brain using [H]CPFPX receptor autoradiography. We also examined the role of nuclear factor-kappaB (NF-kappaB) in transcriptional upregulation of A1R mRNA by use of the inhibitor peptide SN50 to inhibit nuclear translocation of NF-kappaB. We found a significant increase in cholinergic basal forebrain A1R density following 24 h of sleep deprivation and evidence that the upregulation of A1R is mediated by NF-kappaB. The A1R increase may be important in sleep homeostasis, since the increase in A1R density would increase the inhibitory effect of given level of adenosine, thus increasing the gain of the homeostat.


Assuntos
Núcleo Basal de Meynert/metabolismo , Fibras Colinérgicas/metabolismo , Homeostase/fisiologia , Receptor A1 de Adenosina/metabolismo , Privação do Sono/metabolismo , Sono/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Adenosina/metabolismo , Animais , Autorradiografia , Masculino , NF-kappa B/metabolismo , Peptídeos/farmacologia , RNA Mensageiro/metabolismo , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptor A1 de Adenosina/genética , Privação do Sono/fisiopatologia , Ativação Transcricional/fisiologia , Regulação para Cima/fisiologia
18.
Sleep Med ; 8(4): 302-30, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17468046

RESUMO

This paper presents an overview of the current knowledge of the neurophysiology and cellular pharmacology of sleep mechanisms. It is written from the perspective that recent years have seen a remarkable development of knowledge about sleep mechanisms, due to the capability of current cellular neurophysiological, pharmacological and molecular techniques to provide focused, detailed, and replicable studies that have enriched and informed the knowledge of sleep phenomenology and pathology derived from electroencephalographic (EEG) analysis. This chapter has a cellular and neurophysiological/neuropharmacological focus, with an emphasis on rapid eye movement (REM) sleep mechanisms and non-REM (NREM) sleep phenomena attributable to adenosine. The survey of neuronal and neurotransmitter-related brainstem mechanisms of REM includes monoamines, acetylcholine, the reticular formation, a new emphasis on GABAergic mechanisms and a discussion of the role of orexin/hypcretin in diurnal consolidation of REM sleep. The focus of the NREM sleep discussion is on the basal forebrain and adenosine as a mediator of homeostatic control. Control is through basal forebrain extracellular adenosine accumulation during wakefulness and inhibition of wakefulness-active neurons. Over longer periods of sleep loss, there is a second mechanism of homeostatic control through transcriptional modification. Adenosine acting at the A1 receptor produces an up-regulation of A1 receptors, which increases inhibition for a given level of adenosine, effectively increasing the gain of the sleep homeostat. This second mechanism likely occurs in widespread cortical areas as well as in the basal forebrain. Finally, the results of a new series of experimental paradigms in rodents to measure the neurocognitive effects of sleep loss and sleep interruption (modeling sleep apnea) provide animal model data congruent with those in humans.


Assuntos
Sono REM/fisiologia , Acetilcolina/metabolismo , Adenosina/metabolismo , Tronco Encefálico/metabolismo , Fibras Colinérgicas/metabolismo , Eletroencefalografia , Homeostase/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neuropeptídeos/metabolismo , Orexinas , Núcleos da Rafe/metabolismo , Formação Reticular/metabolismo , Fases do Sono/fisiologia , Tálamo/metabolismo , Ácido gama-Aminobutírico/metabolismo
19.
J Neurochem ; 100(5): 1351-63, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17316404

RESUMO

In our investigations related to the homeostatic sleep factor adenosine (AD), we previously demonstrated that the DNA-binding activity of the transcription factor NF-kappaB in rat cholinergic basal forebrain increased following 3 h of sleep deprivation (SD). However, the neurotransmitter nature of the cells and the SD-induced stimuli responsible for NF-kappaB activation were not defined. In this report, we demonstrate, using double labeling immunohistochemistry, that nuclear translocation of NF-kappaB occurs almost exclusively in the cholinergic neurons of the basal forebrain following 3 h of SD. Furthermore, cholinergic basal forebrain microinjection of AD (25 nmol/L) or the A(1) receptor agonist N(6)-cyclo-hexyladenosine (100 nmol/L) induced nuclear translocation of NF-kappaB, thus suggesting that SD-induced increased extracellular concentrations of AD, acting via the A(1) AD receptor, may be responsible for the nuclear translocation of NF-kappaB in cholinergic neurons. Moreover, blocking the nuclear translocation of NF-kappaB by injection of inhibitor peptide, SN50, immediately prior to 6 h SD significantly reduced delta activity (1-4 Hz) during the first two hours of recovery sleep. Together, these data suggest a role in sleep homeostasis for the SD-induced activation of NF-kappaB in cholinergic basal forebrain, and that transcription factor NF-kappaB may code for factor(s) that play a role in sleep homeostasis.


Assuntos
Adenosina/fisiologia , Núcleo Celular/metabolismo , Colina O-Acetiltransferase/metabolismo , NF-kappa B/metabolismo , Prosencéfalo/metabolismo , Privação do Sono , Transporte Ativo do Núcleo Celular , Adenosina/farmacologia , Agonistas do Receptor A1 de Adenosina , Animais , Imuno-Histoquímica , Masculino , Microinjeções , Peptídeos/farmacologia , Ratos , Ratos Long-Evans , Receptor A1 de Adenosina/fisiologia , Transdução de Sinais , Sono/fisiologia , Vigília
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA