Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3661, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38688901

RESUMO

Optochemistry, an emerging pharmacologic approach in which light is used to selectively activate or deactivate molecules, has the potential to alleviate symptoms, cure diseases, and improve quality of life while preventing uncontrolled drug effects. The development of in-vivo applications for optochemistry to render brain cells photoresponsive without relying on genetic engineering has been progressing slowly. The nucleus accumbens (NAc) is a region for the regulation of slow-wave sleep (SWS) through the integration of motivational stimuli. Adenosine emerges as a promising candidate molecule for activating indirect pathway neurons of the NAc expressing adenosine A2A receptors (A2ARs) to induce SWS. Here, we developed a brain-permeable positive allosteric modulator of A2ARs (A2AR PAM) that can be rapidly photoactivated with visible light (λ > 400 nm) and used it optoallosterically to induce SWS in the NAc of freely behaving male mice by increasing the activity of extracellular adenosine derived from astrocytic and neuronal activity.


Assuntos
Adenosina , Núcleo Accumbens , Receptor A2A de Adenosina , Sono de Ondas Lentas , Animais , Núcleo Accumbens/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Masculino , Receptor A2A de Adenosina/metabolismo , Receptor A2A de Adenosina/genética , Camundongos , Adenosina/metabolismo , Adenosina/farmacologia , Regulação Alostérica , Sono de Ondas Lentas/fisiologia , Sono de Ondas Lentas/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/efeitos dos fármacos , Luz , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Humanos , Agonistas do Receptor A2 de Adenosina/farmacologia
2.
Brain Struct Funct ; 226(6): 1755-1778, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33997911

RESUMO

The basal forebrain (BF) is involved in arousal, attention, and reward processing but the role of individual BF neuronal subtypes is still being uncovered. Glutamatergic neurons are the least well-understood of the three main BF neurotransmitter phenotypes. Here we analyzed the distribution, size, calcium-binding protein content and projections of the major group of BF glutamatergic neurons expressing the vesicular glutamate transporter subtype 2 (vGluT2) and tested the functional effect of activating them. Mice expressing Cre recombinase under the control of the vGluT2 promoter were crossed with a reporter strain expressing the red fluorescent protein, tdTomato, to generate vGluT2-cre-tdTomato mice. Immunohistochemical staining for choline acetyltransferase and a cross with mice expressing green fluorescent protein selectively in GABAergic neurons confirmed that cholinergic, GABAergic and vGluT2+ neurons represent distinct BF subpopulations. Subsets of BF vGluT2+ neurons expressed the calcium-binding proteins calbindin or calretinin, suggesting that multiple subtypes of BF vGluT2+ neurons exist. Anterograde tracing using adeno-associated viral vectors expressing channelrhodopsin2-enhanced yellow fluorescent fusion proteins revealed major projections of BF vGluT2+ neurons to neighboring BF cholinergic and parvalbumin neurons, as well as to extra-BF areas involved in the control of arousal or aversive/rewarding behavior such as the lateral habenula and ventral tegmental area. Optogenetic activation of BF vGluT2+ neurons elicited a striking avoidance of the area where stimulation was given, whereas stimulation of BF parvalbumin or cholinergic neurons did not. Together with previous optogenetic findings suggesting an arousal-promoting role, our findings suggest that BF vGluT2 neurons play a dual role in promoting wakefulness and avoidance behavior.


Assuntos
Prosencéfalo Basal , Animais , Aprendizagem da Esquiva , Prosencéfalo Basal/metabolismo , Colinérgicos , Neurônios Colinérgicos/metabolismo , Ácido Glutâmico , Camundongos , Parvalbuminas/metabolismo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Vigília
3.
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
4.
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
5.
J Neurophysiol ; 117(1): 327-335, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27784808

RESUMO

Sleep homeostasis in rats undergoes significant maturational changes during postweaning development, but the underlying mechanisms of this process are unknown. In the present study we tested the hypothesis that the maturation of sleep is related to the functional emergence of adenosine (AD) signaling in the brain. We assessed postweaning changes in 1) wake-related elevation of extracellular AD in the basal forebrain (BF) and adjacent lateral preoptic area (LPO), and 2) the responsiveness of median preoptic nucleus (MnPO) sleep-active cells to increasing homeostatic sleep drive. We tested the ability of exogenous AD to augment homeostatic responses to sleep deprivation (SD) in newly weaned rats. In groups of postnatal day (P)22 and P30 rats, we collected dialysate from the BF/LPO during baseline (BSL) wake-sleep, SD, and recovery sleep (RS). HPLC analysis of microdialysis samples revealed that SD in P30 rats results in significant increases in AD levels compared with BSL. P22 rats do not exhibit changes in AD levels in response to SD. We recorded neuronal activity in the MnPO during BSL, SD, and RS at P22/P30. MnPO neurons exhibited adult-like increases in waking neuronal discharge across SD on both P22 and P30, but discharge rates during enforced wake were higher on P30 vs. P22. Central administration of AD (1 nmol) during SD on P22 resulted in increased sleep time and EEG slow-wave activity during RS compared with saline control. Collectively, these findings support the hypothesis that functional reorganization of an adenosinergic mechanism of sleep regulation contributes to the maturation of sleep homeostasis. NEW & NOTEWORTHY: Brain mechanisms that regulate the maturation of sleep are understudied. The present study generated first evidence about a potential mechanistic role for adenosine in the maturation of sleep homeostasis. Specifically, we demonstrate that early postweaning development in rats, when homeostatic response to sleep loss become adult like, is characterized by maturational changes in wake-related production/release of adenosine in the brain. Pharmacologically increased adenosine signaling in developing brain facilitates homeostatic responses to sleep deprivation.


Assuntos
Adenosina/metabolismo , Homeostase/fisiologia , Área Pré-Óptica/crescimento & desenvolvimento , Área Pré-Óptica/metabolismo , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Sono/fisiologia , Adenosina/farmacologia , Fatores Etários , Envelhecimento/fisiologia , Análise de Variância , Animais , Animais Recém-Nascidos , Cromatografia Líquida de Alta Pressão , Eletroencefalografia , Eletromiografia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Homeostase/efeitos dos fármacos , Área Pré-Óptica/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Privação do Sono/fisiopatologia , Vigília
6.
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
7.
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
8.
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
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.
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
13.
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
14.
J Neurosci ; 27(9): 2410-5, 2007 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-17329439

RESUMO

It is currently hypothesized that adenosine is involved in the induction of sleep after prolonged wakefulness. This effect is partially reversed by the application of caffeine, which is a nonselective blocker of adenosine receptors. Here, we report that the most abundant and highly concentrated A1 subtype of cerebral adenosine receptors is upregulated after 24 h of sleep deprivation. We used the highly selective A1 adenosine receptor (A1AR) radioligand [18F]CPFPX ([18F]8-cyclopentyl-3-(3-fluoropropyl)-1-propylxanthine) and quantitative positron emission tomography to assess cerebral A1ARs before and after sleep deprivation in 12 healthy volunteers and a control group (n = 10) with regular sleep. In sleep deprived subjects, we found an increase of the apparent equilibrium total distribution volume in a region-specific pattern in all examined brain regions with a maximum increase in the orbitofrontal cortex (15.3%; p = 0.014). There were no changes in the control group with regular sleep. This is the first molecular imaging study that provides in vivo evidence for an A1AR upregulation in cortical and subcortical brain regions after prolonged wakefulness, indicating that A1AR expression is contributing to the homeostatic sleep regulation.


Assuntos
Receptor A1 de Adenosina/metabolismo , Privação do Sono/metabolismo , Adulto , Humanos , Masculino , Tomografia por Emissão de Pósitrons , Privação do Sono/diagnóstico por imagem
15.
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
17.
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
18.
J Neurosci ; 22(17): 7680-6, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12196591

RESUMO

In the cholinergic basal forebrain, we found previously that the extracellular adenosine concentration increase that accompanies sleep deprivation, acting via the A1 receptor, led to activation of the transcription factor nuclear factor-kappaB and to the upregulation of A1 adenosine receptor mRNA. We thus began to examine intracellular signaling mechanisms. We report here that adenosine, acting in a dose-dependent manner and predominantly via A1 receptors, stimulated IP3 receptor-regulated calcium release from intracellular stores. To the best of our knowledge, this calcium signaling pathway effect is a novel action of the G(i)-coupled A1 adenosine receptor in neurons. Moreover, this calcium mobilization was not seen at all in noncholinergic neurons but was present in a large proportion of cholinergic neurons. These data suggest a potential role for a calcium-signaling pathway in adenosine-induced long-term effects of sleep deprivation and a key role for cholinergic neurons in this process.


Assuntos
Adenosina/análogos & derivados , Adenosina/farmacologia , Canais de Cálcio/metabolismo , Cálcio/metabolismo , Fibras Colinérgicas/metabolismo , Neurônios/metabolismo , Prosencéfalo/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato , Líquido Intracelular/metabolismo , Masculino , Neurônios/citologia , Neurônios/efeitos dos fármacos , Prosencéfalo/citologia , Agonistas do Receptor Purinérgico P1 , Ratos , Ratos Long-Evans , Receptores Purinérgicos P1/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA