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.
BMC Biol ; 19(1): 65, 2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33823872

RESUMO

BACKGROUND: Homeostatic regulation of sleep is reflected in the maintenance of a daily balance between sleep and wakefulness. Although numerous internal and external factors can influence sleep, it is unclear whether and to what extent the process that keeps track of time spent awake is determined by the content of the waking experience. We hypothesised that alterations in environmental conditions may elicit different types of wakefulness, which will in turn influence both the capacity to sustain continuous wakefulness as well as the rates of accumulating sleep pressure. To address this, we compared the effects of repetitive behaviours such as voluntary wheel running or performing a simple touchscreen task, with wakefulness dominated by novel object exploration, on sleep timing and EEG slow-wave activity (SWA) during subsequent NREM sleep. RESULTS: We find that voluntary wheel running is associated with higher wake EEG theta-frequency activity and results in longer wake episodes, as compared with exploratory behaviour; yet, it does not lead to higher levels of EEG SWA during subsequent NREM sleep in either the frontal or occipital derivation. Furthermore, engagement in a touchscreen task, motivated by food reward, results in lower SWA during subsequent NREM sleep in both derivations, as compared to exploratory wakefulness, even though the total duration of wakefulness is similar. CONCLUSION: Overall, our study suggests that sleep-wake behaviour is highly flexible within an individual and that the homeostatic processes that keep track of time spent awake are sensitive to the nature of the waking experience. We therefore conclude that sleep dynamics are determined, to a large degree, by the interaction between the organism and the environment.


Assuntos
Comportamento Exploratório , Camundongos/fisiologia , Atividade Motora , Corrida , Sono/fisiologia , Vigília , Animais , Masculino , Camundongos Endogâmicos C57BL , Sono de Ondas Lentas/fisiologia
2.
Biochem Pharmacol ; 191: 114515, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33713641

RESUMO

GABA-ergic neurotransmission plays a key role in sleep regulatory mechanisms and in brain oscillations during sleep. Benzodiazepines such as diazepam are known to induce sedation and promote sleep, however, EEG spectral power in slow frequencies is typically reduced after the administration of benzodiazepines or similar compounds. EEG slow waves arise from a synchronous alternation between periods of cortical network activity (ON) and silence (OFF), and represent a sensitive marker of preceding sleep-wake history. Yet it remains unclear how benzodiazepines act on cortical neural activity during sleep. To address this, we obtained chronic recordings of local field potentials and multiunit activity (MUA) from deep cortical layers of the primary motor cortex in freely behaving mice after diazepam injection. We found that the amplitude of individual LFP slow waves was significantly reduced after diazepam injection and was accompanied by a lower incidence and duration of the corresponding neuronal OFF periods. Further investigation suggested that this is due to a disruption in the synchronisation of cortical neurons. Our data suggest that the state of global sleep and local cortical synchrony can be dissociated, and that the brain state induced by benzodiazepines is qualitatively different from spontaneous physiological sleep.


Assuntos
Diazepam/administração & dosagem , Hipnóticos e Sedativos/administração & dosagem , Córtex Motor/efeitos dos fármacos , Rede Nervosa/efeitos dos fármacos , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , Animais , Estudos Cross-Over , Eletroencefalografia/efeitos dos fármacos , Eletroencefalografia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Motor/fisiologia , Rede Nervosa/fisiologia , Distribuição Aleatória , Sono/fisiologia , Vigília/fisiologia
3.
Sleep ; 41(7)2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29697841

RESUMO

Sleep-wake history, wake behaviors, lighting conditions, and circadian time influence sleep, but neither their relative contribution nor the underlying mechanisms are fully understood. The dynamics of electroencephalogram (EEG) slow-wave activity (SWA) during sleep can be described using the two-process model, whereby the parameters of homeostatic Process S are estimated using empirical EEG SWA (0.5-4 Hz) in nonrapid eye movement sleep (NREMS), and the 24 hr distribution of vigilance states. We hypothesized that the influence of extrinsic factors on sleep homeostasis, such as the time of day or wake behavior, would manifest in systematic deviations between empirical SWA and model predictions. To test this hypothesis, we performed parameter estimation and tested model predictions using NREMS SWA derived from continuous EEG recordings from the frontal and occipital cortex in mice. The animals showed prolonged wake periods, followed by consolidated sleep, both during the dark and light phases, and wakefulness primarily consisted of voluntary wheel running, learning a new motor skill or novel object exploration. Simulated SWA matched empirical levels well across conditions, and neither waking experience nor time of day had a significant influence on the fit between data and simulation. However, we consistently observed that Process S declined during sleep significantly faster in the frontal than in the occipital area of the neocortex. The striking resilience of the model to specific wake behaviors, lighting conditions, and time of day suggests that intrinsic factors underpinning the dynamics of Process S are robust to extrinsic influences, despite their major role in shaping the overall amount and distribution of vigilance states across 24 hr.


Assuntos
Lobo Frontal/fisiologia , Atividade Motora , Lobo Occipital/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Ritmo Circadiano , Eletroencefalografia , Homeostase , Masculino , Camundongos
4.
J Neurosci ; 38(16): 3911-3928, 2018 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-29581380

RESUMO

Healthy aging is associated with marked effects on sleep, including its daily amount and architecture, as well as the specific EEG oscillations. Neither the neurophysiological underpinnings nor the biological significance of these changes are understood, and crucially the question remains whether aging is associated with reduced sleep need or a diminished capacity to generate sufficient sleep. Here we tested the hypothesis that aging may affect local cortical networks, disrupting the capacity to generate and sustain sleep oscillations, and with it the local homeostatic response to sleep loss. We performed chronic recordings of cortical neural activity and local field potentials from the motor cortex in young and older male C57BL/6J mice, during spontaneous waking and sleep, as well as during sleep after sleep deprivation. In older animals, we observed an increase in the incidence of non-rapid eye movement sleep local field potential slow waves and their associated neuronal silent (OFF) periods, whereas the overall pattern of state-dependent cortical neuronal firing was generally similar between ages. Furthermore, we observed that the response to sleep deprivation at the level of local cortical network activity was not affected by aging. Our data thus suggest that the local cortical neural dynamics and local sleep homeostatic mechanisms, at least in the motor cortex, are not impaired during healthy senescence in mice. This indicates that powerful protective or compensatory mechanisms may exist to maintain neuronal function stable across the life span, counteracting global changes in sleep amount and architecture.SIGNIFICANCE STATEMENT The biological significance of age-dependent changes in sleep is unknown but may reflect either a diminished sleep need or a reduced capacity to generate deep sleep stages. As aging has been linked to profound disruptions in cortical sleep oscillations and because sleep need is reflected in specific patterns of cortical activity, we performed chronic electrophysiological recordings of cortical neural activity during waking, sleep, and after sleep deprivation from young and older mice. We found that all main hallmarks of cortical activity during spontaneous sleep and recovery sleep after sleep deprivation were largely intact in older mice, suggesting that the well-described age-related changes in global sleep are unlikely to arise from a disruption of local network dynamics within the neocortex.


Assuntos
Envelhecimento/fisiologia , Córtex Motor/fisiologia , Fases do Sono , Animais , Excitabilidade Cortical , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Motor/citologia , Córtex Motor/crescimento & desenvolvimento , Neurônios/fisiologia
5.
Neuropsychopharmacology ; 42(6): 1305-1314, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27658486

RESUMO

Trace amine-associated receptor 1 (TAAR1) agonists have been shown to have procognitive, antipsychotic-like, anxiolytic, weight-reducing, glucose-lowering, and wake-promoting activities. We used Taar1 knockout (KO) and overexpressing (OE) mice and TAAR1 agonists to elucidate the role of TAAR1 in sleep/wake. EEG, EMG, body temperature (Tb), and locomotor activity (LMA) were recorded in Taar1 KO, OE, and WT mice. Following a 24 h recording to characterize basal sleep/wake parameters, mice were sleep deprived (SD) for 6 h. In another experiment, mice were given three doses of the TAAR1 partial agonist RO5263397, caffeine, or vehicle p.o. Baseline wakefulness was modestly increased in OE compared with WT mice. Baseline theta (4.5-9 Hz) and low gamma (30-60 Hz) activity was elevated in KO compared with OE mice in NREM and REM sleep. Following SD, both KO and OE mice exhibited a homeostatic sleep rebound. In WT mice, RO5263397 increased waking and reduced NREM and REM sleep, decreased gamma power during wake and NREM, and decreased Tb without affecting LMA; these effects were absent in KO mice and potentiated in OE mice. In contrast, caffeine increased wake time, NREM gamma power, and LMA in all strains compared with vehicle; this effect was attenuated in KO and potentiated in OE mice. TAAR1 overexpression modestly increases wakefulness, whereas TAAR1 partial agonism increases wakefulness and also reduces NREM and also REM sleep. These results indicate a modulatory role for TAAR1 in sleep/wake and cortical activity and suggest TAAR1 as a novel target for wake-promoting therapeutics.


Assuntos
Comportamento Animal/fisiologia , Ondas Encefálicas/fisiologia , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Oxazóis/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Ondas Encefálicas/efeitos dos fármacos , Cafeína/administração & dosagem , Estimulantes do Sistema Nervoso Central/administração & dosagem , Masculino , Camundongos , Camundongos Knockout , Oxazóis/administração & dosagem , Receptores Acoplados a Proteínas G/agonistas , Fases do Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos
6.
eNeuro ; 3(5)2016.
Artigo em Inglês | MEDLINE | ID: mdl-27822504

RESUMO

Unraveling the roles of distinct neuron types is a fundamental challenge to understanding brain function in health and disease. In the amygdala, a brain structure regulating emotional behavior, the diversity of GABAergic neurons has been only partially explored. We report a novel population of GABAergic amygdala neurons expressing high levels of neuronal nitric oxide synthase (nNOS). These cells are predominantly localized along basolateral amygdala (BLA) boundaries. Performing ex vivo patch-clamp recordings from nNOS+ neurons in Nos1-CreER;Ai9 mice, we observed that nNOS+ neurons located along the external capsule display distinctive electrophysiological properties, axonal and dendritic arborization, and connectivity. Examining their c-Fos expression, we found that paracapsular nNOS+ neurons are activated during a period of undisturbed sleep following sleep deprivation, but not during sleep deprivation. Consistently, we found that dorsal raphe serotonin [5-hydroxytryptamine (5-HT)] neurons, which are involved in sleep-wake regulation, innervate nNOS+ neurons. Bath application of 5-HT hyperpolarizes nNOS+ neurons via 5-HT1A receptors. This hyperpolarization produces a reduction in firing rate and, occasionally, a switch from tonic to burst firing mode, thereby contrasting with the classic depolarizing effect of 5-HT on BLA GABAergic cells reported so far. Thus, nNOS+ cells are a distinct cell type of the amygdala that controls the activity of downstream neurons in both amygdaloid and extra-amygdaloid regions in a vigilance state-dependent fashion. Given the strong links among mood, sleep deprivation, and 5-HT, the recruitment of paracapsular nNOS+ neurons following high sleep pressure may represent an important mechanism in emotional regulation.


Assuntos
Tonsila do Cerebelo/metabolismo , Neurônios GABAérgicos/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Serotonina/metabolismo , Sono/fisiologia , Tonsila do Cerebelo/citologia , Animais , Núcleo Dorsal da Rafe/citologia , Núcleo Dorsal da Rafe/metabolismo , Neurônios GABAérgicos/citologia , Masculino , Potenciais da Membrana/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Privação do Sono/metabolismo , Privação do Sono/patologia , Sinapses/metabolismo , Técnicas de Cultura de Tecidos
7.
Nat Commun ; 7: 13138, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27748455

RESUMO

Prolonged wakefulness is thought to gradually increase 'sleep need' and influence subsequent sleep duration and intensity, but the role of specific waking behaviours remains unclear. Here we report the effect of voluntary wheel running during wakefulness on neuronal activity in the motor and somatosensory cortex in mice. We find that stereotypic wheel running is associated with a substantial reduction in firing rates among a large subpopulation of cortical neurons, especially at high speeds. Wheel running also has longer-term effects on spiking activity across periods of wakefulness. Specifically, cortical firing rates are significantly higher towards the end of a spontaneous prolonged waking period. However, this increase is abolished when wakefulness is dominated by running wheel activity. These findings indicate that wake-related changes in firing rates are determined not only by wake duration, but also by specific waking behaviours.


Assuntos
Atividade Motora/fisiologia , Córtex Motor/fisiologia , Condicionamento Físico Animal/fisiologia , Córtex Somatossensorial/fisiologia , Animais , Eletroencefalografia , Fenômenos Eletrofisiológicos , Masculino , Camundongos Endogâmicos C57BL , Córtex Motor/citologia , Neurônios/fisiologia , Sono/fisiologia , Privação do Sono/fisiopatologia , Córtex Somatossensorial/citologia , Comportamento Estereotipado/fisiologia , Vigília/fisiologia
8.
Sleep ; 39(2): 379-91, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26446107

RESUMO

STUDY OBJECTIVES: Patients with Huntington's disease (HD) show a high prevalence of sleep disorders that typically occur prior to the onset of motoric symptoms and neurodegeneration. Our understanding of the pathophysiological alterations in premanifest HD is limited, hindering the ability to measure disease modification in response to treatment. We used a full-length knock-in HD model to determine early changes in the electroencephalogram (EEG) and sleep that may predict the onset and progression of the disease. METHODS: A 10-month longitudinal study was designed to determine the effect of the HD mutation on the EEG and sleep/wake changes in heterozygous (HET) and homozygous (HOM) zQ175 mice and wild-type (WT) littermates from 8 to 48 w of age. Mice were instrumented with tethered headmounts to record EEG/electromyography signals. Telemeters were implanted to continuously measure locomotor activity (LMA) and body temperature (Tb). Sleep deprivation (SDep) was performed at 8, 12, 16, 24, 32, and 48 w of age. RESULTS: The HD mutation disrupted the EEG field potential from 8-12 w in an age- and mutant huntington dose-dependent manner, prior to changes in sleep/wake states, LMA, and Tb. Prominent effects of the HD mutation on the EEG included a progressive reduction in low frequency power, a slowing of rapid eye movement peak theta frequency, and the emergence of state-dependent beta/gamma oscillations. There was no effect of genotype on the relative increase in nonrapid eye movement delta power or sleep time in response to SDep. CONCLUSIONS: The expression of the Huntington's disease (HD) mutation results in complex EEG alterations that occur prior to deficits in behavioral measures and are one of the earliest phenotypes uncovered in this mouse model. Despite these EEG changes, homeostatic responses to sleep loss were preserved in HET and HOM zQ175 mice. Greater insight into the localization and response of these EEG alterations to novel therapies may enable early intervention and improve outcomes for patients with HD.


Assuntos
Modelos Animais de Doenças , Progressão da Doença , Eletroencefalografia , Técnicas de Introdução de Genes , Doença de Huntington/genética , Doença de Huntington/fisiopatologia , Proteínas do Tecido Nervoso/genética , Transtornos do Sono-Vigília/complicações , Transtornos do Sono-Vigília/fisiopatologia , Envelhecimento , Animais , Temperatura Corporal , Ondas Encefálicas , Eletromiografia , Genótipo , Humanos , Proteína Huntingtina , Doença de Huntington/complicações , Estudos Longitudinais , Masculino , Camundongos , Atividade Motora , Mutação/genética , Fenótipo , Privação do Sono/fisiopatologia , Sono REM/fisiologia , Vigília/fisiologia
10.
Brain ; 136(Pt 7): 2159-72, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23801738

RESUMO

Deficits in sleep and circadian organization have been identified as common early features in patients with Huntington's disease that correlate with symptom severity and may be instrumental in disease progression. Studies in Huntington's disease gene carriers suggest that alterations in the electroencephalogram may reflect underlying neuronal dysfunction that is present in the premanifest stage. We conducted a longitudinal characterization of sleep/wake and electroencephalographic activity in the R6/2 mouse model of Huntington's disease to determine whether analogous electroencephalographic 'signatures' could be identified early in disease progression. R6/2 and wild-type mice were implanted for electroencephalographic recordings along with telemetry for the continuous recording of activity and body temperature. Diurnal patterns of activity and core body temperature were progressively disrupted in R6/2 mice, with a large reduction in the amplitude of these rhythms apparent by 13 weeks of age. The diurnal variation in sleep/wake states was gradually attenuated as sleep became more fragmented and total sleep time was reduced relative to wild-type mice. These genotypic differences were augmented at 17 weeks and evident across the entire 24-h period. Quantitative electroencephalogram analysis revealed anomalous increases in high beta and gamma activity (25-60 Hz) in all sleep/wake states in R6/2 mice, along with increases in theta activity during both non-rapid eye movement and rapid eye movement sleep and a reduction of delta power in non-rapid eye movement sleep. These dramatic alterations in quantitative electroencephalographic measures were apparent from our earliest recording (9 weeks), before any major differences in diurnal physiology or sleep/wake behaviour occurred. In addition, the homeostatic response to sleep deprivation was greatly attenuated with disease progression. These findings demonstrate the sensitivity of quantitative electroencephalographic analysis to identify early pathophysiological alterations in the R6/2 model of Huntington's disease and suggest longitudinal studies in other preclinical Huntington's disease models are needed to determine the generality of these observations as a potential adjunct in therapeutic development.


Assuntos
Ondas Encefálicas/fisiologia , Ritmo Circadiano/fisiologia , Doença de Huntington/complicações , Fases do Sono/fisiologia , Transtornos do Sono-Vigília/etiologia , Análise de Variância , Animais , Temperatura Corporal/genética , Ondas Encefálicas/genética , Ritmo Circadiano/genética , Modelos Animais de Doenças , Progressão da Doença , Eletroencefalografia , Eletromiografia , Humanos , Proteína Huntingtina , Doença de Huntington/genética , Estudos Longitudinais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/genética , Proteínas do Tecido Nervoso/genética , Fenótipo , Privação do Sono/fisiopatologia , Fases do Sono/genética , Análise Espectral , Repetições de Trinucleotídeos/genética , Vigília/genética
11.
Handb Exp Pharmacol ; (217): 157-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23604479

RESUMO

The sleep/wake cycle is arguably the most familiar output of the circadian system, however, sleep is a complex biological process that arises from multiple brain regions and neurotransmitters, which is regulated by numerous physiological and environmental factors. These include a circadian drive for wakefulness as well as an increase in the requirement for sleep with prolonged waking (the sleep homeostat). In this chapter, we describe the regulation of sleep, with a particular emphasis on the contribution of the circadian system. Since their identification, the role of clock genes in the regulation of sleep has attracted considerable interest, and here, we provide an overview of the interplay between specific elements of the molecular clock with the sleep regulatory system. Finally, we summarise the role of the light environment, melatonin and social cues in the modulation of sleep, with a focus on the role of melanopsin ganglion cells.


Assuntos
Ritmo Circadiano/fisiologia , Sono/fisiologia , Fatores de Transcrição ARNTL/genética , Animais , Proteínas CLOCK/genética , Homeostase , Humanos , Melatonina/fisiologia , Saúde Mental , Opsinas de Bastonetes/fisiologia
12.
Sleep ; 36(3): 325-36, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23449602

RESUMO

STUDY OBJECTIVES: Humans with narcolepsy and orexin/ataxin-3 transgenic (TG) mice exhibit extensive, but incomplete, degeneration of hypo-cretin (Hcrt) neurons. Partial Hcrt cell loss also occurs in Parkinson disease and other neurologic conditions. Whether Hcrt antagonists such as almorexant (ALM) can exert an effect on the Hcrt that remains after Hcrt neurodegeneration has not yet been determined. The current study was designed to evaluate the hypnotic and cataplexy-inducing efficacy of a Hcrt antagonist in an animal model with low Hcrt tone and compare the ALM efficacy profile in the disease model to that produced in wild-type (WT) control animals. DESIGN: Counterbalanced crossover study. SETTING: Home cage. PATIENTS OR PARTICIPANTS: Nine TG mice and 10 WT mice. INTERVENTIONS: ALM (30, 100, 300 mg/kg), vehicle and positive control injections, dark/active phase onset. MEASUREMENTS AND RESULTS: During the 12-h dark period after dosing, ALM exacerbated cataplexy in TG mice and increased nonrapid eye movement sleep with heightened sleep/wake fragmentation in both genotypes. ALM showed greater hypnotic potency in WT mice than in TG mice. The 100 mg/kg dose conferred maximal promotion of cataplexy in TG mice and maximal promotion of REM sleep in WT mice. In TG mice, ALM (30 mg/ kg) paradoxically induced a transient increase in active wakefulness. Core body temperature (Tb) decreased after acute Hcrt receptor blockade, but the reduction in Tb that normally accompanies the wake-to-sleep transition was blunted in TG mice. CONCLUSIONS: These complex dose- and genotype-dependent interactions underscore the importance of effector mechanisms downstream from Hcrt receptors that regulate arousal state. Cataplexy promotion by ALM warrants cautious use of Hcrt antagonists in patient populations with Hcrt neurodegeneration, but may also facilitate the discovery of anticataplectic medications. CITATION: Black SW; Morairty SR; Fisher SP; Chen TM; Warrier DR; Kilduff TS. Almorexant promotes sleep and exacerbates cataplexy in a murine model of narcolepsy. SLEEP 2013;36(3):325-336.


Assuntos
Acetamidas/farmacologia , Cataplexia/induzido quimicamente , Isoquinolinas/farmacologia , Narcolepsia/tratamento farmacológico , Sono/efeitos dos fármacos , Análise de Variância , Animais , Estudos Cross-Over , Modelos Animais de Doenças , Eletroencefalografia/efeitos dos fármacos , Eletromiografia/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neuropeptídeos/efeitos dos fármacos , Orexinas , Vigília/efeitos dos fármacos
13.
Curr Biol ; 22(15): 1397-402, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22771039

RESUMO

The change in irradiance at dawn and dusk provides the primary cue for the entrainment of the mammalian circadian pacemaker. Irradiance detection has been ascribed largely to melanopsin-based phototransduction [1-5]. Here we examine the role of ultraviolet-sensitive (UVS) cones in the modulation of circadian behavior, sleep, and suprachiasmatic nucleus (SCN) electrical activity. UV light exposure leads to phase-shifting responses comparable to those of white light. Moreover, UV light exposure induces sleep in wild-type and melanopsin-deficient (Opn4(-/-)) mice with equal efficacy. Electrical recordings from the SCN of wild-type mice show that UV light elicits irradiance-dependent sustained responses that are similar to those induced by white light, with characteristic fast transient components occurring at the light transitions. These responses are retained in Opn4(-/-) mice and preserved under saturating photopic conditions. The sensitivity of phase-shifting responses to UV light is unaffected by the loss of rods but is severely attenuated by the additional loss of cones. Our data show that UVS cones play an important role in circadian and sleep regulation in mice.


Assuntos
Ritmo Circadiano/efeitos da radiação , Células Fotorreceptoras Retinianas Cones/fisiologia , Opsinas de Bastonetes/fisiologia , Núcleo Supraquiasmático/fisiologia , Raios Ultravioleta , Animais , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Sono/efeitos da radiação
14.
J Biol Rhythms ; 27(1): 48-58, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22306973

RESUMO

Sleep is a fundamental biological rhythm involving the interaction of numerous brain structures and diverse neurotransmitter systems. The primary measures used to define sleep are the electroencephalogram (EEG) and electromyogram (EMG). However, EEG-based methods are often unsuitable for use in high-throughput screens as they are time-intensive and involve invasive surgery. As such, the dissection of sleep mechanisms and the discovery of novel drugs that modulate sleep would benefit greatly from further development of rapid behavioral assays to assess sleep in animal models. Here is described an automated noninvasive approach to evaluate sleep duration, latency, and fragmentation using video tracking of mice in their home cage. This approach provides a high correlation with EEG/EMG measures under both baseline conditions and following administration of pharmacological agents. Moreover, the dose-dependent effects of sedatives, stimulants, and light can be readily detected. This approach is robust yet relatively inexpensive to implement and can be easily incorporated into ongoing screening programs to provide a powerful first-pass screen for assessing sleep and allied behaviors.


Assuntos
Comportamento Animal , Monitorização Fisiológica/veterinária , Sono/fisiologia , Gravação em Vídeo/métodos , Vigília/fisiologia , Animais , Automação Laboratorial , Cafeína/farmacologia , Eletroencefalografia , Eletromiografia , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/farmacologia , Sono/efeitos dos fármacos , Sono/efeitos da radiação , Vigília/efeitos dos fármacos , Zolpidem
15.
Curr Biol ; 22(4): 314-9, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22264613

RESUMO

Sleep and circadian rhythm disruption has been widely observed in neuropsychiatric disorders including schizophrenia [1] and often precedes related symptoms [2]. However, mechanistic basis for this association remains unknown. Therefore, we investigated the circadian phenotype of blind-drunk (Bdr), a mouse model of synaptosomal-associated protein (Snap)-25 exocytotic disruption that displays schizophrenic endophenotypes modulated by prenatal factors and reversible by antipsychotic treatment [3, 4]. Notably, SNAP-25 has been implicated in schizophrenia from genetic [5-8], pathological [9-13], and functional studies [14-16]. We show here that the rest and activity rhythms of Bdr mice are phase advanced and fragmented under a light/dark cycle, reminiscent of the disturbed sleep patterns observed in schizophrenia. Retinal inputs appear normal in mutants, and clock gene rhythms within the suprachiasmatic nucleus (SCN) are normally phased both in vitro and in vivo. However, the 24 hr rhythms of arginine vasopressin within the SCN and plasma corticosterone are both markedly phase advanced in Bdr mice. We suggest that the Bdr circadian phenotype arises from a disruption of synaptic connectivity within the SCN that alters critical output signals. Collectively, our data provide a link between disruption of circadian activity cycles and synaptic dysfunction in a model of neuropsychiatric disease.


Assuntos
Arginina Vasopressina/metabolismo , Ritmo Circadiano , Corticosterona/metabolismo , Atividade Motora , Esquizofrenia/metabolismo , Núcleo Supraquiasmático/química , Adulto , Animais , Corticosterona/sangue , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Análise em Microsséries , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Esquizofrenia/genética , Sono , Proteína 25 Associada a Sinaptossoma/genética , Proteína 25 Associada a Sinaptossoma/metabolismo , Gravação de Videoteipe
16.
Sleep ; 33(6): 833-40, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20550025

RESUMO

STUDY OBJECTIVES: Though melatonin and melatonin receptor agonists are in clinical use and under development for treating insomnia, the role of endogenous melatonin in the regulation of the sleep-wake cycle remains uncertain. Some clinical case reports suggest that reduced nocturnal melatonin secretion is linked to sleep disruption, but pineal-gland removal in experimental animals has given variable results. DESIGN: The present study examined the effects of pinealectomy on the diurnal sleep-wake cycle of rats implanted with a radiotransmitter to allow continuous measurement of cortical electroencephalogram, electromyogram, and core temperature (Tc) without restraint in their home cages. MEASUREMENTS AND RESULTS: Tc was slightly (0.2 degrees C) but significantly lower after pineal removal. The total amount and diurnal distribution of locomotor activity, wake, non-rapid eye movement (NREM) sleep, and rapid eye movement (REM) sleep were unaltered in pinealectomized rats compared to sham-operated controls. Sleep consolidation measured by determining wake, NREM sleep, and REM sleep bout length and frequency was also unchanged. The EEG power spectrum during NREM sleep was unchanged, but a significant decrease in theta power (5-8 Hz) during REM sleep episodes was found. CONCLUSIONS: Our data provide no evidence that endogenous circulating melatonin plays a role in regulating the sleep-wake cycle in rats. However, because cortical theta oscillations are generated in the CA1-3 layer of the hippocampus, neurons known to express melatonin receptors, this suggests that a lack of melatonin following pineal removal influences the function of these neurons and is consistent with previous work suggesting that endogenous melatonin is an important regulator of hippocampal physiology.


Assuntos
Depressores do Sistema Nervoso Central/farmacologia , Melatonina/farmacologia , Sono/efeitos dos fármacos , Vigília/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Temperatura Corporal/efeitos dos fármacos , Depressores do Sistema Nervoso Central/urina , Creatinina/urina , Eletroencefalografia/efeitos dos fármacos , Eletromiografia/efeitos dos fármacos , Masculino , Melatonina/urina , Glândula Pineal/cirurgia , Radioimunoensaio/métodos , Ratos , Ratos Sprague-Dawley , Fases do Sono/efeitos dos fármacos
17.
Neurosci Lett ; 457(2): 93-6, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19429170

RESUMO

Several novel melatonin receptor agonists, in addition to various formulations of melatonin itself, are either available or in development for the treatment of insomnia. Melatonin is thought to exert its effects principally through two high affinity, G-protein coupled receptors, MT1 and MT2, though it is not known which subtype is responsible for the sleep-promoting action. The present study used radiotelemetry to record EEG and EMG in un-restrained freely moving rats to monitor the sleep-wake behaviour and examined the acute sleep-promoting activity of an MT2 receptor subtype selective melatonin analog, IIK7. IIK7 is a full agonist at the MT2 receptor subtype but a partial agonist at the MT1 receptor and has approximately 90-fold higher affinity for MT2 than MT1. Like melatonin, IIK7 (10mg/kg i.p.) significantly reduced NREM sleep onset latency and transiently increased the time spent in NREM sleep, but did not alter REM sleep latency or the amount of REM sleep. An analysis of the EEG power spectrum showed no change in delta (1-4 Hz) or theta activity (5-8 Hz) following IIK7 administration. Core body temperature was slightly decreased ( approximately 0.3 degrees C) by IIK7 compared to vehicle-treated rats. The acute and transient changes in the sleep-wake cycle mimic the changes seen with melatonin and suggest that its sleep-promoting activity is mediated by activation of the MT2 receptor subtype.


Assuntos
Encéfalo/efeitos dos fármacos , Isoindóis/farmacologia , Receptor MT2 de Melatonina/agonistas , Sono/efeitos dos fármacos , Animais , Depressores do Sistema Nervoso Central/farmacologia , Eletroencefalografia , Eletromiografia , Masculino , Melatonina/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor MT2 de Melatonina/efeitos dos fármacos
18.
J Pineal Res ; 45(2): 125-32, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18298466

RESUMO

Insomnia, which is severe enough to warrant treatment, occurs in approximately 10% of the general population. It is associated with a range of adverse consequences for human health, economic productivity and quality of life. In animal and human studies, administration of melatonin has been reported to promote sleep, although there has been controversy regarding its effectiveness. The present study used a chronically implanted radiotelemetry transmitter to record electroencephalogram (EEG) and electromyogram (EMG) to enable discrimination of wake (W), nonrapid eye movement (NREM) sleep and rapid eye movement (REM) sleep in un-restrained rats. The acute action of melatonin and ramelteon, a melatonin agonist recently approved for long-term treatment of insomnia in the USA, was examined. Radioligand binding assays on recombinant human MT(1)/MT(2) receptors showed that both the melatonin and ramelteon were both high affinity, nonsubtype selective ligands. Both compounds acted as potent full agonists on a cellular model of melatonin action, the pigment aggregation response in Xenopus laevis melanophores. Both melatonin and ramelteon (10 mg/kg, i/p), administered close to the mid-point of the dark phase of the L:D cycle, significantly reduced NREM sleep latency (time from injection to the appearance of NREM sleep). Both the drugs also produced a short-lasting increase in NREM sleep duration, but the NREM power spectrum was unaltered. Neither drug altered REM latency, REM sleep duration nor power spectrum during REM sleep. In conclusion, ramelteon administration, like melatonin, exerted an acute, short-lasting sleep-promoting effect in the rat, the model most commonly used to evaluate the activity of novel hypnotic drugs.


Assuntos
Indenos/farmacologia , Receptor MT1 de Melatonina/agonistas , Receptor MT2 de Melatonina/agonistas , Animais , Eletroencefalografia , Humanos , Indenos/química , Melatonina/química , Melatonina/farmacologia , Camundongos , Estrutura Molecular , Células NIH 3T3 , Ratos , Sono/efeitos dos fármacos , Sono/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...