Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci Methods ; 402: 110030, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38042303

RESUMO

BACKGROUND: A noninvasive method that can accurately quantify sleep before, during, and after sleep disruption (SD) has not been validated in female rats across their estrous cycle. In female rats, we hypothesized that the duration of physical inactivity (PIA) required to predict sleep would 1) change with the differences in baseline sleep between the circadian and estrous cycle phases and 2) predict sleep and the change in sleep (Δsleep) before, during, and after SD independent of circadian and estrous cycle phase. NEW METHODS: EEG, EMG, physical activity and estrous cycle phase were measured in female Sprague-Dawley rats before, during, and after SD. Sleep was determined by two methods [EEG/EMG and a duration of continuous PIA (i.e., PIA criterion)]. Reliability between the methods was tested with a previously validated criterion (40 s). Sensitivity analyses and criterion-related validity analyses for sleep during SD and recovery were conducted across multiple PIA criteria (10 s-120 s). Predictability between the two methods and Δsleep was calculated. RESULTS/COMPARISON WITH EXISTING METHODS: Three criteria (10 s, 20 s, 30 s) predicted baseline sleep independent of circadian and estrous cycle phase. Sleep during SD and recovery were predicted by two criteria (30 s and 10 s). Δsleep between study periods was not reliably predicted by a single PIA criterion. CONCLUSION: PIA predicted sleep independent of estrous cycle phase in female rats. However, the specific criterion was dependent upon the study period (before, during, and after SD) and circadian phase. Thus, prior work validating a PIA criterion in male rodents is not applicable to the female rat.


Assuntos
Ciclo Estral , Sono , Ratos , Masculino , Feminino , Animais , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Ritmo Circadiano
2.
Neurosci Lett ; 792: 136959, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36370954

RESUMO

Sleep disruption (SD) promotes stress which may mediate the effect of SD induced by noise on bodyweight gain and food intake. We determined if the change in bodyweight during SD caused by noise was driven by stress (assessed by corticosterone) and whether the effects of noise on SD, stress and bodyweight were specific to the method of SD or a consequence of SD per se. We isolated stress from SD due to noise by exposing rats to noise during the darkphase to test whether darkphase noise stimulated weight gain, stress and food intake. Male Sprague-Dawley rats slept undisturbed, were exposed to noise during both circadian phases (lightphase vs darkphase) and lightphase gentle handling. Bodyweight, food intake, physical activity, vigilance states, and plasma corticosterone were determined. Darkphase noise did not affect vigilance states. Unlike lightphase noise, darkphase noise and lightphase gentle handling did not stimulate weight gain or food intake. Only gentle handling significantly increased corticosterone levels. Noise during the lightphase increasesed weight gain and food intake by causing SD and these effects were not driven by stress as assessed by corticosterone. These results may have significant implications for developing translational models of insomnia-induced obesity in humans.


Assuntos
Corticosterona , Distúrbios do Início e da Manutenção do Sono , Humanos , Ratos , Animais , Masculino , Privação do Sono , Ratos Sprague-Dawley , Sono , Aumento de Peso , Peso Corporal
3.
Int J Obes (Lond) ; 43(9): 1759-1768, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30568267

RESUMO

BACKGROUND/OBJECTIVES: Inadequate sleep increases obesity and environmental noise contributes to poor sleep. However, women may be more vulnerable to noise and hence more susceptible to sleep disruption-induced weight gain than men. In male rats, exposure to environmental (i.e. ambient) noise disrupts sleep and increases feeding and weight gain. However, the effects of environmental noise on sleep and weight gain in female rats are unknown. Thus, this study was designed to determine whether noise exposure would disturb sleep, increase feeding and weight gain and alter the length of the estrous cycle in female rats. SUBJECTS/METHODS: Female rats (12 weeks old) were exposed to noise for 17d (8 h/d during the light period) to determine the effects of noise on weight gain and food intake. In a separate set of females, estrous cycle phase and length, EEG, EMG, spontaneous physical activity and energy expenditure were recorded continuously for 27d during baseline (control, 9d), noise exposure (8 h/d, 9d) and recovery (9d) from sleep disruption. RESULTS: Noise exposure significantly increased weight gain and food intake compared to females that slept undisturbed. Noise also significantly increased wakefulness, reduced sleep and resulted in rebound sleep during the recovery period. Total energy expenditure was significantly lower during both noise exposure and recovery due to lower energy expenditure during spontaneous physical activity and sleep. Notably, noise did not alter the estrous cycle length. CONCLUSIONS: As previously observed in male rats, noise exposure disrupted sleep and increased weight gain in females but did not alter the length of the estrous cycle. This is the first demonstration of weight gain in female rats during sleep disruption. We conclude that the sleep disruption caused by exposure to environmental noise is a significant tool for determining how sleep loss contributes to obesity in females.


Assuntos
Metabolismo Energético/efeitos da radiação , Ruído/efeitos adversos , Privação do Sono/etiologia , Aumento de Peso/efeitos da radiação , Animais , Feminino , Hiperfagia/etiologia , Ratos , Ratos Sprague-Dawley , Sono/efeitos da radiação
4.
Proc Natl Acad Sci U S A ; 113(21): 6023-8, 2016 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-27140610

RESUMO

Neuropeptide B (NPB) and neuropeptide W (NPW) are endogenous neuropeptide ligands for the G protein-coupled receptors NPBWR1 and NPBWR2. Here we report that the majority of NPW neurons in the mesolimbic region possess tyrosine hydroxylase immunoreactivity, indicating that a small subset of dopaminergic neurons coexpress NPW. These NPW-containing neurons densely and exclusively innervate two limbic system nuclei in adult mouse brain: the lateral bed nucleus of the stria terminalis and the lateral part of the central amygdala nucleus (CeAL). In the CeAL of wild-type mice, restraint stress resulted in an inhibition of cellular activity, but this stress-induced inhibition was attenuated in the CeAL neurons of NPW(-/-) mice. Moreover, the response of NPW(-/-) mice to either formalin-induced pain stimuli or a live rat (i.e., a potential predator) was abnormal only when they were placed in a novel environment: The mice failed to show the normal species-specific self-protective and aversive reactions. In contrast, the behavior of NPW(-/-) mice in a habituated environment was indistinguishable from that of wild-type mice. These results indicate that the NPW/NPBWR1 system could play a critical role in the gating of stressful stimuli during exposure to novel environments.


Assuntos
Aprendizagem da Esquiva , Comportamento Animal , Núcleo Central da Amígdala/metabolismo , Neurônios Dopaminérgicos/metabolismo , Neuropeptídeos/metabolismo , Dor/metabolismo , Animais , Núcleo Central da Amígdala/patologia , Neurônios Dopaminérgicos/patologia , Camundongos , Camundongos Knockout , Neuropeptídeos/genética , Dor/genética , Dor/patologia , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/genética , Receptores de Neuropeptídeos/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
5.
Sleep ; 38(9): 1361-70, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25845696

RESUMO

STUDY OBJECTIVES: The ventrolateral preoptic area (VLPO) and the orexin/hypocretin neuronal system are key regulators of sleep onset, transitions between vigilance states, and energy homeostasis. Reciprocal projections exist between the VLPO and orexin/hypocretin neurons. Although the importance of the VLPO to sleep regulation is clear, it is unknown whether VLPO neurons are involved in energy balance. The purpose of these studies was to determine if the VLPO is a site of action for orexin-A, and which orexin receptor subtype(s) would mediate these effects of orexin-A. We hypothesized that orexin-A in the VLPO modulates behaviors (sleep and wakefulness, feeding, spontaneous physical activity [SPA]) to increase energy expenditure. DESIGN AND MEASUREMENTS: Sleep, wakefulness, SPA, feeding, and energy expenditure were determined after orexin-A microinjection in the VLPO of male Sprague-Dawley rats with unilateral cannulae targeting the VLPO. We also tested whether pretreatment with a dual orexin receptor antagonist (DORA, TCS-1102) or an OX2R antagonist (JNJ-10397049) blocked the effects of orexin-A on the sleep/wake cycle or SPA, respectively. RESULTS: Orexin-A injected into the VLPO significantly increased wakefulness, SPA, and energy expenditure (SPA-induced and total) and reduced NREM sleep and REM sleep with no effect on food intake. Pretreatment with DORA blocked the increase in wakefulness and the reduction in NREM sleep elicited by orexin-A, and the OX2R antagonist reduced SPA stimulated by orexin-A. CONCLUSIONS: These data show the ventrolateral preoptic area is a site of action for orexin-A, which may promote negative energy balance by modulating sleep/wakefulness and stimulating spontaneous physical activity and energy expenditure.


Assuntos
Metabolismo Energético/fisiologia , Orexinas/metabolismo , Área Pré-Óptica/metabolismo , Vigília/fisiologia , Animais , Atenção/efeitos dos fármacos , Atenção/fisiologia , Dioxanos/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Metabolismo Energético/efeitos dos fármacos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/metabolismo , Compostos de Fenilureia/farmacologia , Área Pré-Óptica/citologia , Área Pré-Óptica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sono/efeitos dos fármacos , Sono/fisiologia , Privação do Sono/fisiopatologia , Fatores de Tempo , Vigília/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 110(25): 10288-93, 2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23716651

RESUMO

Sleep is regulated by homeostatic mechanisms, and the low-frequency power in the electroencephalogram (delta power) during non-rapid eye movement sleep reflects homeostatic sleep need. Additionally, sleep is limited by circadian and environmentally influenced arousal. Little is known, however, about the underlying neural substrates for sleep homeostasis and arousal and about the potential link between them. Here, we subjected C57BL/6 mice to 6 h of sleep deprivation using two different methods: gentle handling and continual cage change. Both groups were deprived of sleep to a similar extent (>99%), and, as expected, the delta power increase during recovery sleep was quantitatively similar in both groups. However, in a multiple sleep latency test, the cage change group showed significantly longer sleep latencies than the gentle handling group, indicating that the cage change group had a higher level of arousal despite the similar sleep loss. To investigate the possible biochemical correlates of these behavioral changes, we screened for arousal-related and sleep need-related phosphoprotein markers from the diencephalon. We found that the abundance of highly phosphorylated forms of dynamin 1, a presynaptic neuronal protein, was associated with sleep latency in the multiple sleep latency test. In contrast, the abundance of highly phosphorylated forms of N-myc downstream regulated gene 2, a glial protein, was increased in parallel with delta power. The changes of these protein species disappeared after 2 h of recovery sleep. These results suggest that homeostatic sleep need and arousal can be dissociated behaviorally and biochemically and that phosphorylated N-myc downstream regulated gene 2 and dynamin 1 may serve as markers of homeostatic sleep need and arousal, respectively.


Assuntos
Nível de Alerta/fisiologia , Homeostase/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Comportamento Animal/fisiologia , Biomarcadores/metabolismo , Ritmo Delta , Diencéfalo/metabolismo , Dinaminas/genética , Dinaminas/metabolismo , Eletroencefalografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/metabolismo , Restrição Física , Privação do Sono/fisiopatologia , Estresse Psicológico/fisiopatologia , Eletroforese em Gel Diferencial Bidimensional
7.
Neurobiol Dis ; 48(1): 92-101, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22683290

RESUMO

Brain glucose supplies most of the carbon required for acetyl-coenzyme A (acetyl-CoA) generation (an important step for myelin synthesis) and for neurotransmitter production via further metabolism of acetyl-CoA in the tricarboxylic acid (TCA) cycle. However, it is not known whether reduced brain glucose transporter type I (GLUT-1) activity, the hallmark of the GLUT-1 deficiency (G1D) syndrome, leads to acetyl-CoA, TCA or neurotransmitter depletion. This question is relevant because, in its most common form in man, G1D is associated with cerebral hypomyelination (manifested as microcephaly) and epilepsy, suggestive of acetyl-CoA depletion and neurotransmitter dysfunction, respectively. Yet, brain metabolism in G1D remains underexplored both theoretically and experimentally, partly because computational models of limited brain glucose transport are subordinate to metabolic assumptions and partly because current hemizygous G1D mouse models manifest a mild phenotype not easily amenable to investigation. In contrast, adult antisense G1D mice replicate the human phenotype of spontaneous epilepsy associated with robust thalamocortical electrical oscillations. Additionally, and in consonance with human metabolic imaging observations, thalamus and cerebral cortex display the lowest GLUT-1 expression and glucose uptake in the mutant mouse. This depletion of brain glucose is associated with diminished plasma fatty acids and elevated ketone body levels, and with decreased brain acetyl-CoA and fatty acid contents, consistent with brain ketone body consumption and with stimulation of brain beta-oxidation and/or diminished cerebral lipid synthesis. In contrast with other epilepsies, astrocyte glutamine synthetase expression, cerebral TCA cycle intermediates, amino acid and amine neurotransmitter contents are also intact in G1D. The data suggest that the TCA cycle is preserved in G1D because reduced glycolysis and acetyl-CoA formation can be balanced by enhanced ketone body utilization. These results are incompatible with global cerebral energy failure or with neurotransmitter depletion as responsible for epilepsy in G1D and point to an unknown mechanism by which glycolysis critically regulates cortical excitability.


Assuntos
Encéfalo/metabolismo , Erros Inatos do Metabolismo dos Carboidratos/metabolismo , Epilepsia/metabolismo , Transportador de Glucose Tipo 1/deficiência , Animais , Encéfalo/fisiopatologia , Erros Inatos do Metabolismo dos Carboidratos/fisiopatologia , Modelos Animais de Doenças , Dopamina/metabolismo , Epilepsia/fisiopatologia , Ácidos Graxos/metabolismo , Feminino , Glucose/metabolismo , Masculino , Camundongos , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/metabolismo , Serotonina/metabolismo
8.
J Neurosci ; 32(17): 5880-90, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539849

RESUMO

Adult neurogenesis persists throughout life in restricted brain regions in mammals and is affected by various physiological and pathological conditions. The tumor suppressor gene Pten is involved in adult neurogenesis and is mutated in a subset of autism patients with macrocephaly; however, the link between the role of PTEN in adult neurogenesis and the etiology of autism has not been studied before. Moreover, the role of hippocampus, one of the brain regions where adult neurogenesis occurs, in development of autism is not clear. Here, we show that ablating Pten in adult neural stem cells in the subgranular zone of hippocampal dentate gyrus results in higher proliferation rate and accelerated differentiation of the stem/progenitor cells, leading to depletion of the neural stem cell pool and increased differentiation toward the astrocytic lineage at later stages. Pten-deleted stem/progenitor cells develop into hypertrophied neurons with abnormal polarity. Additionally, Pten mutant mice have macrocephaly and exhibit impairment in social interactions and seizure activity. Our data reveal a novel function for PTEN in adult hippocampal neurogenesis and indicate a role in the pathogenesis of abnormal social behaviors.


Assuntos
Células-Tronco Adultas/fisiologia , Giro Denteado/citologia , Regulação da Expressão Gênica/genética , Neurogênese/genética , Neurônios/fisiologia , PTEN Fosfo-Hidrolase/deficiência , Adaptação Ocular/genética , Células-Tronco Adultas/efeitos dos fármacos , Fatores Etários , Animais , Bromodesoxiuridina/metabolismo , Diferenciação Celular/genética , Movimento Celular/genética , Células Cultivadas , Relação Dose-Resposta a Droga , Eletroencefalografia , Eletromiografia , Comportamento Exploratório/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Proteínas de Filamentos Intermediários/genética , Relações Interpessoais , Ventrículos Laterais/citologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Nestina , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Propanolaminas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
9.
J Neurosci ; 31(17): 6518-26, 2011 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-21525292

RESUMO

Orexin-A and orexin-B are hypothalamic neuropeptides that play critical roles in the maintenance of wakefulness. Intracerebroventricular (ICV) administration of orexin-A has been shown to promote wakefulness and suppress both rapid eye movement (REM) sleep and non-REM (NREM) sleep through the orexin receptor-1 (OX(1)R) and orexin receptor-2 (OX(2)R). Here, we elucidated the differential roles of orexin receptors in the regulation of sleep and wakefulness by comparing the effects of ICV orexin-A administration in wild-type, OX(1)R(-/-), and OX(2)R(-/-) mice. The effects of orexin-A on wakefulness and NREM sleep were significantly attenuated in both knock-out mice as compared with wild-type mice, with substantially larger attenuation in OX(2)R(-/-) mice than in OX(1)R(-/-) mice. These results suggest that although the OX(2)R-mediated pathway has a pivotal role in the promotion of wakefulness, OX(1)R also plays additional roles in promoting arousal. In contrast, suppression of REM sleep by orexin-A administration was slightly and similarly attenuated in both OX(1)R(-/-) and OX(2)R(-/-) mice, suggesting a comparable contribution of the two receptors to REM sleep suppression. Histological studies demonstrated differential distributions of each receptor subtype in distinct neuronal populations with specific neurotransmitter identities in brainstem cholinergic/monoaminergic neurons. In the laterodorsal tegmental and pedunculopontine tegmental nuclei especially, cholinergic neurons exclusively expressed OX(1)R mRNA, but OX(2)R mRNA was expressed mainly in GABAergic putative interneurons. Thus, each orexin receptor subtype plays differential roles in gating NREM and REM sleep through distinct neuronal pathways.


Assuntos
Antígenos de Superfície/fisiologia , Encéfalo/fisiologia , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Neuropeptídeos/fisiologia , Fases do Sono/fisiologia , Vigília/fisiologia , Análise de Variância , Animais , Antígenos de Superfície/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Eletroencefalografia , Eletromiografia , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Injeções Intraventriculares/métodos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Neurológicos , Neuropeptídeos/farmacologia , Neurotransmissores/farmacologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Receptores de Orexina , Orexinas , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/deficiência , Receptores de Neuropeptídeos/genética , Fases do Sono/efeitos dos fármacos , Fases do Sono/genética , Proteínas Vesiculares de Transporte de Acetilcolina/genética , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo , Proteínas Vesiculares de Transporte de Monoamina/genética , Proteínas Vesiculares de Transporte de Monoamina/metabolismo , Vigília/efeitos dos fármacos , Vigília/genética
10.
Sleep Med Rev ; 15(3): 197-207, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21269851

RESUMO

In the decade since the discovery that pathology of the orexin/hypocretin system is causative for the sleep disorder narcolepsy, considerable progress has been made in understanding the functional role of the neuropeptide. Two, apparently separate functions of orexin have emerged as a consensus from studies to date. The first is the effect on vigilance state boundaries, as exemplified by narcolepsy. Thus the absence of orexin severely limits the ability to maintain prolonged periods of wakefulness or sleep and also allows the unregulated appearance of cataplexy as sudden muscle weakness during wakefulness. The second function is that orexin acts as a signaling molecule in transferring information about physiological disequilibrium to the central nervous system. Orexin activates the central arousal and motor systems during such disequilibrium and so may facilitate the necessary response and adaptation to restore equilibrium. A feasible relationship between these two functions is therefore that the maintenance of prolonged and active wakefulness is an integral part of this adaptive process. Furthermore, the limit placed on the onset of sleep by orexin suggests that these adaptive processes then continue during sleep to become integrated into the development of a coping strategy for the longer term.


Assuntos
Encéfalo/fisiopatologia , Homeostase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Narcolepsia/fisiopatologia , Neuropeptídeos/fisiologia , Adaptação Fisiológica/fisiologia , Animais , Nível de Alerta/fisiologia , Cataplexia/fisiopatologia , Dopamina/fisiologia , Metabolismo Energético/fisiologia , Histamina/fisiologia , Humanos , Orexinas , Formação Reticular/fisiopatologia , Transdução de Sinais/fisiologia , Sono/fisiologia , Vigília/fisiologia , Ácido gama-Aminobutírico/fisiologia
11.
Proc Natl Acad Sci U S A ; 107(42): 18155-60, 2010 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-20921407

RESUMO

Sleep and wakefulness are regulated primarily by inhibitory interactions between the hypothalamus and brainstem. The expression of the states of rapid eye movement (REM) sleep and non-REM (NREM) sleep also are correlated with the activity of groups of REM-off and REM-on neurons in the dorsal brainstem. However, the contribution of ventral brainstem nuclei to sleep regulation has been little characterized to date. Here we examined sleep and wakefulness in mice deficient in a homeobox transcription factor, Goosecoid-like (Gscl), which is one of the genes deleted in DiGeorge syndrome or 22q11 deletion syndrome. The expression of Gscl is restricted to the interpeduncular nucleus (IP) in the ventral region of the midbrain-hindbrain transition. The IP has reciprocal connections with several cell groups implicated in sleep/wakefulness regulation. Although Gscl(-/-) mice have apparently normal anatomy and connections of the IP, they exhibited a reduced total time spent in REM sleep and fewer REM sleep episodes. In addition, Gscl(-/-) mice showed reduced theta power during REM sleep and increased arousability during REM sleep. Gscl(-/-) mice also lacked the expression of DiGeorge syndrome critical region 14 (Dgcr14) in the IP. These results indicate that the absence of Gscl and Dgcr14 in the IP results in altered regulation of REM sleep.


Assuntos
Tronco Encefálico/fisiologia , Síndrome de DiGeorge/genética , Proteínas de Homeodomínio/fisiologia , Proteínas Nucleares/fisiologia , Sono REM , Animais , Eletroencefalografia , Eletromiografia , Proteínas de Homeodomínio/genética , Hibridização In Situ , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética
12.
J Neurosci Methods ; 184(1): 71-8, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19646474

RESUMO

Interrupted sleep, fragmented sleep or restricted sleep is a corollary of many psychiatric, neurological and respiratory disorders and also results from disruptive environments such as that of the intensive care unit (ICU). Recent rodent studies have revealed that sleep interruption (SI) can have more significant consequences for cognitive and neurophysiological variables than were expected and may even be equivalent to those of total sleep deprivation. Results from this research are therefore being increasingly recognized for their implications, which may include delayed recovery from critical illness in the ICU. Here we describe in detail a method for interrupting sleep in a murine model, which we had previously adopted to show an increase in mortality after septic insult. Interrupting sleep for 30s every 2 min over 48 h significantly decreased rapid eye movement (REM) and non-rapid eye movement (NREM) sleep. The technique, which is based on using a standard laboratory orbital shaker to oscillate the cage containing the mouse, can easily be adapted to use different parameters for SI. During recovery, mice exhibited a rebound in REM sleep time and an increase in the depth of NREM sleep as measured by delta (1-4 Hz) power in the electroencephalogram. The changes in sleep both during and after SI showed some differences from those previously observed in the rat using the same SI parameters. In conclusion, the mouse may provide a useful alternative model for studying the effects of SI.


Assuntos
Modelos Animais de Doenças , Equipamentos e Provisões Elétricas , Transtornos do Sono-Vigília , Animais , Encéfalo/fisiopatologia , Ritmo Delta , Eletrodos Implantados , Eletroencefalografia , Eletromiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Periodicidade , Fotoperíodo , Sono/fisiologia , Fases do Sono/fisiologia , Transtornos do Sono-Vigília/fisiopatologia , Fatores de Tempo , Vigília/fisiologia
13.
PLoS One ; 4(6): e5808, 2009 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-19529798

RESUMO

Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase that has been implicated in learning, synaptic plasticity, neurotransmission, and numerous neurological disorders. We previously showed that conditional loss of Cdk5 in adult mice enhanced hippocampal learning and plasticity via modulation of calpain-mediated N-methyl-D-aspartic acid receptor (NMDAR) degradation. In the present study, we characterize the enhanced synaptic plasticity and examine the effects of long-term Cdk5 loss on hippocampal excitability in adult mice. Field excitatory post-synaptic potentials (fEPSPs) from the Schaffer collateral CA1 subregion of the hippocampus (SC/CA1) reveal that loss of Cdk5 altered theta burst topography and enhanced post-tetanic potentiation. Since Cdk5 governs NMDAR NR2B subunit levels, we investigated the effects of long-term Cdk5 knockout on hippocampal neuronal excitability by measuring NMDAR-mediated fEPSP magnitudes and population-spike thresholds. Long-term loss of Cdk5 led to increased Mg(2+)-sensitive potentials and a lower threshold for epileptiform activity and seizures. Biochemical analyses were performed to better understand the role of Cdk5 in seizures. Induced-seizures in wild-type animals led to elevated amounts of p25, the Cdk5-activating cofactor. Long-term, but not acute, loss of Cdk5 led to decreased p25 levels, suggesting that Cdk5/p25 may be activated as a homeostatic mechanism to attenuate epileptiform activity. These findings indicate that Cdk5 regulates synaptic plasticity, controls neuronal and behavioral stimulus-induced excitability and may be a novel pharmacological target for cognitive and anticonvulsant therapies.


Assuntos
Quinase 5 Dependente de Ciclina/fisiologia , Regulação da Expressão Gênica , Hipocampo/metabolismo , Doenças do Sistema Nervoso/metabolismo , Animais , Anticonvulsivantes/farmacologia , Comportamento Animal , Quinase 5 Dependente de Ciclina/metabolismo , Eletromiografia/métodos , Eletrofisiologia/métodos , Magnésio/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos
14.
J Trauma ; 66(1): 50-4, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19131805

RESUMO

BACKGROUND: Sleep deprivation is a common problem in the intensive care unit. Animal models have demonstrated that sleep deprivation alone is associated with increased mortality. We have previously shown that septic insult with sleep deprivation results in increased mortality in a murine model. The aging process is known to reduce the restorative phases of sleep. The purpose of this study was to evaluate the effect of age on mortality with sleep deprivation during recovery from septic insult. METHODS: C57BL/6J male mice aged 2 months (young) or 9 months (old) underwent cecal ligation and puncture (CLP). Animals were randomized to receive sleep interruption (SI) for 48 hours or standard recovery (no SI). Sham animals underwent laparotomy and cecal manipulation without puncture. SI was achieved by securing animal housing to an orbital shaker set to repeatedly cycle at 30 rpm over 120 seconds (30 seconds on/90 seconds off). The primary outcome was survival at 5 days post-CLP. Kaplan-Meier survival analysis with log-rank test was used to explore differences in mortality. RESULTS: SI resulted in an increase in time awake for both light and dark cycles (p < 0.001). Mortality after CLP with SI (n = 30) was 57% and mortality after CLP without SI (controls; n = 33) was 24%. SI was associated with a greater than 3-fold increase in mortality after CLP (RR = 3.29; 95% CI, 1.42-7.63). Young mice (n = 28) had a mortality of 31% with CLP alone increasing to 67% with SI (p = 0.03). Old mice (n = 35) had a mortality of 18% with CLP alone increasing to 50% with SI (p = 0.05). There was no difference in survival between young and old mice undergoing SI (p = 0.49). CONCLUSIONS: Sleep deprivation after septic insult increases mortality in both young and old mice. However, sleep deprivation after septic insult does not have a more profound effect on mortality in either age group. These findings suggest that sleep deprivation experienced in the intensive care unit setting during recovery from critical illness may increase mortality. This effect appears independent of increased age. Further studies evaluating extremes of age are warranted.


Assuntos
Sepse/mortalidade , Privação do Sono/mortalidade , Análise de Variância , Animais , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Distribuição Aleatória
15.
Brain Res ; 1168: 112-23, 2007 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-17706614

RESUMO

Conditional deletion of Pten (phosphatase and tensin homolog on chromosome ten) in differentiated cortical and hippocampal neurons in the mouse results in seizures, macrocephaly, social interaction deficits and anxiety, reminiscent of human autism spectrum disorder. Here we extended our previous examination of these mice using electroencephalogram/electromyogram (EEG/EMG) monitoring and found age-related increases in spontaneous seizures, which were correlated with cellular dispersion in the hippocampal dentate gyrus. Increased spontaneous locomotor activity in the open field on the first and the second day of a 3-day continuous study suggested heightened anxiety in Pten mutant mice. In contrast, the mutants exhibited decreased wheel running activity, which may reflect reduced adaptability to a novel environment. Synchronization to the light-dark cycle was normal, but for up to 28 days under constant darkness, the Pten mutants maintained a significantly lengthened and remarkably constant free-running period of almost exactly 24 h. This result implies the involvement of Pten in the maintenance of circadian rhythms, which we interpret as being due to an effect on the phosphatidylinositol 3-kinase (PI3K) signaling cascade.


Assuntos
PTEN Fosfo-Hidrolase/genética , Periodicidade , Corrida/fisiologia , Convulsões/genética , Convulsões/fisiopatologia , Fatores Etários , Análise de Variância , Animais , Comportamento Animal , Eletroencefalografia/métodos , Eletromiografia/métodos , Comportamento Exploratório/fisiologia , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/deficiência , Fenótipo , Fosfopiruvato Hidratase/genética , Convulsões/patologia
16.
J Neurosci ; 27(22): 5967-75, 2007 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-17537967

RESUMO

The conceptual understanding of hippocampal function has been challenged recently by the finding that new granule cells are born throughout life in the mammalian dentate gyrus (DG). The number of newborn neurons is dynamically regulated by a variety of factors. Kainic acid-induced seizures, a rodent model of human temporal lobe epilepsy, strongly induce the proliferation of DG neurogenic progenitor cells and are also associated with long-term cognitive impairment. We show here that the antiepileptic drug valproic acid (VPA) potently blocked seizure-induced neurogenesis, an effect that appeared to be mainly mediated by inhibiting histone deacetylases (HDAC) and normalizing HDAC-dependent gene expression within the epileptic dentate area. Strikingly, the inhibition of aberrant neurogenesis protected the animals from seizure-induced cognitive impairment in a hippocampus-dependent learning task. We propose that seizure-generated granule cells have the potential to interfere with hippocampal function and contribute to cognitive impairment caused by epileptic activity within the hippocampal circuitry. Furthermore, our data indicate that the effectiveness of VPA as an antiepileptic drug may be partially explained by the HDAC-dependent inhibition of aberrant neurogenesis induced by seizure activity within the adult hippocampus.


Assuntos
Transtornos Cognitivos/patologia , Epigênese Genética/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Convulsões/patologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/prevenção & controle , Epigênese Genética/efeitos dos fármacos , Feminino , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Neurônios/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Convulsões/complicações , Convulsões/prevenção & controle , Ácido Valproico/farmacologia , Ácido Valproico/uso terapêutico
17.
Proc Natl Acad Sci U S A ; 102(28): 9942-7, 2005 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-15983370

RESUMO

Neuropeptide B (NPB) and neuropeptide W (NPW) have been recently identified as ligands for the G protein-coupled receptor (GPR) 7 and GPR8. The precise in vivo role of this neuropeptide-receptor pathway has not been fully demonstrated. In this paper, we report that NPB-deficient mice manifest a mild adult-onset obesity, similar to that reported in GPR7-null mice. NPB-deficient mice also exhibit hyperalgesia in response to inflammatory pain. Hyperalgesia was not observed in response to chemical pain, thermal pain, or electrical stimulation. NPB-deficient mice demonstrated intact behavioral responses to pain, and learning from the negative reinforcement of electrical stimulation was unaltered. Baseline anxiety was also unchanged as measured in both the elevated plus maze and time spent immobile in a novel environment. These data support the idea that NPB is a factor in the modulation of responses to inflammatory pain and body weight homeostasis.


Assuntos
Hiperalgesia/genética , Inflamação/complicações , Neuropeptídeos/genética , Dor/fisiopatologia , Análise de Variância , Animais , Peso Corporal/genética , Formaldeído , Hiperalgesia/etiologia , Hibridização In Situ , Camundongos , Camundongos Knockout , Dor/etiologia
18.
J Neurosci ; 24(46): 10493-501, 2004 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-15548664

RESUMO

Temporal restriction of feeding can entrain circadian behavioral and physiological rhythms in mammals. Considering the critical functions of the hypothalamic orexin (hypocretin) neuropeptides in promoting wakefulness and locomotor activity, we examined the role of orexin neurons in the adaptation to restricted feeding. In orexin neuron-ablated transgenic mice, the food-entrained rhythmicity of mPer2 expression in the brain and liver, the reversal of the sleep-wake cycle, and the recovery of daily food intake were unaltered compared with wild-type littermates. In contrast, orexin neuron-ablated mice had a severe deficit in displaying the normal food-anticipatory increases in wakefulness and locomotor activity under restricted feeding. Moreover, activity of orexin neurons markedly increased during the food-anticipatory period under restricted feeding in wild-type mice. Orexin neurons thus convey an efferent signal from putative food-entrainable oscillator or oscillators to increase wakefulness and locomotor activity.


Assuntos
Ritmo Circadiano , Comportamento Alimentar , Privação de Alimentos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Vigília , Animais , Encéfalo/metabolismo , Proteínas de Ciclo Celular , Cruzamentos Genéticos , Ingestão de Alimentos , Eletroencefalografia , Eletromiografia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Neuropeptídeos/genética , Proteínas Nucleares/biossíntese , Orexinas , Proteínas Circadianas Period , Sono , Fatores de Transcrição
19.
Semin Neurol ; 24(3): 211-23, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15449215

RESUMO

Following a summary of the stages of sleep and wakefulness as monitored with the electroencephalogram and electromyogram, important aspects of the neurophysiology and neuroanatomy of the circuits of vigilance state control are reviewed. A homeostatic drive for sleep and a circadian influence work in concert to determine sleepiness. These processes influence sleep-promoting and central arousing neuronal systems, the former dependent on a group of neurons in the hypothalamic ventrolateral preoptic area and the latter governed by neurons in the pons and basal forebrain. The interactive neuronal circuit that is formed by these cell groups ensures the balance between sleep and wakefulness and the rapid transition to and from sleep. As sleep deepens, the switch to rapid eye movement (REM) sleep occurs. This transition can also be viewed as a balance between one group of pontine neurons that discharge only during REM sleep and another group that cease to discharge during REM sleep. This article concludes with future perspectives based on the recent discovery of the orexin cell group. Orexinergic neurons may be critical both for promoting wakefulness at certain times in the daily cycle and for controlling the switch into REM sleep.


Assuntos
Encéfalo/fisiologia , Eletroencefalografia/métodos , Sono/fisiologia , Vigília/fisiologia , Animais , Humanos
20.
J Neurosci ; 24(18): 4469-77, 2004 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15128861

RESUMO

The sleep disorder narcolepsy has been linked to loss of hypothalamic neurons producing the orexin (hypocretin) neuropeptides. Here, we report the generation of transgenic rats expressing a human ataxin-3 fragment with an elongated polyglutamyl stretch under control of the human prepro-orexin promoter (orexin/ataxin-3 rats). At 17 weeks of age, the transgenic rats exhibited postnatal loss of orexin-positive neurons in the lateral hypothalamus, and orexin-containing projections were essentially undetectable. The loss of orexin production resulted in the expression of a phenotype with fragmented vigilance states, a decreased latency to rapid eye movement (REM) sleep and increased REM sleep time during the dark active phase. Wakefulness time was also reduced during the dark phase, and this effect was concentrated at the photoperiod boundaries. Direct transitions from wakefulness to REM sleep, a defining characteristic of narcolepsy, occurred frequently. Brief episodes of muscle atonia and postural collapse resembling cataplexy were also noted while rats maintained the electroencephalographic characteristics of wakefulness. These findings indicate that the orexin/ataxin-3 transgenic rat could provide a useful model of human narcolepsy.


Assuntos
Proteínas de Transporte/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Narcolepsia/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Transgenes , Animais , Animais Geneticamente Modificados , Nível de Alerta/genética , Ataxina-3 , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Eletroencefalografia , Eletromiografia , Humanos , Masculino , Narcolepsia/metabolismo , Narcolepsia/fisiopatologia , Neuropeptídeos/deficiência , Proteínas Nucleares , Orexinas , Fenótipo , Fotoperíodo , Regiões Promotoras Genéticas/genética , Ratos , Ratos Wistar , Proteínas Repressoras , Sono REM/genética , Vigília/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...