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 Res ; 101(8): 1305-1323, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37012516

RESUMO

Orexin-A (OXA) is a hypothalamic neuropeptide implicated in the regulation of wakefulness, appetite, reward processing, muscle tone, motor activity, and other physiological processes. The broad range of systems affected stems from the widespread projections of orexin neurons toward multiple brain regions regulating numerous physiological processes. Orexin neurons integrate nutritional, energetic, and behavioral cues and modulate the functions of target structures. Orexin promotes spontaneous physical activity (SPA), and we recently showed that orexin injected into the ventrolateral preoptic area (VLPO) of the hypothalamus increases behavioral arousal and SPA in rats. However, the specific mechanisms underlying the role of orexin in physical activity are unknown. Here we tested the hypothesis that OXA injected into the VLPO alters the oscillatory activity in the electroencephalogram (EEG) to reflect an increased excitability of the sensorimotor cortex, which may explain the associated increase in SPA. The results showed that OXA increased wakefulness following injections into the VLPO. In addition, OXA altered the power spectrum of the EEG during the awake state by decreasing the power of 5-19 Hz oscillations and increasing the power of >35 Hz oscillations, which are markers of increased sensorimotor excitability. Consistently, we found that OXA induced greater muscle activity. Furthermore, we found a similar change in power spectrum during slow-wave sleep, which suggests that OXA altered the EEG activity in a fundamental way, even in the absence of physical activity. These results support the idea that OXA increases the excitability of the sensorimotor system, which may explain the corresponding increase in awake time, muscle tone, and SPA.


Assuntos
Tono Muscular , Área Pré-Óptica , Ratos , Animais , Orexinas/farmacologia , Orexinas/metabolismo , Área Pré-Óptica/metabolismo , Sono/fisiologia , Hipotálamo/metabolismo , Vigília/fisiologia
2.
J Neurophysiol ; 127(5): 1221-1229, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-35353632

RESUMO

The regulation of sleep/wake behavior and energy homeostasis is maintained in part by the hypothalamic neuropeptide orexin A (OXA, hypocretin). Reduction in orexin signaling is associated with sleep disorders and obesity, whereas higher lateral hypothalamic (LH) orexin signaling and sensitivity promotes obesity resistance. Similarly, dysregulation of hypothalamic neural networks is associated with onset of age-related diseases, including obesity and several neurological diseases. Despite the association of obesity and aging, and that adult populations are the target for the majority of pharmaceutical and obesity studies, conventional models for neuronal networks utilize embryonic neural cultures rather than adult neurons. Synchronous activity describes correlated changes in neuronal activity between neurons and is a feature of normal brain function, and is a measure of functional connectivity and final output from a given neural structure. Earlier studies show alterations in hypothalamic synchronicity following behavioral perturbations in embryonic neurons obtained from obesity-resistant rats and following application of orexin onto embryonic hypothalamic cultures. Synchronous network dynamics in adult hypothalamic neurons remain largely undescribed. To address this, we established an adult rat hypothalamic culture in multi-electrode-array (MEA) dishes and recorded the field potentials. Then we studied the effect of exogenous orexin on network synchronization of these adult hypothalamic cultures. In addition, we studied the wake promoting effects of orexin in vivo when directly injected into the lateral hypothalamus (LH). Our results showed that the adult hypothalamic cultures are viable for nearly 3 mo in vitro, good quality MEA recordings can be obtained from these cultures in vitro, and finally, that cultured adult hypothalamus is responsive to orexin. These results support that adult rat hypothalamic cultures could be used as a model to study the neural mechanisms underlying obesity. In addition, LH administration of OXA enhanced wakefulness in rats, indicating that OXA enhances wakefulness partly by promoting neural synchrony in the hypothalamus.NEW & NOTEWORTHY This study, for the first time, demonstrates that adult hypothalamic cultures are viable in vitro for a prolonged duration and are electrophysiologically active. In addition, the study shows that orexin enhances neural synchronization in adult hypothalamic cultures.


Assuntos
Região Hipotalâmica Lateral , Hipotálamo , Animais , Região Hipotalâmica Lateral/fisiologia , Neurônios/fisiologia , Obesidade , Orexinas/farmacologia , Ratos
3.
WIREs Mech Dis ; 14(1): e1536, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35023323

RESUMO

The lateral hypothalamus is critical for the control of ingestive behavior and spontaneous physical activity (SPA), as lesion or stimulation of this region alters these behaviors. Evidence points to lateral hypothalamic orexin neurons as modulators of feeding and SPA. These neurons affect a broad range of systems, and project to multiple brain regions such as the dorsal raphe nucleus, which contains serotoninergic neurons (DRN) important to energy homeostasis. Physical activity is comprised of intentional exercise and SPA. These are opposite ends of a continuum of physical activity intensity and structure. Non-goal-oriented behaviors, such as fidgeting, standing, and ambulating, constitute SPA in humans, and reflect a propensity for activity separate from intentional activity, such as high-intensity voluntary exercise. In animals, SPA is activity not influenced by rewards such as food or a running wheel. Spontaneous physical activity in humans and animals burns calories and could theoretically be manipulated pharmacologically to expend calories and protect against obesity. The DRN neurons receive orexin inputs, and project heavily onto cortical and subcortical areas involved in movement, feeding and energy expenditure (EE). This review discusses the function of hypothalamic orexin in energy-homeostasis, the interaction with DRN serotonin neurons, and the role of this orexin-serotonin axis in regulating food intake, SPA, and EE. In addition, we discuss possible brain areas involved in orexin-serotonin cross-talk; the role of serotonin receptors, transporters and uptake-inhibitors in the pathogenesis and treatment of obesity; animal models of obesity with impaired serotonin-function; single-nucleotide polymorphisms in the serotonin system and obesity; and future directions in the orexin-serotonin field. This article is categorized under: Metabolic Diseases > Molecular and Cellular Physiology.


Assuntos
Metabolismo Energético , Serotonina , Animais , Humanos , Região Hipotalâmica Lateral/metabolismo , Hipotálamo/metabolismo , Orexinas/metabolismo
4.
J Med Chem ; 64(12): 8806-8825, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34101446

RESUMO

Loss of orexin-producing neurons results in narcolepsy with cataplexy, and orexin agonists have been shown to increase wakefulness and alleviate narcolepsy symptoms in animal models. Several OX2R agonists have been reported but with little or no activity at OX1R. We conducted structure-activity relationship studies on the OX2R agonist YNT-185 (2) and discovered dual agonists such as RTOXA-43 (40) with EC50's of 24 nM at both OX2R and OX1R. Computational modeling studies based on the agonist-bound OX2R cryogenic electron microscopy structures showed that 40 bound in the same binding pocket and interactions of the pyridylmethyl group of 40 with OX1R may have contributed to its high OX1R potency. Intraperitoneal injection of 40 increased time awake, decreased time asleep, and increased sleep/wake consolidation in 12-month old mice. This work provides a promising dual small molecule agonist and supports development of orexin agonists as potential treatments for orexin-deficient disorders such as narcolepsy.


Assuntos
Receptores de Orexina/agonistas , Sono/efeitos dos fármacos , Sulfonamidas/farmacologia , Vigília/efeitos dos fármacos , Animais , Células CHO , Cricetulus , Feminino , Masculino , Camundongos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Estrutura Molecular , Receptores de Orexina/metabolismo , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/metabolismo
5.
Exp Brain Res ; 239(3): 755-764, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33388905

RESUMO

Synchronous neural activity is a feature of normal brain function, and altered synchronization is observed in several neurological diseases. Dysfunction in hypothalamic pathways leads to obesity, suggesting that hypothalamic neural synchrony is critical for energy homeostasis. The lateral hypothalamic orexin neurons are extensively interconnected with other brain structures and are important for energy balance. Earlier studies show that rats with higher orexin sensitivity are obesity resistant. Similarly, topiramate, an anti-epileptic drug, has been shown to reduce weight in humans. Since orexin enhances neuronal excitation, we hypothesized that obesity-resistant rats with higher orexin sensitivity may exhibit enhanced hypothalamic synchronization. We further hypothesized that anti-obesity agents such as orexin and topiramate will enhance hypothalamic synchronization. To test this, we examined neural synchronicity in primary embryonic hypothalamic cell cultures, obtained from embryonic day 18 (E18) obesity-susceptible Sprague-Dawley (SD) and obesity-resistant rats. Hypothalamic tissue was cultured in multielectrode array (MEA), and recordings were performed twice weekly, from 4th to 32nd day in vitro (DIV). Next, we tested the effects of orexin and topiramate application on neural synchronicity of hypothalamic cultures obtained from SD rat embryos. Signals were analyzed for synchronization using cross correlation. Our results showed that (1) obesity-resistant hypothalamus exhibits significantly higher synchronization compared to obesity-sensitive hypothalamus; and (2) orexin and topiramate enhance hypothalamic synchronization. These results support that enhanced orexin sensitivity is associated with greater neural synchronization, and that anti-obesity treatments enhance network synchronization, thus constrain variability in hypothalamic output signals, to extrahypothalamic structures involved in energy homeostasis.


Assuntos
Doenças Metabólicas , Animais , Hipotálamo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Neurobiol Learn Mem ; 146: 21-30, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29107703

RESUMO

Orexin A is produced in neurons of the lateral, perifornical and dorsomedial regions of the lateral hypothalamic area, which then project widely throughout the central nervous system to regulate arousal state, sleep-wake architecture, energy homeostasis and cognitive processes. Disruption of orexin signaling leads to sleep disturbances and increased body mass index, but recent studies also indicate that orexin neuron activation improves learning and memory. We hypothesized that hippocampal orexin receptor activation improves memory. To test this idea, we obtained orexin/ataxin-3 (O/A3) mice, which become deficient in orexin neurons by about 12 weeks of age. We first measured hippocampal orexin receptor 1 (OX1R) gene expression and protein levels, then tested acquisition and consolidation of two-way active avoidance (TWAA) memory, a hippocampal-dependent learning and memory task. Finally, we determined if exogenous intra-hippocampal OXA treatment could reverse cognitive impairment (as determined by TWAA) in OA/3 mice. We showed that OX1R mRNA expression and protein levels were significantly elevated in O/A3 mice, indicating the potential for preserved orexin responsiveness. The O/A3 mice were significantly impaired in TWAA memory vs. control mice, but OXA treatment (both acute and chronic) reversed these memory deficits. These results demonstrate that orexin plays an important role in hippocampal-dependent consolidation of two-way active avoidance memory, and orexin replacement can rescue the cognitive impairment.


Assuntos
Aprendizagem da Esquiva , Disfunção Cognitiva , Hipocampo , Consolidação da Memória , Transtornos da Memória , Receptores de Orexina/metabolismo , Orexinas/deficiência , Orexinas/farmacologia , Animais , Ataxina-3 , Aprendizagem da Esquiva/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/fisiopatologia , Modelos Animais de Doenças , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Consolidação da Memória/efeitos dos fármacos , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Orexina/genética , Orexinas/administração & dosagem , RNA Mensageiro/metabolismo
7.
J Vis Exp ; (116)2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27805611

RESUMO

Nanoparticles found in air pollutants can alter neurotransmitter profiles, increase neuroinflammation, and alter brain function. Therefore, the assay described here will aid in elucidating the role of microglia in neuroinflammation and neurodegenerative diseases. The use of microglia, resident immune cells of the brain, as a surrogate biosensor provides novel insight into how inflammatory responses mediate neuronal insults. Here, we utilize an immortalized murine microglial cell line, designated BV2, and describe a method for nanoparticle exposure using silver nanoparticles (AgNPs) as a standard. We describe how to expose microglia to nanoparticles, how to remove nanoparticles from supernatant, and how to use supernatant from activated microglia to determine toxicity, using hypothalamic cell survival as a measure. Following AgNP exposure, BV2 microglial activation was validated using a tumor necrosis factor alpha (TNF-α) enzyme linked immunosorbent assay (ELISA). The supernatant was filtered to remove the AgNP and to allow cytokines and other secreted factors to remain in the conditioned media. Hypothalamic cells were then exposed to supernatant from AgNP activated microglia and survival of neurons was determined using a resazurin-based fluorescent assay. This technique is useful for utilizing microglia as a surrogate biomarker of neuroinflammation and determining the effect of neuroinflammation on other cell types.


Assuntos
Técnicas Biossensoriais , Microglia , Nanopartículas/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular , Células Cultivadas , Hipotálamo , Camundongos , Testes de Toxicidade , Fator de Necrose Tumoral alfa
8.
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
9.
Ageing Res Rev ; 20: 63-73, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25462194

RESUMO

The hypothalamic neuropeptides orexin A and B (hypocretin 1 and 2) are important homeostatic mediators of central control of energy metabolism and maintenance of sleep/wake states. Dysregulation or loss of orexin signaling has been linked to narcolepsy, obesity, and age-related disorders. In this review, we present an overview of our current understanding of orexin function, focusing on sleep disorders, energy balance, and aging, in both rodents and humans. We first discuss animal models used in studies of obesity and sleep, including loss of function using transgenic or viral-mediated approaches, gain of function models using exogenous delivery of orexin receptor agonist, and naturally-occurring models in which orexin responsiveness varies by individual. We next explore rodent models of orexin in aging, presenting evidence that orexin loss contributes to age-related changes in sleep and energy balance. In the next section, we focus on clinical importance of orexin in human obesity, sleep, and aging. We include discussion of orexin loss in narcolepsy and potential importance of orexin in insomnia, correlations between animal and human studies of age-related decline, and evidence for orexin involvement in age-related changes in cognitive performance. Finally, we present a summary of recent studies of orexin in neurodegenerative disease. We conclude that orexin acts as an integrative homeostatic signal influencing numerous brain regions, and that this pivotal role results in potential dysregulation of multiple physiological processes when orexin signaling is disrupted or lost.


Assuntos
Envelhecimento/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Neuropeptídeos , Obesidade , Transtornos do Sono-Vigília , Animais , Modelos Animais de Doenças , Metabolismo Energético , Homeostase/fisiologia , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Neurotransmissores/metabolismo , Neurotransmissores/farmacologia , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Orexinas , Transdução de Sinais , Sono/efeitos dos fármacos , Sono/fisiologia , Transtornos do Sono-Vigília/tratamento farmacológico , Transtornos do Sono-Vigília/metabolismo
10.
Neurobiol Learn Mem ; 114: 40-50, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24755094

RESUMO

BACKGROUND: Previous studies have shown that a western diet impairs, whereas physical exercise enhances hippocampus-dependent learning and memory. Both diet and exercise influence expression of hippocampal brain-derived neurotrophic factor (BDNF), which is associated with improved cognition. We hypothesized that exercise reverses diet-induced cognitive decline while increasing hippocampal BDNF. METHODS: To test the effects of exercise on hippocampal-dependent memory, we compared cognitive scores of Sprague-Dawley rats exercised by voluntary running wheel (RW) access or forced treadmill (TM) to sedentary (Sed) animals. Memory was tested by two-way active avoidance test (TWAA), in which animals are exposed to a brief shock in a specific chamber area. When an animal avoids, escapes or has reduced latency to do either, this is considered a measure of memory. In a second experiment, rats were fed either a high-fat diet or control diet for 16 weeks, then randomly assigned to running wheel access or sedentary condition, and TWAA memory was tested once a week for 7 weeks of exercise intervention. RESULTS: Both groups of exercised animals had improved memory as indicated by reduced latency to avoid and escape shock, and increased avoid and escape episodes (p<0.05). Exposure to a high-fat diet resulted in poor performance during both the acquisition and retrieval phases of the memory test as compared to controls. Exercise reversed high-fat diet-induced memory impairment, and increased brain-derived neurotrophic factor (BDNF) in neurons of the hippocampal CA3 region. CONCLUSIONS: These data suggest that exercise improves memory retrieval, particularly with respect to avoiding aversive stimuli, and may be beneficial in protecting against diet induced cognitive decline, likely via elevated BDNF in neurons of the CA3 region.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Região CA3 Hipocampal/fisiologia , Transtornos Cognitivos/prevenção & controle , Dieta/efeitos adversos , Neurônios/metabolismo , Condicionamento Físico Animal/fisiologia , Animais , Aprendizagem da Esquiva/fisiologia , Região CA3 Hipocampal/metabolismo , Cognição/fisiologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
11.
Obesity (Silver Spring) ; 21(7): 1396-405, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23666828

RESUMO

OBJECTIVE: Sleep restriction in humans increases risk for obesity, but previous rodent studies show weight loss following sleep deprivation, possibly due to stressful methods used to prevent sleep. Obesity-resistant (OR) rats exhibit consolidated-sleep and resistance to weight gain. It was hypothesized that sleep disruption by a less-stressful method would increase body weight, and the effect of partial sleep deprivation (PSD) on body weight in OR and Sprague-Dawley (SD) rats was examined. DESIGN AND METHODS: OR and SD rats (n = 12/group) were implanted with transmitters to record sleep/wake. After baseline recording, six SD and six OR rats underwent 8 h PSD during light phase for 9 days. Sleep was reduced using recordings of random noise. Sleep/wake states were scored as wakefulness (W), slow-wave-sleep (SWS), and rapid-eye-movement-sleep (REMS). Total number of transitions between stages, SWS-delta-power, food intake, and body weight were documented. RESULTS: Exposure to noise decreased SWS and REMS time, while increasing W time. Sleep-deprivation increased the number of transitions between stages and SWS-delta-power. Further, PSD during the rest phase increased recovery sleep during the active phase. The PSD SD and OR rats had greater food intake and body weight compared to controls CONCLUSIONS: PSD by less-stressful means increases body weight in rats. Also, PSD during the rest phase increases active period sleep.


Assuntos
Peso Corporal , Ingestão de Alimentos , Ruído/efeitos adversos , Obesidade/fisiopatologia , Privação do Sono/complicações , Animais , Eletroencefalografia , Masculino , Obesidade/etiologia , Ratos , Ratos Sprague-Dawley , Descanso/fisiologia , Fatores de Risco , Sono REM/fisiologia , Vigília/fisiologia
12.
Vitam Horm ; 89: 91-109, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22640610

RESUMO

The orexins/hypocretins are endogenous, modulatory and multifunctional neuropeptides with prominent influence on several physiological processes. The influence of orexins on energy expenditure is highlighted with focus on orexin action on individual components of energy expenditure. As orexin stabilizes and maintains normal states of arousal and the sleep/wake cycle, we also highlight orexin mediation of sleep and how sleep interacts with energy expenditure.


Assuntos
Metabolismo Energético , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Transmissão Sináptica , Animais , Humanos , Atividade Motora , Orexinas , Sono
13.
Neurosci Biobehav Rev ; 36(3): 1015-29, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22266350

RESUMO

The rapid rise in obesity prevalence in the modern world parallels a significant reduction in restorative sleep (Agras et al., 2004; Dixon et al., 2007, 2001; Gangwisch and Heymsfield, 2004; Gupta et al., 2002; Sekine et al., 2002; Vioque et al., 2000; Wolk et al., 2003). Reduced sleep time and quality increases the risk for obesity, but the underlying mechanisms remain unclear (Gangwisch et al., 2005; Hicks et al., 1986; Imaki et al., 2002; Jennings et al., 2007; Moreno et al., 2006). A majority of the theories linking human sleep disturbances and obesity rely on self-reported sleep. However, studies with objective measurements of sleep/wake parameters suggest a U-shaped relationship between sleep and obesity. Studies in animal models are needed to improve our understanding of the association between sleep disturbances and obesity. Genetic and experimenter-induced models mimicking characteristics of human obesity are now available and these animal models will be useful in understanding whether sleep disturbances determine propensity for obesity, or result from obesity. These models exhibit weight gain profiles consistently different from control animals. Thus a careful evaluation of animal models will provide insight into the relationship between sleep disturbances and obesity in humans. In this review we first briefly consider the fundamentals of sleep and key sleep disturbances, such as sleep fragmentation and excessive daytime sleepiness (EDS), observed in obese individuals. Then we consider sleep deprivation studies and the role of circadian alterations in obesity. We describe sleep/wake changes in various rodent models of obesity and obesity resistance. Finally, we discuss possible mechanisms linking sleep disturbances with obesity.


Assuntos
Modelos Animais de Doenças , Camundongos , Obesidade/etiologia , Ratos , Sono/fisiologia , Animais , Relógios Circadianos/fisiologia , Humanos , Obesidade/metabolismo , Privação do Sono , Sono REM/fisiologia
14.
J Neurosci Res ; 80(5): 727-37, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15880522

RESUMO

The aim of this study was to test the hypothesis that the dorsal hippocampus plays a critical role in pontine-wave (P-wave) generator activation-dependent memory processing of two-way active avoidance (TWAA) learning. To achieve this objective, rats were given small bilateral lesions in the CA1, dentate gyrus (DG), or CA3 region of the dorsal hippocampus by microinjecting ibotenic acid. After recovery, lesioned and sham-lesioned rats were trained on a TWAA learning paradigm, allowed a 6-hr period of undisturbed sleep, and then were tested on the same TWAA paradigm. It was found that lesions in the CA3 region impaired retention of avoidance learning. Conversely, lesions in the CA1 and DG regions had no effect on TWAA learning retention. None of the groups showed any changes in the baseline sleep-wake cycle or in the acquisition of TWAA learning. All rats showed increased rapid eye movement (REM) sleep and increased REM sleep P-wave density during the subsequent 6-hr recording period. Impaired retention in the CA3 group occurred despite an increase in REM sleep and P-wave density, suggesting that during REM sleep, the P-wave generator interacts with the CA3 region of the dorsal hippocampus to aid in consolidation of TWAA learning. The results of the present study thus demonstrate that P-wave generator activation-dependent consolidation of memory requires an intact CA3 subfield of the dorsal hippocampus. The results also provide evidence that under mnemonic pressure, the dorsal hippocampus may not be involved directly in regulating the sleep-wake cycle.


Assuntos
Aprendizagem da Esquiva/fisiologia , Hipocampo/fisiologia , Memória/fisiologia , Ponte/fisiologia , Animais , Denervação , Agonistas de Aminoácidos Excitatórios , Hipocampo/citologia , Ácido Ibotênico , Masculino , Vias Neurais , Ponte/citologia , Ratos , Ratos Sprague-Dawley , Sono REM/fisiologia , Vigília/fisiologia
15.
Sleep ; 27(7): 1282-92, 2004 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-15586781

RESUMO

STUDY OBJECTIVES: The aim of this study was to test the hypothesis that the activation of pontine (P)-wave generator is critical for the posttraining rapid eye movement (REM) sleep-dependent memory processing. DESIGN: Ibotenic acid was microinjected (0.5 microg in 0.05 microL) into the functionally identified P-wave generator in order to destroy the cell bodies and thus to study the effects of their destruction upon waking-sleep states, P-waves, and 2-way active avoidance memory. SETTING: Sleep research laboratory at Boston University School of Medicine. PARTICIPANTS: Adult male Sprague-Dawley rats (N = 27). INTERVENTIONS: Chronically implanted for recording polygraphic signs of sleep and bilateral guide tubes for the local microinjections into the P-wave generator. MEASUREMENTS AND RESULTS: The ibotenic acid produced a small spherical area (< or = 0.35 mm in diameter) of nerve cell loss centered on the P-wave generator. Bilateral lesioning of the P-wave generator decreased P-waves during REM sleep by > 95% without significantly changing the amounts of time spent in wake, slow-wave sleep, or REM sleep. In these P-wave generator-lesioned rats, acquisition of avoidance learning and posttraining wake-sleep changes were identical to those of the sham-lesioned rats. However, in the test trials, after 6 hours of undisturbed sleep-wake, P-wave generator-lesioned rats had no retention of avoidance memory. CONCLUSIONS: These findings, for the first time, provide direct evidence that P-wave-generating cells are critical for normal REM sleep-dependent memory processing. This evidence supports our hypothesis that the P-wave generator in the brainstem may act as an on switch to provide activating input to forebrain structures for sleep-dependent memory processing.


Assuntos
Aprendizagem da Esquiva/efeitos dos fármacos , Ácido Ibotênico/farmacologia , Neurotoxinas/farmacologia , Ponte/efeitos dos fármacos , Retenção Psicológica/efeitos dos fármacos , Sono REM/efeitos dos fármacos , Animais , Aprendizagem da Esquiva/fisiologia , Mapeamento Encefálico , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Masculino , Microinjeções , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Polissonografia/efeitos dos fármacos , Ponte/fisiologia , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/fisiologia , Ratos , Ratos Sprague-Dawley , Retenção Psicológica/fisiologia , Sono REM/fisiologia
16.
J Neurosci ; 24(6): 1416-27, 2004 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-14960614

RESUMO

Animal and human studies of sleep and learning have demonstrated that training on various tasks increases subsequent rapid eye movement (REM) sleep and phasic pontine-wave (P-wave) activity, followed by improvement in performance on the learned task. It is well documented that REM sleep deprivation after learning trials blocks the expected improvement in performance on subsequent retesting. Our aim was to test whether experimentally induced P-wave generator activation could eliminate the learning impairment produced by post-training REM sleep deprivation. Rats were trained on a two-way active avoidance-learning task. Immediately thereafter, two groups of those rats received a control vehicle (100 nl saline) microinjection and one group received a carbachol (50 ng in 100 nl saline) microinjection into the P-wave generator. The carbachol-injected group and one of the two control saline microinjected groups were selectively deprived of REM sleep during a 6 hr polygraphic recording session. All rats were then tested on the avoidance-learning task. The rats that received both the control saline injection and REM sleep deprivation showed learning deficits compared with the control saline-injected rats that were allowed to sleep normally. In contrast, the rats that received the carbachol microinjection and REM sleep deprivation demonstrated normal learning. These results demonstrate, for the first time, that carbachol-induced activation of the P-wave generator prevents the memory-impairing effects of post-training REM sleep deprivation. This evidence supports our hypothesis that the activation of the P-wave generator during REM sleep deprivation enhances a physiological process of memory, which occurs naturally during post-training REM sleep.


Assuntos
Deficiências da Aprendizagem/etiologia , Deficiências da Aprendizagem/prevenção & controle , Plasticidade Neuronal/fisiologia , Ponte/fisiologia , Privação do Sono/complicações , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Eletrodos Implantados , Eletroencefalografia , Deficiências da Aprendizagem/fisiopatologia , Masculino , Microinjeções , Polissonografia , Ponte/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Privação do Sono/fisiopatologia , Vigília/fisiologia
17.
J Neurophysiol ; 91(4): 1822-31, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14702336

RESUMO

Considerable evidence suggests that the neurotransmitter gamma-aminobutyric acid (GABA)-ergic system and pedunculopontine tegmentum (PPT) in the brain stem are critically involved in the regulation of rapid-eye-movement (REM) sleep. GABA and its various receptors are normally present in the PPT cholinergic cell compartment. The aim of this study was to identify the role of GABA and its receptors in the regulation of REM sleep. To achieve this aim, specific receptors were activated differentially by local microinjection of selective GABA receptor agonists into the PPT while quantifying its effects on REM sleep in freely moving chronically instrumented rats (n = 21). The results demonstrated that when GABAB receptors were activated by local microinjection of a GABAB receptor selective agonist, baclofen, spontaneous REM sleep was suppressed in a dose-dependent manner. The optimum dose for REM sleep reduction was 1.5 nmol. In contrast, when GABAA and GABAC receptors were activated by microinjecting their receptor selective agonists, isoguvacine (ISGV) and cis-4-aminocrotonic acid (CACA), respectively, the total percentages of REM sleep did not change compared with the control values. In another eight freely moving rats, effects of baclofen application was tested on firing rates of REM-on cells (n = 12). Of those 12 neurons, 11 stopped firing immediately after application of baclofen [latency: 50 +/- 14 s (SD)] and remained almost silent for 130 +/- 12 min. Findings of the present study provide direct evidence that the PPT GABAB receptors and REM-on cells are involved in the regulation of REM sleep.


Assuntos
Núcleo Tegmental Pedunculopontino/fisiologia , Receptores de GABA-B/fisiologia , Sono REM/fisiologia , Vigília/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Relação Dose-Resposta a Droga , Eletroencefalografia/métodos , Eletromiografia/métodos , Eletroculografia/métodos , GABAérgicos/farmacologia , Masculino , Microinjeções/métodos , Núcleo Tegmental Pedunculopontino/citologia , Núcleo Tegmental Pedunculopontino/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de GABA-B/efeitos dos fármacos , Sono REM/efeitos dos fármacos , Fatores de Tempo , Vigília/efeitos dos fármacos
18.
Sleep ; 26(5): 513-20, 2003 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12938803

RESUMO

STUDY OBJECTIVES: Considerable evidence suggests that rapid eye movement (REM) sleep is induced by glutamatergic activation of cholinergic cells within the pedunculopontine tegmentum (PPT). The aim of this study is to test a popular hypothesis that serotonin, norepinephrine, and adenosine act on PPT cells to regulate REM sleep. This study also tests an alternate hypothesis that serotonin may inhibit REM sleep signs by direct action on the individual REM sleep sign generators. DESIGN: Serotonin, norepinephrine, and adenosine were locally microinjected into the PPT and serotonin was microinjected into the pontine-wave (P-wave) generator (dorsal part of the locus subcoeruleus nucleus) while quantifying the effects on REM sleep and P-wave activity in freely moving rats. SETTING: N/A. PARTICIPANTS: N/A. INTERVENTIONS: N/A. MEASUREMENTS AND RESULTS: Local microinjections of serotonin, norepinephrine, and adenosine into the PPT did not change REM sleep. Microinjection of serotonin into the P-wave generator suppressed P-wave activity but not REM sleep. CONCLUSIONS: The present findings provide direct evidence that serotonin, norepinephrine, and adenosine-induced REM sleep suppression in the behaving rat are not mediated by the PPT. The results also provide direct evidence, for the first time, that serotonin suppresses P-wave activity by acting directly on the P-wave generator. These results suggest that the serotonin-induced inhibition of REM sleep in the freely moving rat is probably not mediated through the mesopontine cholinergic cell compartment but, rather, through individual REM sleep sign generators.


Assuntos
Adenosina/farmacologia , Tronco Encefálico/efeitos dos fármacos , Movimentos Oculares/efeitos dos fármacos , Locomoção/efeitos dos fármacos , Norepinefrina/farmacologia , Serotonina/farmacologia , Sono REM/efeitos dos fármacos , Adenosina/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Fibras Colinérgicas/efeitos dos fármacos , Fibras Colinérgicas/metabolismo , Masculino , Microinjeções , Norepinefrina/administração & dosagem , Núcleo Tegmental Pedunculopontino/efeitos dos fármacos , Núcleo Tegmental Pedunculopontino/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato/efeitos dos fármacos , Receptores de Glutamato/metabolismo , Serotonina/administração & dosagem
19.
Behav Neurosci ; 117(4): 751-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12931960

RESUMO

This study examined the effects of intense emotional learning on the sleep-wake state-specific electroencephalographic (EEG) activities of the basolateral (BLA) and central (CeA) nuclei of the amygdala. Rats were trained on a passive-avoidance learning (PAL) protocol that was followed by 6 hr of undisturbed polygraphic recording and a PAL test. After PAL training, the total amount of REM sleep decreased: high-frequency EEG power decreased in the CeA during REM sleep and increased in the BLA during all sleep-wake stages. These results suggest that there is no homeostatic demand for REM sleep after intense emotional learning. However, the PAL-specific changes in the local EEG suggest that some form of memory processing may occur within the amygdala during REM sleep.


Assuntos
Tonsila do Cerebelo/fisiologia , Aprendizagem da Esquiva , Sono REM/fisiologia , Animais , Eletroencefalografia/veterinária , Masculino , Memória , Ratos , Ratos Sprague-Dawley
20.
Eur J Neurosci ; 17(2): 359-70, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12542673

RESUMO

The aim of this study was to test the hypothesis that supplementary activation of the phasic pontine wave (P-wave) generator during rapid eye movement (REM) sleep enhances consolidation and integration of memories, resulting in improved learning. To test this hypothesis, two groups of rats were trained on a two-way active avoidance learning task in the morning. Immediately after training, one group of rats received a carbachol microinjection into the P-wave generator and the other group was microinjected with control saline into the same target area. After training trials and microinjections, rats were allowed a 6-h period of undisturbed sleep in the polygraphic recording chamber. At the end of 6 h of undisturbed sleep-wake recordings, rats were retested in a session of avoidance learning trials. After learning trials, the total percentage of time spent in REM sleep was significantly increased in both saline (15.36%)- and carbachol (17.70%)-microinjected rats. After learning trials, REM sleep P-wave density was significantly greater throughout the 6-h period of recordings in carbachol treated rats than in the saline treated rats. In the retrial session, the improvement in learning task performance was 22.75% higher in the carbachol-microinjected rats than in the saline-microinjected rats. These findings show that the consolidation and integration of memories create a homeostatic demand for P-waves. In addition, these findings provide experimental evidence, for the first time, that activation of the P-wave generator may enhance consolidation and integration of memories, resulting in improved performance on a recently learned task.


Assuntos
Memória/fisiologia , Ponte/fisiologia , Sono REM/fisiologia , Animais , Carbacol/administração & dosagem , Carbacol/farmacologia , Agonistas Colinérgicos/administração & dosagem , Agonistas Colinérgicos/farmacologia , Eletroencefalografia , Injeções Intraventriculares , Masculino , Memória/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Ponte/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Sono REM/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...