Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.545
Filtrar
Más filtros

Intervalo de año de publicación
1.
Nature ; 571(7764): 198-204, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31292557

RESUMEN

Slow-wave sleep and rapid eye movement (or paradoxical) sleep have been found in mammals, birds and lizards, but it is unclear whether these neuronal signatures are found in non-amniotic vertebrates. Here we develop non-invasive fluorescence-based polysomnography for zebrafish, and show-using unbiased, brain-wide activity recording coupled with assessment of eye movement, muscle dynamics and heart rate-that there are at least two major sleep signatures in zebrafish. These signatures, which we term slow bursting sleep and propagating wave sleep, share commonalities with those of slow-wave sleep and paradoxical or rapid eye movement sleep, respectively. Further, we find that melanin-concentrating hormone signalling (which is involved in mammalian sleep) also regulates propagating wave sleep signatures and the overall amount of sleep in zebrafish, probably via activation of ependymal cells. These observations suggest that common neural signatures of sleep may have emerged in the vertebrate brain over 450 million years ago.


Asunto(s)
Neuronas/fisiología , Sueño/fisiología , Pez Cebra/fisiología , Animales , Evolución Biológica , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Encéfalo/fisiopatología , Epéndimo/citología , Movimientos Oculares , Fluorescencia , Frecuencia Cardíaca , Hipnóticos y Sedantes/farmacología , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Neuronas/efectos de los fármacos , Pigmentación/fisiología , Hormonas Hipofisarias/metabolismo , Polisomnografía/métodos , Sueño/efectos de los fármacos , Privación de Sueño/fisiopatología , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Sueño de Onda Lenta/efectos de los fármacos , Sueño de Onda Lenta/fisiología
2.
Behav Pharmacol ; 35(4): 239-252, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38567447

RESUMEN

Rapid-eye movement (REM) sleep deprivation (SD) can induce manic-like behaviors including hyperlocomotion. On the other hand, crocin (one of the main compounds of Crocus sativus L. or Saffron) may be beneficial in the improvement of mental and cognitive dysfunctions. Also, crocin can restore the deleterious effects of SD on mental and cognitive processes. In this study, we investigated the effect of REM SD on female rats' behaviors including depression- and anxiety-like behaviors, locomotion, pain perception, and obsessive-compulsive-like behavior, and also, the potential effect of crocin on REM SD effects. We used female rats because evidence on the role of REM SD in modulating psychological and behavioral functions of female (but not male) rats is limited. REM SD was induced for 14 days (6h/day), and crocin (25, 50, and 75 mg/kg) was injected intraperitoneally. Open field test, forced swim test, hot plate test, and marble burying test were used to assess rats' behaviors. The results showed REM SD-induced manic-like behavior (hyperlocomotion). Also, REM SD rats showed decreased anxiety- and depression-like behavior, pain subthreshold (the duration it takes for the rat to feel pain), and showed obsessive compulsive-like behavior. However, crocin at all doses partially or fully reversed REM SD-induced behavioral changes. In conclusion, our results suggested the possible comorbidity of OCD and REM SD-induced manic-like behavior in female rats or the potential role of REM SD in the etiology of OCD, although more studies are needed. In contrast, crocin can be a possible therapeutic choice for decreasing manic-like behaviors.


Asunto(s)
Carotenoides , Crocus , Privación de Sueño , Animales , Femenino , Ratas , Privación de Sueño/tratamiento farmacológico , Privación de Sueño/complicaciones , Carotenoides/farmacología , Trastorno Obsesivo Compulsivo/tratamiento farmacológico , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Manía/tratamiento farmacológico , Depresión/tratamiento farmacológico , Ratas Wistar , Modelos Animales de Enfermedad , Trastorno Bipolar/tratamiento farmacológico , Sueño REM/efectos de los fármacos , Relación Dosis-Respuesta a Droga
3.
Nature ; 548(7669): 582-587, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28847002

RESUMEN

Multiple populations of wake-promoting neurons have been characterized in mammals, but few sleep-promoting neurons have been identified. Wake-promoting cell types include hypocretin and GABA (γ-aminobutyric-acid)-releasing neurons of the lateral hypothalamus, which promote the transition to wakefulness from non-rapid eye movement (NREM) and rapid eye movement (REM) sleep. Here we show that a subset of GABAergic neurons in the mouse ventral zona incerta, which express the LIM homeodomain factor Lhx6 and are activated by sleep pressure, both directly inhibit wake-active hypocretin and GABAergic cells in the lateral hypothalamus and receive inputs from multiple sleep-wake-regulating neurons. Conditional deletion of Lhx6 from the developing diencephalon leads to decreases in both NREM and REM sleep. Furthermore, selective activation and inhibition of Lhx6-positive neurons in the ventral zona incerta bidirectionally regulate sleep time in adult mice, in part through hypocretin-dependent mechanisms. These studies identify a GABAergic subpopulation of neurons in the ventral zona incerta that promote sleep.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sueño/fisiología , Factores de Transcripción/metabolismo , Zona Incerta/citología , Ácido gamma-Aminobutírico/metabolismo , Animales , Linaje de la Célula , Neuronas GABAérgicas/efectos de los fármacos , Eliminación de Gen , Hipocampo/citología , Hipocampo/fisiología , Proteínas con Homeodominio LIM/deficiencia , Proteínas con Homeodominio LIM/efectos de los fármacos , Proteínas con Homeodominio LIM/genética , Masculino , Ratones , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Orexinas/metabolismo , Terminales Presinápticos/metabolismo , Sueño/efectos de los fármacos , Sueño/genética , Sueño REM/efectos de los fármacos , Sueño REM/genética , Sueño REM/fisiología , Factores de Tiempo , Factores de Transcripción/deficiencia , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Vigilia/efectos de los fármacos , Vigilia/genética , Vigilia/fisiología , Zona Incerta/efectos de los fármacos , Zona Incerta/fisiología
4.
Biochem Biophys Res Commun ; 632: 10-16, 2022 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-36191372

RESUMEN

Paradoxical sleep deprivation (PSD) is prevalent in modern society, and impaired memory is one of its serious consequences. The pathogenic mechanism is still unclear, and the therapeutic strategies for PSD are limited. Here, we found that quercetin treatment ameliorated memory impairments caused by PSD in a dose-dependent manner in an animal model. Quercetin could restore the dynamic changes of the gamma band while the animals performed novel object recognition (NOR) tasks as determined by electroencephalogram analysis. Morphological analysis showed that quercetin, by targeting the hippocampal CA1 region, strikingly ameliorated the overactivation of microglia induced by PSD. Mechanistically, quercetin inhibited the toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), and nuclear factor kappa-b (NF-κB) cascade, which is critical for abnormal microglial activation following PSD stress. Our results provided experimental evidence for the therapeutic effects of quercetin on PSD-related memory impairments by suppressing TLR4/MyD88/NF-κB signaling that mediated abnormal microglia activation in the hippocampus.


Asunto(s)
Trastornos de la Memoria , Microglía , Quercetina , Animales , Ratones , Modelos Animales de Enfermedad , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/etiología , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Quercetina/farmacología , Quercetina/uso terapéutico , Sueño REM/efectos de los fármacos , Receptor Toll-Like 4/metabolismo
5.
Neuroimage ; 231: 117850, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33582277

RESUMEN

Consciousness is a mental characteristic of the human mind, whose exact neural features remain unclear. We aimed to identify the critical nodes within the brain's global functional network that support consciousness. To that end, we collected a large fMRI resting state dataset with subjects in at least one of the following three consciousness states: preserved (including the healthy awake state, and patients with a brain injury history (BI) that is fully conscious), reduced (including the N1-sleep state, and minimally conscious state), and lost (including the N3-sleep state, anesthesia, and unresponsive wakefulness state). We also included a unique dataset of subjects in rapid eye movement sleep state (REM-sleep) to test for the presence of consciousness with minimum movements and sensory input. To identify critical nodes, i.e., hubs, within the brain's global functional network, we used a graph-theoretical measure of degree centrality conjoined with ROI-based functional connectivity. Using these methods, we identified various higher-order sensory and motor regions including the supplementary motor area, bilateral supramarginal gyrus (part of inferior parietal lobule), supragenual/dorsal anterior cingulate cortex, and left middle temporal gyrus, that could be important hubs whose degree centrality was significantly reduced when consciousness was reduced or absent. Additionally, we identified a sensorimotor circuit, in which the functional connectivity among these regions was significantly correlated with levels of consciousness across the different groups, and remained present in the REM-sleep group. Taken together, we demonstrated that regions forming a higher-order sensorimotor integration circuit are involved in supporting consciousness within the brain's global functional network. That offers novel and more mechanism-guided treatment targets for disorders of consciousness.


Asunto(s)
Anestesia/métodos , Estado de Conciencia/fisiología , Red Nerviosa/fisiología , Corteza Sensoriomotora/fisiología , Sueño REM/fisiología , Vigilia/fisiología , Adulto , Anestésicos Intravenosos/administración & dosificación , Estado de Conciencia/efectos de los fármacos , Electroencefalografía/métodos , Femenino , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Red Nerviosa/diagnóstico por imagen , Red Nerviosa/efectos de los fármacos , Corteza Sensoriomotora/diagnóstico por imagen , Corteza Sensoriomotora/efectos de los fármacos , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos , Adulto Joven
6.
Am J Physiol Heart Circ Physiol ; 320(1): H305-H315, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33185112

RESUMEN

Binge alcohol consumption elicits acute and robust increases of muscle sympathetic nerve activity (MSNA), yet the impact of evening binge drinking on morning-after MSNA is unknown. The present study examined the effects of evening binge alcohol consumption on polysomnographic sleep and morning-after MSNA. We hypothesized that evening binge drinking (i.e. 4-5 drink equivalent in <2 h) would reduce sleep quality and increase morning-after blood pressure (BP) and MSNA. Following a familiarization night within the sleep laboratory, 22 participants (12 men, 10 women; 25 ± 1 yr) were examined after simulated binge drinking or fluid control (randomized, crossover design). Morning MSNA was successfully recorded across both conditions in 16 participants (8 men, 8 women) during a 10-min baseline and three Valsalva's maneuvers (VM). Binge drinking reduced rapid eye movement (REM) sleep (15 ± 1 vs. 20 ± 1%, P = 0.003), increased stage II sleep (54 ± 1 vs. 51 ± 1%, P = 0.002), and increased total urine output (2.9 ± 0.2 vs. 2.1 ± 0.1 liters, P < 0.001) but did not alter morning-after urine specific gravity. Binge drinking increased morning-after heart rate [65 (54-72) vs. 58 (51-67) beats/min, P = 0.013] but not resting BP or MSNA. Binge drinking elicited greater sympathoexcitation during VM (38 ± 3 vs. 43 ± 3 bursts/min, P = 0.036). Binge drinking augmented heart rate (P = 0.002), systolic BP (P = 0.022), and diastolic BP (P = 0.037) reactivity to VM phase IV and blunted cardiovagal baroreflex sensitivity during VM phases II (P = 0.028) and IV (P = 0.043). In conclusion, evening binge alcohol consumption disrupted REM sleep and morning-after autonomic function. These findings provide new mechanistic insight into the potential role of binge drinking on cardiovascular risk.NEW & NOTEWORTHY Chronic binge alcohol consumption is associated with future cardiovascular disease (CVD) risk in both men and women. In addition, binge alcohol consumption is known to disrupt normal sleep quality during the early morning hours, coinciding with the morning sympathetic surge. In the present study, an evening of binge alcohol consumption increased baseline morning heart rate and cardiovascular reactivity during the Valsalva maneuver (VM) strain. Specifically, muscle sympathetic nerve activity and phase IV hemodynamic responses increased during VM the morning after binge alcohol consumption. The autonomic dysfunction and increased cardiovascular reactivity during VM suggests a contributing mechanism to CVD risk present in individuals who binge drink.


Asunto(s)
Barorreflejo/efectos de los fármacos , Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Sistema Cardiovascular/inervación , Ritmo Circadiano , Músculo Esquelético/inervación , Sistema Nervioso Simpático/fisiopatología , Adulto , Presión Sanguínea/efectos de los fármacos , Estudios Cruzados , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Masculino , Sueño REM/efectos de los fármacos , Factores de Tiempo , Micción/efectos de los fármacos , Adulto Joven
7.
J Pharmacol Sci ; 145(1): 97-104, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33357785

RESUMEN

Sleep disorders adversely affect daily activities and cause physiological and psychiatric problems. The shortcomings of benzodiazepine hypnotics have led to the development of ramelteon, a melatonin MT1 and MT2 agonist. Although the sleep-promoting effects of ramelteon have been documented, few studies have precisely investigated the structure of sleep and neural oscillatory activities. In this study, we recorded electrocorticograms in the primary motor cortex, the primary somatosensory cortex and the olfactory bulb as well as electromyograms in unrestrained rats treated with either ramelteon or vehicle. A neural-oscillation-based algorithm was used to classify the behavior of the rats into three vigilance states (e.g., awake, rapid eye movement (REM) sleep, and non-REM (NREM) sleep). Moreover, we investigated the region-, frequency- and state-specific modulation of extracellular oscillations in the ramelteon-treated rats. We demonstrated that in contrast to benzodiazepine treatment, ramelteon treatment promoted NREM sleep and enhanced fast gamma power in the primary motor cortex during NREM sleep, while REM sleep was unaffected. Gamma oscillations locally coordinate neuronal firing, and thus, ramelteon modulates neural oscillations in sleep states in a unique manner and may contribute to off-line information processing during sleep.


Asunto(s)
Ritmo Gamma/efectos de los fármacos , Indenos/farmacología , Corteza Motora/fisiología , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Animales , Electrocorticografía , Masculino , Ratas Wistar , Receptor de Melatonina MT1/agonistas , Receptor de Melatonina MT2/agonistas
8.
Biol Pharm Bull ; 44(6): 789-797, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34078810

RESUMEN

Sleep curtailment negatively affects cardiac activities and thus should be ameliorated by pharmacological methods. One of the therapeutic targets is melatonin receptors, which tune circadian rhythms. Ramelteon, a melatonin MT1/MT2 receptor agonist, has recently been developed to modulate sleep-wake rhythms. To date, the sleep-promoting effect of ramelteon has been widely delineated, but whether ramelteon treatment physiologically influences cardiac function is not well understood. To address this question, we recorded electrocardiograms, electromyograms, and electrocorticograms in the frontal cortex and the olfactory bulb of unrestrained rats treated with either ramelteon or vehicle. We detected vigilance states based on physiological measurements and analyzed cardiac and muscular activities. We found that during non-rapid eye movement (non-REM) sleep, heartrate variability was maintained by ramelteon treatment. Analysis of the electromyograms confirmed that neither microarousal during non-REM sleep nor the occupancy of phasic periods during REM sleep was altered by ramelteon. Our results indicate that ramelteon has a remedial effect on cardiac activity by keeping the heartrate variability and may reduce cardiac dysfunction during sleep.


Asunto(s)
Frecuencia Cardíaca/efectos de los fármacos , Indenos/farmacología , Sueño REM/efectos de los fármacos , Animales , Electrocardiografía , Electrocorticografía , Electromiografía , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/fisiología , Masculino , Bulbo Olfatorio/efectos de los fármacos , Bulbo Olfatorio/fisiología , Ratas Wistar , Sueño REM/fisiología
9.
Tijdschr Psychiatr ; 63(1): 70-73, 2021.
Artículo en Holandés | MEDLINE | ID: mdl-33537977

RESUMEN

A 61-year-old woman with suspected schizophrenia has been attending an outpatient geriatrics service for some time, initially with memory complaints and panic attacks. During treatment, the diagnosis schizophrenia was rejected and psychopharmaceuticals were largely phased out, which improved cognitive functions. Eventually, flashbacks of incest experienced in childhood remained together with REM sleep pathology. The flashbacks, nightmares and the REM sleep pathology were responsive to rivastigmine. Rivastigmine use for the treatment of REM sleep pathology is known in the literature, but it has never been described previously that rivastigmine also impacts on flashbacks and nightmares..


Asunto(s)
Sueños/efectos de los fármacos , Alucinaciones/tratamiento farmacológico , Incesto/psicología , Rivastigmina/uso terapéutico , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Sueño REM/efectos de los fármacos , Femenino , Alucinaciones/psicología , Humanos , Persona de Mediana Edad , Trastorno de Pánico/etiología , Trastorno de Pánico/psicología , Sueño REM/fisiología , Resultado del Tratamiento
10.
J Pharmacol Exp Ther ; 372(3): 339-353, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31818916

RESUMEN

The serine hydrolase monoacylglycerol lipase (MAGL) is the rate-limiting enzyme responsible for the degradation of the endocannabinoid 2-arachidonoylglycerol (2-AG) into arachidonic acid and glycerol. Inhibition of 2-AG degradation leads to elevation of 2-AG, the most abundant endogenous agonist of the cannabinoid receptors (CBs) CB1 and CB2. Activation of these receptors has demonstrated beneficial effects on mood, appetite, pain, and inflammation. Therefore, MAGL inhibitors have the potential to produce therapeutic effects in a vast array of complex human diseases. The present report describes the pharmacologic characterization of [1-(4-fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone (JNJ-42226314), a reversible and highly selective MAGL inhibitor. JNJ-42226314 inhibits MAGL in a competitive mode with respect to the 2-AG substrate. In rodent brain, the compound time- and dose-dependently bound to MAGL, indirectly led to CB1 occupancy by raising 2-AG levels, and raised norepinephrine levels in cortex. In vivo, the compound exhibited antinociceptive efficacy in both the rat complete Freund's adjuvant-induced radiant heat hypersensitivity and chronic constriction injury-induced cold hypersensitivity models of inflammatory and neuropathic pain, respectively. Though 30 mg/kg induced hippocampal synaptic depression, altered sleep onset, and decreased electroencephalogram gamma power, 3 mg/kg still provided approximately 80% enzyme occupancy, significantly increased 2-AG and norepinephrine levels, and produced neuropathic antinociception without synaptic depression or decreased gamma power. Thus, it is anticipated that the profile exhibited by this compound will allow for precise modulation of 2-AG levels in vivo, supporting potential therapeutic application in several central nervous system disorders. SIGNIFICANCE STATEMENT: Potentiation of endocannabinoid signaling activity via inhibition of the serine hydrolase monoacylglycerol lipase (MAGL) is an appealing strategy in the development of treatments for several disorders, including ones related to mood, pain, and inflammation. [1-(4-Fluorophenyl)indol-5-yl]-[3-[4-(thiazole-2-carbonyl)piperazin-1-yl]azetidin-1-yl]methanone is presented in this report to be a novel, potent, selective, and reversible noncovalent MAGL inhibitor that demonstrates dose-dependent enhancement of the major endocannabinoid 2-arachidonoylglycerol as well as efficacy in models of neuropathic and inflammatory pain.


Asunto(s)
Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Monoacilglicerol Lipasas/antagonistas & inhibidores , Piperazinas/farmacología , Animales , Unión Competitiva , Encéfalo/enzimología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/sangre , Escherichia coli/enzimología , Escherichia coli/genética , Células HeLa , Humanos , Cinética , Leucocitos Mononucleares/enzimología , Masculino , Ratones Endogámicos C57BL , Estructura Molecular , Monoacilglicerol Lipasas/genética , Dolor/tratamiento farmacológico , Piperazinas/sangre , Unión Proteica , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB2/agonistas , Sueño REM/efectos de los fármacos , Especificidad por Sustrato
11.
J Neurol Neurosurg Psychiatry ; 91(7): 740-749, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32404379

RESUMEN

The rapid eye movement sleep behavioural disorder (RBD) population is an ideal study population for testing disease-modifying treatments for synucleinopathies, since RBD represents an early prodromal stage of synucleinopathy when neuropathology may be more responsive to treatment. While clonazepam and melatonin are most commonly used as symptomatic treatments for RBD, clinical trials of symptomatic treatments are also needed to identify evidence-based treatments. A comprehensive framework for both disease-modifying and symptomatic treatment trials in RBD is described, including potential treatments in the pipeline, cost-effective participant recruitment and selection, study design, outcomes and dissemination of results. For disease-modifying treatment clinical trials, the recommended primary outcome is phenoconversion to an overt synucleinopathy, and stratification features should be used to select a study population at high risk of phenoconversion, to enable more rapid clinical trials. For symptomatic treatment clinical trials, objective polysomnogram-based measurement of RBD-related movements and vocalisations should be the primary outcome measure, rather than subjective scales or diaries. Mobile technology to enable objective measurement of RBD episodes in the ambulatory setting, and advances in imaging, biofluid, tissue, and neurophysiological biomarkers of synucleinopathies, will enable more efficient clinical trials but are still in development. Increasing awareness of RBD among the general public and medical community coupled with timely diagnosis of these diseases will facilitate progress in the development of therapeutics for RBD and associated neurodegenerative disorders.


Asunto(s)
Ensayos Clínicos como Asunto , Trastorno de la Conducta del Sueño REM/tratamiento farmacológico , Sueño REM/efectos de los fármacos , Humanos , Proyectos de Investigación
12.
Alcohol Clin Exp Res ; 44(7): 1378-1388, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424852

RESUMEN

BACKGROUND: Alcohol use is on the rise among women in the United States which is especially concerning since women who drink have a higher risk of alcohol-related problems. Orexin (hypocretin) receptor antagonists may have some therapeutic value for alcohol-induced insomnia; however, the use of this class of drugs following female adolescent binge drinking is limited. The current study will address whether adolescent intermittent ethanol (AIE) in female rats can result in lasting changes in sleep pathology and whether orexin-targeted treatment can alleviate these deficits. METHODS: Following a 5-week AIE vapor model, young adult rats were evaluated on waking event-related oscillations (EROs) and EEG sleep. Subsequently, AIE rats were treated with orexin receptor 2 (OX2 R) antagonist (MK-1064; 10, 20mg/kg) to test for modifications in sleep pathology and waking ERO. RESULTS: Female AIE rats exhibited lasting changes in sleep compared to controls. This was demonstrated by increased fragmentation of slow wave sleep (SWS) and rapid eye movement sleep, as well as reductions in delta and theta power during SWS. There was no impact of AIE on waking EROs. Acute MK-1064 hastened SWS onset and increased the number of SWS episodes, without increasing sleep fragmentation in AIE and controls. While treatment with MK-1064 did not impact sleep EEG spectra, waking ERO energy was increased in delta, theta, and beta frequency bands. CONCLUSIONS: These results demonstrate that AIE can produce lasting changes in sleep in female rats, highly similar to what we previously found in males. Additionally, while the OX2 R antagonist promoted sleep in both alcohol-exposed and unexposed rats, it did not reverse most of the alcohol-induced disruptions in sleep. Thus, OX2 R antagonism may serve as a potential therapeutic strategy for the treatment of insomnia, but not the specific signs of alcohol-induced insomnia.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas , Ondas Encefálicas/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Antagonistas de los Receptores de Orexina/farmacología , Trastornos del Inicio y del Mantenimiento del Sueño , Sueño/efectos de los fármacos , Animales , Ritmo Delta/efectos de los fármacos , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Receptores de Orexina , Ratas , Privación de Sueño , Sueño REM/efectos de los fármacos , Sueño de Onda Lenta/efectos de los fármacos , Ritmo Teta/efectos de los fármacos , Consumo de Alcohol en Menores , Vigilia/efectos de los fármacos
13.
Epilepsy Behav ; 107: 107064, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32320932

RESUMEN

BACKGROUND: Sleep and epilepsy are bedfellows, and they affect each other reciprocally. Despite the well-known relationship between sleep and epilepsy, data about the impact of sleep on seizure control and responsiveness to therapy are scarce. OBJECTIVE: The aim of this work was to study the impact of sleep architecture in drug-naïve patients with idiopathic generalized epilepsy (IGE) on seizure control and responsiveness to treatment. METHODS: This is a prospective cohort study conducted on thirty newly diagnosed patients with IGE attending the epilepsy clinic in Alexandria University Hospital in Egypt and thirty healthy controls. All recruited subjects had a baseline overnight polysomnographic study, then patients were given sodium valproate in therapeutic doses and followed up for six months for assessment of seizure control. After follow-up, they were classified into fully controlled and inadequately controlled patients, and a comparison between them was made. RESULTS: Of the recruited patients, 13 were fully controlled. Rapid eye movement (REM) sleep % was significantly lower among inadequately controlled patients (9.01 ±â€¯6.23) than fully controlled group (19.6 ±â€¯9.01) and controls (18.17 ±â€¯4.85) (p = 0.002), and the REM sleep latency was significantly longer among the inadequately controlled patients (115.7 ±â€¯72.8 min) than fully controlled patients (54.6 ±â€¯77.3 min) and controls (68.75 ±â€¯37.95 min) (p = 011). On univariate regression analysis, the Odd's ratio (OR) for REM sleep percentage was 3.04 (p = 0.001). CONCLUSION: Rapid eye movement sleep percentage and latency can contribute to seizure control in IGE.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia Generalizada/tratamiento farmacológico , Epilepsia Generalizada/fisiopatología , Convulsiones/tratamiento farmacológico , Convulsiones/fisiopatología , Sueño REM/fisiología , Adolescente , Adulto , Anticonvulsivantes/farmacología , Niño , Estudios de Cohortes , Egipto/epidemiología , Epilepsia Generalizada/epidemiología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Estudios Prospectivos , Convulsiones/epidemiología , Sueño REM/efectos de los fármacos , Ácido Valproico/farmacología , Ácido Valproico/uso terapéutico , Adulto Joven
14.
J Cell Mol Med ; 23(10): 6822-6834, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31386303

RESUMEN

Hypocretin 1 and hypocretin 2 (orexin A and B) regulate sleep, wakefulness and emotion. Tumour necrosis factor alpha (TNF-α) is an important neuroinflammation mediator. Here, we examined the effects of TNF-α treatment on hypocretin expression in vivo and behaviour in mice. TNF-α decreased hypocretin 1 and hypocretin 2 expression in a dose-dependent manner in cultured hypothalamic neurons. TNF-α decreased mRNA stability of prepro-hypocretin, the single precursor of hypocretin 1 and hypocretin 2. Mice challenged with TNF-α demonstrated decreased expression of prepro-hypocretin, hypocretin 1 and hypocretin 2 in hypothalamus. In response to TNF-α, prepro-hypocretin mRNA decay was increased in hypothalamus. TNF-α neutralizing antibody restored the expression of prepro-hypocretin, hypocretin 1 and hypocretin 2 in vivo in TNF-α challenged mice, supporting hypocretin system can be impaired by increased TNF-α through decreasing hypocretin expression. Repeated TNF-α challenge induced muscle activity during rapid eye movement sleep and sleep fragmentation, but decreased learning, cognition and memory in mice. TNF-α neutralizing antibody blocked the effects of TNF-α; in contrast, hypocretin receptor antagonist enhanced the effects of TNF-α. The data support that TNF-α is involved in the regulation of hypocretin expression, sleep and cognition. The findings shed some lights on the role of neuroinflammation in neurodegenerative diseases including Alzheimer's disease and Parkinson's disease.


Asunto(s)
Conducta Animal , Orexinas/metabolismo , Sueño , Factor de Necrosis Tumoral alfa/farmacología , Animales , Anticuerpos Neutralizantes/farmacología , Conducta Animal/efectos de los fármacos , Células Cultivadas , Cognición/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/metabolismo , Memoria/efectos de los fármacos , Ratones Endogámicos C57BL , Músculos/efectos de los fármacos , Músculos/fisiología , Neuronas/metabolismo , Orexinas/genética , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Sueño/efectos de los fármacos , Privación de Sueño , Sueño REM/efectos de los fármacos
15.
J Pharmacol Exp Ther ; 371(1): 1-14, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31371483

RESUMEN

For the past 50 years, the clinical efficacy of antipsychotic medications has relied on blockade of dopamine D2 receptors. Drug development of non-D2 compounds, seeking to avoid the limiting side effects of dopamine receptor blockade, has failed to date to yield new medicines for patients. In this work, we report the discovery of SEP-363856 (SEP-856), a novel psychotropic agent with a unique mechanism of action. SEP-856 was discovered in a medicinal chemistry effort utilizing a high throughput, high content, mouse-behavior phenotyping platform, in combination with in vitro screening, aimed at developing non-D2 (anti-target) compounds that could nevertheless retain efficacy across multiple animal models sensitive to D2-based pharmacological mechanisms. SEP-856 demonstrated broad efficacy in putative rodent models relating to aspects of schizophrenia, including phencyclidine (PCP)-induced hyperactivity, prepulse inhibition, and PCP-induced deficits in social interaction. In addition to its favorable pharmacokinetic properties, lack of D2 receptor occupancy, and the absence of catalepsy, SEP-856's broad profile was further highlighted by its robust suppression of rapid eye movement sleep in rats. Although the mechanism of action has not been fully elucidated, in vitro and in vivo pharmacology data as well as slice and in vivo electrophysiology recordings suggest that agonism at both trace amine-associated receptor 1 and 5-HT1A receptors is integral to its efficacy. Based on the preclinical data and its unique mechanism of action, SEP-856 is a promising new agent for the treatment of schizophrenia and represents a new pharmacological class expected to lack the side effects stemming from blockade of D2 signaling. SIGNIFICANCE STATEMENT: Since the discovery of chlorpromazine in the 1950s, the clinical efficacy of antipsychotic medications has relied on blockade of dopamine D2 receptors, which is associated with substantial side effects and little to no efficacy in treating the negative and cognitive symptoms of schizophrenia. In this study, we describe the discovery and pharmacology of SEP-363856, a novel psychotropic agent that does not exert its antipsychotic-like effects through direct interaction with D2 receptors. Although the mechanism of action has not been fully elucidated, our data suggest that agonism at both trace amine-associated receptor 1 and 5-HT1A receptors is integral to its efficacy. Based on its unique profile in preclinical species, SEP-363856 represents a promising candidate for the treatment of schizophrenia and potentially other neuropsychiatric disorders.


Asunto(s)
Psicotrópicos/farmacología , Piranos/farmacología , Esquizofrenia/tratamiento farmacológico , Animales , Excitabilidad Cortical/efectos de los fármacos , Alucinógenos/toxicidad , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos C57BL , Fenciclidina/toxicidad , Psicotrópicos/uso terapéutico , Piranos/química , Piranos/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Esquizofrenia/etiología , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Agonistas del Receptor de Serotonina 5-HT1/uso terapéutico , Sueño REM/efectos de los fármacos
16.
Anesthesiology ; 130(6): 981-994, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30946702

RESUMEN

BACKGROUND: Previous studies suggest that rapid eye movement sleep rebound and disruption of rapid eye movement sleep architecture occur during the first 24 h after general anesthesia with volatile anesthetics in adult rats. However, it is unknown whether rapid eye movement sleep alterations persist beyond the anesthetic recovery phase in neonatal rats. This study tested the hypothesis that rapid eye movement sleep disturbances would be present in adolescent rats treated with anesthesia on postnatal day 7. METHODS: Forty-four neonatal rats were randomly allocated to treatment with anesthesia consisting of midazolam, nitrous oxide, and isoflurane or control conditions for 2 h or 6 h. Electroencephalographic and electromyographic electrodes were implanted and recordings obtained between postnatal days 26 and 34. The primary outcome was time spent in rapid eye movement sleep. Data were analyzed using two-tailed unpaired t tests and two-way repeated measures analysis of variance. RESULTS: Rats treated with midazolam, nitrous oxide, and isoflurane exhibited a significant increase in rapid eye movement sleep three weeks later when compared with control rats, regardless of whether they were treated for 2 h (174.0 ± 7.2 min in anesthetized, 108.6 ± 5.3 in controls, P < 0.0001) or 6 h (151.6 ± 9.9 min in anesthetized, 108.8 ± 7.1 in controls, P = 0.002). CONCLUSIONS: Treatment with midazolam, nitrous oxide, and isoflurane on postnatal day 7 increases rapid eye movement sleep three weeks later in rats.


Asunto(s)
Anestesia General/tendencias , Anestésicos por Inhalación/administración & dosificación , Anestésicos Intravenosos/administración & dosificación , Homeostasis/efectos de los fármacos , Sueño REM/efectos de los fármacos , Anestesia General/efectos adversos , Anestésicos por Inhalación/efectos adversos , Anestésicos Intravenosos/efectos adversos , Animales , Animales Recién Nacidos , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Femenino , Homeostasis/fisiología , Isoflurano/administración & dosificación , Isoflurano/efectos adversos , Masculino , Midazolam/administración & dosificación , Midazolam/efectos adversos , Óxido Nitroso/administración & dosificación , Óxido Nitroso/efectos adversos , Ratas , Ratas Sprague-Dawley , Sueño REM/fisiología
17.
Sleep Breath ; 23(2): 413-423, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30232681

RESUMEN

Rapid eye movement (REM) sleep was discovered nearly 60 years ago. This stage of sleep accounts for approximately a quarter of total sleep time in healthy adults, and it is mostly concentrated in the second half of the sleep period. The majority of research on REM sleep has focused on neurocognition. More recently, however, there has been a growing interest in understanding whether obstructive sleep apnea (OSA) during the two main stages of sleep (REM and non-REM sleep) leads to different cardiometabolic and neurocognitive risk. In this review, we discuss the growing evidence indicating that OSA during REM sleep is a prevalent disorder that is independently associated with adverse cardiovascular, metabolic, and neurocognitive outcomes. From a therapeutic standpoint, we discuss limitations of continuous positive airway pressure (CPAP) therapy given that 3 or 4 h of CPAP use from the beginning of the sleep period would leave 75% or 60% of obstructive events during REM sleep untreated. We also review potential pharmacologic approaches to treating OSA during REM sleep. Undoubtedly, further research is needed to establish best treatment strategies in order to effectively treat REM OSA. Moreover, it is critical to understand whether treatment of REM OSA will translate into better patient outcomes.


Asunto(s)
Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/terapia , Sueño REM , Animales , Clozapina/análogos & derivados , Clozapina/uso terapéutico , Presión de las Vías Aéreas Positiva Contínua , Relación Dosis-Respuesta a Droga , Dronabinol/uso terapéutico , Humanos , Evaluación de Procesos y Resultados en Atención de Salud , Faringe/efectos de los fármacos , Polisomnografía , Ensayos Clínicos Controlados Aleatorios como Asunto , Riesgo , Apnea Obstructiva del Sueño/fisiopatología , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Lengua/efectos de los fármacos
18.
Int J Mol Sci ; 20(3)2019 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-30708948

RESUMEN

Impaired sleep is both a risk factor and a symptom of depression. Objective sleep is assessed using the sleep electroencephalogram (EEG). Characteristic sleep-EEG changes in patients with depression include disinhibition of rapid eye movement (REM) sleep, changes of sleep continuity, and impaired non-REM sleep. Most antidepressants suppress REM sleep both in healthy volunteers and depressed patients. Various sleep-EEG variables may be suitable as biomarkers for diagnosis, prognosis, and prediction of therapy response in depression. In family studies of depression, enhanced REM density, a measure for frequency of rapid eye movements, is characteristic for an endophenotype. Cordance is an EEG measure distinctly correlated with regional brain perfusion. Prefrontal theta cordance, derived from REM sleep, appears to be a biomarker of antidepressant treatment response. Some predictive sleep-EEG markers of depression appear to be related to hypothalamo-pituitary-adrenocortical system activity.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Trastornos del Sueño-Vigilia/diagnóstico , Antidepresivos/farmacología , Depresión/complicaciones , Depresión/etiología , Depresión/genética , Electroencefalografía , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Trastornos del Sueño-Vigilia/complicaciones , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/prevención & control , Sueño REM/efectos de los fármacos
19.
Horm Behav ; 100: 20-28, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29501756

RESUMEN

The basolateral nucleus of the amygdala (BLA) plays a significant role in mediating individual differences in the effects of fear memory on sleep. Here, we assessed the effects of antagonizing corticotropin releasing factor receptor 1 (CRFR1) after shock training (ST) on fear-conditioned behaviors and sleep. Outbred Wistar rats were surgically implanted with electrodes for recording EEG and EMG and with bilateral guide cannulae directed at BLA. Data loggers were placed intraperitoneally to record core body temperature. The CRFR1 antagonist, antalarmin (ANT; 4.82 mM) was microinjected into BLA after shock training (ST: 20 footshocks, 0.8 mA, 0.5 s duration, 60 s interstimulus interval), and the effects on sleep, freezing and the stress response (stress-induced hyperthermia, SIH) were examined after ST and fearful context re-exposure alone at 7 days (CTX1) and 21 days (CTX2) post-ST. EEG and EMG recordings were scored for non-rapid eye movement sleep (NREM), rapid eye movement sleep (REM) and wakefulness. The rats were separated into 4 groups: Vehicle-vulnerable (Veh-Vul; n = 10), Veh-resilient (Veh-Res; n = 11), ANT-vulnerable (ANT-Vul; n = 8) and ANT-resilient (ANT-Res; n = 8) based on whether, compared to baseline, the rats showed a decrease or no change/increase in REM during the first 4 h following ST. Post-ST ANT microinjected into BLA attenuated the fear-conditioned reduction in REM in ANT-Vul rats on CTX1, but did not significantly alter REM in ANT-Res rats. However, compared to Veh treated rats, REM was reduced in ANT treated rats on CTX2. There were no group differences in freezing or SIH across conditions. Therefore, CRFR1 in BLA plays a role in mediating individual differences in sleep responses to stress and in the extinction of fear conditioned changes in sleep.


Asunto(s)
Adaptación Psicológica/efectos de los fármacos , Complejo Nuclear Basolateral/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Miedo/efectos de los fármacos , Reacción Cataléptica de Congelación/efectos de los fármacos , Pirimidinas/farmacología , Pirroles/farmacología , Sueño/efectos de los fármacos , Animales , Complejo Nuclear Basolateral/metabolismo , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Electroencefalografía , Miedo/psicología , Masculino , Memoria/efectos de los fármacos , Condicionamiento Físico Animal/fisiología , Condicionamiento Físico Animal/psicología , Ratas , Ratas Wistar , Sueño/fisiología , Sueño REM/efectos de los fármacos
20.
J Neurochem ; 141(1): 111-123, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28027399

RESUMEN

Rapid eye movement (REM) sleep dysregulation is a symptom of many neuropsychiatric disorders, yet the mechanisms of REM sleep homeostatic regulation are not fully understood. We have shown that, after REM sleep deprivation, the pedunculopontine tegmental nucleus (PPT) plays a critical role in the generation of recovery REM sleep. In this study, we used multidisciplinary techniques to show a causal relationship between brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the PPT and the development of REM sleep homeostatic drive. Rats were randomly assigned to conditions of unrestricted sleep or selective REM sleep deprivation (RSD) with PPT microinjections of vehicle control or a dose of a TrkB receptor inhibitor (2, 3, or 4 nmol K252a or 4 nmol ANA-12). On experimental days, rats received PPT microinjections and their sleep-wake physiological signals were recorded for 3 or 6 h, during which selective RSD was performed in the first 3 h. At the end of all 3 h recordings, rats were killed and the PPT was dissected out for BDNF quantification. Our results show that K252a and ANA-12 dose-dependently reduced the homeostatic responses to selective RSD. Specifically, TrkB receptor inhibition reduced REM sleep homeostatic drive and limited REM sleep rebound. There was also a dose-dependent suppression of PPT BDNF up-regulation, and regression analysis revealed a significant positive relationship between REM sleep homeostatic drive and the level of PPT BDNF expression. These data provide the first direct evidence that activation of BDNF-TrkB signaling in the PPT is a critical step for the development of REM sleep homeostatic drive.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Homeostasis/fisiología , Núcleo Tegmental Pedunculopontino/metabolismo , Receptor trkB/metabolismo , Transducción de Señal/fisiología , Sueño REM/fisiología , Animales , Carbazoles/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Homeostasis/efectos de los fármacos , Alcaloides Indólicos/farmacología , Masculino , Núcleo Tegmental Pedunculopontino/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos , Vigilia/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA