Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Food Funct ; 13(24): 12697-12706, 2022 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-36408594

RESUMEN

Turmeric (Curcuma longa) had been considered as a universal panacea in functional foods and traditional medicines. In recent, the sedative-hypnotic effect of turmeric extract (TE) was reported. However, sleep-promoting compounds in TE have been not yet demonstrated. Curcuminoids (curcumin, demethoxycurcumin, and bisdemethoxycurcumin) are the major constituents of turmeric being responsible for its various biological activities. Therefore, they can be first assumed to be sedative-hypnotic compounds of TE. In the present study, we aimed to investigate the effects and underlying mechanisms of curcuminoids and each constituent on the sleep-wake cycle of mice. Molecular docking studies, histamine H1 receptor (H1R) binding assays, and H1R knockout animal studies were used to investigate the molecular mechanisms underlying the sleep-promoting effects. Curcuminoids and their constituents reduced sleep latency and increased sleep duration in the pentobarbital-induced sleep test in mice. In addition, curcuminoids significantly increased the duration of NREMS and reduced sleep latency without altering the REMS and delta activity. Curcumin, demethoxycurcumin, and bisdemethoxycurcumin were predicted to interact with H1R in the molecular model. In the binding affinity assay, we found that curcuminoids, as well as their constituents, significantly bind to H1R with the Ki value of 1.49 µg mL-1. Furthermore, sleep latency was reduced and NREMS frequency was increased following curcuminoid administration in wild-type mice but not in H1R knockout mice. Therefore, we conclude that curcuminoids reduce sleep latency and enhance the quantity of NREMS by acting as modulators of H1R, indicating their usefulness in treating insomnia.


Asunto(s)
Curcuma , Curcumina , Diarilheptanoides , Receptores Histamínicos H1 , Fármacos Inductores del Sueño , Latencia del Sueño , Sueño REM , Animales , Ratones , Curcuma/química , Curcumina/química , Curcumina/farmacología , Diarilheptanoides/farmacología , Simulación del Acoplamiento Molecular , Extractos Vegetales/química , Extractos Vegetales/farmacología , Receptores Histamínicos H1/genética , Receptores Histamínicos H1/metabolismo , Latencia del Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Fármacos Inductores del Sueño/química , Fármacos Inductores del Sueño/farmacología
2.
J Ethnopharmacol ; 285: 114866, 2022 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-34822960

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Jiaotaiwan (JTW) is good at communicating the heart and kidney. That meets the main mechanism of insomnia in traditional Chinese medicine. But the mechanism of JTW in promoting sleep is not clear. AIM OF THE STUDY: To investigate the mechanism of JTW in promoting sleep and identify the main components. MATERIALS AND METHODS: In this study, we detected the levels of GABA in serum and brain via LC-MS/MS analysis and investigated the hypnotic effect of JTW and its role in promoting sleep via Sleep monitoring and vigilance state analysis. Further, the identification of the main components was carried out by using LC-MS/MS analysis. RESULTS: JTW could increase the GABA levels in serum, FC and BS of SDM rats. JTW reduced the amount of wakefulness, increased the time of NREM sleep and REM sleep. A total of 25 compounds were identified. CONCLUSIONS: The current work provides valuable information on the hypnotic effects of JTW and its regulatory mechanisms in promoting sleep.


Asunto(s)
Encéfalo/efectos de los fármacos , Medicamentos Herbarios Chinos , Sueño REM/efectos de los fármacos , Sueño/efectos de los fármacos , Ácido gamma-Aminobutírico/sangre , Ácido gamma-Aminobutírico/metabolismo , Animales , Encéfalo/metabolismo , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico
3.
J Ethnopharmacol ; 265: 113316, 2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-32866569

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Calea zacatechichi is a plant with an extensive popular and ritual use in Mexico. In healthy volunteers, it induces well-being and tranquility senses, and facilitates superficial stages of sleep. However, anxiolytic, and antidepressant-like effects and changes on the sleep-waking stages have not been explored. AIM: To determine anxiolytic and antidepressant-like effects of an aqueous extract of C. zacatechichi (CZ) in rodents and to analyze their effects on hippocampal activity in the rat sleep-waking cycle. MATERIAL AND METHODS: CZ anxiolytic- and antidepressant-like effects were evaluated in several mice and rat behavioral paradigms. CZ effects on temporal distribution of sleep were described, and hippocampus EEG frequency patterns were analyzed during the sleep-waking cycle; absolute and relative powers were analyzed during Rapid Eye Movements (REM) and non-REM sleep stages. CZ chemical analysis was performed by UPLC-ESI-MS. RESULTS: CZ produced specific and robust anxiolytic- and antidepressant-like effects in mice and rats, similar to those of prototypical drugs, at doses ranging from 0.5 to 50 mg/kg. CZ at 100 mg/kg produced visible mild sedative effects in rats, associated with a significant increase in Slow Wave Sleep episodes during a 6 h recording, and enhanced fast frequencies of hippocampus (gamma-band:31-50 Hz) during REM sleep. CONCLUSION: Results could support the well-being and tranquility senses reported by healthy consumers, and to explain the oneiric content during dreams and some improvements in cognitive processes described by consumers. Anxiolytic- and antidepressant-like effects of this species, reported for first time in this study could improve some aspects of mental health.


Asunto(s)
Ansiolíticos/farmacología , Antidepresivos/farmacología , Asteraceae/química , Extractos Vegetales/farmacología , Animales , Ansiolíticos/administración & dosificación , Ansiolíticos/aislamiento & purificación , Antidepresivos/administración & dosificación , Antidepresivos/aislamiento & purificación , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , México , Ratones , Extractos Vegetales/administración & dosificación , Ratas , Ratas Wistar , Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos
4.
Biomed Res Int ; 2020: 3636874, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33123570

RESUMEN

Previous studies have shown the therapeutic properties of ginseng and ginsenosides on hyperactive and impulsive behaviors in several psychiatric diseases. Herein, we investigated the effect of Panax ginseng Meyer (PG) on hyperactive/impulsive behaviors in a manic-like animal model, sleep deprivation (SD) rats. Male rats were sleep-deprived for 48 h, and PG (200 mg/kg) was administered for 4 days, from 2 days prior to the start of SD to the end date of SD. The elevated plus maze (EPM) test showed that PG alleviated the increased frequency of entries into and spent time within open arms by SD. In order to investigate the molecular mechanism on this effect of PG, we assessed differentially expressed genes (DEGs) in the prefrontal cortex of PG-treated SD rats using RNA sequencing (RNA-seq) and performed gene-enrichment analysis for DEGs. The gene-enrichment analysis showed that PG most prominently affected the glutamatergic synapse pathway. Among the glutamatergic synapse pathway genes, particularly, PG enhanced the expressions of glutamate transporter Slc1a3 and Slc1a2 reduced in SD rats. Moreover, we found that PG could inhibit the SD-induced phosphorylation of the NR2A subunit of the NMDA receptor. These results suggested that PG might have a therapeutic effect against the manic-like behaviors, regulating the glutamatergic neurotransmission.


Asunto(s)
Antimaníacos/farmacología , Ginsenósidos/farmacología , Ácido Glutámico/metabolismo , Panax/química , Privación de Sueño/tratamiento farmacológico , Sueño REM/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Conducta Animal , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Proteínas de Transporte de Glutamato en la Membrana Plasmática/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Privación de Sueño/metabolismo , Sinapsis/efectos de los fármacos , Sinapsis/metabolismo
5.
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
6.
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
7.
Biomed Res Int ; 2019: 6198067, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31183370

RESUMEN

Pinelliae Rhizoma Praeparatum Cum Alumine (PRPCA) is useful for eliminating dampness and phlegm in clinical settings, targeting the main mechanisms of insomnia as defined in traditional Chinese medicine. However, little is known regarding the sedative and hypnotic effects of PRPCA. In the present study, we examined the sedative effects of PRPCA via a locomotor activity test and aimed to determine the most appropriate concentration of PRPCA for achieving these effects. The strongest sedative effects were observed at a PRPCA concentration of 0.45 g/ml. In addition, we investigated the hypnotic effects of PRPCA and its role in promoting sleep via sleep monitoring and vigilance state analysis. PRPCA increased rapid eye movement (REM) sleep and non-REM (NREM) sleep while decreasing wakefulness. In addition, PRPCA decreased the number of bouts of wakefulness (16-32 s and 32-64 s) and increased the number of bouts of NREM sleep (128-256 s). Furthermore, we identified a total of 32 component compounds via chromatography and mass spectrometry. Hence, the current work provides valuable information regarding the sedative and hypnotic effects of PRPCA and its regulatory mechanisms in promoting sleep.


Asunto(s)
Hipnóticos y Sedantes/farmacología , Locomoción/efectos de los fármacos , Pinellia/química , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Animales , Electroencefalografía , Humanos , Hipnóticos y Sedantes/química , Espectrometría de Masas , Medicina Tradicional China , Ratones , Extractos Vegetales/química , Extractos Vegetales/farmacología , Sueño/efectos de los fármacos , Trastornos del Inicio y del Mantenimiento del Sueño/patología , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos
8.
Nat Prod Res ; 33(22): 3326-3329, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29764203

RESUMEN

Pinellia ternata (Thunb.) Makino Preparation (PTP) is widely used to treat insomnia in traditional Chinese medicine; however, its specific role is not clear. In this study, PTP was prepared at three concentrations. For locomotor activity tests, mice were treated with PTP and evaluated for 14 days. For polygraph recordings, mice were treated for 14 days and recorded after treatment. The main chemical constituents in PTP were identified by Ultra performance liquid chromatography/quadrupole time spectrometry (UPLC/Q-TOF-MS). The results showed that 0.9 g/mL PTP significantly reduced locomotor activity. The effect was related to the time of treatment. PTP reduced wakefulness and increased sleep in mice. Furthermore, PTP promoted sleep by increasing the number of REM sleep episodes with a duration of 64-128s and increasing the number of transitions from NREM sleep to REM sleep and from REM sleep to wakefulness. A total of 17 compounds were identified.


Asunto(s)
Medicina Tradicional China , Pinellia/química , Sueño REM/efectos de los fármacos , Animales , Cromatografía Liquida/métodos , Medicamentos Herbarios Chinos/química , Locomoción/efectos de los fármacos , Masculino , Espectrometría de Masas/métodos , Ratones , Sueño/efectos de los fármacos
9.
Biomed Pharmacother ; 99: 913-920, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29710491

RESUMEN

The aim of this study was to investigate the beneficial effect of Valerian/Cascade mixture on sleeping in mammal models. In pentobarbital-induced sleep model, Valerian, Cascade, and Valerian/Cascade mixture significantly reduced the latency time for sleeping, and total sleeping time effectively increased in these sample groups compared with the control. Valerian/Cascade mixture increased sleep duration by 37%. The mixture significantly increased the non-rapid eye movement (NREM) sleep time by 53% compared with the control, while REM sleeping time was decreased by 33% with Valerian/Cascade mixture, in Electroencephalography (EEG) analysis, resulting in the increase of total sleep time and the decrease of awakening. This sleep-promoting effect was obvious in caffeine-induced awakening model; Valerian, Cascade, and the mixture significantly enhanced NREM and total sleep time, which were reduced by caffeine. Caffeine-induced increase of awakening was effectively deceased to the normal level by these three samples. In particular, delta wave responsible for deep sleep in NREM was greatly increased by the mixture in both normal and caffeine-induced awake models. This sleep-promoting effect of Valerian/Cascade mixture was shown to be due to the upregulation of gamma-aminobutyric acid A receptor (GABAAR). Valerian/Cascade mixture showed 91% binding capacity to GABAA-BZD receptor. Two compounds, Valerenic acid and Xanthohumol, were shown to significantly contribute to the binding activity of Valerian/Cascade mixture on the GABA receptor.


Asunto(s)
Humulus/química , Extractos Vegetales/farmacología , Sueño/efectos de los fármacos , Valeriana/química , Animales , Cafeína/farmacología , Electroencefalografía , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/aislamiento & purificación , Hipnóticos y Sedantes/farmacología , Ratones , Ratones Endogámicos ICR , Extractos Vegetales/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/efectos de los fármacos , Receptores de GABA-A/metabolismo , Fases del Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
10.
J Sleep Res ; 26(3): 377-385, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28397310

RESUMEN

Sleep has been postulated to promote brain energy restoration. It is as yet unknown if increasing the energy availability within the brain reduces sleep need. The guanidine amino acid creatine (Cr) is a well-known energy booster in cellular energy homeostasis. Oral Cr-monohydrate supplementation (CS) increases exercise performance and has been shown to have substantial effects on cognitive performance, neuroprotection and circadian rhythms. The effect of CS on cellular high-energy molecules and sleep-wake behaviour is unclear. Here, we examined the sleep-wake behaviour and brain energy metabolism before and after 4-week-long oral administration of CS in the rat. CS decreased total sleep time and non-rapid eye movement (NREM) sleep significantly during the light (inactive) but not during the dark (active) period. NREM sleep and NREM delta activity were decreased significantly in CS rats after 6 h of sleep deprivation. Biochemical analysis of brain energy metabolites showed a tendency to increase in phosphocreatine after CS, while cellular adenosine triphosphate (ATP) level decreased. Microdialysis analysis showed that the sleep deprivation-induced increase in extracellular adenosine was attenuated after CS. These results suggest that CS reduces sleep need and homeostatic sleep pressure in rats, thereby indicating its potential in the treatment of sleep-related disorders.


Asunto(s)
Creatina/farmacología , Homeostasis/efectos de los fármacos , Sueño/efectos de los fármacos , Sueño/fisiología , Adenosina/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Creatina/administración & dosificación , Electroencefalografía , Metabolismo Energético/efectos de los fármacos , Masculino , Microdiálisis , Fosfocreatina/metabolismo , Ratas , Ratas Sprague-Dawley , Privación de Sueño/tratamiento farmacológico , Privación de Sueño/metabolismo , Sueño REM/efectos de los fármacos , Sueño REM/fisiología
11.
Phytomedicine ; 23(14): 1797-1805, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27912882

RESUMEN

BACKGROUND: Radix Polygalae, the dried root of Polygala tenuifolia, has been extensively used as a traditional Chinese medicine for promoting intelligence and tranquilization. Polygalasaponins extracted from the root of P. tenuifolia possess evident anxiolytic and sedative-hypnotic activities. Previous studies have reported that tenuifolin was a major constituent of polygalasaponins. PURPOSE: The currently study aims to investigate the hypnotic effect and possible mechanism of tenuifolin in freely moving mice. DESIGN/METHODS: The hypnotic effects of tenuifolin (20, 40 and 80mg/kg, p.o.) were assessed by electroencephalographic (EEG) and electromyographic (EMG) analysis. Double-staining immunohistochemistry test was performed to evaluate the neuronal activity of sleep-wake regulating brain areas. High performance liquid chromatograph- electrochemical detection (HPLC-ECD) and ultrafast liquid chromatography-mass spectrometry (UFLC-MS) were used for the detection of neurotransmitters. Locomotor activity was measured by Open-field Test. RESULTS: Tenuifolin at doses of 40 and 80mg/kg (p.o.) significantly prolonged the total sleep time by increasing the amount of non-rapid eye movement (NREM) and rapid eye movement (REM) sleep, associated with the significant increase in the bouts of episodes respectively. After administration of tenuifolin, the cortical EEG power spectral densities during NREM and REM sleep were similar to that of natural sleep (vehicle) and thus compatible with physiological sleep. Double-immunohistochemistry staining test showed that tenuifolin increased the c-Fos positive ratios of GABAergic NREM sleep-promoting neurons in ventrolateral preoptic area (VLPO), cholinergic REM sleep-promoting neurons in laterodorsal tegmental area (LDT) and pontomesencephalic tegmental area (PPT) and decreased the c-Fos positive ratios in wake-promoting neurons (locus coeruleus (LC) and perifornical area (Pef)). Neurotransmitter detections revealed that tenuifolin significantly reduced the noradrenaline (NA) levels in LC, VLPO, PPT and LDT, elevated the GABA levels in VLPO, LC and Pef and increased the acetylcholine (Ach) levels in LDT and PPT. In addition, tenuifolin did not cause any change to locomotor activity. CONCLUSION: Taken together, these results provide the first experimental evidence of the significant sleep-enhancing effect of tenuifolin in mice. This effect appears to be mediated, at least in part, by the activation of GABAergic systems and/or by the inhibition of noradrenergic systems. Moreover, this study adds new scientific evidence and highlights the therapeutic potential of the medicinal plant P. tenuifolia in the development of phytomedicines with hypnotic properties.


Asunto(s)
Encéfalo/efectos de los fármacos , Diterpenos de Tipo Kaurano/farmacología , Hipnóticos y Sedantes/farmacología , Extractos Vegetales/farmacología , Polygala/química , Saponinas/farmacología , Sueño/efectos de los fármacos , Acetilcolina/metabolismo , Animales , Ansiolíticos/farmacología , Encéfalo/metabolismo , Electroencefalografía , Masculino , Ratones Endogámicos ICR , Neurotransmisores/metabolismo , Raíces de Plantas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sueño REM/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
12.
Mol Pharmacol ; 90(5): 530-539, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27573669

RESUMEN

α-Pinene is a major monoterpene of the pine tree essential oils. It has been reported that α-pinene shows anxiolytic and hypnotic effects upon inhaled administration. However, hypnotic effect by oral supplementation and the molecular mechanism of α-pinene have not been determined yet. By combining in vivo sleep behavior, ex vivo electrophysiological recording from brain slices, and in silico molecular modeling, we demonstrate that (-)-α-pinene shows sleep enhancing property through a direct binding to GABAA-benzodiazepine (BZD) receptors by acting as a partial modulator at the BZD binding site. The effect of (-)-α-pinene on sleep-wake profiles was evaluated by recording electroencephalogram and electromyogram. The molecular mechanism of (-)-α-pinene was investigated by electrophysiology and molecular docking study. (-)-α-pinene significantly increased the duration of non-rapid eye movement sleep (NREMS) and reduced the sleep latency by oral administration without affecting duration of rapid eye movement sleep and delta activity. (-)-α-pinene potentiated the GABAA receptor-mediated synaptic response by increasing the decay time constant of sIPSCs in hippocampal CA1 pyramidal neurons. These effects of (-)-α-pinene on sleep and inhibitory synaptic response were mimicked by zolpidem, acting as a modulator for GABAA-BZD receptors, and fully antagonized by flumazenil, an antagonist for GABAA-BZD receptor. (-)-α-pinene was found to bind to aromatic residues of α1- and -γ2 subunits of GABAA-BZD receptors in the molecular model. We conclude that (-)-α-pinene enhances the quantity of NREMS without affecting the intensity of NREMS by prolonging GABAergic synaptic transmission, acting as a partial modulator of GABAA-BZD receptors and directly binding to the BZD binding site of GABAA receptor.


Asunto(s)
Benzodiazepinas/metabolismo , Movimientos Oculares/efectos de los fármacos , Monoterpenos/farmacología , Pinus/química , Aceites de Plantas/farmacología , Receptores de GABA-A/metabolismo , Sueño/efectos de los fármacos , Animales , Monoterpenos Bicíclicos , Sitios de Unión , Flumazenil/química , Flumazenil/farmacología , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Modelos Moleculares , Monoterpenos/química , Pentobarbital , Piridinas/química , Piridinas/farmacología , Sueño REM/efectos de los fármacos , Factores de Tiempo , Vigilia/efectos de los fármacos , Zolpidem
13.
J Med Food ; 19(5): 472-80, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26870900

RESUMEN

Sweet dream oral liquid (SDOL), a traditional Chinese herbal compound contains 17 traditional Chinese medicines. It has various pharmacological effects, such as improving brain dysfunction and increasing sleeping quality. This study investigated the neuroprotective effect and the underlying mechanisms of SDOL-impaired hippocampus learning and memory-induced paradoxical sleep deprivation (PSD) in rats. Sixty Male Wistar rats were randomly divided into six groups. Before PSD, SDOL treatment group rats were intragastrically administered SDOL for 25 days at dose of 2.1, 4.2, and 8.4 mL/kg body weight per day. Normal control group, large platform control group, and PSD groups were treated with normal saline instead of SDOL. After 25 days treatment, PSD and SDOL groups were deprived of paradoxical sleep for 72 h. Then two behavioral studies were conducted to test the spatial learning and memory ability using the open field test and Morris water maze test. Expression of the c-fos, c-jun, cyclic AMP response element binding protein (CREB), extracellular signal-regulated protein kinase (ERK), mitogen-activated protein kinases (MAPK)/ERK kinase (MEK), and p-CREB, p-ERK, and p-MEK in the hippocampus were also assayed by western blot. In this study, PSD decreased the levels of p-CREB, p-ERK, p-MEK, c-fos, and c-jun. However, SDOL treatment increased expressions of these proteins. Our results showed that SDOL improved 72-h PSD-induced cognitive impairment. These affects may be mediated by increasing the contents of c-fos, c-jun, and p-CREB/ERK signaling.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Privación de Sueño/tratamiento farmacológico , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Aprendizaje/efectos de los fármacos , Masculino , Memoria/efectos de los fármacos , Trastornos de la Memoria , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Privación de Sueño/metabolismo , Privación de Sueño/fisiopatología , Privación de Sueño/psicología , Sueño REM/efectos de los fármacos
14.
Phytomedicine ; 22(11): 1000-8, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26407942

RESUMEN

BACKGROUND: Many antidepressants are effective in alleviating ethanol withdrawal symptoms. However, most of them suppress rapid eye movement (REM) sleep. Thus, development of antidepressants without undesirable side effects would be preferable. Previously, crude alkaloid extract from Mitragyna speciosa (MS) Korth was found to produce antidepressant activities. It was hypothesized that the alkaloid extract from MS may attenuate ethanol withdrawal without REM sleep disturbance. METHODS: Adult male Wistar rats implanted with electrodes over the frontal and parietal cortices were used for two separated studies. For an acute study, 10 mg/kg fluoxetine or 60 mg/kg alkaloid extract from MS were administered intragastrically. Electroencephalographic (EEG) signals were recorded for 3 h to examine sleep profiles and EEG fingerprints. Another set of animal was used for an ethanol withdrawal study. They were rendered dependent on ethanol via a modified liquid diet (MLD) containing ethanol ad libitum for 28 days. On day 29, fluoxetine (10 mg/kg) or alkaloid extract from MS (60 mg/kg) were administered 15 min before the ethanol-containing MLD was replaced with an isocaloric ethanol-free MLD to induced ethanol withdrawal symptoms. RESULTS: The sleep analysis revealed that alkaloid extract from MS did not change any REM parameters which included average duration of each REM episode, total REM time, number of REM episode and REM latency whereas fluoxetine significantly suppressed all REM parameters and delayed REM latency. However, power spectral analysis revealed similar fingerprints for fluoxetine and alkaloid extract from MS characterized by decreasing powers in the slow frequency range in frontal and parietal cortical EEG. Neither treatment affected spontaneous motor activity. Finally, both alkaloid extract from MS and fluoxetine were found to significantly attenuate ethanol withdrawal-induced hyperexcitability (increases gamma activity) in both cortices and to reduce locomotor activity. CONCLUSION: The present study demonstrated that the alkaloid extract from MS alleviates ethanol withdrawal severity with no side effect on REM sleep. In addition, these data suggest that suppressive effects on slow frequency powers but not REM sleep may be hallmarks of effective antidepressants for ethanol withdrawal treatment.


Asunto(s)
Alcaloides/farmacología , Fluoxetina/farmacología , Mitragyna/química , Extractos Vegetales/farmacología , Sueño REM/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Animales , Electrodos , Electroencefalografía , Etanol/efectos adversos , Masculino , Hojas de la Planta/química , Ratas , Ratas Wistar
15.
Mol Nutr Food Res ; 59(10): 2087-93, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26105624

RESUMEN

Zinc is an essential trace element for humans and animals, being located, among other places, in the synaptic vesicles of cortical glutamatergic neurons and hippocampal mossy fibers in the brain. Extracellular zinc has the potential to interact with and modulate many different synaptic targets, including glutamate and GABA receptors. Because of the central role of these neurotransmitters in brain activity, we examined in this study the sleep-promoting activity of zinc by monitoring locomotor activity and electroencephalogram after its administration to mice. Zinc-containing yeast extract (40 and 80 mg/kg) dose dependently increased the total amount of nonrapid eye movement sleep and decreased the locomotor activity. However, this preparation did not change the amount of rapid eye movement sleep or show any adverse effects such as rebound of insomnia during a period of 24 h following the induction of sleep; whereas the extracts containing other divalent cations (manganese, iron, and copper) did not decrease the locomotor activity. This is the first evidence that zinc can induce sleep. Our data open the way to new types of food supplements designed to improve sleep.


Asunto(s)
Movimientos Oculares/efectos de los fármacos , Sueño/efectos de los fármacos , Levaduras/química , Zinc/farmacología , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Electroencefalografía , Masculino , Ratones Endogámicos C57BL , Sueño/fisiología , Sueño REM/efectos de los fármacos
16.
Psychopharmacology (Berl) ; 232(21-22): 3977-89, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25902875

RESUMEN

While treatment options are available, excessive daytime sleepiness (EDS) remains a significant unmet medical need for many patients. Relatively little rodent behavioural pharmacology has been conducted in this context to assess potential pro-vigilant compounds for their ability to restore functional capacity following experimentally induced sleep loss. Male Wistar rats were prepared for electroencephalographic (EEG) recording and subject to 11 h of sleep restriction using a biofeedback-induced cage rotation protocol. A simple response latency task (SRLT) was used to behaviourally index sleep restriction and the effects of pro-vigilant compounds: modafinil, D-amphetamine, caffeine, and the mGlu5-positive allosteric modulator LSN2814617. Sleep restriction resulted in a consistent, quantified loss of non-rapid eye movement (NREM) and REM sleep that impaired SRLT performance in a manner suggestive of progressive task disengagement. In terms of EEG parameters, all compounds induced wakefulness. Amphetamine treatment further decreased SRLT performance capacity, whereas the other three compounds decreased omissions and allowed animals to re-engage in the task. Caffeine and modafinil also significantly increased premature responses during this period, an effect not observed for LSN2814617. While all compounds caused compensatory sleep responses, the magnitude of compensation observed for LSN2814617 was much smaller than would be predicted to result from the prolongation of wakefulness exhibited. Using simple response latencies to index performance, an mGlu5 PAM dramatically increased wakefulness and improved functional capacity of sleep-restricted animals, without eliciting a proportionate compensatory sleep response. This effect was qualitatively distinct from that of amphetamine, caffeine and modafinil.


Asunto(s)
Nivel de Alerta/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Oxadiazoles/farmacología , Piridinas/farmacología , Receptor del Glutamato Metabotropico 5/agonistas , Triazoles/farmacología , Animales , Compuestos de Bencidrilo/farmacología , Biorretroalimentación Psicológica , Cafeína/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Condicionamiento Operante/efectos de los fármacos , Dextroanfetamina/farmacología , Electroencefalografía/efectos de los fármacos , Masculino , Modafinilo , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Sueño/efectos de los fármacos , Privación de Sueño , Sueño REM/efectos de los fármacos
18.
Sleep ; 38(1): 73-84, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25325438

RESUMEN

STUDY OBJECTIVES: Dexmedetomidine is used clinically to induce states of sedation that have been described as homologous to nonrapid eye movement (NREM) sleep. A better understanding of the similarities and differences between NREM sleep and dexmedetomidine-induced sedation is essential for efforts to clarify the relationship between these two states. This study tested the hypothesis that dexmedetomidine-induced sedation is homologous to sleep. DESIGN: This study used between-groups and within-groups designs. SETTING: University of Michigan. PARTICIPANTS: Adult male Sprague Dawley rats (n = 40). INTERVENTIONS: Independent variables were administration of dexmedetomidine and saline or Ringer's solution (control). Dependent variables included time spent in states of wakefulness, sleep, and sedation, electroencephalographic (EEG) power, adenosine levels in the substantia innominata (SI), and activation of pCREB and c-Fos in sleep related forebrain regions. MEASUREMENTS AND RESULTS: Dexmedetomidine significantly decreased time spent in wakefulness (-49%), increased duration of sedation (1995%), increased EEG delta power (546%), and eliminated the rapid eye movement (REM) phase of sleep for 16 h. Sedation was followed by a rebound increase in NREM and REM sleep. Systemically administered dexmedetomidine significantly decreased (-39%) SI adenosine levels. Dialysis delivery of dexmedetomidine into SI did not decrease adenosine level. Systemic delivery of dexmedetomidine did not alter c-Fos or pCREB expression in the horizontal diagonal band, or ventrolateral, median, and medial preoptic areas of the hypothalamus. CONCLUSIONS: Dexmedetomidine significantly altered normal sleep phenotypes, and the dexmedetomidine-induced state did not compensate for sleep need. Thus, in the Sprague Dawley rat, dexmedetomidine-induced sedation is characterized by behavioral, electrographic, and immunohistochemical phenotypes that are distinctly different from similar measures obtained during sleep.


Asunto(s)
Dexmedetomidina/farmacología , Hipnóticos y Sedantes/farmacología , Fenotipo , Sueño/fisiología , Adenosina/metabolismo , Animales , Prosencéfalo Basal/efectos de los fármacos , Prosencéfalo Basal/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Dexmedetomidina/administración & dosificación , Diálisis , Electroencefalografía , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley , Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Sustancia Innominada/efectos de los fármacos , Sustancia Innominada/metabolismo , Factores de Tiempo , Vigilia/efectos de los fármacos , Vigilia/fisiología
19.
J Tradit Chin Med ; 34(5): 572-5, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25417408

RESUMEN

OBJECTIVE: To investigate the effects of the Sini San at different doses on each sleeping state [slow-wave sleep 1 (SWS 1), slow-wave sleep 2 (SWS2), rapid-eye-movement (REM), wakefulness (W)] in insomnia rats and to identify its mode of action for improving sleep. METHODS: The insomnia rats were randomly divided into a high-, medium- or low-dose group of Sini San (equal to crude drug 8.8, 4.4, or 2.2 g/kg, respectively) for seven consecutive days. RESULTS: Compared with pre-administration, SWS2 was significantly increased after administration of the low dose. Compared with pre-administration, W was significantly decreased and SWS1, SWS2, and the total sleeping time (TST) were markedly increased after administration of the medium dose. Compared with pre-administration, W was significantly decreased and SWS1, SWS2, rapid-eye-movement sleep, and TST were significantly longer after administration of the high dose. The effects of Sini San on sleep-wake cycle are dose-dependent. CONCLUSION: The results suggest that Sini San extends SWS1 and SWS2, which increases the total sleeping time.


Asunto(s)
Medicamentos Herbarios Chinos/administración & dosificación , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología , Animales , Humanos , Masculino , Ratas , Ratas Wistar , Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos
20.
Neuroscience ; 253: 1-8, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-23973403

RESUMEN

Sleep is an essential physiological process that underlies crucial cognitive functions as well as emotional reactivity. Thus, sleep deprivation (SD) may exert various deleterious effects. In this study, we aimed to examine the adverse behavioral and hormonal effects of SD and a potential treatment with Plant-derived nanoparticle treatment - cocc 30c. The study was a 4-arm trial with randomization and double-blinding of verum and placebo treatments. SD was induced by using the Multiple Platform Method for 48 h. The effects of SD were evaluated behaviorally (pre-pulse inhibition (PPI), startle response and rotor-rod) at baseline as well as at 6, 12, 24h, and 14 days post deprivation. cocc 30c treatment was administrated Per Os every three hours starting immediately after baseline tests and for a period of 24h. On day 14, blood samples were taken and serum levels of corticosterone, testosterone, serotonin and leptin were tested. We found that cocc 30c improved PPI 12 and 24h post deprivation, likewise, cocc 30c improved motor learning. On day 14 SD led to increased startle response that was ameliorated by cocc 30c. Likewise, SD led to increased levels of corticosterone and serotonin while decreasing testosterone and leptin. Interestingly, cocc 30c treatment has moderated these hormonal alterations. We conclude that the treatment with cocc 30c recovers both short-term behavioral and the long-term hormonal modulations following SD.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/tratamiento farmacológico , Trastornos del Movimiento/tratamiento farmacológico , Nanopartículas/uso terapéutico , Fitoterapia/métodos , Privación de Sueño/complicaciones , Estimulación Acústica , Animales , Trastorno por Déficit de Atención con Hiperactividad/etiología , Cocculus/química , Modelos Animales de Enfermedad , Método Doble Ciego , Masculino , Trastornos del Movimiento/etiología , Extractos Vegetales/uso terapéutico , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Reflejo de Sobresalto/efectos de los fármacos , Privación de Sueño/sangre , Sueño REM/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA