Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.545
Filtrar
1.
Brain Res ; 1841: 149096, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-38936532

RESUMEN

Drug addiction may result in sleep problems. Importantly, sleep deprivation (SD) is known as an important risk factor for relapse to drug abuse as SD mimics the effects of psychostimulants on dopamine system of the brain. Moreover, aging may affect sleep and drug addiction. This study, therefore, set out to assess the effects of methamphetamine (METH) and REM sleep deprivation (RSD) on locomotor activity, anxiety-like behavior and spatial memory in adult and adolescent rats. Adult and adolescent male Wistar rats received a neurotoxic METH regimen; four subcutaneous injections of 6 mg/kg, at 2 h intervals. Five days later, the animals underwent a 48-h RSD episode using the multiple platforms method. They were then examined using the open field (OF), elevated plus maze (EPM) and Y-maze tasks. We found that the METH and RSD paradigms showed synergistic effects to increase locomotion and risk-taking behavior in both adult and adolescent animals, while only adolescent rats revealed RSD-induced anxiety-like behavior. Moreover, adolescent animals revealed greater sensitization for vertical activity following METH plus RSD episode. In addition, METH and RSD paradigms revealed synergistic effects to impair spatial working memory, but neither METH nor RSD alone affected performance of animals in the Y-maze task. Our findings may indicate that there are important relationships between METH and RSD to induce hyperlocomotion, risk-taking behavior and spatial memory impairment, particularly in adolescent animals. Moreover, it seems that adolescent rats may be more susceptible to anxiety-like behavior and hyperlocomotion than adults.


Asunto(s)
Ansiedad , Estimulantes del Sistema Nervioso Central , Aprendizaje por Laberinto , Metanfetamina , Ratas Wistar , Privación de Sueño , Animales , Metanfetamina/farmacología , Masculino , Estimulantes del Sistema Nervioso Central/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Ratas , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Locomoción/efectos de los fármacos , Locomoción/fisiología , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Actividad Motora/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Envejecimiento/fisiología , Envejecimiento/efectos de los fármacos
2.
Sci Rep ; 14(1): 14060, 2024 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890405

RESUMEN

Isoflurane anesthesia (IA) partially compensates NREM sleep (NREMS) and not REM sleep (REMS) requirement, eliciting post-anesthetic REMS rebound. Sleep deprivation triggers compensatory NREMS rebounds and REMS rebounds during recovery sleep as a result of the body's homeostatic mechanisms. A combination of sleep deprivation and isoflurane anesthesia is common in clinical settings, especially prior to surgeries. This study investigates the effects of pre-anesthetic sleep deprivation on post-anesthetic sleep-wake architecture. The effects of isoflurane exposure (90 min) alone were compared with the effects of isoflurane exposure preceded by experimental sleep deprivation (6 h, gentle handling) on recovery sleep in adult mice by studying the architecture of post-anesthetic sleep for 3 consecutive post-anesthetic days. Effects of isoflurane anesthesia on recovery sleep developed only during the first dark period after anesthesia, the active phase in mice. During this time, mice irrespective of preceding sleep pressure, showed NREMS and REMS rebound and decreased wakefulness during recovery sleep. Additionally, sleep deprivation prior to isoflurane treatment caused a persistent reduction of theta power during post-anesthetic REMS at least for 3 post-anesthetic days. We showed that isoflurane causes NREMS rebound during recovery sleep which suggests that isoflurane may not fully compensate for natural NREMS. The study also reveals that isoflurane exposure preceded by sleep deprivation caused a persistent disruption of REMS quality. We suggest that preoperative sleep deprivation may impair postoperative recovery through lasting disruption in sleep quality.


Asunto(s)
Anestésicos por Inhalación , Isoflurano , Privación de Sueño , Sueño REM , Vigilia , Isoflurano/efectos adversos , Isoflurano/farmacología , Animales , Privación de Sueño/fisiopatología , Ratones , Masculino , Anestésicos por Inhalación/efectos adversos , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos , Vigilia/fisiología , Ratones Endogámicos C57BL , Electroencefalografía , Sueño/efectos de los fármacos , Sueño/fisiología , Anestesia/efectos adversos
3.
J Neurosci ; 44(29)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38744530

RESUMEN

Sleep disorders affect millions of people around the world and have a high comorbidity with psychiatric disorders. While current hypnotics mostly increase non-rapid eye movement sleep (NREMS), drugs acting selectively on enhancing rapid eye movement sleep (REMS) are lacking. This polysomnographic study in male rats showed that the first-in-class selective melatonin MT1 receptor partial agonist UCM871 increases the duration of REMS without affecting that of NREMS. The REMS-promoting effects of UCM871 occurred by inhibiting, in a dose-response manner, the firing activity of the locus ceruleus (LC) norepinephrine (NE) neurons, which express MT1 receptors. The increase of REMS duration and the inhibition of LC-NE neuronal activity by UCM871 were abolished by MT1 pharmacological antagonism and by an adeno-associated viral (AAV) vector, which selectively knocked down MT1 receptors in the LC-NE neurons. In conclusion, MT1 receptor agonism inhibits LC-NE neurons and triggers REMS, thus representing a novel mechanism and target for REMS disorders and/or psychiatric disorders associated with REMS impairments.


Asunto(s)
Locus Coeruleus , Ratas Sprague-Dawley , Receptor de Melatonina MT1 , Sueño REM , Animales , Masculino , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Locus Coeruleus/fisiología , Ratas , Receptor de Melatonina MT1/agonistas , Receptor de Melatonina MT1/metabolismo , Sueño REM/fisiología , Sueño REM/efectos de los fármacos , Norepinefrina/metabolismo , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Neuronas Adrenérgicas/fisiología , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/fisiología
4.
Sleep Med ; 119: 379-388, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38761607

RESUMEN

BACKGROUND: Dopamine agonists (DAs) constitute the standard therapeutic scheme for restless leg syndrome (RLS) because they have been proven to be effective. However, DAs may change sleep parameters, thus having adverse effects on patient condition. This meta-analysis clarified the effects of DAs used in RLS treatment on the sleep architecture. METHODS: PubMed, Embase, and Cochrane Central databases were searched for randomized control trials (RCT) (up to October 2023) that discussed the effects of DAs on sleep architecture in patients with RLS. A meta-analysis employing a random-effects model was conducted. The patients were divided into subgroups according to individual DAs and treatment duration (1 day or ≥4 weeks). RESULTS: Thirteen eligible randomized placebo-controlled trials were included in the assessment. The effects of three DAs (i.e., pramipexole, ropinirole, and rotigotine) on rapid eye movement (REM) sleep, slow-wave sleep (SWS), and sleep efficiency (SE) were analyzed. Overall, pramipexole significantly improved SE but decreased the percentage of REM sleep among treated patients. Ropinirole also enhanced SE compared with the placebo group. Rotigotine did not affect SE and REM sleep. Subgroup analysis found that pramipexole used for 1 day and ≥4 weeks significantly diminished the percentage of REM sleep. Ropinirole used for 1 day showed similar REM sleep patterns. Finally, none of the three DAs affected SWS. CONCLUSIONS: This meta-analysis demonstrated that DAs significantly affect sleep parameters.


Asunto(s)
Agonistas de Dopamina , Pramipexol , Síndrome de las Piernas Inquietas , Síndrome de las Piernas Inquietas/tratamiento farmacológico , Humanos , Agonistas de Dopamina/uso terapéutico , Agonistas de Dopamina/efectos adversos , Pramipexol/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Tetrahidronaftalenos/uso terapéutico , Tetrahidronaftalenos/efectos adversos , Sueño REM/efectos de los fármacos , Indoles , Tiofenos
5.
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
6.
Psychopharmacology (Berl) ; 241(7): 1345-1363, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38430395

RESUMEN

BACKGROUND: Rapid-eye movement (REM) sleep deprivation (SD) can induce manic-like behaviors in rodents. On the other hand, lithium, as one of the oldest drugs used in neuropsychiatric disorders, is still one of the best drugs for the treatment and control of bipolar disorder. In this study, we aimed to investigate the role of chronic short-term REM SD in the induction of manic-like behaviors in female rats. METHODS: The rats were exposed to REM SD for 14 days (6 hours/day). Lithium was intraperitoneally injected at the doses of 10, 50, and 100 mg/kg. RESULTS: REM SD induced hyperactivity and OCD-like behavior, and decreased anxiety, depressive-like behavior, and pain subthreshold. REM SD also impaired passive avoidance memory and decreased hippocampal brain-derived neurotrophic factor (BDNF) expression level. Lithium at the doses of 50 and 100 mg/kg partly and completely abolished these effects, respectively. However, lithium (100 mg/kg) increased BDNF expression level in control and sham REM SD rats with no significant changes in behavior. CONCLUSIONS: Chronic short-term REM SD may induce a mania-like model and lead to OCD-like behavior and irritability. In the present study, we demonstrated a putative rodent model of mania induced by chronic REM SD in female rats. We suggest that future studies should examine behavioral and mood changes following chronic REM SD in both sexes. Furthermore, the relationship between manic-like behaviors and chronic REM SD should be investigated.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Hipocampo , Manía , Privación de Sueño , Animales , Femenino , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Privación de Sueño/metabolismo , Privación de Sueño/complicaciones , Ratas , Manía/metabolismo , Ratas Sprague-Dawley , Trastorno Obsesivo Compulsivo/metabolismo , Modelos Animales de Enfermedad , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sueño REM/efectos de los fármacos
7.
Psychopharmacology (Berl) ; 241(7): 1417-1426, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38467891

RESUMEN

Ibogaine is a potent atypical psychedelic that has gained considerable attention due to its antiaddictive and antidepressant properties in preclinical and clinical studies. Previous research from our group showed that ibogaine suppresses sleep and produces an altered wakefulness state, which resembles natural REM sleep. However, after systemic administration, ibogaine is rapidly metabolized to noribogaine, which also shows antiaddictive effects but with a distinct pharmacological profile, making this drug a promising therapeutic candidate. Therefore, we still ignore whether the sleep/wake alterations depend on ibogaine or its principal metabolite noribogaine. To answer this question, we conducted polysomnographic recordings in rats following the administration of pure noribogaine. Our results show that noribogaine promotes wakefulness while reducing slow-wave sleep and blocking REM sleep, similar to our previous results reported for ibogaine administration. Thus, we shed new evidence on the mechanisms by which iboga alkaloids work in the brain.


Asunto(s)
Ibogaína , Polisomnografía , Sueño REM , Vigilia , Animales , Sueño REM/efectos de los fármacos , Vigilia/efectos de los fármacos , Vigilia/fisiología , Masculino , Ratas , Ibogaína/análogos & derivados , Ibogaína/farmacología , Ibogaína/administración & dosificación , Ratas Sprague-Dawley , Sueño de Onda Lenta/efectos de los fármacos , Sueño de Onda Lenta/fisiología , Alucinógenos/farmacología , Alucinógenos/administración & dosificación , Electroencefalografía/efectos de los fármacos
8.
Psychopharmacology (Berl) ; 241(6): 1213-1225, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38427059

RESUMEN

RATIONALE: Prepulse inhibition (PPI) impairment reflects sensorimotor gating problems, i.e. in schizophrenia. This study aims to enlighten the role of orexinergic regulation on PPI in a psychosis-like model. OBJECTIVES: In order to understand the impact of orexinergic innervation on PPI and how it is modulated by age and baseline PPI (bPPI), chronic orexin A (OXA) injections was carried on non-sleep-deprived and sleep-deprived rats that are grouped by their bPPI. METHODS: bPPI measurements were carried on male Wistar rats on P45 or P90 followed by grouping into low-PPI and high-PPI rats. The rats were injected with OXA twice per day for four consecutive days starting on P49 or P94, while the control groups received saline injections. 72 h REMSD was carried on via modified multiple platform technique on P94 and either OXA or saline was injected during REMSD. PPI tests were carried out 30 min. after the last injection. RESULTS: Our previous study with acute OXA injection after REMSD without bPPI grouping revealed that low OXA doses might improve REMSD-induced PPI impairment. Our current results present three important conclusions: (1) The effect of OXA on PPI is bPPI-dependent and age-dependent. (2) The effect of REMSD is bPPI-dependent. (3) The effect of OXA on PPI after REMSD also depends on bPPI. CONCLUSION: Orexinergic regulation of PPI response with and without REMSD can be predicted by bPPI levels. Our findings provide potential insights into the regulation of sensorimotor gating by sleep/wakefulness systems and present potential therapeutic targets for the disorders, where PPI is disturbed.


Asunto(s)
Orexinas , Inhibición Prepulso , Ratas Wistar , Privación de Sueño , Animales , Orexinas/farmacología , Orexinas/administración & dosificación , Orexinas/metabolismo , Masculino , Privación de Sueño/fisiopatología , Ratas , Inhibición Prepulso/efectos de los fármacos , Inhibición Prepulso/fisiología , Sueño REM/efectos de los fármacos , Filtrado Sensorial/efectos de los fármacos , Factores de Edad , Modelos Animales de Enfermedad
9.
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
10.
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
11.
PLoS One ; 17(2): e0264386, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35213655

RESUMEN

Both chronic pain and sleep disorders are associated with a reduction in the quality of life. They can be both a cause and a consequence of each other, and should therefore be simultaneously treated. However, optimal treatments for chronic pain-related sleep disorders are not well established. Here, we aimed to investigate the effects of suvorexant, a novel sleep drug, and mirtazapine, a noradrenergic and specific serotonergic antidepressant, on pain-related changes in sleep parameters in a preclinical chronic pain mice model, by partial sciatic nerve ligation. We evaluated the quantity, duration, and depth of sleep by analyzing the electroencephalogram and voluntary activity by counting the number of wheel rotations to determine various symptoms of sleep disorders, including reduced total sleep time, fragmentation, low quality, and impaired activity in the daytime. Suvorexant and mirtazapine normalized the reduction in sleep time and fragmented sleep, further regaining the sleep depth at sleep onset in the chronic pain state in nerve-ligated mice. Mirtazapine also increased the percentage of rapid eye movement sleep in mice. Suvorexant decreased voluntary activity, which was prolonged after administration; however, mirtazapine did not decrease it. Although the effects of suvorexant and mirtazapine on sleep and activity are different, both suvorexant and mirtazapine could be potential therapeutic agents for chronic pain-related sleep disorders.


Asunto(s)
Azepinas/farmacología , Mirtazapina/farmacología , Nervio Ciático , Sueño REM/efectos de los fármacos , Triazoles/farmacología , Animales , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/fisiopatología , Masculino , Ratones , Nervio Ciático/lesiones , Nervio Ciático/fisiopatología
12.
Sci Rep ; 12(1): 2117, 2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35136089

RESUMEN

Previous spectral analysis studies on insomnia have shown inconsistent results due to their heterogeneity and small sample sizes. We compared the difference of electroencephalogram (EEG) spectral power during sleep among participants without insomnia, insomniacs with no hypnotic use, hypnotic users with no insomnia complaints, and hypnotic users with insomnia complaints using the Sleep Heart Health Study data, which is large sample size and has good quality control. The fast Fourier transformation was used to calculate the EEG power spectrum for total sleep duration within contiguous 30-s epochs of sleep. For 1985 participants, EEG spectral power was compared among the groups while adjusting for potential confounding factors that could affect sleep EEG. The power spectra during total sleep differed significantly among the groups in all frequency bands (pcorr < 0.001). We found that quantitative EEG spectral power in the beta and sigma bands of total sleep differed (pcorr < 0.001) between participants without insomnia and hypnotic users with insomnia complaints after controlling for potential confounders. The higher beta and sigma power were found in the hypnotic users with insomnia complaints than in the non-insomnia participants. This study suggests differences in the microstructures of polysomnography-derived sleep EEG between the two groups.


Asunto(s)
Ondas Encefálicas/efectos de los fármacos , Polisomnografía , Fármacos Inductores del Sueño/uso terapéutico , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Sueño REM/efectos de los fármacos , Adulto , Anciano , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fármacos Inductores del Sueño/farmacología
13.
Neuropharmacology ; 206: 108940, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-34982973

RESUMEN

The interaction among the acetylcholine (ACh)-ergic REM-ON neurons in the pedunculo-pontine area (PPT), noradrenergic REM-OFF neurons in locus coeruleus (LC) and GABA-ergic neurons in the regulation of rapid eye movement sleep (REMS) have been studied in relative details; however, many questions including the role of dopamine (DA) remain unanswered. The ventral tegmental area (VTA) is rich in DA-ergic neurons, which have been implicated with schizophrenia and depression, when REMS is significantly affected. Also, some of the symptoms of REMS and these diseases are common. As the ACh-ergic REM-ON neurons in the PPT project to VTA, we proposed that such inputs might affect REMS, dreams and hallucinations. We recorded sleep-wake-REMS in freely moving, chronically prepared rats under three controlled experimental conditions. In different sets of experiments, either the ACh-ergic inputs to the VTA were blocked by local microinjection of Scopolamine (Scop) alone, or, the PPT neurons were bilaterally stimulated by Glutamate (Glut), or, the PPT neurons were stimulated by Glut in presence of Scop into the VTA. It was observed that Glut into PPT and Scop into the VTA significantly increased and decreased REMS, respectively. Additionally, PPT stimulation induced increased REMS was prevented in the presence of Scop into the VTA. Based on these findings we propose that inputs from ACh-ergic REM-ON neurons to VTA increase REMS and it could be a possible circuitry for expressions of hallucinations and dreams.


Asunto(s)
Neuronas Colinérgicas/fisiología , Neuronas Dopaminérgicas/fisiología , Tegmento Pontino/fisiología , Sueño REM/fisiología , Área Tegmental Ventral/fisiología , Animales , Antagonistas Colinérgicos/farmacología , Ratas , Escopolamina/farmacología , Sueño REM/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos
14.
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
15.
Neurosci Lett ; 765: 136247, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34530113

RESUMEN

Accumulating evidence has shown that sleep disturbance is a common symptom in Alzheimer's disease (AD), which is regarded as a modifiable risk factor for AD. Orexin is a key modulator of the sleep-wake cycle and has been found to be dysregulated in AD patients. The increased orexin in cerebrospinal fluid (CSF) is associated with decreased sleep efficiency and REM sleep, as well as cognitive impairment in AD patients. The orexin system has profuse projections to brain regions that are implicated in arousal and cognition and has been found to participate in the progression of AD pathology. Conversely the orexin receptor antagonists are able to consolidate sleep and reduce AD pathology. Therefore, improved understanding of the mechanisms linking orexin system, sleep disturbance and AD could make orexin receptor antagonists a promising target for the prevention or treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antagonistas de los Receptores de Orexina/uso terapéutico , Orexinas/metabolismo , Trastornos del Sueño-Vigilia/etiología , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Cognición/efectos de los fármacos , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Modelos Animales de Enfermedad , Humanos , Antagonistas de los Receptores de Orexina/farmacología , Orexinas/antagonistas & inhibidores , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Trastornos del Sueño-Vigilia/patología , Sueño REM/efectos de los fármacos
16.
Sci Rep ; 11(1): 16313, 2021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34381098

RESUMEN

The first night effect (FNE) is a type of sleep disturbance caused by an unfamiliar environment, which leads to difficulty falling asleep and reduced sleep duration. Previously, we reported that Lactobacillus fermentum PS150 (PS150) improves sleep conditions in a pentobarbital-induced sleep mouse model. In this study, we aimed to evaluate the effect of PS150 on the FNE in mice. Briefly, mice were implanted with electrodes and orally administered PS150 for four weeks, and then the FNE was induced by cage changing. Analysis of polysomnographic signals revealed that intervention with PS150 restored non-rapid eye movement (NREM) sleep length under the FNE. Compared to diphenhydramine, a commonly used sleep aid, PS150 had no unwanted side effects, such as rapid eye movement (REM) sleep deprivation and fragmented sleep. Moreover, temporal analysis revealed that PS150 efficiently reduced both sleep latency and time spent restoring normal levels of REM sleep. Taken together, these results suggest that PS150 efficiently ameliorates sleep disturbance caused by the FNE. Additionally, V3-V4 16S rRNA sequencing revealed significant increases in Erysipelotrichia, Actinobacteria, and Coriobacteriia in fecal specimens of the PS150-treated group, indicating that PS150 induces gut microbiota remodeling.


Asunto(s)
Limosilactobacillus fermentum/fisiología , Sueño REM/fisiología , Animales , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Pentobarbital/farmacología , Polisomnografía/métodos , ARN Ribosómico 16S/genética , Privación de Sueño/inducido químicamente , Privación de Sueño/microbiología , Privación de Sueño/fisiopatología , Trastornos del Inicio y del Mantenimiento del Sueño/inducido químicamente , Trastornos del Inicio y del Mantenimiento del Sueño/microbiología , Trastornos del Inicio y del Mantenimiento del Sueño/fisiopatología , Trastornos del Sueño-Vigilia/inducido químicamente , Trastornos del Sueño-Vigilia/microbiología , Trastornos del Sueño-Vigilia/fisiopatología , Sueño REM/efectos de los fármacos
17.
Sci Rep ; 11(1): 16713, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408180

RESUMEN

The effects of chronic antidepressant (AD) treatment on sleep disturbances in rodent chronic stress models have not been thoroughly investigated. Here, we show that chronic social defeat stress (SDS) in rats induces prolonged social avoidance, alterations in sleep architecture (increased total rapid eye movement [REM] sleep duration, bout, and shortened REM latency), and contextual but not cued fear memory deficits, even 1 month after the last SDS. These abnormalities were associated with changes in electroencephalography (EEG) spectral powers, including reduced REM sleep theta power during the light phase. Chronic treatment with two different classes of antidepressants (ADs), imipramine and fluoxetine, significantly ameliorated these behavioral, sleep, and EEG abnormalities. Interestingly, REM theta power was normalized by chronic (1 month) but not 1 week AD administration and solely correlated with the ratio (an objective indicator) of social interaction 1 month after the last SDS. These data suggest that reductions in REM sleep theta power, an EEG parameter that has never been directly investigated in humans, is a core sleep symptom in socially defeated rats, and, potentially, also in patients with stress-related psychiatric disorders, including major depressive and posttraumatic stress disorders.


Asunto(s)
Antidepresivos/efectos adversos , Fluoxetina/efectos adversos , Imipramina/efectos adversos , Sueño REM/efectos de los fármacos , Estrés Psicológico/fisiopatología , Ritmo Teta/efectos de los fármacos , Animales , Antidepresivos/farmacología , Enfermedad Crónica , Electroencefalografía , Fluoxetina/farmacología , Humanos , Imipramina/farmacología , Masculino , Ratas
18.
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
19.
Neuropharmacology ; 193: 108607, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34023337

RESUMEN

Dreams appear intermittently during phasic rapid eye movement sleep (REMS). Although reasonable progress has been made about neuro-physio-pharmacological mechanism of appearance of REMS, appearance of dreams is a mystery. Isolated studies have reported that substantia nigra (SN) withdraws inhibition from pedunculo-pontine tegmentum (PPT) acetylcholine (ACh)-ergic REM-ON neurons to trigger REMS; some REM-ON neurons become phasically active during REMS; amygdala (Amyg), a limbic structure associated with emotions, may be related with dreaming like state; Amyg receives projections from both SN-Dopamine (DA)-ergic and PPT-ACh-ergic neurons. Collating these isolated findings, we proposed that on the background of REMS, SN-DA-ergic and PPT-ACh-ergic inputs phasically activate Amyg-neurons to manifest dreams. In the absence of better criteria, we recorded electrophysiological characteristics of REMS as the closest objective read-out for dreams in surgically prepared, chronic, freely moving rats. Microinjection of either DA-ergic or ACh-ergic agonist [Quinpirole (Qnp) or Carbachol (Carb)] bilaterally into Amyg increased, while antagonists [Haloperidol (Hal) or Scopolamine (Scop)] reduced REMS. Electrical stimulation of either bilateral SN or PPT increased REMS, which however, was prevented when stimulated in presence of Hal or Scop, respectively into the Amyg. These findings confirm and support our contention that SN-DA-ergic and PPT-ACh-ergic inputs integrate in Amyg for REMS regulation. Further, subject to confirmation in humans, we propose that on the background of REMS, some phasic PPT-ACh-ergic-REM-ON neurons intermittently trigger some neurons in Amyg, the area known to be associated with memory and emotions, causing intermittent appearance of REMS-associated dreams and in REMS behavior disorder.


Asunto(s)
Amígdala del Cerebelo/fisiología , Neuronas Dopaminérgicas/fisiología , Tegmento Pontino/fisiología , Sueño REM/fisiología , Sustancia Negra/fisiología , Vigilia/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Carbacol/farmacología , Neuronas Colinérgicas , Estimulación Eléctrica , Haloperidol/farmacología , Masculino , Tegmento Pontino/efectos de los fármacos , Quinpirol/farmacología , Ratas , Ratas Wistar , Escopolamina/farmacología , Sueño REM/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Vigilia/efectos de los fármacos
20.
Clin Pharmacol Ther ; 110(3): 808-815, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33961287

RESUMEN

In contrast to the dose-occupancy relationship in the treatment of schizophrenia, the minimal effective level of dopamine receptor 2 (D2R) blockade for antipsychotics in the treatment of bipolar depression is unknown. Lower doses aimed at reducing extrapyramidal side effects must be balanced against the need to retain the therapeutic benefit of D2R blockade on emergent cycling, mixed, manic, anxiety, and/or psychotic symptoms. Dose-reductions intended to lower D2R blockade, however, could also decrease concomitant serotonin receptor antagonism and its potential benefit on depressive symptoms. Here, we uncoupled the potential antidepressant activity in amisulpride, driven by 5-HT7 receptor (5-HT7R) antagonism, from the D2R-mediated antipsychotic activity by discovering that each enantiomer favors a different receptor. Aramisulpride was more potent at 5-HT7R relative to esamisulpride (Ki 47 vs. 1,900 nM, respectively), whereas esamisulpride was more potent at D2R (4.0 vs. 140 nM). We hypothesized that a nonracemic ratio might achieve greater 5-HT7R-mediated antidepressant effects at a lower level of D2R blockade. The dose-occupancy relationship of esamisulpride at D2R was determined by positron emission tomography (PET) imaging in human volunteers. Separately the dose-relationship of aramisulpride was established in humans using suppression of rapid eye movement (REM) sleep as a marker of 5-HT7R antagonism. These results led to the discovery of an 85:15 ratio of aramisulpride to esamisulpride (SEP-4199) that maximizes the potential for antidepressant benefit of aramisulpride via 5-HT7R and reduces esamisulpride to minimize D2R-related extrapyramidal side effects while still retaining D2R-mediated effects predicted to provide benefit in bipolar depression. The antidepressant efficacy of SEP-4199 was recently confirmed in a proof-of-concept trial for the treatment of bipolar depression (NCT03543410).


Asunto(s)
Amisulprida/efectos adversos , Amisulprida/uso terapéutico , Antipsicóticos/efectos adversos , Antipsicóticos/uso terapéutico , Antagonistas de los Receptores de Dopamina D2/uso terapéutico , Trastornos del Humor/tratamiento farmacológico , Receptores de Serotonina/metabolismo , Adulto , Animales , Antidepresivos/uso terapéutico , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/metabolismo , Femenino , Humanos , Masculino , Trastornos del Humor/metabolismo , Tomografía de Emisión de Positrones/métodos , Ratas , Ratas Wistar , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/metabolismo , Sueño REM/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...