Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Sleep Res ; 19(3): 384-93, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20408925

RESUMEN

The adult hippocampal dentate gyrus (DG) exhibits cell proliferation and neurogenesis throughout life. We examined the effects of daily administration of eszopiclone (Esz), a commonly used hypnotic drug and gamma-aminobutyric acid (GABA) agonist, compared with vehicle, on DG cell proliferation and neurogenesis, and on sleep-wake patterns. Esz was administered during the usual sleep period of rats, to mimic typical use in humans. Esz treatment for 7 days did not affect the rate of cell proliferation, as measured by 5-bromo-2'-deoxyuridine (BrdU) immunostaining. However, twice-daily Esz administration for 2 weeks increased survival of newborn cells by 46%. Most surviving cells exhibited a neuronal phenotype, identified as BrdU-neuronal nuclei (NeuN) double-labeling. NeuN is a marker of neurons. Non-rapid eye movement sleep was increased on day 1, but not on days 7 or 14 of Esz administration. Delta electroencephalogram activity was increased on days 1 and 7 of treatment, but not on day 14. There is evidence that enhancement of DG neurogenesis is a critical component of the effects of antidepressant treatments of major depressive disorder (MDD). Adult-born DG cells are responsive to GABAergic stimulation, which promotes cell maturation. The present study suggests that Esz, presumably acting as a GABA agonist, has pro-neurogenic effects in the adult DG. This result is consistent with evidence that Esz enhances the antidepressant treatment response of patients with MDD with insomnia.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Hipocampo/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Neurogénesis/efectos de los fármacos , Piperazinas/farmacología , Animales , Compuestos de Azabiciclo/administración & dosificación , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Eszopiclona , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Hipnóticos y Sedantes/administración & dosificación , Masculino , Piperazinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Sueño/efectos de los fármacos , Factores de Tiempo
2.
Brain Res ; 1304: 96-104, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19781535

RESUMEN

The perifornical-lateral hypothalamic area (PF-LHA) has been implicated in the regulation of arousal. The PF-LHA contains wake-active neurons that are quiescent during non-REM sleep and in the case of neurons expressing the peptide hypocretin (HCRT), quiescent during both non-REM and REM sleep. Adenosine is an endogenous sleep factor and recent evidence suggests that adenosine via A(1) receptors may act on PF-LHA neurons to promote sleep. We examined the effects of bilateral activation as well as blockade of A(1) receptors in the PF-LHA on sleep-wakefulness in freely behaving rats. The sleep-wake profiles of male Wistar rats were recorded during reverse microdialysis perfusion of artificial cerebrospinal fluid (aCSF) and two doses of adenosine A(1) receptor antagonist, 1,3-dipropyl-8-phenylxanthine (CPDX; 5 microM and 50 microM) or A(1) receptor agonist, N(6)-cyclopentyladenosine (CPA; 5 microM and 50 microM) into the PF-LHA for 2 h followed by 4 h of aCSF perfusion. CPDX perfused into the PF-LHA during lights-on phase produced arousal (F=7.035, p<0.001) and concomitantly decreased both non-REM (F=7.295, p<0.001) and REM sleep (F=3.456, p<0.004). In contrast, CPA perfused into the PF-LHA during lights-off phase significantly suppressed arousal (F=7.891, p<0.001) and increased non-REM (F=8.18, p <0.001) and REM sleep (F=30.036, p<0.001). These results suggest that PF-LHA is one of the sites where adenosine, acting via A(1) receptors, inhibits PF-LHA neurons to promote sleep.


Asunto(s)
Área Hipotalámica Lateral/fisiología , Receptor de Adenosina A1/metabolismo , Sueño/fisiología , Vigilia/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A1 , Antagonistas del Receptor de Adenosina A1 , Animales , Cateterismo , Fármacos del Sistema Nervioso Central/farmacología , Área Hipotalámica Lateral/efectos de los fármacos , Masculino , Estimulación Luminosa , Ratas , Ratas Wistar , Sueño/efectos de los fármacos , Sueño REM/efectos de los fármacos , Sueño REM/fisiología , Factores de Tiempo , Vigilia/efectos de los fármacos , Xantinas/farmacología
3.
Am J Physiol Regul Integr Comp Physiol ; 296(1): R178-84, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18971348

RESUMEN

Previous work showed that sleep is associated with increased brain protein synthesis and that arrest of protein synthesis facilitates sleep. Arrest of protein synthesis is induced during the endoplasmic reticulum (ER) stress response, through phosphorylation of eukaryotic initiation factor 2alpha (p-eIF2alpha). We tested a hypothesis that elevation of p-eIF2alpha would facilitate sleep. We studied the effects of intracerebroventricular infusion of salubrinal (Salub), which increases p-eIF2alpha by inhibiting its dephosphorylation. Salub increased deep slow wave sleep by 255%, while reducing active waking by 49%. Delta power within non-rapid eye movement (NREM) sleep was increased, while power in the sigma, beta, and gamma bands during NREM was reduced. We found that Salub increased expression of p-eIF2alpha in the basal forebrain (BF) area, a sleep-wake regulatory brain region. Therefore, we quantified the p-eIF2alpha-immunolabeled neurons in the BF area; Salub administration increased the number of p-eIF2alpha-expressing noncholinergic neurons in the caudal BF. In addition, Salub also increased the intensity of p-eIF2alpha expression in both cholinergic and noncholinergic neurons, but this was more widespread among the noncholinergic neurons. Our findings support a hypothesis that sleep is facilitated by signals associated with the ER stress response.


Asunto(s)
Cinamatos/farmacología , Hipnóticos y Sedantes/farmacología , Neuronas/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Fases del Sueño/efectos de los fármacos , Tiourea/análogos & derivados , Animales , Fibras Colinérgicas/efectos de los fármacos , Fibras Colinérgicas/metabolismo , Cinamatos/administración & dosificación , Electroencefalografía , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Hipnóticos y Sedantes/administración & dosificación , Infusiones Parenterales , Masculino , Neuronas/metabolismo , Fosforilación , Prosencéfalo/metabolismo , Inhibidores de la Síntesis de la Proteína/administración & dosificación , Ratas , Ratas Sprague-Dawley , Estrés Fisiológico/efectos de los fármacos , Tiourea/administración & dosificación , Tiourea/farmacología , Factores de Tiempo , Regulación hacia Arriba , Vigilia/efectos de los fármacos
4.
J Physiol ; 575(Pt 3): 807-19, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16825295

RESUMEN

Previous work shows that sleep deprivation impairs hippocampal-dependent learning and long-term potentiation (LTP). Brain-derived neurotrophic factor (BDNF), cAMP response-element-binding (CREB) and calcium-calmodulin-dependent protein kinase II (CAMKII) are critical modulators of hippocampal-dependent learning and LTP. In the present study we compared the effects of short- (8 h) and intermediate-term (48 h) sleep deprivation (SD) on the expression of BDNF and its downstream targets, Synapsin I, CREB and CAMKII in the neocortex and the hippocampus. Rats were sleep deprived using an intermittent treadmill system which equated total movement in the SD and control treadmill animals (CT), but permitted sustained periods of rest in CT animals. Animals were divided into SD (treadmill schedule: 3 s on/12 s off) and two treadmill control groups, CT1 (15 min on/60 min off) and CT2 (30 min on/120 min off - permitting more sustained sleep). Real-time Taqman RT-PCR was used to measure changes in mRNA; BDNF protein levels were determined using ELISA. In the hippocampus, 8 h treatments reduced BDNF, Synapsin I, CREB and CAMKII gene expression in both SD and control groups. Following 48 h of experimental procedures, the expression of all these four molecular markers of plasticity was reduced in SD and CT1 groups compared to the CT2 and cage control groups. In the hippocampus, BDNF protein levels after 8 h and 48 h treatments paralleled the changes in mRNA. In neocortex, neither 8 h nor 48 h SD or control treatments had significant effects on BDNF, Synapsin I and CAMKII mRNA levels. Stepwise regression analysis suggested that loss of REM sleep underlies the effects of SD on hippocampal BDNF, Synapsin I and CREB mRNA levels, whereas loss of NREM sleep underlies the effects on CAMKII mRNA.


Asunto(s)
Regulación de la Expresión Génica , Hipocampo/metabolismo , Plasticidad Neuronal/genética , Privación de Sueño/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Masculino , Modelos Animales , Neocórtex/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Privación de Sueño/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Factores de Tiempo , Vigilia/genética
5.
Eur J Neurosci ; 22(8): 2111-6, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16262649

RESUMEN

We reported previously that 96 h of sleep deprivation (SD) reduced cell proliferation in the dentate gyrus (DG) of the hippocampus in adult rats. We now report that SD reduces the number of new cells expressing a mature neuronal marker, neuronal nuclear antigen (NeuN). Rats were sleep-deprived for 96 h, using an intermittent treadmill system. Total sleep time was reduced to 6.9% by this method in SD animals, but total treadmill movement was equated in SD and treadmill control (CT) groups. Rats were allowed to survive for 3 weeks after 5-bromo-2-deoxyuridine (BrdU) injection. The phenotype of BrdU-positive cells in the DG was assessed by immunofluorescence and confocal microscopy. After 3 weeks the number of BrdU-positive cells was reduced by 39.6% in the SD group compared with the CT. The percentage of cells that co-localized BrdU and NeuN was also lower in the SD group (SD: 46.6 +/- 1.8% vs. CT: 71.9 +/- 2.1, P < 0.001). The percentages of BrdU-labeled cells co-expressing markers of immature neuronal (DCX) or glial (S100-beta) cells were not different in SD and CT groups. Thus, SD reduces neurogenesis in the DG by affecting both total proliferation and the percentage of cells expressing a mature neuronal phenotype. We hypothesize that sleep provides anabolic or signaling support for proliferation and cell fate determination.


Asunto(s)
Proliferación Celular , Hipocampo/patología , Neuronas/patología , Privación de Sueño/fisiopatología , Animales , Conducta Animal , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Proteína Doblecortina , Inmunohistoquímica/métodos , Masculino , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Vigilia/fisiología
6.
Am J Physiol Regul Integr Comp Physiol ; 289(6): R1715-23, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16109808

RESUMEN

Evidence suggests that adenosine (AD) is an endogenous sleep factor. The hypnogenic action of AD is mediated through its inhibitory A1 and excitatory A2A receptors. Although AD is thought to be predominantly active in the wake-active region of the basal forebrain (BF), a hypnogenic action of AD has been demonstrated in several other brain areas, including the preoptic area. We hypothesized that in lateral preoptic area (LPOA), a region with an abundance of sleep-active neurons, AD acting via A1 receptors would induce waking by inhibition of sleep-active neurons and that AD acting via A2A receptors would promote sleep by stimulating the sleep-active neurons. To this end, we studied the effects on sleep of an AD transport inhibitor, nitrobenzyl-thio-inosine (NBTI) and A1 and A2A receptor agonists/antagonists by microdialyzing them into the LPOA. The results showed that, in the sleep-promoting area of LPOA: 1) A1 receptor stimulation or inhibition of AD transport by NBTI induced waking and 2) A2A receptor stimulation induced sleep. We also confirmed that NBTI administration in the wake promoting area of the BF increased sleep. The effects of AD could be mediated either directly or indirectly via interaction with other neurotransmitter systems. These observations support a hypothesis that AD mediated effects on sleep-wake cycles are site and receptor dependent.


Asunto(s)
Adenosina/metabolismo , Área Preóptica/fisiología , Receptor de Adenosina A1/metabolismo , Receptor de Adenosina A2A/metabolismo , Sueño/fisiología , Vigilia/fisiología , Animales , Masculino , Microdiálisis/métodos , Área Preóptica/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A1/efectos de los fármacos , Receptor de Adenosina A2A/efectos de los fármacos , Sueño/efectos de los fármacos , Tioinosina/análogos & derivados , Tioinosina/farmacología , Vigilia/efectos de los fármacos
7.
J Physiol ; 563(Pt 2): 569-82, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15613374

RESUMEN

The perifornical-lateral hypothalamic area (PF-LHA) has been implicated in the regulation of behavioural arousal. The PF-LHA contains several cell types including neurones expressing the peptides, hypocretin (HCRT; also called orexin) and melanin-concentrating hormone (MCH). Evidence suggests that most of the PF-LHA neurones, including HCRT neurones, are active during waking and quiescent during non-rapid eye movement (non-NREM) sleep. The PF-LHA contains local GABAergic interneurones and also receives GABAergic inputs from sleep-promoting regions in the preoptic area of the hypothalamus. We hypothesized that increased GABA-mediated inhibition within PF-LHA contributes to the suppression of neuronal activity during non-REM sleep. EEG and EMG activity of rats were monitored for 2 h during microdialytic delivery of artificial cerebrospinal fluid (aCSF) or bicuculline, a GABAA receptor antagonist, into the PF-LHA in spontaneously sleeping rats during the lights-on period. At the end of aCSF or bicuculline perfusion, rats were killed and c-Fos immunoreactivity (Fos-IR) in HCRT, MCH and other PF-LHA neurones was quantified. In response to bicuculline perfusion into the PF-LHA, rats exhibited a dose-dependent decrease in non-REM and REM sleep time and an increase in time awake. The number of HCRT, MCH and non-HCRT/non-MCH neurones exhibiting Fos-IR adjacent to the microdialysis probe also increased dose-dependently in response to bicuculline. However, significantly fewer MCH neurones exhibited Fos-IR in response to bicuculline as compared to HCRT and other PF-LHA neurones. These results support the hypothesis that PF-LHA neurones, including HCRT neurones, are subject to increased endogenous GABAergic inhibition during sleep. In contrast, MCH neurones appear to be subject to weaker GABAergic control during sleep.


Asunto(s)
Hormonas Hipotalámicas/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Melaninas/fisiología , Neuronas/fisiología , Neuropéptidos/fisiología , Hormonas Hipofisarias/fisiología , Fases del Sueño/fisiología , Ácido gamma-Aminobutírico/fisiología , Animales , Bicuculina/farmacología , Ritmo Circadiano , Relación Dosis-Respuesta a Droga , Genes fos/fisiología , Área Hipotalámica Lateral/fisiología , Área Hipotalámica Lateral/ultraestructura , Masculino , Neuronas/efectos de los fármacos , Neuronas/ultraestructura , Orexinas , Ratas , Ratas Sprague-Dawley
8.
Brain Res ; 960(1-2): 165-73, 2003 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-12505669

RESUMEN

Activation of the preoptic area (POA) warm sensitive neurons is known to promote non-REM (NREM) sleep and inhibit neuronal discharge in arousal-related brain structures. The perifornical area of the lateral hypothalamus (PF/LH) was recently recognized to be an additional important arousal promoting region. We studied the behavior of PF/LH neurons in rats during the normal sleep-wake cycle and in response to local POA warming. Most PF/LH neurons were wake-active, and exhibited low discharge throughout NREM. Seventy four percent of these wake-active neurons exhibited moderate or strong activation in REM sleep compared to NREM sleep. A substantial group (26%) exhibited very low discharge in REM as well as NREM sleep. Fifty two percent of units in the PF/LH area were responsive to POA warming; 90% of responsive neurons exhibited a significant reduction (-26.47+/-2.16% for 1 degrees C of POA warming) in their discharge rate. The inhibitory effect of POA warming on PF/LH neurons was not associated with EEG slowing. This study supports the hypothesis that sleep induction by POA warm sensitive neurons is mediated through the inhibition of multiple arousal-related structures.


Asunto(s)
Área Hipotalámica Lateral/fisiología , Neuronas/fisiología , Área Preóptica/fisiología , Vigilia/fisiología , Animales , Temperatura Corporal/fisiología , Electroencefalografía , Electromiografía , Electrofisiología , Área Hipotalámica Lateral/citología , Masculino , Polisomnografía , Ratas , Ratas Sprague-Dawley , Sueño/fisiología , Sueño REM/fisiología , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...