Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
Science ; 194(4264): 537-9, 1976 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-973138

RESUMEN

The relationship between hypothalamic temperature and metabolic heat production was measured during wakefulness, slow-wave sleep, and paradoxical sleep in unrestrained kangaroo rats (Dipodomys). Hypothalamic temperature was manipulated with chronically implanted, water-perfused thermodes while cortical electroencephalogram, electromyogram, metabolic rate, and body movement were continuously recorded. During slow-wave sleep, in comparison to wakefulness, there is a lowered threshold hypothalamic temperature for the metabolic heat production response and a lowered proportionality constant relating rate of metabolic heat production to hypothalamic temperature. During paradoxical sleep no increase in metabolic heat production could be elicited by lowering hypothalamic temperature, which indicates that the thermoregulatory system is inoperative. These results provide a basis for explaining the changes in various body temperatures, metabolic rate, and other thermoregulatory responses during sleep in a variety of mammals.


Asunto(s)
Regulación de la Temperatura Corporal , Hipotálamo/fisiología , Fases del Sueño/fisiología , Animales , Dipodomys , Consumo de Oxígeno , Temperatura , Vigilia/fisiología
2.
Science ; 204(4397): 1098-100, 1979 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-221974

RESUMEN

Estivation (shallow torpor) in the round-tailed ground squirrel (Citellus tereticaudus) is entered through electrophysiologically defined states of sleep. Rapid-eye-movement sleep diminishes as body temperature falls in such a way that, at a body temperature of 26 degrees to 28 degrees C, torpor is characterized by almost continuous slow-wave sleep isomorphic with that observed at euthermic body temperatures.


Asunto(s)
Metabolismo Energético , Hibernación , Roedores/fisiología , Sciuridae/fisiología , Sueño/fisiología , Animales , Regulación de la Temperatura Corporal , Privación de Alimentos , Corazón/fisiología , Frecuencia Cardíaca , Estaciones del Año , Sueño REM/fisiología
3.
Prog Neurobiol ; 44(5): 433-49, 1994 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7886233

RESUMEN

We have hypothesized that REM sleep is functionally and homeostatically related to NREM sleep rather than to waking. In other words, REM sleep rather than to waking. In other words, REM sleep occurs in response to NREM-sleep expression and compensates for some process that takes place during NREM sleep. Under normal conditions, the need for REM sleep does not accrue during waking. The primary basis for this hypothesis is the fact that REM-sleep expression is a function of prior NREM-sleep expression. That is, REM sleep follows NREM sleep within sleep periods, REM-sleep episodes occur at intervals determined by the amount of NREM-sleep time elapsed, and total time spent in REM sleep is consistently about 1/4 of prior NREM-sleep time, regardless of how much time is spent in NREM sleep. Our experimental tests of the hypothesis support it. (1) REM-sleep propensity accumulates quite rapidly during a 2-hr interval spent predominantly in NREM sleep. (2) The timing of individual REM-sleep episodes is controlled homeostatically, by accumulation within NREM sleep of a propensity for REM sleep. The NREM sleep-related model of REM-sleep regulation (Fig. 1) explains a number of phenomena of REM-sleep expression, including the frequent and periodic occurrence of REM-sleep episodes throughout sleep periods, that have been accommodated by the waking-related model but are not functionally accounted for by it. In our opinion, the NREM sleep-related model of REM-sleep regulation recommends itself partly by its simplicity. According to the waking-related model, two independent and competing sleep propensities accumulate during waking and are discharged in two distinct sleep states that perform different waking-related recovery processes. One behaviour, sleep, is thought to perform two independent and competing functions that alternate at regular intervals. In the NREM sleep-related model of REM-sleep regulation, sleep debt simply reflects a need for NREM sleep. That is, the cerebrally less activated state of NREM sleep enables some form of restoration made necessary by the cerebrally activated state of waking. Periodic occurrence of REM-sleep episodes is explained without postulating an oscillatory mechanism to gate expression of NREM sleep versus REM sleep. In assessing the comparative merits of the waking-related and NREM sleep-related models of REM-sleep regulation, one should consider the influence of time-worn habits of thought.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Sueño REM/fisiología , Sueño/fisiología , Vigilia/fisiología , Adulto , Anciano , Animales , Preescolar , Ritmo Circadiano/fisiología , Trastorno Depresivo/fisiopatología , Homeostasis , Humanos , Modelos Biológicos , Narcolepsia/fisiopatología , Privación de Sueño/fisiología
4.
J Neurosci ; 19(19): 8630-6, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10493763

RESUMEN

Temperature compensation and the effects of heat pulses on rhythm phase were assessed in the suprachiasmatic nucleus (SCN). Circadian neuronal rhythms were recorded from the rat SCN at 37 and 31 degrees C in vitro. Rhythm period was 23.9 +/- 0.1 and 23.7 +/- 0.1 hr at 37 and 31 degrees C, respectively; the Q(10) for tau was 0.99. Heat pulses were administered at various circadian times (CTs) by increasing SCN temperature from 34 to 37 degrees C for 2 hr. Phase delays and advances were observed during early and late subjective night, respectively, and no phase shifts were obtained during midsubjective day. Maximum phase delays of 2.2 +/- 0.3 hr were obtained at CT 14, and maximum phase advances of 3.5 +/- 0.2 hr were obtained at CT 20. Phase delays were not blocked by a combination of NMDA [AP-5 (100 microM)] and non-NMDA [CNQX (10 microM)] receptor antagonists or by tetrodotoxin (TTX) at concentrations of 1 or 3 microM. The phase response curve for heat pulses is similar to ones obtained with light pulses for behavioral rhythms. These data demonstrate that circadian pacemaker period in the rat SCN is temperature-compensated over a physiological range of temperatures. Phase delays were not caused by activation of ionotropic glutamate receptors, release of other neurotransmitters, or temperature-dependent increases in metabolism associated with action potentials. Heat pulses may have phase-shifted rhythms by directly altering transcriptional or translational events in SCN pacemaker cells.


Asunto(s)
Ritmo Circadiano/fisiología , Antagonistas de Aminoácidos Excitadores/farmacología , Neuronas/fisiología , Núcleo Supraquiasmático/fisiología , 2-Amino-5-fosfonovalerato/farmacología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Ciclos de Actividad/fisiología , Animales , Calor , Técnicas In Vitro , Masculino , Neuronas/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de N-Metil-D-Aspartato/fisiología , Núcleo Supraquiasmático/efectos de los fármacos , Temperatura , Tetrodotoxina/farmacología
5.
J Neurosci ; 19(10): 3781-90, 1999 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-10234010

RESUMEN

The purpose of this study was to characterize changes in gene expression in the brain of a seasonal hibernator, the golden-mantled ground squirrel, Spermophilus lateralis, during the hibernation season. Very little information is available on molecular changes that correlate with hibernation state, and what has been done focused mainly on seasonal changes in peripheral tissues. We produced over 4000 reverse transcription-PCR products from euthermic and hibernating brain and compared them using differential display. Twenty-nine of the most promising were examined by Northern analysis. Although some small differences were observed across hibernation states, none of the 29 had significant changes. However, a more direct approach, investigating expression of putative hibernation-responsive genes by Northern analysis, revealed an increase in expression of transcription factors c-fos, junB, and c-Jun, but not junD, commencing during late torpor and peaking during the arousal phase of individual hibernation bouts. In contrast, prostaglandin D2 synthase declined during late torpor and arousal but returned to a high level on return to euthermia. Other genes that have putative roles in mammalian sleep or specific brain functions, including somatostatin, enkephalin, growth-associated protein 43, glutamate acid decarboxylases 65/67, histidine decarboxylase, and a sleep-related transcript SD464 did not change significantly during individual hibernation bouts. We also observed no decline in total RNA or total mRNA during torpor; such a decline had been previously hypothesized. Therefore, it appears that the dramatic changes in body temperature and other physiological variables that accompany hibernation involve only modest reprogramming of gene expression or steady-state mRNA levels.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hibernación/fisiología , Animales , Northern Blotting , Femenino , Masculino , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-jun/genética , ARN Mensajero/biosíntesis , Sciuridae
6.
J Biol Rhythms ; 11(2): 126-36, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8744240

RESUMEN

Temperature compensation of circadian rhythms in neuronal firing rate was investigated in the suprachiasmatic nucleus (SCN) of ground squirrels and rats in vitro. A reduction in SCN temperature from 37 to 25 degrees C reduced peak firing rates by > 70% in rats but only by approximately 21% in squirrels; trough firing rates were marginally altered in both species. In the rat SCN at 25 degrees C, the peak in neuronal activity decreased progressively on successive days and circadian rhythms no longer were present by Day 3. There was a 37% reduction in the number of single units detected and an increase in the temporal variability of peak firing rates among individual rat SCN neurons at low temperature. By contrast, single units were readily detected and circadian rhythms were robust in squirrels at 37 and 25 degrees C; a Q10 of 0.927 was associated with a shortening of tau by 2 h and a 5-h phase change after only 48 h at low temperature. These results suggest that temperature can have a substantial impact on circadian organization in a mammalian pacemaker considered to be temperature compensated.


Asunto(s)
Neuronas/fisiología , Ratas Wistar/fisiología , Sciuridae/fisiología , Núcleo Supraquiasmático/fisiología , Potenciales de Acción , Animales , Ritmo Circadiano , Electrofisiología , Técnicas In Vitro , Masculino , Microelectrodos , Ratas , Especificidad de la Especie , Temperatura , Factores de Tiempo
7.
J Biol Rhythms ; 13(6): 506-17, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9850011

RESUMEN

Body temperature (Tb) or activity rhythms were monitored in male Siberian hamsters (Phodopus sungorus) housed in an LD cycle of 16 h light/day from birth. At 3 months of age, rhythms were monitored for 14 days, and then the LD cycle was phase delayed by 1, 3, or 5 h or phase advanced by 5 h in four separate groups of animals. Phase delays were accomplished via a 1- or 3-h extension of the light phase or via a 5-h extension of the dark phase. The phase advance was accomplished via a 5-h shortening of the light phase. After 2 to 3 weeks, hamsters that were phase delayed by 1 or 3 h were then phase advanced by 1 or 3 h, respectively, via a shortening of the light phase. All of the animals reentrained to phase delays of 1 or 3 h and to a 1-h phase advance; 79% reentrained to a 3-h phase advance. In contrast, only 13% of the animals reentrained to the 5-h phase advance, 13% became arrhythmic, and 74% free ran for several weeks. After the 5-h phase delay, however, reentrainment was observed in 50% of the animals although half of them required more than 21 days to reentrain. The response to a phase shift could not be predicted by any parameter of circadian rhythm organization assessed prior to the phase shift. These data demonstrate that a phase shift of the LD cycle can permanently disrupt entrainment mechanisms and eliminate circadian Tb and activity rhythms. Magnitude and direction of a phase shift of the LD cycle determine not only the rate but also the probability of reentrainment. Furthermore, the phase of the LD cycle at which the phase shift is made has a marked effect on the proportion of animals that reentrain. Light exposure during mid-subjective night combined with daily light exposure during the active phase may explain these phenomena.


Asunto(s)
Ritmo Circadiano/fisiología , Animales , Temperatura Corporal/fisiología , Cricetinae , Masculino , Actividad Motora/fisiología , Phodopus , Fotoperiodo
8.
J Biol Rhythms ; 8(1): 1-16, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8490207

RESUMEN

The primary mammalian circadian clock, located in the suprachiasmatic nuclei (SCN), receives a major input from the raphe nuclei. The role of this input is largely unknown, and is the focus of this research. The SCN clock survives in vitro, where it produces a 24-hr rhythm in spontaneous neuronal activity that is sustained for at least three cycles. The sensitivity of the SCN clock to drugs can therefore be tested in vitro by determining whether various compounds alter the phase of this rhythm. We have previously shown that the nonspecific serotonin (5-HT) agonist quipazine resets the SCN clock in vitro, inducing phase advances in the daytime and phase delays at night. These results suggest that the 5-HT-ergic input from the raphe nuclei can modulate the phase of the SCN circadian clock. In this study we began by using autoradiography to determine that the SCN contain abundant 5-HT1A and 5-HT1B receptors, very few 5-HT1C and 5-HT2 receptors, and no 5-HT3 receptors. Next we investigated the ability of 5-HT-ergic agonists and antagonists to reset the clock in vitro, in order to determine what type or types of 5-HT receptor(s) are functionally linked to the SCN clock. We began by providing further evidence of 5-HT-ergic effects in the SCN. We found that 5-HT mimicked the effects of quipazine, whereas the nonspecific 5-HT antagonist metergoline blocked these effects, in both the day and night. Next we found that the 5-HT1A agonist 8-OH-DPAT, and to a lesser extent the 5-HT1A-1B agonist RU 24969, mimicked the effects of quipazine during the subjective daytime, whereas the 5-HT1A antagonist NAN-190 blocked quipazine's effects. None of the other specific agonists or antagonists we tried induced similar effects. This suggests that quipazine acts on 5-HT1A receptors in the daytime to advance the SCN clock. None of the specific agents we tried were able either to mimic or to block the actions of 5-HT or quipazine at circadian time 15. Thus, we were unable to determine the type of 5-HT receptor involved in nighttime phase delays by quipazine or 5-HT. However, since the dose-response curves for quipazine during the day and night are virtually identical, we hypothesize that the nighttime 5-HT receptor is a 5-HT1-like receptor.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Antagonistas de la Serotonina/farmacología , Agonistas de Receptores de Serotonina/farmacología , Serotonina/fisiología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Autorradiografía , Relación Dosis-Respuesta a Droga , Electrofisiología , Técnicas In Vitro , Yodocianopindolol , Dietilamida del Ácido Lisérgico/farmacología , Masculino , Metergolina/farmacología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Pindolol/análogos & derivados , Pindolol/farmacología , Quipazina/farmacología , Ratas , Ratas Wistar , Receptores de Serotonina/efectos de los fármacos , Núcleo Supraquiasmático/efectos de los fármacos , Núcleo Supraquiasmático/fisiología
9.
J Biol Rhythms ; 13(4): 347-57, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9711509

RESUMEN

The genes NGFI-A (also known as EGR-1, zif/268, and Krox-24) and NGFI-B (nur/77) have previously been shown to be induced in the SCN of rats and hamsters by photic stimulation during the subjective night. The purpose of this study is to determine whether these genes are also induced in the SCN of mice and, if so, to characterize the circadian system of animals in which either NGFI-A or both NGFI-A and NGFI-B were eliminated by homologous recombination. In wildtype mice, NGFI-A mRNA was found to be induced in the SCN as in other rodent species. Therefore, wheel-running activity was recorded from null mutants and wildtype controls under LD 12:12 and DD conditions. Mice of all three strains appeared to entrain normally to LD 12:12 and could re-entrain to both phase advances and phase delays of the light cycle. The response of the circadian pacemaker of all three genotypes to acute light pulses appeared to be normal. The retinal innervation of the SCN in NGFI-A-/- mice and the photic induction of Fos in the SCN of both NGFI-A-/- and NGFI-A-/-/B-/- mice were indistinguishable from wildtype mice. These results indicate that induction of NGFI-A and NGFI-B is not required for photic entrainment or phase shifting of the mouse circadian system.


Asunto(s)
Ritmo Circadiano/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Inmediatas-Precoces , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Adaptación Fisiológica/fisiología , Animales , Ritmo Circadiano/genética , Cricetinae , Proteínas de Unión al ADN/genética , Proteína 1 de la Respuesta de Crecimiento Precoz , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Estimulación Luminosa , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides , Núcleo Supraquiasmático/anatomía & histología , Núcleo Supraquiasmático/fisiología , Factores de Transcripción/genética
10.
Neurobiol Aging ; 14(5): 441-6, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8247226

RESUMEN

We investigated age-related changes in a molecular mechanism associated with synchronization of circadian rhythms to the environment. In mammals, the suprachiasmatic nucleus (SCN) of the hypothalamus contains a circadian pacemaker that regulates a variety of physiological and behavioral rhythms. Recent studies have demonstrated photic induction of immediate early genes (IEGs) such as c-fos in the SCN in a circadian-phase dependent manner, suggesting that IEGs may be part of the pathway for entrainment of circadian rhythms. We find that there is a decreased response of the IEGs c-fos, and NGFI-A but not NGFI-B in the SCN of old animals after photic stimulation. Changes in gene transcription indicated by IEGs may provide insights into the molecular machinery of the biological clock and ultimately elucidate mechanisms underlying the age-dependent decay of circadian organization.


Asunto(s)
Envejecimiento/genética , Ritmo Circadiano/fisiología , Expresión Génica/fisiología , Genes Inmediatos-Precoces/fisiología , Luz , Núcleo Supraquiasmático/metabolismo , Envejecimiento/metabolismo , Animales , Sondas de ADN , Genes fos/fisiología , Procesamiento de Imagen Asistido por Computador , Masculino , Factores de Crecimiento Nervioso/biosíntesis , Hibridación de Ácido Nucleico , Estimulación Luminosa , Ratas , Ratas Endogámicas F344 , Péptido Intestinal Vasoactivo/biosíntesis
11.
J Comp Neurol ; 399(4): 541-60, 1998 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-9741482

RESUMEN

In contrast to some previous reports suggesting a delay in synapse formation in vitro, we found that under ideal conditions, most hippocampal and hypothalamic rat neurons were synaptically coupled after 3 or 4 days in vitro. Synaptophysin immunocytochemistry revealed strongly stained presynaptic boutons by 3 days in vitro. Studies with time-lapse laser confocal imaging of FM1-43 revealed that axonal boutons were recycling their synaptic vesicles, an indication of synapse formation, as early as 3 days after plating. To test the hypothesis that neurite outgrowth was enhanced in high-density cultures, thereby increasing the probability of synapse formation, neurons were transfected with the jellyfish green fluorescent protein (GFP) gene. After 2 days in high-density cultures, green fluorescent neurites were about three times longer than in sister neurons plated in low-density cultures. Even in single dishes, GFP-transfected cells in contact with other neurons had neurites that were at least three times longer and grew faster than more isolated cells. Neurons grew longer neurites (+51%) when growing on surface membranes of heat-killed neurons than on polylysine, underlining the importance of plasma membrane contact. Calcium imaging with fura-2 and whole cell recording showed that both GABA and glutamate presynaptic release occurred after 3 or 4 days in vitro in high-density cultures but was absent in low-density cultures at this time. Together, these morphological, cytochemical, and physiological data suggest that the distance an axon must grow to find a postsynaptic partner plays a substantial role in the timing of synapse formation. Although other factors in vitro may also play a role, the distance to a postsynaptic target, which defines the interval during which an axon grows to its target, can probably account for much of the difference in timing of synapse formation previously reported in vitro. A short intercell distance may increase the concentration of limited amounts of trophic factors available to a nearby cell, and once contact is made, a neuronal membrane provides a superior substrate for neuritic elongation.


Asunto(s)
Axones/fisiología , Hipotálamo/citología , Sinapsis/fisiología , 2-Amino-5-fosfonovalerato/farmacología , 6-Ciano 7-nitroquinoxalina 2,3-diona/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Axones/química , Bicuculina/farmacología , Calcio/análisis , Calcio/fisiología , Recuento de Células , Células Cultivadas , Estimulación Eléctrica , Antagonistas de Aminoácidos Excitadores/farmacología , Colorantes Fluorescentes , Fura-2 , Antagonistas del GABA/farmacología , Ácido Glutámico/fisiología , Proteínas Fluorescentes Verdes , Hipocampo/citología , Hipotálamo/química , Hipotálamo/fisiología , Indicadores y Reactivos , Proteínas Luminiscentes , Neuritas/química , Neuritas/efectos de los fármacos , Neuritas/fisiología , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Vesículas Sinápticas/química , Sinaptofisina/análisis , Tetrodotoxina/farmacología , Ácido gamma-Aminobutírico/fisiología
12.
Neuroscience ; 65(3): 797-803, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7609878

RESUMEN

In vivo studies reported that cholinergic agents affect mammalian circadian rhythmicity. To study phase resetting properties of cholinergic compounds more directly, we carried out experiments in rat suprachiasmatic nuclei slices. Compounds were added to the perfusate for 1 h at specific phases of the circadian cycle. On the following day, the time of peak neuronal activity, a measure of the phase of the endogenous circadian pacemaker, was assessed by means of extracellular recording in the suprachiasmatic nuclei. The peak of neuronal activity occurred at circadian time 5.8 +/- 0.7 (mean +/- 95% confidence limits) in the control slice (circadian time 0: lights-on). Ten-micromolar carbachol had no effect on the phase of the circadian rhythm when given at circadian times 6 and 15, while at circadian time 21 a phase advance of one hour was observed. By contrast, 10 microM nicotine significantly phase advanced (> 1 h) the neuronal circadian rhythm at all but one experimental circadian phase. The circadian times of maximal nicotinic phase advances were 15 (+2.6 h) and 21 (+2.8 h). A concentration response curve for nicotine was generated and pharmacological blocking experiments were performed at circadian time 15. The estimated maximum response of nicotine was 3.4 h, and the estimated concentration for half maximal response was 5 microM. The Hill coefficient (= 1.08) indicated that the effects of nicotine may be explained by a single receptor occupancy model. Mecamylamine (20 microM) almost completely antagonized the nicotinic phase-advances, whereas tetrodotoxin (1 microM) or high Mg2+ (10 mM) did not significantly attenuate the nicotinic phase-advances.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Nicotina/farmacología , Núcleo Supraquiasmático/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Magnesio/farmacología , Masculino , Mecamilamina/farmacología , Ratas , Ratas Wistar , Factores de Tiempo
13.
Neuroscience ; 120(4): 1115-24, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12927216

RESUMEN

Previous studies have documented changes in expression of the immediate early gene (IEG) c-fos and Fos protein in the brain between sleep and wakefulness. Such expression differences implicate changes in transcriptional regulation across behavioral states and suggest that other transcription factors may also be affected. In the current study, we examined the expression of seven fos/jun family member mRNAs (c-fos, fosB, fos related antigen (fra)1, fra-2, junB, c-jun, and junD) and three other IEG mRNAs (egr-1, egr-3, and nur77) in mouse brain following short-term (6 h) sleep deprivation (SD) and 4 h recovery sleep (RS) after SD. Gene expression was quantified in seven brain regions by real-time reverse transcription-polymerase chain reaction (RT-PCR). Multivariate analysis of variance revealed statistically significant variation in cerebral cortex, basal forebrain, thalamus and cerebellum. Levels of c-fos and fosB mRNA were elevated during SD in all four of these brain regions. In the cerebral cortex, junB mRNA was also elevated during SD whereas, in the basal forebrain, fra-1 and fra-2 mRNA levels increased in this condition. During RS, the only IEG mRNA to undergo significant increase was fra-2 in the cortex. C-jun and junD mRNAs were invariant across experimental conditions. These results indicate that the expression of fos/jun family members is diverse during SD. Among other IEGs, nur77 mRNA expression across conditions was similar to c-fos and fosB, egr-1 mRNA was elevated during SD in the cortex and basal forebrain, and egr-3 mRNA was elevated in the cortex during both SD and RS. The similarity of fosB and nur77 expression to c-fos expression indicates that these genes might also be useful markers of functional activity. Along with our previous results, the increased levels of fra-2 and egr-3 mRNAs during RS reported here suggest that increased mRNA expression during sleep is rare and may be anatomically restricted.


Asunto(s)
Encéfalo/metabolismo , Expresión Génica , Genes Inmediatos-Precoces , Privación de Sueño/metabolismo , Sueño/fisiología , Análisis de Varianza , Animales , Encéfalo/anatomía & histología , Química Encefálica , Inmunohistoquímica/métodos , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sueño/genética , Privación de Sueño/genética , Factores de Tiempo
14.
Neuroscience ; 116(1): 187-200, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12535952

RESUMEN

Although sleep is thought to be restorative from prior wakeful activities, it is not clear what is being restored. To determine whether the synthesis of macromolecules is increased in the cerebral cortex during sleep, we subjected C57BL/6 mice to 6 hours of sleep deprivation and then screened the expression of 1176 genes of known function by using cDNA arrays. The expression of the heat shock proteins (HSP), endoplasmic reticulum protein (ERp72) and glucose-regulated protein (GRp78), was among the genes whose expression was significantly elevated in the cortex during sleep deprivation, whereas GRp78 and GRp94 mRNAs were elevated in the cortex during recovery sleep after sleep deprivation, as confirmed by conventional and quantitative real-time polymerase chain reaction and/or Northern analyses. A systematic evaluation of the expression of six heat shock protein family members (ERP72, GRp78, GRp94, HSP27, HSP70-1, and HSP84) in seven brain regions revealed increased mRNA levels in cortex, basal forebrain, hypothalamus, cerebellum and medulla during sleep deprivation, whereas increased mRNA levels during recovery sleep were limited to the cortex and medulla. Immunohistochemical studies identified increased numbers of GRp78-, GRp94-, and ERp72-immunoreactive cells in the dorsal and lateral cortex during sleep deprivation but, during recovery sleep, elevated numbers of these cells were found only in the lateral cortex. In the medulla, increased numbers of GRp94-immunoreactive cells were observed in nucleus tractus solitarius, dorsal motor nucleus of the vagus and the rostroventrolateral medulla during recovery sleep. The widespread increase of heat shock protein family mRNAs in brain during sleep deprivation may be a neuroprotective response to prolonged wakefulness. In contrast, the relatively limited heat shock protein family mRNA expression during recovery sleep may be related to the role of heat shock proteins in protein biogenesis and thus to the restorative function of sleep.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Choque Térmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Privación de Sueño/metabolismo , Sueño , Animales , Northern Blotting , Chaperón BiP del Retículo Endoplásmico , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Sleep ; 17(1): 28-36, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8191200

RESUMEN

Algorithms for scoring sleep/waking states and transitions to REM sleep (NRTs) in rats are presented and validated. Both algorithms are based on electroencephalographic (EEG) power in delta (0.5-4.0 Hz), theta (6-9 Hz) and sigma (10-14 Hz) frequency bands, and electromyogram (EMG) intensity. Waking is scored when EMG intensity is high or (sigma power).(theta power) is low. Nonrapid eye movement (NREM) sleep is scored in nonwaking epochs having high (delta power)/(theta power). Rapid eye movement (REM) sleep is scored in nonwaking epochs having low (delta power)/(theta power). NRTs are identified by the EEG phenomena of the pre-REM sleep phase of NREM sleep. Algorithms are validated by comparison with records scored independently by two investigators based on visual examination of EEGs and EMGs. The sleep/waking-state scoring algorithm produces greater than 90% agreement with visual scoring. The NRT-scoring algorithm produces 88-92% agreement with visual scoring. Scoring NRTs based on the phenomena of the pre-REM sleep phase of NREM sleep, instead of relying solely on REM sleep expression for identification of REM sleep onset, reveals a significant population of brief REM sleep episodes that are ignored by most sleep cycle analyses and allows independent quantification of REM sleep timing and maintenance.


Asunto(s)
Encéfalo/fisiología , Fases del Sueño/fisiología , Sueño REM/fisiología , Algoritmos , Animales , Electroencefalografía , Masculino , Ratas , Ratas Wistar , Vigilia/fisiología
16.
Sleep ; 16(1): 1-7, 1993 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8456228

RESUMEN

The two-process model of sleep regulation posits that a homeostatic drive to sleep, referred to as Process S, increases with time spent awake. The purpose of this study was to evaluate whether immediate early gene (IEG) expression increases in the brain in proportion to time spent awake, when Process S would be expected to increase. Rats were deprived of sleep by cage tapping, cage rotation and gentle handling beginning at light onset for 45 minutes, 3 hours or 6 hours. At the end of the deprivation periods, deprived animals and an equal number of controls were decapitated, the brains dissected into subregions and frozen. Northern blots were prepared from cortex, thalamus, cerebellum, pons and hypothalamus and hybridized with cDNA probes to five IEG mRNAs; c-fos, c-jun, junB, NGFI-A and NGFI-B. Basal levels of c-fos mRNA were detectable in all brain regions from all animals. Sleep-deprived animals showed higher expression of c-fos mRNA than control animals following 45 minutes and 6 hours of sleep deprivation in all brain regions examined, with the greatest increases observed in the cerebellum. Surprisingly, only the pons and cerebellum showed clear increases at the 3-hour timepoint. In contrast to c-fos, c-jun mRNA was essentially invariant among the animals while junB mRNA was inconsistently elevated. The expression of NGFI-A and NGFI-B was similar to the c-fos pattern but of lesser magnitude.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Química Encefálica , Encéfalo/fisiopatología , Regulación de la Expresión Génica/fisiología , Privación de Sueño/fisiología , Animales , Northern Blotting , Sondas de ADN , Femenino , Proteínas Proto-Oncogénicas c-fos/análisis , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
Sleep ; 23(7): 867-74, 2000 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11083595

RESUMEN

The hypocretin/orexin ligand-receptor system has recently been implicated in the sleep disorder narcolepsy. During the dark (active) period, null mutants of the prepro-orexin (prepro-hypocretin) gene have cataplectic attacks and increased levels of both rapid eye movement (REM) and non-REM (NREM) sleep. Intracerebroventricular injection of one of the encoded neuropeptides, orexin-A, early in the light period increases wakefulness and reduces REM sleep in the rat, suggesting that this system may be involved in the normal regulation of sleep and wakefulness. To further test this hypothesis, we measured hypocretin (hcrt) mRNA levels by both Northern hybridization and Taqman analysis in mouse and rat hypothalamus after short-term (6 h) sleep deprivation (SD) and 2-4 hours after recovery from SD. Although our SD procedures effectively induced a sleep debt and increased c-fos mRNA expression in the cortex and hypothalamus as described by other investigators, we found that hcrt mRNA levels were not significantly changed in either species either after SD or after recovery from SD. If the hcrt system is involved in normal regulation of sleep and wakefulness, longer periods of SD may be necessary to affect hcrt mRNA levels or changes may occur at the protein rather than mRNA level. Alternatively, this system may also be involved in another function that counterbalances any SD-induced changes in hcrt mRNA levels.


Asunto(s)
Hipotálamo/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Privación de Sueño/metabolismo , Sueño REM/fisiología , Animales , Northern Blotting , Electrodos Implantados , Electroencefalografía , Electromiografía , Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Orexinas , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/genética , Ratas , Vigilia/fisiología
18.
Psychoneuroendocrinology ; 23(2): 161-73, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9621396

RESUMEN

Considerable data support a role for cholinergic influences on the circadian system. The extent to which these influences are mediated by nicotinic acetylcholine receptors (nAChRs) has been controversial, as have the specific actions of nicotine and acetylcholine in the suprachiasmatic nucleus (SCN) of the hypothalamus. In this article we review the existing literature and present new data supporting an important role for nAChRs in both the developing and adult SCN. Specifically, we present data showing that nicotine is capable of causing phase shifts in the circadian rhythms of rats. Like light and carbachol, nicotine appears to cause phase delays in the early subjective night and phase advances in the late subjective night. In the isolated SCN slice, however, only phase advances are seen, and, surprisingly, nicotine appears to cause the inhibition rather than the excitation of neurons. Among nAChR subunit mRNAs, alpha 7 appears to be the most abundant subunit in the adult SCN, whereas in the perinatal period, the more typical nAChRs with higher affinity for nicotine predominate in the SCN. This developmental change in subunit expression may explain the dramatic sensitivity of the perinatal SCN to nicotine that we have previously observed. The effects of nicotine on the SCN may contribute to alterations caused by nicotine in other physiological systems. These effects might also contribute to the dependence properties of nicotine through influences on arousal.


Asunto(s)
Ritmo Circadiano/fisiología , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptores Nicotínicos/fisiología , Animales , Ritmo Circadiano/efectos de los fármacos , Humanos , Ratas , Receptores Nicotínicos/efectos de los fármacos
19.
Brain Res Mol Brain Res ; 25(1-2): 151-6, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7984042

RESUMEN

The mammalian suprachiasmatic nuclei (SCN) contain a circadian clock that produces approximately 24 h rhythms of physiology and behavior even during constant dark. Under such conditions, light stimuli applied during the subjective night induce phase shifts of circadian rhythms and increase immediate early gene expression (c-fos) in the SCN. In vitro preparations of the SCN continue to show circadian rhythms of metabolic rate and neuronal firing rates, which can be phase shifted by non-photic stimuli. This study was designed to investigate whether the SCN display a rhythm of c-fos mRNA levels in vitro and whether quipazine, which phase-shifts the SCN circadian clock, induces c-fos expression in vitro. Levels of c-fos mRNA were found to be significantly higher in the subjective day than subjective night in the SCN in vitro. This rhythm parallels other in vivo and in vitro rhythms in SCN metabolic and neuronal activity and is consistent with previous in vivo work showing higher daytime levels of Fos-like immunoreactivity in animals maintained under constant dark conditions. Quipazine treatment during the subjective day (which phase-advances the circadian rhythm of neuronal firing in the SCN) decreased c-fos mRNA levels in the dorsomedial but not ventrolateral SCN, but quipazine did not affect c-fos levels when administered at night. This effect is consistent with serotonergic agonists inhibiting SCN neuronal activity and is the first evidence that a non-photic phase-shifting stimulus alters c-fos in the SCN at a phase-appropriate time.


Asunto(s)
Ritmo Circadiano/efectos de los fármacos , Genes fos , ARN Mensajero/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Núcleo Supraquiasmático/efectos de los fármacos , Animales , Ritmo Circadiano/genética , Técnicas In Vitro , Masculino , Quipazina/farmacología , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Núcleo Supraquiasmático/metabolismo
20.
Brain Res Mol Brain Res ; 28(2): 239-50, 1995 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7723623

RESUMEN

Identification of the neurotransmitter receptor subtypes within the suprachiasmatic nuclei (SCN) will further understanding of the mechanism of the biological clock and may provide targets to manipulate circadian rhythms pharmacologically. We have focused on the ionotropic GABA and glutamate receptors because these appear to account for the majority of synaptic communication in the SCN. Of the 15 genes known to code for GABA receptor subunits in mammals we have examined the expression of 12 in the SCN, neglecting only the alpha 6, gamma 3, and rho 2 subunits. Among glutamate receptors, we have focused on the five known genes coding for the NMDA receptor subunits, and two subunits which help comprise the kainate-selective receptors. Expression was characterized by Northern analysis with RNA purified from a large number of mouse SCN and compared to expression in the remaining hypothalamus, cortex and cerebellum. This approach provided a uniform source of RNA to generate many replicate blots, each of which was probed repeatedly. The most abundant GABA receptor subunit mRNAs in the SCN were alpha 2, alpha 5, beta 1, beta 3, gamma 1 and gamma 2. The rho 1 (rho 1) subunit, which produces GABAC pharmacology, was expressed primarily in the retina in three different species and was not detectable in the mouse SCN despite a common embryological origin with the retina. For several GABA subunits we detected additional mRNA species not previously described. High expression of both genes coding for glutamic acid decarboxylase (GAD65 and GAD67) was also found in the SCN. Among the NMDA receptor subunits, NR1 was most highly expressed in the SCN followed in order of abundance by NR2B, NR2A, NR2C and NR2D. In addition, both GluR5 and GluR6 show clear expression in the SCN, with GluR5 being the most SCN specific. This approach provides a simple measure of receptor subtype expression, complements in situ hybridization studies, and may suggest novel isoforms of known subunits.


Asunto(s)
Corteza Cerebral/fisiología , ARN Mensajero/genética , Receptores de GABA-A/genética , Receptores de GABA/genética , Receptores de N-Metil-D-Aspartato/genética , Núcleo Supraquiasmático/fisiología , Animales , Relojes Biológicos , Cerebelo/fisiología , Ritmo Circadiano , Hipotálamo/fisiología , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA