Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34764226

RESUMEN

Growing evidence indicates that microglia impact brain function by regulating synaptic pruning and formation as well as synaptic transmission and plasticity. Iba1 (ionized Ca+2-binding adapter protein 1), encoded by the Allograft inflammatory factor 1 (Aif1) gene, is an actin-interacting protein in microglia. Although Iba1 has long been used as a cellular marker for microglia, its functional role remains unknown. Here, we used global, Iba1-deficient (Aif1-/-) mice to characterize microglial activity, synaptic function, and behavior. Microglial imaging in acute hippocampal slices and fixed tissues from juvenile mice revealed that Aif1-/- microglia display reductions in ATP-induced motility and ramification, respectively. Biochemical assays further demonstrated that Aif1-/- brain tissues exhibit an altered expression of microglial-enriched proteins associated with synaptic pruning. Consistent with these changes, juvenile Aif1-/- mice displayed deficits in the excitatory synapse number and synaptic drive assessed by neuronal labeling and whole-cell patch-clamp recording in acute hippocampal slices. Unexpectedly, microglial synaptic engulfment capacity was diminished in juvenile Aif1-/- mice. During early postnatal development, when synapse formation is a predominant event in the hippocampus, the excitatory synapse number was still reduced in Aif1-/- mice. Together, these findings support an overall role of Iba1 in excitatory synaptic growth in juvenile mice. Lastly, postnatal synaptic deficits persisted in adulthood and correlated with significant behavioral changes in adult Aif1-/- mice, which exhibited impairments in object recognition memory and social interaction. These results suggest that Iba1 critically contributes to microglial activity underlying essential neuroglia developmental processes that may deeply influence behavior.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Hipocampo/metabolismo , Proteínas de Microfilamentos/metabolismo , Microglía/metabolismo , Sinapsis/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Neurogénesis/fisiología , Plasticidad Neuronal/fisiología , Neuronas/metabolismo , Fagocitosis/fisiología , Transmisión Sináptica/fisiología
2.
J Sleep Res ; 30(4): e13262, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33403714

RESUMEN

Subtle changes in sleep architecture can accompany and be symptomatic of many diseases or disorders. In order to probe and understand the complex interactions between sleep and health, the ability to model, track, and modulate sleep in preclinical animal models is vital. While various methods have been described for scoring experimental sleep recordings, few are designed to work in real time - a prerequisite for closed-loop sleep manipulation. In the present study, we have developed algorithms and software to classify sleep in real time and validated it on C57BL/6 mice (n = 8). Hidden Markov models of baseline sleep dynamics were fitted using an unsupervised algorithm to electroencephalogram (EEG) and electromyogram (EMG) data for each mouse, and were able to classify sleep in a manner highly concordant with manual scoring (Cohen's Kappa >75%) up to 3 weeks after model construction. This approach produced reasonably accurate estimates of common sleep metrics (proportion, mean duration, and number of bouts). After construction, the models were used to track sleep in real time and accomplish selective rapid eye movement (REM) sleep restriction by triggering non-invasive somatosensory stimulation. During REM restriction trials, REM bout duration was significantly reduced, and the classifier continued to perform satisfactorily despite the disrupted sleep patterns. The software can easily be tailored for use with other commercial or customised methods of sleep disruption (e.g. stir bar, optogenetic stimulation, etc.) and could serve as a robust platform to facilitate closed-loop experimentation. The source code and documentation are freely available upon request from the authors.


Asunto(s)
Algoritmos , Electroencefalografía , Electromiografía , Sueño/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Sueño REM
3.
Am J Physiol Regul Integr Comp Physiol ; 318(4): R781-R789, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32130024

RESUMEN

Sleep loss contributes to the development of cardiovascular, metabolic, and neurological disorders by promoting a systemic proinflammatory phenotype. The neuroendocrine-immune mechanisms contributing to such pathologies are poorly understood. The sympathetic nervous system (SNS) regulates immunity and is often activated following sleep disturbances. The aims of this study were to determine 1) the effect of SNS inhibition on inflammatory responses to sleep fragmentation (SF) and 2) whether homeostasis can be restored after 1 wk of recovery sleep. We measured stress responses (norepinephrine and corticosterone), gene expression levels of pro- and anti-inflammatory cytokines in peripheral (heart, liver, and spleen) tissues, and protein levels of cytokines and chemokines in serum of female mice that were subjected to acute SF for 24 h, chronic SF for 8 wk, or 7 days of recovery after chronic SF. In each experiment, SF and control mice were chemically sympathectomized with 6-hydroxydopamine (6-OHDA) or injected with vehicle. Both acute and chronic SF elevated mRNA and protein levels of cytokines in peripheral tissues. Changes in inflammatory responses mirrored stress-axes activation, with increased corticosterone and norepinephrine in SF mice. 6-OHDA treatment significantly alleviated SF-induced inflammation, thus providing evidence of SNS regulation of peripheral inflammation from SF. Effects of chronic SF were more severe than acute SF, and 1 wk of recovery from SF sufficiently alleviated peripheral inflammatory responses but not NE responses.


Asunto(s)
Inflamación/prevención & control , Privación de Sueño/patología , Simpatectomía Química , Animales , Cortisona/sangre , Femenino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/sangre , Oxidopamina/toxicidad , Estrés Fisiológico , Simpaticolíticos/toxicidad
4.
J Neuroinflammation ; 15(1): 154, 2018 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-29789012

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) begins with the application of mechanical force to the head or brain, which initiates systemic and cellular processes that are hallmarks of the disease. The pathological cascade of secondary injury processes, including inflammation, can exacerbate brain injury-induced morbidities and thus represents a plausible target for pharmaceutical therapies. We have pioneered research on post-traumatic sleep, identifying that injury-induced sleep lasting for 6 h in brain-injured mice coincides with increased cortical levels of inflammatory cytokines, including tumor necrosis factor (TNF). Here, we apply post-traumatic sleep as a physiological bio-indicator of inflammation. We hypothesized the efficacy of novel TNF receptor (TNF-R) inhibitors could be screened using post-traumatic sleep and that these novel compounds would improve functional recovery following diffuse TBI in the mouse. METHODS: Three inhibitors of TNF-R activation were synthesized based on the structure of previously reported TNF CIAM inhibitor F002, which lodges into a defined TNFR1 cavity at the TNF-binding interface, and screened for in vitro efficacy of TNF pathway inhibition (IκB phosphorylation). Compounds were screened for in vivo efficacy in modulating post-traumatic sleep. Compounds were then tested for efficacy in improving functional recovery and verification of cellular mechanism. RESULTS: Brain-injured mice treated with Compound 7 (C7) or SGT11 slept significantly less than those treated with vehicle, suggesting a therapeutic potential to target neuroinflammation. SGT11 restored cognitive, sensorimotor, and neurological function. C7 and SGT11 significantly decreased cortical inflammatory cytokines 3 h post-TBI. CONCLUSIONS: Using sleep as a bio-indicator of TNF-R-dependent neuroinflammation, we identified C7 and SGT11 as potential therapeutic candidates for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Complemento C7/uso terapéutico , Factores Inmunológicos/uso terapéutico , Receptores Tipo I de Factores de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Proteínas de Unión al Calcio/metabolismo , Complemento C7/química , Citocinas/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Factores Inmunológicos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Microglía/patología , Actividad Motora/efectos de los fármacos , Examen Neurológico , Reconocimiento en Psicología/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante , Trastornos del Sueño-Vigilia/tratamiento farmacológico , Trastornos del Sueño-Vigilia/etiología
5.
Brain Behav Immun ; 47: 131-40, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25585137

RESUMEN

Traumatic brain injury (TBI) is induced by mechanical forces which initiate a cascade of secondary injury processes, including inflammation. Therapies which resolve the inflammatory response may promote neural repair without exacerbating the primary injury. Specific derivatives of omega-3 fatty acids loosely grouped as specialized pro-resolving lipid mediators (SPMs) and termed resolvins promote the active resolution of inflammation. In the current study, we investigate the effect of two resolvin molecules, RvE1 and AT-RvD1, on post-traumatic sleep and functional outcome following diffuse TBI through modulation of the inflammatory response. Adult, male C57BL/6 mice were injured using a midline fluid percussion injury (mFPI) model (6-10min righting reflex time for brain-injured mice). Experimental groups included mFPI administered RvE1 (100ng daily), AT-RvD1 (100ng daily), or vehicle (sterile saline) and counterbalanced with uninjured sham mice. Resolvins or saline were administered daily for seven consecutive days beginning 3days prior to TBI to evaluate proof-of-principle to improve outcome. Immediately following diffuse TBI, post-traumatic sleep was recorded for 24h post-injury. For days 1-7 post-injury, motor outcome was assessed by rotarod. Cognitive function was measured at 6days post-injury using novel object recognition (NOR). At 7days post-injury, microglial activation was quantified using immunohistochemistry for Iba-1. In the diffuse brain-injured mouse, AT-RvD1 treatment, but not RvE1, mitigated motor and cognitive deficits. RvE1 treatment significantly increased post-traumatic sleep in brain-injured mice compared to all other groups. RvE1 treated mice displayed a higher proportion of ramified microglia and lower proportion of activated rod microglia in the cortex compared to saline or AT-RvD1 treated brain-injured mice. Thus, RvE1 treatment modulated post-traumatic sleep and the inflammatory response to TBI, albeit independently of improvement in motor and cognitive outcome as seen in AT-RvD1-treated mice. This suggests AT-RvD1 may impart functional benefit through mechanisms other than resolution of inflammation alone.


Asunto(s)
Lesiones Encefálicas/metabolismo , Ácidos Docosahexaenoicos/farmacología , Ácido Eicosapentaenoico/análogos & derivados , Microglía/metabolismo , Sueño/fisiología , Animales , Lesiones Encefálicas/fisiopatología , Cognición/efectos de los fármacos , Cognición/fisiología , Ácido Eicosapentaenoico/farmacología , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Memoria/efectos de los fármacos , Memoria/fisiología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Sueño/efectos de los fármacos
6.
Genome Res ; 21(8): 1223-38, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21734011

RESUMEN

Genetic reference populations in model organisms are critical resources for systems genetic analysis of disease related phenotypes. The breeding history of these inbred panels may influence detectable allelic and phenotypic diversity. The existing panel of common inbred strains reflects historical selection biases, and existing recombinant inbred panels have low allelic diversity. All such populations may be subject to consequences of inbreeding depression. The Collaborative Cross (CC) is a mouse reference population with high allelic diversity that is being constructed using a randomized breeding design that systematically outcrosses eight founder strains, followed by inbreeding to obtain new recombinant inbred strains. Five of the eight founders are common laboratory strains, and three are wild-derived. Since its inception, the partially inbred CC has been characterized for physiological, morphological, and behavioral traits. The construction of this population provided a unique opportunity to observe phenotypic variation as new allelic combinations arose through intercrossing and inbreeding to create new stable genetic combinations. Processes including inbreeding depression and its impact on allelic and phenotypic diversity were assessed. Phenotypic variation in the CC breeding population exceeds that of existing mouse genetic reference populations due to both high founder genetic diversity and novel epistatic combinations. However, some focal evidence of allele purging was detected including a suggestive QTL for litter size in a location of changing allele frequency. Despite these inescapable pressures, high diversity and precision for genetic mapping remain. These results demonstrate the potential of the CC population once completed and highlight implications for development of related populations.


Asunto(s)
Cruzamientos Genéticos , Endogamia , Sitios de Carácter Cuantitativo , Animales , Femenino , Variación Genética , Genotipo , Tamaño de la Camada/genética , Masculino , Ratones , Ratones Endogámicos , Fenotipo , Polimorfismo de Nucleótido Simple
7.
Exp Brain Res ; 232(9): 2709-19, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24760409

RESUMEN

Following mild traumatic brain injury (TBI), patients may self-treat symptoms of concussion, including post-traumatic headache, taking over-the-counter (OTC) analgesics. Administering one dose of OTC analgesics immediately following experimental brain injury mimics the at-home treated population of concussed patients and may accelerate the understanding of the relationship between brain injury and OTC pharmacological intervention. In the current study, we investigate the effect of acute administration of OTC analgesics on neurological function and cortical cytokine levels after experimental diffuse TBI in the mouse. Adult, male C57BL/6 mice were injured using a midline fluid percussion (mFPI) injury model of concussion (6-10 min righting reflex time for brain-injured mice). Experimental groups included mFPI paired with either ibuprofen (60 mg/kg, i.p.; n = 16), acetaminophen (40 mg/kg, i.p.; n = 9), or vehicle (15% ethanol (v/v) in 0.9% saline; n = 13) and sham injury paired OTC medicine or vehicle (n = 7-10 per group). At 24 h after injury, functional outcome was assessed using the rotarod task and a modified neurological severity score. Following behavior assessment, cortical cytokine levels were measured by multiplex ELISA at 24 h post-injury. To evaluate efficacy on acute inflammation, cortical cytokine levels were measured also at 6 h post-injury. In the diffuse brain-injured mouse, immediate pharmacological intervention did not attenuate or exacerbate TBI-induced functional deficits. Cortical cytokine levels were affected by injury, time, or their interaction. However, levels were not affected by treatment at 6 or 24 h post-injury. These data indicate that acute administration of OTC analgesics did not exacerbate or attenuate brain-injury deficits which may inform clinical recommendations for the at-home treated mildly concussed patient.


Asunto(s)
Anestésicos/farmacología , Anestésicos/uso terapéutico , Lesiones Encefálicas/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Medicamentos sin Prescripción/farmacología , Medicamentos sin Prescripción/uso terapéutico , Análisis de Varianza , Animales , Lesiones Encefálicas/complicaciones , Citocinas/metabolismo , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Examen Neurológico , Prueba de Desempeño de Rotación con Aceleración Constante , Factores de Tiempo
8.
Brain Inj ; 28(4): 504-10, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24702469

RESUMEN

PRIMARY OBJECTIVE: To test if the current model of diffuse brain injury produces chronic sleep disturbances similar to those reported by TBI patients. METHODS AND PROCEDURES: Adult male C57BL/6 mice were subjected to moderate midline fluid percussion injury (n = 7; 1.4 atm; 6-10 minutes righting reflex time) or sham injury (n = 5). Sleep-wake activity was measured post-injury using a non-invasive, piezoelectric cage system. Chronic sleep patterns were analysed weekly for increases or decreases in percentage sleep (hypersomnia or insomnia) and changes in bout length (fragmentation). MAIN OUTCOMES AND RESULTS: During the first week after diffuse TBI, brain-injured mice exhibited increased mean percentage sleep and mean bout length compared to sham-injured mice. Further analysis indicated the increase in mean percentage sleep occurred during the dark cycle. Injury-induced changes in sleep, however, did not extend beyond the first week post-injury and were not present in weeks 2-5 post-injury. CONCLUSIONS: Previously, it has been shown that the midline fluid percussion model used in this study immediately increased post-traumatic sleep. The current study extended the timeline of investigation to show that sleep disturbances extended into the first week post-injury, but did not develop into chronic sleep disturbances. However, the clinical prevalence of TBI-related sleep-wake disturbances warrants further experimental investigation.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Interleucina-1beta/metabolismo , Trastornos del Sueño-Vigilia/fisiopatología , Animales , Lesiones Encefálicas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Movimiento , Trastornos del Sueño-Vigilia/metabolismo , Factores de Tiempo , Vigilia
9.
J Neurosci Methods ; 404: 110063, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38301833

RESUMEN

BACKGROUND: Sleep perturbation is widely used to investigate the physiological mechanisms that mediate sleep-wake dynamics, and to isolate the specific roles of sleep in health and disease. However, state-of-the-art methods to accomplish sleep perturbation in preclinical models are limited in their throughput, flexibility, and specificity. NEW METHOD: A system was developed to deliver vibro-tactile somatosensory stimulation aimed at controlled, selective sleep perturbation. The frequency and intensity of stimulation can be tuned to target a variety of experimental applications, from sudden arousal to sub-threshold transitions between light and deep stages of NREM sleep. This device was activated in closed-loop to selectively interrupt REM sleep in mice. RESULTS: Vibro-tactile stimulation effectively and selectively interrupted REM sleep - significantly reducing the average REM bout duration relative to matched, unstimulated baseline recordings. As REM sleep was repeatedly interrupted, homeostatic mechanisms prompted a progressively quicker return to REM sleep. These effects were dependent on the parameters of stimulation applied. COMPARISON WITH EXISTING METHODS: Existing sleep perturbation systems often require moving parts within the cage and/or restrictive housing. The system presented is unique in that it interrupts sleep without invading the animal's space. The ability to vary stimulation parameters is a great advantage over existing methods, as it allows for adaptation in response to habituation and/or circadian/homeostatic changes in arousal threshold. CONCLUSIONS: The proposed method of stimulation demonstrates feasibility in affecting mouse sleep within a standard home cage environment, thus limiting environmental stress. Furthermore, the ability to tune frequency and intensity of stimulation allows for graded control over the extent of sleep perturbation, which potentially expands the utility of this technology beyond applications related to sleep.


Asunto(s)
Sueño REM , Sueño de Onda Lenta , Ratones , Animales , Sueño REM/fisiología , Sueño/fisiología , Nivel de Alerta , Homeostasis , Electroencefalografía
10.
Sleep ; 47(6)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38512801

RESUMEN

Accumulation of amyloid-ß (Aß) plays an important role in Alzheimer's disease (AD) pathology. There is growing evidence that disordered sleep may accelerate AD pathology by impeding the physiological clearance of Aß from the brain that occurs in normal sleep. Therapeutic strategies for improving sleep quality may therefore help slow disease progression. It is well documented that the composition and dynamics of sleep are sensitive to ambient temperature. We therefore compared Aß pathology and sleep metrics derived from polysomnography in 12-month-old female 3xTg-AD mice (n = 8) exposed to thermoneutral temperatures during the light period over 4 weeks to those of age- and sex-matched controls (n = 8) that remained at normal housing temperature (22°C) during the same period. The treated group experienced greater proportions of slow wave sleep (SWS)-i.e. epochs of elevated 0.5-2 Hz EEG slow wave activity during non-rapid eye movement (NREM) sleep-compared to controls. Assays performed on mouse brain tissue harvested at the end of the experiment showed that exposure to thermoneutral temperatures significantly reduced levels of DEA-soluble (but not RIPA- or formic acid-soluble) Aß40 and Aß42 in the hippocampus, though not in the cortex. With both groups pooled together and without regard to treatment condition, NREM sleep continuity and any measure of SWS within NREM at the end of the treatment period were inversely correlated with DEA-soluble Aß40 and Aß42 levels, again in the hippocampus but not in the cortex. These findings suggest that experimental manipulation of SWS could offer useful clues into the mechanisms and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Modelos Animales de Enfermedad , Ratones Transgénicos , Polisomnografía , Sueño de Onda Lenta , Animales , Enfermedad de Alzheimer/fisiopatología , Ratones , Péptidos beta-Amiloides/metabolismo , Sueño de Onda Lenta/fisiología , Femenino , Temperatura , Electroencefalografía , Encéfalo/fisiopatología , Encéfalo/metabolismo
11.
J Affect Disord ; 332: 299-308, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37060954

RESUMEN

BACKGROUND: Bright light therapy (BLT) is the first-line treatment for seasonal affective disorder. However, the neural mechanisms underlying BLT are unclear. To begin filling this gap, the present study examined the impact of BLT on sleep/wakefulness, daily rhythms, and the wakefulness-promoting orexin/hypocretin system in a diurnal rodent, Nile grass rats (Arvicanthis niloticus). METHODS: Male and female grass rats were housed under a 12:12 h light/dark cycle with dim light (50 lx) during the day. The experimental group received daily 1-h early morning BLT (full-spectrum white light, 10,000 lx), while the control group received narrowband red light for 4 weeks. Sleep/wakefulness and in-cage locomotor activity were monitored, followed by examination of hypothalamic prepro-orexin and orexin receptors OX1R and OX2R expression in corticolimbic brain regions. RESULTS: The BLT group had higher wakefulness during light treatment, better nighttime sleep quality, and improved daily rhythm entrainment compared to controls. The impact of BLT on the orexin system was sex- and brain region-specific, with males showing higher OX1R and OX2R in the CA1, while females showed higher prepro-orexin but lower OX1R and OX2R in the BLA, compared to same-sex controls. LIMITATIONS: The present study focused on the orexin system in a limited number of brain regions at a single time point. Sex wasn't a statistical factor, as male and female cohorts were run independently. CONCLUSIONS: The diurnal grass rats show similar behavioral responses to BLT as humans, thus could be a good model for further elucidating the neural mechanisms underlying the therapeutic effects of BLT.


Asunto(s)
Trastorno Afectivo Estacional , Animales , Femenino , Masculino , Ritmo Circadiano/fisiología , Murinae/metabolismo , Orexinas/metabolismo , Fototerapia , Trastorno Afectivo Estacional/terapia , Sueño/fisiología , Vigilia
12.
Behav Brain Funct ; 6: 47, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20670413

RESUMEN

BACKGROUND: A number of benefits from meditation have been claimed by those who practice various traditions, but few have been well tested in scientifically controlled studies. Among these claims are improved performance and decreased sleep need. Therefore, in these studies we assess whether meditation leads to an immediate performance improvement on a well validated psychomotor vigilance task (PVT), and second, whether longer bouts of meditation may alter sleep need. METHODS: The primary study assessed PVT reaction times before and after 40 minute periods of mediation, nap, or a control activity using a within subject cross-over design. This study utilized novice meditators who were current university students (n = 10). Novice meditators completed 40 minutes of meditation, nap, or control activities on six different days (two separate days for each condition), plus one night of total sleep deprivation on a different night, followed by 40 minutes of meditation.A second study examined sleep times in long term experienced meditators (n = 7) vs. non-meditators (n = 23). Experienced meditators and controls were age and sex matched and living in the Delhi region of India at the time of the study. Both groups continued their normal activities while monitoring their sleep and meditation times. RESULTS: Novice meditators were tested on the PVT before each activity, 10 minutes after each activity and one hour later. All ten novice meditators improved their PVT reaction times immediately following periods of meditation, and all but one got worse immediately following naps. Sleep deprivation produced a slower baseline reaction time (RT) on the PVT that still improved significantly following a period of meditation. In experiments with long-term experienced meditators, sleep duration was measured using both sleep journals and actigraphy. Sleep duration in these subjects was lower than control non-meditators and general population norms, with no apparent decrements in PVT scores. CONCLUSIONS: These results suggest that meditation provides at least a short-term performance improvement even in novice meditators. In long term meditators, multiple hours spent in meditation are associated with a significant decrease in total sleep time when compared with age and sex matched controls who did not meditate. Whether meditation can actually replace a portion of sleep or pay-off sleep debt is under further investigation.


Asunto(s)
Meditación , Desempeño Psicomotor , Sueño , Actigrafía , Adulto , Estudios Cruzados , Femenino , Humanos , India , Masculino , Registros Médicos , Persona de Mediana Edad , Pruebas Neuropsicológicas , Tiempo de Reacción , Privación de Sueño , Factores de Tiempo , Adulto Joven
13.
Proc Natl Acad Sci U S A ; 104(50): 20090-5, 2007 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-18077435

RESUMEN

Sleep is regulated by a homeostatic process that determines its need and by a circadian process that determines its timing. By using sleep deprivation and transcriptome profiling in inbred mouse strains, we show that genetic background affects susceptibility to sleep loss at the transcriptional level in a tissue-dependent manner. In the brain, Homer1a expression best reflects the response to sleep loss. Time-course gene expression analysis suggests that 2,032 brain transcripts are under circadian control. However, only 391 remain rhythmic when mice are sleep-deprived at four time points around the clock, suggesting that most diurnal changes in gene transcription are, in fact, sleep-wake-dependent. By generating a transgenic mouse line, we show that in Homer1-expressing cells specifically, apart from Homer1a, three other activity-induced genes (Ptgs2, Jph3, and Nptx2) are overexpressed after sleep loss. All four genes play a role in recovery from glutamate-induced neuronal hyperactivity. The consistent activation of Homer1a suggests a role for sleep in intracellular calcium homeostasis for protecting and recovering from the neuronal activation imposed by wakefulness.


Asunto(s)
Encéfalo/fisiología , Proteínas Portadoras/fisiología , Privación de Sueño/metabolismo , Sueño/fisiología , Animales , Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad , Proteínas de Andamiaje Homer , Ratones , Ratones Endogámicos AKR , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , ARN Mensajero/metabolismo , Sueño/genética , Privación de Sueño/genética
14.
F1000Res ; 92020.
Artículo en Inglés | MEDLINE | ID: mdl-32274013

RESUMEN

Sleep is a ubiquitous and complex behavior in both its manifestation and regulation. Despite its essential role in maintaining optimal performance, health, and well-being, the genetic mechanisms underlying sleep remain poorly understood. Here, we review the forward genetic approaches undertaken in the last four years to elucidate the genes and gene pathways affecting sleep and its regulation. Despite an increasing number of studies and mining large databases, a coherent picture on "sleep" genes has yet to emerge. We highlight the results achieved by using unbiased genetic screens mainly in humans, mice, and fruit flies with an emphasis on normal sleep and make reference to lessons learned from the circadian field.


Asunto(s)
Sueño , Animales , Pruebas Genéticas , Humanos
15.
Sci Rep ; 10(1): 14970, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917924

RESUMEN

In the U.S., opioid prescription for treatment of pain nearly quadrupled from 1999 to 2014. The diversion and misuse of prescription opioids along with increased use of drugs like heroin and fentanyl, has led to an epidemic in addiction and overdose deaths. The most common cause of opioid overdose and death is opioid-induced respiratory depression (OIRD), a life-threatening depression in respiratory rate thought to be caused by stimulation of opioid receptors in the inspiratory-generating regions of the brain. Studies in mice have revealed that variation in opiate lethality is associated with strain differences, suggesting that sensitivity to OIRD is genetically determined. We first tested the hypothesis that genetic variation in inbred strains of mice influences the innate variability in opioid-induced responses in respiratory depression, recovery time and survival time. Using the founders of the advanced, high-diversity mouse population, the Diversity Outbred (DO), we found substantial sex and genetic effects on respiratory sensitivity and opiate lethality. We used DO mice treated with morphine to map quantitative trait loci for respiratory depression, recovery time and survival time. Trait mapping and integrative functional genomic analysis in GeneWeaver has allowed us to implicate Galnt11, an N-acetylgalactosaminyltransferase, as a gene that regulates OIRD.


Asunto(s)
Analgésicos Opioides/efectos adversos , Variación Genética , Morfina/efectos adversos , N-Acetilgalactosaminiltransferasas/genética , Sitios de Carácter Cuantitativo , Insuficiencia Respiratoria/genética , Analgésicos Opioides/farmacología , Animales , Femenino , Masculino , Ratones , Morfina/farmacología , Insuficiencia Respiratoria/inducido químicamente
16.
Sci Rep ; 10(1): 10944, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32616800

RESUMEN

Most published sleep studies use three species: human, house mouse, or Norway rat. The degree to which data from these species captures variability in mammalian sleep remains unclear. To gain insight into mammalian sleep diversity, we examined sleep architecture in the spiny basal murid rodent Acomys cahirinus. First, we used a piezoelectric system validated for Mus musculus to monitor sleep in both species. We also included wild M. musculus to control for alterations generated by laboratory-reared conditions for M. musculus. Using this comparative framework, we found that A. cahirinus, lab M. musculus, and wild M. musculus were primarily nocturnal, but exhibited distinct behavioral patterns. Although the activity of A. cahirinus increased sharply at dark onset, it decreased sharply just two hours later under group and individual housing conditions. To further characterize sleep patterns and sleep-related variables, we set up EEG/EMG and video recordings and found that A. cahirinus sleep significantly more than M. musculus, exhibit nearly three times more REM, and sleep almost exclusively with their eyes open. The observed differences in A. cahirinus sleep architecture raise questions about the evolutionary drivers of sleep behavior.


Asunto(s)
Ritmo Circadiano , Ratones/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Ratones/clasificación
17.
Genetics ; 214(3): 719-733, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31896565

RESUMEN

The microbiome influences health and disease through complex networks of host genetics, genomics, microbes, and environment. Identifying the mechanisms of these interactions has remained challenging. Systems genetics in laboratory mice (Mus musculus) enables data-driven discovery of biological network components and mechanisms of host-microbial interactions underlying disease phenotypes. To examine the interplay among the whole host genome, transcriptome, and microbiome, we mapped QTL and correlated the abundance of cecal messenger RNA, luminal microflora, physiology, and behavior in a highly diverse Collaborative Cross breeding population. One such relationship, regulated by a variant on chromosome 7, was the association of Odoribacter (Bacteroidales) abundance and sleep phenotypes. In a test of this association in the BKS.Cg-Dock7m +/+ Leprdb/J mouse model of obesity and diabetes, known to have abnormal sleep and colonization by Odoribacter, treatment with antibiotics altered sleep in a genotype-dependent fashion. The many other relationships extracted from this study can be used to interrogate other diseases, microbes, and mechanisms.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Factores de Intercambio de Guanina Nucleótido/genética , Obesidad/genética , Receptores de Leptina/genética , Sueño/genética , Animales , Antibacterianos/farmacología , Bacteroides/genética , Cromosomas Humanos Par 7/genética , Microbioma Gastrointestinal/genética , Genómica , Genotipo , Humanos , Ratones , Obesidad/microbiología , Obesidad/fisiopatología
18.
Nat Sci Sleep ; 11: 113-121, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31496853

RESUMEN

BACKGROUND: Sleep disturbances are common maladies associated with human age. Sleep duration is decreased, sleep fragmentation is increased, and the timing of sleep onset and sleep offset is earlier. These disturbances have been associated with several neurodegenerative diseases. Mouse models for human sleep disturbances can be powerful due to the accessibility to neuroscientific and genetic approaches, but these are hampered by the fact that most mouse models employed in sleep research have spontaneous mutations in the biosynthetic pathway(s) regulating the rhythmic production of the pineal hormone melatonin, which has been implicated in human sleep. PURPOSE AND METHOD: The present study employed a non-invasive piezoelectric measure of sleep wake cycles in young, middle-aged and old CBA mice, a strain capable of melatonin biosynthesis, to investigate naturally-occurring changes in sleep and circadian parameters as the result of aging. RESULTS: The results indicate that young mice sleep less than do middle-aged or aged mice, especially during the night, while the timing of activity onset and acrophase is delayed in aged mice compared to younger mice. CONCLUSION: These data point to an effect of aging on the quality and timing of sleep in these mice but also that there are fundamental differences between control of sleep in humans and in laboratory mice.

19.
Front Neurosci ; 13: 969, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31619950

RESUMEN

People with diabetes are more likely to experience sleep disturbance than those without. Sleep disturbance can cause daytime sleepiness in diabetic patients, which may impair their daytime performance or even lead to workplace injuries. Therefore, restoring the normal sleep-wake cycle is critical for diabetic patients who experience daytime sleepiness. Previous data on a diabetic mouse model, the db/db mice, have demonstrated that the total sleep time and sleep fragmentation are increased and the daily rhythm of the sleep-wake cycle is attenuated. Accumulating evidence has shown that active time-restricted feeding (ATRF), in which the timing of food availability is restricted to the active-phase, is beneficial to metabolic health. However, it is unknown whether ATRF restores the normal sleep-wake cycle in diabetes. To test that, we used a non-invasive piezoelectric system to monitor the sleep-wake profile in the db/db mice with ad libitum feeding (ALF) as a baseline and then followed with ATRF. The results showed that at baseline, db/db mice exhibited abnormal sleep-wake patterns: the sleep time percent during the light-phase was decreased, while during the dark-phase it was increased with unusual cycling compared to control mice. In addition, the sleep bout length during both the light-phase and the full 24-h period was shortened in db/db mice. Analysis of the sleep-wake circadian rhythm showed that ATRF effectively restored the circadian but suppressed the ultradian oscillations of the sleep-wake cycle in the db/db mice. In conclusion, ATRF may serve as a novel strategy for treating diabetes-induced irregularity of the sleep-wake cycle.

20.
PLoS One ; 14(8): e0212823, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31461439

RESUMEN

The objective was to determine the effects of sleep or lying deprivation on the behavior of dairy cows. Data were collected from 8 multi- and 4 primiparous cows (DIM = 199 ± 44 (mean ± SD); days pregnant = 77 ± 30). Using a crossover design, each cow experienced: 1) sleep deprivation implemented by noise or physical contact when their posture suggested sleep, and 2) lying deprivation imposed by a grid placed on the pen floor. One day before treatment (baseline), and treatment day (treatment) were followed by a 12-d washout period (with the first 7 d used to evaluate recovery). Study days were organized from 2100 to 2059. During habituation (d -3 and -2 before treatment), baseline (d -1), and trt (d 0), housing was individual boxstalls (mattress with no bedding). After treatment, cows returned to sand-bedded freestalls for a 7-d recovery period (d 1 to 7) where data on lying behaviors were collected. Following the recovery period, an additional 5-d period was provided to allow the cows a 12-d period between exposures to treatments. Daily lying time, number lying bouts, bout duration, and number of steps were recorded by dataloggers attached to the hind leg of cows throughout the study period. Data were analyzed using a mixed model including fixed effects of treatment (sleep deprivation vs. sleep and lying deprivation), day, and their interaction with significant main effects separated using a PDIFF statement (P ≤ 0.05). Interactions between treatment and day were detected for daily lying time and the number of bouts. Lying time was lower for both treatments during the treatment period compared to baseline. Lying time increased during the recovery period for both lying and sleep deprived cows. However, it took 4 d for the lying deprived cows to fully recover their lying time after treatment, whereas it took the sleep deprived cows 2 d for their lying time to return to baseline levels. Results suggest that both sleep and lying deprivation can have impact cow behavior. Management factors that limit freestall access likely reduce lying time and sleep, causing negative welfare implications for dairy cows.


Asunto(s)
Industria Lechera , Lactancia , Postura , Privación de Sueño/fisiopatología , Animales , Bovinos , Electroencefalografía , Femenino , Leche/metabolismo , Embarazo , Privación de Sueño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA