Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Res Sq ; 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38313253

RESUMEN

Sleep and circadian rhythm disruptions are comorbid features of many pathologies and can negatively influence numerous health conditions, including degenerative diseases, metabolic illnesses, cancer, and various neurological disorders. Genetic association studies linking sleep and circadian disturbances with disease susceptibility have mainly focused on changes in gene expression due to mutations, such as single-nucleotide polymorphisms. Thus, associations between sleep and/or circadian rhythm and alternative polyadenylation (APA), particularly in the context of other health challenges, are largely undescribed. APA is a process that generates various transcript isoforms from the same gene, resulting in effects on mRNA translation, stability, localization, and subsequent function. Here, we have identified unique APAs in rat brain that exhibit time-of-day-dependent oscillations in expression as well as APAs that are altered by sleep deprivation and the subsequent recovery period. Genes affected by APA usage include Mapt/Tau, Ntrk2, Homer1A, Sin3band Sorl. Sorl1 has two APAs which cycle with a 24 h period, one additional APA cycles with a 12 h period and one more that is reduced during recovery sleep. Finally, we compared sleep- or circadian-associated APAs with recently described APA-linked brain disorder susceptibility genes and found 46 genes in common.

3.
Front Syst Neurosci ; 17: 1212213, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37404868

RESUMEN

Sleep and circadian rhythms are observed broadly throughout animal phyla and influence neural plasticity and cognitive function. However, the few phylogenetically conserved cellular and molecular pathways that are implicated in these processes are largely focused on neuronal cells. Research on these topics has traditionally segregated sleep homeostatic behavior from circadian rest-activity rhythms. Here we posit an alternative perspective, whereby mechanisms underlying the integration of sleep and circadian rhythms that affect behavioral state, plasticity, and cognition reside within glial cells. The brain-type fatty acid binding protein, FABP7, is part of a larger family of lipid chaperone proteins that regulate the subcellular trafficking of fatty acids for a wide range of cellular functions, including gene expression, growth, survival, inflammation, and metabolism. FABP7 is enriched in glial cells of the central nervous system and has been shown to be a clock-controlled gene implicated in sleep/wake regulation and cognitive processing. FABP7 is known to affect gene transcription, cellular outgrowth, and its subcellular localization in the fine perisynaptic astrocytic processes (PAPs) varies based on time-of-day. Future studies determining the effects of FABP7 on behavioral state- and circadian-dependent plasticity and cognitive processes, in addition to functional consequences on cellular and molecular mechanisms related to neural-glial interactions, lipid storage, and blood brain barrier integrity will be important for our knowledge of basic sleep function. Given the comorbidity of sleep disturbance with neurological disorders, these studies will also be important for our understanding of the etiology and pathophysiology of how these diseases affect or are affected by sleep.

4.
Front Neurosci ; 16: 798994, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35844236

RESUMEN

Fatty acid binding proteins (FABPs) are a family of intracellular lipid chaperone proteins known to play critical roles in the regulation of fatty acid uptake and transport as well as gene expression. Brain-type fatty acid binding protein (FABP7) is enriched in astrocytes and has been implicated in sleep/wake regulation and neurodegenerative diseases; however, the precise mechanisms underlying the role of FABP7 in these biological processes remain unclear. FABP7 binds to both arachidonic acid (AA) and docosahexaenoic acid (DHA), resulting in discrete physiological responses. Here, we propose a dichotomous role for FABP7 in which ligand type determines the subcellular translocation of fatty acids, either promoting wakefulness aligned with Alzheimer's pathogenesis or promoting sleep with concomitant activation of anti-inflammatory pathways and neuroprotection. We hypothesize that FABP7-mediated translocation of AA to the endoplasmic reticulum of astrocytes increases astrogliosis, impedes glutamatergic uptake, and enhances wakefulness and inflammatory pathways via COX-2 dependent generation of pro-inflammatory prostaglandins. Conversely, we propose that FABP7-mediated translocation of DHA to the nucleus stabilizes astrocyte-neuron lactate shuttle dynamics, preserves glutamatergic uptake, and promotes sleep by activating anti-inflammatory pathways through the peroxisome proliferator-activated receptor-γ transcriptional cascade. Importantly, this model generates several testable hypotheses applicable to other neurodegenerative diseases, including amyotrophic lateral sclerosis and Parkinson's disease.

5.
Neuroglia ; 3(2): 73-83, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36909794

RESUMEN

Humans with post-traumatic stress disorder (PTSD) exhibit sleep disturbances that include insomnia, nightmares, and enhanced daytime sleepiness. Sleep disturbances are considered a hallmark feature of PTSD; however, little is known about the cellular and molecular mechanisms regulating trauma-induced sleep disorders. Using a rodent model of PTSD called "Single Prolonged Stress" (SPS) we examined the requirement of the brain-type fatty acid binding protein Fabp7, an astrocyte expressed lipid-signaling molecule, in mediating trauma-induced sleep disturbances. We measured baseline sleep/wake parameters and then exposed Fabp7 knock-out (KO) and wild-type (WT) C57BL/6N genetic background control animals to SPS. Sleep and wake measurements were obtained immediately following the initial trauma exposure of SPS, and again 7 days later. We found that active-phase (dark period) wakefulness was similar in KO and WT at baseline and immediately following SPS; however, it was significantly increased after 7 days. These effects were opposite in the inactive-phase (light period), where KOs exhibited increased wake in baseline and following SPS, but returned to WT levels after 7 days. To examine the effects of Fabp7 on unconditioned anxiety following trauma, we exposed KO and WT mice to the light-dark box test before and after SPS. Prior to SPS, KO and WT mice spent similar amounts of time in the lit compartment. Following SPS, KO mice spent significantly more time in the lit compartment compared to WT mice. These results demonstrate that mutations in an astrocyte-expressed gene (Fabp7) influence changes in stress-dependent sleep disturbances and associated anxiety behavior.

6.
Int J Mol Sci ; 22(23)2021 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-34884544

RESUMEN

Parkinson's Disease (PD) is the most common movement disorder, and the strongest genetic risk factor for PD is mutations in the glucocerebrosidase gene (GBA). Mutations in GBA also lead to the development of Gaucher Disease (GD), the most common type of lysosomal storage disorder. Current therapeutic approaches fail to address neurological GD symptoms. Therefore, identifying therapeutic strategies that improve the phenotypic traits associated with GD/PD in animal models may provide an opportunity for treating neurological manifestations of GD/PD. Thiazolidinediones (TZDs, also called glitazones) are a class of compounds targeted for the treatment of type 2 diabetes, and have also shown promise for the treatment of neurodegenerative disease, including PD. Here, we tested the efficacy of glitazone administration during development in a fly GD model with deletions in the GBA homolog, dGBA1b (GBA1ΔTT/ΔTT). We observed an optimal dose of pioglitazone (PGZ) at a concentration of 1 µM that reduced sleep deficits, locomotor impairments, climbing defects, and restoration of normal protein levels of Ref(2)P, a marker of autophagic flux, in GBA1ΔTT/ΔTT mutant flies, compared to GBA1+/+ control flies. These data suggest that PGZ may represent a potential compound with which to treat GD/PD by improving function of lysosomal-autophagy pathways, a cellular process that removes misfolded or aggregated proteins.


Asunto(s)
Enfermedad de Gaucher/tratamiento farmacológico , Glucosilceramidasa/deficiencia , Enfermedad de Parkinson/tratamiento farmacológico , Tiazolidinedionas/farmacología , Animales , Drosophila melanogaster , Enfermedad de Gaucher/etiología , Enfermedad de Gaucher/patología , Humanos , Masculino , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Fenotipo
7.
Artículo en Inglés | MEDLINE | ID: mdl-34056625

RESUMEN

The astrocyte brain-type fatty-acid binding protein (Fabp7) circadian gene expression is synchronized in the same temporal phase throughout mammalian brain. Cellular and molecular mechanisms that contribute to this coordinated expression are not completely understood, but likely involve the nuclear receptor Rev-erbα (NR1D1), a transcriptional repressor. We performed ChIP-seq on ventral tegmental area (VTA) and identified gene targets of Rev-erbα, including Fabp7. We confirmed that Rev-erbα binds to the Fabp7 promoter in multiple brain areas, including hippocampus, hypothalamus, and VTA, and showed that Fabp7 gene expression is upregulated in Rev-erbα knock-out mice. Compared to Fabp7 mRNA levels, Fabp3 and Fabp5 mRNA were unaffected by Rev-erbα depletion in hippocampus, suggesting that these effects are specific to Fabp7. To determine whether these effects of Rev-erbα depletion occur broadly throughout the brain, we also evaluated Fabp mRNA expression levels in multiple brain areas, including cerebellum, cortex, hypothalamus, striatum, and VTA in Rev-erbα knock-out mice. While small but significant changes in Fabp5 mRNA expression exist in some of these areas, the magnitude of these effects are minimal to that of Fabp7 mRNA expression, which was over 6-fold across all brain regions. These studies suggest that Rev-erbα is a transcriptional repressor of Fabp7 gene expression throughout mammalian brain.

8.
PLoS One ; 16(1): e0243743, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33406143

RESUMEN

Sleep is intimately linked to cognitive performance and exposure to traumatic stress that leads to post-traumatic stress disorder (PTSD) impairs both sleep and cognitive function. However, the contribution of pre-trauma sleep loss to subsequent trauma-dependent fear-associated memory impairment remains unstudied. We hypothesized that sleep deprivation (SD) prior to trauma exposure may increase the severity of a PTSD-like phenotype in rats exposed to single prolonged stress (SPS), a rodent model of PTSD. Rats were exposed to SPS alone, SD alone, or a combination of SPS+SD and measures of fear-associated memory impairments and vigilance state changes were compared to a group of control animals not exposed to SPS or SD. We found that SPS, and SPS+SD animals showed impaired fear-associated memory processing and that the addition of SD to SPS did not further exaggerate the effect of SPS alone. Additionally, the combination of SPS with SD results in a unique homeostatic sleep duration phenotype when compared to SD, SPS, or control animals. SPS exposure following SD represses homeostatic rebound and eliminates sleep-deprivation-induced increases in NREM sleep delta power. This work identifies a unique time frame where trauma exposure and sleep interact and identifies this window of time as a potential therapeutic treatment window for staving off the negative consequences of trauma exposure.


Asunto(s)
Progresión de la Enfermedad , Miedo/psicología , Trastornos de la Memoria/psicología , Índice de Severidad de la Enfermedad , Privación de Sueño/psicología , Sueño/fisiología , Estrés Psicológico/fisiopatología , Heridas y Lesiones/psicología , Animales , Extinción Psicológica , Homeostasis , Masculino , Trastornos de la Memoria/complicaciones , Trastornos de la Memoria/fisiopatología , Recuerdo Mental , Ratas Long-Evans , Privación de Sueño/complicaciones , Fases del Sueño/fisiología , Estrés Psicológico/complicaciones
9.
Front Neurol ; 11: 520, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32714261

RESUMEN

Seizure patterns observed in patients with epilepsy suggest that circadian rhythms and sleep/wake mechanisms play some role in the disease. This review addresses key topics in the relationship between circadian rhythms and seizures in epilepsy. We present basic information on circadian biology, but focus on research studying the influence of both the time of day and the sleep/wake cycle as independent but related factors on the expression of seizures in epilepsy. We review studies investigating how seizures and epilepsy disrupt expression of core clock genes, and how disruption of clock mechanisms impacts seizures and the development of epilepsy. We focus on the overlap between mechanisms of circadian-associated changes in SCN neuronal excitability and mechanisms of epileptogenesis as a means of identifying key pathways and molecules that could represent new targets or strategies for epilepsy therapy. Finally, we review the concept of chronotherapy and provide a perspective regarding its application to patients with epilepsy based on their individual characteristics (i.e., being a "morning person" or a "night owl"). We conclude that better understanding of the relationship between circadian rhythms, neuronal excitability, and seizures will allow both the identification of new therapeutic targets for treating epilepsy as well as more effective treatment regimens using currently available pharmacological and non-pharmacological strategies.

10.
Mol Brain ; 13(1): 26, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32093736

RESUMEN

The astrocyte brain-type fatty acid binding protein (Fabp7) gene expression cycles globally throughout mammalian brain, and is known to regulate sleep in multiple species, including humans. The mechanisms that control circadian Fabp7 gene expression are not completely understood and may include core circadian clock components. Here we examined the circadian expression of Fabp7 mRNA in the hypothalamus of core clock gene Bmal1 knock-out (KO) mice. We observed that the circadian rhythm of Fabp7 mRNA expression is blunted, while overall Fabp7 mRNA levels are significantly higher in Bmal1 KO compared to control (C57BL/6 J) mice. We did not observe any significant changes in levels of hypothalamic mRNA expression of Fabp3 or Fabp5, two other fatty acid binding proteins expressed in mammalian brain, between Bmal1 KO and control mice. These results suggest that Fabp7 gene expression is regulated by circadian processes and may represent a molecular link controlling the circadian timing of sleep with sleep behavior.


Asunto(s)
Factores de Transcripción ARNTL/deficiencia , Ritmo Circadiano/genética , Proteína de Unión a los Ácidos Grasos 7/genética , Regulación de la Expresión Génica , Factores de Transcripción ARNTL/metabolismo , Animales , Proteína de Unión a los Ácidos Grasos 7/metabolismo , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Matters Sel ; 20192019.
Artículo en Inglés | MEDLINE | ID: mdl-31938713

RESUMEN

Sleep is a behavior that exists broadly across animal phyla, from flies to humans, and is necessary for normal brain function. Recent studies in both vertebrates and invertebrates have suggested a role for glial cells in sleep regulatory processes. Changes in neural-glial interactions have been shown to be critical for synaptic plasticity and circuit function. Here, we wanted to test the hypothesis that changes in sleep pressure alters neural-glial interactions. In the fruit fly, Drosophila melanogaster, sleep is known to be regulated by mushroom body (MB) circuits. We used the technique GFP Reconstitution Across Synaptic Partners (GRASP) to test whether changes in sleep pressure affect neural-glial interactions between MB neurons and astrocytes, a specialized glial cell type known to regulate sleep in flies and mammals. The MB-astrocyte GRASP signal was reduced after 24 h of sleep deprivation, whereas the signal returned to baseline levels following 72 h of recovery. Social enrichment, which increases sleep drive, similarly reduced the MB-astrocyte GRASP signal. We did not observe any changes in the MB-astrocyte GRASP signal over time-of-day, or following paraquat exposure or starvation. These data suggest that changes in sleep pressure are linked to dynamic changes in neural-glial interactions between astrocytes and neuronal sleep circuits, which are not caused by normal rest-activity cycles or stressors.

12.
PLoS Genet ; 14(10): e1007724, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30379810

RESUMEN

Sleep contributes to cognitive functioning and is sufficient to alter brain morphology and function. However, mechanisms underlying sleep regulation remain poorly understood. In mammals, tumor necrosis factor-alpha (TNFα) is known to regulate sleep, and cytokine expression may represent an evolutionarily ancient mechanism in sleep regulation. Here we show that the Drosophila TNFα homologue, Eiger, mediates sleep in flies. We show that knockdown of Eiger in astrocytes, but not in neurons, significantly reduces sleep duration, and total loss-of-function reduces the homeostatic response to sleep loss. In addition, we show that neuronal, but not astrocyte, expression of the TNFα receptor superfamily member, Wengen, is necessary for sleep deprivation-induced homeostatic response and for mediating increases in sleep in response to human TNFα. These data identify a novel astrocyte-to-neuron signaling mechanism in the regulation of sleep homeostasis and show that the Drosophila cytokine, Eiger, represents an evolutionarily conserved mechanism of sleep regulation across phylogeny.


Asunto(s)
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Sueño/fisiología , Animales , Astrocitos/metabolismo , Astrocitos/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Evolución Molecular , Neuronas/metabolismo , Receptores del Factor de Necrosis Tumoral , Transducción de Señal , Sueño/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Neurosci ; 38(12): 2901-2910, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29563238

RESUMEN

Sleep-wake abnormalities are common in patients with Alzheimer's disease, and can be a major reason for institutionalization. However, an emerging concept is that these sleep-wake disturbances are part of the causal pathway accelerating the neurodegenerative process. Recently, new findings have provided intriguing evidence for a positive feedback loop between sleep-wake dysfunction and ß-amyloid (Aß) aggregation. Studies in both humans and animal models have shown that extended periods of wakefulness increase Aß levels and aggregation, and accumulation of Aß causes fragmentation of sleep. This perspective is aimed at presenting evidence supporting causal links between sleep-wake dysfunction and aggregation of Aß peptide in Alzheimer's disease, and explores the role of astrocytes, a specialized type of glial cell, in this context underlying Alzheimer's disease pathology. The utility of current animal models and the unexplored potential of alternative animal models for testing mechanisms involved in the reciprocal relationship between sleep disruption and Aß are also discussed.Dual Perspectives Companion Paper: Microglia-Mediated Synapse Loss in Alzheimer's Disease by Lawrence Rajendran and Rosa Paolicelli.


Asunto(s)
Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides/metabolismo , Astrocitos/patología , Trastornos del Sueño-Vigilia/complicaciones , Enfermedad de Alzheimer/patología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Trastornos del Sueño-Vigilia/patología
14.
Sci Adv ; 3(4): e1602663, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28435883

RESUMEN

Sleep is found widely in the animal kingdom. Despite this, few conserved molecular pathways that govern sleep across phyla have been described. The mammalian brain-type fatty acid binding protein (Fabp7) is expressed in astrocytes, and its mRNA oscillates in tandem with the sleep-wake cycle. However, the role of FABP7 in regulating sleep remains poorly understood. We found that the missense mutation FABP7.T61M is associated with fragmented sleep in humans. This phenotype was recapitulated in mice and fruitflies bearing similar mutations: Fabp7-deficient mice and transgenic flies that express the FABP7.T61M missense mutation in astrocytes also show fragmented sleep. These results provide novel evidence for a distinct molecular pathway linking lipid-signaling cascades within astrocytes in sleep regulation among phylogenetically disparate species.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Proteína de Unión a los Ácidos Grasos 7/biosíntesis , Transducción de Señal/fisiología , Sueño/fisiología , Proteínas Supresoras de Tumor/biosíntesis , Animales , Astrocitos/citología , Relojes Biológicos/fisiología , Encéfalo/citología , Drosophila melanogaster , Proteína de Unión a los Ácidos Grasos 7/genética , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación Missense , Proteínas Supresoras de Tumor/genética
15.
J Neurosci Res ; 95(8): 1548-1564, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27320125

RESUMEN

Disruption of sleep/wake activity in Alzheimer's disease (AD) patients significantly affects their quality of life and that of their caretakers and is a major contributing factor for institutionalization. Levels of amyloid-ß (Aß) have been shown to be regulated by neuronal activity and to correlate with the sleep/wake cycle. Whether consolidated sleep can be disrupted by Aß alone is not well understood. We hypothesize that Aß42 can increase wakefulness and disrupt consolidated sleep. Here we report that flies expressing the human Aß42 transgene in neurons have significantly reduced consolidated sleep compared with control flies. Fatty acid binding proteins (Fabp) are small hydrophobic ligand carriers that have been clinically implicated in AD. Aß42 flies that carry a transgene of either the Drosophila Fabp or the mammalian brain-type Fabp show a significant increase in nighttime sleep and long consolidated sleep bouts, rescuing the Aß42-induced sleep disruption. These studies suggest that alterations in Fabp levels and/or activity may be associated with sleep disturbances in AD. Future work to determine the molecular mechanisms that contribute to Fabp-mediated rescue of Aß42-induced sleep loss will be important for the development of therapeutics in the treatment of AD. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Péptidos beta-Amiloides/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/genética , Trastornos del Sueño-Vigilia/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Drosophila , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Antagonistas de Hormonas/toxicidad , Humanos , Locomoción/efectos de los fármacos , Locomoción/genética , Mifepristona/farmacología , Mifepristona/toxicidad , ARN Mensajero/metabolismo , Sueño/efectos de los fármacos , Sueño/genética , Trastornos del Sueño-Vigilia/inducido químicamente , Trastornos del Sueño-Vigilia/fisiopatología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vigilia/efectos de los fármacos , Vigilia/genética
16.
BMC Genomics ; 17(Suppl 8): 727, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27801296

RESUMEN

BACKGROUND: Why we sleep is still one of the most perplexing mysteries in biology. Strong evidence indicates that sleep is necessary for normal brain function and that sleep need is a tightly regulated process. Surprisingly, molecular mechanisms that determine sleep need are incompletely described. Moreover, very little is known about transcriptional changes that specifically accompany the accumulation and discharge of sleep need. Several studies have characterized differential gene expression changes following sleep deprivation. Much less is known, however, about changes in gene expression during the compensatory response to sleep deprivation (i.e. recovery sleep). RESULTS: In this study we present a comprehensive analysis of the effects of sleep deprivation and subsequent recovery sleep on gene expression in the mouse cortex. We used a non-traditional analytical method for normalization of genome-wide gene expression data, Removal of Unwanted Variation (RUV). RUV improves detection of differential gene expression following sleep deprivation. We also show that RUV normalization is crucial to the discovery of differentially expressed genes associated with recovery sleep. Our analysis indicates that the majority of transcripts upregulated by sleep deprivation require 6 h of recovery sleep to return to baseline levels, while the majority of downregulated transcripts return to baseline levels within 1-3 h. We also find that transcripts that change rapidly during recovery (i.e. within 3 h) do so on average with a time constant that is similar to the time constant for the discharge of sleep need. CONCLUSIONS: We demonstrate that proper data normalization is essential to identify changes in gene expression that are specifically linked to sleep deprivation and recovery sleep. Our results provide the first evidence that recovery sleep is comprised of two waves of transcriptional regulation that occur at different times and affect functionally distinct classes of genes.


Asunto(s)
Corteza Cerebral/fisiología , Regulación de la Expresión Génica , Homeostasis/genética , Sueño/genética , Transcriptoma , Animales , Ritmo Circadiano/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica , Masculino , Ratones , Privación de Sueño/genética
18.
Neurodegener Dis Manag ; 4(5): 351-62, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25405649

RESUMEN

Sleep-wake disturbances are a highly prevalent and often disabling feature of Alzheimer's disease (AD). A cardinal feature of AD includes the formation of amyloid plaques, associated with the extracellular accumulation of the amyloid-ß (Aß) peptide. Evidence from animal and human studies suggests that Aß pathology may disrupt the sleep-wake cycle, in that as Aß accumulates, more sleep-wake fragmentation develops. Furthermore, recent research in animal and human studies suggests that the sleep-wake cycle itself may influence Alzheimer's disease onset and progression. Chronic sleep deprivation increases amyloid plaque deposition, and sleep extension results in fewer plaques in experimental models. In this review geared towards the practicing clinician, we discuss possible mechanisms underlying the reciprocal relationship between the sleep-wake cycle and AD pathology and behavior, and present current approaches to therapy for sleep disorders in AD.


Asunto(s)
Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/fisiopatología , Encéfalo/fisiopatología , Trastornos del Sueño del Ritmo Circadiano/complicaciones , Trastornos del Sueño del Ritmo Circadiano/fisiopatología , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Placa Amiloide , Sueño , Trastornos del Sueño del Ritmo Circadiano/patología
19.
Front Syst Neurosci ; 8: 121, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25018707

RESUMEN

The epilepsies are a heterogeneous group of neurological diseases defined by the occurrence of unprovoked seizures which, in many cases, are correlated with diurnal rhythms. In order to gain insight into the biological mechanisms controlling this phenomenon, we characterized time-of-day effects on electrical seizure threshold in mice. Male C57BL/6J wild-type mice were maintained on a 14/10 h light/dark cycle, from birth until 6 weeks of age for seizure testing. Seizure thresholds were measured using a step-wise paradigm involving a single daily electrical stimulus. Results showed that the current required to elicit both generalized and maximal seizures was significantly higher in mice tested during the dark phase of the diurnal cycle compared to mice tested during the light phase. This rhythm was absent in BMAL1 knockout (KO) mice. BMAL1 KO also exhibited significantly reduced seizure thresholds at all times tested, compared to C57BL/6J mice. Results document a significant influence of time-of-day on electrical seizure threshold in mice and suggest that this effect is under the control of genes that are known to regulate circadian behaviors. Furthermore, low seizure thresholds in BMAL1 KO mice suggest that BMAL1 itself is directly involved in controlling neuronal excitability.

20.
J Sleep Res ; 23(1): 22-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23992533

RESUMEN

Sleep symptoms are associated with weight gain and cardiometabolic disease. The potential role of diet has been largely unexplored. Data from the 2007-2008 National Health and Nutrition Examination Survey (NHANES) were used (n = 4552) to determine which nutrients were associated with sleep symptoms in a nationally representative sample. Survey items assessed difficulty falling asleep, sleep maintenance difficulties, non-restorative sleep and daytime sleepiness. Analyses were adjusted for energy intake, other dietary factors, exercise, body mass index (BMI) and sociodemographics. Population-weighted, logistic regression, with backwards-stepwise selection, examined which nutrients were associated with sleep symptoms. Odds ratios (ORs) reflect the difference in odds of sleep symptoms associated with a doubling in nutrient. Nutrients that were associated independently with difficulty falling asleep included (in order): alpha-carotene (OR = 0.96), selenium (OR = 0.80), dodecanoic acid (OR = 0.91), calcium (OR = 0.83) and hexadecanoic acid (OR = 1.10). Nutrients that were associated independently with sleep maintenance difficulties included: salt (OR = 1.19), butanoic acid (0.81), carbohydrate (OR = 0.71), dodecanoic acid (OR = 0.90), vitamin D (OR = 0.84), lycopene (OR = 0.98), hexanoic acid (OR = 1.25) and moisture (OR = 1.27). Nutrients that were associated independently with non-restorative sleep included butanoic acid (OR = 1.09), calcium (OR = 0.81), vitamin C (OR = 0.92), water (OR = 0.98), moisture (OR = 1.41) and cholesterol (OR = 1.10). Nutrients that were associated independently with sleepiness included: moisture (OR = 1.20), theobromine (OR = 1.04), potassium (OR = 0.70) and water (OR = 0.97). These results suggest novel associations between sleep symptoms and diet/metabolism, potentially explaining associations between sleep and cardiometabolic diseases.


Asunto(s)
Encuestas sobre Dietas , Dieta , Trastornos del Sueño-Vigilia/inducido químicamente , Trastornos del Sueño-Vigilia/fisiopatología , Sueño/efectos de los fármacos , Sueño/fisiología , Adulto , Índice de Masa Corporal , Ácido Butírico/farmacología , Calcio/farmacología , Carotenoides/efectos adversos , Carotenoides/farmacología , Colesterol/efectos adversos , Carbohidratos de la Dieta/farmacología , Ejercicio Físico , Femenino , Humanos , Ácidos Láuricos/farmacología , Licopeno , Masculino , Persona de Mediana Edad , Encuestas Nutricionales , Oportunidad Relativa , Ácido Palmítico/farmacología , Selenio/farmacología , Trastornos del Sueño-Vigilia/metabolismo , Cloruro de Sodio Dietético/farmacología , Vitamina D/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...