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










Base de datos
Intervalo de año de publicación
1.
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.

2.
Adv Biol (Weinh) ; 6(11): e2101203, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35822937

RESUMEN

The bi-directional relationship between sleep and stress has been actively researched as sleep disturbances and stress have become increasingly common in society. Interestingly, the brain and underlying neural circuits important for sleep regulation may respond uniquely to stress that leads to post-traumatic stress disorder (PTSD) and stress that does not. In stress that does not lead to PTSD, the hypothalamic-pituitary-adrenal axis (HPA) pathway is activated normally that results in sympathetic nervous system activation that allows the brain and body to return to baseline functioning. However, exposure to stress that leads to PTSD, causes enhanced negative feedback of this same pathway and results in long-term physiological and psychological changes. In this review, how stress regulates glucocorticoid signaling pathways in brain glial cells called astrocytes, and then mediates stress-induced insomnia are examined. Astrocytes are critical sleep regulatory cells and their connections to sleep and stress due to disturbed glucocorticoid signaling provide a novel mechanism to explain how stress leads to insomnia. This review will examine the interactions of stress neurobiology, astrocytes, sleep, and glucocorticoid signaling pathways and will examine the how stress that leads to PTSD and stress that does not impacts sleep-regulatory processes.


Asunto(s)
Trastornos del Inicio y del Mantenimiento del Sueño , Trastornos por Estrés Postraumático , Humanos , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Trastornos del Inicio y del Mantenimiento del Sueño/metabolismo , Glucocorticoides/metabolismo , Trastornos por Estrés Postraumático/metabolismo
3.
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.

4.
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
5.
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.

6.
Physiol Rep ; 9(4): e14716, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33619911

RESUMEN

Physical exercise and fitness may serve as resilience factors to stress exposure. However, the extreme range in human exercise performance suggests that genetic variation for exercise capacity could be a confounding feature to understanding the connection between exercise and stress exposure. To test this idea, we use laboratory rat models selectively bred for a low and high gain in aerobic running capacity in response to training to examine whether an inherent capacity to respond to physical exercise reflects how stress changes neurobiological functioning and regulates fear-associated memory processing. Utilization of this contrasting rat model system of low and high responders has the potential to guide the interpretation of the reported association with exercise involvement and the reduction of stress-induced anxiety disorders. Our data show that aerobic fitness may be linked to the ability to regulate fear-associated memories. We also show that acquired exercise capacity may play a key role in regulating responses to an acute stressor. Exercise sensitivity plays a significant role in the activation of the plasticity-associated molecule extracellular signal-regulated kinase, changes in stress hormone activity, and anatomical modifications to the noradrenergic locus coeruleus. These data identify a unique operational mechanism that may serve as translational targets for lessening symptoms of stress and anxiety.


Asunto(s)
Ansiedad/psicología , Conducta Animal , Miedo , Recuerdo Mental , Condicionamiento Físico Animal , Esfuerzo Físico , Estrés Psicológico/psicología , Adaptación Fisiológica , Hormona Adrenocorticotrópica/sangre , Animales , Ansiedad/metabolismo , Ansiedad/fisiopatología , Ansiedad/prevención & control , Encéfalo/metabolismo , Encéfalo/fisiopatología , Corticosterona/sangre , Activación Enzimática , Extinción Psicológica , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Masculino , Melatonina/sangre , Plasticidad Neuronal , Fosforilación , Ratas Endogámicas , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Estrés Psicológico/prevención & control
7.
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
8.
Sci Rep ; 10(1): 18025, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33093538

RESUMEN

Sleep disturbances are commonly found in trauma-exposed populations. Additionally, trauma exposure results in fear-associated memory impairments. Given the interactions of sleep with learning and memory, we hypothesized that increasing sleep duration following trauma exposure would restore overall function and improve trauma-induced fear-associated memory dysfunction. Here, we utilized single prolonged stress, a validated rodent model of post-traumatic stress disorder, in combination with optogenetic activation of hypothalamic melanin-concentrating hormone containing cells to increase sleep duration. The goal of this work was to ascertain if post-trauma sleep increases are sufficient to improve fear-associated memory function. In our laboratory, optogenetic stimulation after trauma exposure was sufficient to increase REM sleep duration during both the Light and Dark Phase, whereas NREM sleep duration was only increased during the Dark Phase of the circadian day. Interestingly though, animals that received optogenetic stimulation showed significantly improved fear-associated memory processing compared to non-stimulated controls. These results suggest that sleep therapeutics immediately following trauma exposure may be beneficial and that post-trauma sleep needs to be further examined in the context of the development of post-traumatic stress disorder.


Asunto(s)
Miedo , Hormonas Hipotalámicas/administración & dosificación , Melaninas/administración & dosificación , Trastornos de la Memoria/prevención & control , Optogenética , Hormonas Hipofisarias/administración & dosificación , Trastornos del Sueño-Vigilia/prevención & control , Sueño REM , Trastornos por Estrés Postraumático/complicaciones , Animales , Masculino , Consolidación de la Memoria , Trastornos de la Memoria/etiología , Ratas , Ratas Long-Evans , Trastornos del Sueño-Vigilia/etiología
9.
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.

10.
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
11.
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
12.
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
13.
Exp Brain Res ; 233(8): 2335-46, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26019008

RESUMEN

Sleep abnormalities, such as insomnia, nightmares, hyper-arousal, and difficulty initiating or maintaining sleep, are diagnostic criteria of posttraumatic stress disorder (PTSD). The vivid dream state, rapid eye movement (REM) sleep, has been implicated in processing emotional memories. We have hypothesized that REM sleep is maladaptive in those suffering from PTSD. However, the precise neurobiological mechanisms regulating sleep disturbances following trauma exposure are poorly understood. Using single prolonged stress (SPS), a well-validated rodent model of PTSD, we measured sleep alterations in response to stressor exposure and over a subsequent 7-day isolation period during which the PTSD-like phenotype develops. SPS resulted in acute increases in REM sleep and transition to REM sleep, and decreased waking in addition to alterations in sleep architecture. The severity of the PTSD-like phenotype was later assessed by measuring freezing levels on a fear-associated memory test. Interestingly, the change in REM sleep following SPS was significantly correlated with freezing behavior during extinction recall assessed more than a week later. Reductions in theta (4-10 Hz) and sigma (10-15 Hz) band power during transition to REM sleep also correlated with impaired fear-associated memory processing. These data reveal that changes in REM sleep, transition to REM sleep, waking, and theta and sigma power may serve as sleep biomarkers to identify individuals with increased susceptibility to PTSD following trauma exposure.


Asunto(s)
Ondas Encefálicas/fisiología , Miedo/fisiología , Trastornos de la Memoria/fisiopatología , Trastornos del Sueño-Vigilia/fisiopatología , Sueño REM/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Animales , Conducta Animal , Biomarcadores , Modelos Animales de Enfermedad , Extinción Psicológica , Masculino , Trastornos de la Memoria/etiología , Recuerdo Mental , Ratas , Ratas Long-Evans , Trastornos del Sueño-Vigilia/etiología , Trastornos por Estrés Postraumático/complicaciones
14.
J Neurosci Methods ; 251: 83-91, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26014662

RESUMEN

BACKGROUND: Sleep deprivation via gentle handling is time-consuming and personnel-intensive. NEW METHOD: We present here an automated sleep deprivation system via air puffs. Implanted EMG and EEG electrodes were used to assess sleep/waking states in six male Sprague-Dawley rats. Blood samples were collected from an implanted intravenous catheter every 4h during the 12-h light cycle on baseline, 8h of sleep deprivation via air puffs, and 8h of sleep deprivation by gentle handling days. RESULTS: The automated system was capable of scoring sleep and waking states as accurately as our offline version (∼90% for sleep) and with sufficient speed to trigger a feedback response within an acceptable amount of time (1.76s). Manual state scoring confirmed normal sleep on the baseline day and sleep deprivation on the two manipulation days (68% decrease in non-REM, 63% decrease in REM, and 74% increase in waking). No significant differences in levels of ACTH and corticosterone (stress hormones indicative of HPA axis activity) were found at any time point between baseline sleep and sleep deprivation via air puffs. COMPARISON WITH EXISTING METHOD: There were no significant differences in ACTH or corticosterone concentrations between sleep deprivation by air puffs and gentle handling over the 8-h period. CONCLUSIONS: Our system accurately detects sleep and delivers air puffs to acutely deprive rats of sleep with sufficient temporal resolution during the critical 4-5h post learning sleep-dependent memory consolidation period. The system is stress-free and a viable alternative to existing sleep deprivation techniques.


Asunto(s)
Automatización/métodos , Ritmo Circadiano/fisiología , Manejo Psicológico , Privación de Sueño/etiología , Hormona Adrenocorticotrópica/sangre , Movimientos del Aire , Algoritmos , Animales , Automatización/instrumentación , Corticosterona/sangre , Electroencefalografía , Electromiografía/métodos , Masculino , Sistemas en Línea , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Privación de Sueño/sangre , Estrés Psicológico/sangre , Estrés Psicológico/fisiopatología , Factores de Tiempo , Vigilia
15.
Exp Brain Res ; 232(5): 1575-84, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24623353

RESUMEN

Post-traumatic stress disorder (PTSD) is characterized by intrusive memories of a traumatic event, avoidance behavior related to cues of the trauma, emotional numbing, and hyper-arousal. Sleep abnormalities and nightmares are core symptoms of this disorder. In this review, we propose a model which implicates abnormal activity in the locus coeruleus (LC), an important modifier of sleep-wake regulation, as the source of sleep abnormalities and memory abnormalities seen in PTSD. Abnormal LC activity may be playing a key role in symptom formation in PTSD via sleep dysregulation and suppression of hippocampal bidirectional plasticity.


Asunto(s)
Trastornos del Sueño-Vigilia/etiología , Trastornos por Estrés Postraumático/complicaciones , Trastornos por Estrés Postraumático/etiología , Heridas y Lesiones/complicaciones , Animales , Modelos Animales de Enfermedad , Humanos , Locus Coeruleus/fisiopatología , Roedores , Trastornos por Estrés Postraumático/patología
16.
PLoS One ; 8(11): e81554, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24244744

RESUMEN

Given the relationship between sleep and plasticity, we examined the role of Extracellular signal-regulated kinase (ERK) in regulating baseline sleep, and modulating the response to waking experience. Both sleep deprivation and social enrichment increase ERK phosphorylation in wild-type flies. The effects of both sleep deprivation and social enrichment on structural plasticity in the LNvs can be recapitulated by expressing an active version of ERK (UAS-ERK(SEM)) pan-neuronally in the adult fly using GeneSwitch (Gsw) Gsw-elav-GAL4. Conversely, disrupting ERK reduces sleep and prevents both the behavioral and structural plasticity normally induced by social enrichment. Finally, using transgenic flies carrying a cAMP response Element (CRE)-luciferase reporter we show that activating ERK enhances CRE-Luc activity while disrupting ERK reduces it. These data suggest that ERK phosphorylation is an important mediator in transducing waking experience into sleep.


Asunto(s)
Drosophila/enzimología , Drosophila/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Animales , Animales Modificados Genéticamente , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Fosforilación , Sueño/fisiología
17.
J Neurosci ; 32(4): 1383-94, 2012 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-22279223

RESUMEN

The astrocyte brain fatty acid binding protein (Fabp7) has previously been shown to have a coordinated diurnal regulation of mRNA and protein throughout mouse brain, and an age-dependent decline in protein expression within synaptoneurosomal fractions. Mechanisms that control time-of-day changes in expression and trafficking Fabp7 to the perisynaptic process are not known. In this study, we confirmed an enrichment of Fabp7 mRNA and protein in the astrocytic perisynaptic compartment, and observed a diurnal change in the intracellular distribution of Fabp7 mRNA in molecular layers of hippocampus. Northern blotting revealed a coordinated time-of-day-dependent oscillation for the Fabp7 mRNA poly(A) tail throughout murine brain. Cytoplasmic polyadenylation element-binding protein 1 (CPEB1) regulates subcellular trafficking and translation of synaptic plasticity-related mRNAs. Here we show that Fabp7 mRNA coimmunoprecipitated with CPEB1 from primary mouse astrocyte extracts, and its 3'UTR contains phylogenetically conserved cytoplasmic polyadenylation elements (CPEs) capable of regulating translation of reporter mRNAs during Xenopus oocyte maturation. Given that Fabp7 expression is confined to astrocytes and neural progenitors in adult mouse brain, the synchronized cycling pattern of Fabp7 mRNA is a novel discovery among known CPE-regulated transcripts. These results implicate circadian, sleep, and/or metabolic control of CPEB-mediated subcellular trafficking and localized translation of Fabp7 mRNA in the tripartite synapse of mammalian brain.


Asunto(s)
Astrocitos/metabolismo , Ritmo Circadiano/fisiología , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero/metabolismo , Sinapsis/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Proteína de Unión a los Ácidos Grasos 7 , Femenino , Hipocampo/metabolismo , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Poliadenilación/fisiología , Transporte de Proteínas/fisiología , Fracciones Subcelulares/metabolismo , Fracciones Subcelulares/fisiología , Sinapsis/fisiología , Xenopus
18.
Commun Integr Biol ; 4(5): 623-6, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22046481

RESUMEN

We recently reported evidence implicating fatty-acid binding protein (Fabp) in the control of sleep and memory formation. We used Drosophila melanogaster to examine the relationship between sleep and memory through transgenic overexpression of mouse brain-Fabp, Fabp7, or the Drosophila Fabp homolog, (dFabp). The key findings are that 1) a genetically induced increase in daytime consolidated sleep (naps) correlates with an increase in cognitive performance, and 2) a late "window" of memory consolidation occurs days after the traditionally understood "synaptic" consolidation. Exactly how Fabp-signaling may be involved in converting normal to enhanced long-term memory (LTM) is not known. Here we describe additional data which support relative subcellular compartmental localization of Fabp in regulating stage associations of different forms of memory in Drosophila. Anesthesia resistant memory (ARM) is a longer lasting memory that is produced by massed training, but unlike LTM produced by spaced training, it is insensitive to protein synthesis inhibitors and does not persist as long. We observed that the ratio of ARM to LTM performance index of Fabp7-transgenic flies is proportional to the relative cytoplasmic to nuclear Fabp7 expression level. These data suggest a common lipid-signaling cascade exists between phases of memory formation previously thought to be molecularly distinct.

19.
PLoS One ; 6(1): e15890, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21298037

RESUMEN

Sleep is thought to be important for memory consolidation, since sleep deprivation has been shown to interfere with memory processing. However, the effects of augmenting sleep on memory formation are not well known, and testing the role of sleep in memory enhancement has been limited to pharmacological and behavioral approaches. Here we test the effect of overexpressing the brain-type fatty acid binding protein (Fabp7) on sleep and long-term memory (LTM) formation in Drosophila melanogaster. Transgenic flies carrying the murine Fabp7 or the Drosophila homologue dFabp had reduced baseline sleep but normal LTM, while Fabp induction produced increases in both net sleep and LTM. We also define a post-training consolidation "window" that is sufficient for the observed Fabp-mediated memory enhancement. Since Fabp overexpression increases consolidated daytime sleep bouts, these data support a role for longer naps in improving memory and provide a novel role for lipid-binding proteins in regulating memory consolidation concurrently with changes in behavioral state.


Asunto(s)
Drosophila melanogaster/fisiología , Proteínas de Unión a Ácidos Grasos/fisiología , Memoria a Largo Plazo , Proteínas del Tejido Nervioso/fisiología , Sueño , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/fisiología , Proteína de Unión a los Ácidos Grasos 7 , Proteínas de Unión a Ácidos Grasos/química , Ratones , Proteínas del Tejido Nervioso/química
20.
Am J Physiol Regul Integr Comp Physiol ; 292(1): R217-26, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16931647

RESUMEN

It has long been known that central opioid systems play an important role in certain aspects of appetite and food intake, particularly with regard to the hedonic or rewarding impact of calorically dense food, such as fat and sugar. Ventral striatal enkephalin may be a key component of this system, as infusions of mu-opiate agonists into this region strongly increase feeding, whereas infusions of opiate antagonists decrease food intake. While pharmacological analysis has consistently supported such a role, direct measurement of enkephalin gene expression in relation to differing food motivational conditions has not been examined. In this study, the effects of a restricted laboratory chow diet (resulting in negative energy balance) as well has recent consumption of chow (short-term satiety) on striatal preproenkephalin (PPE) and prodynorphin (PD) mRNA expression were measured in rats, using both Northern blot analysis and in situ hybridization methods. As a comparison, hypothalamic (arcuate nucleus) neuropeptide Y (NPY) was also measured in these conditions. PPE expression was broadly downregulated throughout the striatum in animals that had recently consumed a meal, whereas it was unaffected by negative energy balance. Expression of an additional striatal peptide gene, PD, did not follow this pattern, although diet restriction caused a decrease in accumbens core dynorphin mRNA. Conversely, as expected, arcuate nucleus NPY mRNA expression was markedly upregulated by negative energy balance, but was unchanged by recent food consumption. This double dissociation between striatal and hypothalamic peptide systems suggests a specific role for striatal PPE in relatively short-term food motivational states, but not in long-term metabolic responses to diet restriction.


Asunto(s)
Metabolismo Energético/fisiología , Expresión Génica/fisiología , Hipotálamo/fisiología , Neostriado/metabolismo , Neuropéptido Y/fisiología , Péptidos Opioides/biosíntesis , Péptidos Opioides/genética , Respuesta de Saciedad/fisiología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Northern Blotting , Encefalinas/biosíntesis , Encefalinas/genética , Privación de Alimentos/fisiología , Hambre/fisiología , Hibridación in Situ , Masculino , Motivación , Precursores de Proteínas/biosíntesis , Precursores de Proteínas/genética , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...