Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Neurosci ; 44(28)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38830763

RESUMEN

Chronic sleep disruption (CSD), from insufficient or fragmented sleep and is an important risk factor for Alzheimer's disease (AD). Underlying mechanisms are not understood. CSD in mice results in degeneration of locus ceruleus neurons (LCn) and CA1 hippocampal neurons and increases hippocampal amyloid-ß42 (Aß42), entorhinal cortex (EC) tau phosphorylation (p-tau), and glial reactivity. LCn injury is increasingly implicated in AD pathogenesis. CSD increases NE turnover in LCn, and LCn norepinephrine (NE) metabolism activates asparagine endopeptidase (AEP), an enzyme known to cleave amyloid precursor protein (APP) and tau into neurotoxic fragments. We hypothesized that CSD would activate LCn AEP in an NE-dependent manner to induce LCn and hippocampal injury. Here, we studied LCn, hippocampal, and EC responses to CSD in mice deficient in NE [dopamine ß-hydroxylase (Dbh)-/-] and control male and female mice, using a model of chronic fragmentation of sleep (CFS). Sleep was equally fragmented in Dbh -/- and control male and female mice, yet only Dbh -/- mice conferred resistance to CFS loss of LCn, LCn p-tau, and LCn AEP upregulation and activation as evidenced by an increase in AEP-cleaved APP and tau fragments. Absence of NE also prevented a CFS increase in hippocampal AEP-APP and Aß42 but did not prevent CFS-increased AEP-tau and p-tau in the EC. Collectively, this work demonstrates AEP activation by CFS, establishes key roles for NE in both CFS degeneration of LCn neurons and CFS promotion of forebrain Aß accumulation, and, thereby, identifies a key molecular link between CSD and specific AD neural injuries.


Asunto(s)
Péptidos beta-Amiloides , Cisteína Endopeptidasas , Hipocampo , Locus Coeruleus , Norepinefrina , Privación de Sueño , Animales , Péptidos beta-Amiloides/metabolismo , Norepinefrina/metabolismo , Ratones , Hipocampo/metabolismo , Hipocampo/patología , Privación de Sueño/metabolismo , Privación de Sueño/patología , Masculino , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Fragmentos de Péptidos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Dopamina beta-Hidroxilasa/metabolismo , Dopamina beta-Hidroxilasa/genética , Proteínas tau/metabolismo , Femenino , Degeneración Nerviosa/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/genética
2.
J Neurosci ; 38(48): 10255-10270, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30322903

RESUMEN

Brainstem locus ceruleus neurons (LCn) are among the first neurons across the lifespan to evidence tau pathology, and LCn are implicated in tau propagation throughout the cortices. Yet, events influencing LCn tau are poorly understood. Activated persistently across wakefulness, LCn experience significant metabolic stress in response to chronic short sleep (CSS). Here we explored whether CSS influences LCn tau and the biochemical, neuroanatomical, and/or behavioral progression of tauopathy in male and female P301S mice. CSS in early adult life advanced the temporal progression of neurobehavioral impairments and resulted in a lasting increase in soluble tau oligomers. Intriguingly, CSS resulted in an early increase in AT8 and MC1 tau pathology in the LC. Over time tau pathology, including tangles, was evident in forebrain tau-vulnerable regions. Sustained microglial and astrocytic activation was observed as well. Remarkably, CSS resulted in significant loss of neurons in the two regions examined: the basolateral amygdala and LC. A second, distinct form of chronic sleep disruption, fragmentation of sleep, during early adult life also increased tau deposition and imparted early neurobehavioral impairment. Collectively, the findings demonstrate that early life sleep disruption has important lasting effects on the temporal progression in P301S mice, influencing tau pathology and hastening neurodegeneration, neuroinflammation, and neurobehavioral impairments.SIGNIFICANCE STATEMENT Chronic short sleep (CSS) is pervasive in modern society. Here, we found that early life CSS influences behavioral, biochemical, and neuroanatomic aspects of the temporal progression of tauopathy in a mouse model of the P301S tau mutation. Specifically, CSS hastened the onset of motor impairment and resulted in a greater loss of neurons in both the locus ceruleus and basolateral/lateral amygdala. Importantly, despite a protracted recovery opportunity after CSS, mice evidenced a sustained increase in pathogenic tau oligomers, and increased pathogenic tau in the locus ceruleus and limbic system nuclei. These findings unveil early life sleep habits as an important determinant in the progression of tauopathy.


Asunto(s)
Progresión de la Enfermedad , Mutación/fisiología , Privación de Sueño/metabolismo , Tauopatías/metabolismo , Proteínas tau/metabolismo , Amígdala del Cerebelo/metabolismo , Amígdala del Cerebelo/patología , Animales , Femenino , Humanos , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Privación de Sueño/genética , Privación de Sueño/patología , Tauopatías/genética , Tauopatías/patología , Proteínas tau/genética
3.
J Neurosci ; 34(12): 4418-31, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24647961

RESUMEN

Modern society enables a shortening of sleep times, yet long-term consequences of extended wakefulness on the brain are largely unknown. Essential for optimal alertness, locus ceruleus neurons (LCns) are metabolically active neurons that fire at increased rates across sustained wakefulness. We hypothesized that wakefulness is a metabolic stressor to LCns and that, with extended wakefulness, adaptive mitochondrial metabolic responses fail and injury ensues. The nicotinamide adenine dinucleotide-dependent deacetylase sirtuin type 3 (SirT3) coordinates mitochondrial energy production and redox homeostasis. We find that brief wakefulness upregulates SirT3 and antioxidants in LCns, protecting metabolic homeostasis. Strikingly, mice lacking SirT3 lose the adaptive antioxidant response and incur oxidative injury in LCns across brief wakefulness. When wakefulness is extended for longer durations in wild-type mice, SirT3 protein declines in LCns, while oxidative stress and acetylation of mitochondrial proteins, including electron transport chain complex I proteins, increase. In parallel with metabolic dyshomeostasis, apoptosis is activated and LCns are lost. This work identifies mitochondrial stress in LCns upon wakefulness, highlights an essential role for SirT3 activation in maintaining metabolic homeostasis in LCns across wakefulness, and demonstrates that extended wakefulness results in reduced SirT3 activity and, ultimately, degeneration of LCns.


Asunto(s)
Locus Coeruleus/metabolismo , Degeneración Nerviosa/metabolismo , Neuronas/metabolismo , Privación de Sueño/metabolismo , Sueño/fisiología , Vigilia/fisiología , Animales , Corticosterona/sangre , Locus Coeruleus/patología , Masculino , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Degeneración Nerviosa/patología , Neuronas/patología , Estrés Oxidativo/fisiología , Sirtuina 3/genética , Sirtuina 3/metabolismo , Privación de Sueño/patología , Regulación hacia Arriba
4.
Transl Psychiatry ; 13(1): 78, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36869037

RESUMEN

Disrupted sleep is a symptom of many psychiatric disorders, including substance use disorders. Most drugs of abuse, including opioids, disrupt sleep. However, the extent and consequence of opioid-induced sleep disturbance, especially during chronic drug exposure, is understudied. We have previously shown that sleep disturbance alters voluntary morphine intake. Here, we examine the effects of acute and chronic morphine exposure on sleep. Using an oral self-administration paradigm, we show that morphine disrupts sleep, most significantly during the dark cycle in chronic morphine, with a concomitant sustained increase in neural activity in the Paraventricular Nucleus of the Thalamus (PVT). Morphine binds primarily to Mu Opioid Receptors (MORs), which are highly expressed in the PVT. Translating Ribosome Affinity Purification (TRAP)-Sequencing of PVT neurons that express MORs showed significant enrichment of the circadian entrainment pathway. To determine whether MOR + cells in the PVT mediate morphine-induced sleep/wake properties, we inhibited these neurons during the dark cycle while mice were self-administering morphine. This inhibition decreased morphine-induced wakefulness but not general wakefulness, indicating that MORs in the PVT contribute to opioid-specific wake alterations. Overall, our results suggest an important role for PVT neurons that express MORs in mediating morphine-induced sleep disturbance.


Asunto(s)
Morfina , Trastornos del Sueño-Vigilia , Animales , Ratones , Analgésicos Opioides , Receptores Opioides mu , Neuronas , Tálamo
5.
J Neurosci ; 31(11): 4025-36, 2011 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-21411645

RESUMEN

Wake neurons in the basal forebrain and brainstem provide critical inputs to optimize alertness and attention. These neurons, however, evidence heightened vulnerability to a diverse array of metabolic challenges, including aging. SIRT1 is an nicotinamide adenine dinucleotide responsive deacetylase serving diverse adaptive responses to metabolic challenges, yet this metabolic rheostat may be downregulated under conditions of significant oxidative stress. We hypothesized that SIRT1 might serve as a critical neuroprotectant for wake neurons in young animals but that this protectant would be lost upon aging, rendering the neurons more vulnerable to metabolic insults. In this collection of studies, we first established the presence of nuclear SIRT1 in wake neurons throughout the forebrain and brainstem. Supporting functional and behavioral roles for SIRT1 in wake-active neurons, transgenic whole animal, and conditional loss of brain SIRT1 in the adult mouse impart selective impairments in wakefulness, without disrupting non-rapid eye movement or rapid eye movement sleep. Populations of wake neurons, including the orexinergic, locus ceruleus, mesopontine cholinergic, and dopaminergic wake neurons, evidence loss of dendrites and neurotransmitter synthesis enzymes and develop accelerated accumulation of lipofuscin, consistent with a senescence-like phenotype in wake neurons. Normal aging results in a progressive loss of SIRT1 in wake-active neurons, temporally coinciding with lipofuscin accumulation. SIRT1 is a critical age-sensitive neuroprotectant for wake neurons, and its deficiency results in impaired wakefulness.


Asunto(s)
Envejecimiento/fisiología , Neuronas/metabolismo , Sirtuina 1/metabolismo , Vigilia/fisiología , Animales , Western Blotting , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Transgénicos , Prosencéfalo/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirtuina 1/genética , Sueño/fisiología
6.
Front Aging Neurosci ; 14: 1025402, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275002

RESUMEN

Chronic sleep disruption is a risk factor for Alzheimer's disease (AD), yet mechanisms by which sleep disturbances might promote or exacerbate AD are not understood. Short-term sleep loss acutely increases hippocampal amyloid ß (Aß) in wild type (WT) mice and long-term sleep loss increases amyloid plaque in AD transgenic mouse models. Both effects can be influenced by the wake-promoting neuropeptide, hypocretin (HCRT), but whether HCRT influences amyloid accumulation independent of sleep and wake timing modulation remains unclear. Here, we induced chronic fragmentation of sleep (CFS) in WT and HCRT-deficient mice to elicit similar arousal indices, sleep bout lengths and sleep bout numbers in both genotypes. We then examined the roles of HCRT in CFS-induced hippocampal Aß accumulation and injury. CFS in WT mice resulted in increased Aß42 in the hippocampus along with loss of cholinergic projections and loss of locus coeruleus neurons. Mice with HCRT deficiency conferred resistance to CFS Aß42 accumulation and loss of cholinergic projections in the hippocampus yet evidenced similar CFS-induced loss of locus coeruleus neurons. Collectively, the findings demonstrate specific roles for orexin in sleep disruption hippocampal injury. Significance statement: Chronic fragmentation of sleep (CFS) occurs in common conditions, including sleep apnea syndromes and chronic pain disorders, yet CFS can induce neural injury. Our results demonstrate that under conditions of sleep fragmentation, hypocretin/orexin is essential for the accumulation of amyloid-ß and loss of cholinergic projections in the hippocampus observed in response to CFS yet does not influence locus coeruleus neuron response to CFS.

7.
Sleep ; 44(8)2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-33768250

RESUMEN

Chronic short sleep (CSS) is prevalent in modern societies and has been proposed as a risk factor for Alzheimer's disease (AD). In support, short-term sleep loss acutely increases levels of amyloid ß (Aß) and tau in wild type (WT) mice and humans, and sleep disturbances predict cognitive decline in older adults. We have shown that CSS induces injury to and loss of locus coeruleus neurons (LCn), neurons with heightened susceptibility in AD. Yet whether CSS during young adulthood drives lasting Aß and/or tau changes and/or neural injury later in life in the absence of genetic risk for AD has not been established. Here, we examined the impact of CSS exposure in young adult WT mice on late-in-life Aß and tau changes and neural responses in two AD-vulnerable neuronal groups, LCn and hippocampal CA1 neurons. Twelve months following CSS exposure, CSS-exposed mice evidenced reductions in CA1 neuron counts and volume, spatial memory deficits, CA1 glial activation, and loss of LCn. Aß 42 and hyperphosphorylated tau were increased in the CA1; however, amyloid plaques and tau tangles were not observed. Collectively the findings demonstrate that CSS exposure in the young adult mouse imparts late-in-life neurodegeneration and persistent derangements in amyloid and tau homeostasis. These findings occur in the absence of a genetic predisposition to neurodegeneration and demonstrate for the first time that CSS can induce lasting, significant neural injury consistent with some, but not all, features of late-onset AD.


Asunto(s)
Enfermedad de Alzheimer , Proteínas tau , Enfermedad de Alzheimer/etiología , Péptidos beta-Amiloides , Animales , Ratones , Placa Amiloide , Sueño
8.
J Neurosci ; 28(9): 2168-78, 2008 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-18305250

RESUMEN

Obstructive sleep apnea is associated with neural injury and dysfunction. Hypoxia/reoxygenation exposures, modeling sleep apnea, injure select populations of neurons, including hypoglossal motoneurons. The mechanisms underlying this motoneuron injury are not understood. We hypothesize that endoplasmic reticulum injury contributes to motoneuron demise. Hypoxia/reoxygenation exposures across 8 weeks in adult mice upregulated the unfolded protein response as evidenced by increased phosphorylation of PERK [PKR-like endoplasmic reticulum (ER) kinase] in facial and hypoglossal motoneurons and persistent upregulation of CCAAT/enhancer-binding protein-homologous protein (CHOP)/growth arrest and DNA damage-inducible protein (GADD153) with nuclear translocation. Long-term hypoxia/reoxygenation also resulted in cleavage and nuclear translocation of caspase-7 and caspase-3 in hypoglossal and facial motoneurons. In contrast, occulomotor and trigeminal motoneurons showed persistent phosphorylation of eIF-2a across hypoxia/reoxygenation, without activations of CHOP/GADD153 or either caspase. Ultrastructural analysis of rough ER in hypoglossal motoneurons revealed hypoxia/reoxygenation-induced luminal swelling and ribosomal detachment. Protection of eIF-2alpha phosphorylation with systemically administered salubrinal throughout hypoxia/reoxygenation exposure prevented CHOP/GADD153 activation in susceptible motoneurons. Collectively, this work provides evidence that long-term exposure to hypoxia/reoxygenation events, modeling sleep apnea, results in significant endoplasmic reticulum injury in select upper airway motoneurons. Augmentation of eIF-2a phosphorylation minimizes motoneuronal injury in this model. It is anticipated that obstructive sleep apnea results in endoplasmic reticulum injury involving motoneurons, whereas a critical balance of phosphorylated eIF-2a should minimize motoneuronal injury in obstructive sleep apnea.


Asunto(s)
Tronco Encefálico/patología , Factor 2 Eucariótico de Iniciación/metabolismo , Neuronas Motoras/metabolismo , Síndromes de la Apnea del Sueño/patología , Animales , Caspasas/metabolismo , Colina O-Acetiltransferasa/metabolismo , Cinamatos/farmacología , Modelos Animales de Enfermedad , Retículo Endoplásmico , Regulación de la Expresión Génica , Hipoxia/complicaciones , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión/métodos , Neuronas Motoras/ultraestructura , Estrés Oxidativo , Fosforilación/efectos de los fármacos , Síndromes de la Apnea del Sueño/etiología , Tiourea/análogos & derivados , Tiourea/farmacología , Factor de Transcripción CHOP/metabolismo , eIF-2 Quinasa/metabolismo
9.
J Neurosci ; 27(37): 10060-71, 2007 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-17855620

RESUMEN

The presence of refractory wake impairments in many individuals with severe sleep apnea led us to hypothesize that the hypoxia/reoxygenation events in sleep apnea permanently damage wake-active neurons. We now confirm that long-term exposure to hypoxia/reoxygenation in adult mice results in irreversible wake impairments. Functionality and injury were next assessed in major wake-active neural groups. Hypoxia/reoxygenation exposure for 8 weeks resulted in vacuolization in the perikarya and dendrites and markedly impaired c-fos activation response to enforced wakefulness in both noradrenergic locus ceruleus and dopaminergic ventral periaqueductal gray wake neurons. In contrast, cholinergic, histaminergic, orexinergic, and serotonergic wake neurons appeared unperturbed. Six month exposure to hypoxia/reoxygenation resulted in a 40% loss of catecholaminergic wake neurons. Having previously identified NADPH oxidase as a major contributor to wake impairments in hypoxia/reoxygenation, the role of NADPH oxidase in catecholaminergic vulnerability was next addressed. NADPH oxidase catalytic and cytosolic subunits were evident in catecholaminergic wake neurons, where hypoxia/reoxygenation resulted in translocation of p67(phox) to mitochondria, endoplasmic reticulum, and membranes. Treatment with a NADPH oxidase inhibitor, apocynin, throughout hypoxia/reoxygenation exposures conferred protection of catecholaminergic neurons. Collectively, these data show that select wake neurons, specifically the two catecholaminergic groups, can be rendered persistently impaired after long-term exposure to hypoxia/reoxygenation, modeling sleep apnea; wake impairments are irreversible; catecholaminergic neurons are lost; and neuronal NADPH oxidase contributes to this injury. It is anticipated that severe obstructive sleep apnea in humans destroys catecholaminergic wake neurons.


Asunto(s)
Catecolaminas/biosíntesis , Modelos Animales de Enfermedad , Neuronas/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Vigilia/fisiología , Animales , Catecolaminas/deficiencia , Locus Coeruleus/citología , Locus Coeruleus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/biosíntesis , NADPH Oxidasas/deficiencia , Neuronas/patología , Síndromes de la Apnea del Sueño/patología
10.
J Clin Invest ; 127(5): 1889-1904, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28394263

RESUMEN

Mutations in the X-linked gene encoding methyl-CpG-binding protein 2 (MeCP2) cause Rett syndrome (RTT), a neurological disorder affecting cognitive development, respiration, and motor function. Genetic restoration of MeCP2 expression reverses RTT-like phenotypes in mice, highlighting the need to search for therapeutic approaches. Here, we have developed knockin mice recapitulating the most common RTT-associated missense mutation, MeCP2 T158M. We found that the T158M mutation impaired MECP2 binding to methylated DNA and destabilized MeCP2 protein in an age-dependent manner, leading to the development of RTT-like phenotypes in these mice. Genetic elevation of MeCP2 T158M expression ameliorated multiple RTT-like features, including motor dysfunction and breathing irregularities, in both male and female mice. These improvements were accompanied by increased binding of MeCP2 T158M to DNA. Further, we found that the ubiquitin/proteasome pathway was responsible for MeCP2 T158M degradation and that proteasome inhibition increased MeCP2 T158M levels. Together, these findings demonstrate that increasing MeCP2 T158M protein expression is sufficient to mitigate RTT-like phenotypes and support the targeting of MeCP2 T158M expression or stability as an alternative therapeutic approach.


Asunto(s)
Regulación de la Expresión Génica , Proteína 2 de Unión a Metil-CpG , Mutación Missense , Proteolisis , Síndrome de Rett , Sustitución de Aminoácidos , Animales , Humanos , Proteína 2 de Unión a Metil-CpG/biosíntesis , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología , Ubiquitina/genética , Ubiquitina/metabolismo
11.
Sleep ; 29(2): 152-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16494082

RESUMEN

STUDY OBJECTIVES: Adult male mice exposed to long-term intermittent hypoxia (LTIH), modeling sleep apnea oxygenation patterns, develop nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-dependent residual hypersomnolence and oxidative injury in select brain regions, including wake-active regions. Premenopausal females are less susceptible to selective oxidative brain injuries. We sought to determine whether female mice exposed to LTIH would confer resistance to LTIH-induced wake impairments and oxidative injuries. SUBJECTS AND SETTING: Young adult male and female C57BI/6J mice were studied in a university laboratory. INTERVENTIONS: Mice were randomly assigned to either LTIH or sham LTIH for 8 weeks. Total (24-h) wake time and mean sleep latency were measured under 2 conditions: rested and following 6 hours of enforced wakefulness. NADPH oxidase activation, carbonylation, and lipid peroxidation assays were also performed to assess sex differences in oxidative responses to LTIH. RESULTS: In contrast with the significant LTIH-induced wake impairments observed in male mice, females following LTIH showed normal wake times and sleep latencies. Female mice revealed less baseline carbonylation and less carbonylation following LTIH but showed robust NADPH oxidase activation and lipid peroxidation. In contrast with the female relative resistance to LTIH sleepiness, female mice showed more-pronounced sleepiness and delta response after enforced wakefulness. CONCLUSIONS: Despite a robust oxidative response to LTIH, age-matched female mice may be protected, at least temporarily, from LTIH wake impairments by lower basal carbonylation. In contrast, females show greater wake impairments after sleep deprivation. We hypothesize sex differences in polysomnographic predictors of sleepiness and residual sleepiness in humans with sleep apnea.


Asunto(s)
Trastornos de Somnolencia Excesiva/epidemiología , Hipoxia/fisiopatología , Estrés Oxidativo/fisiología , Animales , Trastornos de Somnolencia Excesiva/enzimología , Femenino , Hipoxia/enzimología , Hipoxia/epidemiología , Isoprostanos/fisiología , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/metabolismo , Carbonilación Proteica/fisiología , Distribución Aleatoria , Factores Sexuales , Privación de Sueño/enzimología , Privación de Sueño/epidemiología , Privación de Sueño/fisiopatología , Trastornos del Sueño del Ritmo Circadiano/enzimología , Trastornos del Sueño del Ritmo Circadiano/epidemiología , Vigilia/fisiología
12.
Sleep ; 39(8): 1601-11, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-27306266

RESUMEN

STUDY OBJECTIVES: Intermittent short sleep (ISS) is pervasive among students and workers in modern societies, yet the lasting consequences of repeated short sleep on behavior and brain health are largely unexplored. Wake-activated neurons may be at increased risk of metabolic injury across sustained wakefulness. METHODS: To examine the effects of ISS on wake-activated neurons and wake behavior, wild-type mice were randomized to ISS (a repeated pattern of short sleep on 3 consecutive days followed by 4 days of recovery sleep for 4 weeks) or rested control conditions. Subsets of both groups were allowed a recovery period consisting of 4-week unperturbed activity in home cages with littermates. Mice were examined for immediate and delayed (following recovery) effects of ISS on wake neuron cell metabolics, cell counts, and sleep/wake patterns. RESULTS: ISS resulted in sustained disruption of sleep/wake activity, with increased wakefulness during the lights-on period and reduced wake bout duration and wake time during the lights-off period. Noradrenergic locus coeruleus (LC) and orexinergic neurons showed persistent alterations in morphology, and reductions in both neuronal stereological cell counts and fronto-cortical projections. Surviving wake-activated neurons evidenced persistent reductions in sirtuins 1 and 3 and increased lipofuscin. In contrast, ISS resulted in no lasting injury to the sleep-activated melanin concentrating hormone neurons. CONCLUSIONS: Collectively these findings demonstrate for the first time that ISS imparts significant lasting disturbances in sleep/wake activity, degeneration of wake-activated LC and orexinergic neurons, and lasting metabolic changes in remaining neurons most consistent with premature senescence.


Asunto(s)
Locus Coeruleus/patología , Neuronas/metabolismo , Neuronas/patología , Orexinas/metabolismo , Trastornos del Sueño-Vigilia/fisiopatología , Envejecimiento/metabolismo , Animales , Recuento de Células , Oscuridad , Luz , Lipofuscina/metabolismo , Locus Coeruleus/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/efectos de la radiación , Norepinefrina/metabolismo , Distribución Aleatoria , Sirtuinas/metabolismo , Sueño/fisiología , Sueño/efectos de la radiación , Vigilia/fisiología , Vigilia/efectos de la radiación
13.
Front Neurol ; 6: 109, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26074865

RESUMEN

Chronic sleep disruption (CSD) is a cardinal feature of sleep apnea that predicts impaired wakefulness. Despite effective treatment of apneas and sleep disruption, patients with sleep apnea may have persistent somnolence. Lasting wake disturbances in treated sleep apnea raise the possibility that CSD may induce sufficient degeneration in wake-activated neurons (WAN) to cause irreversible wake impairments. Implementing a stereological approach in a murine model of CSD, we found reduced neuronal counts in representative WAN groups, locus coeruleus (LC) and orexinergic neurons, reduced by 50 and 25%, respectively. Mice exposed to CSD showed shortened sleep latencies lasting at least 4 weeks into recovery from CSD. As CSD results in frequent activation of WAN, we hypothesized that CSD promotes mitochondrial metabolic stress in WAN. In support, CSD increased lipofuscin within select WAN. Further, examining the LC as a representative WAN nucleus, we observed increased mitochondrial protein acetylation and down-regulation of anti-oxidant enzyme and brain-derived neurotrophic factor mRNA. Remarkably, CSD markedly increased tumor necrosis factor-alpha within WAN, and not in adjacent neurons or glia. Thus, CSD, as observed in sleep apnea, results in a composite of lasting wake impairments, loss of select neurons, a pro-inflammatory, pro-oxidative mitochondrial stress response in WAN, consistent with a degenerative process with behavioral consequences.

14.
Sleep ; 27(3): 388-93, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15164889

RESUMEN

STUDY OBJECTIVES: Objectives were to (1) establish a measure of sleep propensity for a more comprehensive characterization of sleepiness in murine genetics and interventional studies and (2) to characterize sample sizes necessary for statistical differences in effect. DESIGN: Average multiple sleep latency values were compared in mice, varying strain, circadian time, and forced-wakefulness conditions. SUBJECTS: Adult male mice of inbred strains were studied. INTERVENTIONS: Mice were implanted with electroencephalographic and electromyographic recording electrodes. Twenty-four-hour periods of stable baseline sleep activity (> 600 minutes) were confirmed prior to baseline sleep-latency testing. Average sleep latencies were obtained across 10- and 20-minute nap opportunities within 4 consecutive 30-minute periods. Forced wakefulness protocols were performed prior to additional sleep-latency tests. MEASUREMENTS AND RESULTS: Sleep-latency testing with 20-minute nap opportunities every 30 minutes revealed a shorter sleep latency in the lights-on period (12.4 minutes +/- 0.9 vs 16.5 +/- 1, P < .001), a substantial reduction in sleep latencies in mice subjected to 6-hour forced wakefulness (eg, C57BL/6J baseline: 12.4 +/- 0.9 minutes, and forced wakefulness, 8.5 +/- 0.9 minutes, P < .01), and strain differences in latencies following short-term forced wakefulness (P < .01). Sample sizes for 85% power to detect a 25% reduction in the 20-minute daytime Murine Multiple Sleep Latency Test require fewer than 20 mice per group for commonly used transgenic background strains. CONCLUSIONS: The Murine Multiple Sleep Latency Test is a robust measure of sleep propensity, and the latency varies with homeostatic and circadian influences. The test requires minimal added time to standard murine sleep recordings, yet yields important additive information.


Asunto(s)
Trastornos de Somnolencia Excesiva/genética , Sueño/fisiología , Animales , Ritmo Circadiano/fisiología , Electrodos Implantados , Electroencefalografía , Electromiografía/instrumentación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Fenotipo , Factores de Tiempo , Vigilia/fisiología
15.
Sleep ; 27(2): 194-201, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15124711

RESUMEN

STUDY OBJECTIVES: This study was designed to test the hypothesis that long-term intermittent hypoxia (LTIH), modeling the hypoxia-reoxygenation events of sleep apnea, results in oxidative neural injury, including wake-promoting neural groups, and that this injury contributes to residual impaired maintenance of wakefulness. DESIGN: Sleep times and oxidative-injury parameters were compared for mice exposed to LTIH and mice exposed to sham LTIH. SUBJECTS: Adult male C57BL/6J mice were studied. INTERVENTIONS: Mice were exposed to LTIH or sham LTIH in the lights-on period daily for 8 weeks. Electrophysiologic sleep-wake recordings and oxidative-injury measures were performed either immediately or 2 weeks following LTIH exposures. MEASUREMENTS AND RESULTS: At both intervals, total sleep time per 24 hours in LTIH-exposed mice was increased by more than 2 hours, (P<.01). Mean sleep latency was reduced in LTIH-exposed mice relative to sham LTIH mice (8.9 +/- 1.0 minutes vs 12.7 +/- 0.5 minutes, respectively, P<.01). Oxidative injury was present 2 weeks following LTIH in wake-promoting regions of the basal forebrain and brainstem: elevated isoprostane 8,12-iso-IPF2alpha-VI, 22%, P<.05; increased protein carbonylation, 50%, P<.05, increased nitration, 200%, P<.05, and induction of antioxidant enzymes glutathione reductase and methionine sulfoxide reductase A, P<.01. CONCLUSIONS: Exposure to LTIH results in an array of significant oxidative injuries in sleep-wake regions of the brain, and these biochemical changes are associated with marked hypersomnolence and increased susceptibility to short-term sleep loss. The residual forebrain redox alterations in wake-promoting brain regions may contribute to persistent sleepiness in a prevalent disorder, obstructive sleep apnea.


Asunto(s)
Trastornos de Somnolencia Excesiva/etiología , Hipoxia Encefálica/complicaciones , Hipoxia Encefálica/fisiopatología , Estrés Oxidativo/fisiología , Trastornos del Sueño del Ritmo Circadiano/fisiopatología , Animales , Electrodos Implantados , Inmunohistoquímica , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Prosencéfalo/fisiopatología
16.
Sleep ; 37(1): 51-64, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24470695

RESUMEN

STUDY OBJECTIVES: Delayed hypercapnic arousals may occur in obstructive sleep apnea. The impaired arousal response is expected to promote more pronounced oxyhemoglobin desaturations. We hypothesized that long-term sleep fragmentation (SF) results in injury to or dysfunction of wake-active neurons that manifests, in part, as a delayed hypercapnic arousal response. DESIGN: Adult male mice were implanted for behavioral state recordings and randomly assigned to 4 weeks of either orbital platform SF (SF4wk, 30 events/h) or control conditions (Ct4wk) prior to behavioral, histological, and locus coeruleus (LC) whole cell electrophysiological evaluations. MEASUREMENTS AND RESULTS: SF was successfully achieved across the 4 week study, as evidenced by a persistently increased arousal index, P < 0.01 and shortened sleep bouts, P < 0.05, while total sleep/wake times and plasma corticosterone levels were unaffected. A multiple sleep latency test performed at the onset of the dark period showed a reduced latency to sleep in SF4wk mice (P < 0.05). The hypercapnic arousal latency was increased, Ct4wk 64 ± 5 sec vs. SF4wk 154 ± 6 sec, P < 0.001, and remained elevated after a 2 week recovery (101 ± 4 sec, P < 0.001). C-fos activation in noradrenergic, orexinergic, histaminergic, and cholinergic wake-active neurons was reduced in response to hypercapnia (P < 0.05-0.001). Catecholaminergic and orexinergic projections into the cingulate cortex were also reduced in SF4wk (P < 0.01). In addition, SF4wk resulted in impaired LC neuron excitability (P < 0.01). CONCLUSIONS: Four weeks of sleep fragmentation (SF4wk) impairs arousal responses to hypercapnia, reduces wake neuron projections and locus coeruleus neuronal excitability, supporting the concepts that some effects of sleep fragmentation may contribute to impaired arousal responses in sleep apnea, which may not reverse immediately with therapy.


Asunto(s)
Nivel de Alerta/fisiología , Hipercapnia/fisiopatología , Neuronas/fisiología , Apnea Obstructiva del Sueño/fisiopatología , Privación de Sueño/patología , Privación de Sueño/fisiopatología , Vigilia/fisiología , Animales , Axones/fisiología , Enfermedad Crónica , Corticosterona/sangre , Electroencefalografía , Hipercapnia/sangre , Hipercapnia/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Locus Coeruleus/citología , Locus Coeruleus/patología , Locus Coeruleus/fisiopatología , Masculino , Ratones , Neuropéptidos/metabolismo , Orexinas , Polisomnografía , Corteza Prefrontal/patología , Corteza Prefrontal/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Sueño/fisiología , Apnea Obstructiva del Sueño/sangre , Apnea Obstructiva del Sueño/patología , Privación de Sueño/sangre , Factores de Tiempo
17.
Sleep ; 36(4): 481-92, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23564995

RESUMEN

STUDY OBJECTIVES: Obstructive sleep apnea (OSA) is associated with cognitive impairment and neuronal injury. Long-term exposure to intermittent hypoxia (LTIH) in rodents, modeling the oxygenation patterns in sleep apnea, results in NADPH oxidase 2 (Nox2) oxidative injury to many neuronal populations. Brainstem motoneurons susceptible to LTIH injury show uncompensated endoplasmic reticulum stress responses with increased (CCAAT/enhancer binding protein homologous protein (CHOP). We hypothesized that CHOP underlies LTIH oxidative injury. In this series of studies, we first determined whether CHOP is upregulated in other brain regions susceptible to LTIH oxidative Nox2 injury and then determined whether CHOP plays an adaptive or injurious role in the LTIH response. To integrate these findings with previous studies examining LTIH neural injury, we examined the role of CHOP in Nox2, hypoxia-inducible factor-1α (HIF-1α) responses, oxidative injury and apoptosis, and neuron loss. DESIGN: Within/between mice subjects. SETTING: Laboratory setting. PARTICIPANTSSUBJECTS: CHOP null and wild-type adult male mice. INTERVENTIONS: LTIH or sham LTIH. MEASUREMENTS AND MAIN RESULTS: Relative to wild-type mice, CHOP-/- mice conferred resistance to oxidative stress (superoxide production/ carbonyl proteins) in brain regions examined: cortex, hippocampus, and motor nuclei. CHOP deletion prevented LTIH upregulation of Nox2 and HIF-1α in the hippocampus, cortex, and brainstem motoneurons and protected mice from neuronal apoptosis and motoneuron loss. CONCLUSIONS: Endogenous CHOP is necessary for LTIH-induced HIF-1α, Nox2 upregulation, and oxidative stress; CHOP influences LTIH-induced apoptosis in neurons and loss of neurons. Findings support the concept that minimizing CHOP may provide neuroprotection in OSA.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/fisiopatología , Neuronas Motoras/metabolismo , Síndromes de la Apnea del Sueño/metabolismo , Síndromes de la Apnea del Sueño/fisiopatología , Factor de Transcripción CHOP/metabolismo , Análisis de Varianza , Animales , Apoptosis , Western Blotting/métodos , Modelos Animales de Enfermedad , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
18.
Aging Cell ; 10(4): 640-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21388495

RESUMEN

Fragmentation of wakefulness and sleep are expected outcomes of advanced aging. We hypothesize that wake neurons develop endoplasmic reticulum dyshomeostasis with aging, in parallel with impaired wakefulness. In this series of experiments, we sought to more fully characterize age-related changes in wakefulness and then, in relevant wake neuronal populations, explore functionality and endoplasmic reticulum homeostasis. We report that old mice show greater sleep/wake transitions in the active period with markedly shortened wake periods, shortened latencies to sleep, and less wake time in the subjective day in response to a novel social encounter. Consistent with sleep/wake instability and reduced social encounter wakefulness, orexinergic and noradrenergic wake neurons in aged mice show reduced c-fos response to wakefulness and endoplasmic reticulum dyshomeostasis with increased nuclear translocation of CHOP and GADD34. We have identified an age-related unfolded protein response injury to and dysfunction of wake neurons. It is anticipated that these changes contribute to sleep/wake fragmentation and cognitive impairment in aging.


Asunto(s)
Envejecimiento/fisiología , Estrés del Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Neuronas/metabolismo , Vigilia/fisiología , Animales , Ritmo Circadiano/fisiología , Masculino , Ratones , Proteína Fosfatasa 1/metabolismo , Transporte de Proteínas/fisiología , Factor de Transcripción CHOP/metabolismo
19.
Am J Respir Crit Care Med ; 171(12): 1414-20, 2005 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15750040

RESUMEN

RATIONALE: Long-term intermittent hypoxia (LTIH) exposure in adult mice, modeling oxygenation patterns of moderate-severe obstructive sleep apnea, results in lasting hypersomnolence and is associated with nitration and oxidation injuries in many brain regions, including wake-active regions. OBJECTIVES: We sought to determine if LTIH activates inducible nitric oxide synthase (iNOS) in sleep/wake regions, and if this source of NO contributes to the LTIH-induced proinflammatory gene response, oxidative injury, and wake impairments. METHODS: Mice with genetic absence of iNOS activity and wild-type control animals were exposed to 6 weeks of long-term hypoxia/reoxygenation before behavioral state recordings, molecular and biochemical assays, and a pharmacologic intervention. MEASUREMENTS AND MAIN RESULTS: Two weeks after recovery from hypoxia/reoxygenation exposures, wild-type mice showed increased iNOS activity in representative wake-active regions, increased sleep times, and shortened sleep latencies. Mutant mice, with higher baseline sleep times, showed no effect of long-term hypoxia/reoxygenation on sleep time latencies and were resistant to hypoxia/reoxygenation increases in lipid peroxidation and proinflammatory gene responses (tumor necrosis factor alpha and cyclooxygenase 2). Inhibition of iNOS after long-term hypoxia/reoxygenation in wild-type mice was effective in reversing the proinflammatory gene response. CONCLUSIONS: These data support a critical role for iNOS activity in the development of LTIH wake impairments, lipid peroxidation, and proinflammatory responses in wake-active brain regions, and suggest a potential role for inducible NO inhibition in protection from proinflammatory responses, oxidative injury, and residual hypersomnolence in obstructive sleep apnea.


Asunto(s)
Trastornos de Somnolencia Excesiva/etiología , Hipoxia Encefálica/enzimología , Isoprostanos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Trastornos de Somnolencia Excesiva/fisiopatología , Ensayo de Inmunoadsorción Enzimática , Hipoxia Encefálica/complicaciones , Peroxidación de Lípido/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Estrés Oxidativo/fisiología , Probabilidad , Distribución Aleatoria , Valores de Referencia , Sensibilidad y Especificidad
20.
Am J Respir Crit Care Med ; 172(7): 921-9, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15994465

RESUMEN

RATIONALE: Persons with obstructive sleep apnea may have significant residual hypersomnolence, despite therapy. Long-term hypoxia/reoxygenation events in adult mice, simulating oxygenation patterns of moderate-severe sleep apnea, result in lasting hypersomnolence, oxidative injury, and proinflammatory responses in wake-active brain regions. We hypothesized that long-term intermittent hypoxia activates brain NADPH oxidase and that this enzyme serves as a critical source of superoxide in the oxidation injury and in hypersomnolence. OBJECTIVES: We sought to determine whether long-term hypoxia/reoxygenation events in mice result in NADPH oxidase activation and whether NADPH oxidase is essential for the proinflammatory response and hypersomnolence. METHODS: NADPH oxidase gene and protein responses were measured in wake-active brain regions in wild-type mice exposed to long-term hypoxia/reoxygenation. Sleep and oxidative and proinflammatory responses were measured in adult mice either devoid of NADPH oxidase activity (gp91phox-null mice) or in which NADPH oxidase activity was systemically inhibited with apocynin osmotic pumps throughout hypoxia/reoxygenation. MAIN RESULTS: Long-term intermittent hypoxia increased NADPH oxidase gene and protein responses in wake-active brain regions. Both transgenic absence and pharmacologic inhibition of NADPH oxidase activity throughout long-term hypoxia/reoxygenation conferred resistance to not only long-term hypoxia/reoxygenation hypersomnolence but also to carbonylation, lipid peroxidation injury, and the proinflammatory response, including inducible nitric oxide synthase activity in wake-active brain regions. CONCLUSIONS: Collectively, these findings strongly support a critical role for NADPH oxidase in the lasting hypersomnolence and oxidative and proinflammatory responses after hypoxia/reoxygenation patterns simulating severe obstructive sleep apnea oxygenation, highlighting the potential of inhibiting NADPH oxidase to prevent oxidation-mediated morbidities in obstructive sleep apnea.


Asunto(s)
Trastornos de Somnolencia Excesiva/fisiopatología , Hipoxia/fisiopatología , NADPH Oxidasas/fisiología , Animales , Modelos Animales de Enfermedad , Expresión Génica , Inmunohistoquímica , Masculino , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2 , Estrés Oxidativo , Prosencéfalo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA