Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
J Neurosci ; 44(28)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38830763

RESUMO

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.


Assuntos
Peptídeos beta-Amiloides , Cisteína Endopeptidases , Hipocampo , Locus Cerúleo , Norepinefrina , Privação do Sono , Animais , Peptídeos beta-Amiloides/metabolismo , Norepinefrina/metabolismo , Camundongos , Hipocampo/metabolismo , Hipocampo/patologia , Privação do Sono/metabolismo , Privação do Sono/patologia , Masculino , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/genética , Fragmentos de Peptídeos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dopamina beta-Hidroxilase/metabolismo , Dopamina beta-Hidroxilase/genética , Proteínas tau/metabolismo , Feminino , Degeneração Neural/patologia , Degeneração Neural/metabolismo , Degeneração Neural/genética
2.
Neurobiol Dis ; 139: 104820, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32087293

RESUMO

Chronic short sleep or extended wake periods are commonly observed in most industrialized countries. Previously neurobehavioral impairment following sleep loss was considered to be a readily reversible occurrence, normalized upon recovery sleep. Recent clinical studies suggest that chronic short sleep and sleep disruption may be risk factors for neurodegeneration. Animal models have been instrumental in determining whether disturbed sleep can injure the brain. We now understand that repeated periods of extended wakefulness across the typical sleep period and/or sleep fragmentation can have lasting effects on neurogenesis and select populations of neurons and glia. Here we provide a comprehensive overview of the advancements made using animal models of sleep loss to understand the extent and mechanisms of chronic short sleep induced neural injury.


Assuntos
Doenças Neurodegenerativas/complicações , Privação do Sono/complicações , Peptídeos beta-Amiloides/metabolismo , Animais , Modelos Animais de Doenças , Sono/fisiologia , Vigília/fisiologia
3.
J Neurosci ; 38(48): 10255-10270, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30322903

RESUMO

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.


Assuntos
Progressão da Doença , Mutação/fisiologia , Privação do Sono/metabolismo , Tauopatias/metabolismo , Proteínas tau/metabolismo , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Animais , Feminino , Humanos , Locus Cerúleo/metabolismo , Locus Cerúleo/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Transgênicos , Privação do Sono/genética , Privação do Sono/patologia , Tauopatias/genética , Tauopatias/patologia , Proteínas tau/genética
4.
Physiol Rev ; 90(1): 47-112, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20086074

RESUMO

Sleep-induced apnea and disordered breathing refers to intermittent, cyclical cessations or reductions of airflow, with or without obstructions of the upper airway (OSA). In the presence of an anatomically compromised, collapsible airway, the sleep-induced loss of compensatory tonic input to the upper airway dilator muscle motor neurons leads to collapse of the pharyngeal airway. In turn, the ability of the sleeping subject to compensate for this airway obstruction will determine the degree of cycling of these events. Several of the classic neurotransmitters and a growing list of neuromodulators have now been identified that contribute to neurochemical regulation of pharyngeal motor neuron activity and airway patency. Limited progress has been made in developing pharmacotherapies with acceptable specificity for the treatment of sleep-induced airway obstruction. We review three types of major long-term sequelae to severe OSA that have been assessed in humans through use of continuous positive airway pressure (CPAP) treatment and in animal models via long-term intermittent hypoxemia (IH): 1) cardiovascular. The evidence is strongest to support daytime systemic hypertension as a consequence of severe OSA, with less conclusive effects on pulmonary hypertension, stroke, coronary artery disease, and cardiac arrhythmias. The underlying mechanisms mediating hypertension include enhanced chemoreceptor sensitivity causing excessive daytime sympathetic vasoconstrictor activity, combined with overproduction of superoxide ion and inflammatory effects on resistance vessels. 2) Insulin sensitivity and homeostasis of glucose regulation are negatively impacted by both intermittent hypoxemia and sleep disruption, but whether these influences of OSA are sufficient, independent of obesity, to contribute significantly to the "metabolic syndrome" remains unsettled. 3) Neurocognitive effects include daytime sleepiness and impaired memory and concentration. These effects reflect hypoxic-induced "neural injury." We discuss future research into understanding the pathophysiology of sleep apnea as a basis for uncovering newer forms of treatment of both the ventilatory disorder and its multiple sequelae.


Assuntos
Síndromes da Apneia do Sono/fisiopatologia , Animais , Sistema Cardiovascular/fisiopatologia , Cognição/fisiologia , Modelos Animais de Doenças , História do Século XIX , História do Século XX , História do Século XXI , Humanos , Resistência à Insulina/fisiologia , Sistema Nervoso/fisiopatologia , Sistema Respiratório/fisiopatologia , Síndromes da Apneia do Sono/história
5.
N Engl J Med ; 381(3): e7, 2019 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-31314987
8.
FASEB J ; 29(6): 2315-26, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25681462

RESUMO

Conventional T (Tcon) cells and Foxp3(+) T-regulatory (Treg) cells are thought to have differing metabolic requirements, but little is known of mitochondrial functions within these cell populations in vivo. In murine studies, we found that activation of both Tcon and Treg cells led to myocyte enhancer factor 2 (Mef2)-induced expression of genes important to oxidative phosphorylation (OXPHOS). Inhibition of OXPHOS impaired both Tcon and Treg cell function compared to wild-type cells but disproportionally affected Treg cells. Deletion of Pgc1α or Sirt3, which are key regulators of OXPHOS, abrogated Treg-dependent suppressive function and impaired allograft survival. Mef2 is inhibited by histone/protein deacetylase-9 (Hdac9), and Hdac9 deletion increased Treg suppressive function. Hdac9(-/-) Treg showed increased expression of Pgc1α and Sirt3, and improved mitochondrial respiration, compared to wild-type Treg cells. Our data show that key OXPHOS regulators are required for optimal Treg function and Treg-dependent allograft acceptance. These findings provide a novel approach to increase Treg function and give insights into the fundamental mechanisms by which mitochondrial energy metabolism regulates immune cell functions in vivo.


Assuntos
Metabolismo Energético/imunologia , Fatores de Transcrição Forkhead/imunologia , Sobrevivência de Enxerto/imunologia , Mitocôndrias/imunologia , Linfócitos T Reguladores/imunologia , Animais , Western Blotting , Metabolismo Energético/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Sobrevivência de Enxerto/genética , Histona Desacetilases/genética , Histona Desacetilases/imunologia , Histona Desacetilases/metabolismo , Fatores de Transcrição MEF2/imunologia , Fatores de Transcrição MEF2/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/imunologia , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 3/genética , Sirtuína 3/imunologia , Sirtuína 3/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Fatores de Transcrição/metabolismo
9.
J Neurosci ; 34(12): 4418-31, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24647961

RESUMO

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.


Assuntos
Locus Cerúleo/metabolismo , Degeneração Neural/metabolismo , Neurônios/metabolismo , Privação do Sono/metabolismo , Sono/fisiologia , Vigília/fisiologia , Animais , Corticosterona/sangue , Locus Cerúleo/patologia , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Degeneração Neural/patologia , Neurônios/patologia , Estresse Oxidativo/fisiologia , Sirtuína 3/genética , Sirtuína 3/metabolismo , Privação do Sono/patologia , Regulação para Cima
10.
Nat Genet ; 32(2): 290-5, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12244319

RESUMO

Rab3a is the most abundant Rab (ras-associated binding) protein in the brain and has a regulatory role in synaptic vesicle trafficking. Mice with a targeted loss-of-function mutation in Rab3a have defects in Ca(2+)-dependent synaptic transmission: the number of vesicles released in response to an action potential is greater than in wildtype mice, resulting in greater synaptic depression and the abolishment of CA3 mossy-fiber long term potentiation. The effect of these changes on behavior is unknown. In a screen for mouse mutants with abnormal rest-activity and sleep patterns, we identified a semidominant mutation, called earlybird, that shortens the circadian period of locomotor activity. Sequence analysis of Rab3a identified a point mutation in the conserved amino acid (Asp77Gly) within the GTP-binding domain of this protein in earlybird mutants, resulting in significantly reduced levels of Rab3a protein. Phenotypic assessment of earlybird mice and a null allele of Rab3a revealed anomalies in circadian period and sleep homeostasis, providing evidence that Rab3a-mediated synaptic transmission is involved in these behaviors.


Assuntos
Ritmo Circadiano/genética , Mutação Puntual , Privação do Sono/fisiopatologia , Proteína rab3A de Ligação ao GTP/genética , Alelos , Animais , Western Blotting , Etilnitrosoureia , Feminino , Homeostase , Locomoção/genética , Locomoção/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Transtornos do Sono-Vigília/genética , Proteína rab3A de Ligação ao GTP/fisiologia
11.
Transl Psychiatry ; 13(1): 78, 2023 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-36869037

RESUMO

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.


Assuntos
Morfina , Transtornos do Sono-Vigília , Animais , Camundongos , Analgésicos Opioides , Receptores Opioides mu , Neurônios , Tálamo
12.
J Neurosci ; 31(11): 4025-36, 2011 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-21411645

RESUMO

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.


Assuntos
Envelhecimento/fisiologia , Neurônios/metabolismo , Sirtuína 1/metabolismo , Vigília/fisiologia , Animais , Western Blotting , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Prosencéfalo/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/genética , Sono/fisiologia
13.
FASEB J ; 25(6): 1922-33, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21368105

RESUMO

Pulmonary arterial vascular smooth muscle (PAVSM) cell proliferation is a key pathophysiological component of vascular remodeling in pulmonary arterial hypertension (PAH) for which cellular and molecular mechanisms are poorly understood. The goal of our study was to determine the role of mammalian target of rapamycin (mTOR) in PAVSM cell proliferation, a major pathological manifestation of vascular remodeling in PAH. Our data demonstrate that chronic hypoxia promoted mTOR(Ser-2481) phosphorylation, an indicator of mTOR intrinsic catalytic activity, mTORC1-specific S6 and mTORC2-specific Akt (Ser-473) phosphorylation, and proliferation of human and rat PAVSM cells that was inhibited by siRNA mTOR. PAVSM cells derived from rats exposed to chronic hypoxia (VSM-H cells) retained increased mTOR(Ser-2481), S6, Akt (Ser-473) phosphorylation, and DNA synthesis compared to cells from normoxia-exposed rats. Suppression of mTORC2 signaling with siRNA rictor, or inhibition of mTORC1 signaling with rapamycin and metformin, while having little effect on other complex activities, inhibited VSM-H and chronic hypoxia-induced human and rat PAVSM cell proliferation. Collectively, our data demonstrate that up-regulation of mTOR activity and activation of both mTORC1 and mTORC2 are required for PAVSM cell proliferation induced by in vitro and in vivo chronic hypoxia and suggest that mTOR may serve as a potential therapeutic target to inhibit vascular remodeling in PAH.


Assuntos
Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Artéria Pulmonar/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Proliferação de Células , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Masculino , Consumo de Oxigênio , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Serina-Treonina Quinases TOR/genética
14.
Trends Neurosci ; 45(9): 678-691, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35691776

RESUMO

Recent studies in both humans and animal models call into question the completeness of recovery after chronic sleep disruption. Studies in humans have identified cognitive domains particularly vulnerable to delayed or incomplete recovery after chronic sleep disruption, including sustained vigilance and episodic memory. These findings, in turn, provide a focus for animal model studies to critically test the lasting impact of sleep loss on the brain. Here, we summarize the human response to sleep disruption and then discuss recent findings in animal models examining recovery responses in circuits pertinent to vigilance and memory. We then propose pathways of injury common to various forms of sleep disruption and consider the implications of this injury in aging and in neurodegenerative disorders.


Assuntos
Memória Episódica , Sono , Envelhecimento , Animais , Encéfalo , Humanos , Sono/fisiologia , Vigília/fisiologia
15.
Front Aging Neurosci ; 14: 1025402, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36275002

RESUMO

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.

16.
Front Neurosci ; 16: 836693, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35250468

RESUMO

The opioid epidemic remains a significant healthcare problem and is attributable to over 100,000 deaths per year. Poor sleep increases sensitivity to pain, impulsivity, inattention, and negative affect, all of which might perpetuate drug use. Opioid users have disrupted sleep during drug use and withdrawal and report poor sleep as a reason for relapse. However, preclinical studies investigating the relationship between sleep loss and substance use and the associated underlying neurobiological mechanisms of potential interactions are lacking. One of the most common forms of sleep loss in modern society is chronic short sleep (CSS) (<7 h/nightly for adults). Here, we used an established model of CSS to investigate the influence of disrupted sleep on opioid reward in male mice. The CSS paradigm did not increase corticosterone levels or depressive-like behavior after a single sleep deprivation session but did increase expression of Iba1, which typically reflects microglial activation, in the hypothalamus after 4 weeks of CSS. Rested control mice developed a morphine preference in a 2-bottle choice test, while mice exposed to CSS did not develop a morphine preference. Both groups demonstrated morphine conditioned place preference (mCPP), but there were no differences in conditioned preference between rested and CSS mice. Taken together, our results show that recovery sleep after chronic sleep disruption lessens voluntary opioid intake, without impacting conditioned reward associated with morphine.

17.
Nephrol Dial Transplant ; 26(7): 2244-50, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21317406

RESUMO

BACKGROUND: Previous studies have found an association between obstructive sleep apnea (OSA) and chronic kidney disease (CKD). However, subjects with confounding factors such as diabetes and hypertension were not excluded. The purpose of the present study was to determine whether patients with OSA without meeting criteria for diabetes or hypertension would also show increased likelihood of CKD. METHODS: We prospectively enrolled adult patients with a chief complaint of habitual snoring. Overnight polysomnography, fasting blood triglyceride, cholesterol, glucose, insulin, creatinine, albumin and hemoglobin A1c, and first voiding urine albumin and creatinine were examined. Estimated glomerular filtration rate (eGFR), urine albumin-to-creatinine ratio (UACR), homeostatic model assessment-insulin resistance and percentage of CKD were calculated. RESULTS: The final analyses involved 40 patients who were middle-aged [44.8 (8.6) years] predominantly male (83%), obese [body mass index, 28.2 (5.1) kg/m(2)] and more severe OSA, with an apnea-hypopnea index (AHI) of 51.6 (39.2)/h. The mean eGFR and UACR were 85.4 (18.3) mL/min/1.73m(2) and 13.4 (23.4) mg/g, respectively. The prevalence of CKD in severe OSA subjects is 18%. With stepwise multivariate linear regression analysis, AHI and desaturation index were the only independent predictor of UACR (ß = 0.26, P = 0.01, R(2) = 0.17) and eGFR (ß = 0.32, P < 0.01, R(2) = 0.32), respectively. CONCLUSIONS: High prevalence of CKD is present in severe OSA patients without hypertension or diabetes. Significantly positive correlations were found between severity of OSA and renal function impairment.


Assuntos
Falência Renal Crônica/complicações , Apneia Obstrutiva do Sono/etiologia , Adulto , Idoso , Albuminúria , Índice de Massa Corporal , Creatinina/metabolismo , Feminino , Seguimentos , Taxa de Filtração Glomerular , Hemoglobinas Glicadas/metabolismo , Humanos , Incidência , Testes de Função Renal , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Prospectivos , Fatores de Risco
18.
Proc Natl Acad Sci U S A ; 105(4): 1309-14, 2008 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18195361

RESUMO

The neural mechanisms through which the state of anesthesia arises and dissipates remain unknown. One common belief is that emergence from anesthesia is the inverse process of induction, brought about by elimination of anesthetic drugs from their CNS site(s) of action. Anesthetic-induced unconsciousness may result from specific interactions of anesthetics with the neural circuits regulating sleep and wakefulness. Orexinergic agonists and antagonists have the potential to alter the stability of the anesthetized state. In this report, we refine the role of the endogenous orexin system in impacting emergence from, but not entry into the anesthetized state, and in doing so, we distinguish mechanisms of induction from those of emergence. We demonstrate that isoflurane and sevoflurane, two commonly used general anesthetics, inhibit c-Fos expression in orexinergic but not adjacent melanin-concentrating hormone (MCH) neurons; suggesting that wake-active orexinergic neurons are inhibited by these anesthetics. Genetic ablation of orexinergic neurons, which causes acquired murine narcolepsy, delays emergence from anesthesia, without changing anesthetic induction. Pharmacologic studies with a selective orexin-1 receptor antagonist confirm a specific orexin effect on anesthetic emergence without an associated change in induction. We conclude that there are important differences in the neural substrates mediating induction and emergence. These findings support the concept that emergence depends, in part, on recruitment and stabilization of wake-active regions of brain.


Assuntos
Anestesia Geral , Anestésicos Inalatórios , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Neuropeptídeos/fisiologia , Anestesia Geral/efeitos adversos , Animais , Feminino , Peptídeos e Proteínas de Sinalização Intracelular/agonistas , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isoflurano , Masculino , Éteres Metílicos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neuropeptídeos/agonistas , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/genética , Orexinas , Transporte Proteico , Proteínas Proto-Oncogênicas c-fos/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Sevoflurano , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
19.
Sleep ; 44(8)2021 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-33768250

RESUMO

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.


Assuntos
Doença de Alzheimer , Proteínas tau , Doença de Alzheimer/etiologia , Peptídeos beta-Amiloides , Animais , Camundongos , Placa Amiloide , Sono
20.
Curr Neurol Neurosci Rep ; 10(1): 47-52, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20425226

RESUMO

Sleepiness has long been recognized as a presenting symptom in obstructive sleep apnea syndrome, but persistent neurocognitive injury from sleep apnea has been appreciated only recently. Although therapy for sleep apnea markedly improves daytime symptoms, cognitive impairments may persist despite long-term therapy with continuous positive airway pressure. We know now that certain groups of neurons, typically those that are more metabolically active, are more vulnerable to injury than others. Animal models of sleep apnea oxygenation patterns have been instrumental in elucidating mechanisms of injury. The hypoxia/reoxygenation events result in oxidative, inflammatory, and endoplasmic reticulum stress responses in susceptible neural groups. With molecular pathways being fleshed out in animal models, it is time to carefully and systematically examine neural injury in humans and test the applicability of findings from animal models. To succeed, however, we cannot view sleep apnea as an isolated process. Rather, injury in sleep apnea is more likely the consequence of overlapping injuries from comorbid conditions. The progress in elucidating mechanisms of neural injury is palpable, and it now seems we indeed are closer to developing therapies to prevent and treat neural injury in obstructive sleep apnea.


Assuntos
Lesões Encefálicas/patologia , Síndromes da Apneia do Sono/complicações , Traumatismos do Sistema Nervoso/etiologia , Animais , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Humanos , Condução Nervosa/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA