Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell ; 163(3): 643-55, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26496606

RESUMO

Thermogenic brown and beige adipose tissues dissipate chemical energy as heat, and their thermogenic activities can combat obesity and diabetes. Herein the functional adaptations to cold of brown and beige adipose depots are examined using quantitative mitochondrial proteomics. We identify arginine/creatine metabolism as a beige adipose signature and demonstrate that creatine enhances respiration in beige-fat mitochondria when ADP is limiting. In murine beige fat, cold exposure stimulates mitochondrial creatine kinase activity and induces coordinated expression of genes associated with creatine metabolism. Pharmacological reduction of creatine levels decreases whole-body energy expenditure after administration of a ß3-agonist and reduces beige and brown adipose metabolic rate. Genes of creatine metabolism are compensatorily induced when UCP1-dependent thermogenesis is ablated, and creatine reduction in Ucp1-deficient mice reduces core body temperature. These findings link a futile cycle of creatine metabolism to adipose tissue energy expenditure and thermal homeostasis. PAPERCLIP.


Assuntos
Tecido Adiposo Marrom/metabolismo , Creatina/metabolismo , Termogênese , Difosfato de Adenosina/metabolismo , Tecido Adiposo/metabolismo , Animais , Metabolismo Energético , Homeostase , Humanos , Canais Iônicos/metabolismo , Camundongos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Obesidade/metabolismo , Proteína Desacopladora 1
2.
Curr Neurol Neurosci Rep ; 23(12): 907-923, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38060134

RESUMO

PURPOSE OF REVIEW: Since the formal discovery of rapid eye movement (REM) sleep in 1953, we have gained a vast amount of knowledge regarding the specific populations of neurons, their connections, and synaptic mechanisms regulating this stage of sleep and its accompanying features. This article discusses REM sleep circuits and their dysfunction, specifically emphasizing recent studies using conditional genetic tools. RECENT FINDINGS: Sublaterodorsal nucleus (SLD) in the dorsolateral pons, especially the glutamatergic subpopulation in this region (SLDGlut), are shown to be indispensable for REM sleep. These neurons appear to be single REM generators in the rodent brain and may initiate and orchestrate all REM sleep events, including cortical and hippocampal activation and muscle atonia through distinct pathways. However, several cell groups in the brainstem and hypothalamus may influence SLDGlut neuron activity, thereby modulating REM sleep timing, amounts, and architecture. Damage to SLDGlut neurons or their projections involved in muscle atonia leads to REM behavior disorder, whereas the abnormal activation of this pathway during wakefulness may underlie cataplexy in narcolepsy. Despite some opposing views, it has become evident that SLDGlut neurons are the sole generators of REM sleep and its associated characteristics. Further research should prioritize a deeper understanding of their cellular, synaptic, and molecular properties, as well as the mechanisms that trigger their activation during cataplexy and make them susceptible in RBD.


Assuntos
Cataplexia , Narcolepsia , Transtorno do Comportamento do Sono REM , Humanos , Sono REM/fisiologia , Encéfalo
3.
PLoS Biol ; 17(3): e3000172, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30893297

RESUMO

Sleep and wakefulness are greatly influenced by various physiological and psychological factors, but the neuronal elements responsible for organizing sleep-wake behavior in response to these factors are largely unknown. In this study, we report that a subset of neurons in the lateral hypothalamic area (LH) expressing the neuropeptide neurotensin (Nts) is critical for orchestrating sleep-wake responses to acute psychological and physiological challenges or stressors. We show that selective activation of NtsLH neurons with chemogenetic or optogenetic methods elicits rapid transitions from non-rapid eye movement (NREM) sleep to wakefulness and produces sustained arousal, higher locomotor activity (LMA), and hyperthermia, which are commonly observed after acute stress exposure. On the other hand, selective chemogenetic inhibition of NtsLH neurons attenuates the arousal, LMA, and body temperature (Tb) responses to a psychological stress (a novel environment) and augments the responses to a physiological stress (fasting).


Assuntos
Febre/metabolismo , Região Hipotalâmica Lateral/metabolismo , Neurotensina/metabolismo , Animais , Temperatura Corporal , Eletroforese , Técnicas de Genotipagem , Locomoção/fisiologia , Masculino , Camundongos , Neurônios/metabolismo
4.
Nature ; 532(7597): 112-6, 2016 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27027295

RESUMO

Brown and beige adipose tissues can dissipate chemical energy as heat through thermogenic respiration, which requires uncoupling protein 1 (UCP1). Thermogenesis from these adipocytes can combat obesity and diabetes, encouraging investigation of factors that control UCP1-dependent respiration in vivo. Here we show that acutely activated thermogenesis in brown adipose tissue is defined by a substantial increase in levels of mitochondrial reactive oxygen species (ROS). Remarkably, this process supports in vivo thermogenesis, as pharmacological depletion of mitochondrial ROS results in hypothermia upon cold exposure, and inhibits UCP1-dependent increases in whole-body energy expenditure. We further establish that thermogenic ROS alter the redox status of cysteine thiols in brown adipose tissue to drive increased respiration, and that Cys253 of UCP1 is a key target. UCP1 Cys253 is sulfenylated during thermogenesis, while mutation of this site desensitizes the purine-nucleotide-inhibited state of the carrier to adrenergic activation and uncoupling. These studies identify mitochondrial ROS induction in brown adipose tissue as a mechanism that supports UCP1-dependent thermogenesis and whole-body energy expenditure, which opens the way to improved therapeutic strategies for combating metabolic disorders.


Assuntos
Cisteína/química , Metabolismo Energético , Canais Iônicos/química , Canais Iônicos/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/química , Proteínas Mitocondriais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Termogênese , Tecido Adiposo Marrom/química , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/metabolismo , Animais , Respiração Celular , Cisteína/genética , Cisteína/metabolismo , Metabolismo Energético/efeitos dos fármacos , Feminino , Humanos , Canais Iônicos/deficiência , Canais Iônicos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Oxirredução , Compostos de Sulfidrila/metabolismo , Termogênese/efeitos dos fármacos , Proteína Desacopladora 1
5.
J Neurosci ; 40(1): 171-190, 2020 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-31694962

RESUMO

Origin and functions of intermittent transitions among sleep stages, including brief awakenings and arousals, constitute a challenge to the current homeostatic framework for sleep regulation, focusing on factors modulating sleep over large time scales. Here we propose that the complex micro-architecture characterizing sleep on scales of seconds and minutes results from intrinsic non-equilibrium critical dynamics. We investigate θ- and δ-wave dynamics in control rats and in rats where the sleep-promoting ventrolateral preoptic nucleus (VLPO) is lesioned (male Sprague-Dawley rats). We demonstrate that bursts in θ and δ cortical rhythms exhibit complex temporal organization, with long-range correlations and robust duality of power-law (θ-bursts, active phase) and exponential-like (δ-bursts, quiescent phase) duration distributions, features typical of non-equilibrium systems self-organizing at criticality. We show that such non-equilibrium behavior relates to anti-correlated coupling between θ- and δ-bursts, persists across a range of time scales, and is independent of the dominant physiologic state; indications of a basic principle in sleep regulation. Further, we find that VLPO lesions lead to a modulation of cortical dynamics resulting in altered dynamical parameters of θ- and δ-bursts and significant reduction in θ-δ coupling. Our empirical findings and model simulations demonstrate that θ-δ coupling is essential for the emerging non-equilibrium critical dynamics observed across the sleep-wake cycle, and indicate that VLPO neurons may have dual role for both sleep and arousal/brief wake activation. The uncovered critical behavior in sleep- and wake-related cortical rhythms indicates a mechanism essential for the micro-architecture of spontaneous sleep-stage and arousal transitions within a novel, non-homeostatic paradigm of sleep regulation.SIGNIFICANCE STATEMENT We show that the complex micro-architecture of sleep-stage/arousal transitions arises from intrinsic non-equilibrium critical dynamics, connecting the temporal organization of dominant cortical rhythms with empirical observations across scales. We link such behavior to sleep-promoting neuronal population, and demonstrate that VLPO lesion (model of insomnia) alters dynamical features of θ and δ rhythms, and leads to significant reduction in θ-δ coupling. This indicates that VLPO neurons may have dual role for both sleep and arousal/brief wake control. The reported empirical findings and modeling simulations constitute first evidences of a neurophysiological fingerprint of self-organization and criticality in sleep- and wake-related cortical rhythms; a mechanism essential for spontaneous sleep-stage and arousal transitions that lays the bases for a novel, non-homeostatic paradigm of sleep regulation.


Assuntos
Sono/fisiologia , Vigília/fisiologia , Animais , Ritmo Delta , Eletroencefalografia , Masculino , Área Pré-Óptica/lesões , Área Pré-Óptica/fisiologia , Ratos , Ratos Sprague-Dawley , Fases do Sono/fisiologia , Organismos Livres de Patógenos Específicos , Ritmo Teta
6.
Eur J Neurosci ; 52(9): 4100-4114, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32588491

RESUMO

Sleep pressure that builds up gradually during the extended wakefulness results in sleep rebound. Several lines of evidence, however, suggest that wake per se may not be sufficient to drive sleep rebound and that rapid eye movement (REM) and non-rapid eye movement (NREM) sleep rebound may be differentially regulated. In this study, we investigated the relative contribution of brain versus physical activities in REM and NREM sleep rebound by four sets of experiments. First, we forced locomotion in rats in a rotating wheel for 4 hr and examined subsequent sleep rebound. Second, we exposed the rats lacking homeostatic sleep response after prolonged quiet wakefulness and arousal brain activity induced by chemoactivation of parabrachial nucleus to the same rotating wheel paradigm and tested if physical activity could rescue the sleep homeostasis. Third, we varied motor activity levels while concurrently inhibiting the cortical activity by administering ketamine or xylazine (motor inhibitor), or ketamine + xylazine mixture and investigated if motor activity in the absence of activated cortex can cause NREM sleep rebound. Fourth and finally, we manipulated cortical activity by administering ketamine (that induced active wakefulness and waking brain) alone or in combination with atropine (that selectively inhibits the cortex) and studied if cortical inhibition irrespective of motor activity levels can block REM sleep rebound. Our results demonstrate that motor activity but not cortical activity determines NREM sleep rebound whereas cortical activity but not motor activity determines REM sleep rebound.


Assuntos
Eletroencefalografia , Sono , Animais , Homeostase , Ratos , Sono REM , Vigília
7.
Eur J Neurosci ; 51(12): 2343-2354, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-30269396

RESUMO

A single phase advance of the light:dark (LD) cycle can temporarily disrupt synchrony of neural circadian rhythms within the suprachiasmatic nucleus (SCN) and between the SCN and peripheral tissues. Compounding this, modern life can involve repeated disruptive light conditions. To model chronic disruption to the circadian system, we exposed male mice to more than a month of a 20-hr light cycle (LD10:10), which mice typically cannot entrain to. Control animals were housed under LD12:12. We measured locomotor activity and body temperature rhythms in vivo, and rhythms of PER2::LUC bioluminescence in SCN and peripheral tissues ex vivo. Unexpectedly, we discovered strong effects of the time of dissection on circadian phase of PER2::LUC bioluminescent rhythms, which varied across tissues. White adipose tissue was strongly reset by dissection, while thymus phase appeared independent of dissection timing. Prior light exposure impacted the SCN, resulting in strong resetting of SCN phase by dissection for mice housed under LD10:10, and weak phase shifts by time of dissection in SCN from control LD12:12 mice. These findings suggest that exposure to circadian disruption may desynchronize SCN neurons, increasing network sensitivity to perturbations. We propose that tissues with a weakened circadian network, such as the SCN under disruptive light conditions, or with little to no coupling, for example, some peripheral tissues, will show increased resetting effects. In particular, exposure to light at inconsistent circadian times on a recurring weekly basis disrupts circadian rhythms and alters sensitivity of the SCN neural pacemaker to dissection time.


Assuntos
Relógios Circadianos , Animais , Ritmo Circadiano , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Núcleo Supraquiasmático/metabolismo
8.
Curr Neurol Neurosci Rep ; 20(12): 55, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-33006677

RESUMO

PURPOSE OF THE REVIEW: Melanin-concentrating hormone (MCH)-expressing neurons located in the lateral hypothalamus are considered as an integral component of sleep-wake circuitry. However, the precise role of MCH neurons in sleep-wake regulation has remained unclear, despite several years of research employing a wide range of techniques. We review recent data on this aspect, which are mostly inconsistent, and propose a novel role for MCH neurons in sleep regulation. RECENT FINDINGS: While almost all studies using "gain-of-function" approaches show an increase in rapid eye movement sleep (or paradoxical sleep; PS), loss-of-function approaches have not shown reductions in PS. Similarly, the reported changes in wakefulness or non-rapid eye movement sleep (slow-wave sleep; SWS) with manipulation of the MCH system using conditional genetic methods are inconsistent. Currently available data do not support a role for MCH neurons in spontaneous sleep-wake but imply a crucial role for them in orchestrating sleep-wake responses to changes in external and internal environments.


Assuntos
Hormônios Hipotalâmicos , Humanos , Hormônios Hipotalâmicos/genética , Melaninas , Neurônios , Hormônios Hipofisários/genética , Sono , Vigília
9.
J Headache Pain ; 21(1): 86, 2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32631251

RESUMO

BACKGROUND: Migraine is a common headache disorder, with cortical spreading depolarization (CSD) considered as the underlying electrophysiological event. CSD is a slowly propagating wave of neuronal and glial depolarization. Sleep disorders are well known risk factors for migraine chronification, and changes in wake-sleep pattern such as sleep deprivation are common migraine triggers. The underlying mechanisms are unknown. As a step towards developing an animal model to study this, we test whether sleep deprivation, a modifiable migraine trigger, enhances CSD susceptibility in rodent models. METHODS: Acute sleep deprivation was achieved using the "gentle handling method", chosen to minimize stress and avoid confounding bias. Sleep deprivation was started with onset of light (diurnal lighting conditions), and assessment of CSD was performed at the end of a 6 h or 12 h sleep deprivation period. The effect of chronic sleep deprivation on CSD was assessed 6 weeks or 12 weeks after lesioning of the hypothalamic ventrolateral preoptic nucleus. All experiments were done in a blinded fashion with respect to sleep status. During 60 min of continuous topical KCl application, we assessed the total number of CSDs, the direct current shift amplitude and duration of the first CSD, the average and cumulative duration of all CSDs, propagation speed, and electrical CSD threshold. RESULTS: Acute sleep deprivation of 6 h (n = 17) or 12 h (n = 11) duration significantly increased CSD frequency compared to controls (17 ± 4 and 18 ± 2, respectively, vs. 14 ± 2 CSDs/hour in controls; p = 0.003 for both), whereas other electrophysiological properties of CSD were unchanged. Acute total sleep deprivation over 12 h but not over 6 h reduced the electrical threshold of CSD compared to controls (p = 0.037 and p = 0.095, respectively). Chronic partial sleep deprivation in contrast did not affect CSD susceptibility in rats. CONCLUSIONS: Acute but not chronic sleep deprivation enhances CSD susceptibility in rodents, possibly underlying its negative impact as a migraine trigger and exacerbating factor. Our findings underscore the importance of CSD as a therapeutic target in migraine and suggest that headache management should identify and treat associated sleep disorders.


Assuntos
Enxaqueca sem Aura/fisiopatologia , Privação do Sono/fisiopatologia , Animais , Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Modelos Animais de Doenças , Masculino , Ratos , Ratos Sprague-Dawley
10.
Neurobiol Dis ; 120: 12-20, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30149182

RESUMO

The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Narcolepsia/metabolismo , Neurônios/fisiologia , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Feminino , Hormônios Hipotalâmicos/antagonistas & inibidores , Hormônios Hipotalâmicos/genética , Masculino , Melaninas/antagonistas & inibidores , Melaninas/genética , Camundongos , Camundongos Knockout , Narcolepsia/genética , Neurônios/efeitos dos fármacos , Piperidinas/farmacologia , Hormônios Hipofisários/antagonistas & inibidores , Hormônios Hipofisários/genética
12.
Cereb Cortex ; 26(4): 1430-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25316334

RESUMO

Lesions of the globus pallidus externa (GPe) produce a profound sleep loss (∼45%) in rats, suggesting that GPe neurons promote sleep. As GPe neuronal activity is enhanced by dopamine (DA) from the substantia nigra pars compacta (SNc), we hypothesized that SNc DA via the GPe promotes sleep. To test this hypothesis, we selectively destroyed the DA afferents to the caudoputamen (CPu) using 6-hydroxydopamine and examined changes in sleep-wake profiles in rats. Rats with 80-90% loss of SNc neurons displayed a significant 33.7% increase in wakefulness (or sleep reduction). This increase significantly correlated with the extent of SNc DA neuron loss. Furthermore, these animals exhibited sleep-wake fragmentation and reduced diurnal variability of sleep. We then optogenetic-stimulated SNc DA terminals in the CPu and found that 20-Hz stimulation from 9 to 10 PM increased total sleep by 69% with high electroencephalograph (EEG) delta power. We finally directly optogenetic-stimulated GPe neurons and found that 20-Hz stimulation of the GPe from 9 to 10 PM increased total sleep by 66% and significantly increased EEG delta power. These findings elucidate a novel circuit for DA control of sleep and the mechanisms of abnormal sleep in BG disorders such as Parkinson's disease and Huntington's disease.


Assuntos
Corpo Estriado/fisiologia , Dopamina/fisiologia , Neurônios Dopaminérgicos/fisiologia , Globo Pálido/fisiologia , Parte Compacta da Substância Negra/fisiologia , Sono , Animais , Masculino , Vias Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Vigília
13.
Sleep Med ; 113: 49-55, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37984017

RESUMO

INTRODUCTION: The basal forebrain (BF) and the medial septum (MS) respectively drive neuronal activity of cerebral cortex and hippocampus (HPC) in sleep-wake cycle. Our previous studies of lesions and neuronal circuit tracing have shown that the pontine parabrachial nucleus (PB) projections to the BF and MS may be a key circuit for cortical and HPC arousal. AIMS: This study aims to demonstrate that PB projections to the BF and MS activate the cerebral cortex and HPC. RESULTS: By using chemogenetic stimulation of the BF, the PB-BF and the PB-MS pathway combined with electroencephalogram (EEG) Fast Fourier Transformation (FFT) analysis in rats, we demonstrated that chemogenetic stimulation of the BF or PB neurons projecting to the BF activated the cerebral cortex while chemogenetic stimulation of the MS or PB neurons projecting to the MS activated HPC activity, in sleep and wake state. These stimulations did not significantly alter sleep-wake amounts. CONCLUSIONS: Our results support that PB projections to the BF and MS specifically regulating cortical and HPC activity.


Assuntos
Prosencéfalo Basal , Núcleos Parabraquiais , Ratos , Animais , Vigília/fisiologia , Prosencéfalo Basal/fisiologia , Nível de Alerta/fisiologia , Eletroencefalografia , Hipocampo
14.
J Neurosci ; 32(50): 17970-6, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23238713

RESUMO

Early transection and stimulation studies suggested the existence of sleep-promoting circuitry in the medullary brainstem, yet the location and identity of the neurons comprising this putative hypnogenic circuitry remains unresolved. In the present study, we sought to uncover the location and identity of medullary neurons that might contribute to the regulation of sleep. Here we show the following in rats: (1) a delimited node of medullary neurons located lateral and dorsal to the facial nerve-a region we termed the parafacial zone (PZ)-project to the wake-promoting medial parabrachial nucleus; (2) PZ neurons express c-Fos after sleep but not after wakefulness and hence are sleep active; and (3) cell-body-specific lesions of the PZ result in large and sustained increases (50%) in daily wakefulness at the expense of slow-wave sleep (SWS). Using transgenic reporter mice [vesicular GABA/glycine transporter (Vgat)-GFP], we then show that >50% of PZ sleep-active neurons are inhibitory (GABAergic/glycinergic, VGAT-positive) in nature. Finally, we used a Cre-expressing adeno-associated viral vector and conditional Vgat(lox/lox) mice to selectively and genetically disrupt GABA/glycinergic neurotransmission from PZ neurons. Disruption of PZ GABAergic/glycinergic neurotransmission resulted in sustained increases (40%) in daily wakefulness at the expense of both SWS and rapid eye movement sleep. These results together reveal the location and neurochemical identity of a delimited node of sleep-active neurons within the rostral medullary brainstem.


Assuntos
Bulbo/citologia , Bulbo/fisiologia , Neurônios/fisiologia , Sono/fisiologia , Animais , Masculino , Camundongos , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley
15.
Aging Brain ; 3: 100068, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36911260

RESUMO

Sleep behavior undergoes significant changes across the lifespan, and aging is associated with marked alterations in sleep amounts and quality. The primary sleep changes in healthy older adults include a shift in sleep timing, reduced slow-wave sleep, and impaired sleep maintenance. However, neurodegenerative and psychiatric disorders are more common among the elderly, which further worsen their sleep health. Irrespective of the cause, insufficient sleep adversely affects various bodily functions including energy metabolism, mood, and cognition. In this review, we will focus on the cognitive changes associated with inadequate sleep during normal aging and the underlying neural mechanisms.

16.
CNS Neurosci Ther ; 29(4): 1192-1196, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36585816

RESUMO

Pontine sub-laterodorsal tegmental nucleus (SLD) is crucial for REM sleep. However, the necessary role of SLD for REM sleep, cataplexy that resembles REM sleep, and emotion memory by REM sleep has remained unclear. To address these questions, we focally ablated SLD neurons using adenoviral diphtheria-toxin (DTA) approach and found that SLD lesions completely eliminated REM sleep accompanied by wake increase, significantly reduced baseline cataplexy amounts by 40% and reward (sucrose) induced cataplexy amounts by 70% and altered cataplexy EEG Fast Fourier Transform (FFT) from REM sleep-like to wake-like in orexin null (OXKO) mice. We then used OXKO animals with absence of REM sleep and OXKO controls and examined elimination of REM sleep in anxiety and fear extinction. Our resulted showed that REM sleep elimination significantly increased anxiety-like behaviors in open field test (OFT), elevated plus maze test (EPM) and defensive aggression and impaired fear extinction. The data indicate that in OXKO mice the SLD is the sole generator for REM sleep; (2) the SLD selectively mediates REM sleep cataplexy (R-cataplexy) that merges with wake cataplexy (W-cataplexy); (3) REM sleep enhances positive emotion (sucrose induced cataplexy) response, reduces negative emotion state (anxiety), and promotes fear extinction.


Assuntos
Cataplexia , Sono REM , Camundongos , Animais , Sono REM/fisiologia , Cataplexia/genética , Extinção Psicológica , Medo , Emoções
17.
CNS Neurosci Ther ; 29(6): 1602-1614, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36794544

RESUMO

AIMS: We often experience dreams of strong irrational and negative emotional contents with postural muscle paralysis during rapid eye movement (REM) sleep, but how REM sleep is generated and its function remain unclear. In this study, we investigate whether the dorsal pontine sub-laterodorsal tegmental nucleus (SLD) is necessary and sufficient for REM sleep and whether REM sleep elimination alters fear memory. METHODS: To investigate whether activation of SLD neurons is sufficient for REM sleep induction, we expressed channelrhodopsin-2 (ChR2) in SLD neurons by bilaterally injecting AAV1-hSyn-ChR2-YFP in rats. We next selectively ablated either glutamatergic or GABAergic neurons from the SLD in mice in order to identify the neuronal subset crucial for REM sleep. We finally  investigated the role of REM sleep in consolidation of fear memory using rat model with complete SLD lesions. RESULTS: We demonstrate the sufficiency of the SLD for REM sleep by showing that photo-activation of ChR2 transfected SLD neurons selectively promotes transitions from non-REM (NREM) sleep to REM sleep in rats. Diphtheria toxin-A (DTA) induced lesions of the SLD in rats or specific deletion of SLD glutamatergic neurons but not GABAergic neurons in mice completely abolish REM sleep, demonstrating the necessity of SLD glutamatergic neurons for REM sleep. We then show that REM sleep elimination by SLD lesions in rats significantly enhances contextual and cued fear memory consolidation by 2.5 and 1.0 folds, respectively, for at least 9 months. Conversely, fear conditioning and fear memory trigger doubled amounts of REM sleep in the following night, and chemo-activation of SLD neurons projecting to the medial septum (MS) selectively enhances hippocampal theta activity in REM sleep; this stimulation immediately after fear acquisition reduces contextual and cued fear memory consolidation by 60% and 30%, respectively. CONCLUSION: SLD glutamatergic neurons generate REM sleep and REM sleep and SLD via the hippocampus particularly down-regulate contextual fear memory.


Assuntos
Medo , Sono REM , Ratos , Camundongos , Animais , Sono REM/fisiologia , Medo/fisiologia , Emoções/fisiologia , Hipocampo , Neurônios GABAérgicos
18.
Metabolism ; 130: 155158, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35150732

RESUMO

BACKGROUND: Nearly 14% of Americans experience chronic circadian disruption due to shift work, increasing their risk of obesity, diabetes, and other cardiometabolic disorders. These disorders are also exacerbated by modern eating habits such as frequent snacking and consumption of high-fat foods. METHODS: We investigated the effects of recurrent circadian disruption (RCD) on glucose metabolism in C57BL/6 mice and in human participants exposed to non-24-h light-dark (LD) schedules vs. those on standard 24-h LD schedules. These LD schedules were designed to induce circadian misalignment between behaviors including rest/activity and fasting/eating with the output of the near-24-h central circadian pacemaker, while minimizing sleep loss, and were maintained for 12 weeks in mice and 3 weeks in humans. We examined interactions of these circadian-disrupted schedules compared to control 24-h schedules with a lower-fat diet (LFD, 13% in mouse and 25-27% in humans) and high-fat diet (HFD, 45% in mouse and 45-50% in humans). We also used young vs. older mice to determine whether they would respond differently to RCD. RESULTS: When combined with a HFD, we found that RCD caused significant weight gain in mice and increased body fat in humans, and significantly impaired glucose tolerance and insulin sensitivity in both mice and humans, but this did not occur when RCD was combined with a LFD. This effect was similar in both young and older mice. CONCLUSION: These results in both humans and a model organism indicate that circadian disruption has an adverse effect on metabolism among individuals eating a high-fat Western-style diet, even in the absence of significant sleep loss, and suggest that reducing dietary fat may protect against the metabolic consequences of a lifestyle (such as shift work) that involves chronic circadian disruption.


Assuntos
Dieta Hiperlipídica , Insulina , Animais , Dieta Hiperlipídica/efeitos adversos , Glucose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia
19.
J Neurosci ; 29(29): 9361-9, 2009 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-19625526

RESUMO

Considerable data support a role for glycinergic ventromedial medulla neurons in the mediation of the postsynaptic inhibition of spinal motoneurons necessary for the motor atonia of rapid-eye movement (REM) sleep in cats. These data are, however, difficult to reconcile with the fact that large lesions of the rostral ventral medulla do not result in loss of REM atonia in rats. In the present study, we sought to clarify which medullary networks in rodents are responsible for REM motor atonia by retrogradely tracing inputs to the spinal ventral horn from the medulla, ablating these medullary sources to determine their effects on REM atonia and using transgenic mice to identify the neurotransmitter(s) involved. Our results reveal a restricted region within the ventromedial medulla, termed here the "supraolivary medulla" (SOM), which contains glutamatergic neurons that project to the spinal ventral horn. Cell-body specific lesions of the SOM resulted in an intermittent loss of muscle atonia, taking the form of exaggerated phasic muscle twitches, during REM sleep. A concomitant reduction in REM sleep time was observed in the SOM-lesioned animals. We next used mice with lox-P modified alleles of either the glutamate or GABA/glycine vesicular transporters to selectively eliminate glutamate or GABA/glycine neurotransmission from SOM neurons. Loss of SOM glutamate release, but not SOM GABA/glycine release, resulted in exaggerated muscle twitches during REM sleep that were similar to those observed after SOM lesions in rats. These findings, together, demonstrate that SOM glutamatergic neurons comprise key elements of the medullary circuitry mediating REM atonia.


Assuntos
Ácido Glutâmico/metabolismo , Bulbo/fisiologia , Tono Muscular/fisiologia , Neurônios/fisiologia , Sono REM/fisiologia , Animais , Glicina/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Movimento/fisiologia , Vias Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Medula Espinal/fisiologia , Fatores de Tempo , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo
20.
Eur J Neurosci ; 31(3): 499-507, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20105243

RESUMO

The basal ganglia (BG) are involved in numerous neurobiological processes that operate on the basis of wakefulness, including motor function, learning, emotion and addictive behaviors. We hypothesized that the BG might play an important role in the regulation of wakefulness. To test this prediction, we made cell body-specific lesions in the striatum and globus pallidus (GP) using ibotenic acid. We found that rats with striatal (caudoputamen) lesions exhibited a 14.95% reduction in wakefulness and robust fragmentation of sleep-wake behavior, i.e. an increased number of state transitions and loss of ultra-long wake bouts (> 120 min). These lesions also resulted in a reduction in the diurnal variation of sleep-wakefulness. On the other hand, lesions of the accumbens core resulted in a 26.72% increase in wakefulness and a reduction in non-rapid eye movement (NREM) sleep bout duration. In addition, rats with accumbens core lesions exhibited excessive digging and scratching. GP lesions also produced a robust increase in wakefulness (45.52%), and frequent sleep-wake transitions and a concomitant decrease in NREM sleep bout duration. Lesions of the subthalamic nucleus or the substantia nigra reticular nucleus produced only minor changes in the amount of sleep-wakefulness and did not alter sleep architecture. Finally, power spectral analysis revealed that lesions of the striatum, accumbens and GP slowed down the cortical electroencephalogram. Collectively, our results suggest that the BG, via a cortico-striato-pallidal loop, are important neural circuitry regulating sleep-wake behaviors and cortical activation.


Assuntos
Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Globo Pálido/fisiologia , Sono/fisiologia , Vigília/fisiologia , Animais , Comportamento Animal/fisiologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/patologia , Eletroencefalografia , Eletromiografia , Agonistas de Aminoácidos Excitatórios/toxicidade , Globo Pálido/efeitos dos fármacos , Globo Pálido/patologia , Ácido Ibotênico/toxicidade , Masculino , Polissonografia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA