这是用户在 2024-4-2 18:14 为 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8602722/ 保存的双语快照页面,由 沉浸式翻译 提供双语支持。了解如何保存?
Skip to main content
跳到主要内容
U.S. flag

An official website of the United States government

Access keys 访问密钥 NCBI Homepage NCBI 主页 MyNCBI Homepage MyNCBI 主页 Main Content 事由 Main Navigation 主导航
2021; 4: 1304.
社区生物学 2021;4: 1304.
Published online 2021 Nov 18. doi: 10.1038/s42003-021-02825-4 IF: 5.9 Q1
2021 年 11 月 18 日在线发布。doi: 10.1038/s42003-021-02825-4IF: 5.9 第一季度
PMCID: PMC8602722 IF: 5.9 Q1
PMCID:PMC8602722IF:5.9 第一季度
PMID: 34795404 IF: 5.9 Q1  PMID: 34795404IF: 5.9 Q1

Role of sleep deprivation in immune-related disease risk and outcomes
睡眠剥夺在免疫相关疾病风险和结局中的作用

Sergio Garbarino,corresponding author1 Paola Lanteri,2 Nicola Luigi Bragazzi,3 Nicola Magnavita,4,5 and Egeria Scoditti6
塞尔吉奥·加巴里诺、 corresponding author 1 保拉·兰特里、 2 尼古拉·路易吉·布拉加齐、 3 尼古拉·马格纳维塔和 4, 5 埃格里亚·斯科迪蒂 6

Associated Data 相关数据

Supplementary Materials 补充材料
Data Availability Statement
数据可用性声明

Abstract 抽象

Modern societies are experiencing an increasing trend of reduced sleep duration, with nocturnal sleeping time below the recommended ranges for health. Epidemiological and laboratory studies have demonstrated detrimental effects of sleep deprivation on health. Sleep exerts an immune-supportive function, promoting host defense against infection and inflammatory insults. Sleep deprivation has been associated with alterations of innate and adaptive immune parameters, leading to a chronic inflammatory state and an increased risk for infectious/inflammatory pathologies, including cardiometabolic, neoplastic, autoimmune and neurodegenerative diseases. Here, we review recent advancements on the immune responses to sleep deprivation as evidenced by experimental and epidemiological studies, the pathophysiology, and the role for the sleep deprivation-induced immune changes in increasing the risk for chronic diseases. Gaps in knowledge and methodological pitfalls still remain. Further understanding of the causal relationship between sleep deprivation and immune deregulation would help to identify individuals at risk for disease and to prevent adverse health outcomes.
现代社会正在经历睡眠时间缩短的趋势,夜间睡眠时间低于健康推荐范围。流行病学和实验室研究表明,睡眠不足对健康有不利影响。睡眠发挥免疫支持功能,促进宿主对感染和炎症性损伤的防御。睡眠不足与先天性和适应性免疫参数的改变有关,导致慢性炎症状态和感染/炎症病理的风险增加,包括心脏代谢、肿瘤、自身免疫性和神经退行性疾病。在这里,我们回顾了实验和流行病学研究、病理生理学以及睡眠剥夺诱导的免疫变化在增加慢性病风险中的作用所证明的对睡眠剥夺的免疫反应的最新进展。知识差距和方法上的缺陷依然存在。进一步了解睡眠剥夺与免疫失调之间的因果关系将有助于识别有疾病风险的个体并防止不良健康结果。

Subject terms: Sleep deprivation, Sleep disorders
主题术语:睡眠剥夺、睡眠障碍

Garbarino et al review recent experimental and epidemiological developments regarding immune responses to sleep deprivation and consider the role for the sleep deprivation induced immune changes in increasing the risk for chronic diseases.
Garbarino等人回顾了最近关于睡眠剥夺免疫反应的实验和流行病学进展,并考虑了睡眠剥夺诱导的免疫变化在增加慢性病风险方面的作用。

Introduction 介绍

Sleep is an active physiological process necessary for life and normally occupying one-third of our lives, playing a fundamental role for physical, mental, and emotional health. Sleep patterns and need are influenced by a complex interplay between chronological age, maturation stage, genetic, behavioral, environmental, and social factors. Adults should sleep a minimum of 7 h per night to promote optimal health,.
睡眠是生命所必需的活跃生理过程,通常占据我们生命的三分之一,对身体、心理和情绪健康 起着重要作用。睡眠模式和需求受到实际年龄、成熟阶段、遗传、行为、环境和社会因素 之间复杂相互作用的影响。成年人每晚应至少睡 7 小时,以促进最佳健康 ,

Besides medical problems including obstructive sleep apnea and insomnia, factors associated mostly with the modern 24/7 society, such as work and social demands, smartphone addiction, and poor diet, contribute to cause the current phenomenon of chronic sleep deprivation, i.e., sleeping less than the recommended amount or, better to say, the intrinsic sleep need.
除了阻塞性睡眠呼吸暂停和失眠等医疗问题外,主要与现代 24/7 社会相关的因素,例如工作和社会需求、智能手机成瘾和不良饮食 习惯,导致当前慢性睡眠剥夺现象,即睡眠少于推荐量,或者更好地说,内在睡眠需求

Sleep deprivation may be categorized as acute or chronic. Acute sleep deprivation refers to no sleep or reduction in the usual total sleep time, usually lasting 1–2 days, with waking time extending beyond the typical 16–18 h. Chronic sleep deprivation is defined by the Third Edition of the International Classification of Sleep Disorders as a disorder characterized by excessive daytime sleepiness caused by routine sleeping less than the amount required for optimal functioning and health maintenance, almost every day for at least 3 months.
睡眠剥夺可分为急性或慢性。急性睡眠剥夺是指没有睡眠或通常的总睡眠时间减少,通常持续 1-2 天,清醒时间超过典型的 16-18 小时。国际睡眠障碍分类第三版将慢性睡眠剥夺定义为一种以日常睡眠少于最佳功能和健康维持所需量而引起的白天过度嗜睡的疾病,几乎每天至少持续 3 个月

Population studies reported a stably increasing prevalence of adults sleeping less than 6 h per night over a long period,,, also affecting children and adolescents,. Sleep duration decline is present not only in high-income and developed countries but also in low-income or racial/ethnic minorities, thus representing a worldwide problem.
人口研究报告称 ,, ,成年人在很长一段时间内每晚睡眠时间少于6小时的患病率稳步上升,这也影响了儿童和青少年 , 。睡眠时间下降不仅存在于高收入和发达国家 ,也存在于低收入或种族/少数民族 中,因此是一个世界性问题。

In addition to fatigue, excessive daytime sleepiness, and impaired cognitive and safety-related performance, sleep deprivation is associated with an increased risk of adverse health outcomes and all-cause mortality. Indeed, epidemiological and experimental data support the association of sleep deprivation with the risk of cardiovascular (CV) (hypertension and coronary artery disease) and metabolic (obesity, type 2 diabetes (T2DM)) diseases. In the United States, sleep deprivation has been linked to 5 of the top 15 leading causes of death including cardio- and cerebrovascular diseases, accidents, T2DM, and hypertension. Data also point to a role for sleep deprivation in the risk of stroke, cancer, and neurodegenerative diseases (NDDs),,. Sleep deprivation is also associated with psychopathological and psychiatric disorders, including negative mood and mood regulation, psychosis, anxiety, suicidal behavior, and the risk for depression.
除了疲劳、白天过度嗜睡以及认知和安全相关表现受损外,睡眠不足还与不良健康结果和全因死亡率 的风险增加有关。事实上,流行病学和实验数据支持睡眠不足与心血管 (CV)(高血压和冠状动脉疾病)和代谢(肥胖、2 型糖尿病 (T2DM))疾病 的风险相关联。在美国,睡眠不足与前 15 大主要死因中的 5 种有关,包括心脑血管疾病、事故、T2DM 和高血压 。数据还指出,睡眠不足在中风、癌症和神经退行性疾病 (NDD) ,, 的风险中起着重要作用。睡眠不足还与精神病理学和精神疾病有关,包括负面情绪和情绪调节、精神病、焦虑、自杀行为和抑郁症 的风险。

Both too short or too long sleep durations have been found to be associated with adverse health outcomes and all-cause mortality with an U-shaped relationship. Although the relation of long sleep duration to adverse health outcomes may be confounded by poor health conditions occurring in older adults, the causal association of sleep deprivation with negative health effects is substantiated by experimental evidence providing biological plausibility,,.
已发现睡眠时间过短或过长都与不良健康结果和全因死亡率有关,呈 U 形关系 。虽然长睡眠时间与不良健康结果的关系可能被老年人的不良健康状况所混淆 ,但睡眠剥夺与负面健康影响的因果关系得到了提供生物学合理性的实验证据的证实 ,,

Sleep profoundly affects endocrine, metabolic, and immune pathways, whose dysfunctions play a determinant role in the development and progression of chronic diseases. Specifically, in many chronic diseases, a deregulated/exacerbated immune response shifts from repair/regulation towards unresolved inflammatory responses.
睡眠深刻影响内分泌、代谢和免疫途径,其功能障碍在慢性疾病 的发展和进展中起着决定性作用。具体来说,在许多慢性疾病中,失调/加剧的免疫反应从修复/调节转变为未解决的炎症反应

Regular sleep is crucial for maintaining immune function integrity and favoring a homeostatic immune defense to microbial or inflammatory insults,. Sleep deprivation may result in deregulated immune responses with increased pro-inflammatory signaling, thus contributing to increase the risk for the onset and/or worsening of infection, as well as inflammation-related chronic diseases.
规律的睡眠对于维持免疫功能完整性和有利于对微生物或炎症损伤的稳态免疫防御至关重要 , 。睡眠不足可能导致免疫反应失调,促炎信号增加,从而增加感染发作和/或恶化的风险,以及与炎症相关的慢性疾病。

Here we reviewed the evidence regarding the impact of sleep deprivation on immune-related diseases by discussing the major points as follows: (1) the immune–sleep relationship; (2) the association of sleep deprivation with the development and/or progression of immune-related chronic diseases; and (3) the immune consequences of sleep deprivation and their implications for diseases. Finally, possible measures to reverse sleep deprivation-associated immune changes were discussed.
在这里,我们回顾了关于睡眠剥夺对免疫相关疾病影响的证据,讨论了以下要点:(1)免疫-睡眠关系;(2)睡眠不足与免疫相关慢性疾病的发展和/或进展的关联;(3)睡眠不足的免疫后果及其对疾病的影响。最后,讨论了逆转睡眠剥夺相关免疫变化的可能措施。

Basic immune mechanisms of sleep regulation
睡眠调节的基本免疫机制

The discovery of muramyl peptide, a bacterial cell wall component that is able to activate the immune system and induce the release of sleep-regulatory cytokines, primary regulators of the inflammatory system, provided the first molecular link between the immune system and sleep. Thereafter, other microbial-derived factors such as the endotoxin lipopolysaccharide (LPS), as well as mediators of inflammation, such as the cytokines interleukin (IL)-1 and tumor necrosis factor (TNF)-α, prostaglandins (PGs), growth hormone-releasing hormone (GHRH), and growth factors, were recognized as sleep-regulating factors.
胞壁肽是一种细菌细胞壁成分,能够激活免疫系统并诱导睡眠调节细胞因子(炎症系统的主要调节因子)的释放,发现了免疫系统和睡眠 之间的第一个分子联系。此后,其他微生物衍生因子如内毒素脂多糖 (LPS) 以及炎症介质,如细胞因子白细胞介素 (IL)-1 和肿瘤坏死因子 (TNF)-α、前列腺素 (PG)、生长激素释放激素 (GHRH) 和生长因子,被认为是睡眠调节因 子。

Along this line, most animal studies have consistently shown a role in particular for IL-1, TNF-α, and PGD2 in the physiologic, homeostatic non-rapid eye movement (NREM) sleep regulation, so that the inhibition of their biological action resulted in decreased spontaneous NREM sleep, whereas their administration enhanced NREM sleep amount and intensity, and suppressed rapid eye movement (REM) sleep. Moreover, the circulating levels of IL-1, IL-6, TNF-α, and PGD2 are highest during sleep. Their effects are dose- and time-of-day-dependent so that, for instance, low doses of IL-1 enhance NREMS, whereas high doses inhibit sleep. Reciprocal effects may be involved in sleep regulation: for instance, the effects of systemic bacterial products such as LPS may also involve TNF-α. Links exist between IL-1β and GHRH/growth hormone (GH) in promoting sleep so that IL-1 induced GH release via GHRH, and hypothalamic γ-aminobutyric acid (GABA)ergic neurons (promoting sleep) are responsive to both GHRH and IL-1β. Instead, anti-inflammatory cytokines, including IL-4, IL-10, and IL-13, inhibited NREM sleep in animal models.
沿着这个思路,大多数动物研究一致表明,IL-1、TNF-α 和 PGD 2 在生理、稳态非快速眼动 (NREM) 睡眠调节中特别起作用,因此抑制它们的生物作用导致自发性 NREM 睡眠减少,而它们的给药增强了 NREM 睡眠量和强度,并抑制了快速眼动 (REM) 睡眠 。此外,IL-1、IL-6、TNF-α 和 PGD 2 的循环水平在睡眠 期间最高。它们的作用与剂量和一天中的时间有关,因此,例如,低剂量的IL-1会增强NREMS,而高剂量会抑制睡眠 。睡眠调节可能涉及相互效应:例如,LPS等全身细菌产物的影响也可能涉及TNF-α 。IL-1β 和 GHRH/生长激素 (GH) 之间存在促进睡眠的联系,因此 IL-1 通过 GHRH 诱导 GH 释放,并且下丘脑γ-氨基丁酸 (GABA) 能神经元(促进睡眠)对 GHRH 和 IL-1β 都有反应 。相反,抗炎细胞因子,包括 IL-4、IL-10 和 IL-13,抑制了动物模型 中的 NREM 睡眠。

Through these substances, the immune system may signal to the brain and interact with other factors involved in sleep regulation such as neurotransmitters (acetylcholine, dopamine, serotonin, norepinephrine, and histamine), neuropeptides (orexin), nucleosides (adenosine), the hormone melatonin, and the hypothalamus-pituitary axis (HPA) axis. Signaling mechanisms to the brain also involve vagal afferents: for instance, vagotomy attenuates intraperitoneal TNF-α-enhanced NREMS responses.
通过这些物质,免疫系统可以向大脑发出信号,并与参与睡眠调节的其他因素相互作用,例如神经递质(乙酰胆碱、多巴胺、血清素、去甲肾上腺素和组胺)、神经肽(食欲素)、核苷(腺苷)、褪黑激素和下丘脑-垂体轴 (HPA) 轴。大脑的信号传导机制也涉及迷走神经传入:例如,迷走神经切开术可减弱腹膜内 TNF-α 增强的 NREMS 反应

Cytokines are produced by a vast array of immune cells, including those resident in the central nervous system (CNS), and non-immune cells, e.g., neurons, astrocytes and microglia, and peripheral tissue cells,. Cytokines interact with the brain through humoral, neural, and cellular pathways, and form a brain cytokine network (Fig. 1) able to produce cytokines, their receptors, and amplify cytokine signals. Peripheral cytokines reach the brain through different non-exclusive mechanisms, including blood–brain barrier (BBB) disruption, penetration of peripheral immune cells, and via afferent nerve fibers, such as the vagus nerve, a bundle of parasympathetic sensory fibers that conveys information from peripheral organs to the CNS.
细胞因子由大量免疫细胞产生,包括驻留在中枢神经系统 (CNS) 中的免疫细胞和非免疫细胞,例如神经元、星形胶质细胞和小胶质细胞以及外周组织细胞 , 。细胞因子通过体液、神经和细胞途径与大脑相互作用,形成脑细胞因子网络(图1),能够产生细胞因子及其受体并放大细胞因子信号 。外周细胞因子通过不同的非排他性机制到达大脑,包括血脑屏障 (BBB) 破坏 、外周免疫细胞的渗透,以及通过传入神经纤维(例如迷走神经),迷走神经是一束副交感神经感觉纤维,将信息从外周器官传递到中枢神经系统

An external file that holds a picture, illustration, etc.
Object name is 42003_2021_2825_Fig1_HTML.jpg
Brain cytokines (CYT, orange circles) network and different suggested routes by which peripherally released inflammatory signals can bypass the blood–brain barrier and circumventricular organs (CVOs), and activate the central nervous system: humoral, cellular, and neural pathways.
脑细胞因子(CYT,橙色圆圈)网络和不同的建议途径,通过这些途径,外周释放的炎症信号可以绕过血脑屏障和脑室周围器官 (CVO),并激活中枢神经系统:体液、细胞和神经通路。

CCL-2: C-C motif chemokine ligand-2; CXCL-10: C-X-C motif chemokine ligand 10; IL-1: interleukin-1; mNTS: medial nucleus tractus solitarius; PGE2: prostaglandin E2; TNF-α: tumor necrosis factor-α.
CCL-2:C-C基序趋化因子配体-2;CXCL-10:C-X-C基序趋化因子配体10;IL-1:白细胞介素-1;mNTS:孤束内侧核;铂族元素 2 :前列腺素E2;TNF-α:肿瘤坏死因子-α。

In the CNS, cytokines mediate a multiplicity of immunological and nonimmunologic biological functions, such as synaptic scaling, synapse formation and elimination, de novo neurogenesis, neuronal apoptosis, brain development, cortical neuron migration, circuit homeostasis and plasticity, and cortical neuron migration, and complex behaviors, sleep, appetite, aging, learning and memory, and mental health status,.
在中枢神经系统中,细胞因子介导多种免疫和非免疫生物学功能 ,如突触缩放、突触形成和消除、从头神经发生、神经元凋亡、大脑发育、皮质神经元迁移 、回路稳态和可塑性 、皮质神经元迁移 、复杂行为、睡眠、食欲、衰老、学习和记忆 以及心理健康状况 ,

A common experimental finding is that after damage to any brain area, if the animal or human survives, sleep always ensues. Recent evidence indicates that sleep is a self-organizing emergent neuronal/glial network property of any viable network regardless of size or location, whether in vivo or in vitro,. Several sleep-regulatory substances, e.g., TNF, IL-1, nitric oxide, PGs, and adenosine are all produced within local cell circuits in response to cell use,.
一个常见的实验发现是,在任何大脑区域受损后,如果动物或人类幸存下来,睡眠总是随之而来 。最近的证据表明,睡眠是任何可行网络的自组织新兴神经元/神经胶质网络特性,无论其大小或位置如何,无论是在体内还是在体外 , 。几种睡眠调节物质,例如 TNF、IL-1、一氧化氮、PG 和腺苷,都是在局部细胞回路内产生的,以响应细胞的使用 ,

From this point of view, TNF-α and IL-1 are closely interconnected and play a similar role in the regulation of sleep. IL-1β and TNF-α self-amplify and increase each other’s mRNA expression in the brain. In rats, IL-1 and TNF-α mRNAs show diurnal variations in different brain areas, with the highest concentrations recorded during increased sleep propensity and peaks occurring at time of sleep period onset in rats and mice.
从这个角度来看,TNF-α和IL-1密切相关,在调节睡眠 中起着相似的作用。IL-1β 和 TNF-α 自我扩增并增加彼此在大脑 中的 mRNA 表达。在大鼠中,IL-1 和TNF-α mRNA在不同的大脑区域显示出昼夜变化,在大鼠和小鼠的睡眠倾向增加期间记录的最高浓度和峰值出现在睡眠期开始时

Sleep-like states in mixed cultures of neurons and glia are dependent in part on the IL-1 receptor accessory protein (AcP),. In the brain, there is an AcP isoform, neuron-specific (AcPb), whose mRNA levels increase with sleep loss,. AcPb is anti-inflammatory, whereas AcP is pro-inflammatory,.
神经元和神经胶质细胞混合培养物中的睡眠样状态部分依赖于 IL-1 受体辅助蛋白 (AcP)。 , 在大脑中,有一种 AcP 亚型,神经元特异性 (AcPb) ,其 mRNA 水平随着睡眠不足 , 而增加。AcPb 具有抗炎作用,而 AcP 具有促炎作用 ,

TNF signaling promotes sleep, whereas reverse TNF-α signaling (the soluble TNF receptor) promotes waking. The brain production of TNF-α is neuron activity-dependent. Afferent activity into the somatosensory cortex enhances TNF expression, and in vitro optogenetic stimulation enhances neuronal expression of TNF immunoreactivity.
TNF 信号转导促进睡眠,而反向 TNF-α 信号转导(可溶性 TNF 受体)促进清醒 。TNF-α的大脑产生是神经元活动依赖 性的。进入躯体感觉皮层的传入活动增强了TNF的表达 ,体外光遗传学刺激增强了TNF免疫反应性的 神经元表达。

Peripheral immune activation following acute or chronic infection or inflammatory diseases is marketed by altered cytokine concentrations and profiles, and is transmitted to the CNS initiating specific adaptive responses. Among these, a sleep response is induced and has been hypothesized to favor recovery from infection and inflammation, supposedly via the timely functional investment of energy into the energy-consuming immune processes,. Accordingly, acute mild immune activation enhances NREM sleep and suppresses REM sleep, whereas severe immune response with an upsurge of cytokine levels causes sleep disturbance with the suppression of both NREM and REM sleep,. This sleep change correlates to the course of the host immune response as observed in bacterial and Trypanosoma infections,. Supportively, the increase in NREM sleep was a favorable prognostic factor for rabbits during infectious diseases.
急性或慢性感染或炎症性疾病后的外周免疫激活通过改变细胞因子浓度和谱进行销售,并传递到中枢神经系统,启动特异性的适应性反应。其中,诱导睡眠反应,并被假设有利于从感染和炎症中恢复,据说是通过及时将能量功能投入到消耗能量的免疫过程中 , 。因此,急性轻度免疫激活可增强 NREM 睡眠并抑制 REM 睡眠,而细胞因子水平激增的严重免疫反应会导致睡眠障碍,同时抑制 NREM 和 REM 睡眠 , 。这种睡眠变化与在细菌和锥虫感染中观察到的宿主免疫反应的过程相关 , 。支持性地,NREM睡眠的增加是兔子在传染病 期间的有利预后因素。

Immune regulators also mediate the complex interrelation between sleep and the circadian systems. Circadian rhythms in behavior and physiology are generated by a molecular clockwork located in the suprachiasmatic nucleus, i.e., the master circadian pacemaker, and peripheral tissues, and involving the so-called clock genes (Clock, Bmals, Npas2, Crys, Pers, Rors, and Rev-erbs). Cytokines, including TNF-α, IL-1β,, and LPS, suppress the peripheral and hypothalamic expression of core clock genes and clock-controlled genes, resulting in reduced locomotor activity accompanied by prolonged rest time.
免疫调节剂还介导睡眠和昼夜节律系统 之间复杂的相互关系。行为和生理学中的昼夜节律是由位于视交叉上核(即主昼夜节律起搏器)和外周组织的分子发条产生的,并涉及所谓的时钟基因(Clock、Bmals、Npas2、Crys、Pers、Rors 和 Rev-erbs)。 细胞因子,包括 TNF-α、IL-1β , 和 LPS ,抑制核心时钟基因和时钟控制基因的外周和下丘脑表达,导致运动活动减少,同时延长休息时间

Sleep deprivation and immune-related disease outcomes
睡眠剥夺和免疫相关疾病结局

In the following section, the association between sleep deprivation and risk or outcomes of immune-related disorders, as observed in human studies (mostly observational) and animal experimentations, will be examined. In this context, considering the sleep–immunity relationship, research has also begun to explore whether and how immune deregulation and inflammation may link sleep deprivation with adverse health outcomes.
在下一节中,将研究在人体研究(主要是观察性)和动物实验中观察到的睡眠剥夺与免疫相关疾病的风险或结果之间的关联。在这种情况下,考虑到睡眠-免疫关系,研究也开始探索免疫失调和炎症是否以及如何将睡眠剥夺与不良健康结果联系起来。

Infection 感染

A breakdown of host defense against microorganisms has been found in sleep-deprived animals, as shown by the increased mortality after septic insult in sleep-deprived mice compared with control mice, or by systemic invasion by opportunistic microorganisms leading to increased morbidity and lethal septicemia in sleep-deprived rats. There is growing evidence associating longer periods of sleep with a substantial reduction in parasitism levels and reduced sleep quality with increased risk of infection and poor infection outcome,. Accordingly, patients with sleep disorders exhibited a 1.23-fold greater risk of herpes zoster than did the comparison cohort. Furthermore, sleep-deprived humans, as those with habitual short sleep (≤5 h) compared with 7–8 h sleep, are more vulnerable to respiratory infections in cross-sectional and prospective studies,, and after an experimental viral challenge,. Similarly, compared with long sleep duration (around 7 h), short sleep duration (around 6 h) is associated with an increased risk of common illnesses, including cold, flu, gastroenteritis, and other common infectious diseases, in adolescents.
在睡眠剥夺的动物中已经发现宿主对微生物的防御崩溃,如与对照小鼠相比,睡眠剥夺小鼠在化脓性损伤后的死亡率增加 ,或者机会性微生物的全身入侵导致睡眠剥夺大鼠的发病率和致死性败血症增加 。越来越多的证据表明,较长的睡眠时间与寄生虫水平 的显着降低和睡眠质量的降低与感染风险增加和感染结果 , 不佳有关。因此,睡眠障碍患者患带状疱疹的风险比对照组高 1.23 倍 。此外,睡眠不足的人,与7-8小时的睡眠相比,睡眠不足的人,作为习惯性睡眠时间短(≤5小时)的人,在横断面和前瞻性研究中 , 更容易受到呼吸道感染,并且在实验性病毒攻击 , 之后。同样,与长睡眠时间(约 7 小时)相比,睡眠时间短(约 6 小时)与青少年患常见疾病的风险增加有关,包括感冒、流感、胃肠炎和其他常见传染病

Compared with non-sleep-deprived mice, REM-sleep-deprived mice failed to control Plasmodium yoelii infection and, consequently, presented a lower survival rate. This was correlated to an impaired T-cell effector activity, characterized by a reduced differentiation of T-helper cells (Th) into Th1 phenotype and following production of pro-inflammatory cytokines, such as interferon (IFN)-γ and TNF-α, and compromised differentiation into T-follicular helper cells (Tfh), essential to B-cell maturation, which therefore resulted to be reduced. Accordingly, both Maf, a Tfh differentiation factor, and T-bet, a pro-Th1 transcription factor, were reduced in the REM-sleep-deprived group. The combination of REM-sleep deprivation and P. yoelii infection resulted in an additive effect on glucocorticoid synthesis, and chemical inhibition of this exacerbated glucocorticoid synthesis reduced parasitemia, death rate, and restored CD4 T-cell, Tfh, and plasma B-cell differentiation in infected sleep-deprived mice, suggesting a role of HPA axis hyperactivation in impairing host immune response under sleep deprivation.
与非睡眠剥夺小鼠相比,REM睡眠剥夺小鼠未能控制约氏疟原虫感染,因此存活率 较低。这与 T 细胞效应活性受损相关,其特征是 T 辅助细胞 (Th) 分化为 Th1 表型的减少,以及促炎细胞因子(如干扰素 (IFN)-γ 和 TNF-α 的产生,以及分化为 T 滤泡辅助细胞 (Tfh) 的受损,这对 B 细胞成熟至关重要,因此导致减少 .因此,在REM睡眠剥夺组 中,Maf(一种Tfh分化因子)和T-bet(一种前Th1转录因子)均减少。REM-睡眠剥夺和约利疟原虫感染的结合导致对糖皮质激素合成的累加作用,并且化学抑制这种加剧的糖皮质激素合成降低了寄生虫血症、死亡率,并恢复了感染睡眠剥夺小鼠的 CD4 T 细胞、Tfh 和血浆 B 细胞分化 ,表明 HPA 轴过度激活在睡眠剥夺下损害宿主免疫反应中的作用。

Seep deprivation may exert detrimental effects on sepsis-induced multi-organ damage. Sleep deprivation (3 days) after LPS administration increased the levels of pro-inflammatory cytokines (IL-6 and TNF-α) in the plasma and organs (lung, liver, and kidney), which could be abrogated by subdiaphragmatic vagotomy or splenectomy 14 days prior to LPS administration. Gut microbiota-vagus nerve axis and gut microbiota-spleen axis may play essential roles in post-septic sleep deprivation-induced aggravation of systemic inflammation and multi-organ injuries.
渗漏剥夺可能对脓毒症诱导的多器官损伤产生不利影响。LPS 给药后睡眠剥夺(3 天)增加了血浆和器官(肺、肝和肾)中促炎细胞因子(IL-6 和 TNF-α)的水平,这可以通过 LPS 给药 前 14 天的膈下迷走神经切开术或脾切除术来消除。肠道菌群-迷走神经轴和肠道菌群-脾轴可能在脓毒症后睡眠剥夺诱导的全身炎症和多器官损伤 加重中起重要作用。

Considering the association between sleep deprivation and immune response to infections, vaccination studies allow to assess the impact of sleep and sleep loss on ongoing immune response and the clinical outcome. Studies in which sleep deprivation (one or few nights) was applied to healthy humans during (mostly after) the immunological challenge of vaccination demonstrate that sleep deprivation reduced both the memory and effector phases of the immune response, as indexed by suppressed antigen-specific antibody and Th cell response compared with undisturbed sleep.
考虑到睡眠剥夺与感染免疫反应之间的关联,疫苗接种研究可以评估睡眠和睡眠不足对持续免疫反应和临床结果的影响。在疫苗接种的免疫挑战期间(主要是在之后)对健康人进行睡眠剥夺(一个或几个晚上)的研究表明,睡眠剥夺降低了免疫反应的记忆和效应阶段,如抑制的抗原特异性抗体和 Th 细胞反应所索引的那样与不受干扰的睡眠 相比。

Congruently, habitual (and hence chronic) short sleep duration (<6 h) compared with longer sleep duration was associated with reduced long-term clinical protection after vaccination against hepatitis B. Sleep deprivation did not exert any impairing effect on mice already immunized. From these studies, it seems that sleep supports—and sleep deprivation impedes—the formation of the immunological memory. Potential mechanisms involved in the beneficial effect of normal sleep on the vaccination response include: (i) the sleep-induced reduction in circulating immune cells that most likely accumulate into lymphatic tissues, increasing the probability to encounter antigens and trigger the immune response; (ii) the sleep-associated profile of inflammatory activation towards Th1 cytokines (increased IL-2, IFN-γ, etc.), which may favor macrophage activation, antigen presentation, and T-cell and B-cell activation; (iii) the effect of sleep stage on the formation of immunological memory through specific immune-active hormones: indeed, during slow wave sleep-rich early sleep, the profile of immune-active hormones, characterized by minimum concentrations of cortisol, endowed with anti-inflammatory activity, and high levels of GH, prolactin, and aldosterone, which support Th1 cell-mediated immunity, may facilitate the mounting of an effective adaptive immune response to a microbial challenge.
同样,与较长的睡眠时间相比,习惯性(因此是慢性)较短的睡眠时间(<6小时)与接种乙 型肝炎疫苗后的长期临床保护作用降低有关。睡眠剥夺对已经免疫的小鼠没有产生任何损害作用 。从这些研究中可以看出,睡眠支持免疫记忆的形成,而睡眠剥夺则阻碍了免疫记忆的形成。正常睡眠对疫苗接种反应的有益影响所涉及的潜在机制包括:(i)睡眠诱导的循环免疫细胞减少,这些免疫细胞最有可能积聚到淋巴组织中,增加遇到抗原和触发免疫反应的可能性;(ii)对Th1细胞因子的炎症激活(IL-2、IFN-γ增加等)的睡眠相关特征,这可能有利于巨噬细胞活化、抗原呈递以及T细胞和B细胞活化;(iii) 睡眠阶段通过特定的免疫活性激素对免疫记忆形成的影响:事实上,在慢波睡眠丰富的早期睡眠期间,免疫活性激素的特征是具有抗炎活性的皮质醇浓度最低,以及支持 Th1 细胞介导的免疫的高水平 GH、催乳素和醛固酮,可能有助于对微生物攻击 产生有效的适应性免疫反应。

Cancer 癌症

Sleep deprivation has increasingly been recognized as a risk factor for impaired anti-tumor response. Epidemiological studies suggest, albeit not consistently, a significant association between short sleep duration and the risk for several cancers, including breast, colorectal, and prostate cancer,. Potential mechanisms underlying this association include a shorter duration of nocturnal secretion of melatonin (putatively due to increased light exposure at night), which exerts anti-cancer properties through antimitotic, antioxidant, apoptotic, anti-estrogenic, and anti-angiogenic mechanisms. Melatonin also plays immunomodulatory and anti-inflammatory effects with relevance for its anti-cancer activity, being able to inhibit the pro-inflammatory nuclear factor-κB (NF-κB)/NLRP3 inflammasome pathways, and to support T/B-cell activation and macrophage function. However, besides melatonin, impaired anti-tumor immune response has been invoked in the sleep deprivation-associated risk for cancer development. A reduced cytotoxic activity of natural killer (NK) cells, which are immune cells with anti-tumor effect, has been reported in 72 h sleep-deprived mice compared with control mice, accompanied by reduced numbers of the cytotoxic cells such as CD8 T cells and NK cells in the tumor microenvironment after chronic sleep deprivation (for 18 h/day during 21 days) in an animal model of experimental pulmonary metastasis,. In this model, the reduced anti-tumor immunity of sleep-deprived animals was also indexed by the reduced number of antigen-presenting cells (dendritic cells) in the lymph nodes, as well as by the decreased effector CD4 T-cell numbers and corresponding cytokine profile (decreased IFN-γ), resulting in lowered Th1 response of Th cells, i.e., the most effective immune response against tumors. Therefore, an immunosuppressive environment develops with sleep deprivation, which could translate into an early onset and increased growth rate of cancer or increased mortality.
睡眠剥夺越来越被认为是抗肿瘤反应受损的危险因素。流行病学研究表明,尽管并非始终如一 但睡眠时间短与多种癌症(包括乳腺癌、结直肠癌和前列腺癌 , )的风险之间存在显着关联。这种关联的潜在机制包括褪黑激素夜间分泌的持续时间较短(推测是由于夜间光照增加), 它通过抗有丝分裂、抗氧化、凋亡、抗雌激素和抗血管生成机制 发挥抗癌特性。褪黑激素还具有与其抗癌活性相关的免疫调节和抗炎作用,能够抑制促炎核因子-κB (NF-κB)/NLRP3 炎症小体通路,并支持 T/B 细胞活化和巨噬细胞功能 。然而,除了褪黑激素之外,抗肿瘤免疫反应受损也被用于与睡眠剥夺相关的癌症发展风险中。据报道,与对照小鼠相比,72小时睡眠剥夺小鼠的自然杀伤(NK)细胞(具有抗肿瘤作用的免疫细胞)的细胞毒性活性降低,伴有细胞毒性细胞(如CD8 T细胞和NK细胞)的数量减少在慢性睡眠剥夺后(21天内18小时/天)在实验性肺转移动物模型 , 中。 在该模型中,睡眠剥夺动物的抗肿瘤免疫力降低也通过淋巴结中抗原呈递细胞(树突状细胞)数量的减少以及效应 CD4 T 细胞数量和相应的细胞因子谱(IFN-γ 降低)来衡量,导致 Th 细胞的 Th1 反应降低,即对肿瘤最有效的免疫反应。因此,免疫抑制环境随着睡眠剥夺而发展,这可能转化为癌症 的早发和生长速度增加或死亡率 增加

An integrated meta-analysis of transcriptomic data showed that circadian rhythm-related genes are downregulated and upregulated in the cortex and hypothalamus samples of mice with sleep deprivation, respectively, with downregulated genes associated with the immune system and upregulated genes associated with oxidative phosphorylation, cancer, and T2DM. Several circadian rhythm-related genes were common to both T2DM and cancer, and seem to associate with malignant transformation and patient outcomes.
转录组学数据的综合荟萃分析显示,在睡眠剥夺小鼠的皮层和下丘脑样本中,昼夜节律相关基因分别下调和上调,与免疫系统相关的基因下调,与氧化磷酸化、癌症和 T2DM 相关的基因上调.几个昼夜节律相关基因在 T2DM 和癌症中都很常见,并且似乎与恶性转化和患者预后 有关。

Hence, although these sleep deprivation-induced immune-mediated mechanisms in cancer warrant further confirmation in humans, the importance of the immune function in the anti-tumor host defense is well recognized, thus suggesting that the impaired immune response after sleep deprivation may represent a plausible mediator of the associated increased risk for cancer as described in animal models and in humans.
因此,尽管这些睡眠剥夺诱导的癌症免疫介导机制值得在人类中得到进一步证实,但免疫功能在抗肿瘤宿主防御中的重要性已得到充分认可 ,从而表明睡眠剥夺后受损的免疫反应可能代表了动物模型和人类中描述的相关癌症风险增加的合理介质。

Neurodegenerative diseases
神经退行性疾病

NDDs are aging-related diseases that selectively target different neuron populations in the CNS, and include Alzheimer’s disease, multiple sclerosis, Parkinson’s disease, Huntington’s disease, and amyotrophic lateral sclerosis. One prevailing hypothesis is that altered sleep habits and specifically sleep deprivation may be a consequence and frequently a marker of the disease. However, human and animal studies have also suggested a causative or contributing role for sleep deprivation in the development and/or worsening of neurodegenerative processes.
NDD 是与衰老相关的疾病,选择性地靶向中枢神经系统中的不同神经元群,包括阿尔茨海默病、多发性硬化症、帕金森病、亨廷顿病和肌萎缩侧索硬化症。一种普遍的假设是,睡眠习惯的改变,特别是睡眠剥夺可能是疾病的结果,并且通常是疾病 的标志。然而,人类和动物研究也表明,睡眠剥夺在神经退行性过程 的发展和/或恶化中起着致病或促成作用。

Potential pathophysiological mechanisms involve, among others, neuro-immune dysregulation. Indeed, a common feature –and a potential therapeutic target- of NDDs is the chronic activation of the immune system, where aspects of peripheral immunity and systemic inflammation integrate with the brain’s immune compartment, leading to neuroinflammation and neuronal damage. Neuroinflammation following sleep deprivation has been studied as a pathogenic mechanism potentially mediating the association between sleep deprivation and neurodegenerative processes. Low-grade neuroinflammation as indexed by heightened levels of pro-inflammatory mediators (e.g., TNF-α, IL-1β, and COX-2) and activation of astrocytes and microglia, main immune cells in the brain, was observed in the hippocampus and piriform cortex regions of the brain of chronic sleep-deprived rats along with neurobehavioral alterations (anxiety, learning, and memory impairments). The sleep deprivation pro-inflammatory milieu was accompanied by oxidative stress in the brain and BBB disruption with consequent increased permeability to blood components. After acute sleep deprivation, there was a significant increased recruitment of B cells in the mouse brain, which could be important given evidence of B cells involvement in NDDs.
潜在的病理生理机制包括神经免疫失调等。事实上,NDD的一个共同特征和潜在的治疗靶点是免疫系统的慢性激活,其中外周免疫和全身炎症的各个方面与大脑的免疫区室整合,导致神经炎症和神经元损伤 。睡眠剥夺后的神经炎症已被研究为一种致病机制,可能介导睡眠剥夺与神经退行性过程之间的关联。在慢性睡眠剥夺大鼠大脑的海马体和梨状皮层区域观察到促炎介质(例如 TNF-α、IL-1β 和 COX-2)水平升高以及星形胶质细胞和小胶质细胞(大脑中的主要免疫细胞)的激活以及神经行为改变(焦虑、学习和记忆障碍)。 睡眠剥夺促炎环境伴随着大脑 中的氧化应激和血脑屏障破坏,从而增加对血液成分 的通透性。在急性睡眠剥夺后,小鼠大脑中B细胞的募集显着增加,鉴于B细胞参与NDD的证据 ,这可能很重要。

Progressive and chronic aggregations of unique proteins in the brain and spinal cord are hallmarks of NDDs and trigger inflammatory responses, gradual loss of physiological functions of the nerve cells, and cell death. Impaired autophagy in humans, a catabolic process of cytoplasmic components, contributes to the aggregation and accumulation of β-amyloid (Aβ), cytoskeleton-related protein τ, and synuclein in neuronal cells and tissues. Sleep plays an important role in the clearance of metabolic waste products accumulated during wakefulness and neural activity. Indeed, the Aβ protein is predominantly cleared from the brain during sleep, possibly through the glymphatic pathway. Congruently, acute and chronic experimental sleep deprivation in animals, and humans resulted in brain Aβ accumulation and plaque formation, a typical pathological change in Alzheimer’s disease process, the most common type of dementia. Imaging studies have revealed that healthy humans with self-reported short sleep were more prone to have cerebral Aβ plaque pathology and disruption of deep sleep (slow wave sleep) increases Aβ in human cerebrospinal fluid (CSF). Likewise, patients with insomnia present higher CSF levels of Aβ.
大脑和脊髓中独特蛋白质的进行性和慢性聚集是 NDD 的标志, 并引发炎症反应、神经细胞生理功能逐渐丧失和细胞死亡 。人类自噬受损是细胞质成分的分解代谢过程,有助于神经元细胞和组织中β-淀粉样蛋白 (Aβ)、细胞骨架相关蛋白 τ 和突触核蛋白的聚集和积累 。睡眠在清除清醒和神经活动期间积累的代谢废物方面起着重要作用。事实上,Aβ蛋白主要在睡眠期间从大脑中清除,可能是通过淋巴途径。同样,动物 , 和人类的 急性和慢性实验性睡眠剥夺导致大脑Aβ积累和斑块形成,这是阿尔茨海默病过程中的典型病理变化,是最常见的痴呆类型。影像学研究表明,自我报告睡眠时间短的健康人更容易出现脑 Aβ 斑块病变 ,深度睡眠中断(慢波睡眠)会增加人脑脊液 (CSF) 中的 Aβ。同样,失眠患者的脑脊液 Aβ 水平也较高

This pathological Aβ accumulation might reflect disrupted balance of Aβ production and clearance after sleep deprivation. On the one hand, sleep deprivation results in reduced clearance as suggested by clinical studies showing that Aβ levels in CSF are the highest before sleep and the lowest after wakening, whereas Aβ clearance from CSF was impaired by sleep deprivation. Impaired clearance might also derive from disrupted peripheral Aβ transport, as suggested by the sleep deprivation-induced downregulation of low-density lipoprotein receptor-related protein-1 (LRP-1), which promotes Aβ efflux from the brain to the peripheral circulation across the BBB, and elevations of receptor of advanced glycation end products (RAGE), which promotes on the contrary the influx of peripheral Aβ into the brain, thus preventing Aβ clearance. On the other hand, apart from impairing Aβ and τ interstitial fluid clearance, sleep deprivation may also have a role in increasing Aβ and τ exocytosis, thereby increasing CSF Aβ and τ levels. In animals, sleep deprivation also leads to upregulation of β-secretase 1 (BACE-1), the most important enzyme regulating Aβ generation in the brain,,, thus opening the hypothesis of increased Aβ production by sleep deprivation. Sleep deprivation-induced neuroinflammatory mediators correlate and could lead to disturbed Aβ clearance and stimulated amyloidogenic pathway, being pro-inflammatory cytokines able to suppress the expression of LRP-1 and to increase RAGE and BACE-1 levels. Likewise, oxidative stress induced by sleep deprivation may also contribute to the neuroinflammatory burden and the increased expression of BACE-1. Furthermore, patients with insomnia, compared with healthy controls, showed decreased serum levels of neurotrophins, including brain-derived neurotrophic factor (BDNF), proteins especially relevant in neuroplasticity, memory and sleep, and this reduction was significantly related to the insomnia severity.
这种病理性 Aβ 积累可能反映了睡眠剥夺后 Aβ 产生和清除平衡的破坏。一方面,睡眠剥夺导致清除率降低,临床研究表明,脑脊液中的Aβ水平在睡眠前最高,醒来后最低,而脑脊液中的Aβ清除率因睡眠剥夺而受损 。清除受损也可能源于外周 Aβ 转运中断,如睡眠剥夺诱导的低密度脂蛋白受体相关蛋白-1 (LRP-1) 下调,其促进 Aβ 从大脑流出到整个 BBB 的外周循环,以及晚期糖基化终产物受体 (RAGE) 的升高,这恰恰促进外周 Aβ 流入大脑, 从而阻止 Aβ 清除 。另一方面,除了损害 Aβ 和 τ 间质液清除外,睡眠剥夺还可能增加 Aβ 和 τ 胞吐作用,从而增加 CSF Aβ 和 τ 水平 。在动物中,睡眠剥夺也会导致β分泌酶1(BACE-1)的上调,BACE-1是大脑 ,, 中调节Aβ生成的最重要酶,从而开启了睡眠剥夺增加Aβ产生的假设。睡眠剥夺诱导的神经炎症介质相互关联,并可能导致 Aβ 清除紊乱和淀粉样蛋白生成途径受刺激 ,是能够抑制 LRP-1 表达并增加 RAGE 和 BACE-1 水平 的促炎细胞因子。同样,睡眠剥夺引起的氧化应激也可能导致神经炎症负荷和 BACE-1 表达增加 。 此外,与健康对照组相比,失眠患者的血清神经营养因子水平降低,包括脑源性神经营养因子 (BDNF),这些蛋白质与神经可塑性、记忆和睡眠特别相关,并且这种降低与失眠严重程度 显着相关。

Sleep deprivation is associated with a rapid decline in circulatory melatonin levels, which may be linked to rapid consumption of melatonin as a first-line defense against the sleep deprivation-associated rise in oxidative stress. Melatonin is a potent antioxidant, interacts with BDNF, and promotes neurogenesis and inhibits apoptosis. The neuroprotective potential of melatonin can target events leading to Alzheimer’s disease development including Aβ pathology, τ hyperphosphorylation, oxidative stress, glutamate excitotoxicity, and calcium dyshomeostasis,. Accordingly, melatonin treatment could restore the autophagy flux, thereby preventing tauopathy and cognitive decline in Alzheimer’s disease mice.
睡眠剥夺与循环褪黑激素水平的快速下降有关,这可能与褪黑激素的快速消耗有关,褪黑激素是抵御睡眠剥夺相关氧化应激 上升的第一道防线。褪黑激素是一种有效的抗氧化剂,与BDNF 相互作用,促进神经发生,抑制细胞凋亡 。褪黑激素的神经保护潜力可以靶向导致阿尔茨海默病发展的事件,包括 Aβ 病理学、τ 过度磷酸化、氧化应激、谷氨酸兴奋性毒性和钙稳态失调 , 。因此,褪黑激素治疗可以恢复自噬通量,从而防止阿尔茨海默病小鼠的tau蛋白病和认知能力下降

Patients with Alzheimer’s disease have an increased incidence of sleep-disordered breathing. In addition, sleep-disordered breathing is associated with an increased risk of mild cognitive impairment or dementia and with earlier onset of Alzheimer’s disease. Sleep-disordered breathing is also associated with altered levels of Alzheimer’s disease biomarkers in CSF, including decreased levels of Aβ and elevated levels of phosphorylated τ. Sleep-disordered breathing possibly via hypoxia, inflammation, and sleep disruption/deprivation could contribute to Alzheimer’s disease processes, e.g., increase of Aβ production and aggregation, suppression of glymphatic clearance of Alzheimer’s disease pathogenic proteins (τ, Aβ) and oxidative stress, inflammation, and synaptic damage,.
阿尔茨海默病患者睡眠呼吸 障碍的发生率增加。此外,睡眠呼吸障碍与轻度认知障碍或痴呆的风险增加以及阿尔茨海默病 的早期发作有关。睡眠呼吸障碍还与脑脊液中阿尔茨海默病生物标志物水平的改变有关,包括 Aβ 水平降低和磷酸化 τ 水平升高 。可能通过缺氧、炎症和睡眠中断/剥夺引起的睡眠呼吸障碍可能导致阿尔茨海默病过程,例如,增加 Aβ 的产生和聚集、抑制阿尔茨海默病致病蛋白(τ、Aβ)的淋巴清除以及氧化应激、炎症和突触损伤 ,

To summarize, the sleep deprivation-associated risk for Alzheimer’s disease could be linked to the induction of inflammation in the brain and disorders of systemic innate and adaptive immunity. However, the relationship of sleep deprivation to inflammation in Alzheimer’s disease is mostly speculative and needs to be confirmed.
总而言之,阿尔茨海默病的睡眠剥夺相关风险可能与大脑炎症的诱导以及全身先天免疫和适应性免疫的紊乱有关 。然而,睡眠剥夺与阿尔茨海默病炎症的关系大多是推测性的,需要确认。

Similar to Aβ in Alzheimer’s disease, abnormal levels of α-synuclein are common to Parkinson’s disease, the second most common NDDs. Sleep disturbances are not only a common comorbidity in Parkinson’s disease, but often precede the onset of classic motor symptoms. The main pathological features of Parkinson’s disease are the reduction of dopaminergic neurons in the extrapyramidal nigrostriatal body and the formation of Lewy bodies formed by the aggregation of α-synuclein and its oligomers surrounded by neurofilaments. Due to the degeneration of the dopaminergic neurons, affected people show muscle stiffness, resting tremors, and posture instability; other pathways involved in sleep, cognition, mental abnormalities, and other non-motor symptoms are also affected. Epidemiological studies also suggest that disturbed sleep may increase the risk of Parkinson’s disease,. Such disease-modifying mechanisms may include activation of inflammatory and immune pathways, abnormal proteostasis, changes in glymphatic clearance, and altered modulation of specific sleep neural circuits that may prime further propagation of α-synucleinopathy in the brain. Melatonin could reduce neurotoxin-induced α-synuclein aggregation in mice. Furthermore, melatonin pretreatment reduced neurotoxin-induced loss of axon and dendritic length in dopaminergic neurons through suppression of autophagy activated by CDK5 and α-synuclein aggregation, thereby reducing dyskinesia symptoms in Parkinson’s disease animal models. A few reports have shown that melatonin exerts protective effects in several experimental models of Parkinson’s disease.
与阿尔茨海默病中的 Aβ 类似,α-突触核蛋白水平异常在帕金森病中很常见,帕金森病是第二常见的 NDD 。睡眠障碍不仅是帕金森病的常见合并症,而且通常先于经典运动症状 的发作。帕金森病的主要病理特征是锥体外系纹状体中多巴胺能神经元的减少以及由α-突触核蛋白及其被神经丝包围的寡聚体聚集形成的路易体的形成。由于多巴胺能神经元的退化,受影响的人表现出肌肉僵硬、静止性震颤和姿势不稳定;涉及睡眠、认知、精神异常和其他非运动症状的其他途径也会受到影响 。流行病学研究还表明,睡眠紊乱可能会增加患帕金森病 , 的风险。这种疾病改变机制可能包括炎症和免疫通路的激活、异常的蛋白稳态、淋巴清除的变化以及特定睡眠神经回路的改变,这些可能引发α突触核蛋白病在大脑 中的进一步传播。褪黑激素可以减少小鼠神经毒素诱导的α-突触核蛋白聚集。此外,褪黑激素预处理通过抑制 CDK5 和 α-突触核蛋白聚集激活的自噬,减少了神经毒素诱导的多巴胺能神经元轴突和树突长度的损失,从而减少了帕金森病动物模型 中的运动障碍症状。一些报告表明,褪黑激素在帕金森病 的几种实验模型中发挥保护作用。

However, although animal experimentations suggest a link between sleep deprivation and immune dysfunction in neurodegenerative processes, no human investigations have yet confirmed the mediating role of immune dysregulation in the association between sleep deprivation and risk or outcomes of NDDs.
然而,尽管动物实验表明睡眠剥夺与神经退行性过程中的免疫功能障碍之间存在联系,但尚未有人体研究证实免疫失调在睡眠剥夺与NDDs风险或结果之间的关联中的中介作用。

Autoimmune diseases 自身免疫性疾病

Sleep disturbances are frequently reported in autoimmune diseases, and immunotherapy in patients with autoimmune pathologies results in sleep improvement. However, knowledge of the immunopathology of autoimmune diseases have disclosed new concepts on the impact of sleep deprivation on autoimmune disease process, showing that sleep deprivation can promote a breakdown of immunologic self-tolerance. Human cohort studies found that non-apnea sleep disorders, including insomnia, were associated with a higher risk of developing autoimmune diseases such as rheumatoid arthritis, ankylosing spondylitis, systemic lupus erythematosus, and systemic sclerosis (adjusted hazard ratio: 1.47, 95% confidence interval (CI) 1.41–1.53) Similarly, in relatives of systemic lupus erythematosus patients, and hence at increased risk for systemic lupus erythematosus, self-reported short sleep duration (<7 h/night) was associated with transitioning to systemic lupus erythematosus (adjusted odds ratio: 2.0, 95% CI 1.1–4.2), independent of early preclinical features that may influence sleep duration such as prednisone use, depression, chronic fatigue, and vitamin D deficiency. This role of sleep deprivation as a risk factor for autoimmune diseases is corroborated by animal studies. In mice genetically predisposed to develop systemic lupus erythematosus, chronic sleep deprivation, applied at an age when animals were yet clinically healthy, caused an early onset of the disease, as indexed by the increased number of antinuclear antibodies, without affecting disease course or severity, according to data on proteinuria, a surrogate marker of autoimmune nephritis, and longevity. Several mechanisms have been postulated to explain the link between sleep deprivation and autoimmune disease risk. Sleep deprivation can accelerate disease development through mechanisms including sleep deprivation-induced increased production of several pro-inflammatory cytokines,, as better discussed below. Indeed, cytokines are synergistically involved in the pathogenesis of autoimmunity, such as IL-6, whose abnormal production results in polyclonal B-cell activation and the occurrence of autoimmune features, and IL-17 and the related Th17-cell response, which require IL-6 for activation and can cause greater amounts of autoantibody production and immune complex formation, or can intensify chronic inflammation by promoting angiogenesis and recruiting of inflammatory cells at inflammation sites as well as cartilage and bone erosion. Furthermore, experimentally sleep-deprived healthy humans showed impaired suppressive activity of CD4 regulatory T cells (Treg), which normally is highest during the night and lowest in the morning. The suppressive function of Treg towards excessive immune response is an important homeostatic mechanism, whose impairment is implicated in autoimmune disease pathogenesis. Hence, sleep deprivation may not be merely an early symptom or a consequence of an autoimmune disease, but may contribute directly to the pathogenesis increasing the susceptibility to develop an autoimmune disease. More studies are warranted in this field.
睡眠障碍在自身免疫性疾病中经常有报道,自身免疫性病变患者的免疫疗法可改善 睡眠。然而,自身免疫性疾病的免疫病理学知识揭示了睡眠剥夺对自身免疫性疾病过程影响的新概念,表明睡眠剥夺可以促进免疫自我耐受的崩溃。人类队列研究发现,非呼吸暂停睡眠障碍(包括失眠)与发生类风湿性关节炎、强直性脊柱炎、系统性红斑狼疮和系统性硬化症等自身免疫性疾病的风险较高有关(校正风险比:1.47,95% 置信区间 (CI) 1.41–1.53) 同样,在系统性红斑狼疮患者的亲属中,因此系统性红斑狼疮的风险增加, 自我报告的睡眠时间短(<7 小时/晚)与过渡到系统性红斑狼疮有关(校正比值比:2.0,95% CI 1.1-4.2),与可能影响睡眠持续时间的早期临床前特征无关,例如泼尼松使用、抑郁、慢性疲劳和维生素 D 缺乏 。动物研究证实了睡眠剥夺作为自身免疫性疾病危险因素的这种作用。在遗传易患系统性红斑狼疮的小鼠中 ,在动物临床健康时应用慢性睡眠剥夺会导致疾病的早期发作,如抗核抗体数量的增加所标记的那样,不影响病程或严重程度,根据蛋白尿(自身免疫性肾炎的替代标志物)和长寿的数据。 已经假设了几种机制来解释睡眠剥夺与自身免疫性疾病风险之间的联系。睡眠剥夺可以通过包括睡眠剥夺诱导的几种促炎细胞因子的产生增加在内的机制加速疾病的发展 , ,如下所述。事实上,细胞因子协同参与自身免疫的发病机制,例如IL-6,其异常产生导致多克隆B细胞活化和自身免疫特征 的发生,以及IL-17和相关的Th17细胞反应 ,需要IL-6激活 ,可以引起更多的自身抗体产生和免疫复合物的形成,或者可以通过促进血管生成和炎症募集来加剧慢性炎症炎症部位的细胞以及软骨和骨侵蚀 。此外,实验中睡眠不足的健康人表现出CD4调节性T细胞(Treg)的抑制活性受损,通常在夜间最高,在早晨 最低。Treg对过度免疫反应的抑制功能是一种重要的稳态机制,其损伤与自身免疫性疾病的发病机制有关 。因此,睡眠不足可能不仅仅是自身免疫性疾病的早期症状或后果,而是可能直接导致发病机制增加患自身免疫性疾病的易感性。在这一领域需要更多的研究。

Metabolic and vascular diseases
代谢和血管疾病

Prospective epidemiological evidence associate sleep deprivation (commonly <7 h/night, often <5 h/night) with the incidence of fatal and non-fatal CV outcomes, with a 48% higher risk of coronary heart disease, a 15% higher risk of stroke, and a 12% increased risk of all-cause mortality, which is mainly due to CV causes, according to some authors. In a recent prospective cohort, a low-stable sleep pattern (<5 h sleep/night) during the 4-year follow-up had the highest risk of death and CV events. Short sleep has also been associated with increased subclinical atherosclerotic burden, the dominant underlying cause of CV diseases.
前瞻性流行病学证据将睡眠剥夺(通常为 <7 小时/晚,通常<5 小时/晚)与致命和非致命性 CV 结果的发生率相关联,冠心病风险增加 48 %,中风 风险增加 15%,全因死亡 风险增加 12%,这主要是由于 CV 原因,根据一些作者 的说法。在最近的一项前瞻性队列中,在 4 年随访期间,低稳定性睡眠模式(<5 小时睡眠/夜间)的死亡和 CV 事件 风险最高。睡眠不足也与亚临床动脉粥样硬化负荷增加有关,亚临床动脉粥样硬化负担是心血管疾病 的主要原因。

In addition, sleep deprivation increases the risk for obesity (about 55% higher risk),, insulin resistance, T2DM (28% higher risk), and hypertension (21% higher risk), which are powerful and preventable risk factors for CV diseases. Notably, the risk for diabetes attributable to sleep deprivation is comparable to that of other established traditional cardiometabolic risk factors, thus underscoring the clinical significance of targeting sleep deprivation in the prevention of cardiometabolic diseases. In contrast with normal nocturnal sleep and in particular NREM sleep characterized by a marked decrease in sympathetic activity, catecholamine plasma levels, and blood pressure, experimental sleep deprivation (acute or chronic) is accompanied by increased sympathetic outflow, with consequent higher blood pressure and heart rate, thus providing a pathogenic link between sleep deprivation and hypertension risk.
此外,睡眠不足会增加肥胖(风险增加约 55%) , 、胰岛素抵抗、T2DM(风险增加 28%) 和高血压(风险增加 21%) 的风险,这些都是心血管疾病的强大且可预防的危险因素。值得注意的是,睡眠剥夺导致的糖尿病风险与其他已确定的传统心脏代谢危险因素 相当,从而强调了针对睡眠剥夺在预防心脏代谢疾病方面的临床意义。与正常的夜间睡眠,特别是以交感神经活动、儿茶酚胺血浆水平和血压显着降低为特征的 NREM 睡眠相比,实验性睡眠剥夺(急性或慢性)伴随着交感神经流出增加,从而导致血压和心率升高,从而在睡眠剥夺和高血压风险 之间提供了致病联系。

Regarding the influence of sleep deprivation on metabolic pathways, studies support a plausible causal link between sleep deprivation and the risk of overweight and obesity, possibly mediated by the effect of sleep deprivation on circulating levels of hormones (leptin, ghrelin) controlling hunger, satiety and energy balance, besides other factors intervening during sleep deprivation, including physical inactivity and overfeeding. Furthermore, human experimental evidence with chronic sleep deprivation protocol demonstrate that sleep deprivation may alter glucose metabolism and insulin sensitivity, thus increasing the risk for obesity and T2DM. The reduction in total body insulin sensitivity observed after sleep deprivation (4.5 h per night for 4 days) in healthy subjects was paralleled by impaired peripheral insulin sensitivity, as demonstrated in subcutaneous fat playing a pivotal role in energy metabolism. Considering a more chronic sleep deprivation, reduced insulin sensitivity was reported in overweight adults after 14 days of experimental sleep deprivation (5.5 h per night) compared with 8.5 h per night of sleep, and after habitual curtailment in sleep duration of 1.5 h (<6 h of sleep per night) in healthy young adults with a family history of T2DM.
关于睡眠剥夺对代谢途径的影响,研究支持睡眠剥夺与超重和肥胖风险之间存在合理的因果关系,可能是由睡眠剥夺对控制饥饿、饱腹感和能量平衡的激素循环水平(瘦素、生长素释放肽)的影响介导的,此外还有睡眠剥夺期间干预的其他因素,包括缺乏身体活动和过度喂养 .此外,具有慢性睡眠剥夺方案的人体实验证据表明,睡眠剥夺可能会改变葡萄糖代谢 和胰岛素敏感性 ,从而增加肥胖和 T2DM 的风险。在健康受试者中睡眠剥夺(每晚 4.5 小时,持续 4 天)后观察到的全身胰岛素敏感性降低与外周胰岛素敏感性受损并行,如皮下脂肪在能量代谢中起关键作用所证明 的那样。考虑到更长期的睡眠剥夺,据报道,在实验性睡眠剥夺 14 天(每晚 5.5 小时)后,超重成年人的胰岛素敏感性降低,而每晚睡眠 8.5 小时,并且在有 T2DM 家族史的健康年轻人中,习惯性地缩短睡眠时间 1.5 小时(每晚 <6 小时)。

Although the mechanisms that underlie most associations between short sleep duration and adverse cardiometabolic outcomes are not fully understood, potential causative mechanisms involving immune-inflammatory activation have been postulated. It is indeed well established that the subclinical inflammatory status induced by sleep deprivation has pathogenic implications for metabolic and CV risk factors (glucose metabolism, diabetes, hypertension, atherogenic lipid profile, endothelial dysfunction, and coronary calcification) and outcomes (stroke and coronary heart disease). Accordingly, most of the markers of systemic and cellular inflammation (leukocyte counts and activation state, cytokines, acute-phase proteins, and adipose tissue-derived adipokines) found to be altered after sleep deprivation have been epidemiologically and pathogenically associated with insulin resistance, T2DM, and vascular complications. In fact, inflammation is an early pathogenic process during the development of obesity and insulin resistance. Many adipose tissue-released inflammatory factors with pro-atherogenic and pro-thrombotic actions have also been regarded as a molecular link between obesity and atherosclerotic CV diseases. Furthermore, chronic inflammatory processes are firmly established as central to the development and clinical complications of CV diseases, form the initiation, promotion and progression of atherosclerotic lesions to plaque instability, and the precipitation of thrombosis, the main underlying cause of myocardial infarction or stroke. Most CV risk factors (adiposity, insulin resistance, T2DM, hypertension, and dyslipidemia) act by inducing or intensifying such underlying inflammatory processes that ultimately promote endothelial dysfunction, altered vascular reactivity, innate and adaptive immune system activation, leukocyte infiltration into the vessel wall, and thus atherogenesis. Experimental sleep deprivation leads to endothelial dysfunction, an early marker of atherosclerosis, as indexed by impaired endothelial-dependent vasodilation or increased levels of endothelial adhesion molecules.
尽管短睡眠时间与不良心脏代谢结局之间大多数关联的机制尚不完全清楚,但已经假设了涉及免疫炎症激活的潜在致病机制。确实可以确定,睡眠剥夺引起的亚临床炎症状态对代谢和 CV 危险因素(葡萄糖代谢、糖尿病、高血压、动脉粥样硬化血脂谱、内皮功能障碍和冠状动脉钙化)和结果(中风和冠心病) 具有致病意义。因此,睡眠剥夺后发现的大多数全身和细胞炎症标志物(白细胞计数和活化状态、细胞因子、急性期蛋白和脂肪组织来源的脂肪因子)在流行病学和发病学上与胰岛素抵抗、T2DM 和血管并发症相关 。事实上,炎症是肥胖和胰岛素抵抗 发展过程中的早期致病过程。许多具有促动脉粥样硬化和促血栓形成作用的脂肪组织释放的炎症因子也被认为是肥胖与动脉粥样硬化性 CV 疾病 之间的分子联系。此外,慢性炎症过程被牢固地确立为心血管疾病发展和临床并发症的核心,形成动脉粥样硬化病变的开始、促进和进展,导致斑块不稳定,以及血栓形成的沉淀,血栓形成是心肌梗死或中风的主要原因。 大多数心血管危险因素(肥胖、胰岛素抵抗、T2DM、高血压和血脂异常)通过诱导或加剧这些潜在的炎症过程起作用,最终促进内皮功能障碍、血管反应性改变、先天性和适应性免疫系统激活、白细胞浸润到血管壁,从而促进动脉粥样硬化形成 .实验性睡眠剥夺导致内皮功能障碍,这是动脉粥样硬化的早期标志物,以内皮依赖性血管舒张受损或内皮粘附分子水平升高为指标

Among the inflammatory markers, besides being a biomarker of future risk for CV diseases and a predictor of clinical response to statin therapy, C-reactiove protein (CRP) has been shown to be involved in the immunologic process that triggers vascular remodeling and atherosclerotic plaque deposition. CRP levels lack diurnal rhythm and its liver production is stimulated by cytokines including IL-6 and IL-17, which are upregulated by sleep deprivation. As such, although limited evidence have found an elevation of circulating CRP following sleep deprivation, CRP is a prototypical inflammatory factor with the potential to mark and—to some extent mediate—CV risk following sleep deprivation. Congruently, elevated and sustained plasma levels of CRP have been observed in healthy humans after prolonged sleep deprivation (5 or 10 nights), in concomitance with increased heart rate,, lymphocyte pro-inflammatory activation, and production of cytokines (e.g., IL-1, IL-6, and IL-17). Similarly, the increase in blood pressure and heart rate observed after acute total sleep deprivation (40 h) was accompanied and even preceded by impaired vasodilation and by increased levels of IL-6 and markers of endothelial dysfunction and activation, such as cellular adhesion molecules (E-selectin, ICAM-1, etc.). The sleep deprivation pro-atherogenic effect in animal model of sleep fragmentation is mediated, at least in part, by reduced hypothalamic release of hypocretin (i.e., orexin), a wake-inducing neuropeptide, which limits the production of leukocytes (monocytes and neutrophils) and atherosclerosis development, and has been inversely associated with the risk of myocardial infarction, heart failure, and obesity. The activation of the sympathetic nervous system (SNS) may be another mechanism for the inflammatory link between sleep loss and atherosclerotic CV disease, because such activation increases the bone marrow release of progenitor cells, the production of innate immune cells (monocytes), and the levels of inflammatory cytokines, and triggers endothelial dysfunction, thereby leading to systemic and vascular inflammation and atherosclerosis,. Playing a key role in instigating inflammatory responses and promoting atherosclerosis, the sleep deprivation-associated oxidative stress may also contribute to CV risk. It has also been hypothesized a role for melatonin suppression following sleep deprivation in the vascular impairment associated with sleep deprivation, given that melatonin inhibits oxidative stress and cytokine production by immune and vascular cells, and represses atherosclerotic lesion formation in vivo.
在炎症标志物中,除了作为未来心血管疾病风险的生物标志物和他汀类药物治疗 临床反应的预测因子外,C-反应蛋白(CRP)已被证明参与触发血管重塑和动脉粥样硬化斑块沉积 的免疫过程。CRP 水平缺乏昼夜节律,其肝脏产生受到细胞因子的刺激,包括 IL-6 和 IL-17,这些细胞因子因睡眠剥夺而上调 。因此,尽管有限的证据表明睡眠剥夺后循环 CRP 升高 ,但 CRP 是一种典型的炎症因子,有可能标记并在一定程度上介导睡眠剥夺后的 CV 风险。同样,在长期睡眠剥夺(5 或 10 晚)后,健康人观察到血浆 CRP 水平升高和持续,同时心率 , 加快、淋巴细胞促炎激活和细胞因子(例如 IL-1、IL-6 和 IL-17) 产生。同样,在急性完全睡眠剥夺(40小时)后观察到的血压和心率的增加伴随着甚至之前血管舒张受损,IL-6水平升高以及内皮功能障碍和激活的标志物,如细胞粘附分子(E-选择素,ICAM-1等) 。睡眠碎片化动物模型中的睡眠剥夺促动脉粥样硬化作用至少部分是由下丘脑下丘脑分泌素(即食欲素)释放减少介导的,下丘脑分泌素是一种诱导觉醒的神经肽,限制白细胞(单核细胞和中性粒细胞)的产生和动脉粥样硬化的发展,并且与心肌梗塞、心力衰竭和肥胖的风险呈负相关 。 交感神经系统(SNS)的激活可能是睡眠不足与动脉粥样硬化性CV疾病之间炎症联系的另一种机制,因为这种激活增加了祖细胞的骨髓释放,先天免疫细胞(单核细胞)的产生以及炎性细胞因子的水平,并引发内皮功能障碍,从而导致全身和血管炎症以及动脉粥样硬化 , .在诱发炎症反应和促进动脉粥样硬化方面起着关键作用 ,睡眠剥夺相关的氧化应激也可能导致心血管风险。鉴于褪黑激素抑制免疫细胞和血管细胞的氧化应激和细胞因子的产生,并抑制体内动脉粥样硬化病变的形成 ,因此还假设睡眠剥夺后褪黑激素抑制在与睡眠剥夺相关的血管损伤中的作用。

Therefore, a significant and consistent association exists between sleep deprivation and cardiometabolic risk and clinical outcomes, with several plausible immune-mediated causative mechanisms explaining this association.
因此,睡眠剥夺与心脏代谢风险和临床结果之间存在显着且一致的关联,有几种合理的免疫介导的致病机制可以解释这种关联。

Immune mechanisms linking sleep deprivation and diseases
睡眠剥夺与疾病相关的免疫机制

As shown above, sleep deprivation has been found to alter inflammatory immune processes via multiple pathways, which could lead to increased susceptibility to chronic inflammatory diseases (Fig. 2). Most of the current knowledge on immune effects of sleep deprivation come from studies using controlled experimental sleep deprivation protocols, among which chronic partial sleep deprivation, lasting 2–15 days, is that mostly resembling the human condition of chronic insufficient sleep.
如上图所示,已发现睡眠剥夺通过多种途径改变炎症免疫过程,这可能导致对慢性炎症性疾病的易感性增加(图2)。目前关于睡眠剥夺的免疫影响的大多数知识来自使用受控实验性睡眠剥夺方案的研究,其中持续 2-15 天的慢性部分睡眠剥夺与人类长期睡眠不足的状况非常相似。

An external file that holds a picture, illustration, etc.
Object name is 42003_2021_2825_Fig2_HTML.jpg
Immune consequences of sleep deprivation.
睡眠剥夺的免疫后果。

Sleep deprivation, as induced experimentally or in the context of habitual short sleep, has been found to be associated with alterations in the circulating numbers and/or activity of total leukocytes and specific cell subsets, elevation of systemic and tissue (e.g., brain) pro-inflammatory markers including cytokines (e.g., interleukins [IL], tumor necrosis factor [TNF]-α), chemokines and acute phase proteins (such as C reactive Protein [CRP]), altered antigen presentation (reduced dendritic cells, altered pattern of activating cytokines, etc.), lowered Th1 response, higher Th2 response, and reduced antibody production. Furthermore, altered monocytes responsiveness to immunological challenges such as lipopolysaccharide (LPS) may contribute to sleep deprivation-associated immune modulation. Hypothesized links between immune dysregulation by sleep deprivation and the risk for immune-related diseases, such as infectious, cardiovascular, metabolic, and neurodegenerative and neoplastic diseases, are shown. The illustrations were modified from Servier Medical Art (http://smart.servier.com/), licensed under a Creative Common Attribution 3.0 Generic License. APC: antigen-presenting cells.
已发现通过实验或习惯性短睡眠诱导的睡眠剥夺与总白细胞和特定细胞亚群的循环数量和/或活性的改变、全身和组织(例如大脑)促炎标志物(例如白细胞介素 [IL]、肿瘤坏死因子 [TNF]-α)、趋化因子和急性期蛋白(如 C 反应蛋白 [CRP])的升高有关, 抗原呈递改变(树突状细胞减少、细胞因子激活模式改变等)、Th1 反应降低、Th2 反应升高和抗体产生减少。此外,单核细胞对脂多糖 (LPS) 等免疫挑战的反应改变可能有助于睡眠剥夺相关的免疫调节。显示了睡眠剥夺导致的免疫失调与免疫相关疾病(如传染病、心血管疾病、代谢疾病、神经退行性疾病和肿瘤性疾病)风险之间的假设联系。插图修改自 Servier Medical Art ( http://smart.servier.com/),根据 Creative Common Attribution 3.0 通用许可获得许可。APC:抗原呈递细胞。

Some studies have observed that sleep deprivation, compared with regular nocturnal sleep, leads to increased circulating numbers of total leukocytes and specific cell subsets mainly neutrophils, monocytes, B cells, CD4 T cells, and decreased circulating numbers and cytotoxic activity of NK cells,. Other studies, however, found contrasting results, including a decrease in CD4 T cells after sleep deprivation,, probably due to differences in sleep deprivation protocol, sampling methodologies, and other factors. Sleep deprivation has also shown to alter circadian rhythm of circulating leukocytes, with higher levels during the night and at awakening and a flattened rhythm,. Additional findings are suggestive of immune deregulation by sleep deprivation, including a decreased neutrophils phagocytic activity, altered lymphocytes adhesion molecule expression, and reduced stimulated production of IL-2 and IL-12, which are important for adaptive immunity,.
一些研究观察到,与正常的夜间睡眠相比,睡眠剥夺导致总白细胞和特定细胞亚群(主要是中性粒细胞、单核细胞、B 细胞、CD4 T 细胞)的循环数量增加,以及 NK 细胞 , 的循环数量和细胞毒性活性降低。然而,其他研究发现了截然不同的结果,包括睡眠剥夺后CD4 T细胞的减少 , ,可能是由于睡眠剥夺协议,采样方法和其他因素的差异。睡眠剥夺也被证明会改变循环白细胞的昼夜节律 ,在夜间和醒来时水平较高,节律扁平 , 。其他发现提示睡眠剥夺导致的免疫失调,包括中性粒细胞吞噬活性 降低、淋巴细胞粘附分子表达 改变以及 IL-2 和 IL-12 的刺激产生减少,这对适应性免疫很重要 ,

Experimental sleep deprivation has been reported to affect systemic markers of inflammation, with studies showing increased circulating pro-inflammatory molecules (IL-1, IL-6, CRP, TNF-α, and MCP-1); this associated in some studies with a subsequent homeostatic increase in endogenous inhibitors, including IL-1 receptor antagonist and TNF receptors,. In agreement with experimental sleep deprivation, population studies found a direct independent association between habitual short sleep duration (generally < 5 or 6 h) and elevated circulating pro-inflammatory markers, e.g., acute phase proteins (CRP and IL-6), cytokines (TNF-α, IFN-γ, IL-1, etc.), adhesion molecules, and leukocyte counts,. Furthermore, a reduced NK cell activity and a decline in naive T cells, compatible with reduced immune competence, was reported in association with habitual short sleep. Shortening of leukocyte telomere length, a cellular senescence marker linked with inflammation, was also associated with shorter sleep duration,.
据报道,实验性睡眠剥夺会影响炎症的全身标志物,研究表明循环促炎分子(IL-1、IL-6、CRP、TNF-α 和 MCP-1)增加;在一些研究中,这与随后内源性抑制剂(包括 IL-1 受体拮抗剂和 TNF 受体)的稳态增加有关 , 。与实验性睡眠剥夺一致,人群研究发现习惯性短睡眠时间(通常< 5 或 6 小时)与循环促炎标志物升高之间存在直接独立关联,例如急性期蛋白(CRP 和 IL-6)、细胞因子(TNF-α、IFN-γ、IL-1 等)、粘附分子和白细胞计数 , 。此外,据报道,NK细胞活性 降低和幼稚T细胞 下降,与免疫能力降低相符,与习惯性短睡眠有关。白细胞端粒长度的缩短是与炎症相关的细胞衰老标志物,也与较短的睡眠时间有关 ,

The reported elevation of systemic inflammation is clinically relevant, because it is suggested to specifically mediate the increased risk of mortality associated with short sleep,, and, as observed, the risk for chronic disease development.
报告的全身炎症升高具有临床意义,因为建议特异性介导与睡眠 ,, 不足相关的死亡风险增加,以及所观察到的慢性疾病发展风险。

Regarding cellular markers of inflammation, some studies found that the ex-vivo LPS-stimulated production of TNF-α,, IL-1β, and IL-6, by human monocytes increased during sleep deprivation but decreased during regular nocturnal sleep,,. However, other studies reported a decrease of TNF-α production by activated monocytes after sleep deprivation compared with regular nocturnal sleep,. These contrasting results need further investigations and may depend on differences in the cytokine sensitivity to different sleep deprivation protocols or sampling methods and time. For instance, it seems that partial acute sleep deprivation increased stimulated monocytic TNF-α production,, whereas more sustained sleep deprivation decreased it,.
关于炎症的细胞标志物,一些研究发现,人单核细胞离体LPS刺激的TNF-α , 、IL-1β和IL-6 , 的产生在睡眠剥夺期间增加,但在正常夜间睡眠 ,, 期间减少。然而,其他研究报告称,与常规夜间睡眠 , 相比,睡眠剥夺后激活的单核细胞产生的 TNF-α 减少。这些对比结果需要进一步研究,并且可能取决于细胞因子对不同睡眠剥夺方案或采样方法和时间的敏感性的差异。例如,似乎部分急性睡眠剥夺增加了刺激的单核细胞TNF-α的产生,而更持久的睡眠剥夺则减少了单 , 核细胞TNF-的产生 ,

Undisturbed sleep is predominantly characterized by a Th1 polarization of Th cells (expressing IFN-γ, IL-2, and TNF-α), and experimental sleep deprivation in humans leads to a shift from a Th1 pattern towards a Th2 pattern (expressing IL-4, IL-5, IL-10, and IL-13),. Accordingly, conditions featured by disturbed sleep with specific deficit in slow wave sleep, as observed in elderly people, alcoholic, and insomnia patients, show a cytokine shift towards Th2. The balance of Th1/Th2 immunity and its shift during sleep deprivation may have crucial implications in anti-microbial and anti-tumor immune responses. Th2 over-activity is known to be involved in some forms of allergic responses, and to increase the susceptibility to infection. Likewise, regarding the anti-tumor immune action, Th1 response supports cytotoxic lymphocytes and tumor cells destruction with the potential of elimination or control of tumor cell growth, so that a type 1 adaptive immune response (increased antigen presentation, IFN-γ signaling, and T-cell receptor signaling) may be associated with an improved survival or prognosis,. In contrast, Th2 over-response is thought to contribute to tumor development and progression, by limiting cytotoxic T lymphocytes proliferation and by the modulation of other inflammatory cell types.
不受干扰的睡眠主要特征是 Th 细胞的 Th1 极化(表达 IFN-γ、IL-2 和 TNF-α),人类的实验性睡眠剥夺导致从 Th1 模式转变为 Th2 模式(表达 IL-4、IL-5、IL-10 和 IL-13)。 , 因此,在老年人 、酗酒 者和失眠 患者中观察到的以慢波睡眠中特定缺陷的睡眠障碍为特征的病症显示细胞因子向 Th2 转移。Th1/Th2免疫的平衡及其在睡眠剥夺期间的转变可能对抗菌和抗肿瘤免疫反应具有至关重要的意义。已知 Th2 过度活动与某些形式的过敏反应有关,并增加对感染 的易感性。同样,关于抗肿瘤免疫作用,Th1 反应支持细胞毒性淋巴细胞和肿瘤细胞破坏,具有消除或控制肿瘤细胞生长的潜力,因此 1 型适应性免疫反应(增加抗原呈递、IFN-γ 信号传导和 T 细胞受体信号传导)可能与生存或 , 预后改善有关.相比之下,Th2 过度反应被认为通过限制细胞毒性 T 淋巴细胞增殖和调节其他炎症细胞类型 来促进肿瘤的发展和进展。

Several cellular and molecular signaling pathways may be involved in mediating the influence of sleep deprivation on immune and inflammatory functions (Fig. 3). Increased oxidative stress markers and/or decreased antioxidant defense have been found after sleep deprivation. Sleep shows an antioxidant function, responsible for eliminating reactive oxygen species produced during wakefulness, and contrarily sleep deprivation may cause oxidative stress, which leads to cell senescence, unbalanced local/systemic inflammation, dysmetabolism, and immune derangements,.
几种细胞和分子信号通路可能参与介导睡眠剥夺对免疫和炎症功能的影响(图3)。睡眠剥夺后发现氧化应激标志物增加和/或抗氧化防御能力下降 。睡眠具有抗氧化功能,负责消除清醒时产生的活性氧,相反,睡眠不足可能会导致氧化应激,从而导致细胞衰老、局部/全身炎症失衡、代谢紊乱和免疫紊乱 ,

An external file that holds a picture, illustration, etc.
Object name is 42003_2021_2825_Fig3_HTML.jpg
Pro-inflammatory molecular pathways induced by sleep deprivation.
睡眠剥夺诱导的促炎分子通路。

A schematic model of potential mechanistic pathways linking sleep deprivation and inflammatory immune activation is depicted. Sleep deprivation is associated with activation of the sympathetic nervous system and release of norepinephrine and epinephrine into the systemic circulation, as well as to some extent with impaired hypothalamus-pituitary axis stimulation. These neuromediators may act along with other potential stimuli accumulated following sleep deprivation including reactive oxygen species (ROS), adenosine, metabolic waste products (e.g., β-amyloid) not cleared during normal sleep, gut microbiota dysbiosis leading to altered local and systemic pattern of metabolic products, as well as with changes in the profile of neuro-endocrine hormones, such as prolactin, growth hormone, and altered circadian rhythm of melatonin secretion. In immune cells located in the brain and the peripheral tissues, these stimuli may in concert trigger inflammatory activation, with release of cytokines, chemokines, acute phase protein, etc. via the recruitment of transcriptional regulators of pro-inflammatory gene expression, mainly nuclear factor (NF)-κB, and disturbing the circadian rhythmicity of gene expression of both clock genes and metabolic, immune and stress response genes (see text for further detail). E: epinephrine; NE: norepinephrine; TLR: Toll-like receptor. Arrows indicate stimulation; lines indicate inhibition. The illustrations were modified from Servier Medical Art (http://smart.servier.com/), licensed under a Creative Common Attribution 3.0 Generic License.
描述了将睡眠剥夺和炎症免疫激活联系起来的潜在机制通路的示意图模型。睡眠剥夺与交感神经系统的激活和去甲肾上腺素和肾上腺素释放到体循环中有关,并且在某种程度上与下丘脑 - 垂体轴刺激受损有关。这些神经介质可能与睡眠剥夺后积累的其他潜在刺激一起起作用,包括活性氧 (ROS)、腺苷、正常睡眠期间未清除的代谢废物(例如β-淀粉样蛋白)、肠道菌群失调导致代谢产物的局部和全身模式改变,以及神经内分泌激素(如催乳素)谱的变化, 生长激素和褪黑激素分泌的昼夜节律改变。在位于大脑和外周组织的免疫细胞中,这些刺激可能协同触发炎症激活,通过募集促炎基因表达的转录调节因子(主要是核因子 (NF)-κB)释放细胞因子、趋化因子、急性期蛋白等,并扰乱时钟基因和代谢基因表达的昼夜节律性, 免疫和应激反应基因(详见正文)。E:肾上腺素;NE:去甲肾上腺素;TLR:Toll样受体。箭头表示刺激;线条表示抑制。插图修改自 Servier Medical Art ( http://smart.servier.com/),根据 Creative Common Attribution 3.0 通用许可获得许可。

Effects of sleep deprivation on the immune response may derive from the activation of the SNS with the corresponding increase in systemic catecholamines,. Catecholamines signal to immune cells via adrenergic receptors, which are primarily α- and β-adrenergic in myeloid cells and β-adrenergic in lymphocytes. The immune outcome of the sympathetic signaling is complex, and includes both stimulatory and inhibitory effects depending on cell and receptor types, cell development/activation states, and local microenvironment,. Some evidence suggest that β-adrenergic signaling inhibits and α-adrenergic signaling promotes excessive inflammation under endotoxemia. Activation of α-adrenergic signaling in peripheral tissues induces the upregulation of pro-inflammatory cytokines,. Sympathetic activation also suppresses the transcription of type I IFNs (IFN-α and IFN-β) genes and interferon response genes, which play a key role in anti-viral immunity, and inhibits via β-adrenergic signaling the anti-tumor cytotoxicity of T lymphocytes. In vitro β-adrenergic stimulation repressed Th1 response and stimulated Th2 response, with varying effects found in vivo,. Although the specific role of SNS activation in the immune phenotype associated with sleep deprivation is not clearly established, data suggest a pro-inflammatory effect of SNS under sleep deprivation. Indeed, chemical sympathectomy has been recently shown to alleviate the inflammatory response following chronic sleep deprivation in mice, and both α- and, to a lesser extent, β-adrenergic receptors seem to contribute to the sympathetic regulation of inflammatory responses to sleep deprivation.
睡眠剥夺对免疫反应的影响可能源于SNS的激活以及全身儿茶酚胺的相应增加 , 。儿茶酚胺通过肾上腺素能受体向免疫细胞发出信号,肾上腺素能受体在髓系细胞中主要是α肾上腺素能和β肾上腺素能,在淋巴细胞中是β肾上腺素能 。交感神经信号转导的免疫结果是复杂的,包括刺激和抑制作用,具体取决于细胞和受体类型、细胞发育/激活状态和局部微环境 , 。一些证据表明,β-肾上腺素能信号传导抑制和α-肾上腺素能信号传导促进内毒素血症下的过度炎症 。外周组织中α肾上腺素能信号传导的激活诱导促炎细胞因子的上调 , 。交感神经激活还抑制I型IFN(IFN-α和IFN-β)基因和干扰素反应基因的转录,这些基因在抗病毒免疫中起关键作用 ,并通过β-肾上腺素能信号抑制T淋巴细胞的抗肿瘤细胞毒性 。体外β肾上腺素能刺激抑制Th1反应并刺激Th2反应,在体内发现不同的效果 , 。尽管SNS激活在与睡眠剥夺相关的免疫表型中的具体作用尚不清楚,但数据表明SNS在睡眠剥夺下具有促炎作用。事实上,化学交感神经切除术最近已被证明可以减轻小鼠慢性睡眠剥夺后的炎症反应 ,并且α和在较小程度上β肾上腺素能受体似乎都有助于对睡眠剥夺的炎症反应的交感神经调节

At the molecular levels, sleep deprivation led to significant gene expression changes in animal tissues and human blood monocytes,,, with affected genes mostly related to immune and inflammatory processes (leukocyte function, Th1/Th2 balance, cytokine regulation, and TLR signaling), oxidative stress, stress response, apoptosis, and circadian system, collectively indicating immune activation and hyperinflammation.
在分子水平上,睡眠剥夺导致动物组织 和人血液单核细胞的基因表达发生显著变化 ,, ,受影响的基因主要与免疫和炎症过程(白细胞功能、Th1/Th2平衡、细胞因子调节和TLR信号传导)、氧化应激、应激反应、细胞凋亡和昼夜节律系统有关,共同表明免疫激活和过度炎症。

Sleep loss and mistimed sleep also led in the blood transcriptome to alteration and reduction in the circadian rhythmicity of gene expression,, which is an integral part of basic biological processes and homeostasis.
睡眠不足和不合时宜的睡眠也导致血液转录组改变和降低基因表达 , 的昼夜节律性,这是基本生物过程和稳态的一个组成部分

The activation of the pro-inflammatory NF-κB/Rel family of transcription factors by sleep deprivation, first demonstrated in the late 1990s in mice, and subsequently widely confirmed,,,, is one of the most consistent findings regarding upstream transcriptional regulation. NF-κB induces the expression of genes (e.g., cytokines/chemokines, growth factors, receptors/transporters, enzymes, adhesion molecules) involved in inflammation, immunity, proliferation, and apoptosis, circadian clock activity, and sleep propensity. Potential signals for NF-κB activation under sleep deprivation include increased adenosine levels, oxidative stress, altered metabolism (adiposity and decreased insulin sensitivity), brain proteins/metabolites (e.g., Aβ), melatonin suppression, circadian clock proteins, and catecholamine surge due to increased sympathetic activity. Given the role of NF-κB in the pathophysiology of inflammatory diseases, its activation under sleep deprivation may be a common pathway for the risk of morbidity and mortality.
1990年代后期,在小鼠中首次发现,通过睡眠剥夺激活促炎性NF-κB/Rel转录因子家族 ,随后被广泛证实 ,,, ,这是关于上游转录调控的最一致的发现之一。NF-κB 诱导参与炎症、免疫、增殖和凋亡 、生物钟活动 和睡眠倾向的基因(例如细胞因子/趋化因子、生长因子、受体/转运蛋白、酶、粘附分子 )的表达。睡眠剥夺下NF-κB激活的潜在信号包括腺苷水平升高、氧化应激、代谢改变(肥胖和胰岛素敏感性降低)、脑蛋白/代谢物(例如Aβ)、褪黑激素抑制 、生物钟蛋白 和儿茶酚胺激增由于交感神经活动 增加。鉴于NF-κB在炎症性疾病 的病理生理学中的作用,它在睡眠剥夺下的激活可能是发病和死亡风险的常见途径。

The intestinal microbiota is also affected by sleep loss, showing indices of dysbiosis (increased Firmicutes:Bacteroidetes ratio; decreased diversity and richness), which may affect the immune system, and are similar to those associated with cardiometabolic diseases.
肠道微生物群也受到睡眠不足 的影响,显示出生态失调的指数(厚壁菌门:拟杆菌门比率增加;多样性和丰富度降低),这可能影响免疫系统 ,并且与心脏代谢疾病 相关。

Countermeasures for sleep deprivation: effect on immune parameters
睡眠剥夺的对策:对免疫参数的影响

Although the impact of strategies to improve sleep duration on neurobehavioral performance and alertness after sleep deprivation have been assessed, sleep deprivation countermeasures to improve immune and inflammatory parameters, and, correspondingly, disease risk and outcomes have been studied to a lesser extent.
尽管已经评估 了改善睡眠持续时间的策略对睡眠剥夺后神经行为表现和警觉性的影响,但改善免疫和炎症参数的睡眠剥夺对策,以及相应的疾病风险和结果的研究程度较小。

Although extension of habitual short sleep did not show to significantly counterbalance the immune consequence of sleep deprivation, mixed results derive from nighttime recovery sleep following sleep deprivation (Table 1), with limited evidence of effectiveness for specific immune parameters,, and mostly after multiple consecutive nights of 8 h sleep recovery or with an extended nocturnal sleep duration,.
虽然习惯性短睡眠的延长并没有显示出显著抵消睡眠剥夺的免疫后果 ,但睡眠剥夺后的夜间恢复睡眠得出了不同的结果(表1),对特定免疫参数 , 的有效性证据有限,而且大多是在连续多个晚上8小时睡眠恢复或夜间睡眠持续时间延长 , 之后。

Table 1 表1

Main human findings on the effects of recovery sleep on sleep deprivation-induced changes in immune and inflammatory parameters.
关于恢复睡眠对睡眠剥夺引起的免疫和炎症参数变化影响的主要人类发现。

Subjects (number and age range or mean)
受试者(数量和年龄范围或平均值)
Sleep deprivation protocol
睡眠剥夺协议
Effect of sleep deprivation on immune parameters compared with baseline
与基线相比,睡眠剥夺对免疫参数的影响
Recovery sleep protocol 恢复睡眠方案Effect of recovery sleep on immune parameters compared with baseline
与基线相比,恢复睡眠对免疫参数的影响
Effect of recovery sleep on immune parameters compared with sleep deprivation
恢复睡眠与睡眠剥夺相比对免疫参数的影响
Reference 参考
Healthy men and women (n = 20, 21–30 yrs)
健康的男性和女性(n = 20,21-30 岁)
64 h TSD 64小时 TSD↑ Granulocytes and monocytes, NK cell activity
↑ 粒细胞和单核细胞,NK细胞活性
1 Night (h sleep not reported)
1 晚(未报告睡眠时间)
↑ Granulocytes, monocytes; = NK cell activity
↑ 粒细胞、单核细胞;= NK细胞活性

= Granulocytes; = 粒细胞;

↓ monocytes; ↓ 单核细胞;

↓ NK cell activity ↓ NK细胞活性

Dinges et al. Dinges等人。
Healthy men (n = 32, 19–29 yrs)
健康男性(n = 32,19-29 岁)
2 Nights TSD or 4 nights of REM SD
2 晚 TSD 或 4 晚 REM SD
TSD: ↑ total leukocytes, neutrophils, CD4 T cells; REM SD: ↓ IgA
TSD:↑ 总白细胞、中性粒细胞、CD4 T 细胞;REM SD:↓ IgA
3 Nights (8 h sleep/night)
3 晚 (8 小时睡眠/晚)
= Total leukocytes, neutrophils; ↑ CD4 T cells ↓ IgA
= 总白细胞、中性粒细胞;↑ CD4 T 细胞 ↓ IgA
NDRuiz et al. Ruiz 等人。
Healthy young men (n = 10, 21–29 yrs)
健康的年轻男性(n = 10,21-29 岁)
1 Night TSD 1 晚 TSD

During SD: ↑ monocytes, lymphocytes, NK cells. The day after SD:
在SD期间:↑单核细胞,淋巴细胞,NK细胞。SD后的第二天:

↓ lymphocytes, NK cells ↓ 淋巴细胞、NK细胞

1 Night (8 h sleep) 1 晚(8 小时睡眠)= Monocytes, lymphocytes; ↓ NK cells
= 单核细胞、淋巴细胞;↓ NK细胞
NDBorn et al. Born等人。
Healthy men (n = 12, mean age 29 yrs)
健康男性(n = 12,平均年龄 29 岁)
40 h TSD 40小时 TSD

↑ Plasma E-selectin; ↑ 血浆E-选择素;

↑ systolic BP, heart rate; plasma norepinephrine;
↑ 收缩压、心率;血浆去甲肾上腺素;

↓ endothelium-dependent and -independent vasodilation
↓ 内皮依赖性和非依赖性血管舒张

1 Night (8 h sleep) 1 晚(8 小时睡眠)↑ Plasma ICAM-1, IL-6, norepinephrine
↑ 血浆ICAM-1、IL-6、去甲肾上腺素
NDSauvet et al. Sauvet 等人。
Healthy men (n = 31, 18–27 yrs)
健康男性(n = 31,18-27 岁)
1 Night with 2 h sleep
1 晚,睡 2 小时
↑ Total leukocytes, neutrophils
↑ 总白细胞、中性粒细胞
1 Night of 8 h sleep or 1 night of 10 h sleep
1 晚 8 小时睡眠或 1 晚 10 小时睡眠
8 h Recovery sleep: ↑ leukocytes, neutrophils; 10 h recovery sleep: = leukocytes, neutrophils
8小时恢复睡眠:↑白细胞,中性粒细胞;10 小时恢复睡眠:= 白细胞、中性粒细胞
8 h Recovery sleep: = leukocytes, neutrophils; 10 h recovery sleep: ↓ leukocytes, neutrophils
8小时恢复睡眠:=白细胞,中性粒细胞;10小时恢复睡眠:↓白细胞,中性粒细胞
Faraut et al. Faraut 等人。
Healthy men (n = 19, 19–29 yrs)
健康男性(n = 19,19-29 岁)
5 Nights with 4 h sleep/night
5 晚,4 小时睡眠/晚

↓ NK cells; ↓ NK细胞;

↑ B cells; ↑ B细胞;

↑ plasma CRP; ↑ 血浆CRP;

↑ IL-17, IL-1β, IL-6 (PBMC mRNA);
↑ IL-17、IL-1β、IL-6(PBMC mRNA);

↓ TNF-α (PBMC protein) ↓ TNF-α(PBMC蛋白)

2 Nights (8 h sleep/night)
2 晚 (8 小时睡眠/晚)
= NK cells; = B cells; ↑ CRP, IL-17; = IL-1β, IL-6, TNF-α
= NK细胞;= B细胞;↑ CRP,IL-17;= IL-1β、IL-6、TNF-α
NDvan Leeuwen et al.
van Leeuwen 等人。
Healthy men (n = 9, 22–27 yrs)
健康男性(n = 9,22-27 岁)
5 Nights with 4 h sleep/night
5 晚,4 小时睡眠/晚
↑ Total leukocytes, monocytes, neutrophils, lymphocytes
↑ 总白细胞、单核细胞、中性粒细胞、淋巴细胞
7 Nights (8 h sleep/night)
7 晚 (8 小时睡眠/晚)
↓ Monocytes, lymphocytes; ↑ neutrophils
↓ 单核细胞、淋巴细胞;↑ 中性粒细胞
NDLasselin et al.
Lasselin等人。
Healthy men and women (n = 24, 36-76 yrs)
健康的男性和女性(n = 24,36-76岁)
1 Night with 4 h sleep
1 晚,睡 4 小时
↑ IL-6 and TNF-α (PBMC protein)
↑ IL-6 和 TNF-α(PBMC 蛋白)
1 Night (8 h sleep) 1 晚(8 小时睡眠)↑ IL-6 and TNF-α ↑ IL-6 和 TNF-αNDIrwin et al. 欧文等人。
Healthy men (n = 10, 22–37 yrs) (Exp. 1) and healthy men and women (n = 10, 26–38 yrs) (Exp. 2)
健康男性(n = 10,22-37 岁)(实验 1)和健康男性和女性(n = 10,26-38 岁)(实验 2)
88 h TSD (Exp. 1) and 10 days with 4.2 h sleep/night (Exp. 2)
88 小时 TSD(经验值 1)和 10 天,睡眠/晚 4.2 小时(经验值 2)
↑ Plasma CRP ↑ 等离子体 CRP3 Nights (assessment only in the first recovery day)
3 晚(仅在第一个恢复日进行评估)
↑ Plasma CRP ↑ 等离子体 CRPNDMeier-Ewert et al.
Meier-Ewert等人。
Healthy men and women (n = 21, 25–39 yrs and n = 49, 60-84 yrs)
健康的男性和女性(n = 21,25-39 岁和 n = 49,60-84 岁)
1 night with 4 h sleep
1 晚,睡 4 小时
↑ IL-6 and TNF-α (PBMC protein) in younger adults
↑ IL-6 和 TNF-α(PBMC 蛋白)在年轻人中的应用
1 night (8 h sleep) 1 晚 (8 小时睡眠)↑ IL-6 and TNF-α ↑ IL-6 和 TNF-αNDCarroll et al. 卡罗尔等人。
Healthy men and women (n = 30, 18–34 yrs)
健康的男性和女性(n = 30,18-34 岁)
6 Nights with 6 h sleep/night
6 晚,6 小时睡眠/晚
↑ Plasma IL-6 ↑ 血浆IL-63 Nights (10 h sleep/night)
3 晚(10 小时睡眠/晚)
= Plasma IL-6 = 血浆 IL-6↓ Plasma IL-6 ↓ 血浆IL-6Pejovic et al. Pejovic 等人。
Healthy men and women (n = 14, 18–35 yrs)
健康的男性和女性(n = 14,18-35 岁)
5 Nights with 4 h sleep/night, for 3 weeks
5 晚,4 小时睡眠/晚,持续 3 周
↑ IL-6 (PBMC protein) ↑ IL-6(PBMC蛋白)2 Nights (8 h sleep/night), for 3 weeks
2 晚(8 小时睡眠/晚),持续 3 周
↑ IL-6 (PBMC protein) ↑ IL-6(PBMC蛋白)NDSimpson et al. 辛普森等人。

BP blood pressure, exp experiment, ICAM-1 intercellular adhesion molecule-1, ND not determined, PBMC peripheral blood mononuclear cells, SD sleep deprivation, TSD total sleep deprivation, VCAM-1 vascular cell adhesion molecule-1, yrs years, significant increase, significant decrease, = no significant change.
血压血压,exp实验,ICAM-1细胞间粘附分子-1,ND未确定,PBMC外周血单核细胞,SD睡眠剥夺,TSD总睡眠剥夺,VCAM-1血管细胞粘附分子-1,年,↑显著增加,↓显著减少,=无显著变化。

Although daytime napping (<20 min) restores alertness, and mental and physical performance without provoking sleep inertia associated with longer nap, the effects of a short nap on immune/inflammatory parameters after sleep deprivation have yet to be firmly established. Differently form population studies, laboratory studies found immune benefit from nap,,,. Regarding immune-related clinical outcomes, controversy exists, with studies finding no association, inverse associations, or positive association, and a J-shaped relationship between napping and CV and metabolic diseases or cancer events and mortality. Whether changes in immune parameters could contribute to the associations between napping and immune-related diseases remains unclear.
虽然白天小睡(<20分钟)可以恢复警觉性,并在不引起与较长午睡相关的睡眠惯性的情况下恢复精神和身体表现 ,但短睡对睡眠剥夺后免疫/炎症参数的影响尚未确定。与不同形式的人群研究 ,实验室研究发现午睡对免疫有益 ,,, 。关于免疫相关的临床结果,存在争议,研究发现没有关联 、负相关 , 或正相关 ,午睡与心血管与代谢疾病或癌症事件和死亡率之间存在 J 型关系 。免疫参数的变化是否有助于午睡与免疫相关疾病之间的关联尚不清楚。

Among the strategies to recover sleep deprivation-induced immune changes, cognitive behavior therapy improves sleep outcomes in insomnia and lowers cellular and systemic inflammatory markers,, and the risk score composed of CV and metabolic risk factors. This highlights the potential role of targeting sleep in reducing the inflammatory risk and the associated chronic diseases.
在恢复睡眠剥夺诱导的免疫变化的策略中,认知行为疗法改善了失眠患者的睡眠结果,降低了细胞和全身炎症标志物 , ,以及由CV和代谢危险因素 组成的风险评分。这凸显了以睡眠为目标在降低炎症风险和相关慢性疾病方面的潜在作用。

Summary and concluding remarks
总结和结束语

Sleep exerts immune-supportive functions and impairments of the immune-inflammatory system are a plausible mechanism mediating the negative health effects of sleep deprivation, and in particular, its role in the risk and outcomes of chronic diseases such as infections, CV, metabolic and autoimmune diseases, NDDs, and cancer. Caution should be exercised in interpreting cellular and molecular outcomes of sleep deprivation in experimental studies conducted till now as a result of an independent effect of sleep deprivation, because other factors may play a role, including extended wakefulness-associated processes, other features of sleep-wakefulness, their temporal and functional segregation or methodologies of sleep manipulation.
睡眠发挥免疫支持功能,免疫炎症系统的损伤是一种合理的机制,介导睡眠剥夺对健康的负面影响,特别是它在慢性疾病(如感染、心血管、代谢和自身免疫性疾病、NDD 和癌症)的风险和结果中的作用。在迄今为止进行的实验研究中,由于睡眠剥夺的独立影响,在解释睡眠剥夺的细胞和分子结果时应谨慎,因为其他因素可能起作用,包括延长的觉醒相关过程、睡眠觉醒的其他特征、它们的时间和功能分离或睡眠操纵方法。

Randomized controlled trials assessing the effect of treatment of sleep deprivation on inflammatory immune dysfunction and/or health outcomes are needed. Knowledge of inflammatory and immunological signatures in response to sleep curtailment may inform not only on the underlying molecular links, but also contribute to refine risk profiles to be used for developing biomarkers of sleep deprivation and sleep disturbance-related health outcomes, which may also represent potential targets of interventions. Recent metabolomic and transcriptomic studies hold promise in biomarker discovery.
需要随机对照试验来评估睡眠剥夺治疗对炎症性免疫功能障碍和/或健康结局的影响。对睡眠减少反应的炎症和免疫特征的了解不仅可以为潜在的分子联系提供信息,还有助于完善风险概况,用于开发睡眠剥夺和睡眠障碍相关健康结果的生物标志物,这也可能代表潜在的干预目标。最近的代谢组学 和转录组 学研究在生物标志物发现 方面很有希望。

These efforts may converge towards a new ground fostering interactions between the sleep research and the medical community to translate scientific knowledge into the clinic, prioritize health issues, and develop strategies and policies for subject risk stratification, to include evidence-based sleep recommendations in guidelines for optimal health and to address sleep hygiene at the individual and the population levels, as a means to prevent the negative health consequences of sleep deprivation. These actions might also foster health literacy and empowerment of individuals to actively better manage their own health and well-being throughout their life course by means of lifestyle, nutritional, and behavioral habits including sleep hygiene.
这些努力可能会汇聚到一个新的领域,促进睡眠研究和医学界之间的互动,将科学知识转化为临床,优先考虑健康问题,并制定受试者风险分层的战略和政策,将循证睡眠建议纳入最佳健康指南,并解决个人和人群层面的睡眠卫生问题, 作为防止睡眠不足对健康造成负面影响的一种手段。这些行动还可以通过生活方式、营养和行为习惯(包括睡眠卫生)来促进个人的健康素养和赋权,从而在整个生命过程中积极更好地管理自己的健康和福祉

Conclusively, in the perspective of staying healthy in this rapidly changing society, the sleep–immunity relationship raises relevant clinical implications for promoting sleep health and, as evidenced here, for improving or therapeutically controlling inflammatory response by targeting sleep. This may ultimately translate, in the era of preventive medicine, into addressing sleep as a lifestyle approach along with diet and physical activity to benefit overall public health.
总而言之,从在这个瞬息万变的社会中保持健康的角度来看,睡眠-免疫关系对促进睡眠健康提出了相关的临床意义,正如本文所证明的那样,通过针对睡眠来改善或治疗控制炎症反应。在预防医学时代,这可能最终转化为将睡眠作为一种生活方式,以及饮食和身体活动,以造福整体公共卫生。

Reporting summary 报告摘要

Further information on research design is available in the Nature Research Reporting Summary linked to this article.
有关研究设计的更多信息,请参阅本文链接的《自然研究报告摘要》。

Supplementary information
补充资料

Transparent Peer Review File(3.3M, pdf)
透明的同行评审文件 (3.3M, pdf)

Reporting Summary(250K, pdf)
报告摘要 (250K, pdf)

Author contributions 作者贡献

E.S. reviewed the literature and wrote the manuscript draft. S.G. and P.L. contributed to writing the manuscript and revised the manuscript draft. N.L.B. and N.M. reviewed the final manuscript.
E.S.审阅了文献并撰写了手稿。S.G. 和 P.L. 参与了手稿的撰写,并修改了手稿草稿。N.L.B. 和 N.M. 审阅了最终手稿。

Data availability 数据可用性

All data generated or analysed during this study are included in this published article.
本研究期间生成或分析的所有数据都包含在这篇已发表的文章中。

Competing interests 利益争夺

The authors declare no competing interests.
作者声明没有竞争利益。

Footnotes 脚注

Peer review information Communications Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Karli Montague-Cardoso. Peer reviewer reports are available.
同行评审信息 Communications Biology 感谢匿名评审员对这项工作的同行评审的贡献。主要处理编辑:Karli Montague-Cardoso。提供同行评审报告。

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
出版商注:施普林格·自然(Springer Nature)对已出版地图和机构隶属关系中的管辖权主张保持中立。

Supplementary information
补充资料

The online version contains supplementary material available at 10.1038/s42003-021-02825-4 IF: 5.9 Q1 .
在线版本包含补充材料,网址为 10.1038/s42003-021-02825-4IF: 5.9 Q1。

References 引用

1. Luyster FS, et al. Sleep: a health imperative. Sleep. 2012;35:727–734. [PMC free article] [PubMed] []
1. Luyster FS 等人。睡眠:健康的当务之急。睡。2012;35:727–734.[ PMC免费商品][ 出版][ 谷歌学术搜索]
2. Grandner MA. Sleep, health, and society. Sleep. Med. Clin. 2017;12:1–22. [PMC free article] [PubMed] []
2. 格兰德纳马。睡眠、健康和社会。睡。医学克林。2017;12:1–22.[ PMC免费商品][ 出版][ 谷歌学术搜索]
3. Ohayon MM, Carskadon MA, Guilleminault C, Vitiello MV. Meta-analysis of quantitative sleep parameters from childhood to old age in healthy individuals: developing normative sleep values across the human lifespan. Sleep. 2004;27:1255–1273. [PubMed] []
3. Ohayon MM、Carskadon MA、Guilleminault C、Vitiello MV。健康个体从童年到老年的定量睡眠参数的荟萃分析:在整个人类生命周期中制定规范的睡眠值。睡。2004;27:1255–1273.[ 出版][ 谷歌学术搜索]
4. Galland BC, Taylor BJ, Elder DE, Herbison P. Normal sleep patterns in infants and children: a systematic review of observational studies. Sleep. Med. Rev. 2012;16:213–222. [PubMed] []
4. Galland BC, Taylor BJ, Elder DE, Herbison P. 婴儿和儿童的正常睡眠模式:观察性研究的系统评价。睡。2012年医学修订版;16:213–222.[ 出版][ 谷歌学术搜索]
5. Galland, B. C. et al. Establishing normal values for pediatric nighttime sleep measured by actigraphy: a systematic review and meta-analysis. Sleep41, 10.1093/sleep/zsy017 IF: 5.6 Q1 (2018). []
5. 加兰德,不列颠哥伦比亚省等人。通过活动记录仪确定儿科夜间睡眠的正常值:系统评价和荟萃分析。Sleep41,10.1093/sleep/zsy017IF:5.6 第一季度(2018 年)。[]
6. Boulos MI, et al. Normal polysomnography parameters in healthy adults: a systematic review and meta-analysis. Lancet Respir. Med. 2019;7:533–543. [PubMed] []
6. Boulos MI 等人。健康成人的正常多导睡眠图参数:系统评价和荟萃分析。柳叶刀呼吸。2019年医学;7:533–543.[ 出版][ 谷歌学术搜索]
7. Consensus Conference P, et al. Joint Consensus Statement of the American Academy of Sleep Medicine and Sleep Research Society on the Recommended Amount of Sleep for a Healthy Adult: methodology and discussion. J. Clin. Sleep. Med. 2015;11:931–952. [PMC free article] [PubMed] []
7. 美国睡眠医学会和睡眠研究学会关于健康成人推荐睡眠量的联合共识声明:方法和讨论。J.克林。睡。2015年医学;11:931–952.[ PMC免费商品][ 出版][ 谷歌学术搜索]
8. Consensus Conference P, et al. Recommended amount of sleep for a healthy adult: a Joint Consensus Statement of the American Academy of Sleep Medicine and Sleep Research Society. J. Clin. Sleep. Med. 2015;11:591–592. [PMC free article] [PubMed] []
8. 共识会议 P 等人。健康成年人的推荐睡眠量:美国睡眠医学会和睡眠研究学会的联合共识声明。J.克林。睡。2015年医学;11:591–592.[ PMC免费商品][ 出版][ 谷歌学术搜索]
9. Carskadon MA, Vieira C, Acebo C. Association between puberty and delayed phase preference. Sleep. 1993;16:258–262. [PubMed] []
9. Carskadon MA, Vieira C, Acebo C. 青春期与延迟阶段偏好之间的关联。睡。1993;16:258–262.[ 出版][ 谷歌学术搜索]
10. Gulia KK, Kumar VM. Sleep disorders in the elderly: a growing challenge. Psychogeriatrics. 2018;18:155–165. [PubMed] []
10. Gulia KK,库马尔 VM。老年人睡眠障碍:日益严峻的挑战。老年精神病学。2018;18:155–165.[ 出版][ 谷歌学术搜索]
11. Garbarino S, Lanteri P, Sannita WG, Bragazzi NL, Scoditti E. Circadian rhythms, sleep, immunity, and fragility in the elderly: the model of the susceptibility to infections. Front. Neurol. 2020;11:558417. [PMC free article] [PubMed] []
11. Garbarino S, Lanteri P, Sannita WG, Bragazzi NL, Scoditti E. 老年人的昼夜节律、睡眠、免疫力和脆弱性:感染易感性模型。前面。神经学 2020;11:558417.[ PMC免费商品][ 出版][ 谷歌学术搜索]
12. Zomers, M. L. et al. Characterizing adult sleep behavior over 20 years-the Population-Based Doetinchem Cohort Study. Sleep40, 10.1093/sleep/zsx085 IF: 5.6 Q1 (2017). [PubMed]
12. Zomers, M. L. 等人。表征 20 年来的成人睡眠行为 - 基于人群的 Doetinchem 队列研究。Sleep40,10.1093/sleep/zsx085IF:5.6 第一季度(2017 年)。[ 出版]
13. American Academy of Sleep Medicine. International Classification Of Sleep Disorders 3rd edn (American Academy of Sleep Medicine, 2014).
13. 美国睡眠医学会。国际睡眠障碍分类第 3 版(美国睡眠医学会,2014 年)。
14. Ford ES, Cunningham TJ, Croft JB. Trends in self-reported sleep duration among US adults from 1985 to 2012. Sleep. 2015;38:829–832. [PMC free article] [PubMed] []
14. 福特 ES、坎宁安 TJ、克罗夫特 JB。1985 年至 2012 年美国成年人自我报告的睡眠持续时间趋势。睡。2015;38:829–832.[ PMC免费商品][ 出版][ 谷歌学术搜索]
15. Gilmour H, et al. Longitudinal trajectories of sleep duration in the general population. Health Rep. 2013;24:14–20. [PubMed] []
15. Gilmour H 等人。一般人群睡眠持续时间的纵向轨迹。2013 年健康代表;24:14–20.[ 出版][ 谷歌学术搜索]
16. Matricciani L, Olds T, Petkov J. In search of lost sleep: secular trends in the sleep time of school-aged children and adolescents. Sleep. Med. Rev. 2012;16:203–211. [PubMed] []
17. Wheaton AG, Jones SE, Cooper AC, Croft JB. Short sleep duration among middle school and high school students - United States, 2015. MMWR Morb. Mortal. Wkly Rep. 2018;67:85–90. [PMC free article] [PubMed] []
18. Kocevska D, et al. Sleep characteristics across the lifespan in 1.1 million people from the Netherlands, United Kingdom and United States: a systematic review and meta-analysis. Nat. Hum. Behav. 2020 doi: 10.1038/s41562-020-00965-x IF: 29.9 Q1 . [PubMed] [CrossRef] []
19. Pandi-Perumal SR, et al. Racial/ethnic and social inequities in sleep medicine: the tip of the iceberg? J. Natl Med. Assoc. 2017;109:279–286. [PubMed] []
20. Irwin MR. Why sleep is important for health: a psychoneuroimmunology perspective. Annu. Rev. Psychol. 2015;66:143–172. [PMC free article] [PubMed] []
21. Vgontzas AN, Liao D, Bixler EO, Chrousos GP, Vela-Bueno A. Insomnia with objective short sleep duration is associated with a high risk for hypertension. Sleep. 2009;32:491–497. [PMC free article] [PubMed] []
22. Vgontzas AN, Fernandez-Mendoza J, Liao D, Bixler EO. Insomnia with objective short sleep duration: the most biologically severe phenotype of the disorder. Sleep. Med. Rev. 2013;17:241–254. [PMC free article] [PubMed] []
23. Smagula SF, et al. Actigraphy- and polysomnography-measured sleep disturbances, inflammation, and mortality among older men. Psychosom. Med. 2016;78:686–696. [PMC free article] [PubMed] []
24. Cappuccio FP, Miller MA. Sleep and cardio-metabolic disease. Curr. Cardiol. Rep. 2017;19:110. [PMC free article] [PubMed] []
25. Cappuccio FP, Cooper D, D’Elia L, Strazzullo P, Miller MA. Sleep duration predicts cardiovascular outcomes: a systematic review and meta-analysis of prospective studies. Eur. Heart J. 2011;32:1484–1492. [PubMed] []
26. von Ruesten A, Weikert C, Fietze I, Boeing H. Association of sleep duration with chronic diseases in the European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam study. PLoS ONE. 2012;7:e30972. [PMC free article] [PubMed] []
27. Tobaldini E, et al. Sleep, sleep deprivation, autonomic nervous system and cardiovascular diseases. Neurosci. Biobehav. Rev. 2017;74:321–329. [PubMed] []
28. Kochanek, K. D., Xu, J. & Arias, E. Mortality in the United States, 2019. NCHS Data Brief No. 395, 1–8 (CDC, 2020). [PubMed]
29. Kakizaki M, et al. Sleep duration and the risk of breast cancer: the Ohsaki Cohort Study. Br. J. Cancer. 2008;99:1502–1505. [PMC free article] [PubMed] []
30. Ahmadian N, Hejazi S, Mahmoudi J, Talebi M. Tau pathology of Alzheimer disease: possible role of sleep deprivation. Basic Clin. Neurosci. 2018;9:307–316. [PMC free article] [PubMed] []
31. Baglioni C, et al. Insomnia as a predictor of depression: a meta-analytic evaluation of longitudinal epidemiological studies. J. Affect. Disord. 2011;135:10–19. [PubMed] []
32. Ben Simon E, Vallat R, Barnes CM, Walker MP. Sleep loss and the socio-emotional brain. Trends Cogn. Sci. 2020;24:435–450. [PubMed] []
33. Bernert RA, Kim JS, Iwata NG, Perlis ML. Sleep disturbances as an evidence-based suicide risk factor. Curr. Psychiatry Rep. 2015;17:554. [PMC free article] [PubMed] []
34. Fredriksen K, Rhodes J, Reddy R, Way N. Sleepless in Chicago: tracking the effects of adolescent sleep loss during the middle school years. Child Dev. 2004;75:84–95. [PubMed] []
35. Tomaso, C. C., Johnson, A. B. & Nelson, T. D. The effect of sleep deprivation and restriction on mood, emotion, and emotion regulation: three meta-analyses in one. Sleep44, 10.1093/sleep/zsaa289 IF: 5.6 Q1 (2021). [PubMed]
36. Waters F, Chiu V, Atkinson A, Blom JD. Severe sleep deprivation causes hallucinations and a gradual progression toward psychosis with increasing time awake. Front. Psychiatry. 2018;9:303. [PMC free article] [PubMed] []
37. Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Sleep duration and all-cause mortality: a systematic review and meta-analysis of prospective studies. Sleep. 2010;33:585–592. [PMC free article] [PubMed] []
38. Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Quantity and quality of sleep and incidence of type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2010;33:414–420. [PMC free article] [PubMed] []
39. Bacaro V, et al. Sleep duration and obesity in adulthood: an updated systematic review and meta-analysis. Obes. Res. Clin. Pract. 2020;14:301–309. [PubMed] []
40. Bishir M, et al. Sleep deprivation and neurological disorders. Biomed. Res. Int. 2020;2020:5764017. [PMC free article] [PubMed] []
41. Mullington JM, Simpson NS, Meier-Ewert HK, Haack M. Sleep loss and inflammation. Best. Pract. Res. Clin. Endocrinol. Metab. 2010;24:775–784. [PMC free article] [PubMed] []
42. Aldabal L, Bahammam AS. Metabolic, endocrine, and immune consequences of sleep deprivation. Open Respir. Med. J. 2011;5:31–43. [PMC free article] [PubMed] []
43. Dantzer R. Neuroimmune interactions: from the brain to the immune system and vice versa. Physiol. Rev. 2018;98:477–504. [PMC free article] [PubMed] []
44. Irwin MR. Sleep and inflammation: partners in sickness and in health. Nat. Rev. Immunol. 2019;19:702–715. [PubMed] []
45. Hand TW, Vujkovic-Cvijin I, Ridaura VK, Belkaid Y. Linking the microbiota, chronic disease, and the immune system. Trends Endocrinol. Metab. 2016;27:831–843. [PMC free article] [PubMed] []
46. Irwin MR, Opp MR. Sleep health: reciprocal regulation of sleep and innate immunity. Neuropsychopharmacology. 2017;42:129–155. [PMC free article] [PubMed] []
47. Miller MA, Cappuccio FP. Inflammation, sleep, obesity and cardiovascular disease. Curr. Vasc. Pharm. 2007;5:93–102. [PubMed] []
48. Krueger JM, Pappenheimer JR, Karnovsky ML. The composition of sleep-promoting factor isolated from human urine. J. Biol. Chem. 1982;257:1664–1669. [PubMed] []
49. Mullington J, et al. Dose-dependent effects of endotoxin on human sleep. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000;278:R947–955. [PubMed] []
50. Zielinski MR, Krueger JM. Sleep and innate immunity. Front. Biosci. (Sch. Ed.) 2011;3:632–642. [PMC free article] [PubMed] []
51. Opp MR. Cytokines and sleep. Sleep. Med. Rev. 2005;9:355–364. [PubMed] []
52. Urade Y, Hayaishi O. Prostaglandin D2 and sleep/wake regulation. Sleep. Med. Rev. 2011;15:411–418. [PubMed] []
53. Krueger JM, Majde JA, Rector DM. Cytokines in immune function and sleep regulation. Handb. Clin. Neurol. 2011;98:229–240. [PMC free article] [PubMed] []
54. Besedovsky L, Lange T, Haack M. The sleep-immune crosstalk in health and disease. Physiol. Rev. 2019;99:1325–1380. [PMC free article] [PubMed] []
55. Opp MR, Obal F, Jr., Krueger JM. Interleukin 1 alters rat sleep: temporal and dose-related effects. Am. J. Physiol. 1991;260:R52–58. [PubMed] []
56. Krueger JM, Obal F., Jr. Growth hormone-releasing hormone and interleukin-1 in sleep regulation. FASEB J. 1993;7:645–652. [PubMed] []
57. De A, Churchill L, Obal F, Jr., Simasko SM, Krueger JM. GHRH and IL1beta increase cytoplasmic Ca(2+) levels in cultured hypothalamic GABAergic neurons. Brain Res. 2002;949:209–212. [PubMed] []
58. Kubota T, Fang J, Kushikata T, Krueger JM. Interleukin-13 and transforming growth factor-beta1 inhibit spontaneous sleep in rabbits. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000;279:R786–792. [PubMed] []
59. Kubota T, Fang J, Guan Z, Brown RA, Krueger JM. Vagotomy attenuates tumor necrosis factor-alpha-induced sleep and EEG delta-activity in rats. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2001;280:R1213–1220. [PubMed] []
60. Curfs JH, Meis JF, Hoogkamp-Korstanje JA. A primer on cytokines: sources, receptors, effects, and inducers. Clin. Microbiol. Rev. 1997;10:742–780. [PMC free article] [PubMed] []
61. Hodo TW, de Aquino MTP, Shimamoto A, Shanker A. Critical neurotransmitters in the neuroimmune network. Front Immunol. 2020;11:1869. [PMC free article] [PubMed] []
62. Huang X, Hussain B, Chang J. Peripheral inflammation and blood-brain barrier disruption: effects and mechanisms. CNS Neurosci. Ther. 2021;27:36–47. [PMC free article] [PubMed] []
63. Grill HJ, Hayes MR. Hindbrain neurons as an essential hub in the neuroanatomically distributed control of energy balance. Cell Metab. 2012;16:296–309. [PMC free article] [PubMed] []
64. Kunz N, Kemper C. Complement has brains-do intracellular complement and immunometabolism cooperate in tissue homeostasis and behavior? Front. Immunol. 2021;12:629986. [PMC free article] [PubMed] []
65. Levin SG, Godukhin OV. Modulating effect of cytokines on mechanisms of synaptic plasticity in the brain. Biochemistry (Mosc.) 2017;82:264–274. [PubMed] []
66. Wang, Y. et al. Astrocyte-secreted IL-33 mediates homeostatic synaptic plasticity in the adult hippocampus. Proc. Natl Acad. Sci. USA118, 10.1073/pnas.2020810118 IF: 11.1 Q1 (2021). [PubMed]
67. Turnbull AV, Rivier C. Regulation of the HPA axis by cytokines. Brain Behav. Immun. 1995;9:253–275. [PubMed] []
68. Besedovsky HO, del Rey A. The cytokine-HPA axis feed-back circuit. Z. Rheumatol. 2000;59(Suppl 2):II/26–30. [PubMed] []
69. Nguyen JT, et al. The neuron-specific interleukin-1 receptor accessory protein alters emergent network state properties in vitro. Neurobiol. Sleep. Circadian Rhythms. 2019;6:35–43. [PMC free article] [PubMed] []
70. Krueger JM, Obal F. A neuronal group theory of sleep function. J. Sleep. Res. 1993;2:63–69. [PubMed] []
71. Rector DM, Topchiy IA, Carter KM, Rojas MJ. Local functional state differences between rat cortical columns. Brain Res. 2005;1047:45–55. [PubMed] []
72. Roy S, Krueger JM, Rector DM, Wan Y. A network model for activity-dependent sleep regulation. J. Theor. Biol. 2008;253:462–468. [PMC free article] [PubMed] []
73. Krueger JM, Huang YH, Rector DM, Buysse DJ. Sleep: a synchrony of cell activity-driven small network states. Eur. J. Neurosci. 2013;38:2199–2209. [PMC free article] [PubMed] []
74. Krueger JM. Sleep and circadian rhythms: evolutionary entanglement and local regulation. Neurobiol. Sleep. Circadian Rhythms. 2020;9:100052. [PMC free article] [PubMed] []
75. Krueger JM, Nguyen JT, Dykstra-Aiello CJ, Taishi P. Local sleep. Sleep. Med. Rev. 2019;43:14–21. [PMC free article] [PubMed] []
76. Taishi P, Churchill L, De A, Obal F, Jr., Krueger JM. Cytokine mRNA induction by interleukin-1beta or tumor necrosis factor alpha in vitro and in vivo. Brain Res. 2008;1226:89–98. [PMC free article] [PubMed] []
77. Rockstrom MD, et al. Tumor necrosis factor alpha in sleep regulation. Sleep. Med. Rev. 2018;40:69–78. [PMC free article] [PubMed] []
78. Krueger JM, Walter J, Dinarello CA, Wolff SM, Chedid L. Sleep-promoting effects of endogenous pyrogen (interleukin-1) Am. J. Physiol. 1984;246:R994–999. [PubMed] []
79. Imeri L, Opp MR. How (and why) the immune system makes us sleep. Nat. Rev. Neurosci. 2009;10:199–210. [PMC free article] [PubMed] []
80. Jewett KA, Krueger JM. Humoral sleep regulation; interleukin-1 and tumor necrosis factor. Vitam. Horm. 2012;89:241–257. [PMC free article] [PubMed] []
81. Davis CJ, et al. The neuron-specific interleukin-1 receptor accessory protein is required for homeostatic sleep and sleep responses to influenza viral challenge in mice. Brain Behav. Immun. 2015;47:35–43. [PMC free article] [PubMed] []
82. Churchill L, et al. Brain distribution of cytokine mRNA induced by systemic administration of interleukin-1beta or tumor necrosis factor alpha. Brain Res. 2006;1120:64–73. [PubMed] []
83. Taishi P, Bredow S, Guha-Thakurta N, Obal F, Jr., Krueger JM. Diurnal variations of interleukin-1 beta mRNA and beta-actin mRNA in rat brain. J. Neuroimmunol. 1997;75:69–74. [PubMed] []
84. Bredow S, Guha-Thakurta N, Taishi P, Obal F, Jr., Krueger JM. Diurnal variations of tumor necrosis factor alpha mRNA and alpha-tubulin mRNA in rat brain. Neuroimmunomodulation. 1997;4:84–90. [PubMed] []
85. Floyd RA, Krueger JM. Diurnal variation of TNF alpha in the rat brain. Neuroreport. 1997;8:915–918. [PubMed] []
86. Garlanda C, Dinarello CA, Mantovani A. The interleukin-1 family: back to the future. Immunity. 2013;39:1003–1018. [PMC free article] [PubMed] []
87. Smith DE, et al. A central nervous system-restricted isoform of the interleukin-1 receptor accessory protein modulates neuronal responses to interleukin-1. Immunity. 2009;30:817–831. [PMC free article] [PubMed] []
88. Taishi P, et al. Brain-specific interleukin-1 receptor accessory protein in sleep regulation. J. Appl Physiol. (1985) 2012;112:1015–1022. [PMC free article] [PubMed] []
89. Oles V, et al. Sleep- and time of day-linked RNA transcript expression in wild-type and IL1 receptor accessory protein-null mice. J. Appl. Physiol. (1985) 2020;128:1506–1522. [PMC free article] [PubMed] []
90. Dykstra-Aiello C, et al. A wake-like state in vitro induced by transmembrane TNF/soluble TNF receptor reverse signaling. Brain Behav. Immun. 2021;94:245–258. [PMC free article] [PubMed] []
91. Guan Y, et al. Astrocytes constitute the major TNF-alpha-producing cell population in the infarct cortex in dMCAO rats receiving intravenous MSC infusion. Biomed. Pharmacother. 2021;142:111971. [PubMed] []
92. Churchill L, et al. Unilateral cortical application of tumor necrosis factor alpha induces asymmetry in Fos- and interleukin-1beta-immunoreactive cells within the corticothalamic projection. Brain Res. 2005;1055:15–24. [PubMed] []
93. Jewett KA, et al. Tumor necrosis factor enhances the sleep-like state and electrical stimulation induces a wake-like state in co-cultures of neurons and glia. Eur. J. Neurosci. 2015;42:2078–2090. [PMC free article] [PubMed] []
94. Schmidt MH. The energy allocation function of sleep: a unifying theory of sleep, torpor, and continuous wakefulness. Neurosci. Biobehav. Rev. 2014;47:122–153. [PubMed] []
95. Sharpley AL, Cooper CM, Williams C, Godlewska BR, Cowen PJ. Effects of typhoid vaccine on inflammation and sleep in healthy participants: a double-blind, placebo-controlled, crossover study. Psychopharmacology (Berl.) 2016;233:3429–3435. [PMC free article] [PubMed] []
96. Toth LA, Tolley EA, Krueger JM. Sleep as a prognostic indicator during infectious disease in rabbits. Proc. Soc. Exp. Biol. Med. 1993;203:179–192. [PubMed] []
97. Toth LA, Krueger JM. Alteration of sleep in rabbits by Staphylococcus aureus infection. Infect. Immun. 1988;56:1785–1791. [PMC free article] [PubMed] []
98. Seke Etet PF, et al. Sleep and rhythm changes at the time of Trypanosoma brucei invasion of the brain parenchyma in the rat. Chronobiol. Int. 2012;29:469–481. [PubMed] []
99. Toth LA, Tolley EA, Broady R, Blakely B, Krueger JM. Sleep during experimental trypanosomiasis in rabbits. Proc. Soc. Exp. Biol. Med. 1994;205:174–181. [PubMed] []
100. Patke A, Young MW, Axelrod S. Molecular mechanisms and physiological importance of circadian rhythms. Nat. Rev. Mol. Cell Biol. 2020;21:67–84. [PubMed] []
101. Cavadini G, et al. TNF-alpha suppresses the expression of clock genes by interfering with E-box-mediated transcription. Proc. Natl Acad. Sci. USA. 2007;104:12843–12848. [PMC free article] [PubMed] []
102. Meier D, Lopez M, Franken P, Fontana A. Twist1 is a TNF-inducible inhibitor of clock mediated activation of period genes. PLoS ONE. 2015;10:e0137229. [PMC free article] [PubMed] []
103. Marpegan L, Bekinschtein TA, Costas MA, Golombek DA. Circadian responses to endotoxin treatment in mice. J. Neuroimmunol. 2005;160:102–109. [PubMed] []
104. Yamamura Y, Yano I, Kudo T, Shibata S. Time-dependent inhibitory effect of lipopolysaccharide injection on Per1 and Per2 gene expression in the mouse heart and liver. Chronobiol. Int. 2010;27:213–232. [PubMed] []
105. Wang Y, et al. Endotoxin disrupts circadian rhythms in macrophages via reactive oxygen species. PLoS ONE. 2016;11:e0155075. [PMC free article] [PubMed] []
106. Friese RS, Bruns B, Sinton CM. Sleep deprivation after septic insult increases mortality independent of age. J. Trauma. 2009;66:50–54. [PubMed] []
107. Everson CA, Toth LA. Systemic bacterial invasion induced by sleep deprivation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2000;278:R905–916. [PubMed] []
108. Opp MR. Sleeping to fuel the immune system: mammalian sleep and resistance to parasites. BMC Evol. Biol. 2009;9:8. [PMC free article] [PubMed] []
109. Prather AA, Janicki-Deverts D, Hall MH, Cohen S. Behaviorally assessed sleep and susceptibility to the common cold. Sleep. 2015;38:1353–1359. [PMC free article] [PubMed] []
110. Fernandes ER, et al. Sleep disturbance during infection compromises Tfh differentiation and impacts host immunity. iScience. 2020;23:101599. [PMC free article] [PubMed] []
111. Chung WS, Lin HH, Cheng NC. The incidence and risk of herpes zoster in patients with sleep disorders: a population-based cohort study. Medicine (Baltimore) 2016;95:e2195. [PMC free article] [PubMed] []
112. Patel SR, et al. A prospective study of sleep duration and pneumonia risk in women. Sleep. 2012;35:97–101. [PMC free article] [PubMed] []
113. Prather AA, Leung CW. Association of insufficient sleep with respiratory infection among adults in the United States. JAMA Intern. Med. 2016;176:850–852. [PMC free article] [PubMed] []
114. Cohen S, Doyle WJ, Alper CM, Janicki-Deverts D, Turner RB. Sleep habits and susceptibility to the common cold. Arch. Intern. Med. 2009;169:62–67. [PMC free article] [PubMed] []
115. Orzech KM, Acebo C, Seifer R, Barker D, Carskadon MA. Sleep patterns are associated with common illness in adolescents. J. Sleep. Res. 2014;23:133–142. [PMC free article] [PubMed] []
116. Zhang Y, Xie B, Chen X, Zhang J, Yuan S. A key role of gut microbiota-vagus nerve/spleen axis in sleep deprivation-mediated aggravation of systemic inflammation after LPS administration. Life Sci. 2021;265:118736. [PubMed] []
117. Lange T, Dimitrov S, Bollinger T, Diekelmann S, Born J. Sleep after vaccination boosts immunological memory. J. Immunol. 2011;187:283–290. [PubMed] []
118. Prather AA, et al. Sleep and antibody response to hepatitis B vaccination. Sleep. 2012;35:1063–1069. [PMC free article] [PubMed] []
119. Renegar KB, Floyd RA, Krueger JM. Effects of short-term sleep deprivation on murine immunity to influenza virus in young adult and senescent mice. Sleep. 1998;21:241–248. [PubMed] []
120. Hurley S, Goldberg D, Bernstein L, Reynolds P. Sleep duration and cancer risk in women. Cancer Causes Control. 2015;26:1037–1045. [PMC free article] [PubMed] []
121. Kakizaki M, et al. Sleep duration and the risk of prostate cancer: the Ohsaki Cohort Study. Br. J. Cancer. 2008;99:176–178. [PMC free article] [PubMed] []
122. Jiao L, et al. Sleep duration and incidence of colorectal cancer in postmenopausal women. Br. J. Cancer. 2013;108:213–221. [PMC free article] [PubMed] []
123. Cao J, et al. Sleep duration and risk of breast cancer: The JACC Study. Breast Cancer Res. Treat. 2019;174:219–225. [PubMed] []
124. Wu AH, et al. Sleep duration, melatonin and breast cancer among Chinese women in Singapore. Carcinogenesis. 2008;29:1244–1248. [PMC free article] [PubMed] []
125. Wehr TA. The durations of human melatonin secretion and sleep respond to changes in daylength (photoperiod) J. Clin. Endocrinol. Metab. 1991;73:1276–1280. [PubMed] []
126. Luo J, et al. Effect of melatonin on T/B cell activation and immune regulation in pinealectomy mice. Life Sci. 2020;242:117191. [PubMed] []
127. De Lorenzo BH, de Oliveira Marchioro L, Greco CR, Suchecki D. Sleep-deprivation reduces NK cell number and function mediated by beta-adrenergic signalling. Psychoneuroendocrinology. 2015;57:134–143. []
128. De Lorenzo BHP, et al. Chronic sleep restriction impairs the antitumor immune response in mice. Neuroimmunomodulation. 2018;25:59–67. [PubMed] []
129. Maragno-Correa JM, et al. Sleep deprivation increases mortality in female mice bearing Ehrlich ascitic tumor. Neuroimmunomodulation. 2013;20:134–140. [PubMed] []
130. Barbosa Vieira TK, et al. Correlation between circadian rhythm related genes, type 2 diabetes, and cancer: insights from metanalysis of transcriptomics data. Mol. Cell Endocrinol. 2021;526:111214. [PubMed] []
131. Gonzalez H, Hagerling C, Werb Z. Roles of the immune system in cancer: from tumor initiation to metastatic progression. Genes Dev. 2018;32:1267–1284. [PMC free article] [PubMed] []
132. Palma JA, Urrestarazu E, Iriarte J. Sleep loss as risk factor for neurologic disorders: a review. Sleep. Med. 2013;14:229–236. [PubMed] []
133. Sabia S, et al. Association of sleep duration in middle and old age with incidence of dementia. Nat. Commun. 2021;12:2289. [PMC free article] [PubMed] []
134. Sadeghmousavi S, Eskian M, Rahmani F, Rezaei N. The effect of insomnia on development of Alzheimer’s disease. J. Neuroinflammation. 2020;17:289. [PMC free article] [PubMed] []
135. Stephenson J, Nutma E, van der Valk P, Amor S. Inflammation in CNS neurodegenerative diseases. Immunology. 2018;154:204–219. [PMC free article] [PubMed] []
136. Manchanda S, Singh H, Kaur T, Kaur G. Low-grade neuroinflammation due to chronic sleep deprivation results in anxiety and learning and memory impairments. Mol. Cell Biochem. 2018;449:63–72. [PubMed] []
137. Xue R, et al. Nicotinic mitigation of neuroinflammation and oxidative stress after chronic sleep deprivation. Front. Immunol. 2019;10:2546. [PMC free article] [PubMed] []
138. Hurtado-Alvarado G, et al. The yin/yang of inflammatory status: blood-brain barrier regulation during sleep. Brain Behav. Immun. 2018;69:154–166. [PubMed] []
139. Korin, B. et al. Short-term sleep deprivation in mice induces B cell migration to the brain compartment. Sleep43, 10.1093/sleep/zsz222 IF: 5.6 Q1 (2020). []
140. Luo, F. et al. Melatonin and autophagy in aging-related neurodegenerative diseases. Int. J. Mol. Sci.21, 10.3390/ijms21197174 IF: 5.6 Q1 (2020). [PubMed]
141. Finkbeiner E, Haindl M, Raman N, Muller S. SUMO routes ribosome maturation. Nucleus. 2011;2:527–532. [PubMed] []
142. Zhao HY, et al. Chronic sleep restriction induces cognitive deficits and cortical beta-amyloid deposition in mice via BACE1-antisense activation. CNS Neurosci. Ther. 2017;23:233–240. [PMC free article] [PubMed] []
143. Liu P, et al. Activation of inflammation is associated with amyloid-beta accumulation induced by chronic sleep restriction in rats. J. Alzheimers Dis. 2020;74:759–773. [PubMed] []
144. Shokri-Kojori E, et al. beta-Amyloid accumulation in the human brain after one night of sleep deprivation. Proc. Natl Acad. Sci. USA. 2018;115:4483–4488. [PMC free article] [PubMed] []
145. Spira AP, et al. Self-reported sleep and beta-amyloid deposition in community-dwelling older adults. JAMA Neurol. 2013;70:1537–1543. [PMC free article] [PubMed] []
146. Ju YS, et al. Slow wave sleep disruption increases cerebrospinal fluid amyloid-beta levels. Brain. 2017;140:2104–2111. [PMC free article] [PubMed] []
147. Chen DW, Wang J, Zhang LL, Wang YJ, Gao CY. Cerebrospinal fluid amyloid-beta levels are increased in patients with insomnia. J. Alzheimers Dis. 2018;61:645–651. [PubMed] []
148. Ooms S, et al. Effect of 1 night of total sleep deprivation on cerebrospinal fluid beta-amyloid 42 in healthy middle-aged men: a randomized clinical trial. JAMA Neurol. 2014;71:971–977. [PubMed] []
149. Zhao B, et al. Chronic sleep restriction induces abeta accumulation by disrupting the balance of abeta production and clearance in rats. Neurochem. Res. 2019;44:859–873. [PubMed] []
150. Wu H, Dunnett S, Ho YS, Chang RC. The role of sleep deprivation and circadian rhythm disruption as risk factors of Alzheimer’s disease. Front. Neuroendocrinol. 2019;54:100764. [PubMed] []
151. Jaeger LB, et al. Lipopolysaccharide alters the blood-brain barrier transport of amyloid beta protein: a mechanism for inflammation in the progression of Alzheimer’s disease. Brain Behav. Immun. 2009;23:507–517. [PMC free article] [PubMed] []
152. Sastre M, et al. Nonsteroidal anti-inflammatory drugs and peroxisome proliferator-activated receptor-gamma agonists modulate immunostimulated processing of amyloid precursor protein through regulation of beta-secretase. J. Neurosci. 2003;23:9796–9804. [PMC free article] [PubMed] []
153. Mouton-Liger F, et al. Oxidative stress increases BACE1 protein levels through activation of the PKR-eIF2alpha pathway. Biochim. Biophys. Acta. 2012;1822:885–896. [PubMed] []
154. Giese M, et al. BDNF: an indicator of insomnia? Mol. Psychiatry. 2014;19:151–152. [PMC free article] [PubMed] []
155. Tan DX, Manchester LC, Terron MP, Flores LJ, Reiter RJ. One molecule, many derivatives: a never-ending interaction of melatonin with reactive oxygen and nitrogen species? J. Pineal Res. 2007;42:28–42. [PubMed] []
156. Zhang L, et al. Melatonin ameliorates cognitive impairment induced by sleep deprivation in rats: role of oxidative stress, BDNF and CaMKII. Behav. Brain Res. 2013;256:72–81. [PubMed] []
157. Fredrich M, Hampel M, Seidel K, Christ E, Korf HW. Impact of melatonin receptor-signaling on Zeitgeber time-dependent changes in cell proliferation and apoptosis in the adult murine. Hippocampus. 2017;27:495–506. [PubMed] []
158. Alghamdi BS. The neuroprotective role of melatonin in neurological disorders. J. Neurosci. Res. 2018;96:1136–1149. [PMC free article] [PubMed] []
159. Luengo E, et al. Pharmacological doses of melatonin impede cognitive decline in tau-related Alzheimer models, once tauopathy is initiated, by restoring the autophagic flux. J. Pineal Res. 2019;67:e12578. [PubMed] []
160. Emamian F, et al. The association between obstructive sleep apnea and Alzheimer’s disease: a meta-analysis perspective. Front. Aging Neurosci. 2016;8:78. [PMC free article] [PubMed] []
161. Osorio RS, et al. Sleep-disordered breathing advances cognitive decline in the elderly. Neurology. 2015;84:1964–1971. [PMC free article] [PubMed] []
162. Frenkel D, et al. Scara1 deficiency impairs clearance of soluble amyloid-beta by mononuclear phagocytes and accelerates Alzheimer’s-like disease progression. Nat. Commun. 2013;4:2030. [PMC free article] [PubMed] []
163. Musiek ES, Holtzman DM. Mechanisms linking circadian clocks, sleep, and neurodegeneration. Science. 2016;354:1004–1008. [PMC free article] [PubMed] []
164. Wang J, Gu BJ, Masters CL, Wang YJ. A systemic view of Alzheimer disease - insights from amyloid-beta metabolism beyond the brain. Nat. Rev. Neurol. 2017;13:703. [PubMed] []
165. Tanner CM, Aston DA. Epidemiology of Parkinson’s disease and akinetic syndromes. Curr. Opin. Neurol. 2000;13:427–430. [PubMed] []
166. Barber A, Dashtipour K. Sleep disturbances in Parkinson’s disease with emphasis on rapid eye movement sleep behavior disorder. Int. J. Neurosci. 2012;122:407–412. [PubMed] []
167. Barnett R. Parkinson’s disease. Lancet. 2016;387:217. [PubMed] []
168. Hsiao YH, et al. Sleep disorders and an increased risk of Parkinson’s disease in individuals with non-apnea sleep disorders: a population-based cohort study. J. Sleep. Res. 2017;26:623–628. [PubMed] []
169. Bohnen NI, Hu MTM. Sleep disturbance as potential risk and progression factor for Parkinson’s disease. J. Parkinsons Dis. 2019;9:603–614. [PMC free article] [PubMed] []
170. Su LY, et al. Melatonin attenuates MPTP-induced neurotoxicity via preventing CDK5-mediated autophagy and SNCA/alpha-synuclein aggregation. Autophagy. 2015;11:1745–1759. [PMC free article] [PubMed] []
171. Srivastava AK, Roy Choudhury S, Karmakar S. Melatonin/polydopamine nanostructures for collective neuroprotection-based Parkinson’s disease therapy. Biomater. Sci. 2020;8:1345–1363. [PubMed] []
172. Abad VC, Sarinas PS, Guilleminault C. Sleep and rheumatologic disorders. Sleep. Med. Rev. 2008;12:211–228. [PubMed] []
173. Hsiao YH, et al. Sleep disorders and increased risk of autoimmune diseases in individuals without sleep apnea. Sleep. 2015;38:581–586. [PMC free article] [PubMed] []
174. Young KA, et al. Less than 7 hours of sleep per night is associated with transitioning to systemic lupus erythematosus. Lupus. 2018;27:1524–1531. [PMC free article] [PubMed] []
175. Palma BD, Gabriel A, Jr., Colugnati FA, Tufik S. Effects of sleep deprivation on the development of autoimmune disease in an experimental model of systemic lupus erythematosus. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2006;291:R1527–1532. [PubMed] []
176. Alarcon-Riquelme ME, Moller G, Fernandez C. Age-dependent responsiveness to interleukin-6 in B lymphocytes from a systemic lupus erythematosus-prone (NZB x NZW)F1 hybrid. Clin. Immunol. Immunopathol. 1992;62:264–269. [PubMed] []
177. Yehuda S, Sredni B, Carasso RL, Kenigsbuch-Sredni D. REM sleep deprivation in rats results in inflammation and interleukin-17 elevation. J. Interferon Cytokine Res. 2009;29:393–398. [PubMed] []
178. Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells. Nat. Immunol. 2007;8:942–949. [PubMed] []
179. Volin, M. V. & Shahrara, S. Role of TH-17 cells in rheumatic and other autoimmune diseases. Rheumatology (Sunnyvale)1, 10.4172/2161-1149.1000104 (2011). [PubMed]
180. Bollinger T, et al. Sleep-dependent activity of T cells and regulatory T cells. Clin. Exp. Immunol. 2009;155:231–238. [PMC free article] [PubMed] []
181. Carbone F, et al. Regulatory T cell proliferative potential is impaired in human autoimmune disease. Nat. Med. 2014;20:69–74. [PubMed] []
182. Leng Y, et al. Sleep duration and risk of fatal and nonfatal stroke: a prospective study and meta-analysis. Neurology. 2015;84:1072–1079. [PMC free article] [PubMed] []
183. Ferrie JE, et al. Associations between change in sleep duration and inflammation: findings on C-reactive protein and interleukin 6 in the Whitehall II Study. Am. J. Epidemiol. 2013;178:956–961. [PMC free article] [PubMed] []
184. Wang YH, et al. Association of longitudinal patterns of habitual sleep duration with risk of cardiovascular events and all-cause mortality. JAMA Netw. Open. 2020;3:e205246. [PMC free article] [PubMed] []
185. Dominguez F, et al. Association of sleep duration and quality with subclinical atherosclerosis. J. Am. Coll. Cardiol. 2019;73:134–144. [PubMed] []
186. Cappuccio FP, et al. Meta-analysis of short sleep duration and obesity in children and adults. Sleep. 2008;31:619–626. [PMC free article] [PubMed] []
187. Meng L, Zheng Y, Hui R. The relationship of sleep duration and insomnia to risk of hypertension incidence: a meta-analysis of prospective cohort studies. Hypertens. Res. 2013;36:985–995. [PMC free article] [PubMed] []
188. Anothaisintawee T, Reutrakul S, Van Cauter E, Thakkinstian A. Sleep disturbances compared to traditional risk factors for diabetes development: systematic review and meta-analysis. Sleep. Med. Rev. 2016;30:11–24. [PubMed] []
189. Zhong X, et al. Increased sympathetic and decreased parasympathetic cardiovascular modulation in normal humans with acute sleep deprivation. J. Appl. Physiol. (1985) 2005;98:2024–2032. [PubMed] []
190. Meier-Ewert HK, et al. Effect of sleep loss on C-reactive protein, an inflammatory marker of cardiovascular risk. J. Am. Coll. Cardiol. 2004;43:678–683. [PubMed] []
191. Sauvet F, et al. Effect of acute sleep deprivation on vascular function in healthy subjects. J. Appl. Physiol. (1985) 2010;108:68–75. [PubMed] []
192. Dettoni JL, et al. Cardiovascular effects of partial sleep deprivation in healthy volunteers. J. Appl. Physiol. (1985) 2012;113:232–236. [PubMed] []
193. Spiegel K, Tasali E, Leproult R, Van Cauter E. Effects of poor and short sleep on glucose metabolism and obesity risk. Nat. Rev. Endocrinol. 2009;5:253–261. [PMC free article] [PubMed] []
194. Buxton OM, et al. Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci. Transl. Med. 2012;4:129ra143. [PMC free article] [PubMed] []
195. Broussard JL, Ehrmann DA, Van Cauter E, Tasali E, Brady MJ. Impaired insulin signaling in human adipocytes after experimental sleep restriction: a randomized, crossover study. Ann. Intern. Med. 2012;157:549–557. [PMC free article] [PubMed] []
196. Nedeltcheva AV, Kilkus JM, Imperial J, Schoeller DA, Penev PD. Insufficient sleep undermines dietary efforts to reduce adiposity. Ann. Intern. Med. 2010;153:435–441. [PMC free article] [PubMed] []
197. Darukhanavala A, et al. Changes in insulin secretion and action in adults with familial risk for type 2 diabetes who curtail their sleep. Diabetes Care. 2011;34:2259–2264. [PMC free article] [PubMed] []
198. Esser N, Legrand-Poels S, Piette J, Scheen AJ, Paquot N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res. Clin. Pract. 2014;105:141–150. [PubMed] []
199. Longo, M. et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int. J. Mol. Sci.20, 10.3390/ijms20092358 IF: 5.6 Q1 (2019). [PubMed]
200. Freitas Lima LC, et al. Adipokines, diabetes and atherosclerosis: an inflammatory association. Front. Physiol. 2015;6:304. [PMC free article] [PubMed] []
201. Geovanini GR, Libby P. Atherosclerosis and inflammation: overview and updates. Clin. Sci. (Lond.) 2018;132:1243–1252. [PubMed] []
202. Yousuf O, et al. High-sensitivity C-reactive protein and cardiovascular disease: a resolute belief or an elusive link? J. Am. Coll. Cardiol. 2013;62:397–408. [PubMed] []
203. van Leeuwen WM, et al. Sleep restriction increases the risk of developing cardiovascular diseases by augmenting proinflammatory responses through IL-17 and CRP. PLoS ONE. 2009;4:e4589. [PMC free article] [PubMed] []
204. Irwin MR, Olmstead R, Carroll JE. Sleep disturbance, sleep duration, and inflammation: a systematic review and meta-analysis of cohort studies and experimental sleep deprivation. Biol. Psychiatry. 2016;80:40–52. [PMC free article] [PubMed] []
205. McAlpine CS, et al. Sleep modulates haematopoiesis and protects against atherosclerosis. Nature. 2019;566:383–387. [PMC free article] [PubMed] []
206. Dutta P, et al. Myocardial infarction accelerates atherosclerosis. Nature. 2012;487:325–329. [PMC free article] [PubMed] []
207. Kadoya, M. & Koyama, H. Sleep, autonomic Nnervous function and atherosclerosis. Int. J. Mol. Sci.20, 10.3390/ijms20040794 IF: 5.6 Q1 (2019). [PubMed]
208. Yang X, et al. Oxidative stress-mediated atherosclerosis: mechanisms and therapies. Front. Physiol. 2017;8:600. [PMC free article] [PubMed] []
209. Li HY, Leu YL, Wu YC, Wang SH. Melatonin inhibits in vitro smooth muscle cell inflammation and proliferation and atherosclerosis in apolipoprotein E-deficient mice. J. Agric. Food Chem. 2019;67:1889–1901. [PubMed] []
210. Born J, Lange T, Hansen K, Molle M, Fehm HL. Effects of sleep and circadian rhythm on human circulating immune cells. J. Immunol. 1997;158:4454–4464. [PubMed] []
211. Dimitrov S, Lange T, Nohroudi K, Born J. Number and function of circulating human antigen presenting cells regulated by sleep. Sleep. 2007;30:401–411. [PubMed] []
212. Lasselin J, Rehman JU, Akerstedt T, Lekander M, Axelsson J. Effect of long-term sleep restriction and subsequent recovery sleep on the diurnal rhythms of white blood cell subpopulations. Brain Behav. Immun. 2015;47:93–99. [PubMed] []
213. Said EA, et al. Sleep deprivation alters neutrophil functions and levels of Th1-related chemokines and CD4(+) T cells in the blood. Sleep. Breath. 2019;23:1331–1339. [PubMed] []
214. Dinges DF, et al. Leukocytosis and natural killer cell function parallel neurobehavioral fatigue induced by 64 hours of sleep deprivation. J. Clin. Invest. 1994;93:1930–1939. [PMC free article] [PubMed] []
215. Scheiermann C, Kunisaki Y, Frenette PS. Circadian control of the immune system. Nat. Rev. Immunol. 2013;13:190–198. [PMC free article] [PubMed] []
216. Redwine L, Dang J, Irwin M. Cellular adhesion molecule expression, nocturnal sleep, and partial night sleep deprivation. Brain Behav. Immun. 2004;18:333–340. [PubMed] []
217. Axelsson J, et al. Effects of sustained sleep restriction on mitogen-stimulated cytokines, chemokines and T helper 1/ T helper 2 balance in humans. PLoS ONE. 2013;8:e82291. [PMC free article] [PubMed] []
218. Shearer WT, et al. Soluble TNF-alpha receptor 1 and IL-6 plasma levels in humans subjected to the sleep deprivation model of spaceflight. J. Allergy Clin. Immunol. 2001;107:165–170. [PubMed] []
219. Hu J, et al. Sleep-deprived mice show altered cytokine production manifest by perturbations in serum IL-1ra, TNFa, and IL-6 levels. Brain Behav. Immun. 2003;17:498–504. [PubMed] []
220. Vgontzas AN, et al. Adverse effects of modest sleep restriction on sleepiness, performance, and inflammatory cytokines. J. Clin. Endocrinol. Metab. 2004;89:2119–2126. [PubMed] []
221. Miller MA, et al. Gender differences in the cross-sectional relationships between sleep duration and markers of inflammation: Whitehall II study. Sleep. 2009;32:857–864. [PMC free article] [PubMed] []
222. Patel SR, et al. Sleep duration and biomarkers of inflammation. Sleep. 2009;32:200–204. [PMC free article] [PubMed] []
223. Perez de Heredia F, et al. Self-reported sleep duration, white blood cell counts and cytokine profiles in European adolescents: the HELENA study. Sleep. Med. 2014;15:1251–1258. [PubMed] []
224. Bakour, C. et al. Sleep duration trajectories and systemic inflammation in young adults: results from the National Longitudinal Study of Adolescent to Adult Health (Add Health). Sleep40, 10.1093/sleep/zsx156 IF: 5.6 Q1 (2017). [PubMed]
225. Richardson MR, Churilla JR. Sleep duration and C-reactive protein in US adults. South Med. J. 2017;110:314–317. [PubMed] []
226. Fondell E, et al. Short natural sleep is associated with higher T cell and lower NK cell activities. Brain Behav. Immun. 2011;25:1367–1375. [PubMed] []
227. Carroll JE, et al. Epigenetic aging and immune senescence in women with insomnia symptoms: findings from the Women’s Health Initiative Study. Biol. Psychiatry. 2017;81:136–144. [PMC free article] [PubMed] []
228. Jackowska M, et al. Short sleep duration is associated with shorter telomere length in healthy men: findings from the Whitehall II cohort study. PLoS ONE. 2012;7:e47292. [PMC free article] [PubMed] []
229. James S, et al. Sleep duration and telomere length in children. J. Pediatr. 2017;187:247–252 e241. [PMC free article] [PubMed] []
230. Hall MH, et al. Association between sleep duration and mortality is mediated by markers of inflammation and health in older adults: the Health, Aging and Body Composition Study. Sleep. 2015;38:189–195. [PMC free article] [PubMed] []
231. Li Y, et al. Hs-CRP and all-cause, cardiovascular, and cancer mortality risk: a meta-analysis. Atherosclerosis. 2017;259:75–82. [PubMed] []
232. Uthgenannt D, Schoolmann D, Pietrowsky R, Fehm HL, Born J. Effects of sleep on the production of cytokines in humans. Psychosom. Med. 1995;57:97–104. [PubMed] []
233. Irwin MR, Wang M, Campomayor CO, Collado-Hidalgo A, Cole S. Sleep deprivation and activation of morning levels of cellular and genomic markers of inflammation. Arch. Intern. Med. 2006;166:1756–1762. [PubMed] []
234. Simpson NS, et al. Repeating patterns of sleep restriction and recovery: do we get used to it? Brain Behav. Immun. 2016;58:142–151. [PMC free article] [PubMed] []
235. Dimitrov S, Besedovsky L, Born J, Lange T. Differential acute effects of sleep on spontaneous and stimulated production of tumor necrosis factor in men. Brain Behav. Immun. 2015;47:201–210. [PubMed] []
236. Dimitrov S, Lange T, Tieken S, Fehm HL, Born J. Sleep associated regulation of T helper 1/T helper 2 cytokine balance in humans. Brain Behav. Immun. 2004;18:341–348. [PubMed] []
237. Ginaldi L, et al. The immune system in the elderly: I. Specific humoral immunity. Immunol. Res. 1999;20:101–108. [PubMed] []
238. Redwine L, Dang J, Hall M, Irwin M. Disordered sleep, nocturnal cytokines, and immunity in alcoholics. Psychosom. Med. 2003;65:75–85. [PubMed] []
239. Sakami S, et al. Coemergence of insomnia and a shift in the Th1/Th2 balance toward Th2 dominance. Neuroimmunomodulation. 2002;10:337–343. [PubMed] []
240. Moser EK, Field NS, Oliver PM. Aberrant Th2 inflammation drives dysfunction of alveolar macrophages and susceptibility to bacterial pneumonia. Cell Mol. Immunol. 2018;15:480–492. [PMC free article] [PubMed] []
241. Disis ML. Immune regulation of cancer. J. Clin. Oncol. 2010;28:4531–4538. [PMC free article] [PubMed] []
242. Lee HL, et al. Inflammatory cytokines and change of Th1/Th2 balance as prognostic indicators for hepatocellular carcinoma in patients treated with transarterial chemoembolization. Sci. Rep. 2019;9:3260. [PMC free article] [PubMed] []
243. Villafuerte G, et al. Sleep deprivation and oxidative stress in animal models: a systematic review. Oxid. Med. Cell Longev. 2015;2015:234952. [PMC free article] [PubMed] []
244. Teixeira KRC, et al. Night workers have lower levels of antioxidant defenses and higher levels of oxidative stress damage when compared to day workers. Sci. Rep. 2019;9:4455. [PMC free article] [PubMed] []
245. Trivedi MS, Holger D, Bui AT, Craddock TJA, Tartar JL. Short-term sleep deprivation leads to decreased systemic redox metabolites and altered epigenetic status. PLoS ONE. 2017;12:e0181978. [PMC free article] [PubMed] []
246. Chen X, Song M, Zhang B, Zhang Y. Reactive oxygen species regulate T cell immune response in the tumor microenvironment. Oxid. Med. Cell Longev. 2016;2016:1580967. [PMC free article] [PubMed] []
247. Liguori I, et al. Oxidative stress, aging, and diseases. Clin. Inter. Aging. 2018;13:757–772. [PMC free article] [PubMed] []
248. Irwin M, Thompson J, Miller C, Gillin JC, Ziegler M. Effects of sleep and sleep deprivation on catecholamine and interleukin-2 levels in humans: clinical implications. J. Clin. Endocrinol. Metab. 1999;84:1979–1985. [PubMed] []
249. Nance DM, Sanders VM. Autonomic innervation and regulation of the immune system (1987-2007) Brain Behav. Immun. 2007;21:736–745. [PMC free article] [PubMed] []
250. Barnes MA, Carson MJ, Nair MG. Non-traditional cytokines: How catecholamines and adipokines influence macrophages in immunity, metabolism and the central nervous system. Cytokine. 2015;72:210–219. [PMC free article] [PubMed] []
251. Grebe KM, et al. Cutting edge: sympathetic nervous system increases proinflammatory cytokines and exacerbates influenza A virus pathogenesis. J. Immunol. 2010;184:540–544. [PMC free article] [PubMed] []
252. Collado-Hidalgo A, Sung C, Cole S. Adrenergic inhibition of innate anti-viral response: PKA blockade of Type I interferon gene transcription mediates catecholamine support for HIV-1 replication. Brain Behav. Immun. 2006;20:552–563. [PubMed] []
253. Kalinichenko VV, Mokyr MB, Graf LH, Jr., Cohen RL, Chambers DA. Norepinephrine-mediated inhibition of antitumor cytotoxic T lymphocyte generation involves a beta-adrenergic receptor mechanism and decreased TNF-alpha gene expression. J. Immunol. 1999;163:2492–2499. [PubMed] []
254. Irwin MR, Cole SW. Reciprocal regulation of the neural and innate immune systems. Nat. Rev. Immunol. 2011;11:625–632. [PMC free article] [PubMed] []
255. Mishra I, et al. Chemical sympathectomy reduces peripheral inflammatory responses to acute and chronic sleep fragmentation. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2020;318:R781–R789. [PMC free article] [PubMed] []
256. Wheeler ND, Ensminger DC, Rowe MM, Wriedt ZS, Ashley NT. Alpha- and beta- adrenergic receptors regulate inflammatory responses to acute and chronic sleep fragmentation in mice. PeerJ. 2021;9:e11616. [PMC free article] [PubMed] []
257. Cirelli C, Gutierrez CM, Tononi G. Extensive and divergent effects of sleep and wakefulness on brain gene expression. Neuron. 2004;41:35–43. [PubMed] []
258. Mackiewicz M, Zimmerman JE, Shockley KR, Churchill GA, Pack AI. What are microarrays teaching us about sleep? Trends Mol. Med. 2009;15:79–87. [PMC free article] [PubMed] []
259. Barclay JL, et al. Circadian desynchrony promotes metabolic disruption in a mouse model of shiftwork. PLoS ONE. 2012;7:e37150. [PMC free article] [PubMed] []
260. Aho V, et al. Partial sleep restriction activates immune response-related gene expression pathways: experimental and epidemiological studies in humans. PLoS ONE. 2013;8:e77184. [PMC free article] [PubMed] []
261. Moller-Levet CS, et al. Effects of insufficient sleep on circadian rhythmicity and expression amplitude of the human blood transcriptome. Proc. Natl Acad. Sci. USA. 2013;110:E1132–1141. [PMC free article] [PubMed] []
262. Irwin MR, Witarama T, Caudill M, Olmstead R, Breen EC. Sleep loss activates cellular inflammation and signal transducer and activator of transcription (STAT) family proteins in humans. Brain Behav. Immun. 2015;47:86–92. [PMC free article] [PubMed] []
263. Archer SN, et al. Mistimed sleep disrupts circadian regulation of the human transcriptome. Proc. Natl Acad. Sci. USA. 2014;111:E682–691. [PMC free article] [PubMed] []
264. Mure, L. S. et al. Diurnal transcriptome atlas of a primate across major neural and peripheral tissues. Science359, 10.1126/science.aao0318 IF: 56.9 Q1 (2018). [PubMed]
265. Sulli G, Manoogian ENC, Taub PR, Panda S. Training the circadian clock, clocking the drugs, and drugging the clock to prevent, manage, and treat chronic diseases. Trends Pharm. Sci. 2018;39:812–827. [PMC free article] [PubMed] []
266. Christou S, et al. Circadian regulation in human white adipose tissue revealed by transcriptome and metabolic network analysis. Sci. Rep. 2019;9:2641. [PMC free article] [PubMed] []
267. Chen Z, Gardi J, Kushikata T, Fang J, Krueger JM. Nuclear factor-kappaB-like activity increases in murine cerebral cortex after sleep deprivation. Am. J. Physiol. 1999;276:R1812–1818. [PubMed] []
268. Williams JA, Sathyanarayanan S, Hendricks JC, Sehgal A. Interaction between sleep and the immune response in Drosophila: a role for the NFkappaB relish. Sleep. 2007;30:389–400. [PMC free article] [PubMed] []
269. Basheer R, Rainnie DG, Porkka-Heiskanen T, Ramesh V, McCarley RW. Adenosine, prolonged wakefulness, and A1-activated NF-kappaB DNA binding in the basal forebrain of the rat. Neuroscience. 2001;104:731–739. [PubMed] []
270. Brandt JA, et al. Sleep deprivation increases the activation of nuclear factor kappa B in lateral hypothalamic cells. Brain Res. 2004;1004:91–97. [PubMed] []
271. Ramesh V, Thatte HS, McCarley RW, Basheer R. Adenosine and sleep deprivation promote NF-kappaB nuclear translocation in cholinergic basal forebrain. J. Neurochem. 2007;100:1351–1363. [PubMed] []
272. Irwin MR, et al. Sleep loss activates cellular inflammatory signaling. Biol. Psychiatry. 2008;64:538–540. [PMC free article] [PubMed] []
273. Madonna R, De Caterina R. Relevance of new drug discovery to reduce NF-kappaB activation in cardiovascular disease. Vasc. Pharm. 2012;57:41–47. [PubMed] []
274. Bellet MM, Zocchi L, Sassone-Corsi P. The RelB subunit of NFkappaB acts as a negative regulator of circadian gene expression. Cell Cycle. 2012;11:3304–3311. [PMC free article] [PubMed] []
275. Kuo TH, Pike DH, Beizaeipour Z, Williams JA. Sleep triggered by an immune response in Drosophila is regulated by the circadian clock and requires the NFkappaB Relish. BMC Neurosci. 2010;11:17. [PMC free article] [PubMed] []
276. Negi G, Kumar A, Sharma SS. Melatonin modulates neuroinflammation and oxidative stress in experimental diabetic neuropathy: effects on NF-kappaB and Nrf2 cascades. J. Pineal Res. 2011;50:124–131. [PubMed] []
277. Narasimamurthy R, et al. Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc. Natl Acad. Sci. USA. 2012;109:12662–12667. [PMC free article] [PubMed] []
278. Kolmus K, Tavernier J, Gerlo S. beta2-Adrenergic receptors in immunity and inflammation: stressing NF-kappaB. Brain Behav. Immun. 2015;45:297–310. [PubMed] []
279. Benedict C, et al. Gut microbiota and glucometabolic alterations in response to recurrent partial sleep deprivation in normal-weight young individuals. Mol. Metab. 2016;5:1175–1186. [PMC free article] [PubMed] []
280. Poroyko VA, et al. Chronic sleep disruption alters gut mmicrobiota, induces systemic and adipose tissue inflammation and insulin resistance in mice. Sci. Rep. 2016;6:35405. [PMC free article] [PubMed] []
281. Gao T, et al. Role of melatonin in sleep deprivation-induced intestinal barrier dysfunction in mice. J. Pineal Res. 2019;67:e12574. [PubMed] []
282. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–141. [PMC free article] [PubMed] []
283. Belenky G, et al. Patterns of performance degradation and restoration during sleep restriction and subsequent recovery: a sleep dose-response study. J. Sleep. Res. 2003;12:1–12. [PubMed] []
284. Rupp TL, Wesensten NJ, Bliese PD, Balkin TJ. Banking sleep: realization of benefits during subsequent sleep restriction and recovery. Sleep. 2009;32:311–321. [PMC free article] [PubMed] []
285. Banks S, Van Dongen HP, Maislin G, Dinges DF. Neurobehavioral dynamics following chronic sleep restriction: dose-response effects of one night for recovery. Sleep. 2010;33:1013–1026. [PMC free article] [PubMed] []
286. Haack M, et al. Increasing sleep duration to lower beat-to-beat blood pressure: a pilot study. J. Sleep. Res. 2013;22:295–304. [PMC free article] [PubMed] []
287. Chennaoui M, et al. Leukocyte expression of type 1 and type 2 purinergic receptors and pro-inflammatory cytokines during total sleep deprivation and/or sleep extension in healthy subjects. Front Neurosci. 2017;11:240. [PMC free article] [PubMed] []
288. Swinbourne, R., Miller, J., Smart, D., Dulson, D. K. & Gill, N. The effects of sleep extension on sleep, performance, immunity and physical stress in rugby players. Sports (Basel)6, 10.3390/sports6020042 IF: 2.7 (2018). [PubMed]
289. Faraut B, et al. Benefits of napping and an extended duration of recovery sleep on alertness and immune cells after acute sleep restriction. Brain Behav. Immun. 2011;25:16–24. [PubMed] []
290. Takahashi M, Arito H. Maintenance of alertness and performance by a brief nap after lunch under prior sleep deficit. Sleep. 2000;23:813–819. [PubMed] []
291. Milner CE, Cote KA. Benefits of napping in healthy adults: impact of nap length, time of day, age, and experience with napping. J. Sleep. Res. 2009;18:272–281. [PubMed] []
292. Keramidas ME, Siebenmann C, Norrbrand L, Gadefors M, Eiken O. A brief pre-exercise nap may alleviate physical performance impairments induced by short-term sustained operations with partial sleep deprivation - A field-based study. Chronobiol. Int. 2018;35:1464–1470. [PubMed] []
293. Leng Y, et al. Daytime napping, sleep duration and serum C reactive protein: a population-based cohort study. BMJ Open. 2014;4:e006071. [PMC free article] [PubMed] []
294. Faraut B, et al. Napping reverses the salivary interleukin-6 and urinary norepinephrine changes induced by sleep restriction. J. Clin. Endocrinol. Metab. 2015;100:E416–426. [PubMed] []
295. Vgontzas AN, et al. Daytime napping after a night of sleep loss decreases sleepiness, improves performance, and causes beneficial changes in cortisol and interleukin-6 secretion. Am. J. Physiol. Endocrinol. Metab. 2007;292:E253–261. [PubMed] []
296. Zhong G, Wang Y, Tao T, Ying J, Zhao Y. Daytime napping and mortality from all causes, cardiovascular disease, and cancer: a meta-analysis of prospective cohort studies. Sleep. Med. 2015;16:811–819. [PubMed] []
297. Naska A, Oikonomou E, Trichopoulou A, Psaltopoulou T, Trichopoulos D. Siesta in healthy adults and coronary mortality in the general population. Arch. Intern. Med. 2007;167:296–301. [PubMed] []
298. Hausler N, Haba-Rubio J, Heinzer R, Marques-Vidal P. Association of napping with incident cardiovascular events in a prospective cohort study. Heart. 2019;105:1793–1798. [PubMed] []
299. Yamada T, Hara K, Shojima N, Yamauchi T, Kadowaki T. Daytime napping and the risk of cardiovascular disease and all-cause mortality: a prospective study and dose-response meta-analysis. Sleep. 2015;38:1945–1953. [PMC free article] [PubMed] []
300. Yamada T, Shojima N, Yamauchi T, Kadowaki T. J-curve relation between daytime nap duration and type 2 diabetes or metabolic syndrome: A dose-response meta-analysis. Sci. Rep. 2016;6:38075. [PMC free article] [PubMed] []
301. Mohammad, Y. Siesta and risk for ischemic stroke: results from a case-control study. Medicina (Kaunas)56, 10.3390/medicina56050222 IF: 2.6 Q3 (2020). [PubMed]
302. Irwin MR, et al. Cognitive behavioral therapy vs. Tai Chi for late life insomnia and inflammatory risk: a randomized controlled comparative efficacy trial. Sleep. 2014;37:1543–1552. [PMC free article] [PubMed] []
303. Irwin MR, et al. Cognitive behavioral therapy and tai chi reverse cellular and genomic markers of inflammation in late-life insomnia: a randomized controlled trial. Biol. Psychiatry. 2015;78:721–729. [PMC free article] [PubMed] []
304. Carroll JE, et al. Improved sleep quality in older adults with insomnia reduces biomarkers of disease risk: pilot results from a randomized controlled comparative efficacy trial. Psychoneuroendocrinology. 2015;55:184–192. [PMC free article] [PubMed] []
305. Weljie AM, et al. Oxalic acid and diacylglycerol 36:3 are cross-species markers of sleep debt. Proc. Natl Acad. Sci. USA. 2015;112:2569–2574. [PMC free article] [PubMed] []
306. Laing, E. E., Moller-Levet, C. S., Dijk, D. J. & Archer, S. N. Identifying and validating blood mRNA biomarkers for acute and chronic insufficient sleep in humans: a machine learning approach. Sleep42, 10.1093/sleep/zsy186 IF: 5.6 Q1 (2019). [PubMed]
307. Garbarino S, Scoditti E. On the role of sleep hygiene in health management during COVID-19 pandemic. Sleep. Med. 2020;77:74. [PMC free article] [PubMed] []
308. Ruiz FS, et al. Immune alterations after selective rapid eye movement or total sleep deprivation in healthy male volunteers. Innate Immun. 2012;18:44–54. [PubMed] []
309. Carroll JE, et al. Sleep deprivation and divergent toll-like receptor-4 activation of cellular inflammation in aging. Sleep. 2015;38:205–211. [PMC free article] [PubMed] []
310. Pejovic S, et al. Effects of recovery sleep after one work week of mild sleep restriction on interleukin-6 and cortisol secretion and daytime sleepiness and performance. Am. J. Physiol. Endocrinol. Metab. 2013;305:E890–896. [PMC free article] [PubMed] []

Articles from Communications Biology are provided here courtesy of Nature Publishing Group

睡眠不足与前 15 大主要死因中的 5 种有关,包括心脑神经退行性疾病 (NDD)的风险中起着重们回顾了关于睡眠剥夺对免疫相关疾病影响的证据,讨论了以下增强了 NREM 睡眠量和强度,并抑制了快速眼动 (RETNF 信号转导促进睡眠B 细胞成熟至关重要但睡眠时间短与多种癌症(包括乳腺癌、结直肠癌和前列腺癌 包括褪黑激素夜间分泌的持续时间较短因此,免疫抑制环境随着睡眠剥夺而发展,这可能转化为癌症 小鼠大脑中B细胞的募集显着增加,鉴于B细胞参与NDD的证自身免疫性病变患者的免疫疗法可改善 172 睡眠例如IL-6,其异常产生导致多克隆B细胞活化和自身免疫特,具有慢性睡眠剥夺方案的人体实验证据表明,睡眠剥夺可能会以确定,睡眠剥夺引起的亚临床炎症状态对代谢和 CV 危险#小鼠大脑中B细胞的募集显着增加,鉴于B细胞参与NDD的证据 139 ,这可能很重要。神经退行性疾病 (NDD)睡眠不足与前 15 大主要死因中的 5 种有关,包括心脑血管疾病、事故、T2DM 和高血压 28 。数据还指出,睡眠不足在中风、癌症和神经退行性疾病 (NDD) 26,29,30 的风险中起着重要作用。的风险中起着重们回顾了关于睡眠剥夺对免疫相关疾病影响的证据,讨论了以下要点:增强了 NREM 睡眠量和强度,并抑制了快速眼动 (REM) 睡眠TNF 信号转导促进睡眠B 细胞成熟至关重要但睡眠时间短与多种癌症(包括乳腺癌、结直肠癌和前列腺癌 29,121–123 )的风险之间存在显着关联包括褪黑激素夜间分泌的持续时间较短因此,免疫抑制环境随着睡眠剥夺而发展,这可能转化为癌症 128 的早发和生长速度增加或死亡率 129 增加自身免疫性病变患者的免疫疗法可改善 172 睡眠例如IL-6,其异常产生导致多克隆B细胞活化和自身免疫特征 176 的发生,,具有慢性睡眠剥夺方案的人体实验证据表明,睡眠剥夺可能会改变葡萄糖代谢 194 和胰岛素敏感性 195 ,从而增加肥胖和 T2DM 的风险。以确定,睡眠剥夺引起的亚临床炎症状态对代谢和 CV 危险因素(葡萄糖代谢、糖尿病、高血压、动脉粥样硬化血脂谱、内皮功能障碍和冠状动脉钙化)和结果(中风和冠心病) 24 具有致病意义