1
|
Zielinski MR, Gibbons AJ. Neuroinflammation, Sleep, and Circadian Rhythms. Front Cell Infect Microbiol 2022; 12:853096. [PMID: 35392608 PMCID: PMC8981587 DOI: 10.3389/fcimb.2022.853096] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022] Open
Abstract
Molecules involved in innate immunity affect sleep and circadian oscillators and vice versa. Sleep-inducing inflammatory molecules are activated by increased waking activity and pathogens. Pathologies that alter inflammatory molecules, such as traumatic brain injury, cancer, cardiovascular disease, and stroke often are associated with disturbed sleep and electroencephalogram power spectra. Moreover, sleep disorders, such as insomnia and sleep disordered breathing, are associated with increased dysregulation of inflammatory processes. Inflammatory molecules in both the central nervous system and periphery can alter sleep. Inflammation can also modulate cerebral vascular hemodynamics which is associated with alterations in electroencephalogram power spectra. However, further research is needed to determine the interactions of sleep regulatory inflammatory molecules and circadian clocks. The purpose of this review is to: 1) describe the role of the inflammatory cytokines interleukin-1 beta and tumor necrosis factor-alpha and nucleotide-binding domain and leucine-rich repeat protein-3 inflammasomes in sleep regulation, 2) to discuss the relationship between the vagus nerve in translating inflammatory signals between the periphery and central nervous system to alter sleep, and 3) to present information about the relationship between cerebral vascular hemodynamics and the electroencephalogram during sleep.
Collapse
Affiliation(s)
- Mark R. Zielinski
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States,Harvard Medical School, West Roxbury, MA, United States,*Correspondence: Mark R. Zielinski,
| | - Allison J. Gibbons
- Veterans Affairs (VA) Boston Healthcare System, West Roxbury, MA, United States
| |
Collapse
|
2
|
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been shown to modulate neuronal signaling in homeostasis and diseases. In homeostasis, IL-1 regulates sleep and memory formation, whereas in diseases, IL-1 impairs memory and alters affect. Interestingly, IL-1 can cause long-lasting changes in behavior, suggesting IL-1 can alter neuroplasticity. The neuroplastic effects of IL-1 are mediated via its cognate receptor, Interleukin-1 Type 1 Receptor (IL-1R1), and are dependent on the distribution and cell type(s) of IL-1R1 expression. Recent reports found that IL-1R1 expression is restricted to discrete subpopulations of neurons, astrocytes, and endothelial cells and suggest IL-1 can influence neural circuits directly through neuronal IL-1R1 or indirectly via non-neuronal IL-1R1. In this review, we analyzed multiple mechanisms by which IL-1/IL-1R1 signaling might impact neuroplasticity based upon the most up-to-date literature and provided potential explanations to clarify discrepant and confusing findings reported in the past.
Collapse
Affiliation(s)
- Daniel P. Nemeth
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, USA
| |
Collapse
|
3
|
Abstract
Interactions between the immune system and the nervous system have been described mostly in the context of diseases. More recent studies have begun to reveal how certain immune cell-derived soluble effectors, the cytokines, can influence host behaviour even in the absence of infection. In this Review, we contemplate how the immune system shapes nervous system function and how it controls the manifestation of host behaviour. Interactions between these two highly complex systems are discussed here also in the context of evolution, as both may have evolved to maximize an organism's ability to respond to environmental threats in order to survive. We describe how the immune system relays information to the nervous system and how cytokine signalling occurs in neurons. We also speculate on how the brain may be hardwired to receive and process information from the immune system. Finally, we propose a unified theory depicting a co-evolution of the immune system and host behaviour in response to the evolutionary pressure of pathogens.
Collapse
|
4
|
Gobbo D, Scheller A, Kirchhoff F. From Physiology to Pathology of Cortico-Thalamo-Cortical Oscillations: Astroglia as a Target for Further Research. Front Neurol 2021; 12:661408. [PMID: 34177766 PMCID: PMC8219957 DOI: 10.3389/fneur.2021.661408] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022] Open
Abstract
The electrographic hallmark of childhood absence epilepsy (CAE) and other idiopathic forms of epilepsy are 2.5-4 Hz spike and wave discharges (SWDs) originating from abnormal electrical oscillations of the cortico-thalamo-cortical network. SWDs are generally associated with sudden and brief non-convulsive epileptic events mostly generating impairment of consciousness and correlating with attention and learning as well as cognitive deficits. To date, SWDs are known to arise from locally restricted imbalances of excitation and inhibition in the deep layers of the primary somatosensory cortex. SWDs propagate to the mostly GABAergic nucleus reticularis thalami (NRT) and the somatosensory thalamic nuclei that project back to the cortex, leading to the typical generalized spike and wave oscillations. Given their shared anatomical basis, SWDs have been originally considered the pathological transition of 11-16 Hz bursts of neural oscillatory activity (the so-called sleep spindles) occurring during Non-Rapid Eye Movement (NREM) sleep, but more recent research revealed fundamental functional differences between sleep spindles and SWDs, suggesting the latter could be more closely related to the slow (<1 Hz) oscillations alternating active (Up) and silent (Down) cortical activity and concomitantly occurring during NREM. Indeed, several lines of evidence support the fact that SWDs impair sleep architecture as well as sleep/wake cycles and sleep pressure, which, in turn, affect seizure circadian frequency and distribution. Given the accumulating evidence on the role of astroglia in the field of epilepsy in the modulation of excitation and inhibition in the brain as well as on the development of aberrant synchronous network activity, we aim at pointing at putative contributions of astrocytes to the physiology of slow-wave sleep and to the pathology of SWDs. Particularly, we will address the astroglial functions known to be involved in the control of network excitability and synchronicity and so far mainly addressed in the context of convulsive seizures, namely (i) interstitial fluid homeostasis, (ii) K+ clearance and neurotransmitter uptake from the extracellular space and the synaptic cleft, (iii) gap junction mechanical and functional coupling as well as hemichannel function, (iv) gliotransmission, (v) astroglial Ca2+ signaling and downstream effectors, (vi) reactive astrogliosis and cytokine release.
Collapse
Affiliation(s)
- Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| |
Collapse
|
5
|
Cellular Effects of Rhynchophylline and Relevance to Sleep Regulation. Clocks Sleep 2021; 3:312-341. [PMID: 34207633 PMCID: PMC8293156 DOI: 10.3390/clockssleep3020020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/25/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Uncaria rhynchophylla is a plant highly used in the traditional Chinese and Japanese medicines. It has numerous health benefits, which are often attributed to its alkaloid components. Recent studies in humans show that drugs containing Uncaria ameliorate sleep quality and increase sleep time, both in physiological and pathological conditions. Rhynchophylline (Rhy) is one of the principal alkaloids in Uncaria species. Although treatment with Rhy alone has not been tested in humans, observations in rodents show that Rhy increases sleep time. However, the mechanisms by which Rhy could modulate sleep have not been comprehensively described. In this review, we are highlighting cellular pathways that are shown to be targeted by Rhy and which are also known for their implications in the regulation of wakefulness and sleep. We conclude that Rhy can impact sleep through mechanisms involving ion channels, N-methyl-d-aspartate (NMDA) receptors, tyrosine kinase receptors, extracellular signal-regulated kinases (ERK)/mitogen-activated protein kinases (MAPK), phosphoinositide 3-kinase (PI3K)/RAC serine/threonine-protein kinase (AKT), and nuclear factor-kappa B (NF-κB) pathways. In modulating multiple cellular responses, Rhy impacts neuronal communication in a way that could have substantial effects on sleep phenotypes. Thus, understanding the mechanisms of action of Rhy will have implications for sleep pharmacology.
Collapse
|
6
|
Zefferino R, Di Gioia S, Conese M. Molecular links between endocrine, nervous and immune system during chronic stress. Brain Behav 2021; 11:e01960. [PMID: 33295155 PMCID: PMC7882157 DOI: 10.1002/brb3.1960] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/17/2020] [Accepted: 10/30/2020] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION The stress response is different in various individuals, however, the mechanisms that could explain these distinct effects are not well known and the molecular correlates have been considered one at the time. Particular harmful conditions occur if the subject, instead to cope the stressful events, succumb to them, in this case, a cascade reaction happens that through different signaling causes a specific reaction named "sickness behaviour." The aim of this article is to review the complex relations among important molecules belonging to Central nervous system (CNS), immune system (IS), and endocrine system (ES) during the chronic stress response. METHODS After having verified the state of art concerning the function of cortisol, norepinephrine (NE), interleukin (IL)-1β and melatonin, we describe as they work together. RESULTS We propose a speculative hypothesis concerning the complex interplay of these signaling molecules during chronic stress, highlighting the role of IL-1β as main biomarker of this effects, indeed, during chronic stress its increment transforms this inflammatory signal into a nervous signal (NE), in turn, this uses the ES (melatonin and cortisol) to counterbalance again IL-1β. During cortisol resistance, a vicious loop occurs that increments all mediators, unbalancing IS, ES, and CNS networks. This IL-1β increase would occur above all when the individual succumbs to stressful events, showing the Sickness Behaviour Symptoms. IL-1β might, through melatonin and vice versa, determine sleep disorders too. CONCLUSION The molecular links here outlined could explain how stress plays a role in etiopathogenesis of several diseases through this complex interplay.
Collapse
Affiliation(s)
- Roberto Zefferino
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Sante Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Massimo Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
7
|
Matenchuk BA, Mandhane PJ, Kozyrskyj AL. Sleep, circadian rhythm, and gut microbiota. Sleep Med Rev 2020; 53:101340. [PMID: 32668369 DOI: 10.1016/j.smrv.2020.101340] [Citation(s) in RCA: 255] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/17/2022]
Abstract
From asthma and heart disease to diabetes and obesity, the human microbiome plays a role in the pathogenesis of each chronic health condition plaguing today's society. Recent work has shown that the gut microbiota and its metabolites exhibit diurnal rhythmicity which predominantly respond to the feeding/fasting cycle. Persistent jet lag, an obesogenic diet, and clock gene deficiency can dampen the oscillatory nature of gut bacterial composition, which can subsequently be rescued by time restricted feeding. Contrastingly, gut microbial metabolites influence central and hepatic clock gene expression and sleep duration in the host and regulate body composition through circadian transcription factors. Both sleep fragmentation and short sleep duration are associated with gut dysbiosis which may be due to activation of the HPA-axis. Metabolic disturbances associated with sleep loss may in fact be mediated through the overgrowth of specific gut bacteria. Reciprocally, the end products of bacterial species which grow in response to sleep loss are able to induce fatigue. Furthermore, probiotic supplementation has been found to improve subjective sleep quality. Sleep quality and duration may be an important target for supporting healthy gut microbiota composition, but the cyclic nature of this relationship should not be overlooked.
Collapse
Affiliation(s)
| | - Piush J Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Anita L Kozyrskyj
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
8
|
Oles V, Koh KMS, Dykstra-Aiello CJ, Savenkova M, Gibbons CM, Nguyen JT, Karatsoreos I, Panchenko A, Krueger JM. 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. [PMID: 32324480 DOI: 10.1152/japplphysiol.00839.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sleep regulation involves interleukin-1β (IL1) family members, TNF, and circadian clock genes. Previously, we characterized spontaneous sleep and sleep after 8 h of sleep deprivation (SD) ending at zeitgeber time (ZT)4 and ZT16 in wild-type (WT) and IL1 receptor accessory protein (AcP)- and brain-specific AcP (AcPb)-knockout (KO) mice. Here, we applied quantitative reverse transcriptase polymerase chain reaction and Spearman gene pair expression correlation methods to characterize IL1, IL1 receptor 1 (IL1R1), AcP, AcPb, Period 1 (Per1), Clock, adenosine deaminase (Ada), peptidoglycan recognition protein 1 (Pglyrp1), and TNF mRNA expressions under conditions with distinct sleep phenotypes. In WT mice, IL1, IL1R1, AcP, Ada, and Clock mRNAs were higher at ZT4 (mid-sleep period) than at ZT16. mRNA expressions differed substantially in AcP and AcPb KO mice at those times. After SD ending at ZT4, only WT mice had a non-rapid eye movement sleep (NREMS) rebound, and AcPb and IL1R1 mRNA increases were unique to WT mice. In AcPb KO mice, which have spontaneous high EEG slow wave power, AcP and Pglyrp1 mRNAs were elevated relative to WT mice at ZT4. At ZT4, the AcPb KO - WT Spearman correlation difference networks showed high positive correlations between IL1R1 and IL1, Per1, and Clock and high negative correlations between TNF and Pglyrp1 and Ada. At ZT16, the WT mice gene pair expression network was mostly negative, whereas in AcP KO mice, which have substantially more rapid eye movement sleep than WT mice, it was all positive. We conclude that gene pair expression correlations depend on the presence of AcP and AcPb.NEW & NOTEWORTHY Spearman gene pair expression correlations depend upon the presence or absence of interleukin-1 receptor accessory protein and upon sleep phenotype.
Collapse
Affiliation(s)
- Vladyslav Oles
- Department of Mathematics and Statistics, Washington State University, Pullman, Washington
| | - Khia Min Sabrina Koh
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | | | - Marina Savenkova
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Cody M Gibbons
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington.,University of Washington School of Medicine, Seattle, Washington
| | - Joseph T Nguyen
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Ilia Karatsoreos
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Alexander Panchenko
- Department of Mathematics and Statistics, Washington State University, Pullman, Washington
| | - James M Krueger
- Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
9
|
Garofalo S, Picard K, Limatola C, Nadjar A, Pascual O, Tremblay MÈ. Role of Glia in the Regulation of Sleep in Health and Disease. Compr Physiol 2020; 10:687-712. [PMID: 32163207 DOI: 10.1002/cphy.c190022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sleep is a naturally occurring physiological state that is required to sustain physical and mental health. Traditionally viewed as strictly regulated by top-down control mechanisms, sleep is now known to also originate locally. Glial cells are emerging as important contributors to the regulation of sleep-wake cycles, locally and among dedicated neural circuits. A few pioneering studies revealed that astrocytes and microglia may influence sleep pressure, duration as well as intensity, but the precise involvement of these two glial cells in the regulation of sleep remains to be fully addressed, across contexts of health and disease. In this overview article, we will first summarize the literature pertaining to the role of astrocytes and microglia in the regulation of sleep under normal physiological conditions. Afterward, we will discuss the beneficial and deleterious consequences of glia-mediated neuroinflammation, whether it is acute, or chronic and associated with brain diseases, on the regulation of sleep. Sleep disturbances are a main comorbidity in neurodegenerative diseases, and in several brain diseases that include pain, epilepsy, and cancer. Identifying the relationships between glia-mediated neuroinflammation, sleep-wake rhythm disruption and brain diseases may have important implications for the treatment of several disorders. © 2020 American Physiological Society. Compr Physiol 10:687-712, 2020.
Collapse
Affiliation(s)
- Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Katherine Picard
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Agnès Nadjar
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France
| | - Olivier Pascual
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Université Claude Bernard Lyon, Lyon, France
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada.,Départment de médecine moleculaire, Faculté de médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
10
|
Szentirmai É, Kapás L. Sleep and body temperature in TNFα knockout mice: The effects of sleep deprivation, β3-AR stimulation and exogenous TNFα. Brain Behav Immun 2019; 81:260-271. [PMID: 31220563 PMCID: PMC6754767 DOI: 10.1016/j.bbi.2019.06.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/06/2019] [Accepted: 06/16/2019] [Indexed: 01/09/2023] Open
Abstract
Increased production of pro-inflammatory cytokines is assumed to mediate increased sleep under inflammatory conditions, such as systemic infections or recovery from sleep loss. The role of cytokines in sleep regulation under normal conditions is less clear. In the present study, we investigated the role of endogenous tumor necrosis factor alpha (TNFα) in sleep regulation using TNFα knockout (KO) mice. Under control conditions at thermoneutral ambient temperature, total sleep time did not differ between TNFα KO and wild-type (WT) mice, but TNFα KO mice had increased rapid-eye-movement sleep (REMS), accompanied by decreased motor activity and body temperature. Exposure to 17 °C induced decreases in total sleep time similarly in both genotypes. Sleep deprivation by gentle handling elicited robust rebound increases in non-rapid-eye movement sleep (NREMS), REMS and electroencephalographic (EEG) slow-wave activity (SWA), accompanied by suppressed motor activity and decreased body temperature; there was no significant difference between the responses of WT and KO mice. Systemic injection of the beta3-adrenergic receptor (β3-AR) agonist CL-316,243 induced increases in NREMS and body temperature. The temperature response, but not the sleep effect, was attenuated in the KO animals. Systemic injection of TNFα induced increased NREMS, reduced REMS and biphasic temperature responses in both genotypes. In the KO mice, the NREMS-promoting effects of exogenously administered TNFα was decreased, while REMS suppression was enhanced, and the first, hypothermic, phase of temperature response was attenuated. Overall, TNFα KO mice did not show any deficiency in sleep regulation which suggests that the role of endogenous TNFα in sleep regulation is less pronounced than previously suggested.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA
| |
Collapse
|
11
|
Nguyen J, Gibbons CM, Dykstra-Aiello C, Ellingsen R, Koh KMS, Taishi P, Krueger JM. Interleukin-1 receptor accessory proteins are required for normal homeostatic responses to sleep deprivation. J Appl Physiol (1985) 2019; 127:770-780. [PMID: 31295066 DOI: 10.1152/japplphysiol.00366.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Interleukin-1β (IL1) is a sleep regulatory substance. The IL1/IL1 type 1 receptor complex requires a receptor accessory protein (AcP) to signal. There are three isoforms of AcP. In the current experiments, mice lacking a neuron-specific isoform, called AcPb knockout (AcPb KO), or mice lacking AcP + AcPb isoforms (AcP KO) or wild-type (WT) mice were used. Spontaneous sleep and sleep responses to sleep deprivation (SD) between zeitgeber time (ZT) 20-ZT4 and ZT8-ZT16 were characterized. Furthermore, somatosensory cortical protein extracts were examined for phosphorylated (p) proto-oncogene tyrosine-protein kinase sarcoma (Src) and p38MAPK levels at ZT4 and ZT16 and after SD. Spontaneous sleep was similar in the three strains, except rapid eye movement sleep (REMS) duration between ZT12-ZT16 was greater in AcP KO than WT mice. After SD at ZT4, only WT mice had non-REMS (NREMS) rebounds. All mouse strains lacked an NREMS rebound after SD at ZT16. All strains after both SD periods had REMS rebounds. AcPb KO mice, but not AcP KO mice, had greater EEG delta wave (0.5-4 Hz) power during NREMS than WT mice. p-Src was very low at ZT16 but high at ZT4, whereas p-p38MAPK was low at ZT4 and high at ZT16. p-p38MAPK levels were not sensitive to SD. In contrast, p-Src levels were less after SD at the P = 0.08 level of significance in the strains lacking AcPb. We conclude that AcPb is required for NREMS responses to sleep loss, but not for SD-induced EEG delta wave or REMS responses.NEW & NOTEWORTHY Interleukin-1β (IL1), a well-characterized sleep regulatory substance, requires an IL1 receptor accessory protein (AcP); one of its isoforms is neuron-specific (called AcPb). We showed that in mice, AcPb is required for nonrapid eye movement sleep responses following 8 h of sleep loss ending 4 h after daybreak but did not affect rapid eye movement sleep rebound. Sleep loss reduced phosphorylation of proto-oncogene tyrosine-protein kinase sarcoma but not of the less sensitive p38MAPK, downstream IL1 signaling molecules.
Collapse
Affiliation(s)
- Joseph Nguyen
- Department Integrative Physiology and Neurobiology, College of Veterinary Medicine, Washington State University, Spokane, Washington
| | - Cody M Gibbons
- School of Medicine University of Washington, Spokane, Washington
| | - Cheryl Dykstra-Aiello
- Department Integrative Physiology and Neurobiology, College of Veterinary Medicine, Washington State University, Spokane, Washington
| | | | - Khia Min Sabrina Koh
- Department Integrative Physiology and Neurobiology, College of Veterinary Medicine, Washington State University, Spokane, Washington
| | - Ping Taishi
- Department Integrative Physiology and Neurobiology, College of Veterinary Medicine, Washington State University, Spokane, Washington
| | - James M Krueger
- Department Integrative Physiology and Neurobiology, College of Veterinary Medicine, Washington State University, Spokane, Washington
| |
Collapse
|
12
|
Gassen J, Hill SE. Why inflammation and the activities of the immune system matter for social and personality psychology (and not only for those who study health). SOCIAL AND PERSONALITY PSYCHOLOGY COMPASS 2019. [DOI: 10.1111/spc3.12471] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
13
|
Interleukin-1β Protects Neurons against Oxidant-Induced Injury via the Promotion of Astrocyte Glutathione Production. Antioxidants (Basel) 2018; 7:antiox7080100. [PMID: 30044427 PMCID: PMC6115796 DOI: 10.3390/antiox7080100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/04/2018] [Accepted: 07/21/2018] [Indexed: 01/13/2023] Open
Abstract
Interleukin-1β (IL-1β), a key cytokine that drives neuroinflammation in the Central Nervous System (CNS), is enhanced in many neurological diseases/disorders. Although IL-1β contributes to and/or sustains pathophysiological processes in the CNS, we recently demonstrated that IL-1β can protect cortical astrocytes from oxidant injury in a glutathione (GSH)-dependent manner. To test whether IL-1β could similarly protect neurons against oxidant stress, near pure neuronal cultures or mixed cortical cell cultures containing neurons and astrocytes were exposed to the organic peroxide, tert-butyl hydroperoxide (t-BOOH), following treatment with IL-1β or its vehicle. Neurons and astrocytes in mixed cultures, but not pure neurons, were significantly protected from the toxicity of t-BOOH following treatment with IL-1β in association with enhanced GSH production/release. IL-1β failed to increase the GSH levels or to provide protection against t-BOOH toxicity in chimeric mixed cultures consisting of IL-1R1+/+ neurons plated on top of IL-1R1−/− astrocytes. The attenuation of GSH release via block of multidrug resistance-associated protein 1 (MRP1) transport also abrogated the protective effect of IL-1β. These protective effects were not strictly an in vitro phenomenon as we found an increased striatal vulnerability to 3-nitropropionic acid-mediated oxidative stress in IL-1R1 null mice. Overall, our data indicate that IL-1β protects neurons against oxidant injury and that this likely occurs in a non-cell-autonomous manner that relies on an increase in astrocyte GSH production and release.
Collapse
|
14
|
Cooper JM, Halter KA, Prosser RA. Circadian rhythm and sleep-wake systems share the dynamic extracellular synaptic milieu. Neurobiol Sleep Circadian Rhythms 2018; 5:15-36. [PMID: 31236509 PMCID: PMC6584685 DOI: 10.1016/j.nbscr.2018.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 03/06/2018] [Accepted: 04/10/2018] [Indexed: 01/23/2023] Open
Abstract
The mammalian circadian and sleep-wake systems are closely aligned through their coordinated regulation of daily activity patterns. Although they differ in their anatomical organization and physiological processes, they utilize overlapping regulatory mechanisms that include an assortment of proteins and molecules interacting within the extracellular space. These extracellular factors include proteases that interact with soluble proteins, membrane-attached receptors and the extracellular matrix; and cell adhesion molecules that can form complex scaffolds connecting adjacent neurons, astrocytes and their respective intracellular cytoskeletal elements. Astrocytes also participate in the dynamic regulation of both systems through modulating neuronal appositions, the extracellular space and/or through release of gliotransmitters that can further contribute to the extracellular signaling processes. Together, these extracellular elements create a system that integrates rapid neurotransmitter signaling across longer time scales and thereby adjust neuronal signaling to reflect the daily fluctuations fundamental to both systems. Here we review what is known about these extracellular processes, focusing specifically on areas of overlap between the two systems. We also highlight questions that still need to be addressed. Although we know many of the extracellular players, far more research is needed to understand the mechanisms through which they modulate the circadian and sleep-wake systems.
Collapse
Key Words
- ADAM, A disintegrin and metalloproteinase
- AMPAR, AMPA receptor
- Astrocytes
- BDNF, brain-derived neurotrophic factor
- BMAL1, Brain and muscle Arnt-like-1 protein
- Bmal1, Brain and muscle Arnt-like-1 gene
- CAM, cell adhesion molecules
- CRY, cryptochrome protein
- Cell adhesion molecules
- Circadian rhythms
- Cry, cryptochrome gene
- DD, dark-dark
- ECM, extracellular matrix
- ECS, extracellular space
- EEG, electroencephalogram
- Endo N, endoneuraminidase N
- Extracellular proteases
- GFAP, glial fibrillary acidic protein
- IL, interleukin
- Ig, immunoglobulin
- LC, locus coeruleus
- LD, light-dark
- LH, lateral hypothalamus
- LRP-1, low density lipoprotein receptor-related protein 1
- LTP, long-term potentiation
- MMP, matrix metalloproteinases
- NCAM, neural cell adhesion molecule protein
- NMDAR, NMDA receptor
- NO, nitric oxide
- NST, nucleus of the solitary tract
- Ncam, neural cell adhesion molecule gene
- Nrl, neuroligin gene
- Nrx, neurexin gene
- P2, purine type 2 receptor
- PAI-1, plasminogen activator inhibitor-1
- PER, period protein
- PPT, peduculopontine tegmental nucleus
- PSA, polysialic acid
- Per, period gene
- REMS, rapid eye movement sleep
- RSD, REM sleep disruption
- SCN, suprachiasmatic nucleus
- SWS, slow wave sleep
- Sleep-wake system
- Suprachiasmatic nucleus
- TNF, tumor necrosis factor
- TTFL, transcriptional-translational negative feedback loop
- VIP, vasoactive intestinal polypeptide
- VLPO, ventrolateral preoptic
- VP, vasopressin
- VTA, ventral tegmental area
- dNlg4, drosophila neuroligin-4 gene
- nNOS, neuronal nitric oxide synthase gene
- nNOS, neuronal nitric oxide synthase protein
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
- uPAR, uPA receptor
Collapse
|
15
|
Szentirmai É, Kapás L. Brown adipose tissue plays a central role in systemic inflammation-induced sleep responses. PLoS One 2018; 13:e0197409. [PMID: 29746591 PMCID: PMC5945014 DOI: 10.1371/journal.pone.0197409] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/01/2018] [Indexed: 01/31/2023] Open
Abstract
We previously identified brown adipose tissue (BAT) as a source of sleep-inducing signals. Pharmacological activation of BAT enhances sleep while sleep loss leads to increased BAT thermogenesis. Recovery sleep after sleep loss is diminished in mice that lack uncoupling protein 1 (UCP-1), and also in wild-type (WT) mice after sensory denervation of the BAT. Systemic inflammation greatly affects metabolism and the function of adipose tissue, and also induces characteristic sleep responses. We hypothesized that sleep responses to acute inflammation are mediated by BAT-derived signals. To test this, we determined the effects of systemic inflammation on sleep and body temperature in UCP-1 knockout (KO) and WT mice. Intraperitoneal injections of lipopolysaccharide, tumor necrosis factor-α, interleukin-1 beta and clodronate containing liposomes were used to induce systemic inflammation. In WT animals, non-rapid-eye movement sleep (NREMS) was elevated in all four inflammatory models. All NREMS responses were completely abolished in UCP-1 KO animals. Systemic inflammation elicited an initial hypothermia followed by fever in WT mice. The hypothermic phase, but not the fever, was abolished in UCP-1 KO mice. The only recognized function of UCP-1 is to promote thermogenesis in brown adipocytes. Present results indicate that the presence of UCP-1 is necessary for increased NREMS but does not contribute to the development of fever in systemic inflammation.
Collapse
Affiliation(s)
- Éva Szentirmai
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America
| | - Levente Kapás
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, Washington, United States of America
- Sleep and Performance Research Center, Washington State University, Spokane, Washington, United States of America
| |
Collapse
|
16
|
Orb Q, Orlandi RR, Alt JA. Sleep dysfunction and its association to chronic rhinosinusitis: Updated review. Laryngoscope Investig Otolaryngol 2017. [PMID: 28630938 PMCID: PMC5473662 DOI: 10.1002/lio2.60] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Poor sleep has significant effects on health contributing to increased morbidity and mortality. The direct and indirect costs of sleep dysfunction total well in to the billions of dollars annually in the United States. Chronic rhinosinusitis (CRS) affects up to 16% of the US population and has been linked to poor sleep quality with up to three quarters of patients with CRS reporting poor sleep quality. There is a growing body of literature evaluating the relationship between sleep and CRS. In this review, we organize and present the current knowledge on the associations between sleep and CRS as well as identify areas for further investigation. Data sources A structured literature search from 1946 to 2016 was conducted in the English language using OVID MEDLINE database, PubMed, and EMBASE. Review methods Abstracts were reviewed for relevance and appropriate studies were included in the narrative review. Results Studies were analyzed and discussed as they pertained to the following categories of CRS and sleep: (1) subjective measures of sleep dysfunction, (2) objective measures of sleep dysfunction, and (3) outcomes on sleep quality following treatment of CRS. Articles on the pathophysiology of sleep dysfunction in CRS were separately reviewed. Conclusions An evolving body of research demonstrates that quality of sleep is compromised in the majority of patients with CRS. Following treatment of CRS, there is significant improvement in subjective sleep quality, but additional research investigating objective measures following treatment is still needed. Additionally, further investigation is required to better elucidate the underlying pathophysiology of the relationship between sleep dysfunction and CRS. Level of Evidence N/A.
Collapse
Affiliation(s)
- Quinn Orb
- Division of Head and Neck Surgery, Rhinology-Sinus & Skull Base Surgery Program, Department of Surgery; University of Utah, Salt Lake City, UT, U.S.A
| | - Richard R Orlandi
- Division of Head and Neck Surgery, Rhinology-Sinus & Skull Base Surgery Program, Department of Surgery; University of Utah, Salt Lake City, UT, U.S.A
| | - Jeremiah A Alt
- Division of Head and Neck Surgery, Rhinology-Sinus & Skull Base Surgery Program, Department of Surgery; University of Utah, Salt Lake City, UT, U.S.A
| |
Collapse
|
17
|
Irwin MR, Opp MR. Sleep Health: Reciprocal Regulation of Sleep and Innate Immunity. Neuropsychopharmacology 2017; 42:129-155. [PMID: 27510422 PMCID: PMC5143488 DOI: 10.1038/npp.2016.148] [Citation(s) in RCA: 324] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Sleep disturbances including insomnia independently contribute to risk of inflammatory disorders and major depressive disorder. This review and overview provides an integrated understanding of the reciprocal relationships between sleep and the innate immune system and considers the role of sleep in the nocturnal regulation of the inflammatory biology dynamics; the impact of insomnia complaints, extremes of sleep duration, and experimental sleep deprivation on genomic, cellular, and systemic markers of inflammation; and the influence of sleep complaints and insomnia on inflammaging and molecular processes of cellular aging. Clinical implications of this research include discussion of the contribution of sleep disturbance to depression and especially inflammation-related depressive symptoms. Reciprocal action of inflammatory mediators on the homeostatic regulation of sleep continuity and sleep macrostructure, and the potential of interventions that target insomnia to reverse inflammation, are also reviewed. Together, interactions between sleep and inflammatory biology mechanisms underscore the implications of sleep disturbance for inflammatory disease risk, and provide a map to guide the development of treatments that modulate inflammation, improve sleep, and promote sleep health.
Collapse
Affiliation(s)
- Michael R Irwin
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience Director and Mindful Awareness Research Center, University of California, Los Angeles, CA, USA
| | - Mark R Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
18
|
Davis CJ, Zielinski MR, Dunbrasky D, Taishi P, Dinarello CA, Krueger JM. Interleukin 37 expression in mice alters sleep responses to inflammatory agents and influenza virus infection. Neurobiol Sleep Circadian Rhythms 2016; 3:1-9. [PMID: 28070566 PMCID: PMC5218600 DOI: 10.1016/j.nbscr.2016.11.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Multiple interactions between the immune system and sleep are known, including the effects of microbial challenge on sleep or the effects of sleep loss on facets of the immune response. Cytokines regulate, in part, sleep and immune responses. Here we examine the role of an anti-inflammatory cytokine, interleukin-37 (IL-37) on sleep in a mouse strain that expresses human IL-37b (IL37tg mice). Constitutive expression of the IL-37 gene in the brains of these mice under resting conditions is low; however, upon an inflammatory stimulus, expression increases dramatically. We measured sleep in three conditions; (a) under baseline conditions and after 6 h of sleep loss, (b) after bolus intraperitoneal administration of lipopolysaccharide (LPS) or IL-1β and (c) after intranasal influenza virus challenge. Under baseline conditions, the IL37tg mice had 7% more spontaneous non-rapid eye movement sleep (NREMS) during the light period than wild-type (WT) mice. After sleep deprivation both WT mice and IL37tg mice slept an extra 21% and 12%, respectively, during the first 6 h of recovery. NREMS responses after sleep deprivation did not significantly differ between WT mice and IL37tg mice. However, in response to either IL-1β or LPS, the increases in time spent in NREMS were about four-fold greater in the WT mice than in the IL37tg mice. In contrast, in response to a low dose of mouse-adapted H1N1 influenza virus, sleep responses developed slowly over the 6 day recording period. By day 6, NREMS increased by 10% and REMS increased by 18% in the IL37tg mice compared to the WT mice. Further, by day 4 IL37tg mice lost less weight, remained more active, and retained their body temperatures closer to baseline values than WT mice. We conclude that conditions that promote IL-37 expression attenuate morbidity to severe inflammatory challenge. Sleep responses to mild acute sleep deprivation are similar in mice transgenic for interleukin-37 (IL37tg) IL37tg and wild type (WT) mice. Sleep responses induced by either IL-β or LPS are greatly attenuated in IL37tg mice compared to WT mice. After influenza virus challenge, IL37tg mice have reduced morbidities and enhanced sleep responses.
Collapse
Affiliation(s)
- Christopher J Davis
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA 99210-1495
| | - Mark R Zielinski
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA 99210-1495; VA Boston Healthcare System, Harvard Medical School, West Roxbury, MA, USA 02312; Department of Psychiatry, Harvard Medical School, West Roxbury, MA, USA 02312
| | - Danielle Dunbrasky
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA 99210-1495
| | - Ping Taishi
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA 99210-1495
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA 80045; Radboud University Medical Center, Nijmegen, The Netherlands
| | - James M Krueger
- Elson S. Floyd College of Medicine, Department of Biomedical Sciences, Washington State University, Spokane, WA, USA 99210-1495
| |
Collapse
|
19
|
Huang TR, Jou SB, Chou YJ, Yi PL, Chen CJ, Chang FC. Interleukin-1 receptor (IL-1R) mediates epilepsy-induced sleep disruption. BMC Neurosci 2016; 17:74. [PMID: 27875989 PMCID: PMC5120515 DOI: 10.1186/s12868-016-0309-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/14/2016] [Indexed: 12/02/2022] Open
Abstract
Background Sleep disruptions are common in epilepsy patients. Our previous study demonstrates that homeostatic factors and circadian rhythm may mediate epilepsy-induced sleep disturbances when epilepsy occurs at different zeitgeber hours. The proinflammatory cytokine, interleukin-1 (IL-1), is a somnogenic cytokine and may also be involved in epileptogenesis; however, few studies emphasize the effect of IL-1 in epilepsy-induced sleep disruption. We herein hypothesized that IL-1 receptor type 1 (IL-1R1) mediates the pathogenesis of epilepsy and epilepsy-induced sleep disturbances. We determined the role of IL-1R1 by using IL-1R1 knockout (IL-1R1 −/− KO) mice. Results Our results elucidated the decrease of non-rapid eye movement (NREM) sleep during the light period in IL-1R −/− mice and confirmed the somnogenic role of IL-1R1. Rapid electrical amygdala kindling was performed to induce epilepsy at the particular zeitgeber time (ZT) point, ZT13. Our results demonstrated that seizure thresholds induced by kindling stimuli, such as the after-discharge threshold and successful kindling rates, were not altered in IL-1R −/− mice when compared to those obtained from the wildtype mice (IL-1R +/+ mice). This result suggests that IL-1R1 is not involved in kindling-induced epileptogenesis. During sleep, ZT13 kindling stimulation significantly enhanced NREM sleep during the subsequent 6 h (ZT13-18) in wildtype mice, and sleep returned to the baseline the following day. However, the kindling-induced sleep alteration was absent in the IL-1R −/− KO mice. Conclusions These results indicate that the IL-1 signal mediates epilepsy-induced sleep disturbance, but dose not participate in kindling-induced epileptogenesis.
Collapse
Affiliation(s)
- Tzu-Rung Huang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4., Roosevelt Road, Taipei, 106, Taiwan
| | - Shuo-Bin Jou
- Department of Neurology, Mackay Memorial Hospital and Mackay Medical College, Taipei, Taiwan
| | - Yu-Ju Chou
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4., Roosevelt Road, Taipei, 106, Taiwan
| | - Pei-Lu Yi
- Department of Sport Management, College of Tourism, Leisure and Sports, Aletheia University, New Taipei City, Taiwan.
| | - Chun-Jen Chen
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Fang-Chia Chang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4., Roosevelt Road, Taipei, 106, Taiwan. .,Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, Taiwan. .,Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
20
|
Abstract
The reciprocal interaction between the immune system and sleep regulation has been widely acknowledged but the cellular mechanisms that underpin this interaction are not completely understood. In the present study, we investigated the role of macrophages in sleep loss- and cold exposure-induced sleep and body temperature responses. Macrophage apoptosis was induced in mice by systemic injection of clodronate-containing liposomes (CCL). We report that CCL treatment induced an immediate and transient increase in non-rapid-eye movement sleep (NREMS) and fever accompanied by decrease in rapid-eye movement sleep, motor activity and NREMS delta power. Chronically macrophage-depleted mice had attenuated NREMS rebound after sleep deprivation compared to normal mice. Cold-induced increase in wakefulness and decrease in NREMS, rapid-eye movement sleep and body temperature were significantly enhanced in macrophage-depleted mice indicating increased cold sensitivity. These findings provide further evidence for the reciprocal interaction among the immune system, sleep and metabolism, and identify macrophages as one of the key cellular elements in this interplay.
Collapse
|
21
|
Yin Y, Liu Y, Pan X, Chen R, Li P, Wu HJ, Zhao ZQ, Li YP, Huang LQ, Zhuang JH, Zhao ZX. Interleukin-1β Promoter Polymorphism Enhances the Risk of Sleep Disturbance in Alzheimer's Disease. PLoS One 2016; 11:e0149945. [PMID: 26937653 PMCID: PMC4777499 DOI: 10.1371/journal.pone.0149945] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 02/07/2016] [Indexed: 01/12/2023] Open
Abstract
Sleep alleviates Alzheimer's disease (AD)-related neuropathological processes, whereas sleep disturbance in AD patients is associated with elevated peripheral inflammatory cytokine levels. In the present study, we assessed interleukin (IL)-1β and APOEε4 polymorphisms for association with susceptibility of sleep disturbances in AD patients. A total of 123 pretreated AD patients and 120 age-, gender- and education level-matched healthy controls were recruited for two consecutive full-night polysomnography and measurement of Epworth Sleepiness Scale (ESS) scores for sleep-wake disturbance. Their genomic DNA was analyzed for IL-1β and APOEε4 SNPs using ligase detection reaction (LDR) technology. Blood levels of IL-1β, IL-6, and tumor necrosis factor alpha (TNF-α) were measured using ELISA after lipopolysaccharide (LPS) stimulation. The odds ratio and 95% confidence interval for genotype-specific risk were calculated using an unconditional logistic regression model and adjusted by age, gender, educational levels, body mass index (BMI), and activities of daily living (ADL). Compared to the non-APOEε4/ε4 genotype, APOEε4/ε4 significantly increased the risk of AD (APOEε4/ε4 vs. non-APOEε4/ε4, adjusted OR = 4.33, 95% CI = 1.33-14.10, p = 0.015). Compared to the IL-1β CC genotype (-31), the TT genotype significantly increased the risk of AD (TT vs. CC, adjusted OR = 1.72, 95% CI = 1.13-2.61, p = 0.010). AD patients carrying the APOEε4 allele and the IL-1β TT genotype showed less time in bed, longer sleep latency and REM latency, more awakenings, and a lower SWS percentage than those carrying CC/CT combined genotypes. In addition, blood IL-1β levels were significantly greater in AD patients carrying both the APOEε4 allele and the IL-1β-31TT genotype than in those carrying the APOEε4 allele and the -31 TC or CC genotype. In conclusion, this study provides the first evidence indicating that the IL-1β-31TT genotype and homozygous APOEε4 combined are associated with increased risk of developing AD with sleep disturbance.
Collapse
Affiliation(s)
- You Yin
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
- Institute of Neuroscience and MOE Key Laboratory of Molecular Neurobiology, Neuroscience Research Center of Changzheng Hospital, The Second Military Medical University Shanghai, Shanghai, China
| | - Yan Liu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiao Pan
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Rui Chen
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Peng Li
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Hui-Juan Wu
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zheng-Qing Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Yan-Peng Li
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Liu-Qing Huang
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jian-Hua Zhuang
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhong-Xin Zhao
- Department of Neurology, Changzheng Hospital, Second Military Medical University, Shanghai, China
- Institute of Neuroscience and MOE Key Laboratory of Molecular Neurobiology, Neuroscience Research Center of Changzheng Hospital, The Second Military Medical University Shanghai, Shanghai, China
| |
Collapse
|
22
|
Geiger SS, Fagundes CT, Siegel RM. Chrono-immunology: progress and challenges in understanding links between the circadian and immune systems. Immunology 2015; 146:349-58. [PMID: 26301993 DOI: 10.1111/imm.12525] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/28/2015] [Accepted: 08/07/2015] [Indexed: 02/06/2023] Open
Abstract
Development of inflammatory diseases, such as metabolic syndrome and cancer, is prevalent in individuals that encounter continuous disruption of their internal clock. Further, daily oscillations in susceptibility to infection as well as a multitude of other immunological processes have been described. Much progress has been made and various mechanisms have been proposed to explain circadian variations in immunity; yet much is still unknown. Understanding the crosstalk between the circadian and the immune systems will allow us to manipulate clock outputs to prevent and treat inflammatory diseases in individuals at risk. This review briefly summarizes current knowledge about circadian rhythms and their role in the immune system and highlights progress and challenges in chrono-immunological research.
Collapse
Affiliation(s)
- Sarah S Geiger
- Immunoregulation Section, Autoimmunity Branch, NIAMS, National Institutes of Health, Bethesda, MD, USA
| | - Caio T Fagundes
- Microorganism/Host Interaction, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Richard M Siegel
- Immunoregulation Section, Autoimmunity Branch, NIAMS, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
Pham LV, Schwartz AR. The pathogenesis of obstructive sleep apnea. J Thorac Dis 2015; 7:1358-72. [PMID: 26380762 DOI: 10.3978/j.issn.2072-1439.2015.07.28] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/17/2015] [Indexed: 12/18/2022]
Abstract
Obstructive sleep apnea (OSA) is a major source of cardiovascular morbidity and mortality, and represents an increasing burden on health care resources. Understanding underlying pathogenic mechanisms of OSA will ultimately allow for the development of rational therapeutic strategies. In this article, we review current concepts about the pathogenesis of OSA. Specifically, we consider the evidence that the upper airway plays a primary role in OSA pathogenesis and provide a framework for modelling its biomechanical properties and propensity to collapse during sleep. Anatomical and neuromuscular factors that modulate upper airway obstruction are also discussed. Finally, we consider models of periodic breathing, and elaborate generalizable mechanisms by which upper airway obstruction destabilizes respiratory patterns during sleep. In our model, upper airway obstruction triggers a mismatch between ventilatory supply and demand. In this model, trade-offs between maintaining sleep stability or ventilation can account for a full range of OSA disease severity and expression. Recurrent arousals and transient increases in airway patency may restore ventilation between periods of sleep, while alterations in neuromuscular and arousal responses to upper airway obstruction may improve sleep stability at still suboptimal levels of ventilation.
Collapse
Affiliation(s)
- Luu V Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins Sleep Disorders Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins Sleep Disorders Center, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Davis CJ, Dunbrasky D, Oonk M, Taishi P, Opp MR, Krueger JM. 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. [PMID: 25449578 PMCID: PMC4418942 DOI: 10.1016/j.bbi.2014.10.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 10/14/2014] [Accepted: 10/23/2014] [Indexed: 12/25/2022] Open
Abstract
Interleukin-1β (IL1) is involved in sleep regulation and sleep responses induced by influenza virus. The IL1 receptor accessory protein (AcP) and an alternatively spliced isoform of AcP found primarily in neurons, AcPb, form part of the IL1 signaling complex. IL1-induced sleep responses depend on injection time. In rat cortex, both IL1 mRNA and AcPb mRNA peak at Zeitgeber Time (ZT) 0 then decline over the daylight hours. Sleep deprivation enhances cortical IL1 mRNA and AcPb mRNA levels, but not AcP mRNA. We used wild type (WT) and AcPb knockout (KO) mice and performed sleep deprivation between ZT10 and 20 or between ZT22 and 8 based on the time of day expression profiles of AcPb and IL1. We hypothesized that the magnitude of the responses to sleep loss would be strain- and time of day-dependent. In WT mice, NREMS and REMS rebounds occurred regardless of when they were deprived of sleep. In contrast, when AcPbKO mice were sleep deprived from ZT10 to 20 NREMS and REMS rebounds were absent. The AcPbKO mice expressed sleep rebound if sleep loss occurred from ZT22 to 8 although the NREMS responses were not as robust as those that occurred in WT mice. We also challenged mice with intranasal H1N1 influenza virus. WT mice exhibited the expected enhanced sleep responses. In contrast, the AcPbKO mice had less sleep after influenza challenge compared to their own baseline values and compared to WT mice. Body temperature and locomotor activity responses after viral challenge were lower and mortality was higher in AcPbKO than in WT mice. We conclude that neuron-specific AcPb plays a critical role in host defenses and sleep homeostasis.
Collapse
Affiliation(s)
- Christopher J. Davis
- College of Medical Sciences and the Sleep and Performance Research Center, Washington State University – Spokane, Spokane, WA 99210,Corresponding Author: Christopher J. Davis, P.O. Box 1495, Spokane, WA 99202, Phone No. 509-358-7820,
| | - Danielle Dunbrasky
- College of Medical Sciences and the Sleep and Performance Research Center, Washington State University – Spokane, Spokane, WA 99210
| | - Marcella Oonk
- College of Medical Sciences and the Sleep and Performance Research Center, Washington State University – Spokane, Spokane, WA 99210
| | - Ping Taishi
- College of Medical Sciences and the Sleep and Performance Research Center, Washington State University – Spokane, Spokane, WA 99210
| | - Mark R. Opp
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA 98104
| | - James M. Krueger
- College of Medical Sciences and the Sleep and Performance Research Center, Washington State University – Spokane, Spokane, WA 99210
| |
Collapse
|
25
|
Haghighi F, Ge Y, Chen S, Xin Y, Umali MU, De Gasperi R, Gama Sosa MA, Ahlers ST, Elder GA. Neuronal DNA Methylation Profiling of Blast-Related Traumatic Brain Injury. J Neurotrauma 2015; 32:1200-9. [PMID: 25594545 DOI: 10.1089/neu.2014.3640] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Long-term molecular changes in the brain resulting from blast exposure may be mediated by epigenetic changes, such as deoxyribonucleic acid (DNA) methylation, that regulate gene expression. Aberrant regulation of gene expression is associated with behavioral abnormalities, where DNA methylation bridges environmental signals to sustained changes in gene expression. We assessed DNA methylation changes in the brains of rats exposed to three 74.5 kPa blast overpressure events, conditions that have been associated with long-term anxiogenic manifestations weeks or months following the initial exposures. Rat frontal cortex eight months post-exposure was used for cell sorting of whole brain tissue into neurons and glia. We interrogated DNA methylation profiles in these cells using Expanded Reduced Representation Bisulfite Sequencing. We obtained data for millions of cytosines, showing distinct methylation profiles for neurons and glia and an increase in global methylation in neuronal versus glial cells (p<10(-7)). We detected DNA methylation perturbations in blast overpressure-exposed animals, compared with sham blast controls, within 458 and 379 genes in neurons and glia, respectively. Differentially methylated neuronal genes showed enrichment in cell death and survival and nervous system development and function, including genes involved in transforming growth factor β and nitric oxide signaling. Functional validation via gene expression analysis of 30 differentially methylated neuronal and glial genes showed a 1.2 fold change in gene expression of the serotonin N-acetyltransferase gene (Aanat) in blast animals (p<0.05). These data provide the first genome-based evidence for changes in DNA methylation induced in response to multiple blast overpressure exposures. In particular, increased methylation and decreased gene expression were observed in the Aanat gene, which is involved in converting serotonin to the circadian hormone melatonin and is implicated in sleep disturbance and depression associated with traumatic brain injury.
Collapse
Affiliation(s)
- Fatemeh Haghighi
- 1 Department of Psychiatry, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Yongchao Ge
- 4 Department of Neurology, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sean Chen
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Yurong Xin
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Michelle U Umali
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Rita De Gasperi
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- 5 Department of Psychiatry, Icahn School of Medicine at Mount Sinai , New York, New York
- 6 Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
| | - Miguel A Gama Sosa
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- 5 Department of Psychiatry, Icahn School of Medicine at Mount Sinai , New York, New York
- 6 Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
| | - Stephen T Ahlers
- 7 Department of Neurotrauma, Operational and Undersea Medicine Directorate Naval Medical Research Center , Silver Spring, Maryland
| | - Gregory A Elder
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- 4 Department of Neurology, Icahn School of Medicine at Mount Sinai , New York, New York
- 5 Department of Psychiatry, Icahn School of Medicine at Mount Sinai , New York, New York
- 8 Neurology Service, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
| |
Collapse
|
26
|
Hiyoshi H, Terao A, Okamatsu-Ogura Y, Kimura K. Characteristics of sleep and wakefulness in wild-derived inbred mice. Exp Anim 2014; 63:205-13. [PMID: 24770646 PMCID: PMC4160977 DOI: 10.1538/expanim.63.205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Genetic variations in the wild-derived inbred mouse strains are more diverse than that
of classical laboratory inbred mouse strains, including C57BL/6J (B6). The sleep/wake and
monoamine properties of six wild-derived inbred mouse strains (PGN2, NJL, BLG2, KJR, MSM,
HMI) were characterized and compared with those of B6 mice. All examined mice were
nocturnal and had a polyphasic sleep pattern with a “main sleep period” identified during
the light period. However, there were three sleep/wake phenotypic differences between the
wild-derived mouse strains and B6 strain. First, the amount of sleep during the dark phase
was comparable with that of B6 mice. However, the amount of sleep during the light phase
was more varied among strains, in particular, NJL and HMI had significantly less sleep
compared with that of B6 mice. Second, PGN2, NJL, BLG2, and KJR mice showed a “highly
awake period” (in which the hourly total sleep time was <10%) immediately after the
onset of the dark period, which was not seen in B6 mice. Third, relative to that of B6
mice, PGN2 and KJR mice showed longer duration of wakefulness episodes during the 12-h
dark phase. Differences in whole brain noradrenaline, dopamine, and 5-hydroxy-tryptamine
contents between the wild-derived mouse strains and B6 strain were also found. These
identified phenotypes might be potentially under strong genetic control. Hence,
wild-derived inbred mice could be useful for identifying the genetic factors underlying
the regulation of sleep and wakefulness.
Collapse
|
27
|
Patil SP, Brown TT, Jacobson LP, Margolick JB, Laffan A, Johnson-Hill L, Godfrey R, Johnson J, Reynolds S, Schwartz AR, Smith PL. Sleep disordered breathing, fatigue, and sleepiness in HIV-infected and -uninfected men. PLoS One 2014; 9:e99258. [PMID: 24991815 PMCID: PMC4084642 DOI: 10.1371/journal.pone.0099258] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 05/12/2014] [Indexed: 12/18/2022] Open
Abstract
Study Objectives We investigated the association of HIV infection and highly active antiretroviral therapy (HAART) with sleep disordered breathing (SDB), fatigue, and sleepiness. Methods HIV-uninfected men (HIV−; n = 60), HIV-infected men using HAART (HIV+/HAART+; n = 58), and HIV-infected men not using HAART (HIV+/HAART−; n = 41) recruited from two sites of the Multicenter AIDS cohort study (MACS) underwent a nocturnal sleep study, anthropometric assessment, and questionnaires for fatigue and the Epworth Sleepiness Scale. The prevalence of SDB in HIV- men was compared to that in men matched from the Sleep Heart Health Study (SHHS). Results The prevalence of SDB was unexpectedly high in all groups: 86.7% for HIV−, 70.7% for HIV+/HAART+, and 73.2% for HIV+/HAART−, despite lower body-mass indices (BMI) in HIV+ groups. The higher prevalence in the HIV− men was significant in univariate analyses but not after adjustment for BMI and other variables. SDB was significantly more common in HIV− men in this study than those in SHHS, and was common in participants with BMIs <25 kg/m2. HIV+ men reported fatigue more frequently than HIV− men (25.5% vs. 6.7%; p = 0.003), but self-reported sleepiness did not differ among the three groups. Sleepiness, but not fatigue, was significantly associated with SDB. Conclusions SDB was highly prevalent in HIV− and HIV+ men, despite a normal or slightly elevated BMI. The high rate of SDB in men who have sex with men deserves further investigation. Sleepiness, but not fatigue, was related to the presence of SDB. Clinicians caring for HIV-infected patients should distinguish between fatigue and sleepiness when considering those at risk for SDB, especially in non-obese men.
Collapse
Affiliation(s)
- Susheel P. Patil
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| | - Todd T. Brown
- Division of Endocrinology and Metabolism, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Lisa P. Jacobson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Joseph B. Margolick
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Alison Laffan
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Lisette Johnson-Hill
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Rebecca Godfrey
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jacquett Johnson
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Sandra Reynolds
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Alan R. Schwartz
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Philip L. Smith
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
28
|
Brigham EP, Patil SP, Jacobson LP, Margolick JB, Godfrey R, Johnson J, Johnson-Hill LM, Reynolds S, Schwartz AR, Smith PL, Brown TT. Association between systemic inflammation and obstructive sleep apnea in men with or at risk for HIV infection. Antivir Ther 2014; 19:725-33. [PMID: 24518040 PMCID: PMC4130807 DOI: 10.3851/imp2745] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND We sought to determine whether markers of systemic inflammation are associated with the presence of moderate/severe obstructive sleep apnea (OSA) and whether this association differs based on HIV and HIV treatment status. METHODS HIV-uninfected men (HIV-; n=60), HIV-infected men receiving HAART (HIV+/HAART; n=58) and HIV-infected men not receiving HAART (HIV+/no HAART; n=41) underwent polysomnograpy and measurement of plasma levels of tumour necrosis factor (TNF)-α, soluble TNF-α receptors I and II (sTNFRI and sTNFRII) and interleukin (IL)-6. The relationship between moderate/severe OSA (respiratory disturbance index ≥15 apnea/hypopnea events/h) and inflammatory markers was assessed with multivariable regression models. RESULTS Compared with the HIV- men, HIV+/HAART men and HIV+/no HAART men had higher levels of TNF-α, sTNFRI and sTNFRII, independent of age, race, smoking status, obstructive lung disease (OLD) and body mass index (BMI). Moderate/severe OSA was present in 48% of the sample (HIV- 57%; HIV+/HAART 41%; HIV+/no HAART 44%). Among the HIV+/no HAART men, but not in the other groups, TNF-α, sTNFRII and IL-6 levels were higher in those with moderate/severe OSA compared to men with no/mild OSA after adjustment for age, race, smoking status, OLD and BMI. Within this group, the association of high TNF-α concentrations with moderate/severe OSA was also independent of CD4(+) T-cell count and plasma HIV RNA concentration. CONCLUSIONS Compared with HIV+/HAART men and HIV- men, markers of systemic inflammation were higher in HIV+/no HAART men. In these men, TNF-α was significantly related to OSA, independent of HIV-related covariates.
Collapse
Affiliation(s)
- Emily P Brigham
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Susheel P Patil
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lisa P Jacobson
- The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Joseph B Margolick
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rebecca Godfrey
- The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jacquett Johnson
- The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Sandra Reynolds
- The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alan R Schwartz
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip L Smith
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Todd T Brown
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Rowe RK, Striz M, Bachstetter AD, Van Eldik LJ, Donohue KD, O'Hara BF, Lifshitz J. Diffuse brain injury induces acute post-traumatic sleep. PLoS One 2014; 9:e82507. [PMID: 24416145 PMCID: PMC3885381 DOI: 10.1371/journal.pone.0082507] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 10/24/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Clinical observations report excessive sleepiness immediately following traumatic brain injury (TBI); however, there is a lack of experimental evidence to support or refute the benefit of sleep following a brain injury. The aim of this study is to investigate acute post-traumatic sleep. METHODS Sham, mild or moderate diffuse TBI was induced by midline fluid percussion injury (mFPI) in male C57BL/6J mice at 9:00 or 21:00 to evaluate injury-induced sleep behavior at sleep and wake onset, respectively. Sleep profiles were measured post-injury using a non-invasive, piezoelectric cage system. In separate cohorts of mice, inflammatory cytokines in the neocortex were quantified by immunoassay, and microglial activation was visualized by immunohistochemistry. RESULTS Immediately after diffuse TBI, quantitative measures of sleep were characterized by a significant increase in sleep (>50%) for the first 6 hours post-injury, resulting from increases in sleep bout length, compared to sham. Acute post-traumatic sleep increased significantly independent of injury severity and time of injury (9:00 vs 21:00). The pro-inflammatory cytokine IL-1β increased in brain-injured mice compared to sham over the first 9 hours post-injury. Iba-1 positive microglia were evident in brain-injured cortex at 6 hours post-injury. CONCLUSION Post-traumatic sleep occurs for up to 6 hours after diffuse brain injury in the mouse regardless of injury severity or time of day. The temporal profile of secondary injury cascades may be driving the significant increase in post-traumatic sleep and contribute to the natural course of recovery through cellular repair.
Collapse
Affiliation(s)
- Rachel K. Rowe
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, United States of America
- Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Martin Striz
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| | - Adam D. Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Linda J. Van Eldik
- Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Kevin D. Donohue
- Department of Electrical and Computer Engineering, College of Engineering, University of Kentucky, Lexington, Kentucky, United States of America
| | - Bruce F. O'Hara
- Department of Biology, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, Arizona, United States of America
- Department of Child Health, University of Arizona College of Medicine–Phoenix, Phoenix, Arizona, United States of America
- Phoenix Veteran Affairs Healthcare System, Phoenix, Arizona, United States of America
- Department of Anatomy and Neurobiology, College of Medicine, University of Kentucky, Lexington, Kentucky, United States of America
- Department of Physical Medicine & Rehabilitation, University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky College of Medicine, Lexington, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
30
|
Mueller AD, Parfyonov M, Pavlovski I, Marchant EG, Mistlberger RE. The inhibitory effect of sleep deprivation on cell proliferation in the hippocampus of adult mice is eliminated by corticosterone clamp combined with interleukin-1 receptor 1 knockout. Brain Behav Immun 2014; 35:182-8. [PMID: 24121035 DOI: 10.1016/j.bbi.2013.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/22/2013] [Accepted: 10/01/2013] [Indexed: 11/17/2022] Open
Abstract
Deprivation or fragmentation of sleep for longer than 2days significantly inhibits cell proliferation and neurogenesis in the hippocampus of adult rats and mice. Signaling pathways that mediate these effects have yet to be clarified. Although deprivation procedures can stimulate adrenal corticosterone (CORT) release, suppression of cell proliferation by sleep deprivation does not require elevated CORT. We examined a role for interleukin-1β (IL-1β), a pro-inflammatory cytokine that is increased by sleep loss and that mediates effects of stress on hippocampal neurogenesis. Wild type (WT) and IL-1 receptor 1 knockout (IL1RI-KO) mice were subjected to rapid-eye-movement sleep deprivation (RSD) for 72-h using the multiple platform-over-water method. Mice were administered BrdU (100mg/kg) i.p. at hour 70 of RSD and were sacrificed 2-h later. New cells were identified by immunoreactivity (ir) for BrdU and Ki67 in the granular cell layer/subgranular zone (GCL/SGZ) and the hilus. In Experiment 1, WT and IL1RI-KO mice, by contrast with respective control groups, exhibited significantly fewer BrdU-ir and Ki67-ir cells. In Experiment 2, WT and IL1RI-KO mice were adrenalectomized (ADX) and maintained on constant low-dose CORT by osmotic minipumps. RSD reduced cell proliferation by 32% (p<0.01) in ADX-WT animals but did not significantly reduce proliferation in ADX IL1RI-KO animals (p>0.1). These results imply that RSD suppresses cell proliferation by the presence of wake-dependent factors (either elevated CORT or IL-1β signaling are sufficient), rather than the absence of a REM sleep-dependent process. The generality of these findings to other sleep deprivation methods and durations remains to be established.
Collapse
Affiliation(s)
- Anka D Mueller
- Cognitive and Neural Sciences Program, Department of Psychology, Simon Fraser University, Burnaby, BC V5A1S6, Canada
| | | | | | | | | |
Collapse
|
31
|
Alt JA, Sautter NB, Mace JC, Detwiller KY, Smith TL. Antisomnogenic cytokines, quality of life, and chronic rhinosinusitis: a pilot study. Laryngoscope 2013; 124:E107-14. [PMID: 24115141 DOI: 10.1002/lary.24412] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 07/24/2013] [Accepted: 08/26/2013] [Indexed: 11/08/2022]
Abstract
OBJECTIVES/HYPOTHESIS Sleep disturbance, reduced quality of life (QOL), and other components of "sickness behavior" in patients with chronic rhinosinusitis (CRS) are poorly understood. These complex changes in central behavior are due to the effects of immune mediators acting in the brain. We hypothesized that immune mediators that have been associated with CRS are also associated with sickness behavior, somnifacient complaints, and CRS disease-specific QOL. STUDY DESIGN Pilot study. METHODS Twenty patients with CRS were prospectively enrolled and completed the Pittsburgh Sleep Quality Index (PSQI), disease-specific QOL, and olfactory instruments. Ethmoid mucosa was obtained and reverse transcription-polymerase chain reaction was performed for the cytokines interleukin (IL)-4, -13, and transforming growth factor-β (TGF-β). Average change in crossover threshold was calculated, and differences in gene expression were correlated with sleep quality, CRS-specific QOL, and disease severity. RESULTS Patients with CRS reported overall poor sleep quality and poor CRS-specific QOL with significant correlations between them. Increased expression of TGF-β (r = -0.443; P = .050) and IL-4 (r = -0.548; P = .012) correlated with sleep dysfunction, whereas IL-13 expression was linearly associated with worse sleep quality (PSQI scores r = -0.417; P = .075). IL-4 and TGF-β expression was not associated with CRS disease severity or QOL, whereas significantly higher levels of IL-13 expression correlated with worse CRS disease severity and QOL. CONCLUSIONS Patients with CRS exhibited behavioral changes commonly referred to as sickness behavior, which include poor sleep quality and reduced QOL. The upregulation of IL-4 and TGF-β may contribute to inflammatory brain-mediated effects on sleep quality, whereas IL-13 may be a pleiotropic signaling molecule influencing sleep, QOL, and CRS disease severity. LEVEL OF EVIDENCE 2b.
Collapse
Affiliation(s)
- Jeremiah A Alt
- Division of Rhinology and Sinus Surgery, Oregon Sinus Center, Department of Otolaryngology-Head and Neck Surgery, Oregon Health and Science University, Portland, Oregon, U.S.A
| | | | | | | | | |
Collapse
|
32
|
Alt JA, Smith TL. Chronic rhinosinusitis and sleep: a contemporary review. Int Forum Allergy Rhinol 2013; 3:941-9. [PMID: 24039230 DOI: 10.1002/alr.21217] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 06/25/2013] [Accepted: 07/26/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Patients with chronic rhinosinusitis (CRS) exhibit centrally mediated behavioral changes commonly referred to as "sickness behavior." Sleep alteration is a component of sickness behavior which is estimated to affect up to 70 million patients annually. Patients with CRS have poor sleep quality, and little is known about the underlying etiology and pathophysiology. This narrative review aims to further organize and present the current knowledge associating sleep and CRS. METHODS A literature search was conducted of the OVID MEDLINE database using key search words including: "chronic rhinosinusitis," "sleep," "sleep disorders," and "sleep dysfunction." Additional keywords "nasal obstruction," "nasal polyp," and "fatigue" were identified and used to further delineate relevant articles. RESULTS The articles that specifically addressed sleep and CRS were dissected and presented as follows: (1) chronic rhinosinusitis and sleep; (2) chronic rhinosinusitis and fatigue; (3) chronic rhinosinusitis, nasal obstruction, and sleep; and (4) pathophysiology of sleep in chronic rhinosinusitis (cytokines in both sleep and chronic rhinosinusitis and their association to the neuroimmune biology of chronic rhinosinusitis). CONCLUSION Patients with CRS have sleep dysfunction that is associated with their disease severity and overall quality of life. The etiology of sleep dysfunction in CRS is most likely multifactorial. Increasing evidence suggests sleep dysfunction in patients with CRS is partly due to the inflammatory disease process, and sleep physiology in patients with CRS may be actively regulated by the inflammatory component of the disease.
Collapse
Affiliation(s)
- Jeremiah A Alt
- Division of Rhinology and Sinus Surgery, Oregon Sinus Center, Department of Otolaryngology-Head and Neck Surgery, Oregon Health and Science University, Portland, OR
| | | |
Collapse
|
33
|
Affiliation(s)
- Christopher J. Davis
- Sleep and Performance Research Center, WWAMI Medical Education and Program in Neuroscience, Washington State University, 412 E Spokane Falls Boulevard, Spokane, WA 99210-1495, USA
| | - James M. Krueger
- Sleep and Performance Research Center, WWAMI Medical Education and Program in Neuroscience, Washington State University, 412 E Spokane Falls Boulevard, Spokane, WA 99210-1495, USA
| |
Collapse
|
34
|
Abstract
This review summarizes the brain mechanisms controlling sleep and wakefulness. Wakefulness promoting systems cause low-voltage, fast activity in the electroencephalogram (EEG). Multiple interacting neurotransmitter systems in the brain stem, hypothalamus, and basal forebrain converge onto common effector systems in the thalamus and cortex. Sleep results from the inhibition of wake-promoting systems by homeostatic sleep factors such as adenosine and nitric oxide and GABAergic neurons in the preoptic area of the hypothalamus, resulting in large-amplitude, slow EEG oscillations. Local, activity-dependent factors modulate the amplitude and frequency of cortical slow oscillations. Non-rapid-eye-movement (NREM) sleep results in conservation of brain energy and facilitates memory consolidation through the modulation of synaptic weights. Rapid-eye-movement (REM) sleep results from the interaction of brain stem cholinergic, aminergic, and GABAergic neurons which control the activity of glutamatergic reticular formation neurons leading to REM sleep phenomena such as muscle atonia, REMs, dreaming, and cortical activation. Strong activation of limbic regions during REM sleep suggests a role in regulation of emotion. Genetic studies suggest that brain mechanisms controlling waking and NREM sleep are strongly conserved throughout evolution, underscoring their enormous importance for brain function. Sleep disruption interferes with the normal restorative functions of NREM and REM sleep, resulting in disruptions of breathing and cardiovascular function, changes in emotional reactivity, and cognitive impairments in attention, memory, and decision making.
Collapse
Affiliation(s)
- Ritchie E Brown
- Laboratory of Neuroscience, VA Boston Healthcare System and Harvard Medical School, Brockton, Massachusetts 02301, USA
| | | | | | | | | |
Collapse
|
35
|
Clinton JM, Davis CJ, Zielinski MR, Jewett KA, Krueger JM. Biochemical regulation of sleep and sleep biomarkers. J Clin Sleep Med 2012; 7:S38-42. [PMID: 22003330 DOI: 10.5664/jcsm.1360] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Symptoms commonly associated with sleep loss and chronic inflammation include sleepiness, fatigue, poor cognition, enhanced sensitivity to pain and kindling stimuli, excess sleep and increases in circulating levels of tumor necrosis factor α (TNF) in humans and brain levels of interleukin-1 β (IL1) and TNF in animals. Cytokines including IL1 and TNF partake in non-rapid eye movement sleep (NREMS) regulation under physiological and inflammatory conditions. Administration of exogenous IL1 or TNF mimics the accumulation of these cytokines occurring during sleep loss to the extent that it induces the aforementioned symptoms. Extracellular ATP associated with neuro- and glio-transmission, acting via purine type 2 receptors, e.g., the P2X7 receptor, has a role in glia release of IL1 and TNF. These substances in turn act on neurons to change their intrinsic membrane properties and sensitivities to neurotransmitters and neuromodulators such as adenosine, glutamate and GABA. These actions change the network input-output properties, i.e., a state shift for the network. State oscillations occur locally within cortical columns and are defined using evoked response potentials. One such state, so defined, shares properties with whole animal sleep in that it is dependent on prior cellular activity--it shows homeostasis. The cortical column sleep-like state is induced by TNF and is associated with experimental performance detriments. ATP released extracellularly as a consequence of cellular activity is posited to initiate a mechanism by which the brain tracks its prior sleep-state history to induce/prohibit sleep. Thus, sleep is an emergent property of populations of local neural networks undergoing state transitions. Specific neuronal groups participating in sleep depend upon prior network use driving local network state changes via the ATP-cytokine-adenosine mechanism. Such considerations add complexity to finding biochemical markers for sleepiness.
Collapse
Affiliation(s)
- James M Clinton
- Sleep and Performance Research Center, WWAMI Medical Education Program, WA State University, Spokane, WA 99164, USA.
| | | | | | | | | |
Collapse
|
36
|
Taishi P, Davis CJ, Bayomy O, Zielinski MR, Liao F, Clinton JM, Smith DE, Krueger JM. Brain-specific interleukin-1 receptor accessory protein in sleep regulation. J Appl Physiol (1985) 2012; 112:1015-22. [PMID: 22174404 PMCID: PMC3311656 DOI: 10.1152/japplphysiol.01307.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 12/14/2011] [Indexed: 12/17/2022] Open
Abstract
Interleukin (IL)-1β is involved in several brain functions, including sleep regulation. It promotes non-rapid eye movement (NREM) sleep via the IL-1 type I receptor. IL-1β/IL-1 receptor complex signaling requires adaptor proteins, e.g., the IL-1 receptor brain-specific accessory protein (AcPb). We have cloned and characterized rat AcPb, which shares substantial homologies with mouse AcPb and, compared with AcP, is preferentially expressed in the brain. Furthermore, rat somatosensory cortex AcPb mRNA varied across the day with sleep propensity, increased after sleep deprivation, and was induced by somnogenic doses of IL-1β. Duration of NREM sleep was slightly shorter and duration of REM sleep was slightly longer in AcPb knockout than wild-type mice. In response to lipopolysaccharide, which is used to induce IL-1β, sleep responses were exaggerated in AcPb knockout mice, suggesting that, in normal mice, inflammation-mediated sleep responses are attenuated by AcPb. We conclude that AcPb has a role in sleep responses to inflammatory stimuli and, possibly, in physiological sleep regulation.
Collapse
Affiliation(s)
- Ping Taishi
- Sleep and Performance Research Center, WWAMI Medical Education Program, Washington State University, Spokane, WA 99210-1495, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Schmidt MA, Wisor JP. Interleukin 1 receptor contributes to methamphetamine- and sleep deprivation-induced hypersomnolence. Neurosci Lett 2012; 513:209-13. [PMID: 22387068 DOI: 10.1016/j.neulet.2012.02.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 01/20/2012] [Accepted: 02/13/2012] [Indexed: 10/28/2022]
Abstract
Methamphetamine-induced wakefulness is dependent on monoamine transporter blockade. Subsequent to methamphetamine-induced wakefulness, the amount of time spent asleep and the depth of sleep are increased relative to baseline sleep. The mechanisms that drive methamphetamine-induced hypersomnolence are not fully understood. We recently observed that methamphetamine exposure elevates the expression of the sleep-promoting cytokine, interleukin-1β in CD11b-positive monocytes within the brain. Here, we sought to determine whether activation of the interleukin 1 receptor (IL1R) drives the increase in the depth and amount of sleep that occurs subsequent to methamphetamine-induced wakefulness. IL1R-deficient mice and wild type control mice were subjected to systemic methamphetamine (1 and 2mg/kg) and saline treatments. The wake-promoting effect of methamphetamine was modestly potentiated by IL1R-deficiency. Additionally, the increase in time spent in NREMS subsequent to methamphetamine-induced wakefulness in wild type mice was abolished in IL1R-deficient mice. The increase in time spent asleep after 3h of behaviorally enforced wakefulness was also abolished in IL1R-deficient mice. Increases in EEG slow wave activity triggered by methamphetamine and sleep deprivation were of equal magnitude in IL1R-deficient and wild type mice. These data demonstrate that IL1R activation contributes to hypersomnolence that occurs after sleep loss, whether that sleep loss is triggered pharmacologically by methamphetamine or through behavioral sleep deprivation.
Collapse
Affiliation(s)
- Michelle A Schmidt
- WWAMI Medical Education Program and Department of Veterinary Comparative Anatomy, Pharmacology and Physiology, Washington State University, Spokane, WA 99202, United States
| | | |
Collapse
|
38
|
Jewett KA, Krueger JM. Humoral sleep regulation; interleukin-1 and tumor necrosis factor. VITAMINS AND HORMONES 2012; 89:241-57. [PMID: 22640617 PMCID: PMC4030541 DOI: 10.1016/b978-0-12-394623-2.00013-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Two substances, the cytokines interleukin-1 beta (IL1β) and tumor necrosis factor alpha (TNFα), known for their many physiological roles, for example, cognition, synaptic plasticity, and immune function, are also well characterized in their actions of sleep regulation. These substances promote non-rapid eye movement sleep and can induce symptoms associated with sleep loss such as sleepiness, fatigue, and poor cognition. IL1β and TNFα are released from glia in response to extracellular ATP. They bind to their receptors on neurons resulting in neuromodulator and neurotransmitter receptor up/downregulation (e.g., adenosine and glutamate receptors) leading to altered neuronal excitability and function, that is, a state change in the local network. Synchronization of state between local networks leads to emergent whole brain oscillations, such as sleep/wake cycles.
Collapse
Affiliation(s)
- Kathryn A Jewett
- WWAMI Medical Education Program, Sleep and Performance Research Center, Washington State University, Spokane, Washington, USA
| | | |
Collapse
|
39
|
Baracchi F, Ingiosi AM, Raymond RM, Opp MR. Sepsis-induced alterations in sleep of rats. Am J Physiol Regul Integr Comp Physiol 2011; 301:R1467-78. [PMID: 21900639 DOI: 10.1152/ajpregu.00354.2011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Sepsis is a systemic immune response to infection that may result in multiple organ failure and death. Polymicrobial infections remain a serious clinical problem, and in the hospital, sepsis is the number-one noncardiac killer. Although the central nervous system may be one of the first systems affected, relatively little effort has been made to determine the impact of sepsis on the brain. In this study, we used the cecal ligation and puncture (CLP) model to determine the extent to which sepsis alters sleep, the EEG, and brain temperature (Tbr) of rats. Sepsis increases the amount of time rats spend in non-rapid eye movement sleep (NREMS) during the dark period, but not during the light period. Rapid eye movements sleep (REMS) of septic rats is suppressed for about 24 h following CLP surgery, after which REMS increases during dark periods for at least three nights. The EEG is dramatically altered shortly after sepsis induction, as evidenced by reductions in slow-frequency components. Furthermore, sleep is fragmented, indicating that the quality of sleep is diminished. Effects on sleep, the EEG, and Tbr persist for at least 84 h after sepsis induction, the duration of our recording period. Immunohistochemical assays focused on brain stem mechanisms responsible for alterations in REMS, as little information is available concerning infection-induced suppression of this sleep stage. Our immunohistochemical data suggest that REMS suppression after sepsis onset may be mediated, in part, by the brain stem GABAergic system. This study demonstrates for the first time that sleep and EEG patterns are altered during CLP-induced sepsis. These data suggest that the EEG may serve as a biomarker for sepsis onset. These data also contribute to our knowledge of potential mechanisms, whereby infections alter sleep and other central nervous system functions.
Collapse
Affiliation(s)
- Francesca Baracchi
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
40
|
Krueger JM, Clinton JM, Winters BD, Zielinski MR, Taishi P, Jewett KA, Davis CJ. Involvement of cytokines in slow wave sleep. PROGRESS IN BRAIN RESEARCH 2011; 193:39-47. [PMID: 21854954 PMCID: PMC3645329 DOI: 10.1016/b978-0-444-53839-0.00003-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cytokines such as tumor necrosis factor alpha (TNFα) and interleukin-1 beta (IL1β) play a role in sleep regulation in health and disease. TNFα or IL1β injection enhances non-rapid eye movement sleep. Inhibition of TNFα or IL1β reduces spontaneous sleep. Mice lacking TNFα or IL1β receptors sleep less. In normal humans and in multiple disease states, plasma levels of TNFα covary with EEG slow wave activity (SWA) and sleep propensity. Many of the symptoms induced by sleep loss, for example, sleepiness, fatigue, poor cognition, enhanced sensitivity to pain, are elicited by injection of exogenous TNFα or IL1β. IL1β or TNFα applied unilaterally to the surface of the cortex induces state-dependent enhancement of EEG SWA ipsilaterally, suggesting greater regional sleep intensity. Interventions such as unilateral somatosensory stimulation enhance localized sleep EEG SWA, blood flow, and somatosensory cortical expression of IL1β and TNFα. State oscillations occur within cortical columns. One such state shares properties with whole animal sleep in that it is dependent on prior cellular activity, shows homeostasis, and is induced by TNFα. Extracellular ATP released during neuro- and gliotransmission enhances cytokine release via purine type 2 receptors. An ATP agonist enhances sleep, while ATP antagonists inhibit sleep. Mice lacking the P2X7 receptor have attenuated sleep rebound responses after sleep loss. TNFα and IL1β alter neuron sensitivity by changing neuromodulator/neurotransmitter receptor expression, allowing the neuron to scale its activity to the presynaptic neurons. TNFα's role in synaptic scaling is well characterized. Because the sensitivity of the postsynaptic neuron is changed, the same input will result in a different network output signal and this is a state change. The top-down paradigm of sleep regulation requires intentional action from sleep/wake regulatory brain circuits to initiate whole-organism sleep. This raises unresolved questions as to how such purposeful action might itself be initiated. In the new paradigm, sleep is initiated within networks and local sleep is a direct consequence of prior local cell activity. Whole-organism sleep is a bottom-up, self-organizing, and emergent property of the collective states of networks throughout the brain.
Collapse
Affiliation(s)
- James M Krueger
- Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Almost 20 years ago, the gene underlying fatal familial insomnia was discovered, and first suggested the concept that a single gene can regulate sleep. In the two decades since, there have been many advances in the field of behavioral genetics, but it is only in the past 10 years that the genetic analysis of sleep has emerged as an important discipline. Major findings include the discovery of a single gene underlying the sleep disorder narcolepsy, and identification of loci that make quantitative contributions to sleep characteristics. The sleep field has also expanded its focus from mammalian model organisms to Drosophila, zebrafish, and worms, which is allowing the application of novel genetic approaches. Researchers have undertaken large-scale screens to identify new genes that regulate sleep, and are also probing questions of sleep circuitry and sleep function on a molecular level. As genetic tools continue to be refined in each model organism, the genes that support a specific function in sleep will become more apparent. Thus, while our understanding of sleep still remains rudimentary, rapid progress is expected from these recently initiated studies.
Collapse
Affiliation(s)
- Amanda Crocker
- Howard Hughes Medical institute, Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
42
|
Hight K, Hallett H, Churchill L, De A, Boucher A, Krueger JM. Time of day differences in the number of cytokine-, neurotrophin- and NeuN-immunoreactive cells in the rat somatosensory or visual cortex. Brain Res 2010; 1337:32-40. [PMID: 20398636 PMCID: PMC2892412 DOI: 10.1016/j.brainres.2010.04.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 03/31/2010] [Accepted: 04/07/2010] [Indexed: 12/22/2022]
Abstract
Sensory input to different cortical areas differentially varies across the light-dark cycle and likely is responsible, in part, for activity-dependent changes in time-of-day differences in protein expression such as Fos. In this study we investigate time-of-day differences between dark (just before light onset) and light (just before dark onset) for the number of immunoreactive (IR) neurons that stained for tumor necrosis factor alpha (TNFalpha), interleukin-1 beta (IL1 beta), nerve growth factor (NGF), the neuronal nuclear protein (NeuN) and Fos in the rat somatosensory cortex (Sctx) and visual cortex (Vctx). Additionally, astrocyte IL1 beta-IR in the Sctx and Vctx was determined. TNFalpha and IL1 beta, as well as the immediate early gene protein Fos, were higher at the end of the dark phase (2300 h) compared to values obtained at the end of the light phase (1100 h) in the Sctx and Vctx. IL1 beta-IR in Sctx and Vctx astrocytes was higher at 2300 h than that observed at 1100 h. . In contrast, the number of NGF-IR neurons was higher in the Vctx than in the Sctx but did not differ in time. However, the density of the NGF-IR neurons in layer V was greater at 2300 h in the Sctx than at 1100 h. NeuN-IR was higher at 2300 h in the Sctx but was lower at this time in the Vctx compared to 1100 h. These data demonstrate that expressions of the molecules examined are dependent on activity, the sleep-wake cycle and brain location. These factors interact to modulate time-of-day expression.
Collapse
Affiliation(s)
- Krista Hight
- Dept. of Veterinary & Comparative Anatomy, Pharmacology and Physiology, Sleep and Performance Research Center, Program in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520
| | - Heather Hallett
- Dept. of Veterinary & Comparative Anatomy, Pharmacology and Physiology, Sleep and Performance Research Center, Program in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520
- WWAMI Program at the University of Washington Medical School, Pullman, WA
| | - Lynn Churchill
- Dept. of Veterinary & Comparative Anatomy, Pharmacology and Physiology, Sleep and Performance Research Center, Program in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520
| | - Alok De
- Dept. of Veterinary & Comparative Anatomy, Pharmacology and Physiology, Sleep and Performance Research Center, Program in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520
- Dept. of OB/ Gyn, School of Medicine, University of Missouri, Kansas City, Kansas City, Missouri 64108
| | - Andrea Boucher
- Dept. of Veterinary & Comparative Anatomy, Pharmacology and Physiology, Sleep and Performance Research Center, Program in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520
| | - James M. Krueger
- Dept. of Veterinary & Comparative Anatomy, Pharmacology and Physiology, Sleep and Performance Research Center, Program in Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520
| |
Collapse
|
43
|
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. [PMID: 20144235 PMCID: PMC2831041 DOI: 10.1186/1471-2202-11-17] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 02/09/2010] [Indexed: 11/27/2022] Open
Abstract
Background Immune challenge impacts behavior in many species. In mammals, this adaptive behavior is often manifested as an increase in sleep. Sleep has therefore been proposed to benefit the host by enhancing immune function and thereby overcome the challenge. To facilitate genetic studies on the relationship between sleep and immune function, we characterized the effect of the immune response on sleep in Drosophila melanogaster. Behavioral features of sleep as well as the innate immune response signaling pathways are well characterized in flies and are highly conserved in mammals. Results An immune response induced by infection with Gram-negative bacteria or by aseptic injury increased sleep in flies. The increase in sleep occurred during the morning hours after treatment and the magnitude of the effect was dependent on the time-of-day of inoculation or injury such that night-time treatment had a stronger effect than that during the daytime. This pattern persisted in constant darkness, indicating a role of the circadian clock. Mutants of the circadian clock gene, period, eliminated the increase in sleep observed in the morning, but instead showed enhanced sleep immediately after injury or infection. Null mutants of the Nuclear Factor κB (NFκB) Relish, which is central to the innate immune response, do not increase sleep in response to injury or infection at any time of day. Instead, they maintain a normal sleep pattern until they die. Expression of a full-length Relish transgene in the fat bodies of Relish mutants restored the morning increase in sleep during an immune response. Fat bodies are a major site of immune signalling in flies and have a key role in host defense. Conclusions These data demonstrate that an immune response increases sleep in flies in a manner that is gated by the circadian clock and that requires the NFκB Relish. These findings support a role of sleep in a recovery process and demonstrate a conserved feature of the Drosophila model of sleep.
Collapse
Affiliation(s)
- Tzu-Hsing Kuo
- Center for Advanced Biotechnology and Medicine, Department of Pharmacology University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
44
|
Schwartz AR, Patil SP, Squier S, Schneider H, Kirkness JP, Smith PL. Obesity and upper airway control during sleep. J Appl Physiol (1985) 2009; 108:430-5. [PMID: 19875707 DOI: 10.1152/japplphysiol.00919.2009] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mechanisms linking obesity with upper airway dysfunction in obstructive sleep apnea are reviewed. Obstructive sleep apnea is due to alterations in upper airway anatomy and neuromuscular control. Upper airway structural alterations in obesity are related to adipose deposition around the pharynx, which can increase its collapsibility or critical pressure (P(crit)). In addition, obesity and, particularly, central adiposity lead to reductions in resting lung volume, resulting in loss of caudal traction on upper airway structures and parallel increases in pharyngeal collapsibility. Metabolic and humoral factors that promote central adiposity may contribute to these alterations in upper airway mechanical function and increase sleep apnea susceptibility. In contrast, neural responses to upper airway obstruction can mitigate these mechanical loads and restore pharyngeal patency during sleep. Current evidence suggests that these responses can improve with weight loss. Improvements in these neural responses with weight loss may be related to a decline in systemic and local pharyngeal concentrations of specific inflammatory mediators with somnogenic effects.
Collapse
Affiliation(s)
- Alan R Schwartz
- Sleep Disorders Center, Johns Hopkins School of Medicine, Baltimore, Maryland 21224, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Clark IA, Budd AC, Alleva LM. Sickness behaviour pushed too far--the basis of the syndrome seen in severe protozoal, bacterial and viral diseases and post-trauma. Malar J 2008; 7:208. [PMID: 18854046 PMCID: PMC2576339 DOI: 10.1186/1475-2875-7-208] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Accepted: 10/14/2008] [Indexed: 12/03/2022] Open
Abstract
Certain distinctive components of the severe systemic inflammatory syndrome are now well-recognized to be common to malaria, sepsis, viral infections, and post-trauma illness. While their connection with cytokines has been appreciated for some time, the constellation of changes that comprise the syndrome has simply been accepted as an empirical observation, with no theory to explain why they should coexist. New data on the effects of the main pro-inflammatory cytokines on the genetic control of sickness behaviour can be extended to provide a rationale for why this syndrome contains many of its accustomed components, such as reversible encephalopathy, gene silencing, dyserythropoiesis, seizures, coagulopathy, hypoalbuminaemia and hypertriglyceridaemia. It is thus proposed that the pattern of pathology that comprises much of the systemic inflammatory syndrome occurs when one of the usually advantageous roles of pro-inflammatory cytokines – generating sickness behaviour by moderately repressing genes (Dbp, Tef, Hlf, Per1, Per2 and Per3, and the nuclear receptor Rev-erbα) that control circadian rhythm – becomes excessive. Although reversible encephalopathy and gene silencing are severe events with potentially fatal consequences, they can be viewed as having survival advantages through lowering energy demand. In contrast, dyserythropoiesis, seizures, coagulopathy, hypoalbuminaemia and hypertriglyceridaemia may best be viewed as unfortunate consequences of extreme repression of these same genetic controls when the pro-inflammatory cytokines that cause sickness behaviour are produced excessively. As well as casting a new light on the previously unrationalized coexistence of these aspects of systemic inflammatory diseases, this concept is consistent with the case for a primary role for inflammatory cytokines in their pathogenesis across this range of diseases.
Collapse
Affiliation(s)
- Ian A Clark
- School of Biochemistry and Molecular Biology, Australian National University, Canberra, Australia.
| | | | | |
Collapse
|
46
|
Baracchi F, Opp MR. Sleep-wake behavior and responses to sleep deprivation of mice lacking both interleukin-1 beta receptor 1 and tumor necrosis factor-alpha receptor 1. Brain Behav Immun 2008; 22:982-93. [PMID: 18329246 PMCID: PMC4164115 DOI: 10.1016/j.bbi.2008.02.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 01/29/2008] [Accepted: 02/03/2008] [Indexed: 01/15/2023] Open
Abstract
Data indicate that interleukin (IL)-1 beta and tumor necrosis factor-alpha (TNFalpha) are involved in the regulation of non-rapid eye movement sleep (NREMS). Previous studies demonstrate that mice lacking the IL-1 beta type 1 receptor spend less time in NREMS during the light period, whereas mice lacking the p55 (type 1) receptor for TNFalpha spend less time in NREMS during the dark period. To further investigate roles for IL-1 beta and TNFalpha in sleep regulation we phenotyped sleep and responses to sleep deprivation of mice lacking both the IL-1 beta receptor 1 and TNFalpha receptor 1 (IL-1R1/TNFR1 KO). Male adult mice (IL-1R1/TNFR1 KO, n=14; B6129SF2/J, n=14) were surgically instrumented with EEG electrodes and with a thermistor to measure brain temperature. After recovery and adaptation to the recording apparatus, 48 h of undisturbed baseline recordings were obtained. Mice were then subjected to 6h sleep deprivation at light onset by gentle handling. IL-1R1/TNFR1 KO mice spent less time in NREMS during the last 6h of the dark period and less time in rapid eye movement sleep (REMS) during the light period. There were no differences between strains in the diurnal timing of delta power during NREMS. However, there were strain differences in the relative power spectra of the NREMS EEG during both the light period and the dark period. In addition, during the light period relative power in the theta frequency band of the REMS EEG differed between strains. After sleep deprivation, control mice exhibited prolonged increases in NREMS and REMS, whereas the duration of the NREMS increase was shorter and there was no increase in REMS of IL-1R1/TNFR1 KO mice. Delta power during NREMS increased in both strains after sleep deprivation, but the increase in delta power during NREMS of IL-1R1/TNFR1 KO mice was of greater magnitude and of longer duration than that observed in control mice. These results provide additional evidence that the IL-1 beta and TNFalpha cytokine systems play a role in sleep regulation and in the alterations in sleep that follow prolonged wakefulness.
Collapse
MESH Headings
- Animals
- Arousal/genetics
- Arousal/physiology
- Body Temperature/physiology
- Electroencephalography
- Interleukin-1beta/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred Strains
- Mice, Knockout
- Polyethylene Glycols
- Receptors, Interleukin-1 Type I/deficiency
- Receptors, Interleukin-1 Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Sleep/genetics
- Sleep/physiology
- Sleep Deprivation/genetics
- Sleep Deprivation/physiopathology
- Sleep Stages/genetics
- Sleep Stages/physiology
- Sleep, REM/genetics
- Sleep, REM/physiology
- Tumor Necrosis Factor-alpha/physiology
- Wakefulness/genetics
- Wakefulness/physiology
Collapse
Affiliation(s)
- Francesca Baracchi
- Department of Anesthesiology, University of Michigan, 7422 Medical Sciences Building I, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-5615, USA
| | - Mark R. Opp
- Department of Anesthesiology, University of Michigan, 7422 Medical Sciences Building I, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-5615, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
47
|
Guan Z, Vgontzas AN, Bixler EO, Fang J. Sleep is increased by weight gain and decreased by weight loss in mice. Sleep 2008; 31:627-33. [PMID: 18517033 DOI: 10.1093/sleep/31.5.627] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To determine whether weight loss could reverse excessive sleep in high-fat diet-induced obesity. DESIGN Three groups of mice participated in the study. A weight gain/loss group was fed with high-fat food for 6 weeks (weight gain), and regular food again for 4 weeks (weight loss). A control group and a weight gain only group were fed with regular food and high-fat food, respectively, for 10 weeks after the baseline. PARTICIPANTS Adult male C57BL/6 mice. MEASUREMENTS The amounts of wake, rapid eye movement sleep (REMS) and non-REM sleep (NREMS) were determined at week 0 (baseline), week 6, and week 10. RESULTS The weight gain/loss group displayed a significant decrease in wakefulness and increases in NREMS and episodes of NREMS during 6 weeks of weight gain, which were reversed during subsequent 4 weeks of weight loss. The weight gain only group displayed significant decrease in wakefulness and increase of NREMS and REMS at both week 6 and week 10. The control group did not show significant sleep alterations during the experiment. CONCLUSION These observations indicate that sleep alterations induced by weight gain are reversed by weight loss in obese animals. These data may shed light on the mechanisms underlying the well-established association between obesity and sleepiness in humans and may lead to new therapeutic strategies for these 2 increasingly prevalent problems in the modern societies.
Collapse
Affiliation(s)
- Zhiwei Guan
- Department of Psychiatry, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
48
|
Olivadoti MD, Opp MR. Effects of i.c.v. administration of interleukin-1 on sleep and body temperature of interleukin-6-deficient mice. Neuroscience 2008; 153:338-48. [PMID: 18367337 PMCID: PMC2396575 DOI: 10.1016/j.neuroscience.2008.02.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 02/11/2008] [Accepted: 02/11/2008] [Indexed: 11/24/2022]
Abstract
Cytokines in brain contribute to the regulation of physiological processes and complex behavior, including sleep. The cytokines that have been most extensively studied with respect to sleep are interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha, and IL-6. Administration of these cytokines into laboratory animals, or in some cases into healthy human volunteers, increases the amount of time spent in non-rapid eye movement (NREM) sleep. Although antagonizing the IL-1 or TNF systems reduces the amount of time laboratory animals spend in NREM sleep, interactions among these three cytokine systems as they pertain to the regulation of physiological NREM sleep are not well understood. To further elucidate mechanisms in brain by which IL-1beta, TNFalpha, and/or IL-6 contribute to NREM sleep regulation, we injected recombinant murine interleukin-1beta (muIL-1beta) into C57BL/6J mice and into IL-6-deficient mice (IL-6 knockout, KO). IL-6 KO (B6.129S6-Il6(tm1Kopf); n=13) and C57BL/6J mice (n=14) were implanted with telemeters to record the electroencephalogram (EEG) and core body temperature, as well as with indwelling guide cannulae targeted to one of the lateral ventricles. After recovery and habituation, mice were injected intracerebroventricularly just prior to dark onset on different days with either 0.5 microl vehicle (pyrogen-free saline; PFS) or with 0.5 microl PFS containing one of four doses of muIL-1beta (2.5 ng, 5 ng, 10 ng, 50 ng). No mouse received more than two doses of muIL-1beta, and administration of muIL-1beta doses was counter-balanced to eliminate potential order effects. Sleep-wake behavior was determined for 24 h after injections. i.c.v. administration of muIL-1beta increased in NREM sleep of both mouse strains in a dose-related fashion, but the maximal increase was of greater magnitude in C57Bl/6J mice. muIL-1beta induced fever in C57Bl/6J mice but not in IL-6 KO mice. Collectively, these data demonstrate IL-6 is necessary for IL-1 to induce fever, but IL-6 is not necessary for IL-1 to alter NREM sleep.
Collapse
Affiliation(s)
| | - Mark R. Opp
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
49
|
Schwartz AR, Patil SP, Laffan AM, Polotsky V, Schneider H, Smith PL. Obesity and obstructive sleep apnea: pathogenic mechanisms and therapeutic approaches. PROCEEDINGS OF THE AMERICAN THORACIC SOCIETY 2008; 5:185-92. [PMID: 18250211 PMCID: PMC2645252 DOI: 10.1513/pats.200708-137mg] [Citation(s) in RCA: 431] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Accepted: 09/21/2007] [Indexed: 12/12/2022]
Abstract
Obstructive sleep apnea is a common disorder whose prevalence is linked to an epidemic of obesity in Western society. Sleep apnea is due to recurrent episodes of upper airway obstruction during sleep that are caused by elevations in upper airway collapsibility during sleep. Collapsibility can be increased by underlying anatomic alterations and/or disturbances in upper airway neuromuscular control, both of which play key roles in the pathogenesis of obstructive sleep apnea. Obesity and particularly central adiposity are potent risk factors for sleep apnea. They can increase pharyngeal collapsibility through mechanical effects on pharyngeal soft tissues and lung volume, and through central nervous system-acting signaling proteins (adipokines) that may affect airway neuromuscular control. Specific molecular signaling pathways encode differences in the distribution and metabolic activity of adipose tissue. These differences can produce alterations in the mechanical and neural control of upper airway collapsibility, which determine sleep apnea susceptibility. Although weight loss reduces upper airway collapsibility during sleep, it is not known whether its effects are mediated primarily by improvement in upper airway mechanical properties or neuromuscular control. A variety of behavioral, pharmacologic, and surgical approaches to weight loss may be of benefit to patients with sleep apnea, through distinct effects on the mass and activity of regional adipose stores. Examining responses to specific weight loss strategies will provide critical insight into mechanisms linking obesity and sleep apnea, and will help to elucidate the humoral and molecular predictors of weight loss responses.
Collapse
Affiliation(s)
- Alan R Schwartz
- Johns Hopkins Sleep Disorders Center, Baltimore, MD 21224, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Interleukin-1 beta (IL1) and tumor necrosis factor alpha (TNF) promote non-rapid eye movement sleep under physiological and inflammatory conditions. Additional cytokines are also likely involved but evidence is insufficient to conclude that they are sleep regulatory substances. Many of the symptoms induced by sleep loss, e.g. sleepiness, fatigue, poor cognition, enhanced sensitivity to pain, can be elicited by injection of exogenous IL1 or TNF. We propose that ATP, released during neurotransmission, acting via purine P2 receptors on glia releases IL1 and TNF. This mechanism may provide the means by which the brain keeps track of prior usage history. IL1 and TNF in turn act on neurons to change their intrinsic properties and thereby change input-output properties (i.e. state shift) of the local network involved. Direct evidence indicates that cortical columns oscillate between states, one of which shares properties with organism sleep. We conclude that sleep is a local use-dependent process influenced by cytokines and their effector molecules such as nitric oxide, prostaglandins and adenosine.
Collapse
Affiliation(s)
- James M Krueger
- Sleep and Performance Research Center, College of Veterinary Medicine, Washington State University, Pullman, WA 99164-6520, USA.
| |
Collapse
|