1
|
Kheirouri S, Alizadeh M. The Association Between Diets With High Inflammatory Potential and Sleep Quality and Its Parameters: A Systematic Review. Nutr Rev 2025; 83:e392-e411. [PMID: 38849315 DOI: 10.1093/nutrit/nuae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024] Open
Abstract
CONTEXT Dietary components or its overall properties can influence an individual's sleep status. OBJECTIVE The aim for this study was to critically search, appraise, and synthesize research evidence on the association between dietary inflammatory index (DII) and sleep quality and its parameters. DATA SOURCES Original published studies on adults were obtained from the PubMed, SCOPUS, ScienceDirect, Cochrane Library, and Google Scholar databases. DATA EXTRACTION The search was conducted without date limitation until April 2023. Duplicated and irrelevant investigations were screened out, and the results of the remaining articles were descriptively summarized, then critically appraised and analyzed. Possible mechanistic pathways regarding diet, systemic inflammation, and sleep status were discussed. DATA ANALYSIS Of the 102 studies searched, 23 articles (n = 4 cohort studies, 18 cross-sectional studies, and 1 intervention study) were included in the final review. The association between DII and sleep status was investigated subjectively in 21 studies and objectively in 6 studies. The main studied sleep outcomes were sleep quality, duration, latency, efficiency, apnea, disturbances, the use of sleeping medications, daytime dysfunctions, wakefulness after sleep onset, and rapid eye movement. CONCLUSIONS According to most of the evidence, DII may not be related to overall sleep quality, sleep duration, latency, efficiency, and the use of sleeping medications. The evidence of positive association was greater between a high DII score (pro-inflammatory diet) with daytime dysfunctions, wakefulness after sleep onset, and sleep apnea. There is insufficient evidence to make any conclusion regarding sleep disturbances and rapid eye movement.
Collapse
Affiliation(s)
- Sorayya Kheirouri
- Department of Nutrition, Tabriz University of Medical Sciences, 5166614711 Tabriz, Iran
| | - Mohammad Alizadeh
- Department of Nutrition, Tabriz University of Medical Sciences, 5166614711 Tabriz, Iran
- Nutrition Research Center, Tabriz University of Medical Sciences, 5166614711 Tabriz, Iran
| |
Collapse
|
2
|
Li Y, Que M, Wang X, Zhan G, Zhou Z, Luo X, Li S. Exploring Astrocyte-Mediated Mechanisms in Sleep Disorders and Comorbidity. Biomedicines 2023; 11:2476. [PMID: 37760916 PMCID: PMC10525869 DOI: 10.3390/biomedicines11092476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Astrocytes, the most abundant cells in the brain, are integral to sleep regulation. In the context of a healthy neural environment, these glial cells exert a profound influence on the sleep-wake cycle, modulating both rapid eye movement (REM) and non-REM sleep phases. However, emerging literature underscores perturbations in astrocytic function as potential etiological factors in sleep disorders, either as protopathy or comorbidity. As known, sleep disorders significantly increase the risk of neurodegenerative, cardiovascular, metabolic, or psychiatric diseases. Meanwhile, sleep disorders are commonly screened as comorbidities in various neurodegenerative diseases, epilepsy, and others. Building on existing research that examines the role of astrocytes in sleep disorders, this review aims to elucidate the potential mechanisms by which astrocytes influence sleep regulation and contribute to sleep disorders in the varied settings of brain diseases. The review emphasizes the significance of astrocyte-mediated mechanisms in sleep disorders and their associated comorbidities, highlighting the need for further research.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Mengxin Que
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Xuan Wang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Gaofeng Zhan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Zhiqiang Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyong Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| |
Collapse
|
3
|
Condoleo V, Bonfrate L, Armentaro G, Pelaia C, Cassano V, De Marco M, Severini G, Pastura CA, Miceli S, Maio R, Perticone M, Arturi F, Sesti G, Sciacqua A. Effects of continuous positive airway pressure on comprehensive geriatric assessment and cognitive function in elderly patients with obstructive sleep apnea syndrome. Intern Emerg Med 2023; 18:769-779. [PMID: 36808594 DOI: 10.1007/s11739-023-03220-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023]
Abstract
Obstructive sleep apnea syndrome (OSAS) can lead to cognitive impairment and depression affecting memory, attention, and executive functions. Continuous positive airway pressure (CPAP) treatment seems to be able to revert changes in brain networks and neuropsychological tests correlated to OSAS. The aim of the present study was to evaluate the effects of a 6-month treatment with CPAP on functional, humoral and cognitive parameters in a cohort of elderly OSAS patients with several comorbidities. We enrolled 360 elderly patients suffering from moderate to severe OSAS and indication for nocturnal CPAP. At baseline the Comprehensive Geriatric Assessment (CGA) revealed a borderline Mini-Mental State Examination (MMSE) score that improved after 6-month treatment with CPAP (25.3 ± 1.6 vs 26 ± 1.5; p < 0.0001), as well as the Montreal Cognitive Assessment (MoCA) showed a mild improvement (24.4 ± 2.3 vs 26.2 ± 1.7; p < 0.0001). Moreover, functionality activities increased after treatment, as documented by a short physical performance battery (SPPB) (6.3 ± 1.5 vs 6.9 ± 1.4; p < 0.0001). Reduction of the Geriatric Depression Scale (GDS) from 6.0 ± 2.5 to 4.6 ± 2.2 (p < 0.0001) was also detected. Changes of homeostasis model assessment (HOMA) index, oxygen desaturation index (ODI), sleep-time spent with saturation below 90% (TC90), peripheral arterial oxyhaemoglobin saturation (SpO2), apnea-hypopnea index (AHI) and estimation of glomerular filtration rate (eGFR), contributed, respectively, to 27.9%, 9.0%, 2.8%, 2.3%, 1.7% and 0.9% of MMSE variability for a total of 44.6% of MMSE variations. GDS score changes were due to the improvement of AHI, ODI and TC90, respectively, for 19.2%, 4.9%, 4.2% of the GDS variability, cumulative responsible for 28.3% of GDS modifications. The present real-world study shows that CPAP treatment is able to improve cognition and depressive symptoms in OSAS elderly patients.
Collapse
Affiliation(s)
- Valentino Condoleo
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Leonilde Bonfrate
- Clinica Medica "A. Murri", Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Armentaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy.
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Velia Cassano
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Mario De Marco
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Giandomenico Severini
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Carlo Alberto Pastura
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Sofia Miceli
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Raffaele Maio
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Maria Perticone
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Franco Arturi
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario "S. Venuta", Viale Europa - Località Germaneto, 88100, Catanzaro, Italy
| |
Collapse
|
4
|
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
|
5
|
Sunkaria A, Bhardwaj S. Sleep Disturbance and Alzheimer's Disease: The Glial Connection. Neurochem Res 2022; 47:1799-1815. [PMID: 35303225 DOI: 10.1007/s11064-022-03578-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/09/2022] [Indexed: 12/28/2022]
Abstract
Poor quality and quantity of sleep are very common in elderly people throughout the world. Growing evidence has suggested that sleep disturbances could accelerate the process of neurodegeneration. Recent reports have shown a positive correlation between sleep deprivation and amyloid-β (Aβ)/tau aggregation in the brain of Alzheimer's patients. Glial cells have long been implicated in the progression of Alzheimer's disease (AD) and recent findings have also suggested their role in regulating sleep homeostasis. However, how glial cells control the sleep-wake balance and exactly how disturbed sleep may act as a trigger for Alzheimer's or other neurological disorders have recently gotten attention. In an attempt to connect the dots, the present review has highlighted the role of glia-derived sleep regulatory molecules in AD pathogenesis. Role of glia in sleep disturbance and Alzheimer's progression.
Collapse
Affiliation(s)
- Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
| | - Supriya Bhardwaj
- Department of Dermatology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| |
Collapse
|
6
|
Vrajová M, Šlamberová R, Hoschl C, Ovsepian SV. Methamphetamine and sleep impairments: neurobehavioral correlates and molecular mechanisms. Sleep 2021; 44:6066541. [PMID: 33406259 DOI: 10.1093/sleep/zsab001] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 11/01/2020] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine is a potent and highly addictive psychostimulant, and one of the most widely used illicit drugs. Over recent years, its global usage and seizure have been on a rapid rise, with growing detrimental effects on mental and physical health, and devastating psychosocial impact pressing for intervention. Among the unwanted effects of methamphetamine, acute and long-term sleep impairments are of major concern, posing a significant therapeutic challenge, and a cause of addiction relapse. Unraveling mechanisms and functional correlates of methamphetamine-related sleep and circadian disruption are, therefore, of key relevance to translational and clinical psychiatry. In this article, we review the mounting evidence for the acute and long-term impairements of sleep-wake behavior and circadian activity caused by single or recurring methamphetamine usage and withdrawal. Factors contributing to the severity of sleep loss and related cognitive deficit, with risks of relapse are discussed. Key molecular players mediating methamphetamine-induced dopamine release and neuromodulation are considered, with wake-promoting effects in mesolimbic circuits. The effects on various sleep phases and related changes in dopamine levels in selected subcortical structures are reviewed and compared to other psychostimulants with similar action mechanisms. A critical appraisal is presented of the therapeutic use of modafinil, countering sleep, and circadian rhythm impairments. Finally, emerging knowledge gaps and methodical limitations are highlighted along with the areas for future research and therapeutic translation.
Collapse
Affiliation(s)
- Monika Vrajová
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Cyril Hoschl
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Klecany, Czech Republic
| | - Saak V Ovsepian
- Department of Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Klecany, Czech Republic
| |
Collapse
|
7
|
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
|
8
|
Ji T, Li X, Qiu Y, Mei L, Jia X, Tai J, Guo Y, Zhang J, Wang S, Ni X. Disease characteristics and neuropathological changes associated with cognitive dysfunction in obstructive sleep apnea. Pediatr Investig 2021; 5:52-57. [PMID: 33778428 PMCID: PMC7984002 DOI: 10.1002/ped4.12247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Obstructive sleep apnea (OSA) is a common sleep-disordered breathing disease that often leads to many comorbidities (e.g., cognitive dysfunction), which adversely affect the quality of life for patients with OSA. Thus far, the underlying mechanisms of this dysfunction remain unclear. Many studies have focused on OSA-related characteristics, including intermittent hypoxemia and sleep fragmentation. There is increasing emphasis on neuroimaging studies to explore underlying relationships between neuropathological changes and cognitive dysfunction. This article reviews recent research progress concerning cognitive dysfunction associated with OSA to reveal potential mechanisms that contribute to this dysfunction.
Collapse
Affiliation(s)
- Tingting Ji
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xiaodan Li
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Yue Qiu
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Lin Mei
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xinbei Jia
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Jun Tai
- Department of OtorhinolaryngologyBeijing Children’s HospitalCapital Institute of PediatricsBeijingChina
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of OtolaryngologyHead and Neck SurgeryBeijing Pediatric Research InstituteBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Shengcai Wang
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| | - Xin Ni
- Department of Otolaryngology, Head and Neck SurgeryBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
- Beijing Key Laboratory for Pediatric Diseases of OtolaryngologyHead and Neck SurgeryBeijing Pediatric Research InstituteBeijing Children’s HospitalCapital Medical UniversityNational Center for Children’s HealthBeijingChina
| |
Collapse
|
9
|
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
|
10
|
Nguyen MH, Bryant K, O'Neill SG. Vitamin D in SLE: a role in pathogenesis and fatigue? A review of the literature. Lupus 2018; 27:2003-2011. [DOI: 10.1177/0961203318796293] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fatigue is a common, disabling problem that is highly prevalent in patients with systemic lupus erythematous (SLE). More recently, vitamin D status has been established as a potential contributor to SLE pathogenesis and manifestations, in particular fatigue. This review summarizes the literature regarding the role of vitamin D in SLE, and provides an overview of the recent literature examining the association between vitamin D and fatigue in patients with SLE. Finally, the role of vitamin D supplementation in the treatment of SLE-related fatigue is considered.
Collapse
Affiliation(s)
- MH Nguyen
- Rheumatology Department, Liverpool Hospital, Australia
- SWS Clinical School, University of New South Wales, and the Ingham Institute for Applied Medical Research, Australia
| | - K Bryant
- SWS Clinical School, University of New South Wales, and the Ingham Institute for Applied Medical Research, Australia
| | - SG O'Neill
- Rheumatology Department, Liverpool Hospital, Australia
- SWS Clinical School, University of New South Wales, and the Ingham Institute for Applied Medical Research, Australia
| |
Collapse
|
11
|
Brenhouse HC, Schwarz JM. Immunoadolescence: Neuroimmune development and adolescent behavior. Neurosci Biobehav Rev 2016; 70:288-299. [PMID: 27260127 PMCID: PMC5412135 DOI: 10.1016/j.neubiorev.2016.05.035] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022]
Abstract
The brain is increasingly appreciated to be a constantly rewired organ that yields age-specific behaviors and responses to the environment. Adolescence in particular is a unique period characterized by continued brain maturation, superimposed with transient needs of the organism to traverse a leap from parental dependence to independence. Here we describe how these needs require immune maturation, as well as brain maturation. Our immune system, which protects us from pathogens and regulates inflammation, is in constant communication with our nervous system. Together, neuro-immune signaling regulates our behavioral responses to the environment, making this interaction a likely substrate for adolescent development. We review here the identified as well as understudied components of neuro-immune interactions during adolescence. Synaptic pruning, neurite outgrowth, and neurotransmitter release during adolescence all regulate-and are regulated by-immune signals, which occur via blood-brain barrier dynamics and glial activity. We discuss these processes, as well as how immune signaling during this transitional period of development confers differential effects on behavior and vulnerability to mental illness.
Collapse
Affiliation(s)
- Heather C Brenhouse
- Northeastern University, Psychology Department, 125 Nightingale Hall, Boston, MA 02115, United States.
| | - Jaclyn M Schwarz
- University of Delaware, Department of Psychological and Brain Sciences, 108 Wolf Hall, Newark, DE 19716, United States
| |
Collapse
|
12
|
Abstract
Sleep is a complex physiological process that is regulated globally, regionally, and locally by both cellular and molecular mechanisms. It occurs to some extent in all animals, although sleep expression in lower animals may be co-extensive with rest. Sleep regulation plays an intrinsic part in many behavioral and physiological functions. Currently, all researchers agree there is no single physiological role sleep serves. Nevertheless, it is quite evident that sleep is essential for many vital functions including development, energy conservation, brain waste clearance, modulation of immune responses, cognition, performance, vigilance, disease, and psychological state. This review details the physiological processes involved in sleep regulation and the possible functions that sleep may serve. This description of the brain circuitry, cell types, and molecules involved in sleep regulation is intended to further the reader's understanding of the functions of sleep.
Collapse
Affiliation(s)
- Mark R. Zielinski
- Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA and Harvard Medical School, Department of Psychiatry
| | - James T. McKenna
- Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA and Harvard Medical School, Department of Psychiatry
| | - Robert W. McCarley
- Veterans Affairs Boston Healthcare System, Brockton, MA 02301, USA and Harvard Medical School, Department of Psychiatry
| |
Collapse
|
13
|
Kuo TH, Williams JA. Acute sleep deprivation enhances post-infection sleep and promotes survival during bacterial infection in Drosophila. Sleep 2014; 37:859-69. [PMID: 24790264 DOI: 10.5665/sleep.3648] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
STUDY OBJECTIVES Sleep is known to increase as an acute response to infection. However, the function of this behavioral response in host defense is not well understood. To address this problem, we evaluated the effect of acute sleep deprivation on post-infection sleep and immune function in Drosophila. SETTING Laboratory. PARTICIPANTS Drosophila melanogaster. METHODS AND RESULTS Flies were subjected to sleep deprivation before (early DEP) or after (late DEP) bacterial infection. Relative to a non-deprived control, flies subjected to early DEP had enhanced sleep after infection as well as increased bacterial clearance and survival outcome. Flies subjected to late DEP experienced enhanced sleep following the deprivation period, and showed a modest improvement in survival outcome. Continuous DEP (early and late DEP) throughout infection also enhanced sleep later during infection and improved survival. However, improved survival in flies subjected to late or continuous DEP did not occur until after flies had experienced sleep. During infection, both early and late DEP enhanced NFκB transcriptional activity as measured by a luciferase reporter (κB-luc) in living flies. Early DEP also increased NFκB activity prior to infection. Flies that were deficient in expression of either the Relish or Dif NFκB transcription factors showed normal responses to early DEP. However, the effect of early DEP on post-infection sleep and survival was abolished in double mutants, which indicates that Relish and Dif have redundant roles in this process. CONCLUSIONS Acute sleep deprivation elevated NFκB-dependent activity, increased post-infection sleep, and improved survival during bacterial infection.
Collapse
Affiliation(s)
- Tzu-Hsing Kuo
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Julie A Williams
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
14
|
Szentirmai É, Krueger JM. Sickness behaviour after lipopolysaccharide treatment in ghrelin deficient mice. Brain Behav Immun 2014; 36:200-6. [PMID: 24309634 PMCID: PMC3951816 DOI: 10.1016/j.bbi.2013.11.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 11/04/2013] [Accepted: 11/26/2013] [Indexed: 12/14/2022] Open
Abstract
Ghrelin is an orexigenic hormone produced mainly by the gastrointestinal system and the brain. Much evidence also indicates a role for ghrelin in sleep and thermoregulation. Further, ghrelin was recently implicated in immune system modulation. Administration of bacterial lipopolysaccharide (LPS) induces fever, anorexia, and increased non-rapid-eye movement sleep (NREMS) and these actions are mediated primarily by proinflammatory cytokines. Ghrelin reduces LPS-induced fever, suppresses circulating levels of proinflammatory cytokines and reduces the severity and mortality of various models of experimental endotoxemia. In the present study, we determined the role of intact ghrelin signaling in LPS-induced sleep, feeding, and thermoregulatory responses in mice. Sleep-wake activity was determined after intraperitoneal, dark onset administration of 0.4, 2 and 10 μg LPS in preproghrelin knockout (KO) and wild-type (WT) mice. In addition, body temperature, motor activity and changes in 24-h food intake and body weight were measured. LPS induced dose-dependent increases in NREMS, and suppressed rapid-eye movement sleep, electroencephalographic slow-wave activity, motor activity, food intake and body weight in both Ppg KO and WT mice. Body temperature changes showed a biphasic pattern with a decrease during the dark period followed by an increase in the light phase. The effects of the low and middle doses of LPS were indistinguishable between the two genotypes. Administration of 10 μg LPS, however, induced significantly larger changes in NREMS and wakefulness amounts, body temperature, food intake and body weight in the Ppg KO mice. These findings support a role for ghrelin as an endogenous modulator of inflammatory responses and a central component of arousal and feeding circuits.
Collapse
Affiliation(s)
- Éva Szentirmai
- Washington, Wyoming, Alaska, Montana and Idaho (WWAMI) Medical Education Program, Washington State University, Spokane, WA, USA; Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, USA.
| | - James M. Krueger
- Washington, Wyoming, Alaska, Montana and Idaho (WWAMI)
Medical Education Program, Washington State University, Spokane, WA, USA,Department of Integrative Physiology and Neuroscience,
Washington State University, Spokane, WA, USA,Sleep and Performance Research Center, Washington State
University, Spokane, WA, USA
| |
Collapse
|
15
|
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: 60] [Impact Index Per Article: 5.0] [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
|
16
|
Krueger JM, Huang YH, Rector DM, Buysse DJ. Sleep: a synchrony of cell activity-driven small network states. Eur J Neurosci 2013; 38:2199-209. [PMID: 23651209 DOI: 10.1111/ejn.12238] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/27/2013] [Accepted: 03/29/2013] [Indexed: 12/14/2022]
Abstract
We posit a bottom-up sleep-regulatory paradigm in which state changes are initiated within small networks as a consequence of local cell activity. Bottom-up regulatory mechanisms are prevalent throughout nature, occurring in vastly different systems and levels of organization. Synchronization of state without top-down regulation is a fundamental property of large collections of small semi-autonomous entities. We posit that such synchronization mechanisms are sufficient and necessary for whole-organism sleep onset. Within the brain we posit that small networks of highly interconnected neurons and glia, for example cortical columns, are semi-autonomous units oscillating between sleep-like and wake-like states. We review evidence showing that cells, small networks and regional areas of the brain share sleep-like properties with whole-animal sleep. A testable hypothesis focused on how sleep is initiated within local networks is presented. We posit that the release of cell activity-dependent molecules, such as ATP and nitric oxide, into the extracellular space initiates state changes within the local networks where they are produced. We review mechanisms of ATP induction of sleep-regulatory substances and their actions on receptor trafficking. Finally, we provide an example of how such local metabolic and state changes provide mechanistic explanations for clinical conditions, such as insomnia.
Collapse
Affiliation(s)
- James M Krueger
- Sleep and Performance Research Center, Washington State University, Pullman, WA, USA.
| | | | | | | |
Collapse
|
17
|
Williamson LL, Bilbo SD. Chemokines and the hippocampus: a new perspective on hippocampal plasticity and vulnerability. Brain Behav Immun 2013; 30:186-94. [PMID: 23376170 DOI: 10.1016/j.bbi.2013.01.077] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/07/2013] [Accepted: 01/15/2013] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is critical for several aspects of learning and memory and is unique among other cortical regions in structure, function and the potential for plasticity. This remarkable region recapitulates development throughout the lifespan with enduring neurogenesis and well-characterized plasticity. The structure and traits of the hippocampus that distinguish it from other brain regions, however, may be the same reasons that this important brain region is particularly vulnerable to insult and injury. The immune system within the brain responds to insult and injury, and the hippocampus and the immune system are extensively interconnected. Immune signaling molecules, cytokines and chemokines (chemotactic cytokines), are well known for their functions during insult or injury. They are also increasingly implicated in normal hippocampal neurogenesis (e.g., CXCR4 on newborn neurons), cellular plasticity (e.g., interleukin-6 in LTP maintenance), and learning and memory (e.g., interleukin-1β in fear conditioning). We provide evidence from the small but growing literature that neuroimmune interactions and immune signaling molecules, especially chemokines, may be a primary underlying mechanism for the coexistence of plasticity and vulnerability within the hippocampus. We also highlight the evidence that the hippocampus exhibits a remarkable resilience in response to diverse environmental events (e.g., enrichment, exercise), which all may converge onto common neuroimmune mechanisms.
Collapse
Affiliation(s)
- Lauren L Williamson
- Duke University, Genome Science Research Building 2, 210 Research Dr., Box 91050, Durham, NC 27710, United States.
| | | |
Collapse
|
18
|
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
|
19
|
Bilbo SD, Schwarz JM. The immune system and developmental programming of brain and behavior. Front Neuroendocrinol 2012; 33:267-86. [PMID: 22982535 PMCID: PMC3484177 DOI: 10.1016/j.yfrne.2012.08.006] [Citation(s) in RCA: 413] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/28/2012] [Accepted: 08/29/2012] [Indexed: 12/16/2022]
Abstract
The brain, endocrine, and immune systems are inextricably linked. Immune molecules have a powerful impact on neuroendocrine function, including hormone-behavior interactions, during health as well as sickness. Similarly, alterations in hormones, such as during stress, can powerfully impact immune function or reactivity. These functional shifts are evolved, adaptive responses that organize changes in behavior and mobilize immune resources, but can also lead to pathology or exacerbate disease if prolonged or exaggerated. The developing brain in particular is exquisitely sensitive to both endogenous and exogenous signals, and increasing evidence suggests the immune system has a critical role in brain development and associated behavioral outcomes for the life of the individual. Indeed, there are associations between many neuropsychiatric disorders and immune dysfunction, with a distinct etiology in neurodevelopment. The goal of this review is to describe the important role of the immune system during brain development, and to discuss some of the many ways in which immune activation during early brain development can affect the later-life outcomes of neural function, immune function, mood and cognition.
Collapse
Affiliation(s)
- Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, 572 Research Drive, Box 91050, Durham, NC 27708, USA.
| | | |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Lu CY, Yi PL, Tsai CH, Cheng CH, Chang HH, Hsiao YT, Chang FC. TNF-NF-kappaB signaling mediates excessive somnolence in hemiparkinsonian rats. Behav Brain Res 2010; 208:484-96. [PMID: 20043954 DOI: 10.1016/j.bbr.2009.12.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 12/11/2009] [Accepted: 12/18/2009] [Indexed: 01/24/2023]
Abstract
Daytime somnolence is common in patients with Parkinson's disease (PD); however there is a lack of understanding of the cellular mechanisms involved in mediating these effects. It has been hypothesized that microglial activation and the subsequent increase of pro-inflammatory cytokines play an important role in the pathogenesis of PD. Because some cytokines are involved in the regulation of sleep, this study was designed to determine if tumor necrosis factor (TNF) and interleukin-1beta (IL-1beta), mediate daytime somnolence in the proteasome inhibitor (MG-132)-induced hemiparkinsonian rat model. Our results indicated that microglial activation caused the loss of dopaminergic neurons in the substantia nigra, and the expression of TNF-alpha, but not IL-1beta, increased in the midbrain and hypothalamus in MG-132-induced hemiparkinsonian rats. Slow-wave sleep (SWS) increased after the induction of hemiparkinsonism, but rapid eye movement (REM) sleep was not consistently altered. Application of the TNF receptor fragment (TNFRF) blocked hemiparkinsonism-induced SWS alteration, whereas the IL-1 receptor antagonist (IL-1ra) exhibited no effect. Increased nuclear translocation of NF-kappaB in the midbrain, and the blockade of SWS enhancement in MG-132-induced hemiparkinsonian rats by an inhibitor of NF-kappaB activation indicate that the TNF-NF-kappaB cascade is a critical mediator of MG-132 hemiparkinsonian-induced sleep alteration. This observation suggests potential therapeutic interventions to target the excessive daytime somnolence in patients with PD.
Collapse
Affiliation(s)
- Chin-Yu Lu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The transcription factor NF-kappaB has diverse functions in the nervous system, depending on the cellular context. NF-kappaB is constitutively activated in glutamatergic neurons. Knockout of p65 or inhibition of neuronal NF-kappaB by super-repressor IkappaB resulted in the loss of neuroprotection and defects in learning and memory. Similarly, p50-/- mice have a lower learning ability and are sensitive to neurotoxins. Activated NF-kappaB can be transported retrogradely from activated synapses to the nucleus to translate short-term processes to long-term changes such as axon growth, which is important for long-term memory. In glia, NF-kappaB is inducible and regulates inflammatory processes that exacerbate diseases such as autoimmune encephalomyelitis, ischemia, and Alzheimer's disease. In summary, inhibition of NF-kappaB in glia might ameliorate disease, whereas activation in neurons might enhance memory. This review focuses on results produced by the analysis of genetic models.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, Universitätsstr. 25, D-33501 Bielefeld.
| | | |
Collapse
|
24
|
Abstract
The psychologic and behavioral components of sickness represent, together with fever response and associated neuroendocrine changes, a highly organized strategy of the organism to fight infection. This strategy, referred to as sickness behavior, is triggered by the proinflammatory cytokines produced by activated cells of the innate immune system in contact with specific pathogen-associated molecular patterns (PAMPs). Interleukin-1 and other cytokines act on the brain via (1) a neural route represented by the primary afferent neurons that innervate the body site where the infectious process takes place and (2) a humoral pathway that involves the production of proinflammatory cytokines. This article presents the current knowledge on the way this communication system is organized and regulated and the implications of these advances for understanding brain physiology and pathology.
Collapse
Affiliation(s)
- Robert Dantzer
- Integrative Immunology and Behavior Program, University of Illinois at Urbana-Champaign, 212 ERML, 1201 W Gregory Drive, Urbana, IL 61801, USA.
| |
Collapse
|
25
|
The influences of cytokines as a possible substrate for the psychological effects of immunomodulation therapy in multiple sclerosis. ACTA ACUST UNITED AC 2008; 39:25-30. [DOI: 10.1007/s11055-008-9087-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
26
|
Methippara MM, Bashir T, Kumar S, Alam N, Szymusiak R, McGinty D. Salubrinal, an inhibitor of protein synthesis, promotes deep slow wave sleep. Am J Physiol Regul Integr Comp Physiol 2008; 296:R178-84. [PMID: 18971348 DOI: 10.1152/ajpregu.90765.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Previous work showed that sleep is associated with increased brain protein synthesis and that arrest of protein synthesis facilitates sleep. Arrest of protein synthesis is induced during the endoplasmic reticulum (ER) stress response, through phosphorylation of eukaryotic initiation factor 2alpha (p-eIF2alpha). We tested a hypothesis that elevation of p-eIF2alpha would facilitate sleep. We studied the effects of intracerebroventricular infusion of salubrinal (Salub), which increases p-eIF2alpha by inhibiting its dephosphorylation. Salub increased deep slow wave sleep by 255%, while reducing active waking by 49%. Delta power within non-rapid eye movement (NREM) sleep was increased, while power in the sigma, beta, and gamma bands during NREM was reduced. We found that Salub increased expression of p-eIF2alpha in the basal forebrain (BF) area, a sleep-wake regulatory brain region. Therefore, we quantified the p-eIF2alpha-immunolabeled neurons in the BF area; Salub administration increased the number of p-eIF2alpha-expressing noncholinergic neurons in the caudal BF. In addition, Salub also increased the intensity of p-eIF2alpha expression in both cholinergic and noncholinergic neurons, but this was more widespread among the noncholinergic neurons. Our findings support a hypothesis that sleep is facilitated by signals associated with the ER stress response.
Collapse
Affiliation(s)
- Melvi M Methippara
- Research Service (151A3), Department of Veterans Affairs of Greater Los Angeles Healthcare System, North Hills, CA 91343, USA
| | | | | | | | | | | |
Collapse
|
27
|
Khatami R, von Büdingen HC, Bassetti CL. Sleep–Wake Disturbances in Neurologic Autoimmune Disorders. Sleep Med Clin 2008. [DOI: 10.1016/j.jsmc.2008.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
28
|
Taishi P, Churchill L, De A, Obal F, Krueger JM. Cytokine mRNA induction by interleukin-1beta or tumor necrosis factor alpha in vitro and in vivo. Brain Res 2008; 1226:89-98. [PMID: 18620339 PMCID: PMC2642478 DOI: 10.1016/j.brainres.2008.05.067] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 05/09/2008] [Accepted: 05/27/2008] [Indexed: 10/22/2022]
Abstract
Hypothalamic and cortical mRNA levels for cytokines such as interleukin-1beta (IL1beta), tumor necrosis factor alpha (TNFalpha), nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are impacted by systemic treatments of IL1beta and TNFalpha. To investigate the time course of the effects of IL1beta and TNFalpha on hypothalamic and cortical cytokine gene expression, we measured mRNA levels for IL1beta, TNFalpha, interleukin-6 (IL-6), interleukin-10 (IL-10), IL1 receptor 1, BDNF, NGF, and glutamate decarboxylase-67 in vitro using hypothalamic and cortical primary cultures. IL1beta and TNFalpha mRNA levels increased significantly in a dose-dependent fashion after exposure to either IL1beta or TNFalpha. IL1beta increased IL1beta mRNA in both the hypothalamic and cortical cultures after 2-6 h while TNFalpha mRNA increased significantly within 30 min and continued to rise up to 2-6 h. Most of the other mRNAs showed significant changes independent of dose in vitro. In vivo, intracerebroventricular (icv) injection of IL1beta or TNFalpha also significantly increased IL1beta, TNFalpha and IL6 mRNA levels in the hypothalamus and cortex. IL1beta icv, but not TNFalpha, increased NGF mRNA levels in both these areas. Results support the hypothesis that centrally active doses of IL1beta and TNFalpha enhance their own mRNA levels as well as affect mRNA levels for other neuronal growth factors.
Collapse
Affiliation(s)
- Ping Taishi
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Programs in Neuroscience, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | | | |
Collapse
|
29
|
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
|
30
|
Puigjaner J, Fábrega J, de Diego I, Subirada F, Durany O, Rivero-Urgell M. Two circadian infant formulas produce differential cerebellum gene expression in lactating rat neonates. GENES & NUTRITION 2007; 2:129-131. [PMID: 18850161 PMCID: PMC2474909 DOI: 10.1007/s12263-007-0037-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Affiliation(s)
- J Puigjaner
- Research and Development Department, Ordesa Group, Sant Boi de Llobregat, Barcelona, Spain,
| | | | | | | | | | | |
Collapse
|
31
|
Lubin FD, Sweatt JD. The IkappaB kinase regulates chromatin structure during reconsolidation of conditioned fear memories. Neuron 2007; 55:942-57. [PMID: 17880897 PMCID: PMC2587178 DOI: 10.1016/j.neuron.2007.07.039] [Citation(s) in RCA: 189] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 05/14/2007] [Accepted: 07/31/2007] [Indexed: 11/22/2022]
Abstract
Previously formed memories are susceptible to disruption immediately after recall due to a necessity to be reconsolidated after retrieval. Protein translation mechanisms have been widely implicated as being necessary for memory reconsolidation, but gene transcription mechanisms have been much less extensively studied in this context. We found that retrieval of contextual conditioned fear memories activates the NF-kappaB pathway to regulate histone H3 phosphorylation and acetylation at specific gene promoters in hippocampus, specifically via IKKalpha and not the NF-kappaB DNA-binding complex. Behaviorally, we found that inhibition of IKKalpha regulation of either chromatin structure or NF-kappaB DNA-binding complex activity leads to impairments in fear memory reconsolidation, and that elevating histone acetylation rescues this memory deficit in the face of IKK blockade. These data provide insights into IKK-regulated transcriptional mechanisms in hippocampus that are necessary for memory reconsolidation.
Collapse
Affiliation(s)
- Farah D Lubin
- The Evelyn F. McKnight Brain Institute, Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
32
|
Lubin FD, Ren Y, Xu X, Anderson AE. Nuclear factor-kappa B regulates seizure threshold and gene transcription following convulsant stimulation. J Neurochem 2007; 103:1381-95. [PMID: 17727632 DOI: 10.1111/j.1471-4159.2007.04863.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We evaluated a role for the nuclear factor-kappa B (NF-kappaB) pathway in the regulation of seizure susceptibility and transcriptional activation during prolonged, continuous seizures (status epilepticus). Using two functionally distinct NF-kappaB inhibitors we observed a decrease in latency to onset of kainate-induced seizures and status epilepticus. To assess NF-kappaB transcriptional activation, we evaluated inhibitor kappa B alpha (IkappaBalpha) and brain-derived neurotrophic factor (bdnf) gene targets. Inhibition of the NF-kappaB signaling pathway significantly attenuated the increases in IkappaBalpha and bdnf mRNA levels that occurred during prolonged seizure activity, suggesting that the NF-kappaB pathway was involved in the up-regulation of these transcripts during status epilepticus. DNA-binding studies and chromatin immunoprecipitation assays using hippocampal extracts from animals with status epilepticus revealed that NF-kappaB subunits were associated with the candidate kappaB-binding elements within promoter 1 of the bdnf gene. The pattern of association was different for the p50 and p65 subunits supporting complex NF-kappaB modifications within promoter 1. In summary, our findings provide additional insights into the role of NF-kappaB transcriptional regulation in hippocampus following status epilepticus and suggest that NF-kappaB pathway activation contributes to seizure susceptibility.
Collapse
Affiliation(s)
- Farah D Lubin
- Cain Foundation Laboratories, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA, and Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | |
Collapse
|
33
|
Cambras T, Weller JR, Anglès-Pujoràs M, Lee ML, Christopher A, Díez-Noguera A, Krueger JM, de la Iglesia HO. Circadian desynchronization of core body temperature and sleep stages in the rat. Proc Natl Acad Sci U S A 2007; 104:7634-9. [PMID: 17452631 PMCID: PMC1863469 DOI: 10.1073/pnas.0702424104] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Proper functioning of the human circadian timing system is crucial to physical and mental health. Much of what we know about this system is based on experimental protocols that induce the desynchronization of behavioral and physiological rhythms within individual subjects, but the neural (or extraneural) substrates for such desynchronization are unknown. We have developed an animal model of human internal desynchrony in which rats are exposed to artificially short (22-h) light-dark cycles. Under these conditions, locomotor activity, sleep-wake, and slow-wave sleep (SWS) exhibit two rhythms within individual animals, one entrained to the 22-h light-dark cycle and the other free-running with a period >24 h (tau(>24 h)). Whereas core body temperature showed two rhythms as well, further analysis indicates this variable oscillates more according to the tau(>24 h) rhythm than to the 22-h rhythm, and that this oscillation is due to an activity-independent circadian regulation. Paradoxical sleep (PS), on the other hand, shows only one free-running rhythm. Our results show that, similarly to humans, (i) circadian rhythms can be internally dissociated in a controlled and predictable manner in the rat and (ii) the circadian rhythms of sleep-wake and SWS can be desynchronized from the rhythms of PS and core body temperature within individual animals. This model now allows for a deeper understanding of the human timekeeping mechanism, for testing potential therapies for circadian dysrhythmias, and for studying the biology of PS and SWS states in a neurologically intact model.
Collapse
Affiliation(s)
- Trinitat Cambras
- *Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona 08028, Spain
- To whom correspondence may be addressed. E-mail: or
| | - John R. Weller
- Department of Biology and Program of Neurobiology and Behavior, Box 351800, University of Washington, Seattle, WA 98195-1800; and
| | | | - Michael L. Lee
- Department of Biology and Program of Neurobiology and Behavior, Box 351800, University of Washington, Seattle, WA 98195-1800; and
| | - Andrea Christopher
- Department of Biology and Program of Neurobiology and Behavior, Box 351800, University of Washington, Seattle, WA 98195-1800; and
| | - Antoni Díez-Noguera
- *Departament de Fisiologia, Facultat de Farmàcia, Universitat de Barcelona, Barcelona 08028, Spain
| | - James M. Krueger
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, Washington State University, Pullman, WA 99164-6520
| | - Horacio O. de la Iglesia
- Department of Biology and Program of Neurobiology and Behavior, Box 351800, University of Washington, Seattle, WA 98195-1800; and
- To whom correspondence may be addressed. E-mail: or
| |
Collapse
|
34
|
Ramesh V, Thatte HS, McCarley RW, Basheer R. Adenosine and sleep deprivation promote NF-kappaB nuclear translocation in cholinergic basal forebrain. J Neurochem 2007; 100:1351-63. [PMID: 17316404 DOI: 10.1111/j.1471-4159.2006.04314.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
In our investigations related to the homeostatic sleep factor adenosine (AD), we previously demonstrated that the DNA-binding activity of the transcription factor NF-kappaB in rat cholinergic basal forebrain increased following 3 h of sleep deprivation (SD). However, the neurotransmitter nature of the cells and the SD-induced stimuli responsible for NF-kappaB activation were not defined. In this report, we demonstrate, using double labeling immunohistochemistry, that nuclear translocation of NF-kappaB occurs almost exclusively in the cholinergic neurons of the basal forebrain following 3 h of SD. Furthermore, cholinergic basal forebrain microinjection of AD (25 nmol/L) or the A(1) receptor agonist N(6)-cyclo-hexyladenosine (100 nmol/L) induced nuclear translocation of NF-kappaB, thus suggesting that SD-induced increased extracellular concentrations of AD, acting via the A(1) AD receptor, may be responsible for the nuclear translocation of NF-kappaB in cholinergic neurons. Moreover, blocking the nuclear translocation of NF-kappaB by injection of inhibitor peptide, SN50, immediately prior to 6 h SD significantly reduced delta activity (1-4 Hz) during the first two hours of recovery sleep. Together, these data suggest a role in sleep homeostasis for the SD-induced activation of NF-kappaB in cholinergic basal forebrain, and that transcription factor NF-kappaB may code for factor(s) that play a role in sleep homeostasis.
Collapse
Affiliation(s)
- Vijay Ramesh
- Department of Psychiatry, VA Boston Healthcare System and Harvard Medical School, West Roxbury, Massachusetts, USA
| | | | | | | |
Collapse
|
35
|
Williams JA, Sathyanarayanan S, Hendricks JC, Sehgal A. Interaction between sleep and the immune response in Drosophila: a role for the NFkappaB relish. Sleep 2007; 30:389-400. [PMID: 17520783 PMCID: PMC2884379 DOI: 10.1093/sleep/30.4.389] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
STUDY OBJECTIVES The regulation of sleep is poorly understood. While some molecules, including those involved in inflammatory/immune responses, have been implicated in the control of sleep, their role in this process remains unclear. The Drosophila model for sleep provides a powerful system to identify and test the role of sleep-relevant molecules. DESIGN We conducted an unbiased screen for molecular candidates involved in sleep regulation by analyzing genome-wide changes in gene expression associated with sleep deprivation in Drosophila. To further examine a role of immune-related genes identified in the screen, we performed molecular assays, analysis of sleep behavior in relevant mutant and transgenic flies, and quantitative analysis of the immune response following sleep deprivation. RESULTS A major class of genes that increased expression with sleep deprivation was that involved in the immune response. We found that immune genes were also upregulated during baseline conditions in the cyc01 sleep mutant. Since the expression of an NFkappaB, Relish, a central player in the inflammatory response, was increased with all manipulations that reduced sleep, we focused on this gene. Flies deficient in, but not lacking, Relish expression exhibited reduced levels of nighttime sleep, supporting a role for Relish in the control of sleep. This mutant phenotype was rescued by expression of a Relish transgene in fat bodies, which are the major site of inflammatory responses in Drosophila. Finally, sleep deprivation also affected the immune response, such that flies deprived of sleep for several hours were more resistant to bacterial infection than those flies not deprived of sleep. CONCLUSION These results demonstrate a conserved interaction between sleep and the immune system. Genetic manipulation of an immune component alters sleep, and likewise, acute sleep deprivation alters the immune response.
Collapse
Affiliation(s)
- Julie A Williams
- Center for Advanced Biotechnology and Medicine and Department of Pharmacology, University of Medicine and Dentistry of New Jersey, Piscataway 08854-5603, USA.
| | | | | | | |
Collapse
|
36
|
Affiliation(s)
- James M Krueger
- Program in Neuroscience, Washington State University, Pullman, WA 99164
| | | | | |
Collapse
|
37
|
Abstract
Sufficient evidence is now available to accept the concept that the brain recognizes cytokines as molecular signals of sickness. Clarifying the way the brain processes information generated by the innate immune system is accompanied by a progressive elucidation of the cellular and molecular components of the intricate system that mediates cytokine-induced sickness behavior. We are still far, however, from understanding the whole. Among the hundreds of genes that proinflammatory cytokines can induce in their cellular targets, only a handful has been examined functionally. In addition, a dynamic view of the cellular interactions that occur at the brain sites of cytokine production and action is missing, together with a clarification of the mechanisms that favor the transition toward pathology.
Collapse
Affiliation(s)
- Robert Dantzer
- Laboratory of Integrative Neurobiology, CNRS, INRA, University of Bordeaux 2, 33077 Bordeaux Cedex, France.
| |
Collapse
|
38
|
Jhaveri KA, Ramkumar V, Trammell RA, Toth LA. Spontaneous, homeostatic, and inflammation-induced sleep in NF-kappaB p50 knockout mice. Am J Physiol Regul Integr Comp Physiol 2006; 291:R1516-26. [PMID: 16793936 DOI: 10.1152/ajpregu.00262.2006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The dimeric transcription factor nuclear factor-kappaB (NF-kappaB) regulates several endogenous sleep-modulatory substances and thereby serves as a pivotal mediator of sleep-wake homeostasis. To further define the role of NF-kappaB in sleep regulation, we monitored sleep and temperature in mice that lack the p50 subunit of NF-kappaB [p50 knockout (KO) mice]. Compared with the control B6129PF2/J strain, p50 KO mice spend more time in slow-wave sleep (SWS) and rapid eye movement sleep (REMS) under normal conditions and show enhanced homeostatic recovery of sleep after sleep loss. p50 KO mice also show increased SWS and reduced REMS and temperature after the administration of lipopolysaccharide, yet they are behaviorally less responsive to challenge with influenza virus. These data support a role for NF-kappaB, and, in particular, for the p50 subunit, in the regulation of sleep in healthy mice and in mice experiencing immune challenge.
Collapse
Affiliation(s)
- K A Jhaveri
- Dept. of Pharmacology, SIU School of Medicine, P.O. Box 19629, Springfield, IL 62794-9629, USA
| | | | | | | |
Collapse
|
39
|
Kushikata T, Fang J, Krueger JM. Platelet activating factor and its metabolite promote sleep in rabbits. Neurosci Lett 2006; 394:233-8. [PMID: 16263215 DOI: 10.1016/j.neulet.2005.10.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2005] [Revised: 10/07/2005] [Accepted: 10/12/2005] [Indexed: 11/21/2022]
Abstract
Platelet activating factor (PAF) is a key inflammatory mediator. PAF and its receptor are found in brain and PAF affects or is affected by the production of sleep promoting cytokines such as interleukin-1. PAF also interacts with several other sleep-regulatory substances such as nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, nitric oxide, prostaglandins, and prolactin. We thus hypothesized that PAF would increase sleep. In these experiments, each rabbit received an injection of 25 microl of 2% DMSO to obtain control values, and on a separate day received PAF or lyso-PAF, a metabolite of PAF. Ten, 100 and 500 nmol for each substance was injected intracerebroventricularly. Both PAF and lyso-PAF enhanced non-rapid eye movement (NREM) sleep but not REM sleep. Lyso-PAF, but not PAF, induced hyperthermia. Results are consistent with the hypothesis that the brain cytokine network is involved in physiological sleep regulation.
Collapse
Affiliation(s)
- Tetsuya Kushikata
- Department of Anesthesiology, University of Hirosaki School of Medicine, Hirosaki 036-8506, Japan
| | | | | |
Collapse
|
40
|
Lubin FD, Johnston LD, Sweatt JD, Anderson AE. Kainate mediates nuclear factor-kappa B activation in hippocampus via phosphatidylinositol-3 kinase and extracellular signal-regulated protein kinase. Neuroscience 2005; 133:969-81. [PMID: 15916859 DOI: 10.1016/j.neuroscience.2005.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2005] [Revised: 03/24/2005] [Accepted: 03/26/2005] [Indexed: 01/01/2023]
Abstract
The transcription factor nuclear factor-kappa B (NF-kappaB) is an inducible regulator of genes that plays a crucial role in the nervous system. Glutamate receptor stimulation is one well-described mechanism for NF-kappaB activation. In the studies presented here we used the glutamate analog, kainate to investigate the signaling mechanisms that couple to NF-kappaB activation in hippocampus. Kainate (250 nM) application to hippocampal slices elicited a time-dependent increase in nuclear NF-kappaB levels in areas CA3 and CA1, but not dentate, compared with controls. Further analysis focused on hippocampal area CA3, revealed increased NF-kappaB DNA binding activity in response to kainate stimulation. Supershift electrophoretic mobility shift assay indicated that the kainate-mediated NF-kappaB complex binding DNA was composed of p65, p50, and c-Rel subunits. Through inhibition studies we found that extracellular signal-regulated protein kinase (ERK) and phosphatidylinositol-3 kinase (PI3K) couple to basal and kainate-mediated NF-kappaB DNA binding activity in area CA3. Kainate elicited decreased total and increased phospho-inhibitor kappa B alpha (IkappaBalpha), suggesting that kainate-mediated activation of NF-kappaB is via the classical IkappaB kinase pathway. Interestingly, inhibition of ERK but not PI3K blocked the kainate-mediated increase in phospho-IkappaBalpha. Thus, our findings support a role for the ERK and PI3K pathways in kainate-mediated NF-kappaB activation in hippocampal area CA3, but these kinases may target the NF-kappaB pathway at different loci.
Collapse
Affiliation(s)
- F D Lubin
- The Cain Foundation Laboratories, Texas Children's Hospital, Department of Pediatrics, MC 3-6365, 1102 Bates Street, Houston, TX 77030-2399, USA
| | | | | | | |
Collapse
|
41
|
Bryant PA, Trinder J, Curtis N. Sick and tired: Does sleep have a vital role in the immune system? Nat Rev Immunol 2004; 4:457-67. [PMID: 15173834 DOI: 10.1038/nri1369] [Citation(s) in RCA: 329] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Penelope A Bryant
- University Department of Paediatrics, Murdoch Childrens Research Institute, and Department of General Medicine, Paediatric Infectious Diseases Unit, Royal Children's Hospital, Flemington Road, Parkville, Victoria 3052, Australia
| | | | | |
Collapse
|
42
|
Marpegan L, Bekinschtein TA, Freudenthal R, Rubio MF, Ferreyra GA, Romano A, Golombek DA. Participation of transcription factors from the Rel/NF-kappa B family in the circadian system in hamsters. Neurosci Lett 2004; 358:9-12. [PMID: 15016422 DOI: 10.1016/j.neulet.2003.12.112] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2003] [Revised: 11/30/2003] [Accepted: 12/13/2003] [Indexed: 11/17/2022]
Abstract
We have studied the presence and activity of components of the nuclear factor-kappaB (NF-kappaB) transcription factor in the hamster circadian system analyzing wheel-running activity, protein expression and DNA binding activity by electrophoresis mobility shift assays (EMSA). Non-rhythmic specific immunoreactive bands corresponding to a NF-kappaB subunit (p65) were found in hamster suprachiasmatic nuclei (SCN) homogenates. The active form of NF-kappaB evidenced by EMSA was clear and specific in SCN nuclear extracts. The administration of the NF-kappaB inhibitor pyrrolidine-dithiocharbamate (PDTC) blocked the light-induced phase advance at circadian time 18 (vehicle+light pulse: 2.08+/-0.46 h, PDTC+light: 0.36+/-0.35 h). These results demonstrate the presence and activity of Rel/NF-kappaB family proteins in the hamster SCN and suggest that these proteins may be related to the entrainment and regulation of circadian rhythms.
Collapse
Affiliation(s)
- Luciano Marpegan
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Roque Sáenz Peña 180, Bernal, B1876BXD Pcia. de Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
43
|
Schulze G. Sleep protects excitatory cortical circuits against oxidative damage. Med Hypotheses 2004; 63:203-7. [PMID: 15236776 DOI: 10.1016/j.mehy.2004.02.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Accepted: 02/25/2004] [Indexed: 11/30/2022]
Abstract
Activity in excitatory cortical pathways increases the oxidative metabolism of the brain and the risk of oxidative damage. Oxyradicals formed during periods of activity are mopped up by neural pools of nuclear factor kappa-B resulting in their activation and translocation to cell nuclei. During waking hours, glucocorticoids inhibit transactivation by nuclear factor kappa-B, increase central norepinephrine release, and elevate expression of prostaglandin D2. The build-up of nuclear factor kappa-B and prostaglandin D2 produces sleep pressures leading to sleep onset, normally gated by circadian melatonin release. During slow wave sleep nuclear factor kappa-B induces transcription of synaptogenic and antioxidant products and synaptic remodeling follows. Synaptically remodeled neural circuits have modified conductivity patterns and timescales and need to be resynchronized with existing unmodified neural circuits. The resynchronization process, mediated by theta rhythm, occurs during rapid eye movement sleep and is orchestrated from pontine centers. Resynchronization of remodeled neural circuits produces dreams. The waking state results upon successful resynchronization. Rapid eye movement sleep deprivation results in a lack of resynchronization and leads to cognitive inefficiencies. The model presented here proposes that the primary purpose of sleep is to protect cortical circuits against oxidative damage by reducing cortical activity and by remodeling and resynchronizing cortical circuits during this period of reduced activity to sustain new patterns of activation more effectively.
Collapse
Affiliation(s)
- Georg Schulze
- Department of Psychology, The University of British Columbia, 2136 West Mall, Vancouver, BC, Canada V6T 1Z4.
| |
Collapse
|
44
|
García-García F, Krueger JM. Intracerebroventricular injection of erythropoietin enhances sleep in the rat. Brain Res Bull 2003; 61:541-6. [PMID: 13679254 DOI: 10.1016/s0361-9230(03)00191-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Systemic injection of erythropoietin (EPO) over several days reduces sleep fragmentation in patients with periodic limb movements in sleep (PLMS). However, there are no studies concerning the effects of EPO on spontaneous sleep. In this study, we determined the effects of intracerebroventricular (i.c.v.) administration of EPO on spontaneous rat sleep. Three doses of EPO (25, 75, and 125 ng) were injected i.c.v. at the onset of the dark period. All doses of EPO increased non-rapid eye movement sleep (NREMS). In addition, high and low doses of EPO (125 and 25 ng) increased rapid eye movement sleep (REMS), but the medium dose of EPO (75 ng) inhibited REMS. Electroencephalogram slow-wave activity during NREMS also increased following the two higher doses of EPO. In contrast, EPO injection during the light period failed to affect sleep. Brain temperature (Tbr) was not affected by any dose of EPO. These results suggest that EPO could be part of the cytokine network involved in sleep regulation.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/metabolism
- Circadian Rhythm/drug effects
- Circadian Rhythm/physiology
- Cytokines/metabolism
- Dose-Response Relationship, Drug
- Erythropoietin/metabolism
- Erythropoietin/pharmacology
- Injections, Intraventricular
- Male
- Photic Stimulation
- Rats
- Rats, Sprague-Dawley
- Receptors, Erythropoietin/drug effects
- Receptors, Erythropoietin/metabolism
- Sleep/drug effects
- Sleep/physiology
- Sleep Disorders, Circadian Rhythm/drug therapy
- Sleep Disorders, Circadian Rhythm/metabolism
- Sleep Disorders, Circadian Rhythm/physiopathology
- Sleep Wake Disorders/drug therapy
- Sleep Wake Disorders/metabolism
- Sleep Wake Disorders/physiopathology
- Sleep, REM/drug effects
- Sleep, REM/physiology
Collapse
Affiliation(s)
- Fabio García-García
- Department of Veterinary and Comparative Anatomy, Pharmacology and Physiology, College of Veterinary Medicine, Washington State University, P.O. Box 99164-6520, Pullman, WA, USA
| | | |
Collapse
|
45
|
Yoshida H, Kubota T, Krueger JM. A cyclooxygenase-2 inhibitor attenuates spontaneous and TNF-alpha-induced non-rapid eye movement sleep in rabbits. Am J Physiol Regul Integr Comp Physiol 2003; 285:R99-109. [PMID: 12623776 DOI: 10.1152/ajpregu.00609.2002] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sleep is regulated in part by the brain cytokine network, including tumor necrosis factor-alpha (TNF-alpha). TNF-alpha activates the transcription factor nuclear factor-kappaB, which in turn promotes transcription of many genes, including cyclooxygenase-2 (COX-2). COX-2 is in the brain and is an enzyme responsible for production of prostaglandin D2. The hypothesis that central COX-2 plays a role in the regulation of spontaneous and TNF-alpha-induced sleep was investigated. Three doses (0.5, 5, and 50 microg) of NS-398, a highly selective COX-2 inhibitor, were injected intracerebroventricularly. The highest dose decreased non-rapid eye movement sleep. The intermediate and highest doses decreased electroencephalographic slow-wave activity; the greatest reduction occurred after 50 microg of NS-398 during the first 3-h postinjection period. Rapid eye movement sleep and brain temperature were not altered by any dose of NS-398. Pretreatment of rabbits with 5 or 50 microg of NS-398 blocked the TNF-alpha-induced increases in non-rapid eye movement sleep, electroencephalographic slow-wave activity, and brain temperature. These data suggest that COX-2 is involved in the regulation of spontaneous and TNF-alpha-induced sleep.
Collapse
Affiliation(s)
- Hitoshi Yoshida
- Dept. of VCAPP, PO Box 646520, Washington State University, Pullman, WA 99164-6520, USA
| | | | | |
Collapse
|
46
|
Fujimori K, Fujitani Y, Kadoyama K, Kumanogoh H, Ishikawa K, Urade Y. Regulation of lipocalin-type prostaglandin D synthase gene expression by Hes-1 through E-box and interleukin-1 beta via two NF-kappa B elements in rat leptomeningeal cells. J Biol Chem 2003; 278:6018-26. [PMID: 12488457 DOI: 10.1074/jbc.m208288200] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The promoter function of the rat lipocalin-type prostaglandin D synthase (L-PGDS) gene was characterized in primary cultures of leptomeningeal cells prepared from the neonatal rat brain. Luciferase reporter assays with deletion and site-directed mutation of the promoter region (-1250 to +77) showed that an AP-2 element at -109 was required for activation and an E-box at +57, for repression. Binding of nuclear factors to each of these cis-elements was demonstrated by an electrophoretic mobility shift assay. Several components of the Notch-Hes signaling pathway, Jagged, Notch1, Notch3, and Hes-1, were expressed in the leptomeningeal cells. Human Hes-1 co-expressed in the leptomeningeal cells bound to the E-box of the rat L-PGDS gene, and repressed the promoter activity of the rat L-PGDS gene in a dose-dependent manner. The L-PGDS gene expression was up-regulated slowly by interleukin-1 beta to the maximum level at 24 h. The reporter assay with deletion and mutation revealed that two NF-kappa B elements at -1106 and -291 were essential for this up-regulation. Binding of two NF-kappa B subunits, p65 and c-Rel, to these two NF-kappa B elements occurred after the interleukin-1 beta treatment. Therefore, the L-PGDS gene is the first gene identified as the target for the Notch-Hes signal through the E-box among a variety of genes involved in the prostanoid biosynthesis, classified to the lipocalin family, and expressed in the leptomeninges. Moreover, the L-PGDS gene is a unique gene that is activated slowly by the NF-kappa B system.
Collapse
Affiliation(s)
- Ko Fujimori
- Core Research for Evolutional Science and Technology, Japan Science and Technology Corporation and the Department of Molecular Behavioral Biology, Osaka Bioscience Institute, 6-2-4 Furuedai, Suita, Osaka 565-0874, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Sickness behaviour represents the expression of the adaptive reorganization of the priorities of the host during an infectious episode. This process is triggered by pro-inflammatory cytokines produced by peripheral phagocytic cells in contact with invading micro-organisms. The peripheral immune message is relayed to the brain via a fast neural pathway and a slower humoral pathway, resulting in the expression of pro-inflammatory cytokines in macrophage-like cells and microglia in the brain. The cellular and molecular components of this previously unsuspected system are being progressively identified. These advances are opening new avenues for understanding brain disorders, including depression.
Collapse
Affiliation(s)
- Jan Pieter Konsman
- Laboratory of Integrative Neurobiology, INRA-INSERM U 394, Rue Camille Saint-Saens, Bordeaux, France
| | | | | |
Collapse
|
48
|
Kubota T, Brown RA, Fang J, Krueger JM. Interleukin-15 and interleukin-2 enhance non-REM sleep in rabbits. Am J Physiol Regul Integr Comp Physiol 2001; 281:R1004-12. [PMID: 11507019 DOI: 10.1152/ajpregu.2001.281.3.r1004] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interleukin (IL)-15 and -2 share receptor- and signal-transduction pathway (Jak-STAT pathway) components. IL-2 is somnogenic in rats but has not been tested in other species. Furthermore, the effects of IL-15 on sleep have not heretofore been described. We investigated the somnogenic actions of IL-15 in rabbits and compared them with those of IL-2. Three doses of IL-15 or -2 (10, 100, and 500 ng) were injected intracerebroventriculary at the onset of the dark period. In addition, 500 ng of IL-15 and -2 were injected 3 h after the beginning of the light period. IL-15 dose dependently increased non-rapid eye movement sleep (NREMS) and induced fever. IL-15 inhibited rapid eye movement sleep (REMS) after its administration during the light period; however, all doses of IL-15 failed to affect REMS if given at dark onset. IL-2 also dose dependently increased NREMS and fever. IL-2 inhibited REMS, and this effect was observed only in the light period. IL-15 and -2 enhanced electroencephalographic (EEG) slow waves during the initial 9-h postinjection period, then, during hours 10-23 postinjection, reduced EEG slow-wave activity. Current data support the notion that the brain cytokine network is involved in the regulation of sleep.
Collapse
Affiliation(s)
- T Kubota
- College of Veterinary Medicine, Department of VCAPP, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | |
Collapse
|
49
|
Kubota T, Fang J, Brown RA, Krueger JM. Interleukin-18 promotes sleep in rabbits and rats. Am J Physiol Regul Integr Comp Physiol 2001; 281:R828-38. [PMID: 11506998 DOI: 10.1152/ajpregu.2001.281.3.r828] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Interleukin (IL)-1beta is involved in physiological sleep regulation. IL-18 is a member of the IL-1 family, and its signal-transduction mechanism is similar to that of IL-1. Therefore, we hypothesized that IL-18 might also be involved in sleep regulation. Three doses of IL-18 (10, 100, and 500 ng) were injected intracerebroventricularly (icv) into rabbits at the onset of the dark period. The two higher doses of IL-18 markedly increased non-rapid eye movement sleep (NREMS), accompanied by increases in brain temperature (Tbr). These effects were lost after the heat inactivation of IL-18. The 500 ng of IL-18 injection during the light period also increased NREMS and Tbr. Similar results were obtained after icv injection of 100 ng of IL-18 into rats. Furthermore, intraperitoneal injection of 30 microg/kg of IL-18 slightly, but significantly, increased NREMS, whereas it significantly decreased electroencephalogram slow-wave activity in rats. Intraperitoneal IL-18 failed to induce fever. An anti-human IL-18 antibody had little effect on spontaneous sleep in rabbits, although the anti-IL-18 antibody significantly attenuated muramyl dipeptide-induced sleep. These data suggest that IL-18 is involved in mechanisms of sleep responses to infection.
Collapse
Affiliation(s)
- T Kubota
- College of Veterinary Medicine, Department of VCAPP, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | |
Collapse
|
50
|
Kubota T, Fang J, Guan Z, Brown RA, Krueger JM. Vagotomy attenuates tumor necrosis factor-alpha-induced sleep and EEG delta-activity in rats. Am J Physiol Regul Integr Comp Physiol 2001; 280:R1213-20. [PMID: 11247847 DOI: 10.1152/ajpregu.2001.280.4.r1213] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Much evidence suggests that tumor necrosis factor-alpha (TNF-alpha) is involved in the regulation of physiological sleep. However, it remains unclear whether peripheral administration of TNF-alpha induces sleep in rats. Furthermore, the role of the vagus nerve in the somnogenic actions of TNF-alpha had not heretofore been studied. Four doses of TNF-alpha were administered intraperitoneally just before the onset of the dark period. The three higher doses of TNF-alpha (50, 100, and 200 microg/kg) dose dependently increased nonrapid eye movement sleep (NREMS), accompanied by increases in electroencephalogram (EEG) slow-wave activity. TNF-alpha increased EEG delta-power and decreased EEG alpha- and beta-power during the initial 3 h after injection. In vagotomized rats, the NREMS responses to 50 or 100 microg/kg of TNF-alpha were attenuated, while significant TNF-alpha-induced increases in NREMS were observed in a sham-operated group. Moreover, the vagotomized rats failed to exhibit the increase in EEG delta-power induced by TNF-alpha intraperitoneally. These results suggest that peripheral TNF-alpha can induce NREMS and vagal afferents play an important role in the effects of peripheral TNF-alpha and EEG synchronization on sleep. Intraperitoneal TNF-alpha failed to affect brain temperature at the doses tested, thereby demonstrating that TNF-alpha-induced sleep effects are, in part, independent from its effects on brain temperature. Results are consistent with the hypothesis that a cytokine network is involved in sleep regulation.
Collapse
Affiliation(s)
- T Kubota
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | | | |
Collapse
|