1
|
Nemeth DP, Liu X, Monet MC, Niu H, Maxey G, Schrier MS, Smirnova MI, McGovern SJ, Herd A, DiSabato DJ, Floyd T, Atluri RR, Nusstein AC, Oliver B, Witcher KG, Juste Ellis JS, Yip J, Crider AD, McKim DB, Gajewski-Kurdziel PA, Godbout JP, Zhang Q, Blakely RD, Sheridan JF, Quan N. Localization of brain neuronal IL-1R1 reveals specific neural circuitries responsive to immune signaling. J Neuroinflammation 2024; 21:303. [PMID: 39563437 PMCID: PMC11575132 DOI: 10.1186/s12974-024-03287-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
Interleukin-1 (IL-1) is a pro-inflammatory cytokine that exerts a wide range of neurological and immunological effects throughout the central nervous system (CNS) and is associated with the etiology of affective and cognitive disorders. The cognate receptor for IL-1, Interleukin-1 Receptor Type 1 (IL-1R1), is primarily expressed on non-neuronal cells (e.g., endothelial cells, choroidal cells, ventricular ependymal cells, astrocytes, etc.) throughout the brain. However, the presence and distribution of neuronal IL-1R1 (nIL-1R1) has been controversial. Here, for the first time, a novel genetic mouse line that allows for the visualization of IL-1R1 mRNA and protein expression (Il1r1GR/GR) was used to map all brain nuclei and determine the neurotransmitter systems which express nIL-1R1 in adult male mice. The direct responsiveness of nIL-1R1-expressing neurons to both inflammatory and physiological levels of IL-1β in vivo was tested. Neuronal IL-1R1 expression across the brain was found in discrete glutamatergic and serotonergic neuronal populations in the somatosensory cortex, piriform cortex, dentate gyrus, and dorsal raphe nucleus. Glutamatergic nIL-1R1 comprises most of the nIL-1R1 expression and, using Vglut2-Cre-Il1r1r/r mice, which restrict IL-1R1 expression to only glutamatergic neurons, an atlas of glutamatergic nIL-1R1 expression across the brain was generated. Analysis of functional outputs of these nIL-1R1-expressing nuclei, in both Il1r1GR/GR and Vglut2-Cre-Il1r1r/r mice, reveals IL-1R1+ nuclei primarily relate to sensory detection, processing, and relay pathways, mood regulation, and spatial/cognitive processing centers. Intracerebroventricular (i.c.v.) injections of IL-1 (20 ng) induces NFκB signaling in IL-1R1+ non-neuronal cells but not in IL-1R1+ neurons, and in Vglut2-Cre-Il1r1r/r mice IL-1 did not change gene expression in the dentate gyrus of the hippocampus (DG). GO pathway analysis of spatial RNA sequencing 1mo following restoration of nIL-1R1 in the DG neurons reveals IL-1R1 expression downregulates genes related to both synaptic function and mRNA binding while increasing select complement markers (C1ra, C1qb). Further, DG neurons exclusively express an alternatively spliced IL-1R Accessory protein isoform (IL-1RAcPb), a known synaptic adhesion molecule. Altogether, this study reveals a unique network of neurons that can respond directly to IL-1 via nIL-1R1 through non-autonomous transcriptional pathways; earmarking these circuits as potential neural substrates for immune signaling-triggered sensory, affective, and cognitive disorders.
Collapse
Affiliation(s)
- Daniel P Nemeth
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA.
| | - Xiaoyu Liu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Marianne C Monet
- The International Max Planck Research School (IMPRS) for Synapses and Circuits, Max Planck Florida Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Haichen Niu
- Department of Genetics, Xuzhou Medical University, Xuzhou, 221004, China
| | - Gabriella Maxey
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
- Wilkes Honors College, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Matt S Schrier
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Maria I Smirnova
- The International Max Planck Research School (IMPRS) for Synapses and Circuits, Max Planck Florida Institute for Neuroscience Jupiter, Jupiter, FL, 33458, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, 33458, USA
| | | | - Anu Herd
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Damon J DiSabato
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Trey Floyd
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Rohit R Atluri
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- College of Medicine and Life Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Alex C Nusstein
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Braedan Oliver
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Kristina G Witcher
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Joshua St Juste Ellis
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Jasmine Yip
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Andrew D Crider
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
| | - Daniel B McKim
- Department of Animal Science, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | | | - Jonathan P Godbout
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Qi Zhang
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
- Department of Biological Sciences, Charles E. Schmidt College of Science, Florida Atlantic University, Jupiter, FL, 33458, USA
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL, 33431, USA
| | - Randy D Blakely
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - John F Sheridan
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, 5353 Parkside Drive, Jupiter, FL, 33458, USA.
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, 33458, USA.
| |
Collapse
|
2
|
Valencia-Sanchez S, Davis M, Martensen J, Hoeffer C, Link C, Opp MR. Sleep-wake behavior and responses to sleep deprivation and immune challenge of protein kinase RNA-activated knockout mice. Brain Behav Immun 2024; 121:74-86. [PMID: 39043346 PMCID: PMC11563030 DOI: 10.1016/j.bbi.2024.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024] Open
Abstract
Protein Kinase RNA-activated (PKR) is an enzyme that plays a role in many systemic processes, including modulation of inflammation, and is implicated in neurodegenerative diseases, such as Alzheimer's disease (AD). PKR phosphorylation results in the production of several cytokines involved in the regulation / modulation of sleep, including interleukin-1β, tumor necrosis factor-α and interferon-γ. We hypothesized targeting PKR would alter spontaneous sleep of mice, attenuate responses to sleep deprivation, and inhibit responses to immune challenge. To test these hypotheses, we determined the sleep-wake phenotype of mice lacking PKR (knockout; PKR-/-) during undisturbed baseline conditions; in responses to six hours of sleep deprivation; and after immune challenge with lipopolysaccharide (LPS). Adult male mice (C57BL/6J, n = 7; PKR-/-, n = 7) were surgically instrumented with EEG recording electrodes and an intraperitoneal microchip to record core body temperature. During undisturbed baseline conditions, PKR -/- mice spent more time in non-rapid eye movement sleep (NREMS) and rapid-eye movement sleep (REMS), and less time awake at the beginning of the dark period of the light:dark cycle. Delta power during NREMS, a measure of sleep depth, was less in PKR-/- mice during the dark period, and core body temperatures were lower during the light period. Both mouse strains responded to sleep deprivation with increased NREMS and REMS, although these changes did not differ substantively between strains. The initial increase in delta power during NREMS after sleep deprivation was greater in PKR-/- mice, suggesting a faster buildup of sleep pressure with prolonged waking. Immune challenge with LPS increased NREMS and inhibited REMS to the same extent in both mouse strains, whereas the initial LPS-induced suppression of delta power during NREMS was greater in PKR-/- mice. Because sleep regulatory and immune responsive systems in brain are redundant and overlapping, other mediators and signaling pathways in addition to PKR are involved in the responses to acute sleep deprivation and LPS immune challenge.
Collapse
Affiliation(s)
- S Valencia-Sanchez
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - M Davis
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - J Martensen
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - C Hoeffer
- Institute for Behavioral Genetics, University of Colorado Boulder, USA
| | - C Link
- Department of Integrative Physiology, University of Colorado Boulder, USA
| | - M R Opp
- Department of Integrative Physiology, University of Colorado Boulder, USA.
| |
Collapse
|
3
|
Kalantari N, Daneault V, Blais H, André C, Sanchez E, Lina JM, Arbour C, Gilbert D, Carrier J, Gosselin N. Cerebral Gray Matter May Not Explain Sleep Slow-Wave Characteristics after Severe Brain Injury. J Neurosci 2024; 44:e1306232024. [PMID: 38844342 PMCID: PMC11308330 DOI: 10.1523/jneurosci.1306-23.2024] [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] [Received: 07/12/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 08/09/2024] Open
Abstract
Sleep slow waves are the hallmark of deeper non-rapid eye movement sleep. It is generally assumed that gray matter properties predict slow-wave density, morphology, and spectral power in healthy adults. Here, we tested the association between gray matter volume (GMV) and slow-wave characteristics in 27 patients with moderate-to-severe traumatic brain injury (TBI, 32.0 ± 12.2 years old, eight women) and compared that with 32 healthy controls (29.2 ± 11.5 years old, nine women). Participants underwent overnight polysomnography and cerebral MRI with a 3 Tesla scanner. A whole-brain voxel-wise analysis was performed to compare GMV between groups. Slow-wave density, morphology, and spectral power (0.4-6 Hz) were computed, and GMV was extracted from the thalamus, cingulate, insula, precuneus, and orbitofrontal cortex to test the relationship between slow waves and gray matter in regions implicated in the generation and/or propagation of slow waves. Compared with controls, TBI patients had significantly lower frontal and temporal GMV and exhibited a subtle decrease in slow-wave frequency. Moreover, higher GMV in the orbitofrontal cortex, insula, cingulate cortex, and precuneus was associated with higher slow-wave frequency and slope, but only in healthy controls. Higher orbitofrontal GMV was also associated with higher slow-wave density in healthy participants. While we observed the expected associations between GMV and slow-wave characteristics in healthy controls, no such associations were observed in the TBI group despite lower GMV. This finding challenges the presumed role of GMV in slow-wave generation and morphology.
Collapse
Affiliation(s)
- Narges Kalantari
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, Quebec H2V 2S9, Canada
| | - Véronique Daneault
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, Quebec H2V 2S9, Canada
| | - Hélène Blais
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
| | - Claire André
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, Quebec H2V 2S9, Canada
| | - Erlan Sanchez
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Cognitive Neurology Research Unit, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | - Jean-Marc Lina
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Department of Electrical Engineering, École de Technologie Supérieure, Montreal, Quebec H3C 1K3, Canada
| | - Caroline Arbour
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Faculty of Nursing, Université de Montréal, Montreal, Quebec H3T 1A8, Canada
| | - Danielle Gilbert
- Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montréal, Montreal, Quebec H3T 1A4, Canada
- Department of Radiology, Hôpital du Sacré-Coeur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
| | - Julie Carrier
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, Quebec H2V 2S9, Canada
| | - Nadia Gosselin
- Center for Advanced Research in Sleep Medicine, Hôpital du Sacré-Cœur de Montréal, Centre intégré universitaire de santé et de services sociaux du Nord-de-l'Île-de Montréal, Montreal, Quebec H4J 1C5, Canada
- Department of Psychology, Université de Montréal, Montreal, Quebec H2V 2S9, Canada
| |
Collapse
|
4
|
Phan TX, Sahibzada N, Ahern GP. Arteries are finely tuned thermosensors regulating myogenic tone and blood flow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.532099. [PMID: 36993664 PMCID: PMC10055355 DOI: 10.1101/2023.03.22.532099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In response to changing blood pressure, arteries adjust their caliber to control perfusion. This vital autoregulatory property, termed vascular myogenic tone, stabilizes downstream capillary pressure. We discovered that tissue temperature critically determines myogenic tone. Heating steeply activates tone in skeletal muscle, gut, brain and skin arteries with temperature coefficients ( Q 10 ) of ∼11-20. Further, arterial thermosensitivity is tuned to resting tissue temperatures, making myogenic tone sensitive to small thermal fluctuations. Interestingly, temperature and intraluminal pressure are sensed largely independently and integrated to trigger myogenic tone. We show that TRPV1 and TRPM4 mediate heat-induced tone in skeletal muscle arteries. Variations in tissue temperature are known to alter vascular conductance; remarkably, thermosensitive tone counterbalances this effect, thus protecting capillary integrity and fluid balance. In conclusion, thermosensitive myogenic tone is a fundamental homeostatic mechanism regulating tissue perfusion. One-Sentence Summary Arterial blood pressure and temperature are integrated via thermosensitve ion channels to produce myogenic tone.
Collapse
|
5
|
Ingiosi AM, Frank MG. Goodnight, astrocyte: waking up to astroglial mechanisms in sleep. FEBS J 2023; 290:2553-2564. [PMID: 35271767 PMCID: PMC9463397 DOI: 10.1111/febs.16424] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/16/2022] [Accepted: 03/07/2022] [Indexed: 01/03/2023]
Abstract
Astrocytes mediate many important aspects of neural homeostasis, but until recently, their role in sleep was largely unknown. The situation has dramatically changed in the last decade. The use of transgenic animals, optogenetics, chemogenetics, brain imaging and sophisticated molecular assays has led to exciting discoveries. Astrocytes dynamically change their activity across the sleep-wake cycle and may encode sleep need via changes in intracellular signalling pathways. Astrocytes also exocytose/secrete sleep-inducing molecules which modulate brain activity, sleep architecture and sleep regulation. Many of these observations have been made in mice and Drosophila melanogaster, indicating that astroglial sleep mechanisms are evolutionarily conserved. We review recent findings and discuss future directions.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Marcos G Frank
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| |
Collapse
|
6
|
Kuna K, Szewczyk K, Gabryelska A, Białasiewicz P, Ditmer M, Strzelecki D, Sochal M. Potential Role of Sleep Deficiency in Inducing Immune Dysfunction. Biomedicines 2022; 10:biomedicines10092159. [PMID: 36140260 PMCID: PMC9496201 DOI: 10.3390/biomedicines10092159] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Sleep deficiency and insomnia deteriorate the quality of patients’ lives, yet the exact influence of these factors on the immune system has only begun to gain interest in recent years. Growing evidence shows that insomnia is a risk factor for numerous diseases, including common infections and autoimmune diseases. Levels of inflammatory markers also seem to be abnormal in sleep deficient individuals, which may lead to low-grade inflammation. The interpretation of studies is difficult due to the equivocal term “sleep disturbances,” as well as due to the various criteria used in studies. This narrative review aims to summarize the available knowledge regarding the bidirectional influence of the immune system and sleep disturbances.
Collapse
Affiliation(s)
- Kasper Kuna
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Krzysztof Szewczyk
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Marta Ditmer
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-213 Lodz, Poland
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland
- Correspondence: ; Tel.: +48-42-678-18-00
| |
Collapse
|
7
|
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: 62] [Impact Index Per Article: 20.7] [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
|
8
|
Hsu CY, Chuang YC, Chang FC, Chuang HY, Chiou TTY, Lee CT. Disrupted Sleep Homeostasis and Altered Expressions of Clock Genes in Rats with Chronic Lead Exposure. TOXICS 2021; 9:toxics9090217. [PMID: 34564368 PMCID: PMC8473409 DOI: 10.3390/toxics9090217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 02/04/2023]
Abstract
Sleep disturbance is one of the neurobehavioral complications of lead neurotoxicity. The present study evaluated the impacts of chronic lead exposure on alteration of the sleep–wake cycle in association with changes of clock gene expression in the hypothalamus. Sprague–Dawley rats with chronic lead exposure consumed drinking water that contained 250 ppm of lead acetate for five weeks. Electroencephalography and electromyography were recorded for scoring the architecture of the sleep–wake cycle in animals. At six Zeitgeber time (ZT) points (ZT2, ZT6, ZT10, ZT14, ZT18, and ZT22), three clock genes, including rPer1, rPer2, and rBmal1b, were analyzed. The rats with chronic lead exposure showed decreased slow wave sleep and increased wakefulness in the whole light period (ZT1 to ZT12) and the early dark period (ZT13 to ZT15) that was followed with a rebound of rapid-eye-movement sleep at the end of the dark period (ZT22 to ZT24). The disturbance of the sleep–wake cycle was associated with changes in clock gene expression that was characterized by the upregulation of rPer1 and rPer2 and the feedback repression of rBmal1b. We concluded that chronic lead exposure has a negative impact on the sleep–wake cycle in rats that predominantly disrupts sleep homeostasis. The disruption of sleep homeostasis was associated with a toxic effect of lead on the clock gene expression in the hypothalamus.
Collapse
Affiliation(s)
- Chung-Yao Hsu
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yao-Chung Chuang
- Department of Neurology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Fang-Chia Chang
- School of Veterinary Medicine, National Taiwan University, Taipei 10617, Taiwan;
| | - Hung-Yi Chuang
- Department of Public Health and Environmental Medicine, College of Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Terry Ting-Yu Chiou
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung 83301, Taiwan
| | - Chien-Te Lee
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, Kaohsiung 83301, Taiwan
- Chang-Gang Kidney Research Center, Kaohsiung 83301, Taiwan
- Correspondence:
| |
Collapse
|
9
|
Tea polyphenols protect learning and memory in sleep-deprived mice by promoting AMPA receptor internalization. Neuroreport 2021; 31:857-864. [PMID: 32453025 PMCID: PMC7368842 DOI: 10.1097/wnr.0000000000001462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic sleep loss caused lots of health problems, also including cognition impairment. Tea is one of the most popular drinks when people stay up late. Nevertheless, the effects of tea on sleep deprivation-induced cognition impairment are still unclear. In the present study, we found 24-h sleep deprivation (S-DEP) increased membrane α-amino-3-hydroxy-5-methyl-4-isoxa-zolep-propionate (AMPA) receptor level through a tumor necrosis factor α (TNFα)-dependent pathway in hippocampi. Blocking elevated TNFα level can protect S-DEP mice from impaired learning ability according to behavioral test. Tea polyphenols, major active compounds in green tea, suppressed TNFα production through downregulating TNFα converting enzyme (TACE) level. Meanwhile, tea polyphenols treatment could ameliorate recognition impairment and anxiety-like behaviors in S-DEP mice. The aforementioned results demonstrate cognition protective effects of tea polyphenols in S-DEP mice model, which provide a theoretical basis for the treatments of S-DEP-induced cognition impairment by targeting the TACE/TNFα/AMPA pathway.
Collapse
|
10
|
Yang Y, Gritton H, Sarter M, Aton SJ, Booth V, Zochowski M. Theta-gamma coupling emerges from spatially heterogeneous cholinergic neuromodulation. PLoS Comput Biol 2021; 17:e1009235. [PMID: 34329297 PMCID: PMC8357148 DOI: 10.1371/journal.pcbi.1009235] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 08/11/2021] [Accepted: 07/01/2021] [Indexed: 11/18/2022] Open
Abstract
Theta and gamma rhythms and their cross-frequency coupling play critical roles in perception, attention, learning, and memory. Available data suggest that forebrain acetylcholine (ACh) signaling promotes theta-gamma coupling, although the mechanism has not been identified. Recent evidence suggests that cholinergic signaling is both temporally and spatially constrained, in contrast to the traditional notion of slow, spatially homogeneous, and diffuse neuromodulation. Here, we find that spatially constrained cholinergic stimulation can generate theta-modulated gamma rhythms. Using biophysically-based excitatory-inhibitory (E-I) neural network models, we simulate the effects of ACh on neural excitability by varying the conductance of a muscarinic receptor-regulated K+ current. In E-I networks with local excitatory connectivity and global inhibitory connectivity, we demonstrate that theta-gamma-coupled firing patterns emerge in ACh modulated network regions. Stable gamma-modulated firing arises within regions with high ACh signaling, while theta or mixed theta-gamma activity occurs at the peripheries of these regions. High gamma activity also alternates between different high-ACh regions, at theta frequency. Our results are the first to indicate a causal role for spatially heterogenous ACh signaling in the emergence of localized theta-gamma rhythmicity. Our findings also provide novel insights into mechanisms by which ACh signaling supports the brain region-specific attentional processing of sensory information.
Collapse
Affiliation(s)
- Yihao Yang
- Department of Physics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Howard Gritton
- Department of Comparative Biosciences and Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Martin Sarter
- Department of Psychology and Neuroscience Program, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Victoria Booth
- Departments of Mathematics and Anesthesiology, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (VB); (MZ)
| | - Michal Zochowski
- Department of Physics and Biophysics Program, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (VB); (MZ)
| |
Collapse
|
11
|
Fang H, Li Z, Graff EC, McCafferty KJ, Judd RL. Niacin increases diet-induced hepatic steatosis in B6129 mice. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158731. [PMID: 32404278 DOI: 10.1016/j.bbalip.2020.158731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a very common disorder affecting between 20 and 30% of adults in the United States. However, there is no effective pharmacotherapy for treating NAFLD. Niacin, a water-soluble vitamin (B3), at pharmacological doses, decreases hepatic triglyceride (TG) content in NAFLD through inhibition of diacylglycerol acyltransferase 2, a key enzyme that catalyzes the final step in TG synthesis. Alternatively, some studies indicate that niacin induces fatty liver in high-fat diet (HFD)-fed rats. Therefore, in this study we investigated whether niacin is beneficial in treating NAFLD in two strains of mice, C57BL/6J (B6) and B6129SF2/J (B6129) mice, with 20 weeks of HFD feeding. Niacin treatment was started from week 5 until the end of the study. Niacin treatment increased normalized liver weight, hepatic TG content and NAFLD score in HFD-fed B6129 mice but had no impact on B6 mice. Metabolomics analysis revealed that in B6129 mice, 4-hydroxyphenylpyruvic acid (4-HPP), which is associated with fatty acid oxidation, did not change with HFD feeding but significantly decreased with niacin treatment. Lipidomics analysis discovered that the abundance of phosphocholine (PC), which is critical for very low-density lipoprotein (VLDL)-TG production and secretion, was decreased in HFD-fed B6129 with niacin treatment. In conclusion, niacin had no impact on diet-induced NAFLD development in B6 mice but potentiated hepatic steatosis in HFD-fed B6129 mice due to impaired fatty acid oxidation and decreased VLDL-TG production and secretion.
Collapse
Affiliation(s)
- Han Fang
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Zhuoyue Li
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Emily C Graff
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Kayleen J McCafferty
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Robert L Judd
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America.
| |
Collapse
|
12
|
Garofalo S, Picard K, Limatola C, Nadjar A, Pascual O, Tremblay MÈ. Role of Glia in the Regulation of Sleep in Health and Disease. Compr Physiol 2020; 10:687-712. [PMID: 32163207 DOI: 10.1002/cphy.c190022] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sleep is a naturally occurring physiological state that is required to sustain physical and mental health. Traditionally viewed as strictly regulated by top-down control mechanisms, sleep is now known to also originate locally. Glial cells are emerging as important contributors to the regulation of sleep-wake cycles, locally and among dedicated neural circuits. A few pioneering studies revealed that astrocytes and microglia may influence sleep pressure, duration as well as intensity, but the precise involvement of these two glial cells in the regulation of sleep remains to be fully addressed, across contexts of health and disease. In this overview article, we will first summarize the literature pertaining to the role of astrocytes and microglia in the regulation of sleep under normal physiological conditions. Afterward, we will discuss the beneficial and deleterious consequences of glia-mediated neuroinflammation, whether it is acute, or chronic and associated with brain diseases, on the regulation of sleep. Sleep disturbances are a main comorbidity in neurodegenerative diseases, and in several brain diseases that include pain, epilepsy, and cancer. Identifying the relationships between glia-mediated neuroinflammation, sleep-wake rhythm disruption and brain diseases may have important implications for the treatment of several disorders. © 2020 American Physiological Society. Compr Physiol 10:687-712, 2020.
Collapse
Affiliation(s)
- Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Katherine Picard
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France.,Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Agnès Nadjar
- Nutrition et Neurobiologie Intégrée, UMR 1286, Institut National de la Recherche Agronomique, Bordeaux University, Bordeaux, France
| | - Olivier Pascual
- INSERM U1028, CNRS UMR5292, Lyon Neuroscience Research Center, Université Claude Bernard Lyon, Lyon, France
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada.,Départment de médecine moleculaire, Faculté de médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
13
|
Ingiosi AM, Schoch H, Wintler T, Singletary KG, Righelli D, Roser LG, Medina E, Risso D, Frank MG, Peixoto L. Shank3 modulates sleep and expression of circadian transcription factors. eLife 2019; 8:e42819. [PMID: 30973326 PMCID: PMC6488297 DOI: 10.7554/elife.42819] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 04/10/2019] [Indexed: 12/30/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is the most prevalent neurodevelopmental disorder in the United States and often co-presents with sleep problems. Sleep problems in ASD predict the severity of ASD core diagnostic symptoms and have a considerable impact on the quality of life of caregivers. Little is known, however, about the underlying molecular mechanisms of sleep problems in ASD. We investigated the role of Shank3, a high confidence ASD gene candidate, in sleep architecture and regulation. We show that mice lacking exon 21 of Shank3 have problems falling asleep even when sleepy. Using RNA-seq we show that sleep deprivation increases the differences in prefrontal cortex gene expression between mutants and wild types, downregulating circadian transcription factors Per3, Bhlhe41, Hlf, Tef, and Nr1d1. Shank3 mutants also have trouble regulating wheel-running activity in constant darkness. Overall, our study shows that Shank3 is an important modulator of sleep and clock gene expression.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Hannah Schoch
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Taylor Wintler
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Kristan G Singletary
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Dario Righelli
- Istituto per le Applicazioni del Calcolo “M. Picone”Consiglio Nazionale della RicercheNapoliItaly
- Dipartimento di Scienze Aziendali Management & Innovation SystemsUniversity of FuscianoFiscianoItaly
| | - Leandro G Roser
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Elizabeth Medina
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Davide Risso
- Department of Statistical SciencesUniversity of PadovaPadovaItaly
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and ResearchWeill Cornell MedicineNew YorkUnited States
| | - Marcos G Frank
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| | - Lucia Peixoto
- Department of Biomedical Sciences, Elson S. Floyd College of MedicineWashington State UniversitySpokaneUnited States
| |
Collapse
|
14
|
Thomasy HE, Opp MR. Hypocretin Mediates Sleep and Wake Disturbances in a Mouse Model of Traumatic Brain Injury. J Neurotrauma 2019; 36:802-814. [PMID: 30136622 PMCID: PMC6387567 DOI: 10.1089/neu.2018.5810] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of disability worldwide. Post-TBI sleep and wake disturbances are extremely common and difficult for patients to manage. Sleep and wake disturbances contribute to poor functional and emotional outcomes from TBI, yet effective therapies remain elusive. A more comprehensive understanding of mechanisms underlying post-TBI sleep and wake disturbance will facilitate development of effective pharmacotherapies. Previous research in human patients and animal models indicates that altered hypocretinergic function may be a major contributor to sleep-wake disturbance after TBI. In this study, we further elucidate the role of hypocretin by determining the impact of TBI on sleep-wake behavior of hypocretin knockout (HCRT KO) mice. Adult male C57BL/6J and HCRT KO mice were implanted with electroencephalography recording electrodes, and pre-injury baseline recordings were obtained. Mice were then subjected to either moderate TBI or sham surgery. Additional recordings were obtained and sleep-wake behavior determined at 3, 7, 15, and 30 days after TBI or sham procedures. At baseline, HCRT KO mice had a significantly different sleep-wake phenotype than control C57BL/6J mice. Post-TBI sleep-wake behavior was altered in a genotype-dependent manner: sleep of HCRT KO mice was not altered by TBI, whereas C57BL/6J mice had more non-rapid eye movement sleep, less wakefulness, and more short wake bouts and fewer long wake bouts. Numbers of hypocretin-positive cells were reduced in C57BL/6J mice by TBI. Collectively, these data indicate that the hypocretinergic system is involved in the alterations in sleep-wake behavior that develop after TBI in this model, and suggest potential therapeutic interventions.
Collapse
Affiliation(s)
- Hannah E. Thomasy
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Mark R. Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, Washington
| |
Collapse
|
15
|
REV-ERBβ is required to maintain normal wakefulness and the wake-inducing effect of dual REV-ERB agonist SR9009. Biochem Pharmacol 2018; 150:1-8. [DOI: 10.1016/j.bcp.2018.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/03/2018] [Indexed: 12/12/2022]
|
16
|
Karlsson C, Schank JR, Rehman F, Stojakovic A, Björk K, Barbier E, Solomon M, Tapocik J, Engblom D, Thorsell A, Heilig M. Proinflammatory signaling regulates voluntary alcohol intake and stress-induced consumption after exposure to social defeat stress in mice. Addict Biol 2017; 22:1279-1288. [PMID: 27273552 DOI: 10.1111/adb.12416] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/22/2016] [Accepted: 05/10/2016] [Indexed: 01/23/2023]
Abstract
Proinflammatory activity has been postulated to play a role in addictive processes and stress responses, but the underlying mechanisms remain largely unknown. Here, we examined the role of interleukin 1 (IL-1) and tumor necrosis factor-α (TNF-α) in regulation of voluntary alcohol consumption, alcohol reward and stress-induced drinking. Mice with a deletion of the IL-1 receptor I gene (IL-1RI KO) exhibited modestly decreased alcohol consumption. However, IL-1RI deletion affected neither the rewarding properties of alcohol, measured by conditioned place preference (CPP), nor stress-induced drinking induced by social defeat stress. TNF-α signaling can compensate for phenotypic consequences of IL1-RI deletion. We therefore hypothesized that double deletion of both IL-1RI and TNF-1 receptors (TNF-1R) may reveal the role of these pathways in regulation of alcohol intake. Double KOs consumed significantly less alcohol than control mice over a range of alcohol concentrations. The combined deletion of TNF-1R and IL-1RI did not influence alcohol reward, but did prevent increased alcohol consumption resulting from exposure to repeated bouts of social defeat stress. Taken together, these data indicate that IL-1RI and TNF-1R contribute to regulation of stress-induced, negatively reinforced drinking perhaps through overlapping signaling events downstream of these receptors, while leaving rewarding properties of alcohol largely unaffected.
Collapse
Affiliation(s)
- Camilla Karlsson
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Jesse R. Schank
- Department of Physiology and Pharmacology; University of Georgia; Athens GA
| | - Faazal Rehman
- Laboratory of Clinical and Translational Studies; National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH); Bethesda MD USA
| | - Andrea Stojakovic
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Karl Björk
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Estelle Barbier
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Matthew Solomon
- Laboratory of Clinical and Translational Studies; National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH); Bethesda MD USA
| | - Jenica Tapocik
- Laboratory of Clinical and Translational Studies; National Institute of Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH); Bethesda MD USA
| | - David Engblom
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Annika Thorsell
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| | - Markus Heilig
- Department of Clinical and Experimental Medicine; Linköping University; Linköping Sweden
| |
Collapse
|
17
|
Meyer CW, Ootsuka Y, Romanovsky AA. Body Temperature Measurements for Metabolic Phenotyping in Mice. Front Physiol 2017; 8:520. [PMID: 28824441 PMCID: PMC5534453 DOI: 10.3389/fphys.2017.00520] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 07/06/2017] [Indexed: 01/01/2023] Open
Abstract
Key Points Rectal probing is subject to procedural bias. This method is suitable for first-line phenotyping, provided probe depth and measurement duration are standardized. It is also useful for detecting individuals with out-of-range body temperatures (during hypothermia, torpor).The colonic temperature attained by inserting the probe >2 cm deep is a measure of deep (core) body temperature.IR imaging of the skin is useful for detecting heat leaks and autonomous thermoregulatory alterations, but it does not measure body temperature.Temperature of the hairy or shaved skin covering the inter-scapular brown adipose tissue can be used as a measure of BAT thermogenesis. However, obtaining such measurements of sufficient quality is very difficult, and interpreting them can be tricky. Temperature differences between the inter-scapular and lumbar areas can be a better measure of the thermogenic activity of inter-scapular brown adipose tissue.Implanted probes for precise determination of BAT temperature (changes) should be fixed close to the Sulzer's vein. For measurement of BAT thermogenesis, core body temperature and BAT temperature should be recorded simultaneously.Tail temperature is suitable to compare the presence or absence of vasoconstriction or vasodilation.Continuous, longitudinal monitoring of core body temperature is preferred over single probing, as the readings are taken in a non-invasive, physiological context.Combining core body temperature measurements with metabolic rate measurements yields insights into the interplay between heat production and heat loss (thermal conductance), potentially revealing novel thermoregulatory phenotypes. Endothermic organisms rely on tightly balanced energy budgets to maintain a regulated body temperature and body mass. Metabolic phenotyping of mice, therefore, often includes the recording of body temperature. Thermometry in mice is conducted at various sites, using various devices and measurement practices, ranging from single-time probing to continuous temperature imaging. Whilst there is broad agreement that body temperature data is of value, procedural considerations of body temperature measurements in the context of metabolic phenotyping are missing. Here, we provide an overview of the various methods currently available for gathering body temperature data from mice. We explore the scope and limitations of thermometry in mice, with the hope of assisting researchers in the selection of appropriate approaches, and conditions, for comprehensive mouse phenotypic analyses.
Collapse
Affiliation(s)
- Carola W Meyer
- Department of Pharmacology, Max-Planck Institute for Heart and Lung ResearchBad Nauheim, Germany
| | - Youichirou Ootsuka
- Centre for Neuroscience, School of Medicine, Flinders University of South AustraliaAdelaide, SA, Australia
| | - Andrej A Romanovsky
- FeverLab, St. Joseph's Hospital and Medical CenterPhoenix, AZ, United States
| |
Collapse
|
18
|
Shestopalov VI, Panchin Y, Tarasova OS, Gaynullina D, Kovalzon VM. Pannexins Are Potential New Players in the Regulation of Cerebral Homeostasis during Sleep-Wake Cycle. Front Cell Neurosci 2017; 11:210. [PMID: 28769767 PMCID: PMC5511838 DOI: 10.3389/fncel.2017.00210] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
During brain homeostasis, both neurons and astroglia release ATP that is rapidly converted to adenosine in the extracellular space. Pannexin-1 (Panx1) hemichannels represent a major conduit of non-vesicular ATP release from brain cells. Previous studies have shown that Panx1−/− mice possess severe disruption of the sleep-wake cycle. Here, we review experimental data supporting the involvement of pannexins (Panx) in the coordination of fundamental sleep-associated brain processes, such as neuronal activity and regulation of cerebrovascular tone. Panx1 hemichannels are likely implicated in the regulation of the sleep-wake cycle via an indirect effect of released ATP on adenosine receptors and through interaction with other somnogens, such as IL-1β, TNFα and prostaglandin D2. In addition to the recently established role of Panx1 in the regulation of endothelium-dependent arterial dilation, similar signaling pathways are the major cellular component of neurovascular coupling. The new discovered role of Panx in sleep regulation may have broad implications in coordinating neuronal activity and homeostatic housekeeping processes during the sleep-wake cycle.
Collapse
Affiliation(s)
- Valery I Shestopalov
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of MedicineMiami, FL, United States.,Microbiology and Bioengineering Laboratory, Department of Genomics and Biotechnology, Vavilov Institute of General Genetics, Russian Academy of SciencesMoscow, Russia
| | - Yuri Panchin
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Mathematical Methods in Biology, Belozersky Institute, M.V. Lomonosov Moscow State UniversityMoscow, Russia
| | - Olga S Tarasova
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State UniversityMoscow, Russia.,State Research Center of the Russian Federation, Institute for Biomedical Problems, Russian Academy of SciencesMoscow, Russia
| | - Dina Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State UniversityMoscow, Russia.,Department of Physiology, Russian National Research Medical UniversityMoscow, Russia
| | - Vladimir M Kovalzon
- Institute for Information Transmission Problems, Russian Academy of SciencesMoscow, Russia.,Severtsov Institute Ecology and Evolution, Russian Academy of SciencesMoscow, Russia
| |
Collapse
|
19
|
The excitatory/inhibitory input to orexin/hypocretin neuron soma undergoes day/night reorganization. Brain Struct Funct 2017; 222:3847-3859. [PMID: 28669028 DOI: 10.1007/s00429-017-1466-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 06/20/2017] [Indexed: 02/07/2023]
Abstract
Orexin (OX)/hypocretin-containing neurons are main regulators of wakefulness stability, arousal, and energy homeostasis. Their activity varies in relation to the animal's behavioral state. We here tested whether such variation is subserved by synaptic plasticity phenomena in basal conditions. Mice were sacrificed during day or night, at times when sleep or wake, respectively, predominates, as assessed by electroencephalography in matched mice. Triple immunofluorescence was used to visualize OX-A perikarya and varicosities containing the vesicular glutamate transporter (VGluT)2 or the vesicular GABA transporter (VGAT) combined with synaptophysin (Syn) as a presynaptic marker. Appositions on OX-A+ somata were quantitatively analyzed in pairs of sections in epifluorescence and confocal microscopy. The combined total number of glutamatergic (Syn+/VGluT2+) and GABAergic (Syn+/VGAT+) varicosities apposed to OX-A somata was similar during day and night. However, glutamatergic varicosities were significantly more numerous at night, whereas GABAergic varicosities prevailed in the day. Triple immunofluorescence in confocal microscopy was employed to visualize synapse scaffold proteins as postsynaptic markers and confirmed the nighttime prevalence of VGluT2+ together with postsynaptic density protein 95+ excitatory contacts, and daytime prevalence of VGAT+ together with gephyrin+ inhibitory contacts, while also showing that they formed synapses on OX-A+ cell bodies. The findings reveal a daily reorganization of axosomatic synapses in orexinergic neurons, with a switch from a prevalence of excitatory innervation at a time corresponding to wakefulness to a prevalence of inhibitory innervations in the antiphase, at a time corresponding to sleep. This reorganization could represent a key mechanism of plasticity of the orexinergic network in basal conditions.
Collapse
|
20
|
Zielinski MR, Gerashchenko D, Karpova SA, Konanki V, McCarley RW, Sutterwala FS, Strecker RE, Basheer R. The NLRP3 inflammasome modulates sleep and NREM sleep delta power induced by spontaneous wakefulness, sleep deprivation and lipopolysaccharide. Brain Behav Immun 2017; 62:137-150. [PMID: 28109896 PMCID: PMC5373953 DOI: 10.1016/j.bbi.2017.01.012] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/20/2016] [Accepted: 01/17/2017] [Indexed: 11/28/2022] Open
Abstract
Both sleep loss and pathogens can enhance brain inflammation, sleep, and sleep intensity as indicated by electroencephalogram delta (δ) power. The pro-inflammatory cytokine interleukin-1 beta (IL-1β) is increased in the cortex after sleep deprivation (SD) and in response to the Gram-negative bacterial cell-wall component lipopolysaccharide (LPS), although the exact mechanisms governing these effects are unknown. The nucleotide-binding domain and leucine-rich repeat protein-3 (NLRP3) inflammasome protein complex forms in response to changes in the local environment and, in turn, activates caspase-1 to convert IL-1β into its active form. SD enhances the cortical expression of the somnogenic cytokine IL-1β, although the underlying mechanism is, as yet, unidentified. Using NLRP3-gene knockout (KO) mice, we provide evidence that NLRP3 inflammasome activation is a crucial mechanism for the downstream pathway leading to increased IL-1β-enhanced sleep. NLRP3 KO mice exhibited reduced non-rapid eye movement (NREM) sleep during the light period. We also found that sleep amount and intensity (δ activity) were drastically attenuated in NLRP3 KO mice following SD (homeostatic sleep response), as well as after LPS administration, although they were enhanced by central administration of IL-1β. NLRP3, ASC, and IL1β mRNA, IL-1β protein, and caspase-1 activity were greater in the somatosensory cortex at the end of the wake-active period when sleep propensity was high and after SD in wild-type but not NLRP3 KO mice. Thus, our novel and converging findings suggest that the activation of the NLRP3 inflammasome can modulate sleep induced by both increased wakefulness and a bacterial component in the brain.
Collapse
Affiliation(s)
- Mark R Zielinski
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA.
| | - Dmitry Gerashchenko
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA
| | - Svetlana A Karpova
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA
| | - Varun Konanki
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA
| | - Robert W McCarley
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, Brockton, MA 02301, USA
| | - Fayyaz S Sutterwala
- Division of Infectious Diseases, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Robert E Strecker
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA
| | - Radhika Basheer
- Department of Psychiatry, Harvard Medical School and Veterans Affairs Boston Healthcare System, West Roxbury, MA 02132, USA
| |
Collapse
|
21
|
Zhang K, Li YJ, Feng D, Zhang P, Wang YT, Li X, Liu SB, Wu YM, Zhao MG. Imbalance between TNFα and progranulin contributes to memory impairment and anxiety in sleep-deprived mice. Sci Rep 2017; 7:43594. [PMID: 28300056 PMCID: PMC5353617 DOI: 10.1038/srep43594] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/25/2017] [Indexed: 12/21/2022] Open
Abstract
Sleep disorder is becoming a widespread problem in current society, and is associated with impaired cognition and emotional disorders. Progranulin (PGRN), also known as granulin epithelin precursor, promotes neurite outgrowth and cell survival, and is encoded by the GRN gene. It is a tumor necrosis factor α receptor (TNFR) ligand which is implicated in many central nervous system diseases. However, the role PGRN in sleep disorder remains unclear. In the present study, we found that sleep deprivation (S-DEP) impaired the memory and produced thigmotaxis/anxiety-like behaviors in mice. S-DEP increased the levels of TNFα but decreased PGRN levels in the hippocampus. The intracerebroventricular (ICV) injection of PGRN or intraperitoneal injection of TNFα synthesis blocker thalidomide (25 mg/kg), prevented the memory impairment and anxiety behaviors induced by S-DEP. PGRN treatment also restored dendritic spine density in the hippocampus CA1 region and neurogenesis in hippocampus dentate gyrus (DG). These results indicate that an imbalance between TNFα and PGRN contributes to memory impairment and thigmotaxis/anxiety caused by sleep deprivation.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.,Precision Pharmacy &Drug Development Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Yu-Jiao Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Dan Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.,Department of Radiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Peng Zhang
- Department of Neurobiology, Capital Medical University, Beijing, 100069, China
| | - Ya-Tao Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Ming-Gao Zhao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.,Precision Pharmacy &Drug Development Center, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| |
Collapse
|
22
|
Irwin MR, Opp MR. Sleep Health: Reciprocal Regulation of Sleep and Innate Immunity. Neuropsychopharmacology 2017; 42:129-155. [PMID: 27510422 PMCID: PMC5143488 DOI: 10.1038/npp.2016.148] [Citation(s) in RCA: 286] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/29/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
Sleep disturbances including insomnia independently contribute to risk of inflammatory disorders and major depressive disorder. This review and overview provides an integrated understanding of the reciprocal relationships between sleep and the innate immune system and considers the role of sleep in the nocturnal regulation of the inflammatory biology dynamics; the impact of insomnia complaints, extremes of sleep duration, and experimental sleep deprivation on genomic, cellular, and systemic markers of inflammation; and the influence of sleep complaints and insomnia on inflammaging and molecular processes of cellular aging. Clinical implications of this research include discussion of the contribution of sleep disturbance to depression and especially inflammation-related depressive symptoms. Reciprocal action of inflammatory mediators on the homeostatic regulation of sleep continuity and sleep macrostructure, and the potential of interventions that target insomnia to reverse inflammation, are also reviewed. Together, interactions between sleep and inflammatory biology mechanisms underscore the implications of sleep disturbance for inflammatory disease risk, and provide a map to guide the development of treatments that modulate inflammation, improve sleep, and promote sleep health.
Collapse
Affiliation(s)
- Michael R Irwin
- Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience Director and Mindful Awareness Research Center, University of California, Los Angeles, CA, USA
| | - Mark R Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
23
|
Abstract
The reciprocal interaction between the immune system and sleep regulation has been widely acknowledged but the cellular mechanisms that underpin this interaction are not completely understood. In the present study, we investigated the role of macrophages in sleep loss- and cold exposure-induced sleep and body temperature responses. Macrophage apoptosis was induced in mice by systemic injection of clodronate-containing liposomes (CCL). We report that CCL treatment induced an immediate and transient increase in non-rapid-eye movement sleep (NREMS) and fever accompanied by decrease in rapid-eye movement sleep, motor activity and NREMS delta power. Chronically macrophage-depleted mice had attenuated NREMS rebound after sleep deprivation compared to normal mice. Cold-induced increase in wakefulness and decrease in NREMS, rapid-eye movement sleep and body temperature were significantly enhanced in macrophage-depleted mice indicating increased cold sensitivity. These findings provide further evidence for the reciprocal interaction among the immune system, sleep and metabolism, and identify macrophages as one of the key cellular elements in this interplay.
Collapse
|
24
|
Effects of sleep disruption and high fat intake on glucose metabolism in mice. Psychoneuroendocrinology 2016; 68:47-56. [PMID: 26943344 PMCID: PMC4851877 DOI: 10.1016/j.psyneuen.2016.02.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 02/22/2016] [Accepted: 02/22/2016] [Indexed: 11/23/2022]
Abstract
Poor sleep quality or quantity impairs glycemic control and increases risk of disease under chronic conditions. Recovery sleep may offset adverse metabolic outcomes of accumulated sleep debt, but the extent to which this occurs is unclear. We examined whether recovery sleep improves glucose metabolism in mice subjected to prolonged sleep disruption, and whether high fat intake during sleep disruption exacerbates glycemic control. Adult male C57BL/6J mice were subjected to 18-h sleep fragmentation daily for 9 days, followed by 1 day of recovery. During sleep disruption, one group of mice was fed a high-fat diet (HFD) while another group was fed standard laboratory chow. Insulin sensitivity and glucose tolerance were assessed by insulin and glucose tolerance testing at baseline, after 3 and 7 days of sleep disruption, and at the end of the protocol after 24h of undisturbed sleep opportunity (recovery). To characterize changes in sleep architecture that are associated with sleep debt and recovery, we quantified electroencephalogram (EEG) recordings during sleep fragmentation and recovery periods from an additional group of mice. We now report that 9 days of 18-h daily sleep fragmentation significantly reduces rapid eye movement sleep (REMS) and non-rapid eye movement sleep (NREMS). Mice respond with increases in REMS, but not NREMS, during the daily 6-h undisturbed sleep opportunity. However, both REMS and NREMS increase significantly during the 24-h recovery period. Although sleep disruption alone has no effect in this protocol, high fat feeding in combination with sleep disruption impairs glucose tolerance, effects that are reversed by recovery sleep. Insulin sensitivity modestly improves after 3 days of sleep fragmentation and after 24h of recovery, with significantly greater improvements in mice exposed to HFD during sleep disruption. Improvements in both glucose tolerance and insulin sensitivity are associated with NREMS rebound, raising the possibility that this sleep phase contributes to restorative effects of recovery sleep on glycemic control.
Collapse
|
25
|
Thomasy HE, Febinger HY, Ringgold KM, Gemma C, Opp MR. Hypocretinergic and cholinergic contributions to sleep-wake disturbances in a mouse model of traumatic brain injury. Neurobiol Sleep Circadian Rhythms 2016; 2:71-84. [PMID: 31236496 PMCID: PMC6575582 DOI: 10.1016/j.nbscr.2016.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/25/2016] [Accepted: 03/28/2016] [Indexed: 12/24/2022] Open
Abstract
Disorders of sleep and wakefulness occur in the majority of individuals who have experienced traumatic brain injury (TBI), with increased sleep need and excessive daytime sleepiness often reported. Behavioral and pharmacological therapies have limited efficacy, in part, because the etiology of post-TBI sleep disturbances is not well understood. Severity of injuries resulting from head trauma in humans is highly variable, and as a consequence so are their sequelae. Here, we use a controlled laboratory model to investigate the effects of TBI on sleep-wake behavior and on candidate neurotransmitter systems as potential mediators. We focus on hypocretin and melanin-concentrating hormone (MCH), hypothalamic neuropeptides important for regulating sleep and wakefulness, and two potential downstream effectors of hypocretin actions, histamine and acetylcholine. Adult male C57BL/6 mice (n=6-10/group) were implanted with EEG recording electrodes and baseline recordings were obtained. After baseline recordings, controlled cortical impact was used to induce mild or moderate TBI. EEG recordings were obtained from the same animals at 7 and 15 days post-surgery. Separate groups of animals (n=6-8/group) were used to determine effects of TBI on the numbers of hypocretin and MCH-producing neurons in the hypothalamus, histaminergic neurons in the tuberomammillary nucleus, and cholinergic neurons in the basal forebrain. At 15 days post-TBI, wakefulness was decreased and NREM sleep was increased during the dark period in moderately injured animals. There were no differences between groups in REM sleep time, nor were there differences between groups in sleep during the light period. TBI effects on hypocretin and cholinergic neurons were such that more severe injury resulted in fewer cells. Numbers of MCH neurons and histaminergic neurons were not altered under the conditions of this study. Thus, we conclude that moderate TBI in mice reduces wakefulness and increases NREM sleep during the dark period, effects that may be mediated by hypocretin-producing neurons and/or downstream cholinergic effectors in the basal forebrain.
Collapse
Affiliation(s)
- Hannah E Thomasy
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Heidi Y Febinger
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| | - Kristyn M Ringgold
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| | - Carmelina Gemma
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| | - Mark R Opp
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States.,Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
26
|
Ingiosi AM, Opp MR. Sleep and immunomodulatory responses to systemic lipopolysaccharide in mice selectively expressing interleukin-1 receptor 1 on neurons or astrocytes. Glia 2016; 64:780-91. [PMID: 26775112 DOI: 10.1002/glia.22961] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/23/2015] [Accepted: 12/16/2015] [Indexed: 12/22/2022]
Abstract
Sleep-wake behavior is altered in response to immune challenge. Although the precise mechanisms that govern sickness-induced changes in sleep are not fully understood, interleukin-1β (IL-1) is one mediator of these responses. To better understand mechanisms underlying sleep and inflammatory responses to immune challenge, we used two transgenic mouse strains that express IL-1 receptor 1 (IL1R1) only in the central nervous system and selectively on neurons or astrocytes. Electroencephalographic recordings from transgenic and wild-type mice reveal that systemic challenge with lipopolysaccharide (LPS) fragments sleep, suppresses rapid eye movement sleep (REMS), increases non-REMS (NREMS), diminishes NREM delta power, and induces fever in all genotypes. However, the magnitude of REMS suppression is greater in mice expressing IL1R1 on astrocytes compared with mice in which IL1R1 is selectively expressed on neurons. Furthermore, there is a delayed increase in NREM delta power when IL1R1 is expressed on astrocytes. LPS-induced sleep fragmentation is reduced in mice expressing IL1R1 on neurons. Although LPS increases IL-1 and IL-6 in brain of all genotypes, this response is attenuated when IL1R1 is expressed selectively on neurons or on astrocytes. Collectively, these data suggest that in these transgenic mice under the conditions of this study it is neuronal IL1R1 that plays a greater role in LPS-induced suppression of REMS and NREM delta power, whereas astroglial IL1R1 is more important for sleep fragmentation after this immune challenge. Thus, aspects of central responses to LPS are modulated by IL1R1 in a cell type-specific manner.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, Michigan.,Program in Biomedical Sciences, University of Michigan, Ann Arbor, Michigan.,Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington
| | - Mark R Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington.,Graduate Program in Neuroscience, University of Washington, Seattle, Washington
| |
Collapse
|
27
|
Ingiosi AM, Raymond RM, Pavlova MN, Opp MR. Selective contributions of neuronal and astroglial interleukin-1 receptor 1 to the regulation of sleep. Brain Behav Immun 2015; 48:244-57. [PMID: 25849975 DOI: 10.1016/j.bbi.2015.03.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/20/2015] [Accepted: 03/28/2015] [Indexed: 12/26/2022] Open
Abstract
Interactions between sleep and immune function are bidirectional. Although the mechanisms that govern these interactions are not fully elucidated, the pro-inflammatory cytokine, interleukin-1β (IL-1), is a known regulator of sleep and mediator of immune responses. To further clarify the underlying substrates of sleep and immune interactions, we engineered two transgenic mouse lines that express interleukin-1 receptor 1 (IL1R1) only in the central nervous system (CNS) and selectively on neurons (NSE-IL1R1) or astrocytes (GFAP-IL1R1). During spontaneous sleep, compared to wild type (WT) animals, NSE-IL1R1 and GFAP-IL1R1 mice have more rapid eye movement sleep (REMS) that is characterized by reduced theta power in the electroencephalogram (EEG) spectra. The non-REM sleep (NREMS) EEG of each of the IL1R1 transgenic mouse strains also is characterized by enhanced power in the delta frequency band. In response to 6h of sleep deprivation, sleep of both IL1R1 transgenic mouse strains is more consolidated than that of WT animals. Additionally, the NREMS EEG of NSE-IL1R1 mice contains less delta power after sleep deprivation, suggesting astroglial IL1R1 activity may modulate sleep homeostasis. Intracerebroventricular injection of IL-1 fails to alter sleep or brain temperature of NSE-IL1R1 or GFAP-IL1R1 mice. These data suggest that selective IL1R1 expression on neurons or on astrocytes is not sufficient for centrally-administered IL-1 to induce sleep or fever. Lack of sleep and febrile responses to IL-1 in these IL1R1 transgenic mouse strains may be due to their inability to produce IL-6 in brain. Overall, these studies demonstrate, through the use of novel transgenic mice, that IL1R1 on neurons and astrocytes differentially mediates aspects of sleep under physiological conditions and in response to central IL-1 administration.
Collapse
Affiliation(s)
- Ashley M Ingiosi
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, United States; Program in Biomedical Sciences, University of Michigan, Ann Arbor, MI, United States; Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States
| | - Richard M Raymond
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States
| | - Maria N Pavlova
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States
| | - Mark R Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, United States; Program of Neurobiology and Behavior, University of Washington, Seattle, WA, United States.
| |
Collapse
|
28
|
Haghighi F, Ge Y, Chen S, Xin Y, Umali MU, De Gasperi R, Gama Sosa MA, Ahlers ST, Elder GA. Neuronal DNA Methylation Profiling of Blast-Related Traumatic Brain Injury. J Neurotrauma 2015; 32:1200-9. [PMID: 25594545 DOI: 10.1089/neu.2014.3640] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Long-term molecular changes in the brain resulting from blast exposure may be mediated by epigenetic changes, such as deoxyribonucleic acid (DNA) methylation, that regulate gene expression. Aberrant regulation of gene expression is associated with behavioral abnormalities, where DNA methylation bridges environmental signals to sustained changes in gene expression. We assessed DNA methylation changes in the brains of rats exposed to three 74.5 kPa blast overpressure events, conditions that have been associated with long-term anxiogenic manifestations weeks or months following the initial exposures. Rat frontal cortex eight months post-exposure was used for cell sorting of whole brain tissue into neurons and glia. We interrogated DNA methylation profiles in these cells using Expanded Reduced Representation Bisulfite Sequencing. We obtained data for millions of cytosines, showing distinct methylation profiles for neurons and glia and an increase in global methylation in neuronal versus glial cells (p<10(-7)). We detected DNA methylation perturbations in blast overpressure-exposed animals, compared with sham blast controls, within 458 and 379 genes in neurons and glia, respectively. Differentially methylated neuronal genes showed enrichment in cell death and survival and nervous system development and function, including genes involved in transforming growth factor β and nitric oxide signaling. Functional validation via gene expression analysis of 30 differentially methylated neuronal and glial genes showed a 1.2 fold change in gene expression of the serotonin N-acetyltransferase gene (Aanat) in blast animals (p<0.05). These data provide the first genome-based evidence for changes in DNA methylation induced in response to multiple blast overpressure exposures. In particular, increased methylation and decreased gene expression were observed in the Aanat gene, which is involved in converting serotonin to the circadian hormone melatonin and is implicated in sleep disturbance and depression associated with traumatic brain injury.
Collapse
Affiliation(s)
- Fatemeh Haghighi
- 1 Department of Psychiatry, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Yongchao Ge
- 4 Department of Neurology, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Sean Chen
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Yurong Xin
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Michelle U Umali
- 2 Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Rita De Gasperi
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- 5 Department of Psychiatry, Icahn School of Medicine at Mount Sinai , New York, New York
- 6 Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
| | - Miguel A Gama Sosa
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- 5 Department of Psychiatry, Icahn School of Medicine at Mount Sinai , New York, New York
- 6 Research and Development Service, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
| | - Stephen T Ahlers
- 7 Department of Neurotrauma, Operational and Undersea Medicine Directorate Naval Medical Research Center , Silver Spring, Maryland
| | - Gregory A Elder
- 3 Friedman Brain Institute, Icahn School of Medicine at Mount Sinai , New York, New York
- 4 Department of Neurology, Icahn School of Medicine at Mount Sinai , New York, New York
- 5 Department of Psychiatry, Icahn School of Medicine at Mount Sinai , New York, New York
- 8 Neurology Service, James J. Peters Department of Veterans Affairs Medical Center , Bronx, New York
| |
Collapse
|
29
|
Bouybayoune I, Mantovani S, Del Gallo F, Bertani I, Restelli E, Comerio L, Tapella L, Baracchi F, Fernández-Borges N, Mangieri M, Bisighini C, Beznoussenko GV, Paladini A, Balducci C, Micotti E, Forloni G, Castilla J, Fiordaliso F, Tagliavini F, Imeri L, Chiesa R. Transgenic fatal familial insomnia mice indicate prion infectivity-independent mechanisms of pathogenesis and phenotypic expression of disease. PLoS Pathog 2015; 11:e1004796. [PMID: 25880443 PMCID: PMC4400166 DOI: 10.1371/journal.ppat.1004796] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/09/2015] [Indexed: 11/18/2022] Open
Abstract
Fatal familial insomnia (FFI) and a genetic form of Creutzfeldt-Jakob disease (CJD178) are clinically different prion disorders linked to the D178N prion protein (PrP) mutation. The disease phenotype is determined by the 129 M/V polymorphism on the mutant allele, which is thought to influence D178N PrP misfolding, leading to the formation of distinctive prion strains with specific neurotoxic properties. However, the mechanism by which misfolded variants of mutant PrP cause different diseases is not known. We generated transgenic (Tg) mice expressing the mouse PrP homolog of the FFI mutation. These mice synthesize a misfolded form of mutant PrP in their brains and develop a neurological illness with severe sleep disruption, highly reminiscent of FFI and different from that of analogously generated Tg(CJD) mice modeling CJD178. No prion infectivity was detectable in Tg(FFI) and Tg(CJD) brains by bioassay or protein misfolding cyclic amplification, indicating that mutant PrP has disease-encoding properties that do not depend on its ability to propagate its misfolded conformation. Tg(FFI) and Tg(CJD) neurons have different patterns of intracellular PrP accumulation associated with distinct morphological abnormalities of the endoplasmic reticulum and Golgi, suggesting that mutation-specific alterations of secretory transport may contribute to the disease phenotype. Genetic prion diseases are degenerative brain disorders caused by mutations in the gene encoding the prion protein (PrP). Different PrP mutations cause different diseases, including Creutzfeldt-Jakob disease (CJD) and fatal familial insomnia (FFI). The reason for this variability is not known, but assembly of the mutant PrPs into distinct aggregates that spread in the brain by promoting PrP aggregation may contribute to the disease phenotype. We previously generated transgenic mice modeling genetic CJD, clinically identified by dementia and motor abnormalities. We have now generated transgenic mice carrying the PrP mutation associated with FFI, and found that they develop severe sleep abnormalities and other key features of the human disorder. Thus, transgenic mice recapitulate the phenotypic differences seen in humans. The mutant PrPs in FFI and CJD mice are aggregated but unable to promote PrP aggregation. They accumulate in different intracellular compartments and cause distinct morphological abnormalities of transport organelles. These results indicate that mutant PrP has disease-encoding properties that are independent of its ability to self-propagate, and suggest that the phenotypic heterogeneity may be due to different effects of aggregated PrP on intracellular transport. Our study provides new insights into the mechanisms of selective neuronal dysfunction due to protein aggregation.
Collapse
Affiliation(s)
- Ihssane Bouybayoune
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Susanna Mantovani
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Federico Del Gallo
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | - Ilaria Bertani
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Elena Restelli
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Liliana Comerio
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Laura Tapella
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Francesca Baracchi
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | | | - Michela Mangieri
- Division of Neuropathology and Neurology, IRCCS Foundation “Carlo Besta” National Neurological Institute, Milan, Italy
| | - Cinzia Bisighini
- Bio-Imaging Unit, Department of Cardiovascular Research, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | | | - Alessandra Paladini
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Claudia Balducci
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Edoardo Micotti
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Gianluigi Forloni
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Joaquín Castilla
- CIC bioGUNE, Parque Tecnológico de Bizkaia, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Fabio Fiordaliso
- Bio-Imaging Unit, Department of Cardiovascular Research, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
| | - Fabrizio Tagliavini
- Division of Neuropathology and Neurology, IRCCS Foundation “Carlo Besta” National Neurological Institute, Milan, Italy
| | - Luca Imeri
- Department of Health Sciences, University of Milan Medical School, Milan, Italy
| | - Roberto Chiesa
- Department of Neuroscience, IRCCS—“Mario Negri” Institute for Pharmacological Research, Milan, Italy
- * E-mail:
| |
Collapse
|
30
|
Dumaine JE, Ashley NT. Acute sleep fragmentation induces tissue-specific changes in cytokine gene expression and increases serum corticosterone concentration. Am J Physiol Regul Integr Comp Physiol 2015; 308:R1062-9. [PMID: 25876653 DOI: 10.1152/ajpregu.00049.2015] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 04/10/2015] [Indexed: 12/13/2022]
Abstract
Sleep deprivation induces acute inflammation and increased glucocorticosteroids in vertebrates, but effects from fragmented, or intermittent, sleep are poorly understood. Considering the latter is more representative of sleep apnea in humans, we investigated changes in proinflammatory (IL-1β, TNF-α) and anti-inflammatory (TGF-β1) cytokine gene expression in the periphery (liver, spleen, fat, and heart) and brain (hypothalamus, prefrontal cortex, and hippocampus) of a murine model exposed to varying intensities of sleep fragmentation (SF). Additionally, serum corticosterone was assessed. Sleep was disrupted in male C57BL/6J mice using an automated sleep fragmentation chamber that moves a sweeping bar at specified intervals (Lafayette Industries). Mice were exposed to bar sweeps every 20 s (high sleep fragmentation, HSF), 120 s (low sleep fragmentation, LSF), or the bar remained stationary (control). Trunk blood and tissue samples were collected after 24 h of SF. We predicted that HSF mice would exhibit increased proinflammatory expression, decreased anti-inflammatory expression, and elevated stress hormones in relation to LSF and controls. SF significantly elevated IL-1β gene expression in adipose tissue, heart (HSF only), and hypothalamus (LSF only) relative to controls. SF did not increase TNF-α expression in any of the tissues measured. HSF increased TGF-β1 expression in the hypothalamus and hippocampus relative to other groups. Serum corticosterone concentration was significantly different among groups, with HSF mice exhibiting the highest, LSF intermediate, and controls with the lowest concentration. This indicates that 24 h of SF is a potent inducer of inflammation and stress hormones in the periphery, but leads to upregulation of anti-inflammatory cytokines in the brain.
Collapse
Affiliation(s)
- Jennifer E Dumaine
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| | - Noah T Ashley
- Department of Biology, Western Kentucky University, Bowling Green, Kentucky
| |
Collapse
|
31
|
Albuquerque RG, Okazaki KM, Hirotsu C, Tomimori J, Tufik S, Andersen ML. Sleep, Hansen's disease and the immune system--a not so harmonic triad. Med Hypotheses 2015; 84:456-9. [PMID: 25686506 DOI: 10.1016/j.mehy.2015.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 01/27/2015] [Accepted: 01/30/2015] [Indexed: 11/26/2022]
Abstract
Hansen's disease is one of the oldest skin diseases in the world characterized by a spectrum of clinical manifestations that are associated with stigmatization and poor quality of life. It is also considered a model disease for investigating the human immune system because of its association with immune reactions, which are thought to be a reflection of the host's immunological response, promoting intense cellular activity or humoral secretion. This relationship between the cellular and microbial components of skin and their regulation by local immune responses may be modulated by a currently neglected behavior: sleep. Recent studies have demonstrated that sleep deprivation may aggravate the progression of chronic dermatological diseases, which in turn can lead to a non-restorative sleep pattern. Indeed, sleep is essential for immune and skin integrity. Thus, we propose here a hypothesis linking Hansen's disease, sleep and immunity in a bidirectional relationship. Hansen's disease patients may demonstrate a worse sleep quality than the general population through the modulation of immunological environment; and sleep restriction, a hallmark of modern society, being a possible predictor of the disease progression.
Collapse
Affiliation(s)
- Rachel Gimenes Albuquerque
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| | - Keity Mey Okazaki
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| | - Camila Hirotsu
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, Vila Clementino, 04024-002 São Paulo, SP, Brazil.
| | - Jane Tomimori
- Department of Dermatology, Universidade Federal de São Paulo, Rua Borges Lagoa, 508, Vila Clementino, 04038-001 São Paulo, Brazil
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| | - Monica Levy Andersen
- Department of Psychobiology, Universidade Federal de São Paulo, Rua Napoleão de Barros, 925, Vila Clementino, 04024-002 São Paulo, SP, Brazil
| |
Collapse
|
32
|
Manzar MD, Zannat W, Hussain ME. Sleep and physiological systems: a functional perspective. BIOL RHYTHM RES 2014. [DOI: 10.1080/09291016.2014.966504] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
33
|
Greenwood BN, Thompson RS, Opp MR, Fleshner M. Repeated exposure to conditioned fear stress increases anxiety and delays sleep recovery following exposure to an acute traumatic stressor. Front Psychiatry 2014; 5:146. [PMID: 25368585 PMCID: PMC4202708 DOI: 10.3389/fpsyt.2014.00146] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 09/30/2014] [Indexed: 01/29/2023] Open
Abstract
Repeated stressor exposure can sensitize physiological responses to novel stressors and facilitate the development of stress-related psychiatric disorders including anxiety. Disruptions in diurnal rhythms of sleep-wake behavior accompany stress-related psychiatric disorders and could contribute to their development. Complex stressors that include fear-eliciting stimuli can be a component of repeated stress experienced by human beings, but whether exposure to repeated fear can prime the development of anxiety and sleep disturbances is unknown. In the current study, adult male F344 rats were exposed to either control conditions or repeated contextual fear conditioning for 22 days followed by exposure to no, mild (10), or severe (100) acute uncontrollable tail shock stress. Exposure to acute stress produced anxiety-like behavior as measured by a reduction in juvenile social exploration and exaggerated shock-elicited freezing in a novel context. Prior exposure to repeated fear enhanced anxiety-like behavior as measured by shock-elicited freezing, but did not alter social exploratory behavior. The potentiation of anxiety produced by prior repeated fear was temporary; exaggerated fear was present 1 day but not 4 days following acute stress. Interestingly, exposure to acute stress reduced rapid eye movement (REM) and non-REM (NREM) sleep during the hours immediately following acute stress. This initial reduction in sleep was followed by robust REM rebound and diurnal rhythm flattening of sleep/wake behavior. Prior repeated fear extended the acute stress-induced REM and NREM sleep loss, impaired REM rebound, and prolonged the flattening of the diurnal rhythm of NREM sleep following acute stressor exposure. These data suggest that impaired recovery of sleep/wake behavior following acute stress could contribute to the mechanisms by which a history of prior repeated stress increases vulnerability to subsequent novel stressors and stress-related disorders.
Collapse
Affiliation(s)
| | - Robert S. Thompson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Mark R. Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Monika Fleshner
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
34
|
Vanini G, Nemanis K, Baghdoyan HA, Lydic R. GABAergic transmission in rat pontine reticular formation regulates the induction phase of anesthesia and modulates hyperalgesia caused by sleep deprivation. Eur J Neurosci 2014; 40:2264-73. [PMID: 24674578 DOI: 10.1111/ejn.12571] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 01/03/2023]
Abstract
The oral part of the pontine reticular formation (PnO) contributes to the regulation of sleep, anesthesia and pain. The role of PnO γ-aminobutyric acid (GABA) in modulating these states remains incompletely understood. The present study used time to loss and time to resumption of righting response (LoRR and RoRR) as surrogate measures of loss and resumption of consciousness. This study tested three hypotheses: (i) pharmacologically manipulating GABA levels in rat PnO alters LoRR, RoRR and nociception; (ii) propofol decreases GABA levels in the PnO; and (iii) inhibiting GABA synthesis in the PnO blocks hyperalgesia caused by sleep deprivation. Administering a GABA synthesis inhibitor [3-mercaptopropionic acid (3-MPA)] or a GABA uptake inhibitor [nipecotic acid (NPA)] into rat PnO significantly altered LoRR caused by propofol. 3-MPA significantly decreased LoRR for propofol (-18%). NPA significantly increased LoRR during administration of propofol (36%). Neither 3-MPA nor NPA altered RoRR following cessation of propofol or isoflurane delivery. The finding that LoRR was decreased by 3-MPA and increased by NPA is consistent with measures showing that extracellular GABA levels in the PnO were decreased (41%) by propofol. Thermal nociception was significantly decreased by 3-MPA and increased by NPA, and 3-MPA blocked the hyperalgesia caused by sleep deprivation. The results demonstrate that GABA levels in the PnO regulate the time for loss of consciousness caused by propofol, extend the concept that anesthetic induction and emergence are not inverse processes, and suggest that GABAergic transmission in the PnO mediates hyperalgesia caused by sleep loss.
Collapse
Affiliation(s)
- Giancarlo Vanini
- Department of Anesthesiology, University of Michigan, 7433 Medical Science Building I, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5615, USA
| | | | | | | |
Collapse
|
35
|
Rachalski A, Freyburger M, Mongrain V. Contribution of transcriptional and translational mechanisms to the recovery aspect of sleep regulation. Ann Med 2014; 46:62-72. [PMID: 24428734 DOI: 10.3109/07853890.2013.866439] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sleep parallels brain functioning and mental health. Neuronal activity during wakefulness leads to a subsequent increase in sleep intensity as measured using electroencephalographic slow-wave activity (SWA; index of neuronal synchrony in the low-frequency range). Wakefulness, and particularly prolonged wakefulness, also drives important changes in brain gene expression and changes in protein regulation. The role of these two cellular mechanisms in sleep-wake regulation has typically been studied independently, and their exact contribution to SWA remains poorly defined. In this review, we highlight that many transcriptional pathways driven by sleep deprivation are associated to protein regulation. We first describe the relationship between cytokines, clock genes, and markers of sleep need with an emphasis on transcriptional processes. Observations regarding the role of protein metabolism in sleep-wake regulation are then depicted while presenting interconnections between transcriptional and translational responses driven by sleep loss. Lastly, a manner by which this integrated response can feed back on neuronal network activity to determine sleep intensity is proposed. Overall, the literature supports that a complex cross-talk between transcriptional and translational regulation during prolonged wakefulness drives the changes in sleep intensity as a function of the sleep/wake history.
Collapse
Affiliation(s)
- Adeline Rachalski
- Center for Advanced Research in Sleep Medicine and Research Center, Hôpital du Sacré-Coeur de Montréal , Montréal, QC , Canada
| | | | | |
Collapse
|
36
|
Trammell RA, Verhulst S, Toth LA. Effects of sleep fragmentation on sleep and markers of inflammation in mice. Comp Med 2014; 64:13-24. [PMID: 24512957 PMCID: PMC3929215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 05/15/2013] [Accepted: 09/11/2013] [Indexed: 06/03/2023]
Abstract
Many people in our society experience curtailment and disruption of sleep due to work responsibilities, care-giving, or life style choice. Delineating the health effect of acute and chronic disruptions in sleep is essential to raising awareness of and creating interventions to manage these prevalent concerns. To provide a platform for studying the health impact and underlying pathophysiologic mechanisms associated with inadequate sleep, we developed and characterized an approach to creating chronic disruption of sleep in laboratory mice. We used this method to evaluate how 3 durations of sleep fragmentation (SF) affect sleep recuperation and blood and lung analyte concentrations in male C57BL/6J mice. Mice housed in environmentally controlled chambers were exposed to automated SF for periods of 6, 12, or 24 h or for 12 h daily during the light (somnolent) phase for 4 sequential days. Sleep time, slow-wave amplitude, or bout lengths were significantly higher when uninterrupted sleep was permitted after each of the 3 SF durations. However, mice did not recover all of the lost slow-wave sleep during the subsequent 12- to 24-h period and maintained a net loss of sleep. Light-phase SF was associated with significant changes in serum and lung levels of some inflammatory substances, but these changes were not consistent or sustained. The data indicate that acute light-phase SF can result in a sustained sleep debt in mice and may disrupt the inflammatory steady-state in serum and lung.
Collapse
Key Words
- dwa, δ wave amplitude
- de, disk environment
- e, time of euthanasia
- g-csf, granulocyte colony-stimulating factor
- hc, home cage
- hpa, hypothalamic–pituitary–adrenal
- ip10, interferon-γ-induced protein 10 (cxcl10)
- kc, keratinocyte-derived chemokine (cxcl1)
- lcn2, lipocalin 2
- mcp1, monocyte chemotactic protein 1 (ccl2)
- m-csf, macrophage colony-stimulating factor
- mip1α, macrophage inflammatory protein
- nrems, non-rapid-eye-movement sleep
- rems, rapid-eye-movement sleep
- sf, sleep fragmentation
- smet, simple main-effects test
- sws, slow-wave sleep
- tpai1, total plasminogen activator inhibitor 1
Collapse
Affiliation(s)
- Rita A Trammell
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Steve Verhulst
- Department of Statistics and Research Informatics, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Linda A Toth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| |
Collapse
|
37
|
Febinger HY, George A, Priestley J, Toth LA, Opp MR. Effects of housing condition and cage change on characteristics of sleep in mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2014; 53:29-37. [PMID: 24411777 PMCID: PMC3894645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 05/17/2013] [Accepted: 08/02/2013] [Indexed: 06/03/2023]
Abstract
Although human subjects are widely used to study sleep and sleep disorders, animals have been invaluable in developing our understanding of the physiology of sleep and underlying mechanisms of sleep disorders. Environmental stimuli are likely to modify sleep in both animals and people, suggesting that environmental stability must be controlled carefully by both husbandry and research staff to allow collection of valid results with minimal numbers of animals. However, few studies have measured the effects of cage condition on sleep parameters in mice. Current guidelines recommend social housing and environmental enrichment for standard rodent housing. Environmental factors such as these create potential confounds in studies for which facets of sleep are outcome measures. We therefore sought to determine whether cage changes, group housing, or single housing with a shelter altered measures of sleep in C57BL/6J mice. The resulting data indicate that 1) cage changing disrupts sleep for approximately 3 h; 2) group housing is associated with shorter bouts of rapid-eye-movement sleep (REMS) and less slow-wave sleep (SWS) during the light phase and with more REMS during the dark phase; and 3) mice housed with a shelter spend less time awake and more time in SWS, with longer bouts of SWS during the dark phase. In additional, both group housing and housing with a shelter were associated with less locomotor activity than occurred in individually housed mice without a shelter. These findings provide evidence for long-held beliefs that housing conditions must be controlled carefully in studies that require assessment of sleep.
Collapse
Affiliation(s)
- Heidi Y Febinger
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, USA
| | - Amrita George
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, USA
| | - Jill Priestley
- Department of Anesthesiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Linda A Toth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Mark R Opp
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
38
|
A neocortical delta rhythm facilitates reciprocal interlaminar interactions via nested theta rhythms. J Neurosci 2013; 33:10750-61. [PMID: 23804097 DOI: 10.1523/jneurosci.0735-13.2013] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Delta oscillations (1-4 Hz) associate with deep sleep and are implicated in memory consolidation and replay of cortical responses elicited during wake states. A potent local generator has been characterized in thalamus, and local generators in neocortex have been suggested. Here we demonstrate that isolated rat neocortex generates delta rhythms in conditions mimicking the neuromodulatory state during deep sleep (low cholinergic and dopaminergic tone). The rhythm originated in an NMDA receptor-driven network of intrinsic bursting (IB) neurons in layer 5, activating a source of GABAB receptor-mediated inhibition. In contrast, regular spiking (RS) neurons in layer 5 generated theta-frequency outputs. In layer 2/3 principal cells, outputs from IB cells associated with IPSPs, whereas those from layer 5 RS neurons related to nested bursts of theta-frequency EPSPs. Both interlaminar spike and field correlations revealed a sequence of events whereby sparse spiking in layer 2/3 was partially reflected back from layer 5 on each delta period. We suggest that these reciprocal, interlaminar interactions may represent a "Helmholtz machine"-like process to control synaptic rescaling during deep sleep.
Collapse
|
39
|
Prolonged sleep fragmentation of mice exacerbates febrile responses to lipopolysaccharide. J Neurosci Methods 2013; 219:104-12. [PMID: 23872243 DOI: 10.1016/j.jneumeth.2013.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/04/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sleep disruption is a frequent occurrence in modern society. Whereas many studies have focused on the consequences of total sleep deprivation, few have investigated the condition of sleep disruption. NEW METHOD We disrupted sleep of mice during the light period for 9 consecutive days using an intermittently rotating disc. RESULTS Electroencephalogram (EEG) data demonstrated that non-rapid eye movement (NREM) sleep was severely fragmented and REM sleep was essentially abolished during the 12h light period. During the dark period, when sleep was not disrupted, neither NREM sleep nor REM sleep times differed from control values. Analysis of the EEG revealed a trend for increased power in the peak frequency of the NREM EEG spectra during the dark period. The fragmentation protocol was not overly stressful as body weights and water consumption remained unchanged, and plasma corticosterone did not differ between mice subjected to 3 or 9 days of sleep disruption and home cage controls. However, mice subjected to 9 days of sleep disruption by this method responded to lipopolysaccharide with an exacerbated febrile response. COMPARISON WITH EXISTING METHODS Existing methods to disrupt sleep of laboratory rodents often subject the animal to excessive locomotion, vibration, or sudden movements. This method does not suffer from any of these confounds. CONCLUSIONS This study demonstrates that prolonged sleep disruption of mice exacerbates febrile responses to lipopolysaccharide. This device provides a method to determine mechanisms by which chronic insufficient sleep contributes to the etiology of many pathologies, particularly those with an inflammatory component.
Collapse
|
40
|
Hypnotic hypersensitivity to volatile anesthetics and dexmedetomidine in dopamine β-hydroxylase knockout mice. Anesthesiology 2013; 117:1006-17. [PMID: 23042227 DOI: 10.1097/aln.0b013e3182700ab9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Multiple lines of evidence suggest that the adrenergic system can modulate sensitivity to anesthetic-induced immobility and anesthetic-induced hypnosis as well. However, several considerations prevent the conclusion that the endogenous adrenergic ligands norepinephrine and epinephrine alter anesthetic sensitivity. METHODS Using dopamine β-hydroxylase knockout (Dbh) mice genetically engineered to lack the adrenergic ligands and their siblings with normal adrenergic levels, we test the contribution of the adrenergic ligands upon volatile anesthetic induction and emergence. Moreover, we investigate the effects of intravenous dexmedetomidine in adrenergic-deficient mice and their siblings using both righting reflex and processed electroencephalographic measures of anesthetic hypnosis. RESULTS We demonstrate that the loss of norepinephrine and epinephrine and not other neuromodulators co-packaged in adrenergic neurons is sufficient to cause hypersensitivity to induction of volatile anesthesia. However, the most profound effect of adrenergic deficiency is retarding emergence from anesthesia, which takes two to three times as long in Dbh mice for sevoflurane, isoflurane, and halothane. Having shown that Dbh mice are hypersensitive to volatile anesthetics, we further demonstrate that their hypnotic hypersensitivity persists at multiple doses of dexmedetomidine. Dbh mice exhibit up to 67% shorter latencies to loss of righting reflex and up to 545% longer durations of dexmedetomidine-induced general anesthesia. Central rescue of adrenergic signaling restores control-like dexmedetomidine sensitivity. A novel continuous electroencephalographic analysis illustrates that the longer duration of dexmedetomidine-induced hypnosis is not due to a motor confound, but occurs because of impaired anesthetic emergence. CONCLUSIONS Adrenergic signaling is essential for normal emergence from general anesthesia. Dexmedetomidine-induced general anesthesia does not depend on inhibition of adrenergic neurotransmission.
Collapse
|
41
|
Toth LA, Bhargava P. Animal models of sleep disorders. Comp Med 2013; 63:91-104. [PMID: 23582416 PMCID: PMC3625050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 10/21/2012] [Accepted: 11/25/2012] [Indexed: 06/02/2023]
Abstract
Problems with sleep affect a large part of the general population, with more than half of all people in the United States reporting difficulties with sleep or insufficient sleep at various times and about 40 million affected chronically. Sleep is a complex physiologic process that is influenced by many internal and environmental factors, and problems with sleep are often related to specific personal circumstances or are based on subjective reports from the affected person. Although human subjects are used widely in the study of sleep and sleep disorders, the study of animals has been invaluable in developing our understanding about the physiology of sleep and the underlying mechanisms of sleep disorders. Historically, the use of animals for the study of sleep disorders has arguably been most fruitful for the condition of narcolepsy, in which studies of dogs and mice revealed previously unsuspected mechanisms for this condition. The current overview considers animal models that have been used to study 4 of the most common human sleep disorders-insomnia, narcolepsy, restless legs syndrome, and sleep apnea-and summarizes considerations relevant to the use of animals for the study of sleep and sleep disorders. Animal-based research has been vital to the elucidation of mechanisms that underlie sleep, its regulation, and its disorders and undoubtedly will remain crucial for discovering and validating sleep mechanisms and testing interventions for sleep disorders.
Collapse
Affiliation(s)
- Linda A Toth
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois, USA.
| | | |
Collapse
|
42
|
Frank MG. Astroglial regulation of sleep homeostasis. Curr Opin Neurobiol 2013; 23:812-8. [PMID: 23518138 DOI: 10.1016/j.conb.2013.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 02/20/2013] [Accepted: 02/21/2013] [Indexed: 12/12/2022]
Abstract
Mammalian sleep is regulated by two distinct mechanisms. A circadian oscillator provides timing signals that organize sleep and wake across the 24 hour day. A homeostatic mechanism increases sleep drive and sleep amounts (or intensity) as a function of prior time awake. The cellular mechanisms of sleep homeostasis are poorly defined, but are thought to be primarily neuronal. According to one view, sleep homeostasis arises from interactions between subcortical neurons that register sleep pressure and other neurons that promote either sleep or wakefulness. Alternatively, sleep drive may arise independently among neurons throughout the brain in a use-dependent fashion. Implicit in both views is the idea that sleep homeostasis is solely the product of neurons. In this article, I discuss an emerging view that glial astrocytes may play an essential role in sleep homeostasis.
Collapse
Affiliation(s)
- Marcos G Frank
- University of Pennsylvania, Perelman School of Medicine, Department of Neuroscience, 215 Stemmler Hall, 35th & Hamilton Walk, Philadelphia, PA 19104-6074, United States.
| |
Collapse
|
43
|
Gast H, Müller A, Lopez M, Meier D, Huber R, Dechent F, Prinz M, Emmenegger Y, Franken P, Birchler T, Fontana A. CD40 activation induces NREM sleep and modulates genes associated with sleep homeostasis. Brain Behav Immun 2013; 27:133-44. [PMID: 23072727 DOI: 10.1016/j.bbi.2012.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 10/02/2012] [Accepted: 10/02/2012] [Indexed: 01/26/2023] Open
Abstract
The T-cell derived cytokine CD40 ligand is overexpressed in patients with autoimmune diseases. Through activation of its receptor, CD40 ligand leads to a tumor necrosis factor (TNF) receptor 1 (TNFR1) dependent impairment of locomotor activity in mice. Here we report that this effect is explained through a promotion of sleep, which was specific to non-rapid eye movement (NREM) sleep while REM sleep was suppressed. The increase in NREM sleep was accompanied by a decrease in EEG delta power during NREM sleep and by a decrease in the expression of transcripts in the cerebral cortex known to be associated with homeostatic sleep drive, such as Homer1a, Early growth response 2, Neuronal pentraxin 2, and Fos-like antigen 2. The effect of CD40 activation was mimicked by peripheral TNF injection and prevented by the TNF blocker etanercept. Our study indicates that sleep-wake dysregulation in autoimmune diseases may result from CD40 induced TNF:TNFR1 mediated alterations of molecular pathways, which regulate sleep-wake behavior.
Collapse
Affiliation(s)
- Heidemarie Gast
- Department of Neurology, Inselspital, University Hospital Berne, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
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
|
45
|
Jakubcakova V, Flachskamm C, Landgraf R, Kimura M. Sleep phenotyping in a mouse model of extreme trait anxiety. PLoS One 2012; 7:e40625. [PMID: 22808211 PMCID: PMC3394752 DOI: 10.1371/journal.pone.0040625] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 06/11/2012] [Indexed: 01/19/2023] Open
Abstract
Background There is accumulating evidence that anxiety impairs sleep. However, due to high sleep variability in anxiety disorders, it has been difficult to state particular changes in sleep parameters caused by anxiety. Sleep profiling in an animal model with extremely high vs. low levels of trait anxiety might serve to further define sleep patterns associated with this psychopathology. Methodology/Principal Findings Sleep-wake behavior in mouse lines with high (HAB), low (LAB) and normal (NAB) anxiety-related behaviors was monitored for 24 h during baseline and recovery after 6 h sleep deprivation (SD). The amounts of each vigilance state, sleep architecture, and EEG spectral variations were compared between the mouse lines. In comparison to NAB mice, HAB mice slept more and exhibited consistently increased delta power during non-rapid eye movement (NREM) sleep. Their sleep patterns were characterized by heavy fragmentation, reduced maintenance of wakefulness, and frequent intrusions of rapid eye movement (REM) sleep. In contrast, LAB mice showed a robust sleep-wake rhythm with remarkably prolonged sleep latency and a long, persistent period of wakefulness. In addition, the accumulation of delta power after SD was impaired in the LAB line, as compared to HAB mice. Conclusions/Significance Sleep-wake patterns were significantly different between HAB and LAB mice, indicating that the genetic predisposition to extremes in trait anxiety leaves a biological scar on sleep quality. The enhanced sleep demand observed in HAB mice, with a strong drive toward REM sleep, may resemble a unique phenotype reflecting not only elevated anxiety but also a depression-like attribute.
Collapse
Affiliation(s)
| | | | | | - Mayumi Kimura
- Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail:
| |
Collapse
|
46
|
Abstract
This review summarizes the brain mechanisms controlling sleep and wakefulness. Wakefulness promoting systems cause low-voltage, fast activity in the electroencephalogram (EEG). Multiple interacting neurotransmitter systems in the brain stem, hypothalamus, and basal forebrain converge onto common effector systems in the thalamus and cortex. Sleep results from the inhibition of wake-promoting systems by homeostatic sleep factors such as adenosine and nitric oxide and GABAergic neurons in the preoptic area of the hypothalamus, resulting in large-amplitude, slow EEG oscillations. Local, activity-dependent factors modulate the amplitude and frequency of cortical slow oscillations. Non-rapid-eye-movement (NREM) sleep results in conservation of brain energy and facilitates memory consolidation through the modulation of synaptic weights. Rapid-eye-movement (REM) sleep results from the interaction of brain stem cholinergic, aminergic, and GABAergic neurons which control the activity of glutamatergic reticular formation neurons leading to REM sleep phenomena such as muscle atonia, REMs, dreaming, and cortical activation. Strong activation of limbic regions during REM sleep suggests a role in regulation of emotion. Genetic studies suggest that brain mechanisms controlling waking and NREM sleep are strongly conserved throughout evolution, underscoring their enormous importance for brain function. Sleep disruption interferes with the normal restorative functions of NREM and REM sleep, resulting in disruptions of breathing and cardiovascular function, changes in emotional reactivity, and cognitive impairments in attention, memory, and decision making.
Collapse
Affiliation(s)
- Ritchie E Brown
- Laboratory of Neuroscience, VA Boston Healthcare System and Harvard Medical School, Brockton, Massachusetts 02301, USA
| | | | | | | | | |
Collapse
|
47
|
|
48
|
Disrupted sleep without sleep curtailment induces sleepiness and cognitive dysfunction via the tumor necrosis factor-α pathway. J Neuroinflammation 2012; 9:91. [PMID: 22578011 PMCID: PMC3411474 DOI: 10.1186/1742-2094-9-91] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 05/11/2012] [Indexed: 12/14/2022] Open
Abstract
Background Sleepiness and cognitive dysfunction are recognized as prominent consequences of sleep deprivation. Experimentally induced short-term sleep fragmentation, even in the absence of any reductions in total sleep duration, will lead to the emergence of excessive daytime sleepiness and cognitive impairments in humans. Tumor necrosis factor (TNF)-α has important regulatory effects on sleep, and seems to play a role in the occurrence of excessive daytime sleepiness in children who have disrupted sleep as a result of obstructive sleep apnea, a condition associated with prominent sleep fragmentation. The aim of this study was to examine role of the TNF-α pathway after long-term sleep fragmentation in mice. Methods The effect of chronic sleep fragmentation during the sleep-predominant period on sleep architecture, sleep latency, cognitive function, behavior, and inflammatory markers was assessed in C57BL/6 J and in mice lacking the TNF-α receptor (double knockout mice). In addition, we also assessed the above parameters in C57BL/6 J mice after injection of a TNF-α neutralizing antibody. Results Mice subjected to chronic sleep fragmentation had preserved sleep duration, sleep state distribution, and cumulative delta frequency power, but also exhibited excessive sleepiness, altered cognitive abilities and mood correlates, reduced cyclic AMP response element-binding protein phosphorylation and transcriptional activity, and increased phosphodiesterase-4 expression, in the absence of AMP kinase-α phosphorylation and ATP changes. Selective increases in cortical expression of TNF-α primarily circumscribed to neurons emerged. Consequently, sleepiness and cognitive dysfunction were absent in TNF-α double receptor knockout mice subjected to sleep fragmentation, and similarly, treatment with a TNF-α neutralizing antibody abrogated sleep fragmentation-induced learning deficits and increases in sleep propensity. Conclusions Taken together, our findings show that recurrent arousals during sleep, as happens during sleep apnea, induce excessive sleepiness via activation of inflammatory mechanisms, and more specifically TNF-α-dependent pathways, despite preserved sleep duration.
Collapse
|
49
|
Zielinski MR, Taishi P, Clinton JM, Krueger JM. 5'-Ectonucleotidase-knockout mice lack non-REM sleep responses to sleep deprivation. Eur J Neurosci 2012; 35:1789-98. [PMID: 22540145 DOI: 10.1111/j.1460-9568.2012.08112.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adenosine and extracellular adenosine triphosphate (ATP) have multiple physiological central nervous system actions including regulation of cerebral blood flow, inflammation and sleep. However, their exact sleep regulatory mechanisms remain unknown. Extracellular ATP and adenosine diphosphate are converted to adenosine monophosphate (AMP) by the enzyme ectonucleoside triphosphate diphosphohydrolase 1, also known as CD39, and extracellular AMP is in turn converted to adenosine by the 5'-ectonuleotidase enzyme CD73. We investigated the role of CD73 in sleep regulation. Duration of spontaneous non-rapid eye movement sleep (NREMS) was greater in CD73-knockout (KO) mice than in C57BL/6 controls whether determined in our laboratory or by others. After sleep deprivation (SD), NREMS was enhanced in controls but not CD73-KO mice. Interleukin-1 beta (IL1β) enhanced NREMS in both strains, indicating that the CD73-KO mice were capable of sleep responses. Electroencephalographic power spectra during NREMS in the 1.0-2.5 Hz frequency range was significantly enhanced after SD in both CD73-KO and WT mice; the increases were significantly greater in the WT mice than in the CD73-KO mice. Rapid eye movement sleep did not differ between strains in any of the experimental conditions. With the exception of CD73 mRNA, the effects of SD on various adenosine-related mRNAs were small and similar in the two strains. These data suggest that sleep is regulated, in part, by extracellular adenosine derived from the actions of CD73.
Collapse
Affiliation(s)
- Mark R Zielinski
- Sleep and Performance Research Center, Programs in Neuroscience, WWAMI Medical Education Program, Washington State University, Spokane, WA 99210-1495, USA
| | | | | | | |
Collapse
|
50
|
Abstract
Scientists are only beginning to fully understand the purpose of sleep and its underlying mechanisms. Lack of sleep is associated with many diseases, including infection, and with increased mortality. Lack of proper sleep is an important problem in the intensive care unit, and interventions have been designed to improve it. Sleep is associated with immune function, and this relationship is partially based on the physiological basis of sleep, sleep architecture, the sleep-wake cycle, cytokines and the hypothalamic-pituitary axis.
Collapse
Affiliation(s)
- Freda DeKeyser Ganz
- Freda DeKeyser Ganz is the head of the masters program, School of Nursing, Hadassah Hebrew University, Jerusalem, Israel
| |
Collapse
|