101
|
Khalyfa A, Carreras A, Almendros I, Hakim F, Gozal D. Sex dimorphism in late gestational sleep fragmentation and metabolic dysfunction in offspring mice. Sleep 2015; 38:545-57. [PMID: 25325475 DOI: 10.5665/sleep.4568] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/17/2014] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Excessive sleep fragmentation (SF) is common in pregnant women. Adult-onset metabolic disorders may begin during early development and exhibit substantial sex dimorphism. We hypothesized that metabolic dysfunction induced by gestational SF in male mice would not be apparent in female littermates. METHODS Body weight and food consumption were measured weekly in male and female offspring after late gestational SF or control sleep (SC). At 20 weeks, plasma leptin, adiponectin, lipid profiles, and insulin and glucose tolerance tests were assessed. Leptin and adiponectin, M1, and M2 macrophage messenger RNA expression and polarity were examined. Adiponectin gene promoter methylation levels in several tissues were assessed. RESULTS Food intake, body weight, visceral fat mass, and insulin resistance were higher, and adiponectin levels lower in male but not female offspring exposed to gestational SF. However, dyslipidemia was apparent in both male and female offspring exposed to SF, albeit of lesser magnitude. In visceral fat, leptin messenger RNA expression was selectively increased and adiponectin expression was decreased in male offspring exposed to gestational SF, but adiponectin was increased in exposed female offspring. Differences in adipokine expression also emerged in liver, subcutaneous fat, and muscle. Increased M1 macrophage markers were present in male offspring exposed to SF (SFOM) while increased M2 markers emerged in SF in female offspring (SFOF). Similarly, significant differences emerged in the methylation patterns of adiponectin promoter in SFOM and SFOF. CONCLUSION Gestational sleep fragmentation increases the susceptibility to obesity and metabolic syndrome in male but not in female offspring, most likely via epigenetic changes. Thus, sleep perturbations impose long-term detrimental effects to the fetus manifesting as sex dimorphic metabolic dysfunction in adulthood.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Isaac Almendros
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Fahed Hakim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
102
|
Baud MO, Magistretti PJ, Petit JM. Sustained sleep fragmentation induces sleep homeostasis in mice. Sleep 2015; 38:567-79. [PMID: 25325477 DOI: 10.5665/sleep.4572] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/14/2014] [Indexed: 01/12/2023] Open
Abstract
STUDY OBJECTIVES Sleep fragmentation (SF) is an integral feature of sleep apnea and other prevalent sleep disorders. Although the effect of repetitive arousals on cognitive performance is well documented, the effects of long-term SF on electroencephalography (EEG) and molecular markers of sleep homeostasis remain poorly investigated. To address this question, we developed a mouse model of chronic SF and characterized its effect on EEG spectral frequencies and the expression of genes previously linked to sleep homeostasis including clock genes, heat shock proteins, and plasticity-related genes. DESIGN N/A. SETTING Animal sleep research laboratory. PARTICIPANTS Sixty-six C57BL6/J adult mice. INTERVENTIONS Instrumental sleep disruption at a rate of 60/h during 14 days. MEASUREMENTS AND RESULTS Locomotor activity and EEG were recorded during 14 days of SF followed by recovery for 2 days. Despite a dramatic number of arousals and decreased sleep bout duration, SF minimally reduced total quantity of sleep and did not significantly alter its circadian distribution. Spectral analysis during SF revealed a homeostatic drive for slow wave activity (SWA; 1-4 Hz) and other frequencies as well (4-40 Hz). Recordings during recovery revealed slow wave sleep consolidation and a transient rebound in SWA, and paradoxical sleep duration. The expression of selected genes was not induced following chronic SF. CONCLUSIONS Chronic SF increased sleep pressure confirming that altered quality with preserved quantity triggers core sleep homeostasis mechanisms. However, it did not induce the expression of genes induced by sleep loss, suggesting that these molecular pathways are not sustainably activated in chronic diseases involving SF.
Collapse
Affiliation(s)
- Maxime O Baud
- Laboratory of Neuroenergetic and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Department of Neurology, University of California at San Francisco (UCSF), San Francisco, CA
| | - Pierre J Magistretti
- Laboratory of Neuroenergetic and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology (KAUST), Thuwal, KSA.,Centre de Neurosciences Psychiatriques, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Prilly, Switzerland
| | - Jean-Marie Petit
- Laboratory of Neuroenergetic and Cellular Dynamics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.,Centre de Neurosciences Psychiatriques, Department of Psychiatry, Centre Hospitalier Universitaire Vaudois, Prilly, Switzerland
| |
Collapse
|
103
|
Cortese R, Khalyfa A, Bao R, Andrade J, Gozal D. Epigenomic profiling in visceral white adipose tissue of offspring of mice exposed to late gestational sleep fragmentation. Int J Obes (Lond) 2015; 39:1135-42. [PMID: 25801690 PMCID: PMC4496299 DOI: 10.1038/ijo.2015.38] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/11/2015] [Accepted: 03/14/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Sleep fragmentation during late gestation (LG-SF) is one of the major perturbations associated with sleep apnea and other sleep disorders during pregnancy. We have previously shown that LG-SF induces metabolic dysfunction in offspring mice during adulthood. OBJECTIVES To investigate the effects of late LG-SF on metabolic homeostasis in offspring and to determine the effects of LG-SF on the epigenome of visceral white adipose tissue (VWAT) in the offspring. METHODS Time-pregnant mice were exposed to LG-SF or sleep control during LG (LG-SC) conditions during the last 6 days of gestation. At 24 weeks of age, lipid profiles and metabolic parameters were assessed in the offspring. We performed large-scale DNA methylation analyses using methylated DNA immunoprecipitation (MeDIP) coupled with microarrays (MeDIP-chip) in VWAT of 24-week-old LG-SF and LG-SC offspring (n=8 mice per group). Univariate multiple-testing adjusted statistical analyses were applied to identify differentially methylated regions (DMRs) between the groups. DMRs were mapped to their corresponding genes, and tested for potential overlaps with biological pathways and gene networks. RESULTS We detected significant increases in body weight (31.7 vs 28.8 g; P=0.001), visceral (642.1 vs 497.0 mg; P=0.002) and subcutaneous (293.1 vs 250.1 mg; P=0.001) fat mass, plasma cholesterol (110.6 vs 87.6 mg dl(-1); P=0.001), triglycerides (87.3 vs 84.1 mg dl(-1); P=0.003) and homeostatic model assessment-insulin resistance values (8.1 vs 6.1; P=0.007) in the LG-SF group. MeDIP analyses revealed that 2148 DMRs (LG-SF vs LG-SC; P<0.0001, model-based analysis of tilling-arrays algorithm). A large proportion of the DMR-associated genes have reported functions that are altered in obesity and metabolic syndrome, such as Cartpt, Akt2, Apoe, Insr1 and so on. Overrepresented pathways and gene networks were related to metabolic regulation and inflammatory response. CONCLUSIONS Our findings show a major role for epigenomic regulation of pathways associated with the metabolic processes and inflammatory responses in VWAT. LG-SF-induced epigenetic alterations may underlie increases in the susceptibility to obesity and metabolic syndrome in the offspring.
Collapse
Affiliation(s)
- R Cortese
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
| | - A Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
| | - R Bao
- Bioinformatics Core Facility, Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - J Andrade
- Bioinformatics Core Facility, Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - D Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
104
|
Zheng J, Almendros I, Wang Y, Zhang SX, Carreras A, Qiao Z, Gozal D. Reduced NADPH oxidase type 2 activity mediates sleep fragmentation-induced effects on TC1 tumors in mice. Oncoimmunology 2015; 4:e976057. [PMID: 25949873 DOI: 10.4161/2162402x.2014.976057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/09/2014] [Indexed: 12/14/2022] Open
Abstract
The molecular mechanisms underlying how sleep fragmentation (SF) influences cancer growth and progression remain largely elusive. Here, we present evidence that SF reduced ROS production by downregulating gp91phox expression and activity in TC1 cell tumor associated macrophages (TAMs), while genetic ablation of phagocytic Nox2 activity increased tumor cell proliferation, motility, invasion, and extravasation in vitro. Importantly, the in vivo studies using immunocompetent syngeneic murine tumor models suggested that Nox2 deficiency mimics SF-induced TAMs infiltration and subsequent tumor growth and invasion. Taken together, these studies reveal that perturbed sleep could adversely affect innate immunity within the tumor by altering Nox2 expression and activity, and indicate that selective potentiation of Nox2 activity may present a novel therapeutic strategy in the treatment of cancer.
Collapse
Key Words
- ANOVA, Analysis of variance
- FBS, fetal bovine serum
- HEPES, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
- MFI, median fluorescence intensities
- NADPH oxidase
- Nox2, NADPH Oxidase Type 2
- PMA, phorbol 12-myristate 13-acetate
- ROS, reactive oxygen species
- SE, standard error
- SF, sleep fragmentation
- TAMs, tumor associated macrophages
- TLR-4, toll like receptor 4
- WT, wild type
- cancer
- reactive oxygen species
- rpm, revolutions per minute
- sleep apnea
- tumor associated macrophage
Collapse
Affiliation(s)
- Jiamao Zheng
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| | - Isaac Almendros
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| | - Yang Wang
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| | - Shelley X Zhang
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| | - Alba Carreras
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| | - David Gozal
- Section of Pediatric Sleep Medicine; Department of Pediatrics; Pritzker School of Medicine; Biological Sciences Division; The University of Chicago ; Chicago, Illinois, USA
| |
Collapse
|
105
|
Mutskov V, Khalyfa A, Wang Y, Carreras A, Nobrega MA, Gozal D. Early-life physical activity reverses metabolic and Foxo1 epigenetic misregulation induced by gestational sleep disturbance. Am J Physiol Regul Integr Comp Physiol 2015; 308:R419-30. [PMID: 25568076 DOI: 10.1152/ajpregu.00426.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sleep disorders are highly prevalent during late pregnancy and can impose adverse effects, such as preeclampsia and diabetes. However, the consequences of sleep fragmentation (SF) on offspring metabolism and epigenomic signatures are unclear. We report that physical activity during early life, but not later, reversed the increased body weight, altered glucose and lipid homeostasis, and increased visceral adipose tissue in offspring of mice subjected to gestational SF (SFo). The reversibility of this phenotype may reflect epigenetic mechanisms induced by SF during gestation. Accordingly, we found that the metabolic master switch Foxo1 was epigenetically misregulated in SFo livers in a temporally regulated fashion. Temporal Foxo1 analysis and its gluconeogenetic targets revealed that the epigenetic abnormalities of Foxo1 precede the metabolic syndrome phenotype. Importantly, regular physical activity early, but not later in life, reversed Foxo1 epigenetic misregulation and altered the metabolic phenotype in gestationally SF-exposed offspring. Thus, we have identified a restricted postnatal period during which lifestyle interventions may reverse the Foxo1 epigenetically mediated risk for metabolic dysfunction later in the life, as induced by gestational sleep disorders.
Collapse
Affiliation(s)
- Vesco Mutskov
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, Illinois; and
| | - Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, Illinois; and
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, Illinois; and
| | - Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, Illinois; and
| | - Marcelo A Nobrega
- Department of Human Genetics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, Illinois
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, Illinois; and
| |
Collapse
|
106
|
Hakim F, Wang Y, Carreras A, Hirotsu C, Zhang J, Peris E, Gozal D. Chronic sleep fragmentation during the sleep period induces hypothalamic endoplasmic reticulum stress and PTP1b-mediated leptin resistance in male mice. Sleep 2015; 38:31-40. [PMID: 25325461 DOI: 10.5665/sleep.4320] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/30/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Sleep fragmentation (SF) is highly prevalent and may constitute an important contributing factor to excessive weight gain and the metabolic syndrome. Increased endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) leading to the attenuation of leptin receptor signaling in the hypothalamus leads to obesity and metabolic dysfunction. METHODS Mice were exposed to SF and sleep control (SC) for varying periods of time during which ingestive behaviors were monitored. UPR pathways and leptin receptor signaling were assessed in hypothalami. To further examine the mechanistic role of ER stress, changes in leptin receptor (ObR) signaling were also examined in wild-type mice treated with the ER chaperone tauroursodeoxycholic acid (TUDCA), as well as in CHOP-/+ transgenic mice. RESULTS Fragmented sleep in male mice induced increased food intake starting day 3 and thereafter, which was preceded by increases in ER stress and activation of all three UPR pathways in the hypothalamus. Although ObR expression was unchanged, signal transducer and activator of transcription 3 (STAT3) phosphorylation was decreased, suggesting reduced ObR signaling. Unchanged suppressor of cytokine signaling-3 (SOCS3) expression and increases in protein-tyrosine phosphatase 1B (PTP1B) expression and activity emerged with SF, along with reduced p-STAT3 responses to exogenous leptin. SF-induced effects were reversed following TUDCA treatment and were absent in CHOP -/+ mice. CONCLUSIONS SF induces hyperphagic behaviors and reduced leptin signaling in hypothalamus that are mediated by activation of ER stress, and ultimately lead to increased PTP1B activity. ER stress pathways are therefore potentially implicated in SF-induced weight gain and metabolic dysfunction, and may represent a viable therapeutic target.
Collapse
Affiliation(s)
- Fahed Hakim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Camila Hirotsu
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Jing Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Eduard Peris
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
107
|
|
108
|
Carreras A, Zhang SX, Peris E, Qiao Z, Gileles-Hillel A, Li RC, Wang Y, Gozal D. Chronic sleep fragmentation induces endothelial dysfunction and structural vascular changes in mice. Sleep 2014; 37:1817-24. [PMID: 25364077 DOI: 10.5665/sleep.4178] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
STUDY OBJECTIVES Sleep fragmentation (SF) is a common occurrence and constitutes a major characteristic of obstructive sleep apnea (OSA). SF has been implicated in multiple OSA-related morbidities, but it is unclear whether SF underlies any of the cardiovascular morbidities of OSA. We hypothesized that long-term SF exposures may lead to endothelial dysfunction and altered vessel wall structure. METHODS AND RESULTS Adult male C57BL/6J mice were fed normal chow and exposed to daylight SF or control sleep (CTL) for 20 weeks. Telemetric blood pressure and endothelial function were assessed weekly using a modified laser-Doppler hyperemic test. Atherosclerotic plaques, elastic fiber disruption, lumen area, wall thickness, foam cells, and macrophage recruitment, as well as expression of senescence-associated markers were examined in excised aortas. Increased latencies to reach baseline perfusion levels during the post-occlusive period emerged in SF mice with increased systemic BP values starting at 8 weeks of SF and persisting thereafter. No obvious atherosclerotic plaques emerged, but marked elastic fiber disruption and fiber disorganization were apparent in SF-exposed mice, along with increases in the number of foam cells and macrophages in the aorta wall. Senescence markers showed reduced TERT and cyclin A and increased p16INK4a expression, with higher IL-6 plasma levels in SF-exposed mice. CONCLUSIONS Long-term sleep fragmentation induces vascular endothelial dysfunction and mild blood pressure increases. Sleep fragmentation also leads to morphologic vessel changes characterized by elastic fiber disruption and disorganization, increased recruitment of inflammatory cells, and altered expression of senescence markers, thereby supporting a role for sleep fragmentation in the cardiovascular morbidity of OSA.
Collapse
Affiliation(s)
- Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Shelley X Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Eduard Peris
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Alex Gileles-Hillel
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Richard C Li
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Pritzker School of Medicine, The University of Chicago, Chicago, IL
| |
Collapse
|
109
|
Khalyfa A, Mutskov V, Carreras A, Khalyfa AA, Hakim F, Gozal D. Sleep fragmentation during late gestation induces metabolic perturbations and epigenetic changes in adiponectin gene expression in male adult offspring mice. Diabetes 2014; 63:3230-41. [PMID: 24812424 PMCID: PMC4171662 DOI: 10.2337/db14-0202] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sleep fragmentation (SF) is a common condition among pregnant women, particularly during late gestation. Gestational perturbations promote the emergence of adiposity and metabolic disease risk in offspring, most likely through epigenetic modifications. Adiponectin (AdipoQ) expression inversely correlates with obesity and insulin resistance. The effects of SF during late gestation on metabolic function and AdipoQ expression in visceral white adipose tissue (VWAT) of offspring mice are unknown. Male offspring mice were assessed at 24 weeks after dams were exposed to SF or control sleep during late gestation. Increased food intake, body weight, VWAT mass, and insulin resistance, with reductions in AdipoQ expression in VWAT, emerged in SF offspring. Increased DNMT3a and -b and global DNA methylation and reduced histone acetyltransferase activity and TET1, -2, and -3 expression were detected in VWAT of SF offspring. Reductions in 5-hydroxymethylcytosine and H3K4m3 and an increase in DNA 5-methylcytosine and H3K9m2 in the promoter and enhancer regions of AdipoQ emerged in adipocytes from VWAT and correlated with AdipoQ expression. SF during late gestation induces epigenetic modifications in AdipoQ in male offspring mouse VWAT adipocytes along with a metabolic syndrome-like phenotype. Thus, altered gestational environments elicited by SF impose the emergence of adverse, long-lasting metabolic consequences in the next generation.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Vesco Mutskov
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Alba Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Ahamed A Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Fahed Hakim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
110
|
Khalyfa A, Wang Y, Zhang SX, Qiao Z, Abdelkarim A, Gozal D. Sleep fragmentation in mice induces nicotinamide adenine dinucleotide phosphate oxidase 2-dependent mobilization, proliferation, and differentiation of adipocyte progenitors in visceral white adipose tissue. Sleep 2014; 37:999-1009. [PMID: 24790279 DOI: 10.5665/sleep.3678] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chronic sleep fragmentation (SF) without sleep curtailment induces increased adiposity. However, it remains unclear whether mobilization, proliferation, and differentiation of adipocyte progenitors (APs) occurs in visceral white adipose tissue (VWAT), and whether nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (Nox2) activity plays a role. METHODS Changes in VWAT depot cell size and AP proliferation were assessed in wild-type and Nox2 null male mice exposed to SF and control sleep (SC). To assess mobilization, proliferation, and differentiation of bone marrow mesenchymal stem cells (BM-MSC), Sca-1+ bone marrow progenitors were isolated from GFP+ or RFP+ mice, and injected intravenously to adult male mice (C57BL/6) previously exposed to SF or SC. RESULTS In comparison with SC, SF was associated with increased weight accrual at 3 w and thereafter, larger subcutaneous and visceral fat depots, and overall adipocyte size at 8 w. Increased global AP numbers in VWAT along with enhanced AP BrDU labeling in vitro and in vivo emerged in SF. Systemic injections of GFP+ BM-MSC resulted in increased AP in VWAT, as well as in enhanced differentiation into adipocytes in SF-exposed mice. No differences occurred between SF and SC in Nox2 null mice for any of these measurements. CONCLUSIONS Chronic sleep fragmentation (SF) induces obesity in mice and increased proliferation and differentiation of adipocyte progenitors (AP) in visceral white adipose tissue (VWAT) that are mediated by increased Nox2 activity. In addition, enhanced migration of bone marrow mesenchymal stem cells from the systemic circulation into VWAT, along with AP differentiation, proliferation, and adipocyte formation occur in SF-exposed wild-type but not in oxidase 2 (Nox2) null mice. Thus, Nox2 may provide a therapeutic target to prevent obesity in the context of sleep disorders.
Collapse
Affiliation(s)
- Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Yang Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Shelley X Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Zhuanhong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Amal Abdelkarim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| |
Collapse
|
111
|
Gozal D, Almendros I, Hakim F. Sleep apnea awakens cancer: A unifying immunological hypothesis. Oncoimmunology 2014; 3:e28326. [PMID: 25050203 PMCID: PMC4063138 DOI: 10.4161/onci.28326] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 02/18/2014] [Accepted: 02/21/2014] [Indexed: 12/02/2022] Open
Abstract
The presence of obstructive sleep apnea (OSA) in patients with cancer appears to be accompanied by poorer outcomes. However, the mechanisms underlying such association are unknown. We hypothesize that the constitutive characteristics of OSA, namely, intermittent hypoxia and sleep fragmentation, promote changes in the tumor microenvironment that ultimately lead to a disadvantageous immunosurveillance, thereby accelerating tumor proliferation and enhancing its invasiveness.
Collapse
Affiliation(s)
- David Gozal
- Department of Pediatrics; Comer Children's Hospital; Pritzker School of Medicine; University of Chicago; Chicago, IL USA
| | - Isaac Almendros
- Department of Pediatrics; Comer Children's Hospital; Pritzker School of Medicine; University of Chicago; Chicago, IL USA
| | - Fahed Hakim
- Department of Pediatrics; Comer Children's Hospital; Pritzker School of Medicine; University of Chicago; Chicago, IL USA
| |
Collapse
|
112
|
Wang Y, Carreras A, Lee S, Hakim F, Zhang SX, Nair D, Ye H, Gozal D. Chronic sleep fragmentation promotes obesity in young adult mice. Obesity (Silver Spring) 2014; 22:758-62. [PMID: 24039209 PMCID: PMC3947647 DOI: 10.1002/oby.20616] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/01/2013] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Short sleep confers a higher risk of obesity in humans. Restricted sleep increases appetite, promotes higher calorie intake from fat and carbohydrate sources, and induces insulin resistance. However, the effects of fragmented sleep (SF), such as occurs in sleep apnea, on body weight, metabolic rates, and adipose tissue distribution are unknown. METHODS C57BL/6 mice were exposed to SF for 8 weeks. Their body weight, food consumption, and metabolic expenditure were monitored over time, and their plasma leptin levels measured after exposure to SF for 1 day as well as for 2 weeks. In addition, adipose tissue distribution was assessed at the end of the SF exposure using MRI techniques. RESULTS Chronic SF-induced obesogenic behaviors and increased weight gain in mice by promoting increased caloric intake without changing caloric expenditure. Plasma leptin levels initially decreased and subsequently increased. Furthermore, increases in both visceral and subcutaneous adipose tissue volumes occurred. CONCLUSIONS These results suggest that SF, a frequent occurrence in many disorders and more specifically in sleep apnea, is a potent inducer of obesity via activation of obesogenic behaviors and possibly leptin resistance, in the absence of global changes in energy expenditure.
Collapse
Affiliation(s)
- Yang Wang
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
- Corresponding author: David Gozal, Department of Pediatrics, The University of Chicago, 5721 S. Maryland Avenue, MC 8000, Suite K-160, Chicago, IL 60637. Tel: (773) 702-6205; (773) 702-4523 – FAX;
| | - Alba Carreras
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
| | - SeungHoon Lee
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Fahed Hakim
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Shelley X. Zhang
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Deepti Nair
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Honggang Ye
- Section of Endocrinology, Department of Medicine, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - David Gozal
- Section of Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL
| |
Collapse
|
113
|
Lim DC, Pack AI. Obstructive sleep apnea and cognitive impairment: addressing the blood-brain barrier. Sleep Med Rev 2014; 18:35-48. [PMID: 23541562 PMCID: PMC3758447 DOI: 10.1016/j.smrv.2012.12.003] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/21/2012] [Accepted: 12/24/2012] [Indexed: 12/14/2022]
Abstract
Increasing data support a connection between obstructive sleep apnea (OSA) and cognitive impairment but a causal link has yet to be established. Although neuronal loss has been linked to cognitive impairment, emerging theories propose that changes in synaptic plasticity can cause cognitive impairment. Studies demonstrate that disruption to the blood-brain barrier (BBB), which is uniquely structured to tightly maintain homeostasis inside the brain, leads to changes in the brain's microenvironment and affects synaptic plasticity. Cyclical intermittent hypoxia is a stressor that could disrupt the BBB via molecular responses already known to occur in either OSA patients or animal models of intermittent hypoxia. However, we do not yet know if or how intermittent hypoxia can cause cognitive impairment by mechanisms operating at the BBB. Therefore, we propose that initially, adaptive homeostatic responses at the BBB occur in response to increased oxygen and nutrient demand, specifically through regulation of influx and efflux BBB transporters that alter microvessel permeability. We further hypothesize that although these responses are initially adaptive, these changes in BBB transporters can have long-term consequences that disrupt the brain's microenvironment and alter synaptic plasticity leading to cognitive impairment.
Collapse
Affiliation(s)
- Diane C Lim
- Department of Medicine, Division of Sleep Medicine, and Center for Sleep and Circadian Neurobiology, University of Pennsylvania, 125 South 31st Street, Suite 2100, Philadelphia, PA 19104, USA.
| | - Allan I Pack
- Department of Medicine, Division of Sleep Medicine, and Center for Sleep and Circadian Neurobiology, University of Pennsylvania, 125 South 31st Street, Suite 2100, Philadelphia, PA 19104, USA.
| |
Collapse
|
114
|
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
|
115
|
Hakim F, Wang Y, Zhang SXL, Zheng J, Yolcu ES, Carreras A, Khalyfa A, Shirwan H, Almendros I, Gozal D. Fragmented sleep accelerates tumor growth and progression through recruitment of tumor-associated macrophages and TLR4 signaling. Cancer Res 2014; 74:1329-37. [PMID: 24448240 DOI: 10.1158/0008-5472.can-13-3014] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sleep fragmentation (SF) is a highly prevalent condition and a hallmark of sleep apnea, a condition that has been associated with increased cancer incidence and mortality. In this study, we examined the hypothesis that sleep fragmentation promotes tumor growth and progression through proinflammatory TLR4 signaling. In the design, we compared mice that were exposed to sleep fragmentation one week before engraftment of syngeneic TC1 or LL3 tumor cells and tumor analysis four weeks later. We also compared host contributions through the use of mice genetically deficient in TLR4 or its effector molecules MYD88 or TRIF. We found that sleep fragmentation enhanced tumor size and weight compared with control mice. Increased invasiveness was apparent in sleep fragmentation tumors, which penetrated the tumor capsule into surrounding tissues, including adjacent muscle. Tumor-associated macrophages (TAM) were more numerous in sleep fragmentation tumors, where they were distributed in a relatively closer proximity to the tumor capsule compared with control mice. Although tumors were generally smaller in both MYD88(-/-) and TRIF(-/-) hosts, the more aggressive features produced by sleep fragmentation persisted. In contrast, these more aggressive features produced by sleep fragmentation were abolished completely in TLR4(-/-) mice. Our findings offer mechanistic insights into how sleep perturbations can accelerate tumor growth and invasiveness through TAM recruitment and TLR4 signaling pathways.
Collapse
Affiliation(s)
- Fahed Hakim
- Authors' Affiliations: Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, The University of Chicago, Chicago, Illinois; and Department of Microbiology and Immunology, The University of Louisville, Louisville, Kentucky
| | | | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Hypersomnolence and reduced activity in pan-leptin receptor knockout mice. J Mol Neurosci 2013; 51:1038-45. [PMID: 23955775 DOI: 10.1007/s12031-013-0093-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/06/2013] [Indexed: 12/26/2022]
Abstract
Excessive obesity correlates with hypersomnolence and impaired cognitive function, presumably induced by metabolic factors and cytokines. Production of the adipokine leptin correlates with the amount of adiposity, and leptin has been shown to promote sleep. To determine whether leptin plays a major role in the hypersomnolence of obesity, we measured sleep architecture in pan-leptin receptor knockout (POKO) mice that do not respond to leptin because of the production of a mutant, non-signaling receptor. The obese POKO mice had more non-rapid eye movement (NREM) sleep and less waking time than their littermate controls. This was mainly seen during the light span, although increased bouts of rapid eye movement sleep were also seen in the dark span. The increase of NREM sleep correlated with the extent of obesity. The POKO mice also had decreased locomotor activity and more immobility in the open field test, but there was no increase of forced immobility nor reduction of sucrose intake as would be seen in depression. The increased NREM sleep and reduced locomotor activity in the POKO mice suggest that it was obesity, rather than leptin signaling, that played a predominant role in altering sleep architecture and activity.
Collapse
|
117
|
Zielinski MR, Dunbrasky DL, Taishi P, Souza G, Krueger JM. Vagotomy attenuates brain cytokines and sleep induced by peripherally administered tumor necrosis factor-α and lipopolysaccharide in mice. Sleep 2013; 36:1227-38, 1238A. [PMID: 23904683 DOI: 10.5665/sleep.2892] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
STUDY OBJECTIVE Systemic tumor necrosis factor-α (TNF-α) is linked to sleep and sleep altering pathologies in humans. Evidence from animals indicates that systemic and brain TNF-α have a role in regulating sleep. In animals, TNF-α or lipopolysaccharide (LPS) enhance brain pro-inflammatory cytokine expression and sleep after central or peripheral administration. Vagotomy blocks enhanced sleep induced by systemic TNF-α and LPS in rats, suggesting that vagal afferent stimulation by TNF-α enhances pro-inflammatory cytokines in sleep-related brain areas. However, the effects of systemic TNF-α on brain cytokine expression and mouse sleep remain unknown. DESIGN We investigated the role of vagal afferents on brain cytokines and sleep after systemically applied TNF-α or LPS in mice. MEASUREMENTS AND RESULTS Spontaneous sleep was similar in vagotomized and sham-operated controls. Vagotomy attenuated TNF-α- and LPS-enhanced non-rapid eye movement sleep (NREMS); these effects were more evident after lower doses of these substances. Vagotomy did not affect rapid eye movement sleep responses to these substances. NREMS electroencephalogram delta power (0.5-4 Hz range) was suppressed after peripheral TNF-α or LPS injections, although vagotomy did not affect these responses. Compared to sham-operated controls, vagotomy did not affect liver cytokines. However, vagotomy attenuated interleukin-1 beta (IL-1β) and TNF-α mRNA brain levels after TNF-α, but not after LPS, compared to the sham-operated controls. CONCLUSIONS We conclude that vagal afferents mediate peripheral TNF-α-induced brain TNF-α and IL-1β mRNA expressions to affect sleep. We also conclude that vagal afferents alter sleep induced by peripheral pro-inflammatory stimuli in mice similar to those occurring in other species.
Collapse
Affiliation(s)
- Mark R Zielinski
- Sleep and Performance Research Center, Washington State University, Spokane, WA 99210-1495, USA
| | | | | | | | | |
Collapse
|
118
|
Zhang SXL, Khalyfa A, Wang Y, Carreras A, Hakim F, Neel BA, Brady MJ, Qiao Z, Hirotsu C, Gozal D. Sleep fragmentation promotes NADPH oxidase 2-mediated adipose tissue inflammation leading to insulin resistance in mice. Int J Obes (Lond) 2013; 38:619-24. [PMID: 23897221 PMCID: PMC3907464 DOI: 10.1038/ijo.2013.139] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 06/24/2013] [Accepted: 07/18/2013] [Indexed: 12/24/2022]
Abstract
Background Short sleep has been implicated in higher risk of obesity in humans, and is associated with insulin resistance. However, the effects of fragmented sleep (SF) rather than curtailed sleep on glucose homeostasis are unknown. Methods Wild type and NADPH oxidase 2 (Nox2) null male mice were subjected to SF or sleep control (SC) conditions for 3 days-3 weeks. Systemic and visceral adipose tissue (VAT) insulin sensitivity tests, glucose tolerance test, FACS and immunohistochemistry for macrophages and sub-types (M1 and M2) and Nox expression and activity were examined. Results Here show that SF in the absence of sleep curtailment induces time-dependent insulin resistance, in vivo and also in vitro in VAT. Oxidative stress pathways were up-regulated by SF in VAT, and were accompanied by M1 macrophage polarization. SF-induced oxidative stress, inflammation, and insulin resistance in VAT were completely abrogated in genetically altered mice lacking Nox2 activity. Conclusions These studies imply that SF, a frequent occurrence in many disorders and more specifically in sleep apnea, is a potent inducer of insulin resistance via activation of oxidative stress and inflammatory pathways, thereby opening the way for therapeutic strategies.
Collapse
Affiliation(s)
- S X L Zhang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - A Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - Y Wang
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - A Carreras
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - F Hakim
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - B A Neel
- Section of Endocrinology, Department of Medicine, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - M J Brady
- Section of Endocrinology, Department of Medicine, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA
| | - Z Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - C Hirotsu
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| | - D Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
119
|
Gozal D. CrossTalk proposal: the intermittent hypoxia attending severe obstructive sleep apnoea does lead to alterations in brain structure and function. J Physiol 2013; 591:379-81. [PMID: 23322286 DOI: 10.1113/jphysiol.2012.241216] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- David Gozal
- Department of Pediatrics, Pritzker School of Medicine, Biological Sciences Division, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
120
|
Obstructive sleep apnea and delirium: exploring possible mechanisms. Sleep Breath 2013; 18:19-29. [PMID: 23584846 DOI: 10.1007/s11325-013-0846-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/19/2013] [Accepted: 03/25/2013] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Obstructive sleep apnea (OSA) is a medical disorder strongly associated with multiple comorbidities and postoperative complications. Current evidence suggests that OSA disturbs fundamental biochemical processes, leading to low-grade systemic inflammation and oxidative stress. Animal models have shown that OSA may lead to apoptosis of central neurons. In clinical studies, oxygen desaturation index and sleep fragmentation have been shown to be independently associated with cognitive dysfunction. Moreover, in several studies, patients with OSA were shown to have decreased brain activation in multiple brain areas. OSA AND DELIRIUM The possibility of an association between OSA and delirium has been highlighted in several case reports. The first prospective study of the possible link between apnea and delirium showed that the presence of OSA was independently associated with the occurrence of delirium after knee replacement surgery. CONCLUSIONS Therefore, we suggest that OSA should be considered as a risk factor for delirium, and clinicians should assess patients for OSA and related risk factors prior to surgery. However, further research is required to shed light on the mechanisms connecting these disorders and on whether the treatment of OSA affects the incidence of delirium.
Collapse
|
121
|
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
|
122
|
|
123
|
Kaushal N, Ramesh V, Gozal D. TNF-α and temporal changes in sleep architecture in mice exposed to sleep fragmentation. PLoS One 2012; 7:e45610. [PMID: 23029133 PMCID: PMC3448632 DOI: 10.1371/journal.pone.0045610] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 08/23/2012] [Indexed: 12/31/2022] Open
Abstract
TNF-α plays critical roles in host-defense, sleep-wake regulation, and the pathogenesis of various disorders. Increases in the concentration of circulating TNF-α after either sleep deprivation or sleep fragmentation (SF) appear to underlie excessive daytime sleepiness in patients with sleep apnea (OSA). Following baseline recordings, mice were subjected to 15 days of SF (daily for 12 h/day from 07.00 h to 19.00 h), and sleep parameters were recorded on days1, 7 and 15. Sleep architecture and sleep propensity were assessed in both C57BL/6J and in TNF-α double receptor KO mice (TNFR KO). To further confirm the role of TNF-α, we also assessed the effect of treatment with a TNF- α neutralizing antibody in C57BL/6J mice. SF was not associated with major changes in global sleep architecture in C57BL/6J and TNFR KO mice. TNFR KO mice showed higher baseline SWS delta power. Further, following 15 days of SF, mice injected with TNF-α neutralizing antibody and TNFR KO mice showed increased EEG SWS activity. However, SWS latency, indicative of increased propensity to sleep, was only decreased in C57BL/6J, and was unaffected in TNFR KO mice as well as in C57BL/6J mice exposed to SF but treated with TNF-α neutralizing antibody. Taken together, our findings show that the excessive sleepiness incurred by recurrent arousals during sleep may be due to activation of TNF-alpha-dependent inflammatory pathways, despite the presence of preserved sleep duration and global sleep architecture.
Collapse
Affiliation(s)
| | | | - David Gozal
- Department of Pediatrics, Section of Pediatric Sleep Medicine, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|