1
|
Chen T, Jia J, Gao C, Zhong Q, Tang L, Sui X, Li S, Chen C, Zhang Z. Integrative metabolomics and transcriptomics analysis of hippocampus reveals taurine metabolism and sphingolipid metabolism dysregulation associated with sleep deprivation-induced memory impairment. Brain Res Bull 2025; 227:111397. [PMID: 40409601 DOI: 10.1016/j.brainresbull.2025.111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 05/06/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
Sleep plays a crucial role in restoring and repairing the body, consolidating memory, regulating emotions, maintaining metabolic and so on. Sleep deprivation is known to impair cognitive functions. In this study, we investigated the mechanisms underlying memory impairment induced by sleep deprivation through a combined metabolomic and transcriptomic analysis of hippocampus. Eight-week-old mice were selected as the study subjects and the sleep deprivation chamber was used to establish a sleep deprivation model. Novel object recognition tests (NOR), and Y-maze tests were used to assess the behavioral outcomes in mice. The hippocampus were extracted and studied using the untargeted metabolomics or transcriptomics high-throughput sequencing method. An integrative analysis was conducted to elucidate the metabolic and genetic changes. Behavioral tests showed that sleep-deprived mice exhibited memory impairment. Metabolomic analysis identified 84 differentially expressed metabolites (DEMs), including 12 under the positive ion mode and 72 under the negative ion mode. The analysis revealed that sleep deprivation caused abnormalities in several metabolic pathways, with particularly pronounced effects observed in glycerophospholipid metabolism, linoieic acid metabolism, alanine, aspartate, glutamate metabolism, taurine and hypotaurine metabolism, and purine metabolism. While transcriptomic analysis releaved 97 differentially expressed genes (DEGs) (51 were down-regulated and 46 were up-regulated DEGs). Integrative analysis of the metabolomic and transcriptomic identified profiles showed that sleep deprivation may regulate taurine and hypotaurine metabolism and sphingolipid metabolism, there by influencing memory. Our results prompt severe metabolic disturbances occur in the hippocampus with sleep deprivation in mice, which can provide a basis for the mechanism research.
Collapse
Affiliation(s)
- Ting Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| | - Junke Jia
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Chenyi Gao
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Department of Anesthesiology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo, Zhejiang 315040, China
| | - Qi Zhong
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Lijuan Tang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Xiaokai Sui
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Shuang Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Sleep Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China
| | - Chang Chen
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| | - Zongze Zhang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China.
| |
Collapse
|
2
|
Dong Y, Lam SM, Li Y, Li MD, Shui G. The circadian clock at the intersection of metabolism and aging - emerging roles of metabolites. J Genet Genomics 2025:S1673-8527(25)00123-7. [PMID: 40306487 DOI: 10.1016/j.jgg.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
The circadian clock is a highly hierarchical network of endogenous pacemakers that primarily maintains and directs oscillations through transcriptional and translational feedback loops, which modulates an approximately 24-hour cycle of endocrine and metabolic rhythms within cells and tissues. While circadian clocks regulate metabolic processes and related physiology, emerging evidence indicates that metabolism and circadian rhythm are intimately intertwined. In this review, we highlight the concept of metabolites, including lipids and other polar metabolites generated from intestinal microbial metabolism and nutrient intake, as circadian pacemakers that drive changes in circadian rhythms, which in turn influence metabolism and aging. Furthermore, we discuss the roles of functional metabolites as circadian pacemakers, paving a new direction on potential intervention targets of circadian disruption, pathological aging, as well as metabolic diseases that are clinically important.
Collapse
Affiliation(s)
- Yue Dong
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; LipidALL Technologies Company Limited, Changzhou, Jiangsu 213022, China
| | - Yan Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China.
| | - Min-Dian Li
- Department of Cardiovascular Medicine, Center for Circadian Metabolism and Cardiovascular Disease, MOE Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease, Southwest Hospital, Army Medical University, Chongqing 400038, China.
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China; Guangzhou National Laboratory, Guangzhou, Guangdong 510005, China.
| |
Collapse
|
3
|
Huang SY, Yang ZJ, Cheng J, Li HY, Chen S, Huang ZH, Chen JD, Xiong RG, Yang MT, Wang C, Li MC, Song S, Huang WG, Wang DL, Li HB, Lan QY. Choline alleviates cognitive impairment in sleep-deprived young mice via reducing neuroinflammation and altering phospholipidomic profile. Redox Biol 2025; 81:103578. [PMID: 40056720 PMCID: PMC11930228 DOI: 10.1016/j.redox.2025.103578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025] Open
Abstract
Cognitive impairment resulting from insufficient sleep poses a significant public health concern, particularly in children. The effects and mechanisms of choline on cognitive impairment caused by sleep deprivation are unknown. Chronic sleep deprivation is induced in young mice in this study, followed by feeding diet containing 11.36 g/kg choline bitartrate. Choline supplementation significantly improves spatial learning ability. Functional MRI results reveal the hippocampus as a key region affected by sleep deprivation, where choline supplementation notably preserves hippocampal structural integrity and enhanced connectivity. Additionally, choline ameliorates hippocampal pathological injury, reduces blood-brain barrier permeability and serum brain injury biomarkers. Choline also reduces inflammation and oxidative stress biomarkers, and mitigates microglial activation in the hippocampus, which preserves synaptic plasticity. A key finding is the changes of hippocampal phospholipidomic profile along with cognitive function, and a total of 313 phospholipid molecules are identified. Choline increases the levels of total phospholipid and sub-classes (particularly PC), which are strongly correlated with reduced neuroinflammation and oxidative stress biomarkers, as well as improved cognitive outcomes. Furthermore, there are similar findings in some phospholipid molecules such as PC 36:1, PC O-33:0, PC p-38:3, PE 36:3, PE p-42:4 and PS 44:12. These findings highlight that choline alleviates cognitive impairment in sleep deprivation via reducing neuroinflammation and oxidative stress as well as altering phospholipidomic profile. This study suggests that choline could develop into functional food or medicine ingredient to prevent and treat cognitive impairment by sleep disturbances, particularly children and adolescents.
Collapse
Affiliation(s)
- Si-Yu Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhi-Jun Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jin Cheng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hang-Yu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Si Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zi-Hui Huang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jie-Dong Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ruo-Gu Xiong
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng-Tao Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Chen Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Meng-Chu Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuang Song
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Wen-Ge Huang
- Center of Experimental Animals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dong-Liang Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hua-Bin Li
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Qiu-Ye Lan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China; School of Public Health, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Dai Y, Sun X, Zhang G, Cui C, Wu X, Aizezi Y, Kadier K. Association between sleep duration, sleep trouble and all-cause mortality in individuals with hyperuricemia in the United States. Front Public Health 2025; 13:1521372. [PMID: 40206179 PMCID: PMC11979105 DOI: 10.3389/fpubh.2025.1521372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/14/2025] [Indexed: 04/11/2025] Open
Abstract
Objectives Despite the crucial role of sleep quality in hyperuricemia onset and progression, there is limited evidence on sleep interventions to improve outcomes for hyperuricemic individuals. This study aims to investigate the effects of sleep duration and sleep difficulties on all-cause mortality in this population. Materials and methods We conducted a secondary analysis of the National Health and Nutrition Examination Survey (NHANES) data from 2007 to 2018, including 5,837 participants. We employed weighted multivariable Cox proportional hazard models to evaluate the independent predictive value of sleep duration and trouble for all-cause mortality. Restricted cubic splines and segmented Cox proportional hazard models were used to examine threshold effects. Results During a mean follow-up of 6.5 years, 906 participants experienced all-cause mortality. After adjusting for confounders, both short (< 7 h; HR = 1.25; 95%CI: 1.04, 1.51; p = 0.018) and long (>9 h; HR = 1.50; 95%CI: 1.10, 2.04; p = 0.011) sleep durations were associated with increased all-cause mortality. The threshold analysis identified an optimal sleep duration of 7.23 h, and when sleep duration was below 7.23 h, it was inversely related to mortality (HR: 0.879; 95% CI: 0.788, 0.981; p = 0.022). Conversely, when sleep duration exceeded 7.23 h, it was positively associated with mortality (HR: 1.187; 95% CI: 1.066, 1.320; p = 0.002). Conclusion Sleep duration is U-shapedly associated with all-cause mortality among individuals with hyperuricemia in the United States. However sleep trouble was not associated with all-cause mortality. Maintaining optimal sleep duration helps improve the prognostic survival rates of those with hyperuricemia.
Collapse
Affiliation(s)
- Yuanhui Dai
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiangyu Sun
- Clinical Medicine College, , Xinjiang Medical University, Urumqi, China
| | - Ge Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunying Cui
- Department of Emergency Medicine, Jining First People's Hospital, Shandong First Medical University, Jining, China
| | - Xiaoli Wu
- Clinical Medicine College, , Xinjiang Medical University, Urumqi, China
| | - Yierzhati Aizezi
- Critical Medicine Center, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Kaisaierjiang Kadier
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
5
|
Ferreira RCM, Ruiz FS, de Mello MT. Human sleep and immunity: The role of circadian patterns. HANDBOOK OF CLINICAL NEUROLOGY 2025; 206:93-103. [PMID: 39864935 DOI: 10.1016/b978-0-323-90918-1.00003-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
It is well established that sleep promotes health and welfare. Literature data suggests that sleep is a recurrent resting state that performs multiple biological functions, such as memory consolidation and regulation of glucose, lipid metabolism, energy metabolism, eating behavior, and blood pressure, besides, regulating the immune system. These immunological functions depend on regular sleep and circadian rhythms, as both impact the magnitude of immune responses. Circadian rhythm is the 24-h internal clock in our brain that regulates cycles of alertness and sleepiness by responding to light changes in our environment. It encompasses physical and behavioral daily oscillations. Sleep deprivation and circadian misalignment affect immunity, and both have been related to adverse health effects and chronic diseases. Studies have shown that individuals with regular and consistent sleep patterns have a more effective immune response. Thus, understanding how sleep disturbance will affect the immune response is vital in developing interventions to prevent the health burden of irregular sleep patterns and circadian misalignment, favoring a homeostatic immune defense to microbial or inflammatory insults. Therefore, the scope of this chapter is to explore evidence that regular circadian rhythms and sleep patterns are needed for optimal resistance to infectious challenges.
Collapse
Affiliation(s)
| | - Francieli S Ruiz
- Escola de Educação Física, Fisioterapia e Terapia Ocupacional, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marco Túlio de Mello
- Escola de Educação Física, Fisioterapia e Terapia Ocupacional, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Faculty of Health Sciences, Universidad Autónoma de Chile, Providencia, Chile.
| |
Collapse
|
6
|
Jiang R, Ohkubo T, Sato T, Sakai N. Stress-Easing Effect of Diacyl Glyceryl Ethers on Anxiety-Related Behavior in Mice. Foods 2024; 13:3765. [PMID: 39682837 DOI: 10.3390/foods13233765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Stress and anxiety are significant psychological challenges in modern society, which have led to a rapidly growing market for functional foods, including those reported to relieve stress, as alternatives to psychoactive drugs. Among these, diacyl glyceryl ethers (DAGE) derived from deep-sea shark liver oil have gained attention for their strong antioxidant properties and potential mental health benefits. Building on preliminary evidence suggesting DAGE's efficacy in enhancing stress resilience and modulating biochemical pathways associated with reduced oxidative stress, the present study aimed to examine their effects on stress responses in two specific mouse strains. Each mouse was fed either a DAGE-infused diet or a control diet for three weeks. Their stress responses were evaluated using three behavioral tests: the elevated plus maze, open-field, and forced swimming tests. The DAGE-fed mice displayed lower stress responses than the control mice in the initial trial of each test. Specifically, DAGE-fed mice demonstrated increased time spent in the open arms in the elevated plus maze and more time spent in the center of the open field, suggesting reduced anxiety. Additionally, in the forced swimming test, DAGE-treated mice displayed reduced immobility times, indicating potential antidepressant effects on the mice. These findings suggest the potential of DAGE to bolster stress resilience in mice, emphasizing their promise for further studies in human stress management.
Collapse
Affiliation(s)
- Rong Jiang
- Department of Psychology, Graduate School of Arts and Letters, Tohoku University, Kawauchi 27-1, Aoba-ku, Sendai 980-8576, Miyagi, Japan
| | - Takeshi Ohkubo
- Faculty of Human Sciences, Department of Health and Nutrition, Sendai Shirayuri Women's College, Honda-cho 6-1, Izumi-ku, Sendai 981-3107, Miyagi, Japan
| | - Toshihiko Sato
- Department of Psychology and Humanities, College of Sociology, Edogawa University, Komagi 474, Nagareyama 270-0198, Chiba, Japan
| | - Nobuyuki Sakai
- Department of Psychology, Graduate School of Arts and Letters, Tohoku University, Kawauchi 27-1, Aoba-ku, Sendai 980-8576, Miyagi, Japan
| |
Collapse
|
7
|
Chen YL, Wang R, Pang R, Sun ZP, He XL, Tang WH, Ou JY, Yi HM, Cheng X, Chen JH, Yu Y, Ren CH, Wang QJ, Zhang ZJ. Transcriptome-Based Revelation of the Effects of Sleep Deprivation on Hepatic Metabolic Rhythms in Tibetan Sheep ( Ovis aries). Animals (Basel) 2024; 14:3165. [PMID: 39595218 PMCID: PMC11591132 DOI: 10.3390/ani14223165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/26/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Sleep deprivation (SD) disrupts circadian rhythms; however, its effects on SD and the mechanisms involved require further investigation. Previous studies on SD were mainly conducted on rodents, such as mice, with few studies on its effects on the liver of large diurnal animals, such as sheep. In this study, we used a Tibetan sheep model for the first time to investigate the effects of SD on the liver by exposing Tibetan sheep (Ovis aries) to 7 days of SD (6 h/day) and performed transcriptome sequencing analysis on liver samples taken at 4 h intervals over 24 h. The results revealed that SD significantly altered the circadian expression of genes and their expression patterns in the liver of Tibetan sheep. Enrichment analysis of the circadian rhythm-altered genes revealed changes in the pathways related to lipid metabolism in the liver. Further evidence from serum markers and gene expression analyses using qualitative real-time polymerase chain reaction and Oil Red O and apoptosis staining indicated that SD leads to abnormal lipid metabolism in the liver, potentially causing liver damage. Therefore, our results suggest that SD disrupts the circadian rhythms of metabolism-related genes in the Tibetan sheep liver, thereby affecting metabolic homeostasis.
Collapse
Affiliation(s)
- Ya-Le Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Ru Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Rui Pang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Zhi-Peng Sun
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China;
| | - Xiao-Long He
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Wen-Hui Tang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Jing-Yu Ou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Huan-Ming Yi
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Xiao Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Jia-Hong Chen
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Chuzhou 233200, China;
| | - Yang Yu
- Qinghai Provincial Key Laboratory of Adaptive Management on Alpine Grassland, Qinghai Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810016, China;
| | - Chun-Huan Ren
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
- Chongqing Key Laboratory of Herbivore Science, College of Animal Science and Technology, Southwest University, Chongqing 400715, China;
| | - Qiang-Jun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
| | - Zi-Jun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.-L.C.); (R.W.); (R.P.); (X.-L.H.); (W.-H.T.); (J.-Y.O.); (H.-M.Y.); (X.C.); (C.-H.R.)
- Center of Agriculture Technology Cooperation and Promotion of Dingyuan County, Chuzhou 233200, China;
| |
Collapse
|
8
|
Goodman LD, Moulton MJ, Lin G, Bellen HJ. Does glial lipid dysregulation alter sleep in Alzheimer's and Parkinson's disease? Trends Mol Med 2024; 30:913-923. [PMID: 38755043 PMCID: PMC11466711 DOI: 10.1016/j.molmed.2024.04.010] [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: 02/17/2024] [Revised: 03/28/2024] [Accepted: 04/10/2024] [Indexed: 05/18/2024]
Abstract
In this opinion article, we discuss potential connections between sleep disturbances observed in Alzheimer's disease (AD) and Parkinson's disease (PD) and the dysregulation of lipids in the brain. Research using Drosophila has highlighted the role of glial-mediated lipid metabolism in sleep and diurnal rhythms. Relevant to AD, the formation of lipid droplets in glia, which occurs in response to elevated neuronal reactive oxygen species (ROS), is required for sleep. In disease models, this process is disrupted, arguing a connection to sleep dysregulation. Relevant to PD, the degradation of neuronally synthesized glucosylceramides by glia requires glucocerebrosidase (GBA, a PD-associated risk factor) and this regulates sleep. Loss of GBA in glia causes an accumulation of glucosylceramides and neurodegeneration. Overall, research primarily using Drosophila has highlighted how dysregulation of glial lipid metabolism may underlie sleep disturbances in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Guang Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Faquih T, Potts K, Yu B, Kaplan R, Isasi CR, Qi Q, Taylor KD, Liu PY, Tracy RP, Johnson C, Rich SS, Clish CB, Gerzsten RE, Rotter JI, Redline S, Sofer T, Wang H. Steroid Hormone Biosynthesis and Dietary Related Metabolites Associated with Excessive Daytime Sleepiness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.12.24313561. [PMID: 39314973 PMCID: PMC11419218 DOI: 10.1101/2024.09.12.24313561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Excessive daytime sleepiness (EDS) is a complex sleep problem that affects approximately 33% of the United States population. Although EDS usually occurs in conjunction with insufficient sleep, and other sleep and circadian disorders, recent studies have shown unique genetic markers and metabolic pathways underlying EDS. Here, we aimed to further elucidate the biological profile of EDS using large scale single- and pathway-level metabolomics analyses. Methods Metabolomics data were available for 877 metabolites in 6,071 individuals from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL) and EDS was assessed using the Epworth Sleepiness Scale (ESS) questionnaire. We performed linear regression for each metabolite on continuous ESS, adjusting for demographic, lifestyle, and physiological confounders, and in sex specific groups. Subsequently, gaussian graphical modelling was performed coupled with pathway and enrichment analyses to generate a holistic interactive network of the metabolomic profile of EDS associations. Findings We identified seven metabolites belonging to steroids, sphingomyelin, and long chain fatty acids sub-pathways in the primary model associated with EDS, and an additional three metabolites in the male-specific analysis. The identified metabolites particularly played a role in steroid hormone biosynthesis. Interpretation Our findings indicate that an EDS metabolomic profile is characterized by endogenous and dietary metabolites within the steroid hormone biosynthesis pathway, with some pathways that differ by sex. Our findings identify potential pathways to target for addressing the causes or consequences of EDS and related sleep disorders. Funding Details regarding funding supporting this work and all studies involved are provided in the acknowledgments section.
Collapse
Affiliation(s)
- Tariq Faquih
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| | - Kaitlin Potts
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| | - Bing Yu
- Department of Epidemiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Robert Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Qibin Qi
- Department of Epidemiology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kent D Taylor
- Department of Paediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Centre, Torrance, CA, USA
| | - Peter Y Liu
- Department of Paediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Centre, Torrance, CA, USA
| | - Russell P Tracy
- Department of Biochemistry, University of Vermont, Burlington, VT, USA
| | - Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, USA
| | - Stephen S Rich
- Centre for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Clary B Clish
- Metabolite Profiling Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert E Gerzsten
- Broad Institute, Cambridge, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jerome I Rotter
- Department of Paediatrics, Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Centre, Torrance, CA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- CardioVascular Institute (CVI), Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Division of Sleep Medicine, Harvard University Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Xie X, Zhang M, Luo H. Regulation of metabolism by circadian rhythms: Support from time-restricted eating, intestinal microbiota & omics analysis. Life Sci 2024; 351:122814. [PMID: 38857654 DOI: 10.1016/j.lfs.2024.122814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/05/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
Circadian oscillatory system plays a key role in coordinating the metabolism of most organisms. Perturbation of genetic effects and misalignment of circadian rhythms result in circadian dysfunction and signs of metabolic disorders. The eating-fasting cycle can act on the peripheral circadian clocks, bypassing the photoperiod. Therefore, time-restricted eating (TRE) can improve metabolic health by adjusting eating rhythms, a process achieved through reprogramming of circadian genomes and metabolic programs at different tissue levels or remodeling of the intestinal microbiota, with omics technology allowing visualization of the regulatory processes. Here, we review recent advances in circadian regulation of metabolism, focus on the potential application of TRE for rescuing circadian dysfunction and metabolic disorders with the contribution of intestinal microbiota in between, and summarize the significance of omics technology.
Collapse
Affiliation(s)
- Ximei Xie
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China
| | - Mengjie Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China
| | - Hailing Luo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, PR China.
| |
Collapse
|
11
|
Scholz M, Steuer AE, Dobay A, Landolt HP, Kraemer T. Assessing the influence of sleep and sampling time on metabolites in oral fluid: implications for metabolomics studies. Metabolomics 2024; 20:97. [PMID: 39112673 PMCID: PMC11306311 DOI: 10.1007/s11306-024-02158-3] [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: 12/15/2023] [Accepted: 07/20/2024] [Indexed: 08/10/2024]
Abstract
INTRODUCTION The human salivary metabolome is a rich source of information for metabolomics studies. Among other influences, individual differences in sleep-wake history and time of day may affect the metabolome. OBJECTIVES We aimed to characterize the influence of a single night of sleep deprivation compared to sufficient sleep on the metabolites present in oral fluid and to assess the implications of sampling time points for the design of metabolomics studies. METHODS Oral fluid specimens of 13 healthy young males were obtained in Salivette® devices at regular intervals in both a control condition (repeated 8-hour sleep) and a sleep deprivation condition (total sleep deprivation of 8 h, recovery sleep of 8 h) and their metabolic contents compared in a semi-targeted metabolomics approach. RESULTS Analysis of variance results showed factor 'time' (i.e., sampling time point) representing the major influencer (median 9.24%, range 3.02-42.91%), surpassing the intervention of sleep deprivation (median 1.81%, range 0.19-12.46%). In addition, we found about 10% of all metabolic features to have significantly changed in at least one time point after a night of sleep deprivation when compared to 8 h of sleep. CONCLUSION The majority of significant alterations in metabolites' abundances were found when sampled in the morning hours, which can lead to subsequent misinterpretations of experimental effects in metabolomics studies. Beyond applying a within-subject design with identical sample collection times, we highly recommend monitoring participants' sleep-wake schedules prior to and during experiments, even if the study focus is not sleep-related (e.g., via actigraphy).
Collapse
Affiliation(s)
- Michael Scholz
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Andrea Eva Steuer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Akos Dobay
- Forensic Machine Learning Technology Center, University of Zurich, Zurich, Switzerland
| | - Hans-Peter Landolt
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Sleep & Health Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Kraemer
- Department of Forensic Pharmacology and Toxicology, Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland.
- Sleep & Health Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Yan H, Li G, Zhang X, Zhang C, Li M, Qiu Y, Sun W, Dong Y, Li S, Li J. Targeted metabolomics-based understanding of the sleep disturbances in drug-naïve patients with schizophrenia. BMC Psychiatry 2024; 24:355. [PMID: 38741058 PMCID: PMC11089724 DOI: 10.1186/s12888-024-05805-0] [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: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Sleep disturbances are a common occurrence in patients with schizophrenia, yet the underlying pathogenesis remain poorly understood. Here, we performed a targeted metabolomics-based approach to explore the potential biological mechanisms contributing to sleep disturbances in schizophrenia. METHODS Plasma samples from 59 drug-naïve patients with schizophrenia and 36 healthy controls were subjected to liquid chromatography-mass spectrometry (LC-MS) targeted metabolomics analysis, allowing for the quantification and profiling of 271 metabolites. Sleep quality and clinical symptoms were assessed using the Pittsburgh Sleep Quality Index (PSQI) and the Positive and Negative Symptom Scale (PANSS), respectively. Partial correlation analysis and orthogonal partial least squares discriminant analysis (OPLS-DA) model were used to identify metabolites specifically associated with sleep disturbances in drug-naïve schizophrenia. RESULTS 16 characteristic metabolites were observed significantly associated with sleep disturbances in drug-naïve patients with schizophrenia. Furthermore, the glycerophospholipid metabolism (Impact: 0.138, p<0.001), the butanoate metabolism (Impact: 0.032, p=0.008), and the sphingolipid metabolism (Impact: 0.270, p=0.104) were identified as metabolic pathways associated with sleep disturbances in drug-naïve patients with schizophrenia. CONCLUSIONS Our study identified 16 characteristic metabolites (mainly lipids) and 3 metabolic pathways related to sleep disturbances in drug-naïve schizophrenia. The detection of these distinct metabolites provide valuable insights into the underlying biological mechanisms associated with sleep disturbances in schizophrenia.
Collapse
Affiliation(s)
- Huiming Yan
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Gang Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
- Chifeng Anding Hospital, NO.18 Gongger Street, Hongshan District, Chifeng City, 024000, Inner Mongolia Autonomous Region, China
| | - Xue Zhang
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
- Chifeng Anding Hospital, NO.18 Gongger Street, Hongshan District, Chifeng City, 024000, Inner Mongolia Autonomous Region, China
| | - Chuhao Zhang
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Meijuan Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Yuying Qiu
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Wei Sun
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Yeqing Dong
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China
| | - Shen Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China.
| | - Jie Li
- Laboratory of Biological Psychiatry, Institute of Mental Health, Tianjin Anding Hospital, Mental Health Center of Tianjin Medical University, 13 Liulin Rd., Hexi District, Tianjin, 300222, China.
| |
Collapse
|
13
|
Jeppe K, Ftouni S, Nijagal B, Grant LK, Lockley SW, Rajaratnam SMW, Phillips AJK, McConville MJ, Tull D, Anderson C. Accurate detection of acute sleep deprivation using a metabolomic biomarker-A machine learning approach. SCIENCE ADVANCES 2024; 10:eadj6834. [PMID: 38457492 PMCID: PMC11094653 DOI: 10.1126/sciadv.adj6834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
Sleep deprivation enhances risk for serious injury and fatality on the roads and in workplaces. To facilitate future management of these risks through advanced detection, we developed and validated a metabolomic biomarker of sleep deprivation in healthy, young participants, across three experiments. Bi-hourly plasma samples from 2 × 40-hour extended wake protocols (for train/test models) and 1 × 40-hour protocol with an 8-hour overnight sleep interval were analyzed by untargeted liquid chromatography-mass spectrometry. Using a knowledge-based machine learning approach, five consistently important variables were used to build predictive models. Sleep deprivation (24 to 38 hours awake) was predicted accurately in classification models [versus well-rested (0 to 16 hours)] (accuracy = 94.7%/AUC 99.2%, 79.3%/AUC 89.1%) and to a lesser extent in regression (R2 = 86.1 and 47.8%) models for within- and between-participant models, respectively. Metabolites were identified for replicability/future deployment. This approach for detecting acute sleep deprivation offers potential to reduce accidents through "fitness for duty" or "post-accident analysis" assessments.
Collapse
Affiliation(s)
- Katherine Jeppe
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
| | - Suzanne Ftouni
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
| | - Brunda Nijagal
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Leilah K. Grant
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Steven W. Lockley
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Shantha M. W. Rajaratnam
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Andrew J. K. Phillips
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
| | - Malcolm J. McConville
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Dedreia Tull
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Australia
| | - Clare Anderson
- School of Psychological Sciences and Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia
- Cooperative Research Centre for Alertness, Safety and Productivity, Melbourne, Australia
- Centre for Human Brain Health, School of Psychology, University of Birmingham, Edgbaston, UK
| |
Collapse
|
14
|
Wang M, Flexeder C, Harris CP, Thiering E, Koletzko S, Bauer CP, Schulte-Körne G, von Berg A, Berdel D, Heinrich J, Schulz H, Schikowski T, Peters A, Standl M. Accelerometry-assessed sleep clusters and cardiometabolic risk factors in adolescents. Obesity (Silver Spring) 2024; 32:200-213. [PMID: 37873587 DOI: 10.1002/oby.23918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/07/2023] [Accepted: 08/18/2023] [Indexed: 10/25/2023]
Abstract
OBJECTIVE This study aimed to identify sleep clusters based on objective multidimensional sleep characteristics and test their associations with adolescent cardiometabolic health. METHODS The authors included 1090 participants aged 14.3 to 16.4 years (mean = 15.2 years) who wore 7-day accelerometers during the 15-year follow-up of the German Infant Study on the influence of Nutrition Intervention PLUS environmental and genetic influences on allergy development (GINIplus) and the Influence of Lifestyle factors on the development of the Immune System and Allergies in East and West Germany (LISA) birth cohorts. K-means cluster analysis was performed across 12 sleep characteristics reflecting sleep quantity, quality, schedule, variability, and regularity. Cardiometabolic risk factors included fat mass index (FMI), blood pressure, triglycerides, high-density lipoprotein cholesterol, high-sensitivity C-reactive protein, and insulin resistance (n = 505). Linear and logistic regression models were examined. RESULTS Five sleep clusters were identified: good sleep (n = 337); delayed sleep phase (n = 244); sleep irregularity and variability (n = 108); fragmented sleep (n = 313); and prolonged sleep latency (n = 88). The "prolonged sleep latency" cluster was associated with increased sex-scaled FMI (β = 0.39, 95% CI: 0.15-0.62) compared with the "good sleep" cluster. The "sleep irregularity and variability" cluster was associated with increased odds of high triglycerides only in male individuals (odds ratio: 9.50, 95% CI: 3.22-28.07), but this finding was not confirmed in linear models. CONCLUSIONS The prolonged sleep latency cluster was associated with higher FMI in adolescents, whereas the sleep irregularity and variability cluster was specifically linked to elevated triglycerides (≥1.7 mmol/L) in male individuals.
Collapse
Affiliation(s)
- Mingming Wang
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Information Processing, Biometry and Epidemiology (IBE), Faculty of Medicine, LMU Munich, Pettenkofer School of Public Health, Munich, Germany
| | - Claudia Flexeder
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Carla P Harris
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospitals, Munich, Germany
| | - Elisabeth Thiering
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospitals, Munich, Germany
| | - Sibylle Koletzko
- Department of Pediatrics, Dr. von Hauner Children's Hospital, LMU University Hospitals, Munich, Germany
- Department of Pediatrics, Gastroenterology and Nutrition, School of Medicine Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | | | - Gerd Schulte-Körne
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Andrea von Berg
- Research Institute, Department of Pediatrics, Marien-Hospital Wesel, Wesel, Germany
| | - Dietrich Berdel
- Research Institute, Department of Pediatrics, Marien-Hospital Wesel, Wesel, Germany
| | - Joachim Heinrich
- Institute and Clinic for Occupational, Social and Environmental Medicine, University Hospital, LMU Munich, Munich, Germany
- Allergy and Lung Health Unit, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Tamara Schikowski
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
15
|
Sinturel F, Chera S, Brulhart-Meynet MC, Montoya JP, Stenvers DJ, Bisschop PH, Kalsbeek A, Guessous I, Jornayvaz FR, Philippe J, Brown SA, D'Angelo G, Riezman H, Dibner C. Circadian organization of lipid landscape is perturbed in type 2 diabetic patients. Cell Rep Med 2023; 4:101299. [PMID: 38016481 PMCID: PMC10772323 DOI: 10.1016/j.xcrm.2023.101299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/26/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023]
Abstract
Lipid homeostasis in humans follows a diurnal pattern in muscle and pancreatic islets, altered upon metabolic dysregulation. We employ tandem and liquid-chromatography mass spectrometry to investigate daily regulation of lipid metabolism in subcutaneous white adipose tissue (SAT) and serum of type 2 diabetic (T2D) and non-diabetic (ND) human volunteers (n = 12). Around 8% of ≈440 lipid metabolites exhibit diurnal rhythmicity in serum and SAT from ND and T2D subjects. The spectrum of rhythmic lipids differs between ND and T2D individuals, with the most substantial changes observed early morning, as confirmed by lipidomics in an independent cohort of ND and T2D subjects (n = 32) conducted at a single morning time point. Strikingly, metabolites identified as daily rhythmic in both serum and SAT from T2D subjects exhibit phase differences. Our study reveals massive temporal and tissue-specific alterations of human lipid homeostasis in T2D, providing essential clues for the development of lipid biomarkers in a temporal manner.
Collapse
Affiliation(s)
- Flore Sinturel
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland
| | - Simona Chera
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland; Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
| | - Marie-Claude Brulhart-Meynet
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Jonathan Paz Montoya
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Dirk Jan Stenvers
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Peter H Bisschop
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Amsterdam Gastroenterology, Endocrinology and Metabolism (AGEM), Amsterdam University Medical Centers, Amsterdam, 1105 AZ, the Netherlands; Laboratory for Endocrinology, Department of Clinical Chemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, 1105 AZ, the Netherlands; Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Arts and Sciences (KNAW), Amsterdam, 1105 BA, the Netherlands
| | - Idris Guessous
- Department and Division of Primary Care Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - François R Jornayvaz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology, Diabetes, Nutrition, and Therapeutic Patient Education, Department of Medicine, University Hospitals of Geneva, 1211 Geneva, Switzerland
| | - Steven A Brown
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Giovanni D'Angelo
- Institute of Bioengineering, School of Life Sciences, EPFL, 1015 Lausanne, Switzerland
| | - Howard Riezman
- Department of Biochemistry, Faculty of Science, NCCR Chemical Biology, University of Geneva, 1211 Geneva, Switzerland
| | - Charna Dibner
- Division of Thoracic and Endocrine Surgery, Department of Surgery, University Hospitals of Geneva, 1211 Geneva, Switzerland; Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland; Institute of Genetics and Genomics in Geneva (iGE3), 1211 Geneva, Switzerland.
| |
Collapse
|
16
|
Mason L, Connolly J, Devenney LE, Lacey K, O’Donovan J, Doherty R. Sleep, Nutrition, and Injury Risk in Adolescent Athletes: A Narrative Review. Nutrients 2023; 15:5101. [PMID: 38140360 PMCID: PMC10745648 DOI: 10.3390/nu15245101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
This narrative review explores the impact of sleep and nutrition on injury risk in adolescent athletes. Sleep is viewed as essential to the recuperation process and is distinguished as an active participant in recovery through its involvement in growth, repair, regeneration, and immunity. Furthermore, the literature has shown that the sleep of athletes impacts elements of athletic performance including both physical and cognitive performance, recovery, injury risk, and mental well-being. For sleep to have a restorative effect on the body, it must meet an individual's sleep needs whilst also lasting for an adequate duration and being of adequate quality, which is age-dependent. The literature has suggested that athletes have increased sleep needs compared to those of the general population and thus the standard recommendations may not be sufficient for athletic populations. Therefore, a more individualised approach accounting for overall sleep health may be more appropriate for addressing sleep needs in individuals including athletes. The literature has demonstrated that adolescent athletes achieve, on average, ~6.3 h of sleep, demonstrating a discrepancy between sleep recommendations (8-10 h) and actual sleep achieved. Sleep-wake cycles undergo development during adolescence whereby adaptation occurs in sleep regulation during this phase. These adaptations increase sleep pressure tolerance and are driven by the maturation of physiological, psychological, and cognitive functioning along with delays in circadian rhythmicity, thus creating an environment for inadequate sleep during adolescence. As such, the adolescent period is a phase of rapid growth and maturation that presents multiple challenges to both sleep and nutrition; consequently, this places a significant burden on an adolescent athletes' ability to recover, thus increasing the likelihood of injury. Therefore, this article aims to provide a comprehensive review of the available literature on the importance of sleep and nutrition interactions in injury risk in adolescent athletes. Furthermore, it provides foundations for informing further investigations exploring the relation of sleep and nutrition interactions to recovery during adolescence.
Collapse
Affiliation(s)
- Lorcán Mason
- Sports Lab North West, Atlantic Technological University Donegal, Port Road, F92 FC93 Letterkenny, Ireland (R.D.)
| | - James Connolly
- Department of Computing, Atlantic Technological University Donegal, Port Road, F92 FC93 Letterkenny, Ireland
| | - Lydia E. Devenney
- Faculty of Arts & Social Sciences, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK
| | - Karl Lacey
- Sports Lab North West, Atlantic Technological University Donegal, Port Road, F92 FC93 Letterkenny, Ireland (R.D.)
| | - Jim O’Donovan
- DCU Glasnevin Campus, Dublin City University, Collins Avenue Extension, Dublin 9, D09 Y8VX Dublin, Ireland
- Sport Ireland Institute, National Sport Campus, Abbotstown, Dublin 15, D15 Y52H Dublin, Ireland
| | - Rónán Doherty
- Sports Lab North West, Atlantic Technological University Donegal, Port Road, F92 FC93 Letterkenny, Ireland (R.D.)
- Sport Ireland Institute, National Sport Campus, Abbotstown, Dublin 15, D15 Y52H Dublin, Ireland
| |
Collapse
|
17
|
Depner CM. Biomarkers linking habitual short sleep duration with risk of cardiometabolic disease: current progress and future directions. FRONTIERS IN SLEEP 2023; 2:1293941. [PMID: 39041043 PMCID: PMC11262587 DOI: 10.3389/frsle.2023.1293941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Approximately one in three adults in the United States sleeps less than the recommended 7 h per night. Decades of epidemiological data and data from experimental sleep restriction studies demonstrate short sleep duration is associated with adverse cardiometabolic risk, including risk of type 2 diabetes and cardiovascular disease. However, the precise mechanisms underlying this risk are not fully elucidated and there is a lack of sleep-based interventions designed to mitigate such risk. One strategy to overcome these limitations is to develop biomarkers that link habitual short sleep duration with adverse cardiometabolic risk. Such biomarkers could inform biochemical mechanisms, identify new targets for interventions, support precision medicine by identifying individuals most likely to benefit from sleep-based interventions, and ultimately lead to improved cardiometabolic health in people with habitual short sleep durations. Early progress demonstrates proof-of-principle that omics-based technologies are a viable approach to create biochemical signatures (biomarkers) of short sleep duration, primarily derived from acute studies of experimental sleep restriction. Yet, much work remains. Notably, studies that translate early findings from experimental sleep restriction to free-living adults with habitual short sleep duration have high potential to advance the field. Such studies also create an exciting opportunity for larger randomized controlled trials that simultaneously identify biomarkers of habitual short sleep duration and evaluate the efficacy of sleep-based interventions. Ultimately, early progress in developing molecular biomarkers of short sleep duration combined with the prior decades of progress in the sleep and metabolism fields provide the foundation for exciting progress in the biomarker development space.
Collapse
Affiliation(s)
- Christopher M. Depner
- Department of Health and Kinesiology, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
18
|
Guo Y, Livelo C, Melkani G. Time-restricted feeding regulates lipid metabolism under metabolic challenges. Bioessays 2023; 45:e2300157. [PMID: 37850554 PMCID: PMC10841423 DOI: 10.1002/bies.202300157] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
Dysregulation of lipid metabolism is a commonly observed feature associated with metabolic syndrome and leads to the development of negative health outcomes such as obesity, diabetes mellitus, non-alcoholic fatty liver disease, or atherosclerosis. Time-restricted feeding/eating (TRF/TRE), an emerging dietary intervention, has been shown to promote pleiotropic health benefits including the alteration of diurnal expression of genes associated with lipid metabolism, as well as levels of lipid species. Although TRF likely induces a response in multiple organs leading to the modulation of lipid metabolism, a majority of the studies related to TRF effects on lipids have focused only on individual tissues, and furthermore there is a lack of insight into potential underlying mechanisms. In this review, we summarize the current insights regarding TRF effects on lipid metabolism and the potential mechanisms in adipose tissue, liver, skeletal muscle, and heart, and conclude by outlining possible avenues for future exploration.
Collapse
Affiliation(s)
- Yiming Guo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher Livelo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Girish Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
19
|
Chen W, Xu Y, Li ZH, Si YC, Wang HY, Bian XL, Li L, Guo ZY, Lai XL. Serum metabolic alterations in peritoneal dialysis patients with excessive daytime sleepiness. Ren Fail 2023; 45:2190815. [PMID: 37051665 PMCID: PMC10116928 DOI: 10.1080/0886022x.2023.2190815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Excessive daytime sleepiness (EDS) is associated with quality of life and all-cause mortality in the end-stage renal disease population. This study aims to identify biomarkers and reveal the underlying mechanisms of EDS in peritoneal dialysis (PD) patients. A total of 48 nondiabetic continuous ambulatory peritoneal dialysis patients were assigned to the EDS group and the non-EDS group according to the Epworth Sleepiness Scale (ESS). Ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UHPLC-Q-TOF/MS) was used to identify the differential metabolites. Twenty-seven (male/female, 15/12; age, 60.1 ± 16.2 years) PD patients with ESS ≥ 10 were assigned to the EDS group, while twenty-one (male/female, 13/8; age, 57.9 ± 10.1 years) PD patients with ESS < 10 were defined as the non-EDS group. With UHPLC-Q-TOF/MS, 39 metabolites with significant differences between the two groups were found, 9 of which had good correlations with disease severity and were further classified into amino acid, lipid and organic acid metabolism. A total of 103 overlapping target proteins of the differential metabolites and EDS were found. Then, the EDS-metabolite-target network and the protein-protein interaction network were constructed. The metabolomics approach integrated with network pharmacology provides new insights into the early diagnosis and mechanisms of EDS in PD patients.
Collapse
Affiliation(s)
- Wei Chen
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Ying Xu
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Zheng-Hao Li
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Military of Education, Naval Medical University, Shanghai, P.R. China
| | - Ya-Chen Si
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Hai-Yan Wang
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Xiao-Lu Bian
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Lu Li
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Zhi-Yong Guo
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| | - Xue-Li Lai
- Department of Nephrology, Shanghai Changhai Hospital, Shanghai, P.R. China
| |
Collapse
|
20
|
Russell KL, Rodman HR, Pak VM. Sleep insufficiency, circadian rhythms, and metabolomics: the connection between metabolic and sleep disorders. Sleep Breath 2023; 27:2139-2153. [PMID: 37147557 DOI: 10.1007/s11325-023-02828-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/06/2023] [Accepted: 04/05/2023] [Indexed: 05/07/2023]
Abstract
PURPOSE US adults who report experiencing insufficient sleep are more likely to suffer from metabolic disorders such as hyperlipidemia, diabetes, and obesity than those with sufficient sleep. Less is understood about the underlying molecular mechanisms connecting these phenomena. A systematic, qualitative review of metabolomics studies exploring metabolic changes in response to sleep insufficiency, sleep deprivation, or circadian disruption was conducted in accordance with PRISMA guidelines. METHODS An electronic literature review in the PubMed database was performed considering publications through May 2021 and screening and eligibility criteria were applied to articles retrieved. The following keywords were used: "metabolomics" and "sleep disorders" or "sleep deprivation" or "sleep disturbance" or "circadian rhythm." After screening and addition of studies included from reference lists of retrieved studies, 16 records were identified for review. RESULTS Consistent changes in metabolites were observed across studies between individuals experiencing sleep deprivation compared to non-sleep deprived controls. Significant increases in phosphatidylcholines, acylcarnitines, sphingolipids, and other lipids were consistent across studies. Increased levels of amino acids such as tryptophan and phenylalanine were also noted. However, studies were limited to small samples of young, healthy, mostly male participants conducted in short inpatient sessions, limiting generalizability. CONCLUSION Changes in lipid and amino acid metabolites accompanying sleep deprivation and/or circadian rhythms may indicate cellular membrane and protein breakdown underlying the connection between sleep disturbance, hyperlipidemia, and other metabolic disorders. Larger epidemiological studies examining changes in the human metabolome in response to chronic insufficient sleep would help elucidate this relationship.
Collapse
Affiliation(s)
| | | | - Victoria M Pak
- Emory Nell Hodgson School of Nursing, Atlanta, GA, USA.
- Emory Rollins School of Public Health, Atlanta, GA, USA.
| |
Collapse
|
21
|
Gombert M, Reisdorph N, Morton SJ, Wright KP, Depner CM. Insufficient sleep and weekend recovery sleep: classification by a metabolomics-based machine learning ensemble. Sci Rep 2023; 13:21123. [PMID: 38036605 PMCID: PMC10689438 DOI: 10.1038/s41598-023-48208-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
Although weekend recovery sleep is common, the physiological responses to weekend recovery sleep are not fully elucidated. Identifying molecular biomarkers that represent adequate versus insufficient sleep could help advance our understanding of weekend recovery sleep. Here, we identified potential molecular biomarkers of insufficient sleep and defined the impact of weekend recovery sleep on these biomarkers using metabolomics in a randomized controlled trial. Healthy adults (n = 34) were randomized into three groups: control (CON: 9-h sleep opportunities); sleep restriction (SR: 5-h sleep opportunities); or weekend recovery (WR: simulated workweek of 5-h sleep opportunities followed by ad libitum weekend recovery sleep and then 2 days with 5-h sleep opportunities). Blood for metabolomics was collected on the simulated Monday immediately following the weekend. Nine machine learning models, including a machine learning ensemble, were built to classify samples from SR versus CON. Notably, SR showed decreased glycerophospholipids and sphingolipids versus CON. The machine learning ensemble showed the highest G-mean performance and classified 50% of the WR samples as insufficient sleep. Our findings show insufficient sleep and recovery sleep influence the plasma metabolome and suggest more than one weekend of recovery sleep may be necessary for the identified biomarkers to return to healthy adequate sleep levels.
Collapse
Affiliation(s)
- Marie Gombert
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010, Valencia, Spain
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - Nichole Reisdorph
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah J Morton
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, 1725 Pleasant Street; Clare Small 114, Boulder, CO, 80309-0354, USA
| | - Kenneth P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, 1725 Pleasant Street; Clare Small 114, Boulder, CO, 80309-0354, USA.
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Christopher M Depner
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, 1725 Pleasant Street; Clare Small 114, Boulder, CO, 80309-0354, USA.
- Department of Health and Kinesiology, University of Utah, 250 S 1850 E; HPER North, RM 206, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
22
|
Barragán R, Zuraikat FM, Cheng B, Scaccia SE, Cochran J, Aggarwal B, Jelic S, St‐Onge M. Paradoxical Effects of Prolonged Insufficient Sleep on Lipid Profile: A Pooled Analysis of 2 Randomized Trials. J Am Heart Assoc 2023; 12:e032078. [PMID: 37815115 PMCID: PMC10757551 DOI: 10.1161/jaha.123.032078] [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: 08/04/2023] [Accepted: 08/15/2023] [Indexed: 10/11/2023]
Abstract
Background Insufficient sleep is associated with increased cardiovascular disease risk, but causality is unclear. We investigated the impact of prolonged mild sleep restriction (SR) on lipid and inflammatory profiles. Methods and Results Seventy-eight participants (56 women [12 postmenopausal]; age, 34.3±12.5 years; body mass index, 25.8±3.5 kg/m2) with habitual sleep duration 7 to 9 h/night (adequate sleep [AS]) underwent two 6-week conditions in a randomized crossover design: AS versus SR (AS-1.5 h/night). Total cholesterol, low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol, triglycerides, and inflammatory markers (CRP [C-reactive protein], interleukin 6, and tumor necrosis factor-α) were assessed. Linear models tested effects of SR on outcomes in the full sample and by sex+menopausal status (premenopausal versus postmenopausal women+men). In the full sample, SR increased high-density lipoprotein cholesterol compared with AS (β=1.2±0.5 mg/dL; P=0.03). Sex+menopausal status influenced the effects of SR on change in total cholesterol (P-interaction=0.04), LDL-C (P-interaction=0.03), and interleukin 6 (P-interaction=0.07). Total cholesterol and LDL-C decreased in SR versus AS in premenopausal women (total cholesterol: β=-4.2±1.9 mg/dL; P=0.03; LDL-C: β=-6.3±2.0 mg/dL; P=0.002). Given paradoxical effects of SR on cholesterol concentrations, we explored associations between changes in inflammation and end point lipids under each condition. Increases in interleukin 6 and tumor necrosis factor-α during SR tended to relate to lower LDL-C in premenopausal women (interleukin 6: β=-5.3±2.6 mg/dL; P=0.051; tumor necrosis factor-α: β=-32.8±14.2 mg/dL; P=0.027). Conclusions Among healthy adults, prolonged insufficient sleep does not increase atherogenic lipids. However, increased inflammation in SR tends to predict lower LDL-C in premenopausal women, resembling the "lipid paradox" in which low cholesterol associates with increased cardiovascular disease risk in proinflammatory conditions. Registration URL: https://www.clinicaltrials.gov; Unique identifiers: NCT02835261, NCT02960776.
Collapse
Affiliation(s)
- Rocío Barragán
- Department of Preventive Medicine and Public HealthUniversity of ValenciaValenciaSpain
- Centro de Investigación Biomédica En Red Fisiopatología de la Obesidad y NutriciónInstituto de Salud Carlos IIIMadridSpain
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
| | - Faris M. Zuraikat
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of General Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
- New York Nutrition Obesity Research CenterColumbia University Irving Medical CenterNew YorkNY
| | - Bin Cheng
- Department of Biostatistics, Mailman School of Public HealthColumbia University Irving Medical CenterNew YorkNY
| | - Samantha E. Scaccia
- Division of Cardiology, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Justin Cochran
- Department of SurgeryColumbia University Irving Medical CenterNew YorkNY
| | - Brooke Aggarwal
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of Cardiology, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Sanja Jelic
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
| | - Marie‐Pierre St‐Onge
- Department of Medicine, Center of Excellence for Sleep and Circadian ResearchColumbia University Irving Medical CenterNew YorkNY
- Division of General Medicine, Department of MedicineColumbia University Irving Medical CenterNew YorkNY
- New York Nutrition Obesity Research CenterColumbia University Irving Medical CenterNew YorkNY
| |
Collapse
|
23
|
Zhuang Z, Dong X, Jia J, Liu Z, Huang T, Qi L. Sleep Patterns, Plasma Metabolome, and Risk of Incident Type 2 Diabetes Mellitus. J Clin Endocrinol Metab 2023; 108:e1034-e1043. [PMID: 37084357 DOI: 10.1210/clinem/dgad218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/06/2023] [Accepted: 04/11/2023] [Indexed: 04/23/2023]
Abstract
CONTEXT A healthy sleep pattern has been related to a lower risk of type 2 diabetes mellitus (T2DM). OBJECTIVE We aimed to identify the metabolomic signature for the healthy sleep pattern and assess its potential causality with T2DM. METHODS This study included 78 659 participants with complete phenotypic data (sleep information and metabolomic measurements) from the UK Biobank study. Elastic net regularized regression was applied to calculate a metabolomic signature reflecting overall sleep patterns. We also performed genome-wide association analysis of the metabolomic signature and one-sample mendelian randomization (MR) with T2DM risk. RESULTS During a median of 8.8 years of follow-up, we documented 1489 incident T2DM cases. Compared with individuals who had an unhealthy sleep pattern, those with a healthy sleep pattern had a 49% lower risk of T2DM (multivariable-adjusted hazard ratio [HR], 0.51; 95% CI, 0.40-0.63). We further constructed a metabolomic signature using elastic net regularized regressions that comprised 153 metabolites, and robustly correlated with sleep pattern (r = 0.19; P = 3×10-325). In multivariable Cox regressions, the metabolomic signature showed a statistically significant inverse association with T2DM risk (HR per SD increment in the signature, 0.56; 95% CI, 0.52-0.60). Additionally, MR analyses indicated a significant causal relation between the genetically predicted metabolomic signature and incident T2DM (P for trend < .001). CONCLUSION In this large prospective study, we identified a metabolomic signature for the healthy sleep pattern, and such a signature showed a potential causality with T2DM risk independent of traditional risk factors.
Collapse
Affiliation(s)
- Zhenhuang Zhuang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Xue Dong
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing 100191, China
| | - Zhonghua Liu
- Department of Biostatistics, Columbia University, New York, NY 10027-6902, USA
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing 100191, China
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing 100191, China
- Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing 100191, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| |
Collapse
|
24
|
Rasaei N, Samadi M, Khadem A, Badrooj N, Hassan Zadeh M, Ghaffarian-Ensaf R, Gholami F, Mirzaei K. The association between cholesterol/saturated fat index (CSI) and quality of sleep, and circadian rhythm among overweight and obese women: a cross-sectional study. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2023; 42:75. [PMID: 37501196 PMCID: PMC10375646 DOI: 10.1186/s41043-023-00414-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND The decline in sleep quality is one of the main public health problems affecting the global population. Some studies have shown that a high-fat diet may be linked to changes in circadian rhythm and sleep quality. The cholesterol/saturated fatty acid index (CSI) determines the amount of cholesterol and saturated fatty acid (SFA) in people's dietary patterns and can affect the quality of sleep and circadian rhythm. However, to date, no studies have investigated the effect of this index on these two variables. Therefore, our aim was to investigate the relationship between CSI on circadian rhythm and sleep quality in obese and overweight women. METHOD This cross-sectional study included 378 adult women who were obese or overweight. Using accepted techniques, anthropometric measurements, blood pressure readings, and biochemical variables were evaluated. A validated and trustworthy semi-quantitative food frequency questionnaire (FFQ 147 items) was used to gauge dietary intake. The CSI was measured to find out how much cholesterol and saturated fats were in the diet. Additionally, to assess circadian rhythm and sleep quality, respectively, the valid morning-evening questionnaire (MEQ) and Pittsburgh sleep quality index (PSQI) questionnaires were utilized. RESULT The results of the multinomial logistic regression model of our analysis showed that a significant association was observed between circadian rhythm status with CSI score, and participants with one higher unit of CSI had a 7.3% more chance of being in the eveningness group than being in morningness category in the crude model (OR: 1.07; 95% CI 1.00, 1.14; P = 0.026). This association remains marginally significant when adjusting for age, energy intake, BMI, job status, thyroid, and smoking status (OR = 1.08; 95% CI 1.00, 1.16; P = 0.051). The binary logistic regression model showed that after controlling for potentially confounding variables, there was no significant association between sleep quality with CSI score, however, those with one higher unit of CSI had 1.6% more chance of having sleep problems (OR: 1.01; 95% CI 0.96, 1.06; P = 0.503). CONCLUSION Our results indicated a direct marginally significant association between CSI with evening type in overweight and obese women. Future studies are needed to clarify the precise link between circadian rhythm and sleep behavior with fatty acid quality index.
Collapse
Affiliation(s)
- Niloufar Rasaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Mahsa Samadi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Alireza Khadem
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Negin Badrooj
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Mohadeseh Hassan Zadeh
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fatemeh Gholami
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), P.O. Box: 14155-6117, Tehran, Iran.
| |
Collapse
|
25
|
Rahman SA, Gathungu RM, Marur VR, St Hilaire MA, Scheuermaier K, Belenky M, Struble JS, Czeisler CA, Lockley SW, Klerman EB, Duffy JF, Kristal BS. Age-related changes in circadian regulation of the human plasma lipidome. Commun Biol 2023; 6:756. [PMID: 37474677 PMCID: PMC10359364 DOI: 10.1038/s42003-023-05102-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 07/06/2023] [Indexed: 07/22/2023] Open
Abstract
Aging alters the amplitude and phase of centrally regulated circadian rhythms. Here we evaluate whether peripheral circadian rhythmicity in the plasma lipidome is altered by aging through retrospective lipidomics analysis on plasma samples collected in 24 healthy individuals (9 females; mean ± SD age: 40.9 ± 18.2 years) including 12 younger (4 females, 23.5 ± 3.9 years) and 12 middle-aged older, (5 females, 58.3 ± 4.2 years) individuals every 3 h throughout a 27-h constant routine (CR) protocol, which allows separating evoked changes from endogenously generated oscillations in physiology. Cosinor regression shows circadian rhythmicity in 25% of lipids in both groups. On average, the older group has a ~14% lower amplitude and a ~2.1 h earlier acrophase of the lipid circadian rhythms (both, p ≤ 0.001). Additionally, more rhythmic circadian lipids have a significant linear component in addition to the sinusoidal across the 27-h CR in the older group (44/56) compared to the younger group (18/58, p < 0.0001). Results from individual-level data are consistent with group-average results. Results indicate that prevalence of endogenous circadian rhythms of the human plasma lipidome is preserved with healthy aging into middle-age, but significant changes in rhythmicity include a reduction in amplitude, earlier acrophase, and an altered temporal relationship between central and lipid rhythms.
Collapse
Grants
- R01 HL128538 NHLBI NIH HHS
- T32 HL007901 NHLBI NIH HHS
- R01 AG006072 NIA NIH HHS
- R01 HD107064 NICHD NIH HHS
- U01 NS114001 NINDS NIH HHS
- R01 HL132556 NHLBI NIH HHS
- UL1 TR001102 NCATS NIH HHS
- UL1 RR025758 NCRR NIH HHS
- R01 HL162102 NHLBI NIH HHS
- R01 HL166205 NHLBI NIH HHS
- R01 HL159207 NHLBI NIH HHS
- U54 AG062322 NIA NIH HHS
- R01 NS114526 NINDS NIH HHS
- R01 HL140335 NHLBI NIH HHS
- R01 HL114088 NHLBI NIH HHS
- R01 NS099055 NINDS NIH HHS
- R21 DA052861 NIDA NIH HHS
- R03 AG071922 NIA NIH HHS
- The work was supported by grants from the NIH: R01-HL132556 (BSK), R01-HL140335 (BSK), R01-HL114088 (EBK), R01-AG06072 (JFD), and R01-HL159207 (SAR). KS was supported by a T32 HL07901 and a NIA F32 AG316902. EBK was supported by NIH R01NS099055, U01NS114001, U54AG062322, R21DA052861, R21DA052861, R01NS114526-02S1, R01-HD107064, DoD W81XWH201076; and Leducq Foundation for Cardiovascular Research. The clinical research projects described were supported by NIH grant 1UL1 TR001102-01, 8UL1TR000170-05, UL1 RR025758, Harvard Clinical and Translational Science Center, from the National Center for Advancing Translational Science. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Center for Research Resources, the National Center for Advancing Translational Science or the National Institutes of Health.
Collapse
Affiliation(s)
- Shadab A Rahman
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Rose M Gathungu
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Enara Bio, The Magdalen Centre, Oxford Science Park, 1 Robert Robinson Avenue, Oxford, OX4 4GA, UK
| | - Vasant R Marur
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Quantitative Biosciences, Merck & Co., Inc, 320 Bent St, Cambridge, MA, 02141, USA
| | - Melissa A St Hilaire
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Department of Computer and Data Sciences, School of Science and Engineering, Merrimack College, 315 Turnpike Street, North Andover, MA, 01845, USA
| | - Karine Scheuermaier
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| | - Marina Belenky
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Jackson S Struble
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Charles A Czeisler
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Steven W Lockley
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Elizabeth B Klerman
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Jeanne F Duffy
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Bruce S Kristal
- Division of Sleep and Circadian Disorders, Departments of Medicine and Neurology, Brigham and Women's Hospital, 221 Longwood Ave, Boston, MA, 02115, USA.
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
26
|
Li Y, Haynes P, Zhang SL, Yue Z, Sehgal A. Ecdysone acts through cortex glia to regulate sleep in Drosophila. eLife 2023; 12:e81723. [PMID: 36719183 PMCID: PMC9928426 DOI: 10.7554/elife.81723] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/30/2023] [Indexed: 02/01/2023] Open
Abstract
Steroid hormones are attractive candidates for transmitting long-range signals to affect behavior. These lipid-soluble molecules derived from dietary cholesterol easily penetrate the brain and act through nuclear hormone receptors (NHRs) that function as transcription factors. To determine the extent to which NHRs affect sleep:wake cycles, we knocked down each of the 18 highly conserved NHRs found in Drosophila adults and report that the ecdysone receptor (EcR) and its direct downstream NHR Eip75B (E75) act in glia to regulate the rhythm and amount of sleep. Given that ecdysone synthesis genes have little to no expression in the fly brain, ecdysone appears to act as a long-distance signal and our data suggest that it enters the brain more at night. Anti-EcR staining localizes to the cortex glia in the brain and functional screening of glial subtypes revealed that EcR functions in adult cortex glia to affect sleep. Cortex glia are implicated in lipid metabolism, which appears to be relevant for actions of ecdysone as ecdysone treatment mobilizes lipid droplets (LDs), and knockdown of glial EcR results in more LDs. In addition, sleep-promoting effects of exogenous ecdysone are diminished in lsd-2 mutant flies, which are lean and deficient in lipid accumulation. We propose that ecdysone is a systemic secreted factor that modulates sleep by stimulating lipid metabolism in cortex glia.
Collapse
Affiliation(s)
- Yongjun Li
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Department of Biology, University of PennsylvaniaPhiladelphiaUnited States
| | - Paula Haynes
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Department of Pharmacology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Shirley L Zhang
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Zhifeng Yue
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Amita Sehgal
- Howard Hughes Medical Institute and Chronobiology and Sleep Institute, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
27
|
Fritz J, Huang T, Depner CM, Zeleznik OA, Cespedes Feliciano EM, Li W, Stone KL, Manson JE, Clish C, Sofer T, Schernhammer E, Rexrode K, Redline S, Wright KP, Vetter C. Sleep duration, plasma metabolites, and obesity and diabetes: a metabolome-wide association study in US women. Sleep 2023; 46:zsac226. [PMID: 36130143 PMCID: PMC9832513 DOI: 10.1093/sleep/zsac226] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/08/2022] [Indexed: 01/16/2023] Open
Abstract
Short and long sleep duration are associated with adverse metabolic outcomes, such as obesity and diabetes. We evaluated cross-sectional differences in metabolite levels between women with self-reported habitual short (<7 h), medium (7-8 h), and long (≥9 h) sleep duration to delineate potential underlying biological mechanisms. In total, 210 metabolites were measured via liquid chromatography-mass spectrometry in 9207 women from the Nurses' Health Study (NHS; N = 5027), the NHSII (N = 2368), and the Women's Health Initiative (WHI; N = 2287). Twenty metabolites were consistently (i.e. praw < .05 in ≥2 cohorts) and/or strongly (pFDR < .05 in at least one cohort) associated with short sleep duration after multi-variable adjustment. Specifically, levels of two lysophosphatidylethanolamines, four lysophosphatidylcholines, hydroxyproline and phenylacetylglutamine were higher compared to medium sleep duration, while levels of one diacylglycerol and eleven triacylglycerols (TAGs; all with ≥3 double bonds) were lower. Moreover, enrichment analysis assessing associations of metabolites with short sleep based on biological categories demonstrated significantly increased acylcarnitine levels for short sleep. A metabolite score for short sleep duration based on 12 LASSO-regression selected metabolites was not significantly associated with prevalent and incident obesity and diabetes. Associations of single metabolites with long sleep duration were less robust. However, enrichment analysis demonstrated significant enrichment scores for four lipid classes, all of which (most markedly TAGs) were of opposite sign than the scores for short sleep. Habitual short sleep exhibits a signature on the human plasma metabolome which is different from medium and long sleep. However, we could not detect a direct link of this signature with obesity and diabetes risk.
Collapse
Affiliation(s)
- Josef Fritz
- Circadian and Sleep Epidemiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Innsbruck, Austria
| | - Tianyi Huang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Christopher M Depner
- Department of Health and Kinesiology, University of Utah, Salt Lake City, UT, USA
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Wenjun Li
- Department of Public Health, School of Health Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Clary Clish
- Metabolomics Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, USA
| | - Eva Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
| | - Kathryn Rexrode
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Division of Women’s Health, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Susan Redline
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kenneth P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Céline Vetter
- Circadian and Sleep Epidemiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
28
|
Tremblay MÈ, Almsherqi ZA, Deng Y. Plasmalogens and platelet-activating factor roles in chronic inflammatory diseases. Biofactors 2022; 48:1203-1216. [PMID: 36370412 DOI: 10.1002/biof.1916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022]
Abstract
Fatty acids and phospholipid molecules are essential for determining the structure and function of cell membranes, and they hence participate in many biological processes. Platelet activating factor (PAF) and its precursor plasmalogen, which represent two subclasses of ether phospholipids, have attracted increasing research attention recently due to their association with multiple chronic inflammatory, neurodegenerative, and metabolic disorders. These pathophysiological conditions commonly involve inflammatory processes linked to an excess presence of PAF and/or decreased levels of plasmalogens. However, the molecular mechanisms underlying the roles of plasmalogens in inflammation have remained largely elusive. While anti-inflammatory responses most likely involve the plasmalogen signal pathway; pro-inflammatory responses recruit arachidonic acid, a precursor of pro-inflammatory lipid mediators which is released from membrane phospholipids, notably derived from the hydrolysis of plasmalogens. Plasmalogens per se are vital membrane phospholipids in humans. Changes in their homeostatic levels may alter cell membrane properties, thus affecting key signaling pathways that mediate inflammatory cascades and immune responses. The plasmalogen analogs of PAF are also potentially important, considering that anti-PAF activity has strong anti-inflammatory effects. Plasmalogen replacement therapy was further identified as a promising anti-inflammatory strategy allowing for the relief of pathological hallmarks in patients affected by chronic diseases with an inflammatory component. The aim of this Short Review is to highlight the emerging roles and implications of plasmalogens in chronic inflammatory disorders, along with the promising outcomes of plasmalogen replacement therapy for the treatment of various PAF-related chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, Canada
- Department of Molecular Medicine, Université de Laval, Québec City, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
| | - Zakaria A Almsherqi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yuru Deng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| |
Collapse
|
29
|
Güldür T. Potential linkages between circadian rhythm and membrane lipids: timekeeper and bilayer. BIOL RHYTHM RES 2022. [DOI: 10.1080/09291016.2022.2096756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Tayfun Güldür
- Medical Biochemistry Department, Faculty of Medicine, Inönü University, Malatya, Turkey
| |
Collapse
|
30
|
Al-Musharaf S. Changes in Sleep Patterns during Pregnancy and Predictive Factors: A Longitudinal Study in Saudi Women. Nutrients 2022; 14:nu14132633. [PMID: 35807814 PMCID: PMC9268456 DOI: 10.3390/nu14132633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to assess sleep patterns during the three trimesters of pregnancy and whether vitamin D concentrations, along with other risk factors, are associated with these alterations. In a longitudinal study, 140 pregnant women (age 18 to 39 years) were followed throughout their first, second, and third trimesters. Sleep was measured using the Pittsburgh Sleep Quality Index (PSQI) at each trimester, along with an assessment of biochemical parameters, including serum vitamin D levels. The information that was collected included anthropometric data, socio-economic status, dietary intake, and physical activity. The PSQI was higher in mid and late pregnancy than in early pregnancy (both p = 0.001), and the sleep duration was also higher in late versus early pregnancy. Linear regression analyses revealed independent predictors of deteriorating sleep quality from early to late pregnancy, including low income (B ± SE −0.60 ± 0.26, p = 0.03) and low serum vitamin D levels in the second trimester (B ± SE −0.20 ± 0.01, p = 0.04). Energy intake and sitting in the second half of pregnancy were positively associated with changes in the PSQI score from the second to third trimesters (B ± SE 0.15 ± 0.07, p = 0.048) and (B ± SE 0.01 ± 0.00, p = 0.044), respectively. Low socio-economic status, low serum vitamin D levels, greater energy intake, and sitting time were associated with worsening patterns of sleep quality from early to late pregnancy.
Collapse
Affiliation(s)
- Sara Al-Musharaf
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
31
|
Circadian rhythm of lipid metabolism. Biochem Soc Trans 2022; 50:1191-1204. [PMID: 35604112 DOI: 10.1042/bst20210508] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Lipids comprise a diverse group of metabolites that are indispensable as energy storage molecules, cellular membrane components and mediators of inter- and intra-cellular signaling processes. Lipid homeostasis plays a crucial role in maintaining metabolic health in mammals including human beings. A growing body of evidence suggests that the circadian clock system ensures temporal orchestration of lipid homeostasis, and that perturbation of such diurnal regulation leads to the development of metabolic disorders comprising obesity and type 2 diabetes. In view of the emerging role of circadian regulation in maintaining lipid homeostasis, in this review, we summarize the current knowledge on lipid metabolic pathways controlled by the mammalian circadian system. Furthermore, we review the emerging connection between the development of human metabolic diseases and changes in lipid metabolites that belong to major classes of lipids. Finally, we highlight the mechanisms underlying circadian organization of lipid metabolic rhythms upon the physiological situation, and the consequences of circadian clock dysfunction for dysregulation of lipid metabolism.
Collapse
|
32
|
Kyle JE, Bramer LM, Claborne D, Stratton KG, Bloodsworth KJ, Teeguarden JG, Gaddameedhi S, Metz TO, Van Dongen HPA. Simulated Night-Shift Schedule Disrupts the Plasma Lipidome and Reveals Early Markers of Cardiovascular Disease Risk. Nat Sci Sleep 2022; 14:981-994. [PMID: 35645584 PMCID: PMC9133431 DOI: 10.2147/nss.s363437] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/28/2022] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION The circadian system coordinates daily rhythms in lipid metabolism, storage and utilization. Disruptions of internal circadian rhythms due to altered sleep/wake schedules, such as in night-shift work, have been implicated in increased risk of cardiovascular disease and metabolic disorders. To determine the impact of a night-shift schedule on the human blood plasma lipidome, an in-laboratory simulated shift work study was conducted. METHODS Fourteen healthy young adults were assigned to 3 days of either a simulated day or night-shift schedule, followed by a 24-h constant routine protocol with fixed environmental conditions, hourly isocaloric snacks, and constant wakefulness to investigate endogenous circadian rhythms. Blood plasma samples collected at 3-h intervals were subjected to untargeted lipidomics analysis. RESULTS More than 400 lipids were identified and quantified across 21 subclasses. Focusing on lipids with low between-subject variation per shift condition, alterations in the circulating plasma lipidome revealed generally increased mean triglyceride levels and decreased mean phospholipid levels after night-shift relative to day-shift. The circadian rhythms of triglycerides containing odd chain fatty acids peaked earlier during constant routine after night-shift. Regardless of shift condition, triglycerides tended to either peak or be depleted at 16:30 h, with chain-specific differences associated with the direction of change. DISCUSSION The simulated night-shift schedule was associated with altered temporal patterns in the lipidome. This may be premorbid to the elevated cardiovascular risk that has been found epidemiologically in night-shift workers.
Collapse
Affiliation(s)
- Jennifer E Kyle
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Lisa M Bramer
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Daniel Claborne
- Computing and Analytics Division, National Security Directorate, PNNL, Richland, WA, 99352, USA
| | - Kelly G Stratton
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Kent J Bloodsworth
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Justin G Teeguarden
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA.,Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, 97331, USA
| | - Shobhan Gaddameedhi
- Department of Biological Sciences and Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Thomas O Metz
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory (PNNL), Richland, WA, 99352, USA
| | - Hans P A Van Dongen
- Sleep and Performance Research Center, Washington State University, Spokane, WA, 99202, USA.,Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| |
Collapse
|
33
|
Role of circadian rhythm and impact of circadian rhythm disturbance on the metabolism and disease. J Cardiovasc Pharmacol 2021; 79:254-263. [PMID: 34840256 DOI: 10.1097/fjc.0000000000001178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 10/23/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Molecular circadian clocks exist in almost all cells of the organism and operate for approximately 24 h, maintain the normal physiological and behavioral body processes and regulate metabolism of many cells related to a variety of disease states. Circadian rhythms regulate metabolism, mainly including neurotransmitters, hormones, amino acids and lipids. Circadian misalignment is related to metabolic syndromes, such as obesity, diabetes and hypertension, which have reached an alarming level in modern society. We reviewed the mechanism of the circadian clock and the interaction between circadian rhythm and metabolism, as well as circadian rhythm disturbance on the metabolism of hypertension, obesity and diabetes. Finally, we discuss how to use the circadian rhythm to prevent diseases. Thus, this review is a micro to macro discussion from the perspective of circadian rhythm and aims to provide basic ideas for circadian rhythm research and disease therapies.
Collapse
|
34
|
Mota MC, Silva CM, Balieiro LCT, Fahmy WM, Marqueze EC, Moreno CRDC, Crispim CA. Social Jetlag Is Associated With Impaired Metabolic Control During a 1-Year Follow-Up. Front Physiol 2021; 12:702769. [PMID: 34539431 PMCID: PMC8445111 DOI: 10.3389/fphys.2021.702769] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/30/2021] [Indexed: 11/13/2022] Open
Abstract
Previous studies have identified social jetlag (SJL) as a risk factor for non-communicable chronic diseases (NCCDs), but its association with metabolic control over time is unclear in the literature. Therefore, we examined the influence of SJL on metabolic parameters and blood pressure (BP) in patients with NCCDs over a 1-year follow-up. This retrospective, longitudinal study included 625 individuals (age: 56.0 +12.0 years; 76% female) with NCCDs [type 2 diabetes mellitus (TD2), systemic arterial hypertension (SHA), obesity, or dyslipidemia]. SJL was calculated based on the absolute difference between mid-sleep time on weekends and weekdays. Current metabolic parameters and BP of the patients were compared with data from a year prior. Generalized estimating equations (GEE) and multiple linear regression analyses were used to examine the association among SJL, metabolic parameters, and BP. Multiple linear regression analyses adjusted for confounders showed that SJL was positively associated with the delta difference of fasting glucose (β = 0.11, p = 0.02) and triglyceride levels (β = 0.09, p = 0.04) among all subjects with NCCDs, and with fasting glucose (β = 0.30, p = 0.0001) and triglyceride levels (β = 0.22, p = 0.01) in the TD2 group. GEE analysis demonstrated an isolated effect of SJL on diastolic BP. High SJL impaired clinical and metabolic control in individuals with NCCDs, leading to a worse profile after a 1-year follow-up, particularly among type II diabetics.
Collapse
Affiliation(s)
- Maria Carliana Mota
- Faculty of Medicine of the Federal University of Uberlândia, Uberlândia, Brazil
| | | | | | | | - Elaine Cristina Marqueze
- Public Health Graduate Program, Department of Epidemiology, Catholic University of Santos, Santos, Brazil.,Department of Health, Life Cycles and Society, School of Public Health, University of São Paulo, São Paulo, Brazil
| | - Claudia Roberta de Castro Moreno
- Department of Health, Life Cycles and Society, School of Public Health, University of São Paulo, São Paulo, Brazil.,Stress Research Institute, Department of Psychology, Stockholm University, Stockholm, Sweden
| | | |
Collapse
|
35
|
Cogswell D, Bisesi P, Markwald RR, Cruickshank-Quinn C, Quinn K, McHill A, Melanson EL, Reisdorph N, Wright KP, Depner CM. Identification of a Preliminary Plasma Metabolome-based Biomarker for Circadian Phase in Humans. J Biol Rhythms 2021; 36:369-383. [PMID: 34182829 PMCID: PMC9134127 DOI: 10.1177/07487304211025402] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Measuring individual circadian phase is important to diagnose and treat circadian rhythm sleep-wake disorders and circadian misalignment, inform chronotherapy, and advance circadian science. Initial findings using blood transcriptomics to predict the circadian phase marker dim-light melatonin onset (DLMO) show promise. Alternatively, there are limited attempts using metabolomics to predict DLMO and no known omics-based biomarkers predict dim-light melatonin offset (DLMOff). We analyzed the human plasma metabolome during adequate and insufficient sleep to predict DLMO and DLMOff using one blood sample. Sixteen (8 male/8 female) healthy participants aged 22.4 ± 4.8 years (mean ± SD) completed an in-laboratory study with 3 baseline days (9 h sleep opportunity/night), followed by a randomized cross-over protocol with 9-h adequate sleep and 5-h insufficient sleep conditions, each lasting 5 days. Blood was collected hourly during the final 24 h of each condition to independently determine DLMO and DLMOff. Blood samples collected every 4 h were analyzed by untargeted metabolomics and were randomly split into training (68%) and test (32%) sets for biomarker analyses. DLMO and DLMOff biomarker models were developed using partial least squares regression in the training set followed by performance assessments using the test set. At baseline, the DLMOff model showed the highest performance (0.91 R2 and 1.1 ± 1.1 h median absolute error ± interquartile range [MdAE ± IQR]), with significantly (p < 0.01) lower prediction error versus the DLMO model. When all conditions (baseline, 9 h, and 5 h) were included in performance analyses, the DLMO (0.60 R2; 2.2 ± 2.8 h MdAE; 44% of the samples with an error under 2 h) and DLMOff (0.62 R2; 1.8 ± 2.6 h MdAE; 51% of the samples with an error under 2 h) models were not statistically different. These findings show promise for metabolomics-based biomarkers of circadian phase and highlight the need to test biomarkers that predict multiple circadian phase markers under different physiological conditions.
Collapse
Affiliation(s)
- D Cogswell
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, Colorado
| | - P Bisesi
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, Colorado
| | - R R Markwald
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, Colorado
| | - C Cruickshank-Quinn
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - K Quinn
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - A McHill
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, Colorado
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon
| | - E L Melanson
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Eastern Colorado Veterans Affairs Geriatric Research, Education, and Clinical Center, Denver, Colorado
| | - N Reisdorph
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - K P Wright
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, Colorado
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - C M Depner
- Sleep and Chronobiology Laboratory, University of Colorado, Boulder, Boulder, Colorado
- Department of Health and Kinesiology, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
36
|
Rukmini AV, Jos AM, Yeo SC, Lee N, Mo D, Mohapatra L, Karamchedu S, Gooley JJ. Circadian regulation of breath alcohol concentration. Sleep 2021; 44:6030924. [PMID: 33305816 DOI: 10.1093/sleep/zsaa270] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/11/2020] [Indexed: 11/13/2022] Open
Abstract
STUDY OBJECTIVES The role of the circadian clock in regulating blood/breath alcohol levels after consuming alcohol is uncertain. Our goal was to evaluate the degree to which the circadian system regulates breath alcohol concentration (BrAC) pharmacokinetic parameters. METHODS Twenty healthy adults aged 21-30 years took part in a 4-day laboratory study. A 40-h constant routine procedure was used to assess circadian rhythms. Every 4 h, participants were given a fixed oral dose of alcohol with breathalyzer measurements taken every 5 min to construct BrAC curves. Sinusoidal models were used to test for circadian variation of the peak BrAC, the time to reach peak BrAC, the absorption rate, the elimination rate, and the time for BrAC to return to zero after alcohol was ingested. RESULTS A significant circadian rhythm was detected for group-averaged peak BrAC values and the time for BrAC to return to zero, but not other BrAC variables. Peak BrAC values were lowest in the evening near the peak of the core body temperature rhythm and nadir of the salivary cortisol rhythm. Peak BrAC values increased during the night and reached their highest levels in the morning and afternoon. The time needed for BrAC to return to zero was also longest in the late morning and afternoon. CONCLUSION The circadian system modulates some BrAC pharmacokinetic parameters. In normally entrained individuals, taking the same oral dose of alcohol at different times of day can result in different BrAC responses. These findings have potential implications for alcohol-related accidents and alcohol toxicity.
Collapse
Affiliation(s)
- A V Rukmini
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Anna Mini Jos
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Sing-Chen Yeo
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Noel Lee
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Di Mo
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Litali Mohapatra
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Swathy Karamchedu
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Joshua J Gooley
- Neuroscience and Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| |
Collapse
|
37
|
Hancox TPM, Skene DJ, Dallmann R, Dunn WB. Tick-Tock Consider the Clock: The Influence of Circadian and External Cycles on Time of Day Variation in the Human Metabolome-A Review. Metabolites 2021; 11:328. [PMID: 34069741 PMCID: PMC8161100 DOI: 10.3390/metabo11050328] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
The past decade has seen a large influx of work investigating time of day variation in different human biofluid and tissue metabolomes. The driver of this daily variation can be endogenous circadian rhythms driven by the central and/or peripheral clocks, or exogenous diurnal rhythms driven by behavioural and environmental cycles, which manifest as regular 24 h cycles of metabolite concentrations. This review, of all published studies to date, establishes the extent of daily variation with regard to the number and identity of 'rhythmic' metabolites observed in blood, saliva, urine, breath, and skeletal muscle. The probable sources driving such variation, in addition to what metabolite classes are most susceptible in adhering to or uncoupling from such cycles is described in addition to a compiled list of common rhythmic metabolites. The reviewed studies show that the metabolome undergoes significant time of day variation, primarily observed for amino acids and multiple lipid classes. Such 24 h rhythms, driven by various factors discussed herein, are an additional source of intra/inter-individual variation and are thus highly pertinent to all studies applying untargeted and targeted metabolomics platforms, particularly for the construction of biomarker panels. The potential implications are discussed alongside proposed minimum reporting criteria suggested to acknowledge time of day variation as a potential influence of results and to facilitate improved reproducibility.
Collapse
Affiliation(s)
- Thomas P. M. Hancox
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Debra J. Skene
- Chronobiology, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK;
| | - Robert Dallmann
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK;
| | - Warwick B. Dunn
- Institute of Metabolism and Systems Research, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
38
|
Reutrakul S, Chen H, Chirakalwasan N, Charoensri S, Wanitcharoenkul E, Amnakkittikul S, Saetung S, Layden BT, Chlipala GE. Metabolomic profile associated with obstructive sleep apnoea severity in obese pregnant women with gestational diabetes mellitus: A pilot study. J Sleep Res 2021; 30:e13327. [PMID: 33792106 DOI: 10.1111/jsr.13327] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/16/2021] [Indexed: 12/16/2022]
Abstract
Obstructive sleep apnoea (OSA) is prevalent in obese women with gestational diabetes mellitus (GDM). The present pilot study explored associations between OSA severity and metabolites in women with GDM. A total of 81 obese women with diet-controlled GDM had OSA assessment (median gestational age [GA] 29 weeks). The metabolic profile was assayed from fasting serum samples via liquid chromatography-mass spectrometry (LC-MS) using an untargeted approach. Metabolites were extracted and subjected to an Agilent 1,290 UPLC coupled to an Agilent 6,545 quadrupole time-of-flight (Q-TOF) MS. Data were acquired using electrospray ionisation in positive and negative ion modes. The raw LC-MS data were processed using the OpenMS toolkit to detect and quantify features, and these features were annotated using the Human Metabolite Database. The feature data were compared with OSA status, apnea-hypopnea index (AHI), body mass index (BMI) and GA using "limma" in R. Correlation analyses of the continuous covariates were performed using Kendall's Tau test. The p values were adjusted for multiple testing using the Benjamini-Hochberg false discovery rate correction. A total of 42 women (51.8%) had OSA, with a median AHI of 9.1 events/hr. There were no significant differences in metabolomics profiles between those with and without OSA. However, differential analyses modelling in GA and BMI found 12 features that significantly associated with the AHI. These features could be annotated to oestradiols, lysophospholipids, and fatty acids, with higher levels related to higher AHI. Metabolites including oestradiols and phospholipids may be involved in pathogenesis of OSA in pregnant women with GDM. A targeted approach may help elucidate our understanding of their role in OSA in this population.
Collapse
Affiliation(s)
- Sirimon Reutrakul
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Hui Chen
- Mass Spectrometry Core, Research Resource Center, Office of Vice Chancellor for Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Naricha Chirakalwasan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Excellence Center for Sleep Disorders, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Suranut Charoensri
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Ekasitt Wanitcharoenkul
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Somvang Amnakkittikul
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sunee Saetung
- Division of Endocrinology and Metabolism, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Brian T Layden
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - George E Chlipala
- Research Informatics Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
39
|
Variability of Lipids in Human Milk. Metabolites 2021; 11:metabo11020104. [PMID: 33670205 PMCID: PMC7916976 DOI: 10.3390/metabo11020104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/02/2021] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Lipids in breastmilk play a critical role in infant growth and development. However, few studies have investigated sources of variability of both high- and low-abundant milk lipids. The objective of our study was to investigate individual and morning-evening differences in the human milk lipidome. In this study, a modified two-phase method (MTBE: Methanol 7:2) was validated for the extraction of lipids from human breastmilk. This method was then applied to samples from a group of 20 healthy women to measure inter- and intra-individual (morning versus evening) variability of the breastmilk lipidome. We report here the levels of 237 lipid species from 13 sub-classes using reversed-phase liquid chromatography mass spectrometry (RP-LCMS) and direct-infusion mass spectrometry (DI-MS). About 85% of lipid species showed stable inter-individual differences across time points. Half of lipid species showed higher concentrations in the evening compared with the morning, with phosphatidylethanolamines (PEs) and triacylglycerols (TAGs) exhibiting the largest changes. In morning and evening samples, the biological variation was greater for diacylglycerols (DAGs) and TAGs compared with phospholipids and sphingolipids, and the variation in DAGs and TAGs was greater in evening samples compared with morning samples. These results demonstrate that variation in the milk lipidome is strongly influenced by individual differences and time of day.
Collapse
|
40
|
Xing C, Huang X, Zhang Y, Zhang C, Wang W, Wu L, Ding M, Zhang M, Song L. Sleep Disturbance Induces Increased Cholesterol Level by NR1D1 Mediated CYP7A1 Inhibition. Front Genet 2020; 11:610496. [PMID: 33424933 PMCID: PMC7793681 DOI: 10.3389/fgene.2020.610496] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 12/03/2020] [Indexed: 01/02/2023] Open
Abstract
Disturbed sleep is closely associated with an increased risk of metabolic diseases. However, the underlying mechanisms of circadian clock genes linking sleep and lipid profile abnormalities have not been fully elucidated. This study aimed to explore the important role of the circadian clock in regulating impaired cholesterol metabolism at an early stage of sleep deprivation (SD). Sleep disturbance was conducted using an SD instrument. Our results showed that SD increased the serum cholesterol levels. Concentrations of serum leptin and resistin were much lower after SD, but other metabolic hormone concentrations (adiponectin, glucagon, insulin, thyroxine, norepinephrine, and epinephrine) were unchanged before and after SD. Warning signs of cardiovascular diseases [decreased high density lipoprotein (HDL)-cholesterol and increased corticosterone and 8-hydroxyguanosine levels] and hepatic cholestasis (elevated total bile acids and bilirubin levels) were observed after SD. Cholesterol accumulation was also observed in the liver after SD. The expression levels of HMGCR, the critical enzyme for cholesterol synthesis, remained unchanged in the liver. However, the expression levels of liver CYP7A1, the enzyme responsible for the conversion of cholesterol into bile acids, significantly reduced after SD. Furthermore, expression of NR1D1, a circadian oscillator and transcriptional regulator of CYP7A1, strikingly decreased after SD. Moreover, NR1D1 deficiency decreased liver CYP7A1 levels, and SD could exacerbate the reduction of CYP7A1 expression in NR1D1-/- mouse livers. Additionally, NR1D1 deficiency could further increase serum cholesterol levels under SD. These results suggest that sleep disturbance can induce increased serum cholesterol levels and liver cholesterol accumulation by NR1D1 mediated CYP7A1 inhibition.
Collapse
Affiliation(s)
- Chen Xing
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Xin Huang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Yifan Zhang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Chongchong Zhang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,School of Basic Medicine, Henan University, Kaifeng, China
| | - Wei Wang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China.,School of Pharmacy, Jiamusi University, Jiamusi, China
| | - Lin Wu
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Mengnan Ding
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Min Zhang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Lun Song
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
41
|
Depner CM, Cogswell DT, Bisesi PJ, Markwald RR, Cruickshank-Quinn C, Quinn K, Melanson EL, Reisdorph N, Wright KP. Developing preliminary blood metabolomics-based biomarkers of insufficient sleep in humans. Sleep 2020; 43:zsz321. [PMID: 31894238 PMCID: PMC7355401 DOI: 10.1093/sleep/zsz321] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/27/2019] [Indexed: 01/20/2023] Open
Abstract
STUDY OBJECTIVE Identify small molecule biomarkers of insufficient sleep using untargeted plasma metabolomics in humans undergoing experimental insufficient sleep. METHODS We conducted a crossover laboratory study where 16 normal-weight participants (eight men; age 22 ± 5 years; body mass index < 25 kg/m2) completed three baseline days (9 hours sleep opportunity per night) followed by 5-day insufficient (5 hours sleep opportunity per night) and adequate (9 hours sleep opportunity per night) sleep conditions. Energy balanced diets were provided during baseline, with ad libitum energy intake provided during the insufficient and adequate sleep conditions. Untargeted plasma metabolomics analyses were performed using blood samples collected every 4 hours across the final 24 hours of each condition. Biomarker models were developed using logistic regression and linear support vector machine (SVM) algorithms. RESULTS The top-performing biomarker model was developed by linear SVM modeling, consisted of 65 compounds, and discriminated insufficient versus adequate sleep with 74% overall accuracy and a Matthew's Correlation Coefficient of 0.39. The compounds in the top-performing biomarker model were associated with ATP Binding Cassette Transporters in Lipid Homeostasis, Phospholipid Metabolic Process, Plasma Lipoprotein Remodeling, and sphingolipid metabolism. CONCLUSION We identified potential metabolomics-based biomarkers of insufficient sleep in humans. Although our current biomarkers require further development and validation using independent cohorts, they have potential to advance our understanding of the negative consequences of insufficient sleep, improve diagnosis of poor sleep health, and could eventually help identify targets for countermeasures designed to mitigate the negative health consequences of insufficient sleep.
Collapse
Affiliation(s)
- Christopher M Depner
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Dasha T Cogswell
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Paul J Bisesi
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | - Rachel R Markwald
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
| | | | - Kevin Quinn
- Skaggs School of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Edward L Melanson
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Eastern Colorado Veterans Affairs Geriatric Research, Education, and Clinical Center, Denver, CO
| | - Nichole Reisdorph
- Skaggs School of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Kenneth P Wright
- Sleep and Chronobiology Laboratory, Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
42
|
Malik DM, Paschos GK, Sehgal A, Weljie AM. Circadian and Sleep Metabolomics Across Species. J Mol Biol 2020; 432:3578-3610. [PMID: 32376454 PMCID: PMC7781158 DOI: 10.1016/j.jmb.2020.04.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023]
Abstract
Under normal circadian function, metabolic control is temporally coordinated across tissues and behaviors with a 24-h period. However, circadian disruption results in negative consequences for metabolic homeostasis including energy or redox imbalances. Yet, circadian disruption has become increasingly prevalent within today's society due to many factors including sleep loss. Metabolic consequences of both have been revealed by metabolomics analyses of circadian biology and sleep. Specifically, two primary analytical platforms, mass spectrometry and nuclear magnetic resonance spectroscopy, have been used to study molecular clock and sleep influences on overall metabolic rhythmicity. For example, human studies have demonstrated the prevalence of metabolic rhythms in human biology, as well as pan-metabolome consequences of sleep disruption. However, human studies are limited to peripheral metabolic readouts primarily through minimally invasive procedures. For further tissue- and organism-specific investigations, a number of model systems have been studied, based upon the conserved nature of both the molecular clock and sleep across species. Here we summarize human studies as well as key findings from metabolomics studies using mice, Drosophila, and zebrafish. While informative, a limitation in existing literature is a lack of interpretation regarding dynamic synthesis or catabolism within metabolite pools. To this extent, future work incorporating isotope tracers, specific metabolite reporters, and single-cell metabolomics may provide a means of exploring dynamic activity in pathways of interest.
Collapse
Affiliation(s)
- Dania M Malik
- Pharmacology Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Georgios K Paschos
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Penn Chronobiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Aalim M Weljie
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
43
|
Huang T, Zeleznik OA, Poole EM, Clish CB, Deik AA, Scott JM, Vetter C, Schernhammer ES, Brunner R, Hale L, Manson JE, Hu FB, Redline S, Tworoger SS, Rexrode KM. Habitual sleep quality, plasma metabolites and risk of coronary heart disease in post-menopausal women. Int J Epidemiol 2020; 48:1262-1274. [PMID: 30371783 DOI: 10.1093/ije/dyy234] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2018] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Epidemiologic studies suggest a strong link between poor habitual sleep quality and increased cardiovascular disease risk. However, the underlying mechanisms are not entirely clear. Metabolomic profiling may elucidate systemic differences associated with sleep quality that influence cardiometabolic health. METHODS We explored cross-sectional associations between sleep quality and plasma metabolites in a nested case-control study of coronary heart disease (CHD) in the Women's Health Initiative (WHI; n = 1956) and attempted to replicate the results in an independent sample from the Nurses' Health Study II (NHSII; n = 209). A sleep-quality score (SQS) was derived from self-reported sleep problems asked in both populations. Plasma metabolomics were assayed using LC-MS with 347 known metabolites. General linear regression was used to identify individual metabolites associated with continuous SQS (false-discovery rate <0.05). Using least absolute shrinkage and selection operator (LASSO) algorithms, a metabolite score was created from replicated metabolites and evaluated with CHD risk in the WHI. RESULTS After adjusting for age, race/ethnicity, body mass index (BMI) and smoking, we identified 69 metabolites associated with SQS in the WHI (59 were lipids). Of these, 16 were replicated in NHSII (15 were lipids), including 6 triglycerides (TAGs), 4 phosphatidylethanolamines (PEs), 3 phosphatidylcholines (PCs), 1 diglyceride (DAG), 1 lysophosphatidylcholine and N6-acetyl-L-lysine (a product of histone acetylation). These metabolites were consistently higher among women with poorer sleep quality. The LASSO selection resulted in a nine-metabolite score (TAGs 45: 1, 48: 1, 50: 4; DAG 32: 1; PEs 36: 4, 38: 5; PCs 30: 1, 40: 6; N6-acetyl-L-lysine), which was positively associated with CHD risk (odds ratio per SD increase in the score: 1.16; 95% confidence interval: 1.05, 1.28; p = 0.0003) in the WHI after adjustment for matching factors and conventional CHD risk factors. CONCLUSIONS Differences in lipid metabolites may be an important pathogenic pathway linking poor habitual sleep quality and CHD risk.
Collapse
Affiliation(s)
- Tianyi Huang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Oana A Zeleznik
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Elizabeth M Poole
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Amy A Deik
- Broad Institute of MIT and Harvard, Boston, MA, USA
| | | | - Céline Vetter
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO, USA
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Center for Public Health, University of Vienna, Vienna, Austria
| | | | - Lauren Hale
- Program in Public Health, Department of Family, Population, and Preventive Medicine, Stony Brook Medicine, Stony Brook, NY, USA
| | - JoAnn E Manson
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Susan Redline
- Departments of Medicine, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shelley S Tworoger
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kathryn M Rexrode
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.,Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
44
|
Free Radical Oxidation and Sleep Disorders in Andro- and Menopause (Literature Review). ACTA BIOMEDICA SCIENTIFICA 2020. [DOI: 10.29413/abs.2020-5.1.4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This review presents data on changes in the physiology of sleep during reproductive aging. It is noted that insomnia and obstructive sleep apnea syndrome (OSAS) are the main sleep disorders. The results of foreign and domestic studies in the field of free radical oxidation during sleep deprivation in animal models are presented, indicating the dependence of processes on the duration of sleep deprivation. The largest number of studies of free radical processes in a person with somnological pathology was carried out in the study of OSAS. Blood, urine, saliva, condensate of exhaled air can be biomaterial for determining the parameters of free radical oxidation. It was shown that the intensity of oxidative stress depends on the severity of OSAS, as evidenced by the positive correlation of the level of active products of thiobarbituric acid, the products of oxidation of proteins and carbonyl groups with the apnea/hypopnea index, determining the development of not only oxidative, but also carbonyl stress in patients with a severe degree OSAS. Biomarkers such as thioredoxin, malondialdehyde, superoxide dismutase, and reduced iron have shown a more stable relationship between increased oxidative stress and OSA. Despite the results obtained, the question of the association of oxidative stress and hypoxia in OSA remains debatable, which is associated with the opposite results of some studies. Insomnia, which occurs mainly in females, is accompanied by a high level of end products of lipid peroxidation with a decrease in the activity of antioxidants such as paraoxonase, an enzymatic component of the glutathione system. Along with this, menopausal women present low levels of uric acid, which correlates with high scores of the Pittsburgh sleep quality index questionnaire. Recent studies have identified an association between the activity of the «lipoperoxidation – antioxidants» system and the Clock 3111T/C gene polymorphism in menopausal Caucasian women, indicating the protective role of the minor allele.
Collapse
|
45
|
Sparks JR, Porter RR, Youngstedt SD, Bowyer KP, Durstine JL, Wang X. Effects of moderate sleep restriction during 8-week calorie restriction on lipoprotein particles and glucose metabolism. SLEEP ADVANCES 2020; 1:zpab001. [PMID: 33644759 PMCID: PMC7898726 DOI: 10.1093/sleepadvances/zpab001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/08/2021] [Indexed: 11/15/2022]
Abstract
Abstract
Study Objectives
This study examined how glucose, glucose regulatory hormones, insulin sensitivity, and lipoprotein subclass particle concentrations and sizes change with sleep restriction during weight loss elicited by calorie restriction.
Methods
Overweight or obese adults were randomized into an 8-week calorie restriction intervention alone (CR, n = 12; 75% female; body mass index = 31.4 ± 2.9 kg/m2) or combined with sleep restriction (CR+SR, n = 16; 75% female; body mass index = 34.5 ± 3.1 kg/m2). Participants in both groups were given the same instructions to reduce calorie intake. Those in the CR+SR group were instructed to reduce their habitual time-in-bed by 30–90 minutes 5 days each week with 2 ad libitum sleep days. Fasting venous blood samples were collected at pre- and post-intervention.
Results
Differential changes were found between the two groups (p = 0.028 for group × time interaction) in glucagon concentration, which decreased in the CR group (p = 0.016) but did not change in CR+SR group. Although changes in mean HDL particle (HDL-P) size and visfatin concentration were not statistically different between groups (p = 0.066 and 0.066 for group×time interaction, respectively), mean HDL-P size decreased only in the CR+SR group (Cohen’s d = 0.50, p = 0.022); visfatin concentrations did not change significantly in either group but appeared to decrease in the CR group (Cohen’s d = 0.67, p = 0.170) but not in the CR+SR group (Cohen’s d = 0.43, p = 0.225).
Conclusion
These results suggest that moderate sleep restriction, despite the presence of periodic ad libitum sleep, influences lipoprotein subclass particles and glucose regulation in individuals undergoing calorie restriction.
Clinical trial registration: ClinicalTrials.gov (NCT02413866, Weight Outlooks by Restriction of Diet and Sleep)
Collapse
Affiliation(s)
- Joshua R Sparks
- Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Ryan R Porter
- Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Shawn D Youngstedt
- Edson College of Nursing and Health Innovation, Arizona State University, Phoenix, AZ
| | - Kimberly P Bowyer
- Department of Exercise Science, University of South Carolina, Columbia, SC
| | - J Larry Durstine
- Department of Exercise Science, University of South Carolina, Columbia, SC
| | - Xuewen Wang
- Department of Exercise Science, University of South Carolina, Columbia, SC
| |
Collapse
|
46
|
Diallo I, Pak VM. Metabolomics, sleepiness, and sleep duration in sleep apnea. Sleep Breath 2020; 24:1327-1332. [PMID: 31955318 DOI: 10.1007/s11325-019-01969-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/21/2019] [Accepted: 10/25/2019] [Indexed: 01/27/2023]
Abstract
PURPOSE Although the mechanism is unclear, daytime sleepiness, a common sequela of obstructive sleep apnea (OSA), has been found to be correlated with a adverse cardiovascular outcomes. Reviewing metabolomics mechanisms of sleep disturbances and cardiovascular disease may help to explain this correlation. METHODS This review examines the current literature on the relationships between sleepiness, sleep duration, and metabolites in sleep apnea. RESULTS Although there is a lack of comprehensive literature in this emerging area, existing studies point to a variety of metabolites in different pathways that are associated with sleepiness and sleep duration. CONCLUSION Advancing metabolomics research in sleep apnea will guide symptom research and provide alternate and novel opportunities for effective treatment for patients with OSA.
Collapse
Affiliation(s)
- Idiatou Diallo
- Department of Global Health, Emory Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Victoria M Pak
- Nell Hodgson Woodruff School of Nursing, Emory University, Atlanta, GA, USA.
| |
Collapse
|
47
|
Pan X, Mota S, Zhang B. Circadian Clock Regulation on Lipid Metabolism and Metabolic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:53-66. [PMID: 32705594 PMCID: PMC8593891 DOI: 10.1007/978-981-15-6082-8_5] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The basic helix-loop-helix-PAS transcription factor (CLOCK, Circadian locomotor output cycles protein kaput) was discovered in 1994 as a circadian clock. Soon after its discovery, the circadian clock, Aryl hydrocarbon receptor nuclear translocator-like protein 1 (ARNTL, also call BMAL1), was shown to regulate adiposity and body weight by controlling on the brain hypothalamic suprachiasmatic nucleus (SCN). Farther, circadian clock genes were determined to exert several of lipid metabolic and diabetes effects, overall indicating that CLOCK and BMAL1 act as a central master circadian clock. A master circadian clock acts through the neurons and hormones, with expression in the intestine, liver, kidney, lung, heart, SCN of brain, and other various cell types of the organization. Among circadian clock genes, numerous metabolic syndromes are the most important in the regulation of food intake (via regulation of circadian clock genes or clock-controlled genes in peripheral tissue), which lead to a variation in plasma phospholipids and tissue phospholipids. Circadian clock genes affect the regulation of transporters and proteins included in the regulation of phospholipid metabolism. These genes have recently received increasing recognition because a pharmacological target of circadian clock genes may be of therapeutic worth to make better resistance against insulin, diabetes, obesity, metabolism syndrome, atherosclerosis, and brain diseases. In this book chapter, we focus on the regulation of circadian clock and summarize its phospholipid effect as well as discuss the chemical, physiology, and molecular value of circadian clock pathway regulation for the treatment of plasma lipids and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA.
| | - Samantha Mota
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA
| | - Boyang Zhang
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA
- Diabetes and Obesity Research Center, New York University Winthrop Hospital, Mineola, NY, USA
| |
Collapse
|
48
|
Rahman SA, Grant LK, Gooley JJ, Rajaratnam SMW, Czeisler CA, Lockley SW. Endogenous Circadian Regulation of Female Reproductive Hormones. J Clin Endocrinol Metab 2019; 104:6049-6059. [PMID: 31415086 PMCID: PMC6821202 DOI: 10.1210/jc.2019-00803] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 08/09/2019] [Indexed: 11/19/2022]
Abstract
CONTEXT Studies suggest that female reproductive hormones are under circadian regulation, although methodological differences have led to inconsistent findings. OBJECTIVE To determine whether circulating levels of reproductive hormones exhibit circadian rhythms. DESIGN Blood samples were collected across ∼90 consecutive hours, including 2 baseline days under a standard sleep-wake schedule and ∼50 hours of extended wake under constant routine (CR) conditions. SETTING Intensive Physiological Monitoring Unit, Brigham and Women's Hospital. PARTICIPANTS Seventeen healthy premenopausal women (22.8 ± 2.6 years; nine follicular; eight luteal). INTERVENTIONS Fifty-hour CR. MAIN OUTCOME MEASURES Plasma estradiol (E2), progesterone (P4), LH, FSH, SHBG, melatonin, and core body temperature. RESULTS All hormones exhibited significant 24-hour rhythms under both standard sleep-wake and CR conditions during the follicular phase (P < 0.05). In contrast, only FSH and SHBG were significantly rhythmic during the luteal phase. Rhythm acrophases and amplitudes were similar between standard sleep-wake and CR conditions. The acrophase occurred in the morning for P4; in the afternoon for FSH, LH, and SHBG; and during the night for E2. CONCLUSIONS Our results confirm previous reports of ∼24-hour rhythms in many female reproductive hormones in humans under ambulatory conditions but demonstrate that these hormones are under endogenous circadian regulation, defined as persisting in the absence of external time cues. These results may have important implications for the effects of circadian disruption on reproductive function.
Collapse
Affiliation(s)
- Shadab A Rahman
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Leilah K Grant
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Joshua J Gooley
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Shantha M W Rajaratnam
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
- Monash Institute of Cognitive and Clinical Neurosciences, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| | - Charles A Czeisler
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
| | - Steven W Lockley
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts
- Monash Institute of Cognitive and Clinical Neurosciences, School of Psychological Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
49
|
Meng M, Jiang Y, Zhu L, Wang G, Lin Q, Sun W, Song Y, Dong S, Deng Y, Rong T, Zhu Q, Mei H, Jiang F. Effect of maternal sleep in late pregnancy on leptin and lipid levels in umbilical cord blood. Sleep Med 2019; 77:376-383. [PMID: 32839086 DOI: 10.1016/j.sleep.2019.11.1194] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVES To study the impact of maternal sleep in late pregnancy on birth weight (BW) and leptin and lipid levels in umbilical cord blood. STUDY DESIGN A total of 277 healthy and singleton pregnancy women were recruited for participation in the Shanghai Sleep Birth Cohort Study (SSBC) during their 36-38 weeks of pregnancy, from May 2012 to July 2013. Maternal night sleep time (NST), sleep efficiency (SE), sleep onset latency (SOL) and the percentage of wake after sleep onset (WASO) in NST and midpoint of sleep (MSF) were measured by actigraphy for seven consecutive days. The leptin and lipid levels were determined in cord blood samples collected from the umbilical vein immediately after delivery. Birth information (birth weight, gender, delivery type, etc.) was extracted from medical records. A multivariable linear regression model was applied to examine the effect of maternal sleep in late pregnancy on newborn leptin and lipid levels in umbilical cord blood. RESULTS A total of 177 women and their infants were included in the analysis. Maternal mean NST was 7.03 ± 1.10 h in late pregnancy, and 48% had a shorter sleep time (NST < 7 h). The average maternal SE was 72.54% ± 9.66%. The mean percentage WASO/NST was 21.62% ± 9.98%; the average MSF was about 3:34 (0:53); and the SOL was 46.78 ± 36.00 min. After adjustment for confounders, both maternal NST and SE were found to be significantly associated with triglyceride levels (β = -0.219, p = 0.006; β = -0.224, p = 0.006) in umbilical cord blood; and maternal NST was also observed to have positive association with newborn leptin levels (β = 0.146, p = 0.047). However, we did not find significant association between other maternal sleep parameters in late pregnancy and leptin and lipid levels and birth weight. CONCLUSIONS Short sleep duration and poor sleep quality during late pregnancy were associated with newborn leptin and lipid levels, and efforts on improving maternal sleep during late pregnancy should be advocated for children's health.
Collapse
Affiliation(s)
- Min Meng
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Yanrui Jiang
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Lixia Zhu
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Guanghai Wang
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Qingmin Lin
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Wanqi Sun
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Yuanjin Song
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Shumei Dong
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Yujiao Deng
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Tingyu Rong
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Qi Zhu
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China
| | - Hao Mei
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Data Science, School of Population Health, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fan Jiang
- Department of Developmental and Behavioral Pediatrics, Pediatric Translational Medicine Institution, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MOE-Shanghai Key Laboratory of Environment and Child Health, Shanghai, China.
| |
Collapse
|
50
|
Noordam R, Bos MM, Wang H, Winkler TW, Bentley AR, Kilpeläinen TO, de Vries PS, Sung YJ, Schwander K, Cade BE, Manning A, Aschard H, Brown MR, Chen H, Franceschini N, Musani SK, Richard M, Vojinovic D, Aslibekyan S, Bartz TM, de las Fuentes L, Feitosa M, Horimoto AR, Ilkov M, Kho M, Kraja A, Li C, Lim E, Liu Y, Mook-Kanamori DO, Rankinen T, Tajuddin SM, van der Spek A, Wang Z, Marten J, Laville V, Alver M, Evangelou E, Graff ME, He M, Kühnel B, Lyytikäinen LP, Marques-Vidal P, Nolte IM, Palmer ND, Rauramaa R, Shu XO, Snieder H, Weiss S, Wen W, Yanek LR, Adolfo C, Ballantyne C, Bielak L, Biermasz NR, Boerwinkle E, Dimou N, Eiriksdottir G, Gao C, Gharib SA, Gottlieb DJ, Haba-Rubio J, Harris TB, Heikkinen S, Heinzer R, Hixson JE, Homuth G, Ikram MA, Komulainen P, Krieger JE, Lee J, Liu J, Lohman KK, Luik AI, Mägi R, Martin LW, Meitinger T, Metspalu A, Milaneschi Y, Nalls MA, O'Connell J, Peters A, Peyser P, Raitakari OT, Reiner AP, Rensen PCN, Rice TK, Rich SS, Roenneberg T, Rotter JI, Schreiner PJ, Shikany J, Sidney SS, Sims M, Sitlani CM, Sofer T, Strauch K, Swertz MA, Taylor KD, Uitterlinden AG, et alNoordam R, Bos MM, Wang H, Winkler TW, Bentley AR, Kilpeläinen TO, de Vries PS, Sung YJ, Schwander K, Cade BE, Manning A, Aschard H, Brown MR, Chen H, Franceschini N, Musani SK, Richard M, Vojinovic D, Aslibekyan S, Bartz TM, de las Fuentes L, Feitosa M, Horimoto AR, Ilkov M, Kho M, Kraja A, Li C, Lim E, Liu Y, Mook-Kanamori DO, Rankinen T, Tajuddin SM, van der Spek A, Wang Z, Marten J, Laville V, Alver M, Evangelou E, Graff ME, He M, Kühnel B, Lyytikäinen LP, Marques-Vidal P, Nolte IM, Palmer ND, Rauramaa R, Shu XO, Snieder H, Weiss S, Wen W, Yanek LR, Adolfo C, Ballantyne C, Bielak L, Biermasz NR, Boerwinkle E, Dimou N, Eiriksdottir G, Gao C, Gharib SA, Gottlieb DJ, Haba-Rubio J, Harris TB, Heikkinen S, Heinzer R, Hixson JE, Homuth G, Ikram MA, Komulainen P, Krieger JE, Lee J, Liu J, Lohman KK, Luik AI, Mägi R, Martin LW, Meitinger T, Metspalu A, Milaneschi Y, Nalls MA, O'Connell J, Peters A, Peyser P, Raitakari OT, Reiner AP, Rensen PCN, Rice TK, Rich SS, Roenneberg T, Rotter JI, Schreiner PJ, Shikany J, Sidney SS, Sims M, Sitlani CM, Sofer T, Strauch K, Swertz MA, Taylor KD, Uitterlinden AG, van Duijn CM, Völzke H, Waldenberger M, Wallance RB, van Dijk KW, Yu C, Zonderman AB, Becker DM, Elliott P, Esko T, Gieger C, Grabe HJ, Lakka TA, Lehtimäki T, North KE, Penninx BWJH, Vollenweider P, Wagenknecht LE, Wu T, Xiang YB, Zheng W, Arnett DK, Bouchard C, Evans MK, Gudnason V, Kardia S, Kelly TN, Kritchevsky SB, Loos RJF, Pereira AC, Province M, Psaty BM, Rotimi C, Zhu X, Amin N, Cupples LA, Fornage M, Fox EF, Guo X, Gauderman WJ, Rice K, Kooperberg C, Munroe PB, Liu CT, Morrison AC, Rao DC, van Heemst D, Redline S. Multi-ancestry sleep-by-SNP interaction analysis in 126,926 individuals reveals lipid loci stratified by sleep duration. Nat Commun 2019; 10:5121. [PMID: 31719535 PMCID: PMC6851116 DOI: 10.1038/s41467-019-12958-0] [Show More Authors] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
Both short and long sleep are associated with an adverse lipid profile, likely through different biological pathways. To elucidate the biology of sleep-associated adverse lipid profile, we conduct multi-ancestry genome-wide sleep-SNP interaction analyses on three lipid traits (HDL-c, LDL-c and triglycerides). In the total study sample (discovery + replication) of 126,926 individuals from 5 different ancestry groups, when considering either long or short total sleep time interactions in joint analyses, we identify 49 previously unreported lipid loci, and 10 additional previously unreported lipid loci in a restricted sample of European-ancestry cohorts. In addition, we identify new gene-sleep interactions for known lipid loci such as LPL and PCSK9. The previously unreported lipid loci have a modest explained variance in lipid levels: most notable, gene-short-sleep interactions explain 4.25% of the variance in triglyceride level. Collectively, these findings contribute to our understanding of the biological mechanisms involved in sleep-associated adverse lipid profiles.
Collapse
Affiliation(s)
- Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Maxime M Bos
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tuomas O Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, 2200, Denmark
- Department of Environmental Medicine and Public Health, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Alisa Manning
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Hugues Aschard
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Michael R Brown
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Public Health & School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Solomon K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Melissa Richard
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stella Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Biostatistics and Medicine, University of Washington, Seattle, WA, USA
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Mary Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrea R Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | | | - Minjung Kho
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Aldi Kraja
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Changwei Li
- Epidemiology and Biostatistics, University of Georgia at Athens College of Public Health, Athens, GA, USA
| | - Elise Lim
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Yongmei Liu
- Public Health Sciences, Epidemiology and Prevention, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, Netherlands
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Salman M Tajuddin
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ashley van der Spek
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Zhe Wang
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Vincent Laville
- Centre de Bioinformatique, Biostatistique et Biologie Intégrative (C3BI), Institut Pasteur, Paris, France
| | - Maris Alver
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Maria E Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Meian He
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Brigitte Kühnel
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pedro Marques-Vidal
- Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Ilja M Nolte
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | | | - Rainer Rauramaa
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harold Snieder
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lisa R Yanek
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Correa Adolfo
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Christie Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, Houston, TX, USA
- Houston Methodist Debakey Heart and Vascular Center, Houston, TX, USA
| | - Larry Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Nienke R Biermasz
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Niki Dimou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | | | - Chuan Gao
- Molecular Genetics and Genomics Program, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Medicine, University of Washington, Seattle, WA, USA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - José Haba-Rubio
- Medicine, Sleep Laboratory, Lausanne University Hospital, Lausanne, Switzerland
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Sami Heikkinen
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
| | - Raphaël Heinzer
- Medicine, Sleep Laboratory, Lausanne University Hospital, Lausanne, Switzerland
| | - James E Hixson
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pirjo Komulainen
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Jiwon Lee
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Jingmin Liu
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Kurt K Lohman
- Public Health Sciences, Biostatistical Sciences, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Annemarie I Luik
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Reedik Mägi
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Lisa W Martin
- Cardiology, School of Medicine and Health Sciences, George Washington University, Washington, D.C., USA
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Andres Metspalu
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Cardiology, School of Medicine and Health Sciences, George Washington University, Washington, D.C., USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Data Tecnica International, Glen Echo, MD, USA
| | - Jeff O'Connell
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
- Program for Personalized and Genomic Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Neuherberg, Germany
| | - Patricia Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- University of Turku, Turku, Finland
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
| | - Treva K Rice
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Till Roenneberg
- Institute of Medical Psychology, Ludwig-Maximilians-Universitat Munchen, Munich, Germany
| | - Jerome I Rotter
- Genomic Outcomes, Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CC, USA
| | - Pamela J Schreiner
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James Shikany
- Division of Preventive Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephen S Sidney
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Mario Sims
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Medicine, University of Washington, Seattle, WA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
- Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Informatics Biometry and Epidemiology, Ludwig-Maximilians-Universitat Munchen, Munich, Germany
| | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Kent D Taylor
- Genomic Outcomes, Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CC, USA
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Neuherberg, Germany
| | - Robert B Wallance
- Department of Epidemiology, University of Iowa College of Public Health, Iowa City, IA, USA
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Caizheng Yu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alan B Zonderman
- Behavioral Epidemiology Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Diane M Becker
- Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Elliott
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- National Institute of Health Research Imperial College London Biomedical Research Centre, London, UK
- UK-DRI Dementia Research Institute at Imperial College London, London, UK
| | - Tõnu Esko
- Estonian Genome Center, Institute of Genomics, University of Tartu, Tartu, Estonia
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Boston, MA, USA
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Hans J Grabe
- Department Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
| | - Timo A Lakka
- Foundation for Research in Health Exercise and Nutrition, Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio Campus, Finland
- Department of Clinical Phsiology and Nuclear Medicine, Kuopia University Hospital, Kuopio, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
- Department of Clinical Chemistry, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health Research Institute, Amsterdam UMC, Vrije Universiteit, Amsterdam, The Netherlands
| | - Peter Vollenweider
- Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | - Lynne E Wagenknecht
- Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Tangchun Wu
- Department of Occupational and Environmental Health and State Key Laboratory of Environmental Health for Incubating, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong-Bing Xiang
- SKLORG & Department of Epidemiology, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P. R. China
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Donna K Arnett
- Dean's Office, University of Kentucky College of Public Health, Lexington, KS, USA
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Michele K Evans
- Health Disparities Research Section, Laboratory of Epidemiology and Population Sciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Sharon Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Rehabilitation, Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health Development Institute, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Mike Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington, Health Research Institute, Seattle, WA, USA
| | - Charles Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaofeng Zhu
- Department of Population Quantitative and Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Najaf Amin
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI Framingham Heart Study, Framingham, MA, USA
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ervin F Fox
- Cardiology, Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiuqing Guo
- Genomic Outcomes, Department of Pediatrics, Institute for Translational Genomics and Population Sciences, LABioMed at Harbor-UCLA Medical Center, Torrance, CC, USA
| | - W James Gauderman
- Biostatistics, Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kenneth Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Charles Kooperberg
- Fred Hutchinson Cancer Research Center, University of Washington School of Public Health, Seattle, WA, USA
| | - Patricia B Munroe
- Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- NIHR Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London, London, UK
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dabeeru C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO, USA
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
- Division of Pulmonary Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|