101
|
Dai J, Wu HY, Jiang XD, Tang YJ, Tang HK, Meng L, Huang N, Gao JY, Li J, Baker JS, Zheng CJ, Yang YD. Association between napping and 24-hour blood pressure variability among university students: A pilot study. Front Pediatr 2023; 11:1062300. [PMID: 36937964 PMCID: PMC10018217 DOI: 10.3389/fped.2023.1062300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Background Blood pressure variability (BPV) has been reported to be a predictor of cardiovascular and some cognitive diseases. However, the association between napping and BPV remains unknown. This study aimed to explore the association between napping and BPV. Materials and methods A cross-sectional study including 105 university students was conducted. Participants' 24 h ambulatory blood pressure monitoring (24 h ABPM) were measured, and napping behaviors were investigated. BPV were measured by the 24 h ABPM, included standard deviation (SD), coefficient of variation (CV), and average real variability (ARV). Results Among the participants, 61.9% reported daytime napping. We found that nap duration was significantly associated with daytime CV of diastolic blood pressure (DBP) (r = 0.250, P = 0.010), nighttime CV of systolic blood pressure (SBP) (r = 0.217, P = 0.026), 24 h WCV of DBP (r = 0.238, P = 0.014), 24 h ARV of SBP (r = 0.246, P = 0.011) and 24 h ARV of DBP (r = 0.291, P = 0.003). Compared with the no napping group, 24 h WCV of DBP, daytime CV of DBP, and daytime SD of DBP were significantly higher in participants with napping duration >60 min. With multiple regression analysis we found that nap duration was an independent predictor for 24 h ARV of SBP (β = 0.859, 95% CI, 0.101-1.616, P = 0.027) and 24 h ARV of DBP (β = 0.674, 95% CI, 0.173-1.175, P = 0.009). Conclusions Napping durations are associated with BPV among university students. Especially those with napping durations >60 min had a significantly higher BPV than those non-nappers.
Collapse
Affiliation(s)
- Jie Dai
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Hua-ying Wu
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Xiao-dong Jiang
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Yong-jie Tang
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Hao-Kai Tang
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Li Meng
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Na Huang
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jing-yu Gao
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Jian Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Julien S. Baker
- Centre for Health and Exercise Science Research, Department of Sport, Physical Education and Health, Hong Kong Baptist University, Hong Kong, Hong Kong SAR, China
| | - Chan-Juan Zheng
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Correspondence: Yi-De Yang Chan-Juan Zheng
| | - Yi-De Yang
- Department of Child and Adolescent Health, School of Medicine, Hunan Normal University, Changsha, China
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
- Correspondence: Yi-De Yang Chan-Juan Zheng
| |
Collapse
|
102
|
Lane JM, Qian J, Mignot E, Redline S, Scheer FAJL, Saxena R. Genetics of circadian rhythms and sleep in human health and disease. Nat Rev Genet 2023; 24:4-20. [PMID: 36028773 PMCID: PMC10947799 DOI: 10.1038/s41576-022-00519-z] [Citation(s) in RCA: 97] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 12/13/2022]
Abstract
Circadian rhythms and sleep are fundamental biological processes integral to human health. Their disruption is associated with detrimental physiological consequences, including cognitive, metabolic, cardiovascular and immunological dysfunctions. Yet many of the molecular underpinnings of sleep regulation in health and disease have remained elusive. Given the moderate heritability of circadian and sleep traits, genetics offers an opportunity that complements insights from model organism studies to advance our fundamental molecular understanding of human circadian and sleep physiology and linked chronic disease biology. Here, we review recent discoveries of the genetics of circadian and sleep physiology and disorders with a focus on those that reveal causal contributions to complex diseases.
Collapse
Affiliation(s)
- Jacqueline M Lane
- Center for Genomic Medicine and Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jingyi Qian
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Emmanuel Mignot
- Center for Narcolepsy, Stanford University, Palo Alto, California, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anaesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital; and Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
103
|
Li L, Zhang Q, Zhu L, Zeng G, Huang H, Zhuge J, Kuang X, Yang S, Yang D, Chen Z, Gan Y, Lu Z, Wu C. Daytime naps and depression risk: A meta-analysis of observational studies. Front Psychol 2022; 13:1051128. [PMID: 36591028 PMCID: PMC9798209 DOI: 10.3389/fpsyg.2022.1051128] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
Background The relationship between daytime napping and depression remains debatable. Thus, a meta-analysis in this study was conducted to evaluate the relationship between daytime napping and depression. Methods The PubMed, Embase, Web of Science, and China National Knowledge Infrastructure databases were searched up to February 2022, and the reference lists of the included studies were also retrieved. A random-effects model was used to estimate the combined effect size. Results Nine studies with 649,111 participants were included in the final analysis. The pooled odds ratio (OR) was 1.15 (95% confidence interval: 1.01-1.31) with a significant heterogeneity (I 2 = 91.3%, P for heterogeneity <0.001), and the results demonstrated an increased risk of depressive symptoms among daytime nappers. Visual inspection of the funnel plot and Egger's and Begg's tests identified no obvious evidence of publication bias. Conclusion This meta-analysis indicates that daytime naps are a predictor of depression. The effects of daytime napping on depression may vary depending on the characteristics of people, the pattern of naps, and the individual's sleep experience. The findings may have significant implications for future research on depression.
Collapse
Affiliation(s)
- Liqing Li
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China,School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhang
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China
| | - Liyong Zhu
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China
| | - Guohua Zeng
- School of Economics and Management, Jiangxi University of Science and Technology, Ganzhou, Jiangxi, China
| | - Hongwei Huang
- Department of Health Management Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Zhuge
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China
| | - Xiaorui Kuang
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China
| | - Sule Yang
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China
| | - Di Yang
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Zhensheng Chen
- Research Center of Health Policy and Innovation, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi, China
| | - Yong Gan
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zuxun Lu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunmei Wu
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, Jiangxi, China,*Correspondence: Chunmei Wu
| |
Collapse
|
104
|
Ma J, Jin C, Yang Y, Li H, Wang Y. Association of daytime napping frequency and schizophrenia: a bidirectional two-sample Mendelian randomization study. BMC Psychiatry 2022; 22:786. [PMID: 36513988 PMCID: PMC9746219 DOI: 10.1186/s12888-022-04431-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The bidirectional causal association between daytime napping frequency and schizophrenia is unclear. METHODS A bidirectional two-sample Mendelian randomization (MR) analysis was conducted with summary statistics of top genetic variants associated with daytime napping frequency and schizophrenia from genome-wide association studies (GWAS). The single nucleotide polymorphisms (SNPs) data of daytime napping frequency GWAS came from the UK Biobank (n = 452,633) and 23andMe study cohort (n = 541,333), while the schizophrenia GWAS came from the Psychiatric Genomics Consortium (PGC, 36,989 cases and 113,075 controls). The inverse variance weighted (IVW) analysis was the primary method, with the weighted median, MR-Robust Adjusted Profile Score (RAPS), Radial MR and MR-Pleiotropy Residual Sum Outlier (PRESSO) as sensitivity analysis. RESULTS The MR analysis showed a bidirectional causal relationship between more frequent daytime napping and the occurrence of schizophrenia, with the odds ratio (OR) for one-unit increase in napping category (never, sometimes, usually) on schizophrenia was 3.38 (95% confidence interval [CI]: 2.02-5.65, P = 3.58 × 10-6), and the beta for the occurrence of schizophrenia on daytime napping frequency was 0.0112 (95%CI: 0.0060-0.0163, P = 2.04 × 10-5). The sensitivity analysis obtained the same conclusions. CONCLUSION Our findings support the bidirectional causal association between more daytime napping frequency and schizophrenia, implying that daytime napping frequency is a potential intervention for the progression and treatment of schizophrenia.
Collapse
Affiliation(s)
- Jun Ma
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Chen Jin
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yan Yang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Haoqi Li
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Wenzhou Medical University, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
105
|
Chen J, Chen Z, Chen R, Feng D, Li T, Han H, Bi X, Wang Z, Li K, Li Y, Li X, Wang L, Li J. HCDT: an integrated highly confident drug-target resource. Database (Oxford) 2022; 2022:6843794. [PMID: 36420558 PMCID: PMC9684616 DOI: 10.1093/database/baac101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/12/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022]
Abstract
Drug-target association plays an important role in drug discovery, drug repositioning, drug synergy prediction, etc. Currently, a lot of drug-related databases, such as DrugBank and BindingDB, have emerged. However, these databases are separate, incomplete and non-uniform with different criteria. Here, we integrated eight drug-related databases; collected, filtered and supplemented drugs, target genes and experimentally validated (highly confident) associations and built a highly confident drug-target (HCDT: http://hainmu-biobigdata.com/hcdt) database. HCDT database includes 500 681 HCDT associations between 299 458 drugs and 5618 target genes. Compared to individual databases, HCDT database contains 1.1 to 254.2 times drugs, 1.8-5.5 times target genes and 1.4-27.7 times drug-target associations. It is normative, publicly available and easy for searching, browsing and downloading. Together with multi-omics data, it will be a good resource in analyzing the drug functional mechanism, mining drug-related biological pathways, predicting drug synergy, etc. Database URL: http://hainmu-biobigdata.com/hcdt.
Collapse
Affiliation(s)
| | | | - Rufei Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Dehua Feng
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Tianyi Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Huirui Han
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Xiaoman Bi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Zhenzhen Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Kongning Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Bioinformatics for Major Diseases Science Innovation Group, College of Biomedical Informatics and Engineering, Hainan Medical University, Haikou 571199, China
| | - Xia Li
- *Corresponding author: Tel: +86-451-86615922; Fax: +86-451-86615922; Correspondence may also be addressed to Limei Wang. Tel: +86-898-66893770; Fax: +86-898-66893770; and Jin Li. Tel: +86-898-66893770; Fax: +86-898-66893770;
| | - Limei Wang
- *Corresponding author: Tel: +86-451-86615922; Fax: +86-451-86615922; Correspondence may also be addressed to Limei Wang. Tel: +86-898-66893770; Fax: +86-898-66893770; and Jin Li. Tel: +86-898-66893770; Fax: +86-898-66893770;
| | - Jin Li
- *Corresponding author: Tel: +86-451-86615922; Fax: +86-451-86615922; Correspondence may also be addressed to Limei Wang. Tel: +86-898-66893770; Fax: +86-898-66893770; and Jin Li. Tel: +86-898-66893770; Fax: +86-898-66893770;
| |
Collapse
|
106
|
Lamri A, De Paoli M, De Souza R, Werstuck G, Anand S, Pigeyre M. Insight into genetic, biological, and environmental determinants of sexual-dimorphism in type 2 diabetes and glucose-related traits. Front Cardiovasc Med 2022; 9:964743. [PMID: 36505380 PMCID: PMC9729955 DOI: 10.3389/fcvm.2022.964743] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2022] Open
Abstract
There is growing evidence that sex and gender differences play an important role in risk and pathophysiology of type 2 diabetes (T2D). Men develop T2D earlier than women, even though there is more obesity in young women than men. This difference in T2D prevalence is attenuated after the menopause. However, not all women are equally protected against T2D before the menopause, and gestational diabetes represents an important risk factor for future T2D. Biological mechanisms underlying sex and gender differences on T2D physiopathology are not yet fully understood. Sex hormones affect behavior and biological changes, and can have implications on lifestyle; thus, both sex-specific environmental and biological risk factors interact within a complex network to explain the differences in T2D risk and physiopathology in men and women. In addition, lifetime hormone fluctuations and body changes due to reproductive factors are generally more dramatic in women than men (ovarian cycle, pregnancy, and menopause). Progress in genetic studies and rodent models have significantly advanced our understanding of the biological pathways involved in the physiopathology of T2D. However, evidence of the sex-specific effects on genetic factors involved in T2D is still limited, and this gap of knowledge is even more important when investigating sex-specific differences during the life course. In this narrative review, we will focus on the current state of knowledge on the sex-specific effects of genetic factors associated with T2D over a lifetime, as well as the biological effects of these different hormonal stages on T2D risk. We will also discuss how biological insights from rodent models complement the genetic insights into the sex-dimorphism effects on T2D. Finally, we will suggest future directions to cover the knowledge gaps.
Collapse
Affiliation(s)
- Amel Lamri
- Department of Medicine, McMaster University, Hamilton, ON, Canada,Population Health Research Institute (PHRI), Hamilton, ON, Canada
| | - Monica De Paoli
- Department of Medicine, McMaster University, Hamilton, ON, Canada,Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton, ON, Canada
| | - Russell De Souza
- Population Health Research Institute (PHRI), Hamilton, ON, Canada,Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Geoff Werstuck
- Department of Medicine, McMaster University, Hamilton, ON, Canada,Thrombosis and Atherosclerosis Research Institute (TaARI), Hamilton, ON, Canada
| | - Sonia Anand
- Department of Medicine, McMaster University, Hamilton, ON, Canada,Population Health Research Institute (PHRI), Hamilton, ON, Canada,Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Marie Pigeyre
- Department of Medicine, McMaster University, Hamilton, ON, Canada,Population Health Research Institute (PHRI), Hamilton, ON, Canada,*Correspondence: Marie Pigeyre
| |
Collapse
|
107
|
Harbison ST. What have we learned about sleep from selective breeding strategies? Sleep 2022; 45:zsac147. [PMID: 36111812 PMCID: PMC9644121 DOI: 10.1093/sleep/zsac147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/19/2022] [Indexed: 09/18/2023] Open
Abstract
Selective breeding is a classic technique that enables an experimenter to modify a heritable target trait as desired. Direct selective breeding for extreme sleep and circadian phenotypes in flies successfully alters these behaviors, and sleep and circadian perturbations emerge as correlated responses to selection for other traits in mice, rats, and dogs. The application of sequencing technologies to the process of selective breeding identifies the genetic network impacting the selected trait in a holistic way. Breeding techniques preserve the extreme phenotypes generated during selective breeding, generating community resources for further functional testing. Selective breeding is thus a unique strategy that can explore the phenotypic limits of sleep and circadian behavior, discover correlated responses of traits having shared genetic architecture with the target trait, identify naturally-occurring genomic variants and gene expression changes that affect trait variability, and pinpoint genes with conserved roles.
Collapse
Affiliation(s)
- Susan T Harbison
- Laboratory of Systems Genetics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD,USA
| |
Collapse
|
108
|
Jia Y, Guo D, Sun L, Shi M, Zhang K, Yang P, Zang Y, Wang Y, Liu F, Zhang Y, Zhu Z. Self-reported daytime napping, daytime sleepiness, and other sleep phenotypes in the development of cardiometabolic diseases: a Mendelian randomization study. Eur J Prev Cardiol 2022; 29:1982-1991. [PMID: 35707994 DOI: 10.1093/eurjpc/zwac123] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 01/11/2023]
Abstract
AIMS Sleep disorders are associated with an increased risk of cardiometabolic diseases in observational studies, but the causality remains unclear. In this study, we leveraged two-sample Mendelian randomization (MR) analyses to assess the causal associations of self-reported daytime napping, daytime sleepiness, and other sleep phenotypes with cardiometabolic diseases including ischaemic stroke (IS), coronary artery disease (CAD), heart failure (HF), and Type 2 diabetes mellitus (T2DM). METHODS AND RESULTS We selected genetic variants as instrumental variables for self-reported daytime napping, daytime sleepiness, morning person, insomnia, short sleep duration, and long sleep duration from European-descent genome-wide association studies (GWASs). Summary statistics for cardiometabolic diseases originated from four different GWASs with a total of 2 500 086 participants. We used the inverse-variance weighted method to explore the role of self-reported sleep phenotypes on the aetiology of cardiometabolic diseases in the main analyses, followed by several sensitivity analyses for robustness validation. Genetically predicted self-reported daytime napping [T2DM: OR, 1.56 (95% confidence interval, 1.21-2.02)], insomnia [IS: OR, 1.07 (1.04-1.11)]; CAD: OR, 1.13 (1.08-1.17); HF: OR, 1.10 (1.07-1.14); T2DM: OR, 1.16 (1.11-1.22); and short sleep duration [CAD: OR, 1.37 (1.21-1.55)] were causally associated with an elevated risk of cardiometabolic diseases. Moreover, genetically determined self-reported daytime sleepiness [CAD: OR, 2.05 (1.18-3.57); HF: OR, 1.82 (1.15-2.87)] and morning person [HF: 1.06 OR, (1.01-1.11)] had potential detrimental effect on cardiometabolic risks. CONCLUSION Self-reported daytime napping, insomnia, and short sleep duration had causal roles in the development of cardiometabolic diseases, while self-reported daytime sleepiness and morning person was the potential risk factor for cardiometabolic diseases.
Collapse
Affiliation(s)
- Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Daoxia Guo
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China.,School of Nursing, Medical College of Soochow University, Suzhou, Jiangsu Province 215006, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Kaixin Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Yuhan Zang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Yu Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Fanghua Liu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, Jiangsu Province 215123, China
| |
Collapse
|
109
|
McAlpine CS, Kiss MG, Zuraikat FM, Cheek D, Schiroli G, Amatullah H, Huynh P, Bhatti MZ, Wong LP, Yates AG, Poller WC, Mindur JE, Chan CT, Janssen H, Downey J, Singh S, Sadreyev RI, Nahrendorf M, Jeffrey KL, Scadden DT, Naxerova K, St-Onge MP, Swirski FK. Sleep exerts lasting effects on hematopoietic stem cell function and diversity. J Exp Med 2022; 219:213487. [PMID: 36129517 PMCID: PMC9499822 DOI: 10.1084/jem.20220081] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 06/21/2022] [Accepted: 08/22/2022] [Indexed: 01/21/2023] Open
Abstract
A sleepless night may feel awful in its aftermath, but sleep's revitalizing powers are substantial, perpetuating the idea that convalescent sleep is a consequence-free physiological reset. Although recent studies have shown that catch-up sleep insufficiently neutralizes the negative effects of sleep debt, the mechanisms that control prolonged effects of sleep disruption are not understood. Here, we show that sleep interruption restructures the epigenome of hematopoietic stem and progenitor cells (HSPCs) and increases their proliferation, thus reducing hematopoietic clonal diversity through accelerated genetic drift. Sleep fragmentation exerts a lasting influence on the HSPC epigenome, skewing commitment toward a myeloid fate and priming cells for exaggerated inflammatory bursts. Combining hematopoietic clonal tracking with mathematical modeling, we infer that sleep preserves clonal diversity by limiting neutral drift. In humans, sleep restriction alters the HSPC epigenome and activates hematopoiesis. These findings show that sleep slows decay of the hematopoietic system by calibrating the hematopoietic epigenome, constraining inflammatory output, and maintaining clonal diversity.
Collapse
Affiliation(s)
- Cameron S. McAlpine
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Friedman Brain Institute and the Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Cameron S. McAlpine:
| | - Máté G. Kiss
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Faris M. Zuraikat
- Sleep Center of Excellence, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - David Cheek
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Giulia Schiroli
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Hajera Amatullah
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Pacific Huynh
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mehreen Z. Bhatti
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Lai-Ping Wong
- Department of Molecular Biology, Massachusetts General Hospital and Department of Genetics, Harvard Medical School, Boston, MA
| | - Abi G. Yates
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Wolfram C. Poller
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - John E. Mindur
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Christopher T. Chan
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Henrike Janssen
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Jeffrey Downey
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Sumnima Singh
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital and Department of Genetics, Harvard Medical School, Boston, MA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Matthias Nahrendorf
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Kate L. Jeffrey
- Division of Gastroenterology and Center for the Study of Inflammatory Bowel Disease, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - David T. Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA
| | - Kamila Naxerova
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Marie-Pierre St-Onge
- Sleep Center of Excellence, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Division of General Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY
- Marie-Pierre St-Onge:
| | - Filip K. Swirski
- Cardiovascular Research Institute and the Department of Medicine, Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Systems Biology and the Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Correspondence to Filip K. Swirski:
| |
Collapse
|
110
|
Sun Y, Cao X, Cao D, Cui Y, Su K, Jia Z, Wu Y, Jiang J. Genetic estimation of correlations and causalities between multifaceted modifiable factors and gastro-oesophageal reflux disease. Front Nutr 2022; 9:1009122. [PMID: 36386930 PMCID: PMC9663808 DOI: 10.3389/fnut.2022.1009122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/17/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Gastro-oesophageal reflux disease (GORD) is a common gastrointestinal dysfunction that significantly affects the quality of daily life, and health interventions are challenging to prevent the risk of GORD. In this study, we used Mendelian randomization framework to genetically determine the causal associations between multifaceted modifiable factors and the risk of GORD. MATERIALS AND METHODS Sixty-six exposures with available instrumental variables (IVs) across 6 modifiable pathways were included in the univariable MR analysis (UVMR). Summary-level genome-wide association studies (GWAS) datasets for GORD were retrieved from the Neale Lab (GORD Neale , Ncases = 29975, Ncontrols = 390556) and FinnGen (GORD Finn , Ncases = 13141, Ncontrols = 89695). Using the METAL software, meta-analysis for single nucleotide polymorphisms (SNPs) from GORD Neale and GORD Finn was conducted with an inverse variance weighted (IVW) fixed-effect model. Moreover, we leveraged partition around medoids (PAM) clustering algorithm to cluster genetic correlation subtypes, whose hub exposures were conditioned for multivariable MR (MVMR) analyses. P-values were adjusted with Bonferroni multiple comparisons. RESULTS Significant causal associations were identified between 26 exposures (15 risk exposures and 11 protective exposures) and the risk of GORD. Among them, 13 risk exposures [lifetime smoking, cigarette consumption, insomnia, short sleep, leisure sedentary behavior (TV watching), body mass index (BMI), body fat percentage, whole body fat mass, visceral adipose tissue, waist circumference, hip circumference, major depressive disorder, and anxious feeling], and 10 protective exposures (leisure sedentary behavior (computer use), sitting height, hand grip strength (left and right), birth weight, life satisfaction, positive affect, income, educational attainment, and intelligence) showed novel significant causal associations with the risk of GORD. Moreover, 13 exposures still demonstrated independent associations with the risk of GORD following MVMR analyses conditioned for hub exposures (educational attainment, smoking initiation and BMI). In addition, 12 exposures showed suggestive causal associations with the risk of GORD. CONCLUSION This study systematically elucidated the modifiable factors causally associated with the risk of GORD from multifaceted perspectives, which provided implications for prevention and treatment of GORD.
Collapse
Affiliation(s)
- Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Donghui Cao
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yingnan Cui
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kaisheng Su
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhifang Jia
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanhua Wu
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jing Jiang
- Department of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
111
|
Influence of DNA-Polymorphisms in Selected Circadian Clock Genes on Clock Gene Expression in Subjects from the General Population and Their Association with Sleep Duration. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58091294. [PMID: 36143969 PMCID: PMC9506325 DOI: 10.3390/medicina58091294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/03/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
Background and Objectives: Circadian rhythms have an important implication in numerous physiological and metabolic processes, including the sleep/wake cycle. Inter-individual differences in factors associated with circadian system may be due to gene differences in gene expression. Although several studies have analyzed the association between DNA polymorphisms and circadian variables, the influence on gene expression has been poorly analyzed. Our goal was to analyze the association of genetic variations in the clock genes and the gene expression level. Materials and Methods: We carried out a cross-sectional study of 102 adults (50.9% women). RNA and DNA were isolated from blood and single-nucleotide polymorphisms (SNPs), and the main circadian clock genes were determined. Gene expression of CLOCK, PER1, and VRK2 genes was measured by Reverse-transcription polymerase chain reaction (RT-PCR). The association between the DNA-SNPs and gene expression was analyzed at the gene level. In addition, a polygenic risk score (PRS), including all the significant SNPs related to gene expression, was created for each gene. Multivariable model analysis was performed. Results: Sex-specific differences were detected in PER1 expression, with these being higher in women (p = 0.034). No significant differences were detected in clock genes expression and lifestyle variables. We observed a significant association between the ARNTL-rs7924734, ARNTL-rs10832027, VRK2- rs2678902 SNPs, and CLOCK gene expression; the PER3-rs228642 and PER3-rs10127838 were related to PER1 expression, and the ARNTL-rs10832027, ARNTL-rs11022778, and MNTR1B-rs10830963 were associated with VRK2 gene expression (p < 0.05). The specific PRS created was significantly associated with each of the gene expressions analyzed (p < 0.001). Finally, sleep duration was associated with PER3-rs238666 (p = 0.008) and CLOCK-rs4580704 (p = 0.023). Conclusion: We detected significant associations between DNA-SNPs in the clock genes and their gene expression level in leukocytes and observed some differences in gene expression per sex. Moreover, we reported for the first time an association between clock gene polymorphisms and CLOCK, PER1, and VRK2 gene expression. These findings need further investigation.
Collapse
|
112
|
Abstract
Genetics is one of the various approaches adopted to understand and control mammalian sleep. Reverse genetics, which is usually applied to analyze sleep in gene-deficient mice, has been the mainstream field of genetic studies on sleep for the past three decades and has revealed that various molecules, including orexin, are involved in sleep regulation. Recently, forward genetic studies in humans and mice have identified gene mutations responsible for heritable sleep abnormalities, such as SIK3, NALCN, DEC2, the neuropeptide S receptor, and β1 adrenergic receptor. Furthermore, the protein kinase A-SIK3 pathway was shown to represent the intracellular neural signaling for sleep need. Large-scale genome-wide analyses of human sleep have been conducted, and many gene loci associated with individual differences in sleep have been found. The development of genome-editing technology and gene transfer by an adeno-associated virus has updated and expanded the genetic studies on mammals. These efforts are expected to elucidate the mechanisms of sleep–wake regulation and develop new therapeutic interventions for sleep disorders.
Collapse
Affiliation(s)
- Hiromasa Funato
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Department of Anatomy, Faculty of Medicine, Toho University, Ota-ku, Tokyo 951-8585, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas 75390, Texas, USA
- Life Science Center, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
113
|
Yang MJ, Zhang Z, Wang YJ, Li JC, Guo QL, Chen X, Wang E. Association of Nap Frequency With Hypertension or Ischemic Stroke Supported by Prospective Cohort Data and Mendelian Randomization in Predominantly Middle-Aged European Subjects. Hypertension 2022; 79:1962-1970. [PMID: 35876003 DOI: 10.1161/hypertensionaha.122.19120] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE The aim of this study was to investigate the association between daytime napping frequency and the incidence of essential hypertension or stroke as well as to validate causality in this relationship via Mendelian randomization (MR). METHODS We conducted Cox regression analysis on 358 451 participants free of hypertension or stroke from UK Biobank. To validate the results of the observational analysis, we conducted a 2-sample MR for daytime napping frequency (123 single-nucleotide polymorphisms) with essential hypertension in FinnGen Biobank, stroke, and ischemic stroke in MEGASTROKE consortium and performed a corresponding 1-sample MR on the UK Biobank data. RESULTS Compared with never napping, usually napping was associated with a higher risk of essential hypertension (hazard ratio, 1.12 [95% CI, 1.08-1.17]), stroke (hazard ratio, 1.24 [95% CI, 1.10-1.39], and ischemic stroke (hazard ratio, 1.20 [95% CI, 1.05-1.36]) in our prospective observational analysis. Both the 1-sample and 2-sample MR results indicated that increased daytime napping frequency was likely to be a potential causal risk factor for essential hypertension in FinnGEN (odds ratio, 1.43 [95% CI, 1.06-1.92]) and UK Biobank (odds ratio, 1.40 [95% CI, 1.28-1.58]). The 2-sample MR results supported the potential causal effect of nap frequency on ischemic stroke in MEGASTROKE (odds ratio, 1.29 [95% CI, 1.04-1.62]). CONCLUSIONS Prospective observational and MR analyses provided evidence that increased daytime nap frequency may represent a potential causal risk factor for essential hypertension. The potential causal association of increased nap frequency with ischemic stroke was supported by 2-sample MR and prospective observational results.
Collapse
Affiliation(s)
- Min-Jing Yang
- Department of Anesthesiology (M.-j.Y., Z.Z., Q.-l.G.) Central South University, Changsha, Hunan, China
| | - Zhong Zhang
- Department of Anesthesiology (M.-j.Y., Z.Z., Q.-l.G.) Central South University, Changsha, Hunan, China
| | - Yi-Jing Wang
- National Clinical Research Center for Geriatric Disorders (Y.-j.W., J.-c.L.) Central South University, Changsha, Hunan, China.,Xiangya Hospital, Centre for Medical Genetics, Hunan Key Laboratory of Medical Genetics, School of Life Sciences (Y.-j.W., J.-c.L.), Central South University, Changsha, Hunan, China
| | - Jin-Chen Li
- National Clinical Research Center for Geriatric Disorders (Y.-j.W., J.-c.L.) Central South University, Changsha, Hunan, China.,Xiangya Hospital, Centre for Medical Genetics, Hunan Key Laboratory of Medical Genetics, School of Life Sciences (Y.-j.W., J.-c.L.), Central South University, Changsha, Hunan, China
| | - Qu-Lian Guo
- Department of Anesthesiology (M.-j.Y., Z.Z., Q.-l.G.) Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Anesthesiology (X.C., E.W.) Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders (X.C., E.W.) Central South University, Changsha, Hunan, China
| | - E Wang
- Department of Anesthesiology (X.C., E.W.) Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders (X.C., E.W.) Central South University, Changsha, Hunan, China
| |
Collapse
|
114
|
Orexin 2 receptor-selective agonist danavorexton improves narcolepsy phenotype in a mouse model and in human patients. Proc Natl Acad Sci U S A 2022; 119:e2207531119. [PMID: 35994639 PMCID: PMC9436334 DOI: 10.1073/pnas.2207531119] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Narcolepsy type 1 (NT1) is a sleep disorder caused by a loss of orexinergic neurons. Narcolepsy type 2 (NT2) is heterogeneous; affected individuals typically have normal orexin levels. Following evaluation in mice, the effects of the orexin 2 receptor (OX2R)-selective agonist danavorexton were evaluated in single- and multiple-rising-dose studies in healthy adults, and in individuals with NT1 and NT2. In orexin/ataxin-3 narcolepsy mice, danavorexton reduced sleep/wakefulness fragmentation and cataplexy-like episodes during the active phase. In humans, danavorexton administered intravenously was well tolerated and was associated with marked improvements in sleep latency in both NT1 and NT2. In individuals with NT1, danavorexton dose-dependently increased sleep latency in the Maintenance of Wakefulness Test, up to the ceiling effect of 40 min, in both the single- and multiple-rising-dose studies. These findings indicate that OX2Rs remain functional despite long-term orexin loss in NT1. OX2R-selective agonists are a promising treatment for both NT1 and NT2.
Collapse
|
115
|
Li S, Liu B, Li QH, Zhang Y, Zhang H, Gao S, Wang L, Wang T, Han Z, Liu G, Wang K. Evaluating the Bidirectional Causal Association Between Daytime Napping and Alzheimer’s Disease Using Mendelian Randomization. J Alzheimers Dis 2022; 89:1315-1322. [DOI: 10.3233/jad-220497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Until now, both cross-sectional and longitudinal studies have identified controversial findings about the association between daytime napping and Alzheimer’s disease (AD) or cognitive decline. Therefore, it remains unclear about the causal association between daytime napping and AD or cognitive decline. Objective: We aim to investigate the causal association between daytime napping and AD. Methods: Here, we conduct a bidirectional Mendelian randomization (MR) analysis to investigate the causal association between daytime napping and AD using large-scale GWAS datasets from daytime napping including 452,633 individuals of European ancestry and AD including 35,274 AD and 59,163 controls of European ancestry. A total of five MR methods are selected including inverse-variance weighted (IVW), weighted median, MR-Egger, MR-PRESSO, and contamination mixture method. Results: MR analysis highlights significant causal association of AD with daytime napping using IVW (beta = -0.006, 95% CI [–0.009, –0.002], p = 2.00E-03), but no significant causal association of daytime napping with AD using IVW (OR = 0.76, 95% CI 0.53-1.10, p = 1.40E-01). Conclusion: Our bidirectional MR analysis demonstrates the causal effect of AD on daytime napping. However, there is no causal effect of daytime napping on AD. Our current findings are consistent with recent evidence from other MR studies that highlight little evidence supporting a causal effect of sleep traits on AD and support the causal effect of AD on sleep traits.
Collapse
Affiliation(s)
- Sijie Li
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Centerfor Brain Disorders, Capital Medical University, Beijing, China
| | - Bian Liu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qing-hao Li
- Children’s Center, the Affiliated Taian City Centeral Hospital of Qingdao University, Taian, Shandong, China
| | - Yan Zhang
- Department of Pathology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Haihua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Centerfor Brain Disorders, Capital Medical University, Beijing, China
| | - Shan Gao
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Centerfor Brain Disorders, Capital Medical University, Beijing, China
| | - Longcai Wang
- Department of Anesthesiology, TheAffiliated Hospital of Weifang Medical University, Weifang, China
| | - Tao Wang
- Chinese Institute for Brain Research, Beijing, China
| | - Zhifa Han
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy ofMedical Sciences, Beijing, China
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Centerfor Brain Disorders, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Key Laboratoryof Cerebral Microcirculation in Universities of Shandong; Departmentof Neurology, Second Affiliated Hospital; Shandong First MedicalUniversity & Shandong Academy of Medical Sciences, Taian, Shandong, China
- Beijing Key Laboratory of HypoxiaTranslational Medicine, National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kun Wang
- Children’s Center, the Affiliated Taian City Centeral Hospital of Qingdao University, Taian, Shandong, China
| |
Collapse
|
116
|
Bao W, Qi L, Bao Y, Wang S, Li W. Alleviating insomnia should decrease the risk of irritable bowel syndrome: Evidence from Mendelian randomization. Front Pharmacol 2022; 13:900788. [PMID: 36071849 PMCID: PMC9442781 DOI: 10.3389/fphar.2022.900788] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 07/06/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Associations have been reported between sleep and irritable bowel syndrome (IBS). However, whether there exists a causation between them is still unknown. Methods: We employed the Mendelian randomization (MR) design to explore the causal relationship between sleep and IBS. All genetic associations with sleep-related traits reached genome-wide significance (p-value < 5 × 10-8). The genetic associations with IBS were obtained from two independent large genome-wide association studies (GWAS), where non-FinnGen GWAS was in the discovery stage and FinnGen GWAS was in the validation stage. Primarily, the inverse-variance weighted method was employed to estimate the causal effects, and a meta-analysis was performed to combine the MR estimates. Results: In the discovery, we observed that genetic liability to the “morning” chronotype could lower the risk of IBS [OR = 0.81 (0.76, 0.86)]. Also, the genetic liability to insomnia can increase the risk of IBS [OR = 2.86 (1.94, 4.23)] and such causation was supported by short sleep duration. In the validation stage, only insomnia displayed statistical significance [OR = 2.22 (1.09, 4.51)]. The meta-analysis suggested two genetically-determined sleep exposures can increase the risk of IBS, including insomnia [OR = 2.70 (1.92, 3.80)] and short sleep duration [OR = 2.46 (1.25, 4.86)]. Furthermore, the multivariable MR analysis suggested insomnia is an independent risk factor for IBS after adjusting for chronotype [OR = 2.32 (1.57, 3.43)] and short sleep duration [OR = 1.45 (1.13, 1.85)]. IBS cannot increase the risk of insomnia in the reverse MR analysis. Conclusion: Genetic susceptibility to insomnia can increase the risk of IBS, and improving sleep quality, especially targeting insomnia, can help to prevent IBS.
Collapse
Affiliation(s)
- Wenzhao Bao
- Department of Anesthesiology, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China
| | - Li Qi
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Yin Bao
- Department of Anesthesiology, The Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, China
| | - Sai Wang
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
| | - Wei Li
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Wei Li,
| |
Collapse
|
117
|
Sun J, Ma C, Zhao M, Magnussen CG, Xi B. Daytime napping and cardiovascular risk factors, cardiovascular disease, and mortality: A systematic review. Sleep Med Rev 2022; 65:101682. [PMID: 36027794 DOI: 10.1016/j.smrv.2022.101682] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 10/15/2022]
Abstract
Associations between night sleep duration and cardiovascular risk factors in adults have been well documented. However, the associations for daytime napping remain unclear. In this review, six databases were searched for eligible publications to April 8, 2022. A total of 11 articles were identified for umbrella review on the association of daytime napping with diabetes, metabolic syndrome (MetS), cardiovascular disease (CVD), and mortality in adults, 97 for systematic review on the association with CVD and several CVD risk factors. Our umbrella review showed that the associations of daytime napping with diabetes, MetS, CVD, and mortality in most meta-analyses were mainly supported by weak or suggestive evidence. Our systematic review showed that long daytime napping (≥1 h/d) was associated with higher odds of several CVD risk factors, CVD, and mortality, but no significant association was found between short daytime napping and most of the abovementioned outcomes. Our dose-response meta-analyses showed that daytime napping <30 min/d was not significantly associated with higher odds of most CVD risk factors and CVD among young and middle-aged adults. However, among older adults aged >60 years, we observed significant dose-response associations of daytime napping with higher odds of diabetes, dyslipidemia, MetS, and mortality starting from 0 min/d.
Collapse
Affiliation(s)
- Jiahong Sun
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuanwei Ma
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Costan G Magnussen
- Baker Heart and Diabetes Institute, Melbourne, Australia; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
118
|
Wang Z, Yang W, Li X, Qi X, Pan KY, Xu W. Association of Sleep Duration, Napping, and Sleep Patterns With Risk of Cardiovascular Diseases: A Nationwide Twin Study. J Am Heart Assoc 2022; 11:e025969. [PMID: 35881527 PMCID: PMC9375484 DOI: 10.1161/jaha.122.025969] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background Although sleep disorders have been linked to cardiovascular diseases (CVDs), the association between sleep characteristics and CVDs remains inconclusive. We aimed to examine the association of nighttime sleep duration, daytime napping, and sleep patterns with CVDs and explore whether genetic and early‐life environmental factors account for this association. Methods and Results In the Swedish Twin Registry, 12 268 CVD‐free twin individuals (mean age=70.3 years) at baseline were followed up to 18 years to detect incident CVDs. Sleep duration, napping, and sleep patterns (assessed by sleep duration, chronotype, insomnia, snoring, and daytime sleepiness) were self‐reported at baseline. CVDs were ascertained through the Swedish National Patient Registry and the Cause of Death Register. Data were analyzed using a Cox model. In the multiadjusted Cox model, compared with 7 to 9 hours/night, the hazard ratios (HRs) of CVDs were 1.14 (95% CI, 1.01–1.28) for <7 hours/night and 1.10 (95% CI, 1.00–1.21) for ≥10 hours/night, respectively. Compared with no napping, napping 1 to 30 minutes (HR, 1.11 [95% CI, 1.03–1.18]) and >30 minutes (HR, 1.23 [95% CI, 1.14–1.33]) were related to CVDs. Furthermore, a poor sleep pattern was associated with CVDs (HR, 1.22 [95% CI, 1.05–1.41]). The co‐twin matched control analyses showed similar results as the unmatched analyses, and there was no significant interaction between sleep characteristics and zygosity (P values >0.05). Conclusions Short or long sleep (<7 or ≥10 hours/night), napping, and poor sleep patterns are associated with an increased CVD risk. Genetic and early‐life environmental factors may not account for the sleep–CVD association.
Collapse
Affiliation(s)
- Zhiyu Wang
- Department of Epidemiology and Biostatistics, School of Public Health Tianjin Medical University Tianjin China.,Tianjin Key Laboratory of Environment Nutrition and Public Health Tianjin China.,Center for International Collaborative Research on Environment Nutrition and Public Health Tianjin China
| | - Wenzhe Yang
- Department of Epidemiology and Biostatistics, School of Public Health Tianjin Medical University Tianjin China.,Tianjin Key Laboratory of Environment Nutrition and Public Health Tianjin China.,Center for International Collaborative Research on Environment Nutrition and Public Health Tianjin China
| | - Xuerui Li
- Department of Epidemiology and Biostatistics, School of Public Health Tianjin Medical University Tianjin China.,Tianjin Key Laboratory of Environment Nutrition and Public Health Tianjin China.,Center for International Collaborative Research on Environment Nutrition and Public Health Tianjin China
| | - Xiuying Qi
- Department of Epidemiology and Biostatistics, School of Public Health Tianjin Medical University Tianjin China.,Tianjin Key Laboratory of Environment Nutrition and Public Health Tianjin China.,Center for International Collaborative Research on Environment Nutrition and Public Health Tianjin China
| | - Kuan-Yu Pan
- Department of Psychiatry, Amsterdam Public Health Amsterdam University Medical Center, Vrije Universiteit Amsterdam The Netherlands
| | - Weili Xu
- Department of Epidemiology and Biostatistics, School of Public Health Tianjin Medical University Tianjin China.,Tianjin Key Laboratory of Environment Nutrition and Public Health Tianjin China.,Center for International Collaborative Research on Environment Nutrition and Public Health Tianjin China.,Aging Research Center, Department of Neurobiology, Health Care Sciences and Society Karolinska Institutet and Stockholm University Stockholm Sweden
| |
Collapse
|
119
|
Nassan M, Daghlas I, Winkelman JW, Dashti HS, Saxena R. Genetic evidence for a potential causal relationship between insomnia symptoms and suicidal behavior: a Mendelian randomization study. Neuropsychopharmacology 2022; 47:1672-1679. [PMID: 35538198 PMCID: PMC9283512 DOI: 10.1038/s41386-022-01319-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 12/04/2022]
Abstract
Insomnia and restless leg syndrome (RLS) are associated with increased risk for suicidal behavior (SB), which is often comorbid with mood or thought disorders; however, it is unclear whether these relationships are causal. We performed a two-sample Mendelian randomization study using summary-level genetic associations with insomnia symptoms and RLS against the outcomes of risk of major depressive disorder (MDD), bipolar disorder (BP), schizophrenia (SCZ), and SB. The inverse-variance weighted method was used in the main analysis. We performed replication and sensitivity analyses to examine the robustness of the results. We identified outcome cohorts for MDD (n = 170,756 cases/329,443 controls), BP (n = 20,352/31,358), SCZ (n = 69,369/236,642), SB-Cohort-2019 (n = 6569/14,996 all with MDD, BP or SCZ; and SB within individual disease categories), and SB-Cohort-2020 (n = 29,782/519,961). Genetically proxied liability to insomnia symptoms significantly associated with increased risk of MDD (odds ratio (OR) = 1.23, 95% confidence interval (CI) = 1.2-1.26, P = 1.37 × 10-61), BP (OR = 1.15, 95% CI = 1.07-1.23, P = 5.11 × 10-5), SB-Cohort-2019 (OR = 1.17, 95% CI = 1.07-1.27, P = 2.30 × 10-4), SB-Cohort-2019 in depressed patients (OR = 1.34, 95% CI = 1.16-1.54, P = 5.97 × 10-5), and SB-Cohort-2020 (OR = 1.24, 95% CI = 1.18-1.3, P = 1.47 × 10-18). Genetically proxied liability to RLS did not significantly influence the risk of any of the outcomes (all corrected P > 0.05). Results were replicated for insomnia with MDD and SB in Mass General Brigham Biobank and were consistent in multiple lines of sensitivity analyses. In conclusion, human genetic evidence supports for the first time a potentially independent and causal effect of insomnia on SB and encourages further clinical investigation of treatment of insomnia for prevention or treatment of SB.
Collapse
Affiliation(s)
- Malik Nassan
- Division of Sleep Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Iyas Daghlas
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - John W Winkelman
- Departments of Psychiatry and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hassan S Dashti
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
120
|
Wang Z, Chen M, Wei YZ, Zhuo CG, Xu HF, Li WD, Ma L. The causal relationship between sleep traits and the risk of schizophrenia: a two-sample bidirectional Mendelian randomization study. BMC Psychiatry 2022; 22:399. [PMID: 35705942 PMCID: PMC9202113 DOI: 10.1186/s12888-022-03946-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 04/19/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Observational studies suggest that sleep disturbances are commonly associated with schizophrenia. However, it is uncertain whether this relationship is causal. To investigate the bidirectional causal relation between sleep traits and schizophrenia, we performed a two-sample bidirectional Mendelian randomization (MR) study with the fixed effects inverse-variance weighted (IVW) method. METHODS As genetic variants for sleep traits, we selected variants from each meta-analysis of genome-wide association studies (GWASs) conducted using data from the UK Biobank (UKB). RESULTS We found that morning diurnal preference was associated with a lower risk of schizophrenia, while long sleep duration and daytime napping were associated with a higher risk of schizophrenia. Multivariable MR analysis also showed that sleep duration was associated with a higher risk of schizophrenia after adjusting for other sleep traits. Furthermore, genetically predicted schizophrenia was negatively associated with morning diurnal preference and short sleep duration and was positively associated with daytime napping and long sleep duration. CONCLUSIONS Therefore, sleep traits were identified as a potential treatment target for patients with schizophrenia.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Cardiovascular Surgery, School of Medicine, the First Affiliated Hospital of Zhejiang University, Number 79 Qingchun Road, Hangzhou, China
| | - Miao Chen
- Department of Cardiovascular Surgery, School of Medicine, the First Affiliated Hospital of Zhejiang University, Number 79 Qingchun Road, Hangzhou, China
| | - Yin-Ze Wei
- Department of Cardiovascular Surgery, School of Medicine, the First Affiliated Hospital of Zhejiang University, Number 79 Qingchun Road, Hangzhou, China
| | - Chen-Gui Zhuo
- Department of Cardiology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Hong-Fei Xu
- Department of Cardiovascular Surgery, School of Medicine, the First Affiliated Hospital of Zhejiang University, Number 79 Qingchun Road, Hangzhou, China
| | - Wei-Dong Li
- Department of Cardiovascular Surgery, School of Medicine, the First Affiliated Hospital of Zhejiang University, Number 79 Qingchun Road, Hangzhou, China.
| | - Liang Ma
- Department of Cardiovascular Surgery, School of Medicine, the First Affiliated Hospital of Zhejiang University, Number 79 Qingchun Road, Hangzhou, China.
| |
Collapse
|
121
|
Liu Z, Luo Y, Su Y, Wei Z, Li R, He L, Yang L, Pei Y, Ren J, Peng X, Hu X. Associations of sleep and circadian phenotypes with COVID-19 susceptibility and hospitalization: an observational cohort study based on the UK Biobank and a two-sample Mendelian randomization study. Sleep 2022; 45:6509040. [PMID: 35034128 PMCID: PMC8807236 DOI: 10.1093/sleep/zsac003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/15/2021] [Indexed: 02/05/2023] Open
Abstract
Study Objectives Sleep and circadian phenotypes are associated with several diseases. The present study aimed to investigate whether sleep and circadian phenotypes were causally linked with coronavirus disease 2019 (COVID-19)-related outcomes. Methods Habitual sleep duration, insomnia, excessive daytime sleepiness, daytime napping, and chronotype were selected as exposures. Key outcomes included positivity and hospitalization for COVID-19. In the observation cohort study, multivariable risk ratios (RRs) and their 95% confidence intervals (CIs) were calculated. Two-sample Mendelian randomization (MR) analyses were conducted to estimate the causal effects of the significant findings in the observation analyses. Beta values and the corresponding 95% CIs were calculated and compared using the inverse variance weighting, weighted median, and MR-Egger methods. Results In the UK Biobank cohort study, both often excessive daytime sleepiness and sometimes daytime napping were associated with hospitalized COVID-19 (excessive daytime sleepiness [often vs. never]: RR=1.24, 95% CI=1.02-1.5; daytime napping [sometimes vs. never]: RR=1.12, 95% CI=1.02-1.22). In addition, sometimes daytime napping was also associated with an increased risk of COVID-19 susceptibility (sometimes vs. never: RR= 1.04, 95% CI=1.01-1.28). In the MR analyses, excessive daytime sleepiness was found to increase the risk of hospitalized COVID-19 (MR IVW method: OR = 4.53, 95% CI = 1.04-19.82), whereas little evidence supported a causal link between daytime napping and COVID-19 outcomes. Conclusions Observational and genetic evidence supports a potential causal link between excessive daytime sleepiness and an increased risk of COVID-19 hospitalization, suggesting that interventions targeting excessive daytime sleepiness symptoms might decrease severe COVID-19 rate.
Collapse
Affiliation(s)
- Zheran Liu
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Yaxin Luo
- Department of Epidemiology and Biostatistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Yonglin Su
- West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | | | - Ruidan Li
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Ling He
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Lianlian Yang
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Yiyan Pei
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Jianjun Ren
- Department of Otolaryngology-Head and Neck Surgery, West China Biomedical Big Data Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy and National Clinical Research Center for Geriatrics, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| | - Xiaolin Hu
- West China School of Nursing, West China Hospital, Sichuan University, Chengdu, Sichuan,China
| |
Collapse
|
122
|
Miyagawa T, Tanaka S, Shimada M, Sakai N, Tanida K, Kotorii N, Kotorii T, Ariyoshi Y, Hashizume Y, Ogi K, Hiejima H, Kanbayashi T, Imanishi A, Ikegami A, Kamei Y, Hida A, Wada Y, Miyamoto M, Takami M, Kondo H, Tamura Y, Taniyama Y, Omata N, Mizuno T, Moriya S, Furuya H, Kato M, Kato K, Ishigooka J, Tsuruta K, Chiba S, Yamada N, Okawa M, Hirata K, Kuroda K, Kume K, Uchimura N, Kitada M, Kodama T, Inoue Y, Nishino S, Mishima K, Tokunaga K, Honda M. A rare genetic variant in the cleavage site of prepro-orexin is associated with idiopathic hypersomnia. NPJ Genom Med 2022; 7:29. [PMID: 35414074 PMCID: PMC9005711 DOI: 10.1038/s41525-022-00298-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/04/2022] [Indexed: 11/08/2022] Open
Abstract
Idiopathic hypersomnia (IH) is a rare, heterogeneous sleep disorder characterized by excessive daytime sleepiness. In contrast to narcolepsy type 1, which is a well-defined type of central disorders of hypersomnolence, the etiology of IH is poorly understood. No susceptibility loci associated with IH have been clearly identified, despite the tendency for familial aggregation of IH. We performed a variation screening of the prepro-orexin/hypocretin and orexin receptors genes and an association study for IH in a Japanese population, with replication (598 patients and 9826 controls). We identified a rare missense variant (g.42184347T>C; p.Lys68Arg; rs537376938) in the cleavage site of prepro-orexin that was associated with IH (minor allele frequency of 1.67% in cases versus 0.32% in controls, P = 2.7 × 10-8, odds ratio = 5.36). Two forms of orexin (orexin-A and -B) are generated from cleavage of one precursor peptide, prepro-orexin. The difference in cleavage efficiency between wild-type (Gly-Lys-Arg; GKR) and mutant (Gly-Arg-Arg; GRR) peptides was examined by assays using proprotein convertase subtilisin/kexin (PCSK) type 1 and PCSK type 2. In both PCSK1 and PCSK2 assays, the cleavage efficiency of the mutant peptide was lower than that of the wild-type peptide. We also confirmed that the prepro-orexin peptides themselves transmitted less signaling through orexin receptors than mature orexin-A and orexin-B peptides. These results indicate that a subgroup of IH is associated with decreased orexin signaling, which is believed to be a hallmark of narcolepsy type 1.
Collapse
Affiliation(s)
- Taku Miyagawa
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Susumu Tanaka
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Mihoko Shimada
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriaki Sakai
- Sleep and Circadian Neurobiology Laboratory, School of Medicine, Stanford University, Stanford, CA, USA
| | - Kotomi Tanida
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nozomu Kotorii
- Department of Neuropsychiatry, Kurume University School of Medicine, Fukuoka, Japan
- Kotorii Isahaya Hospital, Nagasaki, Japan
| | | | | | - Yuji Hashizume
- Department of Neuropsychiatry, Kurume University School of Medicine, Fukuoka, Japan
| | - Kimihiro Ogi
- Department of Neuropsychiatry, Kurume University School of Medicine, Fukuoka, Japan
| | - Hiroshi Hiejima
- Department of Neuropsychiatry, Kurume University School of Medicine, Fukuoka, Japan
| | - Takashi Kanbayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
- Ibaraki Prefectural Medical Center of Psychiatry, Ibaraki, Japan
| | - Aya Imanishi
- Department of Neuropsychiatry, Akita University Graduate School of Medicine, Akita, Japan
| | | | - Yuichi Kamei
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
- Kamisuwa Hospital, Nagano, Japan
| | - Akiko Hida
- Department of Sleep-Wake Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yamato Wada
- Department of Psychiatry, Hannan Hospital, Osaka, Japan
| | | | - Masanori Takami
- Department of Psychiatry, Shiga University of Medical Science, Shiga, Japan
| | - Hideaki Kondo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
| | - Yoshiyuki Tamura
- Department of Psychiatry and Neurology, Asahikawa Medical University, Hokkaido, Japan
| | - Yukari Taniyama
- Department of Neurology, Junwakai Memorial Hospital, Miyazaki, Japan
| | - Naoto Omata
- Department of Nursing, Faculty of Health Science, Fukui Health Science University, Fukui, Japan
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Tomoyuki Mizuno
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Shunpei Moriya
- Department of Psychiatry, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | - Hirokazu Furuya
- Department of Neurology, Neuro-Muscular Center, National Omuta Hospital, Fukuoka, Japan
- Department of Neurology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Mitsuhiro Kato
- Department of Pediatrics, Yamagata University Faculty of Medicine, Yamagata, Japan
- Department of Pediatrics, Showa University School of Medicine, Tokyo, Japan
| | - Kayoko Kato
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | - Kazuhito Tsuruta
- Department of Neurology, Junwakai Memorial Hospital, Miyazaki, Japan
| | - Shigeru Chiba
- Department of Psychiatry and Neurology, Asahikawa Medical University, Hokkaido, Japan
| | - Naoto Yamada
- Department of Psychiatry, Shiga University of Medical Science, Shiga, Japan
| | - Masako Okawa
- Department of Sleep Medicine, Shiga University of Medical Science, Shiga, Japan
- Japan Foundation for Neuroscience and Mental Health, Tokyo, Japan
- Department of Somnology, Tokyo Medical University, Tokyo, Japan
| | - Koichi Hirata
- Department of Neurology, Dokkyo Medical University, Tochigi, Japan
| | - Kenji Kuroda
- Department of Psychiatry, Hannan Hospital, Osaka, Japan
| | - Kazuhiko Kume
- Sleep Center, Kuwamizu Hospital, Kumamoto, Japan
- Department of Stem Cell Biology, Institute of Molecular Genetics and Embryology, Kumamoto University, Kumamoto, Japan
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Aichi, Japan
| | - Naohisa Uchimura
- Department of Neuropsychiatry, Kurume University School of Medicine, Fukuoka, Japan
| | - Masaaki Kitada
- Department of Anatomy, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Tohru Kodama
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuichi Inoue
- Department of Somnology, Tokyo Medical University, Tokyo, Japan
- Yoyogi Sleep Disorder Center, Neuropsychiatric Research Institute, Tokyo, Japan
| | - Seiji Nishino
- Sleep and Circadian Neurobiology Laboratory, School of Medicine, Stanford University, Stanford, CA, USA
| | - Kazuo Mishima
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
- Department of Neuropsychiatry, Akita University Graduate School of Medicine, Akita, Japan
- Department of Sleep-Wake Disorders, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, Tokyo, Japan
| | - Makoto Honda
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Seiwa Hospital, Institute of Neuropsychiatry, Tokyo, Japan
| |
Collapse
|
123
|
Tran S, Prober DA. Validation of Candidate Sleep Disorder Risk Genes Using Zebrafish. Front Mol Neurosci 2022; 15:873520. [PMID: 35465097 PMCID: PMC9021570 DOI: 10.3389/fnmol.2022.873520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/14/2022] [Indexed: 12/31/2022] Open
Abstract
Sleep disorders and chronic sleep disturbances are common and are associated with cardio-metabolic diseases and neuropsychiatric disorders. Several genetic pathways and neuronal mechanisms that regulate sleep have been described in animal models, but the genes underlying human sleep variation and sleep disorders are largely unknown. Identifying these genes is essential in order to develop effective therapies for sleep disorders and their associated comorbidities. To address this unmet health problem, genome-wide association studies (GWAS) have identified numerous genetic variants associated with human sleep traits and sleep disorders. However, in most cases, it is unclear which gene is responsible for a sleep phenotype that is associated with a genetic variant. As a result, it is necessary to experimentally validate candidate genes identified by GWAS using an animal model. Rodents are ill-suited for this endeavor due to their poor amenability to high-throughput sleep assays and the high costs associated with generating, maintaining, and testing large numbers of mutant lines. Zebrafish (Danio rerio), an alternative vertebrate model for studying sleep, allows for the rapid and cost-effective generation of mutant lines using the CRISPR/Cas9 system. Numerous zebrafish mutant lines can then be tested in parallel using high-throughput behavioral assays to identify genes whose loss affects sleep. This process identifies a gene associated with each GWAS hit that is likely responsible for the human sleep phenotype. This strategy is a powerful complement to GWAS approaches and holds great promise to identify the genetic basis for common human sleep disorders.
Collapse
Affiliation(s)
| | - David A. Prober
- Division of Biology and Biological Engineering, Tianqiao and Chrissy Chen Institute for Neuroscience, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
124
|
Ulsa MC, Xi Z, Li P, Gaba A, Wong PM, Saxena R, Scheer FAJL, Rutter M, Akeju O, Hu K, Gao L. Association of Poor Sleep Burden in Middle Age and Older Adults With Risk for Delirium During Hospitalization. J Gerontol A Biol Sci Med Sci 2022; 77:507-516. [PMID: 34558609 PMCID: PMC8893188 DOI: 10.1093/gerona/glab272] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Delirium is a distressing neurocognitive disorder recently linked to sleep disturbances. However, the longitudinal relationship between sleep and delirium remains unclear. This study assessed the associations of poor sleep burden, and its trajectory, with delirium risk during hospitalization. METHODS About 321 818 participants from the UK Biobank (mean age 58 ± 8 years [SD]; range 37-74 years) reported (2006-2010) sleep traits (sleep duration, excessive daytime sleepiness, insomnia-type complaints, napping, and chronotype-a closely related circadian measure for sleep timing), aggregated into a sleep burden score (0-9). New-onset delirium (n = 4 775) was obtained from hospitalization records during a 12-year median follow-up. About 42 291 (mean age 64 ± 8 years; range 44-83 years) had repeat sleep assessment on average 8 years after their first. RESULTS In the baseline cohort, Cox proportional hazards models showed that moderate (aggregate scores = 4-5) and severe (scores = 6-9) poor sleep burden groups were 18% (hazard ratio = 1.18 [95% confidence interval: 1.08-1.28], p < .001) and 57% (1.57 [1.38-1.80], p < .001), more likely to develop delirium, respectively. The latter risk magnitude is equivalent to 2 additional cardiovascular risks. These findings appeared robust when restricted to postoperative delirium and after exclusion of underlying dementia. Higher sleep burden was also associated with delirium in the follow-up cohort. Worsening sleep burden (score increase ≥2 vs no change) further increased the risk for delirium (1.79 [1.23-2.62], p = .002) independent of their baseline sleep score and time lag. The risk was highest in those younger than 65 years at baseline (p for interaction <.001). CONCLUSION Poor sleep burden and worsening trajectory were associated with increased risk for delirium; promotion of sleep health may be important for those at higher risk.
Collapse
Affiliation(s)
- Ma Cherrysse Ulsa
- Medical Biodynamics Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Zheng Xi
- Medical Biodynamics Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Peng Li
- Medical Biodynamics Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Arlen Gaba
- Medical Biodynamics Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Patricia M Wong
- Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Richa Saxena
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Frank A J L Scheer
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Martin Rutter
- Division of Diabetes, Endocrinology & Gastroenterology, The University of Manchester, UK
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Kun Hu
- Medical Biodynamics Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Division of Sleep Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Lei Gao
- Medical Biodynamics Program, Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
125
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 3156] [Impact Index Per Article: 1052.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
126
|
Miyagawa T, Shimada M, Honda Y, Kodama T, Tokunaga K, Honda M. A variant in orexin receptor-2 is associated with self-reported daytime sleepiness in the Japanese population. J Hum Genet 2022; 67:377-380. [PMID: 35034961 DOI: 10.1038/s10038-022-01015-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 12/21/2022]
Abstract
Excessive daytime sleepiness is characterized by a persistent feeling of having trouble staying awake, typically with inappropriate sleep episodes. Orexin (hypocretin) is a neuropeptide that regulates sleep-wake cycles and rapid eye movement sleep. Several large-scale genome-wide association studies (GWASs) in European populations have found genetic variants in orexin receptor-1 (OX1R) and -2 (OX2R) that are associated with sleep traits including daytime sleepiness. To identify genetic variants associated with daytime sleepiness, we performed an association study of genetic variants in prepro-orexin, OX1R, and OX2R in 14,329 Japanese individuals from the Tohoku Medical Megabank Project cohort. A genetic variant in OX2R was significantly associated with self-reported daytime sleepiness after Bonferroni correction (rs188018846: P = 8.4E-05). In addition, a missense variant in OX2R identified by the European GWASs showed a nominally significant association with daytime sleepiness in a Japanese population (p.Ile308Val, rs2653349: P = 0.044). Multiple genetic variants in OX2R can affect daytime sleepiness in general populations.
Collapse
Affiliation(s)
- Taku Miyagawa
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan. .,Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Mihoko Shimada
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, Tokyo, Japan
| | - Yoshiko Honda
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tohru Kodama
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Katsushi Tokunaga
- Department of Human Genetics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Genome Medical Science Project (Toyama), National Center for Global Health and Medicine, Tokyo, Japan
| | - Makoto Honda
- Sleep Disorders Project, Department of Psychiatry and Behavioral Sciences, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Koishikawa Tokyo Hospital, Institute of Neuropsychiatry, Tokyo, Japan
| |
Collapse
|
127
|
Dashti HS, Miranda N, Cade BE, Huang T, Redline S, Karlson EW, Saxena R. Interaction of obesity polygenic score with lifestyle risk factors in an electronic health record biobank. BMC Med 2022; 20:5. [PMID: 35016652 PMCID: PMC8753909 DOI: 10.1186/s12916-021-02198-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Genetic and lifestyle factors have considerable effects on obesity and related diseases, yet their effects in a clinical cohort are unknown. This study in a patient biobank examined associations of a BMI polygenic risk score (PRS), and its interactions with lifestyle risk factors, with clinically measured BMI and clinical phenotypes. METHODS The Mass General Brigham (MGB) Biobank is a hospital-based cohort with electronic health record, genetic, and lifestyle data. A PRS for obesity was generated using 97 genetic variants for BMI. An obesity lifestyle risk index using survey responses to obesogenic lifestyle risk factors (alcohol, education, exercise, sleep, smoking, and shift work) was used to dichotomize the cohort into high and low obesogenic index based on the population median. Height and weight were measured at a clinical visit. Multivariable linear cross-sectional associations of the PRS with BMI and interactions with the obesity lifestyle risk index were conducted. In phenome-wide association analyses (PheWAS), similar logistic models were conducted for 675 disease outcomes derived from billing codes. RESULTS Thirty-three thousand five hundred eleven patients were analyzed (53.1% female; age 60.0 years; BMI 28.3 kg/m2), of which 17,040 completed the lifestyle survey (57.5% female; age: 60.2; BMI: 28.1 (6.2) kg/m2). Each standard deviation increment in the PRS was associated with 0.83 kg/m2 unit increase in BMI (95% confidence interval (CI) =0.76, 0.90). There was an interaction between the obesity PRS and obesity lifestyle risk index on BMI. The difference in BMI between those with a high and low obesogenic index was 3.18 kg/m2 in patients in the highest decile of PRS, whereas that difference was only 1.55 kg/m2 in patients in the lowest decile of PRS. In PheWAS, the obesity PRS was associated with 40 diseases spanning endocrine/metabolic, circulatory, and 8 other disease groups. No interactions were evident between the PRS and the index on disease outcomes. CONCLUSIONS In this hospital-based clinical biobank, obesity risk conferred by common genetic variants was associated with elevated BMI and this risk was attenuated by a healthier patient lifestyle. Continued consideration of the role of lifestyle in the context of genetic predisposition in healthcare settings is necessary to quantify the extent to which modifiable lifestyle risk factors may moderate genetic predisposition and inform clinical action to achieve personalized medicine.
Collapse
Affiliation(s)
- Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA. .,Broad Institute, Cambridge, MA, USA. .,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Nicole Miranda
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Brian E Cade
- Broad Institute, Cambridge, MA, USA.,Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Tianyi Huang
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Elizabeth W Karlson
- Mass General Brigham Personalized Medicine, Mass General Brigham HealthCare, Boston, MA, USA.,Department of Medicines, Brigham and Women's Hospital and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.,Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Broad Institute, Cambridge, MA, USA.,Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.,Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
128
|
Xiao JY, Zhang WS, Jiang CQ, Jin YL, Zhu F, Cheng KK, Lam TH, Xu L. Obesity indicators as mediators of association between daytime napping and type 2 diabetes mellitus: the Guangzhou Biobank Cohort Study. BMC Public Health 2022; 22:56. [PMID: 35012516 PMCID: PMC8744231 DOI: 10.1186/s12889-021-12451-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/21/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To examine the mediating effect of obesity indicators on the association between daytime napping and type 2 diabetes mellitus (T2DM) qualitatively and quantitatively using baseline data from the Guangzhou Biobank Cohort Study. METHODS Twenty-nine thousand three hundred fifty-five participants aged 50+ years were included in this cross-sectional study. Mediation analysis was used to assess the mediating effect of body mass index (BMI), waist circumference (WC), hip circumference (HC), waist-to-hip ratio (WHR) and waist-to-height ratio (WHtR) on the association between daytime napping and T2DM after adjustment for sex, age, education, occupation, smoking status, alcohol use and physical activity. RESULTS The mean (standard deviation) age of participants was 61.5 ( 7.1) years. The prevalence of T2DM and daytime napping was 12.5% and 65.2%, respectively. After adjustment for potential confounders, WC, WHR and WHtR showed partial mediating effects on the association between daytime napping and T2DM, with the proportion (95% confidence interval) of mediation effect being 10.17% (8.14-14.43%), 14.91% (11.95-21.24%) and 9.36% (7.49-13.29%), respectively. No mediating effect of BMI or HC on the association between daytime napping and T2DM was found. CONCLUSIONS Our results showed significant mediating effects of WC, WHR and WHtR on the association between daytime napping and T2DM, suggesting that waist circumference management could be important in daytime nappers.
Collapse
Affiliation(s)
- Jing Yi Xiao
- School of Public Health, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province, China
| | - Wei Sen Zhang
- Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | | | - Ya Li Jin
- Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Feng Zhu
- Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Kar Keung Cheng
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Tai Hing Lam
- School of Public Health, the University of Hong Kong, Hong Kong, Hong Kong
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province, China. .,School of Public Health, the University of Hong Kong, Hong Kong, Hong Kong.
| |
Collapse
|
129
|
Yao Y, Jia Y, Wen Y, Cheng B, Cheng S, Liu L, Yang X, Meng P, Chen Y, Li C, Zhang J, Zhang Z, Pan C, Zhang H, Wu C, Wang X, Ning Y, Wang S, Zhang F. Genome-Wide Association Study and Genetic Correlation Scan Provide Insights into Its Genetic Architecture of Sleep Health Score in the UK Biobank Cohort. Nat Sci Sleep 2022; 14:1-12. [PMID: 35023977 PMCID: PMC8747788 DOI: 10.2147/nss.s326818] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 12/19/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Most previous genetic studies of sleep behaviors were conducted individually, without comprehensive consideration of the complexity of various sleep behaviors. Our aim is to identify the genetic architecture and potential biomarker of the sleep health score, which more powerfully represents overall sleep traits. PATIENTS AND METHODS We conducted a genome-wide association study (GWAS) of sleep health score (overall assessment of sleep duration, snoring, insomnia, chronotype, and daytime dozing) using 336,463 participants from the UK Biobank. Proteome-wide association study (PWAS) and transcriptome-wide association study (TWAS) were then performed to identify candidate genes at the protein and mRNA level, respectively. We finally used linkage disequilibrium score regression (LDSC) to estimate the genetic correlations between sleep health score and other functionally relevance traits. RESULTS GWAS identified multiple variants near known candidate genes associated with sleep health score, such as MEIS1, FBXL13, MED20 and SMAD5. HDHD2 (PPWAS = 0.0146) and GFAP (PPWAS = 0.0236) were identified associated with sleep health score by PWAS. TWAS identified ORC4 (PTWAS = 0.0212) and ZNF732 (PTWAS = 0.0349) considering mRNA expression level. LDSC found significant genetic correlations of sleep health score with 3 sleep behaviors (including insomnia, snoring, dozing), 4 psychiatry disorders (major depressive disorder, attention deficit/hyperactivity disorder, schizophrenia, autism spectrum disorder), and 9 plasma protein (such as Stabilin-1, Stromelysin-2, Cytochrome c) (all LDSC PLDSC < 0.05). CONCLUSION Our results advance the comprehensive understanding of the aetiology and genetic architecture of the sleep health score, refine the understanding of the relationship of sleep health score with other traits and diseases, and may serve as potential targets for future mechanistic studies of sleep phenotype.
Collapse
Affiliation(s)
- Yao Yao
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Peilin Meng
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yujing Chen
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chun'e Li
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Jingxi Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Zhen Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Chuyu Pan
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Huijie Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Cuiyan Wu
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xi Wang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yujie Ning
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Sen Wang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
130
|
Chen M, Peng WY, Tang TC, Zheng H. Differential Sleep Traits Have No Causal Effect on Inflammatory Bowel Diseases: A Mendelian Randomization Study. Front Pharmacol 2021; 12:763649. [PMID: 34916940 PMCID: PMC8669049 DOI: 10.3389/fphar.2021.763649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/03/2021] [Indexed: 12/22/2022] Open
Abstract
Background: Previous studies suggested an association of sleep disorders with inflammatory bowel disease (IBD) and indicated that using pharmacological treatments for the modulation of circadian rhythms might prevent IBD pathogenesis or aggravation, but whether the effect of sleep traits on IBD was causal is inconclusive and, therefore, prevents drug repurposing based on the previous studies. We aimed to examine the causal effect of different sleep traits on the pathogenesis of IBD. Methods: Genetic instruments for sleep traits were selected from the largest GWAS studies available in the UK Biobank (n = 449,734) and the 23andMe Research (n = 541,333). A two-sample Mendelian randomization (MR) study was conducted to examine the association of the genetic instruments with IBD (12,882 cases and 21,770 controls), ulcerative colitis (6,968 cases, 20,464 controls), and Crohn's disease (5,956 cases and 14,927 controls). We applied the inverse-variance weighted (IVW) method to estimate causal effects, and we used the weighted median and MR-Egger method for sensitivity analyses. Results: We found that sleep duration (OR, 1.00, 95% CI 1.00-1.01), short sleep duration (OR, 1.07, 95% CI 0.41-2.83), morningness (OR, 1.05, 95% CI 0.87-1.27), daytime napping (OR, 1.64, 95% CI 0.62-4.4), frequent insomnia (OR, 1.17, 95% CI 0.8-1.72), any insomnia (OR, 1.17, 95% CI 0.69-1.97), and snoring (OR, 0.31, 95% CI 0.06-1.54) had no causal effect on IBD, and these sleep traits had no causal effect on ulcerative colitis and Crohn's disease either. Most of the sensitivity analyses showed consistent results with those of the IVW method. Conclusion: Our MR study did not support the causal effect of sleep traits on IBD. Pharmacological modulation of circadian rhythms for the prevention of IBD pathogenesis was unwarranted.
Collapse
Affiliation(s)
- Min Chen
- Department of Colorectal Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Yan Peng
- The Third Hospital/Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tai-Chun Tang
- Department of Colorectal Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Zheng
- The Third Hospital/Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
131
|
Lin YS, Wang CC, Chen CY. GWAS Meta-Analysis Reveals Shared Genes and Biological Pathways between Major Depressive Disorder and Insomnia. Genes (Basel) 2021; 12:1506. [PMID: 34680902 PMCID: PMC8536096 DOI: 10.3390/genes12101506] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/17/2021] [Accepted: 09/24/2021] [Indexed: 11/27/2022] Open
Abstract
Major depressive disorder (MDD) is one of the most prevalent and disabling mental disorders worldwide. Among the symptoms of MDD, sleep disturbance such as insomnia is prominent, and the first reason patients may seek professional help. However, the underlying pathophysiology of this comorbidity is still elusive. Recently, genome-wide association studies (GWAS) have begun to unveil the genetic background of several psychiatric disorders, including MDD and insomnia. Identifying the shared genomic risk loci between comorbid psychiatric disorders could be a valuable strategy to understanding their comorbidity. This study seeks to identify the shared genes and biological pathways between MDD and insomnia based on their shared genetic variants. First, we performed a meta-analysis based on the GWAS summary statistics of MDD and insomnia obtained from Psychiatric Genomics Consortium and UK Biobank, respectively. Next, we associated shared genetic variants to genes using two gene mapping strategies: (a) positional mapping based on genomic proximity and (b) expression quantitative trait loci (eQTL) mapping based on gene expression linkage across multiple tissues. As a result, a total of 719 shared genes were identified. Over half (51%) of them are protein-coding genes. Functional enrichment analysis shows that the most enriched biological pathways are related to epigenetic modification, sensory perception, and immunologic signatures. We also identified druggable targets using a network approach. Together, these results may provide insights into understanding the genetic predisposition and underlying biological pathways of comorbid MDD and insomnia symptoms.
Collapse
Affiliation(s)
- Yi-Sian Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.L.); (C.-C.W.)
| | - Chia-Chun Wang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.L.); (C.-C.W.)
| | - Cho-Yi Chen
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; (Y.-S.L.); (C.-C.W.)
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
132
|
Pang L, Shah H, Xu Y, Qian S. Delta-5-desaturase: A novel therapeutic target for cancer management. Transl Oncol 2021; 14:101207. [PMID: 34438249 PMCID: PMC8390547 DOI: 10.1016/j.tranon.2021.101207] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
D5D is an independent prognostic factor in cancer. D5D aggravates cancer progression via mediating AA/PGE2 production from DGLA. AA/PGE2 promotes cancer progression via regulating the tumor microenvironment. Inhibition of D5D redirects COX-2 catalyzed DGLA peroxidation, producing 8-HOA. 8-HOA suppress cancer by regulating proliferation, apoptosis, and metastasis.
Delta-5 desaturase (D5D) is a rate-limiting enzyme that introduces double-bonds to the delta-5 position of the n-3 and n-6 polyunsaturated fatty acid chain. Since fatty acid metabolism is a vital factor in cancer development, several recent studies have revealed that D5D activity and expression could be an independent prognostic factor in cancers. However, the mechanistic basis of D5D in cancer progression is still controversial. The classical concept believes that D5D could aggravate cancer progression via mediating arachidonic acid (AA)/prostaglandin E2 production from dihomo-γ-linolenic acid (DGLA), resulting in activation of EP receptors, inflammatory pathways, and immunosuppression. On the contrary, D5D may prevent cancer progression through activating ferroptosis, which is iron-dependent cell death. Suppression of D5D by RNA interference and small-molecule inhibitor has been identified as a promising anti-cancer strategy. Inhibition of D5D could shift DGLA peroxidation pattern from generating AA to a distinct anti-cancer free radical byproduct, 8-hydroxyoctanoic acid, resulting in activation of apoptosis pathway and simultaneously suppression of cancer cell survival, proliferation, migration, and invasion. Hence, understanding the molecular mechanisms of D5D on cancer may therefore facilitate the development of novel therapeutical applications. Given that D5D may serve as a promising target in cancer, in this review, we provide an updated summary of current knowledge on the role of D5D in cancer development and potentially useful therapeutic strategies.
Collapse
Affiliation(s)
- Lizhi Pang
- Department of Pharmaceutical Sciences, North Dakota State University, Sudro 108, 1401 Albrecht Blvd, Fargo, ND, USA.
| | - Harshit Shah
- Department of Pharmaceutical Sciences, North Dakota State University, Sudro 108, 1401 Albrecht Blvd, Fargo, ND, USA
| | - Yi Xu
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Steven Qian
- Department of Pharmaceutical Sciences, North Dakota State University, Sudro 108, 1401 Albrecht Blvd, Fargo, ND, USA
| |
Collapse
|
133
|
De Lillo A, D'Antona S, Pathak GA, Wendt FR, De Angelis F, Fuciarelli M, Polimanti R. Cross-ancestry genome-wide association studies identified heterogeneous loci associated with differences of allele frequency and regulome tagging between participants of European descent and other ancestry groups from the UK Biobank. Hum Mol Genet 2021; 30:1457-1467. [PMID: 33890984 PMCID: PMC8283210 DOI: 10.1093/hmg/ddab114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 04/14/2021] [Accepted: 01/09/2021] [Indexed: 01/28/2023] Open
Abstract
To investigate cross-ancestry genetics of complex traits, we conducted a phenome-wide analysis of loci with heterogeneous effects across African, Admixed-American, Central/South Asian, East Asian, European and Middle Eastern participants of the UK Biobank (N = 441 331). Testing 843 phenotypes, we identified 82 independent genomic regions mapping variants showing genome-wide significant (GWS) associations (P < 5 × 10-8) in the trans-ancestry meta-analysis and GWS heterogeneity among the ancestry-specific effects. These included (i) loci with GWS association in one ancestry and concordant but heterogeneous effects among the other ancestries and (ii) loci with a GWS association in one ancestry group and an experiment-wide significant discordant effect (P < 6.1 × 10-4) in at least another ancestry. Since the trans-ancestry GWS associations were mostly driven by the European ancestry sample size, we investigated the differences of the allele frequency (ΔAF) and linkage disequilibrium regulome tagging (ΔLD) between European populations and the other ancestries. Within loci with concordant effects, the degree of heterogeneity was associated with European-Middle Eastern ΔAF (P = 9.04 × 10-6) and ΔLD of European populations with respect to African, Admixed-American and Central/South Asian groups (P = 8.21 × 10-4, P = 7.17 × 10-4 and P = 2.16 × 10-3, respectively). Within loci with discordant effects, ΔAF and ΔLD of European populations with respect to African and Central/South Asian ancestries were associated with the degree of heterogeneity (ΔAF: P = 7.69 × 10-3 and P = 5.31 × 10-3, ΔLD: P = 0.016 and P = 2.65 × 10-4, respectively). Considering the traits associated with cross-ancestry heterogeneous loci, we observed enrichments for blood biomarkers (P = 5.7 × 10-35) and physical appearance (P = 1.38 × 10-4). This suggests that these specific phenotypic classes may present considerable cross-ancestry heterogeneity owing to large allele frequency and LD variation among worldwide populations.
Collapse
Affiliation(s)
- Antonella De Lillo
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT 06516, USA
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Salvatore D'Antona
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Gita A Pathak
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT 06516, USA
- VA CT Healthcare Center, West Haven, CT 06516, USA
| | - Frank R Wendt
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT 06516, USA
- VA CT Healthcare Center, West Haven, CT 06516, USA
| | - Flavio De Angelis
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT 06516, USA
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
- VA CT Healthcare Center, West Haven, CT 06516, USA
| | - Maria Fuciarelli
- Department of Biology, University of Rome Tor Vergata, Rome 00133, Italy
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, West Haven, CT 06516, USA
- VA CT Healthcare Center, West Haven, CT 06516, USA
| |
Collapse
|
134
|
Dashti HS, Ordovás JM. Genetics of Sleep and Insights into Its Relationship with Obesity. Annu Rev Nutr 2021; 41:223-252. [PMID: 34102077 DOI: 10.1146/annurev-nutr-082018-124258] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Considerable recent advancements in elucidating the genetic architecture of sleep traits and sleep disorders may provide insight into the relationship between sleep and obesity. Despite the considerable involvement of the circadian clock in sleep and metabolism, few shared genes, including FTO, were implicated in genome-wide association studies (GWASs) of sleep and obesity. Polygenic scores composed of signals from GWASs of sleep traits show largely null associations with obesity, suggesting lead variants are unique to sleep. Modest genome-wide genetic correlations are observed between many sleep traits and obesity and are largest for snoring.Notably, U-shaped positive genetic correlations with body mass index (BMI) exist for both short and long sleep durations. Findings from Mendelian randomization suggest robust causal effects of insomnia on higher BMI and, conversely, of higher BMI on snoring and daytime sleepiness. Bidirectional effects between sleep duration and daytime napping with obesity may also exist. Limited gene-sleep interaction studies suggest that achieving favorable sleep, as part of a healthy lifestyle, may attenuate genetic predisposition to obesity, but whether these improvements produce clinically meaningful reductions in obesity risk remains unclear. Investigations of the genetic link between sleep and obesity for sleep disorders other than insomnia and in populations of non-European ancestry are currently limited. Expected final online publication date for the Annual Review of Nutrition, Volume 41 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Hassan S Dashti
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; .,Broad Institute, Cambridge, Massachusetts 02142, USA
| | - José M Ordovás
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, USA.,Precision Nutrition and Obesity Program, IMDEA Alimentación, 28049 Madrid, Spain
| |
Collapse
|
135
|
Martucci VL, Richmond B, Davis LK, Blackwell TS, Cox NJ, Samuels D, Velez Edwards D, Aldrich MC. Fate or coincidence: do COPD and major depression share genetic risk factors? Hum Mol Genet 2021; 30:619-628. [PMID: 33704461 PMCID: PMC8120137 DOI: 10.1093/hmg/ddab068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/24/2021] [Accepted: 02/27/2021] [Indexed: 01/12/2023] Open
Abstract
Major depressive disorder (MDD) is a common comorbidity in chronic obstructive pulmonary disease (COPD), affecting up to 57% of patients with COPD. Although the comorbidity of COPD and MDD is well established, the causal relationship between these two diseases is unclear. A large-scale electronic health record clinical biobank and genome-wide association study summary statistics for MDD and lung function traits were used to investigate potential shared underlying genetic susceptibility between COPD and MDD. Linkage disequilibrium score regression was used to estimate genetic correlation between phenotypes. Polygenic risk scores (PRS) for MDD and lung function traits were developed and used to perform a phenome-wide association study (PheWAS). Multi-trait-based conditional and joint analysis identified single-nucleotide polymorphisms (SNPs) influencing both lung function and MDD. We found genetic correlations between MDD and all lung function traits were small and not statistically significant. A PRS-MDD was significantly associated with an increased risk of COPD in a PheWAS [odds ratio (OR) = 1.12, 95% confidence interval (CI): 1.09-1.16] when adjusting for age, sex and genetic ancestry, but this relationship became attenuated when controlling for smoking history (OR = 1.08, 95% CI: 1.04-1.13). No significant associations were found between the lung function PRS and MDD. Multi-trait-based conditional and joint analysis identified three SNPs that may contribute to both traits, two of which were previously associated with mood disorders and COPD. Our findings suggest that the observed relationship between COPD and MDD may not be driven by a strong shared genetic architecture.
Collapse
Affiliation(s)
- Victoria L Martucci
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Bradley Richmond
- Department of Veterans Affairs Medical Center, Nashville, TN 37212, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lea K Davis
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Timothy S Blackwell
- Department of Veterans Affairs Medical Center, Nashville, TN 37212, USA
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David Samuels
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Digna Velez Edwards
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Melinda C Aldrich
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
136
|
Alimenti K, Chen A, Saxena R, Dashti HS. Habitual Sleep Duration, Daytime Napping, and Dietary Intake: A Mendelian Randomization Study. Curr Dev Nutr 2021; 5:nzab019. [PMID: 34095735 PMCID: PMC8171253 DOI: 10.1093/cdn/nzab019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/12/2021] [Accepted: 02/23/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Chronic inadequate sleep and frequent daytime napping may inflict deleterious health effects including weight gain, cardiometabolic and psychiatric diseases, and cancer. It is plausible that these relations may be partly influenced by the consumption of suboptimal diets. OBJECTIVES The study aimed to identify potential causal links of genetically proxied longer habitual sleep duration and more frequent daytime napping on 61 dietary variables derived from an FFQ. In addition, the study aimed to assess potential bidirectional causal links between habitual sleep duration or daytime napping and macronutrient composition. METHODS Genetic variants robustly associated with habitual sleep duration and daytime napping from published genome-wide association analyses were used. Outcomes included 61 dietary variables estimated from FFQs in the UK Biobank (n = 361,194). For bidirectional associations with macronutrient composition, genetic variants associated with percentage of energy from carbohydrate, fat, and protein were used. Two-sample Mendelian randomization (MR) effects were estimated with inverse-variance weighted (IVW) analysis. RESULTS In 2-sample MR, genetically proxied longer sleep duration was associated with a 0.068 (95% CI: 0.034, 0.103) category increase in salad/raw vegetable intake [P false discovery rate (FDR) = 0.006] per hour of sleep and with "no major dietary changes in the past 5 years" (P FDR = 0.043). No associations were evident for daytime napping on dietary variables (all P FDR > 0.05). In addition, there were no bidirectional associations between habitual sleep duration or daytime napping with the relative intake of carbohydrate, fat, and protein (all P IVW > 0.05). CONCLUSIONS In this MR study, there was modest evidence for associations between habitual sleep duration with dietary intake and no evidence for associations between daytime napping frequency with dietary intake. These preliminary findings suggest that changes to habitual sleep duration or daytime napping frequency may have limited impact on long-term changes in dietary intake.
Collapse
Affiliation(s)
- Kaitlyn Alimenti
- Center for Genomic Medicine, Massachusetts General Hospital,
Boston, MA, USA
| | - Angela Chen
- Center for Genomic Medicine, Massachusetts General Hospital,
Boston, MA, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital,
Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts
General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Sleep Medicine, Harvard Medical School, Boston,
MA, USA
| | - Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital,
Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts
General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|