Skip to main content
Advertisement
  • Loading metrics

Associations of variants In the hexokinase 1 and interleukin 18 receptor regions with oxyhemoglobin saturation during sleep

  • Brian E. Cade ,

    Roles Data curation, Formal analysis, Funding acquisition, Methodology, Software, Visualization, Writing – original draft, Writing – review & editing

    bcade@bwh.harvard.edu

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America

  • Han Chen,

    Roles Formal analysis, Methodology, Writing – original draft, Writing – review & editing

    Affiliations Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX United States of America, Center for Precision Health, School of Public Health and School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX United States of America

  • Adrienne M. Stilp,

    Roles Data curation, Formal analysis, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Tin Louie,

    Roles Data curation, Formal analysis, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Sonia Ancoli-Israel,

    Roles Conceptualization, Writing – review & editing

    Affiliation Department of Psychiatry, University of California, San Diego, CA, United States of America

  • Raanan Arens,

    Roles Conceptualization, Writing – review & editing

    Affiliation The Children’s Hospital at Montefiore, Division of Respiratory and Sleep Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America

  • Richard Barfield,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America

  • Jennifer E. Below,

    Roles Data curation, Writing – review & editing

    Affiliation Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, United States of America

  • Jianwen Cai,

    Roles Conceptualization, Resources, Writing – review & editing

    Affiliation Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States of America

  • Matthew P. Conomos,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Daniel S. Evans,

    Roles Data curation, Writing – review & editing

    Affiliation California Pacific Medical Center Research Institute, San Francisco, CA, United States of America

  • Alexis C. Frazier-Wood,

    Roles Data curation, Writing – review & editing

    Affiliation USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, United States of America

  • Sina A. Gharib,

    Roles Data curation, Writing – review & editing

    Affiliation Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle WA, United States of America

  • Kevin J. Gleason,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Department of Public Health Sciences, University of Chicago, Chicago, IL, United States of America

  • Daniel J. Gottlieb,

    Roles Conceptualization, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, VA Boston Healthcare System, Boston, MA, United States of America

  • David R. Hillman,

    Roles Conceptualization, Funding acquisition, Writing – review & editing

    Affiliation Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia

  • W. Craig Johnson,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • David J. Lederer,

    Roles Data curation, Writing – review & editing

    Affiliation Departments of Medicine and Epidemiology, Columbia University, New York, NY, United States of America

  • Jiwon Lee,

    Roles Data curation, Writing – review & editing

    Affiliation Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America

  • Jose S. Loredo,

    Roles Conceptualization, Writing – review & editing

    Affiliation Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, UC San Diego School of Medicine, La Jolla, CA, United States of America

  • Hao Mei,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Data Science, University of Mississippi Medical Center, Jackson, MS, United States of America

  • Sutapa Mukherjee,

    Roles Data curation, Resources, Writing – review & editing

    Affiliations Sleep Health Service, Respiratory and Sleep Services, Southern Adelaide Local Health Network, Adelaide, South Australia, Adelaide Institute for Sleep Health, Flinders University, Adelaide, South Australia

  • Sanjay R. Patel,

    Roles Conceptualization, Funding acquisition, Writing – review & editing

    Affiliation Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America

  • Wendy S. Post,

    Roles Data curation, Writing – review & editing

    Affiliation Division of Cardiology, Johns Hopkins University, Baltimore, MD, United States of America

  • Shaun M. Purcell,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America

  • Alberto R. Ramos,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States of America

  • Kathryn J. Reid,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America

  • Ken Rice,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Neomi A. Shah,

    Roles Data curation, Writing – review & editing

    Affiliation Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America

  • Tamar Sofer,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Kent D. Taylor,

    Roles Data curation, Writing – review & editing

    Affiliation The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, United States of America

  • Timothy A. Thornton,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Heming Wang,

    Roles Data curation, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America

  • Kristine Yaffe,

    Roles Data curation, Writing – review & editing

    Affiliations Department of Psychiatry, Neurology, and Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, CA, United States of America, San Francisco VA Medical Center, San Francisco, CA, United States of America

  • Phyllis C. Zee,

    Roles Data curation, Writing – review & editing

    Affiliation Department of Neurology, Center for Circadian and Sleep Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States of America

  • Craig L. Hanis,

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliation Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX United States of America

  • Lyle J. Palmer,

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliation School of Public Health, University of Adelaide, South Australia, Australia

  • Jerome I. Rotter,

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliation The Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, CA, United States of America

  • Katie L. Stone,

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliation California Pacific Medical Center Research Institute, San Francisco, CA, United States of America

  • Gregory J. Tranah,

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliation California Pacific Medical Center Research Institute, San Francisco, CA, United States of America

  • James G. Wilson,

    Roles Conceptualization, Funding acquisition, Resources, Writing – review & editing

    Affiliation Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson MS, United States of America

  • Shamil R. Sunyaev,

    Roles Conceptualization, Funding acquisition, Writing – review & editing

    Affiliations Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America, Division of Genetics, Brigham and Women's Hospital, Boston, MA, United States of America, Division of Medical Sciences, Harvard Medical School, Boston, MA, United States of America

  • Cathy C. Laurie,

    Roles Conceptualization, Methodology, Supervision, Writing – review & editing

    Affiliation Department of Biostatistics, University of Washington, Seattle, WA United States of America

  • Xiaofeng Zhu,

    Roles Conceptualization, Funding acquisition, Methodology, Writing – original draft, Writing – review & editing

    Affiliation Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States of America

  • Richa Saxena,

    Roles Conceptualization, Funding acquisition, Methodology, Writing – original draft, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, United States of America, Center for Genomic Medicine and Department of Anesthesia, Pain, and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, United States of America

  • Xihong Lin,

    Roles Conceptualization, Funding acquisition, Methodology, Supervision, Writing – review & editing

    Affiliation Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, United States of America

  •  [ ... ],
  • Susan Redline

    Roles Conceptualization, Data curation, Funding acquisition, Methodology, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    Affiliations Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA, United States of America, Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States of America, Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America

  • [ view all ]
  • [ view less ]

Abstract

Sleep disordered breathing (SDB)-related overnight hypoxemia is associated with cardiometabolic disease and other comorbidities. Understanding the genetic bases for variations in nocturnal hypoxemia may help understand mechanisms influencing oxygenation and SDB-related mortality. We conducted genome-wide association tests across 10 cohorts and 4 populations to identify genetic variants associated with three correlated measures of overnight oxyhemoglobin saturation: average and minimum oxyhemoglobin saturation during sleep and the percent of sleep with oxyhemoglobin saturation under 90%. The discovery sample consisted of 8,326 individuals. Variants with p < 1 × 10−6 were analyzed in a replication group of 14,410 individuals. We identified 3 significantly associated regions, including 2 regions in multi-ethnic analyses (2q12, 10q22). SNPs in the 2q12 region associated with minimum SpO2 (rs78136548 p = 2.70 × 10−10). SNPs at 10q22 were associated with all three traits including average SpO2 (rs72805692 p = 4.58 × 10−8). SNPs in both regions were associated in over 20,000 individuals and are supported by prior associations or functional evidence. Four additional significant regions were detected in secondary sex-stratified and combined discovery and replication analyses, including a region overlapping Reelin, a known marker of respiratory complex neurons.These are the first genome-wide significant findings reported for oxyhemoglobin saturation during sleep, a phenotype of high clinical interest. Our replicated associations with HK1 and IL18R1 suggest that variants in inflammatory pathways, such as the biologically-plausible NLRP3 inflammasome, may contribute to nocturnal hypoxemia.

Author summary

Variation in oxyhemoglobin saturation, the proportion of oxygen-saturated to total hemoglobin in the blood, is associated with numerous disorders and is a predictor of health outcomes including mortality, incident heart failure, and dementia. Despite the fundamental role of oxygen saturation in normal and abnormal physiology, there are few large-scale genetic studies of oxygen saturation performed across populations. Overnight measurements provide more variability than daytime levels due to the “stresses” associated with normal and disordered breathing, and also provide an important measure of sleep apnea severity, a common disorder in the population that is associated with considerable morbidity. In this study, for the first time, we identified multiple replicated genome-significant associations based on up to 22,736 individuals from 10 cohort studies. Our findings suggest a contribution of inflammatory genes such as the Interleukin 18 receptor subunit genes to the genetic architecture of sleep-disordered breathing. These results extend our understanding of the genetics of oxyhemoglobin saturation and sleep-disordered breathing and may provide further insight into the biology of associated diseases.

Introduction

Arterial oxyhemoglobin saturation is a fundamental physiological trait that is tightly regulated at cellular and systemic levels to optimize tissue oxygen delivery. Reduced values, or hypoxemia, occurs secondary to acute and chronic respiratory or cardiovascular diseases, and rarely due to hemoglobin protein mutations. Chronically, low oxygen saturation predicts cognitive deficits in patients with chronic obstructive pulmonary disease (COPD) and in sleep apnea (SA) [1,2].

Oxyhemoglobin saturation (SpO2), the proportion of oxygen-saturated to total hemoglobin in the blood, is most commonly measured using non-invasive equipment (oximeters). Oximetry is used to screen and monitor a wide range of health conditions. Normal SpO2 values range from 95% to 100% during wakefulness and normally fall by 2 to 4% during sleep. Oxygen saturation is reduced in individuals living at high altitude and in patients with cardiopulmonary diseases. However, even within specific disease groups, there is variation in SpO2 that is not explained by factors such as age, obesity, lung function or tobacco exposure [3]. Twin studies indicate that as much as 26% of the variation in waking SpO2 can be explained by genetic factors [4]. Population studies also indicate that genetic effects contribute to variation in waking SpO2 among Tibetan highlanders and in COPD [3,5].

Sleep disordered breathing (SDB) is a common disorder characterized by recurrent falls of SpO2 during sleep due to repetitive episodes of apneas (no airflow) or partial airflow (hypopneas), most often due to recurrent collapse of the upper airway. Our prior family-based studies have indicated that average SpO2 during sleep is significantly heritable [6]. Sleep-related hypoxemia is a key component of the pathophysiology of the disorder and variations in SpO2 during sleep in patients with SDB are predictive of incident atrial fibrillation [7], certain types of cancer [810], and death [7]. Minimum nocturnal SpO2 predicted future carotid plaque burden in the Wisconsin sleep cohort, even after adjusting for traditional risk factors [11]. In cohorts of patients with both heart failure and SDB, overnight hypoxemia is a stronger risk factor for incident cardiovascular events and death than is the apnea hypopnea index (AHI, a count of the number of breathing pauses per sleep hour) [1214]. Mean oxygen saturation and acute hypoxemia during sleep have more significant associations with liver steatosis than the AHI [15]. Sleep-related hypoxemia also may significantly influence prognosis of patients with COPD, asthma, and interstitial lung disease [1619]. Adverse effects of sleep-related hypoxemia include those directly related to tissue ischemia as well as to the effects related to activation of hypoxia-inducible factor-1 (HIF1A) mediated and NF-kB pathways, that then activate the sympathetic nervous system, stimulate release of angiogenic and inflammatory factors, cause oxidative stress, reduce insulin sensitivity, and cause endothelial dysfunction. Therefore, understanding variation in nocturnal SpO2 is important for improving our understanding of variation in risk of a wide range of chronic health outcomes.

In this study, we conducted the first multi-ethnic genome-wide association study (GWAS) of 3 nocturnal oxygen hemoglobin saturation (SpO2) traits: average and minimum SpO2, and the percent of sleep time under 90% SpO2 (Per90). (Lower values of Per90 are better while higher values are better for the other two measures.) These measures provide complementary information on hypoxemic burden across the sleep episode and can be derived from oximetry, which is potentially scalable for large-scale studies.

We analyzed data from 10 cohort studies and four ethnic groups and focused on identifying obesity-independent loci by adjusting for body mass index (BMI). We also considered sex differences in associations given the growing interest in sexual dimorphism in genetic analyses [20]. Furthermore, SDB prevalence varies by sex [21,22]), as do SDB risk factors such as the ventilatory response to arousal and regional fat distributions [23,24], and sex differences have also been reported for the relationship between chronic hypoxemia and cardiovascular events [12]. These analyses extend our prior report of results for SDB in Hispanic/Latino-Americans [25].

Results

Study sample and cross-phenotype correlations

Descriptive characteristics of the discovery and replication study samples are provided in Tables 1 and 2. Collectively, we studied 22,736 individuals. The discovery sample consisted of 8,326 individuals across 6 studies and 4 populations (1,209 African-Americans [AA]; 228 Asian-Americans [AsA]; 5,649 European-Americans [EA]; 1,240 Hispanic/Latino-Americans [HA]). Replication cohorts included 14,410 individuals (681 AAs, 2,378 EAs and European-Australians, and 11,351 HA) from 4 cohorts. Across cohorts, mean age ranged from 37.8 (CFS African-Americans) to 77.7 years (CHS European-Americans). Female participation ranged from 0% (MrOS) to 72% (Starr). The mean BMI ranged from 24.1 kg/m2 (MESA Asian-Americans) to 32.3 (JHS). Waking SpO2 values, which were typically measured prior to the sleep episode on the same equipment, ranged from 94.96% (in the older MrOS cohort) to 97.78% (among relatively young CFS African-Americans). Average SpO2 during sleep ranged from 93.74% (CFS European-Americans) to 96.45% (HCHS/SOL). The median Apnea Hypopnea Index ranged from 1.97 (HCHS/SOL, a relatively young cohort) to 24.60 (WASHS, a sleep clinic-derived cohort). Average forced vital capacity (FVC; percent predicted value) exceeded 90% in cohorts in which these data were available. The prevalence of chronic lung diseases (asthma, COPD) and diabetes varied across cohorts, likely reflecting differences in age, ascertainment and possible disease definitions.

Pairwise correlations among phenotypes and selective demographic variables are shown in S1 Table. As expected, the three overnight oxyhemoglobin saturation traits are strongly correlated (average SpO2 –minimal SpO2 ρ = 0.61; average SpO2 –Per90 ρ = -0.73; minimal SpO2 –Per90 ρ = -0.86). Waking oxyhemoglobin saturation correlates with average nocturnal SpO2 (ρ = 0.59), minimal nocturnal SpO2 (ρ = 0.35) and Per90 (ρ = -0.40). The AHI was also correlated with minimal SpO2 (ρ = -0.71), Per90 (ρ = 0.70), and average SpO2 (ρ = -0.55). Lung function (percent predicted FEV1 and FVC [26]) correlated modestly with each of the overnight oxygen saturation measures (ρ = -0.20 –+0.23).

Meta-analysis results overview

Manhattan and QQ plots for the overall sample and population-specific primary discovery analyses are provided in S1S3 Figs. The maximum lambda value was 1.02, in the multi-ethnic average SpO2 analysis, suggesting that our analysis results were largely free of technical artifacts and corrected appropriately for population structure within each ethnic group. We analyzed SNPs in loci with discovery p-values < 1 × 10−6 in our replication cohorts and identified 6 significant (p < 5.0 × 10−8) and 1 suggestive (p < 1.0 × 10−6) associations in joint discovery and replication analyses spanning 5 regions; 2q12 (IL18R1; Fig 1), 10q22 (HK1; Fig 2), 3p24 (intergenic region; S8 Fig), 4q35 (RP11-242J7.1, S9 Fig); S2 Table), with several associations specific to given population backgrounds. Effect estimates and directions of allelic effect were consistent in the discovery and replication stages for all SNPs (METAL heterogeneity p > 0.1).

thumbnail
Fig 1. Minimum oxygen saturation 2q12 regional association plot.

Physical positions (Build 37 coordinates) are shown on the X-axis. The main graphic shows log-transformed p-values for individual SNPs on the Y-axis. SNP colors indicate the degree of linkage disequilibrium (LD) with the lead SNP rs78136548 (based on combined 1000 Genomes AFR, AMR, and EUR populations). The significance cut-off of p = 5 × 10−8 is shown with a horizontal red line. The blue line denotes recombination rates. Lower tracks indicate positions of SNPs with strong LD with rs78136548, regions of Dnase hypersensitivity sites, and exon positions for local genes.

https://doi.org/10.1371/journal.pgen.1007739.g001

thumbnail
Fig 2. Average oxygen saturation 10q22 regional association plot.

This figure depicts the multi-ethnic average oxygen saturation results in the HK1 (hexokinase 1) region, which had the highest sample size among all significant HK1 region associations (n = 20,676). Other significant and suggestive HK1 regional associations are shown in S7, S11 and S17 Figs.

https://doi.org/10.1371/journal.pgen.1007739.g002

Interleukin 18 Receptor 1 and Hexokinase 1 region meta-analysis results

In the multi-ethnic combined discovery and replication meta-analysis (n>20,000), genome-wide significant associations were identified with: a) minimum SpO2 in the IL18R1 region (rs78136548 discovery, p = 2.66 × 10−7, combined p = 2.70 × 10−10); and b) average SpO2 in the HK1 region (rs72805692 discovery p = 7.20 × 10−8, combined p = 4.58 × 10−8) (Figs 1 and 2, Table 3). Lead population- and sex-specific SNPs from each locus meeting our replication criteria definitions are also presented (S2 Table). Consistent negative effect directionality was observed for the IL18R1 region SNP rs78136548 T across all 13 available association tests in African, European, and Hispanic/Latino ancestral populations (S3 Table). The association was largely driven by males (females beta(se) -0.065 (0.023), p = 0.005, males beta(se) -0.131 (0.022), p = 2.69 × 10−9; S4 Table). Average SpO2 was significantly associated with the HK1 region in a European-American analysis (rs16926246 n = 5,649; p = 2.46 × 10−8; 1000G EUR rs72805692 r2 = 0.625). The HK1 region was also notable for a second European-American significant association using the complementary phenotype Per90 (percentage of sleep with oxyhemoglobin saturation below 90%; rs148471505 p = 3.08 × 10−8; 1000G EUR rs72805692 r2 = 0.679; S2 Table). Minimum SpO2 was also suggestively associated with the HK1 region in European-American males (rs17476364 p = 6.79 × 10−8; 1000G EUR rs72805692 r2 = 0.937; S3 Table).

thumbnail
Table 3. Significant IL18R1 and HK1 region meta-analysis results.

https://doi.org/10.1371/journal.pgen.1007739.t003

Given that sleep disordered breathing and respiratory control vary by sleep state [27,28], we also explored whether associations for oxygen saturation differed when using measurements specific to non rapid eye movement (NREM) versus rapid eye movement (REM) sleep in cohorts with sleep state information (S5 Table). Several SNPs showed associations with lower p-values and/or higher point estimates for stage-specific results. The minimum SpO2 within NREM rs72805692 association result showed the lowest p-value for any HK1 locus SNP across all analyses (p = 1.60 × 10−9) and further indicated that the HK1 region SNPs were significantly associated with all three traits.

Additional analyses

The secondary sex-stratified discovery analyses (Miami plots, S4S6 Figs) identified 6 additional independent loci associated in males with consistent effects in replication and suggestive p-values < 1.0 × 10−6 in joint analyses (S3 Table; S11S17 Figs). A combined secondary meta-analysis of the discovery and replication cohorts for SNPs with initial discovery phase p-values ≥ 1.0 × 10−6 identified four additional significantly associated loci (22q11, 6q25, 17p13 and 7q22 including new candidate genes CHRNE and RELN; S6 Table and S18S22 Figs). Joint analysis results of the lead loci are provided for each SNP in S3 Table, while a comparison of individual SNP results using combined-sex and sex-stratified models can be found in S4 Table.

GWAS results overlap across oxygen saturation traits, apnea hypopnea index, and pulmonary traits

Although overnight oxygen saturation values most strongly correlate with measurements of SDB, they also may be influenced by pulmonary function, waking oxygen saturation, and hemoglobin levels. We therefore tested associations of the lead loci for sleep SpO2 traits with waking SpO2, the AHI (the clinical metric for SDB), forced vital capacity (FVC, percent predicted, a measure of pulmonary function), and hemoglobin concentration (S5 Table). Sample sizes for the comparison traits varied based on the availability of these exploratory phenotypes, with hemoglobin and FVC collected at different exams from the sleep recordings in a subset of cohorts. Consistent with the strong correlation between sleep period SpO2 and AHI phenotypes (S1 Table), several lead oxygen saturation SNPs also displayed modest to strong associations with the AHI (12 of 17 available p-values < 0.05; minimum AHI p = 5.3 × 10−5). Weaker associations were observed with the other traits: waking SpO2 (p generally > 0.01 and < 0.05; minimum p = 0.005); FVC (p generally > 0.05; minimum p = 0.025); and hemoglobin concentration (p generally > 0.05; minimum p = 0.003). We next evaluated whether associations between sleep SpO2 values and lead SNPs persisted after adjusting for AHI, FVC percent predicted, waking SpO2, asthma history, COPD history, current smoking status, and hemoglobin concentration (S8 Table). Analyses restricted to individuals with these available covariates showed that neither FVC, asthma, COPD, current smoker status or hemoglobin concentration changed the estimated SNP effects for sleep SpO2 by more than 10% for any SNP, suggesting that lung function and lung disease did not mediate the associations between sleep oxygen saturation and each SNP. In contrast, adjustment for AHI reduced the SNP association effect estimates by more than 10% in most of the models tested. Adjustment for waking SpO2, which correlated with nocturnal oxygen saturation, reduced the effect estimates by 0.2 to 35% (S8 Table), although the statistical significance of direct associations of these SNPs with waking SpO2 was modest (p = 0.003–0.93; S5 Table).

Correlated functional and eQTL SNPs and cell line enhancer enrichment: Bioinformatics data

We searched for SNPs in our top loci that overlap regulatory regions as determined by the ENCODE and Roadmap Epigenomics Consortia and collated by HaploReg. S9 Table lists the 144 of 227 unique SNPs with p-values < 1 × 10−6 that overlap regulatory regions (promoter or enhancer marks; DNase I hypersensitivity sites; or protein binding regions) in at least 1 cell line. Our lead HK1 region SNP in multi-ethnic meta-analysis, rs72805692 (average SpO2 multi-ethnic p = 4.58 × 10−8) overlapped enhancer marks in 107 cell lines across 21 organs. Other notable genome-level significant SNPs in the HK1 locus include rs16926246 and rs148471505 (overlapping enhancer marks in 91 and 71 cell lines, respectively). We further queried for Blueprint Consortium promoters and enhancers (largely blood cell lines) and Vermunt et al. brain region enhancers (S10 Table). 104 of the 227 unique replication and combined meta-analysis SNPs with p-values < 1 × 10−6 overlapped at least one regulatory region. rs72805692 additionally overlapped 30 Blueprint and 98 Vermunt enhancer regions, and rs16926246 overlapped 106 combined enhancer regions.

We also queried overlap with published expression quantitative trait loci (eQTL) associations. 182 of the 227 unique SNPs with p-values < 1 × 10−6 were eQTL SNPs for at least one of 42 genes (S11 Table). 13 SNPs that were genome-level significant SNPs in the IL18R1 region were also eQTL SNPs for both interleukin 18 receptor subunits in whole blood (IL18R1 eQTL p < 4.9 × 10−11; IL18RAP eQTL p < 1.6 × 10−46), indicating a possible role for interleukin 18 signaling in the as yet unknown causal tissue(s). No significant colocalization was observed when testing this region using Blueprint Consortium eQTL signals. The lead significant HK1 region SNP rs16926246 was also associated with HK1 expression in whole blood (EA average SpO2 p = 2.46 × 10−8; HK1 eQTL p = 9.64 × 10−13).

Gene and pathway analyses

We performed a multi-variate GWAS of the three SpO2 traits in the European-ancestry samples using MTAG [29]. Lead results (p < 1 × 10−6) are shown in S12 Table. No novel genome-level significant loci were detected.

We used our European-American GWAS meta-analysis results to impute gene-level expression differences in a subset of 6 GTEx-assayed tissues and Depression and Genes and Networks (DGN) whole blood using MetaXcan. Tissue-specific results are presented in S13S19 Tables. Three genes were associated at either a Bonferroni-adjusted significance level (p < 4.01 × 10−7) or at a suggestive level within an order of magnitude, all in the minimum SpO2 analysis: CHRNE (minimum p = 7.61 × 10−8 in subcutaneous adipose tissue), C17orf107 (overlapping and antisense to CHRNE; minimum p = 2.68 × 10−7 in visceral omentum adipose tissue), and IL18R1 (minimum p = 6.28 × 10−7 in subcutaneous adipose tissue).

We carried the whole blood MetaXcan results forward for pathway analyses (DGN sample size = 922). GIGSEA analyses of KEGG pathways and Molecular Signatures Database curated microRNAs and transcription factors are presented in S20S22 Tables respectively. The most enriched KEGG pathway was steroid hormone biosynthesis (Average SpO2 empirical p-value = “0” following 10,000 permutations). This pathway was observed twice with empirical p-values < 0.05, as were the KEGG asthma and ribosome pathways. The most significantly observed miRNA binding site was for MIR-380-3P (average SpO2 empirical p-value = 0.006). MIR-140 and MIR-190 displayed empirical p-values < 0.05 in two analyses. PPARG transcription factor binding sites were enriched in all three analyses, while PPAR signaling was the most enriched Per90 KEGG pathway. NHLH1 (formerly HEN1) transcription factor binding sites were enriched in all three analyses, and six transcription factor binding sites were enriched in two analyses.

Discussion

Novel associations were identified for several genetic loci with traits measuring oxyhemoglobin saturation during sleep in a large, multi-ethnic population. The traits examined quantify overnight hypoxemia, a key component of sleep disordered breathing that predicts risk of developing cognitive impairment, cardiovascular disease, atrial fibrillation, and mortality in community and clinical cohorts [2,7,1214]. Although nocturnal hemoglobin oxygen saturation level inversely correlates with the number of breathing pauses at night (apneas, hypopneas), there is much individual variation in sleep-associated hypoxemia that is not well understood. For the first time, this study identified genetic variants associated with oxyhemoglobin saturation traits measured during sleep. Specifically, we identified and replicated variants in two gene regions- hexokinase 1 (HK1) and interleukin 18 receptor (IL18R1)- that individually and together are of potential high relevance to lung and ventilatory-control pathobiology. In addition, we identified several other loci of potential biological significance.

HK1 and IL18R1 regional associations

We identified significant associations between HK1 SNPs and average oxygen saturation (SpO2) during sleep and percentage of sleep with SpO2 < 90% (Table 3). State-specific results also indicate a genome-level association with minimum SpO2 during NREM sleep (S5 Table). Hexokinase is the first enzyme and the rate-limiting step in the glycolysis pathway [30,31] and its activity is regulated by hypoxia inducible factor 1a (HIF1A) [5,32,33]. Obstructive sleep apnea following CPAP withdrawal increases glucose during sleep [34]. Rs16926246 (significantly associated with average SpO2) and rs10159477 (among our suggestive average SpO2 SNPs, S3 Table) are associated with HK1 expression in whole blood ([35], S11 Table) and also have been associated with hemoglobin concentration [36]. Rs16926246 was the study-wide lead SNP and previously found to be highly significantly associated with hemoglobin A1c (HbA1c) levels, a marker of glucose homeostasis, through an erythrocytic pathway [37]. Rs72805692, our lead multi-ethnic average SpO2 SNP, has also been associated with HbA1c levels [38]. Both SNPs overlap enhancer marks in ≥ 197 ENCODE, Roadmap Epigenomics, Blueprint, and Vermunt et al. cell lines and/or brain regions (including erythroblasts; S9 and S10 Tables). The Rapoport–Luebering glycolytic shunt affects erythrocytic oxygen capacity through allosteric binding of 2,3-bisphosphoglycerate (2,3-BPG, also known as 2,3-diphosphoglycerate or 2,3-DPG) to hemoglobin. The concentration of glycolytic pathway intermediates can impact 2,3-BPG concentration, partially mediated by hexokinase [3941]. Rs72805692 was marginally associated with hemoglobin concentration in our sample for individuals with available measurements (p = 0.0034). The A allele was associated with reduced hemoglobin concentration, average sleeping and waking SpO2 and increased sleep time with oxygen saturation under 90%. However, the association with average sleeping SpO2 was not appreciably changed when adjusting for hemoglobin concentration (S5 Table).

Alternatively, HK1, in concert with cytokines including IL18, may influence overnight oxygen saturation through effects on pulmonary inflammation and ventilation-perfusion mismatch. Hexokinase-1 mediated glycolysis regulates the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome [42], a multiprotein complex implicated in obesity-related inflammation [43] as well as several pulmonary diseases [4450]. The NLRP3 inflammasome activates caspase-1, resulting in cleavage of pro-IL1B and pro-IL18 into their mature forms, amplifying inflammation [50,51]. The NLRP3 inflammasome is proposed to play a critical role in lung injury occurring in response to exposures to inflammatory mediators, oxidative stress and mechanical ventilation, including cyclic pulmonary stretching, which induces NLRP3 inflammasome activation in alveolar macrophages [52]. Patients with SDB, particularly obstructive sleep apnea, experience oxidative stress and pulmonary inflammation [53], as well as swings in intrathoracic pressure, potentially causing pulmonary strain. Our data suggest the possibility that variations in HK1 (and possibly IL18) pathways may contribute to individual differences in pulmonary gas exchange occurring during sleep, possibly through pulmonary inflammation and/or subclinical pulmonary injury. Circulating markers of alveolar epithelial injury, including KL-6, surfactant protein-A, and matrix metalloproteinase-7, correlate with degree of overnight hypoxemia and AHI in patients with SDB [54,55]. Chronic intermittent hypoxia induces physiological deficits in rats with allergen-induced airway inflammation, due to collagen deposition and other effects [56]. Although the mechanisms underlying these associations are unclear, lung imaging studies show an increase in subclinical interstitial lung abnormalities in individuals with sleep apnea [55]. Our data suggest the possibility that variations in HK1 pathways may contribute to differences in pulmonary gas exchange that occurs during sleep, possibly through effects on ventilation-perfusion mismatch due to subclinical pulmonary inflammation. Recent population-based studies found that sleep apnea associates with both elevated pulmonary inflammatory markers and imaging evidence of interstitial lung abnormalities [55]. Finally, the NLRP3 inflammasome has been shown to influence brain tissue, and changes in sleep delta power have been observed in knock-out mice [57].

Another and correlated mechanism could be through HIF1A. In addition to oxygen sensing effects in the carotid body [58], HIF1A regulates HK1 in human alveolar cells [59], is regulated by PFKM (a downstream glycolysis enzyme) in macrophages [60], and is involved with metabolic reprogramming of macrophages [61]. Activation of glycolytic enzymes in pulmonary epithelial cells exposed to cyclic mechanical stretching is abrogated with HIF1A repression [62]. Ventilatory differences in responses to intermittent hypoxemia secondary to SDB could influence several measures of overnight SpO2, as observed in our analyses.

The second set of significant SNPs implicated in overnight SpO2 levels were in the IL18R1 region, with external evidence indicating eQTL associations with both IL18 receptor subunit genes (IL18R, IL18RAP) in over a dozen genome-level significant SNPs (S8 Table). Minor alleles were associated with an increase in minimum oxygen saturation, an increase in IL18RAP expression, and a decrease in IL18R1 expression in whole blood [35]. IL18R1 expression was also suggestively associated with minimum oxygen saturation in a gene-level analysis using MetaXcan. These genes are essential for IL18 signaling [63,64], suggesting that the IL18 pathway may partially mediate the association at this locus. IL18 is a pro-inflammatory cytokine produced by macrophages and is involved in multiple inflammatory disorders [65]. This region has been associated with Blautia genus microbiota abundance in the gut (lead locus SNP rs79387448 Min SpO2 p = 8.61 x 10–8 [S3 Table]) [66]. Il18 over-expression in mice leads to chronic pulmonary inflammation, including the increased levels of CD4+, CD8+ CD19+, eosinophils, macrophages, NK1.1+, and neutrophils; along with alveolar destruction, fibrosis, and other effects [67,68]. In humans, IL18 plasma concentration levels are elevated in acute respiratory distress syndrome [69]. As described above, IL18 as well as IL1B are the inflammatory proteins activated by caspase 1 in HK1-regulated NLRP3 inflammasome activation [42]. MTOR, a member of the complex that induces this HK1-mediated activation, is the 6th most associated gene in our analyses of Per90 using MetaXcan whole blood analysis (p = 4.5 × 10−4, S18 Table). Mechanical stretching-induced NLRP3 inflammasome activation induces activated IL18 release from alveolar macrophages [52]. Casp1- (required for Il18 activation) and Nlrp3-knockout mice are protected from hypoxemia accompanying mechanical ventilation [70]. Models of cyclic stretching induce the release of Il18 in mouse alveolar macrophages, mediated by Nlrp3 [52]. Serum IL18 concentrations are also significantly higher in patients with SDB compared to obese controls and correlated with serum concentrations of C reactive protein and interleukin 6 [71]. Il18r1 is among the genes with the most robust circadian rhythmic profiles in healthy mouse lung [72]; it is possible that timing-specific gene expression may influence the nocturnal hypoxemia phenotype we studied. An association between asthma and a SNP in the IL18R1 region is reported [73]. Our lead SNPs had reduced linkage disequilibrium with this SNP (rs3771166 minimum p in any model = 5.6 x 10−5 for minimum SpO2 in EA males). Analyses adjusted for asthma did not significantly attenuate our associations (rs78136548 minimum SpO2 p adjusted for asthma = 1.21 x 10−7, unadjusted p = 1.01 x 10−7 in equivalent samples). Therefore, different variants in the IL18R1 region may influence pulmonary and sleep related hypoxemia traits. The circadian timing of sleep may impact IL18 pathway-specific effects on pulmonary-related traits. The associations with HK1 further also implicate the possibility that variants in both genes may contribute to SpO2 levels. IL18 has also been shown to regulate HIF1A [74].

Other regional associations

The protein coding gene most proximal to the Per90 association with the 4q35 region in African-Americans (S2 Table, S9 Fig) was CASP3, a second caspase gene involved with alveolar wall destruction [75]. Despite the modest sample size, the p-value almost met genome-wide significance (p = 9.39 x 10−7), suggesting the utility of future studies of the role of this gene in influencing nocturnal saturation.

We also detected multiple genome-level significant associations following a combined discovery plus replication cohort meta-analysis (S7 Table). Although these associations require independent evidence for replication, two of these associations are of particular interest. In European-Americans, minimum oxygen saturation was associated with 2 genome-level significant and 65 suggestive SNPs that are associated with CHRNE (acetylcholine receptor, nicotinic epsilon [muscle]) expression in 19 GTEx tissues and monocytes (p < 5 x 10−8; S11 Table). CHRNE and the proximal gene C17orf107 had the lowest p-values in the expression-based MetaXcan gene tests. Phospholipidase D2 (PLD2), another gene associated with the locus through expression (eQTL) SNPs, is required for hypoxia-induced expression of HIF1A. Hypoxia-induced gene expression in mouse lung endothelial cells is reduced in Pld2 knockout mice [76]. A multi-ethnic Per90 association physically overlaps RELN (reelin). While no expression or epigenetic evidence was available, this association is of interest given the suggested respiratory role of reelin within the Pre-Bötzinger complex, a major center for respiratory control [77].

No novel genome-level significant loci were detected in our multi-variate MTAG analyses (S12 Table). As expected from the univariate results, HK1 was significantly associated and the lead SNP rs72805692 had reduced p-values for average and minimum SpO2 (p = 4.0 × 10−9 and 1.1 × 10−9 respectively). Among the genes in novel suggestive regions was WLS (formerly GPR177; rs17481104 minimum SpO2 p = 5.8 × 10−7), which is involved in pulmonary vascular development and has been suggestively associated with airflow obstruction in COPD [78,79]. Enrichment of the KEGG asthma pathway for both minimum SpO2 and Per90 (S20 Table) lends support to the ‘overlap syndrome’ of these two pulmonary diseases [80]. PPAR-gamma transcription factor binding site enrichment in all three analyses (S22 Table) suggests the potential importance of future mechanistic studies of this inflammation-related transcription factor. PPAR signaling was the most enriched KEGG pathway in the Per90 analysis (S20 Table). PPAR signaling and PPARG expression in visceral adipose tissue have previously been associated with obstructive sleep apnea [81].

Sex- and state-specific effects

Sex-stratified analyses identified stronger associations among males compared to females for the IL18R1 signal (rs78136548 p females = 0.005, males = 2.69 × 10−9; S4 Table) and for several SNPs within the HK1 locus (e.g. rs17476364 EA min SpO2 p females = 0.19, males = 6.79 × 10−8). In addition, sex-stratified analyses identified two other loci of interest (S6 Table). Among EA males only, a suggestive association with Per90 in the IL1RAPL1 region of the X chromosome was identified. The region has recently been suggestively associated with asthma in Hispanic/Latino children, with one replication cohort indicating potential male-specific effects [82]. An association in the PPP4R1 region (protein phosphatase 4 regulatory subunit 1) was nearly genome-level significant in EA males (p = 5.4 × 10−8). Ten suggestive SNPs in this locus were also PPP4R1 eQTL SNPs in whole blood (p < 1 × 10−40; S11 Table). PPP4R1 regulates HDAC3 (histone deacetylase 3), an epigenetic modulator of circadian lipid metabolism [83,84].

Respiratory control and neuromuscular activation vary between NREM and REM sleep. Analyses restricting to these states may reduce heterogeneity due to differences in state that influence airway patency or respiratory chemosensitivity. The PPP4R1 locus p-value lowered to genome-level significance when analyzed using NREM sleep data (rs78805840 p = 1.81 × 10−8; S5 Table). The lowest overall p-value in the HK1 locus was a minimum SpO2 within NREM association with rs27805692 (all population combined-sex p = 1.60 × 10−9).

Pulmonary trait effects

The three traits that we analyzed (average and minimum SpO2 during sleep, percent of sleep with SpO2 < 90%) each are commonly measured and reported in evaluation of patients with sleep apnea. Although correlated, they each measure somewhat different aspects of oxygen saturation. Notably, associations for the Hexokinase 1 (HK1) region showed associations with several measures of oxygen saturation. Consistency of these findings across phenotypes suggests the importance of the HK1 pathway in influencing several aspects of oxygenation during sleep, including severity of response to an airway occlusion (i.e., as measured by minimal saturation), overall severity (Per90), and overall level (average). Although overnight hypoxemia can occur with underlying pulmonary disease, our analyses also showed that each SpO2 association typically associated with the AHI, a primary index of sleep apnea, and associations were not appreciably influenced after considering effects of lung disease, tobacco use, lung function, and hemoglobin level (S8 Table). As expected, baseline oxygen saturation also correlated with average SpO2, consistent with an influence of waking SpO2 on overall nocturnal levels. Notably, none of our sleep-related trait associations overlapped published associations for resting oxygen saturation in COPD.

Across our cohorts, average level of lung function was within the normal range, and prevalence of lung diseases was low. These findings, as well as our analyses that adjusted for several factors and independently assessed genetic signals for the other pulmonary traits, indicate that the variations in SpO2 traits in our cohorts predominantly reflected differences in SDB-related levels of hypoxemia. Relevance of our results to SDB is supported by finding that 12 of the 17 available lead loci SNPs across all analyses had at least a nominal association with the AHI (S5 Table), including the associations for HK1 rs72805692 and IL18R1 region rs78136568 SNPs (p = 8.1 × 10−5 and 7.2 × 10−5 respectively). SDB is a common disorder affecting 17% of middle-aged men and 9% of middle-aged women and characterized by repetitive episodes of upper airway obstruction resulting in intermittent hypoxemia, sleep disruption, and profound physiological disturbances [85]. Past genetic analyses of sleep apnea have mainly focused on the AHI, which does not fully describe the broad range of physiologic stressors that occur in SA, including patterns of SpO2 desaturations [86]. Overnight SpO2 analysis provides clinically relevant information [2,79,1214] and can be measured relatively simply, and thus can be scaled for future genetic studies and precision medicine.

Strengths and weaknesses

Our study has several strengths. The sample size of over 22,000 is among the largest available GWAS analyses of any trait associated with objectively recorded sleep disordered breathing. The associations were observed in cohorts with varying demographics and ascertainment strategies (Tables 1 and 2), and as such are likely generalizable to diverse populations. We used a stringent imputation quality threshold (0.88) to reduce random error, using a 1000 Genomes Project or denser template in all studies. Several of our associations are supported by published gene expression, bioinformatics evidence, and/or physiological studies.

Several weaknesses also need to be acknowledged. While we have not performed functional assays as part of the present analysis, the most biologically compelling candidates are supported by several lines of evidence from the literature and will require future experimental validation. Data on potential mediators (e.g., lung function, hemoglobin) were collected in different visits or were available only in a subset of our cohorts. Some promising findings did not meet genome-wide significance criteria or could not be replicated across cohorts. While this first multi-ethnic meta-analysis of the three traits included over 22,000 individuals, weak or population-specific associations were likely to be missed due to power limitations.

In conclusion, we have performed the first genome-wide association analysis of clinically relevant sleep disordered breathing traits, specifically measures of nocturnal oxygen saturation, and identified several novel associations that are of potential biological relevance. Of particular interest were variants in the HK1 and IL18R1 regions. Understanding the genetic underpinnings of these sleep-related traits may guide future studies investigating the contribution of sleep disordered breathing to the hypoxemic burden of pulmonary disorders, and identify common mechanisms such as activation of the NLRP3-inflammasome pathway.

Full meta-analysis results are freely available from http://www.sleepdisordergenetics.org/informational/data.

Material and methods

This research was approved by the Partners Healthcare IRB committee (protocol # 2010P001765). Participant consent was obtained through written documents.

Discovery group studies

The Atherosclerosis Risk in Communities Study (ARIC; n = 1,432) and Framingham Heart Study (FHS; n = 640) cohorts participating in the Sleep Heart Health Study (SHHS) were analyzed with available polysomnography (PSG) and genotype data [8789]. This community-based study included a baseline examination (1995–1998) that included in-home polysomnography, and questionnaires [89]. Polysomnography from the baseline examination was collected using the Compumedics PS-2 system (Abbotsford, AU) [90]. Oxyhemoglobin saturation was measured with finger pulse oximetry over the sleep episode and cleaned of signal artifact. The 3 parent cohorts are described below, with CHS used as a replication cohort in the current study (genetic data from this cohort was obtained after the other two studies). In ARIC, genotyping was performed using the Affymetrix 6.0 array. In FHS, genotyping was performed using the Affymetrix 500k and Illumina Omni 5M arrays (obtained from dbGaP; pht000395.v7.p8).

The Cleveland Family Study (CFS) is examining the genetic and familial basis of sleep apnea with 2,534 African- and European-American individuals from 356 families. Four visits occurred from 1990–2006, with a final visit at a clinical research center (2000–2006). Index probands with confirmed sleep apnea were recruited from sleep centers in northern Ohio. Additional family members and neighborhood control families were also studied [91]. Measurements including sleep apnea monitoring, anthropometry, other related phenotypes, and questionnaires. Before 2000, an Edentrace Type 3 home sleep apnea device was used (Eden Prairie, MN). The final examination used 14-channel polysomnography (Compumedics E series, Abottsford, AU). Genotyping was based on the Affymetrix 6.0 and Illumina OmniExpress, Exome, and IBC chip arrays. Data were based on 1,411 individuals with both genotypes and sleep data from either the home sleep study (n = 784) or the lab-based study (n = 627).

The Multi-Ethnic Study of Atherosclerosis (MESA) is examining the risk factors of clinical cardiovascular disease [92]. The baseline examination in 2000 included 6,814 participants ages 45 to 84 from 6 communities: Baltimore MD, Chicago IL, Los Angeles CA, New York NY, Minneapolis/St. Paul MN, and Winston-Salem NC. Four ethnicities are being studied: African-, Asian-, European-, and Hispanic/Latino-Americans. An ancillary sleep study of 2,060 individuals who did not use nightly CPAP, overnight oxygen, or an oral device for sleep apnea occurred between 2010–2013. Sleep measurements included in-home PSG, actigraphy, and a questionnaire adapted from the SHHS questionnaire [93]. Unattended polysomnography used a 15-channel monitor (Compumedics Somte System, Abbotsford, AU). Final study inclusion for individuals with an Affymetrix 6.0 assay was 1,883.

The Osteoporotic Fractures in Men Study (MrOS) is a prospective cohort study examining the risk factors for fractures, osteoporosis, and prostate cancer [94,95] in males age 65 or older from six U.S. communities. An ancillary sleep study of 3,135 individuals was conducted between 2003 and 2005, including in-home PSG (Compumedics Safiro system; Abbotsford AU), anthropometry, and questionnaires. Genotyping was performed with the Illumina Human Omni 1 Quad v1-0 H array. A total of 2,178 European ancestry individuals had PSG and genotype data.

The Starr County Health Studies (Starr) have been examining the risk factors of diabetes in a predominantly Mexican-American border county in Texas since 1981 [96,97]. The sleep apnea assessment occurred between 2010 and 2014 and included a questionnaire and home sleep apnea testing using the WatchPAT-200 device (Itamar-Medical Ltd., Caesarea, Israel), with recording of finger pulse oximetry, actigraphy, body position, peripheral arterial tonometry, and snoring. It has previously been validated using polysomnography [98]. The current analysis included 782 individuals with valid oximetry and Affymetrix 6.0 data.

Replication group studies

Data from the Sleep Heart Health Study Cardiovascular Health Study (CHS) [99] was available after ARIC and FHS and used in replication analysis. Sleep phenotyping was performed as described earlier. 185 African-American and 731 European-Americans with available polysomnography and Illumina CNV370, and/or Omni1M plus IBC genotypes obtained through dbGaP (pht003699.v1.p1) were analyzed.

The Hispanic Community Health Study/Study of Latinos (HCHS/SOL) is studying risk and protective factors of multiple health conditions in Hispanics/Latinos [100,101]. 16,415 community members from randomly selected households aged 18–74 from 4 cities (Chicago, IL; Miami, FL; Bronx, NY; San Diego, CA) were examined in a baseline exam between 2008–2011. The sample design consisted of a stratified two-stage area probability sample of household addresses. Six cohort backgrounds were represented: Central American (n = 1,730), Cuban (n = 2,348), Dominican (n = 1,460), Mexican (n = 6,471), Puerto-Rican (n = 2,728), and South American (n = 1,068). The exam included anthropometry, questionnaires, and home sleep apnea testing using the ARES Unicorder 5.2 (B-Alert, Carlsbad, CA), which records measurements of airflow using a nasal pressure cannula and pressure transducer; oxyhemoglobin saturation and pulse rate using a forehead-based reflectance oximeter; head movements and position using an accelerometer; and snoring levels using a microphone. The device has undergone previous validation for in-home use [102]. Records were manually scored and cleaned of artifacts at a central sleep reading center [101]. The current study includes 11,351 non-Asian ancestry individuals with oxyhemoglobin saturation values during sleep and Illumina Omni 2.5 genotyping.

The Jackson Heart Study (JHS) is a population-based prospective investigation of cardiovascular disease [103,104]. The ancillary sleep study occurred from 2012–2016, and included home sleep apnea testing with the Embla Embletta Gold, a 6-channel device that includes an oximeter (Broomfield, CO). The device has been validated previously [105]. Additional collected measures include sleep questionnaires and anthropometry. 496 African-American individuals with phenotyping and Affymetrix 6.0 genotyping were included in this study, reflecting a dataset freeze at the time of analysis.

The Western Australian Sleep Health Study (WASHS) is examining the epidemiology and genetics of sleep apnea and related comorbidities [106]. This clinic-based study examines patients presenting to the sole public sleep clinic in Western Australia, located in Perth. 91% of patients were referred for SDB. Data collection for individuals in the current analysis occurred from 2006–2010. In-lab, attended polysomnography was performed using the Compumedics Series E device (Abbotsford, AU). After excluding principal component (PC) outliers (see below), valid oximetry data and genotype data (Illumina Omni 2.5) were available for 1,647 European ancestry patients.

Phenotype and covariate definitions

The quantitative phenotypic outcome was oxyhemoglobin saturation during sleep (SpO2), measured as an average, minimum, or as a percentage of the night with SpO2 < 90% (Per90) measured using finger pulse oximetry (all using NONIN oximetry boards) or transcutaneous oximetry (HCHS/SOL only) measured continuously as part of polysomnography or home sleep apnea testing. Other than the WASHS clinical cohort, all sleep data were scored by a central reading center with high levels of established reliability [107] by scorers blinded to all other data. Intermittent waking and SpO2 artifact were manually edited from all records. Covariates were obtained by questionnaires, direct measurement (BMI), and oximetry (waking oxygen saturation was measured prior to the sleep recording). Secondary measures such as hemoglobin concentration and spirometry were collected from the same visit whenever possible, however this was not possible for all cohorts (most notably hemoglobin was collected years prior to the sleep exam in some cohorts). Potential device differences were minimized by both performing analyses at a cohort level and using a rank-normal phenotype transformation to reduce the impact of phenotypic outliers. Our analysis focused on identifying potential loci operating in obesity-independent pathways. Hispanic/Latino-specific results have been reported previously for average SpO2 [25].

Genotyping and statistical analyses

Genotypes from all cohorts were imputed to at least a 1000 Genomes Phase 1 density. ARIC, JHS, and HCHS/SOL were imputed using a 1000 Genomes Phase 1 version 3 template. WASHS was imputed using a Haplotype Reference Consortium version 1.0 template [108]. All other cohorts were imputed using a 1000 Genomes Phase 3 version 5 template [109,110]. Single nucleotide polymorphisms (SNPs) and insertions/deletions with minor allele frequency < 0.01, minor allele counts < 20 within a cohort, or an IMPUTE2/PBWT info score < 0.88 were removed from the analysis. Sample sizes and variant counts for each cohort for the three primary phenotype analyses are provided in S23 Table.

We explored ancestry-specific associations given past SDB trait differences (e.g. [93,101]) and linkage disequilibrium differences [109]. Data were analyzed at a cohort- and population-specific level (e.g. 4 separate analyses for the MESA cohort). Population structure was controlled for using linear mixed models followed by genomic control. Population structure principal components were calculated for the minimally-admixed, self-reported Asian-American and European-American/Australian population groups within individual studies using TRACE [111]. WASHS initial self-reported European ancestry was based on classification of the patient's parents [106]. Individuals were defined as population outliers and removed from analysis if any coordinate from PC 1–4 was greater than 5 standard deviations from the population mean. Individuals self-reporting into groups with modest sample size within a cohort (e.g. MrOS Asian-Americans) were excluded from study.

Our analysis focused on identifying potential loci operating in obesity-independent pathways. We adjusted for age, age2, sex, age × sex, BMI, and BMI2 to address known demographic factors and potential non-linear effects of age and BMI. Phenotypes, adjusted for age and sex, were rank-normalized. Residuals were then calculated by further adjusting for BMI. Primary analyses were performed using GEMMA, which incorporates linear mixed models that control for the relatedness structure [112]. HCHS/SOL analyses were performed using the GENESIS Bioconductor package [113] (DOI:10.18129/B9.bioc.GENESIS). A fixed effects, inverse variance weighted meta-analysis was performed using METAL with genomic control applied in each analysis [114]. Variants with p-values < 1 × 10−6 in the discovery cohort meta-analysis were carried forward to replication and combined discovery/replication analysis. To reduce the influence of small studies possibly leading to spurious findings, we only present meta-analysis results where 1,000 or more individuals contributed. Individual SNPs in the multi-ethnic analyses were only analyzed if they remained unfiltered in two or more populations. The three traits differed in their final variant counts due to phenotype missingness and ascertainment (no average SpO2 was available for WASHS in this analysis). In aggregate for the discovery cohorts, there were 11,297,250–11,298,080 AA; 8,956,016–8,958,150 EA; and 9,481,040–9,481,751 multi-ethnic variants. The replication cohorts included 12,243,361–12,346,361 AA; 7,448,148–8,707,439 EA; and 10,173,881–10,480,925 multi-ethnic variants. Visualizations were constructed using LocusExplorer and EasyStrata [115,116].

Sleep and sleep disordered breathing may be regulated by multiple tissues [86,117]. Epigenetic database queries were performed using HaploReg version 4 using an imputed model and exact SNPs. HaploReg data included ENCODE and Roadmap Epigenomics consortia data [118120]. Additional queries examined non-cancerous Blueprint Consortium data (largely related to blood cell lines) and Vermunt et al. brain region enhancer data[121123]. Gene expression data used in expression quantitative trait loci (eQTL) lookups were obtained from multiple studies, including a seven-cohort consortium investigating whole blood (Westra et al.) [35,124130]. The Westra data were pruned to include only eQTL SNPs with FDR < 0.05. Moloc was used to test colocalization [131]. Gene-level analyses used MetaXcan to impute expression levels based on GTEx tissues and Depression and Genes and Networks (DGN) whole blood [132]. GIGSEA, which is designed to work with MetaXcan output, was used for pathway analyses using the whole blood results (queried due to improved power from increased sample size) [133]. We used the weighted linear regression model with 10,000 permutations.

Supporting information

S1 Fig. Average oxygen saturation discovery cohorts manhattan and QQ plots.

Top: African-Americans; Middle: European-Americans; Bottom: Multi-ethnic. Variants in the multi-ethnic results had to have results from cohorts in two or more populations.

https://doi.org/10.1371/journal.pgen.1007739.s001

(PDF)

S2 Fig. Minimum oxygen saturation discovery cohorts manhattan and QQ plots.

Top: African-Americans; Middle: European-Americans; Bottom: Multi-ethnic (2 or more populations for each variant).

https://doi.org/10.1371/journal.pgen.1007739.s002

(PDF)

S3 Fig. Per90 discovery cohorts manhattan and QQ plots.

Top: African-Americans; Middle: European-Americans; Bottom: Multi-ethnic (2 or more populations for each variant).

https://doi.org/10.1371/journal.pgen.1007739.s003

(PDF)

S4 Fig. Multi-ethnic discovery cohorts average oxygen saturation miami plots.

Top: Females; Bottom: Males.

https://doi.org/10.1371/journal.pgen.1007739.s004

(PDF)

S5 Fig. Multi-ethnic discovery cohorts minimum oxygen saturation miami plots.

Top: Females; Bottom: Males.

https://doi.org/10.1371/journal.pgen.1007739.s005

(PDF)

S6 Fig. Multi-ethnic discovery cohorts Per90 saturation miami plots.

Top: Females; Bottom: Males.

https://doi.org/10.1371/journal.pgen.1007739.s006

(PDF)

S7 Fig. Average oxygen saturation regional plot (European ancestry, 10q22).

The supplemental regional plot order corresponds to the order found in S2, S3 and S4 Tables, minus the associations shown in Figs 1 and 2.

https://doi.org/10.1371/journal.pgen.1007739.s007

(PDF)

S8 Fig. Per90 regional plot (African-American ancestry, 3p24).

https://doi.org/10.1371/journal.pgen.1007739.s008

(PDF)

S9 Fig. Per90 regional plot (African-American ancestry, 4p35).

https://doi.org/10.1371/journal.pgen.1007739.s009

(PDF)

S10 Fig. Per90 regional plot (European ancestry, 10q22).

https://doi.org/10.1371/journal.pgen.1007739.s010

(PDF)

S11 Fig. Average oxygen saturation regional plot (European ancestry males, 10q22).

https://doi.org/10.1371/journal.pgen.1007739.s011

(PDF)

S12 Fig. Average oxygen saturation regional plot (European ancestry males, 12q14).

https://doi.org/10.1371/journal.pgen.1007739.s012

(PDF)

S13 Fig. Average oxygen saturation regional plot (European ancestry males, 2p21).

https://doi.org/10.1371/journal.pgen.1007739.s013

(PDF)

S14 Fig. Average oxygen saturation regional plot (European ancestry males, 5p15).

https://doi.org/10.1371/journal.pgen.1007739.s014

(PDF)

S15 Fig. Average oxygen saturation regional plot (European ancestry males, 5q21).

https://doi.org/10.1371/journal.pgen.1007739.s015

(PDF)

S16 Fig. Minimum oxygen saturation regional plot (European ancestry males, 10q22).

https://doi.org/10.1371/journal.pgen.1007739.s016

(PDF)

S17 Fig. Minimum oxygen saturation regional plot (European ancestry males, 23p21).

https://doi.org/10.1371/journal.pgen.1007739.s017

(PDF)

S18 Fig. Average oxygen saturation regional plot (Combined discovery/replication African-American ancestry, 22q11).

https://doi.org/10.1371/journal.pgen.1007739.s018

(PDF)

S19 Fig. Minimum oxygen saturation regional plot (Combined discovery/replication African-American ancestry, 6q25).

https://doi.org/10.1371/journal.pgen.1007739.s019

(PDF)

S20 Fig. Minimum oxygen saturation regional plot (Combined discovery/replication European ancestry, 17p13).

https://doi.org/10.1371/journal.pgen.1007739.s020

(PDF)

S21 Fig. Minimum oxygen saturation regional plot (Combined discovery/replication European ancestry, 2p12).

https://doi.org/10.1371/journal.pgen.1007739.s021

(PDF)

S22 Fig. Per90 regional plot (Combined discovery/replication, 7q22).

https://doi.org/10.1371/journal.pgen.1007739.s022

(PDF)

S1 Table. Phenotype and covariate correlations.

Spearman correlations between phenotypes and covariates across all discovery cohorts and ethnic groups are shown. Correlations with the spirometry measures FEV1 and FVC are also shown. Correlations were pooled using Fisher Z-transformations weighted by sample size. 95% confidence intervals are listed in parentheses.

https://doi.org/10.1371/journal.pgen.1007739.s023

(XLS)

S2 Table. Significant and suggestive meta-analysis results.

Lead SNPs are shown for regions with significant (p < 5.0 × 10–8) and suggestive (p < 1.0 × 10–6) p-values. Discovery regions were carried forward if the combined discovery and replication analysis results indicated a lower p-value, retained genome-level significance, or replication was unavailable (i.e. due to frequency or imputation filters). SNPs denotes the count of regional SNPs with p < 1.0 × 10–6. Genes indicates overlapping Ensembl genes within 5 kb of the p < 1 × 10–6 SNPs. CAF indicates coded allele frequency range. Individual regional SNP results are provided in S3 Table. Individual regions were minimally 500 kb apart.

https://doi.org/10.1371/journal.pgen.1007739.s024

(XLS)

S3 Table. All top locus SNP results (p < 1 x 10–6; full sleep combined-sex and sex-stratified analyses).

“Locus Class” indicates “Discovery/Replication” for main analyses or “Combined meta-analysis” for regions not clearing initial discovery p-value thresholds (i.e. S7 Table loci).

https://doi.org/10.1371/journal.pgen.1007739.s025

(XLS)

S4 Table. Sex-stratified comparison of top locus SNPs (p < 1 x 10–6).

Sex-stratified samples with n < 1000 are included here for comparison but were not included in the main analyses. An individual variant present in the combined-sex analysis for a given cohort may be missing from the equivalent sex-stratified analysis due to analysis-specific minor allele count thresholds of 20. “Locus Class” indicates “Discovery/Replication” for main analyses or “Combined meta-analysis” for regions not clearing initial discovery p-value thresholds (i.e. S7 Table loci).

https://doi.org/10.1371/journal.pgen.1007739.s026

(XLS)

S5 Table. Lead loci results for other phenotypes.

Each lead SNP from the “Original Phenotype” and “Original Model” columns was analyzed using equivalent individuals and models for different phenotypes. Available sample size will vary depending on the availability of the phenotype. AHI values are taken from Chen, Cade, et al. (submitted) “Locus Class” indicates “Discovery/Replication” for main analyses or “Combined meta-analysis” for regions not clearing initial discovery p-value thresholds (i.e. S7 Table loci).

https://doi.org/10.1371/journal.pgen.1007739.s027

(XLS)

S6 Table. Suggestive sex-stratified meta-analysis results.

Lead SNPs are shown for regions with suggestive (p < 1.0 × 10–6) p-values. Discovery regions were carried forward if the combined discovery and replication analysis results indicated a lower p-value, retained genome-level significance, or replication was unavailable (i.e. due to frequency or imputation filters). SNPs denotes the count of regional SNPs with p < 1.0 × 10–6. Genes indicates overlapping Ensembl genes within 5 kb of the p < 1 × 10–6 SNPs. CAF indicates coded allele frequency range. Individual regional SNP results are provided in S3 Table. Individual regions were minimally 500 kb apart.

https://doi.org/10.1371/journal.pgen.1007739.s028

(XLS)

S7 Table. Significant combined discovery/replication cohort meta-analysis results.

Lead SNPs are shown for regions with significant (p < 5.0 x 10–8) combined-analysis p-values with p > 1 x 10–6 in the discovery phase. SNPs denotes the count of regional SNPs with p < 1.0 x 10–6. Genes indicates overlapping Ensembl genes within 5 kb of the p < 1 x 10–6 SNPs. CAF indicates coded allele frequency range. Individual regional SNP results are provided in S3 Table. Individual regions were minimally 500 kb apart.

https://doi.org/10.1371/journal.pgen.1007739.s029

(XLS)

S8 Table. Lead loci results adjusted for additional covariates.

Lead variants were re-analyzed with standard models and additional covariates as listed. As these covariates were not collected in all cohorts and individuals, the analysis were also performed with the same individuals for direct comparisons (matched N control columns).

https://doi.org/10.1371/journal.pgen.1007739.s030

(XLS)

S9 Table. Top locus SNP (p < 1 x 10–6) HaploReg (Roadmap Epigenomics and ENCODE) epigenetic evidence.

Data were obtained from HaploReg 4.1 (https://pubs.broadinstitute.org/mammals/haploreg/haploreg.php) using exact SNPs (LD threshold = NA) and a ChromHMM 15-state model, followed by extraction from HTML. “Locus Class” indicates “Discovery/Replication” for main analyses or “Combined meta-analysis” for regions not clearing initial discovery p-value thresholds (i.e. S7 Table loci).

https://doi.org/10.1371/journal.pgen.1007739.s031

(XLS)

S10 Table. Top locus SNP (p < 1 x 10–6) Blueprint and Vermunt epigenetic evidence.

Blueprint cell lines are a non-cancerous sub-set of largely blood cells. Vermunt brain region samples were not assayed for promoter regions. “Locus Class” indicates “Discovery/Replication” for main analyses or “Combined meta-analysis” for regions not clearing initial discovery p-value thresholds (i.e. S7 Table loci).

https://doi.org/10.1371/journal.pgen.1007739.s032

(XLS)

S11 Table. Top locus SNP (p < 1 x 10–6) eQTL evidence.

Data were obtained from Fairfax et al. (monocytes), Geuvadis (LCLs), GTEx, Hao et al. (lung), Muther (adipose, LCLs, skin), Raj et al. (CD4, monocytes), Westra et al. (whole blood), and Zeller et al. (monocytes). “Locus Class” indicates “Discovery/Replication” for main analyses or “Combined meta-analysis” for regions not clearing initial discovery p-value thresholds (i.e. S7 Table loci).

https://doi.org/10.1371/journal.pgen.1007739.s033

(XLS)

S12 Table. Multi-variate MTAG (p < 10 x 10–6) results.

MTAG analysis was performed on European ancestry samples using our summary statistics for the three traits simultaneously. Lead results (p < 1 x 10–6) are shown.

https://doi.org/10.1371/journal.pgen.1007739.s034

(XLS)

S13 Table. MetaXcan gene-level results for subcutaneous adipose tissue in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s035

(XLS)

S14 Table. MetaXcan gene-level results for visceral omentum adipose tissue in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s036

(XLS)

S15 Table. MetaXcan gene-level results for hypothalamus in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s037

(XLS)

S16 Table. MetaXcan gene-level results for liver in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s038

(XLS)

S17 Table. MetaXcan gene-level results for lung in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s039

(XLS)

S18 Table. MetaXcan gene-level results for skeletal muscle in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s040

(XLS)

S19 Table. MetaXcan gene-level results for DGN whole blood in European-Americans.

Lead results (p < 0.05) are shown. Note that Ensembl IDs obtained from MetaXcan are occasionally unavailable.

https://doi.org/10.1371/journal.pgen.1007739.s041

(XLS)

S20 Table. GIGSEA KEGG pathway enrichment in whole blood analyses.

MetaXcan gene-level results for DGN whole blood (S19 Table, all p-values) were used as input.

https://doi.org/10.1371/journal.pgen.1007739.s042

(XLS)

S21 Table. GIGSEA micro RNA enrichment in whole blood analyses.

MetaXcan gene-level results for DGN whole blood (S19 Table, all p-values) were used as input. MicroRNAs are curated by the Molecular Signatures Database (http://software.broadinstitute.org/gsea/msigdb/index.jsp).

https://doi.org/10.1371/journal.pgen.1007739.s043

(XLS)

S22 Table. GIGSEA transcription factor binding site enrichment in whole blood analyses.

MetaXcan gene-level results for DGN whole blood (S19 Table, all p-values) were used as input. Transcription factor binding sites are curated by the Molecular Signatures Database (http://software.broadinstitute.org/gsea/msigdb/index.jsp).

https://doi.org/10.1371/journal.pgen.1007739.s044

(XLS)

S23 Table. Study-level variant counts.

Cohort-level information is shown for the three primary analyses, including discovery/replication category, the number of individuals with available phenotyping and genotyping, genotyping platform, imputation panel, and the number of imputed variants tested in each analysis following info score, MAC, and MAF filtering.

https://doi.org/10.1371/journal.pgen.1007739.s045

(XLS)

Acknowledgments

The authors wish to thank the participants and study staff of all of our cohorts for their important contributions. The authors thank the staff and participants of the ARIC study for their important contribution. This manuscript was not prepared in collaboration with investigators of the ARIC Study, and does not necessarily reflect the opinions or conclusions of the ARIC Study or the NHLBI. The authors thank the staff and participants of HCHS/SOL for their important contributions. Investigators website– http://www.cscc.unc.edu/hchs/. The authors thank the participants and data collection staff of the Jackson Heart Study. The views expressed in this manuscript are those of the authors and do not necessarily represent the views of the National Heart, Lung, and Blood Institute; the National Institutes of Health; or the U.S. Department of Health and Human Services. The authors thank the investigators, the staff, and the participants of the MESA study for their valuable contributions. A full list of participating MESA investigators and institutions can be found at http://www.mesa-nhlbi.org. We thank the field staff in Starr County for their careful collection of these data and are especially grateful to the participants who so graciously cooperated and gave of their time.

References

  1. 1. Antonelli Incalzi R., Marra C., Giordano A., Calcagni M.L., Cappa A., Basso S., Pagliari G., and Fuso L. (2003). Cognitive impairment in chronic obstructive pulmonary disease–a neuropsychological and spect study. J Neurol 250, 325–332. pmid:12638024
  2. 2. Yaffe K., Laffan A.M., Harrison S.L., Redline S., Spira A.P., Ensrud K.E., Ancoli-Israel S., and Stone K.L. (2011). Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 306, 613–619. pmid:21828324
  3. 3. McDonald M.-L.N., Cho M.H., Sørheim I.-C., Lutz S.M., Castaldi P.J., Lomas D.A., Coxson H.O., Edwards L.D., MacNee W., Vestbo J., et al. (2014). Common genetic variants associated with resting oxygenation in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 51, 678–687. pmid:24825563
  4. 4. Tarnoki D.L., Medda E., Tarnoki A.D., Lazar Z., Fagnani C., Stazi M.A., Karlinger K., Torzsa P., Kalabay L., Garami Z., et al. (2014). Genetic influence on capillary oxygen saturation: a twin study. Lung 192, 429–434. pmid:24584632
  5. 5. Beall C.M. (2006). Andean, Tibetan, and Ethiopian patterns of adaptation to high-altitude hypoxia. Integr Comp Biol 46, 18–24. pmid:21672719
  6. 6. Liang J., Cade B.E., Wang H., Chen H., Gleason K.J., Larkin E.K., Saxena R., Lin X., Redline S., and Zhu X. (2016). Comparison of Heritability Estimation and Linkage Analysis for Multiple Traits Using Principal Component Analyses. Genet Epidemiol 40, 222–232. pmid:27027516
  7. 7. Gami A.S., Hodge D.O., Herges R.M., Olson E.J., Nykodym J., Kara T., and Somers V.K. (2007). Obstructive sleep apnea, obesity, and the risk of incident atrial fibrillation. J Am Coll Cardiol 49, 565–571. pmid:17276180
  8. 8. Nieto F.J., Peppard P.E., Young T., Finn L., Hla K.M., and Farré R. (2012). Sleep-disordered breathing and cancer mortality: results from the Wisconsin Sleep Cohort Study. Am J Respir Crit Care Med 186, 190–194. pmid:22610391
  9. 9. Campos-Rodriguez F., Martinez-Garcia M.A., Martinez M., Duran-Cantolla J., Peña M. de la, Masdeu M.J., Gonzalez M., F. del Campo, Gallego I., Marin J.M., et al. (2013). Association between obstructive sleep apnea and cancer incidence in a large multicenter Spanish cohort. Am J Respir Crit Care Med 187, 99–105. pmid:23155146
  10. 10. Gozal D., Ham S.A., and Mokhlesi B. (2016). Sleep Apnea and Cancer: Analysis of a Nationwide Population Sample. Sleep 39, 1493–1500. pmid:27166241
  11. 11. Gunnarsson S.I., Peppard P.E., Korcarz C.E., Barnet J.H., Hagen E.W., Hla K.M., Palta M., Young T., and Stein J.H. (2015). Minimal nocturnal oxygen saturation predicts future subclinical carotid atherosclerosis: the Wisconsin sleep cohort. J. Sleep Res. 24, 680–686. pmid:26096939
  12. 12. Kendzerska T., Gershon A.S., Hawker G., Leung R.S., and Tomlinson G. (2014). Obstructive sleep apnea and risk of cardiovascular events and all-cause mortality: a decade-long historical cohort study. PLoS Med 11, e1001599. pmid:24503600
  13. 13. Oldenburg O., Wellmann B., Buchholz A., Bitter T., Fox H., Thiem U., Horstkotte D., and Wegscheider K. (2016). Nocturnal hypoxaemia is associated with increased mortality in stable heart failure patients. Eur Heart J 37, 1695–1703. pmid:26612581
  14. 14. Gellen B., Canouï-Poitrine F., Boyer L., Drouot X., Le Thuaut A., Bodez D., Covali-Noroc A., D’ortho M.P., Guendouz S., Rappeneau S., et al. (2016). Apnea-hypopnea and desaturations in heart failure with reduced ejection fraction: Are we aiming at the right target? Int J Cardiol 203, 1022–1028. pmid:26630630
  15. 15. Minville C., Hilleret M.-N., Tamisier R., Aron-Wisnewsky J., Clement K., Trocme C., Borel J.-C., Lévy P., Zarski J.-P., and Pépin J.-L. (2014). Nonalcoholic fatty liver disease, nocturnal hypoxia, and endothelial function in patients with sleep apnea. Chest 145, 525–533. pmid:24264333
  16. 16. Lacasse Y., Sériès F., Vujovic-Zotovic N., Goldstein R., Bourbeau J., Lecours R., Aaron S.D., and Maltais F. (2011). Evaluating nocturnal oxygen desaturation in COPD–revised. Respir Med 105, 1331–1337. pmid:21561753
  17. 17. Ross K.R., Storfer-Isser A., Hart M.A., Kibler A.M.V., Rueschman M., Rosen C.L., Kercsmar C.M., and Redline S. (2012). Sleep-disordered breathing is associated with asthma severity in children. J Pediatr 160, 736–742. pmid:22133422
  18. 18. Corte T.J., Wort S.J., Talbot S., Macdonald P.M., Hansel D.M., Polkey M., Renzoni E., Maher T.M., Nicholson A.G., and Wells A.U. (2012). Elevated nocturnal desaturation index predicts mortality in interstitial lung disease. Sarcoidosis Vasc Diffuse Lung Dis 29, 41–50. pmid:23311122
  19. 19. Kolilekas L., Manali E., Vlami K.A., Lyberopoulos P., Triantafillidou C., Kagouridis K., Baou K., Gyftopoulos S., Vougas K.N., Karakatsani A., et al. (2013). Sleep oxygen desaturation predicts survival in idiopathic pulmonary fibrosis. J Clin Sleep Med 9, 593–601. pmid:23772193
  20. 20. Shungin D., Winkler T.W., Croteau-Chonka D.C., Ferreira T., Locke A.E., Mägi R., Strawbridge R.J., Pers T.H., Fischer K., Justice A.E., et al. (2015). New genetic loci link adipose and insulin biology to body fat distribution. Nature 518, 187–196. pmid:25673412
  21. 21. Bixler E.O., Vgontzas A.N., Lin H.M., Ten Have T., Rein J., Vela-Bueno A., and Kales A. (2001). Prevalence of sleep-disordered breathing in women: effects of gender. Am J Respir Crit Care Med 163, 608–613. pmid:11254512
  22. 22. Cairns A., Poulos G., and Bogan R. (2016). Sex differences in sleep apnea predictors and outcomes from home sleep apnea testing. Nat Sci Sleep 8, 197–205. pmid:27418861
  23. 23. Jordan A.S., McEvoy R.D., Edwards J.K., Schory K., Yang C.-K., Catcheside P.G., Fogel R.B., Malhotra A., and White D.P. (2004). The influence of gender and upper airway resistance on the ventilatory response to arousal in obstructive sleep apnoea in humans. J Physiol 558, 993–1004. pmid:15218069
  24. 24. Simpson L., Mukherjee S., Cooper M.N., Ward K.L., Lee J.D., Fedson A.C., Potter J., Hillman D.R., Eastwood P., Palmer L.J., et al. (2010). Sex differences in the association of regional fat distribution with the severity of obstructive sleep apnea. Sleep 33, 467–474. pmid:20394315
  25. 25. Cade B.E., Chen H., Stilp A.M., Gleason K.J., Sofer T., Ancoli-Israel S., Arens R., Bell G.I., Below J.E., Bjonnes A.C., et al. (2016). Genetic Associations with Obstructive Sleep Apnea Traits in Hispanic/Latino Americans. Am J Respir Crit Care Med 194, 886–897. pmid:26977737
  26. 26. Hankinson J.L., Odencrantz J.R., and Fedan K.B. (1999). Spirometric reference values from a sample of the general U.S. population. Am J Respir Crit Care Med 159, 179–187. pmid:9872837
  27. 27. Gugger M., Bögershausen S., and Schäffler L. (1993). Arousal responses to added inspiratory resistance during REM and non-REM sleep in normal subjects. Thorax 48, 125–129. pmid:8493624
  28. 28. Carberry J.C., Jordan A.S., White D.P., Wellman A., and Eckert D.J. (2016). Upper Airway Collapsibility (Pcrit) and Pharyngeal Dilator Muscle Activity are Sleep-Stage Dependent. Sleep 39, 511–521. pmid:26612386
  29. 29. Turley P., Walters R.K., Maghzian O., Okbay A., Lee J.J., Fontana M.A., Nguyen-Viet T.A., Wedow R., Zacher M., Furlotte N.A., et al. (2018). Multi-trait analysis of genome-wide association summary statistics using MTAG. Nat. Genet. 50, 229–237. pmid:29292387
  30. 30. Rapoport S. (1968). The regulation of glycolysis in mammalian erythrocytes. Essays Biochem 4, 69–103. pmid:4308730
  31. 31. Lowry O.H., and Passonneau J.V. (1964). The Relationships between Substrates and Enzymes of Glycolysis in Brain. J Biol Chem 239, 31–42. pmid:14114860
  32. 32. Hoffstein V., Herridge M., Mateika S., Redline S., and Strohl K.P. (1994). Hematocrit levels in sleep apnea. Chest 106, 787–791. pmid:8082360
  33. 33. Wagner P.D., Simonson T.S., Wei G., Wagner H.E., Wuren T., Qin G., Yan M., and Ge R.L. (2015). Sea-level haemoglobin concentration is associated with greater exercise capacity in Tibetan males at 4200 m. Exp Physiol 100, 1256–1262. pmid:26454003
  34. 34. Chopra S., Rathore A., Younas H., Pham L.V., Gu C., Beselman A., Kim I.-Y., Wolfe R.R., Perin J., Polotsky V.Y., et al. (2017). Obstructive Sleep Apnea Dynamically Increases Nocturnal Plasma Free Fatty Acids, Glucose, and Cortisol During Sleep. J. Clin. Endocrinol. Metab. 102, 3172–3181. pmid:28595341
  35. 35. Westra H.-J., Peters M.J., Esko T., Yaghootkar H., Schurmann C., Kettunen J., Christiansen M.W., Fairfax B.P., Schramm K., Powell J.E., et al. (2013). Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat Genet 45, 1238–1243. pmid:24013639
  36. 36. van der Harst P., Zhang W., Mateo Leach I., Rendon A., Verweij N., Sehmi J., Paul D.S., Elling U., Allayee H., Li X., et al. (2012). Seventy-five genetic loci influencing the human red blood cell. Nature 492, 369–375. pmid:23222517
  37. 37. Soranzo N., Sanna S., Wheeler E., Gieger C., Radke D., Dupuis J., Bouatia-Naji N., Langenberg C., Prokopenko I., Stolerman E., et al. (2010). Common variants at 10 genomic loci influence hemoglobin A₁(C) levels via glycemic and nonglycemic pathways. Diabetes 59, 3229–3239. pmid:20858683
  38. 38. An P., Miljkovic I., Thyagarajan B., Kraja A.T., Daw E.W., Pankow J.S., Selvin E., Kao W.H.L., Maruthur N.M., Nalls M.A., et al. (2014). Genome-wide association study identifies common loci influencing circulating glycated hemoglobin (HbA1c) levels in non-diabetic subjects: the Long Life Family Study (LLFS). Metabolism 63, 461–468. pmid:24405752
  39. 39. Benesch R., Benesch R.E., and Yu C.I. (1968). Reciprocal binding of oxygen and diphosphoglycerate by human hemoglobin. Proc Natl Acad Sci U A 59, 526–532. pmid:5238982
  40. 40. van Wijk R., and van Solinge W.W. (2005). The energy-less red blood cell is lost: erythrocyte enzyme abnormalities of glycolysis. Blood 106, 4034–4042. pmid:16051738
  41. 41. Mulquiney P.J., and Kuchel P.W. (1999). Model of 2,3-bisphosphoglycerate metabolism in the human erythrocyte based on detailed enzyme kinetic equations: computer simulation and metabolic control analysis. Biochem J 342 Pt 3, 597–604. pmid:10477270
  42. 42. Moon J.-S., Hisata S., Park M.-A., DeNicola G.M., Ryter S.W., Nakahira K., and Choi A.M.K. (2015). mTORC1-Induced HK1-Dependent Glycolysis Regulates NLRP3 Inflammasome Activation. Cell Rep 12, 102–115. pmid:26119735
  43. 43. Traba J., Kwarteng-Siaw M., Okoli T.C., Li J., Huffstutler R.D., Bray A., Waclawiw M.A., Han K., Pelletier M., Sauve A.A., et al. (2015). Fasting and refeeding differentially regulate NLRP3 inflammasome activation in human subjects. J Clin Invest 125, 4592–4600. pmid:26529255
  44. 44. Lasithiotaki I., Giannarakis I., Tsitoura E., Samara K.D., Margaritopoulos G.A., Choulaki C., Vasarmidi E., Tzanakis N., Voloudaki A., Sidiropoulos P., et al. (2016). NLRP3 inflammasome expression in idiopathic pulmonary fibrosis and rheumatoid lung. Eur Respir J 47, 910–918. pmid:26743485
  45. 45. Grailer J.J., Canning B.A., Kalbitz M., Haggadone M.D., Dhond R.M., Andjelkovic A.V., Zetoune F.S., and Ward P.A. (2014). Critical role for the NLRP3 inflammasome during acute lung injury. J Immunol 192, 5974–5983. pmid:24795455
  46. 46. Sayan M., and Mossman B.T. (2016). The NLRP3 inflammasome in pathogenic particle and fibre-associated lung inflammation and diseases. Part Fibre Toxicol 13, 51. pmid:27650313
  47. 47. Stout-Delgado H.W., Cho S.J., Chu S.G., Mitzel D.N., Villalba J., El-Chemaly S., Ryter S.W., Choi A.M.K., and Rosas I.O. (2016). Age-Dependent Susceptibility to Pulmonary Fibrosis Is Associated with NLRP3 Inflammasome Activation. Am J Respir Cell Mol Biol 55, 252–263. pmid:26933834
  48. 48. Hosseinian N., Cho Y., Lockey R.F., and Kolliputi N. (2015). The role of the NLRP3 inflammasome in pulmonary diseases. Ther Adv Respir Dis 9, 188–197. pmid:26012351
  49. 49. Besnard A.-G., Guillou N., Tschopp J., Erard F., Couillin I., Iwakura Y., Quesniaux V., Ryffel B., and Togbe D. (2011). NLRP3 inflammasome is required in murine asthma in the absence of aluminum adjuvant. Allergy 66, 1047–1057. pmid:21443539
  50. 50. Lee T.-H., Song H.J., and Park C.-S. (2014). Role of inflammasome activation in development and exacerbation of asthma. Asia Pac Allergy 4, 187–196. pmid:25379478
  51. 51. Im H., and Ammit A.J. (2014). The NLRP3 inflammasome: role in airway inflammation. Clin Exp Allergy 44, 160–172. pmid:24118105
  52. 52. Wu J., Yan Z., Schwartz D.E., Yu J., Malik A.B., and Hu G. (2013). Activation of NLRP3 inflammasome in alveolar macrophages contributes to mechanical stretch-induced lung inflammation and injury. J Immunol 190, 3590–3599. pmid:23436933
  53. 53. Lavie L. (2015). Oxidative stress in obstructive sleep apnea and intermittent hypoxia–revisited–the bad ugly and good: implications to the heart and brain. Sleep Med Rev 20, 27–45. pmid:25155182
  54. 54. Lederer D.J., Jelic S., Basner R.C., Ishizaka A., and Bhattacharya J. (2009). Circulating KL-6, a biomarker of lung injury, in obstructive sleep apnoea. Eur Respir J 33, 793–796. pmid:19336590
  55. 55. Kim J.S., Podolanczuk A.J., Borker P., Kawut S.M., Raghu G., Kaufman J.D., Hinckley Stukovsky K.D., Hoffman E.A., Barr R.G., Gottlieb D.J., et al. (2017). Obstructive Sleep Apnea and Subclinical Interstitial Lung Disease in MESA. Ann. Am. Thorac. Soc. 14, 1786–1795. pmid:28613935
  56. 56. Broytman O., Braun R.K., Morgan B.J., Pegelow D.F., Hsu P.-N., Mei L.S., Koya A.K., Eldridge M., and Teodorescu M. (2015). Effects of chronic intermittent hypoxia on allergen-induced airway inflammation in rats. Am. J. Respir. Cell Mol. Biol. 52, 162–170. pmid:25004109
  57. 57. Zielinski M.R., Gerashchenko D., Karpova S.A., Konanki V., McCarley R.W., Sutterwala F.S., Strecker R.E., and Basheer R. (2017). The NLRP3 inflammasome modulates sleep and NREM sleep delta power induced by spontaneous wakefulness, sleep deprivation and lipopolysaccharide. Brain Behav Immun. 62, 137–150. pmid:28109896
  58. 58. Prabhakar N.R., and Semenza G.L. (2016). Regulation of carotid body oxygen sensing by hypoxia-inducible factors. Pflugers Arch. 468, 71–75. pmid:26265380
  59. 59. Luo F., Liu X., Yan N., Li S., Cao G., Cheng Q., Xia Q., and Wang H. (2006). Hypoxia-inducible transcription factor-1alpha promotes hypoxia-induced A549 apoptosis via a mechanism that involves the glycolysis pathway. BMC Cancer 6, 26. pmid:16438736
  60. 60. Palsson-McDermott E.M., Curtis A.M., Goel G., Lauterbach M.A.R., Sheedy F.J., Gleeson L.E., van den Bosch M.W.M., Quinn S.R., Domingo-Fernandez R., Johnston D.G.W., et al. (2015). Pyruvate kinase M2 regulates Hif-1α activity and IL-1β induction and is a critical determinant of the warburg effect in LPS-activated macrophages. Cell Metab 21, 65–80. pmid:25565206
  61. 61. Corcoran S.E., and O’Neill L.A.J. (2016). HIF1α and metabolic reprogramming in inflammation. J Clin Invest 126, 3699–3707. pmid:27571407
  62. 62. Eckle T., Brodsky K., Bonney M., Packard T., Han J., Borchers C.H., Mariani T.J., Kominsky D.J., Mittelbronn M., and Eltzschig H.K. (2013). HIF1A reduces acute lung injury by optimizing carbohydrate metabolism in the alveolar epithelium. PLoS Biol 11, e1001665. pmid:24086109
  63. 63. Hoshino K., Tsutsui H., Kawai T., Takeda K., Nakanishi K., Takeda Y., and Akira S. (1999). Cutting edge: generation of IL-18 receptor-deficient mice: evidence for IL-1 receptor-related protein as an essential IL-18 binding receptor. J Immunol 162, 5041–5044. pmid:10227969
  64. 64. Cheung H., Chen N.-J., Cao Z., Ono N., Ohashi P.S., and Yeh W.-C. (2005). Accessory protein-like is essential for IL-18-mediated signaling. J Immunol 174, 5351–5357. pmid:15843532
  65. 65. Dinarello C.A., Novick D., Kim S., and Kaplanski G. (2013). Interleukin-18 and IL-18 binding protein. Front Immunol 4, 289. pmid:24115947
  66. 66. Wang J., Thingholm L.B., Skiecevičienė J., Rausch P., Kummen M., Hov J.R., Degenhardt F., Heinsen F.-A., Rühlemann M.C., Szymczak S., et al. (2016). Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat. Genet. 48, 1396–1406. pmid:27723756
  67. 67. Hoshino T., Kato S., Oka N., Imaoka H., Kinoshita T., Takei S., Kitasato Y., Kawayama T., Imaizumi T., Yamada K., et al. (2007). Pulmonary inflammation and emphysema: role of the cytokines IL-18 and IL-13. Am J Respir Crit Care Med 176, 49–62. pmid:17400729
  68. 68. Kang M.-J., Choi J.-M., Kim B.H., Lee C.-M., Cho W.-K., Choe G., Kim D.-H., Lee C.G., and Elias J.A. (2012). IL-18 induces emphysema and airway and vascular remodeling via IFN-γ, IL-17A, and IL-13. Am J Respir Crit Care Med 185, 1205–1217. pmid:22383501
  69. 69. Dolinay T., Kim Y.S., Howrylak J., Hunninghake G.M., An C.H., Fredenburgh L., Massaro A.F., Rogers A., Gazourian L., Nakahira K., et al. (2012). Inflammasome-regulated cytokines are critical mediators of acute lung injury. Am J Respir Crit Care Med 185, 1225–1234. pmid:22461369
  70. 70. Jones H.D., Crother T.R., Gonzalez-Villalobos R.A., Jupelli M., Chen S., Dagvadorj J., Arditi M., and Shimada K. (2014). The NLRP3 inflammasome is required for the development of hypoxemia in LPS/mechanical ventilation acute lung injury. Am J Respir Cell Mol Biol 50, 270–280. pmid:24007300
  71. 71. Minoguchi K., Yokoe T., Tazaki T., Minoguchi H., Tanaka A., Oda N., Okada S., Ohta S., Naito H., and Adachi M. (2005). Increased carotid intima-media thickness and serum inflammatory markers in obstructive sleep apnea. Am J Respir Crit Care Med 172, 625–630. pmid:16120716
  72. 72. Haspel J.A., Chettimada S., Shaik R.S., Chu J.-H., Raby B.A., Cernadas M., Carey V., Process V., Hunninghake G.M., Ifedigbo E., et al. (2014). Circadian rhythm reprogramming during lung inflammation. Nat Commun 5, 4753. pmid:25208554
  73. 73. Moffatt M.F., Gut I.G., Demenais F., Strachan D.P., Bouzigon E., Heath S., von Mutius E., Farrall M., Lathrop M., Cookson W.O., et al. (2010). A large-scale, consortium-based genomewide association study of asthma. N Engl J Med 363, 1211–1221. pmid:20860503
  74. 74. Kim J., Shao Y., Kim S.Y., Kim S., Song H.K., Jeon J.H., Suh H.W., Chung J.W., Yoon S.R., Kim Y.S., et al. (2008). Hypoxia-induced IL-18 increases hypoxia-inducible factor-1alpha expression through a Rac1-dependent NF-kappaB pathway. Mol Biol Cell 19, 433–444. pmid:18003981
  75. 75. Petrache I., Fijalkowska I., Medler T.R., Skirball J., Cruz P., Zhen L., Petrache H.I., Flotte T.R., and Tuder R.M. (2006). alpha-1 antitrypsin inhibits caspase-3 activity, preventing lung endothelial cell apoptosis. Am J Pathol 169, 1155–1166. pmid:17003475
  76. 76. Ghim J., Moon J.-S., Lee C.S., Lee J., Song P., Lee A., Jang J.-H., Kim D., Yoon J.H., Koh Y.J., et al. (2014). Endothelial deletion of phospholipase D2 reduces hypoxic response and pathological angiogenesis. Arter. Thromb Vasc Biol 34, 1697–1703. pmid:24947526
  77. 77. Tan W., Sherman D., Turesson J., Shao X.M., Janczewski W.A., and Feldman J.L. (2012). Reelin demarcates a subset of pre-Bötzinger complex neurons in adult rat. J Comp Neurol 520, 606–619. pmid:21858819
  78. 78. Jiang M., Ku W., Fu J., Offermanns S., Hsu W., and Que J. (2013). Gpr177 regulates pulmonary vasculature development. Dev. Camb. Engl. 140, 3589–3594.
  79. 79. Wilk J.B., Shrine N.R.G., Loehr L.R., Zhao J.H., Manichaikul A., Lopez L.M., Smith A.V., Heckbert S.R., Smolonska J., Tang W., et al. (2012). Genome-wide association studies identify CHRNA5/3 and HTR4 in the development of airflow obstruction. Am. J. Respir. Crit. Care Med. 186, 622–632. pmid:22837378
  80. 80. Owens R.L., Macrea M.M., and Teodorescu M. (2017). The overlaps of asthma or COPD with OSA: A focused review. Respirol. Carlton Vic 22, 1073–1083. pmid:28677827
  81. 81. Gharib S.A., Hayes A.L., Rosen M.J., and Patel S.R. (2013). A pathway-based analysis on the effects of obstructive sleep apnea in modulating visceral fat transcriptome. Sleep 36, 23–30. pmid:23288968
  82. 82. Marques C.R., Costa G.N., da Silva T.M., Oliveira P., Cruz A.A., Alcantara-Neves N.M., Fiaccone R.L., Horta B.L., Hartwig F.P., Burchard E.G., et al. (2017). Suggestive association between variants in IL1RAPL and asthma symptoms in Latin American children. Eur J Hum Genet 25, 439–445. pmid:28120837
  83. 83. Zhang X., Ozawa Y., Lee H., Wen Y.-D., Tan T.-H., Wadzinski B.E., and Seto E. (2005). Histone deacetylase 3 (HDAC3) activity is regulated by interaction with protein serine/threonine phosphatase 4. Genes Dev 19, 827–839. pmid:15805470
  84. 84. Feng D., Liu T., Sun Z., Bugge A., Mullican S.E., Alenghat T., Liu X.S., and Lazar M.A. (2011). A circadian rhythm orchestrated by histone deacetylase 3 controls hepatic lipid metabolism. Science 331, 1315–1319. pmid:21393543
  85. 85. Peppard P.E., Young T., Barnet J.H., Palta M., Hagen E.W., and Hla K.M. (2013). Increased prevalence of sleep-disordered breathing in adults. Am J Epidemiol 177, 1006–1014. pmid:23589584
  86. 86. Redline S. (2011). Genetics of Obstructive Sleep Apnea. In Principles and Practice of Sleep Medicine, Kryger Meir H W.C.;Roth, Thomas Dement, ed. (St. Louis, MO: Saunders), pp. 1183–1193.
  87. 87. The ARIC Investigators (1989). The Atherosclerosis Risk in Communities (ARIC) Study: design and objectives. The ARIC investigators. Am. J. Epidemiol. 129, 687–702. pmid:2646917
  88. 88. Feinleib M. (1985). The Framingham Study: sample selection, follow-up, and methods of analyses. Natl Cancer Inst Monogr 67, 59–64. pmid:4047152
  89. 89. Quan S.F., Howard B.V., Iber C., Kiley J.P., Nieto F.J., O’Connor G.T., Rapoport D.M., Redline S., Robbins J., Samet J.M., et al. (1997). The Sleep Heart Health Study: design, rationale, and methods. Sleep 20, 1077–1085. pmid:9493915
  90. 90. Redline S., Sanders M.H., Lind B.K., Quan S.F., Iber C., Gottlieb D.J., Bonekat W.H., Rapoport D.M., Smith P.L., and Kiley J.P. (1998). Methods for obtaining and analyzing unattended polysomnography data for a multicenter study. Sleep Heart Health Research Group. Sleep 21, 759–767. pmid:11300121
  91. 91. Redline S., Tishler P.V., Tosteson T.D., Williamson J., Kump K., Browner I., Ferrette V., and Krejci P. (1995). The familial aggregation of obstructive sleep apnea. Am J Respir Crit Care Med 151, 682–687. pmid:7881656
  92. 92. Bild D.E., Bluemke D.A., Burke G.L., Detrano R., Diez Roux A.V., Folsom A.R., Greenland P., Jacob D.R.J., Kronmal R., Liu K., et al. (2002). Multi-Ethnic Study of Atherosclerosis: objectives and design. Am J Epidemiol 156, 871–881. pmid:12397006
  93. 93. Chen X., Wang R., Zee P., Lutsey P.L., Javaheri S., Alcántara C., Jackson C.L., Williams M.A., and Redline S. (2015). Racial/Ethnic Differences in Sleep Disturbances: The Multi-Ethnic Study of Atherosclerosis (MESA). Sleep 38, 877–888. pmid:25409106
  94. 94. Orwoll E., Blank J.B., Barrett-Connor E., Cauley J., Cummings S., Ensrud K., Lewis C., Cawthon P.M., Marcus R., Marshall L.M., et al. (2005). Design and baseline characteristics of the osteoporotic fractures in men (MrOS) study–a large observational study of the determinants of fracture in older men. Contemp Clin Trials 26, 569–585. pmid:16084776
  95. 95. Mehra R., Stone K.L., Blackwell T., Ancoli Israel S., Dam T.-T.L., Stefanick M.L., and Redline S. (2007). Prevalence and correlates of sleep-disordered breathing in older men: osteoporotic fractures in men sleep study. J Am Geriatr Soc 55, 1356–1364. pmid:17767677
  96. 96. Hanis C.L., Ferrell R.E., Barton S.A., Aguilar L., Garza-Ibarra A., Tulloch B.R., Garcia C.A., and Schull W.J. (1983). Diabetes among Mexican Americans in Starr County, Texas. Am J Epidemiol 118, 659–672. pmid:6637993
  97. 97. Hanis C.L., Redline S., Cade B.E., Bell G.I., Cox N.J., Below J.E., Brown E.L., and Aguilar D. (2016). Beyond type 2 diabetes, obesity and hypertension: an axis including sleep apnea, left ventricular hypertrophy, endothelial dysfunction, and aortic stiffness among Mexican Americans in Starr County, Texas. Cardiovasc Diabetol 15, 86. pmid:27266869
  98. 98. Choi J.H., Kim E.J., Kim Y.S., Choi J., Kim T.H., Kwon S.Y., Lee H.M., Lee S.H., Shin C., and Lee S.H. (2010). Validation study of portable device for the diagnosis of obstructive sleep apnea according to the new AASM scoring criteria: Watch-PAT 100. Acta Otolaryngol. (Stockh.) 130, 838–843. pmid:20082567
  99. 99. Fried L.P., Borhani N.O., Enright P., Furberg C.D., Gardin J.M., Kronmal R.A., Kuller L.H., Manolio T.A., Mittelmark M.B., Newman A., et al. (1991). The Cardiovascular Health Study: design and rationale. Ann Epidemiol 1, 263–276. pmid:1669507
  100. 100. Sorlie P.D., Avilés-Santa L.M., Wassertheil-Smoller S., Kaplan R.C., Daviglus M.L., Giachello A.L., Schneiderman N., Raij L., Talavera G., Allison M., et al. (2010). Design and implementation of the Hispanic Community Health Study/Study of Latinos. Ann Epidemiol 20, 629–641. pmid:20609343
  101. 101. Redline S., Sotres-Alvarez D., Loredo J., Hall M., Patel S.R., Ramos A., Shah N., Ries A., Arens R., Barnhart J., et al. (2014). Sleep-disordered breathing in Hispanic/Latino individuals of diverse backgrounds. The Hispanic Community Health Study/Study of Latinos. Am J Respir Crit Care Med 189, 335–344. pmid:24392863
  102. 102. Westbrook P.R., Levendowski D.J., Cvetinovic M., Zavora T., Velimirovic V., Henninger D., and Nicholson D. (2005). Description and validation of the apnea risk evaluation system: a novel method to diagnose sleep apnea-hypopnea in the home. Chest 128, 2166–2175. pmid:16236870
  103. 103. Taylor H.A.J., Wilson J.G., Jones D.W., Sarpong D.F., Srinivasan A., Garrison R.J., Nelson C., and Wyatt S.B. (2005). Toward resolution of cardiovascular health disparities in African Americans: design and methods of the Jackson Heart Study. Ethn Dis 15, S6-4–17 pmid:16320381
  104. 104. Wilson J.G., Rotimi C.N., Ekunwe L., Royal C.D.M., Crump M.E., Wyatt S.B., Steffes M.W., Adeyemo A., Zhou J., Taylor H.A.J., et al. (2005). Study design for genetic analysis in the Jackson Heart Study. Ethn Dis 15, S6-30–37. pmid:16317983
  105. 105. Ng S.S.S., Chan T.-O., To K.-W., Ngai J., Tung A., Ko F.W.S., and Hui D.S.C. (2010). Validation of Embletta portable diagnostic system for identifying patients with suspected obstructive sleep apnoea syndrome (OSAS). Respirol. Carlton Vic 15, 336–342. pmid:20199644
  106. 106. Mukherjee S., Hillman D., Lee J., Fedson A., Simpson L., Ward K., Love G., Edwards C., Szegner B., and Palmer L.J. (2012). Cohort profile: the Western Australian Sleep Health Study. Sleep Breath 16, 205–215. pmid:21318257
  107. 107. Whitney C.W., Gottlieb D.J., Redline S., Norman R.G., Dodge R.R., Shahar E., Surovec S., and Nieto F.J. (1998). Reliability of scoring respiratory disturbance indices and sleep staging. Sleep 21, 749–757. pmid:11286351
  108. 108. McCarthy S., Das S., Kretzschmar W., Durbin R., Abecasis G., and Marchini J. (2015). A reference panel of 64,976 haplotypes for genotype imputation. BioRxiv 035170.
  109. 109. 1000 Genomes Project Consortium (2015). A global reference for human genetic variation. Nature 526, 68–74. pmid:26432245
  110. 110. Howie B.N., Donnelly P., and Marchini J. (2009). A flexible and accurate genotype imputation method for the next generation of genome-wide association studies. PLoS Genet 5, e1000529. pmid:19543373
  111. 111. Wang C., Zhan X., Liang L., Abecasis G.R., and Lin X. (2015). Improved ancestry estimation for both genotyping and sequencing data using projection procrustes analysis and genotype imputation. Am J Hum Genet 96, 926–937. pmid:26027497
  112. 112. Zhou X., and Stephens M. (2012). Genome-wide efficient mixed-model analysis for association studies. Nat Genet 44, 821–824. pmid:22706312
  113. 113. Conomos, M.P., Thornton, T.A., Gogarten, S.M., and Brown, L. (2017). GENESIS: GENetic EStimation and Inference in Structured samples (GENESIS): Statistical methods for analyzing genetic data from samples with population structure and/or relatedness. R package version 2.8.0. https://doi.org/10.18129/B9.bioc.GENESIS
  114. 114. Willer C.J., Li Y., and Abecasis G.R. (2010). METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191. pmid:20616382
  115. 115. Dadaev T., Leongamornlert D.A., Saunders E.J., Eeles R., and Kote-Jarai Z. (2016). LocusExplorer: a user-friendly tool for integrated visualization of human genetic association data and biological annotations. Bioinformatics 32, 949–951. pmid:26589274
  116. 116. Winkler T.W., Kutalik Z., Gorski M., Lottaz C., Kronenberg F., and Heid I.M. (2015). EasyStrata: evaluation and visualization of stratified genome-wide association meta-analysis data. Bioinformatics 31, 259–261. pmid:25260699
  117. 117. Ehlen J.C., Brager A.J., Baggs J., Pinckney L., Gray C.L., DeBruyne J.P., Esser K.A., Takahashi J.S., and Paul K.N. (2017). Bmal1 function in skeletal muscle regulates sleep. ELife 6, e26557. pmid:28726633
  118. 118. Ward L.D., and Kellis M. (2012). HaploReg: a resource for exploring chromatin states, conservation, and regulatory motif alterations within sets of genetically linked variants. Nucleic Acids Res 40, D930–4. pmid:22064851
  119. 119. ENCODE Project Consortium (2012). An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74. pmid:22955616
  120. 120. Roadmap Epigenomics Consortium, Kundaje A., Meuleman W., Ernst J., Bilenky M., Yen A., Heravi-Moussavi A., Kheradpour P., Zhang Z., Wang J., et al. (2015). Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330. pmid:25693563
  121. 121. Javierre B.M., Burren O.S., Wilder S.P., Kreuzhuber R., Hill S.M., Sewitz S., Cairns J., Wingett S.W., Várnai C., Thiecke M.J., et al. (2016). Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters. Cell 167, 1369–1384.e19. pmid:27863249
  122. 122. Chen L., Ge B., Casale F.P., Vasquez L., Kwan T., Garrido-Martín D., Watt S., Yan Y., Kundu K., Ecker S., et al. (2016). Genetic Drivers of Epigenetic and Transcriptional Variation in Human Immune Cells. Cell 167, 1398–1414.e24. pmid:27863251
  123. 123. Vermunt M.W., Reinink P., Korving J., de Bruijn E., Creyghton P.M., Basak O., Geeven G., Toonen P.W., Lansu N., Meunier C., et al. (2014). Large-scale identification of coregulated enhancer networks in the adult human brain. Cell Rep 9, 767–779. pmid:25373911
  124. 124. Lappalainen T., Sammeth M., Friedländer M.R., ‘t Hoen P.A.C., Monlong J., Rivas M.A., Gonzàlez-Porta M., Kurbatova N., Griebel T., Ferreira P.G., et al. (2013). Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501, 506–511. pmid:24037378
  125. 125. GTEx Consortium (2015). Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660. pmid:25954001
  126. 126. Fairfax B.P., Humburg P., Makino S., Naranbhai V., Wong D., Lau E., Jostins L., Plant K., Andrews R., McGee C., et al. (2014). Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949. pmid:24604202
  127. 127. Raj T., Rothamel K., Mostafavi S., Ye C., Lee M.N., Replogle J.M., Feng T., Lee M., Asinovski N., Frohlich I., et al. (2014). Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science 344, 519–523. pmid:24786080
  128. 128. Zeller T., Wild P., Szymczak S., Rotival M., Schillert A., Castagne R., Maouche S., Germain M., Lackner K., Rossmann H., et al. (2010). Genetics and beyond–the transcriptome of human monocytes and disease susceptibility. PLoS One 5, e10693. pmid:20502693
  129. 129. Hao K., Bossé Y., Nickle D.C., Paré P.D., Postma D.S., Laviolette M., Sandford A., Hackett T.L., Daley D., Hogg J.C., et al. (2012). Lung eQTLs to help reveal the molecular underpinnings of asthma. PLoS Genet. 8, e1003029. pmid:23209423
  130. 130. Grundberg E., Small K.S., Hedman Å.K., Nica A.C., Buil A., Keildson S., Bell J.T., Yang T.-P., Meduri E., Barrett A., et al. (2012). Mapping cis- and trans-regulatory effects across multiple tissues in twins. Nat. Genet. 44, 1084–1089. pmid:22941192
  131. 131. Giambartolomei C., Zhenli Liu J., Zhang W., Hauberg M., Shi H., Boocock J., Pickrell J., Jaffe A.E., CommonMind Consortium, Pasaniuc B., et al. (2018). A Bayesian framework for multiple trait colocalization from summary association statistics. Bioinformatics 34, 2538–2545. pmid:29579179
  132. 132. Barbeira A.N., Dickinson S.P., Bonazzola R., Zheng J., Wheeler H.E., Torres J.M., Torstenson E.S., Shah K.P., Garcia T., Edwards T.L., et al. (2018). Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat Commun. 9, 1825. pmid:29739930
  133. 133. Zhu S., Qian T., Hoshida Y., Shen Y., Yu J., and Hao K. (2018). GIGSEA: Genotype Imputed Gene Set Enrichment Analysis using GWAS Summary Level Data. Bioinformatics 35, 160–163. pmid:30010968