251
|
Luo M, Cai J, Luo S, Hong X, Xu L, Lin H, Chen X, Fu W. Causal effects of gut microbiota on the risk of chronic kidney disease: a Mendelian randomization study. Front Cell Infect Microbiol 2023; 13:1142140. [PMID: 37065213 PMCID: PMC10102584 DOI: 10.3389/fcimb.2023.1142140] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundPrevious studies have reported that gut microbiota is associated with an increased risk of chronic kidney disease (CKD) progression. However, whether gut microbiota has a causal effect on the development of CKD has not been revealed. Thus, we aimed to analyze the potential causal effect of gut microbiota on the risk of CKD using mendelian randomization (MR) study.Materials and MethodsIndependent single nucleotide polymorphisms closely associated with 196 gut bacterial taxa (N = 18340) were identified as instrumental variables. Two-sample MR was performed to evaluate the causal effect of gut microbiota on CKD (N = 480698), including inverse-variance-weighted (IVW) method, weighted median method, MR-Egger, mode-based estimation and MR-PRESSO. The robustness of the estimation was tested by a series of sensitivity analyses including Cochran’s Q test, MR-Egger intercept analysis, leave-one-out analysis and funnel plot. Statistical powers were also calculated.ResultsThe genetically predicted higher abundance of order Desulfovibrionales was causally associated with an increased risk of CKD (odds ratio = 1.15, 95% confidence interval: 1.05-1.26; p = 0.0026). Besides, we also detected potential causalities between nine other taxa (Eubacterium eligens group, Desulfovibrionaceae, Ruminococcaceae UCG-002, Deltaproteobacteria, Lachnospiraceae UCG-010, Senegalimassilia, Peptostreptococcaceae, Alcaligenaceae and Ruminococcus torques group) and CKD (p < 0.05). No heterogeneity or pleiotropy was detected for significant estimates.ConclusionWe found that Desulfovibrionales and nine other taxa are associated with CKD, thus confirming that gut microbiota plays an important role in the pathogenesis of CKD. Our work also provides new potential indicators and targets for screening and prevention of CKD.
Collapse
Affiliation(s)
- Mingli Luo
- Department of Pediatric Urology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jiahao Cai
- Department of Neurology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Shulu Luo
- Department of Prosthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaosi Hong
- Department of Endocrinology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lingxin Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Honghong Lin
- Department of Pediatric Orthopedics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Xiong Chen
- Department of Pediatric Urology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- *Correspondence: Xiong Chen, ; Wen Fu,
| | - Wen Fu
- Department of Pediatric Urology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
- *Correspondence: Xiong Chen, ; Wen Fu,
| |
Collapse
|
252
|
Xu JJ, Zhang XB, Tong WT, Ying T, Liu KQ. Phenome-wide Mendelian randomization study evaluating the association of circulating vitamin D with complex diseases. Front Nutr 2023; 10:1108477. [PMID: 37063319 PMCID: PMC10095159 DOI: 10.3389/fnut.2023.1108477] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/01/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundCirculating vitamin D has been associated with multiple clinical diseases in observational studies, but the association was inconsistent due to the presence of confounders. We conducted a bidirectional Mendelian randomization (MR) study to explore the healthy atlas of vitamin D in many clinical traits and evaluate their causal association.MethodsBased on a large-scale genome-wide association study (GWAS), the single nucleotide polymorphism (SNPs) instruments of circulating 25-hydroxyvitamin D (25OHD) from 443,734 Europeans and the corresponding effects of 10 clinical diseases and 42 clinical traits in the European population were recruited to conduct a bidirectional two-sample Mendelian randomization study. Under the network of Mendelian randomization analysis, inverse-variance weighting (IVW), weighted median, weighted mode, and Mendelian randomization (MR)–Egger regression were performed to explore the causal effects and pleiotropy. Mendelian randomization pleiotropy RESidual Sum and Outlier (MR-PRESSO) was conducted to uncover and exclude pleiotropic SNPs.ResultsThe results revealed that genetically decreased vitamin D was inversely related to the estimated BMD (β = −0.029 g/cm2, p = 0.027), TC (β = −0.269 mmol/L, p = 0.006), TG (β = −0.208 mmol/L, p = 0.002), and pulse pressure (β = −0.241 mmHg, p = 0.043), while positively associated with lymphocyte count (β = 0.037%, p = 0.015). The results did not reveal any causal association of vitamin D with clinical diseases. On the contrary, genetically protected CKD was significantly associated with increased vitamin D (β = 0.056, p = 2.361 × 10−26).ConclusionThe putative causal effects of circulating vitamin D on estimated bone mass, plasma triglyceride, and total cholesterol were uncovered, but not on clinical diseases. Vitamin D may be linked to clinical disease by affecting health-related metabolic markers.
Collapse
Affiliation(s)
- Jin-jian Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, China
- Department of Epidemiology, School of Public Health, Sun Yat-sen University (North Campus), Guangzhou, Guangdong, China
| | - Xiao-bin Zhang
- Department of Hepatobiliary Surgery, Jingdezhen No.1 People's Hospital, Jingdezhen, Jiangxi, China
| | - Wen-tao Tong
- Department of Hepatobiliary Surgery, Jingdezhen No.1 People's Hospital, Jingdezhen, Jiangxi, China
| | - Teng Ying
- Department of Cardiology, The First Affiliated Hospital of Jiangxi Medical College, Shangrao, Jiangxi, China
| | - Ke-qi Liu
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao, Jiangxi, China
- *Correspondence: Ke-qi Liu
| |
Collapse
|
253
|
Li C, Chen Y, Zhao W, Zhang C, Tang L, Ying Z, Chen W, Fu P, Song H, Zhou X, Zeng X. Genetic impact on the association of sleep patterns and chronic kidney disease: A prospective cohort study of 157,175 UK Biobank participants. J Psychosom Res 2023; 169:111323. [PMID: 37037154 DOI: 10.1016/j.jpsychores.2023.111323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/20/2023] [Accepted: 03/22/2023] [Indexed: 04/12/2023]
Abstract
OBJECTIVES The association between sleep pattern and chronic kidney disease (CKD) incidence, and whether the association is dependent on the genetic backgrounds has not been addressed. We sought to investigate the association of multidimensional sleep pattern with CKD in consideration of genetic polymorphisms. METHODS In this prospective cohort study of 157,175 participants from the UK Biobank, sleep patterns were derived by multiple correspondence analysis (MCA) and k-means clustering of individual sleep traits (sleep duration, insomnia, chronotype, daytime sleepiness, snoring, and night shift status). Cox proportional hazard regression was used to estimate the association between sleep patterns and CKD incidence. Gene-environment-wide interaction study (GEWIS) was performed to detect whether gene polymorphisms were modifiers on this association. RESULTS Compared with "healthy sleep" pattern, increased CKD incidence was observed in the clusters with "long sleep duration" (hazard ratios (HR) 1.42, 95% confidence intervals (CI), 1.18-1.72) and "night shift" (HR 1.23, 95% CI, 1.05-1.45) patterns, but not with the "short sleep duration" pattern. By GEWIS, we identified 167 SNPs as suggestive effect modifiers that interacted with unhealthy sleep patterns and affected the risk of CKD. CONCLUSIONS Unhealthy sleep patterns, with features of long sleep duration and night shift, may increase the risk of CKD. The study highlights the interaction of sleep and individual genetic risk to affect health outcomes.
Collapse
Affiliation(s)
- Chunyang Li
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China; Med-X Center for Informatics, Sichuan University, 17 Ren Min Nan Road 3(rd) section, Chengdu, Sichuan 610041, China
| | - Yilong Chen
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China; Med-X Center for Informatics, Sichuan University, 17 Ren Min Nan Road 3(rd) section, Chengdu, Sichuan 610041, China
| | - Weiling Zhao
- School of Biomedical Informatics, The University of Texas Health Science Centre at Houston, 7000 Fannin Street, Houston, TX 77030, USA
| | - Chao Zhang
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China; Med-X Center for Informatics, Sichuan University, 17 Ren Min Nan Road 3(rd) section, Chengdu, Sichuan 610041, China
| | - Lei Tang
- Division of Nephrology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Zhiye Ying
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China; Med-X Center for Informatics, Sichuan University, 17 Ren Min Nan Road 3(rd) section, Chengdu, Sichuan 610041, China
| | - Wenwen Chen
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China
| | - Ping Fu
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China; Med-X Center for Informatics, Sichuan University, 17 Ren Min Nan Road 3(rd) section, Chengdu, Sichuan 610041, China
| | - Huan Song
- Med-X Center for Informatics, Sichuan University, 17 Ren Min Nan Road 3(rd) section, Chengdu, Sichuan 610041, China; Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Xiaobo Zhou
- School of Biomedical Informatics, The University of Texas Health Science Centre at Houston, 7000 Fannin Street, Houston, TX 77030, USA
| | - Xiaoxi Zeng
- Kidney Research Institute, Biomedical Big Data Center, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China; Division of Nephrology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
254
|
Wuttke M, König E, Katsara MA, Kirsten H, Farahani SK, Teumer A, Li Y, Lang M, Göcmen B, Pattaro C, Günzel D, Köttgen A, Fuchsberger C. Imputation-powered whole-exome analysis identifies genes associated with kidney function and disease in the UK Biobank. Nat Commun 2023; 14:1287. [PMID: 36890159 PMCID: PMC9995463 DOI: 10.1038/s41467-023-36864-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
Genome-wide association studies have discovered hundreds of associations between common genotypes and kidney function but cannot comprehensively investigate rare coding variants. Here, we apply a genotype imputation approach to whole exome sequencing data from the UK Biobank to increase sample size from 166,891 to 408,511. We detect 158 rare variants and 105 genes significantly associated with one or more of five kidney function traits, including genes not previously linked to kidney disease in humans. The imputation-powered findings derive support from clinical record-based kidney disease information, such as for a previously unreported splice allele in PKD2, and from functional studies of a previously unreported frameshift allele in CLDN10. This cost-efficient approach boosts statistical power to detect and characterize both known and novel disease susceptibility variants and genes, can be generalized to larger future studies, and generates a comprehensive resource ( https://ckdgen-ukbb.gm.eurac.edu/ ) to direct experimental and clinical studies of kidney disease.
Collapse
Affiliation(s)
- Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| | - Eva König
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Maria-Alexandra Katsara
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | | | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Lang
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Burulca Göcmen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christian Fuchsberger
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy.
| |
Collapse
|
255
|
Zhang T, Yue Y, Jeong SJ, Ryu MS, Wu X, Yang HJ, Li C, Jeong DY, Park S. Improvement of Estrogen Deficiency Symptoms by the Intake of Long-Term Fermented Soybeans (Doenjang) Rich in Bacillus Species through Modulating Gut Microbiota in Estrogen-Deficient Rats. Foods 2023; 12:foods12061143. [PMID: 36981070 PMCID: PMC10048008 DOI: 10.3390/foods12061143] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/30/2023] Open
Abstract
Traditionally made doenjang (TMD) produced by the long-term fermentation of soybeans with salt may improve symptoms of estrogen deficiency. We aimed to evaluate the effects of four TMD types, containing low and high amounts of Bacillus species and biogenic amines (HBHA, HBLA, LBHA, and LBLA), on energy, glucose, and lipid metabolism, by altering the gut microbiota in estrogen-deficient ovariectomized (OVX) rats. Their mechanisms were also examined. The OVX rats were divided into the control, cooked soybean (CSB), HBHA, LBHA, HBLA, and LBLA groups. Sham-operated rats were the normal control group. Serum 17β-estradiol concentrations were similar among all OVX groups. Tail skin temperatures, which are indicative of hot flashes, were higher in the control than the HBHA and HBLA groups and were similar to the normal control group. Weight gain and visceral fat mass were lower in the TMD and CSB intake groups but not as low as in the normal control group. Lean body mass showed a trend opposite to that of visceral fat in the respective groups. The hepatic triglyceride content decreased with the TMD intake compared to the control and CSB groups. mRNA expressions of the peroxisome proliferator-activated receptor-γ (PPAR-γ) and carnitine palmitoyltransferase-1 in the TMD and CSB groups were as high as in the normal control group, and the PPAR-γ mRNA expression was more elevated in the HBLA group than in the normal control group. The morphology of the intestines improved in the TMD groups compared to the control, and the HBHA and HBLA groups showed an enhanced improvement compared to the CSB group. The HBHA, HBLA, and LBHA groups increased the α-diversity of the cecal microbiota compared to the control. Akkermenia and Lactobacillus were higher in the HBLA and LBLA groups compared to the control. The expression of the estrogen, forkhead box proteins of the class-O subgroup, and insulin-signaling pathways were lower in the control group, and HBHA and HBLA prevented their decrement. In conclusion, long-term treatment with TMD containing high amounts of Bacillus potentially improves estrogen deficiency symptoms more than unfermented soybeans.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Bioconvergence, Hoseo University, 20 hoseoro79bun-gil, Baebang-yup, Asan 31499, Republic of Korea
| | - Yu Yue
- Obesity/Diabetes Research Center, Department of Food and Nutrition, Hoseo University, Asan 31499, Republic of Korea
| | - Su-Ji Jeong
- Sunchang Research Center for Fermentation Microbes, Department of R & D, Microbial Institute for Fermentation Industry, 61-27 Minsokmaeul-gil, Sunchang-Gun 56048, Republic of Korea
| | - Myeong-Seon Ryu
- Sunchang Research Center for Fermentation Microbes, Department of R & D, Microbial Institute for Fermentation Industry, 61-27 Minsokmaeul-gil, Sunchang-Gun 56048, Republic of Korea
| | - Xuangao Wu
- Department of Bioconvergence, Hoseo University, 20 hoseoro79bun-gil, Baebang-yup, Asan 31499, Republic of Korea
| | - Hee-Jong Yang
- Sunchang Research Center for Fermentation Microbes, Department of R & D, Microbial Institute for Fermentation Industry, 61-27 Minsokmaeul-gil, Sunchang-Gun 56048, Republic of Korea
| | - Chen Li
- Obesity/Diabetes Research Center, Department of Food and Nutrition, Hoseo University, Asan 31499, Republic of Korea
| | - Do-Youn Jeong
- Sunchang Research Center for Fermentation Microbes, Department of R & D, Microbial Institute for Fermentation Industry, 61-27 Minsokmaeul-gil, Sunchang-Gun 56048, Republic of Korea
| | - Sunmin Park
- Department of Bioconvergence, Hoseo University, 20 hoseoro79bun-gil, Baebang-yup, Asan 31499, Republic of Korea
- Obesity/Diabetes Research Center, Department of Food and Nutrition, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
256
|
Nag A, Dhindsa RS, Middleton L, Jiang X, Vitsios D, Wigmore E, Allman EL, Reznichenko A, Carss K, Smith KR, Wang Q, Challis B, Paul DS, Harper AR, Petrovski S. Effects of protein-coding variants on blood metabolite measurements and clinical biomarkers in the UK Biobank. Am J Hum Genet 2023; 110:487-498. [PMID: 36809768 PMCID: PMC10027475 DOI: 10.1016/j.ajhg.2023.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
Genome-wide association studies (GWASs) have established the contribution of common and low-frequency variants to metabolic blood measurements in the UK Biobank (UKB). To complement existing GWAS findings, we assessed the contribution of rare protein-coding variants in relation to 355 metabolic blood measurements-including 325 predominantly lipid-related nuclear magnetic resonance (NMR)-derived blood metabolite measurements (Nightingale Health Plc) and 30 clinical blood biomarkers-using 412,393 exome sequences from four genetically diverse ancestries in the UKB. Gene-level collapsing analyses were conducted to evaluate a diverse range of rare-variant architectures for the metabolic blood measurements. Altogether, we identified significant associations (p < 1 × 10-8) for 205 distinct genes that involved 1,968 significant relationships for the Nightingale blood metabolite measurements and 331 for the clinical blood biomarkers. These include associations for rare non-synonymous variants in PLIN1 and CREB3L3 with lipid metabolite measurements and SYT7 with creatinine, among others, which may not only provide insights into novel biology but also deepen our understanding of established disease mechanisms. Of the study-wide significant clinical biomarker associations, 40% were not previously detected on analyzing coding variants in a GWAS in the same cohort, reinforcing the importance of studying rare variation to fully understand the genetic architecture of metabolic blood measurements.
Collapse
Affiliation(s)
- Abhishek Nag
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, TX 77030, USA
| | - Lawrence Middleton
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Xiao Jiang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Eleanor Wigmore
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Erik L Allman
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Anna Reznichenko
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Katherine R Smith
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - Benjamin Challis
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Dirk S Paul
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Andrew R Harper
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Early Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Department of Medicine, University of Melbourne, Austin Health, Melbourne, VIC, Australia.
| |
Collapse
|
257
|
Fan S, Spence JP, Feng Y, Hansen MEB, Terhorst J, Beltrame MH, Ranciaro A, Hirbo J, Beggs W, Thomas N, Nyambo T, Mpoloka SW, Mokone GG, Njamnshi A, Folkunang C, Meskel DW, Belay G, Song YS, Tishkoff SA. Whole-genome sequencing reveals a complex African population demographic history and signatures of local adaptation. Cell 2023; 186:923-939.e14. [PMID: 36868214 PMCID: PMC10568978 DOI: 10.1016/j.cell.2023.01.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 10/16/2022] [Accepted: 01/30/2023] [Indexed: 03/05/2023]
Abstract
We conduct high coverage (>30×) whole-genome sequencing of 180 individuals from 12 indigenous African populations. We identify millions of unreported variants, many predicted to be functionally important. We observe that the ancestors of southern African San and central African rainforest hunter-gatherers (RHG) diverged from other populations >200 kya and maintained a large effective population size. We observe evidence for ancient population structure in Africa and for multiple introgression events from "ghost" populations with highly diverged genetic lineages. Although currently geographically isolated, we observe evidence for gene flow between eastern and southern Khoesan-speaking hunter-gatherer populations lasting until ∼12 kya. We identify signatures of local adaptation for traits related to skin color, immune response, height, and metabolic processes. We identify a positively selected variant in the lightly pigmented San that influences pigmentation in vitro by regulating the enhancer activity and gene expression of PDPK1.
Collapse
Affiliation(s)
- Shaohua Fan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, School of Life Science, Fudan University, Shanghai, 200438, China; Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffrey P Spence
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yuanqing Feng
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew E B Hansen
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Terhorst
- Department of Statistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcia H Beltrame
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alessia Ranciaro
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jibril Hirbo
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William Beggs
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Neil Thomas
- Computer Science Division, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Thomas Nyambo
- Department of Biochemistry, Kampala International University in Tanzania, P.O. Box 9790, Dar es Salaam, Tanzania
| | - Sununguko Wata Mpoloka
- Department of Biological Sciences, Faculty of Science, University of Botswana Gaborone, Private Bag UB 0022, Gaborone, Botswana
| | - Gaonyadiwe George Mokone
- Department of Biomedical Sciences, Faculty of Medicine, University of Botswana Gaborone, Private Bag UB 0022, Gaborone, Botswana
| | - Alfred Njamnshi
- Department of Neurology, Central Hospital Yaoundé; Brain Research Africa Initiative (BRAIN), Neuroscience Lab, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 337, Yaoundé, Cameroon
| | - Charles Folkunang
- Department of Pharmacotoxicology and Pharmacokinetics, Faculty of Medicine and Biomedical Sciences, The University of Yaoundé I, P.O. Box 337, Yaoundé, Cameroon
| | - Dawit Wolde Meskel
- Department of Microbial Cellular and Molecular Biology, Addis Ababa University, P.O. Box 1176, Addis Ababa, Ethiopia
| | - Gurja Belay
- Department of Microbial Cellular and Molecular Biology, Addis Ababa University, P.O. Box 1176, Addis Ababa, Ethiopia
| | - Yun S Song
- Computer Science Division, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Sarah A Tishkoff
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
258
|
Kim H, Westerman KE, Smith K, Chiou J, Cole JB, Majarian T, von Grotthuss M, Kwak SH, Kim J, Mercader JM, Florez JC, Gaulton K, Manning AK, Udler MS. High-throughput genetic clustering of type 2 diabetes loci reveals heterogeneous mechanistic pathways of metabolic disease. Diabetologia 2023; 66:495-507. [PMID: 36538063 PMCID: PMC10108373 DOI: 10.1007/s00125-022-05848-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/28/2022] [Indexed: 12/24/2022]
Abstract
AIMS/HYPOTHESIS Type 2 diabetes is highly polygenic and influenced by multiple biological pathways. Rapid expansion in the number of type 2 diabetes loci can be leveraged to identify such pathways. METHODS We developed a high-throughput pipeline to enable clustering of type 2 diabetes loci based on variant-trait associations. Our pipeline extracted summary statistics from genome-wide association studies (GWAS) for type 2 diabetes and related traits to generate a matrix of 323 variants × 64 trait associations and applied Bayesian non-negative matrix factorisation (bNMF) to identify genetic components of type 2 diabetes. Epigenomic enrichment analysis was performed in 28 cell types and single pancreatic cells. We generated cluster-specific polygenic scores and performed regression analysis in an independent cohort (N=25,419) to assess for clinical relevance. RESULTS We identified ten clusters of genetic loci, recapturing the five from our prior analysis as well as novel clusters related to beta cell dysfunction, pronounced insulin secretion, and levels of alkaline phosphatase, lipoprotein A and sex hormone-binding globulin. Four clusters related to mechanisms of insulin deficiency, five to insulin resistance and one had an unclear mechanism. The clusters displayed tissue-specific epigenomic enrichment, notably with the two beta cell clusters differentially enriched in functional and stressed pancreatic beta cell states. Additionally, cluster-specific polygenic scores were differentially associated with patient clinical characteristics and outcomes. The pipeline was applied to coronary artery disease and chronic kidney disease, identifying multiple overlapping clusters with type 2 diabetes. CONCLUSIONS/INTERPRETATION Our approach stratifies type 2 diabetes loci into physiologically interpretable genetic clusters associated with distinct tissues and clinical outcomes. The pipeline allows for efficient updating as additional GWAS become available and can be readily applied to other conditions, facilitating clinical translation of GWAS findings. Software to perform this clustering pipeline is freely available.
Collapse
Affiliation(s)
- Hyunkyung Kim
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kenneth E Westerman
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Kirk Smith
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joshua Chiou
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Joanne B Cole
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Marcin von Grotthuss
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Takeda Pharmaceuticals, Cambridge, MA, USA
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jaegil Kim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- GlaxoSmithKline, Cambridge, MA, USA
| | - Josep M Mercader
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Jose C Florez
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Kyle Gaulton
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
| | - Alisa K Manning
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Miriam S Udler
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
259
|
Hung RKY, Winkler CA, Post FA. Host factors predisposing to kidney disease in people with HIV. Curr Opin HIV AIDS 2023; 18:87-92. [PMID: 36722197 DOI: 10.1097/coh.0000000000000784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE OF REVIEW To highlight advances in understanding of host factors, in particular host genetics, in the development of chronic kidney disease (CKD) in people with HIV. RECENT FINDINGS In Black populations, the G1 and G2 variants of the apolipoprotein L1 (APOL1) gene predispose to HIV-associated nephropathy (HIVAN). The risk of HIVAN is mostly confined to individuals with two APOL1 variants (kidney-risk genotypes). APOL1 kidney-risk genotypes are present in approximately 80% of patients with HIVAN and account for nearly half the burden of end-stage CKD in people of African ancestry with HIV. Progress has been made in elucidating the mechanisms of kidney injury in APOL1 nephropathy, and several targeted molecular therapies are being investigated in clinical trials. Genome- and epigenome-wide association studies are identifying additional genes and pathways that may be involved in the pathogenesis of CKD in people with HIV. SUMMARY Genetic variants of APOL1 are strongly associated with severe CKD and contribute to the high rates of CKD in Black populations with HIV. Most individuals with APOL1 kidney-risk genotypes, however, do not develop kidney disease and further studies are required to understand the role of additional genetic and environmental factors that may affect CKD risk in this population.
Collapse
Affiliation(s)
| | - Cheryl A Winkler
- Frederick National Laboratory for Cancer Research and the National Cancer Institute, Frederick, USA
| | - Frank A Post
- King's College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
260
|
Zhao SS, Bellou E, Verstappen SMM, Cook MJ, Sergeant JC, Warren RB, Barton A, Bowes J. Association between psoriatic disease and lifestyle factors and comorbidities: cross-sectional analysis and Mendelian randomization. Rheumatology (Oxford) 2023; 62:1272-1285. [PMID: 35861400 PMCID: PMC9977114 DOI: 10.1093/rheumatology/keac403] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/03/2022] [Accepted: 07/03/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To examine associations between PsA and psoriasis vs lifestyle factors and comorbidities by triangulating observational and genetic evidence. METHODS We analysed cross-sectional data from the UK Biobank (1836 PsA, 8995 psoriasis, 36 000 controls) to describe the association between psoriatic disease and lifestyle factors (including BMI and smoking) and 15 comorbidities [including diabetes and coronary artery disease (CAD)] using logistic models adjusted for age, sex and lifestyle factors. We applied bidirectional Mendelian randomization (MR) to genome-wide association data (3609 PsA and 7804 psoriasis cases, up to 1.2 million individuals for lifestyle factors and 757 601 for comorbidities) to examine causal direction, using the inverse-variance weighted method. RESULTS BMI was cross-sectionally associated with risk of PsA (OR 1.31 per 5 kg/m2 increase; 95% CI 1.26, 1.37) and psoriasis (OR 1.23; 1.20, 1.26), with consistent MR estimates (PsA OR 1.38; 1.14, 1.67; psoriasis OR 1.36; 1.18, 1.58). In both designs, smoking was more strongly associated with psoriasis than PsA. PsA and psoriasis were cross-sectionally associated with diabetes (OR 1.35 and 1.39, respectively) and CAD (OR 1.56 and 1.38, respective). Genetically predicted glycated haemoglobin (surrogate for diabetes) increased PsA risk (OR 1.18 per 6.7 mmol/mol increase; 1.02, 1.36) but not psoriasis. Genetic liability to PsA (OR 1.05; 1.003, 1.09) and psoriasis (OR 1.03; 1.001, 1.06) were associated with increased risk of CAD. CONCLUSION Observational and genetic evidence converge to suggest that BMI and glycaemic control are associated with increased psoriatic disease risk, while psoriatic disease is associated with increased CAD risk. Further research is needed to understand the mechanism of these associations.
Collapse
Affiliation(s)
| | - Eftychia Bellou
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester.,UK Dementia Research Institute, Cardiff University, Cardiff
| | - Suzanne M M Verstappen
- Centre for Epidemiology Versus Arthritis.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust
| | | | - Jamie C Sergeant
- Centre for Epidemiology Versus Arthritis.,Centre for Biostatistics, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre
| | - Richard B Warren
- Dermatology Centre, Salford Royal NHS Foundation Trust, Manchester NIHR Biomedical Research Centre, University of Manchester, Manchester, UK
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust
| |
Collapse
|
261
|
Kjaergaard AD, Krakauer J, Krakauer N, Teumer A, Winkler TW, Ellervik C. Allometric body shape indices, type 2 diabetes and kidney function: A two-sample Mendelian randomization study. Diabetes Obes Metab 2023. [PMID: 36855799 DOI: 10.1111/dom.15037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/02/2023]
Abstract
AIM To examine the association between body mass index (BMI)-independent allometric body shape indices and kidney function. MATERIALS AND METHODS We performed a two-sample Mendelian randomization (MR) analysis, using summary statistics from UK Biobank, CKDGen and DIAGRAM. BMI-independent allometric body shape indices were: A Body Shape Index (ABSI), Waist-Hip Index (WHI) and Hip Index (HI). Kidney function outcomes were: urinary albumin-to-creatinine ratio (UACR), estimated glomerular filtration rate and blood urea nitrogen. Furthermore, we investigated type 2 diabetes (T2D) as a potential mediator on the pathway to albuminuria. The main analysis was inverse variance-weighted random-effects MR in participants of European ancestry. We also performed several sensitivity MR analyses. RESULTS A 1-standard deviation (SD) increase in genetically predicted ABSI and WHI levels was associated with higher UACR (β = 0.039 [95% confidence interval: 0.016, 0.063] log [UACR], P = 0.001 for ABSI, and β = 0.028 [0.012, 0.044] log [UACR], P = 6 x 10-4 for WHI) in women, but not in men. Meanwhile, a 1-SD increase in genetically predicted HI was associated with lower UACR in women (β = -0.021 [-0.041, 0.000] log [UACR], P = 0.05) and in men (β = -0.026 [-0.058, 0.005] log [UACR], P = 0.10). Corresponding estimates in individuals with diabetes were substantially augmented. Risk of T2D increased for genetically high ABSI and WHI in women (P < 6 x 10-19 ) only, but decreased for genetically high HI in both sexes (P < 9 x 10-3 ). No other associations were observed. CONCLUSIONS Genetically high HI was associated with decreased risk of albuminuria, mediated through decreased T2D risk in both sexes. Opposite associations applied to genetically high ABSI and WHI in women only.
Collapse
Affiliation(s)
- Alisa D Kjaergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Jesse Krakauer
- Associated Physicians/Endocrinology, Berkley, Michigan, USA
| | - Nir Krakauer
- Department of Civil Engineering, City College of New York and Earth and Environmental Sciences, Graduate Center, City University of New York, New York, New York, USA
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Christina Ellervik
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Data and Development, Sorø, Denmark
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
262
|
Hill C, Duffy S, Coulter T, Maxwell AP, McKnight AJ. Harnessing Genomic Analysis to Explore the Role of Telomeres in the Pathogenesis and Progression of Diabetic Kidney Disease. Genes (Basel) 2023; 14:609. [PMID: 36980881 PMCID: PMC10048490 DOI: 10.3390/genes14030609] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The prevalence of diabetes is increasing globally, and this trend is predicted to continue for future decades. Research is needed to uncover new ways to manage diabetes and its co-morbidities. A significant secondary complication of diabetes is kidney disease, which can ultimately result in the need for renal replacement therapy, via dialysis or transplantation. Diabetic kidney disease presents a substantial burden to patients, their families and global healthcare services. This review highlights studies that have harnessed genomic, epigenomic and functional prediction tools to uncover novel genes and pathways associated with DKD that are useful for the identification of therapeutic targets or novel biomarkers for risk stratification. Telomere length regulation is a specific pathway gaining attention recently because of its association with DKD. Researchers are employing both observational and genetics-based studies to identify telomere-related genes associated with kidney function decline in diabetes. Studies have also uncovered novel functions for telomere-related genes beyond the immediate regulation of telomere length, such as transcriptional regulation and inflammation. This review summarises studies that have revealed the potential to harness therapeutics that modulate telomere length, or the associated epigenetic modifications, for the treatment of DKD, to potentially slow renal function decline and reduce the global burden of this disease.
Collapse
Affiliation(s)
- Claire Hill
- Centre for Public Health, Queen’s University of Belfast, Belfast BT12 6BA, UK
| | - Seamus Duffy
- Centre for Public Health, Queen’s University of Belfast, Belfast BT12 6BA, UK
| | - Tiernan Coulter
- Centre for Public Health, Queen’s University of Belfast, Belfast BT12 6BA, UK
| | - Alexander Peter Maxwell
- Centre for Public Health, Queen’s University of Belfast, Belfast BT12 6BA, UK
- Regional Nephrology Unit, Belfast City Hospital, Belfast BT9 7AB, UK
| | - Amy Jayne McKnight
- Centre for Public Health, Queen’s University of Belfast, Belfast BT12 6BA, UK
| |
Collapse
|
263
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation 2023; 147:e93-e621. [PMID: 36695182 DOI: 10.1161/cir.0000000000001123] [Citation(s) in RCA: 2306] [Impact Index Per Article: 1153.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2023 Statistical Update is the product of a full year's worth of effort in 2022 by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. The American Heart Association strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional COVID-19 (coronavirus disease 2019) publications, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
264
|
Sugawara Y, Hirakawa Y, Nagasu H, Narita A, Katayama A, Wada J, Shimizu M, Wada T, Kitamura H, Nakano T, Yokoi H, Yanagita M, Goto S, Narita I, Koshiba S, Tamiya G, Nangaku M, Yamamoto M, Kashihara N. Genome-wide association study of the risk of chronic kidney disease and kidney-related traits in the Japanese population: J-Kidney-Biobank. J Hum Genet 2023; 68:55-64. [PMID: 36404353 DOI: 10.1038/s10038-022-01094-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/22/2022]
Abstract
Chronic kidney disease (CKD) is a syndrome characterized by a gradual loss of kidney function with decreased estimated glomerular filtration rate (eGFR), which may be accompanied by an increase in the urine albumin-to-creatinine ratio (UACR). Although trans-ethnic genome-wide association studies (GWASs) have been conducted for kidney-related traits, there have been few analyses in the Japanese population, especially for the UACR trait. In this study, we conducted a GWAS to identify loci related to multiple kidney-related traits in Japanese individuals. First, to detect loci associated with CKD, eGFR, and UACR, we performed separate GWASs with the following two datasets: 475 cases of CKD diagnosed at seven university hospitals and 3471 healthy subjects (dataset 1) and 3664 cases of CKD-suspected individuals with eGFR <60 ml/min/1.73 m2 or urinary protein ≥ 1+ and 5952 healthy subjects (dataset 2). Second, we performed a meta-analysis between these two datasets and detected the following associated loci: 10 loci for CKD, 9 loci for eGFR, and 22 loci for UACR. Among the loci detected, 22 have never been reported previously. Half of the significant loci for CKD were shared with those for eGFR, whereas most of the loci associated with UACR were different from those associated with CKD or eGFR. The GWAS of the Japanese population identified novel genetic components that were not previously detected. The results also suggest that the group primarily characterized by increased UACR possessed genetically different features from the group characterized by decreased eGFR.
Collapse
Affiliation(s)
- Yuka Sugawara
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Yosuke Hirakawa
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
| | - Akihiro Katayama
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University, Okayama, Japan
| | - Miho Shimizu
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Ishikawa, Japan
| | - Takashi Wada
- Department of Nephrology and Laboratory Medicine, Kanazawa University, Ishikawa, Japan
| | - Hiromasa Kitamura
- Department of Nephrology, Hypertension & Strokology, Kyushu University, Fukuoka, Japan
| | - Toshiaki Nakano
- Department of Nephrology, Hypertension & Strokology, Kyushu University, Fukuoka, Japan
| | - Hideki Yokoi
- Department of Nephrology, Kyoto University, Kyoto, Japan
| | | | - Shin Goto
- Division of Clinical Nephrology and Rheumatology, Niigata University, Niigata, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University, Niigata, Japan
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan.,The Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan.,Graduate School of Medicine, Tohoku University, Sendai, Japan.,Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo, Tokyo, Japan
| | - Masayuki Yamamoto
- Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan.,Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Okayama, Japan.
| |
Collapse
|
265
|
Lin Y, Yang Y, Fu T, Lin L, Zhang X, Guo Q, Chen Z, Liao B, Huang J. Impairment of kidney function and kidney cancer: A bidirectional Mendelian randomization study. Cancer Med 2023; 12:3610-3622. [PMID: 36069056 PMCID: PMC9939186 DOI: 10.1002/cam4.5204] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/21/2022] [Accepted: 08/23/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Many observational epidemiology studies discovered that kidney cancer and impaired kidney function have a bidirectional relationship. However, it remains unclear whether these two kinds of traits are causally linked. In this study, we aimed to investigate the bidirectional causal relation between kidney cancer and kidney function biomarkers (creatinine-based estimated glomerular filtration rate (eGFRcrea), cystatin C-based estimated glomerular filtration rate (eGFRcys), blood urea nitrogen (BUN), serum urate, and urinary albumin-to-creatinine ratio (UACR)). METHODS For both directions, single-nucleotide polymorphisms (SNPs), as genetic instruments, for the five kidney function traits were selected from up to 1,004,040 individuals, and SNPs for kidney cancer were from 408,786 participants(1338 cases). In the main analysis, we applied two state-of-the-art MR methods, namely, contamination mixture and Robust Adjusted Profile Score to downweight the effect of weak instrument bias, pleiotropy, and extreme outliers. We additionally conducted traditional MR analyses as sensitivity analyses. Summary-level data of European ancestry were extracted from UK Biobank, Chronic Kidney Disease Genetics Consortium, and Kaiser Permanente. RESULTS Based on 99 SNPs, we found that the eGFRcrea had a significant negative causal effect on the risk of kidney cancer (OR = 0.007, 95% CI:2.6 × 10-4 -0.569, p = 0.041). After adjusting for body composition or diabetes, urate had a significant negative causal effect on kidney cancer (OR <1, p < 0.05). For UACR, it showed a strong causal effect on kidney cancer, after adjusting for body composition (OR = 14.503, 95% CI: 2.546-96.001, p = 0.032). Due to lacking significant signals and effect power for the reverse MR, further investigations are warranted. CONCLUSIONS Our study suggested a potential causal effect of damaged kidney function on kidney cancer. EGFRcrea and UACR might be causally associated with kidney cancer, especially when patients were comorbid with obesity or diabetes. We called for larger sample-size studies to further unravel the underlying causal relationship and the exact mechanism.
Collapse
Affiliation(s)
- Yifei Lin
- West China Hospital, Sichuan UniversityChengduPeople's Republic of China
- Program in Genetic Epidemiology and Statistical Genetics, Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Yong Yang
- Medical Device Regulatory Research and Evaluation Centre, West China HospitalSichuan UniversityChengduPeople's Republic of China
| | - Tingting Fu
- Medical Device Regulatory Research and Evaluation Centre, West China HospitalSichuan UniversityChengduPeople's Republic of China
| | - Ling Lin
- Medical Device Regulatory Research and Evaluation Centre, West China HospitalSichuan UniversityChengduPeople's Republic of China
| | - Xingming Zhang
- Department of UrologyInstitute of Urology, West China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Qiong Guo
- Medical Device Regulatory Research and Evaluation Centre, West China HospitalSichuan UniversityChengduPeople's Republic of China
| | - Zhenglong Chen
- Medical Device Regulatory Research and Evaluation Centre, West China HospitalSichuan UniversityChengduPeople's Republic of China
| | - Banghua Liao
- Department of UrologyInstitute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan UniversityChengduPeople's Republic of China
| | - Jin Huang
- Medical Device Regulatory Research and Evaluation Centre, West China HospitalSichuan UniversityChengduPeople's Republic of China
| |
Collapse
|
266
|
Yokoyama S, Nakagawa J, Kudo M, Aiuchi N, Seito T, Isida M, Mikami T, Ihara K, Nakaji S, Niioka T. Impact of solute carrier transporter gene polymorphisms on serum creatinine concentrations in healthy volunteers. Pharmacol Res Perspect 2023; 11:e01048. [PMID: 36594679 PMCID: PMC9809111 DOI: 10.1002/prp2.1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/11/2022] [Indexed: 01/04/2023] Open
Abstract
In this study, we investigated the impact of single nucleotide polymorphisms in solute carrier (SLC) transporters, that is, SLC22A7 c.1586 + 206A > G, SLC22A2 c.808G > T, SLC22A3 c.1233G > A, SLC47A1 c.922-158G > A, and SLC47A2 c.-130G > A, on serum creatinine (SCr) concentrations. This cross-sectional study included residents who participated as volunteers in a health promotion study. Lifestyle data, blood chemical analysis data, and SLC gene polymorphism information were collected from each participant. Univariate analyses were carried out to determine differences between groups and correlations in SCr. Stepwise multiple regression analysis was performed to confirm the independence of factors that were significantly different in the univariate analyses. In multiple regression analyses, muscle mass, serum cystatin C concentrations, body fat percentage, serum albumin concentrations, and SLC47A2 c.-130G/G had the highest contribution to SCr concentrations, in that order (standardized regression coefficients = .505, .332, -.234, .123, and .084, respectively). The final model explained 72.2% of the variability in SCr concentrations. The SLC47A2 c.-130G > A polymorphism may affect creatinine dynamics in the proximal tubules. Further studies are needed to determine the effects of SLC transporter gene polymorphisms on SCr concentrations in patients with various diseases in real-world clinical settings.
Collapse
Affiliation(s)
- Satoshi Yokoyama
- Department of Pharmaceutical ScienceHirosaki University Graduate School of MedicineHirosakiAomoriJapan
- Department of PharmacyHirosaki Central HospitalHirosakiAomoriJapan
| | - Junichi Nakagawa
- Department of PharmacyHirosaki University HospitalHirosakiAomoriJapan
| | - Masakiyo Kudo
- Department of PharmacyHirosaki University HospitalHirosakiAomoriJapan
| | - Naoya Aiuchi
- Department of PharmacyHirosaki University HospitalHirosakiAomoriJapan
| | - Tatsuya Seito
- Department of PharmacyHirosaki Central HospitalHirosakiAomoriJapan
| | - Mizuri Isida
- Department of Innovation Center for Health PromotionHirosaki University Graduate School of MedicineHirosakiAomoriJapan
| | - Tatsuya Mikami
- Department of Innovation Center for Health PromotionHirosaki University Graduate School of MedicineHirosakiAomoriJapan
| | - Kazushige Ihara
- Department of Social MedicineHirosaki University Graduate School of MedicineHirosakiAomoriJapan
| | - Shigeyuki Nakaji
- Department of Social MedicineHirosaki University Graduate School of MedicineHirosakiAomoriJapan
| | - Takenori Niioka
- Department of Pharmaceutical ScienceHirosaki University Graduate School of MedicineHirosakiAomoriJapan
- Department of PharmacyHirosaki University HospitalHirosakiAomoriJapan
| |
Collapse
|
267
|
Dellepiane S, Paranjpe I, Rajagopal M, Kamat S, O’Hagan R, Gulamali F, Rein JL, Charney AW, Do R, Coca S, Glicksberg BS, Nadkarni GN. Cannabis Use and CKD: Epidemiological Associations and Mendelian Randomization. Kidney Med 2023; 5:100582. [PMID: 36712313 PMCID: PMC9879977 DOI: 10.1016/j.xkme.2022.100582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Rationale & Objective The association between cannabis use and chronic kidney disease (CKD) is controversial. We aimed to assess association of CKD with cannabis use in a large cohort study and then assess causality using Mendelian randomization with a genome-wide association study (GWAS). Study Design Retrospective cohort study and genome-wide association study. Setting & Participants The retrospective study was conducted on the All of Us cohort (N=223,354). Genetic instruments for cannabis use disorder were identified from 3 GWAS: the Psychiatric Genomics Consortium Substance Use Disorders, iPSYCH, and deCODE (N=384,032). Association between genetic instruments and CKD was investigated in the CKDGen GWAS (N > 1.2 million). Exposure Cannabis consumption. Outcomes CKD outcomes included: cystatin-C and creatinine-based kidney function, proteinuria, and blood urea nitrogen. Analytical Approach We conducted association analyses to test for frequency of cannabis use and CKD. To evaluate causality, we performed a 2-sample Mendelian randomization. Results In the retrospective study, compared to former users, less than monthly (OR, 1.01; 95% CI, 0.87-1.18; P = 0.87) and monthly cannabis users (OR, 1.15; 95% CI, 0.86-1.52; P = 0.33) did not have higher CKD odds. Conversely, weekly (OR, 1.28; 95% CI, 1.01-1.60; P = 0.04) and daily use (OR, 1.25; 95% CI, 1.04-1.50; P = 0.02) was significantly associated with CKD, adjusted for multiple confounders. In Mendelian randomization, genetic liability to cannabis use disorder was not associated with increased odds for CKD (OR, 1.00; 95% CI, 0.99-1.01; P = 0.96). These results were robust across different Mendelian randomization techniques and multiple kidney traits. Limitations Likely underreporting of cannabis use. In Mendelian randomization, genetic instruments were identified in the GWAS that included individuals primarily of European ancestry. Conclusions Despite the epidemiological association between cannabis use and CKD, there was no evidence of a causal effect, indicating confounding in observational studies.
Collapse
Affiliation(s)
- Sergio Dellepiane
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ishan Paranjpe
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Madhumitha Rajagopal
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
| | - Samir Kamat
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ross O’Hagan
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Faris Gulamali
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Joshua L. Rein
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Alexander W. Charney
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ron Do
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Steven Coca
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Benjamin S. Glicksberg
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Girish N. Nadkarni
- Mount Sinai Clinical Intelligence Center (MSCIC), Icahn School of Medicine at Mount Sinai, New York, NY
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine, Division of Data Driven and Precision Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
268
|
Abstract
Hundreds of different genetic causes of chronic kidney disease are now recognized, and while individually rare, taken together they are significant contributors to both adult and pediatric diseases. Traditional genetics approaches relied heavily on the identification of large families with multiple affected members and have been fundamental to the identification of genetic kidney diseases. With the increased utilization of massively parallel sequencing and improvements to genotype imputation, we can analyze rare variants in large cohorts of unrelated individuals, leading to personalized care for patients and significant research advancements. This review evaluates the contribution of rare disorders to patient care and the study of genetic kidney diseases and highlights key advancements that utilize new techniques to improve our ability to identify new gene-disease associations.
Collapse
Affiliation(s)
- Mark D Elliott
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA;
- Center for Precision Medicine and Genomics, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Institute for Genomic Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Hila Milo Rasouly
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA;
- Center for Precision Medicine and Genomics, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA;
- Center for Precision Medicine and Genomics, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
- Institute for Genomic Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
269
|
Cai M, Wei J, Zhang S, Liu W, Wang L, Qian Z, Lin H, Liu E, McMillin SE, Cao Y, Yin P. Short-term air pollution exposure associated with death from kidney diseases: a nationwide time-stratified case-crossover study in China from 2015 to 2019. BMC Med 2023; 21:32. [PMID: 36694165 PMCID: PMC9875429 DOI: 10.1186/s12916-023-02734-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Long-term exposure to air pollution has been associated with the onset and progression of kidney diseases, but the association between short-term exposure to air pollution and mortality of kidney diseases has not yet been reported. METHODS A nationally representative sample of 101,919 deaths from kidney diseases was collected from the Chinese Center for Disease Control and Prevention from 2015 to 2019. A time-stratified case-crossover study was applied to determine the associations. Satellite-based estimates of air pollution were assigned to each case and control day using a bilinear interpolation approach and geo-coded residential addresses. Conditional logistic regression models were constructed to estimate the associations adjusting for nonlinear splines of temperature and relative humidity. RESULTS Each 10 µg/m3 increment in lag 0-1 mean concentrations of air pollutants was associated with a percent increase in death from kidney disease: 1.33% (95% confidence interval [CI]: 0.57% to 2.1%) for PM1, 0.49% (95% CI: 0.10% to 0.88%) for PM2.5, 0.32% (95% CI: 0.08% to 0.57%) for PM10, 1.26% (95% CI: 0.29% to 2.24%) for NO2, and 2.9% (95% CI: 1.68% to 4.15%) for SO2. CONCLUSIONS: Our study suggests that short-term exposure to ambient PM1, PM2.5, PM10, NO2, and SO2 might be important environmental risk factors for death due to kidney diseases in China.
Collapse
Affiliation(s)
- Miao Cai
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Jing Wei
- Department of Atmospheric and Oceanic Science, Earth System Science Interdisciplinary Center, University of Maryland, College Park, MD, 20740, USA
| | - Shiyu Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Wei Liu
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Lijun Wang
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 100050, China
| | - Zhengmin Qian
- Department of Epidemiology and Biostatistics, College for Public Health and Social Justice, Saint Louis University, St. Louis, 63103, USA
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Echu Liu
- Department of Health Management and Policy, College for Public Health and Social Justice, Saint Louis University, St. Louis, MO, 63103, USA
| | - Stephen Edward McMillin
- School of Social Work, College for Public Health and Social Justice, Saint Louis University, St. Louis, MO, 63103, USA
| | - Yu Cao
- Information Center, Chinese Center for Disease Control and Prevention, Beijing, 102206, China.
| | - Peng Yin
- National Center for Chronic and Noncommunicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 100050, China.
| |
Collapse
|
270
|
Lingaas F, Tengvall K, Jansen JH, Pelander L, Hurst MH, Meuwissen T, Karlsson Å, Meadows JRS, Sundström E, Thoresen SI, Arnet EF, Guttersrud OA, Kierczak M, Hytönen MK, Lohi H, Hedhammar Å, Lindblad-Toh K, Wang C. Bayesian mixed model analysis uncovered 21 risk loci for chronic kidney disease in boxer dogs. PLoS Genet 2023; 19:e1010599. [PMID: 36693108 PMCID: PMC9897549 DOI: 10.1371/journal.pgen.1010599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/03/2023] [Accepted: 01/04/2023] [Indexed: 01/25/2023] Open
Abstract
Chronic kidney disease (CKD) affects 10% of the human population, with only a small fraction genetically defined. CKD is also common in dogs and has been diagnosed in nearly all breeds, but its genetic basis remains unclear. Here, we performed a Bayesian mixed model genome-wide association analysis for canine CKD in a boxer population of 117 canine cases and 137 controls, and identified 21 genetic regions associated with the disease. At the top markers from each CKD region, the cases carried an average of 20.2 risk alleles, significantly higher than controls (15.6 risk alleles). An ANOVA test showed that the 21 CKD regions together explained 57% of CKD phenotypic variation in the population. Based on whole genome sequencing data of 20 boxers, we identified 5,206 variants in LD with the top 50 BayesR markers. Following comparative analysis with human regulatory data, 17 putative regulatory variants were identified and tested with electrophoretic mobility shift assays. In total four variants, three intronic variants from the MAGI2 and GALNT18 genes, and one variant in an intergenic region on chr28, showed alternative binding ability for the risk and protective alleles in kidney cell lines. Many genes from the 21 CKD regions, RELN, MAGI2, FGFR2 and others, have been implicated in human kidney development or disease. The results from this study provide new information that may enlighten the etiology of CKD in both dogs and humans.
Collapse
Affiliation(s)
- Frode Lingaas
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Katarina Tengvall
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Johan Høgset Jansen
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Lena Pelander
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Theo Meuwissen
- Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway
| | - Åsa Karlsson
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jennifer R. S. Meadows
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Elisabeth Sundström
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Stein Istre Thoresen
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Ellen Frøysadal Arnet
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Ole Albert Guttersrud
- Faculty of Veterinary Medicine, Department of Preclinical Sciences and Pathology, Norwegian University of Life Sciences, Ås, Norway
| | - Marcin Kierczak
- Department of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Marjo K. Hytönen
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Hannes Lohi
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Center, Helsinki, Finland
| | - Åke Hedhammar
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
- * E-mail: (KL-T); (CW)
| | - Chao Wang
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- * E-mail: (KL-T); (CW)
| |
Collapse
|
271
|
Liu X, Ou YN, Ma YH, Huang LY, Zhang W, Tan L. Renal function and neurodegenerative diseases : a two-sample Mendelian randomization study. Neurol Res 2023; 45:456-464. [PMID: 36692889 DOI: 10.1080/01616412.2022.2158640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Observational studies showed renal function had associations with Alzheimer's disease (AD), Parkinson's disease (PD), Lewy body dementia (LBD) and multiple sclerosis (MS). However, it is unknown whether these associations are causal. METHODS We use a two-sample Mendelian randomization (MR) analysis to investigate causal relationships between renal function and 6 neurodegenerative diseases (NDDs): AD (including familial AD), PD, LBD, frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS) and MS. Blood urea nitrogen (BUN), chronic kidney disease (CKD) and estimated glomerular filtration rate (eGFR) were used to measure renal function. The inverse-variance weighted (IVW) was the predominant estimation method. The results were further validated using sensitivity analysis (i.e. MR Egger regression, Cochran Q statistic of IVW, and leave-one-out method). RESULTS There was no indication of any causative relationship of BUN, CKD, or eGFR with AD, familial AD, PD, LBD, FTD and ALS (all P values >0.05). The IVW analysis demonstrated a causal relationship between eGFR and MS [odds ratio (OR), 4.89; 95% confidence interval (CI), 1.43 to 16.71; P = 0.01] that was not verified in the MR-Egger and weighted median (all P values >0.05). However, no causal association of MS with BUN (OR, 0.91; 95% CI, 0.40-2.07; P = 0.82) and CKD (OR,1.04; 95% CI, 0.88-1.23; P = 0.66) was found. There was no single SNP that affects the overall trend. CONCLUSIONS Our study showed that reduced eGFR was related to MS. The value of this study is that it provides a direction for further research on the relationship between reduced eGFR and MS.
Collapse
Affiliation(s)
- Xue Liu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Hui Ma
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| |
Collapse
|
272
|
Bächle H, Sekula P, Schlosser P, Steinbrenner I, Cheng Y, Kotsis F, Meiselbach H, Stockmann H, Schönherr S, Eckardt KU, Devuyst O, Scherberich J, Köttgen A, Schultheiss UT. Uromodulin and its association with urinary metabolites: the German Chronic Kidney Disease Study. Nephrol Dial Transplant 2023; 38:70-79. [PMID: 35612992 DOI: 10.1093/ndt/gfac187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND The progression of chronic kidney disease (CKD), a global public health burden, is accompanied by a declining number of functional nephrons. Estimation of remaining nephron mass may improve assessment of CKD progression. Uromodulin has been suggested as a marker of tubular mass. We aimed to identify metabolites associated with uromodulin concentrations in urine and serum to characterize pathophysiologic alterations of metabolic pathways to generate new hypotheses regarding CKD pathophysiology. METHODS We measured urinary and serum uromodulin levels (uUMOD, sUMOD) and 607 urinary metabolites and performed cross-sectional analyses within the German Chronic Kidney Disease study (N = 4628), a prospective observational study. Urinary metabolites significantly associated with uUMOD and sUMOD were used to build weighted metabolite scores for urine (uMS) and serum uromodulin (sMS) and evaluated for time to adverse kidney events over 6.5 years. RESULTS Metabolites cross-sectionally associated with uromodulin included amino acids of the tryptophan metabolism, lipids and nucleotides. Higher levels of the sMS [hazard ratio (HR) = 0.73 (95% confidence interval 0.64; 0.82), P = 7.45e-07] and sUMOD [HR = 0.74 (95% confidence interval 0.63; 0.87), P = 2.32e-04] were associated with a lower risk of adverse kidney events over time, whereas uUMOD and uMS showed the same direction of association but were not significant. CONCLUSIONS We identified urinary metabolites associated with urinary and serum uromodulin. The sUMOD and the sMS were associated with lower risk of adverse kidney events among CKD patients. Higher levels of sUMOD and sMS may reflect a higher number of functional nephrons and therefore a reduced risk of adverse kidney outcomes.
Collapse
Affiliation(s)
- Helena Bächle
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Fruzsina Kotsis
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.,Department of Medicine IV-Nephrology and Primary Care, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Heike Meiselbach
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Helena Stockmann
- Department of Nephrology and Medical Intensive Care, Charité, University-Medicine, Berlin, Germany
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Nephrology and Medical Intensive Care, Charité, University-Medicine, Berlin, Germany
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Jürgen Scherberich
- Klinikum München-Harlaching, Nephrology & Clinical Immunology, Teaching Hospital of the Ludwig-Maximilians-University München, Munich, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Ulla T Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.,Department of Medicine IV-Nephrology and Primary Care, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
273
|
Schmidt AF, Joshi R, Gordillo-Marañón M, Drenos F, Charoen P, Giambartolomei C, Bis JC, Gaunt TR, Hughes AD, Lawlor DA, Wong A, Price JF, Chaturvedi N, Wannamethee G, Franceschini N, Kivimaki M, Hingorani AD, Finan C. Biomedical consequences of elevated cholesterol-containing lipoproteins and apolipoproteins on cardiovascular and non-cardiovascular outcomes. COMMUNICATIONS MEDICINE 2023; 3:9. [PMID: 36670186 PMCID: PMC9859819 DOI: 10.1038/s43856-022-00234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Higher concentrations of cholesterol-containing low-density lipoprotein (LDL-C) increase the risk of cardiovascular disease (CVD). The association of LDL-C with non-CVD traits remains unclear, as are the possible independent contributions of other cholesterol-containing lipoproteins and apolipoproteins. METHODS Nuclear magnetic resonance spectroscopy was used to measure the cholesterol content of high density (HDL-C), very low-density (VLDL-C), intermediate-density (IDL-C), as well as low-density lipoprotein fractions, the apolipoproteins Apo-A1 and Apo-B, as well as total triglycerides (TG), remnant-cholesterol (Rem-Chol) and total cholesterol (TC). The causal effects of these exposures were assessed against 33 outcomes using univariable and multivariable Mendelian randomization (MR). RESULTS The majority of cholesterol containing lipoproteins and apolipoproteins affect coronary heart disease (CHD), carotid intima-media thickness, carotid plaque, C-reactive protein (CRP) and blood pressure. Multivariable MR indicated that many of these effects act independently of HDL-C, LDL-C and TG, the most frequently measured lipid fractions. Higher concentrations of TG, VLDL-C, Rem-Chol and Apo-B increased heart failure (HF) risk; often independently of LDL-C, HDL-C or TG. Finally, a subset of these exposures associated with non-CVD traits such as Alzheimer's disease (AD: HDL-C, LDL-C, IDL-C, Apo-B), type 2 diabetes (T2DM: VLDL-C, IDL-C, LDL-C), and inflammatory bowel disease (IBD: LDL-C, IDL-C). CONCLUSIONS The cholesterol content of a wide range of lipoprotein and apolipoproteins associate with measures of atherosclerosis, blood pressure, CRP, and CHD, with a subset affecting HF, T2DM, AD and IBD risk. Many of the observed effects appear to act independently of LDL-C, HDL-C, and TG, supporting the targeting of lipid fractions beyond LDL-C for disease prevention.
Collapse
Affiliation(s)
- Amand F Schmidt
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK.
- UCL BHF Research Accelerator Centre, London, UK.
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | - Roshni Joshi
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Maria Gordillo-Marañón
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Fotios Drenos
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, UK
| | - Pimphen Charoen
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- Department of Tropical Hygiene, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Bangkok, 10400, Thailand
| | - Claudia Giambartolomei
- Istituto Italiano di Tecnologia, Non-coding RNAs and RNA-based Therapeutics, Via Morego, 30, 16163, Genova, Italy
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol National Health Service Foundation Trust and University of Bristol, Bristol, UK
| | - Alun D Hughes
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol National Health Service Foundation Trust and University of Bristol, Bristol, UK
- Population Health, Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrew Wong
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | | | - Nishi Chaturvedi
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing, University College London, London, UK
| | - Goya Wannamethee
- Primary Care and Population Health, University College London, London, UK
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Mika Kivimaki
- Department of Mental Health of Older People, Division of Brain Sciences, University College London, London, UK
| | - Aroon D Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
| | - Chris Finan
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, UK
- UCL BHF Research Accelerator Centre, London, UK
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
274
|
Ong KL, Marklund M, Huang L, Rye KA, Hui N, Pan XF, Rebholz CM, Kim H, Steffen LM, van Westing AC, Geleijnse JM, Hoogeveen EK, Chen YY, Chien KL, Fretts AM, Lemaitre RN, Imamura F, Forouhi NG, Wareham NJ, Birukov A, Jäger S, Kuxhaus O, Schulze MB, de Mello VD, Tuomilehto J, Uusitupa M, Lindström J, Tintle N, Harris WS, Yamasaki K, Hirakawa Y, Ninomiya T, Tanaka T, Ferrucci L, Bandinelli S, Virtanen JK, Voutilainen A, Jayasena T, Thalamuthu A, Poljak A, Bustamante S, Sachdev PS, Senn MK, Rich SS, Tsai MY, Wood AC, Laakso M, Lankinen M, Yang X, Sun L, Li H, Lin X, Nowak C, Ärnlöv J, Risérus U, Lind L, Le Goff M, Samieri C, Helmer C, Qian F, Micha R, Tin A, Köttgen A, de Boer IH, Siscovick DS, Mozaffarian D, Wu JH. Association of omega 3 polyunsaturated fatty acids with incident chronic kidney disease: pooled analysis of 19 cohorts. BMJ 2023; 380:e072909. [PMID: 36653033 PMCID: PMC9846698 DOI: 10.1136/bmj-2022-072909] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To assess the prospective associations of circulating levels of omega 3 polyunsaturated fatty acid (n-3 PUFA) biomarkers (including plant derived α linolenic acid and seafood derived eicosapentaenoic acid, docosapentaenoic acid, and docosahexaenoic acid) with incident chronic kidney disease (CKD). DESIGN Pooled analysis. DATA SOURCES A consortium of 19 studies from 12 countries identified up to May 2020. STUDY SELECTION Prospective studies with measured n-3 PUFA biomarker data and incident CKD based on estimated glomerular filtration rate. DATA EXTRACTION AND SYNTHESIS Each participating cohort conducted de novo analysis with prespecified and consistent exposures, outcomes, covariates, and models. The results were pooled across cohorts using inverse variance weighted meta-analysis. MAIN OUTCOME MEASURES Primary outcome of incident CKD was defined as new onset estimated glomerular filtration rate <60 mL/min/1.73 m2. In a sensitivity analysis, incident CKD was defined as new onset estimated glomerular filtration rate <60 mL/min/1.73 m2 and <75% of baseline rate. RESULTS 25 570 participants were included in the primary outcome analysis and 4944 (19.3%) developed incident CKD during follow-up (weighted median 11.3 years). In multivariable adjusted models, higher levels of total seafood n-3 PUFAs were associated with a lower incident CKD risk (relative risk per interquintile range 0.92, 95% confidence interval 0.86 to 0.98; P=0.009, I2=9.9%). In categorical analyses, participants with total seafood n-3 PUFA level in the highest fifth had 13% lower risk of incident CKD compared with those in the lowest fifth (0.87, 0.80 to 0.96; P=0.005, I2=0.0%). Plant derived α linolenic acid levels were not associated with incident CKD (1.00, 0.94 to 1.06; P=0.94, I2=5.8%). Similar results were obtained in the sensitivity analysis. The association appeared consistent across subgroups by age (≥60 v <60 years), estimated glomerular filtration rate (60-89 v ≥90 mL/min/1.73 m2), hypertension, diabetes, and coronary heart disease at baseline. CONCLUSIONS Higher seafood derived n-3 PUFA levels were associated with lower risk of incident CKD, although this association was not found for plant derived n-3 PUFAs. These results support a favourable role for seafood derived n-3 PUFAs in preventing CKD.
Collapse
Affiliation(s)
- Kwok Leung Ong
- Lipid Research Group, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matti Marklund
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- The Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Liping Huang
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Kerry-Anne Rye
- Lipid Research Group, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Nicholas Hui
- Lipid Research Group, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Xiong-Fei Pan
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan, China
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hyunju Kim
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lyn M Steffen
- University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Anniek C van Westing
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Johanna M Geleijnse
- Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Ellen K Hoogeveen
- Department of Nephrology, Jeroen Bosch Hospital, Den Bosch, The Netherlands
- Institute of Epidemiology and Preventive Medicine College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Kuo-Liong Chien
- Institute of Epidemiology and Preventive Medicine College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Amanda M Fretts
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | | | - Fumiaki Imamura
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Nita G Forouhi
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Nicholas J Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Anna Birukov
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Susanne Jäger
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Olga Kuxhaus
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
- Institute of Nutritional Science, University of Potsdam, Potsdam, Germany
| | - Vanessa Derenji de Mello
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jaakko Tuomilehto
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Matti Uusitupa
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Jaana Lindström
- Population Health Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Nathan Tintle
- The Fatty Acid Research Institute, Sioux Falls, SD, USA
- Department of Population Health Nursing Science, College of Nursing, University of Illinois-Chicago, Chicago, IL, USA
| | - William S Harris
- The Fatty Acid Research Institute, Sioux Falls, SD, USA
- Department of Internal Medicine, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, USA
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Yamasaki
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoichiro Hirakawa
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiharu Ninomiya
- Department of Epidemiology and Public Health, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, National Institute of Health, Baltimore, MD, USA
| | | | - Jyrki K Virtanen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ari Voutilainen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | - Anne Poljak
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Sonia Bustamante
- Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, Australia
| | | | - Mackenzie K Senn
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Stephen S Rich
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Alexis C Wood
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Maria Lankinen
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Xiaowei Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liang Sun
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huaixing Li
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Christoph Nowak
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Sweden
| | - Johan Ärnlöv
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Sweden
| | - Ulf Risérus
- Department of Public Health and Caring Sciences, Clinical Nutrition and Metabolism, Uppsala University, Uppsala, Sweden
| | - Lars Lind
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Mélanie Le Goff
- Bordeaux Population Health Research Centre, INSERM, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Cécilia Samieri
- Bordeaux Population Health Research Centre, INSERM, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Catherine Helmer
- Bordeaux Population Health Research Centre, INSERM, UMR 1219, University of Bordeaux, Bordeaux, France
| | - Frank Qian
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Renata Micha
- Department of Food Science and Nutrition, University of Thessaly, Karditsa, Greece
- The Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Adrienne Tin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Institute of Genetic Epidemiology, Department of Data Driven Medicine, Faculty of Medicine and Medical Centre, University of Freiburg, Freiburg, Germany
| | - Ian H de Boer
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, WA, USA
- Kidney Research Institute, University of Washington, Seattle, WA, USA
- Puget Sound VA Healthcare System, Seattle, WA, USA
| | | | - Dariush Mozaffarian
- The Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Jason Hy Wu
- The George Institute for Global Health, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- School of Population Health, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
275
|
Zhou LY, Sofer T, Horimoto AR, Talavera GA, Lash JP, Cai J, Franceschini N. Polygenic risk scores and kidney traits in the Hispanic/Latino population: The Hispanic Community Health Study/Study of Latinos. HGG ADVANCES 2023; 4:100177. [PMID: 36741942 PMCID: PMC9894917 DOI: 10.1016/j.xhgg.2023.100177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
Estimated glomerular filtration rate (eGFR) is used to evaluate kidney function and determine the presence of chronic kidney disease (CKD), a highly prevalent disease in the US1 , 2 , 3 that varies among subgroups of Hispanic/Latino individuals.4 , 5 The polygenic risk score (PRS) is a popular method that uses large genome-wide association studies (GWASs) to provide a strong estimate of disease risk.7 However, due to the limited availability of summary statistics from GWAS meta-analyses based on Hispanic/Latino populations, PRSs can only be computed using different ancestry GWASs. The performance of eGFR PRSs derived from other GWAS reference populations for Hispanic/Latino population has not been examined. We compared PRS constructions for eGFR prediction in Hispanic/Latino individuals using GWAS-significant variants, clumping and thresholding (C&T),8 and PRS-CS,22 as well as a combination of PRSs calculated with different reference GWAS meta-analyses from European and multi-ethnic studies in Hispanic/Latino individuals from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL). All eGFR PRSs were highly associated with eGFR (p < 1E-20). Additionally, eGFR PRSs were significantly associated with lower risk of prevalent CKD at visit 1 or 2 and incident CKD at visit 2, with the combined PRSs having the best performance. These PRS findings were replicated in an additional dataset of Hispanic/Latino individuals using data from the Women's Health Initiative SNP Health Association Resource (WHI-SHARe).17.
Collapse
Affiliation(s)
- Laura Y. Zhou
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA,Corresponding author
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Gregory A. Talavera
- Graduate School of Public Health, San Diego State University, San Diego, CA, USA
| | - James P. Lash
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
276
|
Li N, Wang Y, Wei P, Min Y, Yu M, Zhou G, Yuan G, Sun J, Dai H, Zhou E, He W, Sheng M, Gao K, Zheng M, Sun W, Zhou D, Zhang L. Causal Effects of Specific Gut Microbiota on Chronic Kidney Diseases and Renal Function-A Two-Sample Mendelian Randomization Study. Nutrients 2023; 15:nu15020360. [PMID: 36678231 PMCID: PMC9863044 DOI: 10.3390/nu15020360] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Targeting the gut microbiota may become a new therapeutic to prevent and delay the progression of chronic kidney disease (CKD). Nonetheless, the causal relationship between specific intestinal flora and CKD is still unclear. MATERIALS AND METHOD To identify genetically predicted microbiota, we used summary data from genome-wide association studies on gut microbiota in 18340 participants from 24 cohorts. Furthermore, we genetically predicted the causal relationship between 211 gut microbiotas and six phenotypes (outcomes) (CKD, estimated glomerular filtration rate (eGFR), urine albumin to creatinine ratio (UACR), dialysis, rapid progress to CKD, and rapid decline of eGFR). Four Mendelian randomization (MR) methods, including inverse variance weighted (IVW), MR-Egger, weighted median, and weighted mode were used to investigate the casual relationship between gut microbiotas and various outcomes. The result of IVW was deemed as the primary result. Then, Cochrane's Q test, MR-Egger, and MR-PRESSO Global test were used to detect heterogeneity and pleiotropy. The leave-one method was used for testing the stability of MR results and Bonferroni-corrected was used to test the strength of the causal relationship between exposure and outcome. RESULTS Through the MR analysis of 211 microbiotas and six clinical phenotypes, a total of 36 intestinal microflora were found to be associated with various outcomes. Among them, Class Bacteroidia (=-0.005, 95% CI: -0.001 to -0.008, p = 0.002) has a strong causality with lower eGFR after the Bonferroni-corrected test, whereas phylum Actinobacteria (OR = 1.0009, 95%CI: 1.0003-1.0015, p = 0.0024) has a strong causal relationship with dialysis. The Cochrane's Q test reveals that there is no significant heterogeneity between various single nucleotide polymorphisms. In addition, no significant level of pleiotropy was found according to MR-Egger and MR-PRESSO Global tests. CONCLUSIONS Through the two-sample MR analysis, we identified the specific intestinal flora that has a causal relationship with the incidence and progression of CKD at the level of gene prediction, which may provide helpful biomarkers for early disease diagnosis and potential therapeutic targets for CKD.
Collapse
Affiliation(s)
- Ning Li
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Yi Wang
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Ping Wei
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Yu Min
- Department of Biotherapy and National Clinical Research Center, Sichuan University, Chengdu 610041, China
| | - Manshu Yu
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Guowei Zhou
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Gui Yuan
- Division of Nephrology, Department of Medicine, University of Connecticut, School of Medicine, Farmington, CT 06030, USA
| | - Jinyi Sun
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Huibo Dai
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Enchao Zhou
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Weiming He
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Meixiao Sheng
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Kun Gao
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Min Zheng
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Wei Sun
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut, School of Medicine, Farmington, CT 06030, USA
- Correspondence: (D.Z.); (L.Z.)
| | - Lu Zhang
- Division of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
- Correspondence: (D.Z.); (L.Z.)
| |
Collapse
|
277
|
Durand A, Winkler CA, Vince N, Douillard V, Geffard E, Binns-Roemer E, Ng DK, Gourraud PA, Reidy K, Warady B, Furth S, Kopp JB, Kaskel FJ, Limou S. Identification of Novel Genetic Risk Factors for Focal Segmental Glomerulosclerosis in Children: Results From the Chronic Kidney Disease in Children (CKiD) Cohort. Am J Kidney Dis 2023; 81:635-646.e1. [PMID: 36623684 DOI: 10.1053/j.ajkd.2022.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/02/2022] [Indexed: 01/09/2023]
Abstract
RATIONALE & OBJECTIVE Focal segmental glomerulosclerosis (FSGS) is a major cause of pediatric nephrotic syndrome, and African Americans exhibit an increased risk for developing FSGS compared with other populations. Predisposing genetic factors have previously been described in adults. Here we performed genomic screening of primary FSGS in a pediatric African American population. STUDY DESIGN Prospective cohort with case-control genetic association study design. SETTING & PARTICIPANTS 140 African American children with chronic kidney disease from the Chronic Kidney Disease in Children (CKiD) cohort, including 32 cases with FSGS. PREDICTORS Over 680,000 common single-nucleotide polymorphisms (SNPs) were tested for association. We also ran a pathway enrichment analysis and a human leucocyte antigen (HLA)-focused association study. OUTCOME Primary biopsy-proven pediatric FSGS. ANALYTICAL APPROACH Multivariate logistic regression models. RESULTS The genome-wide association study revealed 169 SNPs from 14 independent loci significantly associated with FSGS (false discovery rate [FDR]<5%). We observed notable signals for genetic variants within the APOL1 (P=8.6×10-7; OR, 25.8 [95% CI, 7.1-94.0]), ALMS1 (P=1.3×10-7; 13.0% in FSGS cases vs 0% in controls), and FGFR4 (P=4.3×10-6; OR, 24.8 [95% CI, 6.3-97.7]) genes, all of which had previously been associated with adult FSGS, kidney function, or chronic kidney disease. We also highlighted novel, functionally relevant genes, including GRB2 (which encodes a slit diaphragm protein promoting podocyte structure through actin polymerization) and ITGB1 (which is linked to renal injuries). Our results suggest a major role for immune responses and antigen presentation in pediatric FSGS through (1) associations with SNPs in PTPRJ (or CD148, P=3.5×10-7), which plays a role in T-cell receptor signaling, (2) HLA-DRB1∗11:01 association (P=6.1×10-3; OR, 4.5 [95% CI, 1.5-13.0]), and (3) signaling pathway enrichment (P=1.3×10-6). LIMITATIONS Sample size and no independent replication cohort with genomic data readily available. CONCLUSIONS Our genetic study has identified functionally relevant risk factors and the importance of immune regulation for pediatric primary FSGS, which contributes to a better description of its molecular pathophysiological mechanisms.
Collapse
Affiliation(s)
- Axelle Durand
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Cheryl A Winkler
- Basic Research Laboratory, Center for Cancer Research, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland
| | - Nicolas Vince
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Venceslas Douillard
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Estelle Geffard
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Elizabeth Binns-Roemer
- Basic Research Laboratory, Center for Cancer Research, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland
| | - Derek K Ng
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Pierre-Antoine Gourraud
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Kimberley Reidy
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | | | - Susan Furth
- Children's Hospital of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Frederick J Kaskel
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | - Sophie Limou
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France.
| |
Collapse
|
278
|
Lan R, Li X, Chen X, Hu J, Luo W, Lv L, Shen Y, Qin Y, Mao L, Ye H, Li Q, Wang Z. Secondhand smoke, genetic susceptibility, and incident chronic kidney disease in never smokers: A prospective study of a selected population from the UK Biobank. Tob Induc Dis 2023; 21:58. [PMID: 37181462 PMCID: PMC10170651 DOI: 10.18332/tid/162607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
INTRODUCTION A large number of people around the world are exposed to the risks of passive smoking. This prospective study aimed to examine the association between secondhand smoke exposure, exposure time, and the incidence of chronic kidney disease (CKD) and determine whether this association was influenced by genetic susceptibility. METHODS The study included 214244 participants of the UK Biobank who were initially free of CKD. Cox proportional hazards model was used to estimate the associations between secondhand smoke exposure time and the risks of CKD in people who have never smoked. The genetic risk score for CKD was calculated by a weighted method. The likelihood ratio test comparing models was used to examine the cross-product term between secondhand smoke exposure and genetic susceptibility to CKD outcomes. RESULTS During a median of 11.9 years of follow-up, 6583 incidents of CKD were documented. Secondhand smoke exposure increased the risk of CKD (HR=1.09; 95% CI: 1.03-1.16, p<0.01), and a dose-response relationship between CKD prevalence and secondhand smoke exposure time was found (p for trend<0.01). Secondhand smoke exposure increases the risk of CKD even in people who never smoke and have a low genetic risk (HR=1.13; 95% CI: 1.02-1.26, p=0.02). There was no statistically significant interaction between secondhand smoke exposure and genetic susceptibility to CKD (p for interaction=0.80). CONCLUSIONS Secondhand smoke exposure is associated with higher risk of CKD, even in people with low genetic risk, and the relationship is dose dependent. These findings change the belief that people with low genetic susceptibility and without direct participation in smoking activities are not prone to CKD, emphasizing the need to avoid the harm of secondhand smoke in public places.
Collapse
Affiliation(s)
- Rui Lan
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xue Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangjun Chen
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinbo Hu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenjin Luo
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangjing Lv
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Shen
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Qin
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lina Mao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hanwen Ye
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qifu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihong Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
279
|
Chen H, Wang J, Ouyang Q, Peng X, Yu Z, Wang J, Huang J. Alterations of gut microbes and their correlation with clinical features in middle and end-stages chronic kidney disease. Front Cell Infect Microbiol 2023; 13:1105366. [PMID: 37033494 PMCID: PMC10079997 DOI: 10.3389/fcimb.2023.1105366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Gut microecosystem has been shown to play an important role in human health. In recent years, the concept of the gut-kidney axis has been proposed to explain the potential association between gut microbiota and chronic kidney disease (CKD). Here, a cohort of fecal samples collected from patients with CKD (n = 13) were involved. The composition of gut microbial communities and clinical features in CKD and end-stage renal disease (ESRD) were characterized. Our study focused on the changes in gut microbiome and the correlation with clinical features in patients with CKD and ESRD by analyzing high-throughput sequencing results of collected feces. We elucidated the alterations of gut microbiota in CKD patients at different stages of disease and initially identified the gut microbiota associated with CKD progression. We also combined correlation analysis to identify clinical features closely related to the gut microbiome. Our results offered the possibility of using non-invasive gut microbiome in the early diagnosis of course from CKD to ESRD and provide new insights into the association between clinical features and gut microbiota in CKD.
Collapse
Affiliation(s)
- Hao Chen
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jingyan Wang
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Qin Ouyang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xinyue Peng
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jianwen Wang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Jing Huang, ; Jianwen Wang,
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- *Correspondence: Jing Huang, ; Jianwen Wang,
| |
Collapse
|
280
|
Park S, Kim SG, Lee S, Kim Y, Cho S, Kim K, Kim YC, Han SS, Lee H, Lee JP, Joo KW, Lim CS, Kim YS, Kim DK. Genetically predicted body selenium concentration and eGFR: A Mendelian randomization study. Kidney Int Rep 2023; 8:851-859. [PMID: 37069993 PMCID: PMC10105058 DOI: 10.1016/j.ekir.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/29/2022] [Accepted: 01/09/2023] [Indexed: 01/22/2023] Open
Abstract
Introduction Selenium is a trace mineral that is commonly included in micronutrient supplements. The effect of selenium on kidney function remains unclear. A genetically predicted micronutrient and its association with estimated glomerular filtration rate (eGFR) can be used to assess the causal estimates by Mendelian randomization (MR). Methods In this MR study, we instrumented 11 genetic variants associated with blood or total selenium levels from a previous genome-wide association study (GWAS). The association between genetically predicted selenium concentration and eGFR was first assessed by summary-level MR in the chronic kidney disease(CKDGen) GWAS meta-analysis summary statistics, including 567,460 European samples. Inverse-variance weighted and pleiotropy-robust MR analyses were performed, in addition to multivariable MR adjusted for the effects of type 2 diabetes mellitus. Replication analysis was performed with individual-level UK Biobank data, including 337,318 White individuals of British ancestry. Results Summary-level MR analysis indicated that a genetically predicted 1 SD increase in selenium concentration was significantly associated with lower eGFR (-1.05 [-1.28, -0.82] %). The results were similarly reproduced by pleiotropy-robust MR analysis, including MR-Egger and weighted-median methods, and consistent even in the multivariable MR adjusted for diabetes. In the UK Biobank data, genetically predicted higher selenium concentration was also significantly associated with lower eGFR (- 0.36 [-0.52, -0.20] %), and the results were similar when body mass index, waist circumference, hypertension, and diabetes mellitus covariates were adjusted (-0.33 [-0.50, -0.17] %). Conclusion This MR study supports the hypothesis that higher genetically predicted body selenium is causally associated with lower eGFR.
Collapse
|
281
|
Levinsohn J, Li S, Ha E, Susztak K. Combing Genome-Wide Association Studies and Single-Cell Analysis to Elucidate the Mechanisms of Kidney Disease: Proceedings of the Henry Shavelle Professorship. GLOMERULAR DISEASES 2023; 3:258-265. [PMID: 38033715 PMCID: PMC10686632 DOI: 10.1159/000534678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/13/2023] [Indexed: 12/02/2023]
Abstract
Background Kidney diseases pose a significant global health burden; there is an urgent need to deepen our understanding of their underlying mechanisms. Summary This review focuses on new innovative approaches that merge genome-wide association studies (GWAS) and single-cell omics (including transcriptomics) in kidney disease research. We begin by detailing how GWAS has identified numerous genetic risk factors, offering valuable insight into disease susceptibility. Then, we explore the application of scRNA-seq, highlighting its ability to unravel how genetic variants influence cellular phenotypes. Through a synthesis of recent studies, we illuminate the synergy between these two powerful methodologies, demonstrating their potential in elucidating the complex etiology of kidney diseases. Moreover, we discuss how this integrative approach could pave the way for precise diagnostics and personalized treatments. Key Message This review underscores the transformative potential of combining GWAS and scRNA-seq in the journey toward a deeper understanding of kidney diseases.
Collapse
Affiliation(s)
- Jonathan Levinsohn
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, Philadelphia, PA, USA
| | - Shen Li
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, Philadelphia, PA, USA
| | - Eunji Ha
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, Philadelphia, PA, USA
| | - Katalin Susztak
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
- Penn/CHOP Kidney Innovation Center, Philadelphia, PA, USA
| |
Collapse
|
282
|
Yonezawa Y, Guo L, Kakinuma H, Otomo N, Yoshino S, Takeda K, Nakajima M, Shiraki T, Ogura Y, Takahashi Y, Koike Y, Minami S, Uno K, Kawakami N, Ito M, Yonezawa I, Watanabe K, Kaito T, Yanagida H, Taneichi H, Harimaya K, Taniguchi Y, Shigematsu H, Iida T, Demura S, Sugawara R, Fujita N, Yagi M, Okada E, Hosogane N, Kono K, Chiba K, Kotani T, Sakuma T, Akazawa T, Suzuki T, Nishida K, Kakutani K, Tsuji T, Sudo H, Iwata A, Sato T, Inami S, Nakamura M, Matsumoto M, Terao C, Watanabe K, Okamoto H, Ikegawa S. Identification of a Functional Susceptibility Variant for Adolescent Idiopathic Scoliosis that Upregulates Early Growth Response 1 (EGR1)-Mediated UNCX Expression. J Bone Miner Res 2023; 38:144-153. [PMID: 36342191 DOI: 10.1002/jbmr.4738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/23/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Adolescent idiopathic scoliosis (AIS) is a serious health problem affecting 3% of live births all over the world. Many loci associated with AIS have been identified by previous genome wide association studies, but their biological implication remains mostly unclear. In this study, we evaluated the AIS-associated variants in the 7p22.3 locus by combining in silico, in vitro, and in vivo analyses. rs78148157 was located in an enhancer of UNCX, a homeobox gene and its risk allele upregulated the UNCX expression. A transcription factor, early growth response 1 (EGR1), transactivated the rs78148157-located enhancer and showed a higher binding affinity for the risk allele of rs78148157. Furthermore, zebrafish larvae with UNCX messenger RNA (mRNA) injection developed body curvature and defective neurogenesis in a dose-dependent manner. rs78148157 confers the genetic susceptibility to AIS by enhancing the EGR1-regulated UNCX expression. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Yoshiro Yonezawa
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Long Guo
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan.,Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Hisaya Kakinuma
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Brain Science Institute, Saitama, Japan
| | - Nao Otomo
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Soichiro Yoshino
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Takeda
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Nakajima
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Toshiyuki Shiraki
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Brain Science Institute, Saitama, Japan
| | - Yoji Ogura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Takahashi
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshinao Koike
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Department of Orthopedic Surgery, Graduate School of Medical Sciences, Hokkaido University, Sapporo, Japan
| | - Shohei Minami
- Department of Orthopedic Surgery, Seirei Sakura Citizen Hospital, Chiba, Japan
| | - Koki Uno
- Department of Orthopedic Surgery, National Hospital Organization, Kobe Medical Center, Kobe, Japan
| | | | - Manabu Ito
- Department of Orthopedic Surgery, National Hospital Organization Hokkaido Medical Center, Sapporo, Japan
| | - Ikuho Yonezawa
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Kei Watanabe
- Department of Orthopedic Surgery, Niigata University Medical and Dental General Hospital, Niigata, Japan
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Haruhisa Yanagida
- Department of Orthopedic Surgery, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Hiroshi Taneichi
- Department of Orthopedic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Katsumi Harimaya
- Department of Orthopedic Surgery, Kyushu University Beppu Hospital, Beppu, Japan
| | - Yuki Taniguchi
- Department of Orthopedic, Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideki Shigematsu
- Department of Orthopedic Surgery, Nara Medical University, Nara, Japan
| | - Takahiro Iida
- Department of Orthopedic Surgery, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Satoru Demura
- Department of Orthopedic Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Ryo Sugawara
- Department of Orthopedic Surgery, Jichi Medical University, Tochigi, Japan
| | - Nobuyuki Fujita
- Department of Orthopedic Surgery, Fujita Health University, Nagoya, Japan
| | - Mitsuru Yagi
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Eijiro Okada
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Naobumi Hosogane
- Department of Orthopedic Surgery, Kyorin University School of Medicine, Tokyo, Japan
| | - Katsuki Kono
- Department of Orthopedic Surgery, Kono Orthopaedic Clinic, Tokyo, Japan
| | - Kazuhiro Chiba
- Department of Orthopedic Surgery, National Defense Medical College, Saitama, Japan
| | - Toshiaki Kotani
- Department of Orthopedic Surgery, Seirei Sakura Citizen Hospital, Chiba, Japan
| | - Tsuyoshi Sakuma
- Department of Orthopedic Surgery, Seirei Sakura Citizen Hospital, Chiba, Japan
| | - Tsutomu Akazawa
- Department of Orthopedic Surgery, Seirei Sakura Citizen Hospital, Chiba, Japan
| | - Teppei Suzuki
- Department of Orthopedic Surgery, National Hospital Organization, Kobe Medical Center, Kobe, Japan
| | - Kotaro Nishida
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichiro Kakutani
- Department of Orthopedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taichi Tsuji
- Department of Orthopedic Surgery, Meijo Hospital, Nagoya, Japan
| | - Hideki Sudo
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akira Iwata
- Department of Preventive and Therapeutic Research for Metastatic Bone Tumor, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuya Sato
- Department of Orthopedic Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | - Satoshi Inami
- Department of Orthopedic Surgery, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kota Watanabe
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hitoshi Okamoto
- Laboratory for Neural Circuit Dynamics of Decision Making, RIKEN Brain Science Institute, Saitama, Japan
| | - Shiro Ikegawa
- Laboratory for Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| |
Collapse
|
283
|
Huang Y, Wang J, Yang H, Lin Z, Xu L. Causal associations between polyunsaturated fatty acids and kidney function: A bidirectional Mendelian randomization study. Am J Clin Nutr 2023; 117:199-206. [PMID: 36789939 DOI: 10.1016/j.ajcnut.2022.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND PUFAs were suggested to be beneficial for kidney function in observational studies. However, whether these associations are causal remains unclear. OBJECTIVES This study explores the causality between PUFAs and chronic kidney disease (CKD) or estimated glomerular filtration rate (eGFR) using bidirectional 2-sample Mendelian randomization (MR). METHODS Single nucleotide polymorphisms associated with PUFAs and kidney function were obtained from the largest and most recent genome-wide association studies with sample sizes of 13,544, 13,506, 13,499, 13,527, and 13,549 for omega-3 fatty acids, omega-6 fatty acids, DHA, LA, and other PUFAs than 18:2 (otPUFA), and 480,698 and 1,201,909 for CKD and eGFR, respectively. MR inverse-variance weighted (IVW) and pleiotropy residual sum and outlier test (MR-PRESSO) were used for data analysis, supplemented with a weighted median estimator, MR-Egger regression, and multivariable MR, giving β or OR and their 95% CIs. RESULTS There was suggestive evidence that higher omega-6 fatty acids were associated with increased eGFR using MR-PRESSO [β: 0.005 log(mL/min/1.73 m2) per SD increase in omega-6 fatty acids; 95% CI: 0.002, 0.008; P = 0.008]. Higher LA level was also associated with higher eGFR [β: 0.005 log(mL/min/1.73 m2) per SD increase in LA; 95% CI: 0.003, 0.007; P = 0.0007] using MR-PRESSO. Neither association of the other PUFAs, i.e., omega-3 fatty acids, DHA, and otPUFA, with CKD or eGFR nor the association of CKD and eGFR with PUFAs was found. Similar results were found in sensitivity analyses. CONCLUSIONS Our results suggest that higher omega-6 fatty acids and LA may increase eGFR levels. Although the estimated effects were relatively small, the results provide public health and research relevance, indicating the need for further longitudinal cohorts or randomized controlled trials on omega-6 fatty acids in improving kidney function.
Collapse
Affiliation(s)
- Yingyue Huang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jiao Wang
- School of Public Health, Sun Yat-sen University, Guangzhou, China
| | | | - Zihong Lin
- Hezhou Research Institute of Longevity Health Science, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, Guangzhou, China; School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
284
|
Uglebjerg N, Ahmadizar F, Aly DM, Cañadas-Garre M, Hill C, Naber A, Oddsson A, Singh SS, Smyth L, Trégouët DA, Chaker L, Ghanbari M, Steinthorsdottir V, Ahlqvist E, Hadjadj S, Van Hoek M, Kavousi M, McKnight AJ, Sijbrands EJ, Stefansson K, Simons M, Rossing P, Ahluwalia TS. Four missense genetic variants in CUBN are associated with higher levels of eGFR in non-diabetes but not in diabetes mellitus or its subtypes: A genetic association study in Europeans. Front Endocrinol (Lausanne) 2023; 14:1081741. [PMID: 36926036 PMCID: PMC10011651 DOI: 10.3389/fendo.2023.1081741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
AIM Rare genetic variants in the CUBN gene encoding the main albumin-transporter in the proximal tubule of the kidneys have previously been associated with microalbuminuria and higher urine albumin levels, also in diabetes. Sequencing studies in isolated proteinuria suggest that these variants might not affect kidney function, despite proteinuria. However, the relation of these CUBN missense variants to the estimated glomerular filtration rate (eGFR) is largely unexplored. We hereby broadly examine the associations between four CUBN missense variants and eGFRcreatinine in Europeans with Type 1 (T1D) and Type 2 Diabetes (T2D). Furthermore, we sought to deepen our understanding of these variants in a range of single- and aggregate- variant analyses of other kidney-related traits in individuals with and without diabetes mellitus. METHODS We carried out a genetic association-based linear regression analysis between four CUBN missense variants (rs141640975, rs144360241, rs45551835, rs1801239) and eGFRcreatinine (ml/min/1.73 m2, CKD-EPIcreatinine(2012), natural log-transformed) in populations with T1D (n ~ 3,588) or T2D (n ~ 31,155) from multiple European studies and in individuals without diabetes from UK Biobank (UKBB, n ~ 370,061) with replication in deCODE (n = 127,090). Summary results of the diabetes-group were meta-analyzed using the fixed-effect inverse-variance method. RESULTS Albeit we did not observe associations between eGFRcreatinine and CUBN in the diabetes-group, we found significant positive associations between the minor alleles of all four variants and eGFRcreatinine in the UKBB individuals without diabetes with rs141640975 being the strongest (Effect=0.02, PeGFR_creatinine=2.2 × 10-9). We replicated the findings for rs141640975 in the Icelandic non-diabetes population (Effect=0.026, PeGFR_creatinine=7.7 × 10-4). For rs141640975, the eGFRcreatinine-association showed significant interaction with albuminuria levels (normo-, micro-, and macroalbuminuria; p = 0.03). An aggregated genetic risk score (GRS) was associated with higher urine albumin levels and eGFRcreatinine. The rs141640975 variant was also associated with higher levels of eGFRcreatinine-cystatin C (ml/min/1.73 m2, CKD-EPI2021, natural log-transformed) and lower circulating cystatin C levels. CONCLUSIONS The positive associations between the four CUBN missense variants and eGFR in a large population without diabetes suggests a pleiotropic role of CUBN as a novel eGFR-locus in addition to it being a known albuminuria-locus. Additional associations with diverse renal function measures (lower cystatin C and higher eGFRcreatinine-cystatin C levels) and a CUBN-focused GRS further suggests an important role of CUBN in the future personalization of chronic kidney disease management in people without diabetes.
Collapse
Affiliation(s)
- Nicoline Uglebjerg
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Fariba Ahmadizar
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Data Science & Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, Netherlands
| | - Dina M. Aly
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Marisa Cañadas-Garre
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
- GENYO Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Claire Hill
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | - Annemieke Naber
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - Sunny S. Singh
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Laura Smyth
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | - David-Alexandre Trégouët
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale (INSERM), Bordeaux Population Health Research Center, Bordeaux, France
| | - Layal Chaker
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | | - Emma Ahlqvist
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Samy Hadjadj
- Nantes Université, Centre Hospitalier Universitaire Nantes, Centre National de la Recherche Scientifique, INSERM, l’institut du thorax, Nantes, France
| | - Mandy Van Hoek
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Amy Jayne McKnight
- Centre for Public Health, Queen’s University Belfast, Belfast, United Kingdom
| | - Eric J. Sijbrands
- Department of Internal Medicine, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Kari Stefansson
- deCODE Genetics, Amgen, Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Matias Simons
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Rossing
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Tarunveer S. Ahluwalia
- Complications Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- The Bioinformatics Center, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Tarunveer S. Ahluwalia,
| |
Collapse
|
285
|
Devuyst O, Kiryluk K. On the way to translate GWAS into kidney disease mechanisms. Kidney Int 2023; 103:16-18. [PMID: 36309125 DOI: 10.1016/j.kint.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Olivier Devuyst
- Department of Physiology, Mechanisms of Inherited Kidney Disorders, University of Zurich, Zurich, Switzerland.
| | - Krzysztof Kiryluk
- Division of Nephrology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
286
|
Han SK, Muto Y, Wilson PC, Humphreys BD, Sampson MG, Chakravarti A, Lee D. Quality assessment and refinement of chromatin accessibility data using a sequence-based predictive model. Proc Natl Acad Sci U S A 2022; 119:e2212810119. [PMID: 36508674 PMCID: PMC9907136 DOI: 10.1073/pnas.2212810119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022] Open
Abstract
Chromatin accessibility assays are central to the genome-wide identification of gene regulatory elements associated with transcriptional regulation. However, the data have highly variable quality arising from several biological and technical factors. To surmount this problem, we developed a sequence-based machine learning method to evaluate and refine chromatin accessibility data. Our framework, gapped k-mer SVM quality check (gkmQC), provides the quality metrics for a sample based on the prediction accuracy of the trained models. We tested 886 DNase-seq samples from the ENCODE/Roadmap projects to demonstrate that gkmQC can effectively identify "high-quality" (HQ) samples with low conventional quality scores owing to marginal read depths. Peaks identified in HQ samples are more accurately aligned at functional regulatory elements, show greater enrichment of regulatory elements harboring functional variants, and explain greater heritability of phenotypes from their relevant tissues. Moreover, gkmQC can optimize the peak-calling threshold to identify additional peaks, especially for rare cell types in single-cell chromatin accessibility data.
Collapse
Affiliation(s)
- Seong Kyu Han
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston & Harvard Medical School, Boston, MA02115
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA02142
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO63130
| | - Parker C. Wilson
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO63130
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO63130
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO63130
| | - Matthew G. Sampson
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston & Harvard Medical School, Boston, MA02115
- Kidney Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA02142
| | - Aravinda Chakravarti
- Center for Human Genetics and Genomics, New York University Grossman School of Medicine, New York, NY10016
| | - Dongwon Lee
- Department of Pediatrics, Division of Nephrology, Boston Children’s Hospital, Boston & Harvard Medical School, Boston, MA02115
- Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA02115
| |
Collapse
|
287
|
Sun X, Chen L, Zheng L. A Mendelian randomization study to assess the genetic liability of gastroesophageal reflux disease for cardiovascular diseases and risk factors. Hum Mol Genet 2022; 31:4275-4285. [PMID: 35861629 DOI: 10.1093/hmg/ddac162] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/25/2022] [Accepted: 07/11/2022] [Indexed: 01/21/2023] Open
Abstract
Observational studies have reported that gastroesophageal reflux disease (GERD) is a risk factor for cardiovascular diseases (CVD); however, the causal inferences between them remain unknown. We conducted a Mendelian randomization (MR) study to estimate the causal associations between GERD and 10 CVD outcomes, as well as 14 cardiovascular risk factors. We used summary statistics from genome-wide association studies for GERD and the FinnGen consortium for CVD. We further investigated whether GERD correlated with cardiovascular risk factors and performed multivariable MR and mediation analyses to estimate the mediating effects of these risk factors on GERD-CVD progression. Sensitivity analyses and replication analyses were also performed. Our results indicated that GERD was positively associated with seven CVD outcomes with odds ratios of 1.26 [95% confidence interval (CI), 1.15, 1.37] for coronary artery disease, 1.41 (95% CI, 1.28, 1.57) for myocardial infarction, 1.34 (95% CI, 1.19, 1.51) for atrial fibrillation, 1.34 (95% CI, 1.21, 1.50) for heart failure, 1.30 (95% CI, 1.18, 1.43) for any stroke, 1.19 (95% CI, 1.06, 1.34) for ischemic stroke and 1.29 (95% CI, 1.16, 1.44) for venous thromboembolism. Furthermore, GERD was associated with nine cardiovascular risk factors and major depressive disorder demonstrated significant mediation effects on the causal pathway linking GERD and any stroke. This study demonstrates that GERD is associated with seven CVD outcomes and nine cardiovascular risk factors. Importantly, GERD treatment may help prevent common CVD events.
Collapse
Affiliation(s)
- Xingang Sun
- Department of Cardiology and Atrial fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Lu Chen
- Department of Cardiology and Atrial fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Liangrong Zheng
- Department of Cardiology and Atrial fibrillation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
288
|
Hindy G, Tyrrell DJ, Vasbinder A, Wei C, Presswalla F, Wang H, Blakely P, Ozel AB, Graham S, Holton GH, Dowsett J, Fahed AC, Amadi KM, Erne GK, Tekmulla A, Ismail A, Launius C, Sotoodehnia N, Pankow JS, Thørner LW, Erikstrup C, Pedersen OB, Banasik K, Brunak S, Ullum H, Eugen-Olsen J, Ostrowski SR, on behalf of the DBDS Consortium, Haas ME, Nielsen JB, Lotta LA, on behalf of the Regeneron Genetics Center, Engström G, Melander O, Orho-Melander M, Zhao L, Murthy VL, Pinsky DJ, Willer CJ, Heckbert SR, Reiser J, Goldstein DR, Desch KC, Hayek SS. Increased soluble urokinase plasminogen activator levels modulate monocyte function to promote atherosclerosis. J Clin Invest 2022; 132:e158788. [PMID: 36194491 PMCID: PMC9754000 DOI: 10.1172/jci158788] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 09/29/2022] [Indexed: 01/26/2023] Open
Abstract
People with kidney disease are disproportionately affected by atherosclerosis for unclear reasons. Soluble urokinase plasminogen activator receptor (suPAR) is an immune-derived mediator of kidney disease, levels of which are strongly associated with cardiovascular outcomes. We assessed suPAR's pathogenic involvement in atherosclerosis using epidemiologic, genetic, and experimental approaches. We found serum suPAR levels to be predictive of coronary artery calcification and cardiovascular events in 5,406 participants without known coronary disease. In a genome-wide association meta-analysis including over 25,000 individuals, we identified a missense variant in the plasminogen activator, urokinase receptor (PLAUR) gene (rs4760), confirmed experimentally to lead to higher suPAR levels. Mendelian randomization analysis in the UK Biobank using rs4760 indicated a causal association between genetically predicted suPAR levels and atherosclerotic phenotypes. In an experimental model of atherosclerosis, proprotein convertase subtilisin/kexin-9 (Pcsk9) transfection in mice overexpressing suPAR (suPARTg) led to substantially increased atherosclerotic plaques with necrotic cores and macrophage infiltration compared with those in WT mice, despite similar cholesterol levels. Prior to induction of atherosclerosis, aortas of suPARTg mice excreted higher levels of CCL2 and had higher monocyte counts compared with WT aortas. Aortic and circulating suPARTg monocytes exhibited a proinflammatory profile and enhanced chemotaxis. These findings characterize suPAR as a pathogenic factor for atherosclerosis acting at least partially through modulation of monocyte function.
Collapse
Affiliation(s)
- George Hindy
- Department of Clinical Sciences, Lund University, Malmö, Sweden
- Department of Population Medicine, Qatar University College of Medicine, QU Health, Doha, Qatar
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Daniel J. Tyrrell
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Changli Wei
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Feriel Presswalla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hui Wang
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pennelope Blakely
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayse Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Graham
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace H. Holton
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph Dowsett
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Akl C. Fahed
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kingsley-Michael Amadi
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace K. Erne
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Annika Tekmulla
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anis Ismail
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Launius
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jesper Eugen-Olsen
- Department of Clinical Research, Copenhagen University Hospital Amager and Hvidovre, Hvidovre, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Mary E. Haas
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Jonas B. Nielsen
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | - Luca A. Lotta
- Regeneron Genetics Center, Regeneron Pharmaceuticals Inc., Tarrytown, New York, USA
| | | | - Gunnar Engström
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | | | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Venkatesh L. Murthy
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - David J. Pinsky
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Cristen J. Willer
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Susan R. Heckbert
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Karl C. Desch
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Salim S. Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
289
|
Yuan S, Wang L, Sun J, Yu L, Zhou X, Yang J, Zhu Y, Gill D, Burgess S, Denny JC, Larsson SC, Theodoratou E, Li X. Genetically predicted sex hormone levels and health outcomes: phenome-wide Mendelian randomization investigation. Int J Epidemiol 2022; 51:1931-1942. [PMID: 35218343 PMCID: PMC9749729 DOI: 10.1093/ije/dyac036] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sex hormone-binding globulin (SHBG), testosterone and oestradiol have been associated with many diseases in observational studies; however, the causality of associations remains unestablished. METHODS A phenome-wide Mendelian randomization (MR) association study was performed to explore disease outcomes associated with genetically proxied circulating SHBG, testosterone and oestradiol levels by using updated genetic instruments in 339 197 unrelated White British individuals (54% female) in the UK Biobank. Two-sample MR analyses with data from large genetic studies were conducted to replicate identified associations in phenome-wide MR analyses. Multivariable MR analyses were performed to investigate mediation effects of hormone-related biomarkers in observed associations with diseases. RESULTS Phenome-wide MR analyses examined associations of genetically predicted SHBG, testosterone and oestradiol levels with 1211 disease outcomes, and identified 28 and 13 distinct phenotypes associated with genetically predicted SHBG and testosterone, respectively; 22 out of 28 associations for SHBG and 10 out of 13 associations for testosterone were replicated in two-sample MR analyses. Higher genetically predicted SHBG levels were associated with a reduced risk of hypertension, type 2 diabetes, diabetic complications, coronary atherosclerotic outcomes, gout and benign and malignant neoplasm of uterus, but an increased risk of varicose veins and fracture (mainly in females). Higher genetically predicted testosterone levels were associated with a lower risk of type 2 diabetes, coronary atherosclerotic outcomes, gout and coeliac disease mainly in males, but an increased risk of cholelithiasis in females. CONCLUSIONS These findings suggest that sex hormones may causally affect risk of several health outcomes.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lijuan Wang
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Sun
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Yu
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuan Zhou
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jie Yang
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yimin Zhu
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | - Joshua C Denny
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Xue Li
- Corresponding author. School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China. E-mail:
| |
Collapse
|
290
|
Kjaergaard AD, Ellervik C, Witte DR, Nordestgaard BG, Frikke-Schmidt R, Bojesen SE. Kidney function and risk of dementia: Observational study, meta-analysis, and two-sample mendelian randomization study. Eur J Epidemiol 2022; 37:1273-1284. [PMID: 36333541 DOI: 10.1007/s10654-022-00923-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 09/25/2022] [Indexed: 11/06/2022]
Abstract
Whether impaired kidney function is associated with increased risk of developing dementia is unclear. We investigated the association between estimated glomerular filtration rate (eGFR) and dementia. Using a triangulation approach, we performed (1) a prospective study in 90,369 Danes from the Copenhagen General Population Study (CGPS), (2) a meta-analysis in 468,699 Scandinavians (including CGPS) and (3) a two-sample Mendelian randomization study in 218,792-1,004,040 Europeans using summary data from largest publicly available genome wide association studies (GWASs). During up to 15 years of follow-up (CGPS), 2,468 individuals developed dementia. Age and sex standardized percentile of eGFR below versus above the median conferred a multifactorially adjusted hazard ratio of 1.09 (95% confidence interval: 1.01-1.18). In meta-analysis, random-effects risk of dementia was 1.14 (1.06-1.22) for mildly decreased eGFR (60-90 mL/min/1.73 m2), 1.31 (0.92-1.87) for moderately decreased eGFR (30-59 mL/min/1.73 m2) and 1.91 (1.21-3.01) for severely decreased eGFR (< 30 mL/min/1.73 m2), compared to reference eGFR (> 90 mL/min/1.73 m2). Using directly comparable eGFR measures (log[eGFR] scaled to one standard deviation, as well as eGFR below versus above 60 mL/min/1.73 m2), we found no association with risk of dementia in observational CGPS or in Mendelian randomization analyses. In conclusion, impaired kidney function was associated with modestly increased risk of developing dementia. This was not supported by causal, genetic analyses using a Mendelian randomization approach. However, future stronger genetic instruments for kidney function and larger GWASs with more dementia cases, particularly for the vascular dementia subtype, warrant a re-evaluation of the causal hypothesis.
Collapse
Affiliation(s)
- Alisa D Kjaergaard
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Blvd. 11, Indgang A, Aarhus, Denmark.
| | - Christina Ellervik
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
- Department of Data and Development, Sorø, Region Zealand, Denmark
- Department of Pathology, Harvard Medical School and Department of Laboratory Medicine, Boston Children's Hospital, MA-02215, Boston, USA
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Stig E Bojesen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, University of Copenhagen, Herlev, Denmark
| |
Collapse
|
291
|
Al-Hamed MH, Hussein MH, Shah Y, Al-Mojalli H, Alsabban E, Alshareef T, Altayyar A, Elshouny S, Ali W, Abduljabbar M, AlOtaibi A, AlShammasi A, Akili R, Abouelhoda M, Sayer JA, Dasouki MJ, Imtiaz F. Exome sequencing unravels genetic variants associated with chronic kidney disease in Saudi Arabian patients. Hum Mutat 2022; 43:e24-e37. [PMID: 36177613 DOI: 10.1002/humu.24480] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 09/10/2022] [Accepted: 09/26/2022] [Indexed: 01/25/2023]
Abstract
The use of genetic testing within nephrology is increasing and its diagnostic yield depends on the methods utilized, patient selection criteria, and population characteristics. We performed exome sequencing (ES) analysis on 102 chronic kidney disease (CKD) patients with likely genetic kidney disease. Patients had diverse CKD subtypes with/without consanguinity, positive family history, and possible hereditary renal syndrome with extra-renal abnormalities or progressive kidney disease of unknown etiology. The identified genetic variants associated with the observed kidney phenotypes were then confirmed and reported. End-stage kidney disease was reported in 51% of the cohort and a family history of kidney disease in 59%, while known consanguinity was reported in 54%. Pathogenic/likely pathogenic variants were identified in 43 patients with a diagnostic yield of 42%, and clinically associated variants of unknown significance (VUS) were identified in further 21 CKD patients (21%). A total of eight novel predicted pathogenic variants and eight VUS were detected. The clinical utility of ES within the nephrology clinic was demonstrated allowing patient management to be disease-specific. In this cohort, ES detected a diagnostic molecular abnormality in 42% of patients with CKD phenotypes. Positive family history and high rates of consanguinity likely contributed to this high diagnostic yield.
Collapse
Affiliation(s)
- Mohamed H Al-Hamed
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Saudi Diagnostics Laboratory, KFSH&RC, Riyadh, Saudi Arabia
| | - Maged H Hussein
- Medicine Department, Nephrology Section, KFSH&RC, Riyadh, Saudi Arabia
| | - Yaser Shah
- Organ Transplant Centre of Excellence, Adult Transplant Nephrology, KFSH&RC, Riyadh, Saudi Arabia
| | - Hamad Al-Mojalli
- Organ Transplant Centre of Excellence, Adult Transplant Nephrology, KFSH&RC, Riyadh, Saudi Arabia
| | | | | | - Ali Altayyar
- Medicine Department, Nephrology Section, KFSH&RC, Riyadh, Saudi Arabia
| | - Samir Elshouny
- Medicine Department, Nephrology Section, KFSH&RC, Riyadh, Saudi Arabia
| | - Wafaa Ali
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mai Abduljabbar
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Afaf AlOtaibi
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Amal AlShammasi
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Rana Akili
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Mohamed Abouelhoda
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Renal Services, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Majed J Dasouki
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Faiqa Imtiaz
- Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Saudi Diagnostics Laboratory, KFSH&RC, Riyadh, Saudi Arabia
| |
Collapse
|
292
|
Lee S, Han M, Moon S, Kim K, An WJ, Ryu H, Oh KH, Park SK. Identifying Genetic Variants and Metabolites Associated with Rapid Estimated Glomerular Filtration Rate Decline in Korea Based on Genome-Metabolomic Integrative Analysis. Metabolites 2022; 12:1139. [PMID: 36422279 PMCID: PMC9695695 DOI: 10.3390/metabo12111139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
Identifying the predisposing factors to chronic or end-stage kidney disease is essential to preventing or slowing kidney function decline. Therefore, here, we investigated the genetic variants related to a rapid decline in the estimated glomerular filtration rate (eGFR) (i.e., a loss of >5 mL/min/1.73 m2 per year) and verified the relationships between variant-related diseases and metabolic pathway signaling in patients with chronic kidney disease. We conducted a genome-wide association study that included participants with diabetes, hypertension, and rapid eGFR decline from two Korean data sources (N = 115 and 69 for the discovery and the validation cohorts, respectively). We identified a novel susceptibility locus: 4q32.3 (rs10009742 in the MARCHF1 gene, beta = −3.540, P = 4.11 × 10−8). Fine-mapping revealed 19 credible, causal single-nucleotide polymorphisms, including rs10009742. The pimelylcarnitine and octadecenoyl carnitine serum concentrations were associated with rs10009742 (beta = 0.030, P = 7.10 × 10−5, false discovery rate (FDR) = 0.01; beta = 0.167, P = 8.11 × 10−4, FDR = 0.08). Our results suggest that MARCHF1 is associated with a rapid eGFR decline in patients with hypertension and diabetes. Furthermore, MARCHF1 affects the pimelylcarnitine metabolite concentration, which may mediate chronic kidney disease progression by inducing oxidative stress in the endoplasmic reticulum.
Collapse
Affiliation(s)
- Sangjun Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Miyeun Han
- Department of Internal Medicine, National Medical Center, Seoul 04564, Republic of Korea
| | - Sungji Moon
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Interdisciplinary Program in Cancer Biology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Kyungsik Kim
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | - Woo Ju An
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Sue K. Park
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
293
|
Gaziano L, Sun L, Arnold M, Bell S, Cho K, Kaptoge SK, Song RJ, Burgess S, Posner DC, Mosconi K, Robinson-Cohen C, Mason AM, Bolton TR, Tao R, Allara E, Schubert P, Chen L, Staley JR, Staplin N, Altay S, Amiano P, Arndt V, Ärnlöv J, Barr EL, Björkelund C, Boer JM, Brenner H, Casiglia E, Chiodini P, Cooper JA, Coresh J, Cushman M, Dankner R, Davidson KW, de Jongh RT, Donfrancesco C, Engström G, Freisling H, de la Cámara AG, Gudnason V, Hankey GJ, Hansson PO, Heath AK, Hoorn EJ, Imano H, Jassal SK, Kaaks R, Katzke V, Kauhanen J, Kiechl S, Koenig W, Kronmal RA, Kyrø C, Lawlor DA, Ljungberg B, MacDonald C, Masala G, Meisinger C, Melander O, Moreno Iribas C, Ninomiya T, Nitsch D, Nordestgaard BG, Onland-Moret C, Palmieri L, Petrova D, Garcia JRQ, Rosengren A, Sacerdote C, Sakurai M, Santiuste C, Schulze MB, Sieri S, Sundström J, Tikhonoff V, Tjønneland A, Tong T, Tumino R, Tzoulaki I, van der Schouw YT, Monique Verschuren W, Völzke H, Wallace RB, Wannamethee SG, Weiderpass E, Willeit P, Woodward M, Yamagishi K, Zamora-Ros R, Akwo EA, Pyarajan S, Gagnon DR, Tsao PS, Muralidhar S, Edwards TL, Damrauer SM, Joseph J, Pennells L, Wilson PW, Harrison S, et alGaziano L, Sun L, Arnold M, Bell S, Cho K, Kaptoge SK, Song RJ, Burgess S, Posner DC, Mosconi K, Robinson-Cohen C, Mason AM, Bolton TR, Tao R, Allara E, Schubert P, Chen L, Staley JR, Staplin N, Altay S, Amiano P, Arndt V, Ärnlöv J, Barr EL, Björkelund C, Boer JM, Brenner H, Casiglia E, Chiodini P, Cooper JA, Coresh J, Cushman M, Dankner R, Davidson KW, de Jongh RT, Donfrancesco C, Engström G, Freisling H, de la Cámara AG, Gudnason V, Hankey GJ, Hansson PO, Heath AK, Hoorn EJ, Imano H, Jassal SK, Kaaks R, Katzke V, Kauhanen J, Kiechl S, Koenig W, Kronmal RA, Kyrø C, Lawlor DA, Ljungberg B, MacDonald C, Masala G, Meisinger C, Melander O, Moreno Iribas C, Ninomiya T, Nitsch D, Nordestgaard BG, Onland-Moret C, Palmieri L, Petrova D, Garcia JRQ, Rosengren A, Sacerdote C, Sakurai M, Santiuste C, Schulze MB, Sieri S, Sundström J, Tikhonoff V, Tjønneland A, Tong T, Tumino R, Tzoulaki I, van der Schouw YT, Monique Verschuren W, Völzke H, Wallace RB, Wannamethee SG, Weiderpass E, Willeit P, Woodward M, Yamagishi K, Zamora-Ros R, Akwo EA, Pyarajan S, Gagnon DR, Tsao PS, Muralidhar S, Edwards TL, Damrauer SM, Joseph J, Pennells L, Wilson PW, Harrison S, Gaziano TA, Inouye M, Baigent C, Casas JP, Langenberg C, Wareham N, Riboli E, Gaziano J, Danesh J, Hung AM, Butterworth AS, Wood AM, Di Angelantonio E. Mild-to-Moderate Kidney Dysfunction and Cardiovascular Disease: Observational and Mendelian Randomization Analyses. Circulation 2022; 146:1507-1517. [PMID: 36314129 PMCID: PMC9662821 DOI: 10.1161/circulationaha.122.060700] [Show More Authors] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/18/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND End-stage renal disease is associated with a high risk of cardiovascular events. It is unknown, however, whether mild-to-moderate kidney dysfunction is causally related to coronary heart disease (CHD) and stroke. METHODS Observational analyses were conducted using individual-level data from 4 population data sources (Emerging Risk Factors Collaboration, EPIC-CVD [European Prospective Investigation into Cancer and Nutrition-Cardiovascular Disease Study], Million Veteran Program, and UK Biobank), comprising 648 135 participants with no history of cardiovascular disease or diabetes at baseline, yielding 42 858 and 15 693 incident CHD and stroke events, respectively, during 6.8 million person-years of follow-up. Using a genetic risk score of 218 variants for estimated glomerular filtration rate (eGFR), we conducted Mendelian randomization analyses involving 413 718 participants (25 917 CHD and 8622 strokes) in EPIC-CVD, Million Veteran Program, and UK Biobank. RESULTS There were U-shaped observational associations of creatinine-based eGFR with CHD and stroke, with higher risk in participants with eGFR values <60 or >105 mL·min-1·1.73 m-2, compared with those with eGFR between 60 and 105 mL·min-1·1.73 m-2. Mendelian randomization analyses for CHD showed an association among participants with eGFR <60 mL·min-1·1.73 m-2, with a 14% (95% CI, 3%-27%) higher CHD risk per 5 mL·min-1·1.73 m-2 lower genetically predicted eGFR, but not for those with eGFR >105 mL·min-1·1.73 m-2. Results were not materially different after adjustment for factors associated with the eGFR genetic risk score, such as lipoprotein(a), triglycerides, hemoglobin A1c, and blood pressure. Mendelian randomization results for stroke were nonsignificant but broadly similar to those for CHD. CONCLUSIONS In people without manifest cardiovascular disease or diabetes, mild-to-moderate kidney dysfunction is causally related to risk of CHD, highlighting the potential value of preventive approaches that preserve and modulate kidney function.
Collapse
Affiliation(s)
- Liam Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Luanluan Sun
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | | | - Steven Bell
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Stroke Research Group, Department of Clinical Neurosciences (S. Bell), University of Cambridge, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
| | - Kelly Cho
- Division of Aging (K.C., S.P., J.P.C. J.M.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Stephen K. Kaptoge
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Rebecca J. Song
- Department of Epidemiology, Boston University School of Public Health, MA (R.J.S.)
| | - Stephen Burgess
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Medical Research Council Biostatistics Unit (A.M.M., S. Burgess), University of Cambridge, UK
| | - Daniel C. Posner
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
| | - Katja Mosconi
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Cassianne Robinson-Cohen
- Division of Nephrology, Department of Medicine (C.R.-C., E.A.A.), Vanderbilt University Medical Center, Nashville, TN
| | - Amy M. Mason
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Medical Research Council Biostatistics Unit (A.M.M., S. Burgess), University of Cambridge, UK
| | - Thomas R. Bolton
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
| | - Ran Tao
- Department of Biostatistics (R. Tao), Vanderbilt University Medical Center, Nashville, TN
| | - Elias Allara
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
| | - Petra Schubert
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
| | - Lingyan Chen
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - James R. Staley
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Natalie Staplin
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit (N.S., C.B.), Nuffield Department of Population Health, University of Oxford, UK
| | - Servet Altay
- Department of Cardiology, Trakya University School of Medicine, Edirne, Turkey (S.A.)
| | - Pilar Amiano
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain (P.A.)
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain (P.A.)
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain (P.A., A.G.d.l.C., D.P., C. Santiuste)
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research (V.A.), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Johan Ärnlöv
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Medical Research Council Biostatistics Unit (A.M.M., S. Burgess), University of Cambridge, UK
- Stroke Research Group, Department of Clinical Neurosciences (S. Bell), University of Cambridge, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
- MRC Epidemiology Unit, School of Clinical Medicine (C.L., N.W.), University of Cambridge, UK
- Division of Aging (K.C., S.P., J.P.C. J.M.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Cardiovascular Medicine (J.J., T.A.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Epidemiology, Boston University School of Public Health, MA (R.J.S.)
- Division of Nephrology, Department of Medicine (C.R.-C., E.A.A.), Vanderbilt University Medical Center, Nashville, TN
- Department of Biostatistics (R. Tao), Vanderbilt University Medical Center, Nashville, TN
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit (N.S., C.B.), Nuffield Department of Population Health, University of Oxford, UK
- Cancer Epidemiology Unit (T.T.), Nuffield Department of Population Health, University of Oxford, UK
- Department of Cardiology, Trakya University School of Medicine, Edirne, Turkey (S.A.)
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain (P.A.)
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain (P.A.)
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain (P.A., A.G.d.l.C., D.P., C. Santiuste)
- Division of Clinical Epidemiology and Aging Research (V.A.), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cancer Epidemiology (S.K.J., R.K., V.K.), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden (J.A., H.B.)
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.A.)
- Wellbeing & Preventable Chronic Diseases (WPCD) Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia (E.L.M.B.)
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (E.L.M.B., M.I.)
- Institute of Medicine, School of Public Health and Community Medicine (C.B.), Sahlgrenska Academy, University of Gothenburg, Sweden
- Institute of Medicine, Department of Molecular and Clinical Medicine (P.-O.H., A.R.), Sahlgrenska Academy, University of Gothenburg, Sweden
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands (J.M.A.B., W.M.M.V.)
- Network Aging Research (NAR), Heidelberg University, Germany (H.B.)
- Studium Patavinum (E.C.), University of Padua, Italy
- Department of Medicine (V.T.), University of Padua, Italy
- Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania ‘Luigi Vanvitelli’, Caserta, Italy (P.C.)
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, UK (J.A.C.)
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (J.C.)
- Larner College of Medicine, The University of Vermont, Burlington (M.C.)
- The Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Tel Hashomer, Israel (R.D.)
- School of Public Health, Department of Epidemiology and Preventive Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel (R.D.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, NY (R.D., K.W.D.)
- Amsterdam University Medical Centers, VUMC, the Netherlands (R.T.d.J.)
- Department of Cardiovascular, Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy (C.D., L. Palmer)
- Department of Clinical Sciences, Malmö, Lund University, Sweden (G.E., O.M.)
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France (H.F., E.W.)
- 12 Octubre Hospital Research Institute, Madrid, Spain (A.G.d,l,C.)
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland and Icelandic Heart Association, Kopavogur, Iceland (V.G.)
- Medical School Faculty of Health & Medical Sciences, The University of Western Australia, Perth, WA, Australia (G.J.H.)
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Medicine Geriatrics and Emergency Medicine/Östra, Gothenburg, Sweden (P.-O.H., A.R.)
- School of Public Health (A.K.H., I.T., E.R.), Imperial College London, UK
- The George Institute for Global Health (M.W.), Imperial College London, UK
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.J.H.)
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Suita, Japan (H.I.)
- University of Eastern Finland (UEF), Kuopio, Finland (J.K.)
- Department of Neurology & Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria (S.K.)
- Clinical Epidemiology Team, Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria (S.K., P.W.)
- Institute of Epidemiology and Medical Biometry, University of Ulm, Germany (W.K.)
- Deutsches Herzzentrum München, Technische Universität München, Germany (W.K.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (W.K.)
- School of Public Health, University of Washington, Seattle (R.A.K.)
- Danish Cancer Society Research Center, Copenhagen, Denmark (C.K., A.T.)
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, UK (D.A.L.)
- Population Health Science, Bristol Medical School, UK (D.A.L.)
- Department of Surgical and Perioperative sciences, Urology and Andrology, Umeå University, Sweden (B.L.)
- University Paris-Saclay, UVSQ, Inserm, Villejuif, France (C. MacDonald)
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy (G.M.)
- Helmholtz Zentrum München, Munich, Germany (C. Meisinger)
- Navarra Public Health Institute, IdiSNA, Pamplona, Spain (C.M.I.)
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Pamplona, Spain (C.M.I.)
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (T.N.)
- London School of Hygiene & Tropical Medicine, UK (D.N.)
- Herlev and Gentofte Hospital (B.G.N.), Copenhagen University Hospital, Copenhagen, Denmark
- Frederiksberg Hospital B.G.N.), Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences (B.G.N.), University of Copenhagen, Denmark
- Department of Public Health (A.T.), University of Copenhagen, Denmark
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands (C.O.-M., Y.T.v.d.S., W.M.M.V.)
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain (D.P.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain (D.P.)
- Consejería de Sanidad del Principado de Asturias Oviedo, Asturias, Spain (J.R.Q.G.)
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital, Turin, Italy (C. Sacerdote)
- Department of Social and Environmental Medicine, Kanazawa Medical University, Uchinada, Japan (M.S.)
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Spain (C. Santiuste)
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany (M.B.S.)
- German Center for Diabetes Research (DZD), Neuherberg, Germany (M.B.S.)
- Institute of Nutritional Science, University of Potsdam, Germany (M.B.S.)
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy (S.S.)
- Department of Medical Sciences, Uppsala University, Sweden (J.S.)
- Hyblean Association for Epidemiological Reserach AIRE - ONLUS, Ragusa, Italy (R.T.)
- Universitätsmedizin Greifswald, Institut für Community Medicine, Abteilung SHIP/ Klinisch-Epidemiologische Forschung, Germany (H.V.)
- College of Public Health, University of Iowa (R.B.W.)
- University College London, UK (S.G.W.)
- The George Institute for Global Health, Camperdown, NSW, Australia (M.W.)
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Japan (K.Y.)
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain (R.Z.-R.)
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA (S.P.)
- Department of Biostatistics, Boston University School of Public Health, MA (D.R.G.)
- VA Pal Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, CA (P.S.T.)
- Medicine (Cardiovascular Medicine), Stanford University of School of Medicine, CA (P.S.T.)
- Office of Research and Development, Veterans Health Administration, Washington, DC (S.M.)
- Department of Veterans Affairs, Tennessee Valley Health Care System, Vanderbilt University, Nashville (T.L.E.)
- Medicine/Epidemiology, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN (T.L.E.)
- Department of Surgery, Corporal Michael Crescenz VA Medical Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.M.D.)
- Internal Medicine, VA Atlanta Healthcare System, Decatur, GA (P.W.F.W.)
- Emory University School of Medicine (Cardiology), Emory University, Atlanta, GA (P.W.F.W.)
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, Boston, MA (T.A.G.)
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
- The Alan Turing Institute, London, UK (M.I.)
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Germany (C.L.)
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK (J.D.)
- Division of Nephrology & Hypertension, Department of Medicine, Tennessee Valley Health Care System and Vanderbilt University Medical Center, Nashville (A.M.H.)
- Cambridge Centre for AI in Medicine, UK (A.M.W.)
- Health Data Science Centre, Human Technopole, Milan, Italy (E.D.A.)
| | - Elizabeth L.M. Barr
- Wellbeing & Preventable Chronic Diseases (WPCD) Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia (E.L.M.B.)
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (E.L.M.B., M.I.)
| | - Cecilia Björkelund
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit (N.S., C.B.), Nuffield Department of Population Health, University of Oxford, UK
| | - Jolanda M.A. Boer
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands (J.M.A.B., W.M.M.V.)
| | - Hermann Brenner
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden (J.A., H.B.)
- Network Aging Research (NAR), Heidelberg University, Germany (H.B.)
| | | | - Paolo Chiodini
- Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania ‘Luigi Vanvitelli’, Caserta, Italy (P.C.)
| | - Jackie A. Cooper
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, UK (J.A.C.)
| | - Josef Coresh
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (J.C.)
| | - Mary Cushman
- Larner College of Medicine, The University of Vermont, Burlington (M.C.)
| | - Rachel Dankner
- The Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Tel Hashomer, Israel (R.D.)
- School of Public Health, Department of Epidemiology and Preventive Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel (R.D.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, NY (R.D., K.W.D.)
| | - Karina W. Davidson
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, NY (R.D., K.W.D.)
| | | | - Chiara Donfrancesco
- Department of Cardiovascular, Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy (C.D., L. Palmer)
| | - Gunnar Engström
- Department of Clinical Sciences, Malmö, Lund University, Sweden (G.E., O.M.)
| | - Heinz Freisling
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France (H.F., E.W.)
| | - Agustín Gómez de la Cámara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain (P.A., A.G.d.l.C., D.P., C. Santiuste)
- 12 Octubre Hospital Research Institute, Madrid, Spain (A.G.d,l,C.)
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland and Icelandic Heart Association, Kopavogur, Iceland (V.G.)
| | - Graeme J. Hankey
- Medical School Faculty of Health & Medical Sciences, The University of Western Australia, Perth, WA, Australia (G.J.H.)
| | - Per-Olof Hansson
- Institute of Medicine, Department of Molecular and Clinical Medicine (P.-O.H., A.R.), Sahlgrenska Academy, University of Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Medicine Geriatrics and Emergency Medicine/Östra, Gothenburg, Sweden (P.-O.H., A.R.)
| | - Alicia K. Heath
- School of Public Health (A.K.H., I.T., E.R.), Imperial College London, UK
| | - Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.J.H.)
| | - Hironori Imano
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Suita, Japan (H.I.)
| | - Simerjot K. Jassal
- Department of Cancer Epidemiology (S.K.J., R.K., V.K.), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rudolf Kaaks
- Department of Cancer Epidemiology (S.K.J., R.K., V.K.), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Katzke
- Department of Cancer Epidemiology (S.K.J., R.K., V.K.), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jussi Kauhanen
- University of Eastern Finland (UEF), Kuopio, Finland (J.K.)
| | - Stefan Kiechl
- Department of Neurology & Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria (S.K.)
- Clinical Epidemiology Team, Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria (S.K., P.W.)
| | - Wolfgang Koenig
- Institute of Epidemiology and Medical Biometry, University of Ulm, Germany (W.K.)
- Deutsches Herzzentrum München, Technische Universität München, Germany (W.K.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (W.K.)
| | | | - Cecilie Kyrø
- Danish Cancer Society Research Center, Copenhagen, Denmark (C.K., A.T.)
| | - Deborah A. Lawlor
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, UK (D.A.L.)
- Population Health Science, Bristol Medical School, UK (D.A.L.)
| | - Börje Ljungberg
- Department of Surgical and Perioperative sciences, Urology and Andrology, Umeå University, Sweden (B.L.)
| | - Conor MacDonald
- University Paris-Saclay, UVSQ, Inserm, Villejuif, France (C. MacDonald)
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy (G.M.)
| | | | - Olle Melander
- Department of Clinical Sciences, Malmö, Lund University, Sweden (G.E., O.M.)
| | - Conchi Moreno Iribas
- Navarra Public Health Institute, IdiSNA, Pamplona, Spain (C.M.I.)
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Pamplona, Spain (C.M.I.)
| | - Toshiharu Ninomiya
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (T.N.)
| | | | - Børge G. Nordestgaard
- Herlev and Gentofte Hospital (B.G.N.), Copenhagen University Hospital, Copenhagen, Denmark
- Frederiksberg Hospital B.G.N.), Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences (B.G.N.), University of Copenhagen, Denmark
| | - Charlotte Onland-Moret
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands (C.O.-M., Y.T.v.d.S., W.M.M.V.)
| | - Luigi Palmieri
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Medical Research Council Biostatistics Unit (A.M.M., S. Burgess), University of Cambridge, UK
- Stroke Research Group, Department of Clinical Neurosciences (S. Bell), University of Cambridge, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
- MRC Epidemiology Unit, School of Clinical Medicine (C.L., N.W.), University of Cambridge, UK
- Division of Aging (K.C., S.P., J.P.C. J.M.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Cardiovascular Medicine (J.J., T.A.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Epidemiology, Boston University School of Public Health, MA (R.J.S.)
- Division of Nephrology, Department of Medicine (C.R.-C., E.A.A.), Vanderbilt University Medical Center, Nashville, TN
- Department of Biostatistics (R. Tao), Vanderbilt University Medical Center, Nashville, TN
- Medical Research Council Population Health Research Unit, Clinical Trial Service Unit and Epidemiological Studies Unit (N.S., C.B.), Nuffield Department of Population Health, University of Oxford, UK
- Cancer Epidemiology Unit (T.T.), Nuffield Department of Population Health, University of Oxford, UK
- Department of Cardiology, Trakya University School of Medicine, Edirne, Turkey (S.A.)
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, San Sebastián, Spain (P.A.)
- Biodonostia Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, San Sebastián, Spain (P.A.)
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain (P.A., A.G.d.l.C., D.P., C. Santiuste)
- Division of Clinical Epidemiology and Aging Research (V.A.), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Cancer Epidemiology (S.K.J., R.K., V.K.), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden (J.A., H.B.)
- School of Health and Social Studies, Dalarna University, Falun, Sweden (J.A.)
- Wellbeing & Preventable Chronic Diseases (WPCD) Division, Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia (E.L.M.B.)
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (E.L.M.B., M.I.)
- Institute of Medicine, School of Public Health and Community Medicine (C.B.), Sahlgrenska Academy, University of Gothenburg, Sweden
- Institute of Medicine, Department of Molecular and Clinical Medicine (P.-O.H., A.R.), Sahlgrenska Academy, University of Gothenburg, Sweden
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands (J.M.A.B., W.M.M.V.)
- Network Aging Research (NAR), Heidelberg University, Germany (H.B.)
- Studium Patavinum (E.C.), University of Padua, Italy
- Department of Medicine (V.T.), University of Padua, Italy
- Dipartimento di Salute Mentale e Fisica e Medicina Preventiva, Università degli Studi della Campania ‘Luigi Vanvitelli’, Caserta, Italy (P.C.)
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of London, UK (J.A.C.)
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (J.C.)
- Larner College of Medicine, The University of Vermont, Burlington (M.C.)
- The Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Tel Hashomer, Israel (R.D.)
- School of Public Health, Department of Epidemiology and Preventive Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel (R.D.)
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, NY (R.D., K.W.D.)
- Amsterdam University Medical Centers, VUMC, the Netherlands (R.T.d.J.)
- Department of Cardiovascular, Endocrine-metabolic Diseases and Aging, Istituto Superiore di Sanità, Rome, Italy (C.D., L. Palmer)
- Department of Clinical Sciences, Malmö, Lund University, Sweden (G.E., O.M.)
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France (H.F., E.W.)
- 12 Octubre Hospital Research Institute, Madrid, Spain (A.G.d,l,C.)
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland and Icelandic Heart Association, Kopavogur, Iceland (V.G.)
- Medical School Faculty of Health & Medical Sciences, The University of Western Australia, Perth, WA, Australia (G.J.H.)
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Medicine Geriatrics and Emergency Medicine/Östra, Gothenburg, Sweden (P.-O.H., A.R.)
- School of Public Health (A.K.H., I.T., E.R.), Imperial College London, UK
- The George Institute for Global Health (M.W.), Imperial College London, UK
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, the Netherlands (E.J.H.)
- Public Health, Department of Social Medicine, Osaka University Graduate School of Medicine, Suita, Japan (H.I.)
- University of Eastern Finland (UEF), Kuopio, Finland (J.K.)
- Department of Neurology & Neurosurgery, Medical University of Innsbruck, Innsbruck, Austria (S.K.)
- Clinical Epidemiology Team, Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria (S.K., P.W.)
- Institute of Epidemiology and Medical Biometry, University of Ulm, Germany (W.K.)
- Deutsches Herzzentrum München, Technische Universität München, Germany (W.K.)
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (W.K.)
- School of Public Health, University of Washington, Seattle (R.A.K.)
- Danish Cancer Society Research Center, Copenhagen, Denmark (C.K., A.T.)
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, UK (D.A.L.)
- Population Health Science, Bristol Medical School, UK (D.A.L.)
- Department of Surgical and Perioperative sciences, Urology and Andrology, Umeå University, Sweden (B.L.)
- University Paris-Saclay, UVSQ, Inserm, Villejuif, France (C. MacDonald)
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy (G.M.)
- Helmholtz Zentrum München, Munich, Germany (C. Meisinger)
- Navarra Public Health Institute, IdiSNA, Pamplona, Spain (C.M.I.)
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC), Pamplona, Spain (C.M.I.)
- Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (T.N.)
- London School of Hygiene & Tropical Medicine, UK (D.N.)
- Herlev and Gentofte Hospital (B.G.N.), Copenhagen University Hospital, Copenhagen, Denmark
- Frederiksberg Hospital B.G.N.), Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences (B.G.N.), University of Copenhagen, Denmark
- Department of Public Health (A.T.), University of Copenhagen, Denmark
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands (C.O.-M., Y.T.v.d.S., W.M.M.V.)
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain (D.P.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain (D.P.)
- Consejería de Sanidad del Principado de Asturias Oviedo, Asturias, Spain (J.R.Q.G.)
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital, Turin, Italy (C. Sacerdote)
- Department of Social and Environmental Medicine, Kanazawa Medical University, Uchinada, Japan (M.S.)
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Spain (C. Santiuste)
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany (M.B.S.)
- German Center for Diabetes Research (DZD), Neuherberg, Germany (M.B.S.)
- Institute of Nutritional Science, University of Potsdam, Germany (M.B.S.)
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy (S.S.)
- Department of Medical Sciences, Uppsala University, Sweden (J.S.)
- Hyblean Association for Epidemiological Reserach AIRE - ONLUS, Ragusa, Italy (R.T.)
- Universitätsmedizin Greifswald, Institut für Community Medicine, Abteilung SHIP/ Klinisch-Epidemiologische Forschung, Germany (H.V.)
- College of Public Health, University of Iowa (R.B.W.)
- University College London, UK (S.G.W.)
- The George Institute for Global Health, Camperdown, NSW, Australia (M.W.)
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Japan (K.Y.)
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain (R.Z.-R.)
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA (S.P.)
- Department of Biostatistics, Boston University School of Public Health, MA (D.R.G.)
- VA Pal Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, CA (P.S.T.)
- Medicine (Cardiovascular Medicine), Stanford University of School of Medicine, CA (P.S.T.)
- Office of Research and Development, Veterans Health Administration, Washington, DC (S.M.)
- Department of Veterans Affairs, Tennessee Valley Health Care System, Vanderbilt University, Nashville (T.L.E.)
- Medicine/Epidemiology, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN (T.L.E.)
- Department of Surgery, Corporal Michael Crescenz VA Medical Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.M.D.)
- Internal Medicine, VA Atlanta Healthcare System, Decatur, GA (P.W.F.W.)
- Emory University School of Medicine (Cardiology), Emory University, Atlanta, GA (P.W.F.W.)
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, Boston, MA (T.A.G.)
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
- The Alan Turing Institute, London, UK (M.I.)
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Germany (C.L.)
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK (J.D.)
- Division of Nephrology & Hypertension, Department of Medicine, Tennessee Valley Health Care System and Vanderbilt University Medical Center, Nashville (A.M.H.)
- Cambridge Centre for AI in Medicine, UK (A.M.W.)
- Health Data Science Centre, Human Technopole, Milan, Italy (E.D.A.)
| | - Dafina Petrova
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain (P.A., A.G.d.l.C., D.P., C. Santiuste)
- Escuela Andaluza de Salud Pública (EASP), Granada, Spain (D.P.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain (D.P.)
| | | | - Annika Rosengren
- Institute of Medicine, Department of Molecular and Clinical Medicine (P.-O.H., A.R.), Sahlgrenska Academy, University of Gothenburg, Sweden
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Medicine Geriatrics and Emergency Medicine/Östra, Gothenburg, Sweden (P.-O.H., A.R.)
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University-Hospital, Turin, Italy (C. Sacerdote)
| | - Masaru Sakurai
- Department of Social and Environmental Medicine, Kanazawa Medical University, Uchinada, Japan (M.S.)
| | - Carmen Santiuste
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain (P.A., A.G.d.l.C., D.P., C. Santiuste)
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Spain (C. Santiuste)
| | - Matthias B. Schulze
- German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany (M.B.S.)
- German Center for Diabetes Research (DZD), Neuherberg, Germany (M.B.S.)
- Institute of Nutritional Science, University of Potsdam, Germany (M.B.S.)
| | - Sabina Sieri
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano, Italy (S.S.)
| | - Johan Sundström
- Department of Medical Sciences, Uppsala University, Sweden (J.S.)
| | | | - Anne Tjønneland
- Danish Cancer Society Research Center, Copenhagen, Denmark (C.K., A.T.)
- Department of Public Health (A.T.), University of Copenhagen, Denmark
| | - Tammy Tong
- Cancer Epidemiology Unit (T.T.), Nuffield Department of Population Health, University of Oxford, UK
| | - Rosario Tumino
- Hyblean Association for Epidemiological Reserach AIRE - ONLUS, Ragusa, Italy (R.T.)
| | - Ioanna Tzoulaki
- School of Public Health (A.K.H., I.T., E.R.), Imperial College London, UK
| | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands (C.O.-M., Y.T.v.d.S., W.M.M.V.)
| | - W.M. Monique Verschuren
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands (J.M.A.B., W.M.M.V.)
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, the Netherlands (C.O.-M., Y.T.v.d.S., W.M.M.V.)
| | - Henry Völzke
- Universitätsmedizin Greifswald, Institut für Community Medicine, Abteilung SHIP/ Klinisch-Epidemiologische Forschung, Germany (H.V.)
| | | | | | - Elisabete Weiderpass
- International Agency for Research on Cancer (IARC), World Health Organization, Lyon, France (H.F., E.W.)
| | - Peter Willeit
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Clinical Epidemiology Team, Institute of Health Economics, Medical University of Innsbruck, Innsbruck, Austria (S.K., P.W.)
| | - Mark Woodward
- The George Institute for Global Health, Camperdown, NSW, Australia (M.W.)
| | - Kazumasa Yamagishi
- Department of Public Health Medicine, Faculty of Medicine, and Health Services Research and Development Center, University of Tsukuba, Japan (K.Y.)
| | - Raul Zamora-Ros
- Unit of Nutrition and Cancer, Epidemiology Research Program, Catalan Institute of Oncology, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat (Barcelona), Spain (R.Z.-R.)
| | - Elvis A. Akwo
- Division of Nephrology, Department of Medicine (C.R.-C., E.A.A.), Vanderbilt University Medical Center, Nashville, TN
| | - Saiju Pyarajan
- Division of Aging (K.C., S.P., J.P.C. J.M.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Data and Computational Sciences, VA Boston Healthcare System, Boston, MA (S.P.)
| | - David R. Gagnon
- Department of Biostatistics, Boston University School of Public Health, MA (D.R.G.)
| | - Philip S. Tsao
- VA Pal Alto Epidemiology Research and Information Center for Genomics, VA Palo Alto Health Care System, CA (P.S.T.)
- Medicine (Cardiovascular Medicine), Stanford University of School of Medicine, CA (P.S.T.)
| | - Sumitra Muralidhar
- Office of Research and Development, Veterans Health Administration, Washington, DC (S.M.)
| | - Todd L. Edwards
- Department of Veterans Affairs, Tennessee Valley Health Care System, Vanderbilt University, Nashville (T.L.E.)
- Medicine/Epidemiology, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN (T.L.E.)
| | - Scott M. Damrauer
- Department of Surgery, Corporal Michael Crescenz VA Medical Center and Perelman School of Medicine, University of Pennsylvania, Philadelphia (S.M.D.)
| | - Jacob Joseph
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- Division of Cardiovascular Medicine (J.J., T.A.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Lisa Pennells
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Peter W.F. Wilson
- Internal Medicine, VA Atlanta Healthcare System, Decatur, GA (P.W.F.W.)
- Emory University School of Medicine (Cardiology), Emory University, Atlanta, GA (P.W.F.W.)
| | - Seamus Harrison
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Thomas A. Gaziano
- Division of Cardiovascular Medicine (J.J., T.A.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Center for Health Decision Science, Harvard T.H. Chan School of Public Health, Boston, MA (T.A.G.)
| | - Michael Inouye
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia (E.L.M.B., M.I.)
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
- The Alan Turing Institute, London, UK (M.I.)
| | - Colin Baigent
- Institute of Medicine, School of Public Health and Community Medicine (C.B.), Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Juan P. Casas
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- Division of Aging (K.C., S.P., J.P.C. J.M.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Claudia Langenberg
- MRC Epidemiology Unit, School of Clinical Medicine (C.L., N.W.), University of Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Germany (C.L.)
| | - Nick Wareham
- MRC Epidemiology Unit, School of Clinical Medicine (C.L., N.W.), University of Cambridge, UK
| | - Elio Riboli
- The George Institute for Global Health (M.W.), Imperial College London, UK
| | - J.Michael Gaziano
- Massachusetts Veterans Epidemiology Research and Information Center (MAVERIC), VA Boston Healthcare System, Boston, MA (L.G., K.C., R.J.S., D.C.P., P.S., J.J., J.P.C., J.M.G.)
- Division of Aging (K.C., S.P., J.P.C. J.M.G.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - John Danesh
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK (J.D.)
| | - Adriana M. Hung
- Division of Nephrology & Hypertension, Department of Medicine, Tennessee Valley Health Care System and Vanderbilt University Medical Center, Nashville (A.M.H.)
| | - Adam S. Butterworth
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
| | - Angela M. Wood
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Cambridge Centre for AI in Medicine, UK (A.M.W.)
| | - Emanuele Di Angelantonio
- BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care (L.G., L.S., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., T.R.B., E.A., L.C., J.R.S., P.W., L. Pennells, S.H., M.I., J.D., A.S.B., A.M.W., E.D.A.)
- BHF Centre of Research Excellence, School of Clinical Medicine, Addenbrooke’s Hospital (A.M.M., S. Burgess, J.D., A.M.W., A.S.B., E.D.A.)
- Heart and Lung Research Institute, University of Cambridge, Cambridge UK (L.G., S. Bell, S.K.K., S. Burgess, K.M., A.M.M., E.A., L. Pennells, M.I., J.D., A.S.B., A.M.W., E.D.A.)
- NIHR Blood and Transplant Research Unit in Donor Health and Behaviour (S. Bell, T.R.B., E.A., J.D., A.S.B., A.M.W., E.D.A.), University of Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, UK (M.I., J.D., A.S.B., A.M.W., E.D.A.)
- Health Data Science Centre, Human Technopole, Milan, Italy (E.D.A.)
| |
Collapse
|
294
|
Yu Z, Wuttke M. Including APOL1 alleles and ancestry adjustments improve a genome-wide polygenic CKD score. Kidney Int 2022; 102:954-955. [PMID: 35985372 PMCID: PMC10018747 DOI: 10.1016/j.kint.2022.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 12/14/2022]
Affiliation(s)
- Zhi Yu
- Program of Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, Masschusetts, USA
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany.
| |
Collapse
|
295
|
Gulamali FF, Sawant AS, Nadkarni GN. Machine learning for risk stratification in kidney disease. Curr Opin Nephrol Hypertens 2022; 31:548-552. [PMID: 36004937 PMCID: PMC9529795 DOI: 10.1097/mnh.0000000000000832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
PURPOSE OF REVIEW Risk stratification for chronic kidney is becoming increasingly important as a clinical tool for both treatment and prevention measures. The goal of this review is to identify how machine learning tools contribute and facilitate risk stratification in the clinical setting. RECENT FINDINGS The two key machine learning paradigms to predictively stratify kidney disease risk are genomics-based and electronic health record based approaches. These methods can provide both quantitative information such as relative risk and qualitative information such as characterizing risk by subphenotype. SUMMARY The four key methods to stratify chronic kidney disease risk are genomics, multiomics, supervised and unsupervised machine learning methods. Polygenic risk scores utilize whole genome sequencing data to generate an individual's relative risk compared with the population. Multiomic methods integrate information from multiple biomarkers to generate trajectories and prognostic different outcomes. Supervised machine learning methods can directly utilize the growing compendia of electronic health records such as laboratory results and notes to generate direct risk predictions, while unsupervised machine learning methods can cluster individuals with chronic kidney disease into subphenotypes with differing approaches to care.
Collapse
|
296
|
Wang G, Xu Y, Wang Q, Chai Y, Sun X, Yang F, Zhang J, Wu M, Liao X, Yu X, Sheng X, Liu Z, Zhang J. Rare and undiagnosed diseases: From disease-causing gene identification to mechanism elucidation. FUNDAMENTAL RESEARCH 2022; 2:918-928. [PMID: 38933382 PMCID: PMC11197726 DOI: 10.1016/j.fmre.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/04/2022] [Accepted: 09/05/2022] [Indexed: 11/27/2022] Open
Abstract
Rare and undiagnosed diseases substantially decrease patient quality of life and have increasingly become a heavy burden on healthcare systems. Because of the challenges in disease-causing gene identification and mechanism elucidation, patients are often confronted with difficulty obtaining a precise diagnosis and treatment. Due to advances in sequencing and multiomics analysis approaches combined with patient-derived iPSC models and gene-editing platforms, substantial progress has been made in the diagnosis and treatment of rare and undiagnosed diseases. The aforementioned techniques also provide an operational basis for future precision medicine studies. In this review, we summarize recent progress in identifying disease-causing genes based on GWAS/WES/WGS-guided multiomics analysis approaches. In addition, we discuss recent advances in the elucidation of pathogenic mechanisms and treatment of diseases with state-of-the-art iPSC and organoid models, which are improved by cell maturation level and gene editing technology. The comprehensive strategies described above will generate a new paradigm of disease classification that will significantly promote the precision and efficiency of diagnosis and treatment for rare and undiagnosed diseases.
Collapse
Affiliation(s)
- Gang Wang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Yuyan Xu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qintao Wang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yi Chai
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiangwei Sun
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fan Yang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Jian Zhang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mengchen Wu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xufeng Liao
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaomin Yu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xin Sheng
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhihong Liu
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Zhejiang University School of Medicine, Hangzhou 310058, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu 210002, China
| | - Jin Zhang
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou 311121, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, The First Affiliated Hospital, Zhejiang University School of Medicine; Center of Gene/Cell Engineering and Genome Medicine of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
297
|
Zhou C, He P, Ye Z, Zhang Y, Zhang Y, Yang S, Wu Q, Liu M, Nie J, Qin X. Relationships of Serum 25-Hydroxyvitamin D Concentrations, Diabetes, Genetic Susceptibility, and New-Onset Chronic Kidney Disease. Diabetes Care 2022; 45:2518-2525. [PMID: 36102808 DOI: 10.2337/dc22-1194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/13/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The prospective relation of vitamin D status with the risk of chronic kidney diseases (CKD) remains uncertain. We aimed to examine the association of serum 25-hydroxyvitamin D (25OHD) with new-onset CKD in participants with and without diabetes at baseline and examine the potential modifications by genetic susceptibility on the association. RESEARCH DESIGN AND METHODS Included were 348,243 adults from the UK Biobank without prior CKD at baseline. Serum 25OHD concentrations were measured by chemiluminescent immunoassay method. Genetic risk score of CKD was calculated by 263 single nucleotide polymorphisms, which showed significant associations with estimated glomerular filtration rate. The primary outcome was new-onset CKD. RESULTS During a median follow-up duration of 12.1 years, 9,344 new-onset CKD were documented. Overall, there was a significant inverse association between baseline serum 25OHD and new-onset CKD in participants with diabetes (per SD increment, adjusted hazard ratio [HR] 0.91; 95% CI 0.86-0.96), but not in those without diabetes (per SD increment, adjusted HR 0.98; 95% CI 0.96-1.01; P-interaction between serum 25OHD and diabetes = 0.004). Accordingly, among participants with diabetes, compared with those baseline serum 25OHD <25 nmol/L, a significantly lower risk of new-onset CKD was found in those with 25OHD ≥50 nmol/L (adjusted HR 0.77; 95% CI 0.67-0.89). Moreover, the genetic risk of CKD did not significantly modify the association between baseline serum 25OHD and new-onset CKD among participants with diabetes (P-interaction = 0.127). CONCLUSIONS There was an inverse association between baseline serum 25OHD and new-onset CKD in participants with diabetes. The inverse association was not found in participants without diabetes.
Collapse
|
298
|
Mary S, Boder P, Padmanabhan S, McBride MW, Graham D, Delles C, Dominiczak AF. Role of Uromodulin in Salt-Sensitive Hypertension. Hypertension 2022; 79:2419-2429. [PMID: 36378920 PMCID: PMC9553220 DOI: 10.1161/hypertensionaha.122.19888] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The exclusive expression of uromodulin in the kidneys has made it an intriguing protein in kidney and cardiovascular research. Genome-wide association studies discovered variants of uromodulin that are associated with chronic kidney diseases and hypertension. Urinary and circulating uromodulin levels reflect kidney and cardiovascular health as well as overall mortality. More recently, Mendelian randomization studies have shown that genetically driven levels of uromodulin have a causal and adverse effect on kidney function. On a mechanistic level, salt sensitivity is an important factor in the pathophysiology of hypertension, and uromodulin is involved in salt reabsorption via the NKCC2 (Na+-K+-2Cl- cotransporter) on epithelial cells of the ascending limb of loop of Henle. In this review, we provide an overview of the multifaceted physiology and pathophysiology of uromodulin including recent advances in its genetics; cellular trafficking; and mechanistic and clinical studies undertaken to understand the complex relationship between uromodulin, blood pressure, and kidney function. We focus on tubular sodium reabsorption as one of the best understood and pathophysiologically and clinically most important roles of uromodulin, which can lead to therapeutic interventions.
Collapse
Affiliation(s)
- Sheon Mary
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Philipp Boder
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Sandosh Padmanabhan
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Martin W. McBride
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Delyth Graham
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Anna F. Dominiczak
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
299
|
Chen YC, Wong HSC, Wu MY, Chou WH, Kao CC, Chao CH, Chang WC, Wu MS. Genome-Wide Association Study for eGFR in a Taiwanese Population. Clin J Am Soc Nephrol 2022; 17:1598-1608. [PMID: 36223920 PMCID: PMC9718044 DOI: 10.2215/cjn.02180222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 09/16/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND OBJECTIVES Chronic kidney disease (CKD) is a global public health issue associated with large economic burdens. CKD contributes to higher risks of cardiovascular complications, kidney failure, and mortality. The incidence and prevalence rates of kidney failure in Taiwan have remained the highest in the world. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Assessing genetic factors that influence kidney function in specific populations has substantial clinical relevance. We investigated associations of genetic variants with eGFR. The quality control filtering and genotype imputation resulted in 10,008 Taiwan Biobank participants and 6,553,511 variants for final analyses. We examined these loci with in silico replication in individuals of European and African ancestry. RESULTS Our results revealed one significant locus (4q21.1) and three suggestive significant loci (17q23.2, 22q13.2, and 3q29) for eGFR in the Taiwanese population. In total, four conditional-independent single nucleotide polymorphisms were identified as the most important variants within these regions, including rs55948430 (Coiled-Coil Domain Containing 158), rs1010269 (BCAS3), rs56108505 (MKL1), and rs34796810 (upstream of DLG1). By performing a meta-analysis, we found that the 4q21.1 and 17q23.2 loci were successfully replicated in the European population, whereas only the 17q23.2 locus was replicated in African ancestry. Therefore, these two loci are suggested to be transethnic loci, and the other two eGFR-associated loci (22q13.2 and 3q29) are likely population specific. CONCLUSIONS We identified four susceptibility loci on 4q21.1, 17q23.2, 22q13.2, and 3q29 that associated with kidney-related traits in a Taiwanese population. The 22q13.2 (MKL1) and 3q29 (DLG1) were prioritized as critical candidates. Functional analyses delineated novel pathways related to kidney physiology in Taiwanese and East Asian ancestries.
Collapse
Affiliation(s)
- Ying-Chun Chen
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Henry Sung-Ching Wong
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Mei-Yi Wu
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Primary Care Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
| | - Wan-Hsuan Chou
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chih-Chin Kao
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
| | - Ching-Hsuan Chao
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chiao Chang
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Department of Pharmacy, Taipei Medical University–Wan Fang Hospital, Taipei, Taiwan
- Integrative Research Center for Critical Care, Department of Pharmacy, Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Mai-Szu Wu
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Taipei Medical University Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
300
|
Surendran P, Stewart ID, Au Yeung VPW, Pietzner M, Raffler J, Wörheide MA, Li C, Smith RF, Wittemans LBL, Bomba L, Menni C, Zierer J, Rossi N, Sheridan PA, Watkins NA, Mangino M, Hysi PG, Di Angelantonio E, Falchi M, Spector TD, Soranzo N, Michelotti GA, Arlt W, Lotta LA, Denaxas S, Hemingway H, Gamazon ER, Howson JMM, Wood AM, Danesh J, Wareham NJ, Kastenmüller G, Fauman EB, Suhre K, Butterworth AS, Langenberg C. Rare and common genetic determinants of metabolic individuality and their effects on human health. Nat Med 2022; 28:2321-2332. [PMID: 36357675 PMCID: PMC9671801 DOI: 10.1038/s41591-022-02046-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 09/16/2022] [Indexed: 11/12/2022]
Abstract
Garrod's concept of 'chemical individuality' has contributed to comprehension of the molecular origins of human diseases. Untargeted high-throughput metabolomic technologies provide an in-depth snapshot of human metabolism at scale. We studied the genetic architecture of the human plasma metabolome using 913 metabolites assayed in 19,994 individuals and identified 2,599 variant-metabolite associations (P < 1.25 × 10-11) within 330 genomic regions, with rare variants (minor allele frequency ≤ 1%) explaining 9.4% of associations. Jointly modeling metabolites in each region, we identified 423 regional, co-regulated, variant-metabolite clusters called genetically influenced metabotypes. We assigned causal genes for 62.4% of these genetically influenced metabotypes, providing new insights into fundamental metabolite physiology and clinical relevance, including metabolite-guided discovery of potential adverse drug effects (DPYD and SRD5A2). We show strong enrichment of inborn errors of metabolism-causing genes, with examples of metabolite associations and clinical phenotypes of non-pathogenic variant carriers matching characteristics of the inborn errors of metabolism. Systematic, phenotypic follow-up of metabolite-specific genetic scores revealed multiple potential etiological relationships.
Collapse
Affiliation(s)
- Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | | | - Maik Pietzner
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Raffler
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Digital Medicine, University Hospital of Augsburg, Augsburg, Germany
| | - Maria A Wörheide
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Chen Li
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Rebecca F Smith
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Laura B L Wittemans
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Big Data Institute, University of Oxford, Oxford, UK
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Lorenzo Bomba
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Open Targets, Wellcome Genome Campus, Hinxton, UK
| | - Cristina Menni
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Jonas Zierer
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Niccolò Rossi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | | | | | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Pirro G Hysi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Health Data Science Research Centre, Human Technopole, Milan, Italy
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Nicole Soranzo
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Open Targets, Wellcome Genome Campus, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | | | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Luca A Lotta
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Spiros Denaxas
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
- British Heart Foundation Data Science Centre, London, UK
| | - Harry Hemingway
- Institute of Health Informatics, University College London, London, UK
- Health Data Research UK, London, UK
| | - Eric R Gamazon
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Clare Hall & MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| | - Angela M Wood
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
- The Alan Turing Institute, London, UK
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| | - Nicholas J Wareham
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Gabi Kastenmüller
- Institute of Computational Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Eric B Fauman
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Karsten Suhre
- Department of Biophysics and Physiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Hinxton, UK.
- NIHR Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK.
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK.
- Computational Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|