1
|
Lee GY, Lee J, Kim JH, Chung KM, Han SN. Impact of recognition of genetic information related to BMI on changes in physical activity, dietary intake, and blood cholesterol level: a randomized controlled trial. Eur J Nutr 2025; 64:190. [PMID: 40423790 PMCID: PMC12116609 DOI: 10.1007/s00394-025-03713-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 05/11/2025] [Indexed: 05/28/2025]
Abstract
PURPOSE Genes associated with body mass index (BMI), including FTO rs9939609,MC4R rs17782313, and BDNF rs6265, may influence BMI and regulate energy metabolism. While previous studies have explored health-related behavior changes, few have investigated both biochemical and behavior changes resulting from perceived genetic risk. This study investigated whether recognizing BMI-related genes affects health-related behaviors and alters blood metabolite levels. METHODS Normal and overweight adults aged 25-35 years (n = 100) were randomly assigned to an intervention group (n = 65) informed about BMI-related genetic information (FTO rs9939609, MC4R rs17782313, BDNF rs6265) and an uninformed group (n = 35, CON). The intervention group was further divided into Intervention-high risk (IHR, n = 36) and intervention-low risk (ILR, n = 29) subgroups. Dietary intake and physical activity (PA) were assessed using a 3-day dietary record and the IPAQ-short form. Blood metabolites were analyzed through multivariate analyses to identify significant differences among the groups, with measurements taken at baseline, 3 months, and 6 months. RESULTS The IHR group exhibited increased dietary fat and fast foods intake, along with enhanced vigorous and moderate PA. Six metabolites were selected as biomarkers that were distinguishable among groups, and the relative serum cholesterol levels significantly decreased in the IHR group at 3 months. CONCLUSION These results demonstrate that recognizing the BMI-associated genetic risk resulted in a short-term increase in PA but did not improve dietary intake. Increased PA was significantly associated with reduced cholesterol concentration, suggesting the clinical importance of physical activity in the genetically at-risk group. CLINICAL TRIAL AND STUDY REGISTRATION This study was reviewed and approved by the Seoul National University Institutional Review Board (IRB #1901/001-004) and registered on the Clinical Research Information Service (CRIS), KCT0004650 ( https://cris.nih.go.kr/cris/search/detailSearch.do /14091, 2020/01/28).
Collapse
Affiliation(s)
- Ga Young Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Junghak Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jeong-Han Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | | | - Sung Nim Han
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Feuerbacher JF, Cheng R, Sedliak M, Hu M, Finni TJ, Umlauff L, Schumann M, Cheng S. Serum Metabolome Signature Response to Different Types of Resistance Training. Int J Sports Med 2025; 46:22-31. [PMID: 39255827 DOI: 10.1055/a-2412-3410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Pneumatic resistance training (PRT) facilitates a longer time under tension that might lead to greater changes in body composition when compared to traditional resistance training (TRT), possibly enhancing serum metabolite concentrations indicative of healthy metabolic function. To assess the impact of PRT and TRT on muscular strength, body composition, and serum metabolome, 69 men (age: 31.8±7.2 years, height: 179.7±5.4 cm, weight: 81.1±9.9 kg) were randomized into two 10-week intervention groups (PRT:n=24 and TRT:n=24) and one control group (CON:n=21). Serum metabolite concentrations were assessed before and after the training intervention by high-throughput nuclear magnetic resonance. Fat mass and lean mass were obtained by bioimpedance analysis. The training intervention resulted in an increase in lean mass for both PRT (1.85±2.69%; p=0.003) and TRT (2.72±4.53%; p=0.004), while only PRT reduced statistically significantly in body fat percentage (PRT: -5.08±10.76%; p=0.019). Only in PRT and TRT significant increases in small high-density lipoproteins (S-HDL-L) and small HDL particles (S-HDL-P) were observed. When controlling for fat and lean mass, the effects on S-HDL-L/S-HDL-P diminished. Network analysis may suggest that PRT and TRT result in an increase in network connectivity and robustness. It appears that the observed improvements are associated with changes in body composition.
Collapse
Affiliation(s)
- Joshua Frederik Feuerbacher
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
- Department of Sports Medicine and Exercise Therapy, Chemnitz University of Technology, Chemnitz, Germany
| | - Runtan Cheng
- Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
| | - Milan Sedliak
- Department of Biological and Medical Sciences, Faculty of Physical Education, Comenius University in Bratislava, Bratislava, Slovakia
| | - Min Hu
- Guangzhou Sport University, Guangzhou Sport University, Guangzhou, China
| | - Taija Juutinen Finni
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
| | - Lisa Umlauff
- Department of Molecular and Cellular Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Moritz Schumann
- Department of Sports Medicine and Exercise Therapy, Chemnitz University of Technology, Chemnitz, Germany
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shulin Cheng
- Faculty of Physical Education, Shanghai Jiao Tong University, Shanghai, China
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyvaskyla, Finland
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Karjalainen MK, Karthikeyan S, Oliver-Williams C, Sliz E, Allara E, Fung WT, Surendran P, Zhang W, Jousilahti P, Kristiansson K, Salomaa V, Goodwin M, Hughes DA, Boehnke M, Fernandes Silva L, Yin X, Mahajan A, Neville MJ, van Zuydam NR, de Mutsert R, Li-Gao R, Mook-Kanamori DO, Demirkan A, Liu J, Noordam R, Trompet S, Chen Z, Kartsonaki C, Li L, Lin K, Hagenbeek FA, Hottenga JJ, Pool R, Ikram MA, van Meurs J, Haller T, Milaneschi Y, Kähönen M, Mishra PP, Joshi PK, Macdonald-Dunlop E, Mangino M, Zierer J, Acar IE, Hoyng CB, Lechanteur YTE, Franke L, Kurilshikov A, Zhernakova A, Beekman M, van den Akker EB, Kolcic I, Polasek O, Rudan I, Gieger C, Waldenberger M, Asselbergs FW, Hayward C, Fu J, den Hollander AI, Menni C, Spector TD, Wilson JF, Lehtimäki T, Raitakari OT, Penninx BWJH, Esko T, Walters RG, Jukema JW, Sattar N, Ghanbari M, Willems van Dijk K, Karpe F, McCarthy MI, Laakso M, Järvelin MR, Timpson NJ, Perola M, Kooner JS, Chambers JC, van Duijn C, Slagboom PE, Boomsma DI, Danesh J, Ala-Korpela M, Butterworth AS, Kettunen J. Genome-wide characterization of circulating metabolic biomarkers. Nature 2024; 628:130-138. [PMID: 38448586 PMCID: PMC10990933 DOI: 10.1038/s41586-024-07148-y] [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: 11/07/2022] [Accepted: 02/01/2024] [Indexed: 03/08/2024]
Abstract
Genome-wide association analyses using high-throughput metabolomics platforms have led to novel insights into the biology of human metabolism1-7. This detailed knowledge of the genetic determinants of systemic metabolism has been pivotal for uncovering how genetic pathways influence biological mechanisms and complex diseases8-11. Here we present a genome-wide association study for 233 circulating metabolic traits quantified by nuclear magnetic resonance spectroscopy in up to 136,016 participants from 33 cohorts. We identify more than 400 independent loci and assign probable causal genes at two-thirds of these using manual curation of plausible biological candidates. We highlight the importance of sample and participant characteristics that can have significant effects on genetic associations. We use detailed metabolic profiling of lipoprotein- and lipid-associated variants to better characterize how known lipid loci and novel loci affect lipoprotein metabolism at a granular level. We demonstrate the translational utility of comprehensively phenotyped molecular data, characterizing the metabolic associations of intrahepatic cholestasis of pregnancy. Finally, we observe substantial genetic pleiotropy for multiple metabolic pathways and illustrate the importance of careful instrument selection in Mendelian randomization analysis, revealing a putative causal relationship between acetone and hypertension. Our publicly available results provide a foundational resource for the community to examine the role of metabolism across diverse diseases.
Collapse
Affiliation(s)
- Minna K Karjalainen
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland.
- Northern Finland Birth Cohorts, Arctic Biobank, Infrastructure for Population Studies, Faculty of Medicine, University of Oulu, Oulu, Finland.
| | - Savita Karthikeyan
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Clare Oliver-Williams
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Public Health Specialty Training Programme, Cambridge, UK
| | - Eeva Sliz
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Elias Allara
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Wing Tung Fung
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, London, UK
| | - Pekka Jousilahti
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Kati Kristiansson
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Veikko Salomaa
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Matt Goodwin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - David A Hughes
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Lilian Fernandes Silva
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Jiangsu, China
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Genentech, South San Francisco, CA, USA
| | - Matt J Neville
- NIHR Oxford Biomedical Research Centre, OUHFT Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Natalie R van Zuydam
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Ayse Demirkan
- Surrey Institute for People-Centred AI, University of Surrey, Guildford, UK
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford, UK
| | - Jun Liu
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Zhengming Chen
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Christiana Kartsonaki
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - Liming Li
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Peking University Center for Public Health and Epidemic Preparedness and Response, Beijing, China
- Key Laboratory of Epidemiology of Major Diseases, Peking University, Ministry of Education, Beijing, China
| | - Kuang Lin
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Fiona A Hagenbeek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Joyce van Meurs
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Toomas Haller
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital, Tampere, Finland
| | - Pashupati P Mishra
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Peter K Joshi
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Erin Macdonald-Dunlop
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- NIHR Biomedical Research Centre at Guy's and St Thomas' Foundation Trust, London, UK
| | - Jonas Zierer
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ilhan E Acar
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Yara T E Lechanteur
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marian Beekman
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik B van den Akker
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
- Center for Computational Biology, Leiden University Medical Center, Leiden, The Netherlands
- The Delft Bioinformatics Lab, Delft University of Technology, Delft, The Netherlands
| | - Ivana Kolcic
- Department of Public Health, School of Medicine, University of Split, Split, Croatia
| | - Ozren Polasek
- Department of Public Health, School of Medicine, University of Split, Split, Croatia
| | - Igor Rudan
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Folkert W Asselbergs
- Amsterdam University Medical Centers, Department of Cardiology, University of Amsterdam, Amsterdam, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Genomics Research Center, Abbvie, Cambridge, MA, USA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - James F Wilson
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, Scotland
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Terho Lehtimäki
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam Neuroscience and Amsterdam Public Health, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Tonu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Robin G Walters
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- MRC Population Health Research Unit, University of Oxford, Oxford, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Division Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Fredrik Karpe
- NIHR Oxford Biomedical Research Centre, OUHFT Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Genentech, South San Francisco, CA, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Kuopio University Hospital, Kuopio, Finland
| | - Marjo-Riitta Järvelin
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, UK
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
| | - Nicholas J Timpson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Markus Perola
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Jaspal S Kooner
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Cardiology, Ealing Hospital, London North West University Healthcare NHS Trust, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development (AR&D) Research Institute, Amsterdam, The Netherlands
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Mika Ala-Korpela
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research Blood and Transplant Research Unit in Donor Health and Behaviour, University of Cambridge, Cambridge, UK
- Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, University of Cambridge, Cambridge, UK
- Health Data Research UK Cambridge, Wellcome Genome Campus and University of Cambridge, Cambridge, UK
| | - Johannes Kettunen
- Systems Epidemiology, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| |
Collapse
|
4
|
Louck LE, Cara KC, Klatt K, Wallace TC, Chung M. The Relationship of Circulating Choline and Choline-Related Metabolite Levels with Health Outcomes: A Scoping Review of Genome-Wide Association Studies and Mendelian Randomization Studies. Adv Nutr 2024; 15:100164. [PMID: 38128611 PMCID: PMC10819410 DOI: 10.1016/j.advnut.2023.100164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023] Open
Abstract
Choline is essential for proper liver, muscle, brain, lipid metabolism, cellular membrane composition, and repair. Understanding genetic determinants of circulating choline metabolites can help identify new determinants of choline metabolism, requirements, and their link to disease endpoints. We conducted a scoping review to identify studies assessing the association of genetic polymorphisms on circulating choline and choline-related metabolite concentrations and subsequent associations with health outcomes. This study follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement scoping review extension. Literature was searched to September 28, 2022, in 4 databases: Embase, MEDLINE, Web of Science, and the Biological Science Index. Studies of any duration in humans were considered. Any genome-wide association study (GWAS) investigating genetic variant associations with circulating choline and/or choline-related metabolites and any Mendelian randomization (MR) study investigating the association of genetically predicted circulating choline and/or choline-related metabolites with any health outcome were considered. Qualitative evidence is presented in summary tables. From 1248 total reviewed articles, 53 were included (GWAS = 27; MR = 26). Forty-two circulating choline-related metabolites were tested in association with genetic variants in GWAS studies, primarily trimethylamine N-oxide, betaine, sphingomyelins, lysophosphatidylcholines, and phosphatidylcholines. MR studies investigated associations between 52 total unique choline metabolites and 66 unique health outcomes. Of these, 47 significant associations were reported between 16 metabolites (primarily choline, lysophosphatidylcholines, phosphatidylcholines, betaine, and sphingomyelins) and 27 health outcomes including cancer, cardiovascular, metabolic, bone, and brain-related outcomes. Some articles reported significant associations between multiple choline types and the same health outcome. Genetically predicted circulating choline and choline-related metabolite concentrations are associated with a wide variety of health outcomes. Further research is needed to assess how genetic variability influences choline metabolism and whether individuals with lower genetically predicted circulating choline and choline-related metabolite concentrations would benefit from a dietary intervention or supplementation.
Collapse
Affiliation(s)
- Lauren E Louck
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States
| | - Kelly C Cara
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States
| | - Kevin Klatt
- Nutritional Sciences and Toxicology, University of California, Berkeley, CA, United States
| | - Taylor C Wallace
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States; Think Health Group, Inc, Washington, DC, United States
| | - Mei Chung
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States.
| |
Collapse
|
5
|
Ottensmann L, Tabassum R, Ruotsalainen SE, Gerl MJ, Klose C, Widén E, Simons K, Ripatti S, Pirinen M. Genome-wide association analysis of plasma lipidome identifies 495 genetic associations. Nat Commun 2023; 14:6934. [PMID: 37907536 PMCID: PMC10618167 DOI: 10.1038/s41467-023-42532-8] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 10/13/2023] [Indexed: 11/02/2023] Open
Abstract
The human plasma lipidome captures risk for cardiometabolic diseases. To discover new lipid-associated variants and understand the link between lipid species and cardiometabolic disorders, we perform univariate and multivariate genome-wide analyses of 179 lipid species in 7174 Finnish individuals. We fine-map the associated loci, prioritize genes, and examine their disease links in 377,277 FinnGen participants. We identify 495 genome-trait associations in 56 genetic loci including 8 novel loci, with a considerable boost provided by the multivariate analysis. For 26 loci, fine-mapping identifies variants with a high causal probability, including 14 coding variants indicating likely causal genes. A phenome-wide analysis across 953 disease endpoints reveals disease associations for 40 lipid loci. For 11 coronary artery disease risk variants, we detect strong associations with lipid species. Our study demonstrates the power of multivariate genetic analysis in correlated lipidomics data and reveals genetic links between diseases and lipid species beyond the standard lipids.
Collapse
Affiliation(s)
- Linda Ottensmann
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Rubina Tabassum
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanni E Ruotsalainen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | | | - Elisabeth Widén
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | | | - Samuli Ripatti
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Matti Pirinen
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland.
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
6
|
Lee GY, Chung KM, Lee J, Kim JH, Han SN. Changes in anxiety and depression levels and meat intake following recognition of low genetic risk for high body mass index, triglycerides, and lipoproteins: A randomized controlled trial. PLoS One 2023; 18:e0291052. [PMID: 37683016 PMCID: PMC10490956 DOI: 10.1371/journal.pone.0291052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND Psychological status affects dietary intake, and recognizing genetic information can lead to behavior changes by influencing psychological factors such as anxiety or depression. OBJECTIVES In this study, we examined the effects of disclosing genetic information on anxiety or depression levels and the association between these psychological factors and dietary intake. METHODS A total of 100 healthy adults were randomly assigned to an intervention group (n = 65) informed about their genetic test results regarding body mass index and lipid profiles (triglyceride and cholesterol concentrations) and a not-informed control group (CON, n = 35). Based on polygenic risk scores, participants in the intervention group were subclassified into an intervention-low risk (ILR, n = 32) and an intervention-high risk (IHR, n = 33) group. Nutrient and food intakes were assessed via a 3-day dietary record at baseline and at 3 and 6 months. Depression and anxiety levels were measured using PHQ-9 and GAD-7 questionnaires, and the relative levels of blood metabolites were measure using GC-MS/MS analysis. RESULTS Noticeable changes in dietary intake as well as psychological factors were observed in male subjects, with those perceiving their genetic risks as low (ILR) showing a significant increase in protein intake at 3 months compared to baseline (ILR: 3.9 ± 1.4, p<0.05). Meat intake also increased significantly in males in the ILR group at 3 months, but not in the IHR and CON groups (ILR: 49.4 ± 30.8, IHR: -52.2 ± 25.4, CON: -5.3 ± 30.3 g/d). ILR group showed a significant decrease in anxiety levels at 3 months, and their anxiety scores showed a negative association with meat intake (standardized β = -0.321, p<0.05). The meat intake at 3 months was associated with the relative levels of arginine and ornithine (standardized β = 0.452, p<0.05 and standardized β = 0.474, p<0.05, respectively). CONCLUSIONS Taken together, anxiety levels were decreased in male subjects who perceived their genetic risk to be low, and the decrease in anxiety levels was associated with an increase in meat intake. This suggests that recognizing genetic information may affect psychological factors and dietary intake.
Collapse
Affiliation(s)
- Ga Young Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Korea
| | | | - Junghak Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Jeong-Han Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Korea
| | - Sung Nim Han
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul, Korea
- Research Institute of Human Ecology, Seoul National University, Seoul, Korea
| |
Collapse
|
7
|
Sandholm N, Hotakainen R, Haukka JK, Jansson Sigfrids F, Dahlström EH, Antikainen AA, Valo E, Syreeni A, Kilpeläinen E, Kytölä A, Palotie A, Harjutsalo V, Forsblom C, Groop PH. Whole-exome sequencing identifies novel protein-altering variants associated with serum apolipoprotein and lipid concentrations. Genome Med 2022; 14:132. [PMID: 36419110 PMCID: PMC9685920 DOI: 10.1186/s13073-022-01135-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Dyslipidemia is a major risk factor for cardiovascular disease, and diabetes impacts the lipid metabolism through multiple pathways. In addition to the standard lipid measurements, apolipoprotein concentrations provide added awareness of the burden of circulating lipoproteins. While common genetic variants modestly affect the serum lipid concentrations, rare genetic mutations can cause monogenic forms of hypercholesterolemia and other genetic disorders of lipid metabolism. We aimed to identify low-frequency protein-altering variants (PAVs) affecting lipoprotein and lipid traits. METHODS We analyzed whole-exome (WES) and whole-genome sequencing (WGS) data of 481 and 474 individuals with type 1 diabetes, respectively. The phenotypic data consisted of 79 serum lipid and apolipoprotein phenotypes obtained with clinical laboratory measurements and nuclear magnetic resonance spectroscopy. RESULTS The single-variant analysis identified an association between the LIPC p.Thr405Met (rs113298164) and serum apolipoprotein A1 concentrations (p=7.8×10-8). The burden of PAVs was significantly associated with lipid phenotypes in LIPC, RBM47, TRMT5, GTF3C5, MARCHF10, and RYR3 (p<2.9×10-6). The RBM47 gene is required for apolipoprotein B post-translational modifications, and in our data, the association between RBM47 and apolipoprotein C-III concentrations was due to a rare 21 base pair p.Ala496-Ala502 deletion; in replication, the burden of rare deleterious variants in RBM47 was associated with lower triglyceride concentrations in WES of >170,000 individuals from multiple ancestries (p=0.0013). Two PAVs in GTF3C5 were highly enriched in the Finnish population and associated with cardiovascular phenotypes in the general population. In the previously known APOB gene, we identified novel associations at two protein-truncating variants resulting in lower serum non-HDL cholesterol (p=4.8×10-4), apolipoprotein B (p=5.6×10-4), and LDL cholesterol (p=9.5×10-4) concentrations. CONCLUSIONS We identified lipid and apolipoprotein-associated variants in the previously known LIPC and APOB genes, as well as PAVs in GTF3C5 associated with LDLC, and in RBM47 associated with apolipoprotein C-III concentrations, implicated as an independent CVD risk factor. Identification of rare loss-of-function variants has previously revealed genes that can be targeted to prevent CVD, such as the LDL cholesterol-lowering loss-of-function variants in the PCSK9 gene. Thus, this study suggests novel putative therapeutic targets for the prevention of CVD.
Collapse
Affiliation(s)
- Niina Sandholm
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Ronja Hotakainen
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jani K Haukka
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Fanny Jansson Sigfrids
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Emma H Dahlström
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anni A Antikainen
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Erkka Valo
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anna Syreeni
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Elina Kilpeläinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Anastasia Kytölä
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Department of Neurology and Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- The Stanley Center for Psychiatric Research and Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Valma Harjutsalo
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Carol Forsblom
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Research Center, Biomedicum Helsinki, Haartmaninkatu 8, Helsinki, 00290, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
8
|
Tahir UA, Katz DH, Avila-Pachecho J, Bick AG, Pampana A, Robbins JM, Yu Z, Chen ZZ, Benson MD, Cruz DE, Ngo D, Deng S, Shi X, Zheng S, Eisman AS, Farrell L, Hall ME, Correa A, Tracy RP, Durda P, Taylor KD, Liu Y, Johnson WC, Guo X, Yao J, Chen YDI, Manichaikul AW, Ruberg FL, Blaner WS, Jain D, Bouchard C, Sarzynski MA, Rich SS, Rotter JI, Wang TJ, Wilson JG, Clish CB, Natarajan P, Gerszten RE. Whole Genome Association Study of the Plasma Metabolome Identifies Metabolites Linked to Cardiometabolic Disease in Black Individuals. Nat Commun 2022; 13:4923. [PMID: 35995766 PMCID: PMC9395431 DOI: 10.1038/s41467-022-32275-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 07/25/2022] [Indexed: 01/27/2023] Open
Abstract
Integrating genetic information with metabolomics has provided new insights into genes affecting human metabolism. However, gene-metabolite integration has been primarily studied in individuals of European Ancestry, limiting the opportunity to leverage genomic diversity for discovery. In addition, these analyses have principally involved known metabolites, with the majority of the profiled peaks left unannotated. Here, we perform a whole genome association study of 2,291 metabolite peaks (known and unknown features) in 2,466 Black individuals from the Jackson Heart Study. We identify 519 locus-metabolite associations for 427 metabolite peaks and validate our findings in two multi-ethnic cohorts. A significant proportion of these associations are in ancestry specific alleles including findings in APOE, TTR and CD36. We leverage tandem mass spectrometry to annotate unknown metabolites, providing new insight into hereditary diseases including transthyretin amyloidosis and sickle cell disease. Our integrative omics approach leverages genomic diversity to provide novel insights into diverse cardiometabolic diseases.
Collapse
Affiliation(s)
- Usman A Tahir
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Daniel H Katz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | | | | | - Akhil Pampana
- Broad Institute of Harvard and MIT, Cambridge, MA, US
| | - Jeremy M Robbins
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Zhi Yu
- Broad Institute of Harvard and MIT, Cambridge, MA, US
| | - Zsu-Zsu Chen
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Mark D Benson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Daniel E Cruz
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Debby Ngo
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Shuliang Deng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Xu Shi
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Shuning Zheng
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Aaron S Eisman
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Laurie Farrell
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Michael E Hall
- University of Mississippi Medical Center, Jackson, MS, US
| | - Adolfo Correa
- University of Mississippi Medical Center, Jackson, MS, US
| | - Russell P Tracy
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, US
| | - Peter Durda
- Department of Pathology Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, US
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, US
| | - Yongmei Liu
- Department of Medicine, Division of Cardiology, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, US
| | - W Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, US
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, US
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, US
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, US
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, US
- Division of Biostatistics and Epidemiology, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, US
| | - Frederick L Ruberg
- Section of Cardiovascular Medicine, Boston University School of Medicine and Boston Medical Center, Boston, MA, US
| | | | - Deepti Jain
- University of Washington, Seattle, Washington, US
| | - Claude Bouchard
- Human Genomic Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, US
| | - Mark A Sarzynski
- Department of Exercise Science, University of South Carolina, Columbia, SC, US
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, US
- Division of Biostatistics and Epidemiology, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, US
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor UCLA Medical Center, Torrance, CA, US
| | - Thomas J Wang
- Department of Medicine, UT Southwestern Medical Center, Dallas, TX, US
| | - James G Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US
| | - Clary B Clish
- Broad Institute of Harvard and MIT, Cambridge, MA, US
| | - Pradeep Natarajan
- Broad Institute of Harvard and MIT, Cambridge, MA, US
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, US
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, US.
- Broad Institute of Harvard and MIT, Cambridge, MA, US.
| |
Collapse
|
9
|
Ala-Korpela M, Zhao S, Järvelin MR, Mäkinen VP, Ohukainen P. Apt interpretation of comprehensive lipoprotein data in large-scale epidemiology: disclosure of fundamental structural and metabolic relationships. Int J Epidemiol 2022; 51:996-1011. [PMID: 34405869 PMCID: PMC9189959 DOI: 10.1093/ije/dyab156] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/09/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Quantitative lipoprotein analytics using nuclear magnetic resonance (NMR) spectroscopy is currently commonplace in large-scale studies. One methodology has become widespread and is currently being utilized also in large biobanks. It allows the comprehensive characterization of 14 lipoprotein subclasses, clinical lipids, apolipoprotein A-I and B. The details of these data are conceptualized here in relation to lipoprotein metabolism with particular attention on the fundamental characteristics of subclass particle numbers, lipid concentrations and compositional measures. METHODS AND RESULTS The NMR methodology was applied to fasting serum samples from Northern Finland Birth Cohorts 1966 and 1986 with 5651 and 5605 participants, respectively. All results were highly consistent between the cohorts. Circulating lipid concentrations in a particular lipoprotein subclass arise predominantly as the result of the circulating number of those subclass particles. The spherical lipoprotein particle shape, with a radially oriented surface monolayer, imposes size-dependent biophysical constraints for the lipid composition of individual subclass particles and inherently restricts the accommodation of metabolic changes via compositional modifications. The new finding that the relationship between lipoprotein subclass particle concentrations and the particle size is log-linear reveals that circulating lipoprotein particles are also under rather strict metabolic constraints for both their absolute and relative concentrations. CONCLUSIONS The fundamental structural and metabolic relationships between lipoprotein subclasses elucidated in this study empower detailed interpretation of lipoprotein metabolism. Understanding the intricate details of these extensive data is important for the precise interpretation of novel therapeutic opportunities and for fully utilizing the potential of forthcoming analyses of genetic and metabolic data in large biobanks.
Collapse
Affiliation(s)
- Mika Ala-Korpela
- Corresponding author. Computational Medicine, Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland. E-mail:
| | - Siyu Zhao
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, UK
| | - Ville-Petteri Mäkinen
- Australian Centre for Precision Health, University of South Australia, Adelaide, Australia
- Computational and Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
10
|
Li-Gao R, Hughes DA, van Klinken JB, de Mutsert R, Rosendaal FR, Mook-Kanamori DO, Timpson NJ, Willems van Dijk K. Genetic Studies of Metabolomics Change After a Liquid Meal Illuminate Novel Pathways for Glucose and Lipid Metabolism. Diabetes 2021; 70:2932-2946. [PMID: 34610981 DOI: 10.2337/db21-0397] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022]
Abstract
Humans spend the greater part of the day in a postprandial state. However, the genetic basis of postprandial blood measures is relatively uncharted territory. We examined the genetics of variation in concentrations of postprandial metabolites (t = 150 min) in response to a liquid mixed meal through genome-wide association studies (GWAS) performed in the Netherlands Epidemiology of Obesity (NEO) study (n = 5,705). The metabolite response GWAS identified an association between glucose change and rs10830963:G in the melatonin receptor 1B (β [SE] -0.23 [0.03], P = 2.15 × 10-19). In addition, the ANKRD55 locus led by rs458741:C showed strong associations with extremely large VLDL (XXLVLDL) particle response (XXLVLDL total cholesterol: β [SE] 0.17 [0.03], P = 5.76 × 10-10; XXLVLDL cholesterol ester: β [SE] 0.17 [0.03], P = 9.74 × 10-10), which also revealed strong associations with body composition and diabetes in the UK Biobank (P < 5 × 10-8). Furthermore, the associations between XXLVLDL response and insulinogenic index, HOMA-β, Matsuda insulin sensitivity index, and HbA1c in the NEO study implied the role of chylomicron synthesis in diabetes (with false discovery rate-corrected q <0.05). To conclude, genetic studies of metabolomics change after a liquid meal illuminate novel pathways for glucose and lipid metabolism. Further studies are warranted to corroborate biological pathways of the ANKRD55 locus underlying diabetes.
Collapse
Affiliation(s)
- Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - David A Hughes
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Jan B van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Renée de Mutsert
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, the Netherlands
| | - Nicholas J Timpson
- Medical Research Council Integrative Epidemiology Unit, School of Social and Community Medicine, University of Bristol, Bristol, U.K
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Division of Endocrinology, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
11
|
Colaco K, Lee KA, Akhtari S, Winer R, Welsh P, Sattar N, McInnes IB, Chandran V, Harvey P, Cook RJ, Gladman DD, Piguet V, Eder L. Targeted metabolomic profiling and prediction of cardiovascular events: a prospective study of patients with psoriatic arthritis and psoriasis. Ann Rheum Dis 2021; 80:1429-1435. [PMID: 34049856 DOI: 10.1136/annrheumdis-2021-220168] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In patients with psoriatic disease (PsD), we sought serum metabolites associated with cardiovascular (CV) events and investigated whether they could improve CV risk prediction beyond traditional risk factors and the Framingham Risk Score (FRS). METHODS Nuclear magnetic resonance metabolomics identified biomarkers for incident CV events in patients with PsD. The association of each metabolite with incident CV events was analysed using Cox proportional hazards regression models first adjusted for age and sex, and subsequently for traditional CV risk factors. Variable selection was performed using penalisation with boosting after adjusting for age and sex, and the FRS. RESULTS Among 977 patients with PsD, 70 patients had incident CV events. In Cox regression models adjusted for CV risk factors, alanine, tyrosine, degree of unsaturation of fatty acids and high-density lipoprotein particles were associated with decreased CV risk. Glycoprotein acetyls, apolipoprotein B and cholesterol remnants were associated with increased CV risk. The age-adjusted and sex-adjusted expanded model with 13 metabolites significantly improved prediction of CV events beyond the model with age and sex alone, with an area under the receiver operator characteristic curve (AUC) of 79.9 versus 72.6, respectively (p=0.02). Compared with the FRS alone (AUC=73.9), the FRS-adjusted expanded model with 11 metabolites (AUC=75.0, p=0.72) did not improve CV risk discrimination. CONCLUSIONS We identify novel metabolites associated with the development of CV events in patients with PsD. Further study of their underlying causal role may clarify important pathways leading to CV events in this population.
Collapse
Affiliation(s)
- Keith Colaco
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
| | - Ker-Ai Lee
- Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Shadi Akhtari
- Department of Cardiology, Women's College Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Raz Winer
- Department of Neurology, Rambam Health Care Campus, Haifa, Israel
| | - Paul Welsh
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Naveed Sattar
- BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Vinod Chandran
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Paula Harvey
- Department of Cardiology, Women's College Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Richard J Cook
- Department of Statistics and Actuarial Science, University of Waterloo, Waterloo, Ontario, Canada
| | - Dafna D Gladman
- Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Vincent Piguet
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lihi Eder
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Reilly SK, Gosai SJ, Gutierrez A, Mackay-Smith A, Ulirsch JC, Kanai M, Mouri K, Berenzy D, Kales S, Butler GM, Gladden-Young A, Bhuiyan RM, Stitzel ML, Finucane HK, Sabeti PC, Tewhey R. Direct characterization of cis-regulatory elements and functional dissection of complex genetic associations using HCR-FlowFISH. Nat Genet 2021; 53:1166-1176. [PMID: 34326544 PMCID: PMC8925018 DOI: 10.1038/s41588-021-00900-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/23/2021] [Indexed: 12/26/2022]
Abstract
Effective interpretation of genome function and genetic variation requires a shift from epigenetic mapping of cis-regulatory elements (CREs) to characterization of endogenous function. We developed hybridization chain reaction fluorescence in situ hybridization coupled with flow cytometry (HCR-FlowFISH), a broadly applicable approach to characterize CRISPR-perturbed CREs via accurate quantification of native transcripts, alongside CRISPR activity screen analysis (CASA), a hierarchical Bayesian model to quantify CRE activity. Across >325,000 perturbations, we provide evidence that CREs can regulate multiple genes, skip over the nearest gene and display activating and/or silencing effects. At the cholesterol-level-associated FADS locus, we combine endogenous screens with reporter assays to exhaustively characterize multiple genome-wide association signals, functionally nominate causal variants and, importantly, identify their target genes.
Collapse
Affiliation(s)
- Steven K Reilly
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for System Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| | - Sager J Gosai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for System Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Harvard Graduate Program in Biological and Biomedical Science, Boston, MA, USA
| | | | | | - Jacob C Ulirsch
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biological and Biomedical Science, Boston, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Masahiro Kanai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Bioinformatics and Integrative Genomics, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Gina M Butler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut, Farmington, CT, USA
- Institute of Systems Genomics, University of Connecticut, Farmington, CT, USA
| | - Hilary K Finucane
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Pardis C Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for System Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Ryan Tewhey
- The Jackson Laboratory, Bar Harbor, ME, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA.
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
13
|
Helgadottir A, Thorleifsson G, Stefansson K. Increased absorption of phytosterols is the simplest and most plausible explanation for coronary artery disease risk not accounted for by non-HDL cholesterol in high cholesterol absorbers. Eur Heart J 2021; 42:283-284. [PMID: 33167008 PMCID: PMC7819517 DOI: 10.1093/eurheartj/ehaa902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Anna Helgadottir
- deCODE Genetics/Amgen, Inc., Population Genomics, Sturlugata 8, Reykjavik, 102, Iceland; and
| | - Gudmar Thorleifsson
- deCODE Genetics/Amgen, Inc., Population Genomics, Sturlugata 8, Reykjavik, 102, Iceland; and
| | - Kari Stefansson
- deCODE Genetics/Amgen, Inc., Population Genomics, Sturlugata 8, Reykjavik, 102, Iceland; and.,Faculty of Medicine, University of Iceland, Vatnsmyrarvegur, 101 Reykjavik, Iceland
| |
Collapse
|
14
|
Genomic-Metabolomic Associations Support the Role of LIPC and Glycerophospholipids in Age-Related Macular Degeneration. OPHTHALMOLOGY SCIENCE 2021; 1. [PMID: 34382031 PMCID: PMC8353724 DOI: 10.1016/j.xops.2021.100017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Purpose Large-scale genome-wide association studies (GWAS) have reported important single nucleotide polymorphisms (SNPs) with significant associations with age-related macular degeneration (AMD). However, their role in disease development remains elusive. This study aimed to assess SNP–metabolite associations (i.e., metabolite quantitative trait loci [met-QTL]) and to provide insights into the biological mechanisms of AMD risk SNPs. Design Cross-sectional multicenter study (Boston, Massachusetts, and Coimbra, Portugal). Participants Patients with AMD (n = 388) and control participants (n = 98) without any vitreoretinal disease (> 50 years). Methods Age-related macular degeneration grading was performed using color fundus photographs according to the Age-Related Eye Disease Study classification scheme. Fasting blood samples were collected and evaluated with mass spectrometry for metabolomic profiling and Illumina OmniExpress for SNPs profiling. Analyses of met-QTL of endogenous metabolites were conducted using linear regression models adjusted for age, gender, smoking, 10 metabolite principal components (PCs), and 10 SNP PCs. Additionally, we analyzed the cumulative effect of AMD risk SNPs on plasma metabolites by generating genetic risk scores and assessing their associations with metabolites using linear regression models, accounting for the same covariates. Modeling was performed first for each cohort, and then combined by meta-analysis. Multiple comparisons were accounted for using the false discovery rate (FDR). Main Outcome Measures Plasma metabolite levels associated with AMD risk SNPs. Results After quality control, data for 544 plasma metabolites were included. Meta-analysis of data from all individuals (AMD patients and control participants) identified 28 significant met-QTL (β = 0.016–0.083; FDR q-value < 1.14 × 10–2), which corresponded to 5 metabolites and 2 genes: ASPM and LIPC. Polymorphisms in the LIPC gene were associated with phosphatidylethanolamine metabolites, which are glycerophospholipids, and polymorphisms in the ASPM gene with branched-chain amino acids. Similar results were observed when considering only patients with AMD. Genetic risk score–metabolite associations further supported a global impact of AMD risk SNPs on the plasma metabolome. Conclusions This study demonstrated that genomic–metabolomic associations can provide insights into the biological relevance of AMD risk SNPs. In particular, our results support that the LIPC gene and the glycerophospholipid metabolic pathway may play an important role in AMD, thus offering new potential therapeutic targets for this disease.
Collapse
|
15
|
Tabassum R, Ripatti S. Integrating lipidomics and genomics: emerging tools to understand cardiovascular diseases. Cell Mol Life Sci 2021; 78:2565-2584. [PMID: 33449144 PMCID: PMC8004487 DOI: 10.1007/s00018-020-03715-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and morbidity worldwide leading to 31% of all global deaths. Early prediction and prevention could greatly reduce the enormous socio-economic burden posed by CVDs. Plasma lipids have been at the center stage of the prediction and prevention strategies for CVDs that have mostly relied on traditional lipids (total cholesterol, total triglycerides, HDL-C and LDL-C). The tremendous advancement in the field of lipidomics in last two decades has facilitated the research efforts to unravel the metabolic dysregulation in CVDs and their genetic determinants, enabling the understanding of pathophysiological mechanisms and identification of predictive biomarkers, beyond traditional lipids. This review presents an overview of the application of lipidomics in epidemiological and genetic studies and their contributions to the current understanding of the field. We review findings of these studies and discuss examples that demonstrates the potential of lipidomics in revealing new biology not captured by traditional lipids and lipoprotein measurements. The promising findings from these studies have raised new opportunities in the fields of personalized and predictive medicine for CVDs. The review further discusses prospects of integrating emerging genomics tools with the high-dimensional lipidome to move forward from the statistical associations towards biological understanding, therapeutic target development and risk prediction. We believe that integrating genomics with lipidome holds a great potential but further advancements in statistical and computational tools are needed to handle the high-dimensional and correlated lipidome.
Collapse
Affiliation(s)
- Rubina Tabassum
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, PO Box 20, 00014, Helsinki, Finland.
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, PO Box 20, 00014, Helsinki, Finland.
- Department of Public Health, Clinicum, University of Helsinki, Helsinki, Finland.
- Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
16
|
Coltell O, Sorlí JV, Asensio EM, Barragán R, González JI, Giménez-Alba IM, Zanón-Moreno V, Estruch R, Ramírez-Sabio JB, Pascual EC, Ortega-Azorín C, Ordovas JM, Corella D. Genome-Wide Association Study for Serum Omega-3 and Omega-6 Polyunsaturated Fatty Acids: Exploratory Analysis of the Sex-Specific Effects and Dietary Modulation in Mediterranean Subjects with Metabolic Syndrome. Nutrients 2020; 12:E310. [PMID: 31991592 PMCID: PMC7071282 DOI: 10.3390/nu12020310] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 12/13/2022] Open
Abstract
Many early studies presented beneficial effects of polyunsaturated fatty acids (PUFA) on cardiovascular risk factors and disease. However, results from recent meta-analyses indicate that this effect would be very low or nil. One of the factors that may contribute to the inconsistency of the results is that, in most studies, genetic factors have not been taken into consideration. It is known that fatty acid desaturase (FADS) gene cluster in chromosome 11 is a very important determinant of plasma PUFA, and that the prevalence of the single nucleotide polymorphisms (SNPs) varies greatly between populations and may constitute a bias in meta-analyses. Previous genome-wide association studies (GWAS) have been carried out in other populations and none of them have investigated sex and Mediterranean dietary pattern interactions at the genome-wide level. Our aims were to undertake a GWAS to discover the genes most associated with serum PUFA concentrations (omega-3, omega-6, and some fatty acids) in a scarcely studied Mediterranean population with metabolic syndrome, and to explore sex and adherence to Mediterranean diet (MedDiet) interactions at the genome-wide level. Serum PUFA were determined by NMR spectroscopy. We found strong robust associations between various SNPs in the FADS cluster and omega-3 concentrations (top-ranked in the adjusted model: FADS1-rs174547, p = 3.34 × 10-14; FADS1-rs174550, p = 5.35 × 10-14; FADS2-rs1535, p = 5.85 × 10-14; FADS1-rs174546, p = 6.72 × 10-14; FADS2-rs174546, p = 9.75 × 10-14; FADS2- rs174576, p = 1.17 × 10-13; FADS2-rs174577, p = 1.12 × 10-12, among others). We also detected a genome-wide significant association with other genes in chromosome 11: MYRF (myelin regulatory factor)-rs174535, p = 1.49 × 10-12; TMEM258 (transmembrane protein 258)-rs102275, p = 2.43 × 10-12; FEN1 (flap structure-specific endonuclease 1)-rs174538, p = 1.96 × 10-11). Similar genome-wide statistically significant results were found for docosahexaenoic fatty acid (DHA). However, no such associations were detected for omega-6 PUFAs or linoleic acid (LA). For total PUFA, we observed a consistent gene*sex interaction with the DNTTIP2 (deoxynucleotidyl transferase terminal interacting protein 2)-rs3747965 p = 1.36 × 10-8. For adherence to MedDiet, we obtained a relevant interaction with the ME1 (malic enzyme 1) gene (a gene strongly regulated by fat) in determining serum omega-3. The top-ranked SNP for this interaction was ME1-rs3798890 (p = 2.15 × 10-7). In the regional-wide association study, specifically focused on the FADS1/FASD2/FADS3 and ELOVL (fatty acid elongase) 2/ELOVL 5 regions, we detected several statistically significant associations at p < 0.05. In conclusion, our results confirm a robust role of the FADS cluster on serum PUFA in this population, but the associations vary depending on the PUFA. Moreover, the detection of some sex and diet interactions underlines the need for these associations/interactions to be studied in all specific populations so as to better understand the complex metabolism of PUFA.
Collapse
Affiliation(s)
- Oscar Coltell
- Department of Computer Languages and Systems, Universitat Jaume I, 12071 Castellón, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
| | - Jose V. Sorlí
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Eva M. Asensio
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Rocío Barragán
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - José I. González
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Ignacio M. Giménez-Alba
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Vicente Zanón-Moreno
- Area of Health Sciences, Valencian International University, 46002 Valencia, Spain;
- Red Temática de Investigación Cooperativa en Patología Ocular (OFTARED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Ophthalmology Research Unit “Santiago Grisolia”, Dr. Peset University Hospital, 46017 Valencia, Spain
| | - Ramon Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Internal Medicine, Hospital Clinic, Institut d’Investigació Biomèdica August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | | | - Eva C. Pascual
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
- Assisted Reproduction Unit of the University Hospital of Valencia, 46010 Valencia, Spain
| | - Carolina Ortega-Azorín
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Jose M. Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111 USA;
- Department of Cardiovascular Epidemiology and Population Genetics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- IMDEA Alimentación, 28049 Madrid, Spain
| | - Dolores Corella
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 28029 Madrid, Spain; (J.V.S.); (E.M.A.); (R.B.); (J.I.G.); (I.M.G.-A.); (R.E.); (C.O.-A.)
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, 46010 Valencia, Spain;
| |
Collapse
|
17
|
Hagenbeek FA, Pool R, van Dongen J, Draisma HHM, Jan Hottenga J, Willemsen G, Abdellaoui A, Fedko IO, den Braber A, Visser PJ, de Geus EJCN, Willems van Dijk K, Verhoeven A, Suchiman HE, Beekman M, Slagboom PE, van Duijn CM, Harms AC, Hankemeier T, Bartels M, Nivard MG, Boomsma DI. Heritability estimates for 361 blood metabolites across 40 genome-wide association studies. Nat Commun 2020; 11:39. [PMID: 31911595 PMCID: PMC6946682 DOI: 10.1038/s41467-019-13770-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/25/2019] [Indexed: 01/16/2023] Open
Abstract
Metabolomics examines the small molecules involved in cellular metabolism. Approximately 50% of total phenotypic differences in metabolite levels is due to genetic variance, but heritability estimates differ across metabolite classes. We perform a review of all genome-wide association and (exome-) sequencing studies published between November 2008 and October 2018, and identify >800 class-specific metabolite loci associated with metabolite levels. In a twin-family cohort (N = 5117), these metabolite loci are leveraged to simultaneously estimate total heritability (h2total), and the proportion of heritability captured by known metabolite loci (h2Metabolite-hits) for 309 lipids and 52 organic acids. Our study reveals significant differences in h2Metabolite-hits among different classes of lipids and organic acids. Furthermore, phosphatidylcholines with a high degree of unsaturation have higher h2Metabolite-hits estimates than phosphatidylcholines with low degrees of unsaturation. This study highlights the importance of common genetic variants for metabolite levels, and elucidates the genetic architecture of metabolite classes.
Collapse
Affiliation(s)
- Fiona A Hagenbeek
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
| | - René Pool
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Jenny van Dongen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Harmen H M Draisma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jouke Jan Hottenga
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gonneke Willemsen
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Abdel Abdellaoui
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Psychiatry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Iryna O Fedko
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anouk den Braber
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, VU Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Pieter Jelle Visser
- Alzheimer Center Amsterdam, Department of Neurology, VU Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
- Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Alzheimer Center Limburg, Maastricht University, Maastricht, The Netherlands
| | - Eco J C N de Geus
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Ko Willems van Dijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - H Eka Suchiman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marian Beekman
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - P Eline Slagboom
- Department of Biomedical Data Sciences, Section of Molecular Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Amy C Harms
- Division of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University and The Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Leiden Academic Center for Drug Research, Leiden University and The Netherlands Metabolomics Centre, Leiden, The Netherlands
| | - Meike Bartels
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Michel G Nivard
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Dorret I Boomsma
- Department of Biological Psychology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
18
|
Kettunen J, Holmes MV, Allara E, Anufrieva O, Ohukainen P, Oliver-Williams C, Wang Q, Tillin T, Hughes AD, Kähönen M, Lehtimäki T, Viikari J, Raitakari OT, Salomaa V, Järvelin MR, Perola M, Davey Smith G, Chaturvedi N, Danesh J, Di Angelantonio E, Butterworth AS, Ala-Korpela M. Lipoprotein signatures of cholesteryl ester transfer protein and HMG-CoA reductase inhibition. PLoS Biol 2019; 17:e3000572. [PMID: 31860674 PMCID: PMC6944381 DOI: 10.1371/journal.pbio.3000572] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 01/06/2020] [Accepted: 11/29/2019] [Indexed: 02/04/2023] Open
Abstract
Cholesteryl ester transfer protein (CETP) inhibition reduces vascular event risk, but confusion surrounds its effects on low-density lipoprotein (LDL) cholesterol. Here, we clarify associations of genetic inhibition of CETP on detailed lipoprotein measures and compare those to genetic inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR). We used an allele associated with lower CETP expression (rs247617) to mimic CETP inhibition and an allele associated with lower HMGCR expression (rs12916) to mimic the well-known effects of statins for comparison. The study consists of 65,427 participants of European ancestries with detailed lipoprotein subclass profiling from nuclear magnetic resonance spectroscopy. Genetic associations were scaled to 10% reduction in relative risk of coronary heart disease (CHD). We also examined observational associations of the lipoprotein subclass measures with risk of incident CHD in 3 population-based cohorts totalling 616 incident cases and 13,564 controls during 8-year follow-up. Genetic inhibition of CETP and HMGCR resulted in near-identical associations with LDL cholesterol concentration estimated by the Friedewald equation. Inhibition of HMGCR had relatively consistent associations on lower cholesterol concentrations across all apolipoprotein B-containing lipoproteins. In contrast, the associations of the inhibition of CETP were stronger on lower remnant and very-low-density lipoprotein (VLDL) cholesterol, but there were no associations on cholesterol concentrations in LDL defined by particle size (diameter 18–26 nm) (−0.02 SD LDL defined by particle size; 95% CI: −0.10 to 0.05 for CETP versus −0.24 SD, 95% CI −0.30 to −0.18 for HMGCR). Inhibition of CETP was strongly associated with lower proportion of triglycerides in all high-density lipoprotein (HDL) particles. In observational analyses, a higher triglyceride composition within HDL subclasses was associated with higher risk of CHD, independently of total cholesterol and triglycerides (strongest hazard ratio per 1 SD higher triglyceride composition in very large HDL 1.35; 95% CI: 1.18–1.54). In conclusion, CETP inhibition does not appear to affect size-specific LDL cholesterol but is likely to lower CHD risk by lowering concentrations of other atherogenic, apolipoprotein B-containing lipoproteins (such as remnant and VLDLs). Inhibition of CETP also lowers triglyceride composition in HDL particles, a phenomenon reflecting combined effects of circulating HDL, triglycerides, and apolipoprotein B-containing particles and is associated with a lower CHD risk in observational analyses. Our results reveal that conventional composite lipid assays may mask heterogeneous effects of emerging lipid-altering therapies. Inhibition of cholesteryl ester transfer protein does not affect size-specific low-density lipoprotein cholesterol, but may lower coronary heart disease risk by lowering cholesterol concentrations in other apolipoprotein-B containing atherogenic lipoproteins, and by lowering triglyceride content of high-density lipoprotein particles.
Collapse
Affiliation(s)
- Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- National Institute for Health and Welfare, Helsinki, Finland
| | - Michael V. Holmes
- Medical Research Council Population Health Research Unit, University of Oxford, Oxford, United Kingdom
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
- National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, United Kingdom
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
| | - Elias Allara
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, United Kingdom
| | - Olga Anufrieva
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Clare Oliver-Williams
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- Homerton College, University of Cambridge, Cambridge, United Kingdom
| | - Qin Wang
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Therese Tillin
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Alun D. Hughes
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere University Hospital, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technologies, University of Tampere, Tampere, Finland
| | - Jorma Viikari
- Department of Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
| | - Olli T. Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, United Kingdom
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, Finland
- Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
- Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Nish Chaturvedi
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - John Danesh
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
- British Heart Foundation Cambridge Centre of Excellence, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Emanuele Di Angelantonio
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, United Kingdom
| | - Adam S. Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
- National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, United Kingdom
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
19
|
Ohukainen P, Kuusisto S, Kettunen J, Perola M, Järvelin MR, Mäkinen VP, Ala-Korpela M. Data-driven multivariate population subgrouping via lipoprotein phenotypes versus apolipoprotein B in the risk assessment of coronary heart disease. Atherosclerosis 2019; 294:10-15. [PMID: 31931463 DOI: 10.1016/j.atherosclerosis.2019.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/02/2019] [Accepted: 12/12/2019] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND AIMS Population subgrouping has been suggested as means to improve coronary heart disease (CHD) risk assessment. We explored here how unsupervised data-driven metabolic subgrouping, based on comprehensive lipoprotein subclass data, would work in large-scale population cohorts. METHODS We applied a self-organizing map (SOM) artificial intelligence methodology to define subgroups based on detailed lipoprotein profiles in a population-based cohort (n = 5789) and utilised the trained SOM in an independent cohort (n = 7607). We identified four SOM-based subgroups of individuals with distinct lipoprotein profiles and CHD risk and compared those to univariate subgrouping by apolipoprotein B quartiles. RESULTS The SOM-based subgroup with highest concentrations for non-HDL measures had the highest, and the subgroup with lowest concentrations, the lowest risk for CHD. However, apolipoprotein B quartiles produced better resolution of risk than the SOM-based subgroups and also striking dose-response behaviour. CONCLUSIONS These results suggest that the majority of lipoprotein-mediated CHD risk is explained by apolipoprotein B-containing lipoprotein particles. Therefore, even advanced multivariate subgrouping, with comprehensive data on lipoprotein metabolism, may not advance CHD risk assessment.
Collapse
Affiliation(s)
- Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sanna Kuusisto
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland; NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland; National Institute for Health and Welfare, Helsinki, Finland
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, Finland; Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland; Estonian Genome Center, University of Tartu, Tartu, Estonia
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland; Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland; Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK; Department of Life Sciences, College of Health and Life Sciences, Brunel University London, UK
| | - Ville-Petteri Mäkinen
- Computational and Systems Biology Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Australia; Hopwood Centre for Neurobiology, Lifelong Health Theme, SAHMRI, Australia
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland; NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
20
|
Gallois A, Mefford J, Ko A, Vaysse A, Julienne H, Ala-Korpela M, Laakso M, Zaitlen N, Pajukanta P, Aschard H. A comprehensive study of metabolite genetics reveals strong pleiotropy and heterogeneity across time and context. Nat Commun 2019; 10:4788. [PMID: 31636271 PMCID: PMC6803661 DOI: 10.1038/s41467-019-12703-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
Genetic studies of metabolites have identified thousands of variants, many of which are associated with downstream metabolic and obesogenic disorders. However, these studies have relied on univariate analyses, reducing power and limiting context-specific understanding. Here we aim to provide an integrated perspective of the genetic basis of metabolites by leveraging the Finnish Metabolic Syndrome In Men (METSIM) cohort, a unique genetic resource which contains metabolic measurements, mostly lipids, across distinct time points as well as information on statin usage. We increase effective sample size by an average of two-fold by applying the Covariates for Multi-phenotype Studies (CMS) approach, identifying 588 significant SNP-metabolite associations, including 228 new associations. Our analysis pinpoints a small number of master metabolic regulator genes, balancing the relative proportion of dozens of metabolite levels. We further identify associations to changes in metabolic levels across time as well as genetic interactions with statin at both the master metabolic regulator and genome-wide level.
Collapse
Affiliation(s)
- Apolline Gallois
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France
| | - Joel Mefford
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Arthur Ko
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Amaury Vaysse
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France
| | - Hanna Julienne
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France
| | - Mika Ala-Korpela
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Noah Zaitlen
- Department of Medicine, University of California, San Francisco, CA, USA.
| | - Päivi Pajukanta
- Department of Human Genetics, University of California, Los Angeles, CA, USA.
| | - Hugues Aschard
- Department of Computational Biology - USR 3756 CNRS, Institut Pasteur, Paris, France.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
21
|
Deelen J, Kettunen J, Fischer K, van der Spek A, Trompet S, Kastenmüller G, Boyd A, Zierer J, van den Akker EB, Ala-Korpela M, Amin N, Demirkan A, Ghanbari M, van Heemst D, Ikram MA, van Klinken JB, Mooijaart SP, Peters A, Salomaa V, Sattar N, Spector TD, Tiemeier H, Verhoeven A, Waldenberger M, Würtz P, Davey Smith G, Metspalu A, Perola M, Menni C, Geleijnse JM, Drenos F, Beekman M, Jukema JW, van Duijn CM, Slagboom PE. A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals. Nat Commun 2019; 10:3346. [PMID: 31431621 PMCID: PMC6702196 DOI: 10.1038/s41467-019-11311-9] [Citation(s) in RCA: 213] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/08/2019] [Indexed: 11/09/2022] Open
Abstract
Predicting longer-term mortality risk requires collection of clinical data, which is often cumbersome. Therefore, we use a well-standardized metabolomics platform to identify metabolic predictors of long-term mortality in the circulation of 44,168 individuals (age at baseline 18-109), of whom 5512 died during follow-up. We apply a stepwise (forward-backward) procedure based on meta-analysis results and identify 14 circulating biomarkers independently associating with all-cause mortality. Overall, these associations are similar in men and women and across different age strata. We subsequently show that the prediction accuracy of 5- and 10-year mortality based on a model containing the identified biomarkers and sex (C-statistic = 0.837 and 0.830, respectively) is better than that of a model containing conventional risk factors for mortality (C-statistic = 0.772 and 0.790, respectively). The use of the identified metabolic profile as a predictor of mortality or surrogate endpoint in clinical studies needs further investigation.
Collapse
Affiliation(s)
- Joris Deelen
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands. .,Max Planck Institute for Biology of Ageing, PO Box 41 06 23, 50866, Cologne, Germany.
| | - Johannes Kettunen
- National Institute for Health and Welfare, PO Box 30, 00271, Helsinki, Finland.,Computational Medicine, Center for Life Course Health Research and Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland
| | - Krista Fischer
- The Estonian Genome Center, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Ashley van der Spek
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Stella Trompet
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.,German Center for Diabetes Research (DZD), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.,Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Strand, London, WC2R 2LS, UK
| | - Andy Boyd
- ALSPAC, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Jonas Zierer
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.,Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Strand, London, WC2R 2LS, UK.,Novartis Institutes for BioMedical Research, Novartis Campus, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - Erik B van den Akker
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,The Delft Bioinformatics Lab, Delft University of Technology, PO Box 5031, 2600 GA, Delft, The Netherlands
| | - Mika Ala-Korpela
- Computational Medicine, Center for Life Course Health Research and Biocenter Oulu, University of Oulu, PO Box 5000, 90014, Oulu, Finland.,Systems Epidemiology, Baker Heart and Diabetes Institute, PO Box 6492, Melbourne Victoria, 3004, Australia.,Population Health Science, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.,MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1C, Kuopio, 70210, Finland.,Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, The Alfred Hospital, Monash University, Melbourne, Victoria, 3800, Australia
| | - Najaf Amin
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Ayse Demirkan
- Section of Statistical Multi-omics, Department of Clinical and Experimental research, University of Surrey, Guildford, Surrey, GU2 7XH, UK.,Department of Genetics, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, PO Box 91735-951, 9133913716, Mashhad, Iran
| | - Diana van Heemst
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Neurology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Jan Bert van Klinken
- Department of Human Genetics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Simon P Mooijaart
- Department of Internal Medicine, section of Gerontology and Geriatrics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Annette Peters
- German Center for Diabetes Research (DZD), Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.,Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Veikko Salomaa
- National Institute for Health and Welfare, PO Box 30, 00271, Helsinki, Finland
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Strand, London, WC2R 2LS, UK
| | - Henning Tiemeier
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Psychiatry, Erasmus University Medical Center-Sophia Children's Hospital, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Aswin Verhoeven
- Center for Proteomics and Metabolomics, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Melanie Waldenberger
- Institute of Epidemiology II, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany.,Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Ingolstaedter Landstraße 1, 85764, Neuherberg, Germany
| | - Peter Würtz
- Nightingale Health Ltd., Mannerheimintie 164a, 00300, Helsinki, Finland
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Andres Metspalu
- The Estonian Genome Center, University of Tartu, Riia 23b, 51010, Tartu, Estonia.,Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 23b - 134, 51010, Tartu, Estonia
| | - Markus Perola
- Institute for Molecular Medicine Finland, University of Helsinki, Tukholmankatu 8, 00290, Helsinki, Finland.,Clinical and Molecular Metabolism Research Program, Faculty of Medicine, University of Helsinki, PO Box 63, 00014, Helsinki, Finland
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, St Thomas' Hospital, Strand, London, WC2R 2LS, UK
| | - Johanna M Geleijnse
- Division of Human Nutrition, Wageningen University, PO Box 17, 6700 AA, Wageningen, The Netherlands
| | - Fotios Drenos
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.,Department of Life Sciences, Brunel University London, Uxbridge, UB8 3PH, UK
| | - Marian Beekman
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Leiden Academic Centre for Drug Research, Leiden University, PO box 9502, 2300 RA, Leiden, The Netherlands
| | - P Eline Slagboom
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, The Netherlands.
| |
Collapse
|
22
|
Abstract
Extensive research demonstrates unequivocally that nutrition plays a fundamental role in maintaining health and preventing disease. In parallel nutrition research provides evidence that the risks and benefits of diet and lifestyle choices do not affect people equally, as people are inherently variable in their responses to nutrition and associated interventions to maintain health and prevent disease. To simplify the inherent complexity of human subjects and their nutrition, with the aim of managing expectations for dietary guidance required to ensure healthy populations and individuals, nutrition researchers often seek to group individuals based on commonly used criteria. This strategy relies on demonstrating meaningful conclusions based on comparison of group mean responses of assigned groups. Such studies are often confounded by the heterogeneous nutrition response. Commonly used criteria applied in grouping study populations and individuals to identify mechanisms and determinants of responses to nutrition often contribute to the problem of interpreting the results of group comparisons. Challenges of interpreting the group mean using diverse populations will be discussed with respect to studies in human subjects, in vivo and in vitro model systems. Future advances in nutrition research to tackle inter-individual variation require a coordinated approach from funders, learned societies, nutrition scientists, publishers and reviewers of the scientific literature. This will be essential to develop and implement improved study design, data recording, analysis and reporting to facilitate more insightful interpretation of the group mean with respect to population diversity and the heterogeneous nutrition response.
Collapse
|
23
|
Tynkkynen T, Wang Q, Ekholm J, Anufrieva O, Ohukainen P, Vepsäläinen J, Männikkö M, Keinänen-Kiukaanniemi S, Holmes MV, Goodwin M, Ring S, Chambers JC, Kooner J, Järvelin MR, Kettunen J, Hill M, Davey Smith G, Ala-Korpela M. Proof of concept for quantitative urine NMR metabolomics pipeline for large-scale epidemiology and genetics. Int J Epidemiol 2019; 48:978-993. [PMID: 30689875 PMCID: PMC6659374 DOI: 10.1093/ije/dyy287] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Quantitative molecular data from urine are rare in epidemiology and genetics. NMR spectroscopy could provide these data in high throughput, and it has already been applied in epidemiological settings to analyse urine samples. However, quantitative protocols for large-scale applications are not available. METHODS We describe in detail how to prepare urine samples and perform NMR experiments to obtain quantitative metabolic information. Semi-automated quantitative line shape fitting analyses were set up for 43 metabolites and applied to data from various analytical test samples and from 1004 individuals from a population-based epidemiological cohort. Novel analyses on how urine metabolites associate with quantitative serum NMR metabolomics data (61 metabolic measures; n = 995) were performed. In addition, confirmatory genome-wide analyses of urine metabolites were conducted (n = 578). The fully automated quantitative regression-based spectral analysis is demonstrated for creatinine and glucose (n = 4548). RESULTS Intra-assay metabolite variations were mostly <5%, indicating high robustness and accuracy of urine NMR spectroscopy methodology per se. Intra-individual metabolite variations were large, ranging from 6% to 194%. However, population-based inter-individual metabolite variations were even larger (from 14% to 1655%), providing a sound base for epidemiological applications. Metabolic associations between urine and serum were found to be clearly weaker than those within serum and within urine, indicating that urinary metabolomics data provide independent metabolic information. Two previous genome-wide hits for formate and 2-hydroxyisobutyrate were replicated at genome-wide significance. CONCLUSION Quantitative urine metabolomics data suggest broad novelty for systems epidemiology. A roadmap for an open access methodology is provided.
Collapse
Affiliation(s)
- Tuulia Tynkkynen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Qin Wang
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jussi Ekholm
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Olga Anufrieva
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Jouko Vepsäläinen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Minna Männikkö
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Northern Finland Birth Cohorts, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Sirkka Keinänen-Kiukaanniemi
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Medical Research Center Oulu, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit (MRC PHRU), University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), University of Oxford, Oxford, UK
- National Institute for Health Research, Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, UK
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Matthew Goodwin
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, University of Bristol, Bristol, UK
| | - Susan Ring
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, University of Bristol, Bristol, UK
| | - John C Chambers
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jaspal Kooner
- Ealing Hospital NHS Trust, Middlesex, UK
- Imperial College Healthcare NHS Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Marjo-Riitta Järvelin
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Unit of Primary Health Care, Oulu University Hospital, OYS, Oulu, Finland
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
| | - Johannes Kettunen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- THL: National Institute for Health and Welfare, Helsinki, Finland
| | - Michael Hill
- Medical Research Council Population Health Research Unit (MRC PHRU), University of Oxford, Oxford, UK
- Nuffield Department of Population Health, Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), University of Oxford, Oxford, UK
| | - George Davey Smith
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, University of Bristol, Bristol, UK
| | - Mika Ala-Korpela
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
- Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- Population Health Science, University of Bristol, Bristol, UK
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, Alfred Hospital, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Khlebus E, Kutsenko V, Meshkov A, Ershova A, Kiseleva A, Shevtsov A, Shcherbakova N, Zharikova A, Lankin V, Tikhaze A, Chazova I, Yarovaya E, Drapkina O, Boytsov S. Multiple rare and common variants in APOB gene locus associated with oxidatively modified low-density lipoprotein levels. PLoS One 2019; 14:e0217620. [PMID: 31150472 PMCID: PMC6544350 DOI: 10.1371/journal.pone.0217620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 05/15/2019] [Indexed: 01/17/2023] Open
Abstract
Oxidatively modified low-density lipoproteins (oxLDL) play an important role in the occurrence and progression of atherosclerosis. To identify the genetic factors influencing the oxLDL levels, we have genotyped 776 DNA samples of Russian individuals for 196,725 single-nucleotide polymorphisms (SNPs) using the Cardio-MetaboChip (Illumina, USA) and conducted genome-wide association study (GWAS). Fourteen common variants in the locus including APOB gene were significantly associated with the oxLDL levels (P < 2.18 × 10−7). These variants explained only 6% of the variation in the oxLDL levels. Then, we assessed the contribution of rare coding variants of APOB gene to the oxLDL levels. Individuals with the extreme oxLDL levels (48 with the lowest and 48 with the highest values) were selected for targeted sequencing of the region including APOB gene. To evaluate the contribution of the SNPs to the oxLDL levels we used various statistical methods for the association analysis of rare variants: WST, SKAT, and SKAT-O. We revealed that both synonymous and nonsynonymous SNPs affected the oxLDL levels. For the joint analysis of the rare and common variants, we conducted the SKAT-C testing and found a group of 15 SNPs significantly associated with the oxLDL levels (P = 2.14 × 10−9). Our results indicate that the oxLDL levels depend on both common and rare variants of the APOB gene.
Collapse
Affiliation(s)
- Eleonora Khlebus
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Moscow Institute of Physics and Technology (State University), Moscow, Russia
- * E-mail:
| | - Vladimir Kutsenko
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Alexey Meshkov
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Alexandra Ershova
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Anna Kiseleva
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | | | - Natalia Shcherbakova
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Anastasiia Zharikova
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vadim Lankin
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Alla Tikhaze
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Irina Chazova
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | | | - Oksana Drapkina
- Federal State Institution National Medical Research Center for Preventive Medicine of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Sergey Boytsov
- Federal State Budget Organization National Medical Research Center of Cardiology of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| |
Collapse
|
25
|
Han S, Hwang MY, Yoon K, Kim YK, Kim YJ, Kim BJ, Moon S. Exome chip-driven association study of lipidemia in >14,000 Koreans and evaluation of genetic effect on identified variants between different ethnic groups. Genet Epidemiol 2019; 43:617-628. [PMID: 31087446 DOI: 10.1002/gepi.22208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/12/2019] [Accepted: 04/04/2019] [Indexed: 12/18/2022]
Abstract
Lipid levels in blood are widely used to diagnose and monitor chronic diseases. It is essential to identify the genetic traits involved in lipid metabolism for understanding chronic diseases. However, the influence of genetic traits varies depending on race, sex, age, and ethnicity. Therefore, research focusing on populations of individual countries is required, and the results can be used as a basis for comparison of results of other studies at the cross-racial and cross-country levels. In the present study, we selected lipid-related variants and evaluated their effects on lipid-related diseases in more than 14,000 subjects of three cohorts using the Illumina Human Exome Beadchip. A genome-wide association study was conducted using EPACTs after adjusting for age, sex, and recruitment area. A genome-wide significance cutoff was defined as p < 5E-08 in all the three cohorts. Sixteen variants represented the lipid traits and were classified as vulnerable to borderline hypertriglyceridemia, hyper-LDL-cholesterolemia, or hypo-HDL-cholesterolemia. Moreover, we compared the genetic effects of the 16 variants between ethnic groups and identified the missense variants in apolipoprotein A-V, cholesterol ester transfer protein, and apolipoprotein E as Asian-specific. Our study provides candidate genes as markers for chronic diseases through the evaluation of genetic effects.
Collapse
Affiliation(s)
- Sohee Han
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| | - Mi Yeong Hwang
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| | - Kyungheon Yoon
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| | - Yun Kyoung Kim
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| | - Young-Jin Kim
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| | - Bong-Jo Kim
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| | - Sanghoon Moon
- Division of Genome Research, Center for Genome Science, National Institute of Health, Osong Health Technology Administration Complex, Cheongju-si, Republic of Korea
| |
Collapse
|
26
|
Teslovich TM, Kim DS, Yin X, Stancáková A, Jackson AU, Wielscher M, Naj A, Perry JRB, Huyghe JR, Stringham HM, Davis JP, Raulerson CK, Welch RP, Fuchsberger C, Locke AE, Sim X, Chines PS, Narisu N, Kangas AJ, Soininen P, Ala-Korpela M, Gudnason V, Musani SK, Jarvelin MR, Schellenberg GD, Speliotes EK, Kuusisto J, Collins FS, Boehnke M, Laakso M, Mohlke KL. Identification of seven novel loci associated with amino acid levels using single-variant and gene-based tests in 8545 Finnish men from the METSIM study. Hum Mol Genet 2019; 27:1664-1674. [PMID: 29481666 DOI: 10.1093/hmg/ddy067] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 02/15/2018] [Indexed: 12/13/2022] Open
Abstract
Comprehensive metabolite profiling captures many highly heritable traits, including amino acid levels, which are potentially sensitive biomarkers for disease pathogenesis. To better understand the contribution of genetic variation to amino acid levels, we performed single variant and gene-based tests of association between nine serum amino acids (alanine, glutamine, glycine, histidine, isoleucine, leucine, phenylalanine, tyrosine, and valine) and 16.6 million genotyped and imputed variants in 8545 non-diabetic Finnish men from the METabolic Syndrome In Men (METSIM) study with replication in Northern Finland Birth Cohort (NFBC1966). We identified five novel loci associated with amino acid levels (P = < 5×10-8): LOC157273/PPP1R3B with glycine (rs9987289, P = 2.3×10-26); ZFHX3 (chr16:73326579, minor allele frequency (MAF) = 0.42%, P = 3.6×10-9), LIPC (rs10468017, P = 1.5×10-8), and WWOX (rs9937914, P = 3.8×10-8) with alanine; and TRIB1 with tyrosine (rs28601761, P = 8×10-9). Gene-based tests identified two novel genes harboring missense variants of MAF <1% that show aggregate association with amino acid levels: PYCR1 with glycine (Pgene = 1.5×10-6) and BCAT2 with valine (Pgene = 7.4×10-7); neither gene was implicated by single variant association tests. These findings are among the first applications of gene-based tests to identify new loci for amino acid levels. In addition to the seven novel gene associations, we identified five independent signals at established amino acid loci, including two rare variant signals at GLDC (rs138640017, MAF=0.95%, Pconditional = 5.8×10-40) with glycine levels and HAL (rs141635447, MAF = 0.46%, Pconditional = 9.4×10-11) with histidine levels. Examination of all single variant association results in our data revealed a strong inverse relationship between effect size and MAF (Ptrend<0.001). These novel signals provide further insight into the molecular mechanisms of amino acid metabolism and potentially, their perturbations in disease.
Collapse
Affiliation(s)
- Tanya M Teslovich
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Seung Kim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xianyong Yin
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alena Stancáková
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Anne U Jackson
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Matthias Wielscher
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK
| | - Adam Naj
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania, PA 19104, USA.,Departments of Biostatistics, and Epidemiology (DBE) and Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, PA 19104, USA
| | - John R B Perry
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Jeroen R Huyghe
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Heather M Stringham
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - James P Davis
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chelsea K Raulerson
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ryan P Welch
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christian Fuchsberger
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Adam E Locke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xueling Sim
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter S Chines
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Narisu Narisu
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Antti J Kangas
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Pasi Soininen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK.,Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Epidemiology and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, School of Public Health and Preventive Medicine, The Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Vilmundur Gudnason
- Icelandic Heart Association and the Faculty of Medicine, University of Iceland, Kopavogur, Iceland
| | - Solomon K Musani
- University of Mississippi Medical Center, Jackson, MS 39213, USA
| | - Marjo-Riitta Jarvelin
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London, UK.,Center for Life Course Health Research, Faculty of Medicine, University of Oulu, 90014 Oulu, Finland.,Biocenter Oulu, University of Oulu, 90014 Oulu, Finland.,Unit of Primary Care, Oulu University Hospital, Oulu, Finland
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, University of Pennsylvania, PA 19104, USA
| | - Elizabeth K Speliotes
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA.,Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
27
|
Köttgen A, Raffler J, Sekula P, Kastenmüller G. Genome-Wide Association Studies of Metabolite Concentrations (mGWAS): Relevance for Nephrology. Semin Nephrol 2019; 38:151-174. [PMID: 29602398 DOI: 10.1016/j.semnephrol.2018.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metabolites are small molecules that are intermediates or products of metabolism, many of which are freely filtered by the kidneys. In addition, the kidneys have a central role in metabolite anabolism and catabolism, as well as in active metabolite reabsorption and/or secretion during tubular passage. This review article illustrates how the coupling of genomics and metabolomics in genome-wide association analyses of metabolites can be used to illuminate mechanisms underlying human metabolism, with a special focus on insights relevant to nephrology. First, genetic susceptibility loci for reduced kidney function and chronic kidney disease (CKD) were reviewed systematically for their associations with metabolite concentrations in metabolomics studies of blood and urine. Second, kidney function and CKD-associated metabolites reported from observational studies were interrogated for metabolite-associated genetic variants to generate and discuss complementary insights. Finally, insights originating from the simultaneous study of both blood and urine or by modeling intermetabolite relationships are summarized. We also discuss methodologic questions related to the study of metabolite concentrations in urine as well as among CKD patients. In summary, genome-wide association analyses of metabolites using metabolite concentrations quantified from blood and/or urine are a promising avenue of research to illuminate physiological and pathophysiological functions of the kidney.
Collapse
Affiliation(s)
- Anna Köttgen
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Department of Biometry, Epidemiology and Medical Bioinformatics, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
28
|
Mendelian randomization reveals unexpected effects of CETP on the lipoprotein profile. Eur J Hum Genet 2018; 27:422-431. [PMID: 30420679 DOI: 10.1038/s41431-018-0301-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 09/05/2018] [Accepted: 11/01/2018] [Indexed: 01/06/2023] Open
Abstract
According to the current dogma, cholesteryl ester transfer protein (CETP) decreases high-density lipoprotein (HDL)-cholesterol (C) and increases low-density lipoprotein (LDL)-C. However, detailed insight into the effects of CETP on lipoprotein subclasses is lacking. Therefore, we used a Mendelian randomization approach based on a genetic score for serum CETP concentration (rs247616, rs12720922 and rs1968905) to estimate causal effects per unit (µg/mL) increase in CETP on 159 standardized metabolic biomarkers, primarily lipoprotein subclasses. Metabolic biomarkers were measured by nuclear magnetic resonance (NMR) in 5672 participants of the Netherlands Epidemiology of Obesity (NEO) study. Higher CETP concentrations were associated with less large HDL (largest effect XL-HDL-C, P = 6 × 10-22) and more small VLDL components (largest effect S-VLDL cholesteryl esters, P = 6 × 10-6). No causal effects were observed with LDL subclasses. All these effects were replicated in an independent cohort from European ancestry (MAGNETIC NMR GWAS; n ~20,000). Additionally, we assessed observational associations between ELISA-measured CETP concentration and metabolic measures. In contrast to results from Mendelian randomization, observationally, CETP concentration predominantly associated with more VLDL, IDL and LDL components. Our results show that CETP is an important causal determinant of HDL and VLDL concentration and composition, which may imply that the CETP inhibitor anacetrapib decreased cardiovascular disease risk through specific reduction of small VLDL rather than LDL. The contrast between genetic and observational associations might be explained by a high capacity of VLDL, IDL and LDL subclasses to carry CETP, thereby concealing causal effects on HDL.
Collapse
|
29
|
Tremblay BL, Guénard F, Lamarche B, Pérusse L, Vohl MC. Familial resemblances in human plasma metabolites are attributable to both genetic and common environmental effects. Nutr Res 2018; 61:22-30. [PMID: 30683436 DOI: 10.1016/j.nutres.2018.10.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/26/2018] [Accepted: 10/05/2018] [Indexed: 01/06/2023]
Abstract
Metabolites are of great importance for understanding the pathogenesis of several diseases. Understanding the genetic contribution to metabolite concentrations may provide insights into mechanisms of complex diseases. Several studies have investigated heritability of metabolites but none investigated potential influences of genetic and environmental factors on the relationship between metabolites and cardiometabolic (CM) risk factors. Thus, we tested the hypothesis that both genetic and common environmental effects contribute to the variance of plasma metabolite concentrations and that shared genetic and environmental effects explain their phenotypic correlations with CM risk factors. To test this hypothesis, variance component method and bivariate genetic analysis were performed in a family-based sample of 48 French Canadians from 16 families. Familial resemblances were computed for all 147 detected metabolites and 9 (acetylornithine, acylcarnitine C9, arginine, phosphatidylcholine acyl-alkyl C36:4, serotonin, lysophosphatidylcholine acyl C20:4, citrulline, asymmetric dimethylarginine, phosphatidylcholine acyl-alkyl C36:5) showed a significant familial effect (55.7%, 18.7%, and 37.0% for maximal heritability, genetic heritability, and common environmental effect, respectively). Citrulline, phosphatidylcholine acyl-alkyl C36:4, phosphatidylcholine acyl-alkyl C36:5, and serotonin had significant phenotypic correlations with CM risk factors. Citrulline had a positive genetic correlation with apolipoprotein B100, while phosphatidylcholine acyl-alkyl C36:5 had a positive environmental correlation with total cholesterol. In conclusion, familial resemblances in metabolite concentrations were mainly attributable to common environmental effect when considering metabolites with a significant familial effect. Common genetic and environmental factors may also influence the relationship between metabolites and CM risk factors.
Collapse
Affiliation(s)
- Bénédicte L Tremblay
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd, Quebec City, QC, G1V 0A6, Canada.
| | - Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd, Quebec City, QC, G1V 0A6, Canada.
| | - Benoît Lamarche
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd, Quebec City, QC, G1V 0A6, Canada.
| | - Louis Pérusse
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd, Quebec City, QC, G1V 0A6, Canada; CHU de Québec Research Center - Endocrinology and Nephrology, 2705 Laurier Blvd, Quebec City, QC, G1V 4G2, Canada.
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Laval University, 2440 Hochelaga Blvd, Quebec City, QC, G1V 0A6, Canada; CHU de Québec Research Center - Endocrinology and Nephrology, 2705 Laurier Blvd, Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
30
|
Graham E, Lee J, Price M, Tarailo-Graovac M, Matthews A, Engelke U, Tang J, Kluijtmans LAJ, Wevers RA, Wasserman WW, van Karnebeek CDM, Mostafavi S. Integration of genomics and metabolomics for prioritization of rare disease variants: a 2018 literature review. J Inherit Metab Dis 2018; 41:435-445. [PMID: 29721916 PMCID: PMC5959954 DOI: 10.1007/s10545-018-0139-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 12/19/2017] [Accepted: 01/10/2018] [Indexed: 02/08/2023]
Abstract
Many inborn errors of metabolism (IEMs) are amenable to treatment; therefore, early diagnosis and treatment is imperative. Despite recent advances, the genetic basis of many metabolic phenotypes remains unknown. For discovery purposes, whole exome sequencing (WES) variant prioritization coupled with clinical and bioinformatics expertise is the primary method used to identify novel disease-causing variants; however, causation is often difficult to establish due to the number of plausible variants. Integrated analysis of untargeted metabolomics (UM) and WES or whole genome sequencing (WGS) data is a promising systematic approach for identifying disease-causing variants. In this review, we provide a literature-based overview of UM methods utilizing liquid chromatography mass spectrometry (LC-MS), and assess approaches to integrating WES/WGS and LC-MS UM data for the discovery and prioritization of variants causing IEMs. To embed this integrated -omics approach in the clinic, expansion of gene-metabolite annotations and metabolomic feature-to-metabolite mapping methods are needed.
Collapse
Affiliation(s)
- Emma Graham
- Department of Bioinformatics, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Jessica Lee
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Magda Price
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Maja Tarailo-Graovac
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Allison Matthews
- Department of Pediatrics, BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Udo Engelke
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeffrey Tang
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Leo A J Kluijtmans
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Ron A Wevers
- Translational Metabolic Laboratory, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wyeth W Wasserman
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Clara D M van Karnebeek
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
- Department of Pediatrics, BC Children's Hospital Research Institute, Vancouver, BC, Canada.
- Departments of Pediatrics and Clinical Genetics, Emma Children's Hospital, Academic Medical Centre, Amsterdam, The Netherlands.
| | - Sara Mostafavi
- BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- Department of Statistics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
31
|
Holmes MV, Millwood IY, Kartsonaki C, Hill MR, Bennett DA, Boxall R, Guo Y, Xu X, Bian Z, Hu R, Walters RG, Chen J, Ala-Korpela M, Parish S, Clarke RJ, Peto R, Collins R, Li L, Chen Z. Lipids, Lipoproteins, and Metabolites and Risk of Myocardial Infarction and Stroke. J Am Coll Cardiol 2018; 71:620-632. [PMID: 29420958 PMCID: PMC5811927 DOI: 10.1016/j.jacc.2017.12.006] [Citation(s) in RCA: 335] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/24/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Blood lipids are established risk factors for myocardial infarction (MI), but uncertainty persists about the relevance of lipids, lipoprotein particles, and circulating metabolites for MI and stroke subtypes. OBJECTIVES This study sought to investigate the associations of plasma metabolic markers with risks of incident MI, ischemic stroke (IS), and intracerebral hemorrhage (ICH). METHODS In a nested case-control study (912 MI, 1,146 IS, and 1,138 ICH cases, and 1,466 common control subjects) 30 to 79 years of age in China Kadoorie Biobank, nuclear magnetic resonance spectroscopy measured 225 metabolic markers in baseline plasma samples. Logistic regression was used to estimate adjusted odds ratios (ORs) for a 1-SD higher metabolic marker. RESULTS Very low-, intermediate-, and low-density lipoprotein particles were positively associated with MI and IS. High-density lipoprotein (HDL) particles were inversely associated with MI apart from small HDL. In contrast, no lipoprotein particles were associated with ICH. Cholesterol in large HDL was inversely associated with MI and IS (OR: 0.79 and 0.88, respectively), whereas cholesterol in small HDL was not (OR: 0.99 and 1.06, respectively). Triglycerides within all lipoproteins, including most HDL particles, were positively associated with MI, with a similar pattern for IS. Glycoprotein acetyls, ketone bodies, glucose, and docosahexaenoic acid were associated with all 3 diseases. The 225 metabolic markers showed concordant associations between MI and IS, but not with ICH. CONCLUSIONS Lipoproteins and lipids showed similar associations with MI and IS, but not with ICH. Within HDL particles, cholesterol concentrations were inversely associated, whereas triglyceride concentrations were positively associated with MI. Glycoprotein acetyls and several non-lipid-related metabolites associated with all 3 diseases.
Collapse
Affiliation(s)
- Michael V Holmes
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, United Kingdom.
| | - Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Christiana Kartsonaki
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Michael R Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Derrick A Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Ruth Boxall
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Yu Guo
- Chinese Academy of Medical Sciences, Beijing, China
| | - Xin Xu
- Liuyang CDC, Changsha, China
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Beijing, China
| | - Ruying Hu
- NCDs Prevention and Control Department, Zhejiang CDC, Hangzhou, China
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Junshi Chen
- National Center for Food Safety Risk Assessment, Beijing, China
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland; Medical Research Council Integrative Epidemiology Unit at the University of Bristol, Bristol, United Kingdom; Population Health Science, Bristol Medical School, University of Bristol, Bristol, United Kingdom; NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland; Systems Epidemiology, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health Sciences, the Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| | - Sarah Parish
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom; Medical Research Council Population Health Research Unit (MRC PHRU) at the University of Oxford, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Robert J Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Liming Li
- Chinese Academy of Medical Sciences, Beijing, China; Department of Global Health, School of Public Health, Peking University, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
32
|
Würtz P, Kangas AJ, Soininen P, Lawlor DA, Davey Smith G, Ala-Korpela M. Quantitative Serum Nuclear Magnetic Resonance Metabolomics in Large-Scale Epidemiology: A Primer on -Omic Technologies. Am J Epidemiol 2017; 186:1084-1096. [PMID: 29106475 PMCID: PMC5860146 DOI: 10.1093/aje/kwx016] [Citation(s) in RCA: 375] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 01/19/2017] [Indexed: 12/13/2022] Open
Abstract
Detailed metabolic profiling in large-scale epidemiologic studies has uncovered novel biomarkers for cardiometabolic diseases and clarified the molecular associations of established risk factors. A quantitative metabolomics platform based on nuclear magnetic resonance spectroscopy has found widespread use, already profiling over 400,000 blood samples. Over 200 metabolic measures are quantified per sample; in addition to many biomarkers routinely used in epidemiology, the method simultaneously provides fine-grained lipoprotein subclass profiling and quantification of circulating fatty acids, amino acids, gluconeogenesis-related metabolites, and many other molecules from multiple metabolic pathways. Here we focus on applications of magnetic resonance metabolomics for quantifying circulating biomarkers in large-scale epidemiology. We highlight the molecular characterization of risk factors, use of Mendelian randomization, and the key issues of study design and analyses of metabolic profiling for epidemiology. We also detail how integration of metabolic profiling data with genetics can enhance drug development. We discuss why quantitative metabolic profiling is becoming widespread in epidemiology and biobanking. Although large-scale applications of metabolic profiling are still novel, it seems likely that comprehensive biomarker data will contribute to etiologic understanding of various diseases and abilities to predict disease risks, with the potential to translate into multiple clinical settings.
Collapse
Affiliation(s)
- Peter Würtz
- Correspondence to Dr. Peter Würtz, Computational Medicine, Faculty of Medicine, Aapistie 5A, P.O. Box 5000, FI-90014 University of Oulu, Finland (e-mail: ); or Dr. Mika Ala-Korpela, Computational Medicine, Faculty of Medicine, Aapistie 5A, P.O. Box 5000, FI-90014 University of Oulu, Finland (e-mail: )
| | | | | | | | | | - Mika Ala-Korpela
- Correspondence to Dr. Peter Würtz, Computational Medicine, Faculty of Medicine, Aapistie 5A, P.O. Box 5000, FI-90014 University of Oulu, Finland (e-mail: ); or Dr. Mika Ala-Korpela, Computational Medicine, Faculty of Medicine, Aapistie 5A, P.O. Box 5000, FI-90014 University of Oulu, Finland (e-mail: )
| |
Collapse
|
33
|
O'Sullivan JF, Morningstar JE, Yang Q, Zheng B, Gao Y, Jeanfavre S, Scott J, Fernandez C, Zheng H, O'Connor S, Cohen P, Vasan RS, Long MT, Wilson JG, Melander O, Wang TJ, Fox C, Peterson RT, Clish CB, Corey KE, Gerszten RE. Dimethylguanidino valeric acid is a marker of liver fat and predicts diabetes. J Clin Invest 2017; 127:4394-4402. [PMID: 29083323 DOI: 10.1172/jci95995] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/21/2017] [Indexed: 01/11/2023] Open
Abstract
Unbiased, "nontargeted" metabolite profiling techniques hold considerable promise for biomarker and pathway discovery, in spite of the lack of successful applications to human disease. By integrating nontargeted metabolomics, genetics, and detailed human phenotyping, we identified dimethylguanidino valeric acid (DMGV) as an independent biomarker of CT-defined nonalcoholic fatty liver disease (NAFLD) in the offspring cohort of the Framingham Heart Study (FHS) participants. We verified the relationship between DMGV and early hepatic pathology. Specifically, plasma DMGV levels were correlated with biopsy-proven nonalcoholic steatohepatitis (NASH) in a hospital cohort of individuals undergoing gastric bypass surgery, and DMGV levels fell in parallel with improvements in post-procedure cardiometabolic parameters. Further, baseline DMGV levels independently predicted future diabetes up to 12 years before disease onset in 3 distinct human cohorts. Finally, we provide all metabolite peak data consisting of known and unidentified peaks, genetics, and key metabolic parameters as a publicly available resource for investigations in cardiometabolic diseases.
Collapse
Affiliation(s)
- John F O'Sullivan
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Charles Perkins Centre and Heart Research Institute, The University of Sydney, Sydney, Australia
| | - Jordan E Morningstar
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Qiong Yang
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, USA.,Biostatistics Department, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Baohui Zheng
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yan Gao
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Sarah Jeanfavre
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Justin Scott
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Hui Zheng
- Biostatistics Department, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sean O'Connor
- The Rockefeller University, Laboratory of Molecular Metabolism, New York, New York, USA
| | - Paul Cohen
- The Rockefeller University, Laboratory of Molecular Metabolism, New York, New York, USA
| | - Ramachandran S Vasan
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, USA.,Cardiology Division, Boston Medical Center, and
| | - Michelle T Long
- Gastroenterology Division, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - James G Wilson
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Olle Melander
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Center of Emergency Medicine, Skåne University Hospital, Malmö, Sweden
| | - Thomas J Wang
- Cardiology Division, Vanderbilt University, Nashville, Tennessee, USA
| | - Caroline Fox
- Framingham Heart Study of the National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, Massachusetts, USA
| | - Randall T Peterson
- Cardiovascular Research Center, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kathleen E Corey
- Gastroenterology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
34
|
Association of leisure time physical activity and NMR-detected circulating amino acids in peripubertal girls: A 7.5-year longitudinal study. Sci Rep 2017; 7:14026. [PMID: 29070851 PMCID: PMC5656647 DOI: 10.1038/s41598-017-14116-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/02/2017] [Indexed: 11/08/2022] Open
Abstract
This study investigated the longitudinal associations of physical activity and circulating amino acids concentration in peripubertal girls. Three hundred ninety-six Finnish girls participated in the longitudinal study from childhood (mean age 11.2 years) to early adulthood (mean age 18.2 years). Circulating amino acids were assessed by nuclear magnetic resonance spectroscopy. LTPA was assessed by self-administered questionnaire. We found that isoleucine, leucine and tyrosine levels were significantly higher in individuals with lower LTPA than their peers at age 11 (p < 0.05 for all), independent of BMI. In addition, isoleucine and leucine levels increased significantly (~15%) from childhood to early adulthood among the individuals with consistently low LTPA (p < 0.05 for both), while among the individuals with consistently high LTPA the level of these amino acids remained virtually unchanged. In conclusion, high level of physical activity is associated lower serum isoleucine and leucine in peripubertal girls, independent of BMI, which may serve as a mechanistic link between high level of physical activity in childhood and its health benefits later in life. Further studies in peripubertal boys are needed to assess whether associations between physical activity and circulating amino acids in children adolescents are sex-specific.
Collapse
|
35
|
Verbeek R, Boyer M, Boekholdt SM, Hovingh GK, Kastelein JJP, Wareham N, Khaw KT, Arsenault BJ. Carriers of the PCSK9 R46L Variant Are Characterized by an Antiatherogenic Lipoprotein Profile Assessed by Nuclear Magnetic Resonance Spectroscopy-Brief Report. Arterioscler Thromb Vasc Biol 2016; 37:43-48. [PMID: 27856457 DOI: 10.1161/atvbaha.116.307995] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Carriers of the PCSK9 (proprotein convertase subtilisin/kexin 9) R46L genetic variant (rs11591147) are characterized by low levels of low-density lipoprotein cholesterol and a decreased risk of cardiovascular disease. We studied the impact of the R46L variant on lipoprotein size and composition. APPROACH AND RESULTS Lipoprotein size and composition were measured by nuclear magnetic resonance spectroscopy in 2373 participants of the EPIC (European Prospective Investigation into Cancer and Nutrition)-Norfolk study. After adjusting for age, sex, and cardiovascular disease status, carriers of the R46L variant (n=77) were characterized by lower concentrations of very low-density lipoprotein particles (85.8±26.2 versus 99.0±33.3 nmol/L; P<0.001), low-density lipoprotein particles (1479.7±396.8 versus 1662.9±458.3 nmol/L; P<0.001), and lipoprotein(a) (11.1 [7.2-28.6] versus 12.4 [6.7-29.1] mg/dL; P<0.001) compared with noncarriers. Total high-density lipoprotein particle and very low-density lipoprotein, low-density lipoprotein, and high-density lipoprotein particle sizes were comparable in carriers and noncarriers. Carriers were characterized by lower secretory phospholipase A2 (4.2±0.9 versus 4.6±1.3 nmol/mL/min; P=0.004) and lipoprotein-associated phospholipase A2 activity (47.5±14.1 versus 52.4±16.2 nmol/mL/min; P=0.02) compared with noncarriers. CONCLUSIONS Results of this study suggest that carriers of the PCSK9 R46L genetic variant have lower very low-density lipoprotein and low-density lipoprotein particle concentrations, lower lipoprotein(a) levels, and lower secretory phospholipase A2 and lipoprotein-associated phospholipase A2 activity compared with noncarriers.
Collapse
Affiliation(s)
- Rutger Verbeek
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Marjorie Boyer
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - S Matthijs Boekholdt
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - G Kees Hovingh
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - John J P Kastelein
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Nicholas Wareham
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Kay-Tee Khaw
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.)
| | - Benoit J Arsenault
- From the Department of Vascular Medicine (R.V., G.K.H., J.J.P.K.) and Department of Cardiology (S.M.B.), Academic Medical Centre, Amsterdam, The Netherlands; Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Canada (M.B., B.J.A.); Department of Medicine, Faculty of Medicine, Université Laval, Québec, Canada (M.B., B.J.A.); Medical Research Council Epidemiology Unit, Cambridge, United Kingdom (N.W.); and Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge, United Kingdom (K.-T.K.).
| |
Collapse
|
36
|
Yee SW, Giacomini MM, Hsueh CH, Weitz D, Liang X, Goswami S, Kinchen JM, Coelho A, Zur AA, Mertsch K, Brian W, Kroetz DL, Giacomini KM. Metabolomic and Genome-wide Association Studies Reveal Potential Endogenous Biomarkers for OATP1B1. Clin Pharmacol Ther 2016; 100:524-536. [PMID: 27447836 PMCID: PMC6365106 DOI: 10.1002/cpt.434] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/15/2016] [Indexed: 12/17/2022]
Abstract
Transporter-mediated drug-drug interactions (DDIs) are a major cause of drug toxicities. Using published genome-wide association studies (GWAS) of the human metabolome, we identified 20 metabolites associated with genetic variants in organic anion transporter, OATP1B1 (P < 5 × 10-8 ). Of these, 12 metabolites were significantly higher in plasma samples from volunteers dosed with the OATP1B1 inhibitor, cyclosporine (CSA) vs. placebo (q-value < 0.2). Conjugated bile acids and fatty acid dicarboxylates were among the metabolites discovered using both GWAS and CSA administration. In vitro studies confirmed tetradecanedioate (TDA) and hexadecanedioate (HDA) were novel substrates of OATP1B1 as well as OAT1 and OAT3. This study highlights the use of multiple datasets for the discovery of endogenous metabolites that represent potential in vivo biomarkers for transporter-mediated DDIs. Future studies are needed to determine whether these metabolites can serve as qualified biomarkers for organic anion transporters. Quantitative relationships between metabolite levels and modulation of transporters should be established.
Collapse
Affiliation(s)
- S W Yee
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - M M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - C-H Hsueh
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - D Weitz
- Research and Development Drug Disposition, Sanofi-Aventis Deutschland, Frankfurt, Germany
| | - X Liang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - S Goswami
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - J M Kinchen
- Metabolon, Inc., Durham, North Carolina, USA
| | - A Coelho
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - A A Zur
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - K Mertsch
- Research and Development Drug Disposition, Sanofi-Aventis Deutschland, Frankfurt, Germany
| | - W Brian
- Disposition Safety and Animal Research, Sanofi-Aventis, Great Valley, Pennsylvania, USA
| | - D L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| | - K M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA.
- Institute for Human Genetics, University of California, San Francisco, San Francisco, California, USA.
| |
Collapse
|
37
|
Fearnley LG, Inouye M. Metabolomics in epidemiology: from metabolite concentrations to integrative reaction networks. Int J Epidemiol 2016; 45:1319-1328. [PMID: 27118561 PMCID: PMC5100607 DOI: 10.1093/ije/dyw046] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2016] [Indexed: 01/05/2023] Open
Abstract
Metabolomics is becoming feasible for population-scale studies of human disease. In this review, we survey epidemiological studies that leverage metabolomics and multi-omics to gain insight into disease mechanisms. We outline key practical, technological and analytical limitations while also highlighting recent successes in integrating these data. The use of multi-omics to infer reaction rates is discussed as a potential future direction for metabolomics research, as a means of identifying biomarkers as well as inferring causality. Furthermore, we highlight established analysis approaches as well as simulation-based methods currently used in single- and multi-cell levels in systems biology.
Collapse
Affiliation(s)
- Liam G Fearnley
- Centre for Systems Genomics.,School of BioSciences.,Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| | - Michael Inouye
- Centre for Systems Genomics .,School of BioSciences.,Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
38
|
Okazaki M, Yamashita S. Recent Advances in Analytical Methods on Lipoprotein Subclasses: Calculation of Particle Numbers from Lipid Levels by Gel Permeation HPLC Using “Spherical Particle Model”. J Oleo Sci 2016; 65:265-82. [DOI: 10.5650/jos.ess16020] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Shizuya Yamashita
- Rinku General Medical Center
- Department of Community Medicine & Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine
| |
Collapse
|
39
|
Soininen P, Kangas AJ, Würtz P, Suna T, Ala-Korpela M. Quantitative serum nuclear magnetic resonance metabolomics in cardiovascular epidemiology and genetics. ACTA ACUST UNITED AC 2015; 8:192-206. [PMID: 25691689 DOI: 10.1161/circgenetics.114.000216] [Citation(s) in RCA: 573] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Metabolomics is becoming common in epidemiology due to recent developments in quantitative profiling technologies and appealing results from their applications for understanding health and disease. Our team has developed an automated high-throughput serum NMR metabolomics platform that provides quantitative molecular data on 14 lipoprotein subclasses, their lipid concentrations and composition, apolipoprotein A-I and B, multiple cholesterol and triglyceride measures, albumin, various fatty acids as well as on numerous low-molecular-weight metabolites, including amino acids, glycolysis related measures and ketone bodies. The molar concentrations of these measures are obtained from a single serum sample with costs comparable to standard lipid measurements. We have analyzed almost 250 000 samples from around 100 epidemiological cohorts and biobanks and the new international set-up of multiple platforms will allow an annual throughput of more than 250 000 samples. The molecular data have been used to study type 1 and type 2 diabetes etiology as well as to characterize the molecular reflections of the metabolic syndrome, long-term physical activity, diet and lipoprotein metabolism. The results have revealed new biomarkers for early atherosclerosis, type 2 diabetes, diabetic nephropathy, cardiovascular disease and all-cause mortality. We have also combined genomics and metabolomics in diverse studies. We envision that quantitative high-throughput NMR metabolomics will be incorporated as a routine in large biobanks; this would make perfect sense both from the biological research and cost point of view - the standard output of over 200 molecular measures would vastly extend the relevance of the sample collections and make many separate clinical chemistry assays redundant.
Collapse
Affiliation(s)
- Pasi Soininen
- From the Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (P.S., A.J.K., P.W., T.S., M.A.-K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (P.S., M.A.-K.); Oulu University Hospital, Oulu, Finland (M.A.-K.); and Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (M.A.-K.)
| | - Antti J Kangas
- From the Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (P.S., A.J.K., P.W., T.S., M.A.-K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (P.S., M.A.-K.); Oulu University Hospital, Oulu, Finland (M.A.-K.); and Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (M.A.-K.)
| | - Peter Würtz
- From the Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (P.S., A.J.K., P.W., T.S., M.A.-K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (P.S., M.A.-K.); Oulu University Hospital, Oulu, Finland (M.A.-K.); and Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (M.A.-K.)
| | - Teemu Suna
- From the Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (P.S., A.J.K., P.W., T.S., M.A.-K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (P.S., M.A.-K.); Oulu University Hospital, Oulu, Finland (M.A.-K.); and Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (M.A.-K.)
| | - Mika Ala-Korpela
- From the Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland (P.S., A.J.K., P.W., T.S., M.A.-K.); NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland (P.S., M.A.-K.); Oulu University Hospital, Oulu, Finland (M.A.-K.); and Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom (M.A.-K.).
| |
Collapse
|
40
|
Suhre K, Raffler J, Kastenmüller G. Biochemical insights from population studies with genetics and metabolomics. Arch Biochem Biophys 2015; 589:168-76. [PMID: 26432701 DOI: 10.1016/j.abb.2015.09.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/28/2015] [Accepted: 09/28/2015] [Indexed: 12/31/2022]
Abstract
Genome-wide association studies with concentrations of hundreds of small molecules in samples collected from thousands of individuals (mGWAS) access otherwise inaccessible natural genetic experiments and their influence on the metabolic capacities of the human body. By sampling the natural metabolic and genetic variability that is present in the general population, mGWAS identified over 150 associations between genetic variants and variation in the metabolic composition of human body fluids. Many of these genetic variants were found to be located in enzyme or transporter coding genes, whose functions match the biochemical nature of the associated metabolites. Associations identified by mGWAS can reveal novel biochemical knowledge, such as the function of uncharacterized genes, the biochemical identity of small molecules, and the structure of entire biochemical pathways. Here we review findings of recent mGWAS and discuss concrete examples of how their results can be interpreted in a biochemical context. We describe online resources that are available for mining mGWAS results. In this context, we present two concepts that also find more general applications in the field of metabolomics: strengthening of associations by looking at ratios between metabolite pairs and reconstruction of metabolic pathways by Gaussian graphical modeling.
Collapse
Affiliation(s)
- Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medical College - Qatar, Doha, Qatar; Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany.
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; German Center for Diabetes Research, Neuherberg, Germany
| |
Collapse
|
41
|
Burkhardt R, Kirsten H, Beutner F, Holdt LM, Gross A, Teren A, Tönjes A, Becker S, Krohn K, Kovacs P, Stumvoll M, Teupser D, Thiery J, Ceglarek U, Scholz M. Integration of Genome-Wide SNP Data and Gene-Expression Profiles Reveals Six Novel Loci and Regulatory Mechanisms for Amino Acids and Acylcarnitines in Whole Blood. PLoS Genet 2015; 11:e1005510. [PMID: 26401656 PMCID: PMC4581711 DOI: 10.1371/journal.pgen.1005510] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 08/17/2015] [Indexed: 01/23/2023] Open
Abstract
Profiling amino acids and acylcarnitines in whole blood spots is a powerful tool in the laboratory diagnosis of several inborn errors of metabolism. Emerging data suggests that altered blood levels of amino acids and acylcarnitines are also associated with common metabolic diseases in adults. Thus, the identification of common genetic determinants for blood metabolites might shed light on pathways contributing to human physiology and common diseases. We applied a targeted mass-spectrometry-based method to analyze whole blood concentrations of 96 amino acids, acylcarnitines and pathway associated metabolite ratios in a Central European cohort of 2,107 adults and performed genome-wide association (GWA) to identify genetic modifiers of metabolite concentrations. We discovered and replicated six novel loci associated with blood levels of total acylcarnitine, arginine (both on chromosome 6; rs12210538, rs17657775), propionylcarnitine (chromosome 10; rs12779637), 2-hydroxyisovalerylcarnitine (chromosome 21; rs1571700), stearoylcarnitine (chromosome 1; rs3811444), and aspartic acid traits (chromosome 8; rs750472). Based on an integrative analysis of expression quantitative trait loci in blood mononuclear cells and correlations between gene expressions and metabolite levels, we provide evidence for putative causative genes: SLC22A16 for total acylcarnitines, ARG1 for arginine, HLCS for 2-hydroxyisovalerylcarnitine, JAM3 for stearoylcarnitine via a trans-effect at chromosome 1, and PPP1R16A for aspartic acid traits. Further, we report replication and provide additional functional evidence for ten loci that have previously been published for metabolites measured in plasma, serum or urine. In conclusion, our integrative analysis of SNP, gene-expression and metabolite data points to novel genetic factors that may be involved in the regulation of human metabolism. At several loci, we provide evidence for metabolite regulation via gene-expression and observed overlaps with GWAS loci for common diseases. These results form a strong rationale for subsequent functional and disease-related studies.
Collapse
Affiliation(s)
- Ralph Burkhardt
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Holger Kirsten
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- Department for Cell Therapy, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Frank Beutner
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Lesca M. Holdt
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Laboratory Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Arnd Gross
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Andrej Teren
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Heart Center Leipzig, Leipzig, Germany
| | - Anke Tönjes
- Medical Department, Clinic for Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Susen Becker
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Knut Krohn
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Interdisciplinary Centre for Clinical Research, University of Leipzig, Leipzig, Germany
| | - Peter Kovacs
- Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig, Leipzig Germany
| | - Michael Stumvoll
- Medical Department, Clinic for Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
- Integrated Research and Treatment Center Adiposity Diseases, University of Leipzig, Leipzig Germany
| | - Daniel Teupser
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Laboratory Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Joachim Thiery
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Uta Ceglarek
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Markus Scholz
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig Germany
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- * E-mail:
| |
Collapse
|
42
|
Kastenmüller G, Raffler J, Gieger C, Suhre K. Genetics of human metabolism: an update. Hum Mol Genet 2015; 24:R93-R101. [PMID: 26160913 PMCID: PMC4572003 DOI: 10.1093/hmg/ddv263] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/06/2015] [Indexed: 01/01/2023] Open
Abstract
Genome-wide association studies with metabolomics (mGWAS) identify genetically influenced metabotypes (GIMs), their ensemble defining the heritable part of every human's metabolic individuality. Knowledge of genetic variation in metabolism has many applications of biomedical and pharmaceutical interests, including the functional understanding of genetic associations with clinical end points, design of strategies to correct dysregulations in metabolic disorders and the identification of genetic effect modifiers of metabolic disease biomarkers. Furthermore, it has been shown that GIMs provide testable hypotheses for functional genomics and metabolomics and for the identification of novel gene functions and metabolite identities. mGWAS with growing sample sizes and increasingly complex metabolic trait panels are being conducted, allowing for more comprehensive and systems-based downstream analyses. The generated large datasets of genetic associations can now be mined by the biomedical research community and provide valuable resources for hypothesis-driven studies. In this review, we provide a brief summary of the key aspects of mGWAS, followed by an update of recently published mGWAS. We then discuss new approaches of integrating and exploring mGWAS results and finish by presenting selected applications of GIMs in recent studies.
Collapse
Affiliation(s)
- Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, German Center for Diabetes Research, Neuherberg, Germany and
| | - Johannes Raffler
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Christian Gieger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany and Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Karsten Suhre
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany, Department of Physiology and Biophysics, Weill Cornell Medical College-Qatar, Doha, Qatar
| |
Collapse
|
43
|
Männistö VT, Simonen M, Hyysalo J, Soininen P, Kangas AJ, Kaminska D, Matte AK, Venesmaa S, Käkelä P, Kärjä V, Arola J, Gylling H, Cederberg H, Kuusisto J, Laakso M, Yki-Järvinen H, Ala-Korpela M, Pihlajamäki J. Ketone body production is differentially altered in steatosis and non-alcoholic steatohepatitis in obese humans. Liver Int 2015; 35:1853-61. [PMID: 25533197 DOI: 10.1111/liv.12769] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 12/13/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Levels of ketone bodies have been reported to be both increased and decreased in individuals with non-alcoholic fatty liver disease. We investigated whether the metabolism of ketone bodies is different in simple steatosis and in non-alcoholic steatohepatitis (NASH). METHODS Serum low molecular weight molecules including ketone bodies were measured using high-throughput proton (1H) nuclear magnetic resonance in 116 (76 categorized unequivocally to those with normal liver, simple steatosis or NASH) morbidly obese individuals [age 47.3 ± 8.7 (mean ± SD) years, body mass index 45.1 ± 6.1 kg/m(2) , 39 men and 77 women] with histological assessment of NASH and analysis of gene expression in the liver. Finally, we correlated β-hydroxybutyrate (β-OHB) levels with NASH predicting score in Metabolic Syndrome in Men Study (METSIM) population study (n = 8749 non-diabetic men). RESULTS Levels of ketone bodies were lower in individuals with NASH compared to individuals with simple steatosis (P = 0.004 and P = 0.018 for β-OHB and acetoacetate respectively). Lower levels of β-OHB were associated with the NASH predicting score in the METSIM study (P = 0.001). Liver inflammation correlated with mRNA expression of genes regulating ketolysis in the liver (Spearman correlation 0.379-0.388, P < 0.0006 for ACAT1, ACSS2 and BDH1). CONCLUSION Lower levels of ketone bodies in individuals with NASH compared to individuals with simple steatosis suggest a decrease in ketone body metabolism in NASH.
Collapse
Affiliation(s)
- Ville T Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Marko Simonen
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Jenni Hyysalo
- Department of Medicine, University of Helsinki, Helsinki and Minerva Medical Research Institute, Helsinki, Finland
| | - Pasi Soininen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Antti J Kangas
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Dorota Kaminska
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Ananda K Matte
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Sari Venesmaa
- Department of Surgery, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Pirjo Käkelä
- Department of Surgery, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Johanna Arola
- Department of Pathology, The Laboratory of Helsinki University Central Hospital, Helinski, Finland
| | - Helena Gylling
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Department of Medicine, Division of Internal Medicine, University of Helsinki, Helsinki, Finland
| | - Henna Cederberg
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Hannele Yki-Järvinen
- Department of Medicine, University of Helsinki, Helsinki and Minerva Medical Research Institute, Helsinki, Finland
| | - Mika Ala-Korpela
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland.,Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland.,Computational Medicine, School of Social and Community Medicine & Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Jussi Pihlajamäki
- Department of Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.,Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
44
|
Genome-wide association study identifies novel genetic variants contributing to variation in blood metabolite levels. Nat Commun 2015; 6:7208. [PMID: 26068415 DOI: 10.1038/ncomms8208] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 04/20/2015] [Indexed: 01/06/2023] Open
Abstract
Metabolites are small molecules involved in cellular metabolism, which can be detected in biological samples using metabolomic techniques. Here we present the results of genome-wide association and meta-analyses for variation in the blood serum levels of 129 metabolites as measured by the Biocrates metabolomic platform. In a discovery sample of 7,478 individuals of European descent, we find 4,068 genome- and metabolome-wide significant (Z-test, P < 1.09 × 10(-9)) associations between single-nucleotide polymorphisms (SNPs) and metabolites, involving 59 independent SNPs and 85 metabolites. Five of the fifty-nine independent SNPs are new for serum metabolite levels, and were followed-up for replication in an independent sample (N = 1,182). The novel SNPs are located in or near genes encoding metabolite transporter proteins or enzymes (SLC22A16, ARG1, AGPS and ACSL1) that have demonstrated biomedical or pharmaceutical importance. The further characterization of genetic influences on metabolic phenotypes is important for progress in biological and medical research.
Collapse
|
45
|
Shim H, Chasman DI, Smith JD, Mora S, Ridker PM, Nickerson DA, Krauss RM, Stephens M. A multivariate genome-wide association analysis of 10 LDL subfractions, and their response to statin treatment, in 1868 Caucasians. PLoS One 2015; 10:e0120758. [PMID: 25898129 PMCID: PMC4405269 DOI: 10.1371/journal.pone.0120758] [Citation(s) in RCA: 332] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 01/09/2015] [Indexed: 12/27/2022] Open
Abstract
We conducted a genome-wide association analysis of 7 subfractions of low density lipoproteins (LDLs) and 3 subfractions of intermediate density lipoproteins (IDLs) measured by gradient gel electrophoresis, and their response to statin treatment, in 1868 individuals of European ancestry from the Pharmacogenomics and Risk of Cardiovascular Disease study. Our analyses identified four previously-implicated loci (SORT1, APOE, LPA, and CETP) as containing variants that are very strongly associated with lipoprotein subfractions (log10Bayes Factor > 15). Subsequent conditional analyses suggest that three of these (APOE, LPA and CETP) likely harbor multiple independently associated SNPs. Further, while different variants typically showed different characteristic patterns of association with combinations of subfractions, the two SNPs in CETP show strikingly similar patterns - both in our original data and in a replication cohort - consistent with a common underlying molecular mechanism. Notably, the CETP variants are very strongly associated with LDL subfractions, despite showing no association with total LDLs in our study, illustrating the potential value of the more detailed phenotypic measurements. In contrast with these strong subfraction associations, genetic association analysis of subfraction response to statins showed much weaker signals (none exceeding log10Bayes Factor of 6). However, two SNPs (in APOE and LPA) previously-reported to be associated with LDL statin response do show some modest evidence for association in our data, and the subfraction response proles at the LPA SNP are consistent with the LPA association, with response likely being due primarily to resistance of Lp(a) particles to statin therapy. An additional important feature of our analysis is that, unlike most previous analyses of multiple related phenotypes, we analyzed the subfractions jointly, rather than one at a time. Comparisons of our multivariate analyses with standard univariate analyses demonstrate that multivariate analyses can substantially increase power to detect associations. Software implementing our multivariate analysis methods is available at http://stephenslab.uchicago.edu/software.html.
Collapse
Affiliation(s)
- Heejung Shim
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Daniel I. Chasman
- Center for Cardiovascular Disease Prevention, Brigham and Womens Hospital and Harvard Medical School, Boston, MA, USA
| | - Joshua D. Smith
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Samia Mora
- Center for Cardiovascular Disease Prevention, Brigham and Womens Hospital and Harvard Medical School, Boston, MA, USA
| | - Paul M. Ridker
- Center for Cardiovascular Disease Prevention, Brigham and Womens Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Ronald M. Krauss
- Childrens Hospital Oakland Research Institute, Oakland, CA, USA
- * E-mail: (RMK); (MS)
| | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Statistics, University of Chicago, Chicago, IL, USA
- * E-mail: (RMK); (MS)
| |
Collapse
|
46
|
Wahl S, Vogt S, Stückler F, Krumsiek J, Bartel J, Kacprowski T, Schramm K, Carstensen M, Rathmann W, Roden M, Jourdan C, Kangas AJ, Soininen P, Ala-Korpela M, Nöthlings U, Boeing H, Theis FJ, Meisinger C, Waldenberger M, Suhre K, Homuth G, Gieger C, Kastenmüller G, Illig T, Linseisen J, Peters A, Prokisch H, Herder C, Thorand B, Grallert H. Multi-omic signature of body weight change: results from a population-based cohort study. BMC Med 2015; 13:48. [PMID: 25857605 PMCID: PMC4367822 DOI: 10.1186/s12916-015-0282-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/20/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Excess body weight is a major risk factor for cardiometabolic diseases. The complex molecular mechanisms of body weight change-induced metabolic perturbations are not fully understood. Specifically, in-depth molecular characterization of long-term body weight change in the general population is lacking. Here, we pursued a multi-omic approach to comprehensively study metabolic consequences of body weight change during a seven-year follow-up in a large prospective study. METHODS We used data from the population-based Cooperative Health Research in the Region of Augsburg (KORA) S4/F4 cohort. At follow-up (F4), two-platform serum metabolomics and whole blood gene expression measurements were obtained for 1,631 and 689 participants, respectively. Using weighted correlation network analysis, omics data were clustered into modules of closely connected molecules, followed by the formation of a partial correlation network from the modules. Association of the omics modules with previous annual percentage weight change was then determined using linear models. In addition, we performed pathway enrichment analyses, stability analyses, and assessed the relation of the omics modules with clinical traits. RESULTS Four metabolite and two gene expression modules were significantly and stably associated with body weight change (P-values ranging from 1.9 × 10(-4) to 1.2 × 10(-24)). The four metabolite modules covered major branches of metabolism, with VLDL, LDL and large HDL subclasses, triglycerides, branched-chain amino acids and markers of energy metabolism among the main representative molecules. One gene expression module suggests a role of weight change in red blood cell development. The other gene expression module largely overlaps with the lipid-leukocyte (LL) module previously reported to interact with serum metabolites, for which we identify additional co-expressed genes. The omics modules were interrelated and showed cross-sectional associations with clinical traits. Moreover, weight gain and weight loss showed largely opposing associations with the omics modules. CONCLUSIONS Long-term weight change in the general population globally associates with serum metabolite concentrations. An integrated metabolomics and transcriptomics approach improved the understanding of molecular mechanisms underlying the association of weight gain with changes in lipid and amino acid metabolism, insulin sensitivity, mitochondrial function as well as blood cell development and function.
Collapse
|
47
|
Männistö VT, Simonen M, Soininen P, Tiainen M, Kangas AJ, Kaminska D, Venesmaa S, Käkelä P, Kärjä V, Gylling H, Ala-Korpela M, Pihlajamäki J. Lipoprotein subclass metabolism in nonalcoholic steatohepatitis. J Lipid Res 2014; 55:2676-84. [PMID: 25344588 DOI: 10.1194/jlr.p054387] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is associated with increased synthesis of triglycerides and cholesterol coupled with increased VLDL synthesis in the liver. In addition, increased cholesterol content in the liver associates with NASH. Here we study the association of lipoprotein subclass metabolism with NASH. To this aim, liver biopsies from 116 morbidly obese individuals [age 47.3 ± 8.7 (mean ± SD) years, BMI 45.1 ± 6.1 kg/m², 39 men and 77 women] were used for histological assessment. Proton NMR spectroscopy was used to measure lipid concentrations of 14 lipoprotein subclasses in native serum samples at baseline and after obesity surgery. We observed that total lipid concentration of VLDL and LDL subclasses, but not HDL subclasses, associated with NASH [false discovery rate (FDR) < 0.1]. More specifically, total lipid and cholesterol concentration of VLDL and LDL subclasses associated with inflammation, fibrosis, and cell injury (FDR < 0.1), independent of steatosis. Cholesterol concentration of all VLDL subclasses also correlated with total and free cholesterol content in the liver. All NASH-related changes in lipoprotein subclasses were reversed by obesity surgery. High total lipid and cholesterol concentration of serum VLDL and LDL subclasses are linked to cholesterol accumulation in the liver and to liver cell injury in NASH.
Collapse
Affiliation(s)
- Ville T Männistö
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Marko Simonen
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Pasi Soininen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Mika Tiainen
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Antti J Kangas
- Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Dorota Kaminska
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Sari Venesmaa
- Department of Surgery, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Pirjo Käkelä
- Department of Surgery, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Vesa Kärjä
- Department of Pathology, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Helena Gylling
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland Department of Medicine, Division of Internal Medicine, University of Helsinki, Helsinki, Finland
| | - Mika Ala-Korpela
- NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland Computational Medicine, Institute of Health Sciences, University of Oulu, Oulu, Finland Department of Medicine, Oulu University Hospital, Oulu, Finland Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Jussi Pihlajamäki
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
48
|
Frazier-Wood AC, Wojczynski MK, Borecki IB, Hopkins PN, Lai CQ, Ordovas JM, Straka RJ, Tsai MY, Tiwari HK, Arnett DK. Genetic risk scores associated with baseline lipoprotein subfraction concentrations do not associate with their responses to fenofibrate. BIOLOGY 2014; 3:536-50. [PMID: 25157911 PMCID: PMC4192626 DOI: 10.3390/biology3030536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 07/29/2014] [Accepted: 08/05/2014] [Indexed: 12/11/2022]
Abstract
Lipoprotein subclass concentrations are modifiable markers of cardiovascular disease risk. Fenofibrate is known to show beneficial effects on lipoprotein subclasses, but little is known about the role of genetics in mediating the responses of lipoprotein subclasses to fenofibrate. A recent genomewide association study (GWAS) associated several single nucleotide polymorphisms (SNPs) with lipoprotein measures, and validated these associations in two independent populations. We used this information to construct genetic risk scores (GRSs) for fasting lipoprotein measures at baseline (pre-fenofibrate), and aimed to examine whether these GRSs also associated with the responses of lipoproteins to fenofibrate. Fourteen lipoprotein subclass measures were assayed in 817 men and women before and after a three week fenofibrate trial. We set significance at a Bonferroni corrected alpha <0.05 (p < 0.004). Twelve subclass measures changed with fenofibrate administration (each p = 0.003 to <0.0001). Mixed linear models which controlled for age, sex, body mass index (BMI), smoking status, pedigree and study-center, revealed that GRSs were associated with eight baseline lipoprotein measures (p < 0.004), however no GRS was associated with fenofibrate response. These results suggest that the mechanisms for changes in lipoprotein subclass concentrations with fenofibrate treatment are not mediated by the genetic risk for fasting levels.
Collapse
Affiliation(s)
- Alexis C Frazier-Wood
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Ingrid B Borecki
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Paul N Hopkins
- Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA.
| | - Chao-Qiang Lai
- Nutrition and Genomics Laboratory, Jean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA.
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA 02111, USA.
| | - Robert J Straka
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Micheal Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, MN55455, USA.
| | - Hemant K Tiwari
- Section on Statistical Genetics, University of Alabama at Birmingham, School of Public Health, AL 35294, USA.
| | - Donna K Arnett
- USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Auro K, Joensuu A, Fischer K, Kettunen J, Salo P, Mattsson H, Niironen M, Kaprio J, Eriksson JG, Lehtimäki T, Raitakari O, Jula A, Tiitinen A, Jauhiainen M, Soininen P, Kangas AJ, Kähönen M, Havulinna AS, Ala-Korpela M, Salomaa V, Metspalu A, Perola M. A metabolic view on menopause and ageing. Nat Commun 2014; 5:4708. [PMID: 25144627 DOI: 10.1038/ncomms5708] [Citation(s) in RCA: 199] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 07/16/2014] [Indexed: 12/12/2022] Open
Abstract
The ageing of the global population calls for a better understanding of age-related metabolic consequences. Here we report the effects of age, sex and menopause on serum metabolites in 26,065 individuals of Northern European ancestry. Age-specific metabolic fingerprints differ significantly by gender and, in females, a substantial atherogenic shift overlapping the time of menopausal transition is observed. In meta-analysis of 10,083 women, menopause status associates with amino acids glutamine, tyrosine and isoleucine, along with serum cholesterol measures and atherogenic lipoproteins. Among 3,204 women aged 40-55 years, menopause status associates additionally with glycine and total, monounsaturated, and omega-7 and -9 fatty acids. Our findings suggest that, in addition to lipid alterations, menopause may contribute to future metabolic and cardiovascular risk via influencing amino-acid concentrations, adding to the growing evidence of the importance of amino acids in metabolic disease progression. These observations shed light on the metabolic consequences of ageing, gender and menopause at the population level.
Collapse
Affiliation(s)
- Kirsi Auro
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3] Department of Obstetrics and Gynecology, Helsinki University Central Hospital and University of Helsinki, Haartmaninkatu 2, Helsinki 00290, Finland [4]
| | - Anni Joensuu
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3]
| | - Krista Fischer
- Estonian Genome Center, University of Tartu, Riia 23b, Tartu 51010, Estonia
| | - Johannes Kettunen
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland
| | - Perttu Salo
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Hannele Mattsson
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Marjo Niironen
- Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland
| | - Jaakko Kaprio
- 1] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [2] Departmentof Public Health, Hjelt Institute, University of Helsinki, PO Box 41 Mannerheimintie 172, Helsinki 00014, Finland [3] Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, PO Box 30 (Mannerheimintie 166), Helsinki 00300, Finland
| | - Johan G Eriksson
- 1] Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland [2] Department of General Practice and Primary Health Care, University of Helsinki, PL 20, Tukholmankatu 8B, Helsinki 00029, Finland [3] Vasa Central Hospital, Sandviksgatan 2-4, Vasa 65130, Finland [4] Folkhälsan Research Centre, Helsingfors Universitet, PB 63, Helsinki 00014, Finland [5] Unit of General Practice, Helsinki University Central Hospital, Haartmaninkatu 4, Helsinki 00290, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere University, Kalevantie 4, Tampere 33014, Finland
| | - Olli Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, Turku University Hospital, University of Turku, Kiinamyllynkatu 4-8, Turku 20521, Finland
| | - Antti Jula
- Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland
| | - Aila Tiitinen
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital and University of Helsinki, Haartmaninkatu 2, Helsinki 00290, Finland
| | - Matti Jauhiainen
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland
| | - Pasi Soininen
- 1] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland [2] NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 PL 1627, Kuopio 70211, Finland
| | - Antti J Kangas
- 1] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland [2] NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 PL 1627, Kuopio 70211, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere School of Medicine, Tampere University, Kalevantie 4, Tampere 33014, Finland
| | - Aki S Havulinna
- Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland
| | - Mika Ala-Korpela
- 1] Computational Medicine, Institute of Health Sciences, University of Oulu, Pentti Kaiteran katu 1, Oulu 90570, Finland [2] NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 PL 1627, Kuopio 70211, Finland [3] Oulu University Hospital, Kajaanintie 50, Oulu 90220, Finland [4] Computational Medicine, School of Social and Community Medicine and Medical Research Council Integrative Epidemiology Unit, University of Bristol, Senate House, Tyndall Avenue, Bristol, City of Bristol BS8 1TH, UK
| | - Veikko Salomaa
- Chronic Disease Epidemiology and Prevention Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Mannerheimintie 166, Helsinki 00300, Finland
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Riia 23b, Tartu 51010, Estonia
| | - Markus Perola
- 1] Public Health Genomics Unit, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Biomedicum 1, Haartmaninkatu 8, Helsinki 00290, Finland [2] Institute for Molecular Medicine (FIMM), University of Helsinki, Biomedicum 2, Tukholmankatu 8, Helsinki 00290, Finland [3] Estonian Genome Center, University of Tartu, Riia 23b, Tartu 51010, Estonia
| |
Collapse
|
50
|
Marttinen P, Pirinen M, Sarin AP, Gillberg J, Kettunen J, Surakka I, Kangas AJ, Soininen P, O'Reilly P, Kaakinen M, Kähönen M, Lehtimäki T, Ala-Korpela M, Raitakari OT, Salomaa V, Järvelin MR, Ripatti S, Kaski S. Assessing multivariate gene-metabolome associations with rare variants using Bayesian reduced rank regression. Bioinformatics 2014; 30:2026-34. [PMID: 24665129 PMCID: PMC4080737 DOI: 10.1093/bioinformatics/btu140] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/27/2014] [Accepted: 03/04/2014] [Indexed: 01/31/2023] Open
Abstract
MOTIVATION A typical genome-wide association study searches for associations between single nucleotide polymorphisms (SNPs) and a univariate phenotype. However, there is a growing interest to investigate associations between genomics data and multivariate phenotypes, for example, in gene expression or metabolomics studies. A common approach is to perform a univariate test between each genotype-phenotype pair, and then to apply a stringent significance cutoff to account for the large number of tests performed. However, this approach has limited ability to uncover dependencies involving multiple variables. Another trend in the current genetics is the investigation of the impact of rare variants on the phenotype, where the standard methods often fail owing to lack of power when the minor allele is present in only a limited number of individuals. RESULTS We propose a new statistical approach based on Bayesian reduced rank regression to assess the impact of multiple SNPs on a high-dimensional phenotype. Because of the method's ability to combine information over multiple SNPs and phenotypes, it is particularly suitable for detecting associations involving rare variants. We demonstrate the potential of our method and compare it with alternatives using the Northern Finland Birth Cohort with 4702 individuals, for whom genome-wide SNP data along with lipoprotein profiles comprising 74 traits are available. We discovered two genes (XRCC4 and MTHFD2L) without previously reported associations, which replicated in a combined analysis of two additional cohorts: 2390 individuals from the Cardiovascular Risk in Young Finns study and 3659 individuals from the FINRISK study. AVAILABILITY AND IMPLEMENTATION R-code freely available for download at http://users.ics.aalto.fi/pemartti/gene_metabolome/.
Collapse
Affiliation(s)
- Pekka Marttinen
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Matti Pirinen
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Antti-Pekka Sarin
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Jussi Gillberg
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Johannes Kettunen
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Ida Surakka
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Antti J Kangas
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Pasi Soininen
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Paul O'Reilly
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Marika Kaakinen
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Mika Kähönen
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Mika Ala-Korpela
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Olli T Raitakari
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Veikko Salomaa
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, Finland
| | - Marjo-Riitta Järvelin
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Samuli Ripatti
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| | - Samuel Kaski
- Department of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Disease Dynamics, Harvard School of Public Health, Boston, MA, USA Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Unit of Public Health Genomics, National Institute for Health and Welfare, Helsinki, Computational Medicine, Institute of Health Sciences, University of Oulu and Oulu University Hospital, Oulu, NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, Finland, Department of Epidemiology and Biostatistics, MRC Health Protection, Agency (HPA) Centre for Environment and Health, School of Public Health, Imperial College, London, UK, Institute of Health Sciences, Biocenter Oulu, University of Oulu, Oulu, Department of Clinical Physiology, Tampere University Hospital and University of Tampere, Department of Clinical Chemistry, Fimlab Laboratories, University of Tampere School of Medicine, Tampere, Finland, Computational Medicine, School of Social and Community Medicine and the Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK, Department of Clinical Physiology and Nuclear Medicine, Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku and Turku University Hospital, Turku, Department of Chronic Disease Prevention, National Institute for Health and Welfare, Helsinki, Unit of Primary Care, Oulu University Hospital, Department of Children and Young People and Families, National Institute for Health and Welfare, Oulu, Finland, Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK, Hjelt Institute and Department of Computer Science, Helsinki Institute for Information Technology HIIT, University of Helsinki, Helsinki, FinlandDepartment of Information and Computer Science, Helsinki Institute for Information Technology HIIT, Aalto University, Esbo, Finland, Center for Communicable Dise
| |
Collapse
|