1
|
Rajić S, Delerue T, Ronkainen J, Zhang R, Ciantar J, Kostiniuk D, Mishra PP, Lyytikäinen LP, Mononen N, Kananen L, Peters A, Winkelmann J, Kleber ME, Lorkowski S, Kähönen M, Lehtimäki T, Raitakari O, Waldenberger M, Gieger C, März W, Harville EW, Sebert S, Marttila S, Raitoharju E. Regulation of nc886 (vtRNA2-1) RNAs is associated with cardiometabolic risk factors and diseases. Clin Epigenetics 2025; 17:68. [PMID: 40301926 PMCID: PMC12042507 DOI: 10.1186/s13148-025-01871-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/01/2025] [Indexed: 05/01/2025] Open
Abstract
Non-coding 886 (nc886, vtRNA2-1) is a polymorphically imprinted gene. The methylation status of this locus has been shown to be associated with periconceptional conditions, and both the methylation status and the levels of nc886 RNAs have been shown to associate with later-life health traits. We have previously shown that nc886 RNA levels are associated not only with the methylation status of the locus, but also with a genetic polymorphism upstream from the locus. In this study, we describe the genetic and epigenetic regulators that predict lifelong nc886 RNA levels, as well as their association with cardiometabolic disease (CMD) risk factors and events. We utilised six population cohorts and one CMD cohort comprising 9058 individuals in total. The association of nc886 RNA levels, as predicted by epigenetic and genetic regulators, with CMD phenotypes was analysed using regression models, with a meta-analysis of the results. The meta-analysis showed that individuals with upregulated nc886 RNA levels have higher diastolic blood pressure (β = 0.07, p = 0.008), lower HDL levels (β = - 0.07, p = 0.006) and an increased incidence of type 2 diabetes (OR = 1.260, p = 0.013). Moreover, CMD patients with upregulated nc886 RNA levels have an increased incidence of stroke (OR = 1.581, p = 0.006) and death (OR = 1.290, p = 0.046). In conclusion, we show that individuals who are predicted to present elevated nc886 RNA levels have poorer cardiovascular health and are at an elevated risk of complications in secondary prevention. This unique mechanism yields metabolic variation in human populations, constituting a CMD risk factor that cannot be modified through lifestyle choices.
Collapse
Affiliation(s)
- Sonja Rajić
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Thomas Delerue
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Justiina Ronkainen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Ruiyuan Zhang
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Joanna Ciantar
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Daria Kostiniuk
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Laura Kananen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Faculty of Social Sciences (Health Sciences), Gerontology Research Center, Tampere University, Tampere, Finland
- Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institute, Stockholm, Sweden
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Medical Information Sciences, Biometry and Epidemiology, Ludwig-Maximilians-University, Munich, Germany
- German Research Center for Cardiovascular Disease (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Juliane Winkelmann
- Institute of Neurogenomics, Helmholtz Zentrum München, Munich, Germany
- Institute of Human Genetics, Technical University, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Stefan Lorkowski
- Institute of Nutritional Sciences, Friedrich-Schiller-University, Jena, Germany
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Jena, Germany
| | - Mika Kähönen
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- 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
| | - Melanie Waldenberger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Research Center for Cardiovascular Disease (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, University of Heidelberg, Heidelberg, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Augsburg & Mannheim, Germany
| | - Emily W Harville
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Sylvain Sebert
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Saara Marttila
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Gerontology Research Center, Tampere University, Tampere, Finland
| | - Emma Raitoharju
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
- Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland.
| |
Collapse
|
2
|
Burrows K, Heiskala A, Bradfield JP, Balkhiyarova Z, Ning L, Boissel M, Chan YM, Froguel P, Bonnefond A, Hakonarson H, Alves AC, Lawlor DA, Kaakinen M, Järvelin MR, Grant SFA, Tilling K, Prokopenko I, Sebert S, Canouil M, Warrington NM. A framework for conducting GWAS using repeated measures data with an application to childhood BMI. Nat Commun 2024; 15:10067. [PMID: 39567492 PMCID: PMC11579382 DOI: 10.1038/s41467-024-53687-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Genetic effects on changes in human traits over time are understudied and may have important pathophysiological impact. We propose a framework that enables data quality control, implements mixed models to evaluate trajectories of change in traits, and estimates phenotypes to identify age-varying genetic effects in GWAS. Using childhood BMI as an example trait, we included 71,336 participants from six cohorts and estimated the slope and area under the BMI curve within four time periods (infancy, early childhood, late childhood and adolescence) for each participant, in addition to the age and BMI at the adiposity peak and the adiposity rebound. GWAS of the 12 estimated phenotypes identified 28 genome-wide significant variants at 13 loci, one of which (in DAOA) has not been previously associated with childhood or adult BMI. Genetic studies of changes in human traits over time could uncover unique biological mechanisms influencing quantitative traits.
Collapse
Affiliation(s)
- Kimberley Burrows
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Anni Heiskala
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Jonathan P Bradfield
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Quantinuum Research LLC, Wayne, PA, USA
| | - Zhanna Balkhiyarova
- Department of Clinical and Experimental Medicine, School of Biosciences, University of Surrey, Guildford, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Lijiao Ning
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Mathilde Boissel
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Yee-Ming Chan
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Philippe Froguel
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Amelie Bonnefond
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | | | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Marika Kaakinen
- Department of Clinical and Experimental Medicine, School of Biosciences, University of Surrey, Guildford, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Marjo-Riitta Järvelin
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Struan F A Grant
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Divisions of Human Genetics and Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Kate Tilling
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Inga Prokopenko
- Department of Clinical and Experimental Medicine, School of Biosciences, University of Surrey, Guildford, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
| | - Sylvain Sebert
- Research Unit of Population Health, University of Oulu, Oulu, Finland.
| | - Mickaël Canouil
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Nicole M Warrington
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.
- Frazer Institute, University of Queensland, Brisbane, Australia.
| |
Collapse
|
3
|
Passero K, Noll JG, Verma SS, Selin C, Hall MA. Longitudinal method comparison: modeling polygenic risk for post-traumatic stress disorder over time in individuals of African and European ancestry. Front Genet 2024; 15:1203577. [PMID: 38818035 PMCID: PMC11137250 DOI: 10.3389/fgene.2024.1203577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 04/15/2024] [Indexed: 06/01/2024] Open
Abstract
Cross-sectional data allow the investigation of how genetics influence health at a single time point, but to understand how the genome impacts phenotype development, one must use repeated measures data. Ignoring the dependency inherent in repeated measures can exacerbate false positives and requires the utilization of methods other than general or generalized linear models. Many methods can accommodate longitudinal data, including the commonly used linear mixed model and generalized estimating equation, as well as the less popular fixed-effects model, cluster-robust standard error adjustment, and aggregate regression. We simulated longitudinal data and applied these five methods alongside naïve linear regression, which ignored the dependency and served as a baseline, to compare their power, false positive rate, estimation accuracy, and precision. The results showed that the naïve linear regression and fixed-effects models incurred high false positive rates when analyzing a predictor that is fixed over time, making them unviable for studying time-invariant genetic effects. The linear mixed models maintained low false positive rates and unbiased estimation. The generalized estimating equation was similar to the former in terms of power and estimation, but it had increased false positives when the sample size was low, as did cluster-robust standard error adjustment. Aggregate regression produced biased estimates when predictor effects varied over time. To show how the method choice affects downstream results, we performed longitudinal analyses in an adolescent cohort of African and European ancestry. We examined how developing post-traumatic stress symptoms were predicted by polygenic risk, traumatic events, exposure to sexual abuse, and income using four approaches-linear mixed models, generalized estimating equations, cluster-robust standard error adjustment, and aggregate regression. While the directions of effect were generally consistent, coefficient magnitudes and statistical significance differed across methods. Our in-depth comparison of longitudinal methods showed that linear mixed models and generalized estimating equations were applicable in most scenarios requiring longitudinal modeling, but no approach produced identical results even if fit to the same data. Since result discrepancies can result from methodological choices, it is crucial that researchers determine their model a priori, refrain from testing multiple approaches to obtain favorable results, and utilize as similar as possible methods when seeking to replicate results.
Collapse
Affiliation(s)
- Kristin Passero
- Virginia Institute of Psychiatric and Behavioral Genetics, Virginia Commonwealth University, Richmond, VA, United States
| | - Jennie G. Noll
- Department of Psychology, Mount Hope Family Center, University of Rochester, Rochester, NY, United States
| | - Shefali Setia Verma
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Claire Selin
- Center for Childhood Deafness, Language, and Learning, Boys Town National Research Hospital, Omaha, NE, United States
| | - Molly A. Hall
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
4
|
Ma X, Ying F, Li Z, Bai L, Wang M, Zhu D, Liu D, Wen J, Zhao G, Liu R. New insights into the genetic loci related to egg weight and age at first egg traits in broiler breeder. Poult Sci 2024; 103:103613. [PMID: 38492250 PMCID: PMC10959720 DOI: 10.1016/j.psj.2024.103613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
Egg weight (EW) and age at first egg (AFE) are economically important traits in breeder chicken production. The genetic basis of these traits, however, is far from understood, especially for broiler breeders. In this study, genetic parameter estimation, genome-wide association analysis, meta-analysis, and selective sweep analysis were carried out to identify genetic loci associated with EW and AFE in 6,842 broiler breeders. The study found that the heritability of EW ranged from 0.42 to 0.44, while the heritability of AFE was estimated at 0.33 in the maternal line. Meta-analysis and selective sweep analysis identified two colocalized regions on GGA4 that significantly influenced EW at 32 wk (EW32W) and at 43 wk (EW43W) with both paternal and maternal lines. The genes AR, YIPF6, and STARD8 were located within the significant region (GGA4: 366.86-575.50 kb), potentially affecting EW through the regulation of follicle development, cell proliferation, and lipid transfer etc. The promising genes LCORL and NCAPG were positioned within the significant region (GGA4:75.35-75.42 Mb), potentially influencing EW through pleiotropic effects on growth and development. Additionally, 3 significant regions were associated with AFE on chromosomes GGA7, GGA19, and GGA27. All of these factors affected the AFE by influencing ovarian development. In our study, the genomic information from both paternal and maternal lines was used to identify genetic regions associated with EW and AFE. Two genomic regions and eight genes were identified as the most likely candidates affecting EW and AFE. These findings contribute to a better understanding of the genetic basis of egg production traits in broiler breeders and provide new insights into future technology development.
Collapse
Affiliation(s)
- Xiaochun Ma
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Fan Ying
- Foshan Gaoming Xinguang Agricultural and Animal Industrials Corporation, Foshan 528515, China
| | - Zhengda Li
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lu Bai
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Mengjie Wang
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Dan Zhu
- Foshan Gaoming Xinguang Agricultural and Animal Industrials Corporation, Foshan 528515, China
| | - Dawei Liu
- Foshan Gaoming Xinguang Agricultural and Animal Industrials Corporation, Foshan 528515, China
| | - Jie Wen
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guiping Zhao
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ranran Liu
- State Key Laboratory of Animal Biotech Breeding; State Key Laboratory of Animal Nutrition and Feeding; Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture; Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
5
|
Burrows K, Heiskala A, Bradfield JP, Balkhiyarova Z, Ning L, Boissel M, Chan YM, Froguel P, Bonnefond A, Hakonarson H, Alves AC, Lawlor DA, Kaakinen M, Järvelin MR, Grant SF, Tilling K, Prokopenko I, Sebert S, Canouil M, Warrington NM. A framework for conducting time-varying genome-wide association studies: An application to body mass index across childhood in six multiethnic cohorts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.13.24304263. [PMID: 38559031 PMCID: PMC10980110 DOI: 10.1101/2024.03.13.24304263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Genetic effects on changes in human traits over time are understudied and may have important pathophysiological impact. We propose a framework that enables data quality control, implements mixed models to evaluate trajectories of change in traits, and estimates phenotypes to identify age-varying genetic effects in genome-wide association studies (GWASs). Using childhood body mass index (BMI) as an example, we included 71,336 participants from six cohorts and estimated the slope and area under the BMI curve within four time periods (infancy, early childhood, late childhood and adolescence) for each participant, in addition to the age and BMI at the adiposity peak and the adiposity rebound. GWAS on each of the estimated phenotypes identified 28 genome-wide significant variants at 13 loci across the 12 estimated phenotypes, one of which was novel (in DAOA) and had not been previously associated with childhood or adult BMI. Genetic studies of changes in human traits over time could uncover novel biological mechanisms influencing quantitative traits.
Collapse
Affiliation(s)
- Kimberley Burrows
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Anni Heiskala
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Jonathan P. Bradfield
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Quantinuum Research LLC, Wayne, PA, USA
| | - Zhanna Balkhiyarova
- Department of Clinical and Experimental Medicine, School of Biosciences and Medicine, University of Surrey, Guildford, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
- Section of Metabolism, Digestion and Reproduction, Department of Medicine, Imperial College London, London, UK
| | - Lijiao Ning
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Mathilde Boissel
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Yee-Ming Chan
- Division of Endocrinology, Department of Pediatrics, Boston Children’s Hospital
- Department of Pediatrics, Harvard Medical School
| | - Philippe Froguel
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Amelie Bonnefond
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Hakon Hakonarson
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Marika Kaakinen
- Department of Clinical and Experimental Medicine, School of Biosciences and Medicine, University of Surrey, Guildford, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, UK
| | - Marjo-Riitta Järvelin
- Research Unit of Population Health, University of Oulu, Oulu, Finland
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, United Kingdom
| | - Struan F.A. Grant
- Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Divisions of Human Genetics and Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kate Tilling
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
| | - Inga Prokopenko
- Department of Clinical and Experimental Medicine, School of Biosciences and Medicine, University of Surrey, Guildford, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
| | - Sylvain Sebert
- Research Unit of Population Health, University of Oulu, Oulu, Finland
| | - Mickaël Canouil
- Univ Lille, INSERM/CNRS UMR1283/8199, EGID, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Nicole M Warrington
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Frazer Institute, University of Queensland, Brisbane, Australia
| |
Collapse
|
6
|
Kalnytska O, Qvist P, Kunz S, Conrad T, Willnow TE, Schmidt V. SORCS2 activity in pancreatic α-cells safeguards insulin granule formation and release from glucose-stressed β-cells. iScience 2024; 27:108725. [PMID: 38226160 PMCID: PMC10788290 DOI: 10.1016/j.isci.2023.108725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/18/2023] [Accepted: 12/11/2023] [Indexed: 01/17/2024] Open
Abstract
Sorting receptor SORCS2 is a stress-response factor protecting neurons from acute insults, such as during epilepsy. SORCS2 is also expressed in the pancreas, yet its action in this tissue remains unknown. Combining metabolic studies in SORCS2-deficient mice with ex vivo functional analyses and single-cell transcriptomics of pancreatic tissues, we identified a role for SORCS2 in protective stress response in pancreatic islets, essential to sustain insulin release. We show that SORCS2 is predominantly expressed in islet alpha cells. Loss of expression coincides with inability of these cells to produce osteopontin, a secreted factor that facilitates insulin release from stressed beta cells. In line with diminished osteopontin levels, beta cells in SORCS2-deficient islets show gene expression patterns indicative of aggravated cell stress, and exhibit defects in insulin granule maturation and a blunted glucose response. These findings corroborate a function for SORCS2 in protective stress response that extends to metabolism.
Collapse
Affiliation(s)
- Oleksandra Kalnytska
- Molecular Cardiovascular Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Séverine Kunz
- Technology Platform for Electron Microscopy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Thomas Conrad
- Genomics Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Thomas E. Willnow
- Molecular Cardiovascular Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Vanessa Schmidt
- Molecular Cardiovascular Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| |
Collapse
|
7
|
Poliakova T, Wellington CL. Roles of peripheral lipoproteins and cholesteryl ester transfer protein in the vascular contributions to cognitive impairment and dementia. Mol Neurodegener 2023; 18:86. [PMID: 37974180 PMCID: PMC10652636 DOI: 10.1186/s13024-023-00671-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
This narrative review focuses on the role of cholesteryl ester transfer protein (CETP) and peripheral lipoproteins in the vascular contributions to cognitive impairment and dementia (VCID). Humans have a peripheral lipoprotein profile where low-density lipoproteins (LDL) represent the dominant lipoprotein fraction and high-density lipoproteins (HDL) represent a minor lipoprotein fraction. Elevated LDL-cholesterol (LDL-C) levels are well-established to cause cardiovascular disease and several LDL-C-lowering therapies are clinically available to manage this vascular risk factor. The efficacy of LDL-C-lowering therapies to reduce risk of all-cause dementia and AD is now important to address as recent studies demonstrate a role for LDL in Alzheimer's Disease (AD) as well as in all-cause dementia. The LDL:HDL ratio in humans is set mainly by CETP activity, which exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise LDL and lower HDL as CETP activity increases. Genetic and pharmacological studies support the hypothesis that CETP inhibition reduces cardiovascular risk by lowering LDL, which, by extension, may also lower VCID. Unlike humans, wild-type mice do not express catalytically active CETP and have HDL as their major lipoprotein fraction. As HDL has potent beneficial effects on endothelial cells, the naturally high HDL levels in mice protect them from vascular disorders, likely including VCID. Genetic restoration of CETP expression in mice to generate a more human-like lipid profile may increase the relevance of murine models for VCID studies. The therapeutic potential of existing and emerging LDL-lowering therapies for VCID will be discussed. Figure Legend. Cholesteryl Ester Transfer Protein in Alzheimer's Disease. CETP is mainly produced by the liver, and exchanges cholesteryl esters for triglycerides across lipoprotein fractions to raise circulating LDL and lower HDL as CETP activity increases. Low CETP activity is associated with better cardiovascular health, due to decreased LDL and increased HDL, which may also improve brain health. Although most peripheral lipoproteins cannot enter the brain parenchyma due to the BBB, it is increasingly appreciated that direct access to the vascular endothelium may enable peripheral lipoproteins to have indirect effects on brain health. Thus, lipoproteins may affect the cerebrovasculature from both sides of the BBB. Recent studies show an association between elevated plasma LDL, a well-known cardiovascular risk factor, and a higher risk of AD, and considerable evidence suggests that high HDL levels are associated with reduced CAA and lower neuroinflammation. Considering the potential detrimental role of LDL in AD and the importance of HDL's beneficial effects on endothelial cells, high CETP activity may lead to compromised BBB integrity, increased CAA deposits and greater neuroinflammation. Abbreviations: CETP - cholesteryl transfer ester protein; LDL - low-density lipoproteins; HDL - high-density lipoproteins; BBB - blood-brain barrier; CAA - cerebral amyloid angiopathy, SMC - smooth muscle cells, PVM - perivascular macrophages, RBC - red blood cells.
Collapse
Affiliation(s)
- Tetiana Poliakova
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, 2215 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafagian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- International Collaboration On Repair Discoveries, Vancouver, BC, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
8
|
Uncovering the complex genetic architecture of human plasma lipidome using machine learning methods. Sci Rep 2023; 13:3078. [PMID: 36813803 PMCID: PMC9947228 DOI: 10.1038/s41598-023-30168-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Genetic architecture of plasma lipidome provides insights into regulation of lipid metabolism and related diseases. We applied an unsupervised machine learning method, PGMRA, to discover phenotype-genotype many-to-many relations between genotype and plasma lipidome (phenotype) in order to identify the genetic architecture of plasma lipidome profiled from 1,426 Finnish individuals aged 30-45 years. PGMRA involves biclustering genotype and lipidome data independently followed by their inter-domain integration based on hypergeometric tests of the number of shared individuals. Pathway enrichment analysis was performed on the SNP sets to identify their associated biological processes. We identified 93 statistically significant (hypergeometric p-value < 0.01) lipidome-genotype relations. Genotype biclusters in these 93 relations contained 5977 SNPs across 3164 genes. Twenty nine of the 93 relations contained genotype biclusters with more than 50% unique SNPs and participants, thus representing most distinct subgroups. We identified 30 significantly enriched biological processes among the SNPs involved in 21 of these 29 most distinct genotype-lipidome subgroups through which the identified genetic variants can influence and regulate plasma lipid related metabolism and profiles. This study identified 29 distinct genotype-lipidome subgroups in the studied Finnish population that may have distinct disease trajectories and therefore could be useful in precision medicine research.
Collapse
|
9
|
Sultan S, Khan SU, Holden K, Hendi AA, Saeed S, Abbas A, Zaman U, Naeem S, Rehman KU. Reducing the Threshold of Primary Prevention of Cardiovascular Disease to 10% Over 10 Years: The Implications of Altered Intensity "Statin" Therapy Guidance. Curr Probl Cardiol 2023; 48:101486. [PMID: 36336115 DOI: 10.1016/j.cpcardiol.2022.101486] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
Cardiovascular disease (CVD) is a significant noncommunicable disease associated with high long-term mortality. In addition to more effective secondary therapies, the primary prevention of CVD has developed markedly in the past several years. This study aims to investigate the evidence and impact of reducing the threshold for primary CVD risk management to 10% over 10 years with "statin" therapy. To conduct research a systematic review utilizing 5 electronic database searches was completed for studies, analyzing the clinical effect of reducing the threshold of CVD risk to 10% over 10 years for primary prevention with statin therapy. The study included six (6) trials. Statin therapy was allocated to 31,018 participants. The mean age was 61 years and the mean follow-up was 4.6 years. The mean relative reduction in total cholesterol was 19% (from an average of), low-density lipoprotein cholesterol was 28.3% (from mmol/L to mmol/L) and triglycerides were 14.8% (from mmol/L to mmol/L). High-density lipoprotein cholesterol was observed to increase by a mean of 3.3% (from mmol/L to mmol/L). When examining all-cause mortality, statin therapy was associated with a 12% relative risk reduction compared with control, where overall rates were reduced from 1.4% to 1. % There is a 30% risk reduction in general major coronary events (from to %). There is a 19% risk reduction in general major cerebrovascular events with the statin group. While there is undoubtedly statistical evidence that supports the observation of the effectiveness of statin therapy for primary prevention, there is a risk that many hundreds of patients need to be treated to avoid a single adverse clinical outcome.
Collapse
Affiliation(s)
- Salma Sultan
- Faculty of Health Sciences and Wellbeing, University of Sunderland, UK
| | - Shahid Ullah Khan
- Department of Biochemistry, Women Medical and Dental College, Khyber Medical University Khyber, Pakhtunkhwa, Pakistan; National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan 430070, China.
| | - Keith Holden
- Faculty of Health Sciences and Wellbeing, University of Sunderland, UK
| | - Awatif A Hendi
- Department of Physics, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Sumbul Saeed
- National Key Laboratory of Crop Genetic Improvement, Huazhong Agricultural University, Wuhan 430070, China
| | - Ali Abbas
- Peshawar Institute of Cardiology, Peshawar, KPK, Pakistan
| | - Umber Zaman
- Institute of Chemical Sciences, Gomal University, Dera Ismail, Khan, KPK, Pakistan
| | - Sobia Naeem
- Department of Pharmacy, Faculty of Medical and Health Sciences, University of Poonch Rawalakot
| | | |
Collapse
|
10
|
Wuni R, Ventura EF, Curi-Quinto K, Murray C, Nunes R, Lovegrove JA, Penny M, Favara M, Sanchez A, Vimaleswaran KS. Interactions between genetic and lifestyle factors on cardiometabolic disease-related outcomes in Latin American and Caribbean populations: A systematic review. Front Nutr 2023; 10:1067033. [PMID: 36776603 PMCID: PMC9909204 DOI: 10.3389/fnut.2023.1067033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction The prevalence of cardiometabolic diseases has increased in Latin American and the Caribbean populations (LACP). To identify gene-lifestyle interactions that modify the risk of cardiometabolic diseases in LACP, a systematic search using 11 search engines was conducted up to May 2022. Methods Eligible studies were observational and interventional studies in either English, Spanish, or Portuguese. A total of 26,171 publications were screened for title and abstract; of these, 101 potential studies were evaluated for eligibility, and 74 articles were included in this study following full-text screening and risk of bias assessment. The Appraisal tool for Cross-Sectional Studies (AXIS) and the Risk Of Bias In Non-Randomized Studies-of Interventions (ROBINS-I) assessment tool were used to assess the methodological quality and risk of bias of the included studies. Results We identified 122 significant interactions between genetic and lifestyle factors on cardiometabolic traits and the vast majority of studies come from Brazil (29), Mexico (15) and Costa Rica (12) with FTO, APOE, and TCF7L2 being the most studied genes. The results of the gene-lifestyle interactions suggest effects which are population-, gender-, and ethnic-specific. Most of the gene-lifestyle interactions were conducted once, necessitating replication to reinforce these results. Discussion The findings of this review indicate that 27 out of 33 LACP have not conducted gene-lifestyle interaction studies and only five studies have been undertaken in low-socioeconomic settings. Most of the studies were cross-sectional, indicating a need for longitudinal/prospective studies. Future gene-lifestyle interaction studies will need to replicate primary research of already studied genetic variants to enable comparison, and to explore the interactions between genetic and other lifestyle factors such as those conditioned by socioeconomic factors and the built environment. The protocol has been registered on PROSPERO, number CRD42022308488. Systematic review registration https://clinicaltrials.gov, identifier CRD420223 08488.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | - Eduard F. Ventura
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | | | - Claudia Murray
- Department of Real Estate and Planning, University of Reading, Reading, United Kingdom
| | - Richard Nunes
- Department of Real Estate and Planning, University of Reading, Reading, United Kingdom
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
| | - Mary Penny
- Instituto de Investigación Nutricional, Lima, Peru
| | - Marta Favara
- Oxford Department of International Development, University of Oxford, Oxford, United Kingdom
| | - Alan Sanchez
- Grupo de Análisis para el Desarrollo (GRADE), Lima, Peru
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, United Kingdom
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading, United Kingdom
| |
Collapse
|
11
|
Gottschalk WK, Mahon S, Hodgson D, Barrera J, Hill D, Wei A, Kumar M, Dai K, Anderson L, Mihovilovic M, Lutz MW, Chiba-Falek O. The APOE-TOMM40 Humanized Mouse Model: Characterization of Age, Sex, and PolyT Variant Effects on Gene Expression. J Alzheimers Dis 2023; 94:1563-1576. [PMID: 37458041 PMCID: PMC10733864 DOI: 10.3233/jad-230451] [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] [Indexed: 07/18/2023]
Abstract
BACKGROUND The human chromosome 19q13.32 is a gene rich region and has been associated with multiple phenotypes, including late onset Alzheimer's disease (LOAD) and other age-related conditions. OBJECTIVE Here we developed the first humanized mouse model that contains the entire TOMM40 and APOE genes with all intronic and intergenic sequences including the upstream and downstream regions. Thus, the mouse model carries the human TOMM40 and APOE genes and their intact regulatory sequences. METHODS We generated the APOE-TOMM40 humanized mouse model in which the entire mouse region was replaced with the human (h)APOE-TOMM40 loci including their upstream and downstream flanking regulatory sequences using recombineering technologies. We then measured the expression of the human TOMM40 and APOE genes in the mice brain, liver, and spleen tissues using TaqMan based mRNA expression assays. RESULTS We investigated the effects of the '523' polyT genotype (S/S or VL/VL), sex, and age on the human TOMM40- and APOE-mRNAs expression levels using our new humanized mouse model. The analysis revealed tissue specific and shared effects of the '523' polyT genotype, sex, and age on the regulation of the human TOMM40 and APOE genes. Noteworthy, the regulatory effect of the '523' polyT genotype was observed for all studied organs. CONCLUSION The model offers new opportunities for basic science, translational, and preclinical drug discovery studies focused on the APOE genomic region in relation to LOAD and other conditions in adulthood.
Collapse
Affiliation(s)
- William K. Gottschalk
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Scott Mahon
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Dellila Hodgson
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Julio Barrera
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Delaney Hill
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Angela Wei
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Manish Kumar
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Kathy Dai
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| | - Lauren Anderson
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Mirta Mihovilovic
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Michael W. Lutz
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Ornit Chiba-Falek
- Division of Translational Brain Sciences, Department of Neurology, Duke University Medical Center, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
12
|
Zumaraga MP, Borel P, Bott R, Nowicki M, Lairon D, Desmarchelier C. The Interindividual Variability of Phytofluene Bioavailability is Associated with a Combination of Single Nucleotide Polymorphisms. Mol Nutr Food Res 2023; 67:e2200580. [PMID: 36349532 PMCID: PMC10078114 DOI: 10.1002/mnfr.202200580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
SCOPE Phytofluene is a colorless carotenoid with potential health benefits that displays a higher bioavailability compared to carotenoids such as lutein, β-carotene or lycopene. Several studies suggest its bioavailability displays an elevated interindividual variability. The aim of this work is to investigate whether a combination of SNPs is associated with this variability. METHODS AND RESULTS Thirty-seven healthy adult males consume a test meal that provides phytofluene from a tomato puree. Phytofluene concentrations are measured at fast and in chylomicrons at regular time intervals after meal intake. Identification of the combination of SNPs that best explained the interindividual variability of the phytofluene response is assessed by partial least squares regression. There is a large interindividual variability in the phytofluene response, with CV = 88%. Phytofluene bioavailability is positively correlated with fasting plasma phytofluene concentration (r = 0.57; p = 2 × 10-4 ). A robust partial least squares regression model comprising 14 SNPs near or within 11 genes (ABCA1-rs2487059, rs2515629, and rs4149316, APOC1-rs445925, CD36-rs3211881, ELOVL5-rs6941533, FABP1-rs10185660, FADS3-rs1000778, ISX-rs130461, and rs17748559, LIPC-rs17240713, LPL-rs7005359, LYPLAL1-rs1351472, SETD7-rs11936429) explains 51% (adjusted R2 ) of the interindividual variability in phytofluene bioavailability. CONCLUSIONS This study reports a combination of SNPs that is associated with a significant part of the interindividual variability of phytofluene bioavailability in a healthy male adult population.
Collapse
Affiliation(s)
- Mark Pretzel Zumaraga
- C2VN, Aix Marseille Univ, INRAE, INSERM, Faculté de Médecine, 27 boulevard Jean Moulin, Marseille, 13005, France.,Department of Science and Technology, Food and Nutrition Research Institute, Bicutan, Taguig City, NCR 1631, Philippines
| | - Patrick Borel
- C2VN, Aix Marseille Univ, INRAE, INSERM, Faculté de Médecine, 27 boulevard Jean Moulin, Marseille, 13005, France
| | - Romain Bott
- C2VN, Aix Marseille Univ, INRAE, INSERM, Faculté de Médecine, 27 boulevard Jean Moulin, Marseille, 13005, France
| | - Marion Nowicki
- C2VN, Aix Marseille Univ, INRAE, INSERM, Faculté de Médecine, 27 boulevard Jean Moulin, Marseille, 13005, France
| | - Denis Lairon
- C2VN, Aix Marseille Univ, INRAE, INSERM, Faculté de Médecine, 27 boulevard Jean Moulin, Marseille, 13005, France
| | - Charles Desmarchelier
- C2VN, Aix Marseille Univ, INRAE, INSERM, Faculté de Médecine, 27 boulevard Jean Moulin, Marseille, 13005, France.,Institut Universitaire de France (IUF), Paris, France
| |
Collapse
|
13
|
Life-Course Associations between Blood Pressure-Related Polygenic Risk Scores and Hypertension in the Bogalusa Heart Study. Genes (Basel) 2022; 13:genes13081473. [PMID: 36011384 PMCID: PMC9408577 DOI: 10.3390/genes13081473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/30/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Genetic information may help to identify individuals at increased risk for hypertension in early life, prior to the manifestation of elevated blood pressure (BP) values. We examined 369 Black and 832 White Bogalusa Heart Study (BHS) participants recruited in childhood and followed for approximately 37 years. The multi-ancestry genome-wide polygenic risk scores (PRSs) for systolic BP (SBP), diastolic BP (DBP), and hypertension were tested for an association with incident hypertension and stage 2 hypertension using Cox proportional hazards models. Race-stratified analyses were adjusted for baseline age, age2, sex, body mass index, genetic principal components, and BP. In Black participants, each standard deviation increase in SBP and DBP PRS conferred a 38% (p = 0.009) and 22% (p = 0.02) increased risk of hypertension and a 74% (p < 0.001) and 50% (p < 0.001) increased risk of stage 2 hypertension, respectively, while no association was observed with the hypertension PRSs. In Whites, each standard deviation increase in SBP, DBP, and hypertension PRS conferred a 24% (p < 0.05), 29% (p = 0.01), and 25% (p < 0.001) increased risk of hypertension, and a 27% (p = 0.08), 29% (0.01), and 42% (p < 0.001) increased risk of stage 2 hypertension, respectively. The addition of BP PRSs to the covariable-only models generally improved the C-statistics (p < 0.05). Multi-ancestry BP PRSs demonstrate the utility of genomic information in the early life prediction of hypertension.
Collapse
|
14
|
Raitakari O, Kivelä A, Pahkala K, Rovio S, Mykkänen J, Ahola-Olli A, Loo BM, Lyytikäinen LP, Lehtimäki T, Kähönen M, Juonala M, Rönnemaa T, Lamina C, Kronenberg F, Viikari J. Long-term tracking and population characteristics of lipoprotein (a) in the cardiovascular risk in young finns study. Atherosclerosis 2022; 356:18-27. [DOI: 10.1016/j.atherosclerosis.2022.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/01/2022] [Accepted: 07/13/2022] [Indexed: 11/16/2022]
|
15
|
Tölli P, Keltikangas‐Järvinen L, Lehtimäki T, Ravaja N, Hintsanen M, Ahola‐Olli A, Pahkala K, Kähönen M, Hutri‐Kähönen N, Laitinen TT, Tossavainen P, Taittonen L, Dobewall H, Jokinen E, Raitakari O, Cloninger CR, Rovio S, Saarinen A. The relationship between temperament, polygenic score for intelligence and cognition: A population-based study of middle-aged adults. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12798. [PMID: 35170850 PMCID: PMC9744494 DOI: 10.1111/gbb.12798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 01/12/2022] [Accepted: 01/15/2022] [Indexed: 11/30/2022]
Abstract
We investigated whether temperament modifies an association between polygenic intelligence potential and cognitive test performance in midlife. The participants (n = 1647, born between 1962 and 1977) were derived from the Young Finns Study. Temperament was assessed with Temperament and Character Inventory over a 15-year follow-up (1997, 2001, 2007, 2012). Polygenic intelligence potential was assessed with a polygenic score for intelligence. Cognitive performance (visual memory, reaction time, sustained attention, spatial working memory) was assessed with CANTAB in midlife. The PGSI was significantly associated with the overall cognitive performance and performance in visual memory, sustained attention and working memory tests but not reaction time test. Temperament did not correlate with polygenic score for intelligence and did not modify an association between the polygenic score and cognitive performance, either. High persistence was associated with higher visual memory (B = 0.092; FDR-adj. p = 0.007) and low harm avoidance with higher overall cognitive performance, specifically better reaction time (B = -0.102; FDR-adj; p = 0.007). The subscales of harm avoidance had different associations with cognitive performance: higher "anticipatory worry," higher "fatigability," and lower "shyness with strangers" were associated with lower cognitive performance, while the role of "fear of uncertainty" was subtest-related. In conclusion, temperament does not help or hinder one from realizing their genetic potential for intelligence. The overall modest relationships between temperament and cognitive performance advise caution if utilizing temperament-related information e.g. in working-life recruitments. Cognitive abilities may be influenced by temperament variables, such as the drive for achievement and anxiety about test performance, but they involve distinct systems of learning and memory.
Collapse
Affiliation(s)
- Pekka Tölli
- Department of Psychology and Logopedics, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | | | - Terho Lehtimäki
- Department of Clinical ChemistryFimlab Laboratories, and Finnish Cardiovascular Research CenterTampereFinland
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Niklas Ravaja
- Department of Psychology and Logopedics, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| | - Mirka Hintsanen
- Research Unit of Psychology, Faculty of EducationUniversity of OuluOuluFinland
| | - Ari Ahola‐Olli
- Department of Internal MedicineSatasairaala Central HospitalPoriFinland
- Psychiatric and Neurodevelopmental Genetics UnitDepartment of Psychiatry, Massachusetts General HospitalBostonMassachusettsUSA
- Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland
| | - Katja Pahkala
- Research Centre for Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
- Sports Exercise Medicine Unit, Department of Physical Activity and HealthPaavo Nurmi CentreTurkuFinland
| | - Mika Kähönen
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Department of Clinical PhysiologyTampere University HospitalTampereFinland
| | - Nina Hutri‐Kähönen
- Tampere Centre for Skills Training and SimulationTampere UniversityTampereFinland
| | - Tomi T. Laitinen
- Research Centre for Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
- Sports Exercise Medicine Unit, Department of Physical Activity and HealthPaavo Nurmi CentreTurkuFinland
| | - Päivi Tossavainen
- Department of Pediatrics and AdolescentsOulu University HospitalOuluFinland
- PEDEGO Research Unit and Medical Research Center OuluUniversity of OuluOuluFinland
| | - Leena Taittonen
- Vaasa Central HospitalVaasaFinland
- Department of PediatricsUniversity of OuluOuluFinland
| | - Henrik Dobewall
- Research Unit of Psychology, Faculty of EducationUniversity of OuluOuluFinland
| | - Eero Jokinen
- Department of PediatricsUniversity of HelsinkiHelsinkiFinland
- Hospital for Children and AdolescentsHelsinki University HospitalHelsinkiFinland
| | - Olli Raitakari
- Department of Internal MedicineSatasairaala Central HospitalPoriFinland
- Centre for Population Health ResearchUniversity of Turku and Turku University HospitalTurkuFinland
- Department of Clinical Physiology and Nuclear MedicineTurku University HospitalTurkuFinland
| | | | - Suvi Rovio
- Research Centre for Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
| | - Aino Saarinen
- Department of Psychology and Logopedics, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
16
|
Li S, Li S, Su S, Zhang H, Shen J, Wen Y. Gene Region Association Analysis of Longitudinal Quantitative Traits Based on a Function-On-Function Regression Model. Front Genet 2022; 13:781740. [PMID: 35265102 PMCID: PMC8899465 DOI: 10.3389/fgene.2022.781740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
In the process of growth and development in life, gene expressions that control quantitative traits will turn on or off with time. Studies of longitudinal traits are of great significance in revealing the genetic mechanism of biological development. With the development of ultra-high-density sequencing technology, the associated analysis has tremendous challenges to statistical methods. In this paper, a longitudinal functional data association test (LFDAT) method is proposed based on the function-on-function regression model. LFDAT can simultaneously treat phenotypic traits and marker information as continuum variables and analyze the association of longitudinal quantitative traits and gene regions. Simulation studies showed that: 1) LFDAT performs well for both linkage equilibrium simulation and linkage disequilibrium simulation, 2) LFDAT has better performance for gene regions (include common variants, low-frequency variants, rare variants and mixture), and 3) LFDAT can accurately identify gene switching in the growth and development stage. The longitudinal data of the Oryza sativa projected shoot area is analyzed by LFDAT. It showed that there is the advantage of quick calculations. Further, an association analysis was conducted between longitudinal traits and gene regions by integrating the micro effects of multiple related variants and using the information of the entire gene region. LFDAT provides a feasible method for studying the formation and expression of longitudinal traits.
Collapse
Affiliation(s)
- Shijing Li
- College of Computer and Information Science, Fujian Agriculture and Forestry University, Fuzhou, China.,> Institute of Statistics and Application, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Shiqin Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shaoqiang Su
- College of Computer and Information Science, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hui Zhang
- College of Computer and Information Science, Fujian Agriculture and Forestry University, Fuzhou, China.,> Institute of Statistics and Application, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jiayu Shen
- College of Computer and Information Science, Fujian Agriculture and Forestry University, Fuzhou, China.,> Institute of Statistics and Application, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yongxian Wen
- College of Computer and Information Science, Fujian Agriculture and Forestry University, Fuzhou, China.,> Institute of Statistics and Application, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
17
|
Chung W, Cho Y. Bayesian mixed models for longitudinal genetic data: theory, concepts, and simulation studies. Genomics Inform 2022; 20:e8. [PMID: 35399007 PMCID: PMC9001998 DOI: 10.5808/gi.21080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/03/2022] [Indexed: 01/02/2023] Open
Abstract
Despite the success of recent genome-wide association studies investigating longitudinal traits, a large fraction of overall heritability remains unexplained. This suggests that some of the missing heritability may be accounted for by gene-gene and gene-time/environment interactions. In this paper, we develop a Bayesian variable selection method for longitudinal genetic data based on mixed models. The method jointly models the main effects and interactions of all candidate genetic variants and non-genetic factors and has higher statistical power than previous approaches. To account for the within-subject dependence structure, we propose a grid-based approach that models only one fixed-dimensional covariance matrix, which is thus applicable to data where subjects have different numbers of time points. We provide the theoretical basis of our Bayesian method and then illustrate its performance using data from the 1000 Genome Project with various simulation settings. Several simulation studies show that our multivariate method increases the statistical power compared to the corresponding univariate method and can detect gene-time/environment interactions well. We further evaluate our method with different numbers of individuals, variants, and causal variants, as well as different trait-heritability, and conclude that our method performs reasonably well with various simulation settings.
Collapse
Affiliation(s)
- Wonil Chung
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Korea.,Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Youngkwang Cho
- Department of Statistics and Actuarial Science, Soongsil University, Seoul 06978, Korea
| |
Collapse
|
18
|
Schubert R, Geoffroy E, Gregga I, Mulford AJ, Aguet F, Ardlie K, Gerszten R, Clish C, Van Den Berg D, Taylor KD, Durda P, Johnson WC, Cornell E, Guo X, Liu Y, Tracy R, Conomos M, Blackwell T, Papanicolaou G, Lappalainen T, Mikhaylova AV, Thornton TA, Cho MH, Gignoux CR, Lange L, Lange E, Rich SS, Rotter JI, Manichaikul A, Im HK, Wheeler HE. Protein prediction for trait mapping in diverse populations. PLoS One 2022; 17:e0264341. [PMID: 35202437 PMCID: PMC8870552 DOI: 10.1371/journal.pone.0264341] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022] Open
Abstract
Genetically regulated gene expression has helped elucidate the biological mechanisms underlying complex traits. Improved high-throughput technology allows similar interrogation of the genetically regulated proteome for understanding complex trait mechanisms. Here, we used the Trans-omics for Precision Medicine (TOPMed) Multi-omics pilot study, which comprises data from Multi-Ethnic Study of Atherosclerosis (MESA), to optimize genetic predictors of the plasma proteome for genetically regulated proteome-wide association studies (PWAS) in diverse populations. We built predictive models for protein abundances using data collected in TOPMed MESA, for which we have measured 1,305 proteins by a SOMAscan assay. We compared predictive models built via elastic net regression to models integrating posterior inclusion probabilities estimated by fine-mapping SNPs prior to elastic net. In order to investigate the transferability of predictive models across ancestries, we built protein prediction models in all four of the TOPMed MESA populations, African American (n = 183), Chinese (n = 71), European (n = 416), and Hispanic/Latino (n = 301), as well as in all populations combined. As expected, fine-mapping produced more significant protein prediction models, especially in African ancestries populations, potentially increasing opportunity for discovery. When we tested our TOPMed MESA models in the independent European INTERVAL study, fine-mapping improved cross-ancestries prediction for some proteins. Using GWAS summary statistics from the Population Architecture using Genomics and Epidemiology (PAGE) study, which comprises ∼50,000 Hispanic/Latinos, African Americans, Asians, Native Hawaiians, and Native Americans, we applied S-PrediXcan to perform PWAS for 28 complex traits. The most protein-trait associations were discovered, colocalized, and replicated in large independent GWAS using proteome prediction model training populations with similar ancestries to PAGE. At current training population sample sizes, performance between baseline and fine-mapped protein prediction models in PWAS was similar, highlighting the utility of elastic net. Our predictive models in diverse populations are publicly available for use in proteome mapping methods at https://doi.org/10.5281/zenodo.4837327.
Collapse
Affiliation(s)
- Ryan Schubert
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL, United States of America
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| | - Elyse Geoffroy
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| | - Isabelle Gregga
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
| | - Ashley J. Mulford
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| | - Francois Aguet
- Broad Institute, Cambridge, MA, United States of America
| | - Kristin Ardlie
- Broad Institute, Cambridge, MA, United States of America
| | - Robert Gerszten
- Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Clary Clish
- Broad Institute, Cambridge, MA, United States of America
| | - David Van Den Berg
- University of Southern California, Los Angeles, CA, United States of America
| | - 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, United States of America
| | - Peter Durda
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, United States of America
| | - W. Craig Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, United States of America
| | - Elaine Cornell
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, United States of America
| | - 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, United States of America
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC, United States of America
| | - Russell Tracy
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, United States of America
| | - Matthew Conomos
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Tom Blackwell
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, United States of America
| | - George Papanicolaou
- Epidemiology Branch, National Heart, Lung and Blood Institute, Bethesda, MD, United States of America
| | - Tuuli Lappalainen
- New York Genome Center and Department of Systems Biology, Columbia University, New York, NY United States of America
| | - Anna V. Mikhaylova
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Timothy A. Thornton
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Michael H. Cho
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA, United States of America
| | - Christopher R. Gignoux
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Leslie Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Ethan Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States of America
| | - 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, United States of America
| | | | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States of America
| | - Hae Kyung Im
- Section of Genetic Medicine, The University of Chicago, Chicago, IL, United States of America
| | - Heather E. Wheeler
- Department of Biology, Loyola University Chicago, Chicago, IL, United States of America
- Program in Bioinformatics, Loyola University Chicago, Chicago, IL, United States of America
| |
Collapse
|
19
|
Grid-based Gaussian process models for longitudinal genetic data. COMMUNICATIONS FOR STATISTICAL APPLICATIONS AND METHODS 2022. [DOI: 10.29220/csam.2022.29.1.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
20
|
Wuni R, Kuhnle GGC, Wynn-Jones AA, Vimaleswaran KS. A Nutrigenetic Update on CETP Gene–Diet Interactions on Lipid-Related Outcomes. Curr Atheroscler Rep 2022; 24:119-132. [PMID: 35098451 PMCID: PMC8924099 DOI: 10.1007/s11883-022-00987-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2021] [Indexed: 02/08/2023]
Abstract
Purpose of Review An abnormal lipid profile is considered a main risk factor for cardiovascular diseases and evidence suggests that single nucleotide polymorphisms (SNPs) in the cholesteryl ester transfer protein (CETP) gene contribute to variations in lipid levels in response to dietary intake. The objective of this review was to identify and discuss nutrigenetic studies assessing the interactions between CETP SNPs and dietary factors on blood lipids. Recent Findings Relevant articles were obtained through a literature search of PubMed and Google Scholar through to July 2021. An article was included if it examined an interaction between CETP SNPs and dietary factors on blood lipids. From 49 eligible nutrigenetic studies, 27 studies reported significant interactions between 8 CETP SNPs and 17 dietary factors on blood lipids in 18 ethnicities. The discrepancies in the study findings could be attributed to genetic heterogeneity, and differences in sample size, study design, lifestyle and measurement of dietary intake. The most extensively studied ethnicities were those of Caucasian populations and majority of the studies reported an interaction with dietary fat intake. The rs708272 (TaqIB) was the most widely studied CETP SNP, where ‘B1’ allele was associated with higher CETP activity, resulting in lower high-density lipoprotein cholesterol and higher serum triglycerides under the influence of high dietary fat intake. Summary Overall, the findings suggest that CETP SNPs might alter blood lipid profiles by modifying responses to diet, but further large studies in multiple ethnic groups are warranted to identify individuals at risk of adverse lipid response to diet. Supplementary Information The online version contains supplementary material available at 10.1007/s11883-022-00987-y.
Collapse
|
21
|
Grid-based Gaussian process models for longitudinal genetic data. COMMUNICATIONS FOR STATISTICAL APPLICATIONS AND METHODS 2022. [DOI: 10.29220/csam.2022.29.1.745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
22
|
Schmidt V, Horváth C, Dong H, Blüher M, Qvist P, Wolfrum C, Willnow TE. SORLA is required for insulin-induced expansion of the adipocyte precursor pool in visceral fat. J Cell Biol 2021; 220:e202006058. [PMID: 34779857 PMCID: PMC8598079 DOI: 10.1083/jcb.202006058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 06/19/2021] [Accepted: 09/08/2021] [Indexed: 01/24/2023] Open
Abstract
Visceral adipose tissue shows remarkable plasticity, constantly replacing mature adipocytes from an inherent pool of adipocyte precursors. The number of precursors is set in the juvenile organism and remains constant in adult life. Which signals drive precursor pool expansion in juveniles and why they operate in visceral but not in subcutaneous white adipose tissue (WAT) are unclear. Using mouse models, we identified the insulin-sensitizing receptor SORLA as a molecular factor explaining the distinct proliferative capacity of visceral WAT. High levels of SORLA activity in precursors of juvenile visceral WAT prime these cells for nutritional stimuli provided through insulin, promoting mitotic expansion of the visceral precursor cell pool in overfed juvenile mice. SORLA activity is low in subcutaneous precursors, blunting their response to insulin and preventing diet-induced proliferation of this cell type. Our findings provide a molecular explanation for the unique proliferative properties of juvenile visceral WAT, and for the genetic association of SORLA with visceral obesity in humans.
Collapse
Affiliation(s)
- Vanessa Schmidt
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Carla Horváth
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Hua Dong
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Per Qvist
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Genomics and Personalized Medicine, Aarhus University, Aarhus, Denmark
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, ETH Zurich, Schwerzenbach, Switzerland
| | - Thomas E. Willnow
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
23
|
Wang T, Lu H, Zeng P. Identifying pleiotropic genes for complex phenotypes with summary statistics from a perspective of composite null hypothesis testing. Brief Bioinform 2021; 23:6375058. [PMID: 34571531 DOI: 10.1093/bib/bbab389] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/06/2021] [Accepted: 08/28/2021] [Indexed: 12/13/2022] Open
Abstract
Pleiotropy has important implication on genetic connection among complex phenotypes and facilitates our understanding of disease etiology. Genome-wide association studies provide an unprecedented opportunity to detect pleiotropic associations; however, efficient pleiotropy test methods are still lacking. We here consider pleiotropy identification from a methodological perspective of high-dimensional composite null hypothesis and propose a powerful gene-based method called MAIUP. MAIUP is constructed based on the traditional intersection-union test with two sets of independent P-values as input and follows a novel idea that was originally proposed under the high-dimensional mediation analysis framework. The key improvement of MAIUP is that it takes the composite null nature of pleiotropy test into account by fitting a three-component mixture null distribution, which can ultimately generate well-calibrated P-values for effective control of family-wise error rate and false discover rate. Another attractive advantage of MAIUP is its ability to effectively address the issue of overlapping subjects commonly encountered in association studies. Simulation studies demonstrate that compared with other methods, only MAIUP can maintain correct type I error control and has higher power across a wide range of scenarios. We apply MAIUP to detect shared associated genes among 14 psychiatric disorders with summary statistics and discover many new pleiotropic genes that are otherwise not identified if failing to account for the issue of composite null hypothesis testing. Functional and enrichment analyses offer additional evidence supporting the validity of these identified pleiotropic genes associated with psychiatric disorders. Overall, MAIUP represents an efficient method for pleiotropy identification.
Collapse
Affiliation(s)
- Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Haojie Lu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.,Center for Medical Statistics and Data Analysis, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.,Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| |
Collapse
|
24
|
Dobewall H, Keltikangas-Järvinen L, Saarinen A, Lyytikäinen LP, Zwir I, Cloninger R, Raitakari OT, Lehtimäki T, Hintsanen M. Genetic differential susceptibility to the parent-child relationship quality and the life span development of compassion. Dev Psychobiol 2021; 63:e22184. [PMID: 34423428 DOI: 10.1002/dev.22184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 06/26/2021] [Accepted: 07/06/2021] [Indexed: 11/11/2022]
Abstract
The development of compassion for others might be influenced by the social experiences made during childhood and has a genetic component. No research has yet investigated whether the parent-child relationship quality interacts with genetic variation in the oxytocin and dopamine systems in predicting compassion over the life span. In the prospective Young Finns Study (N = 2099, 43.9% men), we examined the interaction between mother-reported emotional warmth and intolerance toward their child assessed in 1980 (age of participants, 3-18 years) and two established genetic risk scores for oxytocin levels and dopamine signaling activity. Dispositional compassion for others was measured with the Temperament and Character Inventory 1997, 2001, and 2012 (age of participants, 20-50 years). We found a gene-environment interaction (p = .031) that remained marginally significant after adjustment for multiple testing. In line with the differential susceptibility hypothesis, only participants who carry alleles associated with low dopamine signaling activity had higher levels of compassion when growing up with emotionally warm parents, whereas they had lower levels of compassion when their parents were emotionally cold. Children's genetic variability in the dopamine system might result in plasticity to early environmental influences that have a long-lasting effect on the development of compassion. However, our findings need replication.
Collapse
Affiliation(s)
- Henrik Dobewall
- Division of Psychology, Faculty of Education, University of Oulu, Oulu, Finland.,Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Aino Saarinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Igor Zwir
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States.,Department of Computer Science, University of Granada, Granada, Spain
| | - Robert Cloninger
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Olli T Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland.,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
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mirka Hintsanen
- Division of Psychology, Faculty of Education, University of Oulu, Oulu, Finland
| |
Collapse
|
25
|
Dobewall H, Saarinen A, Lyytikäinen LP, Keltikangas-Järvinen L, Lehtimäki T, Hintsanen M. Functional Polymorphisms in Oxytocin and Dopamine Pathway Genes and the Development of Dispositional Compassion Over Time: The Young Finns Study. Front Psychol 2021; 12:576346. [PMID: 33897514 PMCID: PMC8060576 DOI: 10.3389/fpsyg.2021.576346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/03/2021] [Indexed: 12/21/2022] Open
Abstract
Background: We define compassion as an enduring disposition that centers upon empathetic concern for another person's suffering and the motivation to act to alleviate it. The contribution of specific candidate genes to the development of dispositional compassion for others is currently unknown. We examine candidate genes in the oxytocin and dopamine signaling pathways. Methods: In a 32-year follow-up of the Young Finns Study (N = 2,130, 44.0% men), we examined with multiple indicators latent growth curve modeling the molecular genetic underpinnings of dispositional compassion for others across the life span. We selected five single nucleotide polymorphisms (SNPs) whose functions are known in humans: rs2268498 (OXTR), rs3796863 (CD38) (related to lower oxytocin levels), rs1800497 (ANKK1/DRD2), rs4680 (COMT), and rs1611115 (DBH) (related to higher dopamine levels). Compassion was measured with Cloninger's Temperament and Character Inventory on three repeated observations spanning 15 years (1997–2012). Differences between gender were tested. Results: We did not find an effect of the five SNPs in oxytocin and dopamine pathway genes on the initial levels of dispositional compassion for others. Individuals who carry one or two copies of the T-allele of DBH rs1611115, however, tend to increase faster in compassion over time than those homozygotes for the C-allele, b = 0.063 (SE = 0.027; p = 0.018). This effect was largely driven by male participants, 0.206 (SE = 0.046; p < 0.001), and was not significant in female participants when analyzed separately. Conclusions: Men who are known to have, on average, lower compassion than women seem to reduce this difference over time if they carry the T-allele of DBH rs1611115. The direction of the association indicates that dopamine signaling activity rather than overall dopamine levels might drive the development of compassion.
Collapse
Affiliation(s)
- Henrik Dobewall
- Research Unit of Psychology, University of Oulu, Oulu, Finland.,Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Aino Saarinen
- Research Unit of Psychology, University of Oulu, Oulu, Finland.,Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Leo-Pekka Lyytikäinen
- Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | | | - Terho Lehtimäki
- Fimlab Laboratories, and Finnish Cardiovascular Research Center - Tampere, Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mirka Hintsanen
- Research Unit of Psychology, University of Oulu, Oulu, Finland
| |
Collapse
|
26
|
Gautier T, Deckert V, Aires V, Le Guern N, Proukhnitzky L, Patoli D, Lemaire S, Maquart G, Bataille A, Xolin M, Magnani C, Masson D, Harscoët E, Da Silva B, Houdebine LM, Jolivet G, Lagrost L. Human apolipoprotein C1 transgenesis reduces atherogenesis in hypercholesterolemic rabbits. Atherosclerosis 2021; 320:10-18. [PMID: 33497863 DOI: 10.1016/j.atherosclerosis.2021.01.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 12/03/2020] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND AND AIMS Apolipoprotein (apo) C1 is a 6.6 kDa protein associated with HDL and VLDL. ApoC1 alters triglyceride clearance, and it also favors cholesterol accumulation in HDL, especially by inhibiting CETP in human plasma. Apart from studies in mice, which lack CETP, the impact of apoC1 on atherosclerosis in animal models expressing CETP, like in humans, is not known. This study aimed at determining the net effect of human apoC1 on atherosclerosis in rabbits, a species with naturally high CETP activity but with endogenous apoC1 without CETP inhibitory potential. METHODS Rabbits expressing a human apoC1 transgene (HuApoC1Tg) were generated and displayed significant amounts of human apoC1 in plasma. RESULTS After cholesterol feeding, atherosclerosis lesions were significantly less extensive (-22%, p < 0.05) and HDL displayed a reduced ability to serve as CETP substrates (-25%, p < 0.05) in HuApoC1Tg rabbits than in WT littermates. It was associated with rises in plasma HDL cholesterol level and PON-1 activity, and a decrease in the plasma level of the lipid oxidation markers 12(S)-HODE and 8(S)HETE. In chow-fed animals, the level of HDL-cholesterol was also significantly higher in HuApoC1Tg than in WT animals (0.83 ± 0.11 versus 0.73 ± 0.11 mmol/L, respectively, p < 0.05), and it was associated with significantly lower CETP activity (cholesteryl ester transfer rate, -10%, p < 0.05; specific CETP activity, -14%, p < 0.05). CONCLUSIONS Constitutive expression of fully functional human apoC1 in transgenic rabbit attenuates atherosclerosis. It was found to relate, at least in part, to the inhibition of plasma CETP activity and to alterations in plasma HDL.
Collapse
Affiliation(s)
- Thomas Gautier
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France.
| | - Valérie Deckert
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Virginie Aires
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Naig Le Guern
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Lil Proukhnitzky
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Danish Patoli
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Stéphanie Lemaire
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Guillaume Maquart
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Amandine Bataille
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Marion Xolin
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - Charlène Magnani
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France
| | - David Masson
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France; University Hospital of Dijon, Dijon, France
| | - Erwana Harscoët
- Université Paris-Saclay, INRAE, ENVA, UVSQ, BREED, 78350, Jouy-en-Josas, France
| | - Bruno Da Silva
- Université Paris-Saclay, INRAE, ENVA, UVSQ, BREED, 78350, Jouy-en-Josas, France; Laboratory of Developmental Biology, CNRS UMR7622, Université Pierre et Marie Curie, Paris, France
| | | | - Geneviève Jolivet
- Université Paris-Saclay, INRAE, ENVA, UVSQ, BREED, 78350, Jouy-en-Josas, France
| | - Laurent Lagrost
- INSERM / University of Bourgogne Franche-Comté LNC UMR1231 and LipSTIC LabEx, UFR Sciences de Santé, Dijon, France; University Hospital of Dijon, Dijon, France
| |
Collapse
|
27
|
Strawbridge RJ, Ward J, Bailey ME, Cullen B, Ferguson A, Graham N, Johnston KJ, Lyall LM, Pearsall R, Pell J, Shaw RJ, Tank R, Lyall DM, Smith DJ. Carotid Intima-Media Thickness: Novel Loci, Sex-Specific Effects, and Genetic Correlations With Obesity and Glucometabolic Traits in UK Biobank. Arterioscler Thromb Vasc Biol 2020; 40:446-461. [PMID: 31801372 PMCID: PMC6975521 DOI: 10.1161/atvbaha.119.313226] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/21/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Atherosclerosis is the underlying cause of most cardiovascular disease, but mechanisms underlying atherosclerosis are incompletely understood. Ultrasound measurement of the carotid intima-media thickness (cIMT) can be used to measure vascular remodeling, which is indicative of atherosclerosis. Genome-wide association studies have identified many genetic loci associated with cIMT, but heterogeneity of measurements collected by many small cohorts have been a major limitation in these efforts. Here, we conducted genome-wide association analyses in UKB (UK Biobank; N=22 179), the largest single study with consistent cIMT measurements. Approach and Results: We used BOLT-LMM software to run linear regression of cIMT in UKB, adjusted for age, sex, and genotyping chip. In white British participants, we identified 5 novel loci associated with cIMT and replicated most previously reported loci. In the first sex-specific analyses of cIMT, we identified a locus on chromosome 5, associated with cIMT in women only and highlight VCAN as a good candidate gene at this locus. Genetic correlations with body mass index and glucometabolic traits were also observed. Two loci influenced risk of ischemic heart disease. CONCLUSIONS These findings replicate previously reported associations, highlight novel biology, and provide new directions for investigating the sex differences observed in cardiovascular disease presentation and progression.
Collapse
Affiliation(s)
- Rona J. Strawbridge
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Joey Ward
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Mark E.S. Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences (M.E.S.B., K.J.A.J.), University of Glasgow, United Kingdom
| | - Breda Cullen
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Amy Ferguson
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Nicholas Graham
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Keira J.A. Johnston
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
- School of Life Sciences, College of Medical, Veterinary and Life Sciences (M.E.S.B., K.J.A.J.), University of Glasgow, United Kingdom
- Division of Psychiatry, College of Medicine, University of Edinburgh, United Kingdom (K.J.A.J.)
| | - Laura M. Lyall
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Robert Pearsall
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Jill Pell
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Richard J. Shaw
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
- Health Data Research United Kingdom (R.J.S.)
- Cardiovascular Medicine Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden (R.J.S.)
| | - Rachana Tank
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Donald M. Lyall
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| | - Daniel J. Smith
- From the Institute of Health and Wellbeing (R.J.S., J.W., B.C., A.F., N.G., K.J.A.J., L.M.L., R.P., J.P., R.J.S., R.T., D.M.L., D.J.S.), University of Glasgow, United Kingdom
| |
Collapse
|
28
|
Abstract
Human personality is 30-60% heritable according to twin and adoption studies. Hundreds of genetic variants are expected to influence its complex development, but few have been identified. We used a machine learning method for genome-wide association studies (GWAS) to uncover complex genotypic-phenotypic networks and environmental interactions. The Temperament and Character Inventory (TCI) measured the self-regulatory components of personality critical for health (i.e., the character traits of self-directedness, cooperativeness, and self-transcendence). In a discovery sample of 2149 healthy Finns, we identified sets of single-nucleotide polymorphisms (SNPs) that cluster within particular individuals (i.e., SNP sets) regardless of phenotype. Second, we identified five clusters of people with distinct profiles of character traits regardless of genotype. Third, we found 42 SNP sets that identified 727 gene loci and were significantly associated with one or more of the character profiles. Each character profile was related to different SNP sets with distinct molecular processes and neuronal functions. Environmental influences measured in childhood and adulthood had small but significant effects. We confirmed the replicability of 95% of the 42 SNP sets in healthy Korean and German samples, as well as their associations with character. The identified SNPs explained nearly all the heritability expected for character in each sample (50 to 58%). We conclude that self-regulatory personality traits are strongly influenced by organized interactions among more than 700 genes despite variable cultures and environments. These gene sets modulate specific molecular processes in brain for intentional goal-setting, self-reflection, empathy, and episodic learning and memory.
Collapse
|
29
|
Zwir I, Arnedo J, Del-Val C, Pulkki-Råback L, Konte B, Yang SS, Romero-Zaliz R, Hintsanen M, Cloninger KM, Garcia D, Svrakic DM, Rozsa S, Martinez M, Lyytikäinen LP, Giegling I, Kähönen M, Hernandez-Cuervo H, Seppälä I, Raitoharju E, de Erausquin GA, Raitakari O, Rujescu D, Postolache TT, Sung J, Keltikangas-Järvinen L, Lehtimäki T, Cloninger CR. Uncovering the complex genetics of human temperament. Mol Psychiatry 2020; 25:2275-2294. [PMID: 30279457 PMCID: PMC7515831 DOI: 10.1038/s41380-018-0264-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 07/21/2018] [Accepted: 08/15/2018] [Indexed: 11/11/2022]
Abstract
Experimental studies of learning suggest that human temperament may depend on the molecular mechanisms for associative conditioning, which are highly conserved in animals. The main genetic pathways for associative conditioning are known in experimental animals, but have not been identified in prior genome-wide association studies (GWAS) of human temperament. We used a data-driven machine learning method for GWAS to uncover the complex genotypic-phenotypic networks and environmental interactions related to human temperament. In a discovery sample of 2149 healthy Finns, we identified sets of single-nucleotide polymorphisms (SNPs) that cluster within particular individuals (i.e., SNP sets) regardless of phenotype. Second, we identified 3 clusters of people with distinct temperament profiles measured by the Temperament and Character Inventory regardless of genotype. Third, we found 51 SNP sets that identified 736 gene loci and were significantly associated with temperament. The identified genes were enriched in pathways activated by associative conditioning in animals, including the ERK, PI3K, and PKC pathways. 74% of the identified genes were unique to a specific temperament profile. Environmental influences measured in childhood and adulthood had small but significant effects. We confirmed the replicability of the 51 Finnish SNP sets in healthy Korean (90%) and German samples (89%), as well as their associations with temperament. The identified SNPs explained nearly all the heritability expected in each sample (37-53%) despite variable cultures and environments. We conclude that human temperament is strongly influenced by more than 700 genes that modulate associative conditioning by molecular processes for synaptic plasticity and long-term memory.
Collapse
Affiliation(s)
- Igor Zwir
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA ,grid.4489.10000000121678994Department of Computer Science, University of Granada, Granada, Spain
| | - Javier Arnedo
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA ,grid.4489.10000000121678994Department of Computer Science, University of Granada, Granada, Spain
| | - Coral Del-Val
- grid.4489.10000000121678994Department of Computer Science, University of Granada, Granada, Spain
| | - Laura Pulkki-Råback
- grid.7737.40000 0004 0410 2071Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Bettina Konte
- grid.9018.00000 0001 0679 2801Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Sarah S. Yang
- grid.31501.360000 0004 0470 5905Department of Epidemiology, School of Public Health, Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Rocio Romero-Zaliz
- grid.4489.10000000121678994Department of Computer Science, University of Granada, Granada, Spain
| | - Mirka Hintsanen
- grid.10858.340000 0001 0941 4873Unit of Psychology, Faculty of Education, University of Oulu, Oulu, Finland
| | | | - Danilo Garcia
- grid.8761.80000 0000 9919 9582Department of Psychology, University of Gothenburg, Gothenburg, Sweden ,grid.435885.70000 0001 0597 1381Blekinge Centre of Competence, Blekinge County Council, Karlskrona, Sweden
| | - Dragan M. Svrakic
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
| | - Sandor Rozsa
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
| | - Maribel Martinez
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA
| | - Leo-Pekka Lyytikäinen
- grid.502801.e0000 0001 2314 6254Fimlab Laboratories, Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, Finnish Cardiovascular Research Center-Tampere, University of Tampere, Tampere, Finland
| | - Ina Giegling
- grid.9018.00000 0001 0679 2801Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany ,grid.5252.00000 0004 1936 973XUniversity Clinic, Ludwig-Maximilian University, Munich, Germany
| | - Mika Kähönen
- grid.502801.e0000 0001 2314 6254Department of Clinical Physiology, Faculty of Medicine and Life Sciences, Tampere University Hospital, University of Tampere, Tampere, Finland
| | - Helena Hernandez-Cuervo
- grid.170693.a0000 0001 2353 285XDepartment of Psychiatry and Neurosurgery, University of South Florida, Tampa, FL USA
| | - Ilkka Seppälä
- grid.502801.e0000 0001 2314 6254Fimlab Laboratories, Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, Finnish Cardiovascular Research Center-Tampere, University of Tampere, Tampere, Finland
| | - Emma Raitoharju
- grid.502801.e0000 0001 2314 6254Fimlab Laboratories, Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, Finnish Cardiovascular Research Center-Tampere, University of Tampere, Tampere, Finland
| | - Gabriel A. de Erausquin
- grid.449717.80000 0004 5374 269XDepartment of Psychiatry and Neurology, Institute of Neurosciences, University of Texas Rio-Grande Valley School of Medicine, Harlingen, TX USA
| | - Olli Raitakari
- grid.410552.70000 0004 0628 215XDepartment of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Dan Rujescu
- grid.9018.00000 0001 0679 2801Department of Psychiatry, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Teodor T. Postolache
- grid.411024.20000 0001 2175 4264Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD USA ,Rocky Mountain Mental Illness, Research, Education and Clinical Center for Veteran Suicide Prevention, Denver, CO USA
| | - Joohon Sung
- grid.31501.360000 0004 0470 5905Department of Epidemiology, School of Public Health, Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Liisa Keltikangas-Järvinen
- grid.7737.40000 0004 0410 2071Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Terho Lehtimäki
- grid.502801.e0000 0001 2314 6254Fimlab Laboratories, Department of Clinical Chemistry, Faculty of Medicine and Life Sciences, Finnish Cardiovascular Research Center-Tampere, University of Tampere, Tampere, Finland
| | - C. Robert Cloninger
- grid.4367.60000 0001 2355 7002Department of Psychiatry, Washington University School of Medicine, St. Louis, MO USA ,grid.4367.60000 0001 2355 7002Department of Psychological and Brain Sciences, School of Arts and Sciences, and Department of Genetics, School of Medicine, Washington University School of Medicine, St. Louis, MO USA
| |
Collapse
|
30
|
Zeng P, Hao X, Zhou X. Pleiotropic mapping and annotation selection in genome-wide association studies with penalized Gaussian mixture models. Bioinformatics 2019; 34:2797-2807. [PMID: 29635306 DOI: 10.1093/bioinformatics/bty204] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 04/02/2018] [Indexed: 12/11/2022] Open
Abstract
Motivation Genome-wide association studies (GWASs) have identified many genetic loci associated with complex traits. A substantial fraction of these identified loci is associated with multiple traits-a phenomena known as pleiotropy. Identification of pleiotropic associations can help characterize the genetic relationship among complex traits and can facilitate our understanding of disease etiology. Effective pleiotropic association mapping requires the development of statistical methods that can jointly model multiple traits with genome-wide single nucleic polymorphisms (SNPs) together. Results We develop a joint modeling method, which we refer to as the integrative MApping of Pleiotropic association (iMAP). iMAP models summary statistics from GWASs, uses a multivariate Gaussian distribution to account for phenotypic correlation, simultaneously infers genome-wide SNP association pattern using mixture modeling and has the potential to reveal causal relationship between traits. Importantly, iMAP integrates a large number of SNP functional annotations to substantially improve association mapping power, and, with a sparsity-inducing penalty, is capable of selecting informative annotations from a large, potentially non-informative set. To enable scalable inference of iMAP to association studies with hundreds of thousands of individuals and millions of SNPs, we develop an efficient expectation maximization algorithm based on an approximate penalized regression algorithm. With simulations and comparisons to existing methods, we illustrate the benefits of iMAP in terms of both high association mapping power and accurate estimation of genome-wide SNP association patterns. Finally, we apply iMAP to perform a joint analysis of 48 traits from 31 GWAS consortia together with 40 tissue-specific SNP annotations generated from the Roadmap Project. Availability and implementation iMAP is freely available at http://www.xzlab.org/software.html. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ping Zeng
- Department of Epidemiology and Biostatistics, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.,Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Xingjie Hao
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.,Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA.,Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
31
|
Samadi S, Mehramiz M, Kelesidis T, Mobarhan MG, Sahebkar AH, Esmaily H, Moohebati M, Farjami Z, Ferns GA, Mohammadpour AH, Avan A. High-density lipoprotein lipid peroxidation as a molecular signature of the risk for developing cardiovascular disease: Results from MASHAD cohort. J Cell Physiol 2019; 234:16168-16177. [PMID: 30784041 PMCID: PMC6699926 DOI: 10.1002/jcp.28276] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 01/17/2019] [Indexed: 01/25/2023]
Abstract
High-density lipoprotein (HDL) function rather than level may better predict cardiovascular disease (CVD). However, the contribution of the impaired antioxidant function of HDL that is associated with increased HDL lipid peroxidation (HDLox) to the development of clinical CVD remains unclear. We have investigated the association between serum HDLox with incident CVD outcomes in Mashhad cohort. Three-hundred and thirty individuals who had a median follow-up period of 7 years were recruited as part of the cohort. The primary end point was cardiovascular event, including myocardial infarction, stable angina, unstable angina, or coronary revascularization. In both univariate/multivariate analyses adjusted for traditional CVD risk factors, HDLox was an independent risk factor for CVD (odds ratio, 1.62; 95% confidence interval, 1.41-1.86; p < 0.001). For every increase in HDLox by 0.1 unit, there was an increase in CVD risk by 1.62-fold. In an adjusted analysis, there was a >2.5-fold increase in cardiovascular risk in individuals with HDLox higher than cutoff point of 1.06 compared to those with lower scores, suggesting HDLox > 1.06 is related to the impaired HDL oxidant function and in turn exposed to elevated risk of CVD outcomes (hazard ratio, 2.72; 95% CI, 1.88-3.94). Higher HDLox is a surrogate measure of reduced HDL antioxidant function that positively associated with cardiovascular events in a population-based cohort.
Collapse
Affiliation(s)
- Sara Samadi
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrane Mehramiz
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Majid Ghayour Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hosein Sahebkar
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Habibollah Esmaily
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Moohebati
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Farjami
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A. Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Sussex, UK
| | - Amir hooshang Mohammadpour
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
32
|
Physical Exercise Affects Adipose Tissue Profile and Prevents Arterial Thrombosis in BDNF Val66Met Mice. Cells 2019; 8:cells8080875. [PMID: 31405230 PMCID: PMC6721716 DOI: 10.3390/cells8080875] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/02/2019] [Accepted: 08/10/2019] [Indexed: 01/04/2023] Open
Abstract
Adipose tissue accumulation is an independent and modifiable risk factor for cardiovascular disease (CVD). The recent CVD European Guidelines strongly recommend regular physical exercise (PE) as a management strategy for prevention and treatment of CVD associated with metabolic disorders and obesity. Although mutations as well as common genetic variants, including the brain-derived neurotrophic factor (BDNF) Val66Met polymorphism, are associated with increased body weight, eating and neuropsychiatric disorders, and myocardial infarction, the effect of this polymorphism on adipose tissue accumulation and regulation as well as its relation to obesity/thrombosis remains to be elucidated. Here, we showed that white adipose tissue (WAT) of humanized knock-in BDNFVal66Met (BDNFMet/Met) mice is characterized by an altered morphology and an enhanced inflammatory profile compared to wild-type BDNFVal/Val. Four weeks of voluntary PE restored the adipocyte size distribution, counteracted the inflammatory profile of adipose tissue, and prevented the prothrombotic phenotype displayed, per se, by BDNFMet/Met mice. C3H10T1/2 cells treated with the Pro-BDNFMet peptide well recapitulated the gene alterations observed in BDNFMet/Met WAT mice. In conclusion, these data indicate the strong impact of lifestyle, in particular of the beneficial effect of PE, on the management of arterial thrombosis and inflammation associated with obesity in relation to the specific BDNF Val66Met mutation.
Collapse
|
33
|
Blauw LL, Li-Gao R, Noordam R, de Mutsert R, Trompet S, Berbée JFP, Wang Y, van Klinken JB, Christen T, van Heemst D, Mook-Kanamori DO, Rosendaal FR, Jukema JW, Rensen PCN, Willems van Dijk K. CETP (Cholesteryl Ester Transfer Protein) Concentration: A Genome-Wide Association Study Followed by Mendelian Randomization on Coronary Artery Disease. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002034. [PMID: 29728394 DOI: 10.1161/circgen.117.002034] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/26/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND We aimed to identify independent genetic determinants of circulating CETP (cholesteryl ester transfer protein) to assess causal effects of variation in CETP concentration on circulating lipid concentrations and cardiovascular disease risk. METHODS A genome-wide association discovery and replication study on serum CETP concentration were embedded in the NEO study (Netherlands Epidemiology of Obesity). Based on the independent identified variants, Mendelian randomization was conducted on serum lipids (NEO study) and coronary artery disease (CAD; CARDIoGRAMplusC4D consortium). RESULTS In the discovery analysis (n=4248), we identified 3 independent variants (P<5×10-8) that determine CETP concentration. These single-nucleotide polymorphisms were mapped to CETP and replicated in a separate subpopulation (n=1458). Per-allele increase (SE) in serum CETP was 0.32 (0.02) µg/mL for rs247616-C, 0.35 (0.02) µg/mL for rs12720922-A, and 0.12 (0.02) µg/mL for rs1968905-G. Combined, these 3 variants explained 16.4% of the total variation in CETP concentration. One microgram per milliliter increase in genetically determined CETP concentration strongly decreased high-density lipoprotein cholesterol (-0.23 mmol/L; 95% confidence interval, -0.26 to -0.20), moderately increased low-density lipoprotein cholesterol (0.08 mmol/L; 95% confidence interval, 0.00-0.16), and was associated with an odds ratio of 1.08 (95% confidence interval, 0.94-1.23) for CAD risk. CONCLUSIONS This is the first genome-wide association study identifying independent variants that largely determine CETP concentration. Although high-density lipoprotein cholesterol is not a causal risk factor for CAD, it has been unequivocally demonstrated that low-density lipoprotein cholesterol lowering is proportionally associated with a lower CAD risk. Therefore, the results of our study are fully consistent with the notion that CETP concentration is causally associated with CAD through low-density lipoprotein cholesterol.
Collapse
Affiliation(s)
- Lisanne L Blauw
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.) .,Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Raymond Noordam
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.)
| | - Renée de Mutsert
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Stella Trompet
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.).,Department of Cardiology (S.T., J.W.J.)
| | - Jimmy F P Berbée
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Yanan Wang
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Jan B van Klinken
- Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.).,Department of Human Genetics (J.B.v.K., K.W.v.D.)
| | - Tim Christen
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | - Diana van Heemst
- Department of Internal Medicine, Division of Gerontology and Geriatrics (R.N., S.T., D.v.H.)
| | - Dennis O Mook-Kanamori
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.).,and Department of Public Health and Primary Care (D.O.M.-K.) Leiden University Medical Center, The Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology (L.L.B., R.L.-G., R.d.M., T.C., D.O.M.-K., F.R.R.)
| | | | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.)
| | - Ko Willems van Dijk
- Department of Internal Medicine, Division of Endocrinology (L.L.B., J.F.P.B., Y.W., P.C.N.R., K.W.v.D.).,Einthoven Laboratory for Experimental Vascular Medicine (L.L.B., J.F.P.B., Y.W., J.B.v.K., P.C.N.R., K.W.v.D.).,Department of Human Genetics (J.B.v.K., K.W.v.D.)
| |
Collapse
|
34
|
Kolifarhood G, Daneshpour MS, Khayat BS, Saadati HM, Guity K, Khosravi N, Akbarzadeh M, Sabour S. Generality of genomic findings on blood pressure traits and its usefulness in precision medicine in diverse populations: A systematic review. Clin Genet 2019; 96:17-27. [PMID: 30820929 DOI: 10.1111/cge.13527] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 01/01/2023]
Abstract
Remarkable findings from genome-wide association studies (GWAS) on blood pressure (BP) traits have made new insights for developing precision medicine toward more effective screening measures. However, generality of GWAS findings in diverse populations is hampered by some technical limitations. There is no comprehensive study to evaluate source(s) of the non-generality of GWAS results on BP traits, so to fill the gap, this systematic review study was carried out. Using MeSH terms, 1545 records were detected through searching in five databases and 49 relevant full-text articles were included in our review. Overall, 749 unique variants were reported, of those, majority of variants have been detected in Europeans and were associated to systolic and diastolic BP traits. Frequency of genetic variants with same position was low in European and non-European populations (n = 38). However, more than 200 (>25%) single nucleotide polymorphisms were found on same loci or linkage disequilibrium blocks (r2 ≥ 80%). Investigating for locus position and linkage disequilibrium of infrequent unique variants showed modest to high reproducibility of findings in Europeans that in some extent was generalizable in other populations. Beyond theoretical limitations, our study addressed other possible sources of non-generality of GWAS findings for BP traits in the same and different origins.
Collapse
Affiliation(s)
- Goodarz Kolifarhood
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam S Daneshpour
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh S Khayat
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein M Saadati
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Guity
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasim Khosravi
- Department of Community Health Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran, Iran
| | - Mahdi Akbarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Siamak Sabour
- Department of Epidemiology, School of Public Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Safety Promotion and Injury Prevention Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Lea A, Subramaniam M, Ko A, Lehtimäki T, Raitoharju E, Kähönen M, Seppälä I, Mononen N, Raitakari OT, Ala-Korpela M, Pajukanta P, Zaitlen N, Ayroles JF. Genetic and environmental perturbations lead to regulatory decoherence. eLife 2019; 8:e40538. [PMID: 30834892 PMCID: PMC6400502 DOI: 10.7554/elife.40538] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 02/14/2019] [Indexed: 01/24/2023] Open
Abstract
Correlation among traits is a fundamental feature of biological systems that remains difficult to study. To address this problem, we developed a flexible approach that allows us to identify factors associated with inter-individual variation in correlation. We use data from three human cohorts to study the effects of genetic and environmental variation on correlations among mRNA transcripts and among NMR metabolites. We first show that environmental exposures (infection and disease) lead to a systematic loss of correlation, which we define as 'decoherence'. Using longitudinal data, we show that decoherent metabolites are better predictors of whether someone will develop metabolic syndrome than metabolites commonly used as biomarkers of this disease. Finally, we demonstrate that correlation itself is under genetic control by mapping hundreds of 'correlation quantitative trait loci (QTLs)'. Together, this work furthers our understanding of how and why coordinated biological processes break down, and points to a potential role for decoherence in disease. Editorial note This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Amanda Lea
- Department of Ecology and EvolutionPrinceton UniversityPrincetonUnited States
- Lewis-Sigler Institute for Integrative GenomicsPrinceton UniversityPrincetonUnited States
| | - Meena Subramaniam
- Department of Medicine, Lung Biology CenterUniversity of California, San FranciscoSan FranciscoUnited States
| | - Arthur Ko
- Department of Medicine, David Geffen School of Medicine at UCLAUniversity of California, Los AngelesLos AngelesUnited States
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Emma Raitoharju
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Mika Kähönen
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Department of Clinical PhysiologyTampere University, Tampere University HospitalTampereFinland
| | - Ilkka Seppälä
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Nina Mononen
- Finnish Cardiovascular Research Center, Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular MedicineUniversity of TurkuTurkuFinland
- Department of Clinical Physiology and Nuclear MedicineTurku University HospitalTurkuFinland
| | - Mika Ala-Korpela
- Systems Epidemiology, Baker Heart and Diabetes InstituteMelbourneAustralia
- Computational Medicine, Faculty of Medicine, Biocenter OuluUniversity of OuluOuluFinland
- NMR Metabolomics Laboratory, School of PharmacyUniversity of Eastern FinlandKuopioFinland
- Population Health Science, Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUnited Kingdom
- Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Faculty of Medicine, Nursing and Health SciencesThe Alfred Hospital, Monash UniversityMelbourneAustralia
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLAUniversity of California, Los AngelesLos AngelesUnited States
| | - Noah Zaitlen
- Department of Medicine, Lung Biology CenterUniversity of California, San FranciscoSan FranciscoUnited States
| | - Julien F Ayroles
- Department of Ecology and EvolutionPrinceton UniversityPrincetonUnited States
- Lewis-Sigler Institute for Integrative GenomicsPrinceton UniversityPrincetonUnited States
| |
Collapse
|
36
|
Dobewall H, Savelieva K, Seppälä I, Knafo-Noam A, Hakulinen C, Elovainio M, Keltikangas-Järvinen L, Pulkki-Råback L, Raitakari OT, Lehtimäki T, Hintsanen M. Gene-environment correlations in parental emotional warmth and intolerance: genome-wide analysis over two generations of the Young Finns Study. J Child Psychol Psychiatry 2019; 60:277-285. [PMID: 30357825 DOI: 10.1111/jcpp.12995] [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] [Accepted: 09/18/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Genomic analysis of the child might offer new potential to illuminate human parenting. We examined whether offspring (G2) genome-wide genotype variation (SNPs) is associated with their mother's (G1) emotional warmth and intolerance, indicating a gene-environment correlation. If this association is stronger than between G2's genes and their emotional warmth and intolerance toward their own children, then this would indicate the presence of an evocative gene-environment correlation. To further understand how G1 mother's parenting has been evoked by genetically influenced characteristics of the child (G2), we examined whether child (G2) temperament partially accounted for the association between offspring genes and parental responses. METHODS Participants were from the Young Finns Study. G1 mothers (N = 2,349; mean age 39 years) self-reported the emotional warmth and intolerance toward G2 in 1980 when the participants were from 3 to 18 years old. G2 participants answered the same parenting scales in 2007/2012 (N = 1,378; mean age = 38 years in 2007; 59% female) when their children were on average 11 years old. Offspring temperament traits were self-reported in 1992 (G2 age range 15-30 years). Estimation of the phenotypic variance explained by the SNPs of G2 was done by genome-wide complex trait analysis with restricted maximum likelihood (GCTA-GREML). RESULTS Results showed that the SNPs of a child (G2) explained 22.6% of the phenotypic variance of maternal intolerance (G1; p-value = .039). G2 temperament trait negative emotionality explained only 2.4% points of this association. G2 genes did not explain G1 emotional warmth or G2's own emotional warmth and intolerance. However, further analyses of a combined measure of both G1 parenting scales found genetic effects. Parent or child gender did not moderate the observed associations. CONCLUSIONS Presented genome-wide evidence is pointing to the important role a child plays in affecting and shaping his/her family environment, though the underlying mechanisms remain unclear.
Collapse
Affiliation(s)
- Henrik Dobewall
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Faculty of Social Sciences, Health Sciences, University of Tampere, Tampere, Finland
| | - Kateryna Savelieva
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Ariel Knafo-Noam
- Department of Psychology, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Christian Hakulinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marko Elovainio
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Laura Pulkki-Råback
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Olli T Raitakari
- Research Collegium for Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | | |
Collapse
|
37
|
Zhao X, Geng X, Srinivasasainagendra V, Chaudhary N, Judd S, Wadley V, Gutiérrez OM, Wang H, Lange EM, Lange LA, Woo D, Unverzagt FW, Safford M, Cushman M, Limdi N, Quarells R, Arnett DK, Irvin MR, Zhi D. A PheWAS study of a large observational epidemiological cohort of African Americans from the REGARDS study. BMC Med Genomics 2019; 12:26. [PMID: 30704471 PMCID: PMC6357353 DOI: 10.1186/s12920-018-0462-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Cardiovascular disease, diabetes, and kidney disease are among the leading causes of death and disability worldwide. However, knowledge of genetic determinants of those diseases in African Americans remains limited. RESULTS In our study, associations between 4956 GWAS catalog reported SNPs and 67 traits were examined among 7726 African Americans from the REasons for Geographic and Racial Differences in Stroke (REGARDS) study, which is focused on identifying factors that increase stroke risk. The prevalent and incident phenotypes studied included inflammation, kidney traits, cardiovascular traits and cognition. Our results validated 29 known associations, of which eight associations were reported for the first time in African Americans. CONCLUSION Our cross-racial validation of GWAS findings provide additional evidence for the important roles of these loci in the disease process and may help identify genes especially important for future functional validation.
Collapse
Affiliation(s)
- Xueyan Zhao
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Xin Geng
- BGI-Shenzhen, Shenzhen, 518083 China
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
| | | | - Ninad Chaudhary
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Suzanne Judd
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Virginia Wadley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Orlando M. Gutiérrez
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233 USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Henry Wang
- Department of Emergency Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Ethan M. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045 USA
| | - Daniel Woo
- Department of Neurology and Rehabilitation Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Frederick W. Unverzagt
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Monika Safford
- Division of General Internal Medicine, Weill Cornell Medical College, Cornell University, New York, NY 10065 USA
| | - Mary Cushman
- Department of Medicine and Pathology, Larner College of Medicine at the University of Vermont, Burlington, VT 05405 USA
| | - Nita Limdi
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Rakale Quarells
- Cardiovascular Research Institute, Department of Community Health and Preventive Medicine, Morehouse School of Medicine, Atlanta, GA 30310 USA
| | - Donna K. Arnett
- College of Public Health, University of Kentucky, Lexington, KY 40506 USA
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Degui Zhi
- School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
| |
Collapse
|
38
|
Pikó P, Fiatal S, Kósa Z, Sándor J, Ádány R. Generalizability and applicability of results obtained from populations of European descent regarding the effect direction and size of HDL-C level-associated genetic variants to the Hungarian general and Roma populations. Gene 2018; 686:187-193. [PMID: 30468910 DOI: 10.1016/j.gene.2018.11.067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/28/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Large-scale association studies that mainly involve European populations identified many genetic loci related to high-density lipoprotein cholesterol (HDL-C) levels, one of the most important indicators of the risk for cardiovascular diseases. The question with intense speculation of whether the effect estimates obtained from European populations for different HDL-C level-related SNPs are applicable to the Roma ethnicity, the largest minority group in Europe with a South Asian origin, was addressed in the present study. DESIGN The associations between 21 SNPs (in the genes LIPC(G), CETP, GALNT2, HMGCP, ABCA1, KCTD10 and WWOX) and HDL-C levels were examined separately in adults of the Hungarian general (N = 1542) and Roma (N = 646) populations by linear regression. Individual effects (direction and size) of single SNPs on HDL-C levels were computed and compared between the study groups and with data published in the literature. RESULTS Significant associations between SNPs and HDL-C levels were more frequently found in general subjects than in Roma subjects (11 SNPs in general vs. 4 SNPs in Roma). The CETP gene variants rs1532624, rs708272 and rs7499892 consistently showed significant associations with HDL-C levels across the study groups (p ˂ 0.05), indicating a possible causal variant(s) in this region. Although nominally significant differences in effect size were found for three SNPs (rs693 in gene APOB, rs9989419 in gene CETP, and rs2548861 in gene WWOX) by comparing the general and Roma populations, most of these SNPs did not have a significant effect on HDL-C levels. The β coefficients for SNPs in the Roma population were found to be identical both in direction and magnitude to the effect obtained previously in large-scale studies on European populations. CONCLUSIONS The effect of the vast majority of the SNPs on HDL-C levels could be replicated in the Hungarian general and Roma populations, which indicates that the effect size measurements obtained from the literature can be used for risk estimation for both populations.
Collapse
Affiliation(s)
- Péter Pikó
- MTA-DE Public Health Research Group of the Hungarian Academy of Sciences, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary
| | - Szilvia Fiatal
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; WHO Collaborating Centre on Vulnerability and Health, Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary
| | - Zsigmond Kósa
- Department of Health Visitor Methodology and Public Health, Faculty of Health, University of Debrecen, Nyíregyháza 4400, Hungary
| | - János Sándor
- Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; WHO Collaborating Centre on Vulnerability and Health, Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary
| | - Róza Ádány
- MTA-DE Public Health Research Group of the Hungarian Academy of Sciences, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary; WHO Collaborating Centre on Vulnerability and Health, Department of Preventive Medicine, Faculty of Public Health, University of Debrecen, Debrecen 4028, Hungary.
| |
Collapse
|
39
|
Dobewall H, Hakulinen C, Keltikangas-Järvinen L, Pulkki-Råback L, Seppälä I, Lehtimäki T, Raitakari OT, Hintsanen M. Oxytocin receptor gene (OXTR) variant rs1042778 moderates the influence of family environment on changes in perceived social support over time. J Affect Disord 2018; 235:480-488. [PMID: 29680729 DOI: 10.1016/j.jad.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 03/02/2018] [Accepted: 04/02/2018] [Indexed: 11/25/2022]
Abstract
BACKGROUND Lack of social support is an established risk factor across health outcomes, making it important to examine its family environmental and genetic determinants. METHODS In a 27-year follow-up of the Young Finns Study (N = 2341), we examined with a latent growth curve model whether genes involved in the oxytocin signaling pathway-namely, oxytocin receptor gene (OXTR) variants rs1042778, rs2254298, and rs53576-moderate the effect of early-life social experiences on perceived social support across the life span. Mothers reported the emotional warmth and acceptance towards their children at baseline when the participants were from 3 to 18 years old (1980). Perceived family support and support from friends and peripheral sources were assessed in five follow-ups 18 years apart (1989-2007). RESULTS Maternal emotional warmth and acceptance predicted the initial level of perceived social support across subscales, while the rate of change in family support was affected by the family environment only if participants carried the T-allele of OXTR rs1042778. This gene-environment interaction was not found for the rate of change in support from friends and peripheral sources and we also did not find associations between latent growth in perceived social support and OXTR variants rs53576 and rs2254298. LIMITATIONS Selective attrition in perceived social support, maternal emotional warmth and acceptance, gender, and SES. Family environment was assessed by a non-standardized measure. CONCLUSIONS OXTR rs1042778 polymorphism seems to contribute to changes in perceived family support in that way that some individuals (T-allele carriers) 'recover', to some extent, from the effects of early-life social experiences, whereas others (G/G genotype carriers) do not.
Collapse
Affiliation(s)
- Henrik Dobewall
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Faculty of Social Sciences, Health Sciences, University of Tampere, Tampere, Finland
| | - Christian Hakulinen
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | - Laura Pulkki-Råback
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, Finland
| | - Olli T Raitakari
- Research Collegium for Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Mirka Hintsanen
- Research Unit of Psychology, University of Oulu, Oulu, Finland; Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
40
|
Ng FL, Warren HR, Caulfield MJ. Hypertension genomics and cardiovascular prevention. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:291. [PMID: 30211179 DOI: 10.21037/atm.2018.06.34] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Hypertension continues to be a major risk factor for global mortality, and recent genome-wide association studies (GWAS) have expanded in size, leading to the identification of further genetic loci influencing blood pressure. In light of the new knowledge from the largest cardiovascular GWAS to date, we review the potential impact of genomics on discovering potential drug targets, risk stratification with genetic risk scores, drug selection with pharmacogenetics, and exploring insights provided by gene-environment interactions.
Collapse
Affiliation(s)
- Fu Liang Ng
- William Harvey Research Institute, The NIHR Biomedical Research Centre at Barts, Queen Mary University London, London, UK.,Barts BP Centre of Excellence, Barts Heart Centre, The NIHR Biomedical Research Centre at Barts, St Bartholomew's Hospital, W Smithfield, London, UK
| | - Helen R Warren
- William Harvey Research Institute, The NIHR Biomedical Research Centre at Barts, Queen Mary University London, London, UK
| | - Mark J Caulfield
- William Harvey Research Institute, The NIHR Biomedical Research Centre at Barts, Queen Mary University London, London, UK.,Barts BP Centre of Excellence, Barts Heart Centre, The NIHR Biomedical Research Centre at Barts, St Bartholomew's Hospital, W Smithfield, London, UK
| |
Collapse
|
41
|
Yashin AI, Fang F, Kovtun M, Wu D, Duan M, Arbeev K, Akushevich I, Kulminski A, Culminskaya I, Zhbannikov I, Yashkin A, Stallard E, Ukraintseva S. Hidden heterogeneity in Alzheimer's disease: Insights from genetic association studies and other analyses. Exp Gerontol 2018; 107:148-160. [PMID: 29107063 PMCID: PMC5920782 DOI: 10.1016/j.exger.2017.10.020] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/20/2017] [Accepted: 10/22/2017] [Indexed: 02/08/2023]
Abstract
Despite evident success in clarifying many important features of Alzheimer's disease (AD) the efficient methods of its prevention and treatment are not yet available. The reasons are likely to be the fact that AD is a multifactorial and heterogeneous health disorder with multiple alternative pathways of disease development and progression. The availability of genetic data on individuals participated in longitudinal studies of aging health and longevity, as well as on participants of cross-sectional case-control studies allow for investigating genetic and non-genetic connections with AD and to link the results of these analyses with research findings obtained in clinical, experimental, and molecular biological studies of this health disorder. The objective of this paper is to perform GWAS of AD in several study populations and investigate possible roles of detected genetic factors in developing AD hallmarks and in other health disorders. The data collected in the Framingham Heart Study (FHS), Cardiovascular Health Study (CHS), Health and Retirement Study (HRS) and Late Onset Alzheimer's Disease Family Study (LOADFS) were used in these analyses. The logistic regression and Cox's regression were used as statistical models in GWAS. The results of analyses confirmed strong associations of genetic variants from well-known genes APOE, TOMM40, PVRL2 (NECTIN2), and APOC1 with AD. Possible roles of these genes in pathological mechanisms resulting in development of hallmarks of AD are described. Many genes whose connection with AD was detected in other studies showed nominally significant associations with this health disorder in our study. The evidence on genetic connections between AD and vulnerability to infection, as well as between AD and other health disorders, such as cancer and type 2 diabetes, were investigated. The progress in uncovering hidden heterogeneity in AD would be substantially facilitated if common mechanisms involved in development of AD, its hallmarks, and AD related chronic conditions were investigated in their mutual connection.
Collapse
Affiliation(s)
- Anatoliy I Yashin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA.
| | - Fang Fang
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Mikhail Kovtun
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Deqing Wu
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Matt Duan
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Konstantin Arbeev
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Igor Akushevich
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Alexander Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Ilya Zhbannikov
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Arseniy Yashkin
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Eric Stallard
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA
| | - Svetlana Ukraintseva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, 2024 W. Main Street, Durham, NC 27705, USA.
| |
Collapse
|
42
|
Shatwan IM, Winther KH, Ellahi B, Elwood P, Ben-Shlomo Y, Givens I, Rayman MP, Lovegrove JA, Vimaleswaran KS. Association of apolipoprotein E gene polymorphisms with blood lipids and their interaction with dietary factors. Lipids Health Dis 2018; 17:98. [PMID: 29712557 PMCID: PMC5928585 DOI: 10.1186/s12944-018-0744-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 04/13/2018] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Several candidate genes have been identified in relation to lipid metabolism, and among these, lipoprotein lipase (LPL) and apolipoprotein E (APOE) gene polymorphisms are major sources of genetically determined variation in lipid concentrations. This study investigated the association of two single nucleotide polymorphisms (SNPs) at LPL, seven tagging SNPs at the APOE gene, and a common APOE haplotype (two SNPs) with blood lipids, and examined the interaction of these SNPs with dietary factors. METHODS The population studied for this investigation included 660 individuals from the Prevention of Cancer by Intervention with Selenium (PRECISE) study who supplied baseline data. The findings of the PRECISE study were further replicated using 1238 individuals from the Caerphilly Prospective cohort (CaPS). Dietary intake was assessed using a validated food-frequency questionnaire (FFQ) in PRECISE and a validated semi-quantitative FFQ in the CaPS. Interaction analyses were performed by including the interaction term in the linear regression model adjusted for age, body mass index, sex and country. RESULTS There was no association between dietary factors and blood lipids after Bonferroni correction and adjustment for confounding factors in either cohort. In the PRECISE study, after correction for multiple testing, there was a statistically significant association of the APOE haplotype (rs7412 and rs429358; E2, E3, and E4) and APOE tagSNP rs445925 with total cholesterol (P = 4 × 10- 4 and P = 0.003, respectively). Carriers of the E2 allele had lower total cholesterol concentration (5.54 ± 0.97 mmol/L) than those with the E3 (5.98 ± 1.05 mmol/L) (P = 0.001) and E4 (6.09 ± 1.06 mmol/L) (P = 2 × 10- 4) alleles. The association of APOE haplotype (E2, E3, and E4) and APOE SNP rs445925 with total cholesterol (P = 2 × 10- 6 and P = 3 × 10- 4, respectively) was further replicated in the CaPS. Additionally, significant association was found between APOE haplotype and APOE SNP rs445925 with low density lipoprotein cholesterol in CaPS (P = 4 × 10- 4 and P = 0.001, respectively). After Bonferroni correction, none of the cohorts showed a statistically significant SNP-diet interaction on lipid outcomes. CONCLUSION In summary, our findings from the two cohorts confirm that genetic variations at the APOE locus influence plasma total cholesterol concentrations, however, the gene-diet interactions on lipids require further investigation in larger cohorts.
Collapse
Affiliation(s)
- Israa M Shatwan
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research (ICMR), Department of Food and Nutritional Sciences, University of Reading, Whiteknights, PO Box 226, Reading, RG6 6AP, UK.,Food and Nutrition Department, Faculty of Home Economics, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Basma Ellahi
- Faculty of Health and Social Care, University of Chester, Chester, CH1 1SL, UK
| | - Peter Elwood
- Department of Epidemiology, Statistics and Public Health, Cardiff University, University Hospital of Wales, Heath Park, Cardiff, CF14 4XW, UK
| | - Yoav Ben-Shlomo
- Population Health Sciences, University of Bristol, Bristol, BS8 2PS, UK
| | - Ian Givens
- Institute for Food, Nutrition and Health, University of Reading, Earley Gate, Reading, RG6 6AR, UK
| | - Margaret P Rayman
- Department of Nutritional Sciences Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research (ICMR), Department of Food and Nutritional Sciences, University of Reading, Whiteknights, PO Box 226, Reading, RG6 6AP, UK
| | - Karani S Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research (ICMR), Department of Food and Nutritional Sciences, University of Reading, Whiteknights, PO Box 226, Reading, RG6 6AP, UK.
| |
Collapse
|
43
|
Staley JR, Suderman M, Simpkin AJ, Gaunt TR, Heron J, Relton CL, Tilling K. Longitudinal analysis strategies for modelling epigenetic trajectories. Int J Epidemiol 2018; 47:516-525. [PMID: 29462323 PMCID: PMC5913606 DOI: 10.1093/ije/dyy012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/22/2017] [Accepted: 01/24/2018] [Indexed: 12/20/2022] Open
Abstract
Background DNA methylation levels are known to vary over time, and modelling these trajectories is crucial for our understanding of the biological relevance of these changes over time. However, due to the computational cost of fitting multilevel models across the epigenome, most trajectory modelling efforts to date have focused on a subset of CpG sites identified through epigenome-wide association studies (EWAS) at individual time-points. Methods We propose using linear regression across the repeated measures, estimating cluster-robust standard errors using a sandwich estimator, as a less computationally intensive strategy than multilevel modelling. We compared these two longitudinal approaches, as well as three approaches based on EWAS (associated at baseline, at any time-point and at all time-points), for identifying epigenetic change over time related to an exposure using simulations and by applying them to blood DNA methylation profiles from the Accessible Resource for Integrated Epigenomics Studies (ARIES). Results Restricting association testing to EWAS at baseline identified a less complete set of associations than performing EWAS at each time-point or applying the longitudinal modelling approaches to the full dataset. Linear regression models with cluster-robust standard errors identified similar sets of associations with almost identical estimates of effect as the multilevel models, while also being 74 times more efficient. Both longitudinal modelling approaches identified comparable sets of CpG sites in ARIES with an association with prenatal exposure to smoking (>70% agreement). Conclusions Linear regression with cluster-robust standard errors is an appropriate and efficient approach for longitudinal analysis of DNA methylation data.
Collapse
Affiliation(s)
- James R Staley
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Matthew Suderman
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Andrew J Simpkin
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Tom R Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Jon Heron
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| |
Collapse
|
44
|
Dobewall H, Hakulinen C, Pulkki-Råback L, Seppälä I, Lehtimäki T, Raitakari OT, Keltikangas-Järvinen L, Hintsanen M. The role of oxytocin receptor gene (OXTR) and mother's emotional warmth in predicting adulthood sociability. PERSONALITY AND INDIVIDUAL DIFFERENCES 2018. [DOI: 10.1016/j.paid.2017.12.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
45
|
Subkhangulova A, Malik AR, Hermey G, Popp O, Dittmar G, Rathjen T, Poy MN, Stumpf A, Beed PS, Schmitz D, Breiderhoff T, Willnow TE. SORCS1 and SORCS3 control energy balance and orexigenic peptide production. EMBO Rep 2018; 19:embr.201744810. [PMID: 29440124 PMCID: PMC5891432 DOI: 10.15252/embr.201744810] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 12/20/2022] Open
Abstract
SORCS1 and SORCS3 are two related sorting receptors expressed in neurons of the arcuate nucleus of the hypothalamus. Using mouse models with individual or dual receptor deficiencies, we document a previously unknown function of these receptors in central control of metabolism. Specifically, SORCS1 and SORCS3 act as intracellular trafficking receptors for tropomyosin-related kinase B to attenuate signaling by brain-derived neurotrophic factor, a potent regulator of energy homeostasis. Loss of the joint action of SORCS1 and SORCS3 in mutant mice results in excessive production of the orexigenic neuropeptide agouti-related peptide and in a state of chronic energy excess characterized by enhanced food intake, decreased locomotor activity, diminished usage of lipids as metabolic fuel, and increased adiposity, albeit at overall reduced body weight. Our findings highlight a novel concept in regulation of the melanocortin system and the role played by trafficking receptors SORCS1 and SORCS3 in this process.
Collapse
Affiliation(s)
| | - Anna R Malik
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Popp
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Gunnar Dittmar
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Thomas Rathjen
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Matthew N Poy
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Alexander Stumpf
- Neuroscience Research Center, Charité - University Medicine, Berlin, Germany
| | - Prateep Sanker Beed
- Neuroscience Research Center, Charité - University Medicine, Berlin, Germany
| | - Dietmar Schmitz
- Neuroscience Research Center, Charité - University Medicine, Berlin, Germany
| | | | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany .,Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
46
|
Seyednasrollah F, Mäkelä J, Pitkänen N, Juonala M, Hutri-Kähönen N, Lehtimäki T, Viikari J, Kelly T, Li C, Bazzano L, Elo LL, Raitakari OT. Prediction of Adulthood Obesity Using Genetic and Childhood Clinical Risk Factors in the Cardiovascular Risk in Young Finns Study. ACTA ACUST UNITED AC 2018; 10:CIRCGENETICS.116.001554. [PMID: 28620069 DOI: 10.1161/circgenetics.116.001554] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Obesity is a known risk factor for cardiovascular disease. Early prediction of obesity is essential for prevention. The aim of this study is to assess the use of childhood clinical factors and the genetic risk factors in predicting adulthood obesity using machine learning methods. METHODS AND RESULTS A total of 2262 participants from the Cardiovascular Risk in YFS (Young Finns Study) were followed up from childhood (age 3-18 years) to adulthood for 31 years. The data were divided into training (n=1625) and validation (n=637) set. The effect of known genetic risk factors (97 single-nucleotide polymorphisms) was investigated as a weighted genetic risk score of all 97 single-nucleotide polymorphisms (WGRS97) or a subset of 19 most significant single-nucleotide polymorphisms (WGRS19) using boosting machine learning technique. WGRS97 and WGRS19 were validated using external data (n=369) from BHS (Bogalusa Heart Study). WGRS19 improved the accuracy of predicting adulthood obesity in training (area under the curve [AUC=0.787 versus AUC=0.744, P<0.0001) and validation data (AUC=0.769 versus AUC=0.747, P=0.026). WGRS97 improved the accuracy in training (AUC=0.782 versus AUC=0.744, P<0.0001) but not in validation data (AUC=0.749 versus AUC=0.747, P=0.785). Higher WGRS19 associated with higher body mass index at 9 years and WGRS97 at 6 years. Replication in BHS confirmed our findings that WGRS19 and WGRS97 are associated with body mass index. CONCLUSIONS WGRS19 improves prediction of adulthood obesity. Predictive accuracy is highest among young children (3-6 years), whereas among older children (9-18 years) the risk can be identified using childhood clinical factors. The model is helpful in screening children with high risk of developing obesity.
Collapse
Affiliation(s)
- Fatemeh Seyednasrollah
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Johanna Mäkelä
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.).
| | - Niina Pitkänen
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Markus Juonala
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Nina Hutri-Kähönen
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Terho Lehtimäki
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Jorma Viikari
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Tanika Kelly
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Changwei Li
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Lydia Bazzano
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Laura L Elo
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| | - Olli T Raitakari
- From the Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland (F.S., J.M., L.L.E.); Department of Mathematics and Statistics (F.S.), Research Centre of Applied and Preventive Cardiovascular Medicine (N.P., O.T.R.), and Department of Medicine (M.J., J.V.), University of Turku, Finland; Division of Medicine (M.J., J.V.) and Clinical Physiology and Nuclear Medicine (O.T.R.), Turku University Hospital, Finland; Department of Pediatrics (N.H.-K.) and School of Medicine (T.L.), University of Tampere, Finland; Tampere University Hospital, Finland (N.H.-K.); Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland (T.L.); Tulane University Health Sciences Center, New Orleans, LA (T.K., L.B.); and Department of Epidemiology and Biostatistics, College of Public Health, University of Georgia, Athens (C.L.)
| |
Collapse
|
47
|
Haeusler RA, McGraw TE, Accili D. Biochemical and cellular properties of insulin receptor signalling. Nat Rev Mol Cell Biol 2018; 19:31-44. [PMID: 28974775 PMCID: PMC5894887 DOI: 10.1038/nrm.2017.89] [Citation(s) in RCA: 470] [Impact Index Per Article: 67.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The mechanism of insulin action is a central theme in biology and medicine. In addition to the rather rare condition of insulin deficiency caused by autoimmune destruction of pancreatic β-cells, genetic and acquired abnormalities of insulin action underlie the far more common conditions of type 2 diabetes, obesity and insulin resistance. The latter predisposes to diseases ranging from hypertension to Alzheimer disease and cancer. Hence, understanding the biochemical and cellular properties of insulin receptor signalling is arguably a priority in biomedical research. In the past decade, major progress has led to the delineation of mechanisms of glucose transport, lipid synthesis, storage and mobilization. In addition to direct effects of insulin on signalling kinases and metabolic enzymes, the discovery of mechanisms of insulin-regulated gene transcription has led to a reassessment of the general principles of insulin action. These advances will accelerate the discovery of new treatment modalities for diabetes.
Collapse
Affiliation(s)
- Rebecca A Haeusler
- Columbia University College of Physicians and Surgeons, Department of Pathology and Cell Biology, New York, New York 10032, USA
| | - Timothy E McGraw
- Weill Cornell Medicine, Departments of Biochemistry and Cardiothoracic Surgery, New York, New York 10065, USA
| | - Domenico Accili
- Columbia University College of Physicians & Surgeons, Department of Medicine, New York, New York 10032, USA
| |
Collapse
|
48
|
Genomic dissection and prediction of feed intake and residual feed intake traits using a longitudinal model in F2 chickens. Animal 2017; 12:1792-1798. [PMID: 29268803 DOI: 10.1017/s1751731117003354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Feed efficiency traits (FETs) are important economic indicators in poultry production. Because feed intake (FI) is a time-dependent variable, longitudinal models can provide insights into the genetic basis of FET variation over time. It is expected that the application of longitudinal models as part of genome-wide association (GWA) and genomic selection (i.e. genome-wide selection (GS)) studies will lead to an increase in accuracy of selection. Thus, the objectives of this study were to evaluate the accuracy of estimated breeding values (EBVs) based on pedigree as well as high-density single nucleotide polymorphism (SNP) genotypes, and to conduct a GWA study on longitudinal FI and residual feed intake (RFI) in a total of 312 chickens with phenotype and genotype in the F2 population. The GWA and GS studies reported in this paper were conducted using β-spline random regression models for FI and RFI traits in a chicken F2 population, with FI and BW recorded for each bird weekly between 2 and 10 weeks of age. A single SNP regression approach was used on spline coefficients for weekly FI and RFI traits, with results showing that two significant SNPs for FI occur in the synuclein (SNCAIP) gene. Results also show that these regions are significantly associated with the spline coefficients (q 2) for 5- and 6-week-old birds, while GWA study results showed no SNP association with RFI in F2 chickens. Estimated breeding value predictions obtained using a pedigree-based best linear unbiased prediction (ABLUP) model were then compared with predictions based on genomic best linear unbiased prediction (GBLUP). The accuracy was measured as correlation between genomic EBV and EBV with the phenotypic value corrected for fixed effects divided by the square root of heritability. The regression of observed on predicted values was used to estimate bias of methods. Results show that prediction accuracies using GBLUP and ABLUP for the FI measured from 2nd to 10th week were between 0.06 and 0.46 and 0.03 and 0.37, respectively. These results demonstrate that genomic methods are able to increase the accuracy of predicted breeding values at later ages on the basis of both traits, and indicate that use of a longitudinal model can improve selection accuracy for the trajectory of traits in F2 chickens when compared with conventional methods.
Collapse
|
49
|
Wang Y, Ding X, Tan Z, Ning C, Xing K, Yang T, Pan Y, Sun D, Wang C. Genome-Wide Association Study of Piglet Uniformity and Farrowing Interval. Front Genet 2017; 8:194. [PMID: 29234349 PMCID: PMC5712316 DOI: 10.3389/fgene.2017.00194] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 11/15/2017] [Indexed: 02/04/2023] Open
Abstract
Piglet uniformity (PU) and farrowing interval (FI) are important reproductive traits related to production and economic profits in the pig industry. However, the genetic architecture of the longitudinal trends of reproductive traits still remains elusive. Herein, we performed a genome-wide association study (GWAS) to detect potential genetic variation and candidate genes underlying the phenotypic records at different parities for PU and FI in a population of 884 Large White pigs. In total, 12 significant SNPs were detected on SSC1, 3, 4, 9, and 14, which collectively explained 1–1.79% of the phenotypic variance for PU from parity 1 to 4, and 2.58–4.11% for FI at different stages. Of these, seven SNPs were located within 16 QTL regions related to swine reproductive traits. One QTL region was associated with birth body weight (related to PU) and contained the peak SNP MARC0040730, and another was associated with plasma FSH concentration (related to FI) and contained the SNP MARC0031325. Finally, some positional candidate genes for PU and FI were identified because of their roles in prenatal skeletal muscle development, fetal energy substrate, pre-implantation, and the expression of mammary gland epithelium. Identification of novel variants and candidate genes will greatly advance our understanding of the genetic mechanisms of PU and FI, and suggest a specific opportunity for improving marker assisted selection or genomic selection in pigs.
Collapse
Affiliation(s)
- Yuan Wang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiangdong Ding
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhen Tan
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chao Ning
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Kai Xing
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ting Yang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yongjie Pan
- Beijing Shunxin Agriculture Co., Ltd., Beijing, China
| | - Dongxiao Sun
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Chuduan Wang
- Key Laboratory of Animal Genetics and Breeding of Ministry of Agriculture, National Engineering Laboratory of Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
50
|
Nath AP, Ritchie SC, Byars SG, Fearnley LG, Havulinna AS, Joensuu A, Kangas AJ, Soininen P, Wennerström A, Milani L, Metspalu A, Männistö S, Würtz P, Kettunen J, Raitoharju E, Kähönen M, Juonala M, Palotie A, Ala-Korpela M, Ripatti S, Lehtimäki T, Abraham G, Raitakari O, Salomaa V, Perola M, Inouye M. An interaction map of circulating metabolites, immune gene networks, and their genetic regulation. Genome Biol 2017; 18:146. [PMID: 28764798 PMCID: PMC5540552 DOI: 10.1186/s13059-017-1279-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 07/14/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Immunometabolism plays a central role in many cardiometabolic diseases. However, a robust map of immune-related gene networks in circulating human cells, their interactions with metabolites, and their genetic control is still lacking. Here, we integrate blood transcriptomic, metabolomic, and genomic profiles from two population-based cohorts (total N = 2168), including a subset of individuals with matched multi-omic data at 7-year follow-up. RESULTS We identify topologically replicable gene networks enriched for diverse immune functions including cytotoxicity, viral response, B cell, platelet, neutrophil, and mast cell/basophil activity. These immune gene modules show complex patterns of association with 158 circulating metabolites, including lipoprotein subclasses, lipids, fatty acids, amino acids, small molecules, and CRP. Genome-wide scans for module expression quantitative trait loci (mQTLs) reveal five modules with mQTLs that have both cis and trans effects. The strongest mQTL is in ARHGEF3 (rs1354034) and affects a module enriched for platelet function, independent of platelet counts. Modules of mast cell/basophil and neutrophil function show temporally stable metabolite associations over 7-year follow-up, providing evidence that these modules and their constituent gene products may play central roles in metabolic inflammation. Furthermore, the strongest mQTL in ARHGEF3 also displays clear temporal stability, supporting widespread trans effects at this locus. CONCLUSIONS This study provides a detailed map of natural variation at the blood immunometabolic interface and its genetic basis, and may facilitate subsequent studies to explain inter-individual variation in cardiometabolic disease.
Collapse
Affiliation(s)
- Artika P Nath
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, 3010, Victoria, Australia.,Systems Genomics Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Scott C Ritchie
- Systems Genomics Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Pathology, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Sean G Byars
- Department of Pathology, The University of Melbourne, Parkville, 3010, Victoria, Australia.,School of BioSciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Liam G Fearnley
- Department of Pathology, The University of Melbourne, Parkville, 3010, Victoria, Australia.,School of BioSciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Aki S Havulinna
- National Institute for Health and Welfare, Helsinki, 00271, Finland.,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, 00014, Finland
| | - Anni Joensuu
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Antti J Kangas
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, 90014, Finland
| | - Pasi Soininen
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, 90014, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland
| | | | - Lili Milani
- University of Tartu, Estonian Genome Center, Tartu, 51010, Estonia
| | - Andres Metspalu
- University of Tartu, Estonian Genome Center, Tartu, 51010, Estonia
| | - Satu Männistö
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Peter Würtz
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, 90014, Finland.,Diabetes and Obesity Research Program, University of Helsinki, Helsinki, Finland
| | - Johannes Kettunen
- National Institute for Health and Welfare, Helsinki, 00271, Finland.,Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, 90014, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland.,Biocenter Oulu, University of Oulu, Oulu, 90014, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, 33014, Tampere, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, University of Tampere and Tampere University Hospital, FI-33521, Tampere, Finland
| | - Markus Juonala
- Department of Medicine, University of Turku and Division of Medicine, Turku University Hospital, FI-20520, Turku, Finland.,Murdoch Childrens Research Institute, Parkville, 3052, Victoria, Australia
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, 00014, Finland.,Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.,Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA.,Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mika Ala-Korpela
- Computational Medicine, Faculty of Medicine, University of Oulu, Oulu, 90014, Finland.,NMR Metabolomics Laboratory, School of Pharmacy, University of Eastern Finland, Kuopio, 70211, Finland.,Biocenter Oulu, University of Oulu, Oulu, 90014, Finland.,Computational Medicine, School of Social and Community Medicine, University of Bristol, Bristol, BS8 1TH, UK.,Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, 00014, Finland.,Department of Public Health, University of Helsinki, Helsinki, 00014, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center-Tampere, Faculty of Medicine and Life Sciences, University of Tampere, 33014, Tampere, Finland
| | - Gad Abraham
- Systems Genomics Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Pathology, The University of Melbourne, Parkville, 3010, Victoria, Australia.,School of BioSciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Olli Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, 20520, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, 20520, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, 00271, Finland
| | - Markus Perola
- National Institute for Health and Welfare, Helsinki, 00271, Finland.,Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, 00014, Finland.,University of Tartu, Estonian Genome Center, Tartu, 51010, Estonia
| | - Michael Inouye
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, 3010, Victoria, Australia. .,Systems Genomics Lab, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia. .,Department of Pathology, The University of Melbourne, Parkville, 3010, Victoria, Australia. .,School of BioSciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| |
Collapse
|