1
|
Parnell LD, McCaffrey KS, Brooks AW, Smith CE, Lai CQ, Christensen JJ, Wiley CD, Ordovas JM. Rate-Limiting Enzymes in Cardiometabolic Health and Aging in Humans. Lifestyle Genom 2023; 16:124-138. [PMID: 37473740 DOI: 10.1159/000531350] [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: 02/14/2023] [Accepted: 05/24/2023] [Indexed: 07/22/2023] Open
Abstract
INTRODUCTION Rate-limiting enzymes (RLEs) are innate slow points in metabolic pathways, and many function in bio-processes related to nutrient sensing. Many RLEs carry causal mutations relevant to inherited metabolic disorders. Because the activity of RLEs in cardiovascular health is poorly characterized, our objective was to assess their involvement in cardiometabolic health and disease and where altered biophysical and biochemical functions can promote disease. METHODS A dataset of 380 human RLEs was compared to protein and gene datasets for factors likely to contribute to cardiometabolic disease, including proteins showing significant age-related altered expression in blood and genetic loci with variants that associate with common cardiometabolic phenotypes. The biochemical reactions catalyzed by RLEs were evaluated for metabolites enriched in RLE subsets associating with various cardiometabolic phenotypes. Most significance tests were based on Z-score enrichment converted to p values with a normal distribution function. RESULTS Of 380 RLEs analyzed, 112 function in mitochondria, and 53 are assigned to inherited metabolic disorders. There was a depletion of RLE proteins known as aging biomarkers. At the gene level, RLEs were assessed for common genetic variants that associated with important cardiometabolic traits of LDL-cholesterol or any of the five outcomes pertinent to metabolic syndrome. This revealed several RLEs with links to cardiometabolic traits, from a minimum of 26 for HDL-cholesterol to a maximum of 45 for plasma glucose. Analysis of these GWAS-linked RLEs for enrichment of the molecular constituents of the catalyzed reactions disclosed a number of significant phenotype-metabolite links. These included blood pressure with acetate (p = 2.2 × 10-4) and NADP+ (p = 0.0091), plasma HDL-cholesterol and triglyceride with diacylglycerol (p = 2.6 × 10-5, 6.4 × 10-5, respectively) and diolein (p = 2.2 × 10-6, 5.9 × 10-6), and waist circumference with d-glucosamine-6-phosphate (p = 1.8 × 10-4). CONCLUSION In the context of cardiometabolic health, aging, and disease, these results highlight key diet-derived metabolites that are central to specific rate-limited processes that are linked to cardiometabolic health. These metabolites include acetate and diacylglycerol, pertinent to blood pressure and triglycerides, respectively, as well as diacylglycerol and HDL-cholesterol.
Collapse
Affiliation(s)
- Laurence D Parnell
- US Department of Agriculture, Nutrition and Genomics Laboratory, Agricultural Research Service, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| | | | | | - Caren E Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| | - Chao-Qiang Lai
- US Department of Agriculture, Nutrition and Genomics Laboratory, Agricultural Research Service, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| | - Jacob J Christensen
- Norwegian National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Oslo, Norway
- Department of Nutrition, University of Oslo, Oslo, Norway
| | - Christopher D Wiley
- Vitamin K Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Suciu I, Pamies D, Peruzzo R, Wirtz PH, Smirnova L, Pallocca G, Hauck C, Cronin MTD, Hengstler JG, Brunner T, Hartung T, Amelio I, Leist M. G × E interactions as a basis for toxicological uncertainty. Arch Toxicol 2023; 97:2035-2049. [PMID: 37258688 PMCID: PMC10256652 DOI: 10.1007/s00204-023-03500-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
To transfer toxicological findings from model systems, e.g. animals, to humans, standardized safety factors are applied to account for intra-species and inter-species variabilities. An alternative approach would be to measure and model the actual compound-specific uncertainties. This biological concept assumes that all observed toxicities depend not only on the exposure situation (environment = E), but also on the genetic (G) background of the model (G × E). As a quantitative discipline, toxicology needs to move beyond merely qualitative G × E concepts. Research programs are required that determine the major biological variabilities affecting toxicity and categorize their relative weights and contributions. In a complementary approach, detailed case studies need to explore the role of genetic backgrounds in the adverse effects of defined chemicals. In addition, current understanding of the selection and propagation of adverse outcome pathways (AOP) in different biological environments is very limited. To improve understanding, a particular focus is required on modulatory and counter-regulatory steps. For quantitative approaches to address uncertainties, the concept of "genetic" influence needs a more precise definition. What is usually meant by this term in the context of G × E are the protein functions encoded by the genes. Besides the gene sequence, the regulation of the gene expression and function should also be accounted for. The widened concept of past and present "gene expression" influences is summarized here as Ge. Also, the concept of "environment" needs some re-consideration in situations where exposure timing (Et) is pivotal: prolonged or repeated exposure to the insult (chemical, physical, life style) affects Ge. This implies that it changes the model system. The interaction of Ge with Et might be denoted as Ge × Et. We provide here general explanations and specific examples for this concept and show how it could be applied in the context of New Approach Methodologies (NAM).
Collapse
Affiliation(s)
- Ilinca Suciu
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany
| | - David Pamies
- Department of Biological Sciences, University of Lausanne, 1005, Lausanne, Switzerland
| | - Roberta Peruzzo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Petra H Wirtz
- Centre for the Advanced Study of Collective Behaviour, University of Konstanz, 78457, Constance, Germany
- Biological Work and Health Psychology, Department of Psychology, University of Konstanz, 78457, Constance, Germany
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | | - Christof Hauck
- Department of Cell Biology, University of Konstanz, 78457, Constance, Germany
| | - Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, 44139, Dortmund, Germany
| | - Thomas Brunner
- Biochemical Pharmacology, Department of Biology, University of Konstanz, 78457, Constance, Germany
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- CAAT Europe, University of Konstanz, 78457, Constance, Germany
| | - Ivano Amelio
- Division for Systems Toxicology, Department of Biology, University of Konstanz, 78457, Constance, Germany
| | - Marcel Leist
- In Vitro Toxicology and Biomedicine, Department Inaugurated By the Doerenkamp-Zbinden Foundation, University of Konstanz, Universitaetsstr. 10, 78457, Constance, Germany.
- CAAT Europe, University of Konstanz, 78457, Constance, Germany.
| |
Collapse
|
3
|
Hu J, Ji Y, Lang X, Zhang XY. Prevalence and clinical correlates of abnormal lipid metabolism in first-episode and drug-naïve patients with major depressive disorder: A large-scale cross-sectional study. J Psychiatr Res 2023; 163:55-62. [PMID: 37201238 DOI: 10.1016/j.jpsychires.2023.05.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/20/2023]
Abstract
OBJECTIVE Studies have shown an association between abnormal lipid profiles and MDD, but there are few studies on the clinical correlates of lipid abnormalities in patients with major depressive disorder (MDD). The purpose of this study was to investigate the prevalence of abnormal lipid metabolism and its correlates in Chinese first-episode and drug-naïve MDD patients, which has not yet been reported. METHODS A total of 1718 outpatients with first-episode and drug-naïve MDD were included. Demographic data were collected by a standardized questionnaire and blood lipid levels were measured, including total cholesterol (TC), triglyceride (TG), low density lipoprotein (LDL-C), high density lipoprotein (HDL-C). The Hamilton Depression Scale (HAMD), Hamilton Anxiety Scale (HAMA), Positive and Negative Syndrome Scale (PANSS) positive subscale, and Clinical Global Impression of Severity Scale (CGI-S) were assessed for each patient. RESULTS The prevalence of abnormal lipid metabolism was 72.73% (1301/1718), and the rates of high TC, high TG, high LDL-C and low HDL-C were 51.05% (877/1718), 61.18% (1051/1718), 30.09% (517/1718), 23.40% (402/1718), respectively. Logistic regression showed the risk factors for abnormal lipid metabolism were severe anxiety, HAMD score, CGI-S score, BMI and systolic blood pressure (SBP). Multiple linear regression analysis showed that age at onset, SBP, HAMD score, HAMA score, PANSS positive subscale score, and CGI-S were independently associated with TC levels. BMI, HAMD score, PANSS positive subscale score and CGI-S score were independently associated with TG levels. SBP, HAMD score, PANSS positive subscale score and CGI-S score were independently associated with LDL-C levels. Age of onset, SBP and CGI-S score were independently associated with HDL-C levels. CONCLUSIONS The prevalence of abnormal lipid metabolism in first-episode and drug-naïve MDD patients is quite high. The severity of psychiatric symptoms may be closely associated with the presence of abnormal lipid metabolism in patients with MDD.
Collapse
Affiliation(s)
- Jieqiong Hu
- Department of Psychosomatic Medicine, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - Yunxin Ji
- Department of Psychosomatic Medicine, Ningbo First Hospital, Ningbo, Zhejiang, China
| | - XiaoE Lang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiang-Yang Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
4
|
Parnell LD, Magadmi R, Zwanger S, Shukitt-Hale B, Lai CQ, Ordovás JM. Dietary Responses of Dementia-Related Genes Encoding Metabolic Enzymes. Nutrients 2023; 15:644. [PMID: 36771351 PMCID: PMC9921944 DOI: 10.3390/nu15030644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
The age-related loss of the cognitive function is a growing concern for global populations. Many factors that determine cognitive resilience or dementia also have metabolic functions. However, this duality is not universally appreciated when the action of that factor occurs in tissues external to the brain. Thus, we examined a set of genes involved in dementia, i.e., those related to vascular dementia, Alzheimer's disease, Parkinson's disease, and the human metabolism for activity in 12 metabolically active tissues. Mining the Genotype-Tissue Expression (GTEx) data showed that most of these metabolism-dementia (MD) genes (62 of 93, 67%) exhibit a higher median expression in any of the metabolically active tissues than in the brain. After identifying that several MD genes served as blood-based biomarkers of longevity in other studies, we examined the impact of the intake of food, nutrients, and other dietary factors on the expression of MD genes in whole blood in the Framingham Offspring Study (n = 2134). We observed positive correlations between flavonoids and HMOX1, taurine and UQCRC1, broccoli and SLC10A2, and myricetin and SLC9A8 (p < 2.09 × 10-4). In contrast, dairy protein, palmitic acid, and pie were negatively correlated, respectively, with the expression of IGF1R, CSF1R, and SLC9A8, among others (p < 2.92 × 10-4). The results of this investigation underscore the potential contributions of metabolic enzyme activity in non-brain tissues to the risk of dementia. Specific epidemiological or intervention studies could be designed using specific foods and nutrients or even dietary patterns focused on these foods and nutrients that influence the expression of some MD genes to verify the findings presented here.
Collapse
Affiliation(s)
- Laurence D Parnell
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, US Department of Agriculture, Boston, MA 02111, USA
| | - Rozana Magadmi
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA 02111, USA
| | | | - Barbara Shukitt-Hale
- Neuroscience and Aging Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, US Department of Agriculture, Boston, MA 02111, USA
| | - Chao-Qiang Lai
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, US Department of Agriculture, Boston, MA 02111, USA
| | - José M Ordovás
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA 02111, USA
| |
Collapse
|
5
|
Lee BY, Ordovás JM, Parks EJ, Anderson CAM, Barabási AL, Clinton SK, de la Haye K, Duffy VB, Franks PW, Ginexi EM, Hammond KJ, Hanlon EC, Hittle M, Ho E, Horn AL, Isaacson RS, Mabry PL, Malone S, Martin CK, Mattei J, Meydani SN, Nelson LM, Neuhouser ML, Parent B, Pronk NP, Roche HM, Saria S, Scheer FAJL, Segal E, Sevick MA, Spector TD, Van Horn L, Varady KA, Voruganti VS, Martinez MF. Research gaps and opportunities in precision nutrition: an NIH workshop report. Am J Clin Nutr 2022; 116:1877-1900. [PMID: 36055772 PMCID: PMC9761773 DOI: 10.1093/ajcn/nqac237] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/06/2022] [Accepted: 08/30/2022] [Indexed: 02/01/2023] Open
Abstract
Precision nutrition is an emerging concept that aims to develop nutrition recommendations tailored to different people's circumstances and biological characteristics. Responses to dietary change and the resulting health outcomes from consuming different diets may vary significantly between people based on interactions between their genetic backgrounds, physiology, microbiome, underlying health status, behaviors, social influences, and environmental exposures. On 11-12 January 2021, the National Institutes of Health convened a workshop entitled "Precision Nutrition: Research Gaps and Opportunities" to bring together experts to discuss the issues involved in better understanding and addressing precision nutrition. The workshop proceeded in 3 parts: part I covered many aspects of genetics and physiology that mediate the links between nutrient intake and health conditions such as cardiovascular disease, Alzheimer disease, and cancer; part II reviewed potential contributors to interindividual variability in dietary exposures and responses such as baseline nutritional status, circadian rhythm/sleep, environmental exposures, sensory properties of food, stress, inflammation, and the social determinants of health; part III presented the need for systems approaches, with new methods and technologies that can facilitate the study and implementation of precision nutrition, and workforce development needed to create a new generation of researchers. The workshop concluded that much research will be needed before more precise nutrition recommendations can be achieved. This includes better understanding and accounting for variables such as age, sex, ethnicity, medical history, genetics, and social and environmental factors. The advent of new methods and technologies and the availability of considerably more data bring tremendous opportunity. However, the field must proceed with appropriate levels of caution and make sure the factors listed above are all considered, and systems approaches and methods are incorporated. It will be important to develop and train an expanded workforce with the goal of reducing health disparities and improving precision nutritional advice for all Americans.
Collapse
Affiliation(s)
- Bruce Y Lee
- Health Policy and Management, City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| | - José M Ordovás
- USDA-Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Elizabeth J Parks
- Nutrition and Exercise Physiology, University of Missouri School of Medicine, MO, USA
| | | | - Albert-László Barabási
- Network Science Institute and Department of Physics, Northeastern University, Boston, MA, USA
| | | | - Kayla de la Haye
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Valerie B Duffy
- Allied Health Sciences, University of Connecticut, Storrs, CT, USA
| | - Paul W Franks
- Novo Nordisk Foundation, Hellerup, Denmark, Copenhagen, Denmark, and Lund University Diabetes Center, Sweden
- The Lund University Diabetes Center, Malmo, SwedenInsert Affiliation Text Here
| | - Elizabeth M Ginexi
- National Institutes of Health, Office of Behavioral and Social Sciences Research, Bethesda, MD, USA
| | - Kristian J Hammond
- Computer Science, Northwestern University McCormick School of Engineering, IL, USA
| | - Erin C Hanlon
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Michael Hittle
- Epidemiology and Clinical Research, Stanford University, Stanford, CA, USA
| | - Emily Ho
- Public Health and Human Sciences, Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Abigail L Horn
- Information Sciences Institute, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | | | | | - Susan Malone
- Rory Meyers College of Nursing, New York University, New York, NY, USA
| | - Corby K Martin
- Ingestive Behavior Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Josiemer Mattei
- Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Simin Nikbin Meydani
- USDA-Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Lorene M Nelson
- Epidemiology and Population Health, Stanford University, Stanford, CA, USA
| | | | - Brendan Parent
- Grossman School of Medicine, New York University, New York, NY, USA
| | | | - Helen M Roche
- UCD Conway Institute, School of Public Health, Physiotherapy, and Sports Science, University College Dublin, Dublin, Ireland
| | - Suchi Saria
- Johns Hopkins University, Baltimore, MD, USA
| | - Frank A J L Scheer
- Brigham and Women's Hospital, Boston, MA, USA
- Medicine and Neurology, Harvard Medical School, Boston, MA, USA
| | - Eran Segal
- Computer Science and Applied Math, Weizmann Institute of Science, Rehovot, Israel
| | - Mary Ann Sevick
- Grossman School of Medicine, New York University, New York, NY, USA
| | - Tim D Spector
- Twin Research and Genetic Epidemiology, King's College London, London, United Kingdom
| | - Linda Van Horn
- Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Krista A Varady
- Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL, USA
| | - Venkata Saroja Voruganti
- Nutrition and Nutrition Research Institute, Gillings School of Public Health, The University of North Carolina, Chapel Hill, NC, USA
| | - Marie F Martinez
- Health Policy and Management, City University of New York Graduate School of Public Health and Health Policy, New York, NY, USA
| |
Collapse
|
6
|
Wang F, Zheng J, Cheng J, Zou H, Li M, Deng B, Luo R, Wang F, Huang D, Li G, Zhang R, Ding X, Li Y, Du J, Yang Y, Kan J. Personalized nutrition: A review of genotype-based nutritional supplementation. Front Nutr 2022; 9:992986. [PMID: 36159456 PMCID: PMC9500586 DOI: 10.3389/fnut.2022.992986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Nutritional disorders have become a major public health issue, requiring increased targeted approaches. Personalized nutrition adapted to individual needs has garnered dramatic attention as an effective way to improve nutritional balance and maintain health. With the rapidly evolving fields of genomics and nutrigenetics, accumulation of genetic variants has been indicated to alter the effects of nutritional supplementation, suggesting its indispensable role in the genotype-based personalized nutrition. Additionally, the metabolism of nutrients, such as lipids, especially omega-3 polyunsaturated fatty acids, glucose, vitamin A, folic acid, vitamin D, iron, and calcium could be effectively improved with related genetic variants. This review focuses on existing literatures linking critical genetic variants to the nutrient and the ways in which these variants influence the outcomes of certain nutritional supplementations. Although further studies are required in this direction, such evidence provides valuable insights for the guidance of appropriate interventions using genetic information, thus paving the way for the smooth transition of conventional generic approach to genotype-based personalized nutrition.
Collapse
Affiliation(s)
| | | | - Junrui Cheng
- Department of Molecular and Structural Biochemistry, North Carolina State University, Kannapolis, NC, United States
| | - Hong Zou
- Sequanta Technologies Co., Ltd, Shanghai, China
| | | | - Bin Deng
- Nutrilite Health Institute, Guangzhou, China
| | - Rong Luo
- Nutrilite Health Institute, Guangzhou, China
| | - Feng Wang
- Nutrilite Health Institute, Guangzhou, China
| | | | - Gang Li
- Nutrilite Health Institute, Shanghai, China
| | - Rao Zhang
- School of Public Health, Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Xin Ding
- School of Public Health, Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Yuan Li
- Sequanta Technologies Co., Ltd, Shanghai, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai, China
- Jun Du
| | - Yuexin Yang
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, China
- Yuexin Yang
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai, China
- *Correspondence: Juntao Kan
| |
Collapse
|
7
|
Westerman KE, Majarian TD, Giulianini F, Jang DK, Miao J, Florez JC, Chen H, Chasman DI, Udler MS, Manning AK, Cole JB. Variance-quantitative trait loci enable systematic discovery of gene-environment interactions for cardiometabolic serum biomarkers. Nat Commun 2022; 13:3993. [PMID: 35810165 PMCID: PMC9271055 DOI: 10.1038/s41467-022-31625-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/24/2022] [Indexed: 11/29/2022] Open
Abstract
Gene-environment interactions represent the modification of genetic effects by environmental exposures and are critical for understanding disease and informing personalized medicine. These often induce differential phenotypic variance across genotypes; these variance-quantitative trait loci can be prioritized in a two-stage interaction detection strategy to greatly reduce the computational and statistical burden and enable testing of a broader range of exposures. We perform genome-wide variance-quantitative trait locus analysis for 20 serum cardiometabolic biomarkers by multi-ancestry meta-analysis of 350,016 unrelated participants in the UK Biobank, identifying 182 independent locus-biomarker pairs (p < 4.5×10-9). Most are concentrated in a small subset (4%) of loci with genome-wide significant main effects, and 44% replicate (p < 0.05) in the Women's Genome Health Study (N = 23,294). Next, we test each locus-biomarker pair for interaction across 2380 exposures, identifying 847 significant interactions (p < 2.4×10-7), of which 132 are independent (p < 0.05) after accounting for correlation between exposures. Specific examples demonstrate interaction of triglyceride-associated variants with distinct body mass- versus body fat-related exposures as well as genotype-specific associations between alcohol consumption and liver stress at the ADH1B gene. Our catalog of variance-quantitative trait loci and gene-environment interactions is publicly available in an online portal.
Collapse
Affiliation(s)
- Kenneth E Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA.
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Timothy D Majarian
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Franco Giulianini
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dong-Keun Jang
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jenkai Miao
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| | - Jose C Florez
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Genetics, Brigham and Women's Hospital, Boston, MA, 02115, USA
- Medical and Population Genetics Program, Broad Institute, Cambridge, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Miriam S Udler
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Alisa K Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Joanne B Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
8
|
Lee YC, Christensen JJ, Parnell LD, Smith CE, Shao J, McKeown NM, Ordovás JM, Lai CQ. Using Machine Learning to Predict Obesity Based on Genome-Wide and Epigenome-Wide Gene-Gene and Gene-Diet Interactions. Front Genet 2022; 12:783845. [PMID: 35047011 PMCID: PMC8763388 DOI: 10.3389/fgene.2021.783845] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity is associated with many chronic diseases that impair healthy aging and is governed by genetic, epigenetic, and environmental factors and their complex interactions. This study aimed to develop a model that predicts an individual's risk of obesity by better characterizing these complex relations and interactions focusing on dietary factors. For this purpose, we conducted a combined genome-wide and epigenome-wide scan for body mass index (BMI) and up to three-way interactions among 402,793 single nucleotide polymorphisms (SNPs), 415,202 DNA methylation sites (DMSs), and 397 dietary and lifestyle factors using the generalized multifactor dimensionality reduction (GMDR) method. The training set consisted of 1,573 participants in exam 8 of the Framingham Offspring Study (FOS) cohort. After identifying genetic, epigenetic, and dietary factors that passed statistical significance, we applied machine learning (ML) algorithms to predict participants' obesity status in the test set, taken as a subset of independent samples (n = 394) from the same cohort. The quality and accuracy of prediction models were evaluated using the area under the receiver operating characteristic curve (ROC-AUC). GMDR identified 213 SNPs, 530 DMSs, and 49 dietary and lifestyle factors as significant predictors of obesity. Comparing several ML algorithms, we found that the stochastic gradient boosting model provided the best prediction accuracy for obesity with an overall accuracy of 70%, with ROC-AUC of 0.72 in test set samples. Top predictors of the best-fit model were 21 SNPs, 230 DMSs in genes such as CPT1A, ABCG1, SLC7A11, RNF145, and SREBF1, and 26 dietary factors, including processed meat, diet soda, French fries, high-fat dairy, artificial sweeteners, alcohol intake, and specific nutrients and food components, such as calcium and flavonols. In conclusion, we developed an integrated approach with ML to predict obesity using omics and dietary data. This extends our knowledge of the drivers of obesity, which can inform precision nutrition strategies for the prevention and treatment of obesity. Clinical Trial Registration: [www.ClinicalTrials.gov], the Framingham Heart Study (FHS), [NCT00005121].
Collapse
Affiliation(s)
- Yu-Chi Lee
- USDA ARS, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Jacob J. Christensen
- Department of Nutrition, Norwegian National Advisory Unit on FH, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Laurence D. Parnell
- USDA ARS, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Caren E. Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| | - Jonathan Shao
- Statistical and Bioinformatics Group, Northeast Area, USDA ARS, Beltsville, MD, United States
| | - Nicola M. McKeown
- Nutritional Epidemiology Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, United States
| | - José M. Ordovás
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
- CEI UAM + CSIC, IMDEA Food Institute, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Chao-Qiang Lai
- USDA ARS, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States
| |
Collapse
|
9
|
San-Cristobal R, de Toro-Martín J, Vohl MC. Appraisal of Gene-Environment Interactions in GWAS for Evidence-Based Precision Nutrition Implementation. Curr Nutr Rep 2022; 11:563-573. [PMID: 35948824 PMCID: PMC9750926 DOI: 10.1007/s13668-022-00430-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW This review aims to analyse the currently reported gene-environment (G × E) interactions in genome-wide association studies (GWAS), involving environmental factors such as lifestyle and dietary habits related to metabolic syndrome phenotypes. For this purpose, the present manuscript reviews the available GWAS registered on the GWAS Catalog reporting the interaction between environmental factors and metabolic syndrome traits. RECENT FINDINGS Advances in omics-related analytical and computational approaches in recent years have led to a better understanding of the biological processes underlying these G × E interactions. A total of 42 GWAS were analysed, reporting over 300 loci interacting with environmental factors. Alcohol consumption, sleep time, smoking habit and physical activity were the most studied environmental factors with significant G × E interactions. The implementation of more comprehensive GWAS will provide a better understanding of the metabolic processes that determine individual responses to environmental exposures and their association with the development of chronic diseases such as obesity and the metabolic syndrome. This will facilitate the development of precision approaches for better prevention, management and treatment of these diseases.
Collapse
Affiliation(s)
- Rodrigo San-Cristobal
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| | - Juan de Toro-Martín
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| | - Marie-Claude Vohl
- grid.23856.3a0000 0004 1936 8390Centre Nutrition, Santé Et Société (NUTRISS), Institut Sur La Nutrition Et Les Aliments Fonctionnels (INAF), Université Laval, Québec, QC Canada ,grid.23856.3a0000 0004 1936 8390School of Nutrition, Université Laval, Quebec, QC G1V 0A6 Canada
| |
Collapse
|
10
|
Zago VHS, Scherrer DZ, Parra ES, Vieira IC, Marson FAL, de Faria EC. Effects of SNVs in ABCA1, ABCG1, ABCG5, ABCG8, and SCARB1 Genes on Plasma Lipids, Lipoproteins, and Adiposity Markers in a Brazilian Population. Biochem Genet 2021; 60:822-841. [PMID: 34505223 DOI: 10.1007/s10528-021-10131-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 08/25/2021] [Indexed: 10/20/2022]
Abstract
Several proteins are involved in cholesterol homeostasis, as scavenger receptor class B type I and ATP-binding cassette (ABC) transporters including ABCA1, ABCG1, ABCG5, and ABCG8. This study aimed to determine the effects of single nucleotide variants (SNVs) rs2275543 (ABCA1), rs1893590 (ABCG1), rs6720173 (ABCG5), rs6544718 (ABCG8), and rs5888 (SCARB1) on plasma lipids, lipoproteins, and adiposity markers in an asymptomatic population and its sex-specific effects. Volunteers (n = 590) were selected and plasma lipids, lipoproteins, and adiposity markers (waist-to-hip and waist-to-height ratios, lipid accumulation product and body adiposity index) were measured. Genomic DNA was isolated from peripheral blood cells according to the method adapted from Gross-Bellard. SNVs were detected in the TaqMan® OpenArray® Real-Time polymerase chain reaction platform and data analyses were performed using the TaqMan® Genotyper Software. The rs2275543*C point to an increase of high-density lipoprotein size in females while in males very-low-density lipoprotein, cholesterol, and triglycerides were statistically lower (P value < 0.05). The rs1893590*C was statistically associated with lower apolipoprotein A-I levels and higher activities of paraoxonase-1 and cholesteryl ester transfer protein (P value < 0.05). The rs6720173 was statistically associated with an increase in cholesterol and low-density lipoprotein cholesterol in males; moreover, rs6544718*T reduced adiposity markers in females (P value < 0.05). Regarding the rs5888, a decreased adiposity marker in the total population and in females occurred (P value < 0.05). Multivariate analysis of variance showed that SNVs could influence components of high-density lipoprotein metabolism, mainly through ABCG1 (P value < 0.05). The ABCA1 and ABCG5 variants showed sex-specific effects on lipids and lipoproteins, while SCARB1 and ABCG8 variants might influence adiposity markers in females. Our data indicate a possible role of ABCG1 on HDL metabolism.
Collapse
Affiliation(s)
- Vanessa Helena Souza Zago
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil
| | - Daniel Zanetti Scherrer
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil
| | - Eliane Soler Parra
- Department of Cardiology, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil
| | - Isabela Calanca Vieira
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil
| | - Fernando Augusto Lima Marson
- Department of Pediatrics, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil. .,Department of Medical Genetics and Genomic Medicine, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil. .,Laboratory of Human and Medical Genetics and Laboratory of Cell and Molecular Tumor Biology and Bioactive Compounds, Post Graduate Program in Health Science, São Francisco University, Avenida São Francisco de Assis, 218, Jardim São José, Bragança Paulista, São Paulo, 12916-900, Brazil.
| | - Eliana Cotta de Faria
- Department of Clinical Pathology, Faculty of Medical Sciences, State University of Campinas, Tessália Vieira de Camargo St, 126, Campinas, São Paulo, 13084-971, Brazil.
| |
Collapse
|
11
|
Riccardi G, Giosuè A, Calabrese I, Vaccaro O. Dietary recommendations for prevention of atherosclerosis. Cardiovasc Res 2021; 118:1188-1204. [PMID: 34229346 DOI: 10.1093/cvr/cvab173] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/07/2021] [Accepted: 05/18/2021] [Indexed: 12/17/2022] Open
Abstract
This review aims at summarizing updated evidence on cardiovascular disease (CVD) risk associated with consumption of specific food items to substantiate dietary strategies for atherosclerosis prevention. A systematic search on PubMed was performed to identify meta-analyses of cohort studies and RCTs with CVD outcomes. The evidence is highly concordant in showing that, for the healthy adult population, low consumption of salt and foods of animal origin, and increased intake of plant-based foods-whole grains, fruits, vegetables, legumes, and nuts-are linked with reduced atherosclerosis risk. The same applies for the replacement of butter and other animal/tropical fats with olive oil and other unsaturated-fat-rich oil. Although the literature reviewed overall endorses scientific society dietary recommendations, some relevant novelties emerge. With regard to meat, new evidence differentiates processed and red meat-both associated with increased CVD risk-from poultry, showing a neutral relationship with CVD for moderate intakes. Moreover, the preferential use of low-fat dairies in the healthy population is not supported by recent data, since both full-fat and low-fat dairies, in moderate amounts and in the context of a balanced diet, are not associated with increased CVD risk; furthermore, small quantities of cheese and regular yogurt consumption are even linked with a protective effect. Among other animal protein sources, moderate fish consumption is also supported by the latest evidence, although there might be sustainability concerns. New data endorse the replacement of most high glycemic index (GI) foods with both whole grain and low GI cereal foods. As for beverages, low consumption not only of alcohol, but also of coffee and tea is associated with a reduced atherosclerosis risk while soft drinks show a direct relationship with CVD risk. This review provides evidence-based support for promoting appropriate food choices for atherosclerosis prevention in the general population.
Collapse
Affiliation(s)
- Gabriele Riccardi
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples, Via Sergio Pansini, 5 - 80131, Naples, Italy
| | - Annalisa Giosuè
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples, Via Sergio Pansini, 5 - 80131, Naples, Italy
| | - Ilaria Calabrese
- Department of Clinical Medicine and Surgery, "Federico II" University of Naples, Via Sergio Pansini, 5 - 80131, Naples, Italy
| | - Olga Vaccaro
- Department of Pharmacy, "Federico II" University of Naples, Via Domenico Montesano, 49 - 80131, Naples, Italy
| |
Collapse
|
12
|
Wendt FR, Koller D, Pathak GA, Jacoby D, Miller EJ, Polimanti R. Biobank Scale Pharmacogenomics Informs the Genetic Underpinnings of Simvastatin Use. Clin Pharmacol Ther 2021; 110:777-785. [PMID: 33837531 DOI: 10.1002/cpt.2260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/01/2021] [Indexed: 12/31/2022]
Abstract
Studying drug-metabolizing enzymes, encoded by pharmacogenes, may inform biological mechanisms underlying the diseases for which a medication is prescribed. Until recently, pharmacogenes could not be studied at biobank scale. In 7,649 unrelated African-ancestry (AFR) and 326,214 unrelated European-ancestry (EUR) participants from the UK Biobank, we associated pharmacogene haplotypes from 50 genes with 265 (EUR) and 17 (AFR) medication use phenotypes using generalized linear models. In EUR, N-acetyltransferase 2 (NAT2) metabolizer phenotype and activity score were associated with simvastatin use. The dose of NAT2*1 was associated with simvastatin use when compared with NAT2*5 (the most common haplotype). This association was robust to effects of low-density lipoprotein cholesterol (LDL-C) concentration (NAT2*1 odds ratio (OR) = 1.07, 95% CI: 1.05-1.09, P = 1.14 × 10-8 ) and polygenic risk for LDL-C concentration (NAT2*1 OR = 1.09, 95% CI: 1.04-1.14, P = 2.26 × 10-4 ). Interactive effects between NAT2*1 and simvastatin use on LDL-C concentration (OR = 0.957, 95% CI: 0.916-0.998, P = 0.045) were replicated in the electronic Medical Records and Genomics Pharmacogenetic Sequencing Pilot (eMERGE-PGx) cohort (OR = 0.987, 95% CI: 0.976-0.998, P = 0.029). We used biobank-scale data to uncover and replicate an association between NAT2 locus variation and better response to statin therapy. Testing NAT2 alleles may be useful for making clinical decisions regarding the potential benefit (e.g., absolute risk reduction) in LDL-C concentration prior to statin treatment.
Collapse
Affiliation(s)
- Frank R Wendt
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Dora Koller
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Gita A Pathak
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Daniel Jacoby
- Section of Cardiovascular Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Edward J Miller
- Section of Cardiovascular Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Renato Polimanti
- Division of Human Genetics, Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| |
Collapse
|
13
|
Zhang W, Guo X, Chen L, Chen T, Yu J, Wu C, Zheng J. Ketogenic Diets and Cardio-Metabolic Diseases. Front Endocrinol (Lausanne) 2021; 12:753039. [PMID: 34795641 PMCID: PMC8594484 DOI: 10.3389/fendo.2021.753039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/13/2021] [Indexed: 12/31/2022] Open
Abstract
While the prevalence of cardio-metabolic diseases (CMDs) has become a worldwide epidemic, much attention is paid to managing CMDs effectively. A ketogenic diet (KD) constitutes a high-fat and low-carbohydrate diet with appropriate protein content and calories. KD has drawn the interests of clinicians and scientists regarding its application in the management of metabolic diseases and related disorders; thus, the current review aimed to examine the evidences surrounding KD and the CMDs to draw the clinical implications. Overall, KD appears to play a significant role in the therapy of various CMDs, which is manifested by the effects of KDs on cardio-metabolic outcomes. KD therapy is generally promising in obesity, heart failure, and hypertension, though different voices still exist. In diabetes and dyslipidemia, the performance of KD remains controversial. As for cardiovascular complications of metabolic diseases, current evidence suggests that KD is generally protective to obese related cardiovascular disease (CVD), while remaining contradictory to diabetes and other metabolic disorder related CVDs. Various factors might account for the controversies, including genetic background, duration of therapy, food composition, quality, and sources of KDs. Therefore, it's crucial to perform more rigorous researches to focus on clinical safety and appropriate treatment duration and plan of KDs.
Collapse
Affiliation(s)
- Weiyue Zhang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Xin Guo
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lulu Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Ting Chen
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Jiayu Yu
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
| | - Chaodong Wu
- Department of Nutrition, Texas A&M University, College Station, TX, United States
- *Correspondence: Juan Zheng, ; Chaodong Wu,
| | - Juan Zheng
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Provincial Clinical Research Center for Diabetes and Metabolic Disorders, Wuhan, China
- *Correspondence: Juan Zheng, ; Chaodong Wu,
| |
Collapse
|
14
|
Williams PT. Quantile-Dependent Expressivity and Gene-Lifestyle Interactions Involving High-Density Lipoprotein Cholesterol. Lifestyle Genom 2020; 14:1-19. [PMID: 33296900 DOI: 10.1159/000511421] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The phenotypic expression of a high-density lipoprotein (HDL) genetic risk score has been shown to depend upon whether the phenotype (HDL-cholesterol) is high or low relative to its distribution in the population (quantile-dependent expressivity). This may be due to the effects of genetic mutations on HDL-metabolism being concentration dependent. METHOD The purpose of this article is to assess whether some previously reported HDL gene-lifestyle interactions could potentially be attributable to quantile-dependent expressivity. SUMMARY Seventy-three published examples of HDL gene-lifestyle interactions were interpreted from the perspective of quantile-dependent expressivity. These included interactive effects of diet, alcohol, physical activity, adiposity, and smoking with genetic variants associated with the ABCA1, ADH3, ANGPTL4, APOA1, APOA4, APOA5, APOC3, APOE, CETP, CLASP1, CYP7A1, GALNT2, LDLR, LHX1, LIPC, LIPG, LPL, MVK-MMAB, PLTP, PON1, PPARα, SIRT1, SNTA1,and UCP1genes. The selected examples showed larger genetic effect sizes for lifestyle conditions associated with higher vis-à-vis lower average HDL-cholesterol concentrations. This suggests these reported interactions could be the result of selecting subjects for conditions that differentiate high from low HDL-cholesterol (e.g., lean vs. overweight, active vs. sedentary, high-fat vs. high-carbohydrate diets, alcohol drinkers vs. abstainers, nonsmokers vs. smokers) producing larger versus smaller genetic effect sizes. Key Message: Quantile-dependent expressivity provides a potential explanation for some reported gene-lifestyle interactions for HDL-cholesterol. Although overall genetic heritability appears to be quantile specific, this may vary by genetic variant and environmental exposure.
Collapse
Affiliation(s)
- Paul T Williams
- Molecular Biophysics & Integrated Bioimaging, Lawrence Berkeley National Laboratory, Berkeley, California, USA,
| |
Collapse
|
15
|
Osazuwa-Peters OL, Waken RJ, Schwander KL, Sung YJ, de Vries PS, Hartz SM, Chasman DI, Morrison AC, Bierut LJ, Xiong C, de las Fuentes L, Rao DC. Identifying blood pressure loci whose effects are modulated by multiple lifestyle exposures. Genet Epidemiol 2020; 44:629-641. [PMID: 32227373 PMCID: PMC7717887 DOI: 10.1002/gepi.22292] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/30/2019] [Accepted: 03/06/2020] [Indexed: 12/27/2022]
Abstract
Although multiple lifestyle exposures simultaneously impact blood pressure (BP) and cardiovascular health, most analysis so far has considered each single lifestyle exposure (e.g., smoking) at a time. Here, we exploit gene-multiple lifestyle exposure interactions to find novel BP loci. For each of 6,254 Framingham Heart Study participants, we computed lifestyle risk score (LRS) value by aggregating the risk of four lifestyle exposures (smoking, alcohol, education, and physical activity) on BP. Using the LRS, we performed genome-wide gene-environment interaction analysis in systolic and diastolic BP using the joint 2 degree of freedom (DF) and 1 DF interaction tests. We identified one genome-wide significant (p < 5 × 10-8 ) and 11 suggestive (p < 1 × 10-6 ) loci. Gene-environment analysis using single lifestyle exposures identified only one of the 12 loci. Nine of the 12 BP loci detected were novel. Loci detected by the LRS were located within or nearby genes with biologically plausible roles in the pathophysiology of hypertension, including KALRN, VIPR2, SNX1, and DAPK2. Our results suggest that simultaneous consideration of multiple lifestyle exposures in gene-environment interaction analysis can identify additional loci missed by single lifestyle approaches.
Collapse
Affiliation(s)
| | - R J Waken
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Karen L Schwander
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sarah M Hartz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel I Chasman
- Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics & Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Laura J Bierut
- Department of Psychiatry, Washington University School of Medicine, St. Louis, Missouri
| | - Chengjie Xiong
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Lisa de las Fuentes
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, Missouri
| | - D C Rao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
16
|
Arouca AB, Meirhaeghe A, Dallongeville J, Moreno LA, Lourenço GJ, Marcos A, Huybrechts I, Manios Y, Lambrinou CP, Gottrand F, Kafatos A, Kersting M, Sjöström M, Widhalm K, Ferrari M, Molnár D, González-Gross M, Forsner M, De Henauw S, Michels N. Interplay between the Mediterranean diet and C-reactive protein genetic polymorphisms towards inflammation in adolescents. Clin Nutr 2020; 39:1919-1926. [PMID: 31500937 DOI: 10.1016/j.clnu.2019.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 07/20/2019] [Accepted: 08/15/2019] [Indexed: 12/16/2022]
Abstract
AIM From a nutrigenetics perspective, we aim to investigate the moderating role of the Mediterranean diet and each of its subgroups in the association between C-reactive protein (CRP) gene polymorphisms and CRP blood concentration in adolescents. METHODS In 562 adolescents (13-17 y) of the European HELENA study, data was available on circulating CRP levels as inflammatory biomarker, three CRP gene SNPs (rs3093068, rs1204, rs1130864), food intake determined by a self-administered computerized 24 h-dietary recall for 2 days, and body composition. A 9-point Mediterranean diet score and each food subgroup were tested as moderator via SNP*diet interaction. Analyzes were adjusted for age, sex, puberty, adiposity and socioeconomic status. RESULTS The minor allele frequencies of rs3093068 and rs1130864 SNPs (GG and TT, respectively) were associated with higher CRP concentrations, while rs1205 (CT/TT) was associated with lower CRP concentrations. There were significant interactions between rs3093068 and Mediterranean diet (B = -0.1139, p = 0.011), or the fish food subgroup (B = -0.0090, p = 0.022), so that those with the highest genetic CRP risk underwent the highest CRP attenuation by a healthier diet. Although the effect of diet and SNP was substantial, the explained variance by interaction was only 1%. CONCLUSION Greater adherence to the Mediterranean diet and particularly its fish component was associated with a lower CRP blood concentrations especially in those at highest genetic risk due to the rs3093068 SNP.
Collapse
Affiliation(s)
- Aline B Arouca
- Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Belgium.
| | - Aline Meirhaeghe
- UMR1167, RID-AGE, Risk Factors and Molecular Determinants of Aging-Related Diseases, Centre Hosp. Univ Lille, Institut Pasteur de Lille, Université de Lille, Lille, France.
| | - Jean Dallongeville
- UMR1167, RID-AGE, Risk Factors and Molecular Determinants of Aging-Related Diseases, Centre Hosp. Univ Lille, Institut Pasteur de Lille, Université de Lille, Lille, France.
| | - Luis A Moreno
- GENUD: "Growth, Exercise, Nutrition and Development" Research Group, Facultad de Ciencias de la Salud, University of Zaragoza, Spain; Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain.
| | - Gustavo Jacob Lourenço
- Laboratory of Cancer Genetics, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil.
| | - Ascensión Marcos
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition, Madrid, Spain.
| | - Inge Huybrechts
- Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Belgium; International Agency for Research on Cancer, Lyon, France.
| | - Yannis Manios
- Department of Nutrition and Dietetics, School of Health Science & Education, Harokopio University, Athens, Greece.
| | - Christina-Paulina Lambrinou
- Department of Nutrition and Dietetics, School of Health Science & Education, Harokopio University, Athens, Greece.
| | | | | | - Mathilde Kersting
- Research Department of Child Nutrition, Pediatric University Clinic, Ruhr-University Bochum, Germany.
| | - Michael Sjöström
- Department of Biosciences, Unit for Preventive Nutrition, Karolinska Institutet, Huddinge, Sweden.
| | - Kurt Widhalm
- Department of Gastroenterology and Hepatology, Medical University of Vienna, Austria.
| | - Marika Ferrari
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Italy.
| | - Denes Molnár
- Department of Pediatrics, Medical School, University of Pécs, Pécs, Hungary.
| | - Marcela González-Gross
- ImFine Research Group, Department of Health and Human Performance, Facultad de Ciencias de la Actividad Física y del Deporte-INEF, Universidad Politécnica de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain.
| | | | - Stefaan De Henauw
- Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Belgium.
| | - Nathalie Michels
- Department of Public Health, Faculty of Medicine and Health Sciences, Ghent University, Belgium.
| |
Collapse
|
17
|
Abstract
Purpose “Quantile-dependent expressivity” describes an effect of the genotype that depends upon the level of the phenotype (e.g., whether a subject’s triglycerides are high or low relative to its population distribution). Prior analyses suggest that the effect of a genetic risk score (GRS) on fasting plasma triglyceride levels increases with the percentile of the triglyceride distribution. Postprandial lipemia is well suited for testing quantile-dependent expressivity because it exposes each individual’s genotype to substantial increases in their plasma triglyceride concentrations. Ninety-seven published papers were identified that plotted mean triglyceride response vs. time and genotype, which were converted into quantitative data. Separately, for each published graph, standard least-squares regression analysis was used to compare the genotype differences at time t (dependent variable) to average triglyceride concentrations at time t (independent variable) to assess whether the genetic effect size increased in association with higher triglyceride concentrations and whether the phenomenon could explain purported genetic interactions with sex, diet, disease, BMI, and drugs. Results Consistent with the phenomenon, genetic effect sizes increased (P≤0.05) with increasing triglyceride concentrations for polymorphisms associated with ABCA1, ANGPTL4, APOA1, APOA2, APOA4, APOA5, APOB, APOC3, APOE, CETP, FABP2, FATP6, GALNT2, GCKR, HL, IL1b, LEPR, LOX-1, LPL, MC4R, MTTP, NPY, SORT1, SULF2, TNFA, TCF7L2, and TM6SF2. The effect size for these polymorphisms showed a progressively increasing dose-response, with intermediate effect sizes at intermediate triglyceride concentrations. Quantile-dependent expressivity provided an alternative interpretation to their interactions with sex, drugs, disease, diet, and age, which have been traditionally ascribed to gene-environment interactions and genetic predictors of drug efficacy (i.e., personalized medicine). Conclusion Quantile-dependent expressivity applies to the majority of genetic variants affecting postprandial triglycerides, which may arise because the impaired functionalities of these variants increase at higher triglyceride concentrations. Purported gene-drug interactions may be the manifestations of quantile-dependent expressivity, rather than genetic predictors of drug efficacy.
Collapse
|
18
|
Hebbar P, Abu-Farha M, Mohammad A, Alkayal F, Melhem M, Abubaker J, Al-Mulla F, Thanaraj TA. FTO Variant rs1421085 Associates With Increased Body Weight, Soft Lean Mass, and Total Body Water Through Interaction With Ghrelin and Apolipoproteins in Arab Population. Front Genet 2020; 10:1411. [PMID: 32076432 PMCID: PMC7006511 DOI: 10.3389/fgene.2019.01411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/31/2019] [Indexed: 12/25/2022] Open
Abstract
Association studies have implicated single nucleotide polymorphisms (SNPs), particularly rs1421085, from the fat mass and obesity-associated (FTO) gene with body composition phenotypes, obesity, dietary intake, and physical activity in European, East Asian, and African populations. However, the impact of the rs1421085 variant has not been sufficiently tested in ethnic populations (such as Arabs) with high levels of obesity. Further, there is a lack of studies identifying biomarkers that interact with FTO. Therefore, we investigated the association of rs1421085 with obesity and body composition traits and metabolic biomarkers in Arab population. We genotyped rs1421085 SNP in 278 Arab individuals, where multiple biomarkers relating to obesity, inflammation, and other metabolic pathways were quantified. We performed genetic association tests under additive mode of inheritance using linear regression models and found association of rs1421085_C allele with higher levels of body weight, soft lean mass (SLM), and total body water. Examination (using linear regression models under dominant mode of inheritance) of correlation among biomarkers and interaction with genotypes at the variant revealed that measures of these three body composition traits were found mediated by interaction between carrier genotypes (TC+CC) and measures of ghrelin, ApoA1, and ApoB48. Lean body mass (LBM), to which SLM contributes, is an important determinant of physical strength and is a focal point in studies on sarcopenia. Low LBM is known to be associated with higher risk of cardiometabolic disorders. Thus, the finding on the FTO variant as a genetic determinant of SLM via interaction with ghrelin, ApoA1, and ApoB48 is important.
Collapse
Affiliation(s)
| | | | - Anwar Mohammad
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fadi Alkayal
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
| | - Motasem Melhem
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
| | - Jehad Abubaker
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
| | - Fahd Al-Mulla
- Research Division, Dasman Diabetes Institute, Dasman, Kuwait
| | | |
Collapse
|
19
|
Zhou B, Ichikawa R, Parnell LD, Noel SE, Zhang X, Bhupathiraju SN, Smith CE, Tucker KL, Ordovas JM, Lai CQ. Metabolomic Links between Sugar-Sweetened Beverage Intake and Obesity. J Obes 2020; 2020:7154738. [PMID: 32399287 PMCID: PMC7211252 DOI: 10.1155/2020/7154738] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Sugar-sweetened beverage (SSB) consumption is highly associated with obesity, but the metabolic mechanism underlying this correlation is not understood. OBJECTIVE Our objective was to examine metabolomic links between SSB intake and obesity to understand metabolic mechanisms. DESIGN We examined the association of plasma metabolomic profiles with SSB intake and obesity risk in 781 participants, aged 45-75 y, in the Boston Puerto Rican Health Study (BPRHS) using generalized linear models, controlling for potential confounding factors. Based on identified metabolites, we conducted pathway enrichment analysis to identify potential metabolic pathways that link SSB intake and obesity risk. Variants in genes encoding enzymes known to function in identified metabolic pathways were examined for their interactions with SSB intake on obesity. RESULTS SSB intake was correlated with BMI (β = 0.607, P=0.045). Among 526 measured metabolites, 86 showed a significant correlation with SSB intake and 148 with BMI (P ≤ 0.05); 28 were correlated with both SSB intake and BMI (P ≤ 0.05). Pathway enrichment analysis identified the phosphatidylcholine and lysophospholipid pathways as linking SSB intake to obesity, after correction for multiple testing. Furthermore, 8 of 10 genes functioning in these two pathways showed strong interaction with SSB intake on BMI. Our results further identified participants who may exhibit an increased risk of obesity when consuming SSB. CONCLUSIONS We identified two key metabolic pathways that link SSB intake to obesity, revealing the potential of phosphatidylcholine and lysophospholipid to modulate how SSB intake can increase obesity risk. The interaction between genetic variants related to these pathways and SSB intake on obesity further supports the mechanism.
Collapse
Affiliation(s)
- Bingjie Zhou
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Reiko Ichikawa
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
- Institute for Innovation, Ajinomoto Co., Inc., Kawasaki, Japan
| | - Laurence D. Parnell
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Sabrina E. Noel
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Xiyuan Zhang
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Shilpa N. Bhupathiraju
- Channing Division of Network Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Caren E. Smith
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Katherine L. Tucker
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA, USA
| | - Jose M. Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
- IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Chao-Qiang Lai
- USDA Agricultural Research Service, Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
20
|
Norde MM, Fisberg RM, Marchioni DML, Rogero MM. Systemic low-grade inflammation-associated lifestyle, diet, and genetic factors: A population-based cross-sectional study. Nutrition 2019; 70:110596. [PMID: 31743813 DOI: 10.1016/j.nut.2019.110596] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/24/2019] [Accepted: 08/25/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVES Systemic low-grade inflammation (SLGI) is an intermediary common condition to the physiopathology of chronic noncommunicable diseases and targeting its determinants could lead to more efficient public health strategies. We aimed to investigate SLGI-independent associations with lifestyle, diet, and genetic factors in a population-based sample of adults using a systemic low-grade inflammation score (SIS). METHODS The study sample is composed of 269 participants from the cross-sectional population-based Health Survey of Sao Paulo (2008-2010), ages 20 to 59 y, whose data on socioeconomic variables, lifestyle, health parameters, and blood samples were available. Diet was assessed by two 24-h recalls, and the Brazilian Health Eating Index-Revised (BHEI-R) was scored. From blood samples, 30 single nucleotide polymorphisms on inflammatory genes were genotyped, and plasma eleven inflammatory biomarkers levels that composed the SIS were determined. A multiple, stepwise, linear regression was used to investigate SIS-independent associated factors. RESULTS Factors independently associated with SIS were BHEI-R score (partial R² = 5.1; β = -0.13; P = 0.003), body mass index (partial R² = 3.4; β = 0.19; P = 0.001), TLR4 rs5030728 GA + AA genotype (partial R² = 3.1; β = -1.37; P = 0.008), age 50 to 59 y (partial R² = 2.5; β = 1.93; P = 0.029) in comparison with the reference category (20 to 29 y), and commuting physical activity >150 min/wk (partial R² = 2.2; β = -1.29; P = 0.043) after adjustment for current smoking status, medication use, and dietary misreporting. CONCLUSIONS Eating a lower quality diet, having a higher body mass index score and age, being GG homozygous for TLR4 rs5030728, and spending <150 min/wk in transportation physical activity are independent determinants of SLGI.
Collapse
Affiliation(s)
- Marina M Norde
- Department of Nutrition, School of Public Health, University of Sao Paulo, Sao Paulo City, SP, Brazil
| | - Regina M Fisberg
- Department of Nutrition, School of Public Health, University of Sao Paulo, Sao Paulo City, SP, Brazil
| | - Dirce M L Marchioni
- Department of Nutrition, School of Public Health, University of Sao Paulo, Sao Paulo City, SP, Brazil
| | - Marcelo Macedo Rogero
- Department of Nutrition, School of Public Health, University of Sao Paulo, Sao Paulo City, SP, Brazil.
| |
Collapse
|
21
|
Taylor JY, Ware EB, Wright ML, Smith JA, Kardia SLR. Using Genetic Burden Scores for Gene-by-Methylation Interaction Analysis on Metabolic Syndrome in African Americans. Biol Res Nurs 2019; 21:279-285. [PMID: 30781968 PMCID: PMC6700897 DOI: 10.1177/1099800419828486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
With the rapid advancement of omics-based research, particularly big data such as genome- and epigenome-wide association studies that include extensive environmental and clinical variables, data analytics have become increasingly complex. Researchers face significant challenges regarding how to analyze multifactorial data and make use of the findings for clinical translation. The purpose of this article is to provide a scientific exemplar for use of genetic burden scores as a data analysis method for studies with both genotype and DNA methylation data in which the goal is to evaluate associations with chronic conditions such as metabolic syndrome (MetS). This study included 739 African American men and women from the Genetic Epidemiology Network of Arteriopathy Study who met diagnostic criteria for MetS and had available genetic and epigenetic data. Genetic burden scores for evaluated genes were not significant after multiple testing corrections, but DNA methylation at 2 CpG sites (dihydroorotate dehydrogenase cg22381196 pFDR = .014; CTNNA3 cg00132141 pFDR = .043) was significantly associated with MetS after controlling for multiple comparisons. Interactions between the marginally significant CpG sites and burden scores, however, were not significant. More work is required in this area to identify intermediate biological pathways influenced by environmental, genetic, and epigenetic variation that may explain the high prevalence of MetS among African Americans. This study does serve, however, as an example of the use of the genetic burden score as an alternative data analysis approach for complex studies involving the analysis of genetic and epigenetic data simultaneously.
Collapse
Affiliation(s)
| | - Erin B. Ware
- Institute for Social Research, University of Michigan, Ann Arbor, MI,
USA
| | | | - Jennifer A. Smith
- School of Public Health and Institute for Social Research, University of
Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
22
|
Hebbar P, Nizam R, Melhem M, Alkayal F, Elkum N, John SE, Tuomilehto J, Alsmadi O, Thanaraj TA. Genome-wide association study identifies novel recessive genetic variants for high TGs in an Arab population. J Lipid Res 2018; 59:1951-1966. [PMID: 30108155 DOI: 10.1194/jlr.p080218] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Abnormal blood lipid levels are influenced by genetic and lifestyle/dietary factors. Although many genetic variants associated with blood lipid traits have been identified in Europeans, similar data in Middle Eastern populations are limited. We performed a genome-wide association study with Arab individuals (discovery cohort: 1,353; replication cohort: 1,176) from Kuwait to identify possible associations of genetic variants with high lipid levels. We used Illumina HumanOmniExpress BeadChip and candidate SNP genotyping in the discovery and replication phases, respectively. For association tests, we used genetic models that were based on additive and recessive modes of inheritance. High triglycerides (TGs) were recessively associated with six risk variants (rs1002487/RPS6KA1, rs11805972/LAD1) rs7761746/Or5v1, rs39745/CTTNBP2-LSM8, rs2934952/PGAP3, and rs9626773/RP11-191L9.4-CERK) at genome-wide significance (P 6.12E-09), and another six variants (rs10873925/ST6GALNAC5, rs4663379/SPP2-ARL4C, rs10033119/NPY1R, rs17709449/LINC00911-FLRT2, rs11654954/CDK12-NEUROD2, and rs9972882/STARD3) were associated at borderline significance (P 5.0E-08). High TG was also additively associated with rs11654954. All of the 12 identified markers are novel and are harbored in runs of homozygosity. Literature evidence supports the involvement of these gene loci in lipid-related processes. This study in an Arab population augments international efforts to identify genetic regulation of lipid traits.
Collapse
Affiliation(s)
- Prashantha Hebbar
- Dasman Diabetes Institute, Dasman 15462, Kuwait.,Faculty of Medicine, Univerisity of Helsinki, Helsinki, Finland
| | | | | | | | - Naser Elkum
- Dasman Diabetes Institute, Dasman 15462, Kuwait
| | | | | | | | | |
Collapse
|
23
|
Genetic and Epigenetic Regulations of Post-prandial Lipemia. CURRENT GENETIC MEDICINE REPORTS 2018. [DOI: 10.1007/s40142-018-0146-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
24
|
Wang DD, Hu FB. Precision nutrition for prevention and management of type 2 diabetes. Lancet Diabetes Endocrinol 2018; 6:416-426. [PMID: 29433995 DOI: 10.1016/s2213-8587(18)30037-8] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 12/02/2017] [Accepted: 12/11/2017] [Indexed: 02/08/2023]
Abstract
Precision nutrition aims to prevent and manage chronic diseases by tailoring dietary interventions or recommendations to one or a combination of an individual's genetic background, metabolic profile, and environmental exposures. Recent advances in genomics, metabolomics, and gut microbiome technologies have offered opportunities as well as challenges in the use of precision nutrition to prevent and manage type 2 diabetes. Nutrigenomics studies have identified genetic variants that influence intake and metabolism of specific nutrients and predict individuals' variability in response to dietary interventions. Metabolomics has revealed metabolomic fingerprints of food and nutrient consumption and uncovered new metabolic pathways that are potentially modified by diet. Dietary interventions have been successful in altering abundance, composition, and activity of gut microbiota that are relevant for food metabolism and glycaemic control. In addition, mobile apps and wearable devices facilitate real-time assessment of dietary intake and provide feedback which can improve glycaemic control and diabetes management. By integrating these technologies with big data analytics, precision nutrition has the potential to provide personalised nutrition guidance for more effective prevention and management of type 2 diabetes. Despite these technological advances, much research is needed before precision nutrition can be widely used in clinical and public health settings. Currently, the field of precision nutrition faces challenges including a lack of robust and reproducible results, the high cost of omics technologies, and methodological issues in study design as well as high-dimensional data analyses and interpretation. Evidence is needed to support the efficacy, cost-effectiveness, and additional benefits of precision nutrition beyond traditional nutrition intervention approaches. Therefore, we should manage unrealistically high expectations and balance the emerging field of precision nutrition with public health nutrition strategies to improve diet quality and prevent type 2 diabetes and its complications.
Collapse
Affiliation(s)
- Dong D Wang
- Department of Nutrition, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Harvard T H Chan School of Public Health, and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Cirillo E, Kutmon M, Gonzalez Hernandez M, Hooimeijer T, Adriaens ME, Eijssen LMT, Parnell LD, Coort SL, Evelo CT. From SNPs to pathways: Biological interpretation of type 2 diabetes (T2DM) genome wide association study (GWAS) results. PLoS One 2018; 13:e0193515. [PMID: 29617380 PMCID: PMC5884486 DOI: 10.1371/journal.pone.0193515] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/13/2018] [Indexed: 12/16/2022] Open
Abstract
Genome-wide association studies (GWAS) have become a common method for discovery of gene-disease relationships, in particular for complex diseases like Type 2 Diabetes Mellitus (T2DM). The experience with GWAS analysis has revealed that the genetic risk for complex diseases involves cumulative, small effects of many genes and only some genes with a moderate effect. In order to explore the complexity of the relationships between T2DM genes and their potential function at the process level as effected by polymorphism effects, a secondary analysis of a GWAS meta-analysis is presented. Network analysis, pathway information and integration of different types of biological information such as eQTLs and gene-environment interactions are used to elucidate the biological context of the genetic variants and to perform an analysis based on data visualization. We selected a T2DM dataset from a GWAS meta-analysis, and extracted 1,971 SNPs associated with T2DM. We mapped 580 SNPs to 360 genes, and then selected 460 pathways containing these genes from the curated collection of WikiPathways. We then created and analyzed SNP-gene and SNP-gene-pathway network modules in Cytoscape. A focus on genes with robust connections to pathways permitted identification of many T2DM pertinent pathways. However, numerous genes lack literature evidence of association with T2DM. We also speculate on the genes in specific network structures obtained in the SNP-gene network, such as gene-SNP-gene modules. Finally, we selected genes relevant to T2DM from our SNP-gene-pathway network, using different sources that reveal gene-environment interactions and eQTLs. We confirmed functions relevant to T2DM for many genes and have identified some-LPL and APOB-that require further validation to clarify their involvement in T2DM.
Collapse
Affiliation(s)
- Elisa Cirillo
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Martina Kutmon
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Manuel Gonzalez Hernandez
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Tom Hooimeijer
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Michiel E. Adriaens
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| | - Lars M. T. Eijssen
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Laurence D. Parnell
- Agricultural Research Service, USDA, Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, United States of America
| | - Susan L. Coort
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Chris T. Evelo
- Department of Bioinformatics – BiGCaT, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
26
|
Pan A, Lin X, Hemler E, Hu FB. Diet and Cardiovascular Disease: Advances and Challenges in Population-Based Studies. Cell Metab 2018; 27:489-496. [PMID: 29514062 PMCID: PMC5844273 DOI: 10.1016/j.cmet.2018.02.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/16/2018] [Accepted: 02/16/2018] [Indexed: 12/12/2022]
Abstract
In this Minireview, we provide an epidemiologist's perspective on the debate and recent advances in determining the relationship between diet and cardiovascular health. We conclude that, in order to reduce the global burden of cardiovascular disease, there should be a greater emphasis on improving overall diet quality and food sources of macronutrients, such as dietary fats and carbohydrates. In addition, building a strong evidence base through high-quality intervention and observational studies is crucial for effective policy changes, which can greatly improve the food environment and population health.
Collapse
Affiliation(s)
- An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health and State Key Laboratory of Environmental Health (incubation), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China.
| | - Xu Lin
- Key Laboratory of Nutrition and Metabolism, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Elena Hemler
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW Postprandial lipemia (PPL), the prolonged increase in plasma triglyceride-rich lipoproteins following food consumption, is an independent risk factor for cardiovascular disease. Genetic variation, environment and the interplay between these direct an individual's postprandial lipid response. From such interplay, inducible and reversible epigenetic changes arise. Increasing evidence suggests epigenetic variation contributes to postprandial response in lipids and risk. RECENT FINDINGS Diet and exercise are central agents affecting postprandial lipemia - triglyceride, but heterogeneity of the findings warrant more and larger studies. Several epigenetic loci identified from a human intervention study account for a substantial proportion of PPL phenotype variation, but the burden to conduct an intervention study of postprandial responses likely limits translation to personalized nutrition. SUMMARY The impact of both DNA methylation patterns and environmental factors such as diet, exercise, sleep and medication on PPL is multifaceted. Discovery of interactions that modify the association between CpG (oligodeoxydinucleotide) methylation and postprandial phenotypes is unfolding.
Collapse
Affiliation(s)
| | - Jose M Ordovas
- Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
- IMDEA Food Institute, CEI UAM + CSIC
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | |
Collapse
|
28
|
Rao DC, Sung YJ, Winkler TW, Schwander K, Borecki I, Cupples LA, Gauderman WJ, Rice K, Munroe PB, Psaty BM. Multiancestry Study of Gene-Lifestyle Interactions for Cardiovascular Traits in 610 475 Individuals From 124 Cohorts: Design and Rationale. ACTA ACUST UNITED AC 2018; 10:CIRCGENETICS.116.001649. [PMID: 28620071 DOI: 10.1161/circgenetics.116.001649] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/14/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Several consortia have pursued genome-wide association studies for identifying novel genetic loci for blood pressure, lipids, hypertension, etc. They demonstrated the power of collaborative research through meta-analysis of study-specific results. METHODS AND RESULTS The Gene-Lifestyle Interactions Working Group was formed to facilitate the first large, concerted, multiancestry study to systematically evaluate gene-lifestyle interactions. In stage 1, genome-wide interaction analysis is performed in 53 cohorts with a total of 149 684 individuals from multiple ancestries. In stage 2 involving an additional 71 cohorts with 460 791 individuals from multiple ancestries, focused analysis is performed for a subset of the most promising variants from stage 1. In all, the study involves up to 610 475 individuals. Current focus is on cardiovascular traits including blood pressure and lipids, and lifestyle factors including smoking, alcohol, education (as a surrogate for socioeconomic status), physical activity, psychosocial variables, and sleep. The total sample sizes vary among projects because of missing data. Large-scale gene-lifestyle or more generally gene-environment interaction (G×E) meta-analysis studies can be cumbersome and challenging. This article describes the design and some of the approaches pursued in the interaction projects. CONCLUSIONS The Gene-Lifestyle Interactions Working Group provides an excellent framework for understanding the lifestyle context of genetic effects and to identify novel trait loci through analysis of interactions. An important and novel feature of our study is that the gene-lifestyle interaction (G×E) results may improve our knowledge about the underlying mechanisms for novel and already known trait loci.
Collapse
|
29
|
Gene-by-Psychosocial Factor Interactions Influence Diastolic Blood Pressure in European and African Ancestry Populations: Meta-Analysis of Four Cohort Studies. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2017; 14:ijerph14121596. [PMID: 29258278 PMCID: PMC5751013 DOI: 10.3390/ijerph14121596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/04/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
Inter-individual variability in blood pressure (BP) is influenced by both genetic and non-genetic factors including socioeconomic and psychosocial stressors. A deeper understanding of the gene-by-socioeconomic/psychosocial factor interactions on BP may help to identify individuals that are genetically susceptible to high BP in specific social contexts. In this study, we used a genomic region-based method for longitudinal analysis, Longitudinal Gene-Environment-Wide Interaction Studies (LGEWIS), to evaluate the effects of interactions between known socioeconomic/psychosocial and genetic risk factors on systolic and diastolic BP in four large epidemiologic cohorts of European and/or African ancestry. After correction for multiple testing, two interactions were significantly associated with diastolic BP. In European ancestry participants, outward/trait anger score had a significant interaction with the C10orf107 genomic region (p = 0.0019). In African ancestry participants, depressive symptom score had a significant interaction with the HFE genomic region (p = 0.0048). This study provides a foundation for using genomic region-based longitudinal analysis to identify subgroups of the population that may be at greater risk of elevated BP due to the combined influence of genetic and socioeconomic/psychosocial risk factors.
Collapse
|
30
|
Grimaldi KA, van Ommen B, Ordovas JM, Parnell LD, Mathers JC, Bendik I, Brennan L, Celis-Morales C, Cirillo E, Daniel H, de Kok B, El-Sohemy A, Fairweather-Tait SJ, Fallaize R, Fenech M, Ferguson LR, Gibney ER, Gibney M, Gjelstad IMF, Kaput J, Karlsen AS, Kolossa S, Lovegrove J, Macready AL, Marsaux CFM, Alfredo Martinez J, Milagro F, Navas-Carretero S, Roche HM, Saris WHM, Traczyk I, van Kranen H, Verschuren L, Virgili F, Weber P, Bouwman J. Proposed guidelines to evaluate scientific validity and evidence for genotype-based dietary advice. GENES & NUTRITION 2017; 12:35. [PMID: 29270237 PMCID: PMC5732517 DOI: 10.1186/s12263-017-0584-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 10/09/2017] [Indexed: 12/13/2022]
Abstract
Nutrigenetic research examines the effects of inter-individual differences in genotype on responses to nutrients and other food components, in the context of health and of nutrient requirements. A practical application of nutrigenetics is the use of personal genetic information to guide recommendations for dietary choices that are more efficacious at the individual or genetic subgroup level relative to generic dietary advice. Nutrigenetics is unregulated, with no defined standards, beyond some commercially adopted codes of practice. Only a few official nutrition-related professional bodies have embraced the subject, and, consequently, there is a lack of educational resources or guidance for implementation of the outcomes of nutrigenetic research. To avoid misuse and to protect the public, personalised nutrigenetic advice and information should be based on clear evidence of validity grounded in a careful and defensible interpretation of outcomes from nutrigenetic research studies. Evidence requirements are clearly stated and assessed within the context of state-of-the-art 'evidence-based nutrition'. We have developed and present here a draft framework that can be used to assess the strength of the evidence for scientific validity of nutrigenetic knowledge and whether 'actionable'. In addition, we propose that this framework be used as the basis for developing transparent and scientifically sound advice to the public based on nutrigenetic tests. We feel that although this area is still in its infancy, minimal guidelines are required. Though these guidelines are based on semi-quantitative data, they should stimulate debate on their utility. This framework will be revised biennially, as knowledge on the subject increases.
Collapse
Affiliation(s)
| | | | - Jose M. Ordovas
- JMUSDA-Human Nutrition Research Center on Aging at Tufts University, Boston, USA
- IMDEA Alimentacion, Madrid, Spain
| | - Laurence D. Parnell
- Agriculture Research Service, USDA, Human Nutrition Research Center on Aging, Boston, MA 02111 USA
| | - John C. Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
| | - Igor Bendik
- DSM Nutritional Products, Kaiseraugst, Switzerland
| | - Lorraine Brennan
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin, Republic of Ireland
| | - Carlos Celis-Morales
- Human Nutrition Research Centre, Institute of Cellular Medicine, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL UK
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Science, University of Glasgow, Glasgow, G12 8TA UK
| | | | - Hannelore Daniel
- Nutritional Physiology, Technische Universität München, 85350 Freising, Germany
| | | | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, 150 College Street, 3rd Floor, Toronto, ON M5S 3E2 Canada
| | | | - Rosalind Fallaize
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Sciences, University of Reading, Whiteknights, PO Box 226, Reading, Berkshire RG6 6AP UK
| | - Michael Fenech
- CSIRO Health and Biosecurity, Gate 13, Kintore Avenue, Adelaide, SA 5000 Australia
| | - Lynnette R. Ferguson
- ACSRC and Discipline of Nutrition and Dietetics, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1184 New Zealand
| | - Eileen R. Gibney
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin, Republic of Ireland
| | - Mike Gibney
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin, Republic of Ireland
| | - Ingrid M. F. Gjelstad
- Department of Nutrition, Universitetet i Oslo, PO Box 1046, Blindern, N-0316 Oslo, Norway
| | - Jim Kaput
- Vydiant Inc, 2330 Gold Meadow Way, Gold River, 95670 CA USA
| | - Anette S. Karlsen
- Department of Nutrition, Universitetet i Oslo, PO Box 1046, Blindern, N-0316 Oslo, Norway
| | - Silvia Kolossa
- Nutritional Physiology, Technische Universität München, 85350 Freising, Germany
| | - Julie Lovegrove
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Sciences, University of Reading, Whiteknights, PO Box 226, Reading, Berkshire RG6 6AP UK
| | - Anna L. Macready
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, Department of Food and Nutritional Sciences, University of Reading, Whiteknights, PO Box 226, Reading, Berkshire RG6 6AP UK
| | - Cyril F. M. Marsaux
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre + (MUMC+), Maastricht, The Netherlands
| | - J. Alfredo Martinez
- IMDEA Alimentacion, Madrid, Spain
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Fermin Milagro
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Santiago Navas-Carretero
- Department of Nutrition, Food Science and Physiology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Helen M. Roche
- Nutrigenomics Research Group, UCD Institute of Food and Health/UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Wim H. M. Saris
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre + (MUMC+), Maastricht, The Netherlands
| | - Iwona Traczyk
- Department of Human Nutrition, Faculty on Health Sciences, Medical University of Warsaw, Warsaw, Poland
| | - Henk van Kranen
- Institute for Public Health Genomics (IPHG), Department of Genetics and Cell Biology, Faculty of Health, Medicine & Life Sciences, University of Maastricht, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands
| | | | - Fabio Virgili
- Council for Agricultural Research and Economics, Food and Nutrition Research Centre, (CREA - AN), via Ardeatina 546, 00178 Rome, Italy
| | - Peter Weber
- DSM Nutritional Products, Kaiseraugst, Switzerland
| | | |
Collapse
|
31
|
Cirillo E, Parnell LD, Evelo CT. A Review of Pathway-Based Analysis Tools That Visualize Genetic Variants. Front Genet 2017; 8:174. [PMID: 29163640 PMCID: PMC5681904 DOI: 10.3389/fgene.2017.00174] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/24/2017] [Indexed: 01/04/2023] Open
Abstract
Pathway analysis is a powerful method for data analysis in genomics, most often applied to gene expression analysis. It is also promising for single-nucleotide polymorphism (SNP) data analysis, such as genome-wide association study data, because it allows the interpretation of variants with respect to the biological processes in which the affected genes and proteins are involved. Such analyses support an interactive evaluation of the possible effects of variations on function, regulation or interaction of gene products. Current pathway analysis software often does not support data visualization of variants in pathways as an alternate method to interpret genetic association results, and specific statistical methods for pathway analysis of SNP data are not combined with these visualization features. In this review, we first describe the visualization options of the tools that were identified by a literature review, in order to provide insight for improvements in this developing field. Tool evaluation was performed using a computational epistatic dataset of gene–gene interactions for obesity risk. Next, we report the necessity to include in these tools statistical methods designed for the pathway-based analysis with SNP data, expressly aiming to define features for more comprehensive pathway-based analysis tools. We conclude by recognizing that pathway analysis of genetic variations data requires a sophisticated combination of the most useful and informative visual aspects of the various tools evaluated.
Collapse
Affiliation(s)
- Elisa Cirillo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, Netherlands
| | - Laurence D Parnell
- Jean Mayer-USDA Human Nutrition Research Center on Aging at Tufts University, Agricultural Research Service, USDA, Boston, MA, United States
| | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
32
|
Mediterranean Diet Adherence and Genetic Background Roles within a Web-Based Nutritional Intervention: The Food4Me Study. Nutrients 2017; 9:nu9101107. [PMID: 29019927 PMCID: PMC5691723 DOI: 10.3390/nu9101107] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/04/2017] [Accepted: 10/09/2017] [Indexed: 01/02/2023] Open
Abstract
Mediterranean Diet (MedDiet) adherence has been proven to produce numerous health benefits. In addition, nutrigenetic studies have explained some individual variations in the response to specific dietary patterns. The present research aimed to explore associations and potential interactions between MedDiet adherence and genetic background throughout the Food4Me web-based nutritional intervention. Dietary, anthropometrical and biochemical data from volunteers of the Food4Me study were collected at baseline and after 6 months. Several genetic variants related to metabolic risk features were also analysed. A Genetic Risk Score (GRS) was derived from risk alleles and a Mediterranean Diet Score (MDS), based on validated food intake data, was estimated. At baseline, there were no interactions between GRS and MDS categories for metabolic traits. Linear mixed model repeated measures analyses showed a significantly greater decrease in total cholesterol in participants with a low GRS after a 6-month period, compared to those with a high GRS. Meanwhile, a high baseline MDS was associated with greater decreases in Body Mass Index (BMI), waist circumference and glucose. There also was a significant interaction between GRS and the MedDiet after the follow-up period. Among subjects with a high GRS, those with a high MDS evidenced a highly significant reduction in total carotenoids, while among those with a low GRS, there was no difference associated with MDS levels. These results suggest that a higher MedDiet adherence induces beneficial effects on metabolic outcomes, which can be affected by the genetic background in some specific markers.
Collapse
|
33
|
Ritchie MD, Davis JR, Aschard H, Battle A, Conti D, Du M, Eskin E, Fallin MD, Hsu L, Kraft P, Moore JH, Pierce BL, Bien SA, Thomas DC, Wei P, Montgomery SB. Incorporation of Biological Knowledge Into the Study of Gene-Environment Interactions. Am J Epidemiol 2017; 186:771-777. [PMID: 28978191 PMCID: PMC5860556 DOI: 10.1093/aje/kwx229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022] Open
Abstract
A growing knowledge base of genetic and environmental information has greatly enabled the study of disease risk factors. However, the computational complexity and statistical burden of testing all variants by all environments has required novel study designs and hypothesis-driven approaches. We discuss how incorporating biological knowledge from model organisms, functional genomics, and integrative approaches can empower the discovery of novel gene-environment interactions and discuss specific methodological considerations with each approach. We consider specific examples where the application of these approaches has uncovered effects of gene-environment interactions relevant to drug response and immunity, and we highlight how such improvements enable a greater understanding of the pathogenesis of disease and the realization of precision medicine.
Collapse
Affiliation(s)
- Marylyn D. Ritchie
- Correspondence to Dr. Stephen B. Montgomery, Departments of Genetics and Pathology, Stanford University School of Medicine, Stanford, CA 94305 (e-mail: ); or Dr. Marylyn D. Ritchie, Geisinger Health System, 205 Hood Center for Health Research, Center Street, Danville, PA 17821(e-mail: )
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Stephen B. Montgomery
- Correspondence to Dr. Stephen B. Montgomery, Departments of Genetics and Pathology, Stanford University School of Medicine, Stanford, CA 94305 (e-mail: ); or Dr. Marylyn D. Ritchie, Geisinger Health System, 205 Hood Center for Health Research, Center Street, Danville, PA 17821(e-mail: )
| |
Collapse
|
34
|
Kim G, Lai CQ, Arnett DK, Parnell LD, Ordovas JM, Kim Y, Kim J. Detection of gene-environment interactions in a family-based population using SCAD. Stat Med 2017; 36:3547-3559. [PMID: 28707299 DOI: 10.1002/sim.7382] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 05/19/2017] [Accepted: 06/02/2017] [Indexed: 11/07/2022]
Abstract
Gene-environment interaction (GxE) is emphasized as one potential source of missing genetic variation on disease traits, and the ultimate goal of GxE research is prediction of individual risk and prevention of complex diseases. However, there are various challenges in statistical analysis of GxE. In this paper, we focus on the three methodological challenges: (i) the high dimensions of genes; (ii) the hierarchical structure between interaction effects and their corresponding main effects; and (iii) the correlation among subjects from family-based population studies. In this paper, we propose an algorithm that approaches all three challenges simultaneously. This is the first penalized method focusing on an interaction search based on a linear mixed effect model. For verification, we compare the empirical performance of our new method with other existing methods in simulation study. The results demonstrate the superiority of our method under overall simulation setup. In particular, the outperformance obviously becomes greater as the correlation among subjects increases. In addition, the new method provides a robust estimate for the correlation among subjects. We also apply the new method on Genetics of Lipid Lowering Drugs and Diet Network study data. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Gwangsu Kim
- Data Science for Knowledge Creation Research Center, Seoul National University, Seoul, Korea
| | - Chao-Qiang Lai
- Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, U.S.A
| | - Donna K Arnett
- University of Kentucky College of Public Health, Lexington, KY, U.S.A
| | - Laurence D Parnell
- Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, U.S.A
| | - Jose M Ordovas
- Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, U.S.A.,Department of Epidemiology and Population Genetics, Centro Nacional Investigacion Cardiovasculares (CNIC) Madrid, Madrid, Spain
| | - Yongdai Kim
- Department of Statistics, Seoul National University, Seoul, Korea
| | - Joungyoun Kim
- Department of Information Statistics, Chungbuk National University, Cheongju, Chungbuk, Korea
| |
Collapse
|
35
|
van Ommen B, van den Broek T, de Hoogh I, van Erk M, van Someren E, Rouhani-Rankouhi T, Anthony JC, Hogenelst K, Pasman W, Boorsma A, Wopereis S. Systems biology of personalized nutrition. Nutr Rev 2017; 75:579-599. [PMID: 28969366 PMCID: PMC5914356 DOI: 10.1093/nutrit/nux029] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Personalized nutrition is fast becoming a reality due to a number of technological, scientific, and societal developments that complement and extend current public health nutrition recommendations. Personalized nutrition tailors dietary recommendations to specific biological requirements on the basis of a person's health status and goals. The biology underpinning these recommendations is complex, and thus any recommendations must account for multiple biological processes and subprocesses occurring in various tissues and must be formed with an appreciation for how these processes interact with dietary nutrients and environmental factors. Therefore, a systems biology-based approach that considers the most relevant interacting biological mechanisms is necessary to formulate the best recommendations to help people meet their wellness goals. Here, the concept of "systems flexibility" is introduced to personalized nutrition biology. Systems flexibility allows the real-time evaluation of metabolism and other processes that maintain homeostasis following an environmental challenge, thereby enabling the formulation of personalized recommendations. Examples in the area of macro- and micronutrients are reviewed. Genetic variations and performance goals are integrated into this systems approach to provide a strategy for a balanced evaluation and an introduction to personalized nutrition. Finally, modeling approaches that combine personalized diagnosis and nutritional intervention into practice are reviewed.
Collapse
Affiliation(s)
- Ben van Ommen
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Tim van den Broek
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Iris de Hoogh
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Marjan van Erk
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Eugene van Someren
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Tanja Rouhani-Rankouhi
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | | | - Koen Hogenelst
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Wilrike Pasman
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - André Boorsma
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| | - Suzan Wopereis
- TNO (The Netherlands Organization for Applied Scientific Research), Zeist, the Netherlands
| |
Collapse
|
36
|
Celis-Morales CA, Lyall DM, Gray SR, Steell L, Anderson J, Iliodromiti S, Welsh P, Guo Y, Petermann F, Mackay DF, Bailey MES, Pell JP, Gill JMR, Sattar N. Dietary fat and total energy intake modifies the association of genetic profile risk score on obesity: evidence from 48 170 UK Biobank participants. Int J Obes (Lond) 2017; 41:1761-1768. [PMID: 28736445 DOI: 10.1038/ijo.2017.169] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/19/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND Obesity is a multifactorial condition influenced by both genetics and lifestyle. The aim of this study was to investigate whether the association between a validated genetic profile risk score for obesity (GPRS-obesity) and body mass index (BMI) or waist circumference (WC) was modified by macronutrient intake in a large general population study. METHODS This study included cross-sectional data from 48 170 white European adults, aged 37-73 years, participating in the UK Biobank. Interactions between GPRS-obesity and macronutrient intake (including total energy, protein, fat, carbohydrate and dietary fibre intake) and its effects on BMI and WC were investigated. RESULTS The 93-single-nucleotide polymorphism (SNP) GPRS was associated with a higher BMI (β: 0.57 kg m-2 per s.d. increase in GPRS (95% confidence interval: 0.53-0.60); P=1.9 × 10-183) independent of major confounding factors. There was a significant interaction between GPRS and total fat intake (P(interaction)=0.007). Among high-fat-intake individuals, BMI was higher by 0.60 (0.52, 0.67) kg m-2 per s.d. increase in GPRS-obesity; the change in BMI with GPRS was lower among low-fat-intake individuals (β: 0.50 (0.44, 0.57) kg m-2). Significant interactions with similar patterns were observed for saturated fat intake (high β: 0.66 (0.59, 0.73) versus low β: 0.49 (0.42, 0.55) kg m-2, P(interaction)=2 × 10-4) and for total energy intake (high β: 0.58 (0.51, 0.64) versus low β: 0.49 (0.42, 0.56) kg m-2, P(interaction)=0.019), but not for protein intake, carbohydrate intake and fibre intake (P(interaction) >0.05). The findings were broadly similar using WC as the outcome. CONCLUSIONS These data suggest that the benefits of reducing the intake of fats and total energy intake may be more important in individuals with high genetic risk for obesity.
Collapse
Affiliation(s)
- C A Celis-Morales
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - D M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - S R Gray
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - L Steell
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - J Anderson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - S Iliodromiti
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - P Welsh
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Y Guo
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - F Petermann
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - D F Mackay
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - M E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - J P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - J M R Gill
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - N Sattar
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
Okhovat M, Chen IC, Dehghani Z, Zheng DJ, Ikpatt JE, Momoh H, Phelps SM. Genetic variation in the developmental regulation of cortical avpr1a among prairie voles. GENES BRAIN AND BEHAVIOR 2017; 17:36-48. [PMID: 28589689 DOI: 10.1111/gbb.12396] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 04/15/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
Early experiences can have enduring impacts on brain and behavior, but the strength of these effects can be influenced by genetic variation. In principle, polymorphic CpGs (polyCpGs) may contribute to gene-by-environment interactions (G × E) by altering DNA methylation. In this study, we investigate the influence of polyCpGs on the development of vasopressin receptor 1a abundance in the retrosplenial cortex (RSC-V1aR) of prairie voles (Microtus ochrogaster). Two alternative alleles ('HI'/'LO') predict RSC avpr1a expression, V1aR abundance and sexual fidelity in adulthood; these alleles differ in the frequency of CpG sites and in methylation at a putative intron enhancer. We hypothesized that the elevated CpG abundance in the LO allele would make homozygous LO/LO voles more sensitive to developmental perturbations. We found that genotype differences in RSC-V1aR abundance emerged early in ontogeny and were accompanied by differences in methylation of the putative enhancer. As predicted, postnatal treatment with an oxytocin receptor antagonist (OTA) reduced RSC-V1aR abundance in LO/LO adults but not their HI/HI siblings. Similarly, methylation inhibition by zebularine increased RSC-V1aR in LO/LO adults, but not in HI/HI siblings. These data show a gene-by-environment interaction in RSC-V1aR. Surprisingly, however, neither OTA nor zebularine altered adult methylation of the intronic enhancer, suggesting that differences in sensitivity could not be explained by CpG density at the enhancer alone. Methylated DNA immunoprecipiation-sequencing showed additional differentially methylated regions between HI/HI and LO/LO voles. Future research should examine the role of these regions and other regulatory elements in the ontogeny of RSC-V1aR and its developmentally induced changes.
Collapse
Affiliation(s)
- M Okhovat
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - I C Chen
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - Z Dehghani
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - D J Zheng
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - J E Ikpatt
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - H Momoh
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - S M Phelps
- Section of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
38
|
Norde MM, Oki E, Carioca AAF, Castro IA, Souza JMP, Marchioni DML, Fisberg RM, Rogero MM. Influence of toll-like receptor 4 gene variants and plasma fatty acid profile on systemic inflammation: A population-based cross-sectional study. Nutrition 2017; 35:106-111. [PMID: 28241976 DOI: 10.1016/j.nut.2016.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 09/30/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the interaction of toll-like receptor 4 (TLR4) gene single nucleotide polymorphism (SNP) and plasma fatty acid (FA) profile in modulating risk for systemic inflammation. METHODS In all, 262 adult (19-59 y) participants of the Health Survey of São Paulo met the inclusion criteria. Anthropometric parameters, blood pressure, plasma inflammatory biomarker concentration, and fatty acid profile were measured and four SNPs of the TLR4 gene (rs4986790, rs4986791, rs11536889, and rs5030728) were genotyped. Multivariate cluster analysis was performed to stratify individuals based on levels of 11 plasma inflammatory biomarkers into two groups: inflammatory (INF) and noninflammatory (NINF). RESULTS No association was found between any of the SNPs studied and systemic inflammation. The INF cluster had higher palmitic acid levels (P = 0.039) and estimated stearoyl coenzyme A desaturase activity (P = 0.045) and lower polyunsaturated fatty acid (P = 0.011), ω-6 fatty acid (P = 0.018), arachidonic acid (P = 0.002) levels, and estimated δ-5 desaturase activity (P = 0.025) compared with the NINF cluster. Statistically significant interaction between rs11536889 and arachidonic acid/eicosapentaenoic acid (AA/EPA) ratio (P = 0.034) was found to increase the odds of belonging to the INF cluster when individuals had the variant allele C and were at the higher percentile of AA/EPA plasma ratio. CONCLUSION Plasma fatty acid profile modulated the odds of belonging to the INF cluster depending on genotypes of TRL4 gene polymorphisms.
Collapse
Affiliation(s)
| | - Erica Oki
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Antonio A F Carioca
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Inar A Castro
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Brazil
| | - José M P Souza
- Department of Epidemiology, School of Public Health, University of São Paulo, Brazil
| | - Dirce M L Marchioni
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Regina M Fisberg
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Marcelo M Rogero
- Nutrition Department, School of Public Health, University of São Paulo, Brazil.
| |
Collapse
|
39
|
Amare AT, Schubert KO, Klingler-Hoffmann M, Cohen-Woods S, Baune BT. The genetic overlap between mood disorders and cardiometabolic diseases: a systematic review of genome wide and candidate gene studies. Transl Psychiatry 2017; 7:e1007. [PMID: 28117839 PMCID: PMC5545727 DOI: 10.1038/tp.2016.261] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 10/21/2016] [Accepted: 10/31/2016] [Indexed: 12/11/2022] Open
Abstract
Meta-analyses of genome-wide association studies (meta-GWASs) and candidate gene studies have identified genetic variants associated with cardiovascular diseases, metabolic diseases and mood disorders. Although previous efforts were successful for individual disease conditions (single disease), limited information exists on shared genetic risk between these disorders. This article presents a detailed review and analysis of cardiometabolic diseases risk (CMD-R) genes that are also associated with mood disorders. First, we reviewed meta-GWASs published until January 2016, for the diseases 'type 2 diabetes, coronary artery disease, hypertension' and/or for the risk factors 'blood pressure, obesity, plasma lipid levels, insulin and glucose related traits'. We then searched the literature for published associations of these CMD-R genes with mood disorders. We considered studies that reported a significant association of at least one of the CMD-R genes and 'depression' or 'depressive disorder' or 'depressive symptoms' or 'bipolar disorder' or 'lithium treatment response in bipolar disorder', or 'serotonin reuptake inhibitors treatment response in major depression'. Our review revealed 24 potential pleiotropic genes that are likely to be shared between mood disorders and CMD-Rs. These genes include MTHFR, CACNA1D, CACNB2, GNAS, ADRB1, NCAN, REST, FTO, POMC, BDNF, CREB, ITIH4, LEP, GSK3B, SLC18A1, TLR4, PPP1R1B, APOE, CRY2, HTR1A, ADRA2A, TCF7L2, MTNR1B and IGF1. A pathway analysis of these genes revealed significant pathways: corticotrophin-releasing hormone signaling, AMPK signaling, cAMP-mediated or G-protein coupled receptor signaling, axonal guidance signaling, serotonin or dopamine receptors signaling, dopamine-DARPP32 feedback in cAMP signaling, circadian rhythm signaling and leptin signaling. Our review provides insights into the shared biological mechanisms of mood disorders and cardiometabolic diseases.
Collapse
Affiliation(s)
- A T Amare
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - K O Schubert
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Northern Adelaide Local Health Network, Mental Health Services, Adelaide, SA, Australia
| | - M Klingler-Hoffmann
- Adelaide Proteomics Centre, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - S Cohen-Woods
- School of Psychology, Faculty of Social and Behavioural Sciences, Flinders University, Adelaide, SA, Australia
| | - B T Baune
- Discipline of Psychiatry, School of Medicine, The University of Adelaide, Adelaide, SA, Australia,Discipline of Psychiatry, School of Medicine, The University of Adelaide, North Terrace, Adelaide, SA 5005, Australia. E-mail:
| |
Collapse
|
40
|
Lai CQ, Wojczynski MK, Parnell LD, Hidalgo BA, Irvin MR, Aslibekyan S, Province MA, Absher DM, Arnett DK, Ordovás JM. Epigenome-wide association study of triglyceride postprandial responses to a high-fat dietary challenge. J Lipid Res 2016; 57:2200-2207. [PMID: 27777315 PMCID: PMC5321216 DOI: 10.1194/jlr.m069948] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/16/2016] [Indexed: 12/18/2022] Open
Abstract
Postprandial lipemia (PPL), the increased plasma TG concentration after consuming a high-fat meal, is an independent risk factor for CVD. Individual responses to a meal high in fat vary greatly, depending on genetic and lifestyle factors. However, only a few loci have been associated with TG-PPL response. Heritable epigenomic changes may be significant contributors to the unexplained inter-individual PPL variability. We conducted an epigenome-wide association study on 979 subjects with DNA methylation measured from CD4+ T cells, who were challenged with a high-fat meal as a part of the Genetics of Lipid Lowering Drugs and Diet Network study. Eight methylation sites encompassing five genes, LPP, CPT1A, APOA5, SREBF1, and ABCG1, were significantly associated with PPL response at an epigenome-wide level (P < 1.1 × 10−7), but no methylation site reached epigenome-wide significance after adjusting for baseline TG levels. Higher methylation at LPP, APOA5, SREBF1, and ABCG1, and lower methylation at CPT1A methylation were correlated with an increased TG-PPL response. These PPL-associated methylation sites, also correlated with fasting TG, account for a substantially greater amount of phenotypic variance (14.9%) in PPL and fasting TG (16.3%) when compared with the genetic contribution of loci identified by our previous genome-wide association study (4.5%). In summary, the epigenome is a large contributor to the variation in PPL, and this has the potential to be used to modulate PPL and reduce CVD.
Collapse
Affiliation(s)
- Chao-Qiang Lai
- USDA Agricultural Research Service, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Laurence D Parnell
- USDA Agricultural Research Service, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Bertha A Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL
| | - Marguerite Ryan Irvin
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL
| | - Stella Aslibekyan
- Department of Epidemiology, School of Public Health, University of Alabama, Birmingham, AL
| | - Michael A Province
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Devin M Absher
- Hudson Alpha Institute for Biotechnology, Huntsville, AL
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY
| | - José M Ordovás
- Nutrition and Genomics Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| |
Collapse
|
41
|
Blanco-Rojo R, Delgado-Lista J, Lee YC, Lai CQ, Perez-Martinez P, Rangel-Zuñiga O, Smith CE, Hidalgo B, Alcala-Diaz JF, Gomez-Delgado F, Parnell LD, Arnett DK, Tucker KL, Lopez-Miranda J, Ordovas JM. Interaction of an S100A9 gene variant with saturated fat and carbohydrates to modulate insulin resistance in 3 populations of different ancestries. Am J Clin Nutr 2016; 104:508-17. [PMID: 27440084 PMCID: PMC4962160 DOI: 10.3945/ajcn.116.130898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND S100 calcium-binding protein A9 (S100A9) has previously been identified as a type 2 diabetes (T2D) gene. However, this finding requires independent validation and more in-depth analyses in other populations and ancestries. OBJECTIVES We aimed to replicate the associations between an S100A9 variant and insulin resistance and T2D and to initiate an investigation of potential interactions with the habitual diet in several independent populations. DESIGN We investigated the association of the S100A9 variant rs3014866 with insulin resistance and T2D risk and its interactions with diet in 3 diverse populations as follows: the CORDIOPREV (Coronary Diet Intervention with Olive Oil and Cardiovascular Prevention; n = 711), which consisted of Spanish white adults; the GOLDN (Genetics of Lipids Lowering Drugs and Diet Network; n = 818), which involved North American non-Hispanic white adults; and Hispanic adults who participated in the BPRHS (Boston Puerto Rican Health Study; n = 1155). RESULTS Meta-analysis indicated that T carriers presented a lower risk of T2D than CC carriers (pooled OR: 0.714; 95% CI: 0.584, 0.845; P = 0.002). In all 3 populations (CORDIOPREV, GOLDN, and BPRHS), we showed a significant interaction between the rs3014866 single nucleotide polymorphism and dietary SFA:carbohydrate ratio intake for the homeostasis model assessment of insulin resistance (HOMA-IR) (P = 0.028, P = 0.017, and P = 0.026, respectively). CC carriers had a significantly higher HOMA-IR only when SFA:carbohydrate intake was high (P = 0.045 for the CORDIOPREV, P = 0.033 for the GOLDN, and P = 0.046 for the BPRHS) but not when SFA:carbohydrate ratio intake was low. CONCLUSIONS The minor allele (T) of the S100A9 variant rs3014866 is associated with lower T2D risk in 3 populations of different ancestries. Note that individuals with the high-risk CC genotype may be more likely to benefit from a low SFA:carbohydrate ratio intake to improve insulin resistance as evaluated with the use of the HOMA-IR. These trials were registered at clinicaltrials.gov as NCT00924937 (CORDIOPREV), NCT00083369 (GOLDN), and NCT01231958 (BPRHS).
Collapse
Affiliation(s)
- Ruth Blanco-Rojo
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain; Nutrition and Genomics Laboratory and
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Chao-Qiang Lai
- Agricultural Research Service, USDA, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Oriol Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Bertha Hidalgo
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Francisco Gomez-Delgado
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Laurence D Parnell
- Agricultural Research Service, USDA, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Donna K Arnett
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - Katherine L Tucker
- Department of Clinical Laboratory and Nutritional Sciences, University of Massachusetts, Lowell, MA
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Reina Sofia University Hospital, Maimonides Institute for Biomedical Research at Cordoba, University of Cordoba, Cordoba, Spain; Centro de Investigación Biomédica en Red (CIBER) Fisiopatologia Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain;
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory and Department of Epidemiology, Spanish National Center for Cardiovascular Research (CNIC), Madrid, Spain; and Madrid Institute for Advanced Studies (IMDEA) Food Institute, Madrid, Spain
| |
Collapse
|
42
|
Genetic susceptibility to dyslipidemia and incidence of cardiovascular disease depending on a diet quality index in the Malmö Diet and Cancer cohort. GENES AND NUTRITION 2016; 11:20. [PMID: 27551321 PMCID: PMC4968442 DOI: 10.1186/s12263-016-0536-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 06/27/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND By taking diet quality into account, we may clarify the relationship between genetically elevated triglycerides (TG) and low-density lipoprotein-cholesterol (LDL-C), and better understand the inconsistent results regarding genetically elevated high-density lipoprotein-cholesterol (HDL-C), and cardiovascular disease (CVD) risk. METHODS We included 24,799 participants (62 % women, age 44-74 years) from the Malmö Diet and Cancer cohort. During a mean follow-up time of 15 years, 3068 incident CVD cases (1814 coronary and 1254 ischemic stroke) were identified. Genetic risk scores (GRSs) were constructed by combining 80 validated genetic variants associated with higher TG and LDL-C or lower HDL-C. The participants' dietary intake, assessed by a modified diet history method, was ranked according to a diet quality index that included six dietary components: saturated fat, polyunsaturated fat, fish, fiber, fruit and vegetables, and sucrose. RESULTS The GRSLDL-C (P = 5 × 10(-6)) and GRSHDL-C (P = 0.02) but not GRSTG (P = 0.08) were significantly associated with CVD risk. No significant interaction between the GRSs and diet quality was observed on CVD risk (P > 0.39). A high compared to a low diet quality attenuated the association between GRSLDL-C and the risk of incident ischemic stroke (P interaction = 0.01). CONCLUSION We found some evidence of an interaction between diet quality and GRSLDL-C on ischemic stroke.
Collapse
|
43
|
Ferguson JF, Allayee H, Gerszten RE, Ideraabdullah F, Kris-Etherton PM, Ordovás JM, Rimm EB, Wang TJ, Bennett BJ. Nutrigenomics, the Microbiome, and Gene-Environment Interactions: New Directions in Cardiovascular Disease Research, Prevention, and Treatment: A Scientific Statement From the American Heart Association. CIRCULATION. CARDIOVASCULAR GENETICS 2016; 9:291-313. [PMID: 27095829 PMCID: PMC7829062 DOI: 10.1161/hcg.0000000000000030] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiometabolic diseases are the leading cause of death worldwide and are strongly linked to both genetic and nutritional factors. The field of nutrigenomics encompasses multiple approaches aimed at understanding the effects of diet on health or disease development, including nutrigenetic studies investigating the relationship between genetic variants and diet in modulating cardiometabolic risk, as well as the effects of dietary components on multiple "omic" measures, including transcriptomics, metabolomics, proteomics, lipidomics, epigenetic modifications, and the microbiome. Here, we describe the current state of the field of nutrigenomics with respect to cardiometabolic disease research and outline a direction for the integration of multiple omics techniques in future nutrigenomic studies aimed at understanding mechanisms and developing new therapeutic options for cardiometabolic disease treatment and prevention.
Collapse
|
44
|
Simonds NI, Ghazarian AA, Pimentel CB, Schully SD, Ellison GL, Gillanders EM, Mechanic LE. Review of the Gene-Environment Interaction Literature in Cancer: What Do We Know? Genet Epidemiol 2016; 40:356-65. [PMID: 27061572 DOI: 10.1002/gepi.21967] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/07/2016] [Accepted: 02/11/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Risk of cancer is determined by a complex interplay of genetic and environmental factors. Although the study of gene-environment interactions (G×E) has been an active area of research, little is reported about the known findings in the literature. METHODS To examine the state of the science in G×E research in cancer, we performed a systematic review of published literature using gene-environment or pharmacogenomic flags from two curated databases of genetic association studies, the Human Genome Epidemiology (HuGE) literature finder and Cancer Genome-Wide Association and Meta Analyses Database (CancerGAMAdb), from January 1, 2001, to January 31, 2011. A supplemental search using HuGE was conducted for articles published from February 1, 2011, to April 11, 2013. A 25% sample of the supplemental publications was reviewed. RESULTS A total of 3,019 articles were identified in the original search. From these articles, 243 articles were determined to be relevant based on inclusion criteria (more than 3,500 interactions). From the supplemental search (1,400 articles identified), 29 additional relevant articles (1,370 interactions) were included. The majority of publications in both searches examined G×E in colon, rectal, or colorectal; breast; or lung cancer. Specific interactions examined most frequently included environmental factors categorized as energy balance (e.g., body mass index, diet), exogenous (e.g., oral contraceptives) and endogenous hormones (e.g., menopausal status), chemical environment (e.g., grilled meats), and lifestyle (e.g., smoking, alcohol intake). In both searches, the majority of interactions examined were using loci from candidate genes studies and none of the studies were genome-wide interaction studies (GEWIS). The most commonly reported measure was the interaction P-value, of which a sizable number of P-values were considered statistically significant (i.e., <0.05). In addition, the magnitude of interactions reported was modest. CONCLUSION Observations of published literature suggest that opportunity exists for increased sample size in G×E research, including GWAS-identified loci in G×E studies, exploring more GWAS approaches in G×E such as GEWIS, and improving the reporting of G×E findings.
Collapse
Affiliation(s)
- Naoko I Simonds
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Armen A Ghazarian
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Camilla B Pimentel
- Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Sheri D Schully
- Office of Disease Prevention, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gary L Ellison
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Elizabeth M Gillanders
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Leah E Mechanic
- Epidemiology and Genomics Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
45
|
Celis-Morales C, Marsaux CFM, Livingstone KM, Navas-Carretero S, San-Cristobal R, O'donovan CB, Forster H, Woolhead C, Fallaize R, Macready AL, Kolossa S, Hallmann J, Tsirigoti L, Lambrinou CP, Moschonis G, Godlewska M, Surwiłło A, Grimaldi K, Bouwman J, Manios Y, Traczyk I, Drevon CA, Parnell LD, Daniel H, Gibney ER, Brennan L, Walsh MC, Gibney M, Lovegrove JA, Martinez JA, Saris WHM, Mathers JC. Physical activity attenuates the effect of the FTO genotype on obesity traits in European adults: The Food4Me study. Obesity (Silver Spring) 2016; 24:962-9. [PMID: 26921105 DOI: 10.1002/oby.21422] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 11/19/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To examine whether the effect of FTO loci on obesity-related traits could be modified by physical activity (PA) levels in European adults. METHODS Of 1,607 Food4Me participants randomized, 1,280 were genotyped for FTO (rs9939609) and had available PA data. PA was measured objectively using accelerometers (TracmorD, Philips), whereas anthropometric measures [BMI and waist circumference (WC)] were self-reported via the Internet. RESULTS FTO genotype was associated with a higher body weight [β: 1.09 kg per risk allele, (95% CI: 0.14-2.04), P = 0.024], BMI [β: 0.54 kg m(-2) , (0.23-0.83), P < 0.0001], and WC [β: 1.07 cm, (0.24-1.90), P = 0.011]. Moderate-equivalent PA attenuated the effect of FTO on BMI (P[interaction] = 0.020). Among inactive individuals, FTO increased BMI by 1.06 kg m(-2) per allele (P = 0.024), whereas the increase in BMI was substantially attenuated among active individuals (0.16 kg m(-2) , P = 0.388). We observed similar effects for WC (P[interaction] = 0.005): the FTO risk allele increased WC by 2.72 cm per allele among inactive individuals but by only 0.49 cm in active individuals. CONCLUSIONS PA attenuates the effect of FTO genotype on BMI and WC. This may have important public health implications because genetic susceptibility to obesity in the presence of FTO variants may be reduced by adopting a physically active lifestyle.
Collapse
Affiliation(s)
- Carlos Celis-Morales
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Cyril F M Marsaux
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Katherine M Livingstone
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Santiago Navas-Carretero
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamploma, Spain
- CIBER Fisiopatología Obesidad Y Nutrición (CIBERobn), Instituto De Salud Carlos III, Madrid, Spain
| | - Rodrigo San-Cristobal
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamploma, Spain
- CIBER Fisiopatología Obesidad Y Nutrición (CIBERobn), Instituto De Salud Carlos III, Madrid, Spain
| | - Clare B O'donovan
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Hannah Forster
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Clara Woolhead
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Rosalind Fallaize
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Anna L Macready
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - Silvia Kolossa
- ZIEL Research Center of Nutrition and Food Sciences, Biochemistry Unit, Technische Universität München, Munich, Germany
| | - Jacqueline Hallmann
- ZIEL Research Center of Nutrition and Food Sciences, Biochemistry Unit, Technische Universität München, Munich, Germany
| | - Lydia Tsirigoti
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | | | - George Moschonis
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | | | | | | | - Jildau Bouwman
- TNO, Microbiology and Systems Biology, Zeist, the Netherlands
| | - Yannis Manios
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Iwona Traczyk
- National Food & Nutrition Institute (IZZ), Warsaw, Poland
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Laurence D Parnell
- Nutrition and Genomics Laboratory, Jean Mayer US Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts, USA
| | - Hannelore Daniel
- ZIEL Research Center of Nutrition and Food Sciences, Biochemistry Unit, Technische Universität München, Munich, Germany
| | - Eileen R Gibney
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Lorraine Brennan
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Marianne C Walsh
- CIBER Fisiopatología Obesidad Y Nutrición (CIBERobn), Instituto De Salud Carlos III, Madrid, Spain
| | - Mike Gibney
- UCD Institute of Food and Health, University College Dublin, Belfield, Dublin, Republic of Ireland
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition and Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, UK
| | - J Alfredo Martinez
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamploma, Spain
- CIBER Fisiopatología Obesidad Y Nutrición (CIBERobn), Instituto De Salud Carlos III, Madrid, Spain
| | - Wim H M Saris
- Department of Human Biology, NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - John C Mathers
- Human Nutrition Research Centre, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
46
|
Wojczynski MK, Parnell LD, Pollin TI, Lai CQ, Feitosa MF, O'Connell JR, Frazier-Wood AC, Gibson Q, Aslibekyan S, Ryan KA, Province MA, Tiwari HK, Ordovas JM, Shuldiner AR, Arnett DK, Borecki IB. Genome-wide association study of triglyceride response to a high-fat meal among participants of the NHLBI Genetics of Lipid Lowering Drugs and Diet Network (GOLDN). Metabolism 2015; 64:1359-71. [PMID: 26256467 PMCID: PMC4573277 DOI: 10.1016/j.metabol.2015.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 05/19/2015] [Accepted: 07/01/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The triglyceride (TG) response to a high-fat meal (postprandial lipemia, PPL) affects cardiovascular disease risk and is influenced by genes and environment. Genes involved in lipid metabolism have dominated genetic studies of PPL TG response. We sought to elucidate common genetic variants through a genome-wide association (GWA) study in the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN). METHODS The GOLDN GWAS discovery sample consisted of 872 participants within families of European ancestry. Genotypes for 2,543,887 variants were measured or imputed from HapMap. Replication of our top results was performed in the Heredity and Phenotype Intervention (HAPI) Heart Study (n = 843). PPL TG response phenotypes were constructed from plasma TG measured at baseline (fasting, 0 hour), 3.5 and 6 hours after a high-fat meal, using a random coefficient regression model. Association analyses were adjusted for covariates and principal components, as necessary, in a linear mixed model using the kinship matrix; additional models further adjusted for fasting TG were also performed. Meta-analysis of the discovery and replication studies (n = 1715) was performed on the top SNPs from GOLDN. RESULTS GOLDN revealed 111 suggestive (p < 1E-05) associations, with two SNPs meeting GWA significance level (p < 5E-08). Of the two significant SNPs, rs964184 demonstrated evidence of replication (p = 1.20E-03) in the HAPI Heart Study and in a joint analysis, was GWA significant (p = 1.26E-09). Rs964184 has been associated with fasting lipids (TG and HDL) and is near ZPR1 (formerly ZNF259), close to the APOA1/C3/A4/A5 cluster. This association was attenuated upon additional adjustment for fasting TG. CONCLUSION This is the first report of a genome-wide significant association with replication for a novel phenotype, namely PPL TG response. Future investigation into response phenotypes is warranted using pathway analyses, or newer genetic technologies such as metabolomics.
Collapse
Affiliation(s)
- Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO.
| | - Laurence D Parnell
- Nutrition and Genomics Laboratory, Jean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Toni I Pollin
- Program in Personalized and Genomic Medicine, and Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD
| | - Chao Q Lai
- Nutrition and Genomics Laboratory, Jean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Mary F Feitosa
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Jeff R O'Connell
- Program in Personalized and Genomic Medicine, and Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD
| | | | - Quince Gibson
- Program in Personalized and Genomic Medicine, and Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD
| | - Stella Aslibekyan
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Kathy A Ryan
- Program in Personalized and Genomic Medicine, and Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD
| | - Michael A Province
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Hemant K Tiwari
- Section on Statistical Genetics, Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA
| | - Alan R Shuldiner
- Program in Personalized and Genomic Medicine, and Departments of Medicine and Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD; Geriatric Research and Education Clinical Center, Veterans Administration Medical Center, Baltimore, MD
| | - Donna K Arnett
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Ingrid B Borecki
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
47
|
Dashti HS, Follis JL, Smith CE, Tanaka T, Garaulet M, Gottlieb DJ, Hruby A, Jacques PF, Kiefte-de Jong JC, Lamon-Fava S, Scheer FAJL, Bartz TM, Kovanen L, Wojczynski MK, Frazier-Wood AC, Ahluwalia TS, Perälä MM, Jonsson A, Muka T, Kalafati IP, Mikkilä V, Ordovás JM, Partonen T, Ebeling T, Hopkins PN, Paternoster L, Lahti J, Hernandez DG, Toft U, Saxena R, Vitezova A, Kanoni S, Raitakari OT, Psaty BM, Perola M, Männistö S, Straka RJ, Hansen T, Räikkönen K, Ferrucci L, Grarup N, Johnson WC, Rallidis L, Kähönen M, Siscovick DS, Havulinna AS, Astrup A, Jørgensen T, Chen TA, Hofman A, Deloukas P, Viikari JS, Mozaffarian D, Pedersen O, Rotter JI, Uitterlinden AG, Seppälä I, Tiemeier H, Salomaa V, Gharib SA, Borecki IB, Arnett DK, Sørensen TI, Eriksson JG, Bandinelli S, Linneberg A, Rich SS, Franco OH, Dedoussis G, Lehtimäki T. Gene-Environment Interactions of Circadian-Related Genes for Cardiometabolic Traits. Diabetes Care 2015; 38:1456-66. [PMID: 26084345 PMCID: PMC4512139 DOI: 10.2337/dc14-2709] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/11/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Common circadian-related gene variants associate with increased risk for metabolic alterations including type 2 diabetes. However, little is known about whether diet and sleep could modify associations between circadian-related variants (CLOCK-rs1801260, CRY2-rs11605924, MTNR1B-rs1387153, MTNR1B-rs10830963, NR1D1-rs2314339) and cardiometabolic traits (fasting glucose [FG], HOMA-insulin resistance, BMI, waist circumference, and HDL-cholesterol) to facilitate personalized recommendations. RESEARCH DESIGN AND METHODS We conducted inverse-variance weighted, fixed-effect meta-analyses of results of adjusted associations and interactions between dietary intake/sleep duration and selected variants on cardiometabolic traits from 15 cohort studies including up to 28,190 participants of European descent from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium. RESULTS We observed significant associations between relative macronutrient intakes and glycemic traits and short sleep duration (<7 h) and higher FG and replicated known MTNR1B associations with glycemic traits. No interactions were evident after accounting for multiple comparisons. However, we observed nominally significant interactions (all P < 0.01) between carbohydrate intake and MTNR1B-rs1387153 for FG with a 0.003 mmol/L higher FG with each additional 1% carbohydrate intake in the presence of the T allele, between sleep duration and CRY2-rs11605924 for HDL-cholesterol with a 0.010 mmol/L higher HDL-cholesterol with each additional hour of sleep in the presence of the A allele, and between long sleep duration (≥9 h) and MTNR1B-rs1387153 for BMI with a 0.60 kg/m(2) higher BMI with long sleep duration in the presence of the T allele relative to normal sleep duration (≥7 to <9 h). CONCLUSIONS Our results suggest that lower carbohydrate intake and normal sleep duration may ameliorate cardiometabolic abnormalities conferred by common circadian-related genetic variants. Until further mechanistic examination of the nominally significant interactions is conducted, recommendations applicable to the general population regarding diet—specifically higher carbohydrate and lower fat composition—and normal sleep duration should continue to be emphasized among individuals with the investigated circadian-related gene variants.
Collapse
Affiliation(s)
- Hassan S Dashti
- Nutrition and Genomics Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Jack L Follis
- Department of Mathematics, Computer Science and Cooperative Engineering, University of St. Thomas, Houston, TX
| | - Caren E Smith
- Nutrition and Genomics Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD
| | - Marta Garaulet
- Department of Physiology, University of Murcia, Murcia, Spain
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA Sleep Disorders Center, VA Boston Healthcare System, Boston, MA
| | - Adela Hruby
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Paul F Jacques
- Nutritional Epidemiology Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Jessica C Kiefte-de Jong
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands Global Public Health, Leiden University College, The Hague, the Netherlands
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Frank A J L Scheer
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Boston, MA Division of Sleep Medicine, Harvard Medical School, Boston, MA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA Department of Biostatistics, University of Washington, Seattle, WA
| | - Leena Kovanen
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Mary K Wojczynski
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Alexis C Frazier-Wood
- U.S. Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Tarunveer S Ahluwalia
- The Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark Copenhagen Prospective Studies on Asthma in Childhood, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark Danish Pediatric Asthma Centre, Gentofte Hospital, The Capital Region, Copenhagen, Denmark
| | - Mia-Maria Perälä
- Department of Chronic Disease Prevention, National Institute for Health and Welfare (THL), Helsinki, Finland
| | - Anna Jonsson
- The Novo Nordisk Foundation Centre for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Taulant Muka
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Ioanna P Kalafati
- Department of Nutrition and Dietetics, Harokopio University, Athens, Greece
| | - Vera Mikkilä
- Department of Food and Environmental Sciences, Division of Nutrition, University of Helsinki, Helsinki, Finland Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - José M Ordovás
- Nutrition and Genomics Laboratory, Jean Mayer U.S. Department of Agriculture Human Nutrition Research Center on Aging at Tufts University, Boston, MA Department of Epidemiology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain Instituto Madrileño de Estudios Avanzados en Alimentación (IMDEA-FOOD), Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|