1
|
Wuni R, Curi-Quinto K, Liu L, Espinoza D, Aquino AI, Del Valle-Mendoza J, Aguilar-Luis MA, Murray C, Nunes R, Methven L, Lovegrove JA, Penny M, Favara M, Sánchez A, Vimaleswaran KS. Interaction between genetic risk score and dietary carbohydrate intake on high-density lipoprotein cholesterol levels: Findings from the study of obesity, nutrition, genes and social factors (SONGS). Clin Nutr ESPEN 2025; 66:83-92. [PMID: 39800136 DOI: 10.1016/j.clnesp.2024.12.027] [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: 09/27/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS Cardiometabolic traits are complex interrelated traits that result from a combination of genetic and lifestyle factors. This study aimed to assess the interaction between genetic variants and dietary macronutrient intake on cardiometabolic traits [body mass index, waist circumference, total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol, triacylglycerol, systolic blood pressure, diastolic blood pressure, fasting serum glucose, fasting serum insulin, and glycated haemoglobin]. METHODS This cross-sectional study consisted of 468 urban young adults aged 20 ± 1 years, and it was conducted as part of the Study of Obesity, Nutrition, Genes and Social factors (SONGS) project, a sub-study of the Young Lives study. Thirty-nine single nucleotide polymorphisms (SNPs) known to be associated with cardiometabolic traits at a genome-wide significance level (P < 5 × 10-8) were used to construct a genetic risk score (GRS). RESULTS There were no significant associations between the GRS and any of the cardiometabolic traits. However, a significant interaction was observed between the GRS and carbohydrate intake on HDL-C concentration (Pinteraction = 0.0007). In the first tertile of carbohydrate intake (≤327 g/day), participants with a high GRS (>37 risk alleles) had a higher concentration of HDL-C than those with a low GRS (≤37 risk alleles) [Beta = 0.06 mmol/L, 95 % confidence interval (CI), 0.01-0.10; P = 0.018]. In the third tertile of carbohydrate intake (>452 g/day), participants with a high GRS had a lower concentration of HDL-C than those with a low GRS (Beta = -0.04 mmol/L, 95 % CI -0.01 to -0.09; P = 0.027). A significant interaction was also observed between the GRS and glycaemic load (GL) on the concentration of HDL-C (Pinteraction = 0.002). For participants with a high GRS, there were lower concentrations of HDL-C across tertiles of GL (Ptrend = 0.017). There was no significant interaction between the GRS and glycaemic index on the concentration of HDL-C, and none of the other GRS∗macronutrient interactions were significant. CONCLUSIONS Our results suggest that young adults who consume a higher carbohydrate diet and have a higher GRS have a lower HDL-C concentration, which in turn is linked to cardiovascular diseases, and indicate that personalised nutrition strategies targeting a reduction in carbohydrate intake might be beneficial for these individuals.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Katherine Curi-Quinto
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru.
| | - Litai Liu
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Dianela Espinoza
- Group for the Analysis of Development (GRADE), Lima, 15063, Peru.
| | - Anthony I Aquino
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru
| | - Juana Del Valle-Mendoza
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru; Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, 15087, Peru.
| | - Miguel Angel Aguilar-Luis
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru; Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, 15087, Peru.
| | - Claudia Murray
- Department of Real Estate and Planning, University of Reading, Reading, RG6 6UD, UK.
| | - Richard Nunes
- Department of Real Estate and Planning, University of Reading, Reading, RG6 6UD, UK.
| | - Lisa Methven
- Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK; Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, RG6 6AP, UK.
| | - Mary Penny
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru.
| | - Marta Favara
- Oxford Department of International Development, University of Oxford, Oxford, OX1 3TB, UK.
| | - Alan Sánchez
- Group for the Analysis of Development (GRADE), Lima, 15063, Peru; Oxford Department of International Development, University of Oxford, Oxford, OX1 3TB, UK.
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK; Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, RG6 6AP, UK.
| |
Collapse
|
2
|
Jaskoll S, Kramer A, Elbaz-Hayoun S, Rinsky B, Eandi CM, Grunin M, Shwartz Y, Tiosano L, Heid IM, Winkler T, Chowers I. Adult Onset Foveomacular Vitelliform Dystrophy Shows Genetic Overlap With Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2024; 65:53. [PMID: 39585675 PMCID: PMC11601137 DOI: 10.1167/iovs.65.13.53] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/04/2024] [Indexed: 11/26/2024] Open
Abstract
Purpose Adult-onset foveomacular vitelliform dystrophy (AFVD) shares phenotypic similarities with age-related macular degeneration (AMD). The genetic factors associated with AFVD are unknown in >80% of cases. This study evaluated the association of known AMD genetic risk variants with AFVD and compared systemic complement activation in these conditions. Methods Clinical, imaging, and genetic data were collected from 50 patients with AFVD (men/women = 25/25, mean age ± SD 73 ± 10 years), 917 patients with AMD (men/women = 377/540, mean age ± SD 77 ± 9 years), and 432 unaffected healthy controls (men/women = 202/230, mean age ± SD 71 ± 8 years). Genotyping focused on 52 single nucleotide polymorphisms (SNPs) linked to AMD. Weighted genetic risk scores (GRS) for 19 complement system associated variants, 7 lipid metabolism associated variants, the remaining 26 variants (other pathways GRS), and for all 52 variants (global score) were derived and correlated with phenotype. Results Of the 52 SNPs evaluated, CFH (rs570618) and C2/CFB/SKIV2L (rs116503776 and rs114254831) were associated with AFVD compared with healthy controls (odds ratio [OR] = 2.73, 95% confidence interval [CI] = 1.32-5.73, P = 0.01; OR = 0.31, 95% CI = 0.14-0.71, P = 0.0036; and OR = 0.41, 95% CI = 0.22-0.74, P = 0.0025, respectively). MIR6130/RORB (rs10781182) was negatively associated with AFVD compared with the healthy controls (OR = 0.13, CI = 0.06-0.25, P < 0.0001) and AMD (OR = 0.19, CI = 0.10-0.34, P < 0.0001). Regression analysis showed complement GRS was positively associated with AFVD compared with controls (OR = 1.42, 95% CI = 1.04-1.95, P = 0.03), whereas the other pathways' GRS was negatively associated (OR = 0.46, 95% CI = 0.21-0.98, P = 0.04). AMD was positively associated with the complement score, global score, and ARMS2/HTRA1 compared with controls. Conclusions Non-monogenic AFVD is associated with AMD risk alleles in the complement cascade, but not in other pathways. Further research is needed to explore complement inhibition for AFVD.
Collapse
Affiliation(s)
- Shlomit Jaskoll
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Military Medicine and “Tzameret”, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel, and Medical Corps, Israel Defense Forces, Israel
| | - Adi Kramer
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sarah Elbaz-Hayoun
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Batya Rinsky
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Chiara M. Eandi
- Department of Surgical Sciences, University of Torino, Torino, Italy
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, Department of Ophthalmology, University of Lausanne, Lausanne, Switzerland
| | - Michelle Grunin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Braun School of Public Health and Community Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yahel Shwartz
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Liran Tiosano
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Iris M. Heid
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Thomas Winkler
- Department of Genetic Epidemiology, University of Regensburg, Regensburg, Germany
| | - Itay Chowers
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, and the Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
3
|
Wuni R, Vimaleswaran KS. Barriers in Translating Existing Nutrigenetics Insights to Precision Nutrition for Cardiometabolic Health in Ethnically Diverse Populations. Lifestyle Genom 2024; 17:122-135. [PMID: 39467522 DOI: 10.1159/000541909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Cardiometabolic diseases pose a significant threat to global public health, with a substantial majority of cardiovascular disease mortality (more than three-quarters) occurring in low- and middle-income countries. There have been remarkable advances in recent years in identifying genetic variants that alter disease susceptibility by interacting with dietary factors. Despite the remarkable progress, several factors need to be considered before the translation of nutrigenetics insights to personalised and precision nutrition in ethnically diverse populations. Some of these factors include variations in genetic predispositions, cultural and lifestyle factors as well as socio-economic factors. SUMMARY This review aimed to explore the factors that need to be considered in bridging the gap between existing nutrigenetics insights and the implementation of personalised and precision nutrition across diverse ethnicities. Several factors might influence variations among individuals with regard to dietary exposures and metabolic responses, and these include genetic diversity, cultural and lifestyle factors as well as socio-economic factors. A multi-omics approach involving disciplines such as metabolomics, epigenetics, and the gut microbiome might contribute to improved understanding of the underlying mechanisms of gene-diet interactions and the implementation of precision nutrition although more research is needed to confirm the practicality and effectiveness of this approach. Conducting gene-diet interaction studies in diverse populations is essential and studies utilising large sample sizes are required as this improves the power to detect interactions with minimal effect sizes. Future studies should focus on replicating initial findings to enhance reliability and promote comparison across studies. Once findings have been replicated in independent samples, dietary intervention studies will be required to further strengthen the evidence and facilitate their application in clinical practice. KEY MESSAGES Nutrigenetics has a potential role to play in the prevention and management of cardiometabolic diseases. Conducting gene-diet interaction studies in diverse populations is essential giving the genetic diversity and variations in dietary patterns. Integrating data from disciplines such as metabolomics, epigenetics, and the gut microbiome could help in early identification of individuals at risk of cardiometabolic diseases as well as the implementation of precise dietary interventions for preventing and managing cardiometabolic diseases.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, UK
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, UK
- Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, UK
| |
Collapse
|
4
|
Wuni R, Amerah H, Ammache S, Cruvinel NT, da Silva NR, Kuhnle GGC, Horst MA, Vimaleswaran KS. Interaction between genetic risk score and dietary fat intake on lipid-related traits in Brazilian young adults. Br J Nutr 2024; 132:575-589. [PMID: 39308196 PMCID: PMC11536265 DOI: 10.1017/s0007114524001594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 11/01/2024]
Abstract
The occurrence of dyslipidaemia, which is an established risk factor for cardiovascular diseases, has been attributed to multiple factors including genetic and environmental factors. We used a genetic risk score (GRS) to assess the interactions between genetic variants and dietary factors on lipid-related traits in a cross-sectional study of 190 Brazilians (mean age: 21 ± 2 years). Dietary intake was assessed by a trained nutritionist using three 24-h dietary recalls. The high GRS was significantly associated with increased concentration of TAG (beta = 0·10 mg/dl, 95 % CI 0·05-0·16; P < 0·001), LDL-cholesterol (beta = 0·07 mg/dl, 95 % CI 0·04, 0·11; P < 0·0001), total cholesterol (beta = 0·05 mg/dl, 95 % CI: 0·03, 0·07; P < 0·0001) and the ratio of TAG to HDL-cholesterol (beta = 0·09 mg/dl, 95 % CI: 0·03, 0·15; P = 0·002). Significant interactions were found between the high GRS and total fat intake on TAG:HDL-cholesterol ratio (Pinteraction = 0·03) and between the high GRS and SFA intake on TAG:HDL-cholesterol ratio (Pinteraction = 0·03). A high intake of total fat (>31·5 % of energy) and SFA (>8·6 % of energy) was associated with higher TAG:HDL-cholesterol ratio in individuals with the high GRS (beta = 0·14, 95 % CI: 0·06, 0·23; P < 0·001 for total fat intake; beta = 0·13, 95 % CI: 0·05, 0·22; P = 0·003 for SFA intake). Our study provides evidence that the genetic risk of high TAG:HDL-cholesterol ratio might be modulated by dietary fat intake in Brazilians, and these individuals might benefit from limiting their intake of total fat and SFA.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, ReadingRG6 6DZ, UK
| | - Heyam Amerah
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, ReadingRG6 6DZ, UK
| | - Serena Ammache
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, ReadingRG6 6DZ, UK
| | - Nathália T. Cruvinel
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás (UFG), Goiania, Brazil
| | - Nara R. da Silva
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás (UFG), Goiania, Brazil
| | - Gunter G. C. Kuhnle
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, ReadingRG6 6DZ, UK
| | - Maria A. Horst
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás (UFG), Goiania, Brazil
| | - Karani S. Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, ReadingRG6 6DZ, UK
- Institute for Food, Nutrition, and Health (IFNH), University of Reading, ReadingRG6 6EU, UK
| |
Collapse
|
5
|
Sekar P, Aji AS, Ariyasra U, Sari SR, Tasrif N, Yani FF, Lovegrove JA, Sudji IR, Lipoeto NI, Vimaleswaran KS. A Novel Interaction between a 23-SNP Genetic Risk Score and Monounsaturated Fatty Acid Intake on HbA1c Levels in Southeast Asian Women. Nutrients 2024; 16:3022. [PMID: 39275336 PMCID: PMC11397529 DOI: 10.3390/nu16173022] [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: 07/26/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Metabolic diseases result from interactions between genetic and lifestyle factors. Understanding the combined influences of single-nucleotide polymorphisms (SNPs) and lifestyle is crucial. This study employs genetic risk scores (GRS) to assess SNPs, providing insight beyond single gene/SNP studies by revealing synergistic effects. Here, we aim to investigate the association of a 23-SNP GRS with metabolic disease-related traits (obesity and type 2 diabetes) to understand if these associations are altered by lifestyle/dietary factors. For this study, 106 Minangkabau women were included and underwent physical, anthropometric, biochemical, dietary and genetic evaluations. The interaction of GRS with lifestyle factors was analyzed using linear regression models, adjusting for potential confounders. No statistically significant associations were observed between GRS and metabolic traits; however, this study demonstrates a novel interaction observed between 13-SNP GRS and monounsaturated fatty acid (MUFA) intake, and that it had an effect on HbA1c levels (p = 0.026). Minangkabau women with low MUFA intake (≤7.0 g/day) and >13 risk alleles had significantly higher HbA1c levels (p = 0.010). This finding has implications for public health, suggesting the need for large-scale studies to confirm our results before implementing dietary interventions in the Indonesian population. Identifying genetic influences on dietary response can inform personalized nutrition strategies to reduce the risk of metabolic disease.
Collapse
Affiliation(s)
- Padmini Sekar
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK
| | - Arif S Aji
- Department of Nutrition, Faculty of Health Sciences, Alma Ata University, Bantul, Yogyakarta 55183, Indonesia
| | - Utami Ariyasra
- Department of Biomedical Science, Faculty of Medicine, Andalas University, Padang 25163, Indonesia
| | - Sri R Sari
- Department of Biomedical Science, Faculty of Medicine, Andalas University, Padang 25163, Indonesia
| | - Nabila Tasrif
- Culinary Study Program, Faculty of Tourism and Hospitality, Universitas Negeri Padang, Padang 25163, Indonesia
| | - Finny F Yani
- Department of Child Health, Faculty of Medicine, Andalas University, Padang 25163, Indonesia
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK
| | - Ikhwan R Sudji
- Biomedical Laboratory, Faculty of Medicine, Andalas University, Padang 25163, Indonesia
| | - Nur I Lipoeto
- Department of Nutrition, Faculty of Medicine, Andalas University, Padang 25163, Indonesia
| | - Karani S Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading RG6 6DZ, UK
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading RG6 6AH, UK
| |
Collapse
|
6
|
AlAnazi MM, Ventura EF, Lovegrove JA, Vimaleswaran KS. A Systematic Review of the Gene-Lifestyle Interactions on Metabolic Disease-Related Outcomes in Arab Populations. Nutrients 2024; 16:2519. [PMID: 39125399 PMCID: PMC11314532 DOI: 10.3390/nu16152519] [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: 06/13/2024] [Revised: 07/18/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
The increased prevalence of metabolic diseases in the Arab countries is mainly associated with genetic susceptibility, lifestyle behaviours, such as physical inactivity, and an unhealthy diet. The objective of this review was to investigate and summarise the findings of the gene-lifestyle interaction studies on metabolic diseases such as obesity and type 2 diabetes in Arab populations. Relevant articles were retrieved from a literature search on PubMed, Web of Science, and Google Scholar starting at the earliest indexing date through to January 2024. Articles that reported an interaction between gene variants and diet or physical activity were included and excluded if no interaction was investigated or if they were conducted among a non-Arab population. In total, five articles were included in this review. To date, among three out of twenty-two Arab populations, fourteen interactions have been found between the FTO rs9939609, TCF7L2 rs7903146, MC4R rs17782313, and MTHFR rs1801133 polymorphisms and diet or physical activity on obesity and type 2 diabetes outcomes. The majority of the reported gene-diet/ gene-physical activity interactions (twelve) appeared only once in the review. Consequently, replication, comparisons, and generalisation of the findings are limited due to the sample size, study designs, dietary assessment tools, statistical analysis, and genetic heterogeneity of the studied sample.
Collapse
Affiliation(s)
- Maria M. AlAnazi
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (M.M.A.); (J.A.L.)
- Department of Human Nutrition, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar
| | - Eduard Flores Ventura
- Institute of Agrochemistry and Food Technology—Spanish National Research Council (IATA-CSIC), 46980 Valencia, Spain;
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (M.M.A.); (J.A.L.)
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (M.M.A.); (J.A.L.)
- Institute for Food, Nutrition and Health (IFNH), University of Reading, Reading RG6 6AH, UK
| |
Collapse
|
7
|
Lu X, Xie T, van Faassen M, Kema IP, van Beek AP, Xu X, Huo X, Wolffenbuttel BHR, van Vliet-Ostaptchouk JV, Nolte IM, Snieder H. Effects of endocrine disrupting chemicals and their interactions with genetic risk scores on cardiometabolic traits. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 914:169972. [PMID: 38211872 DOI: 10.1016/j.scitotenv.2024.169972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/04/2024] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Ubiquitous non-persistent endocrine disrupting chemicals (EDCs) have inconsistent associations with cardiometabolic traits. Additionally, large-scale genome-wide association studies (GWASs) have yielded many genetic risk variants for cardiometabolic traits and diseases. This study aimed to investigate the associations between a wide range of EDC exposures (parabens, bisphenols, and phthalates) and 14 cardiometabolic traits and whether these are moderated by their respective genetic risk scores (GRSs). Data were from 1074 participants aged 18 years or older of the Lifelines Cohort Study, a large population-based biobank. GRSs for 14 cardiometabolic traits were calculated based on genome-wide significant common variants from recent GWASs. The concentrations of 15 EDCs in 24-hour urine were measured by isotope dilution liquid chromatography tandem mass spectrometry technology. The main effects of trait-specific GRSs and each of the EDC exposures and their interaction effects on the 14 cardiometabolic traits were examined in multiple linear regression. The present study confirmed significant main effects for all GRSs on their corresponding cardiometabolic trait. Regarding the main effects of EDC exposures, 26 out of 280 EDC-trait tests were significant with explained variances ranging from 0.43 % (MMP- estimated glomerular filtration rate (eGFR)) to 2.37 % (PrP-waist-hip ratio adjusted body mass index (WHRadjBMI)). We confirmed the association of MiBP and MBzP with WHRadjBMI and body mass index (BMI), and showed that parabens, bisphenol F, and many other phthalate metabolites significantly contributed to the variance of WHRadjBMI, BMI, high-density lipoprotein (HDL), eGFR, fasting glucose (FG), and diastolic blood pressure (DBP). Only one association between BMI and bisphenol F was nominally significantly moderated by the GRS explaining 0.36 % of the variance. However, it did not survive multiple testing correction. We showed that non-persistent EDC exposures exerted effects on BMI, WHRadjBMI, HDL, eGFR, FG, and DBP. However no evidence for a modulating role of GRSs was found.
Collapse
Affiliation(s)
- Xueling Lu
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands; Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 515041, Guangdong, China
| | - Tian Xie
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - André P van Beek
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Xijin Xu
- Laboratory of Environmental Medicine and Developmental Toxicology, Shantou University Medical College, 515041, Guangdong, China
| | - Xia Huo
- Laboratory of Environmental Medicine and Developmental Toxicology, Guangdong Key Laboratory of Environmental Pollution and Health, School of Environment, Jinan University, 510632, Guangdong, China
| | - Bruce H R Wolffenbuttel
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Jana V van Vliet-Ostaptchouk
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, the Netherlands.
| |
Collapse
|
8
|
Janardhanan M, Sen S, Shankarappa B, Purushottam M. Molecular genetics of neuropsychiatric illness: some musings. Front Genet 2023; 14:1203017. [PMID: 38028602 PMCID: PMC10646253 DOI: 10.3389/fgene.2023.1203017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Research into the genetic underpinnings of neuropsychiatric illness has occurred at many levels. As more information accumulates, it appears that many approaches may each offer their unique perspective. The search for low penetrance and common variants, that may mediate risk, has necessitated the formation of many international consortia, to pool resources, and achieve the large sample sizes needed to discover these variants. There has been the parallel development of statistical methods to analyse large datasets and present summary statistics which allows data comparison across studies. Even so, the results of studies on well-characterised clinical datasets of modest sizes can be enlightening and provide important clues to understanding these complex disorders. We describe the use of common variants, at multiallelic loci like TOMM40 and APOE to study dementia, weighted genetic risk scores for alcohol-induced liver cirrhosis and whole exome sequencing to identify rare variants in genes like PLA2G6 in familial psychoses and schizophrenia in our Indian population.
Collapse
Affiliation(s)
| | | | | | - Meera Purushottam
- Molecular Genetics Laboratory, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Bengaluru, India
| |
Collapse
|
9
|
Pledger SL, Ahmadizar F. Gene-environment interactions and the effect on obesity risk in low and middle-income countries: a scoping review. Front Endocrinol (Lausanne) 2023; 14:1230445. [PMID: 37664850 PMCID: PMC10474324 DOI: 10.3389/fendo.2023.1230445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
Background Obesity represents a major and preventable global health challenge as a complex disease and a modifiable risk factor for developing other non-communicable diseases. In recent years, obesity prevalence has risen more rapidly in low- and middle-income countries (LMICs) compared to high-income countries (HICs). Obesity traits are shown to be modulated by an interplay of genetic and environmental factors such as unhealthy diet and physical inactivity in studies from HICs focused on populations of European descent; however, genetic heterogeneity and environmental differences prevent the generalisation of study results to LMICs. Primary research investigating gene-environment interactions (GxE) on obesity in LMICs is limited but expanding. Synthesis of current research would provide an overview of the interactions between genetic variants and environmental factors that underlie the obesity epidemic and identify knowledge gaps for future studies. Methods Three databases were searched systematically using a combination of keywords such as "genes", "obesity", "LMIC", "diet", and "physical activity" to find all relevant observational studies published before November 2022. Results Eighteen of the 1,373 articles met the inclusion criteria, of which one was a genome-wide association study (GWAS), thirteen used a candidate gene approach, and five were assigned as genetic risk score studies. Statistically significant findings were reported for 12 individual SNPs; however, most studies were small-scale and without replication. Conclusion Although the results suggest significant GxE interactions on obesity in LMICs, updated robust statistical techniques with more precise and standardised exposure and outcome measurements are necessary for translatable results. Future research should focus on improved quality replication efforts, emphasising large-scale and long-term longitudinal study designs using multi-ethnic GWAS.
Collapse
Affiliation(s)
- Sophia L. Pledger
- Department of Epidemiology and Global Health, Julius Global Health, University Medical Center Utrecht, Utrecht, Netherlands
| | - Fariba Ahmadizar
- Department of Data Science and Biostatistics, Julius Global Health, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
10
|
Keijer J, Escoté X, Galmés S, Palou-March A, Serra F, Aldubayan MA, Pigsborg K, Magkos F, Baker EJ, Calder PC, Góralska J, Razny U, Malczewska-Malec M, Suñol D, Galofré M, Rodríguez MA, Canela N, Malcic RG, Bosch M, Favari C, Mena P, Del Rio D, Caimari A, Gutierrez B, Del Bas JM. Omics biomarkers and an approach for their practical implementation to delineate health status for personalized nutrition strategies. Crit Rev Food Sci Nutr 2023; 64:8279-8307. [PMID: 37077157 DOI: 10.1080/10408398.2023.2198605] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Personalized nutrition (PN) has gained much attention as a tool for empowerment of consumers to promote changes in dietary behavior, optimizing health status and preventing diet related diseases. Generalized implementation of PN faces different obstacles, one of the most relevant being metabolic characterization of the individual. Although omics technologies allow for assessment the dynamics of metabolism with unprecedented detail, its translatability as affordable and simple PN protocols is still difficult due to the complexity of metabolic regulation and to different technical and economical constrains. In this work, we propose a conceptual framework that considers the dysregulation of a few overarching processes, namely Carbohydrate metabolism, lipid metabolism, inflammation, oxidative stress and microbiota-derived metabolites, as the basis of the onset of several non-communicable diseases. These processes can be assessed and characterized by specific sets of proteomic, metabolomic and genetic markers that minimize operational constrains and maximize the information obtained at the individual level. Current machine learning and data analysis methodologies allow the development of algorithms to integrate omics and genetic markers. Reduction of dimensionality of variables facilitates the implementation of omics and genetic information in digital tools. This framework is exemplified by presenting the EU-Funded project PREVENTOMICS as a use case.
Collapse
Affiliation(s)
- Jaap Keijer
- Human and Animal Physiology, Wageningen University, Wageningen, the Netherlands
| | - Xavier Escoté
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, Reus, Spain
| | - Sebastià Galmés
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Andreu Palou-March
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Francisca Serra
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Group of Nutrigenomics, Biomarkers and Risk Evaluation - NuBE), University of the Balearic Islands, Palma, Spain
- Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Spin-off n.1 of the University of the Balearic Islands, Alimentómica S.L, Palma, Spain
| | - Mona Adnan Aldubayan
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Nutrition, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Kristina Pigsborg
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Ella J Baker
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | - Joanna Góralska
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | - Urszula Razny
- Department of Clinical Biochemistry, Jagiellonian University Medical College, Krakow, Poland
| | | | - David Suñol
- Digital Health, Eurecat, Centre Tecnològic de Catalunya, Barcelona, Spain
| | - Mar Galofré
- Digital Health, Eurecat, Centre Tecnològic de Catalunya, Barcelona, Spain
| | - Miguel A Rodríguez
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, Reus, Spain
| | - Núria Canela
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, Reus, Spain
| | - Radu G Malcic
- Health and Biomedicine, LEITAT Technological Centre, Barcelona, Spain
| | - Montserrat Bosch
- Applied Microbiology and Biotechnologies, LEITAT Technological Centre, Terrassa, Spain
| | - Claudia Favari
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Pedro Mena
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Daniele Del Rio
- Human Nutrition Unit, Department of Food & Drug, University of Parma, Parma, Italy
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology area, Reus, Spain
| | | | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology area, Reus, Spain
| |
Collapse
|
11
|
Wan EYF, Fung WT, Yu EYT, Cheng WHG, Chan KS, Wang Y, Chan EWY, Wong ICK, Lam CLK. Association of genetic variants related to combined exposure to higher BMI and waist-to-hip ratio on lifelong cardiovascular risk in UK Biobank. Public Health Nutr 2023; 26:416-424. [PMID: 35621080 DOI: 10.1017/s1368980022001276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE This study examines the individual and combined association of BMI and waist-to-hip ratio (WHR) with CVD risk using genetic scores of the obesity measurements as proxies. DESIGN A 2 × 2 factorial analysis approach was applied, with participants divided into four groups of lifetime exposure to low BMI and WHR, high BMI, high WHR, and high BMI and WHR based on weighted genetic risk scores. The difference in CVD risk across groups was evaluated using multivariable logistic regression. SETTING Cohort study. PARTICIPANTS A total of 408 003 participants were included from the prospective observational UK Biobank study. RESULTS A total of 58 429 CVD events were recorded. Compared to the low BMI and WHR genetic scores group, higher BMI or higher WHR genetic scores were associated with an increase in CVD risk (high WHR: OR, 1·07; 95 % CI (1·04, 1·10)); high BMI: OR, 1·12; 95 % CI (1·09, 1·16). A weak additive effect on CVD risk was found between BMI and WHR (high BMI and WHR: OR, 1·16; 95 % CI (1·12, 1·19)). Subgroup analysis showed similar patterns between different sex, age (<65, ≥65 years old), smoking status, Townsend deprivation index, fasting glucose level and medication uses, but lower systolic blood pressure was associated with higher CVD risk in obese participants. CONCLUSIONS High BMI and WHR were associated with increased CVD risk, and their effects are weakly additive. Even though there were overlapping of effect, both BMI and WHR are important in assessing the CVD risk in the general population.
Collapse
Affiliation(s)
- Eric Yuk Fai Wan
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Special Administrative Region, China
| | - Wing Tung Fung
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Esther Yee Tak Yu
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Will Ho Gi Cheng
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kam Suen Chan
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yuan Wang
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Esther Wai Yin Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Special Administrative Region, China
| | - Ian Chi Kei Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Laboratory of Data Discovery for Health (D24H), Hong Kong Special Administrative Region, China
- Research Department of Practice and Policy, School of Pharmacy, University College London, London, UK
| | - Cindy Lo Kuen Lam
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Family Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
12
|
Efficient gene-environment interaction testing through bootstrap aggregating. Sci Rep 2023; 13:937. [PMID: 36650248 PMCID: PMC9845231 DOI: 10.1038/s41598-023-28172-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
Gene-environment (GxE) interactions are an important and sophisticated component in the manifestation of complex phenotypes. Simple univariate tests lack statistical power due to the need for multiple testing adjustment and not incorporating potential interplay between several genetic loci. Approaches based on internally constructed genetic risk scores (GRS) require the partitioning of the available sample into training and testing data sets, thus, lowering the effective sample size for testing the GxE interaction itself. To overcome these issues, we propose a statistical test that employs bagging (bootstrap aggregating) in the GRS construction step and utilizes its out-of-bag prediction mechanism. This approach has the key advantage that the full available data set can be used for both constructing the GRS and testing the GxE interaction. To also incorporate interactions between genetic loci, we, furthermore, investigate if using random forests as the GRS construction method in GxE interaction testing further increases the statistical power. In a simulation study, we show that both novel procedures lead to a higher statistical power for detecting GxE interactions, while still controlling the type I error. The random-forests-based test outperforms a bagging-based test that uses the elastic net as its base learner in most scenarios. An application of the testing procedures to a real data set from a German cohort study suggests that there might be a GxE interaction involving exposure to air pollution regarding rheumatoid arthritis.
Collapse
|
13
|
Padilla-Martinez F, Szczerbiński Ł, Citko A, Czajkowski M, Konopka P, Paszko A, Wawrusiewicz-Kurylonek N, Górska M, Kretowski A. Testing the Utility of Polygenic Risk Scores for Type 2 Diabetes and Obesity in Predicting Metabolic Changes in a Prediabetic Population: An Observational Study. Int J Mol Sci 2022; 23:16081. [PMID: 36555722 PMCID: PMC9787993 DOI: 10.3390/ijms232416081] [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: 11/03/2022] [Revised: 12/03/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Prediabetes is an intermediate state of hyperglycemia during which glycemic parameters are above normal levels but below the T2D threshold. T2D and its precursor prediabetes affect 6.28% and 7.3% of the world’s population, respectively. The main objective of this paper was to create and compare two polygenic risk scores (PRSs) versus changes over time (Δ) in metabolic parameters related to prediabetes and metabolic complications. The genetics of 446 prediabetic patients from the Polish Registry of Diabetes cohort were investigated. Seventeen metabolic parameters were measured and compared at baseline and after five years using statistical analysis. Subsequently, genetic polymorphisms present in patients were determined to build a T2D PRS (68 SNPs) and an obesity PRS (21 SNPs). Finally, the association among the two PRSs and the Δ of the metabolic traits was assessed. After a multiple linear regression with adjustment for age, sex, and BMI at a nominal significance of (p < 0.05) and adjustment for multiple testing, the T2D PRS was found to be positively associated with Δ fat mass (FM) (p = 0.025). The obesity PRS was positively associated with Δ FM (p = 0.023) and Δ 2 h glucose (p = 0.034). The comparison of genotype frequencies showed that AA genotype carriers of rs10838738 were significantly higher in Δ 2 h glucose and in Δ 2 h insulin. Our findings suggest that prediabetic individuals with a higher risk of developing T2D experience increased Δ FM, and those with a higher risk of obesity experience increased Δ FM and Δ two-hour postprandial glucose. The associations found in this research could be a powerful tool for identifying prediabetic individuals with an increased risk of developing T2D and obesity.
Collapse
Affiliation(s)
| | - Łukasz Szczerbiński
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Anna Citko
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Marcin Czajkowski
- Faculty of Computer Science, Bialystok University of Technology, Wiejska 45a, 15-351 Bialystok, Poland
| | - Paulina Konopka
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Adam Paszko
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Natalia Wawrusiewicz-Kurylonek
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
- Department of Clinical Genetics, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Maria Górska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
14
|
Wigmann C, Hüls A, Krutmann J, Schikowski T. Estimating the Relative Contribution of Environmental and Genetic Risk Factors to Different Aging Traits by Combining Correlated Variables into Weighted Risk Scores. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16746. [PMID: 36554627 PMCID: PMC9779342 DOI: 10.3390/ijerph192416746] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/06/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
Genetic and exposomal factors contribute to the development of human aging. For example, genetic polymorphisms and exposure to environmental factors (air pollution, tobacco smoke, etc.) influence lung and skin aging traits. For prevention purposes it is highly desirable to know the extent to which each category of the exposome and genetic factors contribute to their development. Estimating such extents, however, is methodologically challenging, mainly because the predictors are often highly correlated. Tackling this challenge, this article proposes to use weighted risk scores to assess combined effects of categories of such predictors, and a measure of relative importance to quantify their relative contribution. The risk score weights are determined via regularized regression and the relative contributions are estimated by the proportion of explained variance in linear regression. This approach is applied to data from a cohort of elderly Caucasian women investigated in 2007-2010 by estimating the relative contribution of genetic and exposomal factors to skin and lung aging. Overall, the models explain 17% (95% CI: [9%, 28%]) of the outcome's variance for skin aging and 23% ([11%, 34%]) for lung function parameters. For both aging traits, genetic factors make up the largest contribution. The proposed approach enables us to quantify and rank contributions of categories of exposomal and genetic factors to human aging traits and facilitates risk assessment related to common human diseases in general. Obtained rankings can aid political decision making, for example, by prioritizing protective measures such as limit values for certain exposures.
Collapse
Affiliation(s)
- Claudia Wigmann
- IUF—Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany
| | - Anke Hüls
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Jean Krutmann
- IUF—Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany
- The Human Phenome Institute, Shanghai 200433, China
| | - Tamara Schikowski
- IUF—Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany
| |
Collapse
|
15
|
Kress S, Kilanowski A, Wigmann C, Zhao Q, Zhao T, Abramson MJ, Gappa M, Standl M, Unfried K, Schikowski T. Airway inflammation in adolescents and elderly women: Chronic air pollution exposure and polygenic susceptibility. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 841:156655. [PMID: 35697214 DOI: 10.1016/j.scitotenv.2022.156655] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIM The fractional exhaled nitric oxide (FeNO) concentration in the exhaled breath is a biomarker for eosinophilic airway inflammation. We explored the interplay between chronic air pollution exposure and polygenic susceptibility to airway inflammation at different critical age stages. METHODS Adolescents (15 yr) enrolled in the GINIplus/LISA birth cohorts (n = 2434) and 220 elderly women (75 yr on average) enrolled in the SALIA cohort with FeNO measurements available were investigated. Environmental main effects of the mean of ESCAPE land-use regression air pollutant concentrations within a time window of 15 years and main effects of the polygenic risk scores (PRS) using internal weights from elastic net regression of genome-wide derived single nucleotide polymorphisms were investigated. Furthermore, we examined gene-environment interaction (GxE) effects on natural log-transformed FeNO levels by adjusted linear regression models. RESULTS While we observed no significant environmental and polygenic main effects on airway inflammation in either age group, we found robust harmful effects of chronic nitrogen dioxide (NO2) exposure in the GxE models for elderly women (16.2 % increase in FeNO, p-value = 0.027). Stratified analyses found GxE effects between the PRS and chronic NO2 exposure in never-smoker elderly women and in adolescents without any inflammatory respiratory conditions. CONCLUSIONS FeNO measurement is a useful biomarker to detect higher risk of NO2-induced eosinophilic airway inflammation in the elderly. There was limited evidence for GxE effects on airway inflammation in adolescents or the elderly. Further GxE studies in subpopulations should be conducted to investigate the assumption that susceptibility to airway inflammation differs between age stages.
Collapse
Affiliation(s)
- Sara Kress
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, Düsseldorf 40225, Germany; Medical Research School Düsseldorf, Heinrich Heine University, Universitätsstraße 1, Düsseldorf 40225, Germany.
| | - Anna Kilanowski
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg 85764, Germany; Institute for Medical Information Processing, Biometry, and Epidemiology; Pettenkofer School of Public Health, LMU Munich, Geschwister-Scholl-Platz 1, Munich 80539, Germany; Division of Metabolic and Nutritional Medicine, Dr. von Hauner Children's Hospital, University of Munich Medical Center, Lindwurmstr. 4, Munich 80337, Germany.
| | - Claudia Wigmann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, Düsseldorf 40225, Germany.
| | - Qi Zhao
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, Düsseldorf 40225, Germany; Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan City 250012, Shandong Province, China; School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Rd, Melbourne, VIC 3004, Australia.
| | - Tianyu Zhao
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg 85764, Germany.
| | - Michael J Abramson
- School of Public Health and Preventive Medicine, Monash University, 553 St Kilda Rd, Melbourne, VIC 3004, Australia.
| | - Monika Gappa
- Department of Paediatrics, Evangelisches Krankenhaus, Kirchfeldstraße 40, Düsseldorf 40217, Germany.
| | - Marie Standl
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg 85764, Germany; German Center for Lung Research (DZL), Aulweg 130, Gießen 35392, Germany.
| | - Klaus Unfried
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, Düsseldorf 40225, Germany.
| | - Tamara Schikowski
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, Düsseldorf 40225, Germany.
| |
Collapse
|
16
|
Kress S, Wigmann C, Zhao Q, Herder C, Abramson MJ, Schwender H, Schikowski T. Chronic air pollution-induced subclinical airway inflammation and polygenic susceptibility. Respir Res 2022; 23:265. [PMID: 36151579 PMCID: PMC9508765 DOI: 10.1186/s12931-022-02179-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Background Air pollutants can activate low-grade subclinical inflammation which further impairs respiratory health. We aimed to investigate the role of polygenic susceptibility to chronic air pollution-induced subclinical airway inflammation. Methods We used data from 296 women (69–79 years) enrolled in the population-based SALIA cohort (Study on the influence of Air pollution on Lung function, Inflammation and Aging). Biomarkers of airway inflammation were measured in induced-sputum samples at follow-up investigation in 2007–2010. Chronic air pollution exposures at residential addresses within 15 years prior to the biomarker assessments were used to estimate main environmental effects on subclinical airway inflammation. Furthermore, we calculated internally weighted polygenic risk scores based on genome-wide derived single nucleotide polymorphisms. Polygenic main and gene-environment interaction (GxE) effects were investigated by adjusted linear regression models. Results Higher exposures to nitrogen dioxide (NO2), nitrogen oxides (NOx), particulate matter with aerodynamic diameters of ≤ 2.5 μm, ≤ 10 μm, and 2.5–10 µm significantly increased the levels of leukotriene (LT)B4 by 19.7% (p-value = 0.005), 20.9% (p = 0.002), 22.1% (p = 0.004), 17.4% (p = 0.004), and 23.4% (p = 0.001), respectively. We found significant effects of NO2 (25.9%, p = 0.008) and NOx (25.9%, p-value = 0.004) on the total number of cells. No significant GxE effects were observed. The trends were mostly robust in sensitivity analyses. Conclusions While this study confirms that higher chronic exposures to air pollution increase the risk of subclinical airway inflammation in elderly women, we could not demonstrate a significant role of polygenic susceptibility on this pathway. Further studies are required to investigate the role of polygenic susceptibility. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02179-3.
Collapse
Affiliation(s)
- Sara Kress
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.,Medical Research School Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Claudia Wigmann
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany
| | - Qi Zhao
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.,Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China.,School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research (DZD), Partner Düsseldorf, München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Michael J Abramson
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Holger Schwender
- Mathematical Institute, Heinrich Heine University, Düsseldorf, Germany
| | - Tamara Schikowski
- IUF - Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225, Düsseldorf, Germany.
| |
Collapse
|
17
|
Kress S, Hara A, Wigmann C, Sato T, Suzuki K, Pham KO, Zhao Q, Areal A, Tajima A, Schwender H, Nakamura H, Schikowski T. The Role of Polygenic Susceptibility on Air Pollution-Associated Asthma between German and Japanese Elderly Women. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:9869. [PMID: 36011501 PMCID: PMC9407879 DOI: 10.3390/ijerph19169869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 06/15/2023]
Abstract
Polygenic susceptibility likely influences individual responses to air pollutants and the risk of asthma. We compared the role of polygenic susceptibility on air pollution-associated asthma between German and Japanese women. We investigated women that were enrolled in the German SALIA cohort (n = 771, mean age = 73 years) and the Japanese Shika cohort (n = 847, mean age = 67 years) with known asthma status. Adjusted logistic regression models were used to assess the associations between (1) particulate matter with a median aerodynamic diameter ≤ 2.5μm (PM2.5) and nitrogen dioxide (NO2), (2) polygenic risk scores (PRS), and (3) gene-environment interactions (G × E) with asthma. We found an increased risk of asthma in Japanese women after exposure to low pollutant levels (PM2.5: median = 12.7µg/m3, p-value < 0.001, NO2: median = 8.5µg/m3, p-value < 0.001) and in German women protective polygenic effects (p-value = 0.008). While we found no significant G × E effects, the direction in both groups was that the PRS increased the effect of PM2.5 and decreased the effect of NO2 on asthma. Our study confirms that exposure to low air pollution levels increases the risk of asthma in Japanese women and indicates polygenic effects in German women; however, there was no evidence of G × E effects. Future genome-wide G × E studies should further explore the role of ethnic-specific polygenic susceptibility to asthma.
Collapse
Affiliation(s)
- Sara Kress
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
- Medical Research School Düsseldorf, Heinrich Heine University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Akinori Hara
- Department of Hygiene and Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8640, Ishikawa, Japan
| | - Claudia Wigmann
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| | - Takehiro Sato
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8640, Ishikawa, Japan
| | - Keita Suzuki
- Department of Hygiene and Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8640, Ishikawa, Japan
| | - Kim-Oanh Pham
- Department of Hygiene and Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8640, Ishikawa, Japan
| | - Qi Zhao
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 West Wenhua Road, Jinan 250012, China
| | - Ashtyn Areal
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
- Medical Research School Düsseldorf, Heinrich Heine University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8640, Ishikawa, Japan
| | - Holger Schwender
- Mathematical Institute, Heinrich Heine University, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Hiroyuki Nakamura
- Department of Hygiene and Public Health, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa 920-8640, Ishikawa, Japan
| | - Tamara Schikowski
- IUF—Leibniz Research Institute for Environmental Medicine, Auf’m Hennekamp 50, 40225 Düsseldorf, Germany
| |
Collapse
|
18
|
Tai KY, Dhaliwal J, Wong K. Risk score prediction model based on single nucleotide polymorphism for predicting malaria: a machine learning approach. BMC Bioinformatics 2022; 23:325. [PMID: 35934714 PMCID: PMC9358850 DOI: 10.1186/s12859-022-04870-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 08/01/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The malaria risk prediction is currently limited to using advanced statistical methods, such as time series and cluster analysis on epidemiological data. Nevertheless, machine learning models have been explored to study the complexity of malaria through blood smear images and environmental data. However, to the best of our knowledge, no study analyses the contribution of Single Nucleotide Polymorphisms (SNPs) to malaria using a machine learning model. More specifically, this study aims to quantify an individual's susceptibility to the development of malaria by using risk scores obtained from the cumulative effects of SNPs, known as weighted genetic risk scores (wGRS). RESULTS We proposed an SNP-based feature extraction algorithm that incorporates the susceptibility information of an individual to malaria to generate the feature set. However, it can become computationally expensive for a machine learning model to learn from many SNPs. Therefore, we reduced the feature set by employing the Logistic Regression and Recursive Feature Elimination (LR-RFE) method to select SNPs that improve the efficacy of our model. Next, we calculated the wGRS of the selected feature set, which is used as the model's target variables. Moreover, to compare the performance of the wGRS-only model, we calculated and evaluated the combination of wGRS with genotype frequency (wGRS + GF). Finally, Light Gradient Boosting Machine (LightGBM), eXtreme Gradient Boosting (XGBoost), and Ridge regression algorithms are utilized to establish the machine learning models for malaria risk prediction. CONCLUSIONS Our proposed approach identified SNP rs334 as the most contributing feature with an importance score of 6.224 compared to the baseline, with an importance score of 1.1314. This is an important result as prior studies have proven that rs334 is a major genetic risk factor for malaria. The analysis and comparison of the three machine learning models demonstrated that LightGBM achieves the highest model performance with a Mean Absolute Error (MAE) score of 0.0373. Furthermore, based on wGRS + GF, all models performed significantly better than wGRS alone, in which LightGBM obtained the best performance (0.0033 MAE score).
Collapse
Affiliation(s)
- Kah Yee Tai
- School of Information Technology, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Jasbir Dhaliwal
- School of Information Technology, Monash University Malaysia, Subang Jaya, Selangor, Malaysia.
| | - KokSheik Wong
- School of Information Technology, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
19
|
Schnurr TM, Katz SF, Justesen JM, O’Sullivan JW, Saliba‐Gustafsson P, Assimes TL, Carcamo‐Orive I, Ahmed A, Ashley EA, Hansen T, Knowles JW. Interactions of physical activity, muscular fitness, adiposity, and genetic risk for NAFLD. Hepatol Commun 2022; 6:1516-1526. [PMID: 35293152 PMCID: PMC9234625 DOI: 10.1002/hep4.1932] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 02/01/2022] [Accepted: 02/19/2022] [Indexed: 11/22/2022] Open
Abstract
Genetic predisposition and unhealthy lifestyle are risk factors for nonalcoholic fatty liver disease (NAFLD). We investigated whether the genetic risk of NAFLD is modified by physical activity, muscular fitness, and/or adiposity. In up to 242,524 UK Biobank participants without excessive alcohol intake or known liver disease, we examined cross-sectional interactions and joint associations of physical activity, muscular fitness, body mass index (BMI), and a genetic risk score (GRS) with alanine aminotransferase (ALT) levels and the proxy definition for suspected NAFLD of ALT levels > 30 U/L in women and >40 U/L in men. Genetic predisposition to NAFLD was quantified using a GRS consisting of 68 loci known to be associated with chronically elevated ALT. Physical activity was assessed using accelerometry, and muscular fitness was estimated by measuring handgrip strength. We found that increased physical activity and grip strength modestly attenuate genetic predisposition to elevation in ALT levels, whereas higher BMI markedly amplifies it (all p values < 0.001). Among those with normal weight and high level of physical activity, the odds of suspected NAFLD were 1.6-fold higher in those with high versus low genetic risk (reference group). In those with high genetic risk, the odds of suspected NAFLD were 12-fold higher in obese participants with low physical activity versus those with normal weight and high physical activity (odds ratio for NAFLD = 19.2 and 1.6, respectively, vs. reference group). Conclusion: In individuals with high genetic predisposition for NAFLD, maintaining a normal body weight and increased physical activity may reduce the risk of NAFLD.
Collapse
Affiliation(s)
- Theresia M. Schnurr
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKobenhavnDenmark
| | - Sophia Figueroa Katz
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve UniversityClevelandOhioUSA
| | - Johanne M. Justesen
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKobenhavnDenmark
| | - Jack W. O’Sullivan
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
| | - Peter Saliba‐Gustafsson
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Cardiovascular Medicine UnitDepartment of MedicineCenter for Molecular Medicine at BioClinicumKarolinska University HospitalKarolinska InstitutetStockholmSweden
| | - Themistocles L. Assimes
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- VA Palo Alto Health Care SystemPalo AltoCaliforniaUSA
| | - Ivan Carcamo‐Orive
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Stanford Diabetes Research CenterStanfordCaliforniaUSA
| | - Aijaz Ahmed
- Division of Gastroenterology and HepatologyStanford University School of MedicineStanfordCaliforniaUSA
| | - Euan A. Ashley
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Department of GeneticsStanford UniversityStanfordCaliforniaUSA
- Department of Biomedical Data ScienceStanford UniversityStanfordCaliforniaUSA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKobenhavnDenmark
| | - Joshua W. Knowles
- Department of MedicineDivision of Cardiovascular Medicine and Cardiovascular InstituteStanford UniversityStanfordCaliforniaUSA
- Stanford Diabetes Research CenterStanfordCaliforniaUSA
- Stanford Prevention Research CenterStanfordCaliforniaUSA
| |
Collapse
|
20
|
Development and validation of an RNA-seq-based transcriptomic risk score for asthma. Sci Rep 2022; 12:8643. [PMID: 35606385 PMCID: PMC9126925 DOI: 10.1038/s41598-022-12199-0] [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: 02/15/2022] [Accepted: 05/04/2022] [Indexed: 11/30/2022] Open
Abstract
Recent progress in RNA sequencing (RNA-seq) allows us to explore whole-genome gene expression profiles and to develop predictive model for disease risk. The objective of this study was to develop and validate an RNA-seq-based transcriptomic risk score (RSRS) for disease risk prediction that can simultaneously accommodate demographic information. We analyzed RNA-seq gene expression data from 441 asthmatic and 254 non-asthmatic samples. Logistic least absolute shrinkage and selection operator (Lasso) regression analysis in the training set identified 73 differentially expressed genes (DEG) to form a weighted RSRS that discriminated asthmatics from healthy subjects with area under the curve (AUC) of 0.80 in the testing set after adjustment for age and gender. The 73-gene RSRS was validated in three independent RNA-seq datasets and achieved AUCs of 0.70, 0.77 and 0.60, respectively. To explore their biological and molecular functions in asthma phenotype, we examined the 73 genes by enrichment pathway analysis and found that these genes were significantly (p < 0.0001) enriched for DNA replication, recombination, and repair, cell-to-cell signaling and interaction, and eumelanin biosynthesis and developmental disorder. Further in-silico analyses of the 73 genes using Connectivity map shows that drugs (mepacrine, dactolisib) and genetic perturbagens (PAK1, GSR, RBM15 and TNFRSF12A) were identified and could potentially be repurposed for treating asthma. These findings show the promise for RNA-seq risk scores to stratify and predict disease risk.
Collapse
|
21
|
Hernandez-Pacheco N, Kere M, Melén E. Gene-environment interactions in childhood asthma revisited; expanding the interaction concept. Pediatr Allergy Immunol 2022; 33:e13780. [PMID: 35616899 PMCID: PMC9325482 DOI: 10.1111/pai.13780] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 01/04/2023]
Abstract
Investigation of gene-environment interactions (GxE) may provide important insights into the gene regulatory framework in response to environmental factors of relevance for childhood asthma. Over the years, different methodological strategies have been applied, more recently using genome-wide approaches. The best example to date is the major asthma locus on the 17q12-21 chromosome region, viral infections, and airway epithelium processes where recent studies have shed much light on mechanisms in childhood asthma. However, there are challenges with the traditional single variant-single exposure interaction models, as they do not encompass the complexity and cumulative effects of multiple exposures or multiple genetic variants. As such, we need to redefine our traditional GxE thinking, and we propose in this review to expand the GxE concept by also evaluating other omics layers, such as epigenetics, transcriptomics, metabolomics, and proteomics. In addition, host factors such as age, gender, and other exposures are very likely to influence GxE effects and need firmly to be considered in future studies.
Collapse
Affiliation(s)
- Natalia Hernandez-Pacheco
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Maura Kere
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden.,Sachs' Children's Hospital, South General Hospital, Stockholm, Sweden
| |
Collapse
|
22
|
Lau M, Wigmann C, Kress S, Schikowski T, Schwender H. Evaluation of tree-based statistical learning methods for constructing genetic risk scores. BMC Bioinformatics 2022; 23:97. [PMID: 35313824 PMCID: PMC8935722 DOI: 10.1186/s12859-022-04634-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/14/2022] [Indexed: 04/11/2024] Open
Abstract
Background Genetic risk scores (GRS) summarize genetic features such as single nucleotide polymorphisms (SNPs) in a single statistic with respect to a given trait. So far, GRS are typically built using generalized linear models or regularized extensions. However, these linear methods are usually not able to incorporate gene-gene interactions or non-linear SNP-response relationships. Tree-based statistical learning methods such as random forests and logic regression may be an alternative to such regularized-regression-based methods and are investigated in this article. Moreover, we consider modifications of random forests and logic regression for the construction of GRS. Results In an extensive simulation study and an application to a real data set from a German cohort study, we show that both tree-based approaches can outperform elastic net when constructing GRS for binary traits. Especially a modification of logic regression called logic bagging could induce comparatively high predictive power as measured by the area under the curve and the statistical power. Even when considering no epistatic interaction effects but only marginal genetic effects, the regularized regression method lead in most cases to inferior results. Conclusions When constructing GRS, we recommend taking random forests and logic bagging into account, in particular, if it can be assumed that possibly unknown epistasis between SNPs is present. To develop the best possible prediction models, extensive joint hyperparameter optimizations should be conducted. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-022-04634-w.
Collapse
Affiliation(s)
- Michael Lau
- Mathematical Institute, Heinrich Heine University, Düsseldorf, Germany. .,IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Claudia Wigmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Sara Kress
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tamara Schikowski
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Holger Schwender
- Mathematical Institute, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
23
|
Tai KY, Dhaliwal J, Balasubramaniam V. Leveraging Mann-Whitney U test on large-scale genetic variation data for analysing malaria genetic markers. Malar J 2022; 21:79. [PMID: 35264165 PMCID: PMC8905822 DOI: 10.1186/s12936-022-04104-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background The malaria risk analysis of multiple populations is crucial and of great importance whilst compressing limitations. However, the exponential growth in diversity and accumulation of genetic variation data obtained from malaria-infected patients through Genome-Wide Association Studies opens up unprecedented opportunities to explore the significant differences between genetic markers (risk factors), particularly in the resistance or susceptibility of populations to malaria risk. Thus, this study proposes using statistical tests to analyse large-scale genetic variation data, comprising 20,854 samples from 11 populations within three continents: Africa, Oceania, and Asia. Methods Even though statistical tests have been utilized to conduct case–control studies since the 1950s to link risk factors to a particular disease, several challenges faced, including the choice of data (ordinal vs. non-ordinal) and test (parametric vs. non-parametric). This study overcomes these challenges by adopting the Mann–Whitney U test to analyse large-scale genetic variation data; to explore the statistical significance of markers between populations; and to further identify the highly differentiated markers. Results The findings of this study revealed a significant difference in the genetic markers between populations (p < 0.01) in all the case groups and most control groups. However, for the highly differentiated genetic markers, a significant difference (p < 0.01) was present for most genetic markers with varying p-values between the populations in the case and control groups. Moreover, several genetic markers were observed to have very significant differences (p < 0.001) across all populations, while others exist between certain specific populations. Also, several genetic markers have no significant differences between populations. Conclusions These findings further support that the genetic markers contribute differently between populations towards malaria resistance or susceptibility, thus showing differences in the likelihood of malaria infection. In addition, this study demonstrated the robustness of the Mann–Whitney U test in analysing genetic markers in large-scale genetic variation data, thereby indicating an alternative method to explore genetic markers in other complex diseases. The findings hold great promise for genetic markers analysis, and the pipeline emphasized in this study can fully be reproduced to analyse new data. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04104-x.
Collapse
Affiliation(s)
- Kah Yee Tai
- School of Information Technology, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| | - Jasbir Dhaliwal
- School of Information Technology, Monash University Malaysia, Subang Jaya, Selangor, Malaysia.
| | - Vinod Balasubramaniam
- Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Subang Jaya, Selangor, Malaysia
| |
Collapse
|
24
|
Alathari BE, Cruvinel NT, da Silva NR, Chandrabose M, Lovegrove JA, Horst MA, Vimaleswaran KS. Impact of Genetic Risk Score and Dietary Protein Intake on Vitamin D Status in Young Adults from Brazil. Nutrients 2022; 14:1015. [PMID: 35267990 PMCID: PMC8912678 DOI: 10.3390/nu14051015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 01/27/2023] Open
Abstract
Given the relationship between vitamin D deficiency (VDD) and adverse outcomes of metabolic diseases, we investigated the interplay of dietary and genetic components on vitamin D levels and metabolic traits in young adults from Brazil. Genetic analysis, dietary intake, and anthropometric and biochemical measurements were performed in 187 healthy young adults (19−24 years). Genetic risk scores (GRS) from six genetic variants associated with vitamin D (vitamin D-GRS) and 10 genetic variants associated with metabolic disease (metabolic-GRS) were constructed. High vitamin D-GRS showed a significant association with low 25(OH)D concentrations (p = 0.001) and high metabolic-GRS showed a significant association with high fasting insulin concentrations (p = 0.045). A significant interaction was found between vitamin D-GRS and total protein intake (g/day) (adjusted for non-animal protein) on 25(OH)D (pinteraction = 0.006), where individuals consuming a high protein diet (≥73 g/d) and carrying >4 risk alleles for VDD had significantly lower 25(OH)D (p = 0.002) compared to individuals carrying ≤4 risk alleles. Even though our study did not support a link between metabolic-GRS and vitamin D status, our study has demonstrated a novel interaction, where participants with high vitamin D-GRS and consuming ≥73 g of protein/day had significantly lower 25(OH)D levels. Further research is necessary to evaluate the role of animal protein consumption on VDD in Brazilians.
Collapse
Affiliation(s)
- Buthaina E. Alathari
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Reading RG6 6DZ, UK; (B.E.A.); (J.A.L.)
- Department of Food Science and Nutrition, Faculty of Health Sciences, The Public Authority for Applied Education and Training, P.O. Box 14281, AlFaiha 72853, Kuwait
| | - Nathália Teixeira Cruvinel
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (N.T.C.); (N.R.d.S.)
| | - Nara Rubia da Silva
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (N.T.C.); (N.R.d.S.)
| | - Mathurra Chandrabose
- Department of Psychology and Clinical Language Sciences, University of Reading, Harry Pitt Building, Earley Gate, Reading RG6 6ES, UK;
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Reading RG6 6DZ, UK; (B.E.A.); (J.A.L.)
- Institute for Food, Nutrition and Health, University of Reading, Reading RG6 6AH, UK
- Institute of Cardiovascular and Metabolic Research, University of Reading, Reading RG6 6AA, UK
| | - Maria A. Horst
- Nutritional Genomics Research Group, Faculty of Nutrition, Federal University of Goiás (UFG), Goiânia 74690-900, Brazil; (N.T.C.); (N.R.d.S.)
| | - Karani S. Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Reading RG6 6DZ, UK; (B.E.A.); (J.A.L.)
- Institute for Food, Nutrition and Health, University of Reading, Reading RG6 6AH, UK
- Institute of Cardiovascular and Metabolic Research, University of Reading, Reading RG6 6AA, UK
| |
Collapse
|
25
|
Isgin-Atici K, Alathari BE, Turan-Demirci B, Sendur SN, Lay I, Ellahi B, Alikasifoglu M, Erbas T, Buyuktuncer Z, Vimaleswaran KS. Interaction between Dietary Fat Intake and Metabolic Genetic Risk Score on 25-Hydroxyvitamin D Concentrations in a Turkish Adult Population. Nutrients 2022; 14:382. [PMID: 35057563 PMCID: PMC8778439 DOI: 10.3390/nu14020382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 12/19/2022] Open
Abstract
Previous studies have pointed out a link between vitamin D status and metabolic traits, however, consistent evidence has not been provided yet. This cross-sectional study has used a nutrigenetic approach to investigate the interaction between metabolic-genetic risk score (GRS) and dietary intake on serum 25-hydroxyvitamin D [25(OH)D] concentrations in 396 unrelated Turkish adults, aged 24-50 years. Serum 25(OH)D concentration was significantly lower in those with a metabolic-GRS ≥ 1 risk allele than those with a metabolic-GRS < 1 risk allele (p = 0.020). A significant interaction between metabolic-GRS and dietary fat intake (energy%) on serum 25(OH)D levels was identified (Pinteraction = 0.040). Participants carrying a metabolic-GRS ≥ 1 risk allele and consuming a high fat diet (≥38% of energy = 122.3 ± 52.51 g/day) had significantly lower serum 25(OH)D concentration (p = 0.006) in comparison to those consuming a low-fat diet (<38% of energy = 82.5 ± 37.36 g/d). In conclusion, our study suggests a novel interaction between metabolic-GRS and dietary fat intake on serum 25(OH)D level, which emphasises that following the current dietary fat intake recommendation (<35% total fat) could be important in reducing the prevalence of vitamin D deficiency in this Turkish population. Nevertheless, further larger studies are needed to verify this interaction, before implementing personalized dietary recommendations for the maintenance of optimal vitamin D status.
Collapse
Affiliation(s)
- Kubra Isgin-Atici
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06230, Turkey; (K.I.-A.); (B.T.-D.)
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Amasya University, Amasya 05000, Turkey
| | - Buthaina E. Alathari
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK;
- Department of Food Science and Nutrition, Faculty of Health Sciences, The Public Authority for Applied Education and Training, AlFaiha 72853, Kuwait
| | - Busra Turan-Demirci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06230, Turkey; (K.I.-A.); (B.T.-D.)
| | - Suleyman Nahit Sendur
- Department of Endocrinology and Metabolism, School of Medicine, Hacettepe University, Ankara 06230, Turkey; (S.N.S.); (T.E.)
| | - Incilay Lay
- Department of Medical Biochemistry, Faculty of Medicine, Hacettepe University, Ankara 06230, Turkey;
- Clinical Pathology Laboratory, Hacettepe University Hospitals, Ankara 06230, Turkey
| | - Basma Ellahi
- Faculty of Health and Social Care, University of Chester, Chester CH1 4DS, UK;
| | - Mehmet Alikasifoglu
- Department of Medical Genetics, School of Medicine, Hacettepe University, Ankara 06230, Turkey;
- Genetics Diagnostic Centre, DAMAGEN, Ankara 06230, Turkey
| | - Tomris Erbas
- Department of Endocrinology and Metabolism, School of Medicine, Hacettepe University, Ankara 06230, Turkey; (S.N.S.); (T.E.)
| | - Zehra Buyuktuncer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Hacettepe University, Ankara 06230, Turkey; (K.I.-A.); (B.T.-D.)
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK;
- Institute for Food, Nutrition, and Health, University of Reading, Reading RG6 6AH, UK
| |
Collapse
|
26
|
Leung C, Ryu MH, Bølling AK, Maestre-Batlle D, Rider CF, Hüls A, Urtatiz O, MacIsaac JL, Lau KSK, Lin DTS, Kobor MS, Carlsten C. Peroxisome proliferator-activated receptor gamma gene variants modify human airway and systemic responses to indoor dibutyl phthalate exposure. Respir Res 2022; 23:248. [PMID: 36114491 PMCID: PMC9482266 DOI: 10.1186/s12931-022-02174-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 09/08/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) of peroxisome proliferator-activated receptor gamma (PPAR-γ; gene: PPARG) and oxidative stress genes are associated with asthma risk. However, whether such variants modulate responses to dibutyl phthalate (DBP), a common plasticizer associated with increased asthma development, remains unknown. The purpose of this study is to investigate how SNPs in PPARG and oxidative stress genes, as represented by two separate genetic risk scores, modify the impact of DBP exposure on lung function and the airway and systemic response after an inhaled allergen challenge. METHODS We conducted a double-blinded human crossover study with sixteen allergen-sensitized participants exposed for three hours to DBP and control air on distinct occasions separated by a 4-week washout. Each exposure was followed by an allergen inhalation challenge; subsequently, lung function was measured, and blood and bronchoalveolar lavage (BAL) were collected and analyzed for cell counts and allergen-specific immunoglobulin E (IgE). Genetic risk scores for PPAR-γ (P-GRS; weighted sum of PPARG SNPs rs10865710, rs709158, and rs3856806) and oxidative stress (OS-GRS; unweighted sum of 16 SNPs across multiple genes) were developed, and their ability to modify DBP effects were assessed using linear mixed-effects models. RESULTS P-GRS and OS-GRS modified DBP effects on allergen-specific IgE in blood at 20 h (interaction effect [95% CI]: 1.43 [1.13 to 1.80], p = 0.005) and 3 h (0.99 [0.98 to 1], p = 0.03), respectively. P-GRS also modified DBP effects on Th2 cells in blood at 3 h (- 25.2 [- 47.7 to - 2.70], p = 0.03) and 20 h (- 39.1 [- 57.9 to - 20.3], p = 0.0005), and Th2 cells in BAL at 24 h (- 4.99 [- 8.97 to - 1.01], p = 0.02). An increasing P-GRS associated with reduced DBP effect on Th2 cells. Neither GRS significantly modified DBP effects on lung function parameters. CONCLUSIONS PPAR-γ variants modulated several airway and systemic immune responses to the ubiquitous chemical plasticizer DBP. Our results suggest that PPAR-γ variants may play a greater role than those in oxidative stress-related genes in airway allergic responses to DBP. TRIAL REGISTRATION This study reports results from The Phthalate-Allergen Immune Response Study that was registered on ClinicalTrials.gov with identification NCT02688478.
Collapse
Affiliation(s)
- Clarus Leung
- grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, 7th Floor, 2775 Laurel St, VancouverVancouver, BC V5Z1M9 Canada
| | - Min Hyung Ryu
- grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, 7th Floor, 2775 Laurel St, VancouverVancouver, BC V5Z1M9 Canada
| | - Anette Kocbach Bølling
- grid.418193.60000 0001 1541 4204Department of Air Pollution and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Danay Maestre-Batlle
- grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, 7th Floor, 2775 Laurel St, VancouverVancouver, BC V5Z1M9 Canada
| | - Christopher F. Rider
- grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, 7th Floor, 2775 Laurel St, VancouverVancouver, BC V5Z1M9 Canada
| | - Anke Hüls
- grid.189967.80000 0001 0941 6502Department of Epidemiology and Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA USA
| | - Oscar Urtatiz
- grid.414137.40000 0001 0684 7788Department of Medical Genetics, University of British Columbia-BC Children’s Hospital Research Institute, Vancouver, BC Canada
| | - Julie L. MacIsaac
- grid.414137.40000 0001 0684 7788Department of Medical Genetics, University of British Columbia-BC Children’s Hospital Research Institute, Vancouver, BC Canada
| | - Kevin Soon-Keen Lau
- grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, 7th Floor, 2775 Laurel St, VancouverVancouver, BC V5Z1M9 Canada
| | - David Tse Shen Lin
- grid.414137.40000 0001 0684 7788Department of Medical Genetics, University of British Columbia-BC Children’s Hospital Research Institute, Vancouver, BC Canada
| | - Michael S. Kobor
- grid.414137.40000 0001 0684 7788Department of Medical Genetics, University of British Columbia-BC Children’s Hospital Research Institute, Vancouver, BC Canada
| | - Chris Carlsten
- grid.17091.3e0000 0001 2288 9830Department of Medicine, University of British Columbia, 7th Floor, 2775 Laurel St, VancouverVancouver, BC V5Z1M9 Canada
| |
Collapse
|
27
|
Natae SF, Kósa Z, Sándor J, Merzah MA, Bereczky Z, Pikó P, Ádány R, Fiatal S. The Higher Prevalence of Venous Thromboembolism in the Hungarian Roma Population Could Be Due to Elevated Genetic Risk and Stronger Gene-Environmental Interactions. Front Cardiovasc Med 2021; 8:647416. [PMID: 34765649 PMCID: PMC8576195 DOI: 10.3389/fcvm.2021.647416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/20/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Interactions between genetic and environmental risk factors (GxE) contribute to an increased risk of venous thromboembolism (VTE). Understanding how these factors interact provides insight for the early identification of at-risk groups within a population and creates an opportunity to apply appropriate preventive and curative measures. Objective: To estimate and compare GxE for VTE risk in the general Hungarian and Roma populations. Methods: The study was based on data extracted from a database consisting of results previously obtained from a complex health survey with three pillars (questionnaire-based, physical, and laboratory examinations) involving 406 general Hungarian and 395 Roma subjects. DNA was genotyped for rs121909567 (SERPINC1), rs1799963 (F2), rs2036914 (F11), rs2066865 (FGG), rs6025 (F5), and rs8176719 (ABO) polymorphisms. After allele frequency comparisons, the odds ratio (OR) was calculated for individual SNPs. Furthermore, genetic risk scores (weighted GRS, unweighted GRS) were computed to estimate the joint effect of the genetic factors. Multivariable linear regression analysis was applied to test the impact of GxE on VTE risk after interaction terms were created between genetic and VTE risk factors [diabetes mellitus (DM), cancer, chronic kidney diseases (CKD), coronary artery diseases (CAD), migraine, depression, obesity, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high density lipoprotein (HDL-C), triglyceride (TG), and smoking]. Results: Interestingly, the rs121909567 (SERPINC1, ATBp3 mutation) SNP was not present in the general population at all. However, the risk allele frequency was 1% among the Roma population, which might suggest a founder effect in this minority. This polymorphism multiplicatively interacted with CAD, CKD, cancer, DM, depression, migraine, and obesity. Even though interactions were not statistically significant, the trend of interaction showed the probability of an incremental VTE risk among the Roma population. The risk of VTE was 4.7 times higher (p > 0.05) for Roma subjects who had ≥3 wGRS (median value) compared with individuals having lower wGRS values but lower for the general subjects (OR = 3.1 × 10−8). Additionally, the risk of VTE was 6.6 times higher in the Roma population that had ≥3 risk alleles (median value) than in individuals with the 0–1 risk allele, and the overall risk was much higher for the Roma population (OR = 6.6; p > 0.05) than for the general Hungarian population (OR = 1.5; p > 0.05). Five positive and significant GxE interactions were identified in the Roma population. The risk of VTE was higher among depressive Roma subjects who carried the risk variant rs2036914 (β = 0.819, p = 0.02); however, this interaction was not significant for the general subjects. The joint presence of high levels of LDL-C and rs2066865 (FGG) increased the VTE risk only among Roma individuals (β = 0.389, p = 0.002). The possibility of VTE risk increment, as a result of a multiplicative interaction between rs8176719 (ABO) and cancer, was identified, which was higher for the Roma population (β = 0.370, p < 0.001) than for the general population (β = −0.042, p = 0.6). The VTE risk increased in the Roma population (β = 0.280, p = 0.001), but was higher in the general population (β = 0.423, p = 0.001) as a result of the multiplicative interaction between CAD and rs2036914 (F11). The presence of a multiplicative interaction between rs2066865 (FGG) and CAD increased the VTE risk for the Roma population (β = 0.143, p = 0.046) but not for the general population (β = −0.329, p < 0.001). Conclusions: rs121909567 (SERPINC1, ATBp3) was confirmed as a founder mutation in the Roma population. Our study revealed some evidence on the burden of the joint presence of genetic and environmental risk factors on VTE, although the finding is highly subjected to the selection and observational biases due to the very small number of VTE cases and the observational nature of the study design, respectively. As a result of higher genetic load and GxE interactions, this minority Roma population is at higher risk of VTE than the general Hungarian population. Thus, our results suggest the need for an intensive search for the rs121909567 (SERPINC1; ATBp3) founder mutation, which might be an important factor for the assessment of thrombotic disease susceptibility among the Roma population. In addition, we strongly recommend further studies among a large number of VTE cases to explore the more precise impact of genetic and environmental risk factors on VTE in the study populations.
Collapse
Affiliation(s)
- Shewaye Fituma Natae
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Zsigmond Kósa
- Department of Health Methodology and Public Health, Faculty of Health, University of Debrecen, Nyíregyháza, Hungary
| | - János Sándor
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Mohammed Abdulridha Merzah
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Bereczky
- Division of Clinical Laboratory Science, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Pikó
- Magyar Tudományos Akadémia-Debreceni Egyetem (MTA-DE) Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Róza Ádány
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary.,Magyar Tudományos Akadémia-Debreceni Egyetem (MTA-DE) Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Szilvia Fiatal
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
28
|
Vimaleswaran KS. GeNuIne (gene-nutrient interactions) Collaboration: towards implementing multi-ethnic population-based nutrigenetic studies of vitamin B 12 and D deficiencies and metabolic diseases. Proc Nutr Soc 2021; 80:1-11. [PMID: 34548115 DOI: 10.1017/s0029665121002822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gene-nutrient interactions (GeNuIne) collaboration, a large-scale collaborative project, has been initiated to investigate the impact of gene-nutrient interactions on cardiometabolic diseases using population-based studies from ethnically diverse populations. In this project, the relationship between deficiencies of vitamins B12 and D, and metabolic diseases was explored using a nutrigenetic approach. A genetic risk score (GRS) analysis was used to examine the combined effect of several genetic variations that have been shown to be associated with metabolic diseases and vitamin B12 and D deficiencies, respectively. In Sri Lankan, Indonesian and Brazilian populations, those carrying a high B12-GRS had an increased risk of metabolic diseases under the influence of dietary protein, fibre and carbohydrate intakes, respectively; however, in Asian Indians, genetically instrumented metabolic disease risk showed a significant association with low vitamin B12 status. With regards to nutrigenetic studies on vitamin D status, although high metabolic-GRS showed an interaction with dietary carbohydrate intake on vitamin D status, the study in Indonesian women demonstrated a vitamin D GRS-carbohydrate interaction on body fat percentage. In summary, these nutrigenetic studies from multiple ethnic groups have provided evidence for the influence of the dietary factors on the relationship between vitamin B12/D deficiency and metabolic outcomes. Furthermore, these studies highlight the existence of genetic heterogeneity in gene-diet interactions across ethnically diverse populations, which further implicates the significance of personalised dietary approaches for the prevention of these micronutrient deficiencies and metabolic diseases.
Collapse
Affiliation(s)
- Karani S Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading, UK
- The Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, UK
| |
Collapse
|
29
|
Lin WY, Chan CC, Liu YL, Yang AC, Tsai SJ, Kuo PH. Sex-specific autosomal genetic effects across 26 human complex traits. Hum Mol Genet 2021; 29:1218-1228. [PMID: 32160288 DOI: 10.1093/hmg/ddaa040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/26/2019] [Accepted: 03/05/2020] [Indexed: 12/28/2022] Open
Abstract
Previous studies have shown that men and women have different genetic architectures across many traits. However, except waist-to-hip ratio (WHR) and waist circumference (WC), it remains unknown whether the genetic effects of a certain trait are weaker or stronger on men/women. With ~18 000 Taiwan Biobank subjects, we comprehensively investigate sexual heterogeneity in autosomal genetic effects, for traits regarding cardiovascular health, diabetes, kidney, liver, anthropometric profiles, blood, etc. 'Gene-by-sex interactions' (G $\times$ S) were detected in 18 out of 26 traits, each with an interaction P-value (${{P}}_{{INT}}$) less than $0.05/104={0.00048}$, where 104 is the number of tests conducted in this study. The most significant evidence of G $\times$ S was found in WHR (${{P}}_{{INT}}$ = 3.2 $\times{{10}}^{-{55}}$) and WC (${{P}}_{{INT}}$ = 2.3$\times{{10}}^{-{41}}$). As a novel G$\times$S investigation for other traits, we here find that the autosomal genetic effects are weaker on women than on men, for low-density lipoprotein cholesterol (LDL-C), uric acid (UA) and diabetes-related traits such as fasting glucose and glycated hemoglobin. For LDL-C and UA, the evidence of G$\times$S is especially notable in subjects aged less than 50 years, where estrogen can play a role in attenuating the autosomal genetic effects of these two traits. Men and women have systematically distinct environmental contexts caused by hormonal milieu and their specific society roles, which may trigger diverse gene expressions despite the same DNA materials. As many environmental exposures are difficult to collect and quantify, sex can serve as a good surrogate for these factors.
Collapse
Affiliation(s)
- Wan-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chang-Chuan Chan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan.,Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Albert C Yang
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA.,Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
30
|
Abstract
It is necessary to consider all aspects of environmental factors when assessing the health impact of an eco-building environment on its occupants. However, the multi-criteria and imprecise nature of the indoor-environment in the eco-buildings has caused difficulties in quantifying the indoor environmental pollution level. This paper describes the optimal classification and priority weight methods, which are particularly useful for assessing the indoor environmental quality (IEQ) of an eco-building to demonstrate its innovative applications. The analytic hierarchy process (AHP) was used to set up the strategic decision-making evaluation system for computing the indoor environment index (IEI) risk ranking of eco-buildings. Combined with this, a Microsoft Delphi-based IEQ intelligent forecasting software simulations package was developed, and the innovative application of indoor environmental comprehensive assessment was verified by a case study in Shanghai. The evaluation result was analyzed by the priority weight methods and the AHP decision-making system noted above. This health assessment method and system provides an innovative way for the indoor environment risk evaluation of eco-buildings and is helpful to standardize the local building market.
Collapse
|
31
|
Alathari BE, Aji AS, Ariyasra U, Sari SR, Tasrif N, Yani FF, Sudji IR, Lovegrove JA, Lipoeto NI, Vimaleswaran KS. Interaction between Vitamin D-Related Genetic Risk Score and Carbohydrate Intake on Body Fat Composition: A Study in Southeast Asian Minangkabau Women. Nutrients 2021; 13:nu13020326. [PMID: 33498618 PMCID: PMC7911469 DOI: 10.3390/nu13020326] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic diseases have been shown to be associated with low vitamin D status; however, the findings have been inconsistent. Hence, the objective of our study was to investigate the relationship between vitamin D status and metabolic disease-related traits in healthy Southeast Asian women and examine whether this relationship was modified by dietary factors using a nutrigenetic study. The study included 110 Minangkabau women (age: 25–60 years) from Padang, Indonesia. Genetic risk scores (GRS) were constructed based on five vitamin D-related single nucleotide polymorphisms (SNPs) (vitamin D-GRS) and ten metabolic disease-associated SNPs (metabolic-GRS). The metabolic-GRS was significantly associated with lower 25-hydroxyvitamin D (25(OH)D) concentrations (p = 0.009) and higher body mass index (BMI) (p = 0.016). Even though the vitamin D-GRS had no effect on metabolic traits (p > 0.12), an interaction was observed between the vitamin D-GRS and carbohydrate intake (g) on body fat percentage (BFP) (pinteraction = 0.049), where those individuals who consumed a high carbohydrate diet (mean ± SD: 319 g/d ± 46) and carried >2 vitamin D-lowering risk alleles had significantly higher BFP (p = 0.016). In summary, we have replicated the association of metabolic-GRS with higher BMI and lower 25(OH)D concentrations and identified a novel interaction between vitamin D-GRS and carbohydrate intake on body fat composition.
Collapse
Affiliation(s)
- Buthaina E. Alathari
- Department of Food Science and Nutrition, Faculty of Health Sciences, The Public Authority for Applied Education and Training, Al Faiha 72853, Kuwait;
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Reading RG6 6DZ, UK;
| | - Arif Sabta Aji
- Department of Public Health, Alma Ata Graduate School of Public Health, University of Alma Ata, Yogyakarta 55183, Indonesia;
- Department of Nutrition, Faculty of Health Sciences, University of Alma Ata, Yogyakarta 55183, Indonesia
| | - Utami Ariyasra
- Biomedical Science Department, Faculty of Medicine, Andalas University, West Sumatra 25172, Indonesia; (U.A.); (S.R.S.)
| | - Sri R. Sari
- Biomedical Science Department, Faculty of Medicine, Andalas University, West Sumatra 25172, Indonesia; (U.A.); (S.R.S.)
| | - Nabila Tasrif
- Public Health Department, Faculty of Medicine, Andalas University, West Sumatra 25172, Indonesia;
| | - Finny F. Yani
- Department of Child Health, Faculty of Medicine, Andalas University, West Sumatra 25172, Indonesia;
| | - Ikhwan R. Sudji
- Department of Medical Laboratory Technology, Faculty of Health Science, University Perintis, Padang 25586, Indonesia;
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Reading RG6 6DZ, UK;
| | - Nur I. Lipoeto
- Department of Nutrition, Faculty of Medicine, Andalas University, West Sumatra 25172, Indonesia;
| | - Karani S. Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Harry Nursten Building, Pepper Lane, Reading RG6 6DZ, UK;
- Correspondence:
| |
Collapse
|
32
|
Maternal antenatal depression and child mental health: Moderation by genomic risk for attention-deficit/hyperactivity disorder. Dev Psychopathol 2021; 32:1810-1821. [PMID: 33427178 DOI: 10.1017/s0954579420001418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal antenatal depression strongly influences child mental health but with considerable inter-individual variation that is, in part, linked to genotype. The challenge is to effectively capture the genotypic influence. We outline a novel approach to describe genomic susceptibility to maternal antenatal depression focusing on child emotional/behavioral difficulties. Two cohorts provided measures of maternal depression, child genetic variation, and child mental health symptoms. We constructed a conventional polygenic risk score (PRS) for attention-deficit/hyperactivity disorder (ADHD) (PRSADHD) that significantly moderated the association between maternal antenatal depression and internalizing problems at 60 months (p = 2.94 × 10-4, R2 = .18). We then constructed an interaction PRS (xPRS) based on a subset of those single nucleotide polymorphisms from the PRSADHD that most accounted for the moderation of the association between maternal antenatal depression and child outcome. The interaction between maternal antenatal depression and this xPRS accounted for a larger proportion of the variance in child emotional/behavioral problems than models based on any PRSADHD (p = 5.50 × 10-9, R2 = .27), with similar findings in the replication cohort. The xPRS was significantly enriched for genes involved in neuronal development and synaptic function. Our study illustrates a novel approach to the study of genotypic moderation on the impact of maternal antenatal depression on child mental health and highlights the utility of the xPRS approach. These findings advance our understanding of individual differences in the developmental origins of mental health.
Collapse
|
33
|
Interaction between the genetic risk score and dietary protein intake on cardiometabolic traits in Southeast Asian. GENES AND NUTRITION 2020; 15:19. [PMID: 33045981 PMCID: PMC7552350 DOI: 10.1186/s12263-020-00678-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 09/30/2020] [Indexed: 12/18/2022]
Abstract
Background Cardiometabolic diseases are complex traits which are influenced by several single nucleotide polymorphisms (SNPs). Thus, analysing the combined effects of multiple gene variants might provide a better understanding of disease risk than using a single gene variant approach. Furthermore, studies have found that the effect of SNPs on cardiometabolic traits can be influenced by lifestyle factors, highlighting the importance of analysing gene-lifestyle interactions. Aims In the present study, we investigated the association of 15 gene variants with cardiometabolic traits and examined whether these associations were modified by lifestyle factors such as dietary intake and physical activity. Methods The study included 110 Minangkabau women [aged 25–60 years and body mass index (BMI) 25.13 ± 4.2 kg/m2] from Padang, Indonesia. All participants underwent a physical examination followed by anthropometric, biochemical and dietary assessments and genetic tests. A genetic risk score (GRS) was developed based on 15 cardiometabolic disease-related SNPs. The effect of GRS on cardiometabolic traits was analysed using general linear models. GRS-lifestyle interactions on continuous outcomes were tested by including the interaction term (e.g. lifestyle factor*GRS) in the regression model. Models were adjusted for age, BMI and location (rural or urban), wherever appropriate. Results There was a significant association between GRS and BMI, where individuals carrying 6 or more risk alleles had higher BMI compared to those carrying 5 or less risk alleles (P = 0.018). Furthermore, there were significant interactions of GRS with protein intake on waist circumference (WC) and triglyceride concentrations (Pinteraction = 0.002 and 0.003, respectively). Among women who had a lower protein intake (13.51 ± 1.18% of the total daily energy intake), carriers of six or more risk alleles had significantly lower WC and triglyceride concentrations compared with carriers of five or less risk alleles (P = 0.0118 and 0.002, respectively). Conclusions Our study confirmed the association of GRS with higher BMI and further showed a significant effect of the GRS on WC and triglyceride levels through the influence of a low-protein diet. These findings suggest that following a lower protein diet, particularly in genetically predisposed individuals, might be an effective approach for addressing cardiometabolic diseases among Southeast Asian women.
Collapse
|
34
|
Lin WY, Huang CC, Liu YL, Tsai SJ, Kuo PH. Polygenic approaches to detect gene-environment interactions when external information is unavailable. Brief Bioinform 2020; 20:2236-2252. [PMID: 30219835 PMCID: PMC6954453 DOI: 10.1093/bib/bby086] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 12/18/2022] Open
Abstract
The exploration of 'gene-environment interactions' (G × E) is important for disease prediction and prevention. The scientific community usually uses external information to construct a genetic risk score (GRS), and then tests the interaction between this GRS and an environmental factor (E). However, external genome-wide association studies (GWAS) are not always available, especially for non-Caucasian ethnicity. Although GRS is an analysis tool to detect G × E in GWAS, its performance remains unclear when there is no external information. Our 'adaptive combination of Bayes factors method' (ADABF) can aggregate G × E signals and test the significance of G × E by a polygenic test. We here explore a powerful polygenic approach for G × E when external information is unavailable, by comparing our ADABF with the GRS based on marginal effects of SNPs (GRS-M) and GRS based on SNP × E interactions (GRS-I). ADABF is the most powerful method in the absence of SNP main effects, whereas GRS-M is generally the best test when single-nucleotide polymorphisms main effects exist. GRS-I is the least powerful test due to its data-splitting strategy. Furthermore, we apply these methods to Taiwan Biobank data. ADABF and GRS-M identified gene × alcohol and gene × smoking interactions on blood pressure (BP). BP-increasing alleles elevate more BP in drinkers (smokers) than in nondrinkers (nonsmokers). This work provides guidance to choose a polygenic approach to detect G × E when external information is unavailable.
Collapse
Affiliation(s)
- Wan-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Ching-Chieh Huang
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli County, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, TaipeiVeterans General Hospital, Taipei, Taiwan.,Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
35
|
Sukcharoen K, Sharp SA, Thomas NJ, Kimmitt RA, Harrison J, Bingham C, Mozere M, Weedon MN, Tyrrell J, Barratt J, Gale DP, Oram RA. IgA Nephropathy Genetic Risk Score to Estimate the Prevalence of IgA Nephropathy in UK Biobank. Kidney Int Rep 2020; 5:1643-1650. [PMID: 33102956 PMCID: PMC7572308 DOI: 10.1016/j.ekir.2020.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/02/2020] [Accepted: 07/10/2020] [Indexed: 11/03/2022] Open
Abstract
Background IgA nephropathy (IgAN) is the commonest glomerulonephritis worldwide. Its prevalence is difficult to estimate, as people with mild disease do not commonly receive a biopsy diagnosis. We aimed to generate an IgA nephropathy genetic risk score (IgAN-GRS) and estimate the proportion of people with hematuria who had IgAN in the UK Biobank (UKBB). Methods We calculated an IgAN-GRS using 14 single-nucleotide polymorphisms (SNPs) drawn from the largest European Genome-Wide Association Study (GWAS) and validated the IgAN-GRS in 464 biopsy-proven IgAN European cases from the UK Glomerulonephritis DNA Bank (UKGDB) and in 379,767 Europeans in the UKBB. We used the mean of IgAN-GRS to calculate the proportion of potential IgAN in 14,181 with hematuria and other nonspecific renal phenotypes from 379,767 Europeans in the UKBB. Results The IgAN-GRS was higher in the IgAN cohort (4.30; 95% confidence interval [95% CI: 4.23–4.38) than in controls (3.98; 3.97–3.98; P < 0.0001). The mean GRS in UKBB participants with hematuria (n = 12,858) was higher (4.04; 4.02–4.06) than UKBB controls (3.98; 3.97–3.98; P < 0.0001) and higher in those with hematuria, hypertension, and microalbuminuria (n = 1323) (4.07; 4.02–4.13) versus (3.98; 3.97–3.98; P = 0.0003). Using the difference in these means, we estimated that IgAN accounted for 19% of noncancer hematuria and 28% with hematuria, hypertension, and microalbuminuria in UKBB. Conclusions We used an IgAN-GRS to estimate the prevalence of IgAN contributing to common phenotypes that are not always biopsied. The noninvasive use of polygenic risk in this setting may have further utility to identify likely etiology of nonspecific renal phenotypes in large population cohorts.
Collapse
Affiliation(s)
- Kittiya Sukcharoen
- The Academic Renal Unit, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - Seth A Sharp
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Nicholas J Thomas
- The Academic Renal Unit, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - Robert A Kimmitt
- The Academic Renal Unit, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - Jamie Harrison
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Coralie Bingham
- The Academic Renal Unit, Royal Devon and Exeter Foundation Trust, Exeter, UK
| | - Monika Mozere
- Department of Renal Medicine, University College London, London, UK
| | - Michael N Weedon
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Jessica Tyrrell
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | - Jonathan Barratt
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Daniel P Gale
- Department of Renal Medicine, University College London, London, UK
| | - Richard A Oram
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
36
|
Lin WY, Lin YS, Chan CC, Liu YL, Tsai SJ, Kuo PH. Using Genetic Risk Score Approaches to Infer Whether an Environmental Factor Attenuates or Exacerbates the Adverse Influence of a Candidate Gene. Front Genet 2020; 11:331. [PMID: 32457790 PMCID: PMC7225361 DOI: 10.3389/fgene.2020.00331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/20/2020] [Indexed: 11/18/2022] Open
Abstract
Some candidate genes have been robustly reported to be associated with complex traits, such as the fat mass and obesity-associated (FTO) gene on body mass index (BMI), and the fibroblast growth factor 5 (FGF5) gene on blood pressure levels. It is of interest to know whether an environmental factor (E) can attenuate or exacerbate the adverse influence of a candidate gene. To this end, we here evaluate the performance of “genetic risk score” (GRS) approaches to detect “gene-environment interactions” (G × E). In the first stage, a GRS is calculated according to the genotypes of variants in a candidate gene. In the second stage, we test whether E can significantly modify this GRS effect. This two-stage procedure can not only provide a p-value for a G × E test but also guide inferences on how E modifies the adverse effect of a gene. With systematic simulations, we compared several ways to construct a GRS. If E exacerbates the adverse influence of a gene, GRS formed by the elastic net (ENET) or the least absolute shrinkage and selection operator (LASSO) is recommended. However, the performance of ENET or LASSO will be compromised if E attenuates the adverse influence of a gene, and using the ridge regression (RIDGE) can be more powerful in this situation. Applying RIDGE to 18,424 subjects in the Taiwan Biobank, we showed that performing regular exercise can attenuate the adverse influence of the FTO gene on four obesity measures: BMI (p = 0.0009), body fat percentage (p = 0.0031), waist circumference (p = 0.0052), and hip circumference (p = 0.0001). As another example, we used RIDGE and found the FGF5 gene has a stronger effect on blood pressure in Han Chinese with a higher waist-to-hip ratio [p = 0.0013 for diastolic blood pressure (DBP) and p = 0.0027 for systolic blood pressure (SBP)]. This study provides an evaluation on the GRS approaches, which is important to infer whether E attenuates or exacerbates the adverse influence of a candidate gene.
Collapse
Affiliation(s)
- Wan-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Shun Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Chang-Chuan Chan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan.,Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan.,Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.,Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Alathari BE, Bodhini D, Jayashri R, Lakshmipriya N, Shanthi Rani CS, Sudha V, Lovegrove JA, Anjana RM, Mohan V, Radha V, Pradeepa R, Vimaleswaran KS. A Nutrigenetic Approach to Investigate the Relationship between Metabolic Traits and Vitamin D Status in an Asian Indian Population. Nutrients 2020; 12:E1357. [PMID: 32397403 PMCID: PMC7285077 DOI: 10.3390/nu12051357] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Studies in Asian Indians have examined the association of metabolic traits with vitamin D status. However, findings have been quite inconsistent. Hence, we aimed to explore the relationship between metabolic traits and 25-hydroxyvitamin D [25(OH)D] concentrations. We investigate whether this relationship was modified by lifestyle factors using a nutrigenetic approach in 545 Asian Indians randomly selected from the Chennai Urban Rural Epidemiology Study (219 normal glucose tolerant individuals, 151 with pre-diabetes and 175 individuals with type 2 diabetes). A metabolic genetic risk score (GRS) was developed using five common metabolic disease-related genetic variants. There was a significant interaction between metabolic GRS and carbohydrate intake (energy%) on 25(OH)D (Pinteraction = 0.047). Individuals consuming a low carbohydrate diet (≤62%) and those having lesser number of metabolic risk alleles (GRS ≤ 1) had significantly higher levels of 25(OH)D (p = 0.033). Conversely, individuals consuming a high carbohydrate diet despite having lesser number of risk alleles did not show a significant increase in 25(OH)D (p = 0.662). In summary, our findings show that individuals carrying a smaller number of metabolic risk alleles are likely to have higher 25(OH)D levels if they consume a low carbohydrate diet. These data support the current dietary carbohydrate recommendations of 50%-60% energy suggesting that reduced metabolic genetic risk increases 25(OH)D.
Collapse
Affiliation(s)
- Buthaina E. Alathari
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AP, UK; (B.E.A.); (J.A.L.)
- Department of Food Science and Nutrition, Faculty of Health Sciences, The Public Authority for Applied Education and Training, PO Box 14281, AlFaiha 72853, Kuwait
| | - Dhanasekaran Bodhini
- Department of Molecular Genetics, Madras Diabetes Research Foundation, Chennai 603103, India; (D.B.); (V.R.)
| | - Ramamoorthy Jayashri
- Department of Biochemistry, Madras Diabetes Research Foundation, Chennai 600086, India;
| | - Nagarajan Lakshmipriya
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India; (N.L.); (V.S.)
| | | | - Vasudevan Sudha
- Department of Foods, Nutrition and Dietetics Research, Madras Diabetes Research Foundation, Chennai 600086, India; (N.L.); (V.S.)
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AP, UK; (B.E.A.); (J.A.L.)
| | - Ranjit Mohan Anjana
- Department of Diabetology, Madras Diabetes Research Foundation & Dr. Mohan′s Diabetes Specialities Centre, WHO Collaborating Centre for Non-communicable Diseases Prevention and Control, ICMR Centre for Advanced Research on Diabetes, Gopalapuram, Chennai 600086, India; (R.M.A.); (V.M.); (R.P.)
| | - Viswanathan Mohan
- Department of Diabetology, Madras Diabetes Research Foundation & Dr. Mohan′s Diabetes Specialities Centre, WHO Collaborating Centre for Non-communicable Diseases Prevention and Control, ICMR Centre for Advanced Research on Diabetes, Gopalapuram, Chennai 600086, India; (R.M.A.); (V.M.); (R.P.)
| | - Venkatesan Radha
- Department of Molecular Genetics, Madras Diabetes Research Foundation, Chennai 603103, India; (D.B.); (V.R.)
| | - Rajendra Pradeepa
- Department of Diabetology, Madras Diabetes Research Foundation & Dr. Mohan′s Diabetes Specialities Centre, WHO Collaborating Centre for Non-communicable Diseases Prevention and Control, ICMR Centre for Advanced Research on Diabetes, Gopalapuram, Chennai 600086, India; (R.M.A.); (V.M.); (R.P.)
| | - Karani S. Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6AP, UK; (B.E.A.); (J.A.L.)
| |
Collapse
|
38
|
Wooding DJ, Ryu MH, Hüls A, Lee AD, Lin DTS, Rider CF, Yuen ACY, Carlsten C. Particle Depletion Does Not Remediate Acute Effects of Traffic-related Air Pollution and Allergen. A Randomized, Double-Blind Crossover Study. Am J Respir Crit Care Med 2020; 200:565-574. [PMID: 30974969 DOI: 10.1164/rccm.201809-1657oc] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rationale: Diesel exhaust (DE), an established model of traffic-related air pollution, contributes significantly to the global burden of asthma and may augment the effects of allergen inhalation. Newer diesel particulate-filtering technologies may increase NO2 emissions, raising questions regarding their effectiveness in reducing harm from associated engine output.Objectives: To assess the effects of DE and allergen coexposure on lung function, airway responsiveness, and circulating leukocytes, and determine whether DE particle depletion remediates these effects.Methods: In this randomized, double-blind crossover study, 14 allergen-sensitized participants (9 with airway hyperresponsiveness) underwent inhaled allergen challenge after 2-hour exposures to DE, particle-depleted DE (PDDE), or filtered air. The control condition was inhaled saline after filtered air. Blood sampling and spirometry were performed before and up to 48 hours after exposures. Airway responsiveness was evaluated at 24 hours.Measurements and Main Results: PDDE plus allergen coexposure impaired lung function more than DE plus allergen, particularly in those genetically at risk. DE plus allergen and PDDE plus allergen each increased airway responsiveness in normally responsive participants. DE plus allergen increased blood neutrophils and was associated with persistent eosinophilia at 48 hours. DE and PDDE each increased total peripheral leukocyte counts in a manner affected by participant genotypes. Changes in peripheral leukocytes correlated with lung function decline.Conclusions: Coexposure to DE and allergen impaired lung function, which was worse after particle depletion (which increased NO2). Thus, particulates are not necessarily the sole or main culprit responsible for all harmful effects of DE. Policies and technologies aimed at protecting public health should be scrutinized in that regard.Clinical trial registered with www.clinicaltrials.gov (NCT02017431).
Collapse
Affiliation(s)
- Denise J Wooding
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, and
| | - Min Hyung Ryu
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, and
| | - Anke Hüls
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Human Genetics, Emory University, Atlanta, Georgia; and.,Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Andrew D Lee
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, and
| | - David T S Lin
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.,Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Christopher F Rider
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, and
| | - Agnes C Y Yuen
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, and
| | - Chris Carlsten
- Air Pollution Exposure Laboratory, Division of Respiratory Medicine, Department of Medicine, and
| |
Collapse
|
39
|
Hüls A, Vanker A, Gray D, Koen N, MacIsaac JL, Lin DTS, Ramadori KE, Sly PD, Stein DJ, Kobor MS, Zar HJ. Genetic susceptibility to asthma increases the vulnerability to indoor air pollution. Eur Respir J 2020; 55:13993003.01831-2019. [PMID: 31949118 PMCID: PMC7931665 DOI: 10.1183/13993003.01831-2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 12/21/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Indoor air pollution and maternal smoking during pregnancy are associated with respiratory symptoms in infants, but little is known about the direct association with lung function or interactions with genetic risk factors. We examined associations of exposure to indoor particulate matter with a 50% cut-off aerodynamic diameter of 10 µm (PM10) and maternal smoking with infant lung function and the role of gene-environment interactions. METHODS Data from the Drakenstein Child Health Study, a South African birth cohort, were analysed (n=270). Lung function was measured at 6 weeks and 1 year of age, and lower respiratory tract infection episodes were documented. We measured pre- and postnatal PM10 exposures using devices placed in homes, and prenatal tobacco smoke exposure using maternal urine cotinine levels. Genetic risk scores determined from associations with childhood-onset asthma in the UK Biobank were used to investigate effect modifications. RESULTS Pre- and postnatal exposure to PM10 as well as maternal smoking during pregnancy were associated with reduced lung function at 6 weeks and 1 year as well as with lower respiratory tract infection in the first year. Due to a significant interaction between the genetic risk score and prenatal exposure to PM10, infants carrying more asthma-related risk alleles were more susceptible to PM10-associated reduced lung function (pinteraction=0.007). This interaction was stronger in infants with Black African ancestry (pinteraction=0.001) and nonexistent in children with mixed ancestry (pinteraction=0.876). CONCLUSIONS PM10 and maternal smoking exposures were associated with reduced lung function, with a higher susceptibility for infants with an adverse genetic predisposition for asthma that also depended on the infant's ancestry.
Collapse
Affiliation(s)
- Anke Hüls
- Depts of Epidemiology and Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA .,Dept of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA.,Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Dept of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Aneesa Vanker
- Dept of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and South African Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Diane Gray
- Dept of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and South African Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Nastassja Koen
- Dept of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.,South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Julia L MacIsaac
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Dept of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - David T S Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Dept of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Katia E Ramadori
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Dept of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Peter D Sly
- Children's Health and Environment Program, Child Health Research Centre, The University of Queensland, Brisbane, Australia
| | - Dan J Stein
- Dept of Psychiatry and Mental Health, University of Cape Town, Cape Town, South Africa.,South African Medical Research Council Unit on Risk and Resilience in Mental Disorders, Cape Town, South Africa
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Dept of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Heather J Zar
- Dept of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital and South African Medical Research Council Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
40
|
Hüls A, Czamara D. Methodological challenges in constructing DNA methylation risk scores. Epigenetics 2020; 15:1-11. [PMID: 31318318 PMCID: PMC6961658 DOI: 10.1080/15592294.2019.1644879] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/28/2019] [Accepted: 07/09/2019] [Indexed: 12/23/2022] Open
Abstract
Polygenic approaches often access more variance of complex traits than is possible by single variant approaches. For genotype data, genetic risk scores (GRS) are widely used for risk prediction as well as in association and interaction studies. Recently, interest has been growing in transferring GRS approaches to DNA methylation data (methylation risk scores, MRS), which can be used 1) as biomarkers for environmental exposures, 2) in association analyses in which single CpG sites do not achieve significance, 3) as dimension reduction approach in interaction and mediation analyses, and 4) to predict individual risks of disease or treatment success. Most GRS approaches can directly be transferred to methylation data. However, since methylation data is more sensitive to confounding, e.g. by age and tissue, it is more complex to find appropriate external weights. In this review, we will outline the adaption of current GRS approaches to methylation data and highlight occurring challenges.
Collapse
Affiliation(s)
- Anke Hüls
- Department of Human Genetics, Emory University, Atlanta, GA, USA
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Darina Czamara
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
41
|
Lin WY, Chan CC, Liu YL, Yang AC, Tsai SJ, Kuo PH. Performing different kinds of physical exercise differentially attenuates the genetic effects on obesity measures: Evidence from 18,424 Taiwan Biobank participants. PLoS Genet 2019; 15:e1008277. [PMID: 31369549 PMCID: PMC6675047 DOI: 10.1371/journal.pgen.1008277] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/26/2019] [Indexed: 12/17/2022] Open
Abstract
Obesity is a worldwide health problem that is closely linked to many metabolic disorders. Regular physical exercise has been found to attenuate the genetic predisposition to obesity. However, it remains unknown what kinds of exercise can modify the genetic risk of obesity. This study included 18,424 unrelated Han Chinese adults aged 30–70 years who participated in the Taiwan Biobank (TWB). A total of 5 obesity measures were investigated here, including body mass index (BMI), body fat percentage (BFP), waist circumference (WC), hip circumference (HC), and waist-to-hip ratio (WHR). Because there have been no large genome-wide association studies on obesity for Han Chinese, we used the TWB internal weights to construct genetic risk scores (GRSs) for each obesity measure, and then test the significance of GRS-by-exercise interactions. The significance level throughout this work was set at 0.05/550 = 9.1x10-5 because a total of 550 tests were performed. Performing regular exercise was found to attenuate the genetic effects on 4 obesity measures, including BMI, BFP, WC, and HC. Among the 18 kinds of self-reported regular exercise, 6 mitigated the genetic effects on at least one obesity measure. Regular jogging blunted the genetic effects on BMI, BFP, and HC. Mountain climbing, walking, exercise walking, international standard dancing, and a longer practice of yoga also attenuated the genetic effects on BMI. Exercises such as cycling, stretching exercise, swimming, dance dance revolution, and qigong were not found to modify the genetic effects on any obesity measure. Across all 5 obesity measures, regular jogging consistently presented the most significant interactions with GRSs. Our findings show that the genetic effects on obesity measures can be decreased to various extents by performing different kinds of exercise. The benefits of regular physical exercise are more impactful in subjects who are more predisposed to obesity. The complex interplay of genetics and lifestyle makes obesity a challenging issue. Previous studies have found performing regular physical exercise could blunt the genetic effects on body mass index (BMI). However, BMI does not take into account lean body mass or identify central obesity. Moreover, it remains unclear what kinds of exercise could more effectively attenuate the genetic effects on obesity measures. With a sample of 18,424 unrelated Han Chinese adults, we comprehensively investigated gene-exercise interactions on 5 obesity measures: BMI, body fat percentage, waist circumference, hip circumference, and waist-to-hip ratio. Moreover, we tested whether the genetic effects on obesity measures could be modified by any of 18 kinds of self-reported regular exercise. Because no large genome-wide association studies on obesity have been done for Han Chinese, we constructed genetic risk scores with internal weights for analyses. Among these exercises, regular jogging consistently presented the strongest evidence to mitigate the genetic effects on all 5 obesity measures. Moreover, mountain climbing, walking, exercise walking, international standard dancing, and a longer practice of yoga attenuated the genetic effects on BMI. The benefits of regularly performing these 6 kinds of exercise are more impactful in subjects who are more predisposed to obesity.
Collapse
Affiliation(s)
- Wan-Yu Lin
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- * E-mail: (WYL); (PHK)
| | - Chang-Chuan Chan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- Institute of Occupational Medicine and Industrial Hygiene, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Yu-Li Liu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Albert C. Yang
- Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, United States of America
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
| | - Shih-Jen Tsai
- Division of Psychiatry, National Yang-Ming University, Taipei, Taiwan
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan
- Department of Psychiatry, Taipei Veterans General Hospital, Beitou District, Taipei, Taiwan
| | - Po-Hsiu Kuo
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
- Department of Public Health, College of Public Health, National Taiwan University, Taipei, Taiwan
- * E-mail: (WYL); (PHK)
| |
Collapse
|
42
|
A genetic approach to examine the relationship between vitamin B12 status and metabolic traits in a South Asian population. Int J Diabetes Dev Ctries 2019. [DOI: 10.1007/s13410-019-00749-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
43
|
Hüls A, Sugiri D, Abramson MJ, Hoffmann B, Schwender H, Ickstadt K, Krämer U, Schikowski T. Benefits of improved air quality on ageing lungs: impacts of genetics and obesity. Eur Respir J 2019; 53:13993003.01780-2018. [PMID: 30765509 DOI: 10.1183/13993003.01780-2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/27/2019] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The beneficial effect of improving air quality on lung function in the elderly remains unclear. We examined associations between decline in air pollutants and lung function, and effect modifications by genetics and body mass index (BMI), in elderly German women. METHODS Data were analysed from the prospective SALIA (Study on the influence of Air pollution on Lung function, Inflammation and Aging) study (n=601). Spirometry was conducted at baseline (1985-1994; age 55 years), in 2007-2010 and in 2012-2013. Air pollution concentrations at home addresses were determined for each time-point using land-use regression models. Global Lung Initiative 2012 z-scores were calculated. Weighted genetic risk scores (GRSs) were determined from lung function-related risk alleles and used to investigate interactions with improved air quality. Multiple linear mixed models were fitted. RESULTS Air pollution levels decreased substantially during the study period. Reduction of air pollution was associated with an increase in z-scores for forced expiratory volume in 1 s (FEV1) and the FEV1/forced vital capacity ratio. For a decrease of 10 µg·m-3 in nitrogen dioxide (NO2), the z-score for FEV1 increased by 0.14 (95% CI 0.01-0.26). However, with an increasing number of lung function-related risk alleles, the benefit from improved air quality decreased (GRS×NO2 interaction: p=0.029). Interactions with BMI were not significant. CONCLUSIONS Reduction of air pollution is associated with a relative improvement of lung function in elderly women, but also depends on their genetic make-up.
Collapse
Affiliation(s)
- Anke Hüls
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.,Dept of Human Genetics, Emory University, Atlanta, GA, USA
| | - Dorothee Sugiri
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Michael J Abramson
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.,School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Barbara Hoffmann
- Institute of Occupational, Social and Environmental Medicine, Centre for Health and Society, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Holger Schwender
- Mathematical Institute, Heinrich Heine University, Düsseldorf, Germany
| | - Katja Ickstadt
- Faculty of Statistics, TU Dortmund University, Dortmund, Germany
| | - Ursula Krämer
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tamara Schikowski
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| |
Collapse
|
44
|
Hüls A, Abramson MJ, Sugiri D, Fuks K, Krämer U, Krutmann J, Schikowski T. Nonatopic eczema in elderly women: Effect of air pollution and genes. J Allergy Clin Immunol 2019; 143:378-385.e9. [DOI: 10.1016/j.jaci.2018.09.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/30/2018] [Accepted: 09/21/2018] [Indexed: 11/29/2022]
|