1
|
Shibata M, Terada A, Kawaguchi T, Kamatani Y, Okada D, Nagashima K, Ohmura K, Matsuda F, Kawaguchi S, Sese J, Yamada R. Identification of epistatic SNP combinations in rheumatoid arthritis using LAMPLINK and Japanese cohorts. J Hum Genet 2024; 69:541-547. [PMID: 39014190 DOI: 10.1038/s10038-024-01269-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/16/2024] [Accepted: 06/20/2024] [Indexed: 07/18/2024]
Abstract
Genome-wide association studies have enabled the identification of important genetic factors in many trait studies. However, only a fraction of the heritability can be explained by known genetic factors, even in the most common diseases. Genetic loci combinations, or epistatic contributions expressed by combinations of single nucleotide polymorphisms (SNPs), have been argued to be one of the critical factors explaining some of the missing heritability, especially in oligogenic/polygenic diseases. Rheumatoid arthritis (RA) is a complex disease with more than 100 reported SNP associations, as well as various HLA haplotypes and amino acids; however, many associations between RA and inter-chromosomal SNP combinations are unknown. To discover novel associations of epistatic interactions with high odds ratios in RA, we applied the LAMPLINK method, a systematic enumerative procedure for identifying high-order SNP combinations, to a Japanese RA cohort (discovery cohort; 4024 patients with RA and 7731 controls). We validated the identified associations in a different Japanese cohort (validation cohort; 810 RA patients and 6303 controls). In this study, we identified 90 significant genetic associations in the discovery cohort. Among these, 74 (82.2%) associations were replicated in the validation cohort, and eight combinations were inter-chromosomal, all of which comprised rs7765379 or rs35265698 located in the HLA region. These two SNPs exhibited strong correlations with valine at amino acid position 11 in HLA-DRB1 (HLA-DRB1-11-Val). Finally, we discovered that rs9624 showed an association with RA through an epistatic interaction with HLA-DRB1-11-Val. Overall, LAMPLINK showed high reliability for identifying epistatic genetic contributions hidden in complex traits.
Collapse
Affiliation(s)
- Mio Shibata
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Takahisa Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Daigo Okada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kazuhisa Nagashima
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shuji Kawaguchi
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Kyoto-McGill International Collaborative School in Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Jun Sese
- Humanome Lab. Inc., Tokyo, Japan.
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.
| | - Ryo Yamada
- Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Hajiaghabozorgi M, Fischbach M, Albrecht M, Wang W, Myers CL. BridGE: a pathway-based analysis tool for detecting genetic interactions from GWAS. Nat Protoc 2024; 19:1400-1435. [PMID: 38514837 PMCID: PMC11311251 DOI: 10.1038/s41596-024-00954-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/22/2023] [Indexed: 03/23/2024]
Abstract
Genetic interactions have the potential to modulate phenotypes, including human disease. In principle, genome-wide association studies (GWAS) provide a platform for detecting genetic interactions; however, traditional methods for identifying them, which tend to focus on testing individual variant pairs, lack statistical power. In this protocol, we describe a novel computational approach, called Bridging Gene sets with Epistasis (BridGE), for discovering genetic interactions between biological pathways from GWAS data. We present a Python-based implementation of BridGE along with instructions for its application to a typical human GWAS cohort. The major stages include initial data processing and quality control, construction of a variant-level genetic interaction network, measurement of pathway-level genetic interactions, evaluation of statistical significance using sample permutations and generation of results in a standardized output format. The BridGE software pipeline includes options for running the analysis on multiple cores and multiple nodes for users who have access to computing clusters or a cloud computing environment. In a cluster computing environment with 10 nodes and 100 GB of memory per node, the method can be run in less than 24 h for typical human GWAS cohorts. Using BridGE requires knowledge of running Python programs and basic shell script programming experience.
Collapse
Affiliation(s)
- Mehrad Hajiaghabozorgi
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Mathew Fischbach
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Bioinformatics and Computational Biology (BICB), University of Minnesota, Minneapolis, MN, USA
| | - Michael Albrecht
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Wen Wang
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA.
| | - Chad L Myers
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA.
- Graduate Program in Bioinformatics and Computational Biology (BICB), University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
3
|
Fu B, Pazokitoroudi A, Xue A, Anand A, Anand P, Zaitlen N, Sankararaman S. A biobank-scale test of marginal epistasis reveals genome-wide signals of polygenic epistasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557084. [PMID: 37745394 PMCID: PMC10515811 DOI: 10.1101/2023.09.10.557084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
The contribution of epistasis (interactions among genes or genetic variants) to human complex trait variation remains poorly understood. Methods that aim to explicitly identify pairs of genetic variants, usually single nucleotide polymorphisms (SNPs), associated with a trait suffer from low power due to the large number of hypotheses tested while also having to deal with the computational problem of searching over a potentially large number of candidate pairs. An alternate approach involves testing whether a single SNP modulates variation in a trait against a polygenic background. While overcoming the limitation of low power, such tests of polygenic or marginal epistasis (ME) are infeasible on Biobank-scale data where hundreds of thousands of individuals are genotyped over millions of SNPs. We present a method to test for ME of a SNP on a trait that is applicable to biobank-scale data. We performed extensive simulations to show that our method provides calibrated tests of ME. We applied our method to test for ME at SNPs that are associated with 53 quantitative traits across ≈ 300 K unrelated white British individuals in the UK Biobank (UKBB). Testing 15, 601 trait-loci associations that were significant in GWAS, we identified 16 trait-loci pairs across 12 traits that demonstrate strong evidence of ME signals (p-value p < 5 × 10 - 8 53 ). We further partitioned the significant ME signals across the genome to identify 6 trait-loci pairs with evidence of local (within-chromosome) ME while 15 show evidence of distal (cross-chromosome) ME. Across the 16 trait-loci pairs, we document that the proportion of trait variance explained by ME is about 12x as large as that explained by the GWAS effects on average (range: 0.59 to 43.89). Our results show, for the first time, evidence of interaction effects between individual genetic variants and overall polygenic background modulating complex trait variation.
Collapse
Affiliation(s)
- Boyang Fu
- Department of Computer Science, UCLA, Los Angeles, CA, USA
| | | | - Albert Xue
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, USA
| | - Aakarsh Anand
- Department of Computer Science, UCLA, Los Angeles, CA, USA
| | - Prateek Anand
- Department of Computer Science, UCLA, Los Angeles, CA, USA
| | - Noah Zaitlen
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Sriram Sankararaman
- Department of Computer Science, UCLA, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| |
Collapse
|
4
|
Li L, Wu X, Chen J, Wang S, Wan Y, Ji H, Wen Y, Zhang J. Genetic Dissection of Epistatic Interactions Contributing Yield-Related Agronomic Traits in Rice Using the Compressed Mixed Model. PLANTS 2022; 11:plants11192504. [PMID: 36235370 PMCID: PMC9571936 DOI: 10.3390/plants11192504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022]
Abstract
Rice (Oryza sativa) is one of the most important cereal crops in the world, and yield-related agronomic traits, including plant height (PH), panicle length (PL), and protein content (PC), are prerequisites for attaining the desired yield and quality in breeding programs. Meanwhile, the main effects and epistatic effects of quantitative trait nucleotides (QTNs) are all important genetic components for yield-related quantitative traits. In this study, we conducted genome-wide association studies (GWAS) for 413 rice germplasm resources, with 36,901 single nucleotide polymorphisms (SNPs), to identify QTNs, QTN-by-QTN interaction (QQI), and their candidate genes, using a multi-locus compressed variance component mixed model, 3VmrMLM. As a result, two significant QTNs and 56 paired QQIs were detected, amongst 5219 genes of these QTNs, and 26 genes were identified as the yield-related confirmed genes, such as LCRN1, OsSPL3, and OsVOZ1 for PH, and LOG and QsBZR1 for PL. To reveal the substantial contributions related to the variation of yield-related agronomic traits in rice, we further implemented an enrichment analysis and expression analysis. As the results showed, 114 genes, nearly all significant QQIs, were involved in 37 GO terms; for example, the macromolecule metabolic process (GO:0043170), intracellular part (GO:0044424), and binding (GO:0005488). It was revealed that most of the QQIs and the candidate genes were significantly involved in the biological process, molecular function, and cellular component of the target traits. The demonstrated genetic interactions play a critical role in yield-related agronomic traits of rice, and such epistatic interactions contributed to large portions of the missing heritability in GWAS. These results help us to understand the genetic basis underlying the inheritance of the three yield-related agronomic traits and provide implications for rice improvement.
Collapse
Affiliation(s)
- Ling Li
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Xinyi Wu
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Juncong Chen
- College of Finance, Nanjing Agricultural University, Nanjing 210095, China
| | - Shengmeng Wang
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Yuxuan Wan
- School of Business Administration, Jiangxi University of Finance and Economics, Nanchang 330013, China
| | - Hanbing Ji
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Yangjun Wen
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (Y.W.); (J.Z.)
| | - Jin Zhang
- College of Science, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China
- Correspondence: (Y.W.); (J.Z.)
| |
Collapse
|
5
|
Trinder M, Brunham LR. Polygenic scores for dyslipidemia: the emerging genomic model of plasma lipoprotein trait inheritance. Curr Opin Lipidol 2021; 32:103-111. [PMID: 33395106 DOI: 10.1097/mol.0000000000000737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Contemporary polygenic scores, which summarize the cumulative contribution of millions of common single-nucleotide variants to a phenotypic trait, can have effects comparable to monogenic mutations. This review focuses on the emerging use of 'genome-wide' polygenic scores for plasma lipoproteins to define the etiology of clinical dyslipidemia, modify the severity of monogenic disease, and inform therapeutic options. RECENT FINDINGS Polygenic scores for low-density lipoprotein cholesterol (LDL-C), triglycerides, and high-density lipoprotein cholesterol are associated with severe hypercholesterolemia, hypertriglyceridemia, or hypoalphalipoproteinemia, respectively. These polygenic scores for LDL-C or triglycerides associate with risk of incident coronary artery disease (CAD) independent of polygenic scores designed specifically for CAD and may identify individuals that benefit most from lipid-lowering medication. Additionally, the severity of hypercholesterolemia and CAD associated with familial hypercholesterolemia-a common monogenic disorder-is modified by these polygenic factors. The current focus of polygenic scores for dyslipidemia is to design predictive polygenic scores for diverse populations and determining how these polygenic scores could be implemented and standardized for use in the clinic. SUMMARY Polygenic scores have shown early promise for the management of dyslipidemias, but several challenges need to be addressed before widespread clinical implementation to ensure that potential benefits are robust and reproducible, equitable, and cost-effective.
Collapse
Affiliation(s)
- Mark Trinder
- Centre for Heart Lung Innovation, University of British Columbia
- Experimental Medicine Program, University of British Columbia
| | - Liam R Brunham
- Centre for Heart Lung Innovation, University of British Columbia
- Experimental Medicine Program, University of British Columbia
- Department of Medicine, University of British Columbia
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
6
|
Functional Haplotype of LIPC Induces Triglyceride-Mediated Suppression of HDL-C Levels According to Genome-Wide Association Studies. Genes (Basel) 2021; 12:genes12020148. [PMID: 33499410 PMCID: PMC7910859 DOI: 10.3390/genes12020148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Hepatic lipase (encoded by LIPC) is a glycoprotein in the triacylglycerol lipase family and mainly synthesized in and secreted from the liver. Previous studies demonstrated that hepatic lipase is crucial for reverse cholesterol transport and modulating metabolism and the plasma levels of several lipoproteins. This study was conducted to investigate the suppression effect of high-density lipoprotein cholesterol (HDL-C) levels in a genome-wide association study and explore the possible mechanisms linking triglyceride (TG) to LIPC variants and HDL-C. Genome-wide association data for TG and HDL-C were available for 4657 Taiwan-biobank participants. The prevalence of haplotypes in the LIPC promoter region and their effects were calculated. The cloned constructs of the haplotypes were expressed transiently in HepG2 cells and evaluated in a luciferase reporter assay. Genome-wide association analysis revealed that HDL-C was significantly associated with variations in LIPC after adjusting for TG. Three haplotypes (H1: TCG, H2: CTA and H3: CCA) in LIPC were identified. H2: CTA was significantly associated with HDL-C levels and H1: TCG suppressed HDL-C levels when a third factor, TG, was included in mediation analysis. The luciferase reporter assay further showed that the H2: CTA haplotype significantly inhibited luciferase activity compared with the H1: TCG haplotype. In conclusion, we identified a suppressive role for TG in the genome-wide association between LIPC and HDL-C. A functional haplotype of hepatic lipase may reduce HDL-C levels and is suppressed by TG.
Collapse
|
7
|
Vrablik M, Tichý L, Freiberger T, Blaha V, Satny M, Hubacek JA. Genetics of Familial Hypercholesterolemia: New Insights. Front Genet 2020; 11:574474. [PMID: 33133164 PMCID: PMC7575810 DOI: 10.3389/fgene.2020.574474] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Familial hypercholesterolemia (FH) is one of the most common monogenic diseases, leading to an increased risk of premature atherosclerosis and its cardiovascular complications due to its effect on plasma cholesterol levels. Variants of three genes (LDL-R, APOB and PCSK9) are the major causes of FH, but in some probands, the FH phenotype is associated with variants of other genes. Alternatively, the typical clinical picture of FH can result from the accumulation of common cholesterol-increasing alleles (polygenic FH). Although the Czech Republic is one of the most successful countries with respect to FH detection, approximately 80% of FH patients remain undiagnosed. The opportunities for international collaboration and experience sharing within international programs (e.g., EAS FHSC, ScreenPro FH, etc.) will improve the detection of FH patients in the future and enable even more accessible and accurate genetic diagnostics.
Collapse
Affiliation(s)
- Michal Vrablik
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Tichý
- Centre of Molecular Biology and Gene Therapy, University Hospital, Brno, Czechia
| | - Tomas Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Vladimir Blaha
- Internal Gerontometabolic Department, Charles University and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Martin Satny
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Jaroslav A Hubacek
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia.,Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| |
Collapse
|
8
|
Cao X, Yu G, Ren W, Guo M, Wang J. DualWMDR: Detecting epistatic interaction with dual screening and multifactor dimensionality reduction. Hum Mutat 2019; 41:719-734. [DOI: 10.1002/humu.23951] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/10/2019] [Accepted: 11/07/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Xia Cao
- College of Computer and Information ScienceSouthwest UniversityChongqing China
| | - Guoxian Yu
- College of Computer and Information ScienceSouthwest UniversityChongqing China
| | - Wei Ren
- College of Computer and Information ScienceSouthwest UniversityChongqing China
| | - Maozu Guo
- School of Electrical and Information EngineeringBeijing University of Civil Engineering and ArchitectureBeijing China
- Beijing Key Laboratory of Intelligent Processing for Building Big DataBeijing China
| | - Jun Wang
- College of Computer and Information ScienceSouthwest UniversityChongqing China
| |
Collapse
|
9
|
Discovering genetic interactions bridging pathways in genome-wide association studies. Nat Commun 2019; 10:4274. [PMID: 31537791 PMCID: PMC6753138 DOI: 10.1038/s41467-019-12131-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 08/20/2019] [Indexed: 12/20/2022] Open
Abstract
Genetic interactions have been reported to underlie phenotypes in a variety of systems, but the extent to which they contribute to complex disease in humans remains unclear. In principle, genome-wide association studies (GWAS) provide a platform for detecting genetic interactions, but existing methods for identifying them from GWAS data tend to focus on testing individual locus pairs, which undermines statistical power. Importantly, a global genetic network mapped for a model eukaryotic organism revealed that genetic interactions often connect genes between compensatory functional modules in a highly coherent manner. Taking advantage of this expected structure, we developed a computational approach called BridGE that identifies pathways connected by genetic interactions from GWAS data. Applying BridGE broadly, we discover significant interactions in Parkinson's disease, schizophrenia, hypertension, prostate cancer, breast cancer, and type 2 diabetes. Our novel approach provides a general framework for mapping complex genetic networks underlying human disease from genome-wide genotype data.
Collapse
|
10
|
Teng MS, Wu S, Hsu LA, Tzeng IS, Chou HH, Su CW, Ko YL. Pleiotropic association of LIPC variants with lipid and urinary 8-hydroxy deoxyguanosine levels in a Taiwanese population. Lipids Health Dis 2019; 18:111. [PMID: 31077211 PMCID: PMC6511151 DOI: 10.1186/s12944-019-1057-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 04/24/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hepatic lipase (HL, encoded by LIPC) is a glycoprotein primarily synthesized and secreted by hepatocytes. Previous studies had demonstrated that HL is crucial for reverse cholesterol transport and affects the metabolism, composition, and level of several lipoproteins. In current study, we investigated the association of LIPC (Lipase C, Hepatic Type) variants with circulating and urinary biomarker levels by using subgroup and mediation analyses. METHODS A total of 572 participants from Taiwan were genotyped for three LIPC single nucleotide polymorphisms (SNPs) by using TaqMan assay. Fasting levels of glucose, lipid profile, inflammation markers, urine creatinine and 8-hydroxy deoxyguanosine (8-OHdG) were measured. The chi-square test, 2-sample t test and Analysis of variance (ANOVA) were used to examine differences among variables and genotype frequencies. RESULTS SNPs rs2043085 and rs1532085 were significantly associated with urinary 8-OHdG levels, whereas all three SNPs were more significantly associated with Triglycerides (TG) or HDL-cholesterol (HDL-C) levels after additional adjustment for HDL-C or TG levels, respectively. Subgroup analyses revealed that the association of the LIPC SNPs with the levels of serum TG, HDL-C, and urinary 8-OHdG were predominantly observed in the men but not in the women. Differential associations of the LIPC SNPs with various lipid levels were observed in participants with different adiposity statuses. Mediation analyses indicated that TG levels acted as a suppressor masking the association of the LIPC genotypes with HDL-C levels, particularly in the men (Sobel test, all P < 0.01). CONCLUSION Our data revealed that interaction and suppression effects mediated the pleiotropic association of the LIPC variants. The effects of the LIPC SNPs depended on sex, adiposity status, and TG levels. Thus, our findings can provide a method for identifying high-risk populations of cardiovascular diseases for clinical diagnosis.
Collapse
Affiliation(s)
- Ming-Sheng Teng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Semon Wu
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan.,Department of Life Science, Chinese Culture University, Taipei, Taiwan
| | - Lung-An Hsu
- The First Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - I-Shiang Tzeng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Hsin-Hua Chou
- The Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Cheng-Wen Su
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan
| | - Yu-Lin Ko
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan. .,The Division of Cardiology, Department of Internal Medicine and Cardiovascular Center, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei city, Taiwan. .,School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
11
|
Ramstein GP, Evans J, Nandety A, Saha MC, Brummer EC, Kaeppler SM, Buell CR, Casler MD. Candidate Variants for Additive and Interactive Effects on Bioenergy Traits in Switchgrass ( Panicum virgatum L.) Identified by Genome-Wide Association Analyses. THE PLANT GENOME 2018; 11:180002. [PMID: 30512032 DOI: 10.3835/plantgenome2018.01.0002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Switchgrass ( L.) is a promising herbaceous energy crop, but further gains in biomass yield and quality must be achieved to enable a viable bioenergy industry. Developing DNA markers can contribute to such progress, but depiction of genetic bases should be reliable, involving simple additive marker effects and also interactions with genetic backgrounds (e.g., ecotypes) or synergies with other markers. We analyzed plant height, C content, N content, and mineral concentration in a diverse panel consisting of 512 genotypes of upland and lowland ecotypes. We performed association analyses based on exome capture sequencing and tested 439,170 markers for marginal effects, 83,290 markers for marker × ecotype interactions, and up to 311,445 marker pairs for pairwise interactions. Analyses of pairwise interactions focused on subsets of marker pairs preselected on the basis of marginal marker effects, gene ontology annotation, and pairwise marker associations. Our tests identified 12 significant effects. Homology and gene expression information corroborated seven effects and indicated plausible causal pathways: flowering time and lignin synthesis for plant height; plant growth and senescence for C content and mineral concentration. Four pairwise interactions were detected, including three interactions preselected on the basis of pairwise marker correlations. Furthermore, a marker × ecotype interaction and a pairwise interaction were confirmed in an independent switchgrass panel. Our analyses identified reliable candidate variants for important bioenergy traits. Moreover, they exemplified the importance of interactive effects for depicting genetic bases and illustrated the usefulness of preselecting marker pairs for identifying pairwise marker interactions in association studies.
Collapse
|
12
|
Dong SS, Yao S, Chen YX, Guo Y, Zhang YJ, Niu HM, Hao RH, Shen H, Tian Q, Deng HW, Yang TL. Detecting epistasis within chromatin regulatory circuitry reveals CAND2 as a novel susceptibility gene for obesity. Int J Obes (Lond) 2018; 43:450-456. [PMID: 29717274 DOI: 10.1038/s41366-018-0069-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/14/2018] [Accepted: 02/13/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Genome-wide association studies have identified many susceptibility loci for obesity. However, missing heritability problem is still challenging and ignorance of genetic interactions is believed to be an important cause. Current methods for detecting interactions usually do not consider regulatory elements in non-coding regions. Interaction analyses within chromatin regulatory circuitry may identify new susceptibility loci. METHODS We developed a pipeline named interaction analyses within chromatin regulatory circuitry (IACRC), to identify genetic interactions impacting body mass index (BMI). Potential interacting SNP pairs were obtained based on Hi-C datasets, PreSTIGE (Predicting Specific Tissue Interactions of Genes and Enhancers) algorithm, and super enhancer regions. SNP × SNP analyses were next performed in three GWAS datasets, including 2286 unrelated Caucasians from Kansas City, 3062 healthy Caucasians from the Gene Environment Association Studies initiative, and 3164 Hispanic subjects from the Women's Health Initiative. RESULTS A total of 16,643,227 SNP × SNP analyses were performed. Meta-analyses showed that two SNP pairs, rs6808450-rs9813534 (combined P = 2.39 × 10-9) and rs6808450-rs3773306 (combined P = 2.89 × 10-9) were associated with BMI after multiple testing corrections. Single-SNP analyses did not detect significant association signals for these three SNPs. In obesity relevant cells, rs6808450 is located in intergenic enhancers, while rs9813534 and rs3773306 are located in the region of strong transcription regions of CAND2 and RPL32, respectively. The expression of CAND2 was significantly downregulated after the differentiation of human Simpson-Golabi-Behmel syndrome (SGBS) preadipocyte cells (P = 0.0241). Functional validation in the International Mouse Phenotyping Consortium database showed that CAND2 was associated with increased lean body mass and decreased total body fat amount. CONCLUSIONS Detecting epistasis within chromatin regulatory circuitry identified CAND2 as a novel obesity susceptibility gene. We hope IACRC could facilitate the interaction analyses for complex diseases and offer new insights into solving the missing heritability problem.
Collapse
Affiliation(s)
- Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shi Yao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yi-Xiao Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yu-Jie Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Hui-Min Niu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Ruo-Han Hao
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Hui Shen
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Qing Tian
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Hong-Wen Deng
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| |
Collapse
|
13
|
Ritchie MD, Van Steen K. The search for gene-gene interactions in genome-wide association studies: challenges in abundance of methods, practical considerations, and biological interpretation. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:157. [PMID: 29862246 DOI: 10.21037/atm.2018.04.05] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
One of the primary goals in this era of precision medicine is to understand the biology of human diseases and their treatment, such that each individual patient receives the best possible treatment for their disease based on their genetic and environmental exposures. One way to work towards achieving this goal is to identify the environmental exposures and genetic variants that are relevant to each disease in question, as well as the complex interplay between genes and environment. Genome-wide association studies (GWAS) have allowed for a greater understanding of the genetic component of many complex traits. However, these genetic effects are largely small and thus, our ability to use these GWAS finding for precision medicine is limited. As more and more GWAS have been performed, rather than focusing only on common single nucleotide polymorphisms (SNPs) and additive genetic models, many researchers have begun to explore alternative heritable components of complex traits including rare variants, structural variants, epigenetics, and genetic interactions. While genetic interactions are a plausible reality that could explain some of the heritabliy that has not yet been identified, especially when one considers the identification of genetic interactions in model organisms as well as our understanding of biological complexity, still there are significant challenges and considerations in identifying these genetic interactions. Broadly, these can be summarized in three categories: abundance of methods, practical considerations, and biological interpretation. In this review, we will discuss these important elements in the search for genetic interactions along with some potential solutions. While genetic interactions are theoretically understood to be important for complex human disease, the body of evidence is still building to support this component of the underlying genetic architecture of complex human traits. Our hope is that more sophisticated modeling approaches and more robust computational techniques will enable the community to identify these important genetic interactions and improve our ability to implement precision medicine in the future.
Collapse
Affiliation(s)
- Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristel Van Steen
- WELBIO, GIGA-R Medical Genomics Unit - BIO3, University of Liège, Liège, Belgium.,Department of Human Genetics, University of Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Verma SS, Ritchie MD. Another Round of "Clue" to Uncover the Mystery of Complex Traits. Genes (Basel) 2018; 9:E61. [PMID: 29370075 PMCID: PMC5852557 DOI: 10.3390/genes9020061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/19/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
A plethora of genetic association analyses have identified several genetic risk loci. Technological and statistical advancements have now led to the identification of not only common genetic variants, but also low-frequency variants, structural variants, and environmental factors, as well as multi-omics variations that affect the phenotypic variance of complex traits in a population, thus referred to as complex trait architecture. The concept of heritability, or the proportion of phenotypic variance due to genetic inheritance, has been studied for several decades, but its application is mainly in addressing the narrow sense heritability (or additive genetic component) from Genome-Wide Association Studies (GWAS). In this commentary, we reflect on our perspective on the complexity of understanding heritability for human traits in comparison to model organisms, highlighting another round of clues beyond GWAS and an alternative approach, investigating these clues comprehensively to help in elucidating the genetic architecture of complex traits.
Collapse
Affiliation(s)
- Shefali Setia Verma
- The Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Marylyn D Ritchie
- The Huck Institute of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
Matalliotakis M, Zervou MI, Matalliotaki C, Rahmioglu N, Koumantakis G, Kalogiannidis I, Prapas I, Zondervan K, Spandidos DA, Matalliotakis I, Goulielmos GN. The role of gene polymorphisms in endometriosis. Mol Med Rep 2017; 16:5881-5886. [PMID: 28901453 PMCID: PMC5865763 DOI: 10.3892/mmr.2017.7398] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/29/2017] [Indexed: 11/06/2022] Open
Abstract
Endometriosis is a benign gynecologic disorder, affecting up to 10% of women, characterized by the presence of functional endometrial tissue at ectopic positions generally within the peritoneum. It is a heritable condition influenced by multiple genetic and environmental factors, with an overall heritability estimated at approximately 50%. In this study, we investigated whether single nucleotide polymorphisms (SNPs) rs7521902, rs10859871 and rs11031006, mapping to WNT4, VEZT and FSHB genetic loci, respectively, are associated with risk for endometriosis in a Greek population. This study included 166 women with histologically confirmed endometriosis diagnosed through surgery and 150 normal controls. Genotyping of the rs7521902, rs10859871 and rs11031006 SNPs was performed with Taqman primer/probe sets. A significant association was detected with the AC genotype of rs7521902 (WNT4) in patients with stage III and IV disease only. Evidence for association with endometriosis was also found for the AC genotype of the rs10859871 of VEZT. Notably, a significant difference in the distribution of the AG genotype and the minor allele A of FSHB rs11031006 SNP was found between the endometriosis patients and controls. We found a genetic association between rs11031006 (FSHB) SNP and endometriosis. WNT4 and VEZT genes constitute the most consistently associated genes with endometriosis. In the present study, an association of rs7521902 (WNT4) and rs10859871 (VEZT) was confirmed in women with endometriosis at the genotypic but not the allelic level.
Collapse
Affiliation(s)
- Michail Matalliotakis
- Department of Obstetrics and Gynecology, Venizeleio General Hospital, Heraklion 714 09, Crete, Greece
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion 710 03, Crete, Greece
| | - Maria I. Zervou
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion 710 03, Crete, Greece
| | - Charoula Matalliotaki
- Department of Obstetrics and Gynecology, Venizeleio General Hospital, Heraklion 714 09, Crete, Greece
| | - Nilufer Rahmioglu
- Wellcome Trust Centre for Human Genetics University of Oxford, Oxford OX1 1JD, UK
- Endometriosis CaRe Centre, Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford OX3 7BN, UK
| | | | - Ioannis Kalogiannidis
- 3rd Department of Obstetrics and Gynaecology, Aristotle University of Thessaloniki, Thessaloniki 541 24, Crete, Greece
| | - Ioannis Prapas
- IAKENTRO, Infertility Treatment Center, Thessaloniki 542 50, Crete, Greece
| | - Krina Zondervan
- Wellcome Trust Centre for Human Genetics University of Oxford, Oxford OX1 1JD, UK
- Endometriosis CaRe Centre, Nuffield Department of Obstetrics and Gynaecology, University of Oxford, Oxford OX3 7BN, UK
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion 710 03, Crete, Greece
| | - Ioannis Matalliotakis
- Department of Obstetrics and Gynecology, Venizeleio General Hospital, Heraklion 714 09, Crete, Greece
| | - George N. Goulielmos
- Section of Molecular Pathology and Human Genetics, Department of Internal Medicine, School of Medicine, University of Crete, Heraklion 710 03, Crete, Greece
| |
Collapse
|
16
|
Wang Z, Manichukal A, Goff DC, Mora S, Ordovas JM, Pajewski NM, Post WS, Rotter JI, Sale MM, Santorico SA, Siscovick D, Tsai MY, Arnett DK, Rich S, Frazier-Wood AC. Genetic associations with lipoprotein subfraction measures differ by ethnicity in the multi-ethnic study of atherosclerosis (MESA). Hum Genet 2017; 136:715-726. [PMID: 28352986 PMCID: PMC5429342 DOI: 10.1007/s00439-017-1782-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/16/2017] [Indexed: 12/25/2022]
Abstract
A recent genome-wide association study associated 62 single nucleotide polymorphisms (SNPs) from 43 genomic loci, with fasting lipoprotein subfractions in European-Americans (EAs) at genome-wide levels of significance across three independent samples. Whether these associations are consistent across ethnicities with a non-European ancestry is unknown. We analyzed 15 lipoprotein subfraction measures, on 1677 African-Americans (AAs), 1450 Hispanic-Americans (HAs), and 775 Chinese-Americans (CHN) participating in the multi-ethnic study of atherosclerosis (MESA). Genome-wide data were obtained using the Affymetrix 6.0 and Illumina HumanOmni chips. Linear regression models between genetic variables and lipoprotein subfractions were adjusted for age, gender, body mass index, smoking, study center, and genetic ancestry (based on principal components), and additionally adjusted for Mexican/Non-Mexican status in HAs. A false discovery rate correction was applied separately within the results for each ethnicity to correct for multiple testing. Power calculations revealed that we did not have the power for SNP-based measures of association, so we analyzed phenotype-specific genetic risk scores (GRSs), constructed as in the original genome-wide analysis. We successfully replicated all 15 GRS-lipoprotein associations in 2527 EAs. Among the 15 significant GRS-lipoprotein associations in EAs, 11 were significant in AAs, 13 in HAs, and 1 in CHNs. Further analyses revealed that ethnicity differences could not be explained by differences in linkage disequilibrium, lipid lowering drugs, diabetes, or gender. Our study emphasizes the importance of ethnicity (here indexing genetic ancestry) in genetic risk for CVD and highlights the need to identify ethnicity-specific genetic variants associated with CVD risk.
Collapse
Affiliation(s)
- Zhe Wang
- Department of Epidemiology, Human Genetics and Environmental Health, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Ani Manichukal
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - David C Goff
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO, 80045, USA
| | - Samia Mora
- Divisions of Preventive Medicine and Cardiovascular Medicine Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, Jean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University, Boston, MA, 02111, USA
- The Department of Epidemiology and Population Genetics, Centro Nacional Investigación Cardiovasculares (CNIC), 28029, Madrid, Spain
- IMDEA Food, 28049, Madrid, Spain
| | - Nicholas M Pajewski
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Jerome I Rotter
- Los Angeles Biomedical Research Institute and Department of Pediatrics, Institute for Translational Genomics and Population Sciences, Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Michele M Sale
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Stephanie A Santorico
- Department of Mathematical and Statistical Sciences, Human Medical Genetics and Genomics Program, Department of Biostatistics & Informatics, University of Colorado Denver, Denver, CO, 80204, USA
| | - David Siscovick
- Cardiovascular Health Research Unit, Department of Medicine and Epidemiology, University of Washington, Seattle, WA, 98195, USA
- The New York Academy of Medicine, New York, NY, 10029, USA
| | - Michael Y Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, 40508, USA
| | - Stephen Rich
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Alexis C Frazier-Wood
- Department of Pediatrics, USDA/ARS Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Jiang J, Shen B, O’Connell JR, VanRaden PM, Cole JB, Ma L. Dissection of additive, dominance, and imprinting effects for production and reproduction traits in Holstein cattle. BMC Genomics 2017; 18:425. [PMID: 28558656 PMCID: PMC5450346 DOI: 10.1186/s12864-017-3821-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/25/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Although genome-wide association and genomic selection studies have primarily focused on additive effects, dominance and imprinting effects play an important role in mammalian biology and development. The degree to which these non-additive genetic effects contribute to phenotypic variation and whether QTL acting in a non-additive manner can be detected in genetic association studies remain controversial. RESULTS To empirically answer these questions, we analyzed a large cattle dataset that consisted of 42,701 genotyped Holstein cows with genotyped parents and phenotypic records for eight production and reproduction traits. SNP genotypes were phased in pedigree to determine the parent-of-origin of alleles, and a three-component GREML was applied to obtain variance decomposition for additive, dominance, and imprinting effects. The results showed a significant non-zero contribution from dominance to production traits but not to reproduction traits. Imprinting effects significantly contributed to both production and reproduction traits. Interestingly, imprinting effects contributed more to reproduction traits than to production traits. Using GWAS and imputation-based fine-mapping analyses, we identified and validated a dominance association signal with milk yield near RUNX2, a candidate gene that has been associated with milk production in mice. When adding non-additive effects into the prediction models, however, we observed little or no increase in prediction accuracy for the eight traits analyzed. CONCLUSIONS Collectively, our results suggested that non-additive effects contributed a non-negligible amount (more for reproduction traits) to the total genetic variance of complex traits in cattle, and detection of QTLs with non-additive effect is possible in GWAS using a large dataset.
Collapse
Affiliation(s)
- Jicai Jiang
- Department of Animal and Avian Sciences, University of Maryland, 2123 Animal Science Building, College Park, MD 20742 USA
| | - Botong Shen
- Department of Animal and Avian Sciences, University of Maryland, 2123 Animal Science Building, College Park, MD 20742 USA
| | | | - Paul M. VanRaden
- Animal Genomics and Improvement Laboratory, USDA, Building 5, Beltsville, MD 20705 USA
| | - John B. Cole
- Animal Genomics and Improvement Laboratory, USDA, Building 5, Beltsville, MD 20705 USA
| | - Li Ma
- Department of Animal and Avian Sciences, University of Maryland, 2123 Animal Science Building, College Park, MD 20742 USA
| |
Collapse
|
18
|
Monir MM, Zhu J. Comparing GWAS Results of Complex Traits Using Full Genetic Model and Additive Models for Revealing Genetic Architecture. Sci Rep 2017; 7:38600. [PMID: 28079101 PMCID: PMC5227710 DOI: 10.1038/srep38600] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/25/2016] [Indexed: 01/09/2023] Open
Abstract
Most of the genome-wide association studies (GWASs) for human complex diseases have ignored dominance, epistasis and ethnic interactions. We conducted comparative GWASs for total cholesterol using full model and additive models, which illustrate the impacts of the ignoring genetic variants on analysis results and demonstrate how genetic effects of multiple loci could differ across different ethnic groups. There were 15 quantitative trait loci with 13 individual loci and 3 pairs of epistasis loci identified by full model, whereas only 14 loci (9 common loci and 5 different loci) identified by multi-loci additive model. Again, 4 full model detected loci were not detected using multi-loci additive model. PLINK-analysis identified two loci and GCTA-analysis detected only one locus with genome-wide significance. Full model identified three previously reported genes as well as several new genes. Bioinformatics analysis showed some new genes are related with cholesterol related chemicals and/or diseases. Analyses of cholesterol data and simulation studies revealed that the full model performs were better than the additive-model performs in terms of detecting power and unbiased estimations of genetic variants of complex traits.
Collapse
Affiliation(s)
- Md Mamun Monir
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| | - Jun Zhu
- Institute of Bioinformatics, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
19
|
Leduc V, Bourque L, Poirier J, Dufour R. Role of rs3846662 and HMGCR alternative splicing in statin efficacy and baseline lipid levels in familial hypercholesterolemia. Pharmacogenet Genomics 2016; 26:1-11. [PMID: 26466344 DOI: 10.1097/fpc.0000000000000178] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To assess the contribution of the rs3846662 polymorphism of HMGCR on serum lipid levels and statin efficacy, we measured in vivo HMGCR mRNA and lipid levels in French Canadian individuals affected by heterozygous familial hypercholesterolemia due to the deletion of more than 15 kb of the LDLR gene. RESULTS Men and women carrying the AA genotype at rs3846662, and no APOE4 allele, had higher levels of total cholesterol (5.43 vs. 4.58 mmol/l, P<0.05) and LDL-cholesterol (5.20 vs. 4.39 mmol/l, P<0.05) at baseline. However, with regard to statin efficacy, the penetrance of the AA genotype was sex dependent. Indeed, the percentage reduction in LDL-cholesterol upon statin treatment was significantly decreased in women with the AA genotype compared with women without it (38.4 vs. 46.2%, P<0.05), whereas this was not observed in men. Although both men and women bearing the AA genotype showed a higher ratio of full-length HMGCR mRNA/total HMGCR mRNA compared with individuals without it (n=37, P<0.05), overall transcription of HMGCR was decreased and increased in men and women carrying this genotype, respectively (n=37, P<0.01 and P<0.05). Finally, in our familial hypercholesterolemia cohort, HMGCR alternative splicing explained between 22 and 55% of the variance in statin response. CONCLUSION rs3846662 polymorphism and the alternative splicing of HMGCR mRNA significantly impact women's response to statin therapy.
Collapse
Affiliation(s)
- Valerie Leduc
- aCentre for Studies in Alzheimer's disease prevention bDouglas Mental Health University Institute, McGill University cDepartment of Nutrition, Clinical Research Institute of Montreal (IRCM), Montreal University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
20
|
Zhou D, Li Z, Yu D, Wan L, Zhu Y, Lai M, Zhang D. Polymorphisms involving gain or loss of CpG sites are significantly enriched in trait-associated SNPs. Oncotarget 2016; 6:39995-40004. [PMID: 26503467 PMCID: PMC4741875 DOI: 10.18632/oncotarget.5650] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/02/2015] [Indexed: 12/30/2022] Open
Abstract
Some single nucleotide polymorphisms (SNPs) influence the existence of CpG sites, the basis of DNA modification such as methylation and hydroxymethylation. These polymorphisms can lead to gain or loss of CpG sites and were defined as CpG site related SNPs (cgSNPs) in this study. The cgSNPs change DNA sequence and might potentially affect DNA modification such as methylation. However, the functional consequence of cgSNPs is poorly understood. We observed that a considerable proportion (23.0%) of common variants were cgSNPs in human genome. Mutations involving loss of CpG sites were associated with reduced levels of methylation (~20.2%) using The Cancer Genome Atlas (TCGA) data. Using public databases (SCAN and seeQTL) of expression quantitative trait loci (eQTLs), we found that the cgSNPs were significantly enriched in eQTLs via logistic regression and simulation test. Furthermore, we observed that cgSNPs were more likely to be trait-associated loci especially cancers using a catalog of published genome-wide association studies (GWAS) recorded by National Human Genome Research Institute (NHGRI). Our results indicated that cgSNP might be meaningful as annotation either in SNP functional prediction or in screening for trait-associated SNPs.
Collapse
Affiliation(s)
- Dan Zhou
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China.,Department of Epidemiology & Biostatistics, Zhejiang University School of Public Health, Hangzhou, Zhejiang, 310058, China
| | - Zhenli Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Dan Yu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Ledong Wan
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, Zhejiang University School of Public Health, Hangzhou, Zhejiang, 310058, China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
21
|
Pathway Analysis Incorporating Protein-Protein Interaction Networks Identified Candidate Pathways for the Seven Common Diseases. PLoS One 2016; 11:e0162910. [PMID: 27622767 PMCID: PMC5021324 DOI: 10.1371/journal.pone.0162910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 08/30/2016] [Indexed: 01/08/2023] Open
Abstract
Pathway analysis has become popular as a secondary analysis strategy for genome-wide association studies (GWAS). Most of the current pathway analysis methods aggregate signals from the main effects of single nucleotide polymorphisms (SNPs) in genes within a pathway without considering the effects of gene-gene interactions. However, gene-gene interactions can also have critical effects on complex diseases. Protein-protein interaction (PPI) networks have been used to define gene pairs for the gene-gene interaction tests. Incorporating the PPI information to define gene pairs for interaction tests within pathways can increase the power for pathway-based association tests. We propose a pathway association test, which aggregates the interaction signals in PPI networks within a pathway, for GWAS with case-control samples. Gene size is properly considered in the test so that genes do not contribute more to the test statistic simply due to their size. Simulation studies were performed to verify that the method is a valid test and can have more power than other pathway association tests in the presence of gene-gene interactions within a pathway under different scenarios. We applied the test to the Wellcome Trust Case Control Consortium GWAS datasets for seven common diseases. The most significant pathway is the chaperones modulate interferon signaling pathway for Crohn’s disease (p-value = 0.0003). The pathway modulates interferon gamma, which induces the JAK/STAT pathway that is involved in Crohn’s disease. Several other pathways that have functional implications for the seven diseases were also identified. The proposed test based on gene-gene interaction signals in PPI networks can be used as a complementary tool to the current existing pathway analysis methods focusing on main effects of genes. An efficient software implementing the method is freely available at http://puppi.sourceforge.net.
Collapse
|
22
|
Verma SS, Cooke Bailey JN, Lucas A, Bradford Y, Linneman JG, Hauser MA, Pasquale LR, Peissig PL, Brilliant MH, McCarty CA, Haines JL, Wiggs JL, Vrabec TR, Tromp G, Ritchie MD. Epistatic Gene-Based Interaction Analyses for Glaucoma in eMERGE and NEIGHBOR Consortium. PLoS Genet 2016; 12:e1006186. [PMID: 27623284 PMCID: PMC5021356 DOI: 10.1371/journal.pgen.1006186] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/22/2016] [Indexed: 12/22/2022] Open
Abstract
Primary open angle glaucoma (POAG) is a complex disease and is one of the major leading causes of blindness worldwide. Genome-wide association studies have successfully identified several common variants associated with glaucoma; however, most of these variants only explain a small proportion of the genetic risk. Apart from the standard approach to identify main effects of variants across the genome, it is believed that gene-gene interactions can help elucidate part of the missing heritability by allowing for the test of interactions between genetic variants to mimic the complex nature of biology. To explain the etiology of glaucoma, we first performed a genome-wide association study (GWAS) on glaucoma case-control samples obtained from electronic medical records (EMR) to establish the utility of EMR data in detecting non-spurious and relevant associations; this analysis was aimed at confirming already known associations with glaucoma and validating the EMR derived glaucoma phenotype. Our findings from GWAS suggest consistent evidence of several known associations in POAG. We then performed an interaction analysis for variants found to be marginally associated with glaucoma (SNPs with main effect p-value <0.01) and observed interesting findings in the electronic MEdical Records and GEnomics Network (eMERGE) network dataset. Genes from the top epistatic interactions from eMERGE data (Likelihood Ratio Test i.e. LRT p-value <1e-05) were then tested for replication in the NEIGHBOR consortium dataset. To replicate our findings, we performed a gene-based SNP-SNP interaction analysis in NEIGHBOR and observed significant gene-gene interactions (p-value <0.001) among the top 17 gene-gene models identified in the discovery phase. Variants from gene-gene interaction analysis that we found to be associated with POAG explain 3.5% of additional genetic variance in eMERGE dataset above what is explained by the SNPs in genes that are replicated from previous GWAS studies (which was only 2.1% variance explained in eMERGE dataset); in the NEIGHBOR dataset, adding replicated SNPs from gene-gene interaction analysis explain 3.4% of total variance whereas GWAS SNPs alone explain only 2.8% of variance. Exploring gene-gene interactions may provide additional insights into many complex traits when explored in properly designed and powered association studies. The complex nature of primary-open angle glaucoma (POAG) has left researchers exploring the genetic architecture and searching for the missing heritability using a number of different study designs. Over the past decade, many studies have been conducted to explain the etiology of POAG; however, a high proportion of estimated heritability still remains unexplained. GWA studies for POAG have identified significant associations but these associations have only explained a small proportion of the genetic risk (odds ratios range between 1–3). In this paper, we sought to confirm the primary genome-wide significant associations that have been discovered so far for glaucoma in phenotypes developed from EMR data in an effort to show that EMR data can be a powerful resource for finding genetic variants influencing POAG susceptibility. Next, we tested for statistical interactions, which can be presented as an important tool in an attempt to explain POAG heritability. We used a reduced list of variants filtered by marginal main effect analysis to look for epistatic interactions. We present our results from replication of gene-based interaction analyses performed in eMERGE and the NEIGHBOR consortium data. Using expression data and annotations from various publicly available databases, the most significant genes that replicated in our analyses show expression in the eye and trabecular meshwork. Analysis for estimation of genetic variance explained by significant associations from previous GWAS and replicated variants from gene-based interactions suggest that these explain 5.6% of variance in eMERGE dataset and also explain 3.4% variance in NEIGHBOR dataset.
Collapse
Affiliation(s)
- Shefali Setia Verma
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, Pennsylvania, United States of America
- The Huck Institute of Life Sciences, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jessica N. Cooke Bailey
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Anastasia Lucas
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Yuki Bradford
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - James G. Linneman
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, United States of America
| | - Michael A. Hauser
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Louis R. Pasquale
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peggy L. Peissig
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, United States of America
| | - Murray H. Brilliant
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, Wisconsin, United States of America
| | | | - Jonathan L. Haines
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Janey L. Wiggs
- Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, United States of America
| | - Tamara R. Vrabec
- Department of Ophthalmology, Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Marylyn D. Ritchie
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, Pennsylvania, United States of America
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| | | | | |
Collapse
|
23
|
A Novel Test for Detecting SNP-SNP Interactions in Case-Only Trio Studies. Genetics 2016; 202:1289-97. [PMID: 26865367 DOI: 10.1534/genetics.115.179846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 01/27/2016] [Indexed: 02/06/2023] Open
Abstract
Epistasis plays a significant role in the genetic architecture of many complex phenotypes in model organisms. To date, there have been very few interactions replicated in human studies due in part to the multiple-hypothesis burden implicit in genome-wide tests of epistasis. Therefore, it is of paramount importance to develop the most powerful tests possible for detecting interactions. In this work we develop a new SNP-SNP interaction test for use in case-only trio studies called the trio correlation (TC) test. The TC test computes the expected joint distribution of marker pairs in offspring conditional on parental genotypes. This distribution is then incorporated into a standard 1 d.f. correlation test of interaction. We show via extensive simulations under a variety of disease models that our test substantially outperforms existing tests of interaction in case-only trio studies. We also demonstrate a bias in a previous case-only trio interaction test and identify its origin. Finally, we show that a previously proposed permutation scheme in trio studies mitigates the known biases of case-only tests in the presence of population stratification. We conclude that the TC test shows improved power to identify interactions in existing, as well as emerging, trio association studies. The method is publicly available at www.github.com/BrunildaBalliu/TrioEpi.
Collapse
|
24
|
Verma SS, Frase AT, Verma A, Pendergrass SA, Mahony S, Haas DW, Ritchie MD. PHENOME-WIDE INTERACTION STUDY (PheWIS) IN AIDS CLINICAL TRIALS GROUP DATA (ACTG). PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2016; 21:57-68. [PMID: 26776173 PMCID: PMC4722952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Association studies have shown and continue to show a substantial amount of success in identifying links between multiple single nucleotide polymorphisms (SNPs) and phenotypes. These studies are also believed to provide insights toward identification of new drug targets and therapies. Albeit of all the success, challenges still remain for applying and prioritizing these associations based on available biological knowledge. Along with single variant association analysis, genetic interactions also play an important role in uncovering the etiology and progression of complex traits. For gene-gene interaction analysis, selection of the variants to test for associations still poses a challenge in identifying epistatic interactions among the large list of variants available in high-throughput, genome-wide datasets. Therefore in this study, we propose a pipeline to identify interactions among genetic variants that are associated with multiple phenotypes by prioritizing previously published results from main effect association analysis (genome-wide and phenome-wide association analysis) based on a-priori biological knowledge in AIDS Clinical Trials Group (ACTG) data. We approached the prioritization and filtration of variants by using the results of a previously published single variant PheWAS and then utilizing biological information from the Roadmap Epigenome project. We removed variants in low functional activity regions based on chromatin states annotation and then conducted an exhaustive pairwise interaction search using linear regression analysis. We performed this analysis in two independent pre-treatment clinical trial datasets from ACTG to allow for both discovery and replication. Using a regression framework, we observed 50,798 associations that replicate at p-value 0.01 for 26 phenotypes, among which 2,176 associations for 212 unique SNPs for fasting blood glucose phenotype reach Bonferroni significance and an additional 9,970 interactions for high-density lipoprotein (HDL) phenotype and fasting blood glucose (total of 12,146 associations) reach FDR significance. We conclude that this method of prioritizing variants to look for epistatic interactions can be used extensively for generating hypotheses for genomewide and phenome-wide interaction analyses. This original Phenome-wide Interaction study (PheWIS) can be applied further to patients enrolled in randomized clinical trials to establish the relationship between patient's response to a particular drug therapy and non-linear combination of variants that might be affecting the outcome.
Collapse
Affiliation(s)
- Shefali S Verma
- Center for System Genomics, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015. [PMID: 26620375 DOI: 10.1007/s11745‐015‐4096‐7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada
- Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada
- Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
26
|
Alphonse PAS, Jones PJH. Revisiting Human Cholesterol Synthesis and Absorption: The Reciprocity Paradigm and its Key Regulators. Lipids 2015; 51:519-36. [PMID: 26620375 DOI: 10.1007/s11745-015-4096-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/09/2015] [Indexed: 12/22/2022]
Abstract
Hypercholesterolemia is a major risk factor for cardiovascular disease. Cholesterol homeostasis in the body is governed by the interplay between absorption, synthesis, and excretion or conversion of cholesterol into bile acids. A reciprocal relationship between cholesterol synthesis and absorption is known to regulate circulating cholesterol in response to dietary or therapeutic interventions. However, the degree to which these factors affect synthesis and absorption and the extent to which one vector shifts in response to the other are not thoroughly understood. Also, huge inter-individual variability exists in the manner in which the two systems act in response to any cholesterol-lowering treatment. Various factors are known to account for this variability and in light of recent experimental advances new players such as gene-gene interactions, gene-environmental effects, and gut microbiome hold immense potential in offering an explanation to the complex traits of inter-individual variability in human cholesterol metabolism. In this context, the objective of the present review is to provide an overview on cholesterol metabolism and discuss the role of potential factors such as genetics, epigenetics, epistasis, and gut microbiome, as well as other regulators in modulating cholesterol metabolism, especially emphasizing the reciprocal relationship between cholesterol synthesis and absorption. Furthermore, an evaluation of the implications of this push-pull mechanism on cholesterol-lowering strategies is presented.
Collapse
Affiliation(s)
- Peter A S Alphonse
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada. .,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.
| | - Peter J H Jones
- Human Nutritional Sciences, University of Manitoba, Winnipeg, MB, Canada.,Richardson Centre for Functional Foods and Nutraceuticals (RCFFN), University of Manitoba, 196, Innovation Drive, SmartPark, Winnipeg, MB, R3T 2N2, Canada.,Food Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
27
|
Yu Z, Demetriou M, Gillen DL. Genome-Wide Analysis of Gene-Gene and Gene-Environment Interactions Using Closed-Form Wald Tests. Genet Epidemiol 2015; 39:446-55. [PMID: 26095143 DOI: 10.1002/gepi.21907] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 02/25/2015] [Accepted: 05/06/2015] [Indexed: 01/31/2023]
Abstract
Despite the successful discovery of hundreds of variants for complex human traits using genome-wide association studies, the degree to which genes and environmental risk factors jointly affect disease risk is largely unknown. One obstacle toward this goal is that the computational effort required for testing gene-gene and gene-environment interactions is enormous. As a result, numerous computationally efficient tests were recently proposed. However, the validity of these methods often relies on unrealistic assumptions such as additive main effects, main effects at only one variable, no linkage disequilibrium between the two single-nucleotide polymorphisms (SNPs) in a pair or gene-environment independence. Here, we derive closed-form and consistent estimates for interaction parameters and propose to use Wald tests for testing interactions. The Wald tests are asymptotically equivalent to the likelihood ratio tests (LRTs), largely considered to be the gold standard tests but generally too computationally demanding for genome-wide interaction analysis. Simulation studies show that the proposed Wald tests have very similar performances with the LRTs but are much more computationally efficient. Applying the proposed tests to a genome-wide study of multiple sclerosis, we identify interactions within the major histocompatibility complex region. In this application, we find that (1) focusing on pairs where both SNPs are marginally significant leads to more significant interactions when compared to focusing on pairs where at least one SNP is marginally significant; and (2) parsimonious parameterization of interaction effects might decrease, rather than increase, statistical power.
Collapse
Affiliation(s)
- Zhaoxia Yu
- Department of Statistics, University of California, Irvine, California, United States of America
| | - Michael Demetriou
- Department of Neurology, University of California, Irvine, California, United States of America.,Department of Microbiology & Molecular Genetics, University of California, Irvine, California, United States of America
| | - Daniel L Gillen
- Department of Statistics, University of California, Irvine, California, United States of America
| |
Collapse
|
28
|
Murk W, Bracken MB, DeWan AT. Confronting the missing epistasis problem: on the reproducibility of gene-gene interactions. Hum Genet 2015; 134:837-49. [PMID: 25998948 DOI: 10.1007/s00439-015-1564-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/06/2015] [Indexed: 11/28/2022]
Abstract
Epistasis (gene-gene interaction) is thought to play an integral role in the genetic basis of complex traits, and a significant amount of research has been invested into identifying this phenomenon in human disease. However, the overall success of empirical studies of epistasis in humans is unclear, as such studies are rarely systematically evaluated. Here, we have selected asthma as an example of a well-studied, complex human disease, and provide a critical analysis and replication attempt of nearly all prior reports of epistasis for this disease. Of 191 previously reported interactions, we find that 39.8% were not originally identified using an explicit test for interaction and thus may not have been true epistatic effects to begin with. Moreover, directions of effect were not described for 46.1% of the interactions, which prevents their rigorous replication. In the original studies, attempts at replication were made for 15.2% of the interactions, and 7.3% were actually replicated. In the current study, we were able to evaluate 85.9% of the interactions using a large asthma dataset from the GABRIEL Consortium. None of these interactions could be replicated based on strict criteria. However, we found nominally significant (p < 0.05) evidence in support of 23.8% of the evaluated interactions. Although many reports of epistasis are not robustly supported in the published literature, our results suggest that at least some of these reports may have been true-positive examples of epistasis. In general, improvements in empirical studies of epistasis are called for, in order to better understand the importance of this phenomenon in human disease.
Collapse
Affiliation(s)
- William Murk
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT, 06510, USA
| | | | | |
Collapse
|
29
|
Hall MA, Verma SS, Wallace J, Lucas A, Berg RL, Connolly J, Crawford DC, Crosslin DR, de Andrade M, Doheny KF, Haines JL, Harley JB, Jarvik GP, Kitchner T, Kuivaniemi H, Larson EB, Carrell DS, Tromp G, Vrabec TR, Pendergrass SA, McCarty CA, Ritchie MD. Biology-Driven Gene-Gene Interaction Analysis of Age-Related Cataract in the eMERGE Network. Genet Epidemiol 2015; 39:376-84. [PMID: 25982363 PMCID: PMC4550090 DOI: 10.1002/gepi.21902] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 02/27/2015] [Accepted: 03/13/2015] [Indexed: 01/19/2023]
Abstract
Bioinformatics approaches to examine gene-gene models provide a means to discover interactions between multiple genes that underlie complex disease. Extensive computational demands and adjusting for multiple testing make uncovering genetic interactions a challenge. Here, we address these issues using our knowledge-driven filtering method, Biofilter, to identify putative single nucleotide polymorphism (SNP) interaction models for cataract susceptibility, thereby reducing the number of models for analysis. Models were evaluated in 3,377 European Americans (1,185 controls, 2,192 cases) from the Marshfield Clinic, a study site of the Electronic Medical Records and Genomics (eMERGE) Network, using logistic regression. All statistically significant models from the Marshfield Clinic were then evaluated in an independent dataset of 4,311 individuals (742 controls, 3,569 cases), using independent samples from additional study sites in the eMERGE Network: Mayo Clinic, Group Health/University of Washington, Vanderbilt University Medical Center, and Geisinger Health System. Eighty-three SNP-SNP models replicated in the independent dataset at likelihood ratio test P < 0.05. Among the most significant replicating models was rs12597188 (intron of CDH1)-rs11564445 (intron of CTNNB1). These genes are known to be involved in processes that include: cell-to-cell adhesion signaling, cell-cell junction organization, and cell-cell communication. Further Biofilter analysis of all replicating models revealed a number of common functions among the genes harboring the 83 replicating SNP-SNP models, which included signal transduction and PI3K-Akt signaling pathway. These findings demonstrate the utility of Biofilter as a biology-driven method, applicable for any genome-wide association study dataset.
Collapse
Affiliation(s)
- Molly A Hall
- Department of Biochemistry and Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Shefali S Verma
- Department of Biochemistry and Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - John Wallace
- Department of Biochemistry and Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Anastasia Lucas
- Department of Biochemistry and Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Richard L Berg
- Marshfield Clinic, Marshfield, Wisconsin, United States of America
| | - John Connolly
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Dana C Crawford
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David R Crosslin
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
| | | | - Kimberly F Doheny
- Center for Inherited Disease Research, IGM, Johns Hopkins University SOM, Baltimore, Maryland, United States of America
| | - Jonathan L Haines
- Department of Epidemiology and Biostatistics, Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - John B Harley
- Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Gail P Jarvik
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America.,Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Terrie Kitchner
- Marshfield Clinic, Marshfield, Wisconsin, United States of America
| | - Helena Kuivaniemi
- Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Eric B Larson
- Group Health Research Institute, Seattle, Washington, United States of America
| | - David S Carrell
- Group Health Research Institute, Seattle, Washington, United States of America
| | - Gerard Tromp
- Geisinger Health System, Danville, Pennsylvania, United States of America
| | - Tamara R Vrabec
- Geisinger Health System, Danville, Pennsylvania, United States of America
| | | | | | - Marylyn D Ritchie
- Department of Biochemistry and Molecular Biology, Center for Systems Genomics, Eberly College of Science, The Pennsylvania State University, University Park, Pennsylvania, United States of America.,Geisinger Health System, Danville, Pennsylvania, United States of America
| |
Collapse
|
30
|
Daya M, van der Merwe L, van Helden PD, Möller M, Hoal EG. Investigating the Role of Gene-Gene Interactions in TB Susceptibility. PLoS One 2015; 10:e0123970. [PMID: 25919455 PMCID: PMC4412713 DOI: 10.1371/journal.pone.0123970] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/24/2015] [Indexed: 11/22/2022] Open
Abstract
Tuberculosis (TB) is the second leading cause of mortality from infectious disease worldwide. One of the factors involved in developing disease is the genetics of the host, yet the field of TB susceptibility genetics has not yielded the answers that were expected. A commonly posited explanation for the missing heritability of complex disease is gene-gene interactions, also referred to as epistasis. In this study we investigate the role of gene-gene interactions in genetic susceptibility to TB using a cohort recruited from a high TB incidence community from Cape Town, South Africa. Our discovery data set incorporates genotypes from a large a number of candidate gene studies as well as genome-wide data. After limiting our search space to pairs of putative TB susceptibility genes, as well as pairs of genes that have been curated in online databases as potential interactors, we use statistical modelling to identify pairs of interacting SNPs. We attempt to validate the top models identified in our discovery data set using an independent genome-wide TB case-control data set from The Gambia. A number of models were successfully validated, indicating that interplay between the NRG1 - NRG3, GRIK1 - GRIK3 and IL23R - ATG4C gene pairs may modify susceptibility to TB. Gene pairs involved in the NF-κB pathway were also identified in the discovery data set (SFTPD - NOD2, ISG15 - TLR8 and NLRC5 - IL12RB1), but could not be tested in the Gambian study group due to lack of overlapping data.
Collapse
Affiliation(s)
- Michelle Daya
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Lize van der Merwe
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Paul D. van Helden
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Marlo Möller
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Eileen G. Hoal
- SA MRC Centre for TB Research, DST/NRF Centre of Excellence for Biomedical Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
31
|
Fitzpatrick DJ, Ryan CJ, Shah N, Greene D, Molony C, Shields DC. Genome-wide epistatic expression quantitative trait loci discovery in four human tissues reveals the importance of local chromosomal interactions governing gene expression. BMC Genomics 2015; 16:109. [PMID: 25765234 PMCID: PMC4345003 DOI: 10.1186/s12864-015-1300-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 01/29/2015] [Indexed: 12/15/2022] Open
Abstract
Background Epistasis (synergistic interaction) among SNPs governing gene expression is likely to arise within transcriptional networks. However, the power to detect it is limited by the large number of combinations to be tested and the modest sample sizes of most datasets. By limiting the interaction search space firstly to cis-trans and then cis-cis SNP pairs where both SNPs had an independent effect on the expression of the most variable transcripts in the liver and brain, we greatly reduced the size of the search space. Results Within the cis-trans search space we discovered three transcripts with significant epistasis. Surprisingly, all interacting SNP pairs were located nearby each other on the chromosome (within 290 kb-2.16 Mb). Despite their proximity, the interacting SNPs were outside the range of linkage disequilibrium (LD), which was absent between the pairs (r2 < 0.01). Accordingly, we redefined the search space to detect cis-cis interactions, where a cis-SNP was located within 10 Mb of the target transcript. The results of this show evidence for the epistatic regulation of 50 transcripts across the tissues studied. Three transcripts, namely, HLA-G, PSORS1C1 and HLA-DRB5 share common regulatory SNPs in the pre-frontal cortex and their expression is significantly correlated. This pattern of epistasis is consistent with mediation via long-range chromatin structures rather than the binding of transcription factors in trans. Accordingly, some of the interactions map to regions of the genome known to physically interact in lymphoblastoid cell lines while others map to known promoter and enhancer elements. SNPs involved in interactions appear to be enriched for promoter markers. Conclusions In the context of gene expression and its regulation, our analysis indicates that the study of cis-cis or local epistatic interactions may have a more important role than interchromosomal interactions. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1300-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Darren J Fitzpatrick
- School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, 4, Ireland.
| | - Colm J Ryan
- School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, 4, Ireland.
| | - Naisha Shah
- School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, 4, Ireland.
| | - Derek Greene
- School of Computer Science and Informatics, University College Dublin, Belfield, Dublin, 4, Ireland.
| | - Cliona Molony
- Merck Research Laboratories, Merck & Co. Inc. 33 Avenue Louis Pasteur, Boston, MA, 02115, USA.
| | - Denis C Shields
- School of Medicine and Medical Sciences, University College Dublin, Belfield, Dublin, 4, Ireland.
| |
Collapse
|
32
|
Musameh MD, Wang WYS, Nelson CP, Lluís-Ganella C, Debiec R, Subirana I, Elosua R, Balmforth AJ, Ball SG, Hall AS, Kathiresan S, Thompson JR, Lucas G, Samani NJ, Tomaszewski M. Analysis of gene-gene interactions among common variants in candidate cardiovascular genes in coronary artery disease. PLoS One 2015; 10:e0117684. [PMID: 25658981 PMCID: PMC4320092 DOI: 10.1371/journal.pone.0117684] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 12/30/2014] [Indexed: 11/19/2022] Open
Abstract
Objective Only a small fraction of coronary artery disease (CAD) heritability has been explained by common variants identified to date. Interactions between genes of importance to cardiovascular regulation may account for some of the missing heritability of CAD. This study aimed to investigate the role of gene-gene interactions in common variants in candidate cardiovascular genes in CAD. Approach and Results 2,101 patients with CAD from the British Heart Foundation Family Heart Study and 2,426 CAD-free controls were included in the discovery cohort. All subjects were genotyped with the Illumina HumanCVD BeadChip enriched for genes and pathways relevant to the cardiovascular system and disease. The primary analysis in the discovery cohort examined pairwise interactions among 913 common (minor allele frequency >0.1) independent single nucleotide polymorphisms (SNPs) with at least nominal association with CAD in single locus analysis. A secondary exploratory interaction analysis was performed among all 11,332 independent common SNPs surviving quality control criteria. Replication analyses were conducted in 2,967 patients and 3,075 controls from the Myocardial Infarction Genetics Consortium. None of the interactions amongst 913 SNPs analysed in the primary analysis was statistically significant after correction for multiple testing (required P<1.2x10-7). Similarly, none of the pairwise gene-gene interactions in the secondary analysis reached statistical significance after correction for multiple testing (required P = 7.8x10-10). None of 36 suggestive interactions from the primary analysis or 31 interactions from the secondary analysis was significant in the replication cohort. Our study had 80% power to detect odds ratios > 1.7 for common variants in the primary analysis. Conclusions Moderately large additive interactions between common SNPs in genes relevant to cardiovascular disease do not appear to play a major role in genetic predisposition to CAD. The role of genetic interactions amongst less common SNPs and with medium and small magnitude effects remain to be investigated.
Collapse
Affiliation(s)
- Muntaser D. Musameh
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
- * E-mail:
| | - William Y. S. Wang
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | | | - Radoslaw Debiec
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Isaac Subirana
- Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain
- Epidemiology and Public Health Network (CIBERESP), Barcelona, Spain
| | - Roberto Elosua
- Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain
| | - Anthony J. Balmforth
- Division of Epidemiology, LIGHT, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Stephen G. Ball
- University of Leeds, MCRC, Leeds Institute of Genetics, Health and Therapeutics, Leeds, United Kingdom
| | - Alistair S. Hall
- Division of Epidemiology, LIGHT, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Sekar Kathiresan
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - John R. Thompson
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
| | - Gavin Lucas
- Cardiovascular Epidemiology and Genetics, IMIM, Barcelona, Spain
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| | - Maciej Tomaszewski
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, United Kingdom
- NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom
| |
Collapse
|
33
|
Ma L, Keinan A, Clark AG. Biological knowledge-driven analysis of epistasis in human GWAS with application to lipid traits. Methods Mol Biol 2015; 1253:35-45. [PMID: 25403526 DOI: 10.1007/978-1-4939-2155-3_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
While the importance of epistasis is well established, specific gene-gene interactions have rarely been identified in human genome-wide association studies (GWAS), mainly due to low power associated with such interaction tests. In this chapter, we integrate biological knowledge and human GWAS data to reveal epistatic interactions underlying quantitative lipid traits, which are major risk factors for coronary artery disease. To increase power to detect interactions, we only tested pairs of SNPs filtered by prior biological knowledge, including GWAS results, protein-protein interactions (PPIs), and pathway information. Using published GWAS and 9,713 European Americans (EA) from the Atherosclerosis Risk in Communities (ARIC) study, we identified an interaction between HMGCR and LIPC affecting high-density lipoprotein cholesterol (HDL-C) levels. We then validated this interaction in additional multiethnic cohorts from ARIC, the Framingham Heart Study, and the Multi-Ethnic Study of Atherosclerosis. Both HMGCR and LIPC are involved in the metabolism of lipids and lipoproteins, and LIPC itself has been marginally associated with HDL-C. Furthermore, no significant interaction was detected using PPI and pathway information, mainly due to the stringent significance level required after correcting for the large number of tests conducted. These results suggest the potential of biological knowledge-driven approaches to detect epistatic interactions in human GWAS, which may hold the key to exploring the role gene-gene interactions play in connecting genotypes and complex phenotypes in future GWAS.
Collapse
Affiliation(s)
- Li Ma
- Department of Animal and Avian Sciences, University of Maryland, Bldg 142, College Park, MD, 20742, USA,
| | | | | |
Collapse
|
34
|
Epistasis between SNPs in genes involved in lipoprotein metabolism influences high- and low-density lipoprotein cholesterol levels. Genes Genomics 2014. [DOI: 10.1007/s13258-014-0216-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Parnell LD, Blokker BA, Dashti HS, Nesbeth PD, Cooper BE, Ma Y, Lee YC, Hou R, Lai CQ, Richardson K, Ordovás JM. CardioGxE, a catalog of gene-environment interactions for cardiometabolic traits. BioData Min 2014; 7:21. [PMID: 25368670 PMCID: PMC4217104 DOI: 10.1186/1756-0381-7-21] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 10/18/2014] [Indexed: 12/29/2022] Open
Abstract
Background Genetic understanding of complex traits has developed immensely over the past decade but remains hampered by incomplete descriptions of contribution to phenotypic variance. Gene-environment (GxE) interactions are one of these contributors and in the guise of diet and physical activity are important modulators of cardiometabolic phenotypes and ensuing diseases. Results We mined the scientific literature to collect GxE interactions from 386 publications for blood lipids, glycemic traits, obesity anthropometrics, vascular measures, inflammation and metabolic syndrome, and introduce CardioGxE, a gene-environment interaction resource. We then analyzed the genes and SNPs supporting cardiometabolic GxEs in order to demonstrate utility of GxE SNPs and to discern characteristics of these important genetic variants. We were able to draw many observations from our extensive analysis of GxEs. 1) The CardioGxE SNPs showed little overlap with variants identified by main effect GWAS, indicating the importance of environmental interactions with genetic factors on cardiometabolic traits. 2) These GxE SNPs were enriched in adaptation to climatic and geographical features, with implications on energy homeostasis and response to physical activity. 3) Comparison to gene networks responding to plasma cholesterol-lowering or regression of atherosclerotic plaques showed that GxE genes have a greater role in those responses, particularly through high-energy diets and fat intake, than do GWAS-identified genes for the same traits. Other aspects of the CardioGxE dataset were explored. Conclusions Overall, we demonstrate that SNPs supporting cardiometabolic GxE interactions often exhibit transcriptional effects or are under positive selection. Still, not all such SNPs can be assigned potential functional or regulatory roles often because data are lacking in specific cell types or from treatments that approximate the environmental factor of the GxE. With research on metabolic related complex disease risk embarking on genome-wide GxE interaction tests, CardioGxE will be a useful resource.
Collapse
Affiliation(s)
- Laurence D Parnell
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Britt A Blokker
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Hassan S Dashti
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Paula-Dene Nesbeth
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Brittany Elle Cooper
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Yiyi Ma
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Yu-Chi Lee
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Ruixue Hou
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Chao-Qiang Lai
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - Kris Richardson
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| | - José M Ordovás
- JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111, USA
| |
Collapse
|
36
|
The challenges of genome-wide interaction studies: lessons to learn from the analysis of HDL blood levels. PLoS One 2014; 9:e109290. [PMID: 25329471 PMCID: PMC4203717 DOI: 10.1371/journal.pone.0109290] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 08/29/2014] [Indexed: 11/23/2022] Open
Abstract
Genome-wide association studies (GWAS) have revealed 74 single nucleotide polymorphisms (SNPs) associated with high-density lipoprotein cholesterol (HDL) blood levels. This study is, to our knowledge, the first genome-wide interaction study (GWIS) to identify SNP×SNP interactions associated with HDL levels. We performed a GWIS in the Rotterdam Study (RS) cohort I (RS-I) using the GLIDE tool which leverages the massively parallel computing power of Graphics Processing Units (GPUs) to perform linear regression on all genome-wide pairs of SNPs. By performing a meta-analysis together with Rotterdam Study cohorts II and III (RS-II and RS-III), we were able to filter 181 interaction terms with a p-value<1 · 10−8 that replicated in the two independent cohorts. We were not able to replicate any of these interaction term in the AGES, ARIC, CHS, ERF, FHS and NFBC-66 cohorts (Ntotal = 30,011) when adjusting for multiple testing. Our GWIS resulted in the consistent finding of a possible interaction between rs774801 in ARMC8 (ENSG00000114098) and rs12442098 in SPATA8 (ENSG00000185594) being associated with HDL levels. However, p-values do not reach the preset Bonferroni correction of the p-values. Our study suggest that even for highly genetically determined traits such as HDL the sample sizes needed to detect SNP×SNP interactions are large and the 2-step filtering approaches do not yield a solution. Here we present our analysis plan and our reservations concerning GWIS.
Collapse
|
37
|
Abstract
Genome-wide association studies (GWASs) have become the focus of the statistical analysis of complex traits in humans, successfully shedding light on several aspects of genetic architecture and biological aetiology. Single-nucleotide polymorphisms (SNPs) are usually modelled as having additive, cumulative and independent effects on the phenotype. Although evidently a useful approach, it is often argued that this is not a realistic biological model and that epistasis (that is, the statistical interaction between SNPs) should be included. The purpose of this Review is to summarize recent directions in methodology for detecting epistasis and to discuss evidence of the role of epistasis in human complex trait variation. We also discuss the relevance of epistasis in the context of GWASs and potential hazards in the interpretation of statistical interaction terms.
Collapse
|
38
|
Al Riyami NB, Banerjee Y, Al-Waili K, Rizvi SG, Al-Yahyaee S, Hassan MO, Albarwani S, Al-Rasadi K, Bayoumi RA. The Effect of Residing Altitude on Levels of High-Density Lipoprotein Cholesterol: A Pilot Study From the Omani Arab Population. Angiology 2014; 66:568-73. [PMID: 25078070 DOI: 10.1177/0003319714544355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lower mortality rates from coronary heart disease and higher levels of serum high-density lipoprotein cholesterol (HDL-C) have been observed in populations residing at high altitude. However, this effect has not been investigated in Arab populations, which exhibit considerable genetic homogeneity. We assessed the relationship between residing altitude and HDL-C in 2 genetically similar Omani Arab populations residing at different altitudes. The association between the levels of HDL-C and other metabolic parameters was also investigated. The levels of HDL-C were significantly higher in the high-altitude group compared with the low-altitude group. Stepwise regression analysis showed that altitude was the most significant factor affecting HDL-C, followed by gender, serum triglycerides, and finally the 2-hour postprandial plasma glucose. This finding is consistent with previously published studies from other populations and should be taken into consideration when comparing cardiovascular risk factors in populations residing at different altitudes.
Collapse
Affiliation(s)
- Nafila B Al Riyami
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Yajnavalka Banerjee
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalid Al-Waili
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Syed G Rizvi
- Department of Family Medicine and Public Health, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Said Al-Yahyaee
- Department of Genetics, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Mohammed O Hassan
- Department of Physiology, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Sulayma Albarwani
- Department of Physiology, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Khalid Al-Rasadi
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| | - Riad A Bayoumi
- Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
39
|
Rodriguez-Fontenla C, Calaza M, Gonzalez A. Genetic distance as an alternative to physical distance for definition of gene units in association studies. BMC Genomics 2014; 15:408. [PMID: 24884992 PMCID: PMC4048458 DOI: 10.1186/1471-2164-15-408] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 05/20/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Some association studies, as the implemented in VEGAS, ALIGATOR, i-GSEA4GWAS, GSA-SNP and other software tools, use genes as the unit of analysis. These genes include the coding sequence plus flanking sequences. Polymorphisms in the flanking sequences are of interest because they involve cis-regulatory elements or they inform on untyped genetic variants trough linkage disequilibrium. Gene extensions have customarily been defined as ±50 Kb. This approach is not fully satisfactory because genetic relationships between neighbouring sequences are a function of genetic distances, which are only poorly replaced by physical distances. RESULTS Standardized recombination rates (SRR) from the deCODE recombination map were used as units of genetic distances. We searched for a SRR producing flanking sequences near the ±50 Kb offset that has been common in previous studies. A SRR≥2 was selected because it led to gene extensions with median length=45.3 Kb and the simplicity of an integer value. As expected, boundaries of the genes defined with the ±50 Kb and with the SRR≥2 rules were rarely concordant. The impact of these differences was illustrated with the interpretation of top association signals from two large studies including many hits and their detailed analysis based in different criteria. The definition based in genetic distance was more concordant with the results of these studies than the based in physical distance. In the analysis of 18 top disease associated loci form the first study, the SRR≥2 genes led to a fully concordant interpretation in 17 loci; the ±50 Kb genes only in 6. Interpretation of the 43 putative functional genes of the second study based in the SRR≥2 definition only missed 4 of the genes, whereas the based in the ±50 Kb definition missed 10 genes. CONCLUSIONS A gene definition based on genetic distance led to results more concordant with expert detailed analyses than the commonly used based in physical distance. The genome coordinates for each gene are provided to maintain a simple use of the new definitions.
Collapse
Affiliation(s)
| | | | - Antonio Gonzalez
- Laboratorio de Investigacion 10 and Rheumatology Unit, Instituto de Investigacion Sanitaria - Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain.
| |
Collapse
|
40
|
Wei WH, Guo Y, Kindt ASD, Merriman TR, Semple CA, Wang K, Haley CS. Abundant local interactions in the 4p16.1 region suggest functional mechanisms underlying SLC2A9 associations with human serum uric acid. Hum Mol Genet 2014; 23:5061-8. [PMID: 24821702 PMCID: PMC4159153 DOI: 10.1093/hmg/ddu227] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human serum uric acid concentration (SUA) is a complex trait. A recent meta-analysis of multiple genome-wide association studies (GWAS) identified 28 loci associated with SUA jointly explaining only 7.7% of the SUA variance, with 3.4% explained by two major loci (SLC2A9 and ABCG2). Here we examined whether gene-gene interactions had any roles in regulating SUA using two large GWAS cohorts included in the meta-analysis [the Atherosclerosis Risk in Communities study cohort (ARIC) and the Framingham Heart Study cohort (FHS)]. We found abundant genome-wide significant local interactions in ARIC in the 4p16.1 region located mostly in an intergenic area near SLC2A9 that were not driven by linkage disequilibrium and were replicated in FHS. Taking the forward selection approach, we constructed a model of five SNPs with marginal effects and three epistatic SNP pairs in ARIC-three marginal SNPs were located within SLC2A9 and the remaining SNPs were all located in the nearby intergenic area. The full model explained 1.5% more SUA variance than that explained by the lead SNP alone, but only 0.3% was contributed by the marginal and epistatic effects of the SNPs in the intergenic area. Functional analysis revealed strong evidence that the epistatically interacting SNPs in the intergenic area were unusually enriched at enhancers active in ENCODE hepatic (HepG2, P = 4.7E-05) and precursor red blood (K562, P = 5.0E-06) cells, putatively regulating transcription of WDR1 and SLC2A9. These results suggest that exploring epistatic interactions is valuable in uncovering the complex functional mechanisms underlying the 4p16.1 region.
Collapse
Affiliation(s)
- Wen-Hua Wei
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK, Arthritis Research UK Centre for Genetics and Genomics, Institute of Inflammation and Repair, Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK,
| | - Yunfei Guo
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Alida S D Kindt
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, PO Box 56, Dunedin, New Zealand
| | - Colin A Semple
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| | - Kai Wang
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, USA
| | - Chris S Haley
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
41
|
Sun X, Lu Q, Mukherjee S, Crane PK, Elston R, Ritchie MD. Analysis pipeline for the epistasis search - statistical versus biological filtering. Front Genet 2014; 5:106. [PMID: 24817878 PMCID: PMC4012196 DOI: 10.3389/fgene.2014.00106] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 04/10/2014] [Indexed: 12/15/2022] Open
Abstract
Gene-gene interactions may contribute to the genetic variation underlying complex traits but have not always been taken fully into account. Statistical analyses that consider gene-gene interaction may increase the power of detecting associations, especially for low-marginal-effect markers, and may explain in part the "missing heritability." Detecting pair-wise and higher-order interactions genome-wide requires enormous computational power. Filtering pipelines increase the computational speed by limiting the number of tests performed. We summarize existing filtering approaches to detect epistasis, after distinguishing the purposes that lead us to search for epistasis. Statistical filtering includes quality control on the basis of single marker statistics to avoid the analysis of bad and least informative data, and limits the search space for finding interactions. Biological filtering includes targeting specific pathways, integrating various databases based on known biological and metabolic pathways, gene function ontology and protein-protein interactions. It is increasingly possible to target single-nucleotide polymorphisms that have defined functions on gene expression, though not belonging to protein-coding genes. Filtering can improve the power of an interaction association study, but also increases the chance of missing important findings.
Collapse
Affiliation(s)
- Xiangqing Sun
- Department of Epidemiology and Biostatistics, Case Western Reserve UniversityCleveland, OH, USA
| | - Qing Lu
- Department of Epidemiology and Biostatistics, Michigan State UniversityEast Lansing, MI, USA
| | | | - Paul K. Crane
- Department of Medicine, University of WashingtonSeattle, WA, USA
| | - Robert Elston
- Department of Epidemiology and Biostatistics, Case Western Reserve UniversityCleveland, OH, USA
| | - Marylyn D. Ritchie
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University ParkPA, USA
| |
Collapse
|
42
|
Zhao Y, Xiong N, Liu Y, Zhou Y, Li N, Qing H, Lin Z. Human dopamine transporter gene: differential regulation of 18-kb haplotypes. Pharmacogenomics 2014; 14:1481-94. [PMID: 24024899 DOI: 10.2217/pgs.13.141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIM Since previous functional studies of short haplotypes and polymorphic sites of SLC6A3 have shown variant-dependent and drug-sensitive promoter activity, this study aimed to understand whether a large SLC6A3 regulatory region, containing these small haplotypes and polymorphic sites, can display haplotype-dependent promoter activity in a drug-sensitive and pathway-related manner. MATERIALS & METHODS By creating and using a single copy number luciferase-reporter vector, we examined regulation of two different SLC6A3 haplotypes (A and B) of the 5´ 18-kb promoter and two known downstream regulatory variable number tandem repeats by 17 drugs in four different cellular models. RESULTS The two regulatory haplotypes displayed up to 3.2-fold difference in promoter activity. The regulations were drug selective (37.5% of the drugs showed effects), and both haplotype and cell type dependent. Pathway analysis revealed at least 13 main signaling hubs targeting SLC6A3, including histone deacetylation, AKT, PKC and CK2 α-chains. CONCLUSION SLC6A3 may be regulated via either its promoter or the variable number tandem repeats independently by specific signaling pathways and in a haplotype-dependent manner. Furthermore, we have developed the first pathway map for SLC6A3 regulation. These findings provide a framework for understanding complex and variant-dependent regulations of SLC6A3.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Psychiatry, Harvard Medical School & Laboratory of Psychiatric Neurogenomics, Division of Alcohol & Drug Abuse, McLean Hospital, Mailstop 318, 115 Mill Street, Belmont, MA 02478, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Ma L, Ballantyne C, Brautbar A, Keinan A. Analysis of multiple association studies provides evidence of an expression QTL hub in gene-gene interaction network affecting HDL cholesterol levels. PLoS One 2014; 9:e92469. [PMID: 24651390 PMCID: PMC3961362 DOI: 10.1371/journal.pone.0092469] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 02/21/2014] [Indexed: 11/18/2022] Open
Abstract
Epistasis has been suggested to underlie part of the missing heritability in genome-wide association studies. In this study, we first report an analysis of gene-gene interactions affecting HDL cholesterol (HDL-C) levels in a candidate gene study of 2,091 individuals with mixed dyslipidemia from a clinical trial. Two additional studies, the Atherosclerosis Risk in Communities study (ARIC; n = 9,713) and the Multi-Ethnic Study of Atherosclerosis (MESA; n = 2,685), were considered for replication. We identified a gene-gene interaction between rs1532085 and rs12980554 (P = 7.1×10−7) in their effect on HDL-C levels, which is significant after Bonferroni correction (Pc = 0.017) for the number of SNP pairs tested. The interaction successfully replicated in the ARIC study (P = 7.0×10−4; Pc = 0.02). Rs1532085, an expression QTL (eQTL) of LIPC, is one of the two SNPs involved in another, well-replicated gene-gene interaction underlying HDL-C levels. To further investigate the role of this eQTL SNP in gene-gene interactions affecting HDL-C, we tested in the ARIC study for interaction between this SNP and any other SNP genome-wide. We found the eQTL to be involved in a few suggestive interactions, one of which significantly replicated in MESA. Importantly, these gene-gene interactions, involving only rs1532085, explain an additional 1.4% variation of HDL-C, on top of the 0.65% explained by rs1532085 alone. LIPC plays a key role in the lipid metabolism pathway and it, and rs1532085 in particular, has been associated with HDL-C and other lipid levels. Collectively, we discovered several novel gene-gene interactions, all involving an eQTL of LIPC, thus suggesting a hub role of LIPC in the gene-gene interaction network that regulates HDL-C levels, which in turn raises the hypothesis that LIPC's contribution is largely via interactions with other lipid metabolism related genes.
Collapse
Affiliation(s)
- Li Ma
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Christie Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Ariel Brautbar
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Department of Medical Genetics, Marshfield Clinic, Marshfield, Wisconsin, United States of America
- * E-mail: (AK); (AB)
| | - Alon Keinan
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
- * E-mail: (AK); (AB)
| |
Collapse
|
44
|
Parnell LD, Casas-Agustench P, Iyer LK, Ordovas JM. How Gene Networks Can Uncover Novel CVD Players. CURRENT CARDIOVASCULAR RISK REPORTS 2014; 8:372. [PMID: 24683432 DOI: 10.1007/s12170-013-0372-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Cardiovascular diseases (CVD) are complex, involving numerous biological entities from genes and small molecules to organ function. Placing these entities in networks where the functional relationships among the constituents are drawn can aid in our understanding of disease onset, progression and prevention. While networks, or interactomes, are often classified by a general term, say lipids or inflammation, it is a more encompassing class of network that is more informative in showing connections among the active entities and allowing better hypotheses of novel CVD players to be formulated. A range of networks will be presented whereby the potential to bring new objects into the CVD milieu will be exemplified.
Collapse
Affiliation(s)
- Laurence D Parnell
- Nutritional Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111
| | - Patricia Casas-Agustench
- Instituto Madrileño de Estudios Avanzados (IMDEA) Alimentación, CEI UAM+CSIC, C/Faraday, 7, 1 planta D1.11, Ciudad Universitaria de Cantoblanco, Ctra. de Colmenar Km.15, Madrid, 28049, Spain
| | - Lakshmanan K Iyer
- Tufts Center for Neuroscience Research, Tufts University School of Medicine, 136 Harrison Ave, Boston, MA 02111, Molecular Cardiology Research Institute, Tufts Medical Center, 15 Kneeland Street, Boston, MA 02111
| | - Jose M Ordovas
- Nutritional Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, 711 Washington Street, Boston, MA 02111 ; Instituto Madrileño de Estudios Avanzados (IMDEA) Alimentación, CEI UAM+CSIC, C/Faraday, 7, 1 planta D1.11, Ciudad Universitaria de Cantoblanco, Ctra. de Colmenar Km.15, Madrid, 28049, Spain
| |
Collapse
|
45
|
Koran MEI, Hohman TJ, Thornton-Wells TA. Genetic interactions found between calcium channel genes modulate amyloid load measured by positron emission tomography. Hum Genet 2014; 133:85-93. [PMID: 24026422 PMCID: PMC4045094 DOI: 10.1007/s00439-013-1354-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/17/2013] [Indexed: 12/16/2022]
Abstract
Late-onset Alzheimer's disease (LOAD) is known to have a complex, oligogenic etiology, with considerable genetic heterogeneity. We investigated the influence of genetic interactions between genes in the Alzheimer's disease (AD) pathway on amyloid-beta (Aβ) deposition as measured by PiB or AV-45 ligand positron emission tomography (PET) to aid in understanding LOAD's genetic etiology. Subsets of the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohorts were used for discovery and for two independent validation analyses. A significant interaction between RYR3 and CACNA1C was confirmed in all three of the independent ADNI datasets. Both genes encode calcium channels expressed in the brain. The results shown here support previous animal studies implicating interactions between these calcium channels in amyloidogenesis and suggest that the pathological cascade of this disease may be modified by interactions in the amyloid-calcium axis. Future work focusing on the mechanisms of such relationships may inform targets for clinical intervention.
Collapse
Affiliation(s)
- Mary Ellen I. Koran
- Center for Human Genetics and Research, Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine 519 Light Hall Nashville, TN 37232-0700
| | - Timothy J. Hohman
- Center for Human Genetics and Research, Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine 519 Light Hall Nashville, TN 37232-0700
| | - Tricia A. Thornton-Wells
- Center for Human Genetics and Research, Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine 519 Light Hall Nashville, TN 37232-0700
| |
Collapse
|
46
|
Su WH, Yao Shugart Y, Chang KP, Tsang NM, Tse KP, Chang YS. How genome-wide SNP-SNP interactions relate to nasopharyngeal carcinoma susceptibility. PLoS One 2013; 8:e83034. [PMID: 24376627 PMCID: PMC3871583 DOI: 10.1371/journal.pone.0083034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 10/29/2013] [Indexed: 11/18/2022] Open
Abstract
This study is the first to use genome-wide association study (GWAS) data to evaluate the multidimensional genetic architecture underlying nasopharyngeal cancer. Since analysis of data from GWAS confirms a close and consistent association between elevated risk for nasopharyngeal carcinoma (NPC) and major histocompatibility complex class 1 genes, our goal here was to explore lesser effects of gene-gene interactions. We conducted an exhaustive genome-wide analysis of GWAS data of NPC, revealing two-locus interactions occurring between single nucleotide polymorphisms (SNPs), and identified a number of suggestive interaction loci which were missed by traditional GWAS analyses. Although none of the interaction pairs we identified passed the genome-wide Bonferroni-adjusted threshold for significance, using independent GWAS data from the same population (Stage 2), we selected 66 SNP pairs in 39 clusters with P<0.01. We identified that in several chromosome regions, multiple suggestive interactions group to form a block-like signal, effectively reducing the rate of false discovery. The strongest cluster of interactions involved the CREB5 gene and a SNP rs1607979 on chromosome 17q22 (P = 9.86×10(-11)) which also show trans-expression quantitative loci (eQTL) association in Chinese population. We then detected a complicated cis-interaction pattern around the NPC-associated HLA-B locus, which is immediately adjacent to copy-number variations implicated in male susceptibility for NPC. While it remains to be seen exactly how and to what degree SNP-SNP interactions such as these affect susceptibility for nasopharyngeal cancer, future research on these questions holds great promise for increasing our understanding of this disease's genetic etiology, and possibly also that of other gene-related cancers.
Collapse
Affiliation(s)
- Wen-Hui Su
- Department of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Chang Gung Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yin Yao Shugart
- Genomic Research Branch, Division of Neuroscience and Behavioral Sciences, National Institute of Mental Health, NIH, Bethesda, Maryland, United States of America
- Department of Gastroenterology, Johns Hopkins Medical School, Baltimore, Maryland, United States of America
| | - Kai-Ping Chang
- Department of Otolaryngology - Head and Neck Surgery, Chang Gung Memorial Hospital at Lin-Kou, Taoyuan, Taiwan
| | - Ngan-Ming Tsang
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Lin-Kou, Taoyuan, Taiwan
| | - Ka-Po Tse
- Chang Gung Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Sun Chang
- Chang Gung Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
47
|
Yang W, Gu C. A whole-genome simulator capable of modeling high-order epistasis for complex disease. Genet Epidemiol 2013; 37:686-94. [PMID: 24114848 PMCID: PMC4143152 DOI: 10.1002/gepi.21761] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 08/09/2013] [Accepted: 08/14/2013] [Indexed: 11/10/2022]
Abstract
Genome-wide association studies (GWAS) have been successful in finding numerous new risk variants for complex diseases, but the results almost exclusively rely on single-marker scans. Methods that can analyze joint effects of many variants in GWAS data are still being developed and trialed. To evaluate the performance of such methods it is essential to have a GWAS data simulator that can rapidly simulate a large number of samples, and capture key features of real GWAS data such as linkage disequilibrium (LD) among single-nucleotide polymorphisms (SNPs) and joint effects of multiple loci (multilocus epistasis). In the current study, we combine techniques for specifying high-order epistasis among risk SNPs with an existing program GWAsimulator [Li and Li, 2008] to achieve rapid whole-genome simulation with accurate modeling of complex interactions. We considered various approaches to specifying interaction models including the following: departure from product of marginal effects for pairwise interactions, product terms in logistic regression models for low-order interactions, and penetrance tables conforming to marginal effect constraints for high-order interactions or prescribing known biological interactions. Methods for conversion among different model specifications are developed using penetrance table as the fundamental characterization of disease models. The new program, called simGWA, is capable of efficiently generating large samples of GWAS data with high precision. We show that data simulated by simGWA are faithful to template LD structures, and conform to prespecified diseases models with (or without) interactions.
Collapse
Affiliation(s)
- Wei Yang
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO
| | - Charles Gu
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
48
|
Wei W, Gyenesei A, Semple CAM, Haley CS. Properties of local interactions and their potential value in complementing genome-wide association studies. PLoS One 2013; 8:e71203. [PMID: 23940718 PMCID: PMC3733963 DOI: 10.1371/journal.pone.0071203] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 07/03/2013] [Indexed: 01/11/2023] Open
Abstract
Local interactions between neighbouring SNPs are hypothesized to be able to capture variants missing from genome-wide association studies (GWAS) via haplotype effects but have not been thoroughly explored. We have used a new high-throughput analysis tool to probe this underexplored area through full pair-wise genome scans and conventional GWAS in diastolic and systolic blood pressure and six metabolic traits in the Northern Finland Birth Cohort 1966 (NFBC1966) and the Atherosclerosis Risk in Communities study cohort (ARIC). Genome-wide significant interactions were detected in ARIC for systolic blood pressure between PLEKHA7 (a known GWAS locus for blood pressure) and GPR180 (which plays a role in vascular remodelling), and also for triglycerides as local interactions within the 11q23.3 region (replicated significantly in NFBC1966), which notably harbours several loci (BUD13, ZNF259 and APOA5) contributing to triglyceride levels. Tests of the local interactions within the 11q23.3 region conditional on the top GWAS signal suggested the presence of two independent functional variants, each with supportive evidence for their roles in gene regulation. Local interactions captured 9 additional GWAS loci identified in this study (3 significantly replicated) and 73 from previous GWAS (24 in the eight traits and 49 in related traits). We conclude that the detection of local interactions requires adequate SNP coverage of the genome and that such interactions are only likely to be detectable between SNPs in low linkage disequilibrium. Analysing local interactions is a potentially valuable complement to GWAS and can provide new insights into the biology underlying variation in complex traits.
Collapse
Affiliation(s)
- Wenhua Wei
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine at the University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | |
Collapse
|
49
|
Tan Q, Soerensen M, Kruse TA, Christensen K, Christiansen L. A novel permutation test for case-only analysis identifies epistatic effects on human longevity in the FOXO gene family. Aging Cell 2013; 12:690-4. [PMID: 23607278 DOI: 10.1111/acel.12092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2013] [Indexed: 01/13/2023] Open
Abstract
Genetic interactions or epistasis could make a substantial contribution to variation in human complex traits including longevity. However, detecting epistatic interactions in high dimensional datasets is difficult due to various reasons including multiple testing of correlated tests. We introduce a novel permutation strategy to the case-only analysis of gene-by-gene interaction using multiple SNPs. The method is applied to genes coding for Forkhead box O transcription factors which recently have been associated with human longevity across different populations hypothesizing that epistatic interaction in the regulation and expression of the FOXO gene family could contribute to the human longevity phenotype. Genotype data were collected from 1088 individuals from the Danish 1905 birth cohort aged over 92-93 years with 12 SNPs in the FOXO1a and 15 SNPs in the FOXO3a genes. Our analysis detected a joint effect between rs9486902 in FOXO3a and rs2701858 in FOXO1a that highly significantly contributes to human longevity (OR = 3.23, 95% CI: 2.93-3.53) which is consistent in both males and females. Our results were compared with published studies, and importance of our novel method and findings was discussed.
Collapse
Affiliation(s)
- Qihua Tan
- Epidemiology Institute of Public Health University of Southern Denmark Odense C Denmark
- Department of Clinical Genetics Odense University Hospital Odense C Denmark
| | - Mette Soerensen
- Epidemiology Institute of Public Health University of Southern Denmark Odense C Denmark
| | - Torben A. Kruse
- Department of Clinical Genetics Odense University Hospital Odense C Denmark
| | - Kaare Christensen
- Epidemiology Institute of Public Health University of Southern Denmark Odense C Denmark
- Department of Clinical Genetics Odense University Hospital Odense C Denmark
- Department of Biochemistry and Pharmacology Odense University Hospital Odense C Denmark
| | - Lene Christiansen
- Epidemiology Institute of Public Health University of Southern Denmark Odense C Denmark
- Department of Clinical Genetics Odense University Hospital Odense C Denmark
| |
Collapse
|
50
|
Rovaris DL, Mota NR, Callegari-Jacques SM, Bau CHD. Approaching "phantom heritability" in psychiatry by hypothesis-driven gene-gene interactions. Front Hum Neurosci 2013; 7:210. [PMID: 23720624 PMCID: PMC3655459 DOI: 10.3389/fnhum.2013.00210] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2013] [Accepted: 05/02/2013] [Indexed: 12/01/2022] Open
Affiliation(s)
- Diego Luiz Rovaris
- Department of Genetics, Instituto de Biociências, Universidade Federal do Rio Grande do Sul Porto Alegre, Brazil
| | | | | | | |
Collapse
|