Next Article in Journal
The Long-Term Effect of COVID-19 Infection on Body Composition
Previous Article in Journal
Differential Activation of TAS2R4 May Recover Ability to Taste Propylthiouracil for Some TAS2R38 AVI Homozygotes
Previous Article in Special Issue
The Impact of Diet on miRNA Regulation and Its Implications for Health: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Integrated Analysis of Genomic and Genome-Wide Association Studies Identified Candidate Genes for Nutrigenetic Studies in Flavonoids and Vascular Health: Path to Precision Nutrition for (Poly)phenols

by
Tatjana Ruskovska
1,
Filip Postolov
1 and
Dragan Milenkovic
2,*
1
Faculty of Medical Sciences, Goce Delcev University, 2000 Stip, North Macedonia
2
Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(9), 1362; https://doi.org/10.3390/nu16091362
Submission received: 21 February 2024 / Revised: 13 April 2024 / Accepted: 24 April 2024 / Published: 30 April 2024

Abstract

:
Flavonoids exert vasculoprotective effects in humans, but interindividual variability in their action has also been reported. This study aims to identify genes that are associated with vascular health effects of flavonoids and whose polymorphisms could explain interindividual variability in response to their intake. Applying the predetermined literature search criteria, we identified five human intervention studies reporting positive effects of flavonoids on vascular function together with global genomic changes analyzed using microarray methods. Genes involved in vascular dysfunction were identified from genome-wide association studies (GWAS). By extracting data from the eligible human intervention studies, we obtained 5807 differentially expressed genes (DEGs). The number of identified upstream regulators (URs) varied across the studies, from 227 to 1407. The search of the GWAS Catalog revealed 493 genes associated with vascular dysfunction. An integrative analysis of transcriptomic data with GWAS genes identified 106 candidate DEGs and 42 candidate URs, while subsequent functional analyses and a search of the literature identified 20 top priority candidate genes: ALDH2, APOE, CAPZA1, CYP11B2, GNA13, IL6, IRF5, LDLR, LPL, LSP1, MKNK1, MMP3, MTHFR, MYO6, NCR3, PPARG, SARM1, TCF20, TCF7L2, and TNF. In conclusion, this integrated analysis identifies important genes to design future nutrigenetic studies for development of precision nutrition for polyphenols.

1. Introduction

(Poly)phenols are the most abundant bioactive compounds of plant origin and are present in the human diet in relatively large amounts, ranging from less than 500 to more than 1500 mg/d [1,2]. In the US adult population, a dietary intake of approximately 900 mg per 1000 kcal/d has recently been reported [3]. (Poly)phenols present extraordinary heterogeneity in their chemical structures, with over 500 chemical entities identified in the human diet, which are divided into two classes: flavonoids and non-flavonoids [4,5]. Following intake of (poly)phenols, they are metabolized by both human enzymes and gastro-intestinal microbiota. Once in the gut and intestine, (poly)phenols can be absorbed by enterocytes, enter the liver, and be converted by Phase I (oxidation, hydrolysis, and reduction) and Phase II (glucuronidation, methylation, and sulfation) metabolic reactions. Derived metabolites enter general circulation and can reach tissues. Non-absorbed (poly)phenols enter the colon where they can be directly metabolized by gut microbiota and give rise to low-molecular-weight metabolites. These gut microbiota-derived metabolites can be absorbed by the enterocytes and be further metabolized by Phase I and Phase II metabolic reactions in the liver before entering general circulation [6].
Flavonoids are among the best studied plant food bioactives in terms of their health-promoting properties. Human studies have shown that a diet rich in flavonoids can reduce type 2 diabetes risk [7], improve insulin sensitivity and blood lipids [8], and have beneficial effects on vascular function [9]. Recently, a long-term, large-scale, randomized, double-blind, placebo-controlled trial with cocoa extract supplementation [500 mg flavanols/d, including 80 mg (−)-epicatechin] showed a significant reduction in cardiovascular disease death by 27% among older adults [10]. In addition, molecular mechanisms underlying vasculoprotective effects of flavonoids have been investigated using omics technologies, such as transcriptomics, epigenomics, proteomics, and metabolomics [11]. These state-of-the-art untargeted analytical methods enable the identification of global molecular modulations, while subsequent bioinformatic analyses of individual omics data indicate key cellular pathways and regulatory mechanisms involved.
Despite the general trend demonstrating positive effects of dietary flavonoids on vascular health in humans, some less convincing results have also been reported in the literature. Such data have allowed for the identification of subgroups of participants where the vasculoprotective effects of flavonoids were more pronounced [12]. Other studies identified factors underlying the interindividual variabilities in health-promoting effects of flavonoids that include sex, age, ethnicity, body mass index, health status, gut microbiome, and genetic factors [13]. Of these, genetic factors have been the least studied.
Vascular function, which is one of the key determinants of overall health, is largely influenced by age [14] and lifestyle [15,16], with genetic factors also playing a significant role. A clinical study has shown the association of endothelial nitric oxide synthase (Nitric Oxide Synthase 3, NOS3) gene G894T polymorphism with hypertension risk and complications [17]. Also, monocyte chemoattractant protein-1 (MCP-1; or C-C Motif Chemokine Ligand 2, CCL2) gene −2578A > G polymorphism has been associated with an increased risk of coronary atherosclerosis in an asymptomatic population [18]. On the other hand, studies on flavonoids, genetic polymorphisms, and vascular health are scarce. The most convincing results were obtained from the studies that were focused on polymorphisms of well-established genes that are directly associated with vascular function or genes involved in the metabolism of circulating lipoproteins. It has been shown that the Glu298Asp single nucleotide polymorphism (SNP) in the NOS3 gene differentially affects the vascular response to acute consumption of fruit and vegetables [19], and that polymorphisms in Apolipoprotein A1 (APOA1_rs964184) and Lipoprotein Lipase (LPL_rs12678919) genes determine the vasculoprotective effects of orange juice [20].
Given the small number of identified genetic polymorphisms that determine the interindividual variabilities in the effects of dietary flavonoids on vascular function, there is a need for additional studies focused on: (a) identification of candidate genes on dietary flavonoids and vascular function, and (b) testing of identified candidate genes in appropriately designed nutrigenetic studies. To address the first goal, i.e., identification of candidate genes for future nutrigenetic studies on dietary flavonoids and vascular function, one approach is through the integration of (i) data from human intervention studies with flavonoids showing modulations in global gene expression together with positive effects on vascular function and (ii) results from genome-wide association studies (GWAS) that have identified variants and risk alleles associated with vascular dysfunction, such as hypertension, atherosclerosis, or arterial stiffness. So far, such an innovative and powerful approach combining genomic and GWAS datasets has enabled the identification of candidate genes associated with, and directly governing, disease pathobiology [21,22], thus facilitating targeted studies to identify functional impact of major causal genes. Indeed, GWAS can identify hundreds of candidate genes associated with the development of disease, revealing a need for a systematic way to understand the causal mechanism(s) of these genes and a means to prioritize them for further study. The integration of genomic and GWAS data has recently allowed to identify candidate genes causal of coronary artery disease (CAD) [23]. The authors performed a comprehensive integrative analysis by combining CAD genome-wide association studies datasets (UK Biobank and CARDIoGRAMplusC4D) with transcriptomic data from the STARNET study (Stockholm–Tartu Atherosclerosis Reverse Network Engineering Task). The same approach has also been recently used to identify genes associated with atrial fibrillation (AF) [24]. The authors concluded that such an integrative omics strategy has improved the power of identifying AF-related genes compared to using GWAS alone.
Therefore, the aim of this study was to identify candidate genes whose polymorphisms potentially determine the interindividual variabilities of the effects of flavonoids on vascular health. To this end, we conducted integrative and functional analyses of genomic data from human intervention studies, presenting vasculoprotective effects of flavonoids and data from GWAS related to vascular dysfunction. Such a novel approach allowed us to identify top-priority candidate genes for future nutrigenetic studies on flavonoids and interindividual variability in vascular health effects, studies that will provide central leads for the development of precision nutrition for (poly)phenols.

2. Materials and Methods

This study is based on our previous systematic literature search and analysis of nutrigenomic effects of (poly)phenols related to cardiometabolic health in humans [25]. However, here we have only included the studies that demonstrated positive effects of flavonoids on vascular function and analyzed their genomic effects using microarray methods [26,27,28]. In addition, two recent studies of relevance for our analyses [29,30] were also included. In all of these studies, the analyses of global gene expression were conducted in samples of peripheral blood.
The analyses of upstream regulators (URs) of each set of differentially expressed genes (DEGs) were conducted using QIAGEN Ingenuity Pathway Analysis (IPA) on-line bioinformatic tool (https://digitalinsights.qiagen.com/, accessed on 28 July 2022, 29 July 2022, 5 August 2022, and 12 September 2022). For identification of genes that are associated with vascular dysfunction across published genome-wide association studies, we searched, in July 2022, GWAS Catalog (https://www.ebi.ac.uk/gwas/home, accessed on 16 July 2022) [31]. Pathway enrichment analyses were conducted using GeneTrail3.2 (https://genetrail.bioinf.uni-sb.de/, accessed on 19 July 2022) [32] as a platform to access the following two databases: Kyoto Encyclopedia of Genes and Genomes (KEGG) [33] and WikiPathways [34,35]. InteractiVenn (http://www.interactivenn.net/, accessed on 23 July 2022) [36] was used as a tool to retrieve elements that different datasets have in common. To identify variants with the highest frequencies of selected top-priority candidate genes, we interrogated the Variation Viewer database, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, U.S. (https://www.ncbi.nlm.nih.gov/variation/view, accessed on 12 September 2022). Pharmacology-relevant variants of selected top-priority candidate genes were identified using the PharmGKB database (https://www.pharmgkb.org, accessed on 12 September 2022) [37,38].
GWAS-reported variants of selected top-priority candidate genes associated with vascular dysfunction were identified back in the GWAS Catalog (https://www.ebi.ac.uk/gwas/home, accessed on 16 July 2022) [31], while their frequencies in the global population and previously reported clinical significance were identified in the dbSNP database, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, U.S. (https://www.ncbi.nlm.nih.gov/snp, accessed on 23 April 2023).
Gene names and symbols were searched in GeneCards database (https://www.genecards.org, accessed on 4 August 2022) [39]. The names of canonical pathways are presented as they appear in the interrogated databases.
A flowchart of the study is presented in Figure 1.

3. Results

3.1. Flavonoids Affect Global Gene Expression in Human Peripheral Blood Cells

3.1.1. General Overview of Selected Studies and DEGs

Based on our previously reported strategy for the systematic literature search [25], we identified five human intervention studies with flavonoids that analyzed global gene expression in peripheral blood cells and demonstrated at least one positive effect on vascular function. General information about each of these studies, referred to as Paper 1 to Paper 5 (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1. In these studies, flavonoids of different subclasses were studied: flavanones in Paper 1 and Paper 5, flavanols in Paper 2 and Paper 4, or anthocyanins in Paper 3. Study populations differed across the selected studies and included overweight men (Paper 1), non-obese healthy male smokers (Paper 2), healthy men (Paper 3 and Paper 4), or postmenopausal women (Paper 5). For each of these studies, at least one positive effect on vascular function was reported, in the same (as for Paper 3) or in an associated paper (Morand et al. [40] for Paper 1; Weseler et al. [41] for Paper 2; Sansone et al. [42] for Paper 4; Habauzit et al. [43] for Paper 5) (Table 1).
The number of DEGs varies across the studies: n = 1693; 717; 554; 2231; and 1401 for Papers 1; 2; 3; 4; and 5 respectively. After removing duplicates, the total number of flavonoid-modulated genes reached n = 5807 (Table S1). Comparative analysis of DEGs across the selected studies showed that n = 720 genes were in common for at least two studies, n = 67 genes were in common for at least three studies, and only two genes were in common for four studies. There were no DEGs that all five studies had in common (Figure 2; Table S2).

3.1.2. Upstream Regulators of DEGs

When analyzing the modulations in global gene expression, for biological interpretation of obtained experimental data, it is of particular importance to predict the upstream regulators (URs) of DEGs. For this analysis, we used the Qiagen IPA on-line bioinformatic tool (https://digitalinsights.qiagen.com/, accessed on 28 July 2022, 29 July 2022, 5 August 2022, and 12 September 2022), applying the default settings suggested by the manufacturer. With this analysis, for each set of DEGs, i.e., for each paper separately, we obtained URs that include not only protein coding genes and miRNAs, but also different chemical compounds, drugs, toxins, etc. The number of identified URs varied across the studies, from 227 to 1407, i.e., n = 227; 503; 508; 1407, and 993 for Papers 1–5, respectively. The lists of URs are presented in Table S3.

3.2. Identification of Genes Associated with Vascular Dysfunction from GWAS Studies

Our next goal was to search for genes for which previous GWAS studies have identified variants and risk alleles that are associated with vascular dysfunction. To this aim, we searched the GWAS Catalog for the following traits: hypertension, atherosclerosis, and arterial stiffness. For hypertension, the initial search retrieved a total number of 575 associations. The search was subsequently refined in terms of excluding studies related to early-onset hypertension, pulmonary arterial hypertension, pseudotumor cerebri, treatment-resistant hypertension, preeclampsia, chemotherapy-induced hypertension, or hypertension risk in short sleep duration, leading to a final list of 461 associations, and 375 genes associated with hypertension across 33 GWAS studies with the following accession numbers: GCST000041, GCST000361, GCST000398, GCST000447, GCST000849, GCST000973, GCST001085, GCST001238, GCST001423, GCST002627, GCST003613, GCST004143, GCST004384, GCST004388, GCST006023, GCST006229, GCST007707, GCST008036, GCST008828, GCST009685, GCST010477, GCST010774, GCST011141, GCST011952, GCST011953, GCST011954, GCST012136, GCST90000060, GCST90000064, GCST90077646, GCST90086091, GCST90086092, GCST90086157. For atherosclerosis, the initial search retrieved a total number of 261 associations. The search was subsequently refined in terms of exclusion of a study on the interaction of traffic-related air pollution with peripheral arterial disease, leading to a final list of 102 associations, and 69 genes associated with atherosclerosis across 19 GWAS studies with the following accession numbers: GCST000720, GCST001231, GCST002504, GCST003154, GCST007425, GCST007435, GCST008474, GCST009134, GCST010549, GCST90013689, GCST90013731, GCST90018670, GCST90018890, GCST90043957, GCST90061371, GCST90061372, GCST90061374, GCST90061375, GCST90061376. For arterial stiffness, a total number of 62 associations and 58 genes were identified in six GWAS studies with the following accession numbers: GCST000370, GCST007846, GCST008403, GCST010654, GCST010655, GCST010656. Pulling together all of these genes, and after the removal of duplicates, we finally obtained a list of n = 493 genes that previous GWAS studies have associated with vascular dysfunction (Table S4).

3.3. Integration of Transcriptomic Data with GWAS Identified Genes

Aiming to identify which of the DEGs from the human intervention studies selected for our analyses may potentially have the capacity to underlie the interindividual variability of the vascular effects in response to flavonoids intake, we conducted an integrative analysis of transcriptomic data and the genes identified from GWAS. To this end, for each of the selected studies, we compared the DEGs with the trait-specific genes identified from GWAS, i.e., genes whose variants are associated with hypertension, atherosclerosis, or arterial stiffness. For Paper 1, we identified 20, 4, and 2 genes associated with hypertension, atherosclerosis, or arterial stiffness, respectively; for Paper 2—13, 2, and 1 genes; for Paper 3—5, 1, and 4 genes; for Paper 4—33, 4 and 9 genes; and for Paper 5—18, 3, and 5 genes associated with hypertension, atherosclerosis, or arterial stiffness, respectively (Figure 3; Table S5).
When pulling together all of these genes, we identified n = 106 DEGs that potentially have the capacity to underlie the interindividual variability of the vascular effects in response to flavonoids intake, as candidate genes for future nutrigenetic studies on flavonoids and vascular health (Table S6A), here referred to as candidate DEGs.

Functional Analysis of Candidate DEGs

To better understand the biological functions of identified candidate DEGs (n = 106) and prioritize some of them for subsequent analyses, we performed functional analyses by determining their place in canonical pathways using pathway enrichment analyses. These analyses pinpointed several pathways of relevance for vascular dysfunction. Some of these pathways are directly involved in vascular dysfunction such as the VEGFA-VEGFR2 signaling pathway, which contains six candidate DEGs, regulation of actin cytoskeleton (four candidate DEGs), adherens junction (three candidate DEGs), focal adhesion (three candidate DEGs), apelin signaling pathway (two candidate DEGs), composition of lipid particles (two candidate DEGs), fluid shear stress and atherosclerosis (two candidate DEGs), or platelet activation (two candidate DEGs), while others are involved in inflammation, cell signaling, or antioxidant protection, such as the chemokine signaling pathway, NF-kappa B signaling pathway, toll-like receptor signaling pathway, MAPK signaling pathway, PI3K-Akt signaling pathway, or the NRF2 pathway. All of these pathways and their associated candidate DEGs are presented in Table 2. In summary, with these pathway enrichment analyses, we identified n = 26 DEGs that are placed in KEGG pathways and n = 25 DEGs that are placed in WikiPathways. After the removal of duplicate genes, there were n = 33 candidate DEGs placed in KEGG or WikiPathways that are relevant to vascular dysfunction (Table S7).
To identify genes with potentially greater influence on the interindividual variability of the vascular effects of flavonoids intake, and prioritize some of them for subsequent analyses, we searched for which of the candidate DEGs are among those that are common in the selected studies. To this end, we conducted a comparative analysis of the DEGs that at least two studies have in common (n = 720) and the candidate DEGs (n = 106) and obtained a list of n = 15 genes (CAPZA1, FSTL4, GNA13, LSP1, MRPL23, MS4A4A, NCR3, NOL10, NUMB, SARM1, SH2B3, SYTL3, TCF20, ZMYM2, ZNF831). These genes are presented in Figure 4. Of note, three of these genes (GNA13, NCR3, SARM1), are associated with pathways related to vascular dysfunction, which are presented in Table S7. In addition, we also conducted a comparative analysis of the DEGs that three or more studies had in common (n = 67) and the candidate DEGs (n = 106) and, in the intersection of the Venn diagram, we obtained only one gene, that is CAPZA1 (Figure 4).
This analysis also pinpointed two interesting candidate DEGs that are associated with two of the analyzed traits each, namely CDKN2B-AS1 in Paper 4, which is associated with both hypertension and atherosclerosis, and HLA-DRB1 in Paper 5, which is associated with both atherosclerosis and arterial stiffness (Table S5).

3.4. Identification of Candidate Genes for Nutrigenetic Studies among the URs of DEGs

After having identified the URs of DEGs for each of the selected studies (Table S3), we aimed to identify for which of these regulators the previous GWAS studies had identified variants and risk alleles that are associated with vascular dysfunction. To this aim, we conducted comparative analyses between all GWAS genes and URs for each study separately, and identified n = 3 candidate URs for Publication 1 (APOE, EBF1, ZBTB10), n = 6 for Publication 2 (H19, KPNA2, LDLR, PBRM1, TNF, ZNF746), n = 7 for Publication 3 (ERAP1, FMN2, FOXO1, HIC1, RPTOR, TCF7L2, ZNF746), n = 25 for Publication 4 (APOE, ARNTL, CERS5, CXCL8, CYP11B2, EBF1, EDNRA, FOXC1, FOXO1, GSE1, IL6, IRF5, MKNK1, MMP3, MYO9B, NFATC2, NPAS3, PLCE1, PPARG, PTPN11, SMARCA4, TCF20, TCF7L2, TNF, ZBTB10), and n = 22 for Publication 5 (APOE, EBF1, FOXC1, IL6, IRF5, KPNA2, LDLR, LSP1, MYO6, NCOR2, NCR3, NFATC2, NR1H3, PNPT1, PPARG, PRDM16, PTPN11, RPTOR, SMARCA4, TNF, USP8, ZBTB10). After pulling together all of these genes and removing duplicates, we obtained n = 42 URs of flavonoid-modulated genes as potential candidate genes for future nutrigenetic studies on vascular effects of these bioactives (Table S6B), here referred to as candidate URs. Notably, the expression of some of these candidate URs is modulated with respective flavonoid interventions, such as: KPNA2 for Publication 2, ARNTL, MKNK1, MYO9B, TCF20 for Publication 4, and IRF5, NCR3 for Publication 5.

3.5. Candidate Genes (DEGs and URs) in Published Studies on Genetic Polymorphisms, Cardiovascular Diseases, and Nutrition

To further prioritize candidate genes for subsequent analyses, we conducted a literature search for published studies that showed associations of these genes’ polymorphisms with cardiovascular diseases and atherosclerosis. For this purpose, in mid-August 2022, we searched PubMed applying the following wording: ((XXX[Title/Abstract]) AND ((SNP[Title/Abstract] OR polymorphism[Title/Abstract]))) AND (cardiovascular[Title/Abstract] OR atherosclerosis[Title/Abstract]), where “XXX” was replaced by the gene of interest. This literature search showed that among candidate DEGs, the number of publications was the largest for MTHFR (n = 394), LPL (n = 92), LDLR (n = 74), ALDH2 (n = 47), and TCF7L2 (n = 34), while among candidate URs, the number of publications was the largest for APOE (n = 383), IL6 (n = 233), TNF (n = 140), LDLR (n = 74), MMP3 (n = 46), CYP11B2 (n = 45), TCF7L2 (n = 34), and PPRG (n = 24).
Furthermore, we also conducted a PubMed search to identify previously published studies that showed associations of candidate genes’ polymorphisms with diet and nutrition. For this purpose, we applied the following wording: ((SNP[Title/Abstract]) AND ((nutrition[Title/Abstract] OR nutrient[Title/Abstract] OR diet[Title/Abstract]))) AND (XXX[Title/Abstract]), where “XXX” was replaced by the gene of interest. This literature search showed that among candidate DEGs, the number of publications was the largest for MTHFR (n = 22), LPL (n = 14), LDLR (n = 5), and TCF7L2 (n = 5), while among candidate URs, the number of publications was the largest for APOE (n = 26), IL6 (n = 13), TNF (n = 10), PPARG (n = 6), LDLR (n = 5), and TCF7L2 (n = 5).

3.6. Selection of Top-Priority Candidate Genes and Their Polymorphisms Potentially Associated with Flavonoids and Vascular Health

Our next step was directed towards final prioritization of a subset of candidate genes. To this end, using the results from the above analyses, we conducted the following additional analyses: (a) intersection between candidate DEGs in two or more papers AND candidate DEGs in canonical pathways; (b) intersection between candidate DEGs in two or more papers AND candidate URs; (c) intersection between candidate DEGs AND top genes in published studies on genetic polymorphisms, cardiovascular diseases, and nutrition; (d) intersection between candidate URs AND top genes in published studies on genetic polymorphisms, cardiovascular diseases, and nutrition; (e) intersection between candidate DEGs in canonical pathways AND candidate URs; (f) intersection between candidate DEGs AND DEGs in three or more papers (i.e., candidate DEGs in three or more papers). The rationale behind conducting these analyses is the following: If a candidate gene takes some of the central positions within these integrative analyses, it is much more likely that it will significantly influence the vascular effects of flavonoids. These analyses allowed us to select n = 20 top-priority candidate genes: ALDH2, APOE, CAPZA1, CYP11B2, GNA13, IL6, IRF5, LDLR, LPL, LSP1, MKNK1, MMP3, MTHFR, MYO6, NCR3, PPARG, SARM1, TCF20, TCF7L2, and TNF.
For each of these top-priority candidate genes, we interrogated the Variation Viewer database to identify their variant types, molecular consequences, most severe clinical significances, and top 10 genetic variants with highest frequencies, which are presented in Table 3. Also, for each of these genes, we interrogated the PharmGKB database with the aim of identifying variants that have already been shown to have specific pharmacologic relevance, and the number of associations is also presented in Table 3.
In addition, GWAS-reported variants of selected top-priority candidate genes associated with vascular dysfunction were identified back in the GWAS Catalog, while their frequencies in the global population were identified in the dbSNP database (Table 4). In the same database, we also searched for the previously reported clinical significance of each of these variants and found that rs671 (ALDH2) is a risk factor, pathogenic, drug response, and protective; rs6418 (CYP11B2 and also GML) is benign; rs1799998 (CYP11B2 and also LY6E-DT) has association and is benign; rs6511720 (LDLR) is benign; rs17367504 (MTHFR) is benign; rs7903146 (TCF7L2) is a likely risk allele and risk factor. For other genetic variants included in Table 4, no clinical significance was reported in the dbSNP database. In addition, the role of each candidate gene, whether it is a DEG, UR, or both, is reported in Table 4.

4. Discussion

Gene–diet interaction has long been considered one of the key determinants of interindividual variability in the effect of a number of dietary factors [44]. Among the classic examples are studies on the interactions between genetic polymorphism, intake of bioactives related to coffee consumption and the risk of acute myocardial infarction. These studies identified caffeine as a key factor for increased risk only for individuals with slow caffeine metabolism [45] that is associated with the −163A > C (rs762551) single nucleotide polymorphism of the Cytochrome P450 Family 1 Subfamily A Member 2 (CYP1A2) gene. This SNP has been shown to alter the inducibility and activity of the CYP1A2 enzyme, which accounts for approximately 95% of caffeine metabolism in the body. Individuals with the AC or CC genotype are categorized as slow metabolizers, while individuals with the AA genotype are categorized as fast metabolizers [46]. Furthermore, a recent study has shown an increased risk of hypertension and renal dysfunction with heavy coffee intake, but only among individuals with the AC and CC genotypes of CYP1A2 at rs762551 [47]. Consequently, an influence of genetic polymorphisms on the vasculoprotective properties of dietary flavonoids can also be expected, as one of the determinants of interindividual variability in the effect. A recent study identified for the first time genetic polymorphisms that determine the effect of orange juice consumption on circulating lipids and blood pressure. In the study group of 46 participants, medium or high excretors of flavanone metabolites, it was observed that for the APOA1_rs964184 polymorphism, the CC genotype is associated with a decrease in circulating triglycerides and blood pressure, both systolic and diastolic. Additionally, for the LPL_rs12678919 polymorphism, the AA genotype was associated with a change in blood lipids [20].
Given that vascular health is governed not only by genes directly associated with vascular tone, vascular permeability, and circulating lipoproteins, but also by genes involved in general metabolic dysregulation, it is realistic to expect that a greater number of genetic polymorphisms determine the interindividual variabilities of the vascular health effects of flavonoids, which highlights the need for their identification and further nutrigenetic studies. To address this issue, GWAS represent a valuable source of information, the approach of which involves genome-wide analysis of genotypes of a large number of individuals to identify variants associated with a specific disease or health-related trait compared to healthy individuals, i.e., identification of genotype–phenotype associations. So far, GWAS have identified hundreds of genetic variants that are associated with different diseases or health-related traits in humans [48]. More importantly, the data from numerous GWAS analyses are aggregated, structured, and standardized into a publicly accessible database [49], allowing them to be utilized in future research. A major limitation of GWAS is that they only provide a statistical association between a specific genetic variant and a given disease or trait. In other words, GWAS provide genes associated with specific diseases or traits and do not necessarily pinpoint causal variants and genes [48]. Understanding potential functional consequences of identified variants represents a considerable challenge, for which various approaches have been proposed [21,22,23,24,50,51]. One approach proposed in a recent study consisted of integrating GWAS and mRNA microarray data to computationally identify key disease pathways, upstream regulators, and downstream therapeutic targets in primary biliary cholangitis [52]. Specifically, GWAS analysis conducted on 1920 patients and 1770 healthy controls identified 261 genes associated with primary biliary cholangitis, in parallel to mRNA microarray analysis that was conducted in liver needle biopsy specimens from 36 patients and 5 controls and identified 1574 DEGs. Subsequent functional analyses, which included signaling networks analyses and analyses of upstream regulators, enabled the prediction of central regulators in disease susceptibility and identified potential downstream therapeutic targets [52].
To address our aim, which is to identify candidate genes which polymorphisms potentially determine the interindividual variabilities in the effects of flavonoids on vascular health, we employed an integrative analysis of GWAS (i.e., genetic) and mRNA microarray (i.e., genomic) data using (a) available global transcriptomic data of published human intervention studies on flavonoids and vascular health demonstrating a positive effect on vascular function and (b) genes associated with vascular dysfunction-related traits (hypertension, atherosclerosis, and arterial stiffness) identified from GWAS. In addition, for each set of genomic data, we identified the URs, molecules capable of regulating the expression of DEGs. Some of these URs are proteins, which are inherently prone to genetic variability, thus potentially serving as a source for significant interindividual variabilities in flavonoids and vascular health. Even though flavonoids are a large and diverse class of (poly)phenols, previous transcriptomic studies have shown that not only (poly)phenols from specific classes, but (poly)phenols in general can exhibit common molecular mechanisms of action [53], most likely because some of them are metabolized by gut microbiota to similar or identical metabolites that mediate their molecular mechanisms of action [54], which was the rationale behind our decision to include human intervention studies conducted with different dietary flavonoids. By employing integrative analysis of transcriptomic and GWAS datasets, we added an important step in the identification of candidate genes for future nutrigenetic studies. This integrative analysis identified 106 candidate DEGs and 42 candidate URs. Subsequent functional analyses and a literature search of these candidate DEGs and URs identified 20 top-priority candidates: ALDH2, APOE, CAPZA1, CYP11B2, GNA13, IL6, IRF5, LDLR, LPL, LSP1, MKNK1, MMP3, MTHFR, MYO6, NCR3, PPARG, SARM1, TCF20, TCF7L2, and TNF. It should be added that our study only focused on genes directly associated with the vascular effects of flavonoids and did not consider genes involved in their absorption and metabolism. Therefore, the results of our study should be verified by carefully designed human nutrigenetic studies that will only include individuals with high levels of circulating metabolites of the tested flavonoids. Such an approach would eliminate the influence of interindividual variabilities in the absorption and metabolism of flavonoids, phenomena that are well-identified but still poorly understood.
Among the top candidate genes and their known SNPs, there is evidence about their functionality related to vascular health. For example, the rs7903146 variant of the Transcription Factor 7 Like 2 (TCF7L2) gene was identified as associated with type 2 diabetes [55], and the T allele of this variant strongly predicts future type 2 diabetes. This allele is associated with enhanced expression of TCF7L2 in human islets as well as with impaired insulin secretion [56]. Furthermore associations between rs7903146 and (a) elevated serum triglycerides in patients with familial combined hyperlipidemia [57], (b) impaired postprandial lipid metabolism in healthy young males and elderly persons [58], (c) inflammation, metabolic dysregulation, and atherosclerotic cardiovascular diseases [59] were observed. TCF7L2 is a transcription factor and the ultimate effector of the Wnt signaling pathway, which plays an important protective role in the development of atherosclerotic cardiovascular diseases [59]. Regarding the results from global gene expression studies on flavonoids and vascular function, TCF7L2 has been identified as a differentially expressed gene in one study and as an upstream regulator in two studies. These observations suggest that the TCF7L2 gene is one of the potential key mediators of the interindividual differences to flavonoid intake.
Another polymorphism with proven functionality in vascular dysfunction is rs6511720 of the Low-Density Lipoprotein Receptor (LDLR) gene, for which a recent study has shown a significant association with susceptibility to coronary artery disease, as well as with regression of carotid intima-media thickness and changes in plasma lipids during rosuvastatin therapy [60]. A single-nucleotide polymorphism, rs1799998, in the aldosterone synthase gene, Cytochrome P450 Family 11 Subfamily B Member 2 (CYP11B2), has also been reported to associate with cardiovascular diseases, such as atrial fibrillation [61] or intracranial large artery stenosis [62]. In addition, it has been shown that this polymorphism is associated with a predisposition to the development of late in-stent restenosis in heterozygous patients with stable coronary artery disease [63]. Other polymorphisms that have not only been statistically associated with vascular dysfunction but have also been functionally related to it include Aldehyde Dehydrogenase 2 Family Member (ALDH2) rs671 and Methylenetetrahydrofolate Reductase (MTHFR) rs17367504. These genes are crucial in alcohol metabolism and folate/homocysteine metabolism, respectively. The rs671 polymorphism in ALDH2 was pinpointed as a risk factor for the occurrence of death from cardio-cerebrovascular complications in patients with type 2 diabetes [64] and has recently been characterized as a novel regulator of cholesterol biosynthesis [65]. For the MTHFR rs17367504, it was not only associated with hypertension in a previous GWAS but was also included in the calculation of genetic risk score (GRS) in a study aiming to evaluate whether the association between GRS and blood pressure was modified by usual coffee consumption. This study revealed that individuals with greater GRS present high blood pressure associated with higher coffee consumption, highlighting the particular importance of reducing coffee intake in individuals who are genetically predisposed to this cardiovascular disease risk factor [66]. Moreover, polymorphisms in LPL and APOE genes were suggested to modulate the effects of orange juice rich in flavanone on vascular function [20]. Taken together, these examples strongly suggest that the candidate genes identified using our integrative analyses of genomic and GWAS data are good candidates, demonstrating the power of such an approach for the identification of novel, still unexplored candidate genes involved in interindividual variability in response to flavonoid intake and vascular health.
There are several limitations to this study. The major limitation is the small number of genomic datasets used, five, corresponding to the only studies available that aimed to assess global genomic change induced by flavonoids in human volunteers associated with positive vascular health effects. Also, the number of studies is limited as we only used genomic data that were obtained using a microarray approach. In addition, recent studies have revealed that (poly)phenols exert their health effects by modulating the expression of not only protein coding genes but also the expression of protein non-coding genes, such as microRNAs or long non-coding RNAs [53,67], and by exerting changes in the DNA methylation profile [68]. Also, GWAS have identified vascular dysfunction-associated variants in the non-coding elements of the genome [69]. Therefore, integration of genomic data with GWAS data in future analyses should include information about changes in the expression of all types of RNA as well as the DNA methylation profile.

5. Conclusions

In conclusion, we performed an integrated bioinformatic analysis with large-scale GWAS and transcriptomic data to generate a refined list of candidate causal genes for interindividual variability in response to flavonoid intake. These results should serve as an important resource, facilitating the focusing of nutrigenetic research in the field of plant food bioactives to identify gene variants associated with a better health response to these bioactives, and therefore build a foundation for precision nutrition research in the field of (poly)phenols.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu16091362/s1, Table S1: Differentially expressed genes (DEGs) from the selected studies; Table S2: Common differentially expressed genes (DEGs) across the selected studies; Table S3: Upstream regulators (URs) across the selected studies; Table S4: Genes associated with vascular dysfunction, identified with previous genome-wide association studies (GWAS); Table S5: Comparative analysis of DEGs and trait-specific genes identified from GWAS; Table S6: Candidate DEGs and candidate URs; Table S7. Candidate DEGs placed in canonical pathways.

Author Contributions

Conceptualization, T.R. and D.M.; data curation, T.R., F.P. and D.M.; formal analysis, T.R., F.P. and D.M.; methodology, T.R. and D.M.; visualization, T.R., F.P. and D.M.; writing—original draft, T.R. and D.M.; writing—review & editing, T.R., F.P. and D.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Material, further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

Acronyms

SNP—single nucleotide polymorphism; CAD—coronary artery disease; AF—atrial fibrillation; GWAS—genome-wide association studies; DEGs—differentially expressed genes; URs—upstream regulators; IPA—Ingenuity Pathway Analysis; KEGG—Kyoto Encyclopedia of Genes and Genomes; GRS—genetic risk score.

References

  1. Grosso, G.; Stepaniak, U.; Topor-Mądry, R.; Szafraniec, K.; Pająk, A. Estimated dietary intake and major food sources of polyphenols in the Polish arm of the HAPIEE study. Nutrition 2014, 30, 1398–1403. [Google Scholar] [CrossRef]
  2. Miranda, A.M.; Steluti, J.; Fisberg, R.M.; Marchioni, D.M. Dietary intake and food contributors of polyphenols in adults and elderly adults of Sao Paulo: A population-based study. Br. J. Nutr. 2016, 115, 1061–1070. [Google Scholar] [CrossRef]
  3. Huang, Q.; Braffett, B.H.; Simmens, S.J.; Young, H.A.; Ogden, C.L. Dietary Polyphenol Intake in US Adults and 10-Year Trends: 2007–2016. J. Acad. Nutr. Diet. 2020, 120, 1821–1833. [Google Scholar] [CrossRef]
  4. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal 2013, 18, 1818–1892. [Google Scholar] [CrossRef]
  5. Neveu, V.; Perez-Jiménez, J.; Vos, F.; Crespy, V.; du Chaffaut, L.; Mennen, L.; Knox, C.; Eisner, R.; Cruz, J.; Wishart, D.; et al. Phenol-Explorer: An online comprehensive database on polyphenol contents in foods. Database 2010, 2010, bap024. [Google Scholar] [CrossRef]
  6. Vivarelli, S.; Costa, C.; Teodoro, M.; Giambò, F.; Tsatsakis, A.M.; Fenga, C. Polyphenols: A route from bioavailability to bioactivity addressing potential health benefits to tackle human chronic diseases. Arch. Toxicol. 2023, 97, 3–38. [Google Scholar] [CrossRef]
  7. Guo, X.F.; Ruan, Y.; Li, Z.H.; Li, D. Flavonoid subclasses and type 2 diabetes mellitus risk: A meta-analysis of prospective cohort studies. Crit. Rev. Food Sci. Nutr. 2019, 59, 2850–2862. [Google Scholar] [CrossRef]
  8. Liu, F.; Sirisena, S.; Ng, K. Efficacy of flavonoids on biomarkers of type 2 diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2023, 63, 4916–4941. [Google Scholar] [CrossRef]
  9. Kay, C.D.; Hooper, L.; Kroon, P.A.; Rimm, E.B.; Cassidy, A. Relative impact of flavonoid composition, dose and structure on vascular function: A systematic review of randomised controlled trials of flavonoid-rich food products. Mol. Nutr. Food Res. 2012, 56, 1605–1616. [Google Scholar] [CrossRef]
  10. Sesso, H.D.; Manson, J.E.; Aragaki, A.K.; Rist, P.M.; Johnson, L.G.; Friedenberg, G.; Copeland, T.; Clar, A.; Mora, S.; Moorthy, M.V.; et al. Effect of cocoa flavanol supplementation for the prevention of cardiovascular disease events: The COcoa Supplement and Multivitamin Outcomes Study (COSMOS) randomized clinical trial. Am. J. Clin. Nutr. 2022, 115, 1490–1500. [Google Scholar] [CrossRef]
  11. Bayram, B.; González-Sarrías, A.; Istas, G.; Garcia-Aloy, M.; Morand, C.; Tuohy, K.; García-Villalba, R.; Mena, P. Breakthroughs in the Health Effects of Plant Food Bioactives: A Perspective on Microbiomics, Nutri(epi)genomics, and Metabolomics. J. Agric. Food Chem. 2018, 66, 10686–10692. [Google Scholar] [CrossRef]
  12. Curtis, P.J.; Potter, J.; Kroon, P.A.; Wilson, P.; Dhatariya, K.; Sampson, M.; Cassidy, A. Vascular function and atherosclerosis progression after 1 y of flavonoid intake in statin-treated postmenopausal women with type 2 diabetes: A double-blind randomized controlled trial. Am. J. Clin. Nutr. 2013, 97, 936–942. [Google Scholar] [CrossRef]
  13. Morand, C.; Tomás-Barberán, F.A. Contribution of plant food bioactives in promoting health effects of plant foods: Why look at interindividual variability? Eur. J. Nutr. 2019, 58, 13–19. [Google Scholar] [CrossRef]
  14. Donato, A.J.; Machin, D.R.; Lesniewski, L.A. Mechanisms of Dysfunction in the Aging Vasculature and Role in Age-Related Disease. Circ. Res. 2018, 123, 825–848. [Google Scholar] [CrossRef]
  15. Barbosa, S.S.; Sousa, L.C.M.; de Oliveira Silva, D.F.; Pimentel, J.B.; Evangelista, K.; Lyra, C.O.; Lopes, M.; Lima, S. A Systematic Review on Processed/Ultra-Processed Foods and Arterial Hypertension in Adults and Older People. Nutrients 2022, 14, 1215. [Google Scholar] [CrossRef]
  16. Koenen, M.; Hill, M.A.; Cohen, P.; Sowers, J.R. Obesity, Adipose Tissue and Vascular Dysfunction. Circ. Res. 2021, 128, 951–968. [Google Scholar] [CrossRef]
  17. ALrefai, A.A.; Habib, M.S.; Yaseen, R.I.; Gabr, M.K.; Habeeb, R.M. Association of endothelial nitric oxide synthase (eNOS) gene G894T polymorphism with hypertension risk and complications. Mol. Cell Biochem. 2016, 421, 103–110. [Google Scholar] [CrossRef]
  18. Kim, M.P.; Wahl, L.M.; Yanek, L.R.; Becker, D.M.; Becker, L.C. A monocyte chemoattractant protein-1 gene polymorphism is associated with occult ischemia in a high-risk asymptomatic population. Atherosclerosis 2007, 193, 366–372. [Google Scholar] [CrossRef]
  19. George, T.W.; Waroonphan, S.; Niwat, C.; Gordon, M.H.; Lovegrove, J.A. The Glu298Asp single nucleotide polymorphism in the endothelial nitric oxide synthase gene differentially affects the vascular response to acute consumption of fruit and vegetable puree based drinks. Mol. Nutr. Food Res. 2012, 56, 1014–1024. [Google Scholar] [CrossRef]
  20. Fraga, L.N.; Milenkovic, D.; Coutinho, C.P.; Rozenbaum, A.C.; Lajolo, F.M.; Hassimotto, N.M.A. Interaction between APOE, APOA1, and LPL Gene Polymorphisms and Variability in Changes in Lipid and Blood Pressure following Orange Juice Intake: A Pilot Study. Mol. Nutr. Food Res. 2023, 67, e2200847. [Google Scholar] [CrossRef]
  21. Li, B.; Ritchie, M.D. From GWAS to Gene: Transcriptome-Wide Association Studies and Other Methods to Functionally Understand GWAS Discoveries. Front. Genet. 2021, 12, 713230. [Google Scholar] [CrossRef]
  22. Zhu, Z.; Zhang, F.; Hu, H.; Bakshi, A.; Robinson, M.R.; Powell, J.E.; Montgomery, G.W.; Goddard, M.E.; Wray, N.R.; Visscher, P.M.; et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 2016, 48, 481–487. [Google Scholar] [CrossRef]
  23. Hao, K.; Ermel, R.; Sukhavasi, K.; Cheng, H.; Ma, L.; Li, L.; Amadori, L.; Koplev, S.; Franzén, O.; d’Escamard, V.; et al. Integrative Prioritization of Causal Genes for Coronary Artery Disease. Circ. Genom. Precis. Med. 2022, 15, e003365. [Google Scholar] [CrossRef]
  24. Wang, B.; Lunetta, K.L.; Dupuis, J.; Lubitz, S.A.; Trinquart, L.; Yao, L.; Ellinor, P.T.; Benjamin, E.J.; Lin, H. Integrative Omics Approach to Identifying Genes Associated With Atrial Fibrillation. Circ. Res. 2020, 126, 350–360. [Google Scholar] [CrossRef]
  25. Ruskovska, T.; Budić-Leto, I.; Corral-Jara, K.F.; Ajdžanović, V.; Arola-Arnal, A.; Bravo, F.I.; Deligiannidou, G.E.; Havlik, J.; Janeva, M.; Kistanova, E.; et al. Systematic analysis of nutrigenomic effects of polyphenols related to cardiometabolic health in humans—Evidence from untargeted mRNA and miRNA studies. Ageing Res. Rev. 2022, 79, 101649. [Google Scholar] [CrossRef]
  26. Milenkovic, D.; Deval, C.; Dubray, C.; Mazur, A.; Morand, C. Hesperidin displays relevant role in the nutrigenomic effect of orange juice on blood leukocytes in human volunteers: A randomized controlled cross-over study. PLoS ONE 2011, 6, e26669. [Google Scholar] [CrossRef]
  27. Milenkovic, D.; Vanden Berghe, W.; Boby, C.; Leroux, C.; Declerck, K.; Szarc vel Szic, K.; Heyninck, K.; Laukens, K.; Bizet, M.; Defrance, M.; et al. Dietary flavanols modulate the transcription of genes associated with cardiovascular pathology without changes in their DNA methylation state. PLoS ONE 2014, 9, e95527. [Google Scholar] [CrossRef]
  28. Rodriguez-Mateos, A.; Istas, G.; Boschek, L.; Feliciano, R.P.; Mills, C.E.; Boby, C.; Gomez-Alonso, S.; Milenkovic, D.; Heiss, C. Circulating Anthocyanin Metabolites Mediate Vascular Benefits of Blueberries: Insights From Randomized Controlled Trials, Metabolomics, and Nutrigenomics. J. Gerontol. A Biol. Sci. Med. Sci. 2019, 74, 967–976. [Google Scholar] [CrossRef]
  29. Krga, I.; Corral-Jara, K.F.; Barber-Chamoux, N.; Dubray, C.; Morand, C.; Milenkovic, D. Grapefruit Juice Flavanones Modulate the Expression of Genes Regulating Inflammation, Cell Interactions and Vascular Function in Peripheral Blood Mononuclear Cells of Postmenopausal Women. Front. Nutr. 2022, 9, 907595. [Google Scholar] [CrossRef] [PubMed]
  30. Milenkovic, D.; Rodriguez-Mateos, A.; Lucosz, M.; Istas, G.; Declerck, K.; Sansone, R.; Deenen, R.; Köhrer, K.; Corral-Jara, K.F.; Altschmied, J.; et al. Flavanol Consumption in Healthy Men Preserves Integrity of Immunological-Endothelial Barrier Cell Functions: Nutri(epi)genomic Analysis. Mol. Nutr. Food Res. 2022, 66, e2100991. [Google Scholar] [CrossRef] [PubMed]
  31. Buniello, A.; MacArthur, J.A.L.; Cerezo, M.; Harris, L.W.; Hayhurst, J.; Malangone, C.; McMahon, A.; Morales, J.; Mountjoy, E.; Sollis, E.; et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res 2019, 47, D1005–D1012. [Google Scholar] [CrossRef] [PubMed]
  32. Gerstner, N.; Kehl, T.; Lenhof, K.; Müller, A.; Mayer, C.; Eckhart, L.; Grammes, N.L.; Diener, C.; Hart, M.; Hahn, O.; et al. GeneTrail 3: Advanced high-throughput enrichment analysis. Nucleic Acids Res. 2020, 48, W515–W520. [Google Scholar] [CrossRef]
  33. Kanehisa, M.; Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000, 28, 27–30. [Google Scholar] [CrossRef]
  34. Agrawal, A.; Balcı, H.; Hanspers, K.; Coort, S.L.; Martens, M.; Slenter, D.N.; Ehrhart, F.; Digles, D.; Waagmeester, A.; Wassink, I.; et al. WikiPathways 2024: Next generation pathway database. Nucleic Acids Res 2024, 52, D679–D689. [Google Scholar] [CrossRef]
  35. Slenter, D.N.; Kutmon, M.; Hanspers, K.; Riutta, A.; Windsor, J.; Nunes, N.; Mélius, J.; Cirillo, E.; Coort, S.L.; Digles, D.; et al. WikiPathways: A multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018, 46, D661–D667. [Google Scholar] [CrossRef] [PubMed]
  36. Heberle, H.; Meirelles, G.V.; da Silva, F.R.; Telles, G.P.; Minghim, R. InteractiVenn: A web-based tool for the analysis of sets through Venn diagrams. BMC Bioinform. 2015, 16, 169. [Google Scholar] [CrossRef] [PubMed]
  37. Whirl-Carrillo, M.; Huddart, R.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Whaley, R.; Klein, T.E. An Evidence-Based Framework for Evaluating Pharmacogenomics Knowledge for Personalized Medicine. Clin. Pharmacol. Ther. 2021, 110, 563–572. [Google Scholar] [CrossRef]
  38. Whirl-Carrillo, M.; McDonagh, E.M.; Hebert, J.M.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Altman, R.B.; Klein, T.E. Pharmacogenomics knowledge for personalized medicine. Clin. Pharmacol. Ther. 2012, 92, 414–417. [Google Scholar] [CrossRef]
  39. Stelzer, G.; Rosen, N.; Plaschkes, I.; Zimmerman, S.; Twik, M.; Fishilevich, S.; Stein, T.I.; Nudel, R.; Lieder, I.; Mazor, Y.; et al. The GeneCards Suite: From Gene Data Mining to Disease Genome Sequence Analyses. Curr. Protoc. Bioinform. 2016, 54, 1.30.31–31.30.33. [Google Scholar] [CrossRef]
  40. Morand, C.; Dubray, C.; Milenkovic, D.; Lioger, D.; Martin, J.F.; Scalbert, A.; Mazur, A. Hesperidin contributes to the vascular protective effects of orange juice: A randomized crossover study in healthy volunteers. Am. J. Clin. Nutr. 2011, 93, 73–80. [Google Scholar] [CrossRef]
  41. Weseler, A.R.; Ruijters, E.J.; Drittij-Reijnders, M.J.; Reesink, K.D.; Haenen, G.R.; Bast, A. Pleiotropic benefit of monomeric and oligomeric flavanols on vascular health--a randomized controlled clinical pilot study. PLoS ONE 2011, 6, e28460. [Google Scholar] [CrossRef]
  42. Sansone, R.; Rodriguez-Mateos, A.; Heuel, J.; Falk, D.; Schuler, D.; Wagstaff, R.; Kuhnle, G.G.; Spencer, J.P.; Schroeter, H.; Merx, M.W.; et al. Cocoa flavanol intake improves endothelial function and Framingham Risk Score in healthy men and women: A randomised, controlled, double-masked trial: The Flaviola Health Study. Br. J. Nutr. 2015, 114, 1246–1255. [Google Scholar] [CrossRef] [PubMed]
  43. Habauzit, V.; Verny, M.A.; Milenkovic, D.; Barber-Chamoux, N.; Mazur, A.; Dubray, C.; Morand, C. Flavanones protect from arterial stiffness in postmenopausal women consuming grapefruit juice for 6 mo: A randomized, controlled, crossover trial. Am. J. Clin. Nutr. 2015, 102, 66–74. [Google Scholar] [CrossRef] [PubMed]
  44. El-Sohemy, A. Nutrigenetics. Forum Nutr. 2007, 60, 25–30. [Google Scholar] [CrossRef] [PubMed]
  45. Cornelis, M.C.; El-Sohemy, A.; Kabagambe, E.K.; Campos, H. Coffee, CYP1A2 genotype, and risk of myocardial infarction. JAMA 2006, 295, 1135–1141. [Google Scholar] [CrossRef] [PubMed]
  46. Guest, N.; Corey, P.; Vescovi, J.; El-Sohemy, A. Caffeine, CYP1A2 Genotype, and Endurance Performance in Athletes. Med. Sci. Sports Exerc. 2018, 50, 1570–1578. [Google Scholar] [CrossRef]
  47. Mahdavi, S.; Palatini, P.; El-Sohemy, A. CYP1A2 Genetic Variation, Coffee Intake, and Kidney Dysfunction. JAMA Netw. Open 2023, 6, e2247868. [Google Scholar] [CrossRef] [PubMed]
  48. Tam, V.; Patel, N.; Turcotte, M.; Bossé, Y.; Paré, G.; Meyre, D. Benefits and limitations of genome-wide association studies. Nat. Rev. Genet. 2019, 20, 467–484. [Google Scholar] [CrossRef]
  49. Sollis, E.; Mosaku, A.; Abid, A.; Buniello, A.; Cerezo, M.; Gil, L.; Groza, T.; Güneş, O.; Hall, P.; Hayhurst, J.; et al. The NHGRI-EBI GWAS Catalog: Knowledgebase and deposition resource. Nucleic Acids Res. 2023, 51, D977–D985. [Google Scholar] [CrossRef]
  50. Cano-Gamez, E.; Trynka, G. From GWAS to Function: Using Functional Genomics to Identify the Mechanisms Underlying Complex Diseases. Front. Genet. 2020, 11, 424. [Google Scholar] [CrossRef]
  51. Zhu, M.; Zhao, S. Candidate gene identification approach: Progress and challenges. Int. J. Biol. Sci. 2007, 3, 420–427. [Google Scholar] [CrossRef] [PubMed]
  52. Ueno, K.; Aiba, Y.; Hitomi, Y.; Shimoda, S.; Nakamura, H.; Gervais, O.; Kawai, Y.; Kawashima, M.; Nishida, N.; Kohn, S.S.; et al. Integrated GWAS and mRNA Microarray Analysis Identified IFNG and CD40L as the Central Upstream Regulators in Primary Biliary Cholangitis. Hepatol. Commun. 2020, 4, 724–738. [Google Scholar] [CrossRef] [PubMed]
  53. Milenkovic, D.; Deval, C.; Gouranton, E.; Landrier, J.F.; Scalbert, A.; Morand, C.; Mazur, A. Modulation of miRNA expression by dietary polyphenols in apoE deficient mice: A new mechanism of the action of polyphenols. PLoS ONE 2012, 7, e29837. [Google Scholar] [CrossRef] [PubMed]
  54. Williamson, G.; Clifford, M.N. Colonic metabolites of berry polyphenols: The missing link to biological activity? Br. J. Nutr. 2010, 104 (Suppl. S3), S48–S66. [Google Scholar] [CrossRef] [PubMed]
  55. Saxena, R.; Gianniny, L.; Burtt, N.P.; Lyssenko, V.; Giuducci, C.; Sjögren, M.; Florez, J.C.; Almgren, P.; Isomaa, B.; Orho-Melander, M.; et al. Common single nucleotide polymorphisms in TCF7L2 are reproducibly associated with type 2 diabetes and reduce the insulin response to glucose in nondiabetic individuals. Diabetes 2006, 55, 2890–2895. [Google Scholar] [CrossRef] [PubMed]
  56. Lyssenko, V.; Lupi, R.; Marchetti, P.; Del Guerra, S.; Orho-Melander, M.; Almgren, P.; Sjögren, M.; Ling, C.; Eriksson, K.F.; Lethagen, A.L.; et al. Mechanisms by which common variants in the TCF7L2 gene increase risk of type 2 diabetes. J. Clin. Investig. 2007, 117, 2155–2163. [Google Scholar] [CrossRef] [PubMed]
  57. Huertas-Vazquez, A.; Plaisier, C.; Weissglas-Volkov, D.; Sinsheimer, J.; Canizales-Quinteros, S.; Cruz-Bautista, I.; Nikkola, E.; Herrera-Hernandez, M.; Davila-Cervantes, A.; Tusie-Luna, T.; et al. TCF7L2 is associated with high serum triacylglycerol and differentially expressed in adipose tissue in families with familial combined hyperlipidaemia. Diabetologia 2008, 51, 62–69. [Google Scholar] [CrossRef] [PubMed]
  58. Perez-Martinez, P.; Perez-Caballero, A.I.; Garcia-Rios, A.; Yubero-Serrano, E.M.; Camargo, A.; Gomez-Luna, M.J.; Marin, C.; Gomez-Luna, P.; Dembinska-Kiec, A.; Rodriguez-Cantalejo, F.; et al. Effects of rs7903146 variation in the Tcf7l2 gene in the lipid metabolism of three different populations. PLoS ONE 2012, 7, e43390. [Google Scholar] [CrossRef] [PubMed]
  59. Li, J.; Zhou, L.; Ouyang, X.; He, P. Transcription Factor-7-Like-2 (TCF7L2) in Atherosclerosis: A Potential Biomarker and Therapeutic Target. Front. Cardiovasc. Med. 2021, 8, 701279. [Google Scholar] [CrossRef]
  60. Kononov, S.; Mal, G.; Azarova, I.; Klyosova, E.; Bykanova, M.; Churnosov, M.; Polonikov, A. Pharmacogenetic loci for rosuvastatin are associated with intima-media thickness change and coronary artery disease risk. Pharmacogenomics 2022, 23, 15–34. [Google Scholar] [CrossRef]
  61. Gouissem, I.; Midani, F.; Soualmia, H.; Bouchemi, M.; Ouali, S.; Kallele, A.; Romdhane, N.B.; Mourali, M.S.; Feki, M. Contribution of the ACE (rs1799752) and CYP11B2 (rs1799998) Gene Polymorphisms to Atrial Fibrillation in the Tunisian Population. Biol. Res. Nurs. 2022, 24, 31–39. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, M.; Sariya, S.; Khasiyev, F.; Tosto, G.; Dueker, N.D.; Cheung, Y.K.; Wright, C.B.; Sacco, R.L.; Rundek, T.; Elkind, M.S.V.; et al. Genetic determinants of intracranial large artery stenosis in the northern Manhattan study. J. Neurol. Sci. 2022, 436, 120218. [Google Scholar] [CrossRef] [PubMed]
  63. Azova, M.; Timizheva, K.; Ait Aissa, A.; Blagonravov, M.; Gigani, O.; Aghajanyan, A.; Tskhovrebova, L. Gene Polymorphisms of the Renin-Angiotensin-Aldosterone System as Risk Factors for the Development of In-Stent Restenosis in Patients with Stable Coronary Artery Disease. Biomolecules 2021, 11, 763. [Google Scholar] [CrossRef] [PubMed]
  64. He, Q.; Pan, J.; Wang, L.; Fang, Y.; Hu, R. Prospective study: Aldehyde dehydrogenase 2 gene is associated with cardio-cerebrovascular complications in type 2 diabetes patients. J. Diabetes Investig. 2021, 12, 1845–1854. [Google Scholar] [CrossRef] [PubMed]
  65. Zhong, S.; Li, L.; Liang, N.; Zhang, L.; Xu, X.; Chen, S.; Yin, H. Acetaldehyde Dehydrogenase 2 regulates HMG-CoA reductase stability and cholesterol synthesis in the liver. Redox Biol. 2021, 41, 101919. [Google Scholar] [CrossRef] [PubMed]
  66. Miranda, A.M.; Steluti, J.; Norde, M.M.; Fisberg, R.M.; Marchioni, D.M. The association between genetic risk score and blood pressure is modified by coffee consumption: Gene-diet interaction analysis in a population-based study. Clin. Nutr. 2019, 38, 1721–1728. [Google Scholar] [CrossRef]
  67. Jiang, Y.; Sun-Waterhouse, D.; Chen, Y.; Li, F.; Li, D. Epigenetic mechanisms underlying the benefits of flavonoids in cardiovascular health and diseases: Are long non-coding RNAs rising stars? Crit. Rev. Food Sci. Nutr. 2022, 62, 3855–3872. [Google Scholar] [CrossRef] [PubMed]
  68. Rajendran, P.; Abdelsalam, S.A.; Renu, K.; Veeraraghavan, V.; Ben Ammar, R.; Ahmed, E.A. Polyphenols as Potent Epigenetics Agents for Cancer. Int. J. Mol. Sci. 2022, 23, 11712. [Google Scholar] [CrossRef]
  69. Dechamethakun, S.; Muramatsu, M. Long noncoding RNA variations in cardiometabolic diseases. J. Hum. Genet. 2017, 62, 97–104. [Google Scholar] [CrossRef]
Figure 1. Flowchart of the study.
Figure 1. Flowchart of the study.
Nutrients 16 01362 g001
Figure 2. Venn diagram representing the number of common genes across the selected studies. Sets 1–5 refer to the sets of DEGs extracted from Papers 1–5, respectively. General information about the papers (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1. The online tool InteractiVenn was used for conducting the analysis and visualizing the results.
Figure 2. Venn diagram representing the number of common genes across the selected studies. Sets 1–5 refer to the sets of DEGs extracted from Papers 1–5, respectively. General information about the papers (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1. The online tool InteractiVenn was used for conducting the analysis and visualizing the results.
Nutrients 16 01362 g002
Figure 3. Comparative analysis of DEGs and trait-specific genes identified from GWAS. For each of the selected papers (Papers 1–5), DEGs were compared with trait-specific genes identified from GWAS, i.e., genes whose variants are associated with hypertension, atherosclerosis, or arterial stiffness (Table S5). Papers 1–5 refer to the papers included in this integrative analysis. General information about the papers (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1.
Figure 3. Comparative analysis of DEGs and trait-specific genes identified from GWAS. For each of the selected papers (Papers 1–5), DEGs were compared with trait-specific genes identified from GWAS, i.e., genes whose variants are associated with hypertension, atherosclerosis, or arterial stiffness (Table S5). Papers 1–5 refer to the papers included in this integrative analysis. General information about the papers (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1.
Nutrients 16 01362 g003
Figure 4. Identification of candidate DEGs which are among those that are common in the selected studies. A comparative analysis of DEGs that at least two studies have in common (n = 720) and candidate DEGs (n = 106) retrieved n = 15 genes with potentially greater influence on interindividual variability of the vascular effects of flavonoids intake: CAPZA1, FSTL4, GNA13, LSP1, MRPL23, MS4A4A, NCR3, NOL10, NUMB, SARM1, SH2B3, SYTL3, TCF20, ZMYM2, ZNF831. Papers 1–5 refer to the papers included in this integrative analysis. General information about these papers (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1. The online tool InteractiVenn was used for conducting the analysis and visualizing the results.
Figure 4. Identification of candidate DEGs which are among those that are common in the selected studies. A comparative analysis of DEGs that at least two studies have in common (n = 720) and candidate DEGs (n = 106) retrieved n = 15 genes with potentially greater influence on interindividual variability of the vascular effects of flavonoids intake: CAPZA1, FSTL4, GNA13, LSP1, MRPL23, MS4A4A, NCR3, NOL10, NUMB, SARM1, SH2B3, SYTL3, TCF20, ZMYM2, ZNF831. Papers 1–5 refer to the papers included in this integrative analysis. General information about these papers (Paper 1 [26]; Paper 2 [27]; Paper 3 [28]; Paper 4 [30]; Paper 5 [29]) is presented in Table 1. The online tool InteractiVenn was used for conducting the analysis and visualizing the results.
Nutrients 16 01362 g004
Table 1. Human intervention studies (Papers 1–5) included in this integrative analysis, and associated papers reporting vascular function-related outcomes.
Table 1. Human intervention studies (Papers 1–5) included in this integrative analysis, and associated papers reporting vascular function-related outcomes.
PaperTitle and ReferenceStudy PopulationBioactivesOutcomesAssociated Paper for Outcomes
1.Hesperidin displays relevant role in the nutrigenomic effect of orange juice on blood leukocytes in human volunteers: a randomized controlled cross-over study [26]Healthy, middle-aged, moderately overweight menHesperidinDecreased diastolic blood pressureHesperidin contributes to the vascular protective effects of orange juice: a randomized crossover study in healthy volunteers [40]
2.Dietary flavanols modulate the transcription of genes associated with cardiovascular pathology without changes in their DNA methylation state [27]Non-obese, healthy male smokers, smoking 10 and more cigarettes per day for at least 5 yearsMonomeric and oligomeric flavanols from grape seedsImproved vascular health indexPleiotropic benefit of monomeric and oligomeric flavanols on vascular health: a randomized controlled clinical pilot study [41]
3.Circulating anthocyanin metabolites mediate vascular benefits of blueberries: insights from randomized controlled trials, metabolomics, and nutrigenomics [28]Healthy male volunteersWild blueberry anthocyaninsIncreased flow-mediated vasodilatation
Decreased 24 h systolic blood pressure
/
4.Flavanol consumption in healthy men preserves integrity of immunological–endothelial barrier cell functions: nutri(epi)genomic analysis [30]Healthy middle-aged menCocoa flavanolsIncreased flow-mediated vasodilatation
Decreased systolic and diastolic blood pressure
Decreased pulse wave velocity
Cocoa flavanol intake improves endothelial function and Framingham Risk Score in healthy men and women: a randomised, controlled, double-masked trial: the Flaviola Health Study [42]
5.Grapefruit juice flavanones modulate the expression of genes regulating inflammation, cell interactions and vascular function in peripheral blood mononuclear cells of postmenopausal women [29]Healthy, non-smoking women, 3 to 10 years after menopauseGrapefruit juice flavanonesDecreased carotid–femoral pulse wave velocityFlavanones protect from arterial stiffness in postmenopausal women consuming grapefruit juice for 6 mo: a randomized, controlled, crossover trial [43]
Papers 1–5 report human intervention studies with flavonoids demonstrating beneficial effects on vascular function together with modulations in global gene expression in peripheral blood cells.
Table 2. Functional analysis of candidate DEGs: candidate DEGs that are placed in canonical pathways relevant for vascular dysfunction.
Table 2. Functional analysis of candidate DEGs: candidate DEGs that are placed in canonical pathways relevant for vascular dysfunction.
Canonical PathwaysNumber of HitsGenes
Pathways directly involved in vascular dysfunction
VEGFA-VEGFR2 Signaling Pathway *6CSK, MKNK1, MYO6, PTPRJ, SMARCA2, TNXB
Regulation of actin cytoskeleton **4BRK1, CSK, GNA13, SOS2
Adherens junction3PTPRJ, TCF7L2, YES1
Angiopoietin Like Protein 8 Regulatory Pathway *3LPL, PRKAG2, SOS2
ECM-receptor interaction3NPNT, TNXB, VTN
Focal adhesion3SOS2, TNXB, VTN
Apelin signaling pathway2GNA13, PRKAG2
Cholesterol metabolism2LDLR, LPL
Composition of Lipid Particles *2LDLR, LPL
Fluid shear stress and atherosclerosis2BMPR1B, GSTA4
Glycerolipid metabolism2ALDH2, LPL
Metabolic pathway of LDL, HDL and TG, including diseases *2LDLR, LPL
Platelet activation2GNA13, LYN
Statin Pathway *2LDLR, LPL
Pathways involved in inflammation
Chemokine signaling pathway **3CSK, LYN, SOS2
Cytokine-cytokine receptor interaction3BMPR1B, GDF10, LTB
NF-kappa B signaling pathway **3LTB, LYN, TAB2
Regulation of toll-like receptor signaling pathway *3IRF5, SARM1, TAB2
B cell receptor signaling pathway2LYN, SOS2
Interleukin-11 Signaling Pathway *2FES, YES1
Natural killer cell mediated cytotoxicity2NCR3, SOS2
Structural Pathway of Interleukin 1 (IL-1) *2MKNK1, TAB2
TNF signaling pathway2DAB2IP, TAB2
Toll-like receptor signaling pathway **2IRF5, TAB2
Cell signaling pathways
MAPK signaling pathway **4MKNK1, SOS2, STK3, TAB2
PI3K-Akt signaling pathway **4MCL1, SOS2, TNXB, VTN
EGF/EGFR Signaling Pathway *3CSK, SOS2, TWIST1
Insulin signaling pathway3MKNK1, PRKAG2, SOS2
Sterol Regulatory Element-Binding Proteins (SREBP) signalling *3LDLR, LPL, PRKAG2
cGMP-PKG signaling pathway2ATP2B1, GNA13
FoxO signaling pathway2PRKAG2, SOS2
Jak-STAT signaling pathway2MCL1, SOS2
Phospholipase D signaling pathway2GNA13, SOS2
Wnt Signaling Pathway and Pluripotency *2LDLR, TCF7L2
Antioxidant protection
NRF2 pathway *2GSTA4, SLC39A8
Legend: No asterisk—KEGG pathways; * WikiPathways; ** both KEGG and WikiPathways.
Table 3. Variant types, molecular consequences, most severe clinical significances, and top 10 genetic variants with highest frequencies, as well as the number of associations with pharmacologic relevance in selected n = 20 top-priority candidate genes.
Table 3. Variant types, molecular consequences, most severe clinical significances, and top 10 genetic variants with highest frequencies, as well as the number of associations with pharmacologic relevance in selected n = 20 top-priority candidate genes.
GeneVariant Type (Number)Molecular Consequences (Number)Most Severe Clinical Significance (Number)Number of Associations with Pharmacological Significance10 Variants with Highest Frequencies
Variant IDMolecular ConsequencesAllelesAlleles with Highest FrequenciesFrequency
ALDH2single nucleotide variant (172)
deletion (3)
insertion (2)
indel (22)
missense variant (1)
intron variant (131)
3 prime UTR variant (27)
500 B downstream variant (1)
2 KB upstream variant (14)
pathogenic (1)9rs7296651
rs6489793
rs2106697
rs10774638
rs886205
rs4767939
rs10774637
rs9971942
rs10774639
rs11066028
intron variant
3 prime UTR variant
not specified
intron variant
2 KB upstream variant
intron variant
intron variant
not specified
not specified
intron variant
C,A,G
T,G
T,A,C,G
T,A,C
A,C,G,T
A,G
C,G,T
C,T
G,A,C,T
A,C,G,T
C
T
T
T
A
G
T
T
A
A
0.497804
0.497604
0.496406
0.492612
0.491214
0.419728
0.419129
0.410743
0.410743
0.372804
APOEsingle nucleotide variant (8)missense variant (3)
synonymous variant (1)
intron variant (4)
5 prime UTR variant (1)
500 B downstream variant (1)
2 KB upstream variant (1)
pathogenic (2)
drug-response (2)
33rs405509
rs440446
rs769450
rs429358
rs7412
rs769449
rs1081105
rs877973
2 KB upstream variant
missense variant, intron variant, synonymous variant
intron variant
missense variant
missense variant
intron variant
500 B downstream variant
5 prime UTR variant, intron variant
T,G
C,G,T
G,A
T,C
C,T
G,A
A,C,G
C,A,T
T
C
A
C
T
A
C
A
0.471845
0.373802
0.327276
0.150559
0.0750799
0.0648962
0.0301518
0.0159744
CAPZA1single nucleotide variant (136)
deletion (1)
insertion (1)
indel (26)
missense variant (1)
synonymous variant (2)
intron variant (149)
nc transcript variant (1)
5 prime UTR variant (3)
3 prime UTR variant (1)
500 B downstream variant (2)
2 KB upstream variant (22)
//rs3013440
rs7524494
rs7415820
rs3103450
rs2932536
rs3013439
rs12046329
rs12046466
rs12046208
rs9429486
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
G,A
A,G,T
G,A
G,A,T
G,A,T
T,A,G
T,A,C,G
T,A,C
C,T
T,A,C,G
G
A
G
G
G
T
T
T
T
T
0.478435
0.478435
0.478035
0.478035
0.478035
0.477835
0.477835
0.477835
0.436502
0.435703
CYP11B2single nucleotide variant (86)
insertion (2)
indel (3)
missense variant (9)
synonymous variant (6)
intron variant (66)
3 prime UTR variant (7)
500 B downstream variant (2)
2 KB upstream variant (5)
pathogenic (1)
association (1)
benign (17)
3rs10110732
rs28615142
rs28366703
rs13263682
rs6421
rs79201878
rs80062072
rs74838461
rs28394055
rs6429
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
C,T
T,C
A,G
A,C
T,C
A,T
A,G
T,C
C,T
G,A,C,T
C
C
G
C
C
T
G
C
T
C
0.495208
0.483227
0.482628
0.479433
0.460064
0.45607
0.45607
0.45607
0.445887
0.44389
GNA13single nucleotide variant (164)
deletion (2)
insertion (1)
indel (18)
missense variant (2)
synonymous variant (1)
intron variant (133)
5 prime UTR variant (2)
3 prime UTR variant (11)
500 B downstream variant (1)
2 KB upstream variant (6)
//rs9911189
rs2011307
rs6504271
rs12944877
rs12939956
rs7501452
rs3960369
rs8082708
rs12945514
rs4791243
intron variant
intron variant
intron variant
intron variant
not specified
not specified
not specified
intron variant
intron variant
intron variant
A,G
C,T
C,T
G,A,C,T
C,T
T,C
C,A,G,T
T,C
G,C,T
T,A,C,G
A
C
C
G
C
T
C
T
G
T
0.440096
0.244209
0.244209
0.238618
0.236621
0.235024
0.234026
0.233826
0.228235
0.228035
IL6single nucleotide variant (38)
insertion (1)
indel (3)
missense variant (4)
synonymous variant (2)
intron variant (27)
nc transcript variant (5)
3 prime UTR variant (6)
500 B downstream variant (10)
2 KB upstream variant (16)
risk-factor (1)
benign (2)
11rs7802308
rs34328912
rs1800796
rs1524107
rs2066992
rs2069845
rs1554606
rs2069840
rs1474347
rs367801961
intron variant
500 B downstream variant
nc transcript variant, intron variant
intron variant, 2 KB upstream variant
intron variant, 2 KB upstream variant
3 prime UTR variant, intron variant
intron variant, 2 KB upstream variant
intron variant, 2 KB upstream variant
intron variant, 2 KB upstream variant
500 B downstream variant
T,A
A,C,T
G,A,C
C,G,T
G,A,C,T
G,A,C,T
T,A,G
C,G
C,A,G
G,A,T
A
C
C
T
T
G
T
G
C
A
0.454473
0.336062
0.313898
0.308307
0.308107
0.252596
0.249401
0.185503
0.168331
0.167133
IRF5single nucleotide variant (59)
indel (6)
intron variant (44)
splice donor variant (1)
5 prime UTR variant (8)
3 prime UTR variant (6)
500 B downstream variant (3)
2 KB upstream variant (4)
risk-factor (2)
benign (1)
/rs3757385
rs3807135
rs752637
rs3757388
rs10954213
rs13242262
rs7808907
rs11770589
rs1874327
rs10954214
5 prime UTR variant
intron variant
intron variant
2 KB upstream variant
3 prime UTR variant
not specified
intron variant
3 prime UTR variant
intron variant
3 prime UTR variant
T,G
T,C
T,A,C
G,A
G,A
A,G,T
T,C
G,A,C,T
A,C,T
C,T
T
T
T
A
G
A
C
A
A
C
0.497604
0.49361
0.491014
0.471845
0.464058
0.463858
0.460264
0.453474
0.400559
0.399561
LDLRsingle nucleotide variant (297)
deletion (1)
indel (41)
missense variant (4)
nonsense (stop gained) (1)
synonymous variant (8)
intron variant (232)
nc transcript variant (41)
3 prime UTR variant (28)
500 B downstream variant (11)
2 KB upstream variant (19)
pathogenic (1)
conflicting-interpretations-of-pathogenicity (1)
uncertain-significance (2)
likely-benign (8)
benign-likely-benign (7)
benign (30)
5rs17248931
rs73017023
rs73017025
rs4804145
rs8102912
rs2738459
rs10422256
rs3180023
rs2738456
rs2738458
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
nc transcript variant, 3 prime UTR variant
intron variant
intron variant
G,A,C
A,T
A,G
G,A
G,A,C
A,C,G,T
G,A,C,T
C,A,G,T
T,A,C
T,C,G
A
T
G
A
A
A
A
C
C
C
0.453674
0.453474
0.453275
0.451278
0.446486
0.425519
0.421526
0.41274
0.40655
0.40635
LPLsingle nucleotide variant (196)
deletion (2)
indel (18)
missense variant (1)
nonsense (stop gained) (1)
synonymous variant (3)
intron variant (161)
3 prime UTR variant (16)
500 B downstream variant (6)
2 KB upstream variant (9)
other (1)
association (2)
likely-benign (1)
benign (21)
3rs253
rs1534649
rs10104051
rs2197089
rs258
rs285
rs314
rs301
rs326
rs56321069
intron variant
intron variant
intron variant
not specified
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
C,G,T
G,A,T
C,T
G,A
G,A,C,T
C,G,T
G,A
T,C
A,G
T,A,G
C
T
T
G
G
C
A
C
G
A
0.487819
0.480032
0.476038
0.457867
0.440695
0.439297
0.390176
0.381989
0.349441
0.346246
LSP1single nucleotide variant (269)
deletion (2)
insertion (1)
indel (18)
missense variant (4)
synonymous variant (2)
intron variant (238)
nc transcript variant (1)
5 prime UTR variant (10)
3 prime UTR variant (2)
500 B downstream variant (8)
2 KB upstream variant (42)
//rs7396311
rs810021
rs28971510
rs1092608
rs542605
rs10734623
rs517101
rs3781961
rs3817197
rs7122680
not specified
not specified
not specified
not specified
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
A,G
G,A,C,T
G,A,C,T
T,A,G
A,C,G,T
C,T
A,G,T
C,G,T
G,A,C
G,A,C,T
G
G
G
T
A
T
A
C
G
C
0.499002
0.482628
0.48143
0.480831
0.479633
0.475639
0.470647
0.449081
0.447484
0.447284
MKNK1single nucleotide variant (200)
deletion (2)
indel (28)
missense variant (2)
synonymous variant (4)
intron variant (206)
nc transcript variant (14)
5 prime UTR variant (35)
3 prime UTR variant (6)
500 B downstream variant (9)
2 KB upstream variant (17)
//rs11211303
rs3766243
rs7543083
rs1258049
rs2181414
rs3766240
rs11211319
rs11211320
rs12022855
rs12136479
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant, 500 B downstream variant
intron variant
intron variant
intron variant
intron variant
T,A,C
A,G,T
G,A
G,A,T
T,A,C
G,A,C,T
T,A,C
T,C,G
C,T
A,C,G,T
C
T
A
A
T
G
T
T
C
A
0.496006
0.49381
0.49361
0.479633
0.472244
0.446086
0.413339
0.413339
0.390375
0.380791
MMP3single nucleotide variant (37)
deletion (1)
indel (2)
missense variant (2)
nonsense (stop gained) (1)
synonymous variant (4)
intron variant (32)
nc transcript variant (3)
3 prime UTR variant (2)
500 B downstream variant (1)
2 KB upstream variant (3)
benign (6)4rs650108
rs538161727
rs639752
rs602128
rs575027
rs520540
rs678815
rs591058
rs679620
rs617819
intron variant
intron variant
nc transcript variant, intron variant
missense variant, synonymous variant
intron variant, 500 B downstream variant
synonymous variant
intron variant
intron variant
missense variant, nonsense (stop gained)
2 KB upstream variant
G,A,T
C,A,T
C,A,T
A,C,G
A,C,G,T
A,G,T
G,A,C,T
T,A,C
T,A,C,G
C,A,G
A
T
C
A
A
A
G
T
T
C
0.442692
0.390974
0.385383
0.38139
0.378994
0.377596
0.35643
0.355232
0.347843
0.347244
MTHFRsingle nucleotide variant (133)
insertion (1)
indel (17)
missense variant (13)
nonsense (stop gained) (1)
synonymous variant (7)
intron variant (134)
nc transcript variant (10)
5 prime UTR variant (10)
3 prime UTR variant (23)
500 B downstream variant (5)
2 KB upstream variant (24)
pathogenic (1)
likely-pathogenic (1)
other (1)
uncertain-significance (1)
benign (5)
139rs10864543
rs4846052
rs6541005
rs3737966
rs1994798
rs7526128
rs6541003
rs4846049
rs11586659
rs2151655
synonymous variant
intron variant
intron variant
3 prime UTR variant, intron variant
intron variant
intron variant
intron variant
3 prime UTR variant, 500 B downstream variant
intron variant
intron variant, 500 B downstream variant
C,G,T
T,A,C
A,T
C,A,G,T
G,A
C,A,G,T
G,A,C
T,A,G
T,A,C,G
G,A,C,T
T
T
A
C
G
C
G
T
T
G
0.498003
0.492612
0.452276
0.441494
0.420727
0.417931
0.408946
0.371605
0.345248
0.294329
MYO6single nucleotide variant (701)
deletion (8)
insertion (3)
indel (104)
missense variant (6)
synonymous variant (3)
intron variant (715)
nc transcript variant (29)
5 prime UTR variant (1)
3 prime UTR variant (28)
500 B downstream variant (2)
2 KB upstream variant (10)
uncertain-significance (2)
likely-benign (15)
benign (14)
/rs276696
rs9360941
rs2748949
rs2842550
rs2647404
rs9360958
rs7742137
rs2842554
rs6920348
rs6903077
intron variant
intron variant
not specified
2 KB upstream variant
intron variant
intron variant
nc transcript variant, 3 prime UTR variant
not specified
intron variant
intron variant
C,T
A,G,T
C,A,G
G,A,T
G,A
A,C,G
C,A,T
C,A,T
T,A,G
A,C,G
C
A
G
A
A
A
C
C
G
G
0.48103
0.476238
0.471645
0.470847
0.470847
0.465455
0.449481
0.422324
0.421526
0.421526
NCR3single nucleotide variant (27)
deletion (1)
indel (3)
missense variant (2)
synonymous variant (4)
intron variant (17)
nc transcript variant (2)
5 prime UTR variant (2)
3 prime UTR variant (5)
500 B downstream variant (5)
2 KB upstream variant (4)
risk-factor (1)/rs1052248
rs2736191
rs2736190
rs3087617
rs986475
rs11575842
rs3896375
rs41268892
rs11575836
rs41268888
nc transcript variant, 3 prime UTR variant, 500 B downstream variant
2 KB upstream variant
2 KB upstream variant
nc transcript variant, 3 prime UTR variant, 500 B downstream variant
3 prime UTR variant, 500 B downstream variant
intron variant
intron variant
intron variant
5 prime UTR variant
intron variant
T,A,C
C,G
T,A,C,G
A,T
A,G,T
G,A
G,A
G,A,C
A,G
G,C
A
G
T
T
G
A
A
A
G
C
0.301717
0.228235
0.174121
0.105232
0.104832
0.0782748
0.0778754
0.0778754
0.0776757
0.0760783
PPARGsingle nucleotide variant (633)
deletion (8)
insertion (1)
indel (65)
missense variant (1)
synonymous variant (1)
intron variant (556)
5 prime UTR variant (4)
3 prime UTR variant (27)
500 B downstream variant (2)
2 KB upstream variant (6)
likely-benign (2)7rs147070788
rs7618026
rs7618046
rs17819328
rs1152003
rs4684104
rs10602803
rs4684854
rs2960420
rs2959269
not specified
not specified
not specified
not specified
not specified
not specified
not specified
not specified
not specified
intron variant
G,A,T
T,C
T,A,C
T,A,G
G,C
A,C,G,T
G,A,T
G,A,C,T
C,G
T,A,C
A
C
C
G
C
A
A
C
G
C
0.495407
0.491414
0.491014
0.489816
0.480232
0.478035
0.460863
0.458866
0.457867
0.455471
SARM1single nucleotide variant (70)
deletion (1)
indel (10)
intron variant (64)
5 prime UTR variant (1)
3 prime UTR variant (17)
500 B downstream variant (1)
2 KB upstream variant (9)
uncertain-significance (1)
benign (11)
/rs2027993
rs967645
rs2239911
rs2239907
rs7212349
rs7212510
rs2239908
rs4795434
rs4795433
rs4794828
intron variant
intron variant
3 prime UTR variant
3 prime UTR variant
2 KB upstream variant
intron variant
3 prime UTR variant
intron variant
intron variant
intron variant
G,C,T
C,A,G,T
G,C,T
T,A,C,G
T,A,C
T,A
G,A,C,T
G,T
C,A,G,T
G,A,T
G
C
G
T
T
T
G
G
C
G
0.469449
0.46845
0.466054
0.451478
0.423522
0.405351
0.394768
0.394369
0.389776
0.389177
TCF20single nucleotide variant (496)
deletion (6)
insertion (4)
indel (67)
missense variant (5)
synonymous variant (5)
intron variant (438)
nc transcript variant (7)
splice donor variant (1)
5 prime UTR variant (4)
3 prime UTR variant (2)
500 B downstream variant (7)
2 KB upstream variant (36)
benign (4)/rs134885
rs134886
rs760648
rs134867
rs134899
rs134891
rs134889
rs134888
rs6002655
rs86669
intron variant
intron variant
intron variant
intron variant
not specified
intron variant
intron variant
intron variant
intron variant
2 KB upstream variant
C,A,G,T
A,C,G,T
G,A,C,T
T,A,C
T,A,G
T,C
A,C,G,T
C,G,T
C,A,G,T
C,G,T
C
A
A
T
T
T
A
C
C
T
0.47484
0.474641
0.467252
0.462061
0.460264
0.454273
0.453275
0.448083
0.441693
0.439497
TCF7L2single nucleotide variant (846)
deletion (11)
insertion (9)
indel (129)
missense variant (4)
frameshift variant (1)
synonymous variant (1)
intron variant (844)
5 prime UTR variant (6)
3 prime UTR variant (8)
500 B downstream variant (5)
2 KB upstream variant (15)
drug-response (1)
risk-factor (2)
benign (2)
14rs720785
rs7918976
rs11196171
rs11196170
rs2296784
rs720784
rs7897438
rs290476
rs10885399
rs61875109
intron variant
not specified
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
intron variant
G,A,C
C,A,G,T
A,C,G
G,A,C,T
T,C
A,C,G,T
C,A,G,T
G,A,C,T
T,A,G
C,A,G,T
G
A
A
G
T
A
A
T
A
A
0.499401
0.498602
0.495607
0.494609
0.494409
0.49401
0.478235
0.477835
0.477436
0.477236
TNFsingle nucleotide variant (9)
indel (4)
synonymous variant (1)
intron variant (7)
3 prime UTR variant (1)
500 B downstream variant (1)
2 KB upstream variant (3)
benign (1)39rs1800610
rs3093662
rs3093664
rs361525
rs3093661
rs673
rs3093665
rs2228088
rs41297589
intron variant
intron variant
intron variant
2 KB upstream variant
intron variant
2 KB upstream variant
3 prime UTR variant
synonymous variant
2 KB upstream variant
G,A
A,G
A,G
G,A
G,A,C
G,A
A,C
G,A,C,T
T,A
A
G
G
A
A
A
C
T
A
0.100439
0.0798722
0.0788738
0.0609026
0.0521166
0.0191693
0.01877
0.0175719
0.0105831
Variant types, molecular consequences, most severe clinical significances, and top 10 genetic variants with highest frequencies were identified in the Variation Viewer database. The number of variants that have already been shown to have specific pharmacologic relevance was retrieved from the PharmGKB database.
Table 4. GWAS-reported variants of selected top-priority candidate genes (DEGs, URs, or both) and their frequencies in the global population.
Table 4. GWAS-reported variants of selected top-priority candidate genes (DEGs, URs, or both) and their frequencies in the global population.
Variant and Risk AlleleMapped Gene/s in GWASGene: Consequence in dbSNPGlobal Frequency in
1000 Genomes
Associated Trait in GWASSNP Identified in GWASGene Identified in Flavonoid Study/-iesNumber of Citations in dbSNP
rs671; AALDH2ALDH2: Missense VariantA = 0.0357HypertensionGCST011141ALDH2: DEG in Paper 1293
rs445925; G
rs445925; A
APOE (also APOC1)
APOE (also APOC1)
APOC1: 2KB Upstream Variant
APOC1: 2KB Upstream Variant
G = 0.8502
A = 0.1498
Atherosclerosis
Atherosclerosis
GCST001231
GCST001231
APOE: UR in Papers 1, 4, and 528
rs10745332; A
rs17030613; A
CAPZA1
CAPZA1
CAPZA1: Intron Variant
CAPZA1: Intron Variant
A = 0.8131
A = 0.7678
Hypertension
Hypertension
GCST002627
GCST007707
CAPZA1: DEG in Papers 2, 3, and 40
2
rs62524579; A
rs12679242; T
rs6418; A
rs1799998; G
CYP11B2 (also LY6E-DT)
CYP11B2
CYP11B2 (also GML)
CYP11B2 (also LY6E-DT)
None
CYP11B2: Intron Variant
CYP11B2: Intron Variant
CYP11B2: 2KB Upstream Variant
A = 0.4794
T = 0.3470
A = 0.6450
G = 0.3472
Hypertension
Hypertension
Hypertension
Hypertension
GCST007707
GCST007707
GCST007707
GCST011141
CYP11B2: UR in Paper 41
0
0
35
rs12941507; CGNA13 (also AMZ2P1)NoneC = 0.0647HypertensionGCST011952; GCST011953GNA13: DEG in Papers 2 and 30
rs4722172; GIL6 (also MTCYBP42)NoneG = 0.0595AtherosclerosisGCST008474; GCST90061371IL6: UR in Papers 4 and 51
rs4728142; AIRF5 (also KCP)NoneA = 0.2945HypertensionGCST006023IRF5: DEG in Paper 5; UR in Papers 4 and 554
rs6511720; T
rs138294113; C
LDLR
LDLR (also SMARCA4)
LDLR: Intron Variant; LDLR-AS1: 2KB Upstream Variant
None
T = 0.0917
C = 0.9095
Atherosclerosis
Atherosclerosis
GCST001231
GCST008474; GCST90061371
LDLR: DEG in Paper 1; UR in Papers 2 and 573
0
rs322; ALPLLPL: Intron VariantA = 0.7079AtherosclerosisGCST008474; GCST90061371LPL: DEG in Paprer 20
rs1973765; T
rs569550; T
rs661348; T
rs4980389; A
LSP1
LSP1
LSP1
LSP1
LSP1: Intron Variant
LSP1: Intron Variant *
LSP1: Intron Variant
LSP1: Intron Variant **
T = 0.5641
T = 0.5765
T = 0.6182
A = 0.4267
Hypertension
Hypertension
Hypertension
Hypertension
GCST007707
GCST007707
GCST007707
GCST007707
LSP1: DEG in Papers 3 and 4; UR in Paper 50
1
7
0
rs139537100; CMKNK1 (also MOB3C)MOB3C: Intron Variant ***
Allele Frequency Aggregator
C = 0.999938HypertensionGCST010477MKNK1: DEG in Paper 4; UR in Paper 40
rs566125; TMMP3MMP3: Intron VariantT = 0.0755AtherosclerosisGCST008474; GCST90061371MMP3: UR in Paper 42
rs17367504; not reportedMTHFRMTHFR: Intron Variant/HypertensionGCST009685MTHFR: DEG in Paper 533
rs3798440; A x rs9350602; Cno mapped genes x MYO6
(SNP x SNP interaction)
rs3798440; MYO6: Intron Variant ****
rs9350602; MYO6: Intron Variant ****
rs3798440; A = not present
rs9350602; C = 0.8972
HypertensionGCST001085MYO6: DEG in Paper 4; UR in Paper 50
0
rs2515920; TNCR3 (also UQCRHP1)NCR3: 2KB Upstream VariantT = 0.0495HypertensionGCST010477NCR3: DEG in Papers 2 and 5; UR in Paper 50
rs17036160; CPPARGPPARG: Intron Variant *****C = 0.9319Arterial stiffnessGCST008403PPARG: UR in Papers 4 and 53
rs704; ASARM1 (also VTN)VTN: Missense VariantA = 0.5551HypertensionGCST90000064SARM1: DEG in Papers 4 and 59
rs17478227; not reportedTCF20TCF20: Intron Variant/Arterial stiffnessGCST007846TCF20: DEG in Papers 1 and 4; UR in Paper 41
rs7903146; TTCF7L2TCF7L2: Intron Variant ******T = 0.2278AtherosclerosisGCST008474; GCST90061371
TCF7L2: DEG in Paper 1; UR in Papers 3 and 4660
rs769177; GTNF (also LTB)Noners769177; G = not presentHypertensionGCST010477TNF: UR in Papers 2, 4 and 57
* genic_upstream_transcript_variant, intron_variant; ** intron_variant, 5_prime_UTR_variant; *** 5_prime_UTR_variant, intron_variant; **** intron_variant, genic_downstream_transcript_variant; ***** genic_upstream_transcript_variant, intron_variant, upstream_transcrip; ****** intron_variantgenic_upstream_transcript_variant.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ruskovska, T.; Postolov, F.; Milenkovic, D. Integrated Analysis of Genomic and Genome-Wide Association Studies Identified Candidate Genes for Nutrigenetic Studies in Flavonoids and Vascular Health: Path to Precision Nutrition for (Poly)phenols. Nutrients 2024, 16, 1362. https://doi.org/10.3390/nu16091362

AMA Style

Ruskovska T, Postolov F, Milenkovic D. Integrated Analysis of Genomic and Genome-Wide Association Studies Identified Candidate Genes for Nutrigenetic Studies in Flavonoids and Vascular Health: Path to Precision Nutrition for (Poly)phenols. Nutrients. 2024; 16(9):1362. https://doi.org/10.3390/nu16091362

Chicago/Turabian Style

Ruskovska, Tatjana, Filip Postolov, and Dragan Milenkovic. 2024. "Integrated Analysis of Genomic and Genome-Wide Association Studies Identified Candidate Genes for Nutrigenetic Studies in Flavonoids and Vascular Health: Path to Precision Nutrition for (Poly)phenols" Nutrients 16, no. 9: 1362. https://doi.org/10.3390/nu16091362

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop