Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jan 24.
Published in final edited form as: Circ Cardiovasc Genet. 2016 Apr 19;9(3):291–313. doi: 10.1161/HCG.0000000000000030

Nutrigenomics, the Microbiome, and Gene-Environment Interactions: New Directions in Cardiovascular Disease Research, Prevention, and Treatment

A Scientific Statement From the American Heart Association

Jane F Ferguson, Hooman Allayee, Robert E Gerszten, Folami Ideraabdullah, Penny M Kris-Etherton, José M Ordovás, Eric B Rimm, Thomas J Wang, Brian J Bennett, on behalf of the American Heart Association Council on Functional Genomics and Translational Biology, Council on Epidemiology and Prevention, and Stroke Council
PMCID: PMC7829062  NIHMSID: NIHMS1564051  PMID: 27095829

Abstract

Cardiometabolic diseases are the leading cause of death worldwide and are strongly linked to both genetic and nutritional factors. The field of nutrigenomics encompasses multiple approaches aimed at understanding the effects of diet on health or disease development, including nutrigenetic studies investigating the relationship between genetic variants and diet in modulating cardiometabolic risk, as well as the effects of dietary components on multiple “omic” measures, including transcriptomics, metabolomics, proteomics, lipidomics, epigenetic modifications, and the microbiome. Here, we describe the current state of the field of nutrigenomics with respect to cardiometabolic disease research and outline a direction for the integration of multiple omics techniques in future nutrigenomic studies aimed at understanding mechanisms and developing new therapeutic options for cardiometabolic disease treatment and prevention.

Keywords: AHA Scientific Statements, diet, metagenomics, microbiota, nutrigenetics, nutrigenomics, nutritional status


Nutrigenomics is a growing field that has received increased attention over the past decade.15 The interpretation and scope of nutrigenomics may vary, but it can be thought to encompass the spectrum of nutritional genomics research, including classic nutrigenetics studies of gene-diet interactions and molecular nutrition, in vitro and in vivo models, human nutrition studies, and the application of large-scale, unbiased studies using high-throughput “omics” techniques to study the effects of nutrients on the body.6 As the Figure shows, diet and the genome may influence cardiometabolic health through a variety of interconnected intermediates, perturbations in which can be measured through omics technologies, including RNA expression (transcriptome), epigenetic modifications (epigenome), metabolites (metabolome), lipids (lipidome), proteins (proteome), and resident microbial communities (microbiome). Although it is clear that both nutrients and genes play a distinct role in determining health, the complex interactions among genes, diet, and downstream networks are not well understood. The application of nutrigenomics approaches to questions of human health and disease is an important component in understanding the complexities of the interplay between basic metabolic processes and external influences in disease processes and has important implications for the development of more targeted strategies in disease prevention and treatment. Analogous to pharmacogenomics, nutrigenomics has the potential to identify genetic predictors of disease-relevant responses to diet, and this potential and its applicability in the context of personalized nutrition have popular appeal. However, nutrigenomics has also been the subject of much hyperbole and has been ascribed much promise, particularly in the arenas of personalized nutrition, functional foods, and nutraceuticals. Unfortunately, the science has not yet fully delivered on this unrealized potential. More than a third of the searchable articles in PubMed on nutrigenomics are review articles, and despite enthusiasm about possible clinical applications, the evidence base remains limited. In this American Heart Association scientific statement, we consider the state of the field of nutrigenomics with respect to cardiometabolic disease, highlight what we know and what is lacking, and propose future directions required to advance the field.

Figure.

Figure.

Potential molecular mechanisms for nutrigenomic/nutrigenetic interactions in cardiovascular disease (CVD) risk. Specific food consumption alters CVD risk through multiple distinct and interrelated mechanisms: (1) differential intestinal metabolism and uptake of nutrients, depending on gut microbiome composition; (2) differential absorption and nutrient binding, depending on individual genotype; (3) modulation of gene expression through specific transcription factor binding; (4) specific effects on methylation and epigenetic modification; and (5) modulation of metabolic signaling through lipids, metabolites, and proteins. SNP indicates single-nucleotide polymorphism.

Nutritional Epidemiology

The study of nutrition and cardiometabolic diseases began in the 1950s with the classic diet-heart hypothesis based on ecological studies linking saturated fat and cardiovascular disease (CVD) mortality.7 These conclusions were supported by small short-term human feeding studies designed to show that replacement of carbohydrate or polyunsaturated fatty acids (PUFAs) with saturated fatty acids (SFAs) increased total cholesterol.8 A growing evidence base that includes millions of participants who have provided detailed dietary and lifestyle data and biological specimens has shaped our understanding of biological pathways for micronutrient and macronutrient metabolism and their effects on disease. Findings from these efforts serve as the foundation for the evolution of contemporary dietary guidelines,9 which are refined as new technologies and methods permit ever more rigorous standards for defining optimal nutritional guidelines. For example, the earlier Dietary Guidelines for Americans (2000)10 recommended a low-fat diet by replacing dietary SFAs with carbohydrate. However, on the basis of hundreds of metabolic intervention studies, cohort studies, and long-term, large, clinical trials of moderate– to high–monounsaturated fatty acid and moderate- to high-PUFA diets, updated guidelines do not emphasize total fat but instead advise replacing foods high in SFAs with food sources of unsaturated fatty acids.

The inclusion of biospecimens in observational and intervention studies enhances the potential to apply nutrigenomics within large-scale human studies, but with the knowledge that nutrition assessment is not without limitations. The limitations of observational cohort studies and of randomized, clinical trials have been discussed at length and are beyond the scope of this review,11 but we highlight important aspects to provide context about why additional insights can be gained through incorporation of measures of the genome, epigenome, metabolome, proteome, microbiome, and other relevant factors.

Observational epidemiological studies have been instrumental in our understanding of nutrition and CVD because many have been ongoing for decades and include hundreds of thousands of participants, as reviewed elsewhere.12,13 Importantly, they capture intake of foods and nutrients as customarily consumed rather than as a supplement or from controlled diets prepared for experimental purposes, but they generally do not have repeated measures of diet over the life course and may be confounded by changes in the food supply, limiting inferences that can be made. The inability to account for other diet, lifestyle, and health characteristics of participants is potentially confounding, but this can be addressed directly through randomization in intervention trials. However, unlike randomized trials of pharmaceutical interventions, trials of nutrients or dietary patterns can give biased or uninformative results when secular trends in the background diet or fortification dilute or mask the effect of the intervention. For example, in the 1990s, many trials were initiated to study folate supplementation on the effects of CVD, but by the mid-1990s, >50 countries worldwide initiated folate fortification, adding 200 to 400 μg/d to the average diet, which ultimately may have been enough to increase folate status in trial participants at highest risk for CVD. Trials are also limited, especially those with clinical end points, because such studies typically test only a single dose, are limited in duration by cost and adherence, and create an artificial environment to test a nutrient or food pattern. Most midlength dietary pattern trials of 1 to 4 years must either provide all foods for consumption (which is simple for the participant but not feasible as a long-term solution) or use weekly or monthly training sessions with dietitians, health coaches, and wellness programs, all of which may be cost-prohibitive. Even trials with the most intensive support systems can experience poor adherence as participants grow weary of the prescribed dietary pattern and either revert to previous dietary habits or adhere to the experimental diets only before clinical examination assessments.

Studies of nutrition and cardiometabolic disease have taught us much over the past half-century, but most important, they have taught us that conclusions are most sound when supported by short-term dietary intervention studies to understand plausible biological mechanisms and long-term observational epidemiological studies to understand the impact of decades of exposure. In some unique situations in which results from both study types are consistent, large, long-term trials are initiated. The most conclusive trials for cardiometabolic disease have been based on dietary pattern such as the Dietary Approaches to Stop Hypertension (DASH) dietary patterns trials,14 the Diabetes Prevention Program,15 the Dietary Intervention Randomized Controlled Trial for weight loss,16 and the Lyon Diet Heart study,17 as well as the Preventión con Dieta Mediterránea (PREDIMED) trial for CVD reduction.18 In PREDIMED, the reduction in CVD among individuals randomized to a Mediterranean diet plus extravirgin olive oil or mixed nuts corroborates decades-long cohort studies and short-term trials reporting benefits of the Mediterranean diet and its components, including olive oil, nuts, fruits and vegetables, whole grains, and wine.19 These highly successful initiatives have greatly enhanced our knowledge of diet at a population level, and the incorporation of omics into future nutrition studies will help us understand the mechanisms of action underlying a healthy diet.

Nutritional Assessment

The importance of using appropriate nutrition assessment tools should not be underestimated, and a combination of diet and biochemical measures may be required to provide a comprehensive assessment of short-term and long-term exposure. The best tools provide an integrated measure of exposure because, for nutrition (except for mega-dose supplements or accidental contamination or poisoning), variation in exposure is modest and complex metabolic systems buffer extreme exposures. The major dietary tools that have been incorporated into large observational studies include 24-hour recalls, diet records, and food frequency questionnaires. Each of these has strengths and weaknesses, but each also provides insights and a foundation of research methods applicable to studies.

The 24-hour recall and single-day diet record are the best methods to assess a 1-day intake. They allow direct quantification of specific foods and thus limit error resulting from estimation of intake. The 24-hour recall may be more susceptible to error because of memory recall or modifications in responses to be more socially acceptable to the interviewer, but careful scripting of a well-trained interviewer can minimize this problem, and online methods that are computer assisted with multistep quality control have reduced errors.20,21 Diet records, in which the participant weighs and records everything consumed in a day as it is eaten, generally are considered the gold standard for assessing short-term intake22 because they do not rely on memory or estimation of portion size, but with multiday assessments, they can be burdensome on study subjects, thus leading to underassessment or changes in diet to reduce the burden of recording.

A single 24-hour recall or diet record is not a representative measure of average intake because of the great fluctuations in nutrient estimates caused by consumption of a single nutrient-dense food, for example, carrots and liver, fish, and berries and tea, which could lead to substantial misclassification of average intake of vitamin A, omega-3 fatty acids, or polyphenols, respectively. Recalls or records are frequently spread out over several months or even a year to better estimate fluctuations in intake. For large studies, multiple assessments can be burdensome or cost-prohibitive, which has led many studies to shift these measures to computerized protocols.2325 Alternatively, the semiquantitative food frequency questionnaire (SFFQ) can be used as the primary method for estimating nutrient intake over a longer period of time. Participants are provided with a list of 100 to 150 commonly consumed foods and portion sizes and are usually asked to estimate intake over a 3- to 12-month period, individually incorporating fluctuations in weekly or seasonal patterns into their grid of fixed response categories. The SFFQ can be developed to be culturally specific, readily computerized and inexpensive, anonymizable, and less burdensome to study subjects than other dietary methods. The quality of the assessment tool is determined by the comprehensiveness of the food list provided, the clarity of provided instructions, and the ability of subjects to recall and properly estimate average intake of listed foods. The SFFQ is best used to rank individuals in a population by their intake of a nutrient or food item because of systematic underestimation or overestimation. When SFFQs are validated against gold standards such as multiple days of diet records or biochemical markers in blood, adipose tissue, or hair, some foods and nutrients are better estimated than others. The SFFQ has been well validated for assessing micronutrients, fatty acids, and many other important nutritional determinants of disease. However, the SFFQ is not good at estimating sodium intake because of great fluctuations in similar foods (homemade food versus highly processed store-bought version) or trace metals such as selenium that are dependent on soil composition. This limitation in the granularity of dietary studies highlights the need for additional methods of characterizing nutrients that are not well characterized by current dietary tools. As discussed in the following sections, there is great promise in omics profiling for better assessing risk of CVD and related traits and advancing our understanding of how genetic variation affects diet response and potentially the underlying mechanisms, thus improving our ability to design better targeted interventions. New omics platforms integrated with dietary measures can better quantify biological systems by incorporating not only a single circulating vitamin but also a family of downstream metabolites and other metabolically relevant nutritional factors. Collectively, advances in these areas will bring us closer to individualized lifestyle and pharmacotherapy interventions that are more effective for preventing and treating CVD.

Genetic Variation

Studies that have focused on identifying the genes and genetic variants associated with the different types of intermediate and CVD phenotypes in humans have been more consistent and successful than studies undertaken to identify genetic variants associated with other complex traits. What may have contributed to this greater success is that the cardiovascular phenotypes studied, both intermediate (plasma lipid concentrations, blood pressure, etc) and CVD end points (myocardial infarction [MI], stroke, and other CVD), have a standardized clinical definition and are easier to measure than other complex phenotypes.

A 2013 American Heart Association statement26 supports the notion that for rare and familial forms of CVD, we are identifying single-gene mutations that impart relatively large effects on individual phenotype. For these cases, progress has led to several clinically useful diagnostic tests. However, the prevalence of monogenic disorders typically accounts for only a small proportion of the total CVD observed in the population. There has been less progress in developing genetic testing for complex CVD because individual common variants usually have only a modest impact on risk. Exome sequencing approaches may begin to address this; rare coding mutations in LDLR and APOA5 were found to affect MI risk in the general population through modulation of low-density lipoprotein (LDL) and triglyceride metabolism.27,28 However, the study of the genomics of complex CVD is further challenged by the influence of environmental variables, phenotypic heterogeneity, and pathogenic complexity.29

Gene-Diet Interactions in Determining CVD Risk in Humans

Multiple gene-environment interactions (GxEs) exist in determining risk of CVD in humans, including factors such as smoking, physical activity, drugs, diet, and social context. An outstanding initiative to unravel this complexity is the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium, which was formed to facilitate meta-analyses of genome-wide association studies (GWAS) and replication opportunities among multiple large, population-based cohort studies.30 While acknowledging the importance of other environmental factors, we focus here on gene-diet interactions because diet is one of the most important exogenous factors that humans are exposed to every day.31 Gene-diet interactions in CVD have been analyzed for many years, with many reports demonstrating that those interactions have an impact on determining both intermediate and final CVD phenotypes,3267 as cataloged in detail.68 Some of these genetic variants have been highlighted through GWAS. For example, variation in the TCF7L2 gene, previously implicated in type 2 diabetes mellitus,69 was shown to interact with diet through intervention with the Mediterranean diet and played a role in determining stroke risk.34 The effects of other loci such as 9p21 and FTO, which are robustly associated with CVD and obesity, respectively, have also been reported to modulate CVD risk through interaction with diet.44,70 Gene-diet relationships are complex, with multiple dietary components potentially interacting with genotype to determine CVD risk. For example, elevated total homocysteine, a marker of CVD risk,71 is modulated by dietary folate and alcohol consumption72,73 and interaction with MTHFR genotype.74 Thus, a better understanding of these gene-nutrient interactions with information on both folate and alcohol intake is essential to understand why total homocysteine levels are elevated (and the underlying reason why CVD risk is elevated).

A comprehensive review of cardiometabolic GxEs from 386 publications,68 including blood lipids, glycemic traits, obesity anthropometrics, vascular measures, inflammation, and metabolic syndrome, allowed the following conclusions. First, the GxE single-nucleotide polymorphisms (SNPs) showed little overlap with variants identified by a main-effect GWAS, indicating the importance of environmental interactions with genetic factors on cardiometabolic traits. Second, these GxE SNPs were enriched in adaptation to climatic and geographical features, with implications for energy homeostasis and response to physical activity. Third, a comparison with 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. Collectively, these studies demonstrate that SNPs supporting cardiometabolic GxEs often exhibit transcriptional effects or are under positive selection. However, not all SNPs can be assigned potential functional or regulatory roles because data are often lacking in specific cell types.

One limitation in the study of gene-diet interactions is that most of the studies are observational, so they provide a lower level of evidence than experimental studies. In addition, those experimental studies have often had an inadequate sample size. Therefore, it is necessary to increase the level of evidence in support of the gene-diet interactions in CVD. Furthermore, relatively few studies have focused on CVD end points, an area of investigation where future efforts should be focused.34,57

Clinical Application of the Gene-Diet Interactions

Research into gene-diet interactions is crucial to obtain information that will allow us to undertake clinical applications of early genetic diagnosis. If a genetic variant is associated with CVD or an intermediate phenotype (eg, greater risk of hypertension, dyslipidemias, or diabetes mellitus) and it is known that a certain diet can counteract that genetic risk, then one can reason that disease risk could be reduced through prescription of a personalized diet. To date, most studies have focused on primary prevention, but there is also great interest in discovering gene-diet interactions in secondary CVD prevention to provide appropriate dietary recommendations for individuals who have already had a nonfatal CVD event. As next-generation sequencing technologies continue to improve, they will contribute to advancing gene-diet interaction studies in particular and GxE studies in general. The results of these efforts can then provide us with new knowledge to be applied in achieving more efficacious prevention of CVD.

Nutrient Effects on RNA Expression

We expect that many of the biological effects of diet on cardiovascular risk and outcomes are mediated by changes in gene expression, whether as a result of genetic variation or environmental influences. In this regard, the use of global transcriptional profiling is a powerful tool in nutrigenomic studies. For example, measuring dynamic changes in gene expression before and after a short-term dietary challenge can highlight novel nutrient-responsive genes, and profiling gene expression after a diet intervention can identify genes responsive to the intervention. The overall goal of these studies is to identify novel candidates for cardiometabolic risk.

Candidate Gene Studies

Some of the best evidence for direct effects of specific nutrients on gene expression comes from studies of orphan nuclear receptors.75 Of these nutrient-responsive transcription factors, the peroxisome proliferator-activated receptor (PPAR) family has been particularly well studied.76,77 As lipid-responsive genes, with effects on lipoprotein metabolisms and inflammation, the PPARs have been recognized and extensively studied for their relevance to cardiometabolic disease. SNPs in PPAR genes have been implicated in cardiometabolic risk in gene-diet interaction studies.78,79 The PPAR family members α, δ, and γ are responsive to specific dietary fatty acids (PUFAs, monounsaturated fatty acids, and SFAs)76,80 and fatty acid–derived eicosanoids, as well as to physiological states induced by nutrient status (eg, fasting). PPARs are also responsive to a number of synthetic ligands, including fibrates and thiazolidinediones, and thus are attractive drug targets. PPAR agonism is responsible for activating and inhibiting a number of downstream genes, which remain to be fully characterized,8183 and may act as a key regulator of hepatic fatty acid metabolism, response to fasting, and inflammatory responses.8486 Although PPARα agonism through fenofibrate does not appear to have effects on systemic inflammatory responses in healthy humans,87 agonism of PPARγ and other targets by n-3 PUFA supplementation has an effect on systemic inflammatory response.88 PPARγ is crucial for adipose tissue development and adipocyte differentiation and potentially increasing browning of adipose tissue.89 Thus, PPARγ agonism can increase the lipid storage capacity of adipose tissue while increasing fatty acid oxidation capacity, leading to improvements in insulin sensitivity and cardiometabolic risk. Thiazolidinediones have been used extensively as insulin-sensitizing agents through PPARγ agonism to treat type 2 diabetes mellitus, although concerns have arisen about the safety of this class of drugs, with potential for increased MI and exacerbation of heart failure.90,91 Given the importance of diet-derived ligands in PPAR agonism, differences in nutrient status may be important in mediating drug effects, with potential differences in the effects of PPAR agonism, through physiological versus pharmacological ligands. These genes thus remain interesting targets for further nutrigenomics study.

In addition to PPARs, there are other clear examples of gene-diet interaction, and similarly, several additional nuclear receptors have been shown to be important nutrient regulators, including the bile acid–responsive farnesoid X receptor (FXR or NR1H4), which may influence fatty acid and glucose homeostasis92; the retinoic acid–responsive retinoid X receptor (RXR)93; and the oxysterol-responsive liver X receptor (LXRα/β, NR1H3/2), which may alter atherosclerosis and diabetes mellitus risk.94 The nuclear receptor superfamily members form both homodimers and heterodimers and likely act cooperatively to regulate nutrient metabolism and cardiometabolic risk.95 Each of these highly specific receptors has diet-derived ligands; however, how seemingly divergent signals from each of these receptors respond to a meal remains unclear.

Unbiased Transcriptomics

Although studying candidate gene expression has utility in confirming hypotheses and defining mechanisms, large-scale, unbiased techniques are required to further advance our knowledge. Unbiased approaches to assessing transcriptional changes in response to dietary intervention have used microarray and RNASeq approaches. These technologies and their application to CVD have been reviewed elsewhere.96,97 We focus here on how these technologies can be applied to the field of nutrigenomics.

In understanding the interaction between nutritional components and gene expression changes, selecting the relevant cell type is of great importance. The majority of studies in humans have focused on blood-derived samples, for example, whole blood or peripheral blood mononuclear cells (PBMCs), or on adipose tissue, with a smaller number in skeletal muscle. These represent relatively accessible and relevant tissues and give important, albeit incomplete, insight, which leaves other disease-relevant tissues to be studied in animal models. Transcriptomic studies of nutrition have to date mainly focused broadly on dietary patterns such as Mediterranean diets, on macronutrients, or on specific dietary components, primarily fish oil–derived fatty acids and some phytonutrients. These studies are further categorized into those implementing a dietary intervention versus those examining habitual dietary status. Associations between PBMC gene expression patterns and habitual consumption of a prudent versus Western diet were reported in healthy subjects participating in the PREDIMED study (n=30).98 A Mediterranean diet intervention was also found to alter PBMC expression of genes in cardiovascular pathways.99 Changes in gene expression in blood and adipose tissue from the same individuals (n=3) were reported after an intervention to alter macronutrient content,100 with divergent transcriptomic responses in the 2 tissues. Adipose tissue gene expression changes were characterized in response to both overfeeding in healthy subjects (n=6) and caloric restriction in obese subjects (n=18),101 with >100 genes identified as responding to caloric intake. Several studies have profiled PBMC expression before and after intervention with n-3 PUFA,102104 and the adipose tissue transcriptome by n-3 PUFA status has also been studied.105 Furthermore, the response to an 8-week high-fat diet intervention of SFAs or monounsaturated fatty acids was characterized in adipose tissue from overweight subjects (n=20).106 Other studies have included additional short-term interventions, which increase the power to detect dynamic metabolically relevant changes. For example, skeletal muscle was profiled in obese subjects with insulin resistance (n=15) at 4 time points after supplementation with n-3 PUFA or n-3 PUFA with fish gelatin, in both cases before and after a euglycemic-hyperinsulinemic clamp.107 This approach allowed identification of genes that were robustly altered by the clamp and by supplementation itself. This group has also used response to intervention as a tool to understand genetic differences in transcriptomic responses. High and low responders to n-3 PUFA supplementation as defined by change in triglycerides were identified and shown to differ in their transcriptomic responses.108

Despite increasing availability of transcriptomic data, many limitations still apply. We expect the transcriptome to vary, depending on time of day; cell type composition; age, race, and sex of the individual; and the health or disease status and habitual diet. Given these many confounders, extracting biologically meaningful data is a challenge. In general, the majority of transcriptomic studies will find statistically significant changes in the expression of some genes or transcripts, even in relatively small numbers, but it is difficult to decipher which of these are truly meaningful and functional. A key constraint is the need to collapse data into intelligible results. To reduce the long lists of genes identified in transcriptomics experiments, common approaches include some sort of pathway analysis and functional clustering. Although this approach is certainly valid, it is biased toward genes with known functions, meaning that many interesting but unstudied candidates will likely be missed among the vast amount of available data. The use of unbiased methods to construct interaction networks may overcome these limitations.109 The first step to understanding the health implications of diet-related gene expression patterns on a global scale is to characterize the transcriptome of multiple tissues under different diet and temporal conditions. However, at present, the field is still in a developmental stage in which we are generating data but struggling to obtain meaningful insight. Many studies remain underpowered with small numbers of subjects and many confounders. With decreasing costs, analyzing the transcriptomes of larger numbers of individuals over multiple time points is now feasible. Furthermore, public data repositories and large-scale collaborations are allowing the meta-analysis of multiple data sets,81 increasing the value of the information that can be extracted from existing data. Thus, the next step for the field will be broad integration of available transcriptomic information with in-depth functional and mechanistic interrogation of novel candidates, as well as increased use of repeated-measures and acute “evoked phenotype” study design interventions. Beyond this, integration of transcriptomic data with other information, both in the reverse direction of genomic and expression quantitative trait locus analysis and in the forward direction toward proteins and metabolites, is crucial to obtain a cohesive view of the metabolic response to nutrients.

Noncoding RNA

As next-generation sequencing has become increasingly affordable and feasible, our knowledge of noncoding RNA (ncRNA) has improved.110113 A large proportion of the transcriptome is not translated into protein yet is functional, with both long ncRNA and small RNAs exhibiting specific functionality.114 Of particular relevance to nutrigenomics, microRNAs and other classes of small RNA have received particular attention for their possible ability to signal not just across species but across kingdoms. Plant-derived dietary microRNAs may potentially cross the intestinal barrier and not only remain intact as they enter the bloodstream but also retain functionality and regulate host gene expression.115,116 Although this remains highly controversial,117119 host-derived small RNAs are known to regulate many processes of relevance to cardiometabolic risk120 and to be altered in response to dietary intervention and specific nutrients.121,122 For example, several microRNAs were reported to be changed in the PBMC transcriptome after 1 year of supplementation with resveratrol-containing grapeseed extract in subjects with type 2 diabetes mellitus.123 Thus, the potential functional role of diet acting directly or indirectly through small RNA is intriguing.

Epigenetics

As discussed above, dietary components can directly modify gene expression. However, many of the effects of diet and nutritional status on gene expression and regulation are mediated through epigenetic mechanisms. Dysregulation of epigenetic states plays a major role in disease, including CVD.96,124 Although our awareness of this role is expanding rapidly as we learn more about the nature of the endogenous epigenetic regulatory machinery and the ways in which it can be perturbed, much remains to be elucidated. Importantly, the reversible nature of epigenetic mechanisms provides a unique opportunity for the development of treatment measures targeting the perturbed marks or pathways. Although the list of epigenetic mechanisms and their functions continues to grow, those with roles in disease include DNA methylation, the addition of a methyl group (CH3) to the cytosine of a CpG dinucleotide; DNA hydroxymethylation, the presence of a hydroxymethyl group (CH2OH) at the cytosine of a CpG dinucleotide; and posttranslational modifications to histone tails of nucleosomes such as methylation, acetylation, ubiquitination, phosphorylation, sumoylation, and biotinylation. However, our understanding of how these epigenetic mechanisms regulate gene expression and how disruption leads to disease remains incomplete. Major functions include recruitment and accessibility of transcriptional machinery to target genes and in stability and cellular localization of RNA and proteins. Furthermore, studies show an additional role for epigenetic mechanisms in determining genomic stability.125,126 Although there are seemingly distinct roles for each of these mechanisms acting individually· we more often observe redundant and cooperative functions for which several mechanisms must work together for normal cellular function and development.127,128 Likewise, multiple mechanisms are simultaneously perturbed in disease states.

Role of Nutrition in Epigenetic Perturbation

There is some evidence relating diet, including micronutrients and macronutrients and diet composition· to epigenetic changes that may alter cardiometabolic risk. A classic example of a seemingly direct role of nutrients in determining epigenetic states is the role of “methyl-donor” nutrients such as choline, methionine, betaine, folate, vitamin B12, and zinc. These dietary components promote the formation of S-adenosyl methionine, a metabolite of the 1-carbon pathway,129131 which contributes the methyl group required for methylation of DNA and histones. Bioavailability of these nutrients is a limiting factor in the proper establishment and maintenance of these epigenetic marks. Indeed, in vivo studies show that deficiency in or oversupplementation of these nutrients can result in altered epigenetic states, including changes in DNA methylation, histone modifications, and ncRNA.132134 In addition, acetyl-CoA, a byproduct of fatty acid metabolism, acts as a cofactor for histone acetylation carried out by members of the histone acetyltransferase gene family.135,136 High-fat diet-induced obesity in mice leads to distinct changes in promoter methylation, potentially linked to the downstream health consequences associated with obesity.137 Similarly, research in nonhuman primates has highlighted a link between diet-induced obesity and epigenetic changes.138 Such diets have long been linked to CVD, but the finding that they act via epigenetic mechanisms is intriguing and represents an area for future investigation. Several studies have found evidence of epigenetic changes that alter cardiometabolic disease risk,139141 including defects in endothelial cell function, abnormal blood flow, inflammation, and plaque formation.142147 Identification and characterization of common epigenetic outcomes linked to disease manifestations and their interaction with nutrient status may allow the development of specific treatment and intervention measures.148151

Interestingly, the effect of limited or oversaturated nutrient bioavailability on epigenetic states is complex and not necessarily direct. For example, deficiency of methyl donors results in hypermethylation at some loci and hypomethylation at others. Furthermore, the contribution of oversupplementation to epigenetic perturbation remains largely unexplained. To fully understand the mechanism(s) of nutrient involvement in epigenetic states, we must take into account the complexity of metabolic pathways that involve nutrients and their metabolites, which often act simultaneously in multiple divergent pathways. Different metabolic pathways playing roles in different cellular functions often use common genetic pathways (eg, metabolic enzymes). These effects highlight the importance of distinguishing the difference between epigenetic responses that result from defective gene expression regulation in the cell and lead to disease phenotypes and epigenetic changes that reflect an adaptive response to insult and are not directly causal. Controlled studies are thus required to elucidate the directionality of effects and to distinguish between causal and reactive changes. Finally, we must also consider the demonstrated role of other environmental influences in epigenetic perturbation and the potential for aggregate effects involving nutrition.152 These include well-studied epigenetic effects caused by environmental toxicants and less elucidated environmental stimuli such as stress and general socioeconomic factors, which have been shown to influence the risk of CVD.153 Future research will clarify the overlapping roles that nutrition and other environmental factors play in modulating CVD risk.

Timing of Epigenetic Perturbation: Developmental, Postnatal, and Transgenerational Origins

Maternal nutritional status has important and direct effects on the offspring in utero. Paternal diet has also been shown to play an important role in epigenetic status of the offspring, although specific links to CVD risk have not yet been determined.154 Furthermore, grandmaternal nutritional status may also affect subsequent generations of offspring through epigenetic modification of fetal germ cells.155 Epigenetic programs are especially susceptible to change during fetal development because patterns of epigenetic states required for future development are established during early embryogenesis.152 Although some epigenetic states change throughout fetal development and during postnatal development and aging, epigenetic states that are programmed during embryonic development set the stage for future epigenetic outcomes.156,157 Maternal dietary deficiency altering epigenetic programming can result in severe fetal and infant outcomes such as neural tube defects caused by a folic acid deficiency.158 However, maternal diet may also affect long-term risk of chronic disease development. Notable examples demonstrating maternal undernutrition and disease-relevant epigenetic changes in the resulting offspring include the Dutch Hunger Winter studies.159 People conceived or born in the Netherlands during the World War II Dutch Hunger Winter (1944–1945) were at increased risk of diseases, including diabetes mellitus and CVD.160 These diseases were linked to epigenetic changes, supporting the notion that diet-induced epigenetic defects occurring during fetal development or even before conception161 can alter cardiometabolic disease risk and progression throughout life. Epigenetic perturbation occurring before or during gestation coincides with developmental origins of CVD.162 Investigators cleverly took advantage of seasonal nutritional changes among pregnant women in The Gambia to demonstrate an association between naturally occurring maternal nutrient metabolite levels and DNA methylation patterns.163

Postnatal diet may compensate for some effects occurring in utero. For example, post-weaning supplementation with folate and selenium after a maternal high-fat diet leads to alterations in hepatic methylation.164 These and other studies demonstrate that postnatal nutrition also plays a critical role in maintaining epigenetic programs and that perturbation of these systems after birth contributes to disease; postnatal supplementation may be used to alleviate these incurred effects.

Because of the development of both somatic and germline epigenetic programs during embryonic development, diet-induced epigenetic perturbations occurring in utero are also implicated in the transmission of disease to successive generations in a multigenerational or transgenerational manner.165 Although multigenerational transmission of disease is not a novel concept, it has previously been studied mostly with respect to genetic changes in DNA sequence, leaving the role of epigenetic causes largely unexplored. Thus, this major breakthrough unveiled the involvement of nongenetic changes that by definition occur in the absence of changes in DNA sequence, are susceptible to environmental/exogenous perturbation, may increase in severity with age, may be transmitted to successive generations, and may play a significant role in disease. Animal models make up the majority of the body of work demonstrating these changes, but some human studies also support these findings.124 Little is known about the potential for repeated insult and how that would affect heritable outcomes, and even less is understood about the contributions of effects that are transmitted paternally. It remains difficult to determine causality among nutrition, epigenetic changes, and disease in human studies, given the presence of multiple confounding factors. Thus, animal and cell-based models will play critical roles in identifying the causal mechanisms and characterizing the pathways involved.

Epigenetic Biomarkers

Epigenetic biomarkers allow the determination of risk of CVD and the diagnostic and treatment purposes. However, accurately characterizing epigenetic biomarkers for any disease requires careful measurements and an acknowledgement of the limitations of their use. One major challenge in human studies is the limited sources of biological materials. Most in vivo human studies are limited to blood or plasma collection, the latter of which is lacking cells and thus is not an option for accurately measuring epigenetic states. Because epigenetic states vary between cell types, without cell sorting, mixed cell types present in the blood may create difficulties in accurately measuring epigenetic changes between individuals. To fill in the gaps in disease pathogenesis in humans, human cell lines or animal models are often substituted to identify tissue-relevant biomarkers of change. In addition, because epigenetic states often retain some level of plasticity within a lineage of cells, studies must measure states along a time course sufficient for determining whether a change is stable or transient, a distinction that is necessary for determining the therapeutic use of the biomarker. Furthermore, it is important to note the distinction between epigenetic biomarkers that are causal to the disease mechanism, epigenetic biomarkers that are byproducts of the disease state itself and thus not causal, and epigenetic biomarkers that are initially byproducts of the disease state but contribute to later stage effects as the disease progresses or even to other unrelated diseases/symptoms. Distinguishing these classes of biomarkers is critical to their accurate use in disease diagnoses and prevention or treatment of CVD. Many of the epigenetic biomarkers currently identified are epigenetic changes in genes known to play a role in the initiation or progression of CVD, many of which were already identified as mRNA or protein biomarkers.120,166,167 Alternatively, other biomarkers that are indicative of epigenetic changes may be used as a proxy for changed epigenetic states.168

Therapeutics

Identification of epigenetic perturbations that contribute to CVD provides the potential for gene therapeutic approaches that take advantage of the potentially reversible nature of epigenetic mechanisms. Current studies are investigating the use of therapeutic agents that target histone deacetylases and ncRNA via chemical inhibitors and molecular interference.169171 However, given the wide-ranging effects of epigenetic modifications, the potential for off-target effects remains an important concern. Understanding the links between these epigenetic mechanisms and nutrition may allow more effective dietary interventions to address the development and progression of CVD. Although there is excitement about the potential for clinical applications of therapeutic epigenetic remodeling, much research remains to be done before this could realistically be implemented.172

Metabolomics, Lipidomics, and Proteomics

Metabolomics

Because metabolites and proteins are downstream of genetic variation and transcriptional changes, they are attractive “proximal” reporters of a given metabolic phenotype, particularly because they directly integrate the breakdown products of our dietary intake. Metabolomics offers significant potential to better understand how different dietary patterns or specific foods affect metabolic pathways.173 Specifically, metabolomics provides important information on perturbations in metabolic pathways that affect disease onset and treatment. In addition, metabolomics may soon be used to identify biomarkers of dietary intake in a way that overcomes some of the shortcomings of diet assessment tools.173 Although there is significant circadian and day-to-day variation in metabolite profiles, in a reproducibility study from a single blood collection, 90% of 257 metabolites were at least moderately reproducible 1 year later,174 much better than for nutrients assessed from 2 single days of dietary assessment repeated 1 year apart.22 Importantly, metabolomics can be used to identify patterns of metabolic profiles among individuals that reflect differences in dietary intake or individual metabolic activity that could explain variation in diet response. Aggregating individuals by differences in their metabolome provides a strategy for evaluating the effects of diet. Metabolites span a variety of compound classes, with significant differences in size and polarity across a wide range of concentrations. As a consequence, no single analytical method is able to accommodate the chemical diversity of the entire metabolome. Although various methodologies have been used, 2 core technologies have prevailed as the workhorses of metabolite profiling: nuclear magnetic resonance spectroscopy and mass spectrometry, the latter coupled to an array of separation techniques, including gas and liquid chromatography. However, with an estimated 5000 currently detectable human serum metabolites (a number likely to increase with advancement in technology), a comprehensive map of the entire metabolome remains an unattained goal. However, by using a combined analytical approach, one can build a more inclusive picture of the metabolome by overcoming the limitations of individual techniques.

With improved throughput, an increasing number of studies have begun to apply these technologies to large population-based studies to identify novel predictors of cardiometabolic disease. For instance, studies in epidemiological cohorts have demonstrated that selected amino acids and amino acid derivatives are elevated more than a decade before the onset of type 2 diabetes mellitus.175,176 Some of these metabolites such as 2-aminoadipic acid may directly modulate glucose homeostasis. Furthermore, the experimental demonstration that 2-aminoadipic acid is influenced by both diet and genetic background highlights the potential role for nutrigenomic interventions.177 It is anticipated that as sufficiently large human data sets are acquired, investigators will begin to be able to parse the relative contributions of diet and genes in this pathway and others in disease development.

Given the possibility of nutritional manipulation of circulating metabolites, it is important to understand which ones have direct causal roles in disease protection or susceptibility rather than simply serving as markers. Mendelian randomization studies offer one approach to investigating causality. Given the independent assortment of alleles, individuals are randomized to carrying the major or minor allele at polymorphic sites. Metabolite concentrations can have a strong heritable component, and it has been shown that common genetic variants explain part of this heritability.178 If a SNP is associated with levels of a metabolite, then examining the association of this SNP with a downstream clinical trait may be informative in terms of the causal role of the metabolite. For instance, it is known that elevated LDL cholesterol is associated with increased risk of MI. LDL is also a heritable trait with a number of rare and common genetic determinants. Variants that determine LDL would also be expected to relate to MI risk (which they do) if LDL is causal (which it is). Studies testing the causal links of novel metabolites with cardiometabolic diseases are underway but require very large sample sizes given the complex genetic determination of such traits.

Lipidomics

Lipidomics, closely linked to metabolomics, uses mass spectrometry–based profiling to evaluate the comprehensive lipid profile in a sample179,180 and is increasingly being applied to cardiometabolic disease studies.181 Circulating lipids can have complex structures, with multiple different classes, and specific composition. For example, triacylglycerols are composed of a glycerol backbone attached to 3 acyl chains, all of which can vary in length and saturation, making measurement of specific species challenging.182 Despite the importance of circulating lipids and lipoproteins in cardiometabolic disease, well-known lipid species such as LDL, high-density lipoprotein (HDL), and total cholesterol explain only a small portion of atherogenic risk. The relationship among intake of dietary fat, circulating lipids, and cardiometabolic outcomes is influenced not only by genetically determined host metabolism but also by metabolic action of intestinal microbiota and interaction with other dietary and metabolic components. We have a good understanding of the scientific evidence informing specific dietary guidelines on SFA, monounsaturated fatty acid, and PUFA intake,9,183 but questions remain about the functions and interactions of lipid subspecies and metabolites. Thus, whereas triacylglycerols with certain SFAs may be pathogenic in cardiometabolic disease, triacylglycerols with long-chain PUFAs are thought to be protective.182 However, chain length and saturation may also affect the bioavailability of other nutrients, with SFAs generating smaller micelles with greater bioaccessibility.184 Profiling the many structurally similar yet biologically distinct lipid subspecies is challenging but is becoming possible through improved lipidomics approaches.185,186 These newly developed methods facilitate the application of functional lipidomics to fully profile and understand the specific role of each unique lipid subspecies.187 Application of lipidomics to human plasma has direct utility, with potential to identify novel and predictive biomarkers of disease,188191 including coronary disease progression,192,193 as well as biomarkers of responsiveness to therapeutic intervention.194,195 As many specific lipid species are being generated in vivo from dietary precursors, lipidomics is a promising avenue for understanding the complex relationship between dietary lipids and cardiometabolic risk and identifying lipid biomarkers of dietary intake.

Proteomics

Given the complexity of specific dimensional configuration and posttranslational modifications of proteins, accurately profiling the proteome is particularly challenging. Several methods exist for proteomic characterization, including gel based,196 liquid chromatography–coupled mass spectrometry based,197 and aptamer based,198 with broad applicability to a wide variety of sample types, disease states, and desired biomarkers. However, the complexity of data obtained from proteomic studies makes translation of putative biomarkers into clinically useful prognostic tools difficult. As has been reviewed extensively elsewhere, proteomics has been successfully applied as a discovery tool to CVD in plasma, urine, and tissue199202 in the context of both animal models aimed at understanding cardiac function and development203 and human biomarkers of atherosclerosis,204 dyslipidemia,205 and cardiometabolic disease.206 Proteomics has also been applied in the context of nutritional studies to identify biomarkers of diet and nutritional status.207209 Although the proteome may be the most complex entity to assay accurately, the ultimate application is similar to other omics. As with metabolomics and lipidomics, increased application of proteomics methodologies will be required to fully understand the relationships between dietary and genetic factors in determining the unique cell, tissue, and circulating profile that results in health or disease. With some reduction in costs and increases in the resolution of instrumentation and methods allowing the increased use of proteomics, perhaps the greatest hurdle remaining will be the informatic integration and analysis of vast amounts of data into intelligible results.

Limitations of Disease Biomarkers

The measurement of intermediate biomarkers, including metabolites, proteins, and lipids, is a powerful approach both to understand intermediate signaling processes and to estimate cardiovascular risk in the absence of clinical end points. However, the use of biomarkers as surrogates for clinical outcomes is not without limitations. Dissecting causal and bystander effects is particularly challenging, and current measurement approaches may be misleading. For example, HDL cholesterol, originally considered a causal marker of CVD, is now understood to have a far more complex role in disease pathogenesis.210 Thus, although HDL function may be a causal mediator of disease risk, measurement of HDL concentration may not be an ideal risk predictor, and interventions aimed specifically at reducing HDL cholesterol concentrations have proven ineffective at reducing cardiovascular risk. Large, long-term, randomized, controlled trials; mendelian randomization approaches; and greater inclusion of both biomarkers and clinical end points may be required to establish the utility of many putative disease biomarkers.

The Microbiome

The microbiome, in particular the gut microbiome, has been identified as a potential risk factor for susceptibility to several chronic metabolic diseases, including diabetes mellitus,211 obesity,212 and CVD.213,214 Initial studies have focused primarily on inflammation, and a developing body of literature indicates that microbial dysbiosis215217 in the digestive tract may also influence systemic inflammation by altering gut permeability and thus increasing circulating lipopolysaccharide,218220 a powerful trigger of the immune response. An alternative line of literature is developing that views the microbiome as a metabolically active, complex organ, producing many metabolites that can directly influence host phenotype.

Inflammation and the Microbiome

Inflammation plays a unifying role in cardiometabolic disease,221 with inflammatory elements being observed in atherosclerosis,222 insulin resistance,223,224 and obesity. Obesity, in turn, is characterized by chronic, low-grade systemic inflammation, with adipocytes serving as key mediators for metabolic and cardiovascular sequelae. In addition, there is growing recognition of the importance of the gut in immune system regulation, with subsequent metabolic effects. A large proportion of quantitative trait locus regions reported to regulate microbial abundance contain genes related to immunity and maintenance of barrier function.225227 The influence of immune-related genes is further evidenced by the dramatic effects on microbial community structure caused by mutations in single genes related to host immunity.228,229 As an example, mice genetically deficient in the gut mucosal expression of innate immune system modulators (Toll-like receptor 5) developed insulin resistance even in the absence of increased obesity,217 and high fecal calprotectin levels have been found to be indicative of an inflammatory colonic environment.230 Several taxa such as members of the Clostridiales order are known to be decreased in intestinal inflammatory environments,231 suggesting that differences in immune response and chronic inflammatory disease susceptibility may result from differences in microbiota composition.

Host Genetics and Microbial Composition

Considering the interindividual variability at the level of the microbiome,232,233 detailed studies integrating the intestinal microbiome with disease risk complement current GWAS approaches and other efforts seeking to understand heterogeneity in health and disease status. Importantly, understanding how microbial diversity and specific microbial species affect clinical phenotypes and risk of CVD will be beneficial as we begin to focus on personalized approaches to nutrition and medicine. As our interest in the role of the microbiome role in chronic disease has expanded, so has interest in how host genetics influences microbial diversity. Several groups have reported that enteric microbial composition is a heritable trait,234,235 although results from twin studies have shown discordant evidence of heritability.236 However, studies using naturally occurring genetic variation among panels of inbred mouse strains and single gene mutations in genetically modified mice have consistently shown an effect of host genetics on intestinal microbial community structure225,226,237240 and may have increased power to detect genotype-driven microbial differences. This is especially relevant because murine studies allow tight control over environmental factors, including diet.226 For example, numerous genetic studies in mice, including those with using genetic reference panels,225,226,230,241 have demonstrated an effect of genetic background on microbial diversity. Interestingly, Benson and coworkers226 demonstrated that multiple taxa can colocalize to a single genetic locus, suggesting that a single genetic locus may regulate the abundance of several taxa. Inbred strain surveys in mice also demonstrate a significant effect of host genetic makeup on microbial diversity,241 and some of these differences have been linked to cardiometabolic phenotypes.242

However, the relative strength of environmental versus genetic signals on microbial regulation is unclear. Much of our knowledge of the environmental effects on the microbiome has been derived from studies in mice.239 Several studies have shown how the maternal environment, litter effects, cage mates, the location that the mice are housed, and the commercial vendor can influence microbial populations.226,230,241,243,244 Uterine implantation studies have also shown that mice of different genetic backgrounds have similar microbial composition when reared by the same foster mother, indicating that, in certain circumstances, environmental drivers can overpower genetic influences at least for nonadherent bacterial populations.243 These studies are further supported by studies that have demonstrated that bacteria from diverse sources can colonize the gut of gnotobiotic mice and compete with “normal” microbiota.245 Clearly, more work is needed to understand the interactions between host genetics and microbial diversity.

Effect of Diet

The human microbiome uses both dietary and host-derived nutrients for survival. Thus, changes in diet have a profound impact on the microbiome,239,246 including altering the overall bacterial composition. Host dietary factors can alter the intestinal environment, promoting a bloom or inhibition of certain taxa, as evident by the dynamic changes in both mouse and human microbial populations in response to dietary intervention.239 Interestingly, alterations early in life may have long-lasting effects on multiple phenotypes,247 but it is unclear how these effects influence the regulation of microbial diversity by GxE interactions237 and subsequent risk of disease.227,237

Although the study of GxE interactions influencing the microbiome is relatively new,248 studies using mouse genetic reference populations237 or single gene knockout models249 have demonstrated an interaction between microbiota and diet that is influenced by host genotype. These effects are less clear in humans, and a clinical study reported that, despite retained variation in taxonomy after dietary intervention, microbial gene expression, as assessed by RNASeq, clustered by diet group and exhibited less between-subject variation than at baseline.250

Reports have highlighted interactions between the microbiome and metabolism of dietary components such as phosphatidylcholine and carnitine on modulating CVD risk251253 through the metabolite trimethylamine N-oxide (TMAO). Collectively, these studies demonstrated that increased plasma TMAO levels were positively associated with aortic lesion formation in mice and with increased risk of prevalent CVD and incident adverse cardiac events and mortality in humans in the setting of heart failure,254,255 diabetes mellitus,256 and chronic kidney disease.257 TMAO is formed from trimethylamine via hepatic flavin mono-oxygenase 3.258 Mechanistic studies in mice have identified that modulation of flavin mono-oxygenase 3 levels affect TMAO levels and glucose and lipid metabolism,259,260 further complicating the identification of the precise mechanism by which TMAO affects CVD. The microbiome plays an obligate role in the formation of trimethylamine (from the trimethylamine-containing nutrients choline and carnitine), and antibiotic knockdown studies clearly show that TMAO is not formed in the absence of the microbiome.253 Bacterial species harboring putative choline utilization gene clusters (cut-c) have been suggested to play a central role in enteric trimethylamine formation261 (and therefore downstream TMAO production). The specific microbiota capable of generating trimethylamine have not been fully identified, but previous reports have indicated a relationship between plasma TMAO and members of the Tenericutes phylum,242·253 whereas species within the Desulfovibrio genus have also been demonstrated to degrade choline to trimethylamine.261

Despite the compelling evidence of the microbiome as a critical mediator of CVD risk, we still do not understand the factors responsible. For example, a comparative GWAS approach to discover loci for plasma TMAO levels identified a locus for TMAO levels on mouse chromosome 3 harboring Slc30a7, a gene that encodes a zinc transporter.262 In comparison, no significant loci were identified in a GWAS of ≈2000 subjects undergoing elective cardiac evaluation at the Cleveland Clinic.262 Notably, similar results were also reported in the population-based Framingham Heart Study.178 The relatively limited genetic signals observed for TMAO levels, at least in humans, thus far is consistent with the concept that interpersonal differences in diet and the repertoire of gut microbial species, more so than host genetic variants, likely serve as the primary determinants of plasma TMAO levels. Studies in mice have supported that both TMAO levels and atherosclerosis susceptibility are transmittable via the microbiome.263 Future studies are critical to better understand the relationship among diet, genetics, the microbiome, and ultimately cardiovascular risk.

Evoked Phenotypes

There are important physiological differences between individuals in the fasted state compared with a prandial or postprandial state or in response to pharmacological or pathogenic challenge. The functional changes that occur in response to a meal or other challenge may be more relevant to disease processes than resting metabolism,264,265 highlighting the degree of metabolic and phenotypic flexibility of an individual.266 Thus, although studying individuals in a rested fasting state has important utility in minimizing noise and improving reproducibility, resting biomarkers may not accurately reflect the physiological milieu of the more relevant dynamic and potentially disease-promoting state. The use of evoked phenotypes as a research tool for understanding dynamic physiology has considerable utility in cardiometabolic disease and is particularly applicable to nutrigenomic studies.

Dietary intervention to evoke phenotypes acutely has long been used in nutrition research, principally in the form of carbohydrate challenge (oral glucose tolerance test, frequently sampled intravenous glucose tolerance test), or fat challenge (oral lipid tolerance test), sometimes in combination with longer-term dietary interventions such as modification of dietary fat intake267,268 or sodium restriction.269 The addition of genetic or genomic information such as SNP genotype270273 can further increase the utility of these evoked challenge studies by highlighting subgroups of individuals with differing responses. Relatively few studies to date have used other omics approaches in combination with evoked dietary challenge, although the efficacy of the approach has been demonstrated, with examples of transcriptomic,5,274 metabolomic,275,276 proteomic,277 and lipidomic278 studies highlighting the many powerful potential applications of these approaches.

The effects of longer-term dietary interventions or of habitual dietary patterns are often extremely difficult to detect, with only very subtle changes in resting biomarkers expected from a dietary intervention or supplementation. Thus, even for substances such as n-3 PUFA that are well studied with multiple lines of evidence supporting health effects, detecting biological changes after intervention is difficult. In such studies, additional pharmacological challenges may be required to evoke a measurable and disease-relevant phenotype. The use of evoked endotoxemia (low-dose lipopolysaccharide) as a model of cardiometabolic disease is well documented279282 and has shown great utility as a discovery tool both in omic283,284 and nutritional contexts.88,285,286 The combination of nutritional and omic profiling in the context of evoked endotoxemia has great promise for understanding nutritional effects on inflammatory responses. Other challenges have also been successfully used in an integrative omic context, for example, the use of vaccination as a model for activation of immune response by Franco et al,287 which revealed novel genes acting in response to immune activation. Although this study did not include dietary analysis, the design has obvious utility for a nutrigenomic context. As an alternative to a direct pharmacological or acute dietary challenge, the use of repeated measures in longitudinal studies allows natural interventions to provoke phenotypes, including naturally occurring infection. This was highlighted in a single individual in a personalized omic profiling approach by Chen et al,288 which established a proof of principle that would be extremely valuable to apply to larger numbers of individuals.

Although the use of human evoked phenotypes as models of cardiometabolic disease is still in its infancy, the potential benefits, particularly in the nutritional context, are considerable. Evoked phenotypes not only are more biologically relevant to disease processes but also reveal a greater dynamic range, allowing statistical power for discovery with smaller numbers of individuals. Controlling the provocation can reduce or eliminate the issues of confounding and reverse causation inherent to observational studies. Increased use of phenotype challenges in combination with nutrigenomic approaches is a powerful and pragmatic approach that can yield disease-relevant data from smaller numbers of individuals. Given the historical difficulties in establishing dietary links with disease processes, in-depth phenotyping of dynamic nutrient-responsive physiology and integration with omics-scale data will likely yield significant advances in cardiometabolic disease research.

Personalized Nutrition

The ability to use evidence-based personalized or precision medicine through dietary intervention is a worthy healthcare goal with significant potential. Some examples of routine genetics-based dietary modification with large effects exist, for example, in inborn errors of metabolism such as phenylketonuria,289 mutations in human leukocyte antigen complex and other genes leading to celiac disease or gluten sensitivity,290 or variants in the LCT gene affecting lactase persistence.291 However, such clear examples do not yet exist for complex cardiometabolic disease. We caution that this area in particular is subject to popular claims that reach beyond the evidence base, with several companies offering direct-to-consumer genetic testing promising nutrigenetics-guided personalized dietary advice. The genetics-based ABO blood group diet292 became highly popular after its publication in 1996 and claimed to cure or prevent many chronic diseases, but it was not based on scientific evidence.293 Although the prescribed dietary patterns may be associated with health benefits, these occur independently of ABO blood group status.294 Although the scientific evidence to make personalized dietary recommendations is not yet convincingly established, there is evidence that consumers are receptive to personalized dietary advice. In a randomized trial of genetic-based personalized nutrition advice, individuals receiving personalized advice were more likely to understand the advice given and to judge it as useful.295 In this study, with the exception of sodium intake, participants were no more likely to adhere to the advice given compared with general dietary advice, but this was attributable to the fact that participants were already broadly adhering to dietary guidelines at baseline.296 An intervention to improve diet quality based on personalized advice for genotype at APOE found that high-risk individuals who received personalized dietary advice were more likely to make short-term dietary improvements compared with low-risk or control subjects.297 Although consumers are more engaged when receiving personalized dietary advice, the higher initial motivation may not lead to sustained long-term implementation of dietary changes. However, even if the hurdles of implementation and long-term patient motivation are overcome, the main obstacle to personalized nutrition lies in establishing sufficient scientific evidence to make informed and efficacious recommendations.

Pharmacogenomics trials are starting to study the impact and implementation of personalized drug treatment recommendations based on genotype at known functional variants.298,299 This approach would also be beneficial to advance the field of nutrigenetics. As discussed earlier, there are published reports of genetic variants that interact with dietary composition to modulate biomarkers and health outcomes, including within the context of randomized trials.57,300 However, most have not been validated through prospective, genotype-guided, randomized, controlled dietary intervention trials. Historical limitations in nutrition research limited the ability to develop clear evidence-based guidelines; personalized nutrition presents an opportunity to greatly improve on these recommendations using current standards and technologies. Although some gene-nutrient interaction variants with large effects exist, many of the reported gene-nutrient interaction variants have relatively small individual effects. Genetic risk scores may represent an alternative to single-variant analysis. Such scores have been applied in a predictive capacity to assess disease risk301303 and could be useful within the context of a gene-nutrient risk score. In a study assessing the effect on weight loss of genotype-guided nutrigenetics intervention using genotype information from 7 published SNPs,304 there was no difference in weight loss between the group assigned to personalized diet and the group assigned to a standard balanced diet, although adherence to diet correlated with weight loss in the nutrigenetics-diet group. Although personalized nutrition may prove to become an effective tool in disease prevention and management, current evidence does not yet demonstrate that personalized nutritional advice leads to improved health outcomes compared with following current dietary guidelines. However, the evidence that individuals are both receptive to personalized dietary advice and more motivated to implement personalized recommendations is very encouraging. Many of our existing nutritional guidelines were introduced before stringent standards for level of evidence; personalized nutrition research using optimal current methods allows considerable improvement and refinement of nutritional guidelines on both a personalized and a population level. Large, randomized, controlled trials guided by additional mechanistic research may allow personalized nutrition to become a realistic option for CVD management.

Limitations and Future Directions

Nutrition is a crucial component in the prevention of cardiometabolic disease, but dietary studies are limited by difficulties in accurately assessing dietary intake in free-living subjects and heterogeneity in habitual diet. Furthermore, the richness of dietary options and vast numbers of possible interactions between dietary components make dissecting relative contributions of various nutrients extremely difficult. Omic profiling represents a feasible albeit challenging method to address these issues, with the potential to use biomarkers coupled with genomic knowledge to obtain accurate and comprehensive assessments of nutritional input. However, to relate nutrient intake to nutritional biomarkers, well-conducted studies are required that use currently available nutritional profiling methods coupled with comprehensive unbiased biomarker profiling to discover and validate markers of metabolically active dietary components. Currently, many human studies of biomarkers and disease do not collect dietary information, and many dietary studies do not have resources for omic profiling. Increased collaboration among researchers with nutritional and omics expertise during planning and development stages would allow the efficient collection of the additional data and samples required to provide maximum benefit and allow nutrigenomics to be applied in human disease studies. An increased awareness of nutrition among cardiometabolic disease researchers would be beneficial to the field. Moreover, the collection of dietary information and additional samples for nutritional profiling does not add substantially to the cost of a human trial. Similarly, increased awareness of important dietary distinctions would be beneficial in rodent models of cardiometabolic disease, in which researchers often disregard the complexity of diet, for example, attributing differences in a Western diet and chow solely to the higher fat content despite many other differences in nutrient composition, including sucrose and micronutrients.305

Limitations in computational approaches remain a major bottleneck in nutrigenomics studies. As data collection becomes less arduous and less expensive, a limitation is the ability to analyze and make sense of the resulting data. Development of improved methods and standardized approaches for data reduction and integrative data analysis is crucial.

Although omics profiling methods will allow efficient discovery of new biomarkers, this approach is biased toward hypothesis generation. Even when biomarkers have direct clinical application, in most cases, focused functional and mechanistic interrogation will be required to fully understand the mechanisms of action. Thus, to avoid a glut of underinterpreted data, the research climate and standards in the field should encourage omics researchers to follow up findings with attempts at functional interrogation and translation.

Despite many challenges and limitations, the application of nutrigenomics approaches should be promoted and encouraged, given the potential for discovery and progress, with direct application to human health. Cooperation among researchers from many different disciplines combining diverse expertise will be required to move toward the common goal of an integrated omics approach to nutrition in cardiometabolic disease.

Summary and Conclusions

Despite the known importance of genetics in cardiometabolic disease, environment plays a large role in determining to what extent a genetic predisposition to disease will manifest. Although multiple environmental exposures are known to modify risk, diet is one of the most important. Smoking, another key modifiable risk factor, has already demonstrated improvements, with a marked reduction in smoking rates in the United States since the 1960s that has been accompanied by a reduction in cardiovascular events.306 However, as smoking rates have declined, diet-related obesity has increased. Diet is both essential and directly modifiable, meaning that improved knowledge of optimal nutrition has the potential to improve quality of life and to reduce global disease morbidity and mortality. The use of integrated omics approaches, together with nutritional information, will improve the ability to identify relationships between diet and health, including the interactions among diet, genetic background, and the microbiome that modify these relationships. This will allow the development of new therapeutic approaches, including targeted modification of dietary intake, pharmacotherapies, and new strategies in modulating the microbiome, aimed at the prevention and treatment of cardiometabolic disease.

Footnotes

The American Heart Association makes every effort to avoid any actual or potential conflicts of interest that may arise as a result of an outside relationship or a personal, professional, or business interest of a member of the writing panel. Specifically, all members of the writing group are required to complete and submit a Disclosure Questionnaire showing all such relationships that might be perceived as real or potential conflicts of interest.

This statement was approved by the American Heart Association Science Advisory and Coordinating Committee on January 22, 2016, and the American Heart Association Executive Committee on February 23, 2016. A copy of the document is available at http://professional.heart.org/statements by using either “Search for Guidelines & Statements” or the “Browse by Topic” area. To purchase additional reprints, call 843-216-2533 or kelle.ramsay@wolterskluwer.com.

Expert peer review of AHA Scientific Statements is conducted by the AHA Office of Science Operations. For more on AHA statements and guidelines development, visit http://professional.heart.org/statements. Select the “Guidelines & Statements” drop-down menu, then click “Publication Development.”

Disclosures
Writing Group Disclosures
Writing Group Member Employment Research Grant Other Research Support Speakers’ Bureau/Honoraria Expert Witness Ownership Interest Consultant/Advisory Board Other
Jane F. Ferguson Vanderbilt University Medical Center AHA None None None None None None
Hooman Allayee Institute for Genetic Medicine, University of Southern California Keck School of Medicine NIH None None None None None None
Brian J. Bennett University of North Carolina NHLBI None None None None Egg Nutrition Center* None
Robert E. Gerszten Massachusetts General Hospital None None None None None None None
Folami Ideraabdullah University of North Carolina School of Medicine NIH-NIEHS; UNC Chapel Hill*; UNC Chapel Hill NCTraCs* None None None None None UNC Chapel Hill; NIH NIEHS
Penny M. Kris-Etherton Pennsylvania State University None None None None None None None
José M. Ordovás Tufts University/Jean Mayer-USDA Human Nutrition Research Center on Aging NIH*; USDA* None None None None None None
Eric B. Rimm Harvard T.H. Chan School of Public Health US Blueberry Highbush Council/USDA None None None None IKEA* None
Thomas J. Wang Vanderbilt University School of Medicine NIH None None None None None None
This table represents the relationships of writing group members that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all members of the writing group are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.
*
Modest.
Significant.
Reviewer Disclosures
Reviewer Employment Research Grant Other Research Support Speakers’ Bureau/Honoraria Expert Witness Ownership Interest Consultant/Advisory Board Other
Ahmed El-Sohemy University of Toronto (Canada) CIHR and other funding agencies in Canada (nutrigenomics is the focus of my research, and I hold and have held several major grants on this topic)* None None None I hold shares in Nutrigenomix Inc, a genetic testing company for personalized nutrition* None None
Ronald M. Krauss Children’s Hospital Oakland Research Institute Dairy Research Institute (microbiome analyses supported)†; Almond Board of California (microbiome analyses supported)* NIH (microbiome analyses supported)* None None None None None
Joseph Loscalzo Brigham and Women’s Hospital None None None None None None None
This table represents the relationships of reviewers that may be perceived as actual or reasonably perceived conflicts of interest as reported on the Disclosure Questionnaire, which all reviewers are required to complete and submit. A relationship is considered to be “significant” if (a) the person receives $10 000 or more during any 12-month period, or 5% or more of the person’s gross income; or (b) the person owns 5% or more of the voting stock or share of the entity, or owns $10 000 or more of the fair market value of the entity. A relationship is considered to be “modest” if it is less than “significant” under the preceding definition.
*
Significant.

References

  • 1.Müller M, Kersten S. Nutrigenomics: goals and strategies. Nat Rev Genet. 2003;4:315–322. doi: 10.1038/nrg1047. [DOI] [PubMed] [Google Scholar]
  • 2.Corthésy-Theulaz I, den Dunnen JT, Ferré P, Geurts JM, Müller M, van Belzen N, van Ommen B. Nutrigenomics: the impact of biomics technology on nutrition research. Ann Nutr Metab. 2005;49:355–365. doi: 10.1159/000088315. [DOI] [PubMed] [Google Scholar]
  • 3.Ordovas JM. Genetic interactions with diet influence the risk of cardiovascular disease. Am J Clin Nutr. 2006;83:443S–446S. [DOI] [PubMed] [Google Scholar]
  • 4.Kaput J, Noble J, Hatipoglu B, Kohrs K, Dawson K, Bartholomew A. Application of nutrigenomic concepts to type 2 diabetes mellitus. Nutr Metab Cardiovasc Dis. 2007;17:89–103. doi: 10.1016/j.numecd.2006.11.006. [DOI] [PubMed] [Google Scholar]
  • 5.Afman LA, Müller M. Human nutrigenomics of gene regulation by dietary fatty acids. Prog Lipid Res. 2012;51:63–70. doi: 10.1016/j.plipres.2011.11.005. [DOI] [PubMed] [Google Scholar]
  • 6.Afman L, Müller M. Nutrigenomics: from molecular nutrition to prevention of disease. J Am Diet Assoc. 2006;106:569–576. doi: 10.1016/j.jada.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 7.Keys A, Grande F. Role of dietary fat in human nutrition, III: diet and the epidemiology of coronary heart disease. Am J Public Health Nations Health. 1957;47:1520–1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Keys A, Anderson JT, Grande F. Serum cholesterol in man: diet fat and intrinsic responsiveness. Circulation. 1959;19:201–214. [DOI] [PubMed] [Google Scholar]
  • 9.US Department of Agriculture. Dietary Guidelines for Americans. 2010. health.gov/dietaryguidelines/dga2010/dietaryguidelines2010.pdf. Accessed October 31, 2014.
  • 10.US Department of Agriculture, US Department of Health and Human Services. Nutrition and Your Health: Dietary Guidelines for Americans, Fifth Edition. 2000. http://health.gov/dietaryguidelines/dga95/. Accessed October 31, 2014.
  • 11.Satija A, Yu E, Willett WC, Hu FB. Understanding nutritional epidemiology and its role in policy. Adv Nutr. 2015;6:5–18. doi: 10.3945/an.114.007492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Krauss RM, Eckel RH, Howard B, Appel LJ, Daniels SR, Deckelbaum RJ, Erdman JW Jr, Kris-Etherton P, Goldberg IJ, Kotchen TA, Lichtenstein AH, Mitch WE, Mullis R, Robinson K, Wylie-Rosett J, St Jeor S, Suttie J, Tribble DL, Bazzarre TL. AHA Dietary Guidelines: revision 2000: a statement for healthcare professionals from the Nutrition Committee of the American Heart Association. Circulation. 2000;102:2284–2299. [DOI] [PubMed] [Google Scholar]
  • 13.Kris-Etherton PM, Harris WS, Appel LJ; for the Nutrition Committee. Fish consumption, fish oil, omega-3 fatty acids, and cardiovascular disease [published correction appears in Circulation. 2003;107:512] Circulation. 2002;106:2747–2757. [DOI] [PubMed] [Google Scholar]
  • 14.Sacks FM, Obarzanek E, Windhauser MM, Svetkey LP, Vollmer WM, McCullough M, Karanja N, Lin PH, Steele P, Proschan MA, Evans MA, Appel LJ, Bray GA, Vogt TM, Moore TJ; DASH Investigators. Rationale and design of the Dietary Approaches to Stop Hypertension trial (DASH): a multicenter controlled-feeding study of dietary patterns to lower blood pressure. Ann Epidemiol 1995;5:108–118. [DOI] [PubMed] [Google Scholar]
  • 15.Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM; Diabetes Prevention Program Research Group. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346:393–403. doi: 10.1056/NEJMoa012512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Greenberg I, Stampfer MJ, Schwarzfuchs D, Shai I; DIRECT Group. Adherence and success in long-term weight loss diets: the Dietary Intervention Randomized Controlled Trial (DIRECT). J Am Coll Nutr. 2009;28:159–168. [DOI] [PubMed] [Google Scholar]
  • 17.de Lorgeril M, Salen P, Martin JL, Monjaud I, Delaye J, Mamelle N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation. 1999;99:779–785. [DOI] [PubMed] [Google Scholar]
  • 18.Martínez-González MÁ, Corella D, Salas-Salvadó J, Ros E, Covas MI, Fiol M, Wärnberg J, Arós F, Ruíz-Gutiérrez V, Lamuela-Raventós RM, Lapetra J, Muñoz MÁ, Martínez JA, Sáez G, Serra-Majem L, Pintó X, Mitjavila MT, Tur JA, Portillo MP, Estruch R; PREDIMED Study Investigators. Cohort profile: design and methods of the PREDIMED study. Int J Epidemiol. 2012;41:377–385. doi: 10.1093/ije/dyq250. [DOI] [PubMed] [Google Scholar]
  • 19.Estruch R, Ros E, Salas-Salvadó J, Covas MI, Corella D, Arós F, Gómez-Gracia E, Ruiz-Gutiérrez V, Fiol M, Lapetra J, Lamuela-Raventos RM, Serra-Majem L, Pintó X, Basora J, Muñoz MA, Sorlí JV, Martínez JA, Martínez-González MA; PREDIMED Study Investigators. Primary prevention of cardiovascular disease with a Mediterranean diet [published correction appears in N Engl J Med. 2014;370:886] N Engl J Med. 2013;368:1279–1290. doi: 10.1056/NEJMoa1200303. [DOI] [PubMed] [Google Scholar]
  • 20.Subar AF, Dodd KW, Guenther PM, Kipnis V, Midthune D, McDowell M, Tooze JA, Freedman LS, Krebs-Smith SM. The food propensity questionnaire: concept, development, and validation for use as a covariate in a model to estimate usual food intake. J Am Diet Assoc. 2006;106:1556–1563. doi: 10.1016/j.jada.2006.07.002. [DOI] [PubMed] [Google Scholar]
  • 21.Kirkpatrick SI, Subar AF, Douglass D, Zimmerman TP, Thompson FE, Kahle LL, George SM, Dodd KW, Potischman N. Performance of the Automated Self-Administered 24-hour Recall relative to a measure of true intakes and to an interviewer-administered 24-h recall. Am J Clin Nutr. 2014;100:233–240. doi: 10.3945/ajcn.114.083238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Willett WC. Nutritional Epidemiology. 3rd ed. New York, NY: Oxford University Press; 2012. [Google Scholar]
  • 23.Subar AF, Crafts J, Zimmerman TP, Wilson M, Mittl B, Islam NG, McNutt S, Potischman N, Buday R, Hull SG, Baranowski T, Guenther PM, Willis G, Tapia R, Thompson FE. Assessment of the accuracy of portion size reports using computer-based food photographs aids in the development of an automated self-administered 24-hour recall. J Am Diet Assoc. 2010;110:55–64. doi: 10.1016/j.jada.2009.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Thompson FE, Subar AF, Loria CM, Reedy JL, Baranowski T. Need for technological innovation in dietary assessment. J Am Diet Assoc. 2010;110:48–51. doi: 10.1016/j.jada.2009.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Subar AF, Kirkpatrick SI, Mittl B, Zimmerman TP, Thompson FE, Bingley C, Willis G, Islam NG, Baranowski T, McNutt S, Potischman N. The Automated Self-Administered 24-hour dietary recall (ASA24): a resource for researchers, clinicians, and educators from the National Cancer Institute. J Acad Nutr Diet. 2012;112:1134–1137. doi: 10.1016/j.jand.2012.04.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ganesh SK, Arnett DK, Assimes TL, Assimes TL, Basson CT, Chakravarti A, Ellinor PT, Engler MB, Goldmuntz E, Herrington DM, Hershberger RE, Hong Y, Johnson JA, Kittner SJ, McDermott DA, Meschia JF, Mestroni L, O’Donnell CJ, Psaty BM, Vasan RS, Ruel M, Shen W-K, Terzic A, Waldman SA; on behalf of the American Heart Association Council on Functional Genomics and Translational Biology, Council on Epidemiology and Prevention, Council on Basic Cardiovascular Sciences, Council on Cardiovascular Disease in the Young, Council on Cardiovascular Surgery and Anesthesia, Council on Clinical Cardiology, Council on Cardiovascular and Stroke Nursing, and Stroke Council. Genetics and genomics for the prevention and treatment of cardiovascular disease: update: a scientific statement from the American Heart Association [published correction appears in Circulation. 2014;129:e398] Circulation. 2013;128:2813–2851. doi: 10.1161/01.cir.0000437913.98912.1d. [DOI] [PubMed] [Google Scholar]
  • 27.Do R, Stitziel NO, Won HH, Jørgensen AB, Duga S, Angelica Merlini P, Kiezun A, Farrall M, Goel A, Zuk O, Guella I, Asselta R, Lange LA, Peloso GM, Auer PL, NHLBI Exome Sequencing Project, Girelli D, Martinelli N, Farlow DN, DePristo MA, Roberts R, Stewart AF, Saleheen D, Danesh J, Epstein SE, Sivapalaratnam S, Hovingh GK, Kastelein JJ, Samani NJ, Schunkert H, Erdmann J, Shah SH, Kraus WE, Davies R, Nikpay M, Johansen CT, Wang J, Hegele RA, Hechter E, Marz W, Kleber ME, Huang J, Johnson AD, Li M, Burke GL, Gross M, Liu Y, Assimes TL, Heiss G, Lange EM, Folsom AR, Taylor HA, Olivieri O, Hamsten A, Clarke R, Reilly DF, Yin W, Rivas MA, Donnelly P, Rossouw JE, Psaty BM, Herrington DM, Wilson JG, Rich SS, Bamshad MJ, Tracy RP, Cupples LA, Rader DJ, Reilly MP, Spertus JA, Cresci S, Hartiala J, Tang WH, Hazen SL, Allayee H, Reiner AP, Carlson CS, Kooperberg C, Jackson RD, Boerwinkle E, Lander ES, Schwartz SM, Siscovick DS, McPherson R, Tybjaerg-Hansen A, Abecasis GR, Watkins H, Nickerson DA, Ardissino D, Sunyaev SR, O’Donnell CJ, Altshuler D, Gabriel S, Kathiresan S. Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature. 2015;518:102–106. doi: 10.1038/nature13917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.TG and HDL Working Group of the Exome Sequencing Project, National Heart, Lung, and Blood Institute, Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, Lu Y, Tang ZZ, Zhang H, Hindy G, Masca N, Stirrups K, Kanoni S, Do R, Jun G, Hu Y, Kang HM, Xue C, Goel A, Farrall M, Duga S, Merlini PA, Asselta R, Girelli D, Olivieri O, Martinelli N, Yin W, Reilly D, Speliotes E, Fox CS, Hveem K, Holmen OL, Nikpay M, Farlow DN, Assimes TL, Franceschini N, Robinson J, North KE, Martin LW, DePristo M, Gupta N, Escher SA, Jansson JH, Van Zuydam N, Palmer CN, Wareham N, Koch W, Meitinger T, Peters A, Lieb W, Erbel R, Konig IR, Kruppa J, Degenhardt F, Gottesman O, Bottinger EP, O’Donnell CJ, Psaty BM, Ballantyne CM, Abecasis G, Ordovas JM, Melander O, Watkins H, Orho-Melander M, Ardissino D, Loos RJ, McPherson R, Willer CJ, Erdmann J, Hall AS, Samani NJ, Deloukas P, Schunkert H, Wilson JG, Kooperberg C, Rich SS, Tracy RP, Lin DY, Altshuler D, Gabriel S, Nickerson DA, Jarvik GP, Cupples LA, Reiner AP, Boerwinkle E, Kathiresan S. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med. 2014;371:22–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Corella D, Ordovas JM. Nutrigenomics in cardiovascular medicine. Circ Cardiovasc Genet. 2009;2:637–651. doi: 10.1161/CIRCGENETICS.109.891366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bis JC, Glazer NL, Psaty BM. Genome-wide association studies of cardiovascular risk factors: design, conduct and interpretation. J Thromb Haemost. 2009;7(suppl 1):308–311. doi: 10.1111/j.1538-7836.2009.03392.x. [DOI] [PubMed] [Google Scholar]
  • 31.Ordovas JM, Corella D. Nutritional genomics. Annu Rev Genomics Hum Genet. 2004;5:71–118. doi: 10.1146/annurev.genom.5.061903.180008. [DOI] [PubMed] [Google Scholar]
  • 32.Allayee H, Baylin A, Hartiala J, Wijesuriya H, Mehrabian M, Lusis AJ, Campos H. Nutrigenetic association of the 5-lipoxygenase gene with myocardial infarction. Am J Clin Nutr. 2008;88:934–940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Campos H, D’Agostino M, Ordovás JM. Gene-diet interactions and plasma lipoproteins: role of apolipoprotein E and habitual saturated fat intake. Genet Epidemiol. 2001;20:117–128. doi: . [DOI] [PubMed] [Google Scholar]
  • 34.Corella D, Carrasco P, Sorlí JV, Estruch R, Rico-Sanz J, Martínez-González MÁ, Salas-Salvadó J, Covas MI, Coltell O, Arós F, Lapetra J, Serra-Majem L, Ruiz-Gutiérrez V, Warnberg J, Fiol M, Pintó X, Ortega-Azorin C, Muñoz MÁ, Martínez JA, Gómez-Gracia E, González JI, Ros E, Ordovás JM. Mediterranean diet reduces the adverse effect of the TCF7L2-rs7903146 polymorphism on cardiovascular risk factors and stroke incidence: a randomized controlled trial in a high-cardiovascular-risk population. Diabetes Care. 2013;36:3803–3811. doi: 10.2337/dc13-0955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Corella D, Guillén M, Sáiz C, Portolés O, Sabater A, Cortina S, Folch J, González JI, Ordovas JM. Environmental factors modulate the effect of the APOE genetic polymorphism on plasma lipid concentrations: ecogenetic studies in a Mediterranean Spanish population. Metabolism. 2001;50:936–944. [DOI] [PubMed] [Google Scholar]
  • 36.Corella D, Peloso G, Arnett DK, Demissie S, Cupples LA, Tucker K, Lai CQ, Parnell LD, Coltell O, Lee YC, Ordovas JM. APOA2, dietary fat, and body mass index: replication of a gene-diet interaction in 3 independent populations. Arch Intern Med. 2009;169:1897–1906. doi: 10.1001/archinternmed.2009.343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Corella D, Portolés O, Arriola L, Chirlaque MD, Barrricarte A, Francés F, Huerta JM, Larrañaga N, Martínez C, Martinez-Camblor P, Molina E, Navarro C, Quirós JR, Rodríguez L, Sánchez MJ, Ros E, Sala N, González CA, Moreno-Iribas C. Saturated fat intake and alcohol consumption modulate the association between the APOE polymorphism and risk of future coronary heart disease: a nested case-control study in the Spanish EPIC cohort. J Nutr Biochem. 2011;22:487–494. doi: 10.1016/j.jnutbio.2010.04.003. [DOI] [PubMed] [Google Scholar]
  • 38.Corella D, Qi L, Tai ES, Deurenberg-Yap M, Tan CE, Chew SK, Ordovas JM. Perilipin gene variation determines higher susceptibility to insulin resistance in Asian women when consuming a high-saturated fat, low-carbohydrate diet. Diabetes Care. 2006;29:1313–1319. doi: 10.2337/dc06-0045. [DOI] [PubMed] [Google Scholar]
  • 39.Corella D, Tucker K, Lahoz C, Coltell O, Cupples LA, Wilson PW, Schaefer EJ, Ordovas JM. Alcohol drinking determines the effect of the APOE locus on LDL-cholesterol concentrations in men: the Framingham Offspring Study. Am J Clin Nutr. 2001;73:736–745. [DOI] [PubMed] [Google Scholar]
  • 40.Cornelis MC, El-Sohemy A, Campos H. GSTT1 genotype modifies the association between cruciferous vegetable intake and the risk of myocardial infarction. Am J Clin Nutr. 2007;86:752–758. [DOI] [PubMed] [Google Scholar]
  • 41.Cornelis MC, El-Sohemy A, Kabagambe EK, Campos H. Coffee, CYP1A2 genotype, and risk of myocardial infarction. JAMA. 2006;295:1135–1141. doi: 10.1001/jama.295.10.1135. [DOI] [PubMed] [Google Scholar]
  • 42.D’Angelo A, Coppola A, Madonna P, Fermo I, Pagano A, Mazzola G, Galli L, Cerbone AM. The role of vitamin B12 in fasting hyperhomocysteinemia and its interaction with the homozygous C677T mutation of the methylenetetrahydrofolate reductase (MTHFR) gene: a case-control study of patients with early-onset thrombotic events. Thromb Haemost. 2000;83:563–570. [PubMed] [Google Scholar]
  • 43.Dedoussis GV, Panagiotakos DB, Chrysohoou C, Pitsavos C, Zampelas A, Choumerianou D, Stefanadis C. Effect of interaction between adherence to a Mediterranean diet and the methylenetetrahydrofolate reductase 677C–>T mutation on homocysteine concentrations in healthy adults: the ATTICA Study. Am J Clin Nutr. 2004;80:849–854. [DOI] [PubMed] [Google Scholar]
  • 44.Do R, Xie C, Zhang X, Männistö S, Harald K, Islam S, Bailey SD, Rangarajan S, McQueen MJ, Diaz R, Lisheng L, Wang X, Silander K, Peltonen L, Yusuf S, Salomaa V, Engert JC, Anand SS; INTERHEART Investigators. The effect of chromosome 9p21 variants on cardiovascular disease may be modified by dietary intake: evidence from a case/control and a prospective study. PLoS Med. 2011;8:e1001106. doi: 10.1371/journal.pmed.1001106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Dwyer JH, Allayee H, Dwyer KM, Fan J, Wu H, Mar R, Lusis AJ, Mehrabian M. Arachidonate 5-lipoxygenase promoter genotype, dietary arachidonic acid, and atherosclerosis. N Engl J Med. 2004;350:29–37. doi: 10.1056/NEJMoa025079. [DOI] [PubMed] [Google Scholar]
  • 46.Fumeron F, Betoulle D, Luc G, Behague I, Ricard S, Poirier O, Jemaa R, Evans A, Arveiler D, Marques-Vidal P. Alcohol intake modulates the effect of a polymorphism of the cholesteryl ester transfer protein gene on plasma high density lipoprotein and the risk of myocardial infarction. J Clin Invest. 1995;96:1664–1671. doi: 10.1172/JCI118207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Garcia-Rios A, Delgado-Lista J, Perez-Martinez P, Phillips CM, Ferguson JF, Gjelstad IM, Williams CM, Karlström B, Kieć-Wilk B, Blaak EE, Lairon D, Planells R, Malczewska-Malec M, Defoort C, Riserus U, Saris WH, Lovegrove JA, Drevon CA, Roche HM, Lopez-Miranda J. Genetic variations at the lipoprotein lipase gene influence plasma lipid concentrations and interact with plasma n-6 polyunsaturated fatty acids to modulate lipid metabolism. Atherosclerosis. 2011;218:416–422. doi: 10.1016/j.atherosclerosis.2011.07.092. [DOI] [PubMed] [Google Scholar]
  • 48.Hartiala J, Gilliam E, Vikman S, Campos H, Allayee H. Association of PLA2G4A with myocardial infarction is modulated by dietary PUFAs. Am J Clin Nutr. 2012;95:959–965. doi: 10.3945/ajcn.111.032094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hines LM, Stampfer MJ, Ma J, Gaziano JM, Ridker PM, Hankinson SE, Sacks F, Rimm EB, Hunter DJ. Genetic variation in alcohol dehydrogenase and the beneficial effect of moderate alcohol consumption on myocardial infarction. N Engl J Med. 2001;344:549–555. doi: 10.1056/NEJM200102223440802. [DOI] [PubMed] [Google Scholar]
  • 50.Jansen S, Lopez-Miranda J, Salas J, Ordovas JM, Castro P, Marin C, Ostos MA, Lopez-Segura F, Jimenez-Pereperez JA, Blanco A, Perez-Jimenez F. Effect of 347-serine mutation in apoprotein A-IV on plasma LDL cholesterol response to dietary fat. Arterioscler Thromb Vasc Biol. 1997;17:1532–1538. [DOI] [PubMed] [Google Scholar]
  • 51.Jensen MK, Mukamal KJ, Overvad K, Rimm EB. Alcohol consumption, TaqIB polymorphism of cholesteryl ester transfer protein, high-density lipoprotein cholesterol, and risk of coronary heart disease in men and women. Eur Heart J. 2008;29:104–112. doi: 10.1093/eurheartj/ehm517. [DOI] [PubMed] [Google Scholar]
  • 52.Markus HS, Ali N, Swaminathan R, Sankaralingam A, Molloy J, Powell J. A common polymorphism in the methylenetetrahydrofolate reductase gene, homocysteine, and ischemic cerebrovascular disease. Stroke. 1997;28:1739–1743. [DOI] [PubMed] [Google Scholar]
  • 53.Nettleton JA, Volcik KA, Hoogeveen RC, Boerwinkle E. Carbohydrate intake modifies associations between ANGPTL4[E40K] genotype and HDL-cholesterol concentrations in white men from the Atherosclerosis Risk in Communities (ARIC) study. Atherosclerosis. 2009;203:214–220. doi: 10.1016/j.atherosclerosis.2008.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Norat T, Bowman R, Luben R, Welch A, Khaw KT, Wareham N, Bingham S. Blood pressure and interactions between the angiotensin polymorphism AGT M235T and sodium intake: a cross-sectional population study. Am J Clin Nutr. 2008;88:392–397. [DOI] [PubMed] [Google Scholar]
  • 55.Ordovas JM, Corella D, Demissie S, Cupples LA, Couture P, Coltell O, Wilson PW, Schaefer EJ, Tucker KL. Dietary fat intake determines the effect of a common polymorphism in the hepatic lipase gene promoter on high-density lipoprotein metabolism: evidence of a strong dose effect in this gene-nutrient interaction in the Framingham study. Circulation. 2002;106:2315–2321. [DOI] [PubMed] [Google Scholar]
  • 56.Pérez-Martínez P, Adarraga-Cansino MD, Fernández de la Puebla RA, Blanco-Molina A, Delgado-Lista J, Marín C, Ordovás JM, López-Miranda J, Pérez-Jiménez F. The -675 4G/5G polymorphism at the plasminogen activator inhibitor 1 (PAI-1) gene modulates plasma plasminogen activator inhibitor 1 concentrations in response to dietary fat consumption. Br J Nutr. 2008;99:699–702. doi: 10.1017/S0007114507831710. [DOI] [PubMed] [Google Scholar]
  • 57.Ortega-Azorín C, Sorlí JV, Estruch R, Asensio EM, Coltell O, González JI, Martinez-González MÁ, Ros E, Salas-Salvadó J, Fitó M, Arós F, Lapetra J, Serra-Majem L, Ruiz-Gutiérrez V, Gómez-Gracia E, Fiol M, Flores G, Pintó X, Saiz C, Ordovás JM, Corella D. Amino acid change in the carbohydrate response element binding protein is associated with lower triglycerides and myocardial infarction incidence depending on level of adherence to the Mediterranean diet in the PREDIMED trial. Circ Cardiovasc Genet. 2014;7:49–58. doi: 10.1161/CIRCGENETICS.113.000301. [DOI] [PubMed] [Google Scholar]
  • 58.Richardson K, Louie-Gao Q, Arnett DK, Parnell LD, Lai CQ, Davalos A, Fox CS, Demissie S, Cupples LA, Fernandez-Hernando C, Ordovas JM. The PLIN4 variant rs8887 modulates obesity related phenotypes in humans through creation of a novel miR-522 seed site. PLoS One. 2011;6:e17944. doi: 10.1371/journal.pone.0017944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ruiz-Narváez EA, Kraft P, Campos H. Ala12 variant of the peroxisome proliferator-activated receptor-gamma gene (PPARG) is associated with higher polyunsaturated fat in adipose tissue and attenuates the protective effect of polyunsaturated fat intake on the risk of myocardial infarction. Am J Clin Nutr. 2007;86:1238–1242. [DOI] [PubMed] [Google Scholar]
  • 60.Tai ES, Corella D, Demissie S, Cupples LA, Coltell O, Schaefer EJ, Tucker KL, Ordovas JM; Framingham Heart Study. Polyunsaturated fatty acids interact with the PPARA-L162V polymorphism to affect plasma triglyceride and apolipoprotein C-III concentrations in the Framingham Heart Study. J Nutr. 2005;135:397–403. [DOI] [PubMed] [Google Scholar]
  • 61.Vinukonda G, Shaik Mohammad N, Md Nurul Jain J, Prasad Chintakindi K, Rama Devi Akella R. Genetic and environmental influences on total plasma homocysteine and coronary artery disease (CAD) risk among South Indians. Clin Chim Acta. 2009;405:127–131. doi: 10.1016/j.cca.2009.04.015. [DOI] [PubMed] [Google Scholar]
  • 62.Yang Y, Ruiz-Narvaez E, Kraft P, Campos H. Effect of apolipoprotein E genotype and saturated fat intake on plasma lipids and myocardial infarction in the Central Valley of Costa Rica. Hum Biol. 2007;79:637–647. doi: 10.1353/hub.2008.0010. [DOI] [PubMed] [Google Scholar]
  • 63.Younis J, Cooper JA, Miller GJ, Humphries SE, Talmud PJ. Genetic variation in alcohol dehydrogenase 1C and the beneficial effect of alcohol intake on coronary heart disease risk in the Second Northwick Park Heart Study. Atherosclerosis. 2005;180:225–232. doi: 10.1016/j.atherosclerosis.2004.12.010. [DOI] [PubMed] [Google Scholar]
  • 64.Zhang L, Miyaki K, Araki J, Song Y, Kimura T, Omae K, Muramatsu M. Interaction of angiotensin I-converting enzyme insertion-deletion polymorphism and daily salt intake influences hypertension in Japanese men. Hypertens Res. 2006;29:751–758. doi: 10.1291/hypres.29.751. [DOI] [PubMed] [Google Scholar]
  • 65.Yoo JH, Park SC. Low plasma folate in combination with the 677 C–>T methylenetetrahydrofolate reductase polymorphism is associated with increased risk of coronary artery disease in Koreans. Thromb Res. 2000;97:77–84. [DOI] [PubMed] [Google Scholar]
  • 66.Zheng JS, Arnett DK, Parnell LD, Smith CE, Li D, Borecki IB, Tucker KL, Ordovas JM, Lai CQ. Modulation by dietary fat and carbohydrate of IRS1 association with type 2 diabetes traits in two populations of different ancestries. Diabetes Care. 2013;36:2621–2627. doi: 10.2337/dc12-2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Zheng JS, Parnell LD, Smith CE, Lee YC, Jamal-Allial A, Ma Y, Li D, Tucker KL, Ordovás JM, Lai CQ. Circulating 25-hydroxyvitamin D, IRS1 variant rs2943641, and insulin resistance: replication of a gene-nutrient interaction in 4 populations of different ancestries. Clin Chem. 2014;60:186–196. doi: 10.1373/clinchem.2013.215251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.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. doi: 10.1186/1756-0381-7-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Weedon MN, McCarthy MI, Hitman G, Walker M, Groves CJ, Zeggini E, Rayner NW, Shields B, Owen KR, Hattersley AT, Frayling TM. Combining information from common type 2 diabetes risk polymorphisms improves disease prediction. PLoS Med. 2006;3:e374. doi: 10.1371/journal.pmed.0030374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Sonestedt E, Roos C, Gullberg B, Ericson U, Wirfält E, Orho-Melander M. Fat and carbohydrate intake modify the association between genetic variation in the FTO genotype and obesity. Am J Clin Nutr. 2009;90:1418–1425. doi: 10.3945/ajcn.2009.27958. [DOI] [PubMed] [Google Scholar]
  • 71.Boushey CJ, Beresford SA, Omenn GS, Motulsky AG. A quantitative assessment of plasma homocysteine as a risk factor for vascular disease: probable benefits of increasing folic acid intakes. JAMA. 1995;274:1049–1057. [DOI] [PubMed] [Google Scholar]
  • 72.Barak AJ, Beckenhauer HC, Hidiroglou N, Camilo ME, Selhub J, Tuma DJ. The relationship of ethanol feeding to the methyl folate trap. Alcohol. 1993;10:495–497. [DOI] [PubMed] [Google Scholar]
  • 73.Shaw S, Jayatilleke E, Herbert V, Colman N. Cleavage of folates during ethanol metabolism: role of acetaldehyde/xanthine oxidase-generated superoxide. Biochem J. 1989;257:277–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Chiuve SE, Giovannucci EL, Hankinson SE, Hunter DJ, Stampfer MJ, Willett WC, Rimm EB. Alcohol intake and methylenetetrahydrofolate reductase polymorphism modify the relation of folate intake to plasma homocysteine. Am J Clin Nutr. 2005;82:155–162. [DOI] [PubMed] [Google Scholar]
  • 75.Schupp M, Lazar MA. Endogenous ligands for nuclear receptors: digging deeper. J Biol Chem. 2010;285:40409–40415. doi: 10.1074/jbc.R110.182451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Daynes RA, Jones DC. Emerging roles of PPARs in inflammation and immunity. Nat Rev Immunol. 2002;2:748–759. doi: 10.1038/nri912. [DOI] [PubMed] [Google Scholar]
  • 77.Göttlicher M, Widmark E, Li Q, Gustafsson JA. Fatty acids activate a chimera of the clofibric acid-activated receptor and the glucocorticoid receptor. Proc Natl Acad Sci U S A. 1992;89:4653–4657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Volcik KA, Nettleton JA, Ballantyne CM, Boerwinkle E. Peroxisome proliferator-activated receptor [alpha] genetic variation interacts with n-6 and long-chain n-3 fatty acid intake to affect total cholesterol and LDL-cholesterol concentrations in the Atherosclerosis Risk in Communities Study. Am J Clin Nutr. 2008;87:1926–1931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Alsaleh A, O’Dell SD, Frost GS, Griffin BA, Lovegrove JA, Jebb SA, Sanders TA; RISCK Study Investigators. Interaction of PPARG Pro12Ala with dietary fat influences plasma lipids in subjects at cardiometabolic risk. J Lipid Res. 2011;52:2298–2303. doi: 10.1194/jlr.P019281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, Sternbach DD, Lehmann JM, Wisely GB, Willson TM, Kliewer SA, Milburn MV. Molecular recognition of fatty acids by peroxisome proliferator-activated receptors. Mol Cell. 1999;3:397–403. [DOI] [PubMed] [Google Scholar]
  • 81.Cavalieri D, Calura E, Romualdi C, Marchi E, Radonjic M, Van Ommen B, Möller M. Filling gaps in PPAR-alpha signaling through comparative nutrigenomics analysis. BMC Genomics. 2009;10:596. doi: 10.1186/1471-2164-10-596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Georgiadi A, Boekschoten MV, Müller M, Kersten S. Detailed transcriptomics analysis of the effect of dietary fatty acids on gene expression in the heart. Physiol Genomics. 2012;44:352–361. doi: 10.1152/physiolgenomics.00115.2011. [DOI] [PubMed] [Google Scholar]
  • 83.Laudet V Evolution of the nuclear receptor superfamily: early diversification from an ancestral orphan receptor. J Mol Endocrinol. 1997;19:207–226. [DOI] [PubMed] [Google Scholar]
  • 84.Sanderson LM, Boekschoten MV, Desvergne B, Müller M, Kersten S. Transcriptional profiling reveals divergent roles of PPARalpha and PPARbeta/delta in regulation of gene expression in mouse liver [published correction appears in Physiol Genomics. 2010;42:317] Physiol Genomics. 2010;41:42–52. doi: 10.1152/physiolgenomics.00127.2009. [DOI] [PubMed] [Google Scholar]
  • 85.Sanderson LM, Degenhardt T, Koppen A, Kalkhoven E, Desvergne B, Müller M, Kersten S. Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) but not PPARalpha serves as a plasma free fatty acid sensor in liver [published correction appears in Mol Cell Biol. 2010;30:4977] Mol Cell Biol. 2009;29:6257–6267. doi: 10.1128/MCB.00370-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, Wahli W. The PPARalpha-leukotriene B4 pathway to inflammation control. Nature. 1996;384:39–43. doi: 10.1038/384039a0. [DOI] [PubMed] [Google Scholar]
  • 87.Mulvey CK, Ferguson JF, Tabita-Martinez J, Kong S, Shah RY, Patel PN, Master SR, Usman MH, Propert KJ, Shah R, Mehta NN, Reilly MP. Peroxisome proliferator-activated receptor-α agonism with fenofibrate does not suppress inflammatory responses to evoked endotoxemia. J Am Heart Assoc. 2012;1:e002923. doi: 10.1161/JAHA.112.002923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ferguson JF, Mulvey CK, Patel PN, Shah RY, Doveikis J, Zhang W, Tabita-Martinez J, Terembula K, Eiden M, Koulman A, Griffin JL, Mehta NN, Shah R, Propert KJ, Song WL, Reilly MP. Omega-3 PUFA supplementation and the response to evoked endotoxemia in healthy volunteers. Mol Nutr Food Res. 2014;58:601–613. doi: 10.1002/mnfr.201300368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Vernochet C, Peres SB, Davis KE, McDonald ME, Qiang L, Wang H, Scherer PE, Farmer SR. C/EBPalpha and the corepressors CtBP1 and CtBP2 regulate repression of select visceral white adipose genes during induction of the brown phenotype in white adipocytes by peroxisome proliferator-activated receptor gamma agonists. Mol Cell Biol. 2009;29:4714–4728. doi: 10.1128/MCB.01899-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes [published correction appears in N Engl J Med. 2007;357:100] N Engl J Med. 2007;356:2457–2471. doi: 10.1056/NEJMoa072761. [DOI] [PubMed] [Google Scholar]
  • 91.Soccio RE, Chen ER, Lazar MA. Thiazolidinediones and the promise of insulin sensitization in type 2 diabetes. Cell Metab. 2014;20:573–591. doi: 10.1016/j.cmet.2014.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Heni M, Wagner R, Ketterer C, Böhm A, Linder K, Machicao F, Machann J, Schick F, Hennige AM, Stefan N, Häring HU, Fritsche A, Staiger H. Genetic variation in NR1H4 encoding the bile acid receptor FXR determines fasting glucose and free fatty acid levels in humans. J Clin Endocrinol Metab. 2013;98:E1224–E1229. doi: 10.1210/jc.2013-1177. [DOI] [PubMed] [Google Scholar]
  • 93.Zhang R, Wang Y, Li R, Chen G. Transcriptional factors mediating retinoic acid signals in the control of energy metabolism. Int J Mol Sci. 2015;16:14210–14244. doi: 10.3390/ijms160614210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Parikh M, Patel K, Soni S, Gandhi T. Liver X receptor: a cardinal target for atherosclerosis and beyond. J Atheroscler Thromb. 2014;21:519–531. [PubMed] [Google Scholar]
  • 95.Rankinen T, Sarzynski MA, Ghosh S, Bouchard C. Are there genetic paths common to obesity, cardiovascular disease outcomes, and cardiovascular risk factors? Circ Res. 2015;116:909–922. doi: 10.1161/CIRCRESAHA.116.302888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Schnabel RB, Baccarelli A, Lin H, Ellinor PT, Benjamin EJ. Next steps in cardiovascular disease genomic research: sequencing, epigenetics, and transcriptomics. Clin Chem. 2012;58:113–126. doi: 10.1373/clinchem.2011.170423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Churko JM, Mantalas GL, Snyder MP, Wu JC. Overview of high throughput sequencing technologies to elucidate molecular pathways in cardiovascular diseases. Circ Res. 2013;112:1613–1623. doi: 10.1161/CIRCRESAHA.113.300939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Bouchard-Mercier A, Paradis AM, Rudkowska I, Lemieux S, Couture P, Vohl MC. Associations between dietary patterns and gene expression profiles of healthy men and women: a cross-sectional study. Nutr J. 2013;12:24. doi: 10.1186/1475-2891-12-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Castañer O, Corella D, Covas MI, Sorlí JV, Subirana I, Flores-Mateo G, Nonell L, Bulló M, de la Torre R, Portolés O, Fitó M; PREDIMED Study Investigators. In vivo transcriptomic profile after a Mediterranean diet in high-cardiovascular risk patients: a randomized controlled trial. Am J Clin Nutr. 2013;98:845–853. doi: 10.3945/ajcn.113.060582. [DOI] [PubMed] [Google Scholar]
  • 100.Brattbakk HR, Arbo I, Aagaard S, Lindseth I, de Soysa AK, Langaas M, Kulseng B, Lindberg F, Johansen B. Balanced caloric macronutrient composition downregulates immunological gene expression in human blood cells-adipose tissue diverges. OMICS. 2013;17:41–52. doi: 10.1089/omi.2010.0124. [DOI] [PubMed] [Google Scholar]
  • 101.Franck N, Gummesson A, Jernås M, Glad C, Svensson PA, Guillot G, Rudemo M, Nyström FH, Carlsson LM, Olsson B. Identification of adipocyte genes regulated by caloric intake. J Clin Endocrinol Metab. 2011;96:E413–E418. doi: 10.1210/jc.2009-2534. [DOI] [PubMed] [Google Scholar]
  • 102.Vedin I, Cederholm T, Freund-Levi Y, Basun H, Garlind A, Irving GF, Eriksdotter-Jönhagen M, Wahlund LO, Dahlman I, Palmblad J. Effects of DHA-rich n-3 fatty acid supplementation on gene expression in blood mononuclear leukocytes: the OmegAD study. PLoS One. 2012;7:e35425. doi: 10.1371/journal.pone.0035425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rudkowska I, Paradis AM, Thifault E, Julien P, Tchernof A, Couture P, Lemieux S, Barbier O, Vohl MC. Transcriptomic and metabolomic signatures of an n-3 polyunsaturated fatty acids supplementation in a normolipidemic/normocholesterolemic Caucasian population. J Nutr Biochem. 2013;24:54–61. doi: 10.1016/j.jnutbio.2012.01.016. [DOI] [PubMed] [Google Scholar]
  • 104.Bouwens M, van de Rest O, Dellschaft N, Bromhaar MG, de Groot LC, Geleijnse JM, Müller M, Afman LA. Fish-oil supplementation induces antiinflammatory gene expression profiles in human blood mononuclear cells. Am J Clin Nutr. 2009;90:415–424. doi: 10.3945/ajcn.2009.27680. [DOI] [PubMed] [Google Scholar]
  • 105.Morine MJ, Tierney AC, van Ommen B, Daniel H, Toomey S, Gjelstad IM, Gormley IC, Pérez-Martinez P, Drevon CA, López-Miranda J, Roche HM. Transcriptomic coordination in the human metabolic network reveals links between n-3 fat intake, adipose tissue gene expression and metabolic health. PLoS Comput Biol. 2011;7:e1002223. doi: 10.1371/journal.pcbi.1002223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.van Dijk SJ, Feskens EJ, Bos MB, Hoelen DW, Heijligenberg R, Bromhaar MG, de Groot LC, de Vries JH, Müller M, Afman LA. A saturated fatty acid-rich diet induces an obesity-linked proinflammatory gene expression profile in adipose tissue of subjects at risk of metabolic syndrome. Am J Clin Nutr. 2009;90:1656–1664. doi: 10.3945/ajcn.2009.27792. [DOI] [PubMed] [Google Scholar]
  • 107.Rudkowska I, Jacques H, Weisnagel SJ, Marette A, Vohl MC. Transcriptomic profiles of skeletal muscle tissue following an euglycemic-hyperinsulinemic clamp in insulin-resistant obese subjects. Genes Nutr. 2013;8:91–98. doi: 10.1007/s12263-012-0298-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Rudkowska I, Paradis AM, Thifault E, Julien P, Barbier O, Couture P, Lemieux S, Vohl MC. Differences in metabolomic and transcriptomic profiles between responders and non-responders to an n-3 polyunsaturated fatty acids (PUFAs) supplementation. Genes Nutr. 2013;8:411–423. doi: 10.1007/s12263-012-0328-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Menche J, Sharma A, Kitsak M, Ghiassian SD, Vidal M, Loscalzo J, Barabási AL. Disease networks: uncovering disease-disease relationships through the incomplete interactome. Science. 2015;347:1257601. doi: 10.1126/science.1257601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Khalil AM, Guttman M, Huarte M, Garber M, Raj A, Rivea Morales D, Thomas K, Presser A, Bernstein BE, van Oudenaarden A, Regev A, Lander ES, Rinn JL. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc Natl Acad Sci USA. 2009;106:11667–11672. doi: 10.1073/pnas.0904715106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Cabili MN, Trapnell C, Goff L, Koziol M, Tazon-Vega B, Regev A, Rinn JL. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Genes Dev. 2011;25:1915–1927. doi: 10.1101/gad.17446611. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ulitsky I, Shkumatava A, Jan CH, Sive H, Bartel DP. Conserved function of lincRNAs in vertebrate embryonic development despite rapid sequence evolution [published correction appears in Cell. 2012;151:684–686] Cell. 2011;147:1537–1550. doi: 10.1016/j.cell.2011.11.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hangauer MJ, Vaughn IW, McManus MT. Pervasive transcription of the human genome produces thousands of previously unidentified long intergenic noncoding RNAs. PLoS Genet. 2013;9:e1003569. doi: 10.1371/journal.pgen.1003569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Vickers KC, Roteta LA, Hucheson-Dilks H, Han L, Guo Y. Mining diverse small RNA species in the deep transcriptome. Trends Biochem Sci. 2015;40:4–7. doi: 10.1016/j.tibs.2014.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zhang L, Hou D, Chen X, Li D, Zhu L, Zhang Y, Li J, Bian Z, Liang X, Cai X, Yin Y, Wang C, Zhang T, Zhu D, Zhang D, Xu J, Chen Q, Ba Y, Liu J, Wang Q, Chen J, Wang J, Wang M, Zhang Q, Zhang J, Zen K, Zhang CY. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: evidence of cross-kingdom regulation by microRNA [published correction appears in Cell Res. 2012;22:273–274] Cell Res. 2012;22:107–126. doi: 10.1038/cr.2011.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Liang H, Zen K, Zhang J, Zhang CY, Chen X. New roles for microRNAs in cross-species communication. RNA Biol. 2013;10:367–370. doi: 10.4161/rna.23663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Snow JW, Hale AE, Isaacs SK, Baggish AL, Chan SY. Ineffective delivery of diet-derived microRNAs to recipient animal organisms. RNA Biol. 2013;10:1107–1116. doi: 10.4161/rna.24909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Witwer KW, McAlexander MA, Queen SE, Adams RJ. Real-time quantitative PCR and droplet digital PCR for plant miRNAs in mammalian blood provide little evidence for general uptake of dietary miRNAs: limited evidence for general uptake of dietary plant xenomiRs. RNA Biol. 2013;10:1080–1086. doi: 10.4161/rna.25246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Dickinson B, Zhang Y, Petrick JS, Heck G, Ivashuta S, Marshall WS. Lack of detectable oral bioavailability of plant microRNAs after feeding in mice. Nat Biotechnol. 2013;31:965–967. doi: 10.1038/nbt.2737. [DOI] [PubMed] [Google Scholar]
  • 120.Zhang E, Wu Y. MicroRNAs: important modulators of oxLDL-mediated signaling in atherosclerosis. J Atheroscler Thromb. 2013;20:215–227. [DOI] [PubMed] [Google Scholar]
  • 121.Palmer JD, Soule BP, Simone BA, Zaorsky NG, Jin L, Simone NL. MicroRNA expression altered by diet: can food be medicinal? Ageing Res Rev. 2014;17:16–24. doi: 10.1016/j.arr.2014.04.005. [DOI] [PubMed] [Google Scholar]
  • 122.Yang WM, Jeong HJ, Park SY, Lee W. Induction of miR-29a by saturated fatty acids impairs insulin signaling and glucose uptake through translational repression of IRS-1 in myocytes. FEBS Lett. 2014;588:2170–2176. doi: 10.1016/j.febslet.2014.05.011. [DOI] [PubMed] [Google Scholar]
  • 123.Tomé-Carneiro J, Larrosa M, Yáñez-Gascón MJ, Dávalos A, Gil-Zamorano J, Gonzálvez M, García-Almagro FJ, Ruiz Ros JA, Tomás-Barberán FA, Espín JC, García-Conesa MT. One-year supplementation with a grape extract containing resveratrol modulates inflammatory-related microRNAs and cytokines expression in peripheral blood mononuclear cells of type 2 diabetes and hypertensive patients with coronary artery disease. Pharmacol Res. 2013;72:69–82. doi: 10.1016/j.phrs.2013.03.011. [DOI] [PubMed] [Google Scholar]
  • 124.Sun C, Burgner DP, Ponsonby AL, Saffery R, Huang RC, Vuillermin PJ, Cheung M, Craig JM. Effects of early-life environment and epigenetics on cardiovascular disease risk in children: highlighting the role of twin studies. Pediatr Res. 2013;73(pt 2):523–530. doi: 10.1038/pr.2013.6. [DOI] [PubMed] [Google Scholar]
  • 125.Keller TE, Yi SV. DNA methylation and evolution of duplicate genes. Proc Natl Acad Sci USA. 2014;111:5932–5937. doi: 10.1073/pnas.1321420111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Li J, Harris RA, Cheung SW, Coarfa C, Jeong M, Goodell MA, White LD, Patel A, Kang SH, Shaw C, Chinault AC, Gambin T, Gambin A, Lupski JR, Milosavljevic A. Genomic hypomethylation in the human germline associates with selective structural mutability in the human genome. PLoS Genet. 2012;8:e1002692. doi: 10.1371/journal.pgen.1002692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Weissmann F, Lyko F. Cooperative interactions between epigenetic modifications and their function in the regulation of chromosome architecture. Bioessays. 2003;25:792–797. doi: 10.1002/bies.10314. [DOI] [PubMed] [Google Scholar]
  • 128.Rose NR, Klose RJ. Understanding the relationship between DNA methylation and histone lysine methylation. Biochim Biophys Acta. 2014;1839:1362–1372. doi: 10.1016/j.bbagrm.2014.02.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Niculescu MD, Zeisel SH. Diet, methyl donors and DNA methylation: interactions between dietary folate, methionine and choline. J Nutr. 2002;132(suppl 1):2333S–2335S. [DOI] [PubMed] [Google Scholar]
  • 130.Wallwork JC, Duerre JA. Effect of zinc deficiency on methionine metabolism, methylation reactions and protein synthesis in isolated perfused rat liver. J Nutr. 1985;115:252–262. [DOI] [PubMed] [Google Scholar]
  • 131.Anderson OS, Sant KE, Dolinoy DC. Nutrition and epigenetics: an interplay of dietary methyl donors, one-carbon metabolism and DNA methylation. J Nutr Biochem. 2012;23:853–859. doi: 10.1016/j.jnutbio.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Dobosy JR, Fu VX, Desotelle JA, Srinivasan R, Kenowski ML, Almassi N, Weindruch R, Svaren J, Jarrard DF. A methyl-deficient diet modifies histone methylation and alters Igf2 and H19 repression in the prostate. Prostate. 2008;68:1187–1195. doi: 10.1002/pros.20782. [DOI] [PubMed] [Google Scholar]
  • 133.Tian X, Diaz FJ. Acute dietary zinc deficiency before conception compromises oocyte epigenetic programming and disrupts embryonic development. Dev Biol. 2013;376:51–61. doi: 10.1016/j.ydbio.2013.01.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Dolinoy DC, Das R, Weidman JR, Jirtle RL. Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr Res. 2007;61(pt 2):30R–37R. doi: 10.1203/pdr.0b013e31804575f7. [DOI] [PubMed] [Google Scholar]
  • 135.Maksimoska J, Segura-Peña D, Cole PA, Marmorstein R. Structure of the p300 histone acetyltransferase bound to acetyl-coenzyme A and its analogues. Biochemistry. 2014;53:3415–3422. doi: 10.1021/bi500380f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Galdieri L, Vancura A. Acetyl-CoA carboxylase regulates global histone acetylation. J Biol Chem. 2012;287:23865–23876. doi: 10.1074/jbc.M112.380519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Fan C, Dong H, Yan K, Shen W, Wang C, Xia L, Zhan D, Qi K. Genome-wide screen of promoter methylation identifies novel markers in diet-induced obese mice. Nutr Hosp. 2014;30:42–52. doi: 10.3305/nh.2014.30.1.7521. [DOI] [PubMed] [Google Scholar]
  • 138.Pheiffer C, Dias S, Muller C, Louw J. Decreased global DNA methylation in the white blood cells of high fat diet fed vervet monkeys (Chlorocebus aethiops). J Physiol Biochem. 2014;70:725–733. doi: 10.1007/s13105-014-0341-4. [DOI] [PubMed] [Google Scholar]
  • 139.Abi Khalil C The emerging role of epigenetics in cardiovascular disease. Ther Adv Chronic Dis. 2014;5:178–187. doi: 10.1177/2040622314529325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Udali S, Guarini P, Moruzzi S, Choi SW, Friso S. Cardiovascular epigenetics: from DNA methylation to microRNAs. Mol Aspects Med. 2013;34:883–901. doi: 10.1016/j.mam.2012.08.001. [DOI] [PubMed] [Google Scholar]
  • 141.Whayne TF. Epigenetics in the development, modification, and prevention of cardiovascular disease. Mol Biol Rep. 2015;42:765–776. doi: 10.1007/s11033-014-3727-z. [DOI] [PubMed] [Google Scholar]
  • 142.Zaina S, Heyn H, Carmona FJ, Varol N, Sayols S, Condom E, Ramírez-Ruz J, Gomez A, Gonçalves I, Moran S, Esteller M. DNA methylation map of human atherosclerosis. Circ Cardiovasc Genet. 2014;7:692–700. doi: 10.1161/CIRCGENETICS.113.000441. [DOI] [PubMed] [Google Scholar]
  • 143.Davies PF, Manduchi E, Stoeckert CJ, Jiménez JM, Jiang YZ. Emerging topic: flow-related epigenetic regulation of endothelial phenotype through DNA methylation. Vascul Pharmacol. 2014;62:88–93. doi: 10.1016/j.vph.2014.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Pandey D, Sikka G, Bergman Y, Kim JH, Ryoo S, Romer L, Berkowitz D. Transcriptional regulation of endothelial arginase 2 by histone deacetylase 2. Arterioscler Thromb Vasc Biol. 2014;34:1556–1566. doi: 10.1161/ATVBAHA.114.303685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Jiang YZ, Jiménez JM, Ou K, McCormick ME, Zhang LD, Davies PF. Hemodynamic disturbed flow induces differential DNA methylation of endothelial Kruppel-like factor 4 promoter in vitro and in vivo. Circ Res. 2014;115:32M–3. doi: 10.1161/CIRCRESAHA.115.303883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Maroski J, Vorderwulbecke BJ, Fiedorowicz K, Da Silva-Azevedo L, Siegel G, Marki A, Pries AR, Zakrzewicz A. Shear stress increases endothelial hyaluronan synthase 2 and hyaluronan synthesis especially in regard to an atheroprotective flow profile. Exp Physiol. 2011;96:977–986. doi: 10.1113/expphysiol.2010.056051. [DOI] [PubMed] [Google Scholar]
  • 147.Vigetti D, Deleonibus S, Moretto P, Bowen T, Fischer JW, Grandoch M, Oberhuber A, Love DC, Hanover JA, Cinquetti R, Karousou E, Viola M, D’Angelo ML, Hascall VC, De Luca G, Passi A. Natural antisense transcript for hyaluronan synthase 2 (HAS2-AS1) induces transcription of HAS2 via protein O-GlcNAcylation. J Biol Chem. 2014;289:28816–28826. doi: 10.1074/jbc.M114.597401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Jia SJ, Niu PP, Cong JZ, Zhang BK, Zhao M. TLR4 signaling: a potential therapeutic target in ischemic coronary artery disease. Int Immunopharmacol. 2014;23:54–59. doi: 10.1016/j.intimp.2014.08.011. [DOI] [PubMed] [Google Scholar]
  • 149.Khachatoorian R, Dawson D, Maloney EM, Wang J, French BA, French SW, French SW. SAMe treatment prevents the ethanol-induced epigenetic alterations of genes in the Toll-like receptor pathway. Exp Mol Pathol. 2013;94:243–246. doi: 10.1016/j.yexmp.2012.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Cao Q, Rong S, Repa JJ, St Clair R, Parks JS, Mishra N. Histone deacetylase 9 represses cholesterol efflux and alternatively activated macrophages in atherosclerosis development. Arterioscler Thromb Vasc Biol. 2014;34:1871–1879. doi: 10.1161/ATVBAHA.114.303393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Hoeksema MA, Gijbels MJ, Van den Bossche J, van der Velden S, Sijm A, Neele AE, Seijkens T, Stöger JL, Meiler S, Boshuizen MC, Dallinga-Thie GM, Levels JH, Boon L, Mullican SE, Spann NJ, Cleutjens JP, Glass CK, Lazar MA, de Vries CJ, Biessen EA, Daemen MJ, Lutgens E, de Winther MP. Targeting macrophage Histone deacetylase 3 stabilizes atherosclerotic lesions. EMBO Mol Med. 2014;6:1124–1132. doi: 10.15252/emmm.201404170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Oakes CC, Claus R, Gu L, Assenov Y, Hüllein J, Zucknick M, Bieg M, Brocks D, Bogatyrova O, Schmidt CR, Rassenti L, Kipps TJ, Mertens D, Lichter P, Döhner H, Stilgenbauer S, Byrd JC, Zenz T, Plass C. Evolution of DNA methylation is linked to genetic aberrations in chronic lymphocytic leukemia. Cancer Discov. 2014;4:348–361. doi: 10.1158/2159-8290.CD-13-0349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Saban KL, Mathews HL, DeVon HA, Janusek LW. Epigenetics and social context: implications for disparity in cardiovascular disease. Aging Dis. 2014;5:346–355. doi: 10.14336/AD.2014.0500346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lambrot R, Xu C, Saint-Phar S, Chountalos G, Cohen T, Paquet M, Suderman M, Hallett M, Kimmins S. Low paternal dietary folate alters the mouse sperm epigenome and is associated with negative pregnancy outcomes. Nat Commun. 2013;4:2889. doi: 10.1038/ncomms3889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Nilsson EE, Skinner MK. Environmentally induced epigenetic transgenerational inheritance of disease susceptibility. Transl Res. 2015;165:12–17. doi: 10.1016/j.trsl.2014.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Reik W, Dean W, Walter J. Epigenetic reprogramming in mammalian development. Science. 2001;293:1089–1093. doi: 10.1126/science.1063443. [DOI] [PubMed] [Google Scholar]
  • 157.Reik W, Walter J. Genomic imprinting: parental influence on the genome. Nat Rev Genet. 2001;2:21–32. doi: 10.1038/35047554. [DOI] [PubMed] [Google Scholar]
  • 158.Scott JM, Weir DG, Molloy A, McPartlin J, Daly L, Kirke P. Folic acid metabolism and mechanisms of neural tube defects. Ciba Found Symp. 1994;181:180–187. [DOI] [PubMed] [Google Scholar]
  • 159.Heijmans BT, Tobi EW, Stein AD, Putter H, Blauw GJ, Susser ES, Slagboom PE, Lumey LH. Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc Natl Acad Sci U S A. 2008;105:17046–17049. doi: 10.1073/pnas.0806560105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Roseboom T, de Rooij S, Painter R. The Dutch famine and its long-term consequences for adult health. Early Hum Dev. 2006;82:485–491. doi: 10.1016/j.earlhumdev.2006.07.001. [DOI] [PubMed] [Google Scholar]
  • 161.Casas E, Vavouri T. Sperm epigenomics: challenges and opportunities. Front Genet. 2014;5:330. doi: 10.3389/fgene.2014.00330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Martinez SR, Gay MS, Zhang L. Epigenetic mechanisms in heart development and disease. Drug Discov Today. 2015;20:799–811. doi: 10.1016/j.drudis.2014.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Dominguez-Salas P, Moore SE, Cole D, da Costa KA, Cox SE, Dyer RA, Fulford AJ, Innis SM, Waterland RA, Zeisel SH, Prentice AM, Hennig BJ. DNA methylation potential: dietary intake and blood concentrations of one-carbon metabolites and cofactors in rural African women. Am J Clin Nutr. 2013;97:1217–1227. doi: 10.3945/ajcn.112.048462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Bermingham EN, Bassett SA, Young W, Roy NC, McNabb WC, Cooney JM, Brewster DT, Laing WA, Barnett MP. Post-weaning selenium and folate supplementation affects gene and protein expression and global DNA methylation in mice fed high-fat diets. BMC Med Genomics. 2013;6:7. doi: 10.1186/1755-8794-6-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Radford EJ, Ito M, Shi H, Corish JA, Yamazawa K, Isganaitis E, Seisenberger S, Hore TA, Reik W, Erkek S, Peters AH, Patti ME, Ferguson-Smith AC. In utero effects: in utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science. 2014;345:1255903. doi: 10.1126/science.1255903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Starke RM, Ali MS, Jabbour PM, Tjoumakaris SI, Gonzalez F, Hasan DM, Rosenwasser RH, Owens GK, Koch WJ, Dumont AS. Cigarette smoke modulates vascular smooth muscle phenotype: implications for carotid and cerebrovascular disease. PLoS One. 2013;8:e71954. doi: 10.1371/journal.pone.0071954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Guay SP, Brisson D, Lamarche B, Marceau P, Vohl MC, Gaudet D, Bouchard L. DNA methylation variations at CETP and LPL gene promoter loci: new molecular biomarkers associated with blood lipid profile variability. Atherosclerosis. 2013;228:413–420. doi: 10.1016/j.atherosclerosis.2013.03.033. [DOI] [PubMed] [Google Scholar]
  • 168.Castro R, Rivera I, Martins C, Struys EA, Jansen EE, Clode N, Graç LM, Blom HJ, Jakobs C, de Almeida IT. Intracellular S-adenosylhomocysteine increased levels are associated with DNA hypomethylation in HUVEC. J Mol Med (Berl). 2005;83:831–836. doi: 10.1007/s00109-005-0679-8. [DOI] [PubMed] [Google Scholar]
  • 169.Cao Q, Mani RS, Ateeq B, Dhanasekaran SM, Asangani IA, Prensner JR, Kim JH, Brenner JC, Jing X, Cao X, Wang R, Li Y, Dahiya A, Wang L, Pandhi M, Lonigro RJ, Wu YM, Tomlins SA, Palanisamy N, Qin Z, Yu J, Maher CA, Varambally S, Chinnaiyan AM. Coordinated regulation of polycomb group complexes through microRNAs in cancer. Cancer Cell. 2011;20:187–199. doi: 10.1016/j.ccr.2011.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Kee HJ, Kwon JS, Shin S, Ahn Y, Jeong MH, Kook H. Trichostatin A prevents neointimal hyperplasia via activation of Kruppel like factor 4. Vascul Pharmacol. 2011;55:127–134. doi: 10.1016/j.vph.2011.07.001. [DOI] [PubMed] [Google Scholar]
  • 171.Hinkel R, Ng JK, Kupatt C. Targeting microRNAs for cardiovascular therapeutics in coronary artery disease. Curr Opin Cardiol. 2014;29:586–594. doi: 10.1097/HCO.0000000000000107. [DOI] [PubMed] [Google Scholar]
  • 172.Aslibekyan S, Claas SA, Arnett DK. Clinical applications of epigenetics in cardiovascular disease: the long road ahead. Transl Res. 2015;165:143–153. doi: 10.1016/j.trsl.2014.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Brennan L Metabolomics in nutrition research: current status and perspectives. Biochem Soc Trans. 2013;41:670–673. doi: 10.1042/BST20120350. [DOI] [PubMed] [Google Scholar]
  • 174.Townsend MK, Clish CB, Kraft P, Wu C, Souza AL, Deik AA, Tworoger SS, Wolpin BM. Reproducibility of metabolomic profiles among men and women in 2 large cohort studies. Clin Chem. 2013;59:1657–1667. doi: 10.1373/clinchem.2012.199133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Wang TJ, Larson MG, Vasan RS, Cheng S, Rhee EP, McCabe E, Lewis GD, Fox CS, Jacques PF, Fernandez C, O’Donnell CJ, Carr SA, Mootha VK, Florez JC, Souza A, Melander O, Clish CB, Gerszten RE. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–453. doi: 10.1038/nm.2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Wang TJ, Ngo D, Psychogios N, Dejam A, Larson MG, Vasan RS, Ghorbani A, O’Sullivan J, Cheng S, Rhee EP, Sinha S, McCabe E, Fox CS, O’Donnell CJ, Ho JE, Florez JC, Magnusson M, Pierce KA, Souza AL, Yu Y, Carter C, Light PE, Melander O, Clish CB, Gerszten RE. 2-Aminoadipic acid is a biomarker for diabetes risk. J Clin Invest. 2013;123:4309–4317. doi: 10.1172/JCI64801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Wu Y, Williams EG, Dubuis S, Mottis A, Jovaisaite V, Houten SM, Argmann CA, Faridi P, Wolski W, Kutalik Z, Zamboni N, Auwerx J, Aebersold R. Multilayered genetic and omics dissection of mitochondrial activity in a mouse reference population. Cell. 2014;158:1415–1430. doi: 10.1016/j.cell.2014.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Rhee EP, Ho JE, Chen MH, Shen D, Cheng S, Larson MG, Ghorbani A, Shi X, Helenius IT, O’Donnell CJ, Souza AL, Deik A, Pierce KA, Bullock K, Walford GA, Vasan RS, Florez JC, Clish C, Yeh JR, Wang TJ, Gerszten RE. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013;18:130–143. doi: 10.1016/j.cmet.2013.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Weir JM, Wong G, Barlow CK, Greeve MA, Kowalczyk A, Almasy L, Comuzzie AG, Mahaney MC, Jowett JB, Shaw J, Curran JE, Blangero J, Meikle PJ. Plasma lipid profiling in a large population-based cohort. J Lipid Res. 2013;54:2898–2908. doi: 10.1194/jlr.P035808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Wolf C, Quinn PJ. Lipidomics: practical aspects and applications. Prog Lipid Res. 2008;47:15–36. doi: 10.1016/j.plipres.2007.09.001. [DOI] [PubMed] [Google Scholar]
  • 181.Brown JM, Hazen SL. Seeking a unique lipid signature predicting cardiovascular disease risk. Circulation. 2014;129:1799–1803. doi: 10.1161/CIRCULATIONAHA.n4.009224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Rhee EP, Cheng S, Larson MG, Walford GA, Lewis GD, McCabe E, Yang E, Farrell L, Fox CS, O’Donnell CJ, Carr SA, Vasan RS, Florez JC, Clish CB, Wang TJ, Gerszten RE. Lipid profiling identifies a triacylglycerol signature of insulin resistance and improves diabetes prediction in humans. J Clin Invest. 2011;121:1402–1411. doi: 10.1172/JCI44442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Eckel RH, Jakicic JM, Ard JD, de Jesus JM, Houston Miller N, Hubbard VS, Lee IM, Lichtenstein AH, Loria CM, Millen BE, Nonas CA, Sacks FM, Smith SC Jr, Svetkey LP, Wadden TA, Yanovski SZ, Kendall KA, Morgan LC, Trisolini MG, Velasco G, Wnek J, Anderson JL, Halperin JL, Albert NM, Bozkurt B, Brindis RG, Curtis LH, DeMets D, Hochman JS, Kovacs RJ, Ohman EM, Pressler SJ, Sellke FW, Shen WK, Smith SC Jr, Tomaselli GF. 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines [published corrections appear in Circulation. 2014;129:S100-S101 and Circulation. 2015;131:e326] Circulation. 2014;129(suppl 2):S76–S99. doi: 10.1161/01.cir.0000437740.48606.d1. [DOI] [PubMed] [Google Scholar]
  • 184.Gleize B, Tourniaire F, Depezay L, Bott R, Nowicki M, Albino L, Lairon D, Kesse-Guyot E, Galan P, Hercberg S, Borel P. Effect of type of TAG fatty acids on lutein and zeaxanthin bioavailability. Br J Nutr. 2013;110:1–10. doi: 10.1017/S0007114512004813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Baker PR, Armando AM, Campbell JL, Quehenberger O, Dennis EA. Three-dimensional enhanced lipidomics analysis combining UPLC, differential ion mobility spectrometry, and mass spectrometric separation strategies. J Lipid Res. 2014;55:2432–2442. doi: 10.1194/jlr.D051581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Gross RW, Han X. Shotgun lipidomics of neutral lipids as an enabling technology for elucidation of lipid-related diseases. Am J Physiol Endocrinol Metab. 2009;297:E297–E303. doi: 10.1152/ajpendo.90970.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Masoodi M, Volmer DA. Comprehensive quantitative determination of PUFA-related bioactive lipids for functional lipidomics using high-resolution mass spectrometry. Methods Mol Biol. 2014;1198:221–232. doi: 10.1007/978-1-4939-1258-2_14. [DOI] [PubMed] [Google Scholar]
  • 188.Stübiger G, Aldover-Macasaet E, Bicker W, Sobal G, Willfort-Ehringer A, Pock K, Bochkov V, Widhalm K, Belgacem O. Targeted profiling of atherogenic phospholipids in human plasma and lipoproteins of hyperlipidemic patients using MALDI-QIT-TOF-MS/MS. Atherosclerosis. 2012;224:177–186. doi: 10.1016/j.atherosclerosis.2012.06.010. [DOI] [PubMed] [Google Scholar]
  • 189.Stegemann C, Pechlaner R, Willeit P, Langley SR, Mangino M, Mayr U, Menni C, Moayyeri A, Santer P, Rungger G, Spector TD, Willeit J, Kiechl S, Mayr M. Lipidomics profiling and risk of cardiovascular disease in the prospective population-based Bruneck study. Circulation. 2014;129:1821–1831. doi: 10.1161/CIRCULATIONAHA.113.002500. [DOI] [PubMed] [Google Scholar]
  • 190.Meikle PJ, Wong G, Barlow CK, Weir JM, Greeve MA, MacIntosh GL, Almasy L, Comuzzie AG, Mahaney MC, Kowalczyk A, Haviv I, Grantham N, Magliano DJ, Jowett JB, Zimmet P, Curran JE, Blangero J, Shaw J. Plasma lipid profiling shows similar associations with prediabetes and type 2 diabetes. PLoS One. 2013;8:e74341. doi: 10.1371/journal.pone.0074341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Sigruener A, Kleber ME, Heimerl S, Liebisch G, Schmitz G, Maerz W. Glycerophospholipid and sphingolipid species and mortality: the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. PLoS One. 2014;9:e85724. doi: 10.1371/journal.pone.0085724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Kostara CE, Papathanasiou A, Psychogios N, Cung MT, Elisaf MS, Goudevenos J, Bairaktari ET. NMR-based lipidomic analysis of blood lipoproteins differentiates the progression of coronary heart disease. J Proteome Res. 2014;13:2585–2598. doi: 10.1021/pr500061n. [DOI] [PubMed] [Google Scholar]
  • 193.Williams PT, Zhao XQ, Marcovina SM, Otvos JD, Brown BG, Krauss RM. Comparison of four methods of analysis of lipoprotein particle subfractions for their association with angiographic progression of coronary artery disease. Atherosclerosis. 2014;233:713–720. doi: 10.1016/j.atherosclerosis.2014.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Ban RH, Kamvissi V, Schulte KM, Bornstein SR, Rubino F, Graessler J. Lipidomic profiling at the interface of metabolic surgery and cardiovascular disease. Curr Atheroscler Rep. 2014;16:455. doi: 10.1007/s11883-014-0455-8. [DOI] [PubMed] [Google Scholar]
  • 195.Hu C, Kong H, Qu F, Li Y, Yu Z, Gao P, Peng S, Xu G. Application of plasma lipidomics in studying the response of patients with essential hypertension to antihypertensive drug therapy. Mol Biosyst. 2011;7:3271–3279. doi: 10.1039/c1mb05342f. [DOI] [PubMed] [Google Scholar]
  • 196.Magdeldin S, Enany S, Yoshida Y, Xu B, Zhang Y, Zureena Z, Lokamani I, Yaoita E, Yamamoto T. Basics and recent advances of two dimensional-polyacrylamide gel electrophoresis. Clin Proteomics. 2014;11:16. doi: 10.1186/1559-0275-11-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Thaysen-Andersen M, Packer NH. Advances in LC-MS/MS-based glycoproteomics: getting closer to system-wide site-specific mapping of the N- and O-glycoproteome. Biochim Biophys Acta. 2014;1844:1437–1452. doi: 10.1016/j.bbapap.2014.05.002. [DOI] [PubMed] [Google Scholar]
  • 198.Gold L, Ayers D, Bertino J, Bock C, Bock A, Brody EN, Carter J, Dalby AB, Eaton BE, Fitzwater T, Flather D, Forbes A, Foreman T, Fowler C, Gawande B, Goss M, Gunn M, Gupta S, Halladay D, Heil J, Heilig J, Hicke B, Husar G, Janjic N, Jarvis T, Jennings S, Katilius E, Keeney TR, Kim N, Koch TH, Kraemer S, Kroiss L, Le N, Levine D, Lindsey W, Lollo B, Mayfield W, Mehan M, Mehler R, Nelson SK, Nelson M, Nieuwlandt D, Nikrad M, Ochsner U, Ostroff RM, Otis M, Parker T, Pietrasiewicz S, Resnicow DI, Rohloff J, Sanders G, Sattin S, Schneider D, Singer B, Stanton M, Sterkel A, Stewart A, Stratford S, Vaught JD, Vrkljan M, Walker JJ, Watrobka M, Waugh S, Weiss A, Wilcox SK, Wolfson A, Wolk SK, Zhang C, Zichi D. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS One. 2010;5:e15004. doi: 10.1371/journal.pone.0015004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Napoli C, Zullo A, Picascia A, Infante T, Mancini FP. Recent advances in proteomic technologies applied to cardiovascular disease. J Cell Biochem. 2013;114:7–20. doi: 10.1002/jcb.24307. [DOI] [PubMed] [Google Scholar]
  • 200.Barallobre-Barreiro J, Chung YL, Mayr M. Proteomics and metabolomics for mechanistic insights and biomarker discovery in cardiovascular disease. Rev Esp Cardiol (Engl Ed). 2013;66:657–661. doi: 10.1016/j.rec.2013.04.009. [DOI] [PubMed] [Google Scholar]
  • 201.Langley SR, Dwyer J, Drozdov I, Yin X, Mayr M. Proteomics: from single molecules to biological pathways. Cardiovasc Res. 2013;97:612–622. doi: 10.1093/cvr/cvs346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Delles C, Diez J, Dominiczak AF. Urinary proteomics in cardiovascular disease: achievements, limits and hopes. Proteomics Clin Appl. 2011;5:222–232. doi: 10.1002/prca.201000125. [DOI] [PubMed] [Google Scholar]
  • 203.Cammarato A, Ahrens CH, Alayari NN, Qeli E, Rucker J, Reedy MC, Zmasek CM, Gucek M, Cole RN, Van Eyk JE, Bodmer R, O’Rourke B, Bernstein SI, Foster DB. A mighty small heart: the cardiac proteome of adult Drosophila melanogaster. PLoS One. 2011;6:e18497. doi: 10.1371/journal.pone.0018497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Bleijerveld OB, Zhang YN, Beldar S, Hoefer IE, Sze SK, Pasterkamp G, de Kleijn DP. Proteomics of plaques and novel sources of potential biomarkers for atherosclerosis. Proteomics Clin Appl. 2013;7:490–503. doi: 10.1002/prca.201200119. [DOI] [PubMed] [Google Scholar]
  • 205.Birner-Gruenberger R, Schittmayer M, Holzer M, Marsche G. Understanding high-density lipoprotein function in disease: recent advances in proteomics unravel the complexity of its composition and biology. Prog Lipid Res. 2014;56:36–46. doi: 10.1016/j.plipres.2014.07.003. [DOI] [PubMed] [Google Scholar]
  • 206.Drost JT, Maas AH, Holewijn S, Joosten LA, van Eyck J, van der Schouw YT, de Graaf J. Novel cardiovascular biomarkers in women with a history of early preeclampsia. Atherosclerosis. 2014;237:117–122. doi: 10.1016/j.atherosclerosis.2014.09.009. [DOI] [PubMed] [Google Scholar]
  • 207.de Roos B Nutrition proteomics and biomarker discovery. Expert Rev Proteomics. 2009;6:349–351. doi: 10.1586/epr.09.52. [DOI] [PubMed] [Google Scholar]
  • 208.de Roos B, Geelen A, Ross K, Rucklidge G, Reid M, Duncan G, Caslake M, Horgan G, Brouwer IA. Identification of potential serum biomarkers of inflammation and lipid modulation that are altered by fish oil supplementation in healthy volunteers. Proteomics. 2008;8:1965–1974. doi: 10.1002/pmic.200700457. [DOI] [PubMed] [Google Scholar]
  • 209.Ganesh V, Hettiarachchy NS. Nutriproteomics: a promising tool to link diet and diseases in nutritional research. Biochim Biophys Acta. 2012;1824:1107–1117. doi: 10.1016/j.bbapap.2012.06.006. [DOI] [PubMed] [Google Scholar]
  • 210.Tuteja S, Rader DJ. High-density lipoproteins in the prevention of cardiovascular disease: changing the paradigm. Clin Pharmacol Ther. 2014;96:48–56. doi: 10.1038/clpt.2014.79. [DOI] [PubMed] [Google Scholar]
  • 211.Larsen N, Vogensen FK, van den Berg FW, Nielsen DS, Andreasen AS, Pedersen BK, Al-Soud WA, Sørensen SJ, Hansen LH, Jakobsen M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5:e9085. doi: 10.1371/journal.pone.0009085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Turnbaugh PJ, Bäckhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–223. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Karlsson FH, Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, Bäckhed F, Nielsen J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun. 2012;3:1245. doi: 10.1038/ncomms2266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Robles Alonso V, Guarner F. Linking the gut microbiota to human health. Br J Nutr. 2013;109(suppl 2):S21–S26. doi: 10.1017/S0007114512005235. [DOI] [PubMed] [Google Scholar]
  • 215.Vrieze A, Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JF, Dallinga-Thie GM, Ackermans MT, Serlie MJ, Oozeer R, Derrien M, Druesne A, Van Hylckama Vlieg JE, Bloks VW, Groen AK, Heilig HG, Zoetendal EG, Stroes ES, de Vos WM, Hoekstra JB, Nieuwdorp M. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome [published correction appears in Gastroenterology. 2013; 144:250] Gastroenterology. 2012;143:913–916.e7. doi: 10.1053/j.gastro.2012.06.031. [DOI] [PubMed] [Google Scholar]
  • 216.Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K, Hayashi DK, Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, Walters WA, Knight R, Newgard CB, Heath AC, Gordon JI. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013. ;341:1241214. doi: 10.1126/science.1241214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Vijay-Kumar M, Aitken JD, Carvalho FA, Cullender TC, Mwangi S, Srinivasan S, Sitaraman SV, Knight R, Ley RE, Gewirtz AT. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 2010;328:228–231. doi: 10.1126/science.1179721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470–1481. doi: 10.2337/db07-1403. [DOI] [PubMed] [Google Scholar]
  • 219.Cani PD, Possemiers S, Van de Wiele T, Guiot Y, Everard A, Rottier O, Geurts L, Naslain D, Neyrinck A, Lambert DM, Muccioli GG, Delzenne NM. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut. 2009;58:1091–1103. doi: 10.1136/gut.2008.165886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Ostos MA, Recalde D, Zakin MM, Scott-Algara D. Implication of natural killer T cells in atherosclerosis development during a LPS-induced chronic inflammation. FEBS Lett. 2002;519:23–29. [DOI] [PubMed] [Google Scholar]
  • 221.Gorjão R, Takahashi HK, Pan JA, Massao Hirabara S. Molecular mechanisms involved in inflammation and insulin resistance in chronic diseases and possible interventions. J Biomed Biotechnol. 2012;2012:841983. doi: 10.1155/2012/841983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874. doi: 10.1038/nature01323. [DOI] [PubMed] [Google Scholar]
  • 223.Romeo GR, Lee J, Shoelson SE. Metabolic syndrome, insulin resistance, and roles of inflammation: mechanisms and therapeutic targets. Arterioscler Thromb Vasc Biol. 2012;32:1771–1776. doi: 10.1161/ATVBAHA.111.241869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Glass CK, Olefsky JM. Inflammation and lipid signaling in the etiology of insulin resistance. Cell Metab. 2012;15:635–645. doi: 10.1016/j.cmet.2012.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.McKnite AM, Perez-Munoz ME, Lu L, Williams EG, Brewer S, Andreux PA, Bastiaansen JW, Wang X, Kachman SD, Auwerx J, Williams RW, Benson AK, Peterson DA, Ciobanu DC. Murine gut microbiota is defined by host genetics and modulates variation of metabolic traits. PLoS One. 2012;7:e39191. doi: 10.1371/journal.pone.0039191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Benson AK, Kelly SA, Legge R, Ma F, Low SJ, Kim J, Zhang M, Oh PL, Nehrenberg D, Hua K, Kachman SD, Moriyama EN, Walter J, Peterson DA, Pomp D. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc Natl Acad Sci USA. 2010;107:18933–18938. doi: 10.1073/pnas.1007028107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Srinivas G, Möller S, Wang J, Kunzel S, Zillikens D, Baines JF, Ibrahim SM. Genome-wide mapping of gene-microbiota interactions in susceptibility to autoimmune skin blistering. Nat Commun. 2013;4:2462. doi: 10.1038/ncomms3462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA, Gordon JI, Chervonsky AV. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature. 2008;455:1109–1113. doi: 10.1038/nature07336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Thompson CL, Hofer MJ, Campbell IL, Holmes AJ. Community dynamics in the mouse gut microbiota: a possible role for IRF9-regulated genes in community homeostasis. PLoS One. 2010;5:e10335. doi: 10.1371/journal.pone.0010335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Hildebrand F, Nguyen TL, Brinkman B, Yunta RG, Cauwe B, Vandenabeele P, Liston A, Raes J. Inflammation-associated enterotypes, host genotype, cage and inter-individual effects drive gut microbiota variation in common laboratory mice. Genome Biol. 2013;14:R4. doi: 10.1186/gb-2013-14-1-r4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Schwab C, Berry D, Rauch I, Rennisch I, Ramesmayer J, Hainzl E, Heider S, Decker T, Kenner L, Muller M, Strobl B, Wagner M, Schleper C, Loy A, Urich T. Longitudinal study of murine microbiota activity and interactions with the host during acute inflammation and recovery. ISME J. 2014;8:1101–1114. doi: 10.1038/ismej.2013.223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Eckburg PB, Bik EM, Bernstein CN, Purdom E, Dethlefsen L, Sargent M, Gill SR, Nelson KE, Relman DA. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638. doi: 10.1126/science.1110591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu H, Yu C, Li S, Jian M, Zhou Y, Li Y, Zhang X, Li S, Qin N, Yang H, Wang J, Brunak S, Dore J, Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J; MetaHIT Consortium, Bork P, Ehrlich SD, Wang J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59–65. doi: 10.1038/nature08821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Turnbaugh PJ, Quince C, Faith JJ, McHardy AC, Yatsunenko T, Niazi F, Affourtit J, Egholm M, Henrissat B, Knight R, Gordon JI. Organismal, genetic, and transcriptional variation in the deeply sequenced gut microbiomes of identical twins. Proc Natl Acad Sci USA. 2010;107:7503–7508. doi: 10.1073/pnas.1002355107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Tims S, Derom C, Jonkers DM, Vlietinck R, Saris WH, Kleerebezem M, de Vos WM, Zoetendal EG. Microbiota conservation and BMI signatures in adult monozygotic twins. ISME J. 2013;7:707–717. doi: 10.1038/ismej.2012.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–484. doi: 10.1038/nature07540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Parks BW, Nam E, Org E, Kostem E, Norheim F, Hui ST, Pan C, Civelek M, Rau CD, Bennett BJ, Mehrabian M, Ursell LK, He A, Castellani LW, Zinker B, Kirby M, Drake TA, Drevon CA, Knight R, Gargalovic P, Kirchgessner T, Eskin E, Lusis AJ. Genetic control of obesity and gut microbiota composition in response to high-fat, high-sucrose diet in mice. Cell Metab. 2013;17:141–152. doi: 10.1016/j.cmet.2012.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Kovacs A, Ben-Jacob N, Tayem H, Halperin E, Iraqi FA, Gophna U. Genotype is a stronger determinant than sex of the mouse gut microbiota. Microb Ecol. 2011;61:423–428. doi: 10.1007/s00248-010-9787-2. [DOI] [PubMed] [Google Scholar]
  • 239.Spor A, Koren O, Ley R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat Rev Microbiol. 2011;9:279–290. doi: 10.1038/nrmicro2540. [DOI] [PubMed] [Google Scholar]
  • 240.Toivanen P, Vaahtovuo J, Eerola E. Influence of major histocompatibility complex on bacterial composition of fecal flora. Infect Immun. 2001;69:2372–2377. doi: 10.1128/IAI.69.4.2372-2377.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Campbell JH, Foster CM, Vishnivetskaya T, Campbell AG, Yang ZK, Wymore A, Palumbo AV, Chesler EJ, Podar M. Host genetic and environmental effects on mouse intestinal microbiota. ISME J. 2012;6:2033–2044. doi: 10.1038/ismej.2012.54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.O’Connor A, Quizon PM, Albright JE, Lin FT, Bennett BJ. Responsiveness of cardiometabolic-related microbiota to diet is influenced by host genetics. Mamm Genome. 2014;25:583–599. doi: 10.1007/s00335-014-9540-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Friswell MK, Gika H, Stratford IJ, Theodoridis G, Telfer B, Wilson ID, McBain AJ. Site and strain-specific variation in gut microbiota profiles and metabolism in experimental mice. PLoS One. 2010;5:e8584. doi: 10.1371/journal.pone.0008584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.McCafferty J, Mühlbauer M, Gharaibeh RZ, Arthur JC, Perez-Chanona E, Sha W, Jobin C, Fodor AA. Stochastic changes over time and not founder effects drive cage effects in microbial community assembly in a mouse model. ISME J. 2013;7:2116–2125. doi: 10.1038/ismej.2013.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Seedorf H, Griffin NW, Ridaura VK, Reyes A, Cheng J, Rey FE, Smith MI, Simon GM, Scheffrahn RH, Woebken D, Spormann AM, Van Treuren W, Ursell LK, Pirrung M, Robbins-Pianka A, Cantarel BL, Lombard V, Henrissat B, Knight R, Gordon JI. Bacteria from diverse habitats colonize and compete in the mouse gut. Cell. 2014;159:253–266. doi: 10.1016/j.cell.2014.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, Almeida M, Quinquis B, Levenez F, Galleron N, Gougis S, Rizkalla S, Batto JM, Renault P; ANR MicroObes Consortium, Doré J, Zucker JD, Clément K, Ehrlich SD. Dietary intervention impact on gut microbial gene richness [published correction appears in Nature. 2013;502:580] Nature. 2013;500:585–588. doi: 10.1038/nature12480. [DOI] [PubMed] [Google Scholar]
  • 247.Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG, Li H, Gao Z, Mahana D, Zárate Rodriguez JG, Rogers AB, Robine N, Loke P, Blaser MJ. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158:705–721. doi: 10.1016/j.cell.2014.05.052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Joseph J, Loscalzo J. Nutri(meta)genetics and cardiovascular disease: novel concepts in the interaction of diet and genomic variation. Curr Atheroscler Rep. 2015;17:505. doi: 10.1007/s11883-015-0505-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Kashyap PC, Marcobal A, Ur sell LK, Smits SA, Sonnenburg ED, Costello EK, Higginbottom SK, Domino SE, Holmes SP, Relman DA, Knight R, Gordon JI, Sonnenburg JL. Genetically dictated change in host mucus carbohydrate landscape exerts a diet-dependent effect on the gut microbiota. Proc Natl Acad Sci USA. 2013;110:17059–17064. doi: 10.1073/pnas.1306070110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–563. doi: 10.1038/nature12820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ, Hazen SL. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–585. doi: 10.1038/nm.3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–1584. doi: 10.1056/NEJMoa1109400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Tang WH, Wang Z, Shrestha K, Borowski AG, Wu Y, Troughton RW, Klein AL, Hazen SL. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J Card Fail. 2015;21:91–96. doi: 10.1016/j.cardfail.2014.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Trøseid M, Ueland T, Hov JR, Svardal A, Gregersen I, Dahl CP, Aakhus S, Gude E, Bjørndal B, Halvorsen B, Karlsen TH, Aukrust P, Gullestad L, Berge RK, Yndestad A. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J Intern Med. 2015;277:717–726. doi: 10.1111/joim.12328. [DOI] [PubMed] [Google Scholar]
  • 256.Lever M, George PM, Slow S, Bellamy D, Young JM, Ho M, McEntyre CJ, Elmslie JL, Atkinson W, Molyneux SL, Troughton RW, Frampton CM, Richards AM, Chambers ST. Betaine and trimethylamine-N-oxide as predictors of cardiovascular outcomes show different patterns in diabetes mellitus: an observational study. PLoS One. 2014;9:e114969. doi: 10.1371/journal.pone.0114969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, Li XS, Levison BS, Hazen SL. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res. 2015;116:448–455. doi: 10.1161/CIRCRESAHA.116.305360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Bennett BJ, de Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, Allayee H, Lee R, Graham M, Crooke R, Edwards PA, Hazen SL, Lusis AJ. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013;17:49–60. doi: 10.1016/j.cmet.2012.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Shih DM, Wang Z, Lee R, Meng Y, Che N, Charugundla S, Qi H, Wu J, Pan C, Brown JM, Vallim T, Bennett BJ, Graham M, Hazen SL, Lusis AJ. Flavin containing monooxygenase 3 exerts broad effects on glucose and lipid metabolism and atherosclerosis. J Lipid Res. 2015;56:22–37. doi: 10.1194/jlr.M051680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260.Warrier M, Shih DM, Burrows AC, Ferguson D, Gromovsky AD, Brown AL, Marshall S, McDaniel A, Schugar RC, Wang Z, Sacks J, Rong X, Vallim TA, Chou J, Ivanova PT, Myers DS, Brown HA, Lee RG, Crooke RM, Graham MJ, Liu X, Parini P, Tontonoz P, Lusis AJ, Hazen SL, Temel RE, Brown JM. The TMAO-generating enzyme flavin monooxygenase 3 is a central regulator of cholesterol balance. Cell Rep. 2015:10:326–338. doi: 10.1016/j.celrep.2014.12.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A. 2012;109:21307–21312. doi: 10.1073/pnas.1215689109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Hartiala J, Bennett BJ, Tang WH, Wang Z, Stewart AF, Roberts R, McPherson R, Lusis AJ, Hazen SL, Allayee H; CARDIoGRAM Consortium. Comparative genome-wide association studies in mice and humans for trimethylamine N-oxide, a proatherogenic metabolite of choline and L-carnitine. Arterioscler Thromb Vasc Biol. 2014;34:1307–1313. doi: 10.1161/ATVBAHA.114.303252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, Wagner MA, Bennett BJ, Li L, DiDonato JA, Lusis AJ, Hazen SL. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem. 2015;290:5647–5660. doi: 10.1074/jbc.M114.618249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.AlipourA Elte JW, van Zaanen HC, Rietveld AP, Castro Cabezas M. Novel aspects of postprandial lipemia in relation to atherosclerosis. Atheroscler Suppl. 2008;9:39–44. doi: 10.1016/j.atherosclerosissup.2008.05.007. [DOI] [PubMed] [Google Scholar]
  • 265.van Oostrom AJ, Sijmonsma TP, Verseyden C, Jansen EH, de Koning EJ, Rabelink TJ, Castro Cabezas M. Postprandial recruitment of neutrophils may contribute to endothelial dysfunction. J Lipid Res. 2003;44:576–583. doi: 10.1194/jlr.M200419-JLR200. [DOI] [PubMed] [Google Scholar]
  • 266.van Ommen B, van der Greef J, Ordovas JM, Daniel H. Phenotypic flexibility as key factor in the human nutrition and health relationship. Genes Nutr. 2014;9:423. doi: 10.1007/s12263-014-0423-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Giacco R, Cuomo V, Vessby B, Uusitupa M, Hermansen K, Meyer BJ, Riccardi G, Rivellese AA; KANWU Study Group. Fish oil, insulin sensitivity, insulin secretion and glucose tolerance in healthy people: is there any effect of fish oil supplementation in relation to the type of background diet and habitual dietary intake of n-6 and n-3 fatty acids? Nutr Metab Cardiovasc Dis. 2007;17:572–580. doi: 10.1016/j.numecd.2006.06.006. [DOI] [PubMed] [Google Scholar]
  • 268.Tierney AC, McMonagle J, Shaw DI, Gulseth HL, Helal O, Saris WH, Paniagua JA, Gołąbek-Leszczyñska I, Defoort C, Williams CM, Karsltröm B, Vessby B, Dembinska-Kiec A, López-Miranda J, Blaak EE, Drevon CA, Gibney MJ, Lovegrove JA, Roche HM. Effects of dietary fat modification on insulin sensitivity and on other risk factors of the metabolic syndrome: LIPGENE: a European randomized dietary intervention study. Int J Obes (Lond). 2011;35:800–809. doi: 10.1038/ijo.2010.209. [DOI] [PubMed] [Google Scholar]
  • 269.Luther JM, Byrne LM, Yu C, Wang TJ, Brown NJ. Dietary sodium restriction decreases insulin secretion without affecting insulin sensitivity in humans. J Clin Endocrinol Metab. 2014;99:E1895–E1902. doi: 10.1210/jc.2014-2122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Arora P, Wu C, Khan AM, Bloch DB, Davis-Dusenbery BN, Ghorbani A, Spagnolli E, Martinez A, Ryan A, Tainsh LT, Kim S, Rong J, Huan T, Freedman JE, Levy D, Miller KK, Hata A, Del Monte F, Vandenwijngaert S, Swinnen M, Janssens S, Holmes TM, Buys ES, Bloch KD, Newton-Cheh C, Wang TJ. Atrial natriuretic peptide is negatively regulated by microRNA-425. J Clin Invest. 2013;123:3378–3382. doi: 10.1172/JCI67383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Heni M, Ketterer C, Wagner R, Linder K, Böhm A, Herzberg-Schäfer SA, Machicao F, Knoch KP, Fritsche A, Staiger H, Haring HU, Solimena M. Polymorphism rs11085226 in the gene encoding polypyrimidine tractbinding protein 1 negatively affects glucose-stimulated insulin secretion. PLoS One. 2012;7:e46154. doi: 10.1371/journal.pone.0046154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Perez-Martinez P, Delgado-Lista J, Garcia-Rios A, Ferguson JF, Gulseth HL, Williams CM, Karlström B, Kieć-Wilk B, Blaak EE, Helal O, Malczewska-Malec M, Defoort C, Risérus U, Saris WH, Lovegrove JA, Drevon CA, Roche HM, Lopez-Miranda J. Calpain-10 interacts with plasma saturated fatty acid concentrations to influence insulin resistance in individuals with the metabolic syndrome. Am J Clin Nutr. 2011;93:1136–1141. doi: 10.3945/ajcn.110.010512. [DOI] [PubMed] [Google Scholar]
  • 273.An P, Straka RJ, Pollin TI, Feitosa MF, Wojczynski MK, Daw EW, O’Connell JR, Gibson Q, Ryan KA, Hopkins PN, Tsai MY, Lai CQ, Province MA, Ordovas JM, Shuldiner AR, Arnett DK, Borecki IB. Genome-wide association studies identified novel loci for non-high-density lipoprotein cholesterol and its postprandial lipemic response. Hum Genet. 2014;133:919–930. doi: 10.1007/s00439-014-1435-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Bouwens M, Grootte Bromhaar M, Jansen J, Müller M, Afman LA. Postprandial dietary lipid-specific effects on human peripheral blood mononuclear cell gene expression profiles. Am J Clin Nutr. 2010;91:208–217. doi: 10.3945/ajcn.2009.28586. [DOI] [PubMed] [Google Scholar]
  • 275.Krug S, Kastenmüller G, Stückler F, Rist MJ, Skurk T, Sailer M, Raffler J, Römisch-Margl W, Adamski J, Prehn C, Frank T, Engel KH, Hofmann T, Luy B, Zimmermann R, Moritz F, Schmitt-Kopplin P, Krumsiek J, Kremer W, Huber F, Oeh U, Theis FJ, Szymczak W, Hauner H, Suhre K, Daniel H. The dynamic range of the human metabolome revealed by challenges. FASEB J. 2012;26:2607–2619. doi: 10.1096/fj.11-198093. [DOI] [PubMed] [Google Scholar]
  • 276.Pellis L, van Erk MJ, van Ommen B, Bakker GC, Hendriks HF, Cnubben NH, Kleemann R, van Someren EP, Bobeldijk I, Rubingh CM, Wopereis S. Plasma metabolomics and proteomics profiling after a postprandial challenge reveal subtle diet effects on human metabolic status. Metabolomics. 2012;8:347–359. doi: 10.1007/s11306-011-0320-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Dejeans N, Herosimczyk A, Sayd T, Chambon C, Martin JF, Maier JA, Tauveron I, Mazur A. Effect of a high-fat challenge on the proteome of human postprandial plasma. ClinNutr. 2013;32:468–471. doi: 10.1016/j.clnu.2012.05.017. [DOI] [PubMed] [Google Scholar]
  • 278.Irvin MR, Zhi D, Aslibekyan S, Claas SA, Absher DM, Ordovas JM, Tiwari HK, Watkins S, Arnett DK. Genomics of post-prandial lipidomic phenotypes in the Genetics of Lipid lowering Drugs and Diet Network (GOLDN) study. PLoS One. 2014;9:e99509. doi: 10.1371/journal.pone.0099509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Ferguson JF, Patel PN, Shah RY, Mulvey CK, Gadi R, Nijjar PS, Usman HM, Mehta NN, Shah R, Master SR, Propert KJ, Reilly MP. Race and gender variation in response to evoked inflammation. J Transl Med. 2013;11:63. doi: 10.1186/1479-5876-11-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Mehta NN, Heffron SP, Patel PN, Ferguson J, Shah RD, Hinkle CC, Krishnamoorthy P, Shah R, Tabita-Martinez J, Terembula K, Master SR, Rickels MR, Reilly MP. A human model of inflammatory cardio-metabolic dysfunction; a double blind placebo-controlled crossover trial. J Transl Med. 2012;10:124. doi: 10.1186/1479-5876-10-124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Suffredini AF, Fromm RE, Parker MM, Brenner M, Kovacs JA, Wesley RA, Parrillo JE. The cardiovascular response of normal humans to the administration of endotoxin. N Engl J Med. 1989;321:280–287. doi: 10.1056/NEJM198908033210503. [DOI] [PubMed] [Google Scholar]
  • 282.Patel PN, Shah RY, Ferguson JF, Reilly MP. Human experimental endotoxemia in modeling the pathophysiology, genomics, and therapeutics of innate immunity in complex cardiometabolic diseases. Arterioscler Thromb Vasc Biol. 2015;35:525–534. doi: 10.1161/ATVBAHA.114.304455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Shah R, Lu Y, Hinkle CC, McGillicuddy FC, Kim R, Hannenhalli S, Cappola TP, Heffron S, Wang X, Mehta NN, Putt M, Reilly MP. Gene profiling of human adipose tissue during evoked inflammation in vivo. Diabetes. 2009;58:2211–2219. doi: 10.2337/db09-0256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Liu Y, Ferguson JF, Xue C, Ballantyne RL, Silverman IM, Gosai SJ, Serfecz J, Morley MP, Gregory BD, Li M, Reilly MP. Tissue-specific RNA-Seq in human evoked inflammation identifies blood and adipose LincRNA signatures of cardiometabolic diseases. Arterioscler Thromb Vasc Biol. 2014;34:902–912. doi: 10.1161/ATVBAHA.113.303123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Ferguson JF, Ryan MF, Gibney ER, Brennan L, Roche HM, Reilly MP. Dietary isoflavone intake is associated with evoked responses to inflammatory cardiometabolic stimuli and improved glucose homeostasis in healthy volunteers. Nutr Metab Cardiovasc Dis. 2014;24:996–1003. doi: 10.1016/j.numecd.2014.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Lubbers T, Kox M, de Haan JJ, Greve JW, Pompe JC, Ramakers BP, Pickkers P, Buurman WA. Continuous administration of enteral lipid- and protein-rich nutrition limits inflammation in a human endotoxemia model. Crit Care Med. 2013;41:1258–1265. doi: 10.1097/CCM.0b013e31827c0a17. [DOI] [PubMed] [Google Scholar]
  • 287.Franco LM, Bucasas KL, Wells JM, Nino D, Wang X, Zapata GE, Arden N, Renwick A, Yu P, Quarles JM, Bray MS, Couch RB, Belmont JW, Shaw CA. Integrative genomic analysis of the human immune response to influenza vaccination. Elife. 2013;2:e00299. doi: 10.7554/eLife.00299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Chen R, Mias GI, Li-Pook-Than J, Jiang L, Lam HY, Chen R, Miriami E, Karczewski KJ, Hariharan M, Dewey FE, Cheng Y, Clark MJ, Im H, Habegger L, Balasubramanian S, O’Huallachain M, Dudley JT, Hillenmeyer S, Haraksingh R, Sharon D, Euskirchen G, Lacroute P, Bettinger K, Boyle AP, Kasowski M, Grubert F, Seki S, Garcia M, Whirl-Carrillo M, Gallardo M, Blasco MA, Greenberg PL, Snyder P, Klein TE, Altman RB, Butte AJ, Ashley EA, Gerstein M, Nadeau KC, Tang H, Snyder M. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148:1293–1307. doi: 10.1016/j.cell.2012.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 289.Feillet F, Agostoni C. Nutritional issues in treating phenylketonuria. J Inherit Metab Dis. 2010;33:659–664. doi: 10.1007/s10545-010-9043-4. [DOI] [PubMed] [Google Scholar]
  • 290.Abadie V, Sollid LM, Barreiro LB, Jabri B. Integration of genetic and immunological insights into a model of celiac disease pathogenesis. Annu Rev Immunol. 2011;29:493–525. doi: 10.1146/annurevimmunol-040210-092915. [DOI] [PubMed] [Google Scholar]
  • 291.Järvelä I, Torniainen S, Kolho KL. Molecular genetics of human lactase deficiencies. Ann Med. 2009;41:568–575. doi: 10.1080/07853890903121033. [DOI] [PubMed] [Google Scholar]
  • 292.D’Adamo P, Whitney C. Eat Right 4 (for) Your Type: The Individualized Diet Solution to Staying Healthy, Living Longer & Achieving Your Ideal Weight: 4 Blood Types, 4 Diets. New York, NY: G.P. Putnam’s Sons; 1996. [Google Scholar]
  • 293.Cusack L, De Buck E, Compernolle V, Vandekerckhove P. Blood type diets lack supporting evidence: a systematic review. Am J Clin Nutr. 2013;98:99–104. doi: 10.3945/ajcn.113.058693. [DOI] [PubMed] [Google Scholar]
  • 294.Wang J, García-Bailo B, Nielsen DE, El-Sohemy A. ABO genotype, ‘blood-type’ diet and cardiometabolic risk factors. PLoS One. 2014;9:e84749. doi: 10.1371/journal.pone.0084749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Nielsen DE, El-Sohemy A. A randomized trial of genetic information for personalized nutrition. Genes Nutr. 2012;7:559–566. doi: 10.1007/s12263-012-0290-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Nielsen DE, El-Sohemy A. Disclosure of genetic information and change in dietary intake: a randomized controlled trial. PLoS One. 2014;9:e112665. doi: 10.1371/journal.pone.0112665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Hietaranta-Luoma HL, Luomala HT, Puolijoki H, Hopia A. Using ApoE genotyping to promote healthy lifestyles in Finland: psychological impacts: randomized controlled trial. J Genet Couns. 2015;24:908–921. doi: 10.1007/s10897-015-9826-8. [DOI] [PubMed] [Google Scholar]
  • 298.O’Donnell PH, Danahey K, Jacobs M, Wadhwa NR, Yuen S, Bush A, Sacro Y, Sorrentino MJ, Siegler M, Harper W, Warrick A, Das S, Saner D, Corless CL, Ratain MJ. Adoption of a clinical pharmacogenomics implementation program during outpatient care: initial results of the University of Chicago “1,200 Patients Project.” Am J Med Genet C Semin Med Genet. 2014;166C:68–75. doi: 10.1002/ajmg.c.31385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Caudle KE, Klein TE, Hoffman JM, Muller DJ, Whirl-Carrillo M, Gong L, McDonagh EM, Sangkuhl K, Thorn CF, Schwab M, Agundez JA, Freimuth RR, Huser V, Lee MT, Iwuchukwu OF, Crews KR, Scott SA, Wadelius M, Swen JJ, Tyndale RF, Stein CM, Roden D, Relling MV, Williams MS, Johnson SG. Incorporation of pharmacogenomics into routine clinical practice: the Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline development process. Curr Drug Metab. 2014;15:209–217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300.Corella D, Sorlí JV, Estruch R, Coltell O, Ortega-Azorín C, Portolés O, Martínez-González MÁ, Bulló M, Fito M, Arós F, Lapetra J, Asensio EM, Saez GT, Serra-Majem L, Muñoz-Bravo C, Ruiz-Gutiérrez V, Fiol M, Vinyoles E, Pintó X, Richardson K, Ros E, Ordovás JM. MicroRNA-410 regulated lipoprotein lipase variant rs13702 is associated with stroke incidence and modulated by diet in the randomized controlled PREDIMED trial. Am J Clin Nutr. 2014;100:719–731. doi: 10.3945/ajcn.113.076992. [DOI] [PubMed] [Google Scholar]
  • 301.Havulinna AS, Kettunen J, Ukkola O, Osmond C, Eriksson JG, Kesaniemi YA, Jula A, Peltonen L, Kontula K, Salomaa V, Newton-Cheh C. A blood pressure genetic risk score is a significant predictor of incident cardiovascular events in 32,669 individuals. Hypertension. 2013;61:987–994. doi: 10.1161/HYPERTENSIONAHA.111.00649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Thanassoulis G, Peloso GM, Pencina MJ, Hoffmann U, Fox CS, Cupples LA, Levy D, D’Agostino RB, Hwang SJ, O’Donnell CJ. A genetic risk score is associated with incident cardiovascular disease and coronary artery calcium: the Framingham Heart Study. Circ Cardiovasc Genet. 2012;5:113–121. doi: 10.1161/CIRCGENETICS.111.961342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 303.Casas-Agustench P, Arnett DK, Smith CE, Lai CQ, Parnell LD, Borecki IB, Frazier-Wood AC, Allison M, Chen YD, Taylor KD, Rich SS, Rotter JI, Lee YC, Ordovas JM. Saturated fat intake modulates the association between an obesity genetic risk score and body mass index in two US populations. J Acad Nutr Diet. 2014;114:1954–1966. doi: 10.1016/j.jand.2014.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304.Frankwich KA, Egnatios J, Kenyon ML, Rutledge TR, Liao PS, Gupta S, Herbst KL, Zarrinpar A. Differences in weight loss between persons on standard balanced vs nutrigenetic diets in a randomized controlled trial. Clin Gastroenterol Hepatol. 2015;13:1625–1632.e1; quiz e145-e146. doi: 10.1016/j.cgh.2015.02.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305.Warden CH, Fisler JS. Comparisons of diets used in animal models of high-fat feeding. Cell Metab. 2008;7:277. doi: 10.1016/j.cmet.2008.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306.The Health Consequences of Smoking: 50 Years of Progress: A Report of the Surgeon General. Atlanta, GA: US Department of Health and Human Services, Centers for Disease Control and Prevention; 2014. http://www.surgeongeneral.gov/library/reports/50-years-of-progress/index.html. Accessed October 31, 2014. [Google Scholar]

RESOURCES