Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Dec 1.
Published in final edited form as: Curr Nutr Rep. 2013 Dec;2(4):10.1007/s13668-013-0052-4. doi: 10.1007/s13668-013-0052-4

Systems Epidemiology: A New Direction in Nutrition and Metabolic Disease Research

Marilyn C Cornelis 1, Frank B Hu 1
PMCID: PMC3837346  NIHMSID: NIHMS520818  PMID: 24278790

Abstract

Systems epidemiology applied to the field of nutrition has potential to provide new insight into underlying mechanisms and ways to study the health effects of specific foods more comprehensively. Human intervention and population-based studies have identified i) common genetic factors associated with several nutrition-related traits and ii) dietary factors altering the expression of genes and levels of proteins and metabolites related to inflammation, lipid metabolism and/or gut microbial metabolism, results of high relevance to metabolic disease. System-level tools applied type 2 diabetes and related conditions have revealed new pathways that are potentially modified by diet and thus offer additional opportunities for nutritional investigations. Moving forward, harnessing the resources of existing large prospective studies within which biological samples have been archived and diet and lifestyle have been measured repeatedly within individual will enable systems-level data to be integrated, the outcome of which will be improved personalized optimal nutrition for prevention and treatment of disease.

Keywords: nutrition, systems, genomics, transcriptomics, proteomics, metabolomics, type 2 diabetes, obesity, metabolic disease, epidemiology, populations, diet, network, nutrigenetics, nutrigenomics

Introduction

Traditional epidemiology has made important contributions to the identification of many key lifestyle and environmental risk factors for chronic disease. Technical advances that now allow high-throughput measurements of genomic, transcriptomic, proteomic, and metabolomic traits in combination with more sophisticated bioinformatics and statistical methods provide epidemiologists an unprecedented opportunity to unlock the full potential of their research approach. ‘Systems Epidemiology’ couples traditional epidemiologic methods with modern high-throughput technologies to enhance biological understanding of metabolic pathways in humans [14]. Nutrition research is a potentially ideal field for the application of systems approaches [5]. The dietary record and food frequency questionnaire have been instrumental to developing knowledge of the role diet plays in population health. However, these tools along with other aspects of epidemiological methods have well-known limitations that impede further advancements in the human nutrition field. Foods are mixtures of known and unknown constituents and separating as well as characterizing the effects each in the context of individual intrinsic variances is an on-going challenge in classical nutritional epidemiology.

The purpose of this review is to provide an overview of systems epidemiology with application to nutrition. We discuss progress in applying system-level tools to the study of both type 2 diabetes (T2D) and nutrition. Results of each highlight important connections between them and underscore the great potential for systems epidemiology in advancing nutrition research for disease prevention.

System Level I: Human Genome

Enthusiasm for systems epidemiology is fueled, in part, by the marked recent successes in human genomics: the study of an individual’s entire gene set, including gene-gene and gene-environment interactions. Genome-wide association studies (GWAS) of T2D have contributed to the identification of approximately 65 susceptibility loci [6]; which is an astounding improvement upon the small handful of loci described pre-GWAS [7]. Genome-wide analytical approaches have also proven successful for a number of important nutrition-related traits (Table 1). For example, single nucleotide polymorphisms (SNPs) have been associated with variation in plasma levels of iron and omega-3 fatty acids and consumption of dietary protein, alcohol, and coffee, all of which are dietary factors implicated in T2D [813]. Knowledge of the genetic determinants of diet response or behavior may provide insight into underlying mechanisms and ways to study the potential health effects of diet more comprehensively by using genetic determinants as instrumental variables or by taking into consideration gene-diet interactions. Other products of the GWAS era, besides the newly identified variants, such as consortium and innovative statistical design have greatly advanced the way such investigations are currently being conducted. Recent work aiming to confirm or refute causal effects and/or gene-environment interactions has involved cohort collections that are far larger than those utilized in the pre-GWAS era; this work has been facilitated by advanced meta-analysis techniques to combine data, and/or the availability of extensive replication materials [1417]. The majority of published GWAS have reported on disease-associated loci, but more recently GWAS has been extended to examine variation in nutrient-response or behavior. Adverse effects of several established T2D-associated loci may be attenuated by adopting healthy lifestyle behaviors such as high physical activity, whereas low physical activity and a Western dietary pattern have been found to augment genetic risk [15]. In a recent study among three independent cohorts of adults predisposed to obesity by virtue of a high genetic score of established obesity risk loci, the genetic effects were significantly more pronounced in those who consumed high quantities of sugar-sweetened beverages, compared with those who did not [18].

Table 1.

Genome-wide significant loci associated with nutrition-related traits1

Trait Locus2 Genes in Region Ref
Plasma amino acids
alanine, isoleucine 2p24 GCKR, SNX17, PMG1, NRBP, FNDC4 [135]
glutamine 10q24
12q13
HOGA1, MORN4
TIMELESS, MIP, SPRYD4, GLS2, BAZ2A
histidine 4q35 CYP4V2, KLKB1, F11
phenylalanine 5q33-q35 F12, GRK6
tyrosine 16q22 CHST4, TAT, PHLPPL, MARVELD3, AP1G,
SNORD71, ZNF821, PKD1L3
valine 2p15-p13
4q22
SLC1A4
PPM1K, HERC6, ABCG2
betaine 12p13.33
5q14.1
SLC6A12, SLC6A13
BHMT, BHMT2, DMGDH
[136]
glycine 2q34 CPS1
serine 1p12 PHGDH
Plasma fatty acids
linoleic acid, omega-3 FA 11q12-q13 MYRF, FEN1, FADS(1-3), DAGLA, BEST,
FTH1
[135]
linoleic acid
omega-6 & omega-7 FA
11q23
1p31.3
BUD13, ZNF259, APOA5, APOA4
DOCK7, ANGPTL3
linoleic acid, omega-6 & omega-7 FA, omega-9 & saturated FA,
other PUFA than linoleic
15q21-q23 LIPC, ADAM10
omega-9 & saturated FA 2p24 GCKR, SNX17, PMG1, NRBP1, FNDC4,
other PUFA than linoleic 11q12 CD6, CD5, VPS37C, PGA3
Plasma vitamins & minerals
beta-carotene 16q23.2 PKD1L2, BCMO1 [137]
calcium 3q21.1 CASR,CSTA,WDR5B,KPNA1,CCDC58 [138, 139]
carbohydrate-deficient transferrin (and %), transferrin, total iron binding capacity 3q22.1 TF, SRPRB [140143]
carbohydrate-deficient transferrin (%) 1p31 PGM1,RPL19P3 [140]
erythrocyte mean cell volume, iron
soluble transferrin receptor
22q12.3 TMPRSS6 [143146]
erythrocyte mean cell volume, ferritin, iron, soluble transferrin receptor, transferrin, transferring
saturation, unsaturated iron binding capacity, total iron binding capacity
6p21.3 HFE [140144, 146]
ferritin 6p22.2 SLC17A1 [143]
soluble transferrin receptor 11q23
11q23.3
BUD13, ZNF259, APOA5, APOA4
PCSK7
[146]
transferrin saturation 6p22.1 HIST1H2BJ, VN1R13P, VN1R11P [142]
magnesium 11p14.1
12q21.33
1q22
3q26.2
4q21.1
9q21.13
DCDC5, MPPED2
ATP2B1, MRPL2P1
MUC1
MDS1, MECOM
SHROOM3
TRPM6
[147]
retinol 10q23.33
18q12.1
RBP4, FFAR4
TTR,B4GALT6
[148]
vitamin B12 11q12.1
13q32.3
19p13.3
19q13.33
5q32
MS4A3
CLYBL
FUT6
FUT2
ASS1P10, PRELID2
[149151]
vitamin B6 1p36.12 ALPL, NBPF3 [150]
vitamin D (and insufficiency) 11p15.2
11q13.4
4q13.3
CYP2R1
NADSYN1,DHCR7
GC
[152154]
alpha-tocopherol, vitamin E response to vitamin E supplementation 11q23 BUD13, ZNF259, APOA5, APOA4 [137, 155, 156]
vitamin E 12q24.31
19p13.12
SCARB1
CYP4F2
[156]
Dietary Behaviors
habitual alcohol consumption 12q24.11-13
7q11.22
CCDC63, MYL2, ALDH2, BRAP, CUX2,
AUTS2
[157160]
habitual caffeine consumption, habitual coffee consumption 7p21
15q24.1
AHR
LMAN1L, EDC3, CYP1A2, CYP1A1, CSK
[20, 161, 162]
habitual protein consumption 19q13.33 FGF21, FUT1, FUT2, IZUMO1, RASIP1 [163]
1

Data were obtained by queries of the NHGRI catalogue of GWAS [164] (www.genome.gov/gwastudies, accessed June 2013) with additional data reported in supplementary material published by Kettunen et al [135]. For the latter, we restricted tabulations to SNP-metabolite associations although several SNP-metabolite ratios were also identified.

2

For most traits multiple significant SNPs have been identified at each locus. Interested readers should review the study reference provided for a list of SNPs and study-level details.

The currently known susceptibility loci for T2D explain only 5–10% of the previously estimated heritability of the disease [6] and add only modestly to traditional disease risk factors in prediction models 19]. . Likewise, SNPs associated with dietary behaviors explain very little of the trait variance and/or heritability [20]. This partly reflects the complexity of behaviors and metabolic disease, which is further compounded by measurement error ascribed to their assessment within- and between populations [21]. Knowledge gleaned by other system-level data may greatly facilitate continued progress in these areas of human genomics.

System Level II: Human Transcriptome

The transcriptome is the complete set of messenger RNA molecules in a cell or a tissue at a given time. Unlike the genome, which changes very little during a person’s lifetime, the transcriptome (as well as the proteome and metabolome) can vary by developmental stage and environment [22]. Because transcript quantification requires sufficient volumes of high-quality homogenous cellular material, human studies are often restricted to biopsies from accessible tissues such as subcutaneous adipose tissue, skeletal muscle and peripheral blood mononuclear cells[23]. Blood is the most feasibly collected in a population setting but its use in gene expression assumes that it provides general information on transcription in different cells and tissues, including those more relevant for the phenotype or disease of interest. GW expression studies of T2D have been predominately small, cross-sectional by design and without replication [24]. Only recently has the approach been applied to incident T2D: a GW screen of miRNAs in the Bruneck cohort revealed a plasma miRNA signature for incident T2D that includes loss of endothelial miR-126 [25]. The latter plays a pivotal role in maintaining endothelial homeostasis and vascular integrity [26].

Numerous human intervention studies have used transcriptomics to characterize molecular response to dietary factors such as dietary fatty acids [2731], carbohydrates [32], probiotics [33], folic acid [34], olive oil [27, 35], creatine monohydrate [36], vitamin E and selenium [37], CoQ10 [38], soy isoflavones [39] and different diets [4044]. Most reveal significant yet subtle changes in transcript levels. Convincing human evidence suggests that many of the biological effects of unsaturated fatty acids are mediated through modulation of gene transcription by regulating the activity of transcription factors such as peroxisome proliferator activated receptors, retinoid X receptor, liver X receptor and sterol regulatory binding proteins [23]. In light of emerging data showing gut flora compositions (or ‘microbiota’) differ between healthy individuals and diabetics or obese [4547], results from gene expression measures of duodenal mucosa following supplementation of Lactobacillus GG are encouraging. Lactobacillus GG, also found in yogurt and other fermented foods, altered the expression of genes involved in immune response and inflammation, apoptosis, cell growth and cell differentiation, cell–cell signaling, cell adhesion, and signal transcription and transduction [33].

Altered transcript levels in response to diet can inform mechanisms of action but whether such changes observed in nutritional intervention studies have relevance to a population setting and/or are large enough to have any impact on disease development is an open question. Little is known about whether or how specific dietary factors induce changes in gene expression of disease target tissue. To acquire such knowledge our only feasible option might be to complement human data with that obtained from experimental models. Genome-wide expression profiles collected from mouse liver and adipose tissue are notably enriched for altered transcripts of immunity-related genes when their high fat diets are supplemented with coffee [48]. Coffee suppressed expression of liver cytokine interleukin (IL)-1β and MCP-1 gene expression in white adipose tissue [48]. Given the role inflammation may have in development of T2D [49], these observations suggest a relevant mechanism by which coffee might reduces risk of the disease in humans. Nearly all GW scale studies concerning diet-induced changes in gene expression of pancreatic β-cells, a key target tissue for T2D, originate from rodent animal models or cultured cell lines [50]. While offering insight to tissue-specific gene-expression not easily obtained from humans, experimental studies are not without limitations and will warrant caution when translating to humans.

System Level III: Human Proteome

Transcript levels do not always correspond to protein levels, stressing the need to study proteins directly. The proteome may contain over a million structurally different proteins performing distinct functions in an individual and the aim of proteomics is to simultaneously quantify these proteins in biological samples [51, 52]. Currently, no single analytical platform is able to capture the full spectrum of proteins; those present at low concentrations and abundance are especially difficult to detect and characterize [53]. Metabolic diseases such as diabetes involve various affected tissue and since all are in contact with blood, tissue-specific proteins with diagnostic potential might be recovered in the circulation [52]. Blood is therefore an attractive biospecimen for protein profiling in population settings but is also the most complex to study [52]. Many individual serum proteins such as interleukin-6, resistan, leptin and adiponectin have been shown to vary between healthy and insulin resistant (IR) or diabetic individuals [5457], lending some support to the potential for global protein profiling. Indeed, the few small-scale studies that have applied proteomics have revealed potentially novel biomarkers of IR or T2D [58]. These include blood circulating flotillin-1, arginase, syntaxin 1C, haptoglobin and complement C3[59, 60]; mitochondrial, cytoskeletal, proteasome, and chaperone proteins in skeletal muscle[61]; and structural and stress/unfolded protein response proteins in subcutaneous adipose tissue [62]. Proteomics applied to human nutritional intervention studies have ranged from 1 to 12 weeks in duration and have, for example, tested soy isoflavones [63], flaxseed [64], fish oil [65] folic acid [66] and anti-inflammatory mixtures [67]. Serum levels of inflammatory and lipid proteins such as APOA1, zinc-a-2-glycoprotein, haptoglobin precursor, amyloid P component, and hemopexin are down regulated by 6 weeks of fish oil compared to sunflower oil supplementation [65]. Following 8 weeks of 25 mg of isoflavone supplementation, blood cell levels of proteins promoting fibrinolysis (i.e. alpha-enolase) were higher, while those mediating adhesion, migration, and proliferation of vascular smooth muscle cells (i.e. galectin-1) were lower[63].

Thus far proteomics has had limited application to population studies of human metabolic disease or nutrition. To our knowledge, no prospective proteomic studies of T2D have been conducted, and the few population-based studies of nutritional factors have been small and have all targeted <60 proteins [6871], most of which are from the inflammatory and/or oxidative stress classes of proteins. However, with continued advancements in the field such as high-throughput tools for the fractionation of biological samples and growing databases housing tissue-specific protein reference maps, this pattern promises to change in the near future.

Systems Level IV: Human Metabolome

Metabolomics involves the comprehensive analysis of all measurable metabolite concentrations under a given set of conditions [72, 73]. These metabolites are the final products of preceding ‘omic’ processes and their interaction with intrinsic and extrinsic factors [72, 73]. Thus far over 37,000 unique metabolites have been identified in human specimens [74, 75] (http://www.hmdb.ca, accessed June 2013). Similar to the proteome, no single analytical platform captures the full spectrum of metabolites. Analyses can be conducted in an untargeted manner, typically optimized for broad coverage of the metabolome to enhance opportunities of discovering discriminatory features of a disease/trait, or in a targeted manner, profiling only a defined set of known metabolites to enable improved sensitivity and efficiency [76]. Metabolomics is increasingly applied in the clinical and population setting. Detailed technological and conceptual challenges as well as progress in the broader field have been discussed elsewhere [77].

Recent metabolomic studies of T2D have considerably improved upon the earlier cross-sectional reports [78, 79]. Among adult population-based studies involving up to 800 incident cases, targeted metabolite profiling has identified an array of novel metabolite classes linked to T2D onset such as short- and medium-chain acylcarnitines, the specific lipid classes of sphingomyelins (SMs), lysophosphatidylcholines, phosphatidylcholines (PCs), and lysophosphatidylethanolamines, and branched-chain amino acids (BCAAs) [8084]. Some of these classes have also been linked to obesity and other T2D-related traits [8590]. Patterns linked with T2D among adults have not always been replicated in younger populations [91, 92], supporting the need for longitudinal monitoring to enable trajectory analysis of metabolic responses. Further work is needed to determine whether these metabolites are elevated because of an individual’s genetic or diet characteristics, and/or the actions of gut microbes [93]. Interestingly, high dietary protein intake is associated with T2D and higher plasma levels of BCAAs [8, 94]. PCs occur in animal and plant tissues and are therefore present in the diet (e.g. eggs and soy beans). Recent work suggests these phospholipids are further metabolized by intestinal microbiota, producing the proatherosclerotic metabolite trimethylamine-N-oxide [9598].

Metabolomics has been used to characterize the complex human metabolic effects of specific foods, nutrients and dietary patterns in both the clinical and population setting [99107]. Intervention studies of coffee [108111], tea [112, 113], cocoa [114, 115], nuts [116], dietary fiber[117], vitamins and selected nutraceuticals [67, 118], for example, have incorporated either targeted or untargeted metabolite profiling. Earlier reports have favored the presentation of exogenously-derived metabolites. Confirmed metabolite markers of coffee exposure, for example, include mainly methylxanthines and reduced, sulfated and methylated forms of hydroxycinnamates [108111], which are coffee-derived metabolites. There is, however, an increasing interest in the impact foods have on the endogenous metabolome (known as the ‘host response’)[115]. Potential markers of nut intake include conjugated fatty acids, serotonin metabolites and microbial-derived phenolic metabolites [116]. Metabolites linked with carnitine metabolism, sulfation of tyrosine and gut microbial metabolism are among the set of metabolites linked to cocoa intake[115]. Evidence for diet-induced altered intestinal microbiota metabolism is particularly relevant in light of results from metabolomic studies of T2D and other system-level results discussed above.

Population-based studies aimed at identifying metabolite signatures of self-reported dietary intake have targeted a similar set of metabolites to that of recent metabolomic studies of T2D (i.e. lipids, amino acids and their derivatives). In a population sample of men, coffee intake was positively associated with specific classes of SMs and negatively associated with long- and medium-chain acylcarnitines in plasma[119], classes previously linked to reduced and increased risk of T2D, respectively[80, 81, 120122]. The negative association between coffee and medium-chain acylcarnitines was later confirmed in an independent study of women from the TwinsUK cohort [123], which additionally identified metabolites associated with garlic, fruit and vegetable intake and hypo-caloric dieting [123]. Of relevance to the utility and interpretation of metabolomic approaches applied to populations is recent work by Heinzmann et al [124] and Krug et al [101], showing that interindividual metabolic differences influence proportionally more of the spectrum of metabolites than dietary modulation, although certain individuals display a greater stability of metabolic phenotypes than others [101, 124]. Some of this stability is likely heritable; in the TwinsUK study mentioned above, two thirds of the metabolites associated with nutritional patterns had a significant genetic contribution, and the remaining third were solely environmentally determined[123].

Metabolomics has already provided new insight to chronic disease development and response to diet. Its application in a population setting might further enable epidemiologists to separate metabolites resulting from inherited patterns of metabolism from those that are entirely due to environment[123]. This particular task will greatly benefit from findings of other system-level analysis, the most apparent being genomics. Indeed, several GW-confirmed loci are associated with variation in plasma levels of metabolites implicated in T2D and additionally influenced by diet (Table 1)[125]. Recent metabolomic successes have been based on only a fraction or two of the full spectrum of metabolites. Ongoing efforts to characterize the existing ‘unknowns’ will soon enable more exhaustive investigations.

Future Directions: Human Systems Integration

As outlined above, each system level has potential to reveal insight to disease development and response to diet. Besides single SNP-, transcript-, protein- and metabolite-trait testing, network analysis (knowledge-driven) or computational modeling of all system-level data-points (data-driven) may also be performed. Integrating system-level data from external resources is another feasible and highly efficient approach epidemiologists are taking to maximize ‘omic’ data [126128]. However, extrapolating results from an integrative analysis of potentially diverse datasets does not provide a complete understanding of the human system and its responses to diet as a whole. Intermediate molecular quantitative trait locus (iQTLs) analysis has been one advancement towards integrating large-scale omic data [22, 129]. In iQTL mapping, transcripts, proteins, metabolites or other heritable physiological factors serve as traits for genetic association analysis, the rationale being that these intermediates (or endophenotypes) more closely underlie genetic risk than heterogeneous behavioral or disease phenotypes [129, 130].

The ideal system-wide study would have multi-omic data for an individual collected at multiple time points along with exposure details and longitudinal follow-up. Chen et al [131] recently explored this concept of an ‘integrative personal omics profile’ (iPOP) by combining several omic-profiles from a single individual over a 14-month period. iPOP revealed various health risks, including T2D and uncovered extensive, dynamic changes in diverse molecular components and biological pathways across healthy and diseased conditions. This iPOP frame-work might easily be up-scaled to a nutritional intervention study. The most ambitious systems epidemiology approach would apply this iPOP frame-work to a population-setting. Harnessing the resources of existing large prospective studies of lifestyle and chronic diseases which have archived biological samples and repeated measures of diet and lifestyle would be the most efficient approach to reaching this goal [1]. This strategy was recently applied to a subset of individuals participating in the Finnish population-based Dietary, Lifestyle, and Genetic determinants of Obesity and Metabolic syndrome (DILGOM) study [132] and preliminary cross-sectional analysis are promising. Network analysis of transcriptomic and metabolomic data available from 518 individuals identified genes from the lipid–leukocyte (LL) module as having a key role in over 80 metabolites, including lipoprotein subclasses, lipids, and amino acids. Genomic variation was used to infer this module’s reactivity to fatty acids and high/low/intermediate-density lipoprotein fractions. Parallel associations with plasma IL-1 receptor antagonist, C-reactive protein, and adiponectin suggested the LL module as a possible link between inflammation, metabolism, and adiposity [132].

While beyond the scope of the current paper, there are several important issues that still need resolving before considering system tools in the broader field of systems epidemiology. For example, a common goal in epidemiology is to relate a "usual" level of an exposure with the risk of disease [133] and an outstanding concern pertains to the fact that transcript, protein and metabolite levels vary with time and recent environmental factors. Quantifying and identifying the source of this variation and the extent to which this variation impacts power for detecting associations warrants further study[134]. How repeated measures may remedy this concern is also an open question. Another issue is that the multiple-testing problem is likely to be magnified in multi-omic studies, which will have significant impacts on the power and data interpretation. Epidemiologists will need to be mindful of these and other limitations when designing and interpreting system-wide studies.

Summary and Conclusions

Nutritional systems epidemiology has potential to provide new insight into underlying mechanisms and ways to study the health effects of specific foods more comprehensively. Coincident with advancing nutrition research, it has incredible potential to finally realize the concept of personalized nutrition. Continued technological advances in sensitive high-throughput methods, enhanced bioinformatics and analytical tools, and reduced costs will enable more widespread use of these techniques in nutrition and epidemiological research. However, critical to the success of this approach is well-designed prospective cohort studies with very large size, long-term follow-up, high rates of follow-up, availability of archived biological samples, and detailed measures of diet and lifestyle. Our brief summary of selected studies across systems already yield impressive results and when merged with equivalent applications to T2D provide compelling evidence that the goal of personalized optimal nutrition for prevention and treatment of disease will become increasingly attainable by systems epidemiological approaches.

Footnotes

Compliance with Ethics Guidelines

Conflict of Interest

Marilyn C. Cornelis declares that she has no conflict of interest.

Frank B. Hu has received compensation for serving as a consultant from Bunge Ltd. and Novo Nordisk, and he is supported by grants from Merck and the California Walnut Commission.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

References

  • 1.Hu FB. Metabolic profiling of diabetes: from black-box epidemiology to systems epidemiology. Clin Chem. 2011;57(9):1224–1226. doi: 10.1373/clinchem.2011.167056. [DOI] [PubMed] [Google Scholar]
  • 2.Haring R, Wallaschofski H. Diving Through the "-Omics": The Case for Deep Phenotyping and Systems Epidemiology. Omics. 2012 doi: 10.1089/omi.2011.0108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lund E, Dumeaux V. Systems epidemiology in cancer. Cancer Epidemiol Biomarkers Prev. 2008;17(11):2954–2957. doi: 10.1158/1055-9965.EPI-08-0519. [DOI] [PubMed] [Google Scholar]
  • 4.Bictash M, Ebbels TM, Chan Q, et al. Opening up the "Black Box": metabolic phenotyping and metabolome-wide association studies in epidemiology. J Clin Epidemiol. 2010;63(9):970–979. doi: 10.1016/j.jclinepi.2009.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Keusch GT. What do -omics mean for the science and policy of the nutritional sciences? Am J Clin Nutr. 2006;83(2):520S–522S. doi: 10.1093/ajcn/83.2.520S. [DOI] [PubMed] [Google Scholar]
  • 6. Morris AP, Voight BF, Teslovich TM, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat Genet. 2012;44(9):981–990. doi: 10.1038/ng.2383. This article is the latest in a series of successful genome-wide association studies of type 2 diabetes that demonstrate the power of an agnostic systemwide approach to susceptibility loci discovery.
  • 7.McCarthy MI. Genomics, type 2 diabetes, and obesity. N Engl J Med. 2010;363(24):2339–2350. doi: 10.1056/NEJMra0906948. [DOI] [PubMed] [Google Scholar]
  • 8.Ericson U, Sonestedt E, Gullberg B, et al. High intakes of protein and processed meat associate with increased incidence of type 2 diabetes. Br J Nutr. 2013;109(6):1143–1153. doi: 10.1017/S0007114512003017. [DOI] [PubMed] [Google Scholar]
  • 9.Hu FB, Manson JE, Stampfer MJ, et al. Diet, lifestyle, and the risk of type 2 diabetes mellitus in women. N Engl J Med. 2001;345(11):790–797. doi: 10.1056/NEJMoa010492. [DOI] [PubMed] [Google Scholar]
  • 10.Djousse L, Gaziano JM, Buring JE, et al. Dietary omega-3 fatty acids and fish consumption and risk of type 2 diabetes. Am J Clin Nutr. 2011;93(1):143–150. doi: 10.3945/ajcn.110.005603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.van Dam RM, Hu FB. Coffee consumption and risk of type 2 diabetes: a systematic review. Jama. 2005;294(1):97–104. doi: 10.1001/jama.294.1.97. [DOI] [PubMed] [Google Scholar]
  • 12.Koppes LL, Dekker JM, Hendriks HF, et al. Moderate alcohol consumption lowers the risk of type 2 diabetes: a meta-analysis of prospective observational studies. Diabetes Care. 2005;28(3):719–725. doi: 10.2337/diacare.28.3.719. [DOI] [PubMed] [Google Scholar]
  • 13.Jiang R, Manson JE, Meigs JB, et al. Body iron stores in relation to risk of type 2 diabetes in apparently healthy women. Jama. 2004;291(6):711–717. doi: 10.1001/jama.291.6.711. [DOI] [PubMed] [Google Scholar]
  • 14.Nettleton JA, Hivert MF, Lemaitre RN, et al. Meta-analysis investigating associations between healthy diet and fasting glucose and insulin levels and modification by loci associated with glucose homeostasis in data from 15 cohorts. Am J Epidemiol. 2013;177(2):103–115. doi: 10.1093/aje/kws297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Cornelis MC, Hu FB. Gene-Environment Interactions in the Development of Type 2 Diabetes: Recent Progress and Continuing Challenges. Annu Rev Nutr. 2012;32:245–259. doi: 10.1146/annurev-nutr-071811-150648. This article discusses recent progress, continuing challenges, evolving approaches, and recommendations for future efforts pertaining to studies of gene-environment interactions of type 2 diabetes.
  • 16.Vimaleswaran KS, Berry DJ, Lu C, et al. Causal relationship between obesity and vitamin D status: bi-directional Mendelian randomization analysis of multiple cohorts. PLoS Med. 2013;10(2):e1001383. doi: 10.1371/journal.pmed.1001383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Franks PW, Pearson E, Florez JC. Gene-environment and gene-treatment interactions in type 2 diabetes: progress, pitfalls, and prospects. Diabetes Care. 2013;36(5):1413–1421. doi: 10.2337/dc12-2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Qi Q, Chu AY, Kang JH, et al. Sugar-sweetened beverages and genetic risk of obesity. N Engl J Med. 2012;367(15):1387–1396. doi: 10.1056/NEJMoa1203039. This article tests and validates an interaction between a genetic score of established obesity risk loci and sugar-sweetened beverages for risk of obesity.
  • 19. Herder C, Karakas M, Koenig W. Biomarkers for the prediction of type 2 diabetes and cardiovascular disease. Clin Pharmacol Ther. 2011;90(1):52–66. doi: 10.1038/clpt.2011.93. This article provides an exhaustive review of system-level studies for prediction of type 2 diabetes and cardiovascular disease.
  • 20.Cornelis MC, Monda KL, Yu K, et al. Genome-wide meta-analysis identifies regions on 7p21 (AHR) and 15q24 (CYP1A2) as determinants of habitual caffeine consumption. PLoS Genet. 2011;7(4):e1002033. doi: 10.1371/journal.pgen.1002033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Agrawal A, Freedman ND, Cheng YC, et al. Measuring alcohol consumption for genomic meta-analyses of alcohol intake: opportunities and challenges. Am J Clin Nutr. 2012;95(3):539–547. doi: 10.3945/ajcn.111.015545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Meng Q, Makinen VP, Luk H, et al. Systems Biology Approaches and Applications in Obesity, Diabetes, and Cardiovascular Diseases. Current cardiovascular risk reports. 2013;7(1):73–83. doi: 10.1007/s12170-012-0280-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Afman LA, Muller M. Human nutrigenomics of gene regulation by dietary fatty acids. Progress in lipid research. 2012;51(1):63–70. doi: 10.1016/j.plipres.2011.11.005. [DOI] [PubMed] [Google Scholar]
  • 24.Takamura T, Honda M, Sakai Y, et al. Gene expression profiles in peripheral blood mononuclear cells reflect the pathophysiology of type 2 diabetes. Biochem Biophys Res Commun. 2007;361(2):379–384. doi: 10.1016/j.bbrc.2007.07.006. [DOI] [PubMed] [Google Scholar]
  • 25.Zampetaki A, Kiechl S, Drozdov I, et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circulation research. 2010;107(6):810–817. doi: 10.1161/CIRCRESAHA.110.226357. [DOI] [PubMed] [Google Scholar]
  • 26.Wang S, Aurora AB, Johnson BA, et al. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Developmental cell. 2008;15(2):261–271. doi: 10.1016/j.devcel.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Camargo A, Ruano J, Fernandez JM, et al. Gene expression changes in mononuclear cells in patients with metabolic syndrome after acute intake of phenol-rich virgin olive oil. BMC Genomics. 2010;11:253. doi: 10.1186/1471-2164-11-253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bouwens M, Grootte Bromhaar M, Jansen J, et al. Postprandial dietary lipid-specific effects on human peripheral blood mononuclear cell gene expression profiles. Am J Clin Nutr. 2010;91(1):208–217. doi: 10.3945/ajcn.2009.28586. [DOI] [PubMed] [Google Scholar]
  • 29.van Dijk SJ, Feskens EJ, Bos MB, et al. 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(6):1656–1664. doi: 10.3945/ajcn.2009.27792. [DOI] [PubMed] [Google Scholar]
  • 30.Bouwens M, van de Rest O, Dellschaft N, et al. Fish-oil supplementation induces antiinflammatory gene expression profiles in human blood mononuclear cells. Am J Clin Nutr. 2009;90(2):415–424. doi: 10.3945/ajcn.2009.27680. [DOI] [PubMed] [Google Scholar]
  • 31.Kabir M, Skurnik G, Naour N, et al. Treatment for 2 mo with n 3 polyunsaturated fatty acids reduces adiposity and some atherogenic factors but does not improve insulin sensitivity in women with type 2 diabetes: a randomized controlled study. Am J Clin Nutr. 2007;86(6):1670–1679. doi: 10.1093/ajcn/86.5.1670. [DOI] [PubMed] [Google Scholar]
  • 32.Kallio P, Kolehmainen M, Laaksonen DE, et al. Dietary carbohydrate modification induces alterations in gene expression in abdominal subcutaneous adipose tissue in persons with the metabolic syndrome: the FUNGENUT Study. Am J Clin Nutr. 2007;85(5):1417–1427. doi: 10.1093/ajcn/85.5.1417. [DOI] [PubMed] [Google Scholar]
  • 33.Di Caro S, Tao H, Grillo A, et al. Effects of Lactobacillus GG on genes expression pattern in small bowel mucosa. Dig Liver Dis. 2005;37(5):320–329. doi: 10.1016/j.dld.2004.12.008. [DOI] [PubMed] [Google Scholar]
  • 34.van Oostrom O, de Kleijn DP, Fledderus JO, et al. Folic acid supplementation normalizes the endothelial progenitor cell transcriptome of patients with type 1 diabetes: a casecontrol pilot study. Cardiovasc Diabetol. 2009;8:47. doi: 10.1186/1475-2840-8-47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Khymenets O, Fito M, Covas MI, et al. Mononuclear cell transcriptome response after sustained virgin olive oil consumption in humans: an exploratory nutrigenomics study. Omics. 2009;13(1):7–19. doi: 10.1089/omi.2008.0079. [DOI] [PubMed] [Google Scholar]
  • 36.Safdar A, Yardley NJ, Snow R, et al. Global and targeted gene expression and protein content in skeletal muscle of young men following short-term creatine monohydrate supplementation. Physiol Genomics. 2008;32(2):219–228. doi: 10.1152/physiolgenomics.00157.2007. [DOI] [PubMed] [Google Scholar]
  • 37.Tsavachidou D, McDonnell TJ, Wen S, et al. Selenium and vitamin E: cell type- and intervention-specific tissue effects in prostate cancer. J Natl Cancer Inst. 2009;101(5):306–320. doi: 10.1093/jnci/djn512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Linnane AW, Kopsidas G, Zhang C, et al. Cellular redox activity of coenzyme Q10: effect of CoQ10 supplementation on human skeletal muscle. Free Radic Res. 2002;36(4):445–453. doi: 10.1080/10715760290021306. [DOI] [PubMed] [Google Scholar]
  • 39.Niculescu MD, Pop EA, Fischer LM, et al. Dietary isoflavones differentially induce gene expression changes in lymphocytes from postmenopausal women who form equol as compared with those who do not. J Nutr Biochem. 2007;18(6):380–390. doi: 10.1016/j.jnutbio.2006.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Crujeiras AB, Parra D, Milagro FI, et al. Differential expression of oxidative stress and inflammation related genes in peripheral blood mononuclear cells in response to a lowcalorie diet: a nutrigenomics study. Omics. 2008;12(4):251–261. doi: 10.1089/omi.2008.0001. [DOI] [PubMed] [Google Scholar]
  • 41.Dahlman I, Linder K, Arvidsson Nordstrom E, et al. Changes in adipose tissue gene expression with energy-restricted diets in obese women. Am J Clin Nutr. 2005;81(6):1275–1285. doi: 10.1093/ajcn/81.6.1275. [DOI] [PubMed] [Google Scholar]
  • 42.Ong KR, Sims AH, Harvie M, et al. Biomarkers of dietary energy restriction in women at increased risk of breast cancer. Cancer prevention research (Philadelphia, Pa. 2009;2(8):720–731. doi: 10.1158/1940-6207.CAPR-09-0008. [DOI] [PubMed] [Google Scholar]
  • 43.Capel F, Viguerie N, Vega N, et al. Contribution of energy restriction and macronutrient composition to changes in adipose tissue gene expression during dietary weight-loss programs in obese women. J Clin Endocrinol Metab. 2008;93(11):4315–4322. doi: 10.1210/jc.2008-0814. [DOI] [PubMed] [Google Scholar]
  • 44.van Erk MJ, Blom WA, van Ommen B, et al. High-protein and high-carbohydrate breakfasts differentially change the transcriptome of human blood cells. Am J Clin Nutr. 2006;84(5):1233–1241. doi: 10.1093/ajcn/84.5.1233. [DOI] [PubMed] [Google Scholar]
  • 45.Diamant M, Blaak EE, de Vos WM. Do nutrient-gut-microbiota interactions play a role in human obesity, insulin resistance and type 2 diabetes? Obes Rev. 2011;12(4):272–281. doi: 10.1111/j.1467-789X.2010.00797.x. [DOI] [PubMed] [Google Scholar]
  • 46.Wu X, Ma C, Han L, et al. Molecular characterisation of the faecal microbiota in patients with type II diabetes. Current microbiology. 2010;61(1):69–78. doi: 10.1007/s00284-010-9582-9. [DOI] [PubMed] [Google Scholar]
  • 47.Kalliomaki M, Collado MC, Salminen S, et al. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr. 2008;87(3):534–538. doi: 10.1093/ajcn/87.3.534. [DOI] [PubMed] [Google Scholar]
  • 48.Fukushima Y, Kasuga M, Nakao K, et al. Effects of coffee on inflammatory cytokine gene expression in mice fed high-fat diets. J Agric Food Chem. 2009;57(23):11100–11105. doi: 10.1021/jf901278u. [DOI] [PubMed] [Google Scholar]
  • 49.Schulze MB, Hoffmann K, Manson JE, et al. Dietary pattern, inflammation, and incidence of type 2 diabetes in women. Am J Clin Nutr. 2005;82(3):675–684. doi: 10.1093/ajcn.82.3.675. quiz 714-675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Chadt A, Yeo GS, Al-Hasani H. Nutrition-/diet-induced changes in gene expression in pancreatic beta-cells. Diabetes Obes Metab. 2012;14(Suppl 3):57–67. doi: 10.1111/j.1463-1326.2012.01640.x. This article reviews the application of targeted and genome-wide approaches to diet-induced changes in gene expression in type 2 diabetes target tissue and supports the important utility of experimental studies to address scientific questions that cannot be feasibly answered by human studies.
  • 51.Tyers M, Mann M. From genomics to proteomics. Nature. 2003;422(6928):193–197. doi: 10.1038/nature01510. [DOI] [PubMed] [Google Scholar]
  • 52.Anderson NL, Polanski M, Pieper R, et al. The human plasma proteome: a nonredundant list developed by combination of four separate sources. Mol Cell Proteomics. 2004;3(4):311–326. doi: 10.1074/mcp.M300127-MCP200. [DOI] [PubMed] [Google Scholar]
  • 53.Wittwer J, Rubio-Aliaga I, Hoeft B, et al. Nutrigenomics in human intervention studies: current status, lessons learned and future perspectives. Mol Nutr Food Res. 2011;55(3):341–358. doi: 10.1002/mnfr.201000512. [DOI] [PubMed] [Google Scholar]
  • 54.Considine RV, Sinha MK, Heiman ML, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. N Engl J Med. 1996;334(5):292–295. doi: 10.1056/NEJM199602013340503. [DOI] [PubMed] [Google Scholar]
  • 55.Hotta K, Funahashi T, Arita Y, et al. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol. 2000;20(6):1595–1599. doi: 10.1161/01.atv.20.6.1595. [DOI] [PubMed] [Google Scholar]
  • 56.Norata GD, Ongari M, Garlaschelli K, et al. Plasma resistin levels correlate with determinants of the metabolic syndrome. Eur J Endocrinol. 2007;156(2):279–284. doi: 10.1530/eje.1.02338. [DOI] [PubMed] [Google Scholar]
  • 57.Vozarova B, Weyer C, Hanson K, et al. Circulating interleukin-6 in relation to adiposity, insulin action, and insulin secretion. Obes Res. 2001;9(7):414–417. doi: 10.1038/oby.2001.54. [DOI] [PubMed] [Google Scholar]
  • 58.Sundsten T, Ortsater H. Proteomics in diabetes research. Mol Cell Endocrinol. 2009;297(1–2):93–103. doi: 10.1016/j.mce.2008.06.018. [DOI] [PubMed] [Google Scholar]
  • 59.Jiang M, Jia L, Jiang W, et al. Protein disregulation in red blood cell membranes of type 2 diabetic patients. Biochem Biophys Res Commun. 2003;309(1):196–200. doi: 10.1016/s0006-291x(03)01559-6. [DOI] [PubMed] [Google Scholar]
  • 60.Zhang R, Barker L, Pinchev D, et al. Mining biomarkers in human sera using proteomic tools. Proteomics. 2004;4(1):244–256. doi: 10.1002/pmic.200300495. [DOI] [PubMed] [Google Scholar]
  • 61.Hwang H, Bowen BP, Lefort N, et al. Proteomics analysis of human skeletal muscle reveals novel abnormalities in obesity and type 2 diabetes. Diabetes. 2010;59(1):33–42. doi: 10.2337/db09-0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Boden G, Duan X, Homko C, et al. Increase in endoplasmic reticulum stress-related proteins and genes in adipose tissue of obese, insulin-resistant individuals. Diabetes. 2008;57(9):2438–2444. doi: 10.2337/db08-0604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fuchs D, Vafeiadou K, Hall WL, et al. Proteomic biomarkers of peripheral blood mononuclear cells obtained from postmenopausal women undergoing an intervention with soy isoflavones. Am J Clin Nutr. 2007;86(5):1369–1375. doi: 10.1093/ajcn/86.5.1369. [DOI] [PubMed] [Google Scholar]
  • 64.Fuchs D, Piller R, Linseisen J, et al. The human peripheral blood mononuclear cell proteome responds to a dietary flaxseed-intervention and proteins identified suggest a protective effect in atherosclerosis. Proteomics. 2007;7(18):3278–3288. doi: 10.1002/pmic.200700096. [DOI] [PubMed] [Google Scholar]
  • 65.de Roos B, Geelen A, Ross K, et al. Identification of potential serum biomarkers of inflammation and lipid modulation that are altered by fish oil supplementation in healthy volunteers. Proteomics. 2008;8(10):1965–1974. doi: 10.1002/pmic.200700457. [DOI] [PubMed] [Google Scholar]
  • 66.Duthie SJ, Horgan G, de Roos B, et al. Blood folate status and expression of proteins involved in immune function, inflammation, and coagulation: biochemical and proteomic changes in the plasma of humans in response to long-term synthetic folic acid supplementation. J Proteome Res. 2010;9(4):1941–1950. doi: 10.1021/pr901103n. [DOI] [PubMed] [Google Scholar]
  • 67.Bakker GC, van Erk MJ, Pellis L, et al. An antiinflammatory dietary mix modulates inflammation and oxidative and metabolic stress in overweight men: a nutrigenomics approach. Am J Clin Nutr. 2010;91(4):1044–1059. doi: 10.3945/ajcn.2009.28822. [DOI] [PubMed] [Google Scholar]
  • 68.Esposito K, Marfella R, Ciotola M, et al. Effect of a mediterranean-style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. Jama. 2004;292(12):1440–1446. doi: 10.1001/jama.292.12.1440. [DOI] [PubMed] [Google Scholar]
  • 69.Nettleton JA, Matijevic N, Follis JL, et al. Associations between dietary patterns and flow cytometry-measured biomarkers of inflammation and cellular activation in the Atherosclerosis Risk in Communities (ARIC) Carotid Artery MRI Study. Atherosclerosis. 2010;212(1):260–267. doi: 10.1016/j.atherosclerosis.2010.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lopez-Garcia E, Schulze MB, Fung TT, et al. Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr. 2004;80(4):1029–1035. doi: 10.1093/ajcn/80.4.1029. [DOI] [PubMed] [Google Scholar]
  • 71.Garcia-Bailo B, Brenner DR, Nielsen D, et al. Dietary patterns and ethnicity are associated with distinct plasma proteomic groups. Am J Clin Nutr. 2012;95(2):352–361. doi: 10.3945/ajcn.111.022657. [DOI] [PubMed] [Google Scholar]
  • 72.Norheim F, Gjelstad IM, Hjorth M, et al. Molecular nutrition research: the modern way of performing nutritional science. Nutrients. 2012;4(12):1898–1944. doi: 10.3390/nu4121898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Rezzi S, Ramadan Z, Fay LB, et al. Nutritional metabonomics: applications and perspectives. J Proteome Res. 2007;6(2):513–525. doi: 10.1021/pr060522z. [DOI] [PubMed] [Google Scholar]
  • 74.Wishart DS. Human Metabolome Database: completing the 'human parts list'. Pharmacogenomics. 2007;8(7):683–686. doi: 10.2217/14622416.8.7.683. [DOI] [PubMed] [Google Scholar]
  • 75.The Human Metabolome Project. [ www.metabolomics.ca]
  • 76.Scalbert A, Brennan L, Fiehn O, et al. Mass-spectrometry-based metabolomics: limitations and recommendations for future progress with particular focus on nutrition research. Metabolomics. 2009;5(4):435–458. doi: 10.1007/s11306-009-0168-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Koek MM, Jellema RH, van der Greef J, et al. Quantitative metabolomics based on gas chromatography mass spectrometry: status and perspectives. Metabolomics. 2011;7(3):307–328. doi: 10.1007/s11306-010-0254-3. This article provides technological and conceptual challenges as well as progress in the broader field of metabolomics.
  • 78.Gall WE, Beebe K, Lawton KA, et al. alpha-hydroxybutyrate is an early biomarker of insulin resistance and glucose intolerance in a nondiabetic population. PLoS One. 2010;5(5):e10883. doi: 10.1371/journal.pone.0010883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Xu F, Tavintharan S, Sum CF, et al. Metabolic Signature Shift in Type 2 Diabetes Mellitus Revealed by Mass Spectrometry-based Metabolomics. J Clin Endocrinol Metab. 2013 doi: 10.1210/jc.2012-4132. [DOI] [PubMed] [Google Scholar]
  • 80.Mihalik SJ, Goodpaster BH, Kelley DE, et al. Increased levels of plasma acylcarnitines in obesity and type 2 diabetes and identification of a marker of glucolipotoxicity. Obesity (Silver Spring) 2010;18(9):1695–1700. doi: 10.1038/oby.2009.510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rhee EP, Cheng S, Larson MG, et al. Lipid profiling identifies a triacylglycerol signature of insulin resistance and improves diabetes prediction in humans. J Clin Invest. 2011;121(4):1402–1411. doi: 10.1172/JCI44442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Floegel A, Stefan N, Yu Z, et al. Identification of serum metabolites associated with risk of type 2 diabetes using a targeted metabolomic approach. Diabetes. 2013;62(2):639–648. doi: 10.2337/db12-0495. This article describes the largest prospective metabolomic study of type 2 diabetes conducted to date.
  • 83.Suhre K, Meisinger C, Doring A, et al. Metabolic footprint of diabetes: a multiplatform metabolomics study in an epidemiological setting. PLoS One. 2010;5(11):e13953. doi: 10.1371/journal.pone.0013953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang TJ, Larson MG, Vasan RS, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17(4):448–453. doi: 10.1038/nm.2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wurtz P, Makinen VP, Soininen P, et al. Metabolic Signatures of Insulin Resistance in 7,098 Young Adults. Diabetes. 2012 doi: 10.2337/db11-1355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wurtz P, Tiainen M, Makinen VP, et al. Circulating metabolite predictors of glycemia in middle-aged men and women. Diabetes Care. 2012;35(8):1749–1756. doi: 10.2337/dc11-1838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Newgard CB, An J, Bain JR, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell metabolism. 2009;9(4):311–326. doi: 10.1016/j.cmet.2009.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Shah SH, Crosslin DR, Haynes CS, et al. Branched-chain amino acid levels are associated with improvement in insulin resistance with weight loss. Diabetologia. 2012;55(2):321–330. doi: 10.1007/s00125-011-2356-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Cheng S, Rhee EP, Larson MG, et al. Metabolite Profiling Identifies Pathways Associated with Metabolic Risk in Humans. Circulation. 2012 doi: 10.1161/CIRCULATIONAHA.111.067827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Huffman KM, Shah SH, Stevens RD, et al. Relationships between circulating metabolic intermediates and insulin action in overweight to obese, inactive men and women. Diabetes Care. 2009;32(9):1678–1683. doi: 10.2337/dc08-2075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Mihalik SJ, Michaliszyn SF, de Las Heras J, et al. Metabolomic Profiling of Fatty Acid and Amino Acid Metabolism in Youth With Obesity and Type 2 Diabetes: Evidence for enhanced mitochondrial oxidation. Diabetes Care. 2012;35(3):605–611. doi: 10.2337/DC11-1577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Boyle KE, Canham JP, Consitt LA, et al. A high-fat diet elicits differential responses in genes coordinating oxidative metabolism in skeletal muscle of lean and obese individuals. J Clin Endocrinol Metab. 2011;96(3):775–781. doi: 10.1210/jc.2010-2253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Shah SH, Svetkey LP, Newgard CB. Branching out for detection of type 2 diabetes. Cell metabolism. 2011;13(5):491–492. doi: 10.1016/j.cmet.2011.04.003. [DOI] [PubMed] [Google Scholar]
  • 94.Jakobsen LH, Kondrup J, Zellner M, et al. Effect of a high protein meat diet on muscle and cognitive functions: a randomised controlled dietary intervention trial in healthy men. Clinical nutrition (Edinburgh, Scotland) 2010;30(3):303–311. doi: 10.1016/j.clnu.2010.12.010. [DOI] [PubMed] [Google Scholar]
  • 95.Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–585. doi: 10.1038/nm.3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57–63. doi: 10.1038/nature09922. This article presents the first of a series of investigations confirming an important role of gut flora metabolism (potentially modified by diet) in cardiovascular disease development.
  • 97.Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–1584. doi: 10.1056/NEJMoa1109400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Bennett BJ, de Aguiar Vallim TQ, Wang Z, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell metabolism. 2013;17(1):49–60. doi: 10.1016/j.cmet.2012.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Pellis L, van Erk MJ, van Ommen B, et al. Plasma metabolomics and proteomics profiling after a postprandial challenge reveal subtle diet effects on human metabolic status. Metabolomics. 2012;8(2):347–359. doi: 10.1007/s11306-011-0320-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lankinen M, Schwab U, Gopalacharyulu PV, et al. Dietary carbohydrate modification alters serum metabolic profiles in individuals with the metabolic syndrome. Nutr Metab Cardiovasc Dis. 2010;20(4):249–257. doi: 10.1016/j.numecd.2009.04.009. [DOI] [PubMed] [Google Scholar]
  • 101. Krug S, Kastenmuller G, Stuckler F, et al. The dynamic range of the human metabolome revealed by challenges. Faseb J. 2012 doi: 10.1096/fj.11-198093. This article presents important insight to the potential application of metabolomics for nutrition research in a clinical and population setting.
  • 102.Altmaier E, Kastenmuller G, Romisch-Margl W, et al. Questionnaire-based self-reported nutrition habits associate with serum metabolism as revealed by quantitative targeted metabolomics. Eur J Epidemiol. 2011;26(2):145–156. doi: 10.1007/s10654-010-9524-7. [DOI] [PubMed] [Google Scholar]
  • 103.Solanky KS, Bailey NJ, Beckwith-Hall BM, et al. Biofluid 1H NMR-based metabonomic techniques in nutrition research - metabolic effects of dietary isoflavones in humans. J Nutr Biochem. 2005;16(4):236–244. doi: 10.1016/j.jnutbio.2004.12.005. [DOI] [PubMed] [Google Scholar]
  • 104.Stella C, Beckwith-Hall B, Cloarec O, et al. Susceptibility of human metabolic phenotypes to dietary modulation. J Proteome Res. 2006;5(10):2780–2788. doi: 10.1021/pr060265y. [DOI] [PubMed] [Google Scholar]
  • 105.Walsh MC, Brennan L, Pujos-Guillot E, et al. Influence of acute phytochemical intake on human urinary metabolomic profiles. Am J Clin Nutr. 2007;86(6):1687–1693. doi: 10.1093/ajcn/86.5.1687. [DOI] [PubMed] [Google Scholar]
  • 106.Schwab U, Seppanen-Laakso T, Yetukuri L, et al. Triacylglycerol fatty acid composition in diet-induced weight loss in subjects with abnormal glucose metabolism--the GENOBIN study. PLoS One. 2008;3(7):e2630. doi: 10.1371/journal.pone.0002630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Chorell E, Moritz T, Branth S, et al. Predictive metabolomics evaluation of nutritionmodulated metabolic stress responses in human blood serum during the early recovery phase of strenuous physical exercise. J Proteome Res. 2009;8(6):2966–2977. doi: 10.1021/pr900081q. [DOI] [PubMed] [Google Scholar]
  • 108.Redeuil K, Smarrito-Menozzi C, Guy P, et al. Identification of novel circulating coffee metabolites in human plasma by liquid chromatography-mass spectrometry. J Chromatogr A. 2011 doi: 10.1016/j.chroma.2011.05.050. [DOI] [PubMed] [Google Scholar]
  • 109.Nagy K, Redeuil K, Williamson G, et al. First identification of dimethoxycinnamic acids in human plasma after coffee intake by liquid chromatography-mass spectrometry. J Chromatogr A. 2011;1218(3):491–497. doi: 10.1016/j.chroma.2010.11.076. [DOI] [PubMed] [Google Scholar]
  • 110.Stalmach A, Mullen W, Barron D, et al. Metabolite profiling of hydroxycinnamate derivatives in plasma and urine after the ingestion of coffee by humans: identification of biomarkers of coffee consumption. Drug Metab Dispos. 2009;37(8):1749–1758. doi: 10.1124/dmd.109.028019. [DOI] [PubMed] [Google Scholar]
  • 111.Kempf K, Herder C, Erlund I, et al. Effects of coffee consumption on subclinical inflammation and other risk factors for type 2 diabetes: a clinical trial. Am J Clin Nutr. 2010;91(4):950–957. doi: 10.3945/ajcn.2009.28548. [DOI] [PubMed] [Google Scholar]
  • 112.Daykin CA, Van Duynhoven JP, Groenewegen A, et al. Nuclear magnetic resonance spectroscopic based studies of the metabolism of black tea polyphenols in humans. J Agric Food Chem. 2005;53(5):1428–1434. doi: 10.1021/jf048439o. [DOI] [PubMed] [Google Scholar]
  • 113.van Velzen EJ, Westerhuis JA, van Duynhoven JP, et al. Phenotyping tea consumers by nutrikinetic analysis of polyphenolic end-metabolites. J Proteome Res. 2009;8(7):3317–3330. doi: 10.1021/pr801071p. [DOI] [PubMed] [Google Scholar]
  • 114.Martin FP, Rezzi S, Pere-Trepat E, et al. Metabolic effects of dark chocolate consumption on energy, gut microbiota, and stress-related metabolism in free-living subjects. J Proteome Res. 2009;8(12):5568–5579. doi: 10.1021/pr900607v. [DOI] [PubMed] [Google Scholar]
  • 115.Llorach R, Urpi-Sarda M, Tulipani S, et al. Metabolomic fingerprint in patients at high risk of cardiovascular disease by cocoa intervention. Mol Nutr Food Res. 2013 doi: 10.1002/mnfr.201200736. [DOI] [PubMed] [Google Scholar]
  • 116.Tulipani S, Llorach R, Jauregui O, et al. Metabolomics unveils urinary changes in subjects with metabolic syndrome following 12-week nut consumption. J Proteome Res. 2011;10(11):5047–5058. doi: 10.1021/pr200514h. [DOI] [PubMed] [Google Scholar]
  • 117.Johansson-Persson A, Barri T, Ulmius M, et al. LC-QTOF/MS metabolomic profiles in human plasma after a 5-week high dietary fiber intake. Anal Bioanal Chem. 2013;405(14):4799–4809. doi: 10.1007/s00216-013-6874-5. [DOI] [PubMed] [Google Scholar]
  • 118.McCombie G, Browning LM, Titman CM, et al. omega-3 oil intake during weight loss in obese women results in remodelling of plasma triglyceride and fatty acids. Metabolomics. 2009;5(3):363–374. doi: 10.1007/s11306-009-0161-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Altmaier E, Kastenmuller G, Romisch-Margl W, et al. Variation in the human lipidome associated with coffee consumption as revealed by quantitative targeted metabolomics. Mol Nutr Food Res. 2009;53(11):1357–1365. doi: 10.1002/mnfr.200900116. [DOI] [PubMed] [Google Scholar]
  • 120.Adams SH, Hoppel CL, Lok KH, et al. Plasma acylcarnitine profiles suggest incomplete long-chain fatty acid beta-oxidation and altered tricarboxylic acid cycle activity in type 2 diabetic African-American women. J Nutr. 2009;139(6):1073–1081. doi: 10.3945/jn.108.103754. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Fiehn O, Garvey WT, Newman JW, et al. Plasma metabolomic profiles reflective of glucose homeostasis in non-diabetic and type 2 diabetic obese African-American women. PLoS One. 2010;5(12):e15234. doi: 10.1371/journal.pone.0015234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kim JY, Park JY, Kim OY, et al. Metabolic profiling of plasma in overweight/obese and lean men using ultra performance liquid chromatography and Q-TOF mass spectrometry (UPLC-Q-TOF MS) J Proteome Res. 2010;9(9):4368–4375. doi: 10.1021/pr100101p. [DOI] [PubMed] [Google Scholar]
  • 123.Menni C, Zhai G, Macgregor A, et al. Targeted metabolomics profiles are strongly correlated with nutritional patterns in women. Metabolomics. 2013;9(2):506–514. doi: 10.1007/s11306-012-0469-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Heinzmann SS, Merrifield CA, Rezzi S, et al. Stability and robustness of human metabolic phenotypes in response to sequential food challenges. J Proteome Res. 2012;11(2):643–655. doi: 10.1021/pr2005764. This article presents important insight to the potential application of metabolomics for nutrition research in a clinical and population setting.
  • 125.Demirkan A, van Duijn CM, Ugocsai P, et al. Genome-wide association study identifies novel loci associated with circulating phospho- and sphingolipid concentrations. PLoS Genet. 2012;8(2):e1002490. doi: 10.1371/journal.pgen.1002490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Small KS, Hedman AK, Grundberg E, et al. Identification of an imprinted master trans regulator at the KLF14 locus related to multiple metabolic phenotypes. Nat Genet. 2011;43(6):561–564. doi: 10.1038/ng.833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zhong H, Yang X, Kaplan LM, et al. Integrating pathway analysis and genetics of gene expression for genome-wide association studies. Am J Hum Genet. 2010;86(4):581–591. doi: 10.1016/j.ajhg.2010.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Greenawalt DM, Sieberts SK, Cornelis MC, et al. Integrating genetic association, genetics of gene expression, and single nucleotide polymorphism set analysis to identify susceptibility Loci for type 2 diabetes mellitus. Am J Epidemiol. 2012;176(5):423–430. doi: 10.1093/aje/kws123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Yang X. Use of functional genomics to identify candidate genes underlying human genetic association studies of vascular diseases. Arterioscler Thromb Vasc Biol. 2012;32(2):216–222. doi: 10.1161/ATVBAHA.111.232702. [DOI] [PubMed] [Google Scholar]
  • 130.Robinette SL, Holmes E, Nicholson JK, et al. Genetic determinants of metabolism in health and disease: from biochemical genetics to genome-wide associations. Genome medicine. 2012;4(4):30. doi: 10.1186/gm329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Chen R, Mias GI, Li-Pook-Than J, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148(6):1293–1307. doi: 10.1016/j.cell.2012.02.009. This article features the first ‘integrative personal omics profile’ (iPOP) by combining several omic-profiles from a single individual over a 14 month period.
  • 132.Inouye M, Kettunen J, Soininen P, et al. Metabonomic, transcriptomic, and genomic variation of a population cohort. Mol Syst Biol. 2010;6:441. doi: 10.1038/msb.2010.93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Dodd KW, Guenther PM, Freedman LS, et al. Statistical methods for estimating usual intake of nutrients and foods: a review of the theory. J Am Diet Assoc. 2006;106(10):1640–1650. doi: 10.1016/j.jada.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 134. Sampson JN, Boca SM, Shu XO, et al. Metabolomics in epidemiology: sources of variability in metabolite measurements and implications. Cancer Epidemiol Biomarkers Prev. 2013;22(4):631–640. doi: 10.1158/1055-9965.EPI-12-1109. This article discusses important limitations of metabolomics applied to a population setting.
  • 135.Kettunen J, Tukiainen T, Sarin AP, et al. Genome-wide association study identifies multiple loci influencing human serum metabolite levels. Nat Genet. 2012;44(3):269–276. doi: 10.1038/ng.1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Xie W, Wood AR, Lyssenko V, et al. Genetic variants associated with glycine metabolism and their role in insulin sensitivity and type 2 diabetes. Diabetes. 2013;62(6):2141–2150. doi: 10.2337/db12-0876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Ferrucci L, Perry JR, Matteini A, et al. Common variation in the beta-carotene 15,15'-monooxygenase 1 gene affects circulating levels of carotenoids: a genome-wide association study. Am J Hum Genet. 2009;84(2):123–133. doi: 10.1016/j.ajhg.2008.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kapur K, Johnson T, Beckmann ND, et al. Genome-wide meta-analysis for serum calcium identifies significantly associated SNPs near the calcium-sensing receptor (CASR) gene. PLoS Genet. 2010;6(7):e1001035. doi: 10.1371/journal.pgen.1001035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.O'Seaghdha CM, Yang Q, Glazer NL, et al. Common variants in the calcium-sensing receptor gene are associated with total serum calcium levels. Hum Mol Genet. 2010;19(21):4296–4303. doi: 10.1093/hmg/ddq342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Kutalik Z, Benyamin B, Bergmann S, et al. Genome-wide association study identifies two loci strongly affecting transferrin glycosylation. Hum Mol Genet. 2011;20(18):3710–3717. doi: 10.1093/hmg/ddr272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.McLaren CE, Garner CP, Constantine CC, et al. Genome-wide association study identifies genetic loci associated with iron deficiency. PLoS One. 2011;6(3):e17390. doi: 10.1371/journal.pone.0017390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Benyamin B, McRae AF, Zhu G, et al. Variants in TF and HFE explain approximately 40% of genetic variation in serum-transferrin levels. Am J Hum Genet. 2009;84(1):60–65. doi: 10.1016/j.ajhg.2008.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Pichler I, Minelli C, Sanna S, et al. Identification of a common variant in the TFR2 gene implicated in the physiological regulation of serum iron levels. Hum Mol Genet. 2011;20(6):1232–1240. doi: 10.1093/hmg/ddq552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Benyamin B, Ferreira MA, Willemsen G, et al. Common variants in TMPRSS6 are associated with iron status and erythrocyte volume. Nat Genet. 2009;41(11):1173–1175. doi: 10.1038/ng.456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Tanaka T, Roy CN, Yao W, et al. A genome-wide association analysis of serum iron concentrations. Blood. 2010;115(1):94–96. doi: 10.1182/blood-2009-07-232496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Oexle K, Ried JS, Hicks AA, et al. Novel association to the proprotein convertase PCSK7 gene locus revealed by analysing soluble transferrin receptor (sTfR) levels. Hum Mol Genet. 2011;20(5):1042–1047. doi: 10.1093/hmg/ddq538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Meyer TE, Verwoert GC, Hwang SJ, et al. Genome-wide association studies of serum magnesium, potassium, and sodium concentrations identify six Loci influencing serum magnesium levels. PLoS Genet. 2010;6(8) doi: 10.1371/journal.pgen.1001045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Mondul AM, Yu K, Wheeler W, et al. Genome-wide association study of circulating retinol levels. Hum Mol Genet. 2011;20(23):4724–4731. doi: 10.1093/hmg/ddr387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Hazra A, Kraft P, Selhub J, et al. Common variants of FUT2 are associated with plasma vitamin B12 levels. Nat Genet. 2008;40(10):1160–1162. doi: 10.1038/ng.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tanaka T, Scheet P, Giusti B, et al. Genome-wide association study of vitamin B6, vitamin B12, folate, and homocysteine blood concentrations. Am J Hum Genet. 2009;84(4):477–482. doi: 10.1016/j.ajhg.2009.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Lin X, Lu D, Gao Y, et al. Genome-wide association study identifies novel loci associated with serum level of vitamin B12 in Chinese men. Hum Mol Genet. 2012;21(11):2610–2617. doi: 10.1093/hmg/dds062. [DOI] [PubMed] [Google Scholar]
  • 152.Ahn J, Yu K, Stolzenberg-Solomon R, et al. Genome-wide association study of circulating vitamin D levels. Hum Mol Genet. 2010;19(13):2739–2745. doi: 10.1093/hmg/ddq155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Wang TJ, Zhang F, Richards JB, et al. Common genetic determinants of vitamin D insufficiency: a genome-wide association study. Lancet. 2010;376(9736):180–188. doi: 10.1016/S0140-6736(10)60588-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Lasky-Su J, Lange N, Brehm JM, et al. Genome-wide association analysis of circulating vitamin D levels in children with asthma. Hum Genet. 2012;131(9):1495–1505. doi: 10.1007/s00439-012-1185-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Major JM, Yu K, Chung CC, et al. Genome-wide association study identifies three common variants associated with serologic response to vitamin E supplementation in men. J Nutr. 2012;142(5):866–871. doi: 10.3945/jn.111.156349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Major JM, Yu K, Wheeler W, et al. Genome-wide association study identifies common variants associated with circulating vitamin E levels. Hum Mol Genet. 2011;20(19):3876–3883. doi: 10.1093/hmg/ddr296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Baik I, Cho NH, Kim SH, et al. Genome-wide association studies identify genetic loci related to alcohol consumption in Korean men. Am J Clin Nutr. 2011;93(4):809–816. doi: 10.3945/ajcn.110.001776. [DOI] [PubMed] [Google Scholar]
  • 158.Takeuchi F, Isono M, Nabika T, et al. Confirmation of ALDH2 as a Major locus of drinking behavior and of its variants regulating multiple metabolic phenotypes in a Japanese population. Circ J. 2011;75(4):911–918. doi: 10.1253/circj.cj-10-0774. [DOI] [PubMed] [Google Scholar]
  • 159.Yang X, Lu X, Wang L, et al. Common variants at 12q24 are associated with drinking behavior in Han Chinese. Am J Clin Nutr. 2013;97(3):545–551. doi: 10.3945/ajcn.112.046482. [DOI] [PubMed] [Google Scholar]
  • 160.Schumann G, Coin LJ, Lourdusamy A, et al. Genome-wide association and genetic functional studies identify autism susceptibility candidate 2 gene (AUTS2) in the regulation of alcohol consumption. Proc Natl Acad Sci U S A. 2011;108(17):7119–7124. doi: 10.1073/pnas.1017288108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Sulem P, Gudbjartsson DF, Geller F, et al. Sequence variants at CYP1A1-CYP1A2 and AHR associate with coffee consumption. Hum Mol Genet. 2011 doi: 10.1093/hmg/ddr086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Amin N, Byrne E, Johnson J, et al. Genome-wide association analysis of coffee drinking suggests association with CYP1A1/CYP1A2 and NRCAM. Molecular psychiatry. 2011 doi: 10.1038/mp.2011.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Chu AY, Workalemahu T, Paynter NP, et al. Novel locus including FGF21 is associated with dietary macronutrient intake. Hum Mol Genet. 2013 doi: 10.1093/hmg/ddt032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Hindorff LA, Sethupathy P, Junkins HA, et al. Potential etiologic and functional implications of genome-wide association loci for human diseases and traits. Proc Natl Acad Sci U S A. 2009;106(23):9362–9367. doi: 10.1073/pnas.0903103106. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES