Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 1.
Published in final edited form as: Curr Atheroscler Rep. 2015 May;17(5):505. doi: 10.1007/s11883-015-0505-x

Nutri(meta)genetics and cardiovascular disease: novel concepts in the interaction of diet and genomic variation

Jacob Joseph *,ǂ, Joseph Loscalzo *
PMCID: PMC4401039  NIHMSID: NIHMS680732  PMID: 25782777

Abstract

In addition to the interaction of nutrition and genetic variation on the genesis and natural history of cardiovascular disease, recent studies have revealed an entire, new genome that resides in the trillions of microbes that exist in various human habitats, predominantly in the gut that may also contribute to the pathogenesis of cardiovascular disease. This microbial genome and the proteins for which it codes have important functions in homeostatic adaptations to the past and present changes in diet and environment accompanying human civilization. Both preclinical and clinical investigations suggest the role of commensal microbiota in promoting adverse cardiovascular risk. Specifically, microbial metabolism of methylated amines leads to direct pro-atherogenic effects in humans. Further investigations are needed to understand the complex relationships among nutritional status, genetic variation, and the microbial genome, which may explain the recent negative results of clinical trials of nutritional interventions such as B-vitamin therapy to lower plasma homocysteine levels. The results of such contemporary genomic investigations would allow us to utilize personalized nutritional interventions to reduce cardiovascular risk.

Keywords: genetic variation, genome microbiome, metagenome, nutrition, trimethylamine oxide, choline, phosphatidyl choline, betaine, flavin monooxygenase, cardiovascular risk, obesity, diabetes, atherosclerosis, homocysteine, B-vitamins

Introduction

The last decade has shown that the human gene pool associates with that of microbes that live as commensals in the human body, predominantly in the gastrointestinal system [1, 2]. In fact, the genetic repertoire of microbes far exceeds the number of human genes. Using high-throughput sequencing to analyze the microbiome in fecal samples obtained from 124 European individuals, Qin and colleagues characterized 3.3 million non-redundant microbial gene sequences (approximately 150 times the number of human genes) derived from 576.7 gigabases of DNA sequence [3]. High-throughput sequencing methods have obviated the need for stool culture to decipher the microbial genome, and have allowed identification of the taxonomy and genetics of the human microbiota. The two phyla, Bacteroides and Firmicutes (which are gram positive bacteria and include both anaerobic bacteria such as Clostridia and aerobic bacteria such as Bacilli), account for approximately 90% of microbial species inhabiting the human gut. The Human Microbiome Project Consortium recently reported a comprehensive analysis of 4,788 specimens from multiple habitats obtained from 242 healthy Western adults [4]. Similar to a previous small study [5], microbiomes were found to be fairly unique to individuals even in a somewhat homogeneous population. There were significant differences in percentages of bacterial phyla between habitats in an individual; however, metabolic pathways related to the microbiome were more concordant between habitats. Establishment of a stable gut microbial composition may occur over the initial years of life [6]. Although antibiotic treatment disrupts gut microbial ecology, there is a return towards the previous state within a week of discontinuation of antibiotics, although the return is not complete and results in a 'new' microbiome [7]. It is not clear as to what would be the impact of multiple courses on antibiotic treatment on the individual and the population.

Studies as described below clearly demonstrate the importance of these microbial genes in human health, for which reason the combination of human and microbial genomes has been termed the human metagenome. In this review, we will detail current knowledge about the human metagenome as it relates to human health and cardiovascular disease.

Human metagenome: herald of health and harbinger of disease

Through human history and continuing into the present day, the human diet has varied temporally and geographically as food sources have changed. The gut microbiota have evolved in concert with these changes, and is crucial for metabolizing certain nutrients for which human intestinal enzymes do not exist, for harvesting energy from nutrients, and for synthesizing vitamins [8, 9]. It is not clear whether metabolism of various xenobiotics protects humans from diseases or increase susceptibility to disease; however, microbiota do affect the metabolism of common drugs. In addition, metabolism of intestinal contents, such as bile acids, by gut microbiota affects lipid metabolism and modulate the effects of lipid-modifying drugs, such as statins [10, 11]. In addition, the gut is an area of significant antigen exposure, and the human gut microbiota are thought to play crucial roles in innate and adaptive immunity as a result [1214]. This view suggests an important role for gut microbiota in inflammatory bowel diseases [1517] and extra-intestinal diseases with an immune component, such as asthma [18]. Recent studies also show an important role for microbiota in regulating neural and behavioral responses [19]. An intriguing relationship between the microbiome and cardiovascular disease was suggested by the observation that germ free mice have lower oxygen consumption, lower cardiac output, and smaller hearts [2022]. Further studies are needed to determine if the microbiome influences remodeling of the heart.

Nutrition, metagenome, and cardiovascular risk factors

Le Chatelier and colleagues demonstrated that low bacterial species diversity is associated with adiposity, insulin resistance, dyslipidemia and inflammatory phenotype in Danish individuals [23]. A small number of bacterial species accounted for the differences between individuals with low and high bacterial diversity. Preclinical studies have also focused on this relationship between obesity and microbial genome. For example, Backhed and colleagues showed that transplantation of normal cecal microbiota from normal mice to adult germ free mice led to a 60% increase in body fat content as well as insulin resistance despite a lack of increase in caloric intake [24]. This study suggested that increased absorption of monosaccharides and consequent hepatic lipogenesis and suppression of production of fasting-induced adipocyte factor, a circulating lipoprotein lipase, may be responsible for this adiposity response. Studies of genetically obese and lean mice have shown that obese gut microbiota are characterized by a decrease in Bacteroides and an increase in Firmicutes compared to lean mice [25]. The same group also demonstrated that obese microbiota conferred an increase in energy harvested from the diet, and that colonization of germ free mice with gut microbiota from obese mice led to increased adiposity compared to gut microbiota from lean animals [26].

In addition to the association with insulin resistance mentioned above, multiple pre-clinical and human studies demonstrate a role for the microbiome in the pathogenesis of diabetes. The microbiome signature in children with type I diabetes is significantly different from that of healthy children [27]. The interaction between innate immunity and the microbiome has been shown to be crucial for development of type I diabetes [28]. In a study of adult onset diabetes, Karlsson and colleagues examined the microbiome from fecal samples of European women with normal or impaired glucose control and overt diabetes [29]. They observed significant differences in the microbiome composition of women with diabetes, and were able to develop a mathematical model of the microbiome to predict a diabetic metabolic profile in women with impaired glucose tolerance. The authors applied this model to a Chinese cohort and discovered that microbiomic markers for diabetes differed between Chinese and European subjects.

Nutrition, metagenome, and atherosclerosis

A recent meta-analysis of 21 prospective epidemiological studies with a cumulative population of approximately 350,000 subjects and 5–23 years of follow-up data showed that there is no significant association between dietary intake of saturated fat and risk of coronary artery disease, stroke, or cardiovascular disease (CVD) [30]. A likely explanation, as the authors contend in another publication, could be that saturated fat was replaced by other nutrients that increased atherogenic risk [31]. Alternatively, it is possible that genetic or metagenomic variation interacts with nutrients to promote atherogenesis. For example, in a study of 13 controls and 12 subjects with symptomatic carotid atherosclerosis, analysis of the gut microbiome demonstrated a preponderance of the genus Colinsella (belonging to the phylum Actinobacteria) in patients with atherosclerosis and of the genuses Roseburia (butyrate-producing bacteria that belongs to the phylum Firmicutes) and Eubacterium (Firmicutes phylum) in healthy controls [32].

One of the more exciting developments in this area has been the demonstration of how the gut microbiome interacts with nutrients to promote atherosclerosis. Wang and colleagues demonstrated that metabolism of choline, a common nutrient, by gut microbiota promotes atherosclerosis ˙˙[33]. Metabolomic studies were utilized to show that trimethylamine oxide (TMAO), a common metabolite of choline and phosphatidyl choline, was associated with an increased risk of CVD. Utilizing germ-free mice, the investigators proved the necessity of gut microbial flora for production of TMAO from choline. TMAO was found to increase the accumulation of cholesterol in macrophages and consequent foam cell formation. In addition to TMAO, plasma levels of choline as well as betaine, a product of choline oxidation, were associated with an increased risk of atherosclerosis in humans. The same group further examined the clinical implications of these findings in human subjects ˙˙[34]. In a study of 40 healthy volunteers, they demonstrated that dietary challenge with phosphatidyl choline increased the plasma levels of TMAO. Antibiotic treatment to suppress gut flora markedly decreased plasma levels of TMAO. Analysis of TMAO levels in 4,007 patients undergoing elective coronary angiography showed a significant relationship between an elevated TMAO level and the risk of major cardiovascular events during three years of follow-up.

In a separate study, Dr. Hazen’s group also examined whether gut microbial metabolism of L-carnitine, a trimethylamine found abundantly in red meat and essential to fatty acid transport, promotes atherosclerosis ˙˙[35]. In this study, human vegans given a carnitine load (8 oz. steak) did not have an elevation of plasma TMAO levels while omnivorous human volunteers demonstrated a significant elevation in TMAO levels. Antibiotic treatment to suppress microbiota attenuated TMAO production after carnitine challenge. The composition of gut microbiota varied between omnivores/individuals with higher plasma TMAO levels vs. vegans/individuals with lower TMAO levels. In experiments performed in mice, supplementation of L-carnitine was found to alter gut microbiota, increase synthesis of TMAO, and promote atherogenesis by inhibiting reverse cholesterol transport in vivo. A novel Rieske-type protein with oxygenase/reductase function has been shown to effect the conversion of carnitine to trimethylamine (TMA), the precursor of TMAO, by human gut microbiota [36]. This discovery suggests the potential utility of specific genes in the microbiome as markers of atherosclerotic risk or as therapeutic targets. Another potential target is flavin monooxygenase (FMO). Since an FMO3 mutation and decreased function is responsible for the fish odor syndrome or trimethylaminuria due to impaired metabolism and accumulation of trimethylamine (TMA) [37], it is likely that FMOs are responsible for oxidation of gut microbiota-derived trimethylamine (TMA) to TMAO. A study by Bennett and colleagues showed that FMO3 was the key enzyme responsible for TMAO production in mice [38]. In a study of French-Canadian adults, 3 polymorphisms of FMO3 were identified that were associated with variations in TMAO production [39].

These studies clearly demonstrate that gut microbiota promote atherosclerosis by modification of dietary components that segregate with known dietary risk factors such as fat [40]. Gut microbiota composition is dynamic, with enrichment of microbial species that metabolize specific dietary components. These studies clearly establish gut microbiota as a target for cardiovascular prevention and treatment using nutritional manipulation. Genetic variation in enzymes that modify microbiota-derived metabolites would be crucial in determining the effects of nutritional modulation.

Microbiome, methionine-homocysteine cycle, and methylation

Interaction of nutritional status with genetic variation in the methionine-homocysteine cycle has been investigated as a pathogenic factor in atherosclerosis since the initial reports by Dr. McCully linking homocystinuria to arterial vascular disease [1]. Although multiple observational studies have shown a relationship between plasma homocysteine levels and atherothrombotic risk, nutritional intervention utilizing combinations of B-vitamins to decrease the plasma homocysteine level has not been demonstrated to improve cardiovascular outcomes (reviewed in detail in [42, 43]). One potential reason for lack of effect of B-vitamins could be genetic variability. For example, a study of folate supplementation in healthy female identical and non-identical twins showed that changes in plasma homocysteine level in response to folate was highly heritable [44]. The variability was not based on polymorphisms of methylene tetrahydrofolate, a well-known heritable factor affecting homocysteine metabolism, underscoring the presence of unidentified genetic factors affecting methionine-homocysteine cycle in humans.

Changes in plasma homocysteine occur due to a variety of genetic and environmental factors, suggesting that the methionine-homocysteine cycle is crucial to health and perturbed in various pathological conditions [42, 43]. The major function of the methionine-homocysteine cycle is the regulation of methylation reactions, with S-adenosylmethionine donating methyl groups for all methylation reactions of the body except remethylation of homocysteine to methionine. Choline and its oxidation product, betaine trimethylglycine, provide exogenous methyl groups, are crucial for methyl supply, and interact closely with the methionine-homocysteine cycle. In fact, betaine is the methyl donor for remethylation of homocysteine by the enzyme betaine-homocysteine methyl transferase. Plasma levels of homocysteine, choline, and betaine have been linked to cardiovascular risk. A recent report examined the associations of plasma choline, betaine, and TMAO with the risk of major adverse cardiac events in over 3,000 subjects undergoing coronary angiography [45]. Choline and betaine were associated with TMAO levels and with increased risk of cardiac events; however, the relationships of choline and betaine with cardiovascular risk were rendered non-significant when TMAO was added as a covariate, indicating that the predictive value of choline and betaine were due to microbiota-derived TMAO production.

In the RISTOMED study, a European study that aimed to use dietary modifications to reduce aging-induced diseases in older healthy adults, a personalized diet with or without probiotics was administered and the effects on inflammatory markers, oxidant stress, B-vitamins, and homocysteine were measured [46]. Administration of probiotics increased the prevalence of bifidobacteria in the gut microbiota, as well as folate, and B12 levels, and decreased plasma homocysteine levels, suggesting a close relationship between gut microbiome composition and the methionine-homocysteine cycle. These studies suggest that the gut microbiome interacts closely with the methionine-homocysteine cycle and methyl balance in vivo, and highlights the need to study the gut microbiome in investigations of the relationship between hyperhomocysteinemia and CVD.

Human gut microbial genome: a modifiable risk factor?

Differences in gut microbiota in response to dietary variation and interventions suggest that the microbiome is modifiable. A study of children from Africa (Burkina Faso) and Europe (Italy) showed the impact of diet on gut microbiota across populations [47]. Children from Burkina Faso who eat a fiber-rich diet had over-representation of Bacteroides and less Firmicutes compared to European children on a western diet. In another study, weight loss induced by caloric restriction and high fiber intake was shown concomitantly to increase gut microbial diversity and improve lipid and glucose metabolism in obese individuals [48]. In studies using germ-free mice, Backhed and colleagues demonstrated that germ-free mice are resistant to obesity induced by a western diet while colonization of germ-free mice with gut microbiota of normal mice led to increased fat deposition [49]. Gut microbiota have also been proposed to possess specific anti-atherogenic effects – metabolism of an anthocyanin (pigments derived from grapes and responsible for the color of red wine) by microbes and generation of protocatechuic acid has been shown to promote reverse cholesterol transport in mice [50]. Overall, these studies suggest the close relationship between diet and the microbiome, and the potential for modifying the gut microbiome by diet to induce a favorable diet-metabolism interaction.

Nutrition, human microbial genome and genetics: an integrated target for cardiovascular prevention?

Since metagenomic variation among humans includes not only human genomic variation, but also variation in the microbial genome, it is likely that the human genome, microbiome, and nutritional status interact to influence health and disease. For example, mice with genetic deficiency of Toll-like receptor 5, an important component of the innate immune response in the gut, develop characteristic gut microbiota that confer susceptibility to metabolic syndrome [51]. In humans, bile acids derived from gut microbiota metabolism have been shown to affect host response to simvastatin [11]. This pharmacogenetic study demonstrated that three secondary bile acids derived from microbiota could be used to predict a good response to simvastatin, and that levels of simvastatin and bile acids were correlated. Interestingly, a single nucleotide polymorphism in a gene coding for an organic anion transporter was associated with levels of bile acids. In terms of atherogenic TMAO levels, a human genome-wide association study did not reveal a definitive relationship of genetic variation with plasma TMAO levels [52], which, in light of earlier experimental work reviewed here, suggests that other metagenomic and/or environmental/nutritional factors must be considered to define disease risk optimally.

Twins who are discordant for the obesity phenotype provide a unique opportunity to investigate the relationship between the gut microbiome and nutritional status independent of genetic variation. Gut microbial composition is similar between monozygotic and dizygotic twins and was also similar in mothers indicating the effect of familial environment in establishing the microbiota signature [53]. Ridaura and colleagues identified four twin pairs who had sustained differences in body mass index, and collected fecal samples from them [54]. When fecal microbiota from obese and lean twin pairs were introduced by gavage into germ free mice, the mice that received gut microbiota from obese individuals developed an obesity phenotype. Housing mice that had received obese and lean microbiota in the same cage led to reestablishment of the lean phenotype in these coprophagic mice, indicating that the gut microbiome is a rapidly modifiable factor that modifies phenotype.

Another clinically relevant situation in which nutritional status and the microbiome interact longitudinally is that following gastric bypass surgery in obese patients. Furet and colleagues analyzed the gut microbiota from 13 lean individuals and from 30 obese individuals before and after gastric bypass surgery [55]. In addition to changes in the microbiota profile at baseline between lean and obese subjects, the investigators noticed significant changes in microbiota composition at three months after surgery that was correlated with fat mass independent of food intake.

Similar to genome-wide association studies, microbiome-wide association studies have been attempted to identify linkage between microbiome variation and disease. Qin and coworkers performed shotgun sequencing of the gut microbiome in a Chinese population and identified approximately 60,000 microbiome markers associated with type-2 diabetes mellitus [56]. Gut microbiome markers were found to be useful in differentiating healthy and diabetic subjects with a high level of specificity.

Systems biology or network medicine approaches hold the potential for improved understanding of biological determinants of phenotype owing to the great power of these approaches in holistically evaluating the complexity of the multiple genomic and environmental determinants of phenotype 57, 58]. Greenblum and coworkers obtained microbiome data from 124 unrelated European individuals [59]. A metabolic network was derived by identifying genes coding for enzymes and pooling data from all individual microbiomes studied. In this network, nodes represented enzymes and connections between nodes (or directed edges) represented enzymes that catalyzed sequential reactions. Differences in organization of the network and of gene distribution within the network varied between lean and obese individuals, indicating the potential of network approaches to identify differences in microbiomes between health and disease.

Conclusions

The human microbiome is a complex ecosystem that co-exists with the human genome and is subject to rapid change depending on individual and population variations in dietary intake. In addition to diet, genetic variation, especially in immune mechanisms, may also affect gut microbiota. Perpetuation of major cardiovascular risk factors such as obesity and diabetes may be dependent on the close interaction between diet and microbial augmentation of energy harvesting from foodstuffs. Further studies are needed to understand the many potential interrelationships among nutritional status, genetic variation, and the microbiome. Modulation of these interactions may lead to great strides in the prevention and treatment of CVD.

Acknowledgements

This work was supported by NIH grants R37 HL 061795, U01 HG007690, and HL048743.

Footnotes

The authors have no conflicts of interest.

Compliance with Ethical Guidelines:

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

References

Important references published in the past 3 years are highlighted as follows:

˙ Of importance

˙˙ Of major importance

  • 1.Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489:220–230. doi: 10.1038/nature11550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.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]
  • 3.Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, et al. 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]
  • 4.Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. doi: 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Costello EK, Lauber CL, Hamady M, Fierer N, Gordon JI, Knight R. Bacterial community variation in human body habitats across space and time. Science. 2009;326:1694–1697. doi: 10.1126/science.1177486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, Angenent LT, Ley RE. Succession of microbial consortia in the developing infant gut microbiome. Proc Nat Acad Sci U S A. 2011;108(Suppl 1):4578–4585. doi: 10.1073/pnas.1000081107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4554–4561. doi: 10.1073/pnas.1000087107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449:804–810. doi: 10.1038/nature06244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hehemann JH, Correc G, Barbeyron T, Helbert W, Czjzek M, Michel G. Transfer of carbohydrate-active enzymes from marine bacteria to Japanese gut microbiota. Nature. 2010;464:908–912. doi: 10.1038/nature08937. [DOI] [PubMed] [Google Scholar]
  • 10.Martin FP, Dumas ME, Wang Y, Legido-Quigley C, Yap IK, Tang H, Zirah S, Murphy GM, Cloarec O, Lindon JC, et al. A top-down systems biology view of microbiome-mammalian metabolic interactions in a mouse model. Molec Syst Biol. 2007;3:112. doi: 10.1038/msb4100153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kaddurah-Daouk R, Baillie RA, Zhu H, Zeng ZB, Wiest MM, Nguyen UT, Wojnoonski K, Watkins SM, Trupp M, Krauss RM. Enteric microbiome metabolites correlate with response to simvastatin treatment. PloS One. 2011;6:e25482. doi: 10.1371/journal.pone.0025482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Round JL, Lee SM, Li J, Tran G, Jabri B, Chatila TA, Mazmanian SK. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science. 2011;332:974–977. doi: 10.1126/science.1206095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Lee YK, Mazmanian SK. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science. 2010;330:1768–1773. doi: 10.1126/science.1195568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Slack E, Hapfelmeier S, Stecher B, Velykoredko Y, Stoel M, Lawson MA, Geuking MB, Beutler B, Tedder TF, Hardt WD, et al. Innate and adaptive immunity cooperate flexibly to maintain host-microbiota mutualism. Science. 2009;325:617–620. doi: 10.1126/science.1172747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sokol H, Seksik P, Rigottier-Gois L, Lay C, Lepage P, Podglajen I, Marteau P, Dore J. Specificities of the fecal microbiota in inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:106–111. doi: 10.1097/01.MIB.0000200323.38139.c6. [DOI] [PubMed] [Google Scholar]
  • 16.Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474:298–306. doi: 10.1038/nature10208. [DOI] [PubMed] [Google Scholar]
  • 17.Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, Vandamme P, Vermeire S. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut. 2011;60:631–637. doi: 10.1136/gut.2010.223263. [DOI] [PubMed] [Google Scholar]
  • 18.Russell SL, Gold MJ, Hartmann M, Willing BP, Thorson L, Wlodarska M, Gill N, Blanchet MR, Mohn WW, McNagny KM, et al. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep. 2012;13:440–447. doi: 10.1038/embor.2012.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bravo JA, Forsythe P, Chew MV, Escaravage E, Savignac HM, Dinan TG, Bienenstock J, Cryan JF. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci USA. 2011;108:16050–16055. doi: 10.1073/pnas.1102999108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wostmann BS, Bruckner-Kardoss E, Knight PL., Jr Cecal enlargement, cardiac output, and O2 consumption in germfree rats. Proc Soc Exp Biol Med. 1968;128:137–141. doi: 10.3181/00379727-128-32962. [DOI] [PubMed] [Google Scholar]
  • 21.Bruckner-Kardoss E, Wostmann BS. Oxygen consumption of germfree and conventional mice. Lab Animal Sci. 1978;28:282–286. [PubMed] [Google Scholar]
  • 22.Wostmann BS, Bruckner-Kardoss E, Pleasants JR. Oxygen consumption and thyroid hormones in germfree mice fed glucose-amino acid liquid diet. J Nutr. 1982;112:552–559. doi: 10.1093/jn/112.3.552. [DOI] [PubMed] [Google Scholar]
  • 23.Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, Almeida M, Arumugam M, Batto JM, Kennedy S, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–546. doi: 10.1038/nature12506. [DOI] [PubMed] [Google Scholar]
  • 24.Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 2004;101:15718–15723. doi: 10.1073/pnas.0407076101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ley RE, Backhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci USA. 2005;102:11070–11075. doi: 10.1073/pnas.0504978102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–1031. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
  • 27.Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyoty H, et al. Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 2011;5:82–91. doi: 10.1038/ismej.2010.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA, et al. 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]
  • 29.Karlsson FH, Tremaroli V, Nookaew I, Bergstrom G, Behre CJ, Fagerberg B, Nielsen J, Backhed F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498:99–103. doi: 10.1038/nature12198. [DOI] [PubMed] [Google Scholar]
  • 30.Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Meta-analysis of prospective cohort studies evaluating the association of saturated fat with cardiovascular disease. Am J Clin Nutr. 2010;91:535–546. doi: 10.3945/ajcn.2009.27725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Siri-Tarino PW, Sun Q, Hu FB, Krauss RM. Saturated fatty acids and risk of coronary heart disease: modulation by replacement nutrients. Curr Athero Rep. 2010;12:384–390. doi: 10.1007/s11883-010-0131-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Karlsson FH, Fak F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, Backhed 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]
  • 33. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922. ˙˙This "bedside to bench" study utilized metabolomics approaches to identify a plasma metabolite associated with increased cardiovascular risk, and, subsequently, using germ-free mice, demonstrated that the metabolite trimethylamine oxide was derived from metabolism of phosphatidyl choline, a common dietary component, by gut microbiota, and promoted atherosclerosis.
  • 34. 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. ˙˙ This seminal human study demonstrated that human gut microbiota were crucial in generating trimethylamine oxide from dietary phosphatidylcholine. This report also showed the association of elevated plasma levels of trimethylamine oxide with the risk of cardiovascular events.
  • 35. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–585. doi: 10.1038/nm.3145. ˙˙ This original report demonstrated that omnivorous humans generated more trimethylamine from dietary carnitine compared to vegans and that gut microbiota were crucial to this effect. Using a mouse model, the investigators also showed the interaction of dietary carnitine and gut microbiota in promoting atherosclerosis. This study demonstrated the role of microbiota in the link between consumption of red meat and the risk of atherosclerosis.
  • 36.Zhu Y, Jameson E, Crosatti M, Schafer H, Rajakumar K, Bugg TD, Chen Y. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc Natl Acad Sci USA. 2014;111:4268–4273. doi: 10.1073/pnas.1316569111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Treacy EP, Akerman BR, Chow LM, Youil R, Bibeau C, Lin J, Bruce AG, Knight M, Danks DM, Cashman JR, et al. Mutations of the flavin-containing monooxygenase gene (FMO3) cause trimethylaminuria, a defect in detoxication. Hum Molec Genet. 1998;7:839–845. doi: 10.1093/hmg/7.5.839. [DOI] [PubMed] [Google Scholar]
  • 38.Bennett BJ, de Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, Allayee H, Lee R, Graham M, Crooke R, et al. 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]
  • 39.Lambert DM, Mamer OA, Akerman BR, Choiniere L, Gaudet D, Hamet P, Treacy EP. In vivo variability of TMA oxidation is partially mediated by polymorphisms of the FMO3 gene. Molec Genet Metab. 2001;73:224–229. doi: 10.1006/mgme.2001.3189. [DOI] [PubMed] [Google Scholar]
  • 40.Loscalzo J. Lipid metabolism by gut microbes and atherosclerosis. Circ Res. 2011;109:127–129. doi: 10.1161/RES.0b013e3182290620. [DOI] [PubMed] [Google Scholar]
  • 41.McCully KS. Vascular pathology of homocysteinemia: implications for the pathogenesis of arteriosclerosis. Am J Pathol. 1969;56:111–128. [PMC free article] [PubMed] [Google Scholar]
  • 42.Joseph J, Handy DE, Loscalzo J. Quo vadis: whither homocysteine research? Cardiovasc Toxicol. 2009;9:53–63. doi: 10.1007/s12012-009-9042-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Joseph J, Loscalzo J. Methoxistasis: integrating the roles of homocysteine and folic acid in cardiovascular pathobiology. Nutrients. 2013;5:3235–3256. doi: 10.3390/nu5083235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cotlarciuc I, Andrew T, Dew T, Clement G, Gill R, Surdulescu G, Sherwood R, Ahmadi KR. The basis of differential responses to folic acid supplementation. J Nutrigenet Nutrigenomics. 2011;4:99–109. doi: 10.1159/000327768. [DOI] [PubMed] [Google Scholar]
  • 45.Wang Z, Tang WH, Buffa JA, Fu X, Britt EB, Koeth RA, Levison BS, Fan Y, Wu Y, Hazen SL. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur Heart J. 2014;35:904–910. doi: 10.1093/eurheartj/ehu002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Valentini L, Pinto A, Bourdel-Marchasson I, Ostan R, Brigidi P, Turroni S, Hrelia S, Hrelia P, Bereswill S, Fischer A, et al. Impact of personalized diet and probiotic supplementation on inflammation, nutritional parameters and intestinal microbiota - The "RISTOMED project": Randomized controlled trial in healthy older people. Clin Nutr. 2014 doi: 10.1016/j.clnu.2014.09.023. [Epub ahead of print 12/03/14]. [DOI] [PubMed] [Google Scholar]
  • 47.De Filippo C, Cavalieri D, Di Paola M, Ramazzotti M, Poullet JB, Massart S, Collini S, Pieraccini G, Lionetti P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci USA. 2010;107:14691–14696. doi: 10.1073/pnas.1005963107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, Almeida M, Quinquis B, Levenez F, Galleron N, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500:585–588. doi: 10.1038/nature12480. [DOI] [PubMed] [Google Scholar]
  • 49.Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci USA. 2007;104:979–984. doi: 10.1073/pnas.0605374104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang D, Xia M, Yan X, Li D, Wang L, Xu Y, Jin T, Ling W. Gut microbiota metabolism of anthocyanin promotes reverse cholesterol transport in mice via repressing miRNA-10b. Circ Res. 2012;111:967–981. doi: 10.1161/CIRCRESAHA.112.266502. [DOI] [PubMed] [Google Scholar]
  • 51.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]
  • 52.Hartiala J, Bennett BJ, Tang WH, Wang Z, Stewart AF, Roberts R, McPherson R, Lusis AJ, Hazen SL, Allayee H. Comparative genome-wide association studies in mice and humans for trimethylamine N-oxide, a proatherogenic metabolite of choline and L-carnitine. Arterioscl Thromb Vasc Biol. 2014;34:1307–1313. doi: 10.1161/ATVBAHA.114.303252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, et al. 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]
  • 54.Ridaura VK, Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, et al. Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science. 2013;341 doi: 10.1126/science.1241214. 1241214-1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Furet JP, Kong LC, Tap J, Poitou C, Basdevant A, Bouillot JL, Mariat D, Corthier G, Dore J, Henegar C, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59:3049–3057. doi: 10.2337/db10-0253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. doi: 10.1038/nature11450. [DOI] [PubMed] [Google Scholar]
  • 57.Barabasi AL, Gulbahce N, Loscalzo J. Network medicine: a network-based approach to human disease. Nat Rev Genet. 2011;12:56–68. doi: 10.1038/nrg2918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Borenstein E. Computational systems biology and in silico modeling of the human microbiome. Brief Bionform. 2012;13:769–780. doi: 10.1093/bib/bbs022. [DOI] [PubMed] [Google Scholar]
  • 59.Greenblum S, Turnbaugh PJ, Borenstein E. Metagenomic systems biology of the human gut microbiome reveals topological shifts associated with obesity and inflammatory bowel disease. Proc Natl Acad Sci USA. 2012;109:594–599. doi: 10.1073/pnas.1116053109. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES