Abstract
Over the past decade, the gut microbiome has emerged as a novel and largely untapped source of variability for metabolic and cardiovascular disease risk, including diabetes. Animal and human studies support several possible pathways through which the gut microbiome may impact health, including the production of health-related metabolites from dietary sources. Diet is considered important to shaping the gut microbiota; in addition, gut microbiota influence the metabolism of many dietary components. In the present paper, we address the distinction between compositional and functional analysis of the gut microbiota. We focus on literature that highlights the value of moving beyond surveys of microbial composition to measuring gut microbial functioning to delineate mechanisms related to the interplay between diet and gut microbiota in cardiometabolic health.
Keywords: Gut microbiota, metabolites, nutrition, diabetes, cardiovascular disease
Introduction
There is an intimate relation between diet and the gut flora. Gut microbial communities have co-evolved with host species to provide unique metabolic functions, as reflected in broad patterns of food consumption, with distinct microbiota among carnivores, omnivores, and herbivores[1, 2]. Geographic surveys in humans reveal differences in microbiota corresponding to the extent of plant and animal consumption or Westernization of diet[3–5]. On an individual level, dietary composition contributes, at least in part, to shaping the gut microbiota through the delivery of energy for microbial growth[6–8]. In turn, gut microbiota influence the production of health-related metabolites from dietary components[9, 10], with the potential for personal variability in metabolite production based on differences in gut microbiota[11, 12]. A growing literature supports a role for gut microbiota in the development and progression of metabolic and cardiovascular disease risk[13–16], including type 2 diabetes[17–19], obesity[20–22], insulin resistance[23], inflammation[24], atherosclerosis[12, 25], hypertension[26, 27], dyslipidemia[28], and cardiovascular disease events[29, 30]. The purpose of the current review is to frame a discussion around the complex interplay between diet and and available assessments of the gut microbiota in pathways to metabolic disease. Additionally, we will review evidence supporting the growing emphasis in the field to move from surveys of taxonomic composition of the microbial community to measuring microbiota functional potential and activity for improved understanding of health-related pathways through microbiota.
Studying the gut microbiota, diet, and cardiometabolic health: From microbiota composition to function
Measuring the gut microbiota: a brief introduction
Historically, study of the gut microbiota focused on distinct organisms or a set of specific organisms using culture-dependent approaches[31–33]. However, time- and labor-intensive culture-based methods require the development of appropriate media for the laboratory cultivation of specific microorganisms, limiting the scope of the gut community that can be studied. The introduction of culture-independent methods allowed the direct isolation of microbial DNA from collected samples, such as fecal samples for study of the gut microbiota, and enabled researchers to begin to characterize the larger gut microbial community[34]. This work provided key insights into taxonomic composition and functionality of the gut microbiota as a whole. Past-decade advances in high-throughput (next-generation) sequencing technology have dramatically improved the ease and cost of sequencing, contributing to a rapid acceleration of research in the area[35, 36]. Culture-independent DNA-based approaches have revealed a vast and more diverse community than previously appreciated based on culture-dependent approaches, since many gut microbiota identified through culture-independent methods had not previously been cultivated.
Gut microbiota composition: Challenges for studies of diet and health
To date, microbiome research has largely focused on measures of microbial composition based on sequencing one or more regions of the 16S rRNA marker gene[37–40]. 16S rRNA is an evolutionarily well-conserved prokaryotic gene found in all bacteria and archaea, with sufficient variability in gene regions to allow taxonomic assignment of microbiota to the genus level[41]. Characterization of the gut microbiota based on 16S rRNA include the relative abundance of specific taxonomic groups, as well as ecologic measures of the microbiome as a whole, such as diversity and richness[38].
16S rRNA remains easiest, fastest, and most affordable approach for characterizing the microbial community with respect to taxonomic composition, but it is limited in its ability to provide detailed understanding of gut microbiota functioning. High-throughput sequencing technology has made it economically feasible for researchers to further examine the role of the gut microbiota in health by obtaining a comprehensive survey of genes in the microbiota, using whole genome sequencing (WGS), termed metagenomics[40]. WGS data allows taxonomic assignment at a finer level than 16S rRNA and profiles the genes present in the microbiota, which can be mapped to metabolic pathways, providing a measure of functional potential[42].
The NIH-funded Human Microbiome Project (HMP) generated 16S rRNA and WGS data for microbial communities from multiple body locations, including gut, in roughly 300 U.S. adults[43]. A primary goal of the HMP was to establish the taxonomic entities and composition that constitute a healthy gut microbiota in humans[44]. However, a clearly-defined core microbiota has defied easy identification; rather, the HMP and other studies have found large variation in the taxonomic composition of the gut microbiota among healthy individuals[43]. For example, initial animal and human studies supported associations between obesity and the ratio of Firmicutes to Bacteroidetes, the two most abundant phyla in the human gut microbiota. In one study, ob/ob mice had a greater abundance of Firmicutes relative to Bacteroidetes (F/B ratio) than lean mice[45]. A study of 12 human subjects also found that obese individuals had a higher F/B ratio[20]. In an intervention of calorie-restricted diets, weight loss was associated with decreases in F/B, irrespective of macronutrient composition (low-fat or low-carbohydrate)[20]. Although several subsequent studies reported similar findings, however, other studies showed no F/B difference between obese and lean individuals, the reverse association (lower F/B in obese vs. lean), or no change in F/B with weight loss[46].
Other research has revealed large within-phyla variability that may help explain inconsistent findings. In a study of obese mice fed a Western-style diet, weight gain was accompanied by increases in specific members of Firmicutes from the Mollicutes class[47]. In another animal study, a high-fat diet was associated with increases in Mollicutes, as well as Firmicutes classes Clostridiales and Delta-Proteobacteria[48]. In a dietary intervention among 14 overweight men, carbohydrate subtype did not reveal a change in F/B, but did show changes at lower taxonomic levels[49]. Additional evidence of the importance of more refined taxonomic analysis can be illustrated by findings related to Prevotella, one of the two primary genera in the phylum Bacteroidetes. Prevotella has been associated with plant-based diets[50, 51] and improved glucose metabolism[52], but also with chronic inflammation[53]. Such apparent discordance in health associations may reflect within-genus variability. Indeed, gene-level diversity has been observed among genomes of strains within a single Prevotella species[52]. These data support the need for finer compositional analysis than has been standard, as well as motivate increased focus on measures of microbial function.
In contrast to the large compositional variability, functional potential measured through WGS is relatively stable among healthy individuals[21, 43]. These findings support a model of redundancy in the presence of genes for core metabolic functions across a diverse gut microbial composition. Additional work is needed to improve our understanding of how gene sets align with specific microorganisms, particularly to inform our assessment of the functional impact of microorganism-based targeted interventions. Redundancy in gene presence across microbiota may underlie the finding that greater microbial diversity is generally positively associated with health status[21], with some exceptions, as a diverse microbiota may be more likely to capture the core set of necessary genes.
Metagenomics studies have revealed significant differences in bacterial genes and metabolic pathways in obese and lean twins[21] and among individuals with and without T2D[17, 18]. In a study of 345 Chinese adults with and without T2D, WGS was used to derive species-level compositional information as well as to functionally annotate the gut community[17]. Although specific microbiota were differentially enriched in the T2D and control samples, there were even greater differences in functional potential, most notably with respect to decreased butyrate-producing bacteria among individuals with T2D[17]. Other metagenomics studies of T2D also support a role for butyrate production in distinguishing T2D from controls[18, 54].
There is growing consensus that to truly delineate the role of the gut microbiota in human physiology, we must pursue measures of microbial functional activity. This shift is reflected in Phase 2 of the HMP, the integrated Human Microbiome Project (iHMP), which includes a collection of multiple functional measures, including transcriptomics (RNA-seq), metabolomics, and proteomics[55] The trend towards functional measures in studies of the gut microbiota mirrors the current emphasis on integrating host-level functional measures into animal models and human studies[56, 57]. Functional measures reflect the biologic activity of ultimate physiologic interest; with respect to the gut community, functional redundancy across taxonomic groups limit the ability to infer functional activity from community composition. Thus, functional analysis will be increasingly important for defining microbial pathways to metabolic disease, and for determining targets for intervention.
Production of dietary metabolites as a functional measure of gut microbiota
The gut microbiota has evolved complementary metabolic capacity to its human host, and a substantial proportion of circulating metabolites have microbial origin[9, 10]. A subset of microbiota-related metabolites derive from dietary components[58], several of which have been studied with respect to cardiometabolic outcomes. For example, short-chain fatty acids generated from microbial metabolism of non-digestible polysaccharides, and in particular butyrate, have been shown to have positive effects on glucose and lipid metabolism[59]. Phenolic compounds undergo extensive gut microbiota metabolism, and these metabolites may help explain the health benefits associated with a plant-based diet[60]. Microbiota-dependent metabolites may also have adverse effects on cardiometabolic health, such as trimethylamine N-oxide (discussed below)[12, 29, 30].
The extent to which metabolites from dietary components are dependent on the gut microbiota for production can be estimated in germ-free (gnotobiotic) animal models, which lack gut microbiota. Comparing germ-free and conventional mice, Wikoff and colleagues reported variable production of metabolites[10]. For example, hippuric acid, produced from gut microbial metabolism of phenolic compounds such as in tea, wine, and fruit juices, was 17.4 times higher in conventional as compared to germ-free mice. Equol sulfate and p-cresol sulfate, produced from isoflavones and amino acids, respectively, were only observed in conventional mice.
Based on this and related work, circulating concentrations of microbiota-dependent metabolites in host urine or blood have been proposed as relatively efficient measures of microbial function, particularly at this stage of the science where metabolic pathways through the gut microbiota remain to be defined. Personalized metabolite profiles have been identified under controlled dietary delivery of precursors, and this measure of microbial function is maintained upon transfer of host microbiota into germ-free mice (termed, humanized germ-free model)[61]. That is, the production of metabolites varied among humanized mice in a way that reflected the human host, even when mice were maintained on the same diet[61]. Humanized germ-free animals provide an experimental model for illustrating the role of human gut microbiota in metabolic of dietary components, and have been used to demonstrate rapid shifts in the expression of microbial genes in response to a dietary change[62].
Integrated approaches to functional understanding of the interplay of diet and gut microbiota in cardiometabolic health
We present two examples that we consider illustrative of integrative approaches to the study of diet, gut microbiota function, and cardiometabolic health. These examples highlight what will likely be the translational and transdisciplinary nature of future work to define targeted interventions related to the gut microbiota.
Example 1: Trimethylamine N-oxide (TMAO)
The potential for nutrient metabolite trimethylamine N-oxide (TMAO) to increase CVD risk was identified in a metabolomics screen and confirmed in an independent sample[30]. Subsequent work has supported associations between TMAO and cardiovascular disease[29], chronic kidney disease[63, 64], and type 2 diabetes[65–67], though significant findings have not been consistently observed[68, 69]. Proposed mechanisms for TMAO in cardiometabolic risk include platelet activation[70], cholesterol efflux[12], uremic toxicity[63], and glucose metabolism[67].
The production of TMA from nutrient precursors, choline[29] and L-carnitine[12], is dependent on gut microbiota, with conversion of TMA to TMAO in the liver through flavin monooxygenase 3 (FMO3)[71]. Variability in circulating TMAO will thus reflect: 1) consumption of dietary precursors, 2) gut microbiota metabolic capability for production of TMA, and 3) FMO3 genotype for conversion of TMA to TMAO. A GWAS of mouse and human studies revealed that variability in TMAO production is likely more reflective of differences in diet and gut microbiota, rather than genetic variation in FMO3[72]. Several distinct gut microbial pathways for TMAO production have been identified[73–75] or proposed[76].
The production of TMAO varies among individuals exposed to a dietary challenge of TMAO precursors[12, 29, 74, 77]. In a well-controlled egg challenge study, individual-level dynamics of plasma TMAO was highly variable over a 24-hour period across 6 study participants[77]. Larger choline doses yielded a greater spike in TMAO production over the 24-hour observation period, but considerable variation in TMAO production was observed even at high doses (4–6 eggs, or 476–714 mg choline). Participants were provided standardized meals throughout the testing periods to maintain equal precursor contribution through diet.
Many sources of choline and L-carnitine are animal products, including fish, eggs, and red meat, and it is not surprising that TMAO measured in fasting samples is significantly lower among vegans and vegetarians as compared to omnivores[12] and among individuals with high, as compared to low, adherence to the Mediterranean diet[51]. Interestingly, long-term diet may influence the capacity for TMAO to be produced, perhaps through selection of gut microbiota based on nutrient availability. In an L-carnitine challenge in 10 individuals, TMAO was produced among omnivores, but not among vegans and vegetarians[12]. In addition to microbiota pathways related specifically to TMAO precursors, it is intriguing to consider the possibility that consumption of dietary components that are not direct precursors of TMAO may impact metabolite production through effects on the gut microbiota. For example, recent data indicate that TMAO production may be decreased with increased resveratrol[78], a phenol found in red wine, or allicin[79], an organosulfur compound found in garlic. These data may be consistent with findings from another dietary challenge showing that TMAO production was lower in individuals with less gut microbiota richness[80]. Another possibility is that other nutritional components may serve to offset an adverse impact of TMAO. For example, fish oil appeared to lessen the adverse effect of TMAO on glucose tolerance in mice on a high-fat diet[67], providing a possible explanation for fish being both cardioprotective as well as a major contributor of TMA directly and of TMA precursors.
This example illustrates the combination of discovery and targeted metabolite analysis in observational cohorts, controlled mechanistic work in animal models, and nutrient challenge studies in small human samples to elucidate health effects of the gut microbiota-dependent metabolite TMAO and pathways to TMAO production from dietary precursors through the gut microbiota.
Example 2: Polyphenols: Isoflavones and lignans
Isoflavones and lignans are polyphenols, which are a large class of compounds from plant sources[60]. It has been estimated that 90% of polyphenols have incomplete absorption in the small intestine and undergo metabolism by microbiota in the lower intestine[81, 82]. There is large individual variability in the production of metabolites following consumption of dietary polyphenolic precursors, which may reflect differences in gut microbiota. Plant consumption and many polyphenols, including isoflavones and lignans, have been proposed to be related to a range of health outcomes, including cardiovascular disease and diabetes[83, 84]. It is hypothesized that polyphenol metabolites may explain, at least in part, the health benefits of plant-based diets. Soy is a rich source of the isoflavone daidzein, from which metabolites equol and O-desmethylangolensin are generated; lignans are found in a variety of foods, including rye, berries, and flaxseed, and yield metabolites enterodiol and enterolactone. In two cohorts of U.S. women, higher urinary levels of isoflavones and lignans metabolites have been inversely associated with T2D[85, 86].
The gut microbiota-dependence of isoflavone and lignan metabolite production was demonstrated in a humanized germ-free rat model. Human subjects were classified as high or low producers of equol based on their response following consumption of a soy meal[87]. Equol, O-desmethylangolensin, and enterolactone were not produced in germ-free rats, but were detectable upon introduction of human fecal microbiota. The equol production phenotype was transferrable to germ-free rats: rats produced equol when they received a fecal microbiota transplant from individuals with high equol production, but not when they received a transplant from individuals with low equol production. There is a lack of data on microbial pathways for isoflavone and lignan metabolite production, but one recent study reported a positive association between gut microbiota diversity, based on 16S rRNA analysis, and enterolignan production in 115 premenopausal U.S. women[11].
Equol production is higher in Asian populations and in vegetarians, potentially supporting a role for long-term diet in gut microbiota-dependent metabolite production. In one study, equol production was 51% in Korean-American women as compared to 36% in non-Korean-American women[88]. In a U.S. sample of 41 healthy individuals, equol production varied from 25% among non-vegetarians (n=12) to 59% among vegetarians (n=29)[89]. However, studies of long-term dietary intake and equol production have yielded inconsistent results[90, 91]. In addition, research is needed into the extent to which these phenotypes are inducible with dietary change. A 1-month soy intervention did not alter ability to produce isoflavone metabolites[92]. It is possible that diet is important, but that effects of diet on microbiota are not restricted to the direct metabolite precursor.
Metabolite production studies have generally been small-scale highly controlled challenge studies. Designs that would allow the measurement of metabolite-producer phenotypes in large-scale population-based studies would provide data on the distribution of metabolite production in the population, and allow improved study of covariates that influence metabolite production, including diet and other lifestyle factors. In addition, larger samples are needed to power discovery analysis of microbial components related to metabolite production. In a recent study, Melby and Watanabe addressed some of the necessary pharmacokinetic considerations for integrating a challenge component into an epidemiologic study[93], including the optimal time window for post-challenge sample collection and the dose required to distinguish producers from non-producers.
Challenges for delineating the interplay among diet, gut microbiota, and cardiometabolic health
The rapid growth of microbiome research has significantly advanced our appreciation of the potential for the gut microbiota to impact metabolic and cardiovascular disease risk, but there remain several key challenges for future work.
First, despite the enormous progress in microbiome research, there is a paucity of data that establish causality. Alongside broad enthusiasm for the untapped potential of gut microbiota for understanding human physiology, there are calls for cautious interpretation of findings until data are available that delineate mechanistic pathways through the gut microbiota to health[94]. Findings that microbiota functional measures change upon changes in diet are compelling, but we do not know whether gut microbiota function is altered through targeted dietary interventions or whether changes in metabolite production and gene expression reflect existing functional potential that is activated through the delivery of dietary precursors. TMAO and isoflavones/lignans case studies are useful illustrations of an integrated approach to understanding pathways through diet, but there are few such examples in the literature. With respect to health outcomes, data indicate the potential for complex inter-connections; for example, obesity may be both influenced by the gut microbiota[22] as well as influence the gut microbiota[45]. Most observational studies of the microbiome have included a limited set of covariates to control for potential confounders. A recent analysis of T2D demonstrates the value of adjustment for medication use to clarifying discordant findings in observational studies[54].
Second, there is need for richer microbiota data from well-designed human studies. Model systems are exceptional experimental resources, but they may not reflect human biology. Germ-free animal models have illustrated the relevance of gut microbiota for health, including through the production of certain dietary metabolites[10]. Humanized microbiota-associated mice have been proposed as an efficient method to determine causality by incorporating human-derived gut microbiota in a controlled model organism, but there are many questions about the extent to which these models will reflect the active microbiota of the source human hosts[95]. Human studies are needed to address the potential lack of representation in animal models, as well as to capture the breadth of microbiota existing in human populations. For example, the HMP covered a relatively small sample (n~300) with little racial/ethnic diversity. To date, most observational studies have been cross-sectional comparisons of individuals with and without a health condition, and there is need to incorporate microbiota sample collection in ongoing cohort studies. Human experiments have been informative, but may not always reflect typical behavior, such as a study comparing plant- and animal-based diets[96], which are rare extremes in most societies.
Third, there is increasing recognition of the need for standards in the collection and processing of samples, and the analysis of microbiome data. Using a set of generated communities with known composition, the extent of possible bias was demonstrated in a recent study of vaginal microbiome. This work revealed the potential for large and differential-by-organism errors, largely attributable to DNA extraction and PCR amplification steps of sample processing[97]. The newly-established Microbiome Quality Control project will contribute to elucidating differences stemming from variability in methods in the field, the laboratory, and bioinformatics; and providing guidance in the future for microbiome researchers[98]. In the interim, protocols from the HMP have been published and their use allows comparison with HMP data and other data from researchers using the HMP protocols[39].
Summary and Conclusions
A growing body of literature supports a role of gut microbiota in the development and progression of metabolic and cardiovascular disease risk, but precise causal mechanisms remain to be elucidated. The microbiota-dependent production of health-related nutrient metabolites may be an important pathway through which the microbiota mediates the impact of diet on disease risk; however, the complex interplay between diet and the gut microbiota is only beginning to be understood. Diet appears to be important to shaping the gut microbiota, though the extent to which gut microbiota can be changed for improved health through targeted dietary interventions is not known. Multiple dietary components rely on gut microbiota for metabolism, and there is large individual variability in the gut microbiota-dependent production of health-related metabolites. A direction for future research is integrating gut microbiota measures into approaches for personalized nutrition[99]. Future advances in the field will be aided by shifting to a focus on microbiota functional activity, as well as deeper compositional measures to capture important within-species variability, along with increased attention to study designs for causal inference, a greater number of well-designed human studies, and standards for the conduct of microbiome studies.
Footnotes
Compliance with Ethics Guidelines
Conflicts of Interest
Katie A. Meyer has received research grants from the Egg Nutrition Center and from the National Institute of Health (K01-HL127159).
Brian J. Bennett has received research grants from the National Institutes of Health.
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.Ley RE, Hamady M, Lozupone C, et al. Evolution of mammals and their gut microbes. Science. 2008;320:1647–51. doi: 10.1126/science.1155725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Muegge BD, Kuczynski J, Knights D, et al. Diet drives convergence in gut microbiome functions across mammalian phylogeny and within humans. Science. 2011;332:970–4. doi: 10.1126/science.1198719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.De Filippo C, Cavalieri D, Di Paola M, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc Natl Acad Sci U S A. 2010;107:14691–6. doi: 10.1073/pnas.1005963107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–7. doi: 10.1038/nature11053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Schnorr SL, Candela M, Rampelli S, et al. Gut microbiome of the Hadza hunter-gatherers. Nat Commun. 2014;5:3654. doi: 10.1038/ncomms4654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Xu Z, Knight R. Dietary effects on human gut microbiome diversity. Br J Nutr. 2015;113:S1–5. doi: 10.1017/S0007114514004127. •• This is a review of how diet may influence the composition of the gut microbiota. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wong JM. Gut microbiota and cardiometabolic outcomes: influence of dietary patterns and their associated components. Am J Clin Nutr. 2014;100:369S–77S. doi: 10.3945/ajcn.113.071639. ••A review of how diet may influence the composition of the gut microbiota. [DOI] [PubMed] [Google Scholar]
- 8.Graf D, Di Cagno R, Fak F, et al. Contribution of diet to the composition of the human gut microbiota. Microb Ecol Health Dis. 2015;26:26164. doi: 10.3402/mehd.v26.26164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7. doi: 10.1126/science.1223813. [DOI] [PubMed] [Google Scholar]
- 10.Wikoff WR, Anfora AT, Liu J, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106:3698–703. doi: 10.1073/pnas.0812874106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Hullar MA, Lancaster SM, Li F, et al. Enterolignan-producing phenotypes are associated with increased gut microbial diversity and altered composition in premenopausal women in the United States. Cancer Epidemiol Biomarkers Prev. 2015;24:546–54. doi: 10.1158/1055-9965.EPI-14-0262. • A population-based study of the association between metabolite production and gut microbiota composition. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–85. doi: 10.1038/nm.3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Delzenne NM, Cani PD. Gut microbiota and the pathogenesis of insulin resistance. Curr Diab Rep. 2011;11:154–9. doi: 10.1007/s11892-011-0191-1. [DOI] [PubMed] [Google Scholar]
- 14.Li D, Kirsop J, Tang WH. Listening to our Gut: Contribution of gut microbiota and cardiovascular risk in diabetes pathogenesis. Curr Diab Rep. 2015;15:63. doi: 10.1007/s11892-015-0634-1. •• An excellent review of possible pathways from gut microbiota to cardiovascular disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Cox AJ, West NP, Cripps AW. Obesity, inflammation, and the gut microbiota. Lancet Diabetes Endocrinol. 2015;3:207–15. doi: 10.1016/S2213-8587(14)70134-2. [DOI] [PubMed] [Google Scholar]
- 16.Hartstra AV, Bouter KE, Backhed F, et al. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care. 2015;38:159–65. doi: 10.2337/dc14-0769. •• An excellent review of possible pathways from gut microbiota to obesity and type 2 diabetes. [DOI] [PubMed] [Google Scholar]
- 17.Qin J, Li Y, Cai Z, 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]
- 18.Karlsson FH, Tremaroli V, Nookaew I, et al. 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]
- 19.Larsen N, Vogensen FK, van den Berg FW, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5:e9085. doi: 10.1371/journal.pone.0009085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ley RE, Turnbaugh PJ, Klein S, et al. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444:1022–3. doi: 10.1038/4441022a. [DOI] [PubMed] [Google Scholar]
- 21.Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480–4. doi: 10.1038/nature07540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Turnbaugh PJ, Ley RE, Mahowald MA, et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027–31. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]
- 23.Dumas ME, Barton RH, Toye A, et al. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proc Natl Acad Sci U S A. 2006;103:12511–6. doi: 10.1073/pnas.0601056103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cani PD, Bibiloni R, Knauf C, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470–81. doi: 10.2337/db07-1403. [DOI] [PubMed] [Google Scholar]
- 25.Karlsson FH, Fak F, Nookaew I, et al. 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]
- 26.Yang T, Santisteban MM, Rodriguez V, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015;65:1331–40. doi: 10.1161/HYPERTENSIONAHA.115.05315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Holmes E, Loo RL, Stamler J, et al. Human metabolic phenotype diversity and its association with diet and blood pressure. Nature. 2008;453:396–400. doi: 10.1038/nature06882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Fu J, Bonder MJ, Cenit MC, et al. The gut microbiome contributes to a substantial proportion of the variation in blood lipids. Circ Res. 2015;117:817–24. doi: 10.1161/CIRCRESAHA.115.306807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–84. doi: 10.1056/NEJMoa1109400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zoetendal EG, Collier CT, Koike S, et al. Molecular ecological analysis of the gastrointestinal microbiota: a review. J Nutr. 2004;134:465–72. doi: 10.1093/jn/134.2.465. [DOI] [PubMed] [Google Scholar]
- 32.Savage DC. Microbial ecology of the gastrointestinal tract. Annu Rev Microbiol. 1977;31:107–33. doi: 10.1146/annurev.mi.31.100177.000543. [DOI] [PubMed] [Google Scholar]
- 33.Finegold SM, Attebery HR, Sutter VL. Effect of diet on human fecal flora: comparison of Japanese and American diets. Am J Clin Nutr. 1974;27:1456–69. doi: 10.1093/ajcn/27.12.1456. [DOI] [PubMed] [Google Scholar]
- 34.Suau A, Bonnet R, Sutren M, et al. Direct analysis of genes encoding 16S rRNA from complex communities reveals many novel molecular species within the human gut. Appl Environ Microbiol. 1999;65:4799–807. doi: 10.1128/aem.65.11.4799-4807.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Eckburg PB, Bik EM, Bernstein CN, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–8. doi: 10.1126/science.1110591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Tringe SG, von Mering C, Kobayashi A, et al. Comparative metagenomics of microbial communities. Science. 2005;308:554–7. doi: 10.1126/science.1107851. [DOI] [PubMed] [Google Scholar]
- 37.Olsen GJ, Lane DJ, Giovannoni SJ, et al. Microbial ecology and evolution: a ribosomal RNA approach. Annu Rev Microbiol. 1986;40:337–65. doi: 10.1146/annurev.mi.40.100186.002005. [DOI] [PubMed] [Google Scholar]
- 38.Caporaso JG, Kuczynski J, Stombaugh J, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7:335–6. doi: 10.1038/nmeth.f.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Caporaso JG, Lauber CL, Walters WA, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 2012;6:1621–4. doi: 10.1038/ismej.2012.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Morgan XC, Huttenhower C. Chapter 12: Human microbiome analysis. PLoS Comput Biol. 2012;8:e1002808. doi: 10.1371/journal.pcbi.1002808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Weisburg WG, Barns SM, Pelletier DA, et al. 16S ribosomal DNA amplification for phylogenetic study. J Bacteriol. 1991;173:697–703. doi: 10.1128/jb.173.2.697-703.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gill SR, Pop M, Deboy RT, et al. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312:1355–9. doi: 10.1126/science.1124234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–14. doi: 10.1038/nature11234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Turnbaugh PJ, Ley RE, Hamady M, et al. The human microbiome project. Nature. 2007;449:804–10. doi: 10.1038/nature06244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Ley RE, Backhed F, Turnbaugh P, et al. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–5. doi: 10.1073/pnas.0504978102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Walters WA, Xu Z, Knight R. Meta-analyses of human gut microbes associated with obesity and IBD. FEBS Lett. 2014;588:4223–33. doi: 10.1016/j.febslet.2014.09.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Turnbaugh PJ, Backhed F, Fulton L, et al. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3:213–23. doi: 10.1016/j.chom.2008.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, et al. High-fat diet determines the composition of the murine gut microbiome independently of obesity. Gastroenterology. 2009;137:1716–24. doi: 10.1053/j.gastro.2009.08.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Walker AW, Ince J, Duncan SH, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011;5:220–30. doi: 10.1038/ismej.2010.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Wu GD, Chen J, Hoffmann C, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105–8. doi: 10.1126/science.1208344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.De Filippis F, Pellegrini N, Vannini L, et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2015 doi: 10.1136/gutjnl-2015-309957. [DOI] [PubMed] [Google Scholar]
- 52.Kovatcheva-Datchary P, Nilsson A, Akrami R, et al. Dietary fiber-induced improvement in glucose metabolism is associated with increased abundance of prevotella. Cell Metab. 2015;22:971–82. doi: 10.1016/j.cmet.2015.10.001. [DOI] [PubMed] [Google Scholar]
- 53.Dillon SM, Lee EJ, Kotter CV, et al. Gut dendritic cell activation links an altered colonic microbiome to mucosal and systemic T-cell activation in untreated HIV-1 infection. Mucosal Immunol. 2016;9:24–37. doi: 10.1038/mi.2015.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Forslund K, Hildebrand F, Nielsen T, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528:262–6. doi: 10.1038/nature15766. • A study illustrating the potential for medication to confound associations between the gut microbiota and health outcomes. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Integrative HMP Research Network Consortium. The integrative human microbiome project: Dynamic analysis of microbime-host omics profiles during periods of human health and disease. Cell Host & Microbe. 2014;16:276–89. doi: 10.1016/j.chom.2014.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chen R, Mias GI, Li-Pook-Than J, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148:1293–307. doi: 10.1016/j.cell.2012.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kussmann M, Raymond F, Affolter M. OMICS-driven biomarker discovery in nutrition and health. J Biotechnol. 2006;124:758–87. doi: 10.1016/j.jbiotec.2006.02.014. [DOI] [PubMed] [Google Scholar]
- 58.Duffy LC, Raiten DJ, Hubbard VS, et al. Progress and challenges in developing metabolic footprints from diet in human gut microbial cometabolism. J Nutr. 2015;145:1123S–30S. doi: 10.3945/jn.114.194936. • A review of pathways to dietary metabolites through gut microbiota metabolism. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.den Besten G, van Eunen K, Groen AK, et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J Lipid Res. 2013;54:2325–40. doi: 10.1194/jlr.R036012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Manach C, Scalbert A, Morand C, et al. Polyphenols: food sources and bioavailability. Am J Clin Nutr. 2004;79:727–47. doi: 10.1093/ajcn/79.5.727. [DOI] [PubMed] [Google Scholar]
- 61.Marcobal A, Kashyap PC, Nelson TA, et al. A metabolomic view of how the human gut microbiota impacts the host metabolome using humanized and gnotobiotic mice. ISME J. 2013;7:1933–43. doi: 10.1038/ismej.2013.89. • An example of the integration of human samples and animal models for mechanistic understanding of gut microbiota-metabolome pathways. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Turnbaugh PJ, Ridaura VK, Faith JJ, et al. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1:6ra14. doi: 10.1126/scitranslmed.3000322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tang WH, Wang Z, Kennedy DJ, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res. 2015;116:448–55. doi: 10.1161/CIRCRESAHA.116.305360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Rhee EP, Ho JE, Chen MH, et al. A genome-wide association study of the human metabolome in a community-based cohort. Cell Metab. 2013;18:130–43. doi: 10.1016/j.cmet.2013.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lever M, George PM, Slow S, et al. Betaine and trimethylamine-N-oxide as predictors of cardiovascular outcomes show different patterns in diabetes mellitus: An observational study. PLoS One. 2014;9:e114969. doi: 10.1371/journal.pone.0114969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Miao J, Ling AV, Manthena PV, et al. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nat Commun. 2015;6:6498. doi: 10.1038/ncomms7498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gao X, Xu J, Jiang C, et al. Fish oil ameliorates trimethylamine N-oxide-exacerbated glucose intolerance in high-fat diet-fed mice. Food Funct. 2015;6:1117–25. doi: 10.1039/c5fo00007f. [DOI] [PubMed] [Google Scholar]
- 68.Mueller DM, Allenspach M, Othman A, et al. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis. 2015;243:638–44. doi: 10.1016/j.atherosclerosis.2015.10.091. [DOI] [PubMed] [Google Scholar]
- 69.Wang TJ, Larson MG, Vasan RS, et al. Metabolite profiles and the risk of developing diabetes. Nat Med. 2011;17:448–53. doi: 10.1038/nm.2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165:111–24. doi: 10.1016/j.cell.2016.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.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 Metab. 2013;17:49–60. doi: 10.1016/j.cmet.2012.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Hartiala J, Bennett BJ, Tang WH, et al. Comparative genome-wide association studies in mice and humans for trimethylamine N-oxide, a proatherogenic metabolite of choline and L-carnitine. Arterioscler Thromb Vasc Biol. 2014;34:1307–13. doi: 10.1161/ATVBAHA.114.303252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A. 2012;109:21307–12. doi: 10.1073/pnas.1215689109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Koeth RA, Levison BS, Culley MK, et al. gamma-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab. 2014;20:799–812. doi: 10.1016/j.cmet.2014.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zhu Y, Jameson E, Crosatti M, et al. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc Natl Acad Sci U S A. 2014;111:4268–73. doi: 10.1073/pnas.1316569111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Falony G, Vieira-Silva S, Raes J. Microbiology meets big data: The case of gut microbiota-derived trimethylamine. Annu Rev Microbiol. 2015;69:305–21. doi: 10.1146/annurev-micro-091014-104422. [DOI] [PubMed] [Google Scholar]
- 77.Miller CA, Corbin KD, da Costa KA, et al. Effect of egg ingestion on trimethylamine-N-oxide production in humans: a randomized, controlled, dose-response study. Am J Clin Nutr. 2014;100:778–86. doi: 10.3945/ajcn.114.087692. • A controlled feeding study demonstrating variable production of trimethylamine N-oxide, a gut microbiota-dependent nutrient metabolite. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Chen ML, Yi L, Zhang Y, et al. Resveratrol attenuates trimethylamine-N-oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. MBio. 2016;7:e02210–15. doi: 10.1128/mBio.02210-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wu WK, Panyod S, Ho CT, et al. Dietary allicin reduces transformation of L-carnitine to TMAO through impact on gut microbiota. Journal of Functional Foods. 2015;15:408–17. [Google Scholar]
- 80.Cho CE, Taesuwan S, Malysheva OV, et al. Trimethylamine-N-oxide biomarker response is a function of dietary precursor intake and gut microbiota composition in healthy young men. FASEB J. 2016;30(1) Supplement 406.6. [Google Scholar]
- 81.Manach C, Williamson G, Morand C, et al. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am J Clin Nutr. 2005;81:230S–42S. doi: 10.1093/ajcn/81.1.230S. [DOI] [PubMed] [Google Scholar]
- 82.Williamson G, Manach C. Bioavailability and bioefficacy of polyphenols in humans. II. Review of 93 intervention studies. Am J Clin Nutr. 2005;81:243S–55S. doi: 10.1093/ajcn/81.1.243S. [DOI] [PubMed] [Google Scholar]
- 83.Scalbert A, Manach C, Morand C, et al. Dietary polyphenols and the prevention of diseases. Crit Rev Food Sci Nutr. 2005;45:287–306. doi: 10.1080/1040869059096. [DOI] [PubMed] [Google Scholar]
- 84.Manach C, Mazur A, Scalbert A. Polyphenols and prevention of cardiovascular diseases. Curr Opin Lipidol. 2005;16:77–84. doi: 10.1097/00041433-200502000-00013. [DOI] [PubMed] [Google Scholar]
- 85.Ding M, Franke AA, Rosner BA, et al. Urinary isoflavonoids and risk of type 2 diabetes: a prospective investigation in US women. Br J Nutr. 2015;114:1694–701. doi: 10.1017/S0007114515003359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Sun Q, Wedick NM, Pan A, et al. Gut microbiota metabolites of dietary lignans and risk of type 2 diabetes: a prospective investigation in two cohorts of U.S. women. Diabetes Care. 2014;37:1287–95. doi: 10.2337/dc13-2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bowey E, Adlercreutz H, Rowland I. Metabolism of isoflavones and lignans by the gut microflora: a study in germ-free and human flora associated rats. Food Chem Toxicol. 2003;41:631–6. doi: 10.1016/s0278-6915(02)00324-1. [DOI] [PubMed] [Google Scholar]
- 88.Song KB, Atkinson C, Frankenfeld CL, et al. Prevalence of daidzein-metabolizing phenotypes differs between Caucasian and Korean American women and girls. J Nutr. 2006;136:1347–51. doi: 10.1093/jn/136.5.1347. [DOI] [PubMed] [Google Scholar]
- 89.Setchell KD, Cole SJ. Method of defining equol-producer status and its frequency among vegetarians. J Nutr. 2006;136:2188–93. doi: 10.1093/jn/136.8.2188. [DOI] [PubMed] [Google Scholar]
- 90.Rowland IR, Wiseman H, Sanders TA, et al. Interindividual variation in metabolism of soy isoflavones and lignans: influence of habitual diet on equol production by the gut microflora. Nutr Cancer. 2000;36:27–32. doi: 10.1207/S15327914NC3601_5. [DOI] [PubMed] [Google Scholar]
- 91.Atkinson C, Newton KM, Bowles EJ, et al. Demographic, anthropometric, and lifestyle factors and dietary intakes in relation to daidzein-metabolizing phenotypes among premenopausal women in the United States. Am J Clin Nutr. 2008;87:679–87. doi: 10.1093/ajcn/87.3.679. [DOI] [PubMed] [Google Scholar]
- 92.Lampe JW, Skor HE, Li S, et al. Wheat bran and soy protein feeding do not alter urinary excretion of the isoflavan equol in premenopausal women. J Nutr. 2001;131:740–4. doi: 10.1093/jn/131.3.740. [DOI] [PubMed] [Google Scholar]
- 93.Melby MK, Watanabe S. Soy isoflavones in epidemiologic serum samples: What are the optimal time window and concentration cutoffs for assignment of equol producer status? Austin Journal of Nutrition and Food Sciences. 2014;2:id1034. [Google Scholar]
- 94.Hanage WP. Microbiology: Microbiome science needs a healthy dose of scepticism. Nature. 2014;512:247–8. doi: 10.1038/512247a. • An excellent summary of current challenges in microbiome research. [DOI] [PubMed] [Google Scholar]
- 95.Arrieta MC, Walter J, Finlay BB. Human microbiota-associated mice: A model with challenges. Cell Host Microbe. 2016;19:575–8. doi: 10.1016/j.chom.2016.04.014. [DOI] [PubMed] [Google Scholar]
- 96.David LA, Maurice CF, Carmody RN, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. doi: 10.1038/nature12820. • A controlled feeding study demonstrating changes in microbial metabolite production and gene expression within 24-hours of shifting between plant- and animal-based diets. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Brooks JP, Edwards DJ, Harwich MD, Jr, et al. The truth about metagenomics: quantifying and counteracting bias in 16S rRNA studies. BMC Microbiol. 2015;15:66. doi: 10.1186/s12866-015-0351-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Sinha R, Abnet CC, White O, et al. The microbiome quality control project: baseline study design and future directions. Genome Biol. 2015;16:276. doi: 10.1186/s13059-015-0841-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zeevi D, Korem T, Zmora N, et al. Personalized nutrition by prediction of glycemic responeses. Cell. 2015;163:1079–94. doi: 10.1016/j.cell.2015.11.001. [DOI] [PubMed] [Google Scholar]