Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort

Elin Org, Yuna Blum, Silva Kasela, Margarete Mehrabian, Johanna Kuusisto, Antti J Kangas, Pasi Soininen, Zeneng Wang, Mika Ala-Korpela, Stanley L Hazen, Markku Laakso, Aldons J Lusis, Elin Org, Yuna Blum, Silva Kasela, Margarete Mehrabian, Johanna Kuusisto, Antti J Kangas, Pasi Soininen, Zeneng Wang, Mika Ala-Korpela, Stanley L Hazen, Markku Laakso, Aldons J Lusis

Abstract

Background: The gut microbiome is a complex and metabolically active community that directly influences host phenotypes. In this study, we profile gut microbiota using 16S rRNA gene sequencing in 531 well-phenotyped Finnish men from the Metabolic Syndrome In Men (METSIM) study.

Results: We investigate gut microbiota relationships with a variety of factors that have an impact on the development of metabolic and cardiovascular traits. We identify novel associations between gut microbiota and fasting serum levels of a number of metabolites, including fatty acids, amino acids, lipids, and glucose. In particular, we detect associations with fasting plasma trimethylamine N-oxide (TMAO) levels, a gut microbiota-dependent metabolite associated with coronary artery disease and stroke. We further investigate the gut microbiota composition and microbiota-metabolite relationships in subjects with different body mass index and individuals with normal or altered oral glucose tolerance. Finally, we perform microbiota co-occurrence network analysis, which shows that certain metabolites strongly correlate with microbial community structure and that some of these correlations are specific for the pre-diabetic state.

Conclusions: Our study identifies novel relationships between the composition of the gut microbiota and circulating metabolites and provides a resource for future studies to understand host-gut microbiota relationships.

Keywords: Host-microbiota interactions; Metabolic traits; Serum metabolites; TMAO; Type 2 diabetes.

Figures

Fig. 1
Fig. 1
Microbial community variation in the METSIM cohort. a Top contributors to community variation as determined by canonical correspondence analysis on unscaled genera abundances, plotted on the first principal component (PC) dimensions (arrows scaled to contribution). b The top seven metabolite contributors to microbiome community variation. c A total of 32 out of 60 factors (60%) explained a total of 13% of variance of the gut microbiome according to Bray–Curtis distance. TG triglycerides, FA fatty acids, DHA docosahexaenoic acid, MUFA monounsaturated fatty acid, PUFA polyunsaturated fatty acid
Fig. 2
Fig. 2
Associations between microbiota taxa and circulating serum metabolites. Serum metabolite concentrations were measured from fasting samples using NMR or LC-MS/MS. They were then associated with microbial taxa following adjustment for age and treatment of the subjects. *Ruminococcus from the family Lachnospiraceae; **Ruminococcus from the family Ruminococcaceae. CI confidence interval, DHA docosahexaenoic acid, MUFA monounsaturated fatty acid, PUFA polyunsaturated fatty acid
Fig. 3
Fig. 3
OTU co-occurrence network and module-trait associations. a OTU co-occurrence network where OTU (nodes) are colored according to the phyla to which they belong. Blue edges correspond to positive correlations and red edges to negative correlations. Any resulting correlations with p value ≥0.01 and abs(r) <0.3 were removed. b OTU co-occurrence network were OTU (nodes) are colored according to WGCNA module colors. c Module–trait associations are shown. Each cell of the matrix contains the correlation between one OTU module and a metabolic trait, and the corresponding p value. The table is color-coded by correlation according to the color legend (red for positive correlations and green for negative correlations). FA fatty acids, DHA docosahexaenoic acid, MUFA monounsaturated fatty acid, PUFA polyunsaturated fatty acid, GlycA glycoprotein acetyls, TMAO trimethyl N-oxide
Fig. 4
Fig. 4
The gut microbiota differences in metabolic phenotypes. Mean proportions of significantly different taxa between individuals with different BMI values (a) and individuals with different 2-h glucose tolerance test (NGT and pre-T2D) (b). Examples of BMI/OGGT metabolite–microbiota interaction, where the abundances of Bacteroidales, Collinsella (c) and Coprobacillus (d) exhibit opposite associations in individuals with low versus high BMI levels or individuals with impaired glucose tolerance test (c). *P ≤ 0.01, **P ≤ 0.001, ***P ≤ 0.0001. BMI body mass index, GT glucose tolerance, NGT normal glucose tolerance, T2D type 2 diabetes

References

    1. Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ, Fagerberg B, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498:99–103. doi: 10.1038/nature12198.
    1. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60. doi: 10.1038/nature11450.
    1. Parks BW, Nam E, Org E, Kostem E, Norheim F, Hui ST, et al. Genetic control of obesity and gut microbiota composition in response to high-fat, high-sucrose diet in mice. Cell Metab. 2013;17:141–52. doi: 10.1016/j.cmet.2012.12.007.
    1. Dumas M-E, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell 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.
    1. Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BAH, et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535:376–81. doi: 10.1038/nature18646.
    1. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500:541–6. doi: 10.1038/nature12506.
    1. Ussar S, Griffin NW, Bezy O, Fujisaka S, Vienberg S, Softic S, et al. Interactions between gut microbiota, host genetics and diet modulate the predisposition to obesity and metabolic syndrome. Cell Metab. 2015;22:516–30. doi: 10.1016/j.cmet.2015.07.007.
    1. Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, 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.
    1. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922.
    1. Tang WHW, Wang Z, Shrestha K, Borowski AG, Wu Y, Troughton RW, et al. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J Card Fail. 2015;21:91–6. doi: 10.1016/j.cardfail.2014.11.006.
    1. Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los Reyes-Gavilán CG, Salazar N. Intestinal short chain fatty acids and their link with diet and human health. Front Microbiol. 2016;7:185. doi: 10.3389/fmicb.2016.00185.
    1. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, 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.
    1. Tang WHW, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, 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.
    1. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen Y-Y, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science. 2011;334:105–8. doi: 10.1126/science.1208344.
    1. Org E, Parks BW, Joo JWJ, Emert B, Schwartzman W, Kang EY, et al. Genetic and environmental control of host-gut microbiota interactions. Genome Res. 2015;25:1558–69. doi: 10.1101/gr.194118.115.
    1. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, et al. Human genetics shape the gut microbiome. Cell. 2014;159:789–99. doi: 10.1016/j.cell.2014.09.053.
    1. Tremaroli V, Bäckhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489:242–9. doi: 10.1038/nature11552.
    1. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, et al. Population-level analysis of gut microbiome variation. Science. 2016;352:560–4. doi: 10.1126/science.aad3503.
    1. Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352:565–9. doi: 10.1126/science.aad3369.
    1. Fu J, Bonder MJ, Cenit MC, Tigchelaar EF, Maatman A, Dekens JAM, 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.
    1. Stancáková A, Javorský M, Kuulasmaa T, Haffner SM, Kuusisto J, Laakso M. Changes in insulin sensitivity and insulin release in relation to glycemia and glucose tolerance in 6,414 Finnish men. Diabetes. 2009;58:1212–21. doi: 10.2337/db08-1607.
    1. Zoetendal EG, Rajilic-Stojanovic M, de Vos WM. High-throughput diversity and functionality analysis of the gastrointestinal tract microbiota. Gut. 2008;57:1605–15. doi: 10.1136/gut.2007.133603.
    1. Arumugam M, Raes J, Pelletier E, Le Paslier D, Yamada T, Mende DR, et al. Enterotypes of the human gut microbiome. Nature. 2011;473:174–80. doi: 10.1038/nature09944.
    1. Knights D, Ward TL, McKinlay CE, Miller H, Gonzalez A, McDonald D, et al. Rethinking “enterotypes”. Cell Host Microbe. 2014;16:433–7. doi: 10.1016/j.chom.2014.09.013.
    1. Soininen P, Kangas AJ, Würtz P, Suna T, Ala-Korpela M. Quantitative serum nuclear magnetic resonance metabolomics in cardiovascular epidemiology and genetics. Circ Cardiovasc Genet. 2015;8:192–206. doi: 10.1161/CIRCGENETICS.114.000216.
    1. Risérus U, Willett WC, Hu FB. Dietary fats and prevention of type 2 diabetes. Prog Lipid Res. 2009;48:44–51. doi: 10.1016/j.plipres.2008.10.002.
    1. Cani PD, Delzenne NM. Interplay between obesity and associated metabolic disorders: new insights into the gut microbiota. Curr Opin Pharmacol. 2009;9:737–43. doi: 10.1016/j.coph.2009.06.016.
    1. Karlsson F, Tremaroli V, Nielsen J, Bäckhed F. Assessing the human gut microbiota in metabolic diseases. Diabetes. 2013;62:3341–9. doi: 10.2337/db13-0844.
    1. de Souza AZZ, Zambom AZ, Abboud KY, Reis SK, Tannihão F, Guadagnini D, et al. Oral supplementation with L-glutamine alters gut microbiota of obese and overweight adults: A pilot study. Nutrition. 2015;31:884–9. doi: 10.1016/j.nut.2015.01.004.
    1. Cheng S, Rhee EP, Larson MG, Lewis GD, McCabe EL, Shen D, et al. Metabolite profiling identifies pathways associated with metabolic risk in humans. Circulation. 2012;125:2222–31. doi: 10.1161/CIRCULATIONAHA.111.067827.
    1. Koeth RA, Levison BS, Culley MK, Buffa JA, Wang Z, Gregory JC, et al. γ-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.
    1. Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem. 2015;290:5647–60. doi: 10.1074/jbc.M114.618249.
    1. Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, et al. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J Am Heart Assoc. 2016;5. .
    1. Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–84. doi: 10.1056/NEJMoa1109400.
    1. Chen M, Yi L, Zhang Y, Zhou X, Ran L, Yang J, 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:e02210–5.
    1. Graessler J, Qin Y, Zhong H, Zhang J, Licinio J, Wong M-L, et al. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: correlation with inflammatory and metabolic parameters. Pharmacogenomics J. 2013;13:514–22. doi: 10.1038/tpj.2012.43.
    1. Rajilić–Stojanović M, Biagi E, Heilig HGHJ, Kajander K, Kekkonen RA, Tims S, et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011;141:1792–801. doi: 10.1053/j.gastro.2011.07.043.
    1. Genuth S, Alberti KGMM, Bennett P, Buse J, Defronzo R, Kahn R, et al. Follow-up report on the diagnosis of diabetes mellitus. Diabetes Care. 2003;26:3160–7. doi: 10.2337/diacare.26.12.3331.
    1. Million M, Angelakis E, Maraninchi M, Henry M, Giorgi R, Valero R, et al. Correlation between body mass index and gut concentrations of Lactobacillus reuteri, Bifidobacterium animalis, Methanobrevibacter smithii and Escherichia coli. Int J Obes (Lond) 2013;37:1460–6. doi: 10.1038/ijo.2013.20.
    1. Louis P, Scott KP, Duncan SH, Flint HJ. Understanding the effects of diet on bacterial metabolism in the large intestine. J Appl Microbiol. 2007;102:1197–208. doi: 10.1111/j.1365-2672.2007.03322.x.
    1. 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;444:1027–31. doi: 10.1038/nature05414.
    1. Schwiertz A, Taras D, Schäfer K, Beijer S, Bos NA, Donus C, et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring) 2010;18:190–5. doi: 10.1038/oby.2009.167.
    1. Everard A, Lazarevic V, Gaïa N, Johansson M, Ståhlman M, Backhed F, et al. Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J. 2014;8:2116–30. doi: 10.1038/ismej.2014.45.
    1. Frost G, Sleeth ML, Sahuri-Arisoylu M, Lizarbe B, Cerdan S, Brody L, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun. 2014;5:3611. doi: 10.1038/ncomms4611.
    1. Perry RJ, Peng L, Barry NA, Cline GW, Zhang D, Cardone RL, et al. Acetate mediates a microbiome-brain-β-cell axis to promote metabolic syndrome. Nature. 2016;534:213–7. doi: 10.1038/nature18309.
    1. Hui ST, Parks BW, Org E, Norheim F, Che N, Pan C, et al. The genetic architecture of NAFLD among inbred strains of mice. Elife. 2015;4. .
    1. Baylin A, Campos H. The use of fatty acid biomarkers to reflect dietary intake. Curr Opin Lipidol. 2006;17:22–7. doi: 10.1097/01.mol.0000199814.46720.83.
    1. Daniel H, Gholami AM, Berry D, Desmarchelier C, Hahne H, Loh G, et al. High-fat diet alters gut microbiota physiology in mice. ISME J. 2014;8:295–308. doi: 10.1038/ismej.2013.155.
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63. doi: 10.1038/nature12820.
    1. Alcock J, Lin HC. Fatty acids from diet and microbiota regulate energy metabolism. F1000Research. 2015;4:738.
    1. Mani V, Hollis JH, Gabler NK. 2013. Nutr Metab (Lond) 2013;10:6. doi: 10.1186/1743-7075-10-6.
    1. Summers LKM, Fielding BA, Bradshaw HA, Ilic V, Beysen C, Clark ML, et al. Substituting dietary saturated fat with polyunsaturated fat changes abdominal fat distribution and improves insulin sensitivity. Diabetologia. 2002;45:369–77. doi: 10.1007/s00125-001-0768-3.
    1. Kasai C, Sugimoto K, Moritani I, Tanaka J, Oya Y, Inoue H, et al. Comparison of the gut microbiota composition between obese and non-obese individuals in a Japanese population, as analyzed by terminal restriction fragment length polymorphism and next-generation sequencing. BMC Gastroenterol. 2015;15:100. doi: 10.1186/s12876-015-0330-2.
    1. Juste C, Kreil DP, Beauvallet C, Guillot A, Vaca S, Carapito C, et al. Bacterial protein signals are associated with Crohn’s disease. Gut. 2014;63:1566–77. doi: 10.1136/gutjnl-2012-303786.
    1. Bonder MJ, Kurilshikov A, Tigchelaar EF, Mujagic Z, Imhann F, Vila AV, et al. The effect of host genetics on the gut microbiome. Nat Genet. 2016.
    1. Eren AM, Sogin ML, Morrison HG, Vineis JH, Fisher JC, Newton RJ, et al. A single genus in the gut microbiome reflects host preference and specificity. ISME J. 2015;9:90–100. doi: 10.1038/ismej.2014.97.
    1. McHardy IH, Goudarzi M, Tong M, Ruegger PM, Schwager E, Weger JR, et al. Integrative analysis of the microbiome and metabolome of the human intestinal mucosal surface reveals exquisite inter-relationships. Microbiome. 2013;1:17. doi: 10.1186/2049-2618-1-17.
    1. Poretsky R, Rodriguez-R LM, Luo C, Tsementzi D, Konstantinidis KT, Costello E, et al. Strengths and limitations of 16S rRNA gene amplicon sequencing in revealing temporal microbial community dynamics. PLoS One. 2014;9 doi: 10.1371/journal.pone.0093827.
    1. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, et al. Human genetics shape the gut microbiome. Cell. 2014;159:789–99. doi: 10.1016/j.cell.2014.09.053.
    1. Zhou X, Meile L, Kreuzer M, Zeitz JO. The effect of saturated fatty acids on methanogenesis and cell viability of Methanobrevibacter ruminantium. Archaea. 2013;2013:106916. doi: 10.1155/2013/106916.
    1. Bolnick DI, Snowberg LK, Hirsch PE, Lauber CL, Org E, Parks B, et al. Individual diet has sex-dependent effects on vertebrate gut microbiota. Nat Commun. 2014;5:4500. doi: 10.1038/ncomms5500.
    1. Org E, Mehrabian M, Parks BW, Shipkova P, Liu X, Drake TA, et al. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes. 2016;7:313–22. doi: 10.1080/19490976.2016.1203502.
    1. Haro C, Rangel-Zúñiga OA, Alcalá-Díaz JF, Gómez-Delgado F, Pérez-Martínez P, Delgado-Lista J, et al. Intestinal microbiota is influenced by gender and body mass index. PLoS One. 2016;11 doi: 10.1371/journal.pone.0154090.
    1. Caporaso JG, Kuczynski J, Stombaugh J, Bittinger K, Bushman FD, Costello EK, et al. QIIME allows analysis of high-throughput community sequencing data. Nat Methods. 2010;7:335–6. doi: 10.1038/nmeth.f.303.
    1. Bokulich NA, Subramanian S, Faith JJ, Gevers D, Gordon JI, Knight R, et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat Methods. 2013;10:57–9. doi: 10.1038/nmeth.2276.
    1. Edgar RC. Quality measures for protein alignment benchmarks. Nucleic Acids Res. 2010;38:2145–53. doi: 10.1093/nar/gkp1196.
    1. McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A, et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J. 2012;6:610–8. doi: 10.1038/ismej.2011.139.
    1. Würtz P, Havulinna AS, Soininen P, Tynkkynen T, Prieto-Merino D, Tillin T, et al. Metabolite profiling and cardiovascular event risk: a prospective study of 3 population-based cohorts. Circulation. 2015;131:774–85. doi: 10.1161/CIRCULATIONAHA.114.013116.
    1. Ritchie SC, Würtz P, Nath AP, Abraham G, Havulinna AS, Fearnley LG, et al. The biomarker GlycA is associated with chronic inflammation and predicts long-term risk of severe infection. Cell Syst. 2015;1:293–301. doi: 10.1016/j.cels.2015.09.007.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559. doi: 10.1186/1471-2105-9-559.
    1. Langfelder P, Zhang B, Horvath S. Defining clusters from a hierarchical cluster tree: the Dynamic Tree Cut package for R. Bioinformatics. 2008;24:719–20. doi: 10.1093/bioinformatics/btm563.
    1. Langfelder P, Luo R, Oldham MC, Horvath S. Is my network module preserved and reproducible? PLoS Comput Biol. 2011;7 doi: 10.1371/journal.pcbi.1001057.

Source: PubMed

3
Tilaa