Fecal Microbiome Composition Does Not Predict Diet-Induced TMAO Production in Healthy Adults

Marc Ferrell, Peter Bazeley, Zeneng Wang, Bruce S Levison, Xinmin S Li, Xun Jia, Ronald M Krauss, Rob Knight, Aldons J Lusis, J C Garcia-Garcia, Stanley L Hazen, W H Wilson Tang, Marc Ferrell, Peter Bazeley, Zeneng Wang, Bruce S Levison, Xinmin S Li, Xun Jia, Ronald M Krauss, Rob Knight, Aldons J Lusis, J C Garcia-Garcia, Stanley L Hazen, W H Wilson Tang

Abstract

Background Trimethylamine-N-oxide (TMAO) is a small molecule derived from the metabolism of dietary nutrients by gut microbes and contributes to cardiovascular disease. Plasma TMAO increases following consumption of red meat. This metabolic change is thought to be partly because of the expansion of gut microbes able to use nutrients abundant in red meat. Methods and Results We used data from a randomized crossover study to estimate the degree to which TMAO can be estimated from fecal microbial composition. Healthy participants received a series of 3 diets that differed in protein source (red meat, white meat, and non-meat), and fecal, plasma, and urine samples were collected following 4 weeks of exposure to each diet. TMAO was quantitated in plasma and urine, while shotgun metagenomic sequencing was performed on fecal DNA. While the cai gene cluster was weakly correlated with plasma TMAO (rho=0.17, P=0.0007), elastic net models of TMAO were not improved by abundances of bacterial genes known to contribute to TMAO synthesis. A global analysis of all taxonomic groups, genes, and gene families found no meaningful predictors of TMAO. We postulated that abundances of known genes related to TMAO production do not predict bacterial metabolism, and we measured choline- and carnitine-trimethylamine lyase activity during fecal culture. Trimethylamine lyase genes were only weakly correlated with the activity of the enzymes they encode. Conclusions Fecal microbiome composition does not predict systemic TMAO because, in this case, gene copy number does not predict bacterial metabolic activity. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT01427855.

Keywords: fecal microbiome; metagenomics; trimethylamine N‐oxide; trimethylamine lyase.

Figures

Figure 1. Overview of analyses.
Figure 1. Overview of analyses.
A, Healthy adults were provided non‐meat, white meat, or red meat diets for 4‐week periods and donated plasma, urine, and stool samples during clinic visits. Trimethylamine N‐oxide (TMAO) and related metabolites were measured in plasma and urine samples. A subset of urine samples (n=38) was taken after oral challenge with isotope labeled choline and carnitine. Isotope‐labeled TMAO was measured in these urine samples. Stool was used for both anoxic cultures to determine the activities of enzymes related to trimethylamine production and metagenomics sequencing. Metagenomic sequence data were used to compute metagenomic operational taxonomic units, as well as global gene and gene family abundances. Low abundance trimethylamine‐related genes were quantified using a gene cluster‐base d approach. B, Plasma and urine TMAO were not correlated with any stool data set, and stool trimethylamine ‐related enzyme activities were not correlated with related genes or with TMAO production during the oral challenge. C, Elastic net models trained with trimethylamine‐related gene abundances did not predict TMAO or stool enzyme activities. CV indicates cardiovascular; FMO, flavin mono‐oxygenases; GBB, gamma‐butyrobetaine; HPLC/MS/MS, high performance liquid chromatography with tandem mass spectrometry; MI, myocardial infarction; mOTUs, metagenomic operational taxonomic units; and TMAO, trimethylamine‐N‐oxide.
Figure 2. Synthesis of trimethylamine N ‐oxide…
Figure 2. Synthesis of trimethylamine N‐oxide requires dietary substrate as well as host and microbial enzymes.
Nutrients including choline and carnitine, which are abundant in red meat, are converted to trimethylamine via multiple synthetic pathways. The copy number of related genes is altered by diet and may contribute to trimethylamine N‐oxide production by host enzymes. caiA/B/C, crotonobetainyl‐CoA dehydrogenase; cntA/B, carnitine oxygenase/reductase; cutC/D, choline utilization gene cluster; FMOs, flavin monooxygenases; GBB, gamma‐butyrobetaine; grdH, betaine reductase complex component B subunit beta; MI, myocardial infarction; TMA, trimethylamine; TMAO, trimethylamine‐N‐oxide; torA, trimethylamine‐N‐oxide reductase (cytochrome c); and yeaX/W, carnitine monooxygenase subunit YeaX.
Figure 3. cutC predicts trimethylamine N ‐oxide…
Figure 3. cutC predicts trimethylamine N‐oxide in those assigned to non‐meat before red meat.
Plasma trimethylamine N‐oxide is higher during red meat consumption vs a meat‐free diet regardless of diet order. Although cutC is not differentially abundant in red meat vs meat‐free, cutC abundance is correlated with plasma trimethylamine N‐oxide among those assigned to non‐meat before red meat. P values represent Wilcox tests. RPKM indicates reads per kilobase per million reads; TMAO, trimethylamine‐N‐oxide.
Figure 4. Abundance of genes related to…
Figure 4. Abundance of genes related to trimethylamine synthesis do not improve models of trimethylamine N‐oxide (TMAO).
Elastic net was used to train and test linear models of plasma TMAO, as well as percent changes in TMAO from non‐meat to red meat diets. Models were tested with 100 randomly selected training/testing groups, using adjusted R2 and root‐mean‐squared error as performance metrics. The base model, using data summarized in Table 1, predicted 41% of the variance in TMAO, and the addition of trimethylamine‐related gene abundances did not improve model performance. TMAO fractional excretion rate outperformed the base model in predicting changes in plasma TMAO, in terms of root‐mean‐squared error (P<0.001, Wilcox test). FER, fractional excretion rate; RMSE indicates root‐mean‐squared error; and TMAO, trimethylamine‐N‐oxide.
Figure 5. Fecal trimethylamine‐lyase activities are not…
Figure 5. Fecal trimethylamine‐lyase activities are not correlated with trimethylamine N‐oxide (TMAO) production in host.
In a single visit, 13 participants randomly selected from the APPROACH cohort donated a fecal sample and participated in a heavy‐isotope‐labeled choline and carnitine challenge (38 visits total). A, Participants were dosed orally with 250mg d6‐choline and d3‐carnitine. Labeled TMAO and synthetic intermediates were quantitated in 24‐hour urine collection. B, Paired fecal samples were cultured with 200 µmol/L d6‐choline and d9‐carnitine and labeled TMAO and synthetic intermediates were quantitated after 18‐ and 36‐hour incubations. Analytes not detected in >30% of samples were excluded. C, The heatmap shows Spearman correlations between fecal enzyme activities and the indicated isotopologue in 24‐hour urine collection. TMAO produced from carnitine in the host was not highly correlated with carnitine‐derived trimethylamine in fecal cultures. Urine choline‐derived TMAO was negatively correlated with choline‐trimethylamine‐lyase activity in feces (ρ=−0.46, P=0.003). Cr, creatinine; RMSE indicates root‐mean‐squared error; and TMAO, trimethylamine‐N‐oxide. * Adjusted P<0.05. P values adjusted with the method of Benjamani and Hochberg.
Figure 6. cutC abundance marginally outperforms clinical…
Figure 6. cutC abundance marginally outperforms clinical data in predicting fecal choline‐trimethylamine lyase activity.
Fecal samples were cultured with 200 µmol/L d6‐choline and labeled trimethylamine and synthetic intermediates were quantitated after 18‐hour incubation. Elastic net models were trained to predict 18‐hour d6‐choline with 70% of samples and tested on the remaining 30%, and testing was iterated over 100 different training/testing sets. Although the cutC models had less error than models trained with clinical data (Table 1), cutC models did not result in higher correlations between predicted and actual d6‐choline concentration. RMSE indicates root‐mean‐squared error.

References

    1. Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S, Chiuve SE, Cushman M, Delling FN, Deo R, et al. Heart disease and stroke statistics—2018 update: a report from the American Heart Association. Circulation. 2018;137:67–492. doi: 10.1161/CIR.0000000000000558
    1. 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;472:57–65. doi: 10.1038/nature09922
    1. Shan Z, Clish CB, Hua S, Scott JM, Hanna DB, Burk RD, Haberlen SA, Shah SJ, Margolick JB, Sears CL, et al. Gut microbial‐related choline metabolite trimethylamine‐N‐oxide is associated with progression of carotid artery atherosclerosis in HIV infection. Top Antivir Med. 2018;218:1474–1479. doi: 10.1093/infdis/jiy356
    1. Seldin MM, Meng Y, Qi H, Zhu WF, Wang Z, Hazen SL, Lusis AJ, Shih DM. Trimethylamine N‐oxide promotes vascular inflammation through signaling of mitogen‐activated protein kinase and nuclear factor‐κb. J Am Heart Assoc. 2016;5:e002767. doi: 10.1161/JAHA.115.002767
    1. Zhu W, Zeneng W, Wilson Tang WH, Hazen SL. Gut microbe‐generated TMAO from dietary choline is prothrombotic in subjects. Circulation. 2017;135:1671–1673.
    1. Zhu W, Buffa JA, Wang Z, Warrier M, Schugar R, Shih DM, Gupta N, Gregory JC, Org E, Fu X, et al. Flavin monooxygenase 3, the host hepatic enzyme in the metaorganismal trimethylamine N‐oxide‐generating pathway, modulates platelet responsiveness and thrombosis risk. J Thromb Haemost. 2018;16:1857–1872. doi: 10.1111/jth.14234
    1. Stubbs JR, House JA, Ocque AJ, Zhang S, Johnson C, Kimber C, Schmidt K, Gupta A, Wetmore JB, Nolin TD, et al. Serum trimethylamine‐N‐oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J Am Soc Nephrol. 2016;27:305–313.
    1. Cho CE, Taesuwan S, Malysheva OV, Bender E, Tulchinsky NF, Yan J, Sutter JL, Caudill MA. Trimethylamine‐N‐oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: a randomized controlled trial. Mol Nutr Food Res. 2017;61:1600324.
    1. Koeth RA, Wang Z, Levia R, Garcia‐Garcia JC, Tang WHW, Hazen SL. Gut microbiota metabolism of l‐carnitine, a nutrient in red meat, and the atherogenic metabolites y‐gammabutyrobetaine and TMAO in humans. Circulation. 2017;136:A16758.
    1. Koeth RA, Lam‐Galvez BR, Kirsop J, Wang Z, Levison BS, Gu X, Copeland MF, Bartlett D, Cody DB, Dai HJ, et al. L‐Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J Clin Invest. 2019;129:373–387. doi: 10.1172/JCI94601
    1. 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
    1. Wang Z, Roberts A, Buffa J, Levison B, Zhu W, Org E, Gu X, Huang Y, Zamanian‐Daryoush M, Culley M, et al. Non‐lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163:1585–1595. doi: 10.1016/j.cell.2015.11.055
    1. Gupta N, Buffa JA, Roberts AB, Sangwan N, Skye SM, Li L, Ho KJ, Varga J, DiDonato JA, Tang WHW, et al. Targeted inhibition of gut microbial TMAO production reduces renal tubulointerstitial fibrosis and functional impairment in a murine model of chronic kidney disease. Arterioscler Thromb Vasc Biol. 2020;40:1239–1255.
    1. De Filippis F, Pellegrini N, Vannini L, Jeffery IB, La Storia A, Laghi L, Serrazanetti DI, Di Cagno R, Ferrocino I, Lazzi C, et al. High‐level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 2016;65:1812–1821. doi: 10.1136/gutjnl-2015-309957
    1. Crimarco A, Springfield S, Petlura C, Streaty T, Cunanan K, Lee J, Fielding‐Singh P, Carter MM, Topf MA, Wastyk HC, et al. A randomized crossover trial on the effect of plant‐based compared with animal‐based meat on trimethylamine‐N‐oxide and cardiovascular disease risk factors in generally healthy adults: Study With Appetizing Plantfood—Meat Eating Alternative Trial (SWAP‐MEAT). Am J Clin Nutr. 2020;112:1188–1199. doi: 10.1093/ajcn/nqaa203
    1. Wu WK, Chen CC, Liu PY, Panyod S, Liao BY, Chen PC, Kao HL, Kuo HC, Kuo CH, Chiu THT, et al. Identification of TMAO‐producer phenotype and host‐diet‐gut dysbiosis by carnitine challenge test in human and germ‐free mice. Gut. 2019;68:1439–1449. doi: 10.1136/gutjnl-2018-317155
    1. Smits LP, Kootte RS, Levin E, Prodan A, Fuentes S, Zoetendal EG, Wang Z, Levison BS, Cleophas MCP, Kemper EM, et al. Effect of vegan fecal microbiota transplantation on carnitine‐ and choline‐derived trimethylamine‐N‐oxide production and vascular inflammation in patients with metabolic syndrome. J Am Heart Assoc. 2018;7:e008342. doi: 10.1161/JAHA.117.008342
    1. Lang JM, Pan C, Cantor RM, Tang WHW, Garcia‐Garcia JC, Kurtz I, Hazen SL, Bergeron N, Krauss RM, Lusis AJ. Impact of individual traits, saturated fat, and protein source on the gut microbiome. MBio. 2018;9. doi: 10.1128/mBio.01604-18
    1. Manor O, Zubair N, Conomos MP, Xu X, Rohwer JE, Krafft CE, Lovejoy JC, Magis AT. A multi‐omic association study of trimethylamine N‐oxide. Cell Rep. 2018;24:935–946. doi: 10.1016/j.celrep.2018.06.096
    1. Jie Z, Xia H, Zhong SL, Feng Q, Li S, Liang S, Zhong H, Liu Z, Gao Y, Zhao H, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun. 2017;8:845. doi: 10.1038/s41467-017-00900-1
    1. Bergeron N, Chiu S, Williams PT, King MS, Krauss RM. Effects of red meat, white meat, and nonmeat protein sources on atherogenic lipoprotein measures in the context of low compared with high saturated fat intake: a randomized controlled trial. Am J Clin Nutr. 2019;110:24–33. doi: 10.1093/ajcn/nqz035
    1. Wang Z, Bergeron N, Levison BS, Li XS, Chiu S, Jia X, Koeth RA, Li L, Wu Y, Tang WHW, et al. Impact of chronic dietary red meat, white meat, or non‐meat protein on trimethylamine N‐oxide metabolism and renal excretion in healthy men and women. Eur Heart J. 2019;40:583–594. doi: 10.1093/eurheartj/ehy799
    1. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva‐Datchary P, Li Y, Xia Y, Xie H, Zhong H, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe. 2015;17:690–703. doi: 10.1016/j.chom.2015.04.004
    1. Cox MP, Peterson DA, Biggs PJ. SolexaQA: at‐a‐glance quality assessment of Illumina second‐generation sequencing data. BMC Bioinformatics. 2010;11:485. doi: 10.1186/1471-2105-11-485
    1. Li R, Li Y, Kristiansen K, Wang J. SOAP: short oligonucleotide alignment program. Bioinformatics. 2008;24:713–714. doi: 10.1093/bioinformatics/btn025
    1. Kultima JR, Coelho LP, Forslund K, Huerta‐Cepas J, Li SS, Driessen M, Voigt AY, Zeller G, Sunagawa S, Bork P. MOCAT2: a metagenomic assembly, annotation and profiling framework. Bioinformatics. 2016;32:2520–2523. doi: 10.1093/bioinformatics/btw183
    1. Li J, Jia H, Cai X, Zhong H, Feng Q, Sunagawa S, Arumugam M, Kultima JR, Prifti E, Nielsen T, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol. 2014;32:834–841. doi: 10.1038/nbt.2942
    1. Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M, Weingart G, Lipson KS, Knight R, Caporaso JG, Segata N, et al. Species‐level functional profiling of metagenomes and metatranscriptomes. Nat Methods. 2018;15:962–968. doi: 10.1038/s41592-018-0176-y
    1. Kanehisa M, Furumichi M, Tanabe M, Sato Y, Morishima K. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45:D353–D361. doi: 10.1093/nar/gkw1092
    1. Manor O, Borenstein E. MUSiCC: a marker genes based framework for metagenomic normalization and accurate profiling of gene abundances in the microbiome. Genome Biol. 2015;16:15. doi: 10.1186/s13059-015-0610-8
    1. Zou H, Hastie T. Regularization and variable selection via the elastic net. J R Stat Soc Ser B. 2005;67:301–320. doi: 10.1111/j.1467-9868.2005.00503.x
    1. Friedman J, Hastie T, Tibshirani R. Regularization paths for generalized linear models via coordinate descent. J Statistical Softw. 2010;33. doi: 10.18637/jss.v033.i01
    1. Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci USA. 2012;109:21307–21312. doi: 10.1073/pnas.1215689109
    1. Skye SM, Zhu W, Romano KA, Guo CJ, Wang Z, Jia X, Kirsop J, Haag B, Lang JM, DiDonato JA, et al. Microbial transplantation with human gut commensals containing CutC is sufficient to transmit enhanced platelet reactivity and thrombosis potential. Circ Res. 2018;123:1164–1176. doi: 10.1161/CIRCRESAHA.118.313142
    1. Koeth R, Levison B, Culley M, Buffa J, Wang Z, Gregory J, Org E, Wu Y, Li L, Smith J, 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. Falony G, Vieira‐Silva S, Raes J. Microbiology meets big data: the case of gut microbiota–derived trimethylamine. Annu Rev Microbiol. 2015;69:305–321. doi: 10.1146/annurev-micro-091014-104422
    1. Zhu Y, Jameson E, Crosatti M, Schäfer H, Rajakumar K, Bugg TDH, 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
    1. Wagner M, Sonntag D, Grimm R, Pich A, Eckerskorn C, Söhling B, Andreesen JR. Substrate‐specific selenoprotein B of glycine reductase from Eubacterium acidaminophilum. Biochemical and molecular analysis. Eur J Biochem. 1999;260:38–49. doi: 10.1046/j.1432-1327.1999.00107.x
    1. Meyer M, Granderath K, Andreesen JR. Purification and characterization of protein PB of Betaine Reductase and its relationship to the corresponding proteins Glycine Reductase and Sarcosine Reductase from Eubacterium acidaminophilum . Eur J Biochem. 1995;234:184–191. doi: 10.1111/j.1432-1033.1995.184_c.x
    1. Borrel G, McCann A, Deane J, Neto MC, Lynch DB, Brugère JF, O’Toole PW. Genomics and metagenomics of trimethylamine‐utilizing Archaea in the human gut microbiome. ISME J. 2017;11:2059–2074. doi: 10.1038/ismej.2017.72
    1. Méjean V, Lobbi‐Nivol C, Lepelletier M, Giordano G, Chippaux M, Pascal M‐C. TMAO anaerobic respiration in Escherichia coli: involvement of the tor operon. Mol Microbiol. 1994;11:1169–1179. doi: 10.1111/j.1365-2958.1994.tb00393.x
    1. Eichler K, Bourgis F, Buchet A, Kleber H‐P, Mandrand‐Berthelot M‐A. Molecular characterization of the cai operon necessary for carnitine metabolism in Escherichia coli . Mol Microbiol. 1994;13:775–786.
    1. Engemann C, Elssner T, Pfeifer S, Krumbholz C, Maier T, Kleber HP. Identification and functional characterisation of genes and corresponding enzymes involved in carnitine metabolism of Proteus sp. Arch Microbiol. 2005;183:176–189. doi: 10.1007/s00203-005-0760-2
    1. Arense P, Bernal V, Charlier D, Iborra JL, Foulquié‐Moreno MR, Cánovas M. Metabolic engineering for high yielding L(‐)‐carnitine production in Escherichia coli . Microb Cell Fact. 2013;12:56. doi: 10.1186/1475-2859-12-56
    1. Rath S, Rud T, Pieper DH, Vital M. Potential TMA‐producing bacteria are ubiquitously found in mammalia. Front Microbiol. 2020;10. doi: 10.3389/fmicb.2019.02966
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B. 1995;57:289–300. doi: 10.1111/j.2517-6161.1995.tb02031.x
    1. Jo J, Oh J, Park C. Microbial community analysis using high‐throughput sequencing technology: a beginner’s guide for microbiologists. J Microbiol. 2020;58:176–192. doi: 10.1007/s12275-020-9525-5
    1. Cani PD. Human gut microbiome: hopes, threats and promises. Gut. 2018;67:1716–1725. doi: 10.1136/gutjnl-2018-316723
    1. Lloyd‐Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila‐Pacheco J, Poon TW, Andrews E, Ajami NJ, Bonham KS, Brislawn CJ, et al. Multi‐omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569:655–662. doi: 10.1038/s41586-019-1237-9
    1. Mallick H, Franzosa EA, Mclver LJ, Banerjee S, Sirota‐Madi A, Kostic AD, Clish CB, Vlamakis H, Xavier RJ, Huttenhower C. Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences. Nat Commun. 2019;10:3136. doi: 10.1038/s41467-019-10927-1
    1. Sczyrba A, Hofmann P, Belmann P, Koslicki D, Janssen S, Dröge J, Gregor I, Majda S, Fiedler J, Dahms E, et al. Critical assessment of metagenome interpretation—a benchmark of metagenomics software. Nat Methods. 2017;14:1063–1071. doi: 10.1038/nmeth.4458
    1. Bouchemal N, Ouss L, Brassier A, Barbier V, Gobin S, Hubert L, De Lonlay P, Le Moyec L. Diagnosis and phenotypic assessment of trimethylaminuria, and its treatment with riboflavin: 1H NMR spectroscopy and genetic testing. Orphanet J Rare Dis. 2019;14:222. doi: 10.1186/s13023-019-1174-6
    1. Schirmer M, Franzosa EA, Lloyd‐Price J, McIver LJ, Schwager R, Poon TW, Ananthakrishnan AN, Andrews E, Barron G, Lake K, et al. Dynamics of metatranscription in the inflammatory bowel disease gut microbiome. Nat Microbiol. 2018;3:337–346. doi: 10.1038/s41564-017-0089-z
    1. Qi J, You T, Li J, Pan T, Xiang L, Han Y, Zhu L. Circulating trimethylamine N‐oxide and the risk of cardiovascular diseases: a systematic review and meta‐analysis of 11 prospective cohort studies. J Cell Mol Med. 2018;22:185–194. doi: 10.1111/jcmm.13307
    1. Yao ME, Da LP, Zhao XJ, Wang L. Trimethylamine‐N‐oxide has prognostic value in coronary heart disease: a meta‐analysis and dose‐response analysis. BMC Cardiovasc Disord. 2020;20:7. doi: 10.1186/s12872-019-01310-5
    1. Garza DR, van Verk MC, Huynen MA, Dutilh BE. Towards predicting the environmental metabolome from metagenomics with a mechanistic model. Nat Microbiol. 2018;3:456–460. doi: 10.1038/s41564-018-0124-8

Source: PubMed

Подписаться