The use of an in-vitro batch fermentation (human colon) model for investigating mechanisms of TMA production from choline, L-carnitine and related precursors by the human gut microbiota

Priscilla Day-Walsh, Emad Shehata, Shikha Saha, George M Savva, Barbora Nemeckova, Jasmine Speranza, Lee Kellingray, Arjan Narbad, Paul A Kroon, Priscilla Day-Walsh, Emad Shehata, Shikha Saha, George M Savva, Barbora Nemeckova, Jasmine Speranza, Lee Kellingray, Arjan Narbad, Paul A Kroon

Abstract

Purpose: Plasma trimethylamine-N-oxide (TMAO) levels have been shown to correlate with increased risk of metabolic diseases including cardiovascular diseases. TMAO exposure predominantly occurs as a consequence of gut microbiota-dependent trimethylamine (TMA) production from dietary substrates including choline, carnitine and betaine, which is then converted to TMAO in the liver. Reducing microbial TMA production is likely to be the most effective and sustainable approach to overcoming TMAO burden in humans. Current models for studying microbial TMA production have numerous weaknesses including the cost and length of human studies, differences in TMA(O) metabolism in animal models and the risk of failing to replicate multi-enzyme/multi-strain pathways when using isolated bacterial strains. The purpose of this research was to investigate TMA production from dietary precursors in an in-vitro model of the human colon.

Methods: TMA production from choline, L-carnitine, betaine and γ-butyrobetaine was studied over 24-48 h using an in-vitro human colon model with metabolite quantification performed using LC-MS.

Results: Choline was metabolised via the direct choline TMA-lyase route but not the indirect choline-betaine-TMA route, conversion of L-carnitine to TMA was slower than that of choline and involves the formation of the intermediate γ-BB, whereas the Rieske-type monooxygenase/reductase pathway for L-carnitine metabolism to TMA was negligible. The rate of TMA production from precursors was choline > carnitine > betaine > γ-BB. 3,3-Dimethyl-1-butanol (DMB) had no effect on the conversion of choline to TMA.

Conclusion: The metabolic routes for microbial TMA production in the colon model are consistent with observations from human studies. Thus, this model is suitable for studying gut microbiota metabolism of TMA and for screening potential therapeutic targets that aim to attenuate TMA production by the gut microbiota.

Trial registration number: NCT02653001 ( http://www.clinicaltrials.gov ), registered 12 Jan 2016.

Keywords: Betaine; Cardiovascular disease; Carnitine; Fish odour syndrome; Human gut microbiota; Lecithin; Metabolic disease; Phosphatidylcholine; TMAO; γ-Butyrobetaine.

Conflict of interest statement

None of the authors declared a conflict of interest.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Change in TMA between 0 and 24 h in each batch fermentation seeded with faecal samples of five different donors. Fermentation stratified by substrate added. Colours correspond to donors and marker shapes to individual experiments. There is a high intra-class correlation between replicate fermentations within experiments, but little intra-class correlation within donors beyond this. Variation in the levels of TMA produced across experiments is high for each substrate, despite similar levels of substrate being utilised
Fig. 2
Fig. 2
Estimated mean TMA produced from each substrate over 24 h. Estimates were obtained by mixed effects regression models of the difference between TMA concentration at 0 and 24 h (as described in the statistical methods). The effect of each substrate is calculated with correction for the production of TMA observed in blank vessel without added substrates. Error bars represent 95% confidence intervals
Fig. 3
Fig. 3
The average trajectory of all metabolites. With no added substrate (a) and following supplementation with each substrate (be). Error bars represent standard errors
Fig. 4
Fig. 4
The effects of DMB on TMA production from choline in three independent experiments. The graph is stratified by experiment to enable a direct comparison between paired fermentations with and without DMB added. There was no effect of adding DMB on TMA production or choline concentration at any stage over the time period tested
Fig. 5
Fig. 5
The fermentation of TMA substrates under anaerobic conditions without pH control. There was no fermentation of any of the substrate and no TMA production in anaerobic conditions without pH control, although only a few experiments were carried out (n = 3 for choline and 2 for betaine, l-carnitine and γ-BB)
Fig. 6
Fig. 6
Pathways for the metabolism of choline, betaine, l-carnitine and γ-BB by human gut microbiota. This is based on data reported here and previously by others [, , , –22, 32]. In humans, choline is metabolised to TMA via the choline TMA-lyase pathway, betaine is not formed as an intermediate of choline to produce TMA via the choline dehydrogenase (CHDH)/betaine aldehyde dehydrogenase (BADH) > betaine reductase pathway, nor is it formed as an intermediate of l-carnitine via the l-carnitine dehydrogenase pathway. There is no direct conversion of l-carnitine to TMA via the Rieske-type C l-carnitine oxygenase/reductase pathway, instead l-carnitine is first converted to γ-BB by γ-butyrobetainyl-CoA:carnitine CoA transferases which is then converted to TMA by the l-carnitine TMA lyases. It is possible that betaine may be converted to dimethylglycine by glycine betaine transmethylase and then to TMA by decarboxylation although the evidence for this is weak. Dashed black lines are pathways shown not to be functional in this model; solid green lines indicate pathways we have demonstrated to be important for TMA production in the in-vitro human colonic fermentation model

References

    1. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Nat Acad Sci U S A. 2004;101:15718–15723. doi: 10.1073/pnas.0407076101.
    1. Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BAH, Forslund K, Hildebrand F, Prifti E, Falony G, Le Chatelier E, Levenez F, Doré J, Mattila I, Plichta DR, Pöhö P, Hellgren LI, Arumugam M, Sunagawa S, Vieira-Silva S, Jørgensen T, Holm JB, Trošt K, Kristiansen K, Brix S, Raes J, Wang J, Hansen T, Bork P, Brunak S, Oresic M, Ehrlich SD, Pedersen O, Consortium M Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 2016;535:376–381. doi: 10.1038/nature18646.
    1. Chang Y, Chen Y, Zhou Q, Wang C, Chen L, Di W, Zhang Y. Short-chain fatty acids accompanying changes in the gut microbiome contribute to the development of hypertension in patients with preeclampsia. Clin Sci (Lond) 2020;134:289–302. doi: 10.1042/cs20191253.
    1. Suzuki T, Yazaki Y, Voors AA, Jones DJL, Chan DCS, Anker SD, Cleland JG, Dickstein K, Filippatos G, Hillege HL, Lang CC, Ponikowski P, Samani NJ, van Veldhuisen DJ, Zannad F, Zwinderman AH, Metra M, Ng LL. Association with outcomes and response to treatment of trimethylamine N-oxide in heart failure: results from BIOSTAT-CHF. Eur J Heart Fail. 2019;21:877–886. doi: 10.1002/ejhf.1338.
    1. Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, Li XS, Levison BS, Hazen SL. 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;116:448–455. doi: 10.1161/circresaha.116.305360.
    1. Reiner MF, Muller D, Gobbato S, Stalder O, Limacher A, Bonetti NR, Pasterk L, Mean M, Rodondi N, Aujesky D, Angelillo-Scherrer A, Matter CM, Luscher TF, Camici GG, von Eckardstein A, Beer JH. Gut microbiota-dependent trimethylamine-N-oxide (TMAO) shows a U-shaped association with mortality but not with recurrent venous thromboembolism. Thromb Res. 2019;174:40–47. doi: 10.1016/j.thromres.2018.12.011.
    1. Collins HL, Drazul-Schrader D, Sulpizio AC, Koster PD, Williamson Y, Adelman SJ, Owen K, Sanli T, Bellamine A. L-Carnitine intake and high trimethylamine N-oxide plasma levels correlate with low aortic lesions in ApoE(-/-) transgenic mice expressing CETP. Atherosclerosis. 2016;244:29–37. doi: 10.1016/j.atherosclerosis.2015.10.108.
    1. Nowinski A, Ufnal M. Trimethylamine N-oxide: a harmful, protective or diagnostic marker in lifestyle diseases? Nutrition. 2018;46:7–12. doi: 10.1016/j.nut.2017.08.001.
    1. Narath SH, Mautner SI, Svehlikova E, Schultes B, Pieber TR, Sinner FM, Gander E, Libiseller G, Schimek MG, Sourij H, Magnes C. An untargeted metabolomics approach to characterize short-term and long-term metabolic changes after bariatric surgery. PLoS One. 2016;11:e0161425. doi: 10.1371/journal.pone.0161425.
    1. Tang WHW, Wang ZE, Levison BS, Koeth RA, Britt EB, Fu XM, Wu YP, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. New Engl J Med. 2013;368:1575–1584. doi: 10.1056/nejmoa1109400.
    1. Janeiro MH, Ramirez MJ, Milagro FI, Martinez JA, Solas M. Implication of Trimethylamine N-Oxide (TMAO) in disease: potential biomarker or new therapeutic target. Nutrients. 2018;10:1398. doi: 10.3390/nu10101398.
    1. Rath S, Heidrich B, Pieper DH, Vital M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome. 2017;5:54. doi: 10.1186/s40168-017-0271-9.
    1. Martinez-del Campo A, Bodea S, Hamer HA, Marks JA, Haiser HJ, Turnbaugh PJ, Balskus EP. Characterization and detection of a widely distributed gene cluster that predicts anaerobic choline utilization by human gut bacteria. mBio. 2015;6(2):e00042. doi: 10.1128/mbio.00042-15.
    1. Zhu Y, Jameson E, Crosatti M, Schafer H, Rajakumar K, Bugg TD, Chen Y. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc Natl Acad Sci U S A. 2014;111:4268–4273. doi: 10.1073/pnas.1316569111.
    1. Romano KA, Vivas EI, Amador-Noguez D, Rey FE. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. mBio. 2015;6:e02481. doi: 10.1128/mbio.02481-14.
    1. Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A. 2012;109:21307–21312. doi: 10.1073/pnas.1215689109.
    1. Kalnins G, Kuka J, Grinberga S, Makrecka-Kuka M, Liepinsh E, Dambrova M, Tars K. Structure and function of CutC choline lyase from human microbiota bacterium Klebsiella pneumoniae. J Biol Chem. 2015;290:21732–21740. doi: 10.1074/jbc.M115.670471.
    1. Jameson E, Fu T, Brown IR, Paszkiewicz K, Purdy KJ, Frank S, Chen Y. Anaerobic choline metabolism in microcompartments promotes growth and swarming of Proteus mirabilis. Environ Microbiol. 2016;18:2886–2898. doi: 10.1111/1462-2920.13059.
    1. Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922.
    1. Wang Z, Tang WH, Buffa JA, Fu X, Britt EB, Koeth RA, Levison BS, Fan Y, Wu Y, Hazen SL. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur Heart J. 2014;35:904–910. doi: 10.1093/eurheartj/ehu002.
    1. Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, Gu X, Huang Y, Zamanian-Daryoush M, Culley MK, DiDonato AJ, Fu X, Hazen JE, Krajcik D, DiDonato JA, Lusis AJ, Hazen SL. 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. Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, Smith JD, DiDonato JA, Chen J, Li H, Wu GD, Lewis JD, Warrier M, Brown JM, Krauss RM, Tang WH, Bushman FD, Lusis AJ, Hazen SL. 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. Rebouche CJ, Mack DL, Edmonson PF. l-Carnitine dissimilation in the gastrointestinal tract of the rat. Biochemistry. 1984;23:6422–6426. doi: 10.1021/bi00321a022.
    1. Parmanand BA, Kellingray L, Le Gall G, Basit AW, Fairweather-Tait S, Narbad A. A decrease in iron availability to human gut microbiome reduces the growth of potentially pathogenic gut bacteria; an in vitro colonic fermentation study. J Nutr Biochem. 2019;67:20–27. doi: 10.1016/j.jnutbio.2019.01.010.
    1. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14:e1002533–e1002533. doi: 10.1371/journal.pbio.1002533.
    1. Takagi R, Sasaki K, Sasaki D, Fukuda I, Tanaka K, Yoshida K, Kondo A, Osawa R. A Single-batch fermentation system to simulate human colonic microbiota for high-throughput evaluation of prebiotics. PLoS One. 2016;11:e0160533. doi: 10.1371/journal.pone.0160533.
    1. Lenth R, Singmann H, Love J, Buerkner P, Herve M (2019) Package ‘emmeans’(version 1.3. 4). stimated marginal means, aka least-squares means. . Accessed 2 Feb 2021
    1. Pinheiro J, Bates D, DebRoy S, Sarkar D, R Core Team (2019). nlme: Linear and nonlinear mixed effects models. R package version 3.1–139. . Accessed 2 Feb 2021
    1. Team RC (2018) R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing. . Accessed 2 Feb 2021
    1. Fennema D, Phillips IR, Shephard EA. Trimethylamine and Trimethylamine N-Oxide, a Flavin-containing Monooxygenase 3 (FMO3)-Mediated Host-microbiome metabolic axis implicated in health and disease. Drug Metab Dispos. 2016;44:1839–1850. doi: 10.1124/dmd.116.070615.
    1. Serra AL, Mariscotti JF, Barra JL, Lucchesi GI, Domenech CE, Lisa AT. Glycine betaine transmethylase mutant of Pseudomonas aeruginosa. J Bacteriol. 2002;184:4301–4303. doi: 10.1128/jb.184.15.4301-4303.2002.
    1. Jameson E, Doxey AC, Airs R, Purdy KJ, Murrell JC, Chen Y. Metagenomic data-mining reveals contrasting microbial populations responsible for trimethylamine formation in human gut and marine ecosystems. Microb Genom. 2016;2:e000080. doi: 10.1099/mgen.0.000080.
    1. Rath S, Rud T, Pieper DH, Vital M. Potential TMA-producing bacteria are ubiquitously found in mammalia. Front Microbiol. 2020;10:2966. doi: 10.3389/fmicb.2019.02966.
    1. Kühn T, Rohrmann S, Sookthai D, Johnson T, Katzke V, Kaaks R, von Eckardstein A, Müller D. Intra-individual variation of plasma trimethylamine-N-oxide (TMAO), betaine and choline over 1 year. Clin Chem Lab Med. 2017;55:261–268. doi: 10.1515/cclm-2016-0374.
    1. McEntyre CJ, Lever M, Chambers ST, George PM, Slow S, Elmslie JL, Florkowski CM, Lunt H, Krebs JD. Variation of betaine, N, N-dimethylglycine, choline, glycerophosphorylcholine, taurine and trimethylamine-N-oxide in the plasma and urine of overweight people with type 2 diabetes over a two-year period. Ann Clin Biochem. 2015;52:352–360. doi: 10.1177/0004563214545346.

Source: PubMed

Подписаться