Gut microbiota-dependent metabolite trimethylamine N-oxide (TMAO) and cardiovascular risk in patients with suspected functionally relevant coronary artery disease (fCAD)

Melissa Amrein, Xinmin S Li, Joan Walter, Zeneng Wang, Tobias Zimmermann, Ivo Strebel, Ursina Honegger, Kathrin Leu, Ibrahim Schäfer, Raphael Twerenbold, Christian Puelacher, Noemi Glarner, Thomas Nestelberger, Luca Koechlin, Benjamin Ceresa, Philip Haaf, Adam Bakula, Michael Zellweger, Stanley L Hazen, Christian Mueller, Melissa Amrein, Xinmin S Li, Joan Walter, Zeneng Wang, Tobias Zimmermann, Ivo Strebel, Ursina Honegger, Kathrin Leu, Ibrahim Schäfer, Raphael Twerenbold, Christian Puelacher, Noemi Glarner, Thomas Nestelberger, Luca Koechlin, Benjamin Ceresa, Philip Haaf, Adam Bakula, Michael Zellweger, Stanley L Hazen, Christian Mueller

Abstract

Background: Trimethylamine N-oxide (TMAO) has been associated with cardiovascular outcomes. However, the diagnostic value of TMAO and its precursors have not been assessed for functionally relevant coronary artery disease (fCAD) and its prognostic potential in this setting needs to be evaluated.

Methods: Among 1726 patients with suspected fCAD serum TMAO, and its precursors betaine, choline and carnitine, were quantified using liquid chromatography tandem mass spectrometry. Diagnosis of fCAD was performed by myocardial perfusion single photon emission tomography (MPI-SPECT) and coronary angiography blinded to marker concentrations. Incident all-cause death, cardiovascular death (CVD) and myocardial infarction (MI) were assessed during 5-years follow-up.

Results: Concentrations of TMAO, betaine, choline and carnitine were significantly higher in patients with fCAD versus those without (TMAO 5.33 μM vs 4.66 μM, p < 0.001); however, diagnostic accuracy was low (TMAO area under the receiver operating curve [AUC]: 0.56, 95% CI [0.53-0.59], p < 0.001). In prognostic analyses, TMAO, choline and carnitine above the median were associated with significantly (p < 0.001 for all) higher cumulative events for death and CVD during 5-years follow-up. TMAO remained a significant predictor for death and CVD even in full models adjusted for renal function (HR = 1.58 (1.16, 2.14), p = 0.003; HR = 1.66 [1.07, 2.59], p = 0.025). Prognostic discriminative accuracy for TMAO was good and robust for death and CVD (2-years AUC for CVD 0.73, 95% CI [0.65-0.80]).

Conclusion: TMAO and its precursors, betaine, choline and carnitine were significantly associated with fCAD, but with limited diagnostic value. TMAO was a strong predictor for incident death and CVD in patients with suspected fCAD.

Clinical trial registration: NCT01838148.

Keywords: Cardiovascular death; Functionally relevant coronary artery disease (fCAD); Gut microbiota; Incident major adverse cardiac events (mace); Myocardial infarction; Trimethylamine N-oxide (TMAO).

Conflict of interest statement

Dr. Walter reports a research grant from the Swiss Academy of Medical Sciences and the Bangerter Foundation (YTCR 23/17). Dr. Twerenbold reports receiving research support from the Swiss National Science Foundation (P300PB_167803), the Swiss Heart Foundation, the Swiss Society of Cardiology, the University Hospital of Basel, as well as speaker honoraria/consulting honoraria from Roche Diagnostics, Abbott Diagnostics, Siemens, Singulex and Brahms. Dr. Nestelberger received speaker honoraria from Beckman-Coulter. Dr. Koechlin has received a research grant from the University of Basel, the Swiss Academy of Medical Sciences and the Gottfried and Julia Bangerter-Rhyner Foundation, as well as the “Freiwillige Akademische Gesellschaft Basel”, outside the submitted work. Professor Mueller reports receiving research support from the Swiss National Science Foundation, the Swiss Heart Foundation, the KTI, the European Union, the Stiftung für kardiovaskuläre Forschung Basel, the University of Basel, the University Hospital Basel, Abbott, Beckman Coulter, Biomerieux, Brahms, Ortho Diagnostics, Roche, Siemens, Singulex, Sphingotec, as well as speaker honoraria/consulting honoraria from Abbott, Amgen, Astra Zeneca, Biomerieux, Boehringer Ingelheim, BMS, Brahms, Cardiorentis, Novartis, Roche, Sanofi, Siemens, and Singulex. Professor Hazen and Professor Wang were supported in part by grants from the National Institutes of Health and the Office of Dietary Supplements (P01HL147823, HL103866, HL126827, HL130819 and the Leducq Foundation). Mass spectrometry studies were performed on instruments housed in a facility supported in part by a Center of Excellence Award by Shimadzu Scientific Instruments. Professor Hazen and Professor Wang report being named as co-inventor on pending and issued patents held by the Cleveland Clinic relating to cardiovascular diagnostics and therapeutics and being eligible to receive royalty payments for inventions or discoveries related to cardiovascular diagnostics or therapeutics from Cleveland HeartLab, Quest Diagnostics, and Procter & Gamble. SL Hazen also reports being a paid consultant for Procter & Gamble and having received research funds from Procter & Gamble and Roche Diagnostics.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Levels of TMAO, betaine, choline and carnitine compared between patients adjudicated to have fCAD (A, C, E, G) and compared between patients adjudicated to have fCAD within subgroups of patients with and without history of CAD (B, D, F, H)
Fig. 2
Fig. 2
Kaplan Meier survival analysis of TMAO combined with low and high levels hs-Troponin (below/above URL of 14 ng/L; panel A) and TMAO combined with low and normal ranges of eGFR (> 60 mL/min/1.73m2 considered normal eGFR; panel B—in patients with available eGFR data (n = 921))
Fig. 3
Fig. 3
Time-dependent AUC of ROC of the four markers and over 5-years for all-cause death (223 events), cardiovascular death (115 events), AMI (88 events) and mace (203 events). Each step represents a change in AUC due to an event or censoring. AUC in the first days (ca. 180) vary due to small number of events in early stages

References

    1. 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.
    1. Tang WHW, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–1584. doi: 10.1056/NEJMoa1109400.
    1. Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of l-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–585. doi: 10.1038/nm.3145.
    1. Roncal C, Martínez-Aguilar E, Orbe J, et al. Trimethylamine (Tma) and trimethylamine-N-oxide (Tmao) as predictors of cardiovascular mortality in peripheral artery disease. Atherosclerosis. 2019 doi: 10.1016/j.atherosclerosis.2019.06.716.
    1. Jonsson AL, Bäckhed F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 2017;14:79–87. doi: 10.1038/nrcardio.2016.183.
    1. Yang S, Li X, Yang F, et al (2019) Gut microbiota-dependent marker TMAO in promoting cardiovascular disease: inflammation mechanism, clinical prognostic, and potential as a therapeutic target. Front Pharmacol 10:1360. 10.3389/fphar.2019.01360. PMID: 31803054; PMCID: PMC6877687
    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. Zhu W, Wang Z, Tang WHW, Hazen SL. Gut microbe-generated trimethylamine N-oxide from dietary choline is prothrombotic in subjects. Circulation. 2017;135:1671–1673. doi: 10.1161/CIRCULATIONAHA.116.025338.
    1. Tang WHW, 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. 2014;116:448–455. doi: 10.1161/CIRCRESAHA.116.305360.
    1. Li XS, Obeid S, Wang Z, et al. Trimethyllysine, a trimethylamine N-oxide precursor, provides near- and long-term prognostic value in patients presenting with acute coronary syndromes. Eur Heart J. 2019;40:2700–2709. doi: 10.1093/eurheartj/ehz259.
    1. Senthong V, Wang Z, Li XS, et al. Intestinal microbiota-generated metabolite trimethylamine-N-oxide and 5-year mortality risk in stable coronary artery disease: the contributory role of intestinal microbiota in a COURAGE-like patient cohort. J Am Heart Assoc. 2016 doi: 10.1161/JAHA.115.002816.
    1. Koeth RA, Levison BS, Culley MK, 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. Seldin MM, Meng Y, Qi H, et al. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J Am Heart Assoc. 2016 doi: 10.1161/JAHA.115.002767.
    1. Ma G, Pan B, Chen Y et al (2017) Trimethylamine N-oxide in atherogenesis: impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci Rep. 10.1042/BSR20160244
    1. Rohrmann S, Linseisen J, Allenspach M, et al. Plasma concentrations of trimethylamine-N-oxide are directly associated with dairy food consumption and low-grade inflammation in a German adult population. J Nutr. 2016 doi: 10.3945/jn.115.220103.
    1. Chen M-L, Zhu X-H, Ran L, et al. Trimethylamine-N-oxide induces vascular inflammation by activating the NLRP3 inflammasome through the SIRT3-SOD2-mtROS signaling pathway. J Am Heart Assoc. 2017 doi: 10.1161/JAHA.117.006347.
    1. Zhu W, Gregory JC, Org E, et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016 doi: 10.1016/j.cell.2016.02.011.
    1. Li XS, Wang Z, Cajka T, et al. Untargeted metabolomics identifies trimethyllysine, a TMAO-producing nutrient precursor, as a predictor of incident cardiovascular disease risk. JCI Insight. 2018 doi: 10.1172/jci.insight.99096.
    1. Roberts AB, Gu X, Buffa JA, et al. Development of a gut microbe–targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med. 2018;24:1407–1417. doi: 10.1038/s41591-018-0128-1.
    1. Heianza Y, Ma W, Manson JE, et al. Gut microbiota metabolites and risk of major adverse cardiovascular disease events and death: a systematic review and meta-analysis of prospective studies. J Am Heart Assoc. 2017 doi: 10.1161/JAHA.116.004947.
    1. Schiattarella GG, Sannino A, Toscano E, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J. 2017;38:2948–2956. doi: 10.1093/eurheartj/ehx342.
    1. Farhangi MA, Vajdi M, Asghari-Jafarabadi M. Gut microbiota-associated metabolite trimethylamine N-Oxide and the risk of stroke: a systematic review and dose-response meta-analysis. Nutr J. 2020;19:76. doi: 10.1186/s12937-020-00592-2.
    1. Walter J, du Fay de Lavallaz J, Koechlin L, et al. Using high-sensitivity cardiac troponin for the exclusion of inducible myocardial ischemia in symptomatic patients: a cohort study. Ann Intern Med. 2020;172:175–185. doi: 10.7326/M19-0080.
    1. Meyer KA, Benton TZ, Bennett BJ, et al. Microbiota-dependent metabolite trimethylamine n-oxide and coronary artery calcium in the coronary artery risk development in young adults study (CARDIA) J Am Heart Assoc. 2016 doi: 10.1161/JAHA.116.003970.
    1. Ladapo JA, Blecker S, Douglas PS. Physician decision making and trends in the use of cardiac stress testing in the united states: an analysis of repeated cross-sectional data. Ann Intern Med. 2014;161:482. doi: 10.7326/M14-0296.
    1. Mueller D, Puelacher C, Honegger U, et al. Direct comparison of cardiac troponin T and I using a uniform and a sex-specific approach in the detection of functionally relevant coronary artery disease. Clin Chem. 2018;64:1596–1606. doi: 10.1373/clinchem.2018.286971.
    1. Walter JE, Honegger U, Puelacher C, et al. Prospective validation of a biomarker-based rule out strategy for functionally relevant coronary artery disease. Clin Chem. 2018;64:386–395. doi: 10.1373/clinchem.2017.277210.
    1. Bossuyt PM, Reitsma JB, Bruns DE, et al. STARD 2015: an updated list of essential items for reporting diagnostic accuracy studies. Clin Chem. 2015;61:1446–1452. doi: 10.1373/clinchem.2015.246280.
    1. Giannitsis E, Becker M, Kurz K, et al. High-sensitivity cardiac troponin T for early prediction of evolving non–ST-segment elevation myocardial infarction in patients with suspected acute coronary syndrome and negative troponin results on admission. Clin Chem. 2010;56:642–650. doi: 10.1373/clinchem.2009.134460.
    1. SomaLogic I (2015) SOMAscan Proteomic Assay Technical White Paper. SomaLogic 1–14
    1. Lee G, Twerenbold R, Tanglay Y, et al. Clinical benefit of high-sensitivity cardiac troponin I in the detection of exercise-induced myocardial ischemia. Am Heart J. 2016;173:8–17. doi: 10.1016/j.ahj.2015.11.010.
    1. Tanglay Y, Twerenbold R, Lee G, et al. Incremental value of a single high-sensitivity cardiac troponin I measurement to rule out myocardial ischemia. Am J Med. 2015;128:638–646. doi: 10.1016/j.amjmed.2015.01.009.
    1. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics. 1988;44:837. doi: 10.2307/2531595.
    1. Kanitsoraphan C, Rattanawong P, Charoensri S, Senthong V. Trimethylamine N-oxide and risk of cardiovascular disease and mortality. Curr Nutr Rep. 2018;7:207–213. doi: 10.1007/s13668-018-0252-z.
    1. Blanche P, Dartigues J-F, Jacqmin-Gadda H. Estimating and comparing time-dependent areas under receiver operating characteristic curves for censored event times with competing risks. Stat Med. 2013;32:5381–5397. doi: 10.1002/sim.5958.
    1. Li XS, Obeid S, Klingenberg R, et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: a prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur Heart J. 2017;38:814–824. doi: 10.1093/eurheartj/ehw582.
    1. Roncal C, Martínez-Aguilar E, Orbe J, et al. Trimethylamine-N-Oxide (TMAO) predicts cardiovascular mortality in peripheral artery disease. Sci Rep. 2019;9:15580. doi: 10.1038/s41598-019-52082-z.
    1. Skye SM, Zhu W, Romano KA, et al. Microbial transplantation with human gut commensals containing cut C is sufficient to transmit enhanced platelet reactivity and thrombosis potential. Circ Res. 2018;123:1164–1176. doi: 10.1161/CIRCRESAHA.118.313142.
    1. Haghikia A, Li XS, Liman TG, et al. Gut microbiota-dependent trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke and is related to proinflammatory monocytes. Arterioscler Thromb Vasc Biol. 2018;38:2225–2235. doi: 10.1161/ATVBAHA.118.311023.
    1. Senthong V, Wang Z, Fan Y, et al. Trimethylamine N-oxide and mortality risk in patients with peripheral artery disease. J Am Heart Assoc. 2016;5:1–8. doi: 10.1161/JAHA.116.004237.
    1. Koay YC, Chen Y-C, Wali JA, et al. Plasma levels of TMAO can be increased with “healthy” and “unhealthy” diets and do not correlate with the extent of atherosclerosis but with plaque instability. Cardiovasc Res. 2020 doi: 10.1093/cvr/cvaa094.
    1. Bain MA, Faull R, Fornasini G, et al. Accumulation of trimethylamine and trimethylamine-N-oxide in end-stage renal disease patients undergoing haemodialysis. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc. 2006;21:1300–1304. doi: 10.1093/ndt/gfk056.
    1. Stubbs JR, House JA, Ocque AJ, et al. Serum trimethylamine-N-oxide is elevated in CKD and correlates with coronary atherosclerosis burden. J Am Soc Nephrol. 2016;27:305–313. doi: 10.1681/asn.2014111063.
    1. Mafune A, Iwamoto T, Tsutsumi Y, et al. Associations among serum trimethylamine-N-oxide (TMAO) levels, kidney function and infarcted coronary artery number in patients undergoing cardiovascular surgery: a cross-sectional study. Clin Exp Nephrol. 2016;20:731–739. doi: 10.1007/s10157-015-1207-y.
    1. Gruppen EG, Garcia E, Connelly MA, et al. TMAO is associated with mortality: impact of modestly impaired renal function. Sci Rep. 2017;7:13781. doi: 10.1038/s41598-017-13739-9.
    1. Turin TC, James MT, Jun M, et al. Short‐term change in eGFR and risk of cardiovascular events. J Am Heart Assoc. 2014;3:e000997. doi: 10.1161/JAHA.114.000997.
    1. Chen Q, Zhang Y, Ding D, et al. Estimated glomerular filtration rate and mortality among patients with coronary heart disease. PLoS One. 2016;11:e0161599. doi: 10.1371/journal.pone.0161599.
    1. Matsushita K, Selvin E, Bash LD, et al. Change in estimated GFR associates with coronary heart disease and mortality. J Am Soc Nephrol. 2009;20:2617–2624. doi: 10.1681/ASN.2009010025.
    1. Puelacher C, Wagener M, Honegger U, et al. Combining high-sensitivity cardiac troponin and B-type natriuretic peptide in the detection of inducible myocardial ischemia. Clin Biochem. 2018;52:33–40. doi: 10.1016/j.clinbiochem.2017.10.014.
    1. Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-oxide: the good, the bad and the unknown. Toxins (Basel) 2016;8:326. doi: 10.3390/toxins8110326.

Source: PubMed

3
Subskrybuj