Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Development of a gut microbe–targeted nonlethal therapeutic to inhibit thrombosis potential

This article has been updated

Abstract

Trimethylamine N-oxide (TMAO) is a gut microbiota–derived metabolite that enhances both platelet responsiveness and in vivo thrombosis potential in animal models, and TMAO plasma levels predict incident atherothrombotic event risks in human clinical studies. TMAO is formed by gut microbe–dependent metabolism of trimethylamine (TMA) moiety-containing nutrients, which are abundant in a Western diet. Here, using a mechanism-based inhibitor approach targeting a major microbial TMA-generating enzyme pair, CutC and CutD (CutC/D), we developed inhibitors that are potent, time-dependent, and irreversible and that do not affect commensal viability. In animal models, a single oral dose of a CutC/D inhibitor significantly reduced plasma TMAO levels for up to 3 d and rescued diet-induced enhanced platelet responsiveness and thrombus formation, without observable toxicity or increased bleeding risk. The inhibitor selectively accumulated within intestinal microbes to millimolar levels, a concentration over 1-million-fold higher than needed for a therapeutic effect. These studies reveal that mechanism-based inhibition of gut microbial TMA and TMAO production reduces thrombosis potential, a critical adverse complication in heart disease. They also offer a generalizable approach for the selective nonlethal targeting of gut microbial enzymes linked to host disease limiting systemic exposure of the inhibitor in the host.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Proof of concept that microbial choline TMA lyase inhibition can attenuate choline diet–enhanced platelet aggregation and in vivo thrombus formation.
Fig. 2: IMC and FMC are nonlethal, irreversible, and noncompetitive CutC/D inhibitors.
Fig. 3: The in vivo pharmacokinetic and pharmacodynamic properties of FMC and IMC.
Fig. 4: The mechanism-based CutC/D inhibitors IMC and FMC reverse choline diet–enhanced platelet responsiveness and thrombus formation.
Fig. 5: A microbial choline TMA lyase inhibitor reverses diet-induced changes in cecal microbial community composition associated with plasma TMAO levels, platelet responsiveness, and in vivo thrombosis potential.

Similar content being viewed by others

Change history

  • 21 September 2018

    In the Supplementary Information originally published with this article, Supplementary Fig. 1 was inadvertently omitted. The full version of the Supplementary Information is now available online.

References

  1. Kau, A. L., Ahern, P. P., Griffin, N. W., Goodman, A. L. & Gordon, J. I. Human nutrition, the gut microbiome and the immune system. Nature 474, 327–336 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Blaser, M. J. The microbiome revolution. J. Clin. Invest. 124, 4162–4165 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Fischbach, M. A. & Segre, J. A. Signaling in host-associated microbial communities. Cell 164, 1288–1300 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Aron-Wisnewsky, J. & Clément, K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders. Nat. Rev. Nephrol. 12, 169–181 (2016).

    CAS  PubMed  Google Scholar 

  5. Schroeder, B. O. & Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 22, 1079–1089 (2016).

    CAS  PubMed  Google Scholar 

  6. Koopen, A. M., Groen, A. K. & Nieuwdorp, M. Human microbiome as therapeutic intervention target to reduce cardiovascular disease risk. Curr. Opin. Lipidol. 27, 615–622 (2016).

    CAS  PubMed  Google Scholar 

  7. Wang, Z. et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472, 57–63 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Tang, W. H. W. et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 368, 1575–1584 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Koeth, R. A. et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 19, 576–585 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Wang, Z. et al. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur. Heart J. 35, 904–910 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Tang, W. H. W. et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J. Am. Coll. Cardiol. 64, 1908–1914 (2014).

    CAS  PubMed  Google Scholar 

  12. Tang, W. H. W. 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. 116, 448–455 (2015).

    CAS  PubMed  Google Scholar 

  13. Tang, W. H. W. et al. Intestinal microbiota–dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J. Card. Fail. 21, 91–96 (2015).

    CAS  PubMed  Google Scholar 

  14. Organ, C. L. et al. Choline diet and its gut microbe–derived metabolite, trimethylamine N-oxide, exacerbate pressure overload–induced heart failure. Circ. Heart Fail. 9, e002314 (2016).

    CAS  PubMed  Google Scholar 

  15. Zhu, W. et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 165, 111–124 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Senthong, V. 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. 5, e002816 (2016).

    PubMed  PubMed Central  Google Scholar 

  17. Warrier, M. et al. The TMAO-generating enzyme flavin monooxygenase 3 is a central regulator of cholesterol balance. Cell Rep. 10, 326–338 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Seldin, M. M. et al. Trimethylamine N-oxide promotes vascular inflammation through signaling of mitogen-activated protein kinase and nuclear factor-κB. J. Am. Heart Assoc. 5, e002767 (2016).

    PubMed  PubMed Central  Google Scholar 

  19. Li, T., Chen, Y., Gua, C. & Li, X. Elevated circulating trimethylamine N-oxide levels contribute to endothelial dysfunction in aged rats through vascular inflammation and oxidative stress. Front. Physiol. 8, 350 (2017).

    PubMed  PubMed Central  Google Scholar 

  20. Yue, C. et al. Trimethylamine N-oxide prime NLRP3 inflammasome via inhibiting ATG16L1-induced autophagy in colonic epithelial cells. Biochem. Biophys. Res. Commun. 490, 541–551 (2017).

    CAS  PubMed  Google Scholar 

  21. Yano, J. M. et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 161, 264–276 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Fusaro, M. et al. Vitamin K plasma levels determination in human health. Clin. Chem. Lab. Med 55, 789–799 (2017).

    CAS  PubMed  Google Scholar 

  23. Jäckel, S. et al. Gut microbiota regulate hepatic von Willebrand factor synthesis and arterial thrombus formation via Toll-like receptor-2. Blood 130, 542–553 (2017).

    PubMed  Google Scholar 

  24. Zhu, W., Wang, Z., Tang, W. H. W. & Hazen, S. L. Gut microbe-generated trimethylamine N-oxide from dietary choline is prothrombotic in subjects. Circulation 135, 1671–1673 (2017).

    PubMed  PubMed Central  Google Scholar 

  25. Heianza, Y., Ma, W., Manson, J. E., Rexrode, K. M. & Qi, L. 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. 6, e004947 (2017).

    PubMed  PubMed Central  Google Scholar 

  26. Schiattarella, G. G. et al. Gut microbe–generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose–response meta-analysis. Eur. Heart J. 38, 2948–2956 (2017).

    CAS  PubMed  Google Scholar 

  27. Qi, J. et al. 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. 22, 185–194 (2018).

    CAS  PubMed  Google Scholar 

  28. Brown, J. M. & Hazen, S. L. Targeting of microbe-derived metabolites to improve human health: The next frontier for drug discovery. J. Biol. Chem. 292, 8560–8568 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Dolphin, C. T., Janmohamed, A., Smith, R. L., Shephard, E. A. & Phillips, I. R. Missense mutation in flavin-containing mono-oxygenase 3 gene, FMO3, underlies fish-odour syndrome. Nat. Genet. 17, 491–494 (1997).

    CAS  PubMed  Google Scholar 

  30. Bennett, B. J. et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 17, 49–60 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Phillips, G. B. The lipid composition of human bile. Biochim. Biophys. Acta 41, 361–363 (1960).

    CAS  PubMed  Google Scholar 

  32. Craciun, S., Marks, J. A. & Balskus, E. P. Characterization of choline trimethylamine-lyase expands the chemistry of glycyl radical enzymes. ACS Chem. Biol. 9, 1408–1413 (2014).

    CAS  PubMed  Google Scholar 

  33. Romano, K. A., Vivas, E. I., Amador-Noguez, D. & Rey, F. E. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio 6, e02481 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. Martínez-del Campo, A. et al. Characterization and detection of a widely distributed gene cluster that predicts anaerobic choline utilization by human gut bacteria. MBio 6, e00042–15 (2015).

    PubMed  PubMed Central  Google Scholar 

  35. Wang, Z. et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 163, 1585–1595 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Furie, B. & Furie, B. C. Mechanisms of thrombus formation. N. Engl. J. Med. 359, 938–949 (2008).

    CAS  PubMed  Google Scholar 

  37. Bobadilla, R. V. Acute coronary syndrome: focus on antiplatelet therapy. Crit. Care Nurse 36, 15–27 (2016).

    PubMed  Google Scholar 

  38. Levine, G. N. et al. 2016 ACC/AHA guideline focused update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Thorac. Cardiovasc. Surg. 152, 1243–1275 (2016).

    PubMed  Google Scholar 

  39. Jennings, L. K. Mechanisms of platelet activation: need for new strategies to protect against platelet-mediated atherothrombosis. Thromb. Haemost. 102, 248–257 (2009).

    CAS  PubMed  Google Scholar 

  40. Manchikanti, L. et al. Assessment of bleeding risk of interventional techniques: a best evidence synthesis of practice patterns and perioperative management of anticoagulant and antithrombotic therapy. Pain. Physician 16, SE261–SE318 (2013).

    PubMed  Google Scholar 

  41. Cohen, M. Expanding the recognition and assessment of bleeding events associated with antiplatelet therapy in primary care. Mayo Clin. Proc. 84, 149–160 (2009).

    PubMed  PubMed Central  Google Scholar 

  42. Bodea, S., Funk, M. A., Balskus, E. P. & Drennan, C. L. Molecular basis of C–N bond cleavage by the glycyl radical enzyme choline trimethylamine-lyase. Cell Chem. Biol. 23, 1206–1216 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Chen, M. L. 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 7, e02210–e02215 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Walsh, C. T. Suicide substrates, mechanism-based enzyme inactivators: recent developments. Annu. Rev. Biochem. 53, 493–535 (1984).

    CAS  PubMed  Google Scholar 

  45. Hazen, S. L., Zupan, L. A., Weiss, R. H., Getman, D. P. & Gross, R. W. Suicide inhibition of canine myocardial cytosolic calcium-independent phospholipase A2. Mechanism-based discrimination between calcium-dependent and -independent phospholipases A2. J. Biol. Chem. 266, 7227–7232 (1991).

    CAS  PubMed  Google Scholar 

  46. Sandhu, S. S. & Chase, T. Jr. Aerobic degradation of choline by Proteus mirabilis: enzymatic requirements and pathway. Can. J. Microbiol. 32, 743–750 (1986).

    CAS  PubMed  Google Scholar 

  47. Romano, K. A. et al. Metabolic, epigenetic, and transgenerational effects of gut bacterial choline consumption. Cell Host Microbe 22, 279–290.e7 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Nemzek, J. A., Bolgos, G. L., Williams, B. A. & Remick, D. G. Differences in normal values for murine white blood cell counts and other hematological parameters based on sampling site. Inflamm. Res. 50, 523–527 (2001).

    CAS  PubMed  Google Scholar 

  49. Koeth, R. A. et al. γ-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of l-carnitine to TMAO. Cell Metab. 20, 799–812 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu, Y., Jennings, N. L., Dart, A. M. & Du, X.-J. Standardizing a simpler, more sensitive and accurate tail bleeding assay in mice. World J. Exp. Med. 2, 30–36 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Derrien, M., Belzer, C. & de Vos, W. M. Akkermansia muciniphila and its role in regulating host functions. Microb. Pathog. 106, 171–181 (2017).

    PubMed  Google Scholar 

  52. Gachet, C. Antiplatelet drugs: which targets for which treatments? J. Thromb. Haemost. 13, S313–S322 (2015).

    CAS  PubMed  Google Scholar 

  53. Osbourn, A. E. & Field, B. Operons. Cell. Mol. Life Sci. 66, 3755–3775 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Craciun, S. & Balskus, E. P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl Acad. Sci. USA 109, 21307–21312 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Berthoumieux, S. et al. Shared control of gene expression in bacteria by transcription factors and global physiology of the cell. Mol. Syst. Biol. 9, 634 (2013).

    PubMed  PubMed Central  Google Scholar 

  56. Clarke, D. D. Fluoroacetate and fluorocitrate: mechanism of action. Neurochem. Res. 16, 1055–1058 (1991).

    CAS  PubMed  Google Scholar 

  57. Imhann, F. et al. Proton pump inhibitors affect the gut microbiome. Gut 65, 740–748 (2016).

    CAS  PubMed  Google Scholar 

  58. Rogers, M. A. M. & Aronoff, D. M. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome. Clin. Microbiol. Infect. 22, 178.e1–178.e9 (2016).

    CAS  Google Scholar 

  59. Maier, L. et al. Extensive impact of nonantibiotic drugs on human gut bacteria. Nature 555, 623–628 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Wu, H. et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat. Med. 23, 850–858 (2017).

    CAS  PubMed  Google Scholar 

  61. Wang, Z. et al. Measurement of trimethylamine-N-oxide by stable isotope dilution liquid chromatography tandem mass spectrometry. Anal. Biochem. 455, 35–40 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Rath, S., Heidrich, B., Pieper, D. H. & Vital, M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 5, 54 (2017).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We appreciate the aid of J. A. Drazba and G. Deshpande of the Lerner Research Institute Imaging Core in studies using the Cellix microfluidic system. This research was supported by grants from the National Institutes of Health (NIH) and the Office of Dietary Supplements (HL103866, HL126827 and DK106000 (to S.L.H.), HL122283 and AA024333 (to J.M.B.), and HL28481 and HL30568 (to A.J.L.)). S.L.H. reports being supported in part by a grant from the Leducq Foundation. V.G. acknowledges a Faculty Research Development Award from Cleveland State University. A.B.R. was supported in part by a grant from the American Heart Association (15POST25750053). W.Z. was supported in part by an AHA Scientist Development Grant and an NIH StrokeNet Clinical Research and Training Grant. S.M.S. was supported in part by training grant T32DK007470 from the National Institute of Diabetes and Digestive and Kidney Disease (NIDDK) of the NIH. Some of the toxicology and safety studies were performed by Pharmaron. Mass spectrometry studies were performed on instrumentation housed in a facility supported in part through a Shimadzu Center of Excellence award. Computational resources were provided by the Extreme Science and Engineering Discovery Environment (National Science Foundation).

Author information

Authors and Affiliations

Authors

Contributions

A.B.R, J.A.B., and X.G designed, performed, and analyzed data from most of the studies. They also helped write the manuscript with input from all authors. A.G.H., A.D., V.G., A.J.M., and B.S.L aided in chemical synthesis and characterization of all compounds, computational drug design efforts, docking analyses, and quantum mechanical calculations. Z.W. and X.F. helped with design and performance of mass spectrometry analyses. W.Z., N.G., and M.W.R. helped in the design and performance of platelet functional studies, in vivo thrombosis, and other mouse experiments. S.M.S., J.M.L., L.L, W.T.B., and A.J.L. participated in microbe composition analyses and cut gene cluster transcription quantification studies. D.B.C., J.M.R., and J.C.G.-G. helped design and perform some of the human commensal and polymicrobial bioreactor studies characterizing inhibitor efficacy. J.A.D. provided overall advice, as well as performing plasmid cloning and construction use in multiple bacterial inhibitor studies. S.R., J.M.B., and J.C.G.-G. provided critical scientific input and discussions. S.L.H conceived, designed, and supervised all studies, and participated in the drafting and editing of the manuscript. All authors contributed to the critical review of the manuscript.

Corresponding author

Correspondence to Stanley L. Hazen.

Ethics declarations

Competing interests

S.L.H., X.G., Z.W., and B.S.L. are named as co-inventors on pending and issued patents held by the Cleveland Clinic relating to cardiovascular diagnostics or therapeutics. S.L.H., Z.W., and B.S.L. report having the right to receive royalty payment for inventions or discoveries related to cardiovascular diagnostics from Cleveland Heart Lab, Inc. and Quest Diagnostics. S.L.H. also reports having been paid as a consultant for P&G and receiving research funds from Astra Zeneca, P&G, Pfizer Inc., and Roche Diagnostics.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–19, Supplementary Table 1 and Supplementary Text

Reporting Summary

Supplementary Dataset

Combined source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roberts, A.B., Gu, X., Buffa, J.A. et al. Development of a gut microbe–targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med 24, 1407–1417 (2018). https://doi.org/10.1038/s41591-018-0128-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-018-0128-1

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research