Skip to main content
Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease logoLink to Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease
editorial
. 2020 May 5;9(10):e016553. doi: 10.1161/JAHA.120.016553

It's a “Gut Feeling”: Association of Microbiota, Trimethylamine N‐Oxide and Cardiovascular Outcomes

Gopi K Kolluru 1, Christopher G Kevil 1,
PMCID: PMC7660890  PMID: 32366196

Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide. Major adverse cardiac and cerebrovascular events (MACE), including chronic heart failure, myocardial infarction (MI), and stroke, are a major cause of premature death.1 Atherosclerosis is a classic hallmark of CVD and other associated complications, including coronary artery disease, peripheral artery disease, diabetes mellitus, stroke, hyperlipidemia, hypertension, and aging, which poses a substantial threat to human health.2, 3 Importantly, rupture of an atherosclerotic plaque results in occlusion of distal artery territories, thereby critically reducing blood flow to vital tissues.

Several established and novel emerging biomarkers of vascular disease, including hs‐CRP (high‐sensitivity C‐reactive protein), hs‐TnT (high‐sensitivity troponin T), and NT‐proBNP (N‐terminal pro‐B‐type natriuretic peptide), indicate various pathophysiological associations of CVD, including cardiovascular ischemic events and platelet activation.4 Prospective cohort studies have also suggested an increase in circulating trimethylamine N‐oxide (TMAO), a microbe‐dependent metabolite biomarker with enhanced thrombotic potential, to be associated with MACE.5, 6 Increasing evidence suggests a strong association between TMAO and cardiometabolic defects, atherosclerosis, platelet activation, proinflammatory signaling, and chronic kidney disease.7, 8, 9, 10 The effects of gut microbiota and subsequent changes in TMAO levels have been involved in the underlying mechanisms contributing to CVD risk. Various therapeutic strategies targeting TMAO metabolism have been explored with various degrees of success.11 Despite the accumulating evidence, the question remains whether TMAO is critically involved with MACE, independently involved with MI as a risk factor, and has a higher risk reduction for MACE with prolonged protective drugs.

Previous observations from the PEGASUS‐TIMI 54 (Prevention of Cardiovascular Events in Patients With Prior Heart Attack Using Ticagrelor Compared to Placebo on a Background of Aspirin Thrombolysis in Myocardial Infarction 54) trial, a randomized case–control study, suggests that ticagrelor reduces cardiovascular events among aspirin‐treated individuals with prior MI.12 This issue of the Journal of the American Heart Association (JAHA) includes a study by Gencer et al that analyzed data from the PEGASUS‐TIMI 54 trial revealing a strong association between TMAO and cardiovascular death or stroke.13 The authors reveal the prognostic value of TMAO as a biomarker for risk assessment in patients with CVD, and whether TMAO levels subside over prolonged treatment with ticagrelor after a MI. Interestingly, associations of TMAO were stronger for patients with normal renal function compared with those who had an estimated glomerular filtration rate of >60 mL/min per 1.73 m². While ticagrelor has shown consistent early and late beneficial effects for secondary prevention after MI, this study revealed that the efficacy of ticagrelor was independent of the baseline TMAO (N=1803).

Established reports suggest a positive correlation between increased TMAO and adverse cardiovascular effects.14, 15, 16 This study used odds ratios to determine the association of TMAO quartiles with the incidence of cardiovascular events and death.13 After adjustment for potential confounding variables, including baseline clinical characteristics such as age, sex, estimated glomerular filtration rate, region, body mass index, hypertension, hypercholesterolemia, diabetes mellitus, smoking, peripheral artery disease, index event, aspirin dosage, and treatment arm, the findings show a significant association between higher TMAO levels and incident cardiovascular death or stroke. However, further adjustment for cardiovascular biomarkers—hs‐TnT, hs‐CRP, and NT‐proBNP—attenuated the significance of associations between TMAO and cardiovascular death or stroke. Importantly, a significant reduction in MACE with ticagrelor was observed across TMAO quartiles.

TMAO is a metabolite of gut microflora, generated from dietary choline, betaine, and l‐carnitine.11, 16 As such, dietary components significantly influence the levels of TMAO. Animal‐origin foods such as meat (especially red meat) and eggs are a rich source of l‐carnitine, and choline can trigger proatherosclerotic conditions, thereby elevating TMAO production. Multiple studies have established the association between chronic red meat consumption to elevated TMAO levels and the risk of heart disease alike in both men and women.17, 18 Interestingly, γ‐butyrobetaine, a metabolite of dietary l‐carnitine, can convert l‐carnitine to the TMAO that is pro‐atherogenic.19 Previous studies establish an association between increased plasma choline/betaine levels and the risk of MACE with elevated TMAO.15, 20 Conversely, in the current study no such significance was observed between plasma choline/betaine and the risks of MACE. It would be informative to identify the changes in precursor levels, including γ‐butyrobetaine or l‐carnitine levels that influence TMAO production, as it is evident that diet influences the association between TMAO and atherosclerosis. Subsequently, information on nutritional habits of the study population has to be considered to clarify the association between TMAO–diet–CV outcome, which was not identified in this study.

Overall, Gencer et al13 establish a positive correlation between elevated TMAO and an increased risk of cardiovascular events. Their study further demonstrates prolonged ticagrelor treatment as an effective therapeutic measure with beneficial effects for cardiovascular outcomes, independent of TMAO levels. This study further establishes that elevated systemic TMAO levels in patients with prior MI could lead to increased risk of cardiovascular complications, including stroke and ultimately death. However, considering TMAO levels as a biomarker in the case of repeated MI is less robust compared with traditional clinical risk factors and previously established biomarkers.

Sources of Funding

This work was supported by an Institutional Development Award from the National Institutes of General Medical Sciences of the National Institutes of Health under grant No. P20GM121307 to Kevil.

Disclosures

None.

(J Am Heart Assoc. 2020;9:e016553 DOI: 10.1161/JAHA.120.016553.)

The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.

For Sources of Funding and Disclosures, see page 2.

References

  • 1. Reindl M, Reinstadler SJ, Feistritzer HJ, Mayr A, Klug G, Marschang P, Metzler B. Acute myocardial infarction as a manifestation of systemic vasculitis. Wien Klin Wochenschr. 2016;128:841–843. [DOI] [PubMed] [Google Scholar]
  • 2. Libby P, Theroux P. Pathophysiology of coronary artery disease. Circulation. 2005;111:3481–3488. [DOI] [PubMed] [Google Scholar]
  • 3. Dokken BB. The pathophysiology of cardiovascular disease and diabetes: beyond blood pressure and lipids. Diabetes Spectr. 2008;21:160–165. [Google Scholar]
  • 4. Ibrahim NE, Januzzi JL Jr. Established and emerging roles of biomarkers in heart failure. Circ Res. 2018;123:614–629. [DOI] [PubMed] [Google Scholar]
  • 5. Heianza Y, Ma W, Manson JE, Rexrode KM, 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. 2017;6:e004947 DOI: 10.1161/JAHA.116.004947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Ma J, Li H. The role of gut microbiota in atherosclerosis and hypertension. Front Pharmacol. 2018;9:1082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, Lusis AJ, Shih DM. Trimethylamine N‐oxide promotes vascular inflammation through signaling of mitogen‐activated protein kinase and nuclear factor‐kappaB. J Am Heart Assoc. 2016;5:e002767 DOI: 10.1161/JAHA.115.002767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. 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;472:57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Tomlinson JAP, Wheeler DC. The role of trimethylamine N‐oxide as a mediator of cardiovascular complications in chronic kidney disease. Kidney Int. 2017;92:809–815. [DOI] [PubMed] [Google Scholar]
  • 11. Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N‐oxide: the good, the bad and the unknown. Toxins (Basel). 2016;8:E326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Bonaca MP, Storey RF, Theroux P, Steg PG, Bhatt DL, Cohen MC, Im K, Murphy SA, Magnani G, Ophuis TO, et al. Efficacy and safety of ticagrelor over time in patients with prior MI in PEGASUS‐TIMI 54. J Am Coll Cardiol. 2017;70:1368–1375. [DOI] [PubMed] [Google Scholar]
  • 13. Gencer B, Li XS, Gurmu Y, Bonaca MP, Morrow DA, Cohen M, Bhatt DL, Steg GP, Storey RF, Johanson P, et al. Gut microbiota‐dependent trimethylamine N‐oxide and cardiovascular outcomes in patients with prior myocardial infarction: a nested case control study from the PEGASUS‐TIMI 54 trial. J Am Heart Assoc. 2020;9:e015331 DOI: 10.1161/JAHA.119.015331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Heianza Y, Ma W, DiDonato JA, Sun Q, Rimm EB, Hu FB, Rexrode KM, Manson JE, Qi L. Long‐term changes in gut microbial metabolite trimethylamine N‐oxide and coronary heart disease risk. J Am Coll Cardiol. 2020;75:763–772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. 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:E1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Yao ME, Liao PD, 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] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bernstein AM, Sun Q, Hu FB, Stampfer MJ, Manson JE, Willett WC. Major dietary protein sources and risk of coronary heart disease in women. Circulation. 2010;122:876–883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. 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] [PMC free article] [PubMed] [Google Scholar]
  • 19. 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] [PMC free article] [PubMed] [Google Scholar]
  • 20. Meyer KA, Shea JW. Dietary choline and betaine and risk of CVD: A systematic review and meta‐analysis of prospective studies. Nutrients. 2017;9:E711. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of the American Heart Association: Cardiovascular and Cerebrovascular Disease are provided here courtesy of Wiley

RESOURCES