Nothing Special   »   [go: up one dir, main page]

Next Article in Journal
Gene Therapy for Inherited Liver Disease: To Add or to Edit
Previous Article in Journal
Impairment of Skeletal Muscle Contraction by Inhibitors of GABA Transporters
Previous Article in Special Issue
The Microbiome–Genetics Axis in Autism Spectrum Disorders: A Probiotic Perspective
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbiome-Derived Trimethylamine N-Oxide (TMAO) as a Multifaceted Biomarker in Cardiovascular Disease: Challenges and Opportunities

Laboratory of Centre for Preclinical Research, Department of Experimental Physiology and Pathophysiology, Medical University of Warsaw, Banacha 1B, 02-091 Warsaw, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(23), 12511; https://doi.org/10.3390/ijms252312511
Submission received: 22 October 2024 / Revised: 19 November 2024 / Accepted: 19 November 2024 / Published: 21 November 2024

Abstract

:
Biomarkers play a crucial role in various stages of disease management, including screening, diagnosis, prediction, prognosis, treatment, and safety monitoring. Although they are powerful tools in disease diagnosis, management, and drug development, identifying and validating reliable biomarkers remains a significant challenge. Among potential microbiome-derived biomarkers, trimethylamine N-oxide (TMAO) has gained notable attention for its link to atherosclerosis and cardiovascular risk. However, despite the growing body of research on TMAO, its practical application in clinical settings for disease management and patient outcome enhancement is still not a reality. This paper presents recent data on the utility of TMAO as a cardiovascular biomarker, categorized by its various roles: diagnostic, prognostic, susceptibility/risk, monitoring, pharmacodynamic/response, predictive, and safety. It also briefly discusses research on TMAO’s potential role in cardiovascular disease development. While TMAO shows promise, particularly in prognostic applications, its reliability as a biomarker has been inconsistent across studies. These variances may result from several confounding factors that affect TMAO plasma levels, including diet, kidney function, and demographic variables. The review aims to elucidate the specific contexts in which TMAO can be valuable, potentially leading to more personalized and effective management of cardiovascular disease.

Graphical Abstract">

Graphical Abstract

1. Introduction

Recent decades have witnessed a surge in research activity focused on biomarkers. As defined by the FDA and NIH in the Biomarkers, EndpointS, and other Tools (BEST) Resource, a biomarker is a defined characteristic measured as an indicator of normal biological processes, pathogenic processes, or responses to exposure or intervention, including therapeutic interventions [1]. In practical terms, biomarkers are tools and technologies designed for various functions corresponding to different disease stages: screening, diagnosis, prediction, prognosis, treatment monitoring, and addressing safety concerns. While offering a powerful approach in disease management and drug development, identifying and properly validating biomarkers remains a challenge. Validation involves establishing if a biomarker’s performance is acceptable for its intended purpose (fit-for-purpose), i.e., whether it measures or predicts the clinical concept of interest [1]. There are many quantitative measures of biomarker performance such as sensitivity, specificity, positive and negative predictive values, likelihood ratios, receiver operating characteristic curve, etc. Actually, the desired properties of a biomarker may vary depending on its intended use. Either way, a clinically valuable biomarker has to significantly add to a patient assessment independently of already established predictors [2]. Its use should translate into improved patient outcomes. Therefore, while many biomarker candidates appear promising, very few advance into widespread clinical practice.
Abundant evidence indicates a robust mutual relationship between the gut microbiome and its host, encompassing cardiovascular and metabolic regulation [3,4]. Consequently, evaluating ‘microbial status’ or bacterial-origin compounds emerges as a potential tool for enhancing diagnostic accuracy and therapeutic response. Significantly, the rapid advancement in metagenomics and metabolomics has introduced numerous potential biomarker candidates, such as short-chain fatty acids and taurine [5]. In the context of the circulatory system, microbiome-derived trimethylamine N-oxide (TMAO) has garnered significant attention due to its association with atherosclerosis burden [6] and cardiovascular risk [7].

2. TMAO Metaorganismal Pathway

The human body generates TMAO through two primary pathways. The first involves the metabolism of dietary choline, carnitine, and their derivatives by the gut microbiome into trimethylamine (TMA). This metabolic process is facilitated by specific bacterial enzymes known as choline TMA lyases (CutC). TMA then crosses the gut–blood barrier, entering the portal circulation. In the liver, most of the TMA is oxidized into TMAO by the enzyme hepatic flavin-containing monooxygenase (FMO3). The second pathway is the direct intake of TMA and TMAO, which are abundantly found in fish and other seafood [8]. In fact, consuming fish may result in a greater increase in circulating TMAO levels compared to foods rich in choline and carnitine [9]. The major route of TMAO elimination is via the kidneys but to a small extent it can also be excreted in sweat and breath [10]. It is important to emphasize that the tissue distribution and kinetics of methylamines in the blood are largely unknown, and various factors must be considered when assessing TMAO levels. First, the absorption of methylamines in the intestine is influenced by diet, as well as the composition and metabolic activity of the gut microbiota. Additionally, the rate of TMA oxidation is contingent on the activity of the enzyme FMO3, which can vary based on sex, race, genetic polymorphisms, or the overall condition of the liver [11,12]. Lastly, kidney function, particularly the rate of excretion, significantly influences TMAO concentration [13]. These factors, along with others, lead to relatively high inter- and intra-individual variations [14] that can significantly impede the utility of TMAO as a biomarker (Figure 1).

3. TMAO as a Biomarker

Plasma TMAO has been suggested as a potential biomarker in cardiovascular disease. However, the robustness of TMAO as a biomarker has yielded conflicting results in various studies. These discrepancies may be partly due to numerous confounding factors that influence TMAO plasma levels, such as kidney function, diet, and demographic variables. In this paper, we present an overview of the latest data on the utility of TMAO as a biomarker, ensuring clarity and consistency in terminology based on definitions from the BEST Resource [2].

3.1. Diagnostic Biomarker

Diagnostic biomarkers are tools used to detect or confirm the presence of a disease (or its subtype) or other condition for which treatment may be indicated [2]. Elevated plasma TMAO levels have recently been associated with various cardiovascular disorders, including acute ischemic stroke [15,16,17], coronary artery disease [18], heart failure [19,20,21], and renal dysfunction [19] (Table 1). Additionally, several studies have indicated that TMAO levels may vary with the severity of these conditions. For instance, TMAO has shown diagnostic utility in distinguishing critical limb ischemia among patients with peripheral artery disease [22] and its levels correlate with the severity of coronary atherosclerosis in acute coronary syndrome patients [23,24,25]. Furthermore, in patients with ischemic stroke, TMAO was suggested to differentiate between mild and moderate to severe strokes [17] and identifying early neurological deterioration [26]. TMAO has also been explored for its potential in detecting complications in cardiovascular patients, such as thrombus formation in atrial fibrillation [27], kidney disease in hypertensive patients [28], and subclinical myocardial damage in high-risk patients [29]. It may even detect subclinical atherosclerosis in children with obesity [30]. While some studies have shown statistical differences in TMAO concentrations between affected and unaffected individuals, they often lack assessment using standard diagnostic methods like receiver operator characteristic curves [31,32,33,34,35]. Conversely, certain studies have not confirmed the diagnostic accuracy of TMAO in conditions like coronary artery disease [36] or atrial fibrillation [37].
Currently, there is no conclusive evidence that evaluating plasma TMAO levels significantly enhances established clinical assessments. For example, combining TMAO measurements with N-terminal prohormone of brain natriuretic peptide (NT-proBNP) yielded similar diagnostic results to NT-proBNP alone in heart failure patients with preserved ejection fraction [21].
Therefore, the data on the diagnostic utility of TMAO are promising and its validation as a diagnostic biomarker in the near future calls for further research.

3.2. Prognostic Biomarker

Prognostic biomarkers are employed to determine the likelihood of specific outcomes, such as future clinical events, disease recurrence, or progression, in populations with a disease [2]. Generally, assessing their usefulness requires a prospective study and a larger sample size than that needed for diagnostic biomarkers. In the case of TMAO, studies commonly report the odds or hazard ratios for specified outcomes, like all-cause mortality or major cardiac events, based on the quartiles of TMAO levels. The prognostic utility of TMAO has been suggested in conditions such as acute ischemic stroke, heart failure, peripheral and coronary artery disease, chronic kidney disease, among others (see Table 2 for references). Observations have linked high-plasma TMAO with long-term outcomes [38,39,40], and several studies have demonstrated its incremental prognostic value in conventional risk stratification [31,39,41,42,43]. For instance, incorporating TMAO into a model of traditional cardiovascular risk factors significantly improved the 5-year all-cause mortality risk assessment in stable coronary artery disease patients [42]. It has been postulated that measuring TMAO may be particularly useful in patients with heart failure with preserved ejection fraction (HFpEF), where natriuretic peptide concentrations are relatively lower than in heart failure with reduced ejection fraction (HFrEF), making risk stratification more challenging [44]. Research suggests that, in addition to hard clinical endpoints, plasma TMAO levels may also predict poor functional outcomes in stroke patients [31,45]; however, the data are not consistently supportive [32] (see Table 2). Unfortunately, many cited studies evaluating the prognostic value of plasma TMAO level did not adjust for estimated glomerular filtration rate (eGFR), i.e., renal function. It should be highlighted that low eGFR is a well-established risk factor for cardiovascular mortality [46,47]. In this context, many studies that included eGFR adjustment in their statistical model showed partial or complete attenuation of the associations between TMAO and adverse outcomes [43,48,49,50,51,52,53,54]. Moreover, several studies have linked TMAO to certain study endpoints but not others [32,55,56]. For example, TMAO was associated with cardiovascular death but not with major adverse cardiac events [56]. A study by Li et al. underscores the critical importance of timing in assessing TMAO concentrations, showing that only patients with high TMAO levels at both initial admission and at a one-month follow-up exhibited an increased risk of major adverse cardiovascular events (MACE) [57]. Future research should focus on identifying the patient populations, timepoints, and outcomes where circulating TMAO may have the most promising prognostic performance. Lastly, it remains unclear whether TMAO is a modifiable biomarker. Currently, there is scant clinical data on the effect of treatment on TMAO concentration (discussed further in Section 3.5, Pharmacodynamic/Response Biomarker).
In conclusion, most of the studies point to TMAO’s prognostic utility in various cardiovascular diseases; however, there is some inconsistency, probably due to variances in kidney function, patient populations, and timepoints of TMAO assessment.

3.3. Susceptibility/Risk Biomarker

A susceptibility/risk biomarker is one that indicates the potential for developing a disease or medical condition in individuals with no clinically apparent disease [2]. Conceptually, assessing TMAO could identify a population particularly susceptible to cardiovascular disease and encourage more aggressive modification of risk factors. There are a few studies that have evaluated the prognostic potential of TMAO in the general population. Zheng et al. prospectively investigated the interaction between circulating TMAO and future risk of incident cardiovascular disease in a sample from community-based Chinese adults. TMAO was associated with a higher risk, and analysis confirmed that a model including TMAO had better discrimination than one containing only traditional risk factors [97]. Two other studies on population-based cohorts, one Chinese and one US, revealed the association of TMAO with a higher stroke risk [98,99]. Similarly, TMAO provided incremental risk prediction for future coronary artery disease in the community-based EPIC-Norfolk population [100]. Other Norwegian cohorts have shown that plasma levels of TMAO are positively associated with a risk of incident atrial fibrillation [101]. Conversely, the prospective PREDIMED study on participants at high cardiovascular risk revealed that TMAO level was not associated with a risk of atrial fibrillation or heart failure [102]. Several other studies also do not support the role of TMAO as a susceptibility/risk biomarker. In a prospective nested case–control study conducted among individuals without diabetes, cardiovascular disease, or cancer, there was no association between TMAO and the risk of coronary artery disease [103]. Similarly, a cross-sectional study on elderly Japanese adults showed that TMAO was not a risk factor for atherosclerosis in this population [104]. In addition, in a large cohort of older US adults, a significant interaction between TMAO and the risk of future cardiovascular disease was found only in patients with impaired kidney function [105,106]. Incidentally, a recent study by Heianza et al. in the Nurses’ Health Study cohort showed that a 10-year increase in TMAO concentration corresponded to an increased risk of coronary heart disease, regardless of the initial level of TMAO and other conventional factors [107].
On the whole, data are contradictory, and the utility of TMAO as a risk biomarker in subjects with no clinically apparent disease requires further investigation.

3.4. Monitoring Biomarker

A monitoring biomarker is one that is measured repeatedly and is generally used to assess disease progression, including the occurrence of new disease effects, response to treatment, or evidence of exposure to an environmental agent [2]. Currently, there are no data specifically evaluating TMAO performance as a monitoring biomarker per se, but several studies have provided repeated measurements of TMAO. For instance, the study by Heianza et al., which was already mentioned, aimed to assess the association between temporal changes in plasma TMAO and the incidence of coronary heart disease. Therefore, they measured TMAO at two time points, approximately 10 years apart [107]. In another case, there was inconsistency in data regarding TMAO levels in stroke patients, and two studies assessed fluctuations in TMAO by determining its concentration at three different time points [32,47]. Specifically, they revealed that TMAO levels decreased significantly 48 h after stroke onset and increased again three months later.
All in all, current data do not provide sufficient evidence to evaluate TMAO performance as a monitoring biomarker.

3.5. Pharmacodynamic/Response Biomarker

A pharmacodynamic/response biomarker demonstrates a biological response to treatment or exposure to an environmental agent. Variations in this biomarker type offer preliminary evidence of a treatment’s effect on a desired clinical endpoint. It also provides insights into whether to continue a treatment or adjust the dosage [2]. Numerous animal studies have indicated the causative role of TMAO in cardiovascular pathology [6,108,109]. TMAO is postulated as a mediator linking disturbances in gut microbiota (i.e., dysbiosis) with adverse clinical outcomes. Accordingly, various studies have aimed at modifying the intestinal microbiome to subsequently lower TMAO levels [6,110]. In this context, TMAO can be considered a candidate pharmacodynamic/response biomarker, especially in microbiota-targeted therapies. However, human research in this area is scant. Early results from a small randomized placebo-controlled study suggest that polyphenols may reduce TMAO concentration [111]. Moreover, Park et al. conducted a dietary intervention study, observing a significant increase in TMAO levels during short-term popular diets [112]. In contrast, prolonged, high-dose probiotic supplementation did not alter TMAO concentrations [113,114,115,116] nor did treatment with rifaximin [117]. TMAO was also evaluated in studies assessing response to oral l-carnitine therapy in mitochondrial disorders [118,119]. While these studies presuppose TMAO’s pathological role, the data are conflicting and warrant further investigation [120].
As a potential response biomarker in cardiovascular diseases, the modifiability of TMAO levels by guideline-recommended therapy remains uncertain. The BIOSTAT-CHF study showed that standard heart failure treatment does not affect TMAO levels [57]. In patients with symptomatic heart failure, TMAO levels stayed elevated over the long term, even after left ventricular assist device implantation and heart transplant [121]. Conversely, Yang et al. observed a dynamic decrease in TMAO levels among stable and improved patients with pulmonary hypertension following treatment, but an upward trend among those with worsened risk status [122]. Data regarding the impact of statins and Sodium–Glucose Transport Protein 2 (SGLT2) inhibitors on TMAO concentrations are contradictory. Some studies suggest a causal relationship between statins and decreased TMAO levels [123] with statin use inversely associated with TMAO levels in adults at risk for atherosclerosis [124]. However, long-term statin therapy in diabetic patients did not affect plasma TMAO levels [125]. Experimental studies have suggested the inhibitory effect of SGLT2 inhibitors on TMAO precursors [126], but a secondary analysis of the EMMY trial indicated a significant increase in TMAO levels post-myocardial infarction in patients receiving Empagliflozin compared to those on standard treatment [127].
Therefore, the conflicting results underscore the need for further research into the effects of cardiovascular disease-modifying drugs on TMAO levels and their predictive value.

3.6. Predictive Biomarker

A predictive biomarker is instrumental in identifying individuals who may experience favorable or unfavorable effects from exposure to a treatment or environmental agent. Such biomarkers are commonly utilized to ascertain who might benefit from a specific treatment or to choose among various therapeutic options. Establishing a predictive biomarker necessitates comparing the intervention with a control treatment in individuals both with and without the biomarker [2]. To date, only a limited number of studies have evaluated the influence of TMAO on treatment efficacy. Specifically, Gencer et al. investigated how TMAO impacts the effectiveness of ticagrelor in patients with myocardial infarction. Their findings indicated that the reduction in major adverse cardiac events due to ticagrelor was consistent, irrespective of TMAO levels [75]. Their observation raises questions, especially if TMAO is indeed a mediator in cardiovascular diseases as some theories suggest. The limited experimental data from animal studies seem to support the potential role of TMAO [128], highlighting the need for more extensive research in this area to clarify its impact on therapeutic outcomes.
In conclusion, current data are not sufficient to evaluate TMAO performance as a predictive biomarker.

3.7. Safety Biomarker

A safety biomarker is crucial for detecting or predicting adverse effects from drug usage or environmental exposure. Ideally, such a biomarker would indicate the onset of toxicity before clinical symptoms or irreversible damage occur [2]. Currently, there are limited data on using TMAO as a safety biomarker. However, TMAO is recognized as a uremic toxin whose levels significantly increase with kidney function decline [13,58], suggesting its potential utility as a biomarker for renal toxicity. Notably, the direct linear relationship between the eGFR and TMAO levels remains unclear [129]. Additionally, there is a notable lack of data on TMAO concentration changes in cases of acute kidney injury [130]. This gap in knowledge highlights the need for further research to establish TMAO’s efficacy and reliability as a safety biomarker, particularly in renal toxicity contexts.
To sum up, there are limited data on using TMAO as a safety biomarker and this calls for further research.

4. TMAO as a Therapeutic Target

The role of TMAO in disease pathology, beyond its potential as a biomarker, remains a subject of debate (Figure 2). Animal studies have supported TMAO’s detrimental role by demonstrating various adverse effects following supplementation of TMAO or its dietary precursors [131]. Notably, TMAO intake has been linked to the exacerbation of heart failure [109,132,133], atherosclerosis development [134], impaired glucose tolerance [135], and liver dysfunction [136]. Dietary TMAO also increased platelet hyperreactivity and thrombosis risk [137], with evidence indicating that microbial CutC-dependent TMA production is sufficient to induce these effects [138]. There are several potential mechanisms of TMAO’s detrimental role. TMAO may promote vascular inflammation via NLRP3 inflammasome activation, which is partly mediated through the inhibition of the SIRT3–SOD2–mitochondrial ROS signaling pathway [139]. In this regard, TMAO has also been shown to impair β-oxidation and pyruvate metabolism in cardiac mitochondria [140]. It has been demonstrated that TMAO accelerate endothelial dysfunction, including decreasing endothelial self-repair and increasing monocyte adhesion via PKC/NF-κB/VCAM-1 [141,142]. In addition, TMAO’s pathological role in atrial fibrillation has been raised. Namely, TMAO, by activating p65 NF-κB signaling, altered the stability of atrial electrophysiology in normal canines and aggravated acute electrical remodeling in an atrial fibrillation canine model [143].
However, contrasting studies challenge TMAO’s pathological role [144]. Aldana-Hernández et al. found no association between TMAO and atherosclerosis development [145] and some research suggests a protective effect of TMAO precursors against atherosclerotic lesions [146]. Some studies indicate that TMAO supplementation leads to reduced fibrosis in the heart [147] and reduced mortality in heart failure rats alongside diuretic, natriuretic, and hypotensive effects [148]. Videja et al. suggest that TMAO accumulation in cardiac tissue improves mitochondrial energy metabolism [149]. In vitro studies present mixed results: while TMAO induces inflammation and endothelial dysfunction [139,150], it appears to be non-harmful to cardiomyocytes [151] or vascular smooth muscle cells [152]. These inconsistent findings leave TMAO’s role in cardiovascular pathology uncertain.
In contrast, TMA, the direct precursor of TMAO, has long been recognized for its toxic properties. As an industrial pollutant, TMA can cause various harmful effects, including eye or skin irritation, developmental toxicity, and neurological disorders at relatively low levels [153,154,155,156]. Surprisingly, its role in cardiovascular pathology is largely unexplored, though evidence suggests that TMA, not TMAO, may be the mediator in cardiovascular issues [129,157,158].
Targeting microbial-dependent TMA synthesis holds promise as a therapeutic approach. Reducing intestinal TMA production through antibiotics [7,133], probiotics [159], or non-lethal TMA lyase inhibitors [134,160,161,162,163] has shown potential. Studies confirm that inhibiting bacterial TMA production can mitigate atherosclerosis and related lipid disorders [134,159] and improve cardiac function [164,165]. These findings suggest TMAO as a modifiable risk factor, although further research, particularly in humans, is essential for conclusive evidence.

5. Future Directions

The burgeoning interest in TMAO research over the past decade underscores the necessity for high-quality data evaluating its potential as a biomarker. Despite the exponential growth in studies focusing on TMAO, its integration into general clinical practice for disease management and patient outcome improvement remains a distant goal. A significant gap in our current understanding involves the biological role of TMAO, which requires consensus and comprehensive investigation. Further research is essential to determine whether TMAO could serve as a superior marker compared to established cardiovascular markers like NT-proBNP or eGFR, particularly in specific patient populations. This necessitates comparative studies that not only measure the predictive accuracy of TMAO against these traditional markers but also explore its utility across diverse patient demographics, including those with varying cardiovascular risk profiles and comorbidities. Such investigations would help clarify the contexts in which TMAO could provide additional or enhanced prognostic value, potentially leading to more personalized and effective cardiovascular disease management strategies.

Author Contributions

M.U.: Conceptualization, funding acquisition, project administration, supervision, writing—original draft, and writing—review and editing. K.J.: Data curation, investigation, writing—original draft, and writing—review and editing. W.K.: Data curation, investigation, and writing—original draft. M.K.: Data curation, investigation, and writing—original draft. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by the National Science Centre, Poland, grant no: UMO-2020/37/B/NZ5/00366.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and Other Tools) Resource; Food and Drug Administration (US): Silver Spring, MD, USA, 2016.
  2. Vasan, R.S. Biomarkers of cardiovascular disease: Molecular basis and practical considerations. Circulation 2006, 113, 2335–2362. [Google Scholar] [CrossRef] [PubMed]
  3. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef] [PubMed]
  4. Witkowski, M.; Weeks, T.L.; Hazen, S.L. Gut microbiota and cardiovascular disease. Circ. Res. 2020, 127, 553–570. [Google Scholar] [CrossRef]
  5. Johnson, C.H.; Ivanisevic, J.; Siuzdak, G. Metabolomics: Beyond biomarkers and towards mechanisms. Nat. Rev. Mol. Cell Biol. 2016, 17, 451–459. [Google Scholar] [CrossRef]
  6. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef]
  7. Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [PubMed]
  8. Zeisel, S.H.; Warrier, M. Trimethylamine N-oxide, the microbiome, and heart and kidney disease. Annu. Rev. Nutr. 2017, 37, 157–181. [Google Scholar] [CrossRef]
  9. Cho, C.E.; Taesuwan, S.; Malysheva, O.V.; Bender, E.; Tulchinsky, N.F.; Yan, J.; Sutter, J.L.; Caudill, M.A. 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. [Google Scholar] [CrossRef]
  10. Al-Waiz, M.; Mitchell, S.; Idle, J.; Smith, R. The metabolism of 14C-labelled trimethylamine and its N-oxide in man. Xenobiotica 1987, 17, 551–558. [Google Scholar] [CrossRef]
  11. Janmohamed, A.; Hernandez, D.; Phillips, I.R.; Shephard, E.A. Cell-, tissue-, sex-and developmental stage-specific expression of mouse flavin-containing monooxygenases (Fmos). Biochem. Pharmacol. 2004, 68, 73–83. [Google Scholar] [CrossRef]
  12. Lattard, V.; Lachuer, J.; Buronfosse, T.; Garnier, F.; Benoit, E. Physiological factors affecting the expression of FMO1 and FMO3 in the rat liver and kidney. Biochem. Pharmacol. 2002, 63, 1453–1464. [Google Scholar] [CrossRef] [PubMed]
  13. Stubbs, J.R.; House, J.A.; Ocque, A.J.; Zhang, S.; Johnson, C.; Kimber, C.; Schmidt, K.; Gupta, A.; Wetmore, J.B.; Nolin, T.D.; et al. Serum Trimethylamine-N-Oxide is Elevated in CKD and Correlates with Coronary Atherosclerosis Burden. J. Am. Soc. Nephrol. JASN 2016, 27, 305–313. [Google Scholar] [CrossRef] [PubMed]
  14. Kuhn, T.; Rohrmann, S.; Sookthai, D.; Johnson, T.; Katzke, V.; Kaaks, R.; von Eckardstein, A.; Muller, D. Intra-individual variation of plasma trimethylamine-N-oxide (TMAO), betaine and choline over 1 year. Clin. Chem. Lab. Med. 2017, 55, 261–268. [Google Scholar] [CrossRef] [PubMed]
  15. Rexidamu, M.; Li, H.; Jin, H.; Huang, J. Serum levels of Trimethylamine-N-oxide in patients with ischemic stroke. Biosci. Rep. 2019, 39, BSR20190515. [Google Scholar] [CrossRef]
  16. Sun, T.; Zhang, Y.; Yin, J.; Peng, X.; Zhou, L.; Huang, S.; Wen, Y.; Cao, B.; Chen, L.; Li, X. Association of Gut Microbiota-Dependent Metabolite Trimethylamine N-Oxide with First Ischemic Stroke. J. Atheroscler. Thromb. 2020, 28, 320–328. [Google Scholar] [CrossRef]
  17. Wu, C.; Xue, F.; Lian, Y.; Zhang, J.; Wu, D.; Xie, N.; Chang, W.; Chen, F.; Wang, L.; Wei, W. Relationship between elevated plasma trimethylamine N-oxide levels and increased stroke injury. Neurology 2020, 94, e667–e677. [Google Scholar] [CrossRef]
  18. Guo, F.; Zhou, J.; Li, Z.; Yu, Z.; Ouyang, D. The association between trimethylamine N-oxide and its predecessors choline, L-carnitine, and betaine with coronary artery disease and artery stenosis. Cardiol. Res. Pract. 2020, 2020, 5854919. [Google Scholar] [CrossRef]
  19. Guo, F.; Qiu, X.; Tan, Z.; Li, Z.; Ouyang, D. Plasma trimethylamine n-oxide is associated with renal function in patients with heart failure with preserved ejection fraction. BMC Cardiovasc. Disord. 2020, 20, 394. [Google Scholar] [CrossRef]
  20. Dong, Z.; Liang, Z.; Wang, X.; Liu, W.; Zhao, L.; Wang, S.; Hai, X.; Yu, K. The correlation between plasma trimethylamine N-oxide level and heart failure classification in northern Chinese patients. Ann. Palliat. Med. 2020, 9, 2862–2871. [Google Scholar] [CrossRef]
  21. Dong, Z.; Zheng, S.; Shen, Z.; Luo, Y.; Hai, X. Trimethylamine N-Oxide is Associated with Heart Failure Risk in Patients with Preserved Ejection Fraction. Lab. Med. 2021, 52, 346–351. [Google Scholar] [CrossRef]
  22. Roncal, C.; Martínez-Aguilar, E.; Orbe, J.; Ravassa, S.; Fernandez-Montero, A.; Saenz-Pipaon, G.; Ugarte, A.; de Mendoza, A.E.-H.; Rodriguez, J.A.; Fernández-Alonso, S. Trimethylamine-N-Oxide (TMAO) predicts cardiovascular mortality in peripheral artery disease. Sci. Rep. 2019, 9, 15580. [Google Scholar] [CrossRef]
  23. Waleed, K.B.; Tse, G.; Lu, Y.-K.; Peng, C.-N.; Tu, H.; Ding, L.-G.; Xia, Y.-L.; Wu, S.-L.; Li, X.-T.; Zhou, H.-Q. Trimethylamine N-oxide is associated with coronary atherosclerotic burden in non-ST-segment myocardial infarction patients: SZ-NSTEMI prospective cohort study. Rev. Cardiovasc. Med. 2021, 22, 231–238. [Google Scholar] [CrossRef]
  24. Kong, W.; Ma, J.; Lin, Y.; Chen, W. Positive Association of Plasma Trimethylamine-N-Oxide and Atherosclerosis in Patient with Acute Coronary Syndrome. Cardiovasc. Ther. 2022, 2022, 2484018. [Google Scholar] [CrossRef]
  25. Li, J.; Tan, Y.; Zhou, P.; Liu, C.; Zhao, H.; Song, L.; Zhou, J.; Chen, R.; Wang, Y.; Zhao, X. Association of trimethylamine N-oxide levels and calcification in culprit lesion segments in patients with ST-segment–elevation myocardial infarction evaluated by optical coherence tomography. Front. Cardiovasc. Med. 2021, 8, 628471. [Google Scholar] [CrossRef]
  26. Kang, Y.; Cheng, H.; Shi, Y.; Liu, J.; Wang, Y.; Wan, D. Utility of Trimethylamine Oxide (TMAO) in Predicting Early Neurological Deterioration after Acute Ischemic Stroke. J. Coll. Physicians Surg. Pak. JCPSP 2023, 33, 861–865. [Google Scholar]
  27. Gong, D.; Zhang, L.; Zhang, Y.; Wang, F.; Zhao, Z.; Zhou, X. Gut microbial metabolite trimethylamine N-oxide is related to thrombus formation in atrial fibrillation patients. Am. J. Med. Sci. 2019, 358, 422–428. [Google Scholar] [CrossRef] [PubMed]
  28. Zhou, J.; Wang, D.; Li, B.; Li, X.; Lai, X.; Lei, S.; Li, N.; Zhang, X. Relationship between plasma trimethylamine N-oxide levels and renal dysfunction in patients with hypertension. Kidney Blood Press. Res. 2021, 46, 421–432. [Google Scholar] [CrossRef] [PubMed]
  29. Senthong, V.; Kiatchoosakun, S.; Wongvipaporn, C.; Phetcharaburanin, J.; Tatsanavivat, P.; Sritara, P.; Phrommintikul, A. Gut microbiota-generated metabolite, trimethylamine-N-oxide, and subclinical myocardial damage: A multicenter study from Thailand. Sci. Rep. 2021, 11, 14963. [Google Scholar] [CrossRef] [PubMed]
  30. Mihuta, M.S.; Paul, C.; Borlea, A.; Roi, C.M.; Pescari, D.; Velea-Barta, O.-A.; Mozos, I.; Stoian, D. Connections between serum Trimethylamine N-Oxide (TMAO), a gut-derived metabolite, and vascular biomarkers evaluating arterial stiffness and subclinical atherosclerosis in children with obesity. Front. Endocrinol. 2023, 14, 1253584. [Google Scholar] [CrossRef]
  31. Zhang, J.; Wang, L.; Cai, J.; Lei, A.; Liu, C.; Lin, R.; Jia, L.; Fu, Y. Gut microbial metabolite TMAO portends prognosis in acute ischemic stroke. J. Neuroimmunol. 2021, 354, 577526. [Google Scholar] [CrossRef]
  32. Schneider, C.; Okun, J.G.; Schwarz, K.V.; Hauke, J.; Zorn, M.; Nürnberg, C.; Ungerer, M.; Ringleb, P.A.; Mundiyanapurath, S. Trimethylamine-N-oxide is elevated in the acute phase after ischaemic stroke and decreases within the first days. Eur. J. Neurol. 2020, 27, 1596–1603. [Google Scholar] [CrossRef] [PubMed]
  33. Wen, Y.; Peng, L.; Xu, R.; Zang, N.; Huang, Q.; Zhong, M. Maternal serum trimethylamine-N-oxide is significantly increased in cases with established preeclampsia. Pregnancy Hypertens. 2019, 15, 114–117. [Google Scholar] [CrossRef]
  34. Chen, C.; Qiao, X.; Guo, J.; Yang, T.; Wang, M.; Ma, Y.; Zhao, S.; Ding, L.; Liu, H.; Wang, J. Related factors based on non-targeted metabolomics methods in minor ischaemic stroke. Front. Endocrinol. 2022, 13, 952918. [Google Scholar] [CrossRef] [PubMed]
  35. Ahmad, A.F.; Caparrós-Martin, J.A.; Gray, N.; Lodge, S.; Wist, J.; Lee, S.; O’Gara, F.; Shah, A.; Ward, N.C.; Dwivedi, G. Insights into the associations between the gut microbiome, its metabolites and heart failure. Am. J. Physiol.-Heart Circ. Physiol. 2023, 325, H1325–H1336. [Google Scholar] [CrossRef]
  36. Bordoni, L.; Samulak, J.J.; Sawicka, A.K.; Pelikant-Malecka, I.; Radulska, A.; Lewicki, L.; Kalinowski, L.; Gabbianelli, R.; Olek, R.A. Trimethylamine N-oxide and the reverse cholesterol transport in cardiovascular disease: A cross-sectional study. Sci. Rep. 2020, 10, 18675. [Google Scholar] [CrossRef] [PubMed]
  37. Büttner, P.; Okun, J.G.; Hauke, J.; Holzwirth, E.; Obradovic, D.; Hindricks, G.; Thiele, H.; Kornej, J. Trimethylamine N-oxide in atrial fibrillation progression. IJC Heart Vasc. 2020, 29, 100554. [Google Scholar] [CrossRef]
  38. Chen, Y.-Y.; Ye, Z.-S.; Xia, N.-G.; Xu, Y. TMAO as a novel predictor of major adverse vascular events and recurrence in patients with large artery atherosclerotic ischemic stroke. Clin. Appl. Thromb./Hemost. 2022, 28, 10760296221090503. [Google Scholar] [CrossRef] [PubMed]
  39. Huang, Y.; Zhu, Z.; Huang, Z.; Zhou, J. Elevated serum trimethylamine oxide levels as potential biomarker for diabetic kidney disease. Endocr. Connect. 2023, 12, e220542. [Google Scholar] [CrossRef]
  40. Israr, M.Z.; Bernieh, D.; Salzano, A.; Cassambai, S.; Yazaki, Y.; Heaney, L.M.; Jones, D.J.; Ng, L.L.; Suzuki, T. Association of gut-related metabolites with outcome in acute heart failure. Am. Heart J. 2021, 234, 71–80. [Google Scholar] [CrossRef]
  41. Suzuki, T.; Heaney, L.M.; Jones, D.J.L.; Ng, L.L. Trimethylamine N-oxide and Risk Stratification after Acute Myocardial Infarction. Clin. Chem. 2017, 63, 420–428. [Google Scholar] [CrossRef]
  42. Eyileten, C.; Jarosz-Popek, J.; Jakubik, D.; Gasecka, A.; Wolska, M.; Ufnal, M.; Postula, M.; Toma, A.; Lang, I.M.; Siller-Matula, J.M. Plasma trimethylamine-N-oxide is an independent predictor of long-term cardiovascular mortality in patients undergoing percutaneous coronary intervention for acute coronary syndrome. Front. Cardiovasc. Med. 2021, 8, 728724. [Google Scholar] [CrossRef]
  43. Senthong, V.; Wang, Z.; Fan, Y.; Wu, Y.; Hazen, S.L.; Tang, W.H. Trimethylamine N-Oxide and Mortality Risk in Patients With Peripheral Artery Disease. J. Am. Heart Assoc. 2016, 5, e004237. [Google Scholar] [CrossRef] [PubMed]
  44. Senthong, V.; Wang, Z.; Li, X.S.; Fan, Y.; Wu, Y.; Tang, W.H.; Hazen, S.L. 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, 5, e002816. [Google Scholar] [CrossRef] [PubMed]
  45. Tan, Y.; Zhou, J.; Yang, S.; Li, J.; Zhao, H.; Song, L.; Yan, H. Addition of plasma myeloperoxidase and trimethylamine N-Oxide to the GRACE score improves prediction of near-term major adverse cardiovascular events in patients with ST-segment elevation myocardial infarction. Front. Pharmacol. 2021, 12, 632075. [Google Scholar] [CrossRef] [PubMed]
  46. Salzano, A.; Israr, M.Z.; Yazaki, Y.; Heaney, L.M.; Kanagala, P.; Singh, A.; Arnold, J.R.; Gulsin, G.S.; Squire, I.B.; McCann, G.P. Combined use of trimethylamine N-oxide with BNP for risk stratification in heart failure with preserved ejection fraction: Findings from the DIAMONDHFpEF study. Eur. J. Prev. Cardiol. 2020, 27, 2159–2162. [Google Scholar] [CrossRef] [PubMed]
  47. Tan, C.; Wang, H.; Gao, X.; Xu, R.; Zeng, X.; Cui, Z.; Zhu, J.; Wu, Q.; Xia, G.; Zhou, H. Dynamic changes and prognostic value of gut microbiota-Dependent trimethylamine-N-Oxide in acute ischemic stroke. Front. Neurol. 2020, 11, 29. [Google Scholar] [CrossRef] [PubMed]
  48. Manjunath, G.; Tighiouart, H.; Coresh, J.; Macleod, B.; Salem, D.N.; Griffith, J.L.; Levey, A.S.; Sarnak, M.J. Level of kidney function as a risk factor for cardiovascular outcomes in the elderly. Kidney Int. 2003, 63, 1121–1129. [Google Scholar] [CrossRef]
  49. Meisinger, C.; Döring, A.; Löwel, H. Chronic kidney disease and risk of incident myocardial infarction and all-cause and cardiovascular disease mortality in middle-aged men and women from the general population. Eur. Heart J. 2006, 27, 1245–1250. [Google Scholar] [CrossRef]
  50. Winther, S.A.; Øllgaard, J.C.; Tofte, N.; Tarnow, L.; Wang, Z.; Ahluwalia, T.S.; Jorsal, A.; Theilade, S.; Parving, H.-H.; Hansen, T.W. Utility of plasma concentration of trimethylamine N-oxide in predicting cardiovascular and renal complications in individuals with type 1 diabetes. Diabetes Care 2019, 42, 1512–1520. [Google Scholar] [CrossRef]
  51. Skagen, K.; Trøseid, M.; Ueland, T.; Holm, S.; Abbas, A.; Gregersen, I.; Kummen, M.; Bjerkeli, V.; Reier-Nilsen, F.; Russell, D. The Carnitine-butyrobetaine-trimethylamine-N-oxide pathway and its association with cardiovascular mortality in patients with carotid atherosclerosis. Atherosclerosis 2016, 247, 64–69. [Google Scholar] [CrossRef]
  52. Gruppen, E.G.; Garcia, E.; Connelly, M.A.; Jeyarajah, E.J.; Otvos, J.D.; Bakker, S.J.; Dullaart, R.P. TMAO is associated with mortality: Impact of modestly impaired renal function. Sci. Rep. 2017, 7, 13781. [Google Scholar] [CrossRef] [PubMed]
  53. Wang, M.; Wang, Z.; Lee, Y.; Lai, H.T.; de Oliveira Otto, M.C.; Lemaitre, R.N.; Fretts, A.; Sotoodehnia, N.; Budoff, M.; DiDonato, J.A. Dietary meat, trimethylamine N-oxide-related metabolites, and incident cardiovascular disease among older adults: The cardiovascular health study. Arterioscler. Thromb. Vasc. Biol. 2022, 42, e273–e288. [Google Scholar] [CrossRef] [PubMed]
  54. Sanchez-Gimenez, R.; Peiro, O.M.; Bonet, G.; Carrasquer, A.; Fragkiadakis, G.A.; Bulló, M.; Papandreou, C.; Bardaji, A. Plasma trimethylamine-N-oxide, its precursors and risk of cardiovascular events in patients with acute coronary syndrome: Mediating effects of renal function. Front. Cardiovasc. Med. 2022, 9, 1000815. [Google Scholar] [CrossRef] [PubMed]
  55. Konieczny, R.; Żurawska-Płaksej, E.; Kaaz, K.; Czapor-Irzabek, H.; Bombała, W.; Mysiak, A.; Kuliczkowski, W. All-Cause Mortality and Trimethylamine N-Oxide Levels in Patients with Cardiovascular Disease. Cardiology 2022, 147, 443–452. [Google Scholar] [CrossRef]
  56. Yu, N.; Gu, N.; Wang, Y.; Zhou, B.; Lu, D.; Li, J.; Ma, X.; Zhang, J.; Guo, X. The association of plasma trimethylamine N-oxide with coronary atherosclerotic burden in patients with type 2 diabetes among a Chinese north population. Diabetes Metab. Syndr. Obes. 2022, 15, 69–78. [Google Scholar] [CrossRef]
  57. Suzuki, T.; Yazaki, Y.; Voors, A.A.; Jones, D.J.; Chan, D.C.; Anker, S.D.; Cleland, J.G.; Dickstein, K.; Filippatos, G.; Hillege, H.L. 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. [Google Scholar] [CrossRef]
  58. Kim, R.B.; Morse, B.L.; Djurdjev, O.; Tang, M.; Muirhead, N.; Barrett, B.; Holmes, D.T.; Madore, F.; Clase, C.M.; Rigatto, C.; et al. Advanced chronic kidney disease populations have elevated trimethylamine N-oxide levels associated with increased cardiovascular events. Kidney Int. 2016, 89, 1144–1152. [Google Scholar] [CrossRef]
  59. Missailidis, C.; Hällqvist, J.; Qureshi, A.R.; Barany, P.; Heimbürger, O.; Lindholm, B.; Stenvinkel, P.; Bergman, P. Serum trimethylamine-N-oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS ONE 2016, 11, e0141738. [Google Scholar] [CrossRef]
  60. Stubbs, J.R.; Stedman, M.R.; Liu, S.; Long, J.; Franchetti, Y.; West, R.E.; Prokopienko, A.J.; Mahnken, J.D.; Chertow, G.M.; Nolin, T.D. Trimethylamine N-Oxide and Cardiovascular Outcomes in Patients with ESKD Receiving Maintenance Hemodialysis. Clin. J. Am. Soc. Nephrol. 2019, 14, 261–267. [Google Scholar] [CrossRef]
  61. Tang, W.H.; Wang, Z.; Kennedy, D.J.; Wu, Y.; Buffa, J.A.; Agatisa-Boyle, B.; Li, X.S.; Levison, B.S.; Hazen, S.L. 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. [Google Scholar] [CrossRef]
  62. Zhang, P.; Zou, J.-Z.; Chen, J.; Tan, X.; Xiang, F.-F.; Shen, B.; Hu, J.-C.; Wang, J.-L.; Wang, Y.-Q.; Yu, J.-B. Association of trimethylamine N-Oxide with cardiovascular and all-cause mortality in hemodialysis patients. Ren. Fail. 2020, 42, 1004–1014. [Google Scholar] [CrossRef] [PubMed]
  63. Yuan, X.; Wei, X.; Liu, J.; Tang, C.; Peng, L.; Wang, H.; Huang, Y. Association of Circulating Trimethylamine-N Oxide With Malnutrition and the Risk of Coronary Artery Disease in Patients With Maintenance Hemodialysis. J. Ren. Nutr. 2023, 33, 465–471. [Google Scholar] [CrossRef] [PubMed]
  64. Schrauben, S.J.; Sapa, H.; Xie, D.; Zhang, X.; Anderson, A.H.; Shlipak, M.G.; Hsu, C.-y.; Shafi, T.; Mehta, R.; Bhat, Z. Association of urine and plasma ADMA with atherosclerotic risk in DKD cardiovascular disease risk in diabetic kidney disease: Findings from the Chronic Renal Insufficiency Cohort (CRIC) study. Nephrol. Dial. Transplant. 2023, 38, 2809–2815. [Google Scholar] [CrossRef]
  65. Fu, D.; Shen, J.; Li, W.; Wang, Y.; Zhong, Z.; Ye, H.; Huang, N.; Fan, L.; Yang, X.; Yu, X. Elevated Serum Trimethylamine N-Oxide Levels Are Associated with Mortality in Male Patients on Peritoneal Dialysis. Blood Purif. 2021, 50, 837–847. [Google Scholar] [CrossRef] [PubMed]
  66. Nie, J.; Xie, L.; Zhao, B.; Li, Y.; Qiu, B.; Zhu, F.; Li, G.; He, M.; Wang, Y.; Wang, B. Serum trimethylamine N-oxide concentration is positively associated with first stroke in hypertensive patients. Stroke 2018, 49, 2021–2028. [Google Scholar] [CrossRef] [PubMed]
  67. Lever, M.; George, P.M.; Slow, S.; Bellamy, D.; Young, J.M.; Ho, M.; McEntyre, C.J.; Elmslie, J.L.; Atkinson, W.; Molyneux, S.L.; et al. Betaine and Trimethylamine-N-Oxide as Predictors of Cardiovascular Outcomes Show Different Patterns in Diabetes Mellitus: An Observational Study. PLoS ONE 2014, 9, e114969. [Google Scholar] [CrossRef]
  68. Cardona, A.; O’Brien, A.; Bernier, M.C.; Somogyi, A.; Wysocki, V.H.; Smart, S.; He, X.; Ambrosio, G.; Hsueh, W.A.; Raman, S.V. Trimethylamine N-oxide and incident atherosclerotic events in high-risk individuals with diabetes: An ACCORD trial post hoc analysis. BMJ Open Diabetes Res. Care 2019, 7, e000718. [Google Scholar] [CrossRef]
  69. Tang, W.H.; Wang, Z.; Li, X.S.; Fan, Y.; Li, D.S.; Wu, Y.; Hazen, S.L. Increased Trimethylamine N-Oxide Portends High Mortality Risk Independent of Glycemic Control in Patients with Type 2 Diabetes Mellitus. Clin. Chem. 2017, 63, 297–306. [Google Scholar] [CrossRef]
  70. Winther, S.A.; Øllgaard, J.C.; Hansen, T.W.; Von Scholten, B.J.; Reinhard, H.; Ahluwalia, T.S.; Wang, Z.; Gæde, P.; Parving, H.-H.; Hazen, S. Plasma trimethylamine N-oxide and its metabolic precursors and risk of mortality, cardiovascular and renal disease in individuals with type 2-diabetes and albuminuria. PLoS ONE 2021, 16, e0244402. [Google Scholar] [CrossRef]
  71. Croyal, M.; Saulnier, P.-J.; Aguesse, A.; Gand, E.; Ragot, S.; Roussel, R.; Halimi, J.-M.; Ducrocq, G.; Cariou, B.; Montaigne, D. Plasma trimethylamine N-oxide and risk of cardiovascular events in patients with type 2 diabetes. J. Clin. Endocrinol. Metab. 2020, 105, 2371–2380. [Google Scholar] [CrossRef]
  72. Li, X.S.; Obeid, S.; Klingenberg, R.; Gencer, B.; Mach, F.; Raber, L.; Windecker, S.; Rodondi, N.; Nanchen, D.; Muller, O.; 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. [Google Scholar] [CrossRef] [PubMed]
  73. Witkowski, M.; Witkowski, M.; Friebel, J.; Buffa, J.A.; Li, X.S.; Wang, Z.; Sangwan, N.; Li, L.; DiDonato, J.A.; Tizian, C. Vascular endothelial Tissue Factor contributes to trimethylamine N-oxide-enhanced arterial thrombosis. Cardiovasc. Res. 2022, 118, 2367–2384. [Google Scholar] [CrossRef] [PubMed]
  74. Li, N.; Zhou, J.; Wang, Y.; Chen, R.; Li, J.; Zhao, X.; Zhou, P.; Liu, C.; Song, L.; Liao, Z. Association between trimethylamine N-oxide and prognosis of patients with acute myocardial infarction and heart failure. ESC Heart Fail. 2022, 9, 3846–3857. [Google Scholar] [CrossRef] [PubMed]
  75. Gencer, B.; Li, X.S.; Gurmu, Y.; Bonaca, M.P.; Morrow, D.A.; Cohen, M.; Bhatt, D.L.; Steg, P.G.; Storey, R.F.; Johanson, P. 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. [Google Scholar] [CrossRef]
  76. Li, N.; Wang, Y.; Zhou, J.; Chen, R.; Li, J.; Zhao, X.; Zhou, P.; Liu, C.; Chen, Y.; Song, L. Association between the Changes in Trimethylamine N-Oxide-Related Metabolites and Prognosis of Patients with Acute Myocardial Infarction: A Prospective Study. J. Cardiovasc. Dev. Dis. 2022, 9, 380. [Google Scholar] [CrossRef]
  77. Zhao, S.; Tian, Y.; Wang, S.; Yang, F.; Xu, J.; Qin, Z.; Liu, X.; Cao, M.; Zhao, P.; Zhang, G. Prognostic value of gut microbiota-derived metabolites in patients with ST-segment elevation myocardial infarction. Am. J. Clin. Nutr. 2023, 117, 499–508. [Google Scholar] [CrossRef]
  78. Aldujeli, A.; Patel, R.; Grabauskyte, I.; Hamadeh, A.; Lieponyte, A.; Tatarunas, V.; Khalifeh, H.; Briedis, K.; Skipskis, V.; Aldujeili, M. The Impact of Trimethylamine N-Oxide and Coronary Microcirculatory Dysfunction on Outcomes following ST-Elevation Myocardial Infarction. J. Cardiovasc. Dev. Dis. 2023, 10, 197. [Google Scholar] [CrossRef]
  79. Zhou, J.; Yu, S.; Tan, Y.; Zhou, P.; Liu, C.; Li, J.; Chen, R.; Zhao, S.; Yan, H. Trimethylamine N-oxide was not associated with 30-day left ventricular systolic dysfunction in patients with a first anterior ST-segment elevation myocardial infarction after primary revascularization: A sub-analysis from an optical coherence tomography registry. Front. Cardiovasc. Med. 2020, 7, 353. [Google Scholar]
  80. Haghikia, A.; Li, X.S.; Liman, T.G.; Bledau, N.; Schmidt, D.; Zimmermann, F.; Kränkel, N.; Widera, C.; Sonnenschein, K.; Haghikia, A. 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. [Google Scholar] [CrossRef]
  81. Zhai, Q.; Wang, X.; Chen, C.; Tang, Y.; Wang, Y.; Tian, J.; Zhao, Y.; Liu, X. Prognostic value of plasma trimethylamine N-oxide levels in patients with acute ischemic stroke. Cell. Mol. Neurobiol. 2019, 39, 1201–1206. [Google Scholar] [CrossRef]
  82. Nam, H.S.; Ha, J.; Ji, D.; Kwon, I.; Lee, H.S.; Han, M.; Kim, Y.D.; Heo, J.H.; Lee, S.-G. Elevation of the gut microbiota metabolite trimethylamine N-oxide predicts stroke outcome. J. Stroke 2019, 21, 350. [Google Scholar] [CrossRef] [PubMed]
  83. Zhu, C.; Li, G.; Lv, Z.; Li, J.; Wang, X.; Kang, J.; Zhan, C. Association of plasma trimethylamine-N-oxide levels with post-stroke cognitive impairment: A 1-year longitudinal study. Neurol. Sci. 2020, 41, 57–63. [Google Scholar] [CrossRef] [PubMed]
  84. Xu, J.; Cheng, A.; Song, B.; Zhao, M.; Xue, J.; Wang, A.; Dai, L.; Jing, J.; Meng, X.; Li, H. Trimethylamine N-oxide and stroke recurrence depends on ischemic stroke subtypes. Stroke 2022, 53, 1207–1215. [Google Scholar] [CrossRef] [PubMed]
  85. Xue, J.; Xu, J.; Zhao, M.; Jin, A.; Cheng, A.; Jiang, X.; Li, K.; Lin, J.; Meng, X.; Li, H. Residual Risk of Trimethylamine-N-Oxide and Choline for Stroke Recurrence in Patients With Intensive Secondary Therapy. J. Am. Heart Assoc. 2022, 11, e027265. [Google Scholar] [CrossRef] [PubMed]
  86. Chou, P.-S.; Yang, I.-H.; Kuo, C.-M.; Wu, M.-N.; Lin, T.-C.; Fong, Y.-O.; Juan, C.-H.; Lai, C.-L. The Prognostic Biomarkers of Plasma Trimethylamine N-Oxide and Short-Chain Fatty Acids for Recanalization Therapy in Acute Ischemic Stroke. Int. J. Mol. Sci. 2023, 24, 10796. [Google Scholar] [CrossRef]
  87. Bjørnestad, E.Ø.; Olset, H.; Dhar, I.; Løland, K.; Pedersen, E.K.; Svingen, G.F.; Svardal, A.; Berge, R.K.; Ueland, P.M.; Tell, G.S. Circulating trimethyllysine and risk of acute myocardial infarction in patients with suspected stable coronary heart disease. J. Intern. Med. 2020, 288, 446–456. [Google Scholar] [CrossRef]
  88. Amrein, M.; Li, X.S.; Walter, J.; Wang, Z.; Zimmermann, T.; Strebel, I.; Honegger, U.; Leu, K.; Schäfer, I.; Twerenbold, R. Gut microbiota-dependent metabolite trimethylamine N-oxide (TMAO) and cardiovascular risk in patients with suspected functionally relevant coronary artery disease (fCAD). Clin. Res. Cardiol. 2022, 111, 692–704. [Google Scholar] [CrossRef]
  89. Bjørnestad, E.Ø.; Dhar, I.; Svingen, G.F.; Pedersen, E.R.; Ørn, S.; Svenningsson, M.M.; Tell, G.S.; Ueland, P.M.; Sulo, G.; Laaksonen, R. Circulating trimethylamine N-oxide levels do not predict 10-year survival in patients with or without coronary heart disease. J. Intern. Med. 2022, 292, 915–924. [Google Scholar] [CrossRef]
  90. Chou, R.-H.; Chen, C.-Y.; Chen, I.C.; Huang, H.-L.; Lu, Y.-W.; Kuo, C.-S.; Chang, C.-C.; Huang, P.-H.; Chen, J.-W.; Lin, S.-J. Trimethylamine N-Oxide, Circulating Endothelial Progenitor Cells, and Endothelial Function in Patients with Stable Angina. Sci. Rep. 2019, 9, 4249. [Google Scholar] [CrossRef]
  91. Berger, M.; Kleber, M.E.; Delgado, G.E.; März, W.; Andreas, M.; Hellstern, P.; Marx, N.; Schuett, K.A. Trimethylamine N-Oxide and Adenosine Diphosphate–Induced Platelet Reactivity Are Independent Risk Factors for Cardiovascular and All-Cause Mortality. Circ. Res. 2020, 126, 660–662. [Google Scholar] [CrossRef]
  92. Tang, W.H.; Wang, Z.; Shrestha, K.; Borowski, A.G.; Wu, Y.; Troughton, R.W.; Klein, A.L.; Hazen, S.L. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J. Card. Fail. 2015, 21, 91–96. [Google Scholar] [CrossRef] [PubMed]
  93. Zhou, X.; Jin, M.; Liu, L.; Yu, Z.; Lu, X.; Zhang, H. Trimethylamine N-oxide and cardiovascular outcomes in patients with chronic heart failure after myocardial infarction. ESC Heart Fail. 2020, 7, 189–194. [Google Scholar] [CrossRef] [PubMed]
  94. Kinugasa, Y.; Nakamura, K.; Kamitani, H.; Hirai, M.; Yanagihara, K.; Kato, M.; Yamamoto, K. Trimethylamine N-oxide and outcomes in patients hospitalized with acute heart failure and preserved ejection fraction. ESC Heart Fail. 2021, 8, 2103–2110. [Google Scholar] [CrossRef]
  95. Guo, Y.; Xu, S.; Zhan, H.; Chen, H.; Hu, P.; Zhou, D.; Dai, H.; Liu, X.; Hu, W.; Zhu, G. Trimethylamine N-Oxide Levels Are Associated with Severe Aortic Stenosis and Predict Long-Term Adverse Outcome. J. Clin. Med. 2023, 12, 407. [Google Scholar] [CrossRef]
  96. Luciani, M.; Müller, D.; Vanetta, C.; Diteepeng, T.; von Eckardstein, A.; Aeschbacher, S.; Rodondi, N.; Moschovitis, G.; Reichlin, T.; Sinnecker, T. Trimethylamine-N-oxide is associated with cardiovascular mortality and vascular brain lesions in patients with atrial fibrillation. Heart 2023, 109, 396–404. [Google Scholar] [CrossRef] [PubMed]
  97. Zheng, L.; Zheng, J.; Xie, Y.; Li, Z.; Guo, X.; Sun, G.; Sun, Z.; Xing, F.; Sun, Y. Serum gut microbe-dependent trimethylamine N-oxide improves the prediction of future cardiovascular disease in a community-based general population. Atherosclerosis 2019, 280, 126–131. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, D.; Gu, S.; Zhou, Z.; Ma, Z.; Zuo, H. Associations of plasma TMAO and its precursors with stroke risk in the general population: A nested case-control study. J. Intern. Med. 2023, 293, 110–120. [Google Scholar] [CrossRef]
  99. Lemaitre, R.N.; Jensen, P.N.; Wang, Z.; Fretts, A.M.; Sitlani, C.M.; Nemet, I.; Sotoodehnia, N.; de Oliveira Otto, M.C.; Zhu, W.; Budoff, M. Plasma Trimethylamine-N-Oxide and Incident Ischemic Stroke: The Cardiovascular Health Study and the Multi-Ethnic Study of Atherosclerosis. J. Am. Heart Assoc. 2023, 12, e8711. [Google Scholar] [CrossRef]
  100. Tang, W.W.; Li, X.S.; Wu, Y.; Wang, Z.; Khaw, K.-T.; Wareham, N.J.; Nieuwdorp, M.; Boekholdt, S.M.; Hazen, S.L. Plasma trimethylamine N-oxide (TMAO) levels predict future risk of coronary artery disease in apparently healthy individuals in the EPIC-Norfolk prospective population study. Am. Heart J. 2021, 236, 80–86. [Google Scholar] [CrossRef]
  101. Svingen, G.F.; Zuo, H.; Ueland, P.M.; Seifert, R.; Løland, K.H.; Pedersen, E.R.; Schuster, P.M.; Karlsson, T.; Tell, G.S.; Schartum-Hansen, H. Increased plasma trimethylamine-N-oxide is associated with incident atrial fibrillation. Int. J. Cardiol. 2018, 267, 100–106. [Google Scholar] [CrossRef]
  102. Papandreou, C.; Bulló, M.; Hernández-Alonso, P.; Ruiz-Canela, M.; Li, J.; Guasch-Ferré, M.; Toledo, E.; Clish, C.; Corella, D.; Estruch, R. Choline metabolism and risk of atrial fibrillation and heart failure in the PREDIMED study. Clin. Chem. 2021, 67, 288–297. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, G.; Li, J.; Li, Y.; Hu, Y.; Franke, A.A.; Liang, L.; Hu, F.B.; Chan, A.T.; Mukamal, K.J.; Rimm, E.B. Gut microbiota–derived metabolites and risk of coronary artery disease: A prospective study among US men and women. Am. J. Clin. Nutr. 2021, 114, 238–247. [Google Scholar] [CrossRef] [PubMed]
  104. Bhuiya, J.; Notsu, Y.; Kobayashi, H.; Shibly, A.Z.; Sheikh, A.M.; Okazaki, R.; Yamaguchi, K.; Nagai, A.; Nabika, T.; Abe, T. Neither Trimethylamine-N-Oxide nor Trimethyllysine Is Associated with Atherosclerosis: A Cross-Sectional Study in Older Japanese Adults. Nutrients 2023, 15, 759. [Google Scholar] [CrossRef] [PubMed]
  105. Lee, Y.; Nemet, I.; Wang, Z.; Lai, H.T.M.; Otto, M.C.d.O.; Lemaitre, R.N.; Fretts, A.M.; Sotoodehnia, N.; Budoff, M.; Didonato, J.A.; et al. Longitudinal Measures of Trimethylamine N-oxide and Incident Atherosclerotic Cardiovascular Disease Events in Older Adults: The Cardiovascular Health Study. Curr. Dev. Nutr. 2020, 4 (Suppl. S2), 1434. [Google Scholar] [CrossRef]
  106. Fretts, A.M.; Hazen, S.L.; Jensen, P.; Budoff, M.; Sitlani, C.M.; Wang, M.; de Oliveira Otto, M.C.; DiDonato, J.A.; Lee, Y.; Psaty, B.M. Association of Trimethylamine N-oxide and metabolites with mortality in older adults. JAMA Netw. Open 2022, 5, e2213242. [Google Scholar] [CrossRef]
  107. Heianza, Y.; Ma, W.; DiDonato, J.A.; Sun, Q.; Rimm, E.B.; Hu, F.B.; Rexrode, K.M.; Manson, J.E.; 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. [Google Scholar] [CrossRef]
  108. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; 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. [Google Scholar] [CrossRef]
  109. Organ, C.L.; Otsuka, H.; Bhushan, S.; Wang, Z.; Bradley, J.; Trivedi, R.; Polhemus, D.J.; Tang, W.H.; Wu, Y.; Hazen, S.L.; et al. Choline Diet and Its Gut Microbe-Derived Metabolite, Trimethylamine N-Oxide, Exacerbate Pressure Overload-Induced Heart Failure. Circ. Heart Fail. 2016, 9, e002314. [Google Scholar] [CrossRef]
  110. Gregory, J.C.; Buffa, J.A.; Org, E.; Wang, Z.; Levison, B.S.; Zhu, W.; Wagner, M.A.; Bennett, B.J.; Li, L.; DiDonato, J.A. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J. Biol. Chem. 2015, 290, 5647–5660. [Google Scholar] [CrossRef]
  111. Annunziata, G.; Maisto, M.; Schisano, C.; Ciampaglia, R.; Narciso, V.; Hassan, S.T.; Tenore, G.C.; Novellino, E. Effect of grape pomace polyphenols with or without pectin on TMAO serum levels assessed by LC/MS-based assay: A preliminary clinical study on overweight/obese subjects. Front. Pharmacol. 2019, 10, 575. [Google Scholar] [CrossRef]
  112. Park, J.; Miller, M.; Rhyne, J.; Wang, Z.; Hazen, S. Differential effect of short-term popular diets on TMAO and other cardio-metabolic risk markers. Nutr. Metab. Cardiovasc. Dis. 2019, 29, 513–517. [Google Scholar] [CrossRef]
  113. Montrucchio, C.; De Nicolò, A.; D’Ettorre, G.; D’Ascenzo, F.; Lazzaro, A.; Tettoni, M.; D’Avolio, A.; Bonora, S.; Celani, L.; Di Perri, G. Serum trimethylamine-N-oxide concentrations in people living with HIV and the effect of probiotic supplementation. Int. J. Antimicrob. Agents 2020, 55, 105908. [Google Scholar] [CrossRef] [PubMed]
  114. Tripolt, N.J.; Leber, B.; Triebl, A.; Köfeler, H.; Stadlbauer, V.; Sourij, H. Effect of Lactobacillus casei Shirota supplementation on trimethylamine-N-oxide levels in patients with metabolic syndrome: An open-label, randomized study. Atherosclerosis 2015, 242, 141–144. [Google Scholar] [CrossRef] [PubMed]
  115. Boutagy, N.E.; Neilson, A.P.; Osterberg, K.L.; Smithson, A.T.; Englund, T.R.; Davy, B.M.; Hulver, M.W.; Davy, K.P. Probiotic supplementation and trimethylamine-N-oxide production following a high-fat diet. Obesity 2015, 23, 2357–2363. [Google Scholar] [CrossRef] [PubMed]
  116. Borges, N.A.; Stenvinkel, P.; Bergman, P.; Qureshi, A.; Lindholm, B.; Moraes, C.; Stockler-Pinto, M.; Mafra, D. Effects of probiotic supplementation on trimethylamine-N-oxide plasma levels in hemodialysis patients: A pilot study. Probiotics Antimicrob. Proteins 2019, 11, 648–654. [Google Scholar] [CrossRef]
  117. Kimber, C.; Zhang, S.; Johnson, C.; West III, R.E.; Prokopienko, A.J.; Mahnken, J.D.; Yu, A.S.; Hoofnagle, A.N.; Ir, D.; Robertson, C.E. Randomized, placebo-controlled trial of rifaximin therapy for lowering gut-derived cardiovascular toxins and inflammation in CKD. Kidney360 2020, 1, 1206. [Google Scholar] [CrossRef]
  118. Vallance, H.; Koochin, A.; Branov, J.; Rosen-Heath, A.; Bosdet, T.; Wang, Z.; Hazen, S.; Horvath, G. Marked elevation in plasma trimethylamine-N-oxide (TMAO) in patients with mitochondrial disorders treated with oral l-carnitine. Mol. Genet. Metab. Rep. 2018, 15, 130–133. [Google Scholar] [CrossRef]
  119. Miller, M.J.; Bostwick, B.L.; Kennedy, A.D.; Donti, T.R.; Sun, Q.; Sutton, V.R.; Elsea, S.H. Chronic oral L-carnitine supplementation drives marked plasma TMAO elevations in patients with organic acidemias despite dietary meat restrictions. JIMD Rep. 2016, 30, 39–44. [Google Scholar]
  120. Onyszkiewicz, M.; Jaworska, K.; Ufnal, M. Short chain fatty acids and methylamines produced by gut microbiota as mediators and markers in the circulatory system. Exp. Biol. Med. 2020, 245, 166–175. [Google Scholar] [CrossRef]
  121. Yuzefpolskaya, M.; Bohn, B.; Javaid, A.; Mondellini, G.M.; Braghieri, L.; Pinsino, A.; Onat, D.; Cagliostro, B.; Kim, A.; Takeda, K. Levels of Trimethylamine N-Oxide Remain Elevated Long Term After Left Ventricular Assist Device and Heart Transplantation and Are Independent From Measures of Inflammation and Gut Dysbiosis. Circ. Heart Fail. 2021, 14, e007909. [Google Scholar] [CrossRef]
  122. Yang, Y.; Yang, B.; Li, X.; Xue, L.; Liu, B.; Liang, Y.; Zhao, Z.; Luo, Q.; Liu, Z.; Zeng, Q. Higher circulating Trimethylamine N-oxide levels are associated with worse severity and prognosis in pulmonary hypertension: A cohort study. Respir. Res. 2022, 23, 344. [Google Scholar] [CrossRef] [PubMed]
  123. Kummen, M.; Solberg, O.G.; Storm-Larsen, C.; Holm, K.; Ragnarsson, A.; Trøseid, M.; Vestad, B.; Skårdal, R.; Yndestad, A.; Ueland, T. Rosuvastatin alters the genetic composition of the human gut microbiome. Sci. Rep. 2020, 10, 5397. [Google Scholar] [CrossRef] [PubMed]
  124. Milks, M.; He, X.; Sharkey-Toppen, T.; Smart, S.; McCarthy, B.; Somogyi, A.; Belury, M.; Raman, S. Statin Use is Associated with Lower Trimethylamine-N-oxide (TMAO) Level in Adults at Risk of Atherosclerotic Cardiovascular Disease, Independent of Serum Cholesterol and Renal Function. J. Clin. Lipidol. 2018, 12, 567–568. [Google Scholar] [CrossRef]
  125. Liang, H.; Yu, A.; Wang, Z.; Zhang, N.; Wang, Q.; Gao, H.; Gao, J.; Wang, X.; Wang, H. Atherosclerotic patients with diabetes mellitus may break through the threshold of healthy TMAO levels formed by long-term statins therapy. Heliyon 2023, 9, e13657. [Google Scholar] [CrossRef]
  126. Xu, M.; Zheng, J.; Hou, T.; Lin, H.; Wang, T.; Wang, S.; Lu, J.; Zhao, Z.; Li, M.; Xu, Y.J.D.C. SGLT2 inhibition, choline metabolites, and cardiometabolic diseases: A mediation Mendelian randomization study. Diabetes Care 2022, 45, 2718–2728. [Google Scholar] [CrossRef] [PubMed]
  127. Aziz, F.; Tripolt, N.J.; Pferschy, P.N.; Kolesnik, E.; Mangge, H.; Curcic, P.; Hermann, M.; Meinitzer, A.; von Lewinski, D.; Sourij, H. Alterations in trimethylamine-N-oxide in response to Empagliflozin therapy: A secondary analysis of the EMMY trial. Cardiovasc. Diabetol. 2023, 22, 184. [Google Scholar] [CrossRef]
  128. Ma, R.; Fu, W.; Zhang, J.; Hu, X.; Yang, J.; Jiang, H. TMAO: A potential mediator of clopidogrel resistance. Sci. Rep. 2021, 11, 6580. [Google Scholar] [CrossRef]
  129. Jaworska, K.; Hering, D.; Mosieniak, G.; Bielak-Zmijewska, A.; Pilz, M.; Konwerski, M.; Gasecka, A.; Kaplon-Cieslicka, A.; Filipiak, K.; Sikora, E.; et al. TMA, A Forgotten Uremic Toxin, but Not TMAO, Is Involved in Cardiovascular Pathology. Toxins 2019, 11, 490. [Google Scholar] [CrossRef] [PubMed]
  130. Gong, J.; Noel, S.; Pluznick, J.L.; Hamad, A.R.A.; Rabb, H. Gut microbiota-kidney cross-talk in acute kidney injury. Semin. Nephrol. 2019, 39, 107–116. [Google Scholar] [CrossRef]
  131. Xie, G.; Yan, A.; Lin, P.; Wang, Y.; Guo, L. Trimethylamine N-oxide—A marker for atherosclerotic vascular disease. Rev. Cardiovasc. Med. 2021, 22, 787–797. [Google Scholar] [CrossRef]
  132. Li, X.; Geng, J.; Zhao, J.; Ni, Q.; Zhao, C.; Zheng, Y.; Chen, X.; Wang, L. Trimethylamine N-oxide exacerbates cardiac fibrosis via activating the NLRP3 inflammasome. Front. Physiol. 2019, 10, 866. [Google Scholar] [CrossRef] [PubMed]
  133. Li, Z.; Wu, Z.; Yan, J.; Liu, H.; Liu, Q.; Deng, Y.; Ou, C.; Chen, M. Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Lab. Investig. 2019, 99, 346–357. [Google Scholar] [CrossRef] [PubMed]
  134. Wang, Z.; Roberts, A.B.; Buffa, J.A.; Levison, B.S.; Zhu, W.; Org, E.; Gu, X.; Huang, Y.; Zamanian-Daryoush, M.; Culley, M.K. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef]
  135. Gao, X.; Liu, X.; Xu, J.; Xue, C.; Xue, Y.; Wang, Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J. Biosci. Bioeng. 2014, 118, 476–481. [Google Scholar] [CrossRef]
  136. Tan, X.; Liu, Y.; Long, J.; Chen, S.; Liao, G.; Wu, S.; Li, C.; Wang, L.; Ling, W.; Zhu, H. Trimethylamine N-oxide aggravates liver steatosis through modulation of bile acid metabolism and inhibition of farnesoid X receptor signaling in nonalcoholic fatty liver disease. Mol. Nutr. Food Res. 2019, 63, 1900257. [Google Scholar] [CrossRef] [PubMed]
  137. Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef]
  138. Skye, S.M.; Zhu, W.; Romano, K.A.; Guo, C.-J.; Wang, Z.; Jia, X.; Kirsop, J.; Haag, B.; Lang, J.M.; DiDonato, J.A. Microbial transplantation with human gut commensals containing CutC is sufficient to transmit enhanced platelet reactivity and thrombosis potential. Circ. Res. 2018, 123, 1164–1176. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, M.-L.; Zhu, X.-H.; Ran, L.; Lang, H.-D.; Yi, L.; Mi, M.-T. Trimethylamine-N-Oxide induces vascular inflammation by activating the NLRP3 inflammasome through the SIRT3-SOD2-mtROS signaling pathway. J. Am. Heart Assoc. 2017, 6, e006347. [Google Scholar] [CrossRef]
  140. Makrecka-Kuka, M.; Volska, K.; Antone, U.; Vilskersts, R.; Grinberga, S.; Bandere, D.; Liepinsh, E.; Dambrova, M. Trimethylamine N-oxide impairs pyruvate and fatty acid oxidation in cardiac mitochondria. Toxicol. Lett. 2017, 267, 32–38. [Google Scholar] [CrossRef]
  141. Ma, G.; Pan, B.; Chen, Y.; Guo, C.; Zhao, M.; Zheng, L.; Chen, B. Trimethylamine N-oxide in atherogenesis: Impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci. Rep. 2017, 37, BSR20160244. [Google Scholar] [CrossRef]
  142. Liu, X.; Shao, Y.; Tu, J.; Sun, J.; Li, L.; Tao, J.; Chen, J. Trimethylamine-N-oxide-stimulated hepatocyte-derived exosomes promote inflammation and endothelial dysfunction through nuclear factor-kappa B signaling. Ann. Transl. Med. 2021, 9, 1670. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, L.; Meng, G.; Huang, B.; Zhou, X.; Stavrakis, S.; Wang, M.; Li, X.; Zhou, L.; Wang, Y.; Wang, M. A potential relationship between gut microbes and atrial fibrillation: Trimethylamine N-oxide, a gut microbe-derived metabolite, facilitates the progression of atrial fibrillation. Int. J. Cardiol. 2018, 255, 92–98. [Google Scholar] [CrossRef] [PubMed]
  144. Zhao, Z.-H.; Xin, F.-Z.; Zhou, D.; Xue, Y.-Q.; Liu, X.-L.; Yang, R.-X.; Pan, Q.; Fan, J.-G. Trimethylamine N-oxide attenuates high-fat high-cholesterol diet-induced steatohepatitis by reducing hepatic cholesterol overload in rats. World J. Gastroenterol. 2019, 25, 2450. [Google Scholar] [CrossRef] [PubMed]
  145. Aldana-Hernández, P.; Leonard, K.-A.; Zhao, Y.-Y.; Curtis, J.M.; Field, C.J.; Jacobs, R.L. Dietary choline or trimethylamine N-oxide supplementation does not influence atherosclerosis development in LDLR−/− and ApoE−/− male mice. J. Nutr. 2020, 150, 249–255. [Google Scholar] [CrossRef]
  146. Collins, H.L.; Drazul-Schrader, D.; Sulpizio, A.C.; Koster, P.D.; Williamson, Y.; Adelman, S.J.; 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. [Google Scholar] [CrossRef]
  147. Huc, T.; Drapala, A.; Gawrys, M.; Konop, M.; Bielinska, K.; Zaorska, E.; Samborowska, E.; Wyczalkowska-Tomasik, A.; Pączek, L.; Dadlez, M. Chronic, low-dose TMAO treatment reduces diastolic dysfunction and heart fibrosis in hypertensive rats. Am. J. Physiol.-Heart Circ. Physiol. 2018, 315, H1805–H1820. [Google Scholar] [CrossRef]
  148. Gawrys-Kopczynska, M.; Konop, M.; Maksymiuk, K.; Kraszewska, K.; Derzsi, L.; Sozanski, K.; Holyst, R.; Pilz, M.; Samborowska, E.; Dobrowolski, L.; et al. TMAO, a seafood-derived molecule, produces diuresis and reduces mortality in heart failure rats. Elife 2020, 9, e57028. [Google Scholar] [CrossRef]
  149. Videja, M.; Vilskersts, R.; Korzh, S.; Cirule, H.; Sevostjanovs, E.; Dambrova, M.; Makrecka-Kuka, M. Microbiota-derived metabolite trimethylamine N-oxide protects mitochondrial energy metabolism and cardiac functionality in a rat model of right ventricle heart failure. Front. Cell Dev. Biol. 2021, 8, 1808. [Google Scholar] [CrossRef]
  150. Sun, X.; Jiao, X.; Ma, Y.; Liu, Y.; Zhang, L.; He, Y.; Chen, Y. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem. Biophys. Res. Commun. 2016, 481, 63–70. [Google Scholar] [CrossRef]
  151. Querio, G.; Antoniotti, S.; Levi, R.; Gallo, M.P. Trimethylamine N-Oxide Does Not Impact Viability, ROS Production, and Mitochondrial Membrane Potential of Adult Rat Cardiomyocytes. Int. J. Mol. Sci. 2019, 20, 3045. [Google Scholar] [CrossRef]
  152. Jaworska, K.; Konop, M.; Hutsch, T.; Perlejewski, K.; Radkowski, M.; Grochowska, M.; Bielak-Zmijewska, A.; Mosieniak, G.; Sikora, E.; Ufnal, M. TMA but not TMAO increases with age in rat plasma and affects smooth muscle cells viability. J. Gerontol. A Biol. Sci. Med. Sci. 2020, 75, 1276–1283. [Google Scholar] [CrossRef]
  153. Brieger, H.; Hodes, W.A. Toxic effects of exposure to vapors of aliphatic amines. AMA Arch. Ind. Hyg. Occup. Med. 1951, 3, 287–291. [Google Scholar]
  154. Friemann, W.; Overhoff, W.; Wolter, J.R. Eye diseases in the fishing industry. Arch. Gewerbepathol. Gewerbehyg. 1959, 17, 1–56. [Google Scholar]
  155. Guest, I.; Varma, D.R. Selective growth inhibition of the male progeny of mice treated with trimethylamine during pregnancy. Can. J. Physiol. Pharmacol. 1993, 71, 185–187. [Google Scholar] [CrossRef]
  156. Kinney, L.; Burgess, B.; Chen, H.; Kennedy, G. Inhalation toxicology of trimethylamine. Inhal. Toxicol. 1990, 2, 41–51. [Google Scholar] [CrossRef]
  157. Restini, C.B.A.; Fink, G.D.; Watts, S.W. Vascular reactivity stimulated by TMA and TMAO: Are perivascular adipose tissue and endothelium involved? Pharmacol. Res. 2021, 163, 105273. [Google Scholar] [CrossRef]
  158. Jaworska, K.; Bielinska, K.; Gawrys-Kopczynska, M.; Ufnal, M. TMA (trimethylamine), but not its oxide TMAO (trimethylamine-oxide), exerts haemodynamic effects: Implications for interpretation of cardiovascular actions of gut microbiome. Cardiovasc. Res. 2019, 115, 1948–1949. [Google Scholar] [CrossRef]
  159. Liang, X.; Zhang, Z.; Lv, Y.; Tong, L.; Liu, T.; Yi, H.; Zhou, X.; Yu, Z.; Tian, X.; Cui, Q. Reduction of intestinal trimethylamine by probiotics ameliorated lipid metabolic disorders associated with atherosclerosis. Nutrition 2020, 79, 110941. [Google Scholar] [CrossRef]
  160. Bollenbach, M.; Ortega, M.; Orman, M.; Drennan, C.L.; Balskus, E.P. Discovery of a cyclic choline analog that inhibits anaerobic choline metabolism by human gut bacteria. ACS Med. Chem. Lett. 2020, 11, 1980–1985. [Google Scholar] [CrossRef]
  161. Gabr, M.T.; Deganutti, G.; Reynolds, C.A. Peptidomimetic-based approach toward inhibitors of microbial trimethylamine lyases. Chem. Biol. Drug Des. 2021, 97, 231–236. [Google Scholar] [CrossRef]
  162. Gabr, M.T.; Machalz, D.; Pach, S.; Wolber, G. A benzoxazole derivative as an inhibitor of anaerobic choline metabolism by human gut microbiota. RSC Med. Chem. 2020, 11, 1402–1412. [Google Scholar] [CrossRef] [PubMed]
  163. Pathak, P.; Helsley, R.N.; Brown, A.L.; Buffa, J.A.; Choucair, I.; Nemet, I.; Gogonea, C.B.; Gogonea, V.; Wang, Z.; Garcia-Garcia, J.C. Small molecule inhibition of gut microbial choline trimethylamine lyase activity alters host cholesterol and bile acid metabolism. Am. J. Physiol.-Heart Circ. Physiol. 2020, 318, H1474–H1486. [Google Scholar] [CrossRef] [PubMed]
  164. Chen, K.; Zheng, X.; Feng, M.; Li, D.; Zhang, H. Gut microbiota-dependent metabolite trimethylamine N-oxide contributes to cardiac dysfunction in western diet-induced obese mice. Front. Physiol. 2017, 8, 139. [Google Scholar] [CrossRef] [PubMed]
  165. Organ, C.L.; Li, Z.; Sharp III, T.E.; Polhemus, D.J.; Gupta, N.; Goodchild, T.T.; Tang, W.W.; Hazen, S.L.; Lefer, D.J. Nonlethal Inhibition of Gut Microbial Trimethylamine N-oxide Production Improves Cardiac Function and Remodeling in a Murine Model of Heart Failure. J. Am. Heart Assoc. 2020, 9, e016223. [Google Scholar] [CrossRef]
Figure 1. TMA/TMAO metaorganismal pathway. TMA—trimethylamine; TMAO—trimethylamine N-oxide; FMO3—flavin-containing monooxygenase 3.
Figure 1. TMA/TMAO metaorganismal pathway. TMA—trimethylamine; TMAO—trimethylamine N-oxide; FMO3—flavin-containing monooxygenase 3.
Ijms 25 12511 g001
Figure 2. Hypothetical TMAO effects on cardiovascular pathology. ER stress—endoplasmic reticulum stress; FOXO1—Forkhead box protein O1; IL—interleukin; mt ROS—mitochondrial reactive oxygen species; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3—NOD-, LRR-, and pyrin domain-containing protein 3; PERK—protein kinase R-like endoplasmic reticulum kinase; TMAO—trimethylamine N-oxide; TNFα—tumor necrosis factor alpha; VCAM-1—vascular cell adhesion molecule 1.
Figure 2. Hypothetical TMAO effects on cardiovascular pathology. ER stress—endoplasmic reticulum stress; FOXO1—Forkhead box protein O1; IL—interleukin; mt ROS—mitochondrial reactive oxygen species; NF-κB—nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP3—NOD-, LRR-, and pyrin domain-containing protein 3; PERK—protein kinase R-like endoplasmic reticulum kinase; TMAO—trimethylamine N-oxide; TNFα—tumor necrosis factor alpha; VCAM-1—vascular cell adhesion molecule 1.
Ijms 25 12511 g002
Table 1. TMAO as a diagnostic biomarker in disease detection.
Table 1. TMAO as a diagnostic biomarker in disease detection.
Detected DiseaseDiagnostic AccuracyProposed TMAO Cutoff (μmol/L)ReferenceComments
Acute ischemic strokeModerate (AUC = 0.75)6.6[15]No adjustment to GFR
Moderate (AUC = 0.78)0.014[38]Unexpectedly low cut-off value
Moderate (AUC = 0.729)4.95[17]No adjustment to GFR
Atrial fibrillationNo differences between AF and healthy [37]
Coronary artery diseaseLow (AUC = 0.6)Not provided[18]
Low (AUC = 0.56)Not provided[24]No adjustment to GFR
No differences between CAD and healthy [36]
Diabetic kidney diseaseLow/moderate (AUC 0.691)227[39]
Heart failureModerate (AUC = 0.881)Not provided[20]
Moderate (AUC = 0.817)Not provided[21]HFpEF
Low (AUC  =  0.63)0.094[19]HFpEF; no adjustment to GFR
Footnote. High/moderate/low accuracy by receiver operator characteristics (ROC) curves [Fisher 2003]; AF—atrial fibrillation; AUC—area under ROC curve; CAD—coronary artery disease; HFpEF—heart failure with preserved ejection fraction; GFR—glomerular filtration rate; HF—heart failure.
Table 2. TMAO as a prognostic biomarker.
Table 2. TMAO as a prognostic biomarker.
DiseasePrognosed OutcomePrognostic AccuracyMedian/Mean Follow-Up DurationReferenceComments
Peripheral artery diseaseMortalityHR 2.065 years [43]
MACE, MortalityOR 1.684 years[22]
Chronic kidney diseaseMACEHR 1.233 years[58]
MortalityHR 4.325 years[59]
Mortality, MACENo association5.3 years (max)[60]
MortalityHR 1.935 years[61]
CV Mortality, MortalityHR 1.13; HR 1.146.1 years[62]Patients on hemodialysis
MACEAUC = 0.682 years[63]Patients on hemodialysis
MI, Stroke, or Peripheral Artery Disease EventNo association3.5 years[64]Patients with diabetes
Mortality, CV MortalitySHR 1.22;
SHR 1.41 (Men only)
5.3 years[65]Patients on peritoneal dialysis
HypertensionStrokeOR 1.224.5 years[66]
DiabetesMortalityHR 2.74.8 years[67]
MACENo association3.5 years[68]Type 2 diabetes with atherosclerosis risk factors
MACE, MortalityHR 2.05; HR 2.073 and 5 years[69]
Mortality, CV Mortality, MACENo association6.8 years; 6.8 years;
6.5 years
[70]
MACE, MortalityHR 1.29; HR 1.167.1 years[71]Type 2 diabetes
Carotid atherosclerosisCV MortalityNo association5.3 years[51]
Acute coronary syndromeMortalityHR 1.817 years[72]
MACENo association6.7 years[54]No association after adjustment to GFR
MortalityNo association5 years[55]
MACEHR 1.731 year[73]Patients on anti-platelet therapy
MACEHR: 1.852 years[74]Acute myocardial infarction and heart failure
CV Mortality, StrokeOR 1.89; OR 2.0133 months[75]Prior myocardial infarction
MACEHR 1.592 years[76]High TMAO levels at both time points; no adjustment to GFR
CV MortalityHR 11.627 years[42]
MACEHR 2.611 year[77]STEMI
New-Onset AFOR 1.291 year[78]No adjustment to GFR
Left Ventricular Systolic DysfunctionNo association30 days[79]STEMI
Acute ischemic strokeMACEHR 3.31 year[80]
Poor Functional Outcome, MortalityOR 3.09; OR 5.643 months[81]
Poor Functional Outcome, MortalityOR 1.21; OR 1.363 months[31]
MACEHR 1.691.9 years[82]
Post-Stroke Cognitive ImpairmentOR 3.301 year[83]
Stroke Recurrence, MACEHR 1.371 year[84]
Stroke RecurrenceHR 1.281 year[85]
Functional OutcomeNo associationAt discharge[86]
MACEHR 3.1283 month[38]
Major Ischemic Event, Poor Functional OutcomeOR 3.59; OR 2.581 year[47]
Coronary artery diseaseMortalityHR 1.955 years[44]
Acute Myocardial InfarctionNo association4.9 years[87]Stable angina pectoris
Mortality, CV MortalityHR 1.58; HR 1.665 years[88]
Mortality, CV MortalityNo association9.8 and 10.5 years[89]Two cohorts: CAD and community-based adults
MACElog rank p  =  0.0041.5 years[90]Stable angina pectoris
Individuals with and without CV diseaseCardiovascular Mortality, MortalityHR 1.8; HR 1.99.7 years[91]
Heart failureMortality or HF HospitalizationHR 3.825 years[46]HF with preserved EF
Mortality/Heart TxHR 1.465 years[92]
MACE, MortalityHR 1.57; HR 1.531.8 years[93]
Cardiovascular Mortality, HF HospitalizationHR 2.03; HR 1.962.4 years[94]HF with preserved EF
Mortality, Mortality/HFHR 1.26; 1.251 year[40]
Sever aortic stenosisMortalityHR 1.794.2 years[95]
Atrial fibrillationMortality, CV Mortality, Stroke;HR 1.65
HR 1.86
No association
4 years[96]
Footnote. AF—atrial fibrillation; CAD—coronary artery disease; EF—ejection fraction; GFR—glomerular filtration rate; HF—heart failure; HR—hazard ratio; MACE—major adverse cardiovascular events; OR—odds ratio; Tx—transplantation.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jaworska, K.; Kopacz, W.; Koper, M.; Ufnal, M. Microbiome-Derived Trimethylamine N-Oxide (TMAO) as a Multifaceted Biomarker in Cardiovascular Disease: Challenges and Opportunities. Int. J. Mol. Sci. 2024, 25, 12511. https://doi.org/10.3390/ijms252312511

AMA Style

Jaworska K, Kopacz W, Koper M, Ufnal M. Microbiome-Derived Trimethylamine N-Oxide (TMAO) as a Multifaceted Biomarker in Cardiovascular Disease: Challenges and Opportunities. International Journal of Molecular Sciences. 2024; 25(23):12511. https://doi.org/10.3390/ijms252312511

Chicago/Turabian Style

Jaworska, Kinga, Wojciech Kopacz, Mateusz Koper, and Marcin Ufnal. 2024. "Microbiome-Derived Trimethylamine N-Oxide (TMAO) as a Multifaceted Biomarker in Cardiovascular Disease: Challenges and Opportunities" International Journal of Molecular Sciences 25, no. 23: 12511. https://doi.org/10.3390/ijms252312511

APA Style

Jaworska, K., Kopacz, W., Koper, M., & Ufnal, M. (2024). Microbiome-Derived Trimethylamine N-Oxide (TMAO) as a Multifaceted Biomarker in Cardiovascular Disease: Challenges and Opportunities. International Journal of Molecular Sciences, 25(23), 12511. https://doi.org/10.3390/ijms252312511

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop