IMR Press / RCM / Volume 22 / Issue 3 / DOI: 10.31083/j.rcm2203085
Open Access Review
Trimethylamine N-oxide—a marker for atherosclerotic vascular disease
Guinan Xie1,†An Yan1,†Peng Lin2Yi Wang1,*Liping Guo3,*
Show Less
1 Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
2 Graduate School, Tianjin University of Traditional Chinese Medicine, 301617 Tianjin, China
3 Department of Cardiology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, 300120 Tianjin, China
*Correspondence: wangyi@tjutcm.edu.cn (Yi Wang); lpgtjn@163.com (Liping Guo)
These authors contributed equally.
Academic Editor: Gianluca Rigatelli
Rev. Cardiovasc. Med. 2021, 22(3), 787–797; https://doi.org/10.31083/j.rcm2203085
Submitted: 8 July 2021 | Revised: 2 August 2021 | Accepted: 6 August 2021 | Published: 24 September 2021
(This article belongs to the Special Issue Coronary Artery Atherosclerosis: Translation from Basic to Clinic)
Copyright: © 2021 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license (https://creativecommons.org/licenses/by/4.0/).
Abstract

As a potential causative factor in various cardiovascular diseases, the gut microbe-generated metabolite trimethylamine N-oxide (TMAO) has courted considerable research interest as a potential biomarker. TMAO is a small molecule considered to be beneficial for the health of deep-water animals due to its ability to protect proteins against hydrostatic pressure stress. However, it may cause deleterious effects in humans as mounting evidence suggests that TMAO may enhance atherosclerosis, independent of traditional risk factors. This may be mediated by its capacity to enhance inflammation, platelet activation and thrombosis, and inhibit reverse cholesterol transport. In humans, circulating levels of TMAO have been found to be associated with increased risk of developing atherosclerotic diseases such as carotid atherosclerosis, coronary atherosclerotic heart disease, stroke, and peripheral arteriosclerosis. This review aims to discuss the current role of TMAO in the atherosclerosis process, using animal models and clinical studies, with special attention to determining whether TMAO could be used as a marker for monitoring severity and prognosis in atherosclerosis and to evaluate evidence for its role as a mediator in the pathogenesis of atherosclerotic vascular disease.

Keywords
Atherosclerosis
Trimethylamine N-oxide
Coronary heart disease
Cardiovascular risk
1. Introduction

Atherosclerosis is a chronic inflammatory state and the main pathological basis of cardiovascular disease (CVD) [1]. Within the past ten years, the intestinal flora has been recognized as an independent and important metabolic organ, which closely links the metabolism of nutrients with molecules that cause chronic diseases [2]. Intestinal micro-ecological imbalance can promote the development of atherosclerosis by increasing systemic inflammation [3, 4, 5] and recently, a metabolite of intestinal flora TMAO, has received widespread attention as a marker for and potential mediator of many diseases.

Professor Tang WH of the Cleveland Medical Center, was the first researcher to report a role for elevated TMAO levels as a predictor of major adverse cardiac events such as myocardial infarction, stroke, and mortality [6]. Since then many comparable studies have shown that increased concentrations of TMAO and its precursors (L-carnitine, choline, and betaine) are associated with increased risks of cardiovascular adverse events and mortality [7, 8, 9, 10, 11, 12]. Furthermore, TMAO is thought to be more accurate at predicting these events than traditional CVD risk factors such as blood lipid, C-reactive protein (CRP) levels, or renal function [13]. In a meta-analysis of 17 clinical studies, an every 10 μmol/L increase in TMAO increased the risk of cardiovascular mortality by 7.6% [8]. High levels of TMAO are not only associated with the risk of CVD in humans, but also increase the risk of atherosclerosis in animal models. TMAO is believed to be involved in the complex pathological process of atherosclerotic lesions, such as the promotion of blood vessel inflammation [14], enhanced expression of macrophage scavenger receptors, and the formation of foam cells [6], as well as exacerbated platelet hyperreactivity enhanced thrombosis [15], and inhibition of bile acid synthesis [16].

Here we review the dietary sources, synthesis, and metabolic pathways of TMAO, and provided the latest evidence on the ability of TMAO to be used as a potential marker for atherosclerotic vascular disease, and finally tested TMAO as a target for the treatment of atherosclerosis.

2. Source, synthesis, and metabolism of TMAO

The main sources of TMAO are dietary choline, carnitine, and betaine. Choline is found in beef, chicken liver, bacon and soybeans, and betaine is found in wheat bran, wheat germ and spinach [17]. Meat products such as lean pork, lamb, and beef are the main sources of carnitine [18]. TMAO can also be obtained directly from fish [19] and high protein diets [20]; for example, two large eggs (65 grams) contain 320 mg of phosphatidylcholine, which represents the same amount of TMAO precursor as a 12-ounce beef burgers [21]. Trimethylamine (TMA) is an intermediate of TMAO which is mainly metabolized by the gut microbiota from dietary nutrients having a TMA like structure. It quickly reaches the liver through the portal circulation and flavin-containing monooxygenase 3 (FMO3) can oxidize TMA to form TMAO where it is released into the circulatory system and finally excreted by the kidney [6, 22] (Fig. 1). Consuming 50 mg of deuterium-labeled methyl d9-TMAO can be detected as early as 15 min in plasma, and 96% of the dose is excreted through urine instead of feces by 24 h [23]. The remaining TMAO is reduced to TMA by the action of TMAO reductase [24].

Fig. 1.

Role of TMAO in atherosclerosis progression. Annotation: Trimethylamine (TMA) is generated by the action of TMA lyases in the gut microbiota from Choline, Carnitine and betaine. TMA quickly reaches the liver through the portal circulation, flavin-containing monooxygenase 3 (FMO3) significantly oxidize TMA to form TMAO into the circulatory system. TMAO promotes inflammation of blood vessels, enhances the formation of foam cells, exacerbates platelet hyperreactivity and thrombosis potential, alteres bile acids and cholesterol transport. These factors are associated with increased risk of developing Atherosclerotic disease such as Carotid atherosclerosis, Coronary atherosclerotic heart disease, Stroke, and Peripheral arteriosclerosis.

Microbial enzymes involved in the formation of TMA involve two main pathways, one is the TMA lyase system (CUTC/D) which degrades choline [25]. The pathway encoded by the CUTC gene cluster is the main pathway for TMA production from human choline [26, 27] and the identified CUTC amplicons are associated with various taxa especially Clostridium XIVa strains and Eubacterium sp [28]. Furthermore, Kymberleigh A. Romano identified eight species representing two different phyla (Firmicutes and Proteobacteria) that showed significant choline consumption and TMA accumulation [29]. The other pathway is the decomposition of carnitine (CntA/B and YeaW/X) [30]. Here, CntA amplicons displayed high identity (~99%) to Gammaproteobacteria-derived references, primarily from Escherichia coli [28]. Recently, Robert A Koeth proposed that the conversion of dietary carnitine to TMAO also involves the L-carnitineγBBTMA/TMAO pathway [31]. However, only one type of Clostridium has been found to be able to convert γBB to TMA [31]. Therefore, it is necessary to conduct further research to investigate the composition and activity of the intestinal flora to detect potential correlations.

3. TMAO as an enhancer of atherosclerosis-experimental model
3.1 TMAO and endothelial inflammation

Inflammatory injury of the vascular endothelium is widely regarded as the initial stage of atherosclerosis [32]. In vitro studies have shown an up-regulated IL-6, CRP, TNF-α and reactive oxygen species (ROS), and reduced Nitric Oxide (NO) production in TMAO-treated endothelial cells (ECs), suggesting that TMAO can directly impair NO-mediated endothelial function [33]. TMAO can also up-regulate the expression of vascular cell adhesion molecule 1 (VCAM-1), promote the adhesion of monocytes, and inhibit the self-repair capability of ECs through protein kinase C (PKC) and NF-κB signaling, while enhancing macrophage adhesion [14, 34]. Furthermore, TMAO reduces the expression of the anti-inflammatory cytokine IL-10, which can protect ECs from damage caused by inflammation and increased oxidative stress [35] (Fig. 2).

Fig. 2.

Putative molecular mechanisms underlying TMAO-induced atherogenic effects. Annotation: High concentration of TMAO plays a vital role in endothelial hyper permeability and inflammation, oxidative stress, cholesterol metabolism, plaque formation, activation and thrombosis.

In young mice, six months of dietary supplementation with TMAO can induced aging of the arterial endothelium. This effect was accompanied by increased vascular nitrotyrosine, a marker of oxidative stress [36]. In a different study, aged rats exhibited higher plasma TMAO, TNF-α, and IL-1β, and increased eNOS expression in the aorta when compared to young rats, all of these effects were restored to control levels by treatment with DMB (an inhibitor of TMA formation) [37]. These finding clearly indicated that the increase in circulating TMAO levels caused by aging can lead to endothelial dysfunction.

It has been reported that there are several mechanisms underlying the activation of inflammation, including lysosomal rupture, altered K+ channel gating, and activation of ROS [38]. Krishna proved that endotheliitis caused by TMAO is activated both in vitro and in vivo and in the presence of blockers or inhibitors of individual pathways, and found that the formation and activation of the NLRP3 inflammasomes induced by TMAO in ECs were markedly attenuated or abolished by the ROS scavenger, N-acetyl-L-cysteine and cathepsin B inhibitor. These results showed that TMAO can activate NLRP3 inflammasomes in ECs through at least two different pathways, involving increased ROS production, destabilized lysosomes, and enhanced cathepsin B activity [39]. Sun also confirmed that TMAO activates the thioredoxin interacting protein (ROS-TXNIP-NLRP3) signaling pathway through oxidative stress and promotes the expression of the inflammasome NLRP3 [40]. A different mechanism for TMAO action involves a reduction in Adenosine triphosphate(ATP) production and induced endothelial cell apoptosis by damaging the structure and function of mitochondria, and the succinate dehydrogenase complex subunit B (SDHB)/ROS signaling pathway has been found to play an important role in this process [41].

It is well known that loss of integrity of tight junctions found in the endothelium helps to enhance the permeability of the quasi-cellular endothelium. Stimulating ECs with TMAO can reduce the expression of the tight junction protein ZO-1 in the ECs monolayer and change ECs permeability, which was prevented by silencing NLRP3 in the ECs [39]. The high-mobility basal box protein 1 (HMGB1) is also an inflammatory mediator, which can destroy cell-to-cell connections, resulting in vascular endothelial hyper permeability [42]. In a study conducted by Singh, it was discovered that TMAO significantly reduced the expression of junctional proteins such as ZO-2, VE-kadrin, and Okrudin in the EC monolayer, while this effect was markedly attenuated by glycyrrhizin, an HMGB1 binding compound [42]. Furthermore, TMAO can upregulate HMGB1, and HMGB1 causing further activation of toll-like receptor 4(TLR4, an important receptor for HMGB1 binding where HMGB1 can activate inflammatory pathways). TLR4 siRNA can protect ECs from the interference by TMAO-related tight junction proteins [42] (Fig. 2).

3.2 TMAO and reverse cholesterol transport (RCT)

High TMAO levels were also found to be accompanied by low HDL levels in patients with cardiovascular disease [43]. The accumulation of oxidized low-density lipoprotein (ox-LDL) in LDL cholesterol macrophages can be transported to the liver for recirculation or excreted in the form of bile acid [44]. Previous studies have confirmed that the RCT of choline-added APOE-/-mice was reduced by about 30%, and total bile acids in mice and the expression of key bile acid synthases Cyp7a1 and Cyp27a1 in the liver were also significantly reduced relative to normal chow (P < 0.05) [6, 22], resulting in changes to the main mechanism of removing cholesterol in the body (Fig. 2).

3.3 TMAO and atherosclerotic plaque

Several experimental models have demonstrated a role for TMAO in the process of atherosclerosis. A TMAO-diet significantly promoted plaque progression in APOE-/- mice fed with high-fat for 20 weeks when compared to C57BL/6J mice fed with a normal chow diet. However, the levels of blood sugar, cholesterol and triglycerides had no significant effect on the plaque area [6], and this effect could be reversed by reducing the level of TMAO [45]. TMAO can also increase the formation of foam cells by up-regulating the macrophage scavenger receptors CD36 and SR-A1, which promote the continuous development of plaques [6]. Furthermore, the CD36/MAPK/JNK pathway may play a crucial role in the TMAO-induced formation of foam cells [46]. In addition, it was also found that TMAO can induce the expression of heat shock proteins GRP94 and HSP70 in J774A.1 mouse macrophages, leading to endoplasmic reticulum stress [47] (Fig. 2). TMAO not only promotes plaque formation, but also contributes to plaque instability. In the tandem stenosis mouse model, which reflects plaque instability as typically seen in patients, TMAO levels correlated with several characteristics of plaque instability, such as markers of inflammation, platelet activation, and intraplaque hemorrhage [48].

3.4 TMAO and Thrombus

Platelets contribute to the formation of thrombus and foam cells and play an important role in the occurrence and development of atherosclerosis [49]. After 2 months of choline supplementation, healthy subjects showed a significant >10-fold increase in plasma TMAO levels at both 1- and 2-month periods, with a corresponding enhanced platelet aggregation response to submaximal adenosine diphosphate [50]. Furthermore, large scale clinical association studies (n >4000 subjects) independently demonstrated that plasma TMAO levels are associated with risk of thrombotic events [15]. The exact mechanisms by which TMAO activates platelets however are still unclear.

TMAO can induce the release of calcium from platelets by changing the inositol 1,4,5-triphosphate (IP3) signal transduction pathway, and directly enhance the platelet response to a variety of different agonists (ADP, thrombin and collagen) and promote platelet hyperresponsiveness by enhancing stimulus-dependent Ca2+ release [51]. Tissue factors are essential mediators of hemostasis and trigger thrombosis [52]. Xiaoye Cheng demonstrated that TMAO promoted tissue factor (TF) expression via activation of the NF-κB signaling pathway in primary HCAECs [53]. TMAO can also attenuate the inhibitory effect of clopidogrel on platelet aggregation by inhibiting P2Y12 receptor inhibitors [54] (Fig. 2). Recently, Sarah M Skye found that the expression of the CutC gene from intestinal microbes is sufficient to transmit enhanced platelet reactivity and thrombosis potential in a host via TMA/TMAO generation [55]. These studies implicate TMAO as a potential molecular target for the treatment of atherosclerosis and thrombotic.

4. The role of TMAO in vasculopathy induced by atherosclerotic risk factors

The role of TMAO in atherosclerotic vasculopathy has been demonstrated in various models of atherosclerotic risk factors, namely: diabetes, hypertension, and chronic kidney disease.

4.1 Diabetes

High levels of circulating TMAO are associated with an increased risk of both type 1 and type 2 diabetes (T2DM) [56, 57, 58], as well as pre-diabetes prevalence [59], and diabetic complications such as retinal neuropathy [60]. Miao found that LIRKO mice suffered from severe hyperglycemia after 4 months Pigen diet, which was completely prevented by the knockdown of FMO3 (TMAO synthase) [61]. Similar results were obtained on obese/insulin resistant subjects, where FMO3 was significantly increased in the morbidly obese group (P < 0.05). However, this effect was smaller than that seen in the mouse model, possibly because diabetic patients were treated with insulin and/or other drugs to increase insulin sensitivity [61].

The exact biological mechanism of how plasma TMAO concentration is involved in glucose metabolism remains to be clarified. PI3K is a key protein that transduces insulin signals into the regulation of glucose metabolism, while Akt is an important molecule downstream of the PI3K pathway, and its stimulation will in turn activate glycogen synthetase (a critical regulatory enzyme for the regulation of hepatic glycogen storage) [62]. In a study conducted by Gao it was shown that dietary TMAO reduces the mRNA levels of PI3K and Akt, indicating that dietary TMAO may exacerbate the blocked insulin signaling pathway [63].

Another mechanism involving TMAO promotes the regulation of transcription factor FoxO1 caused by PERK, and inhibition of FMO3 or manipulation of the gut microbiota can hinder metabolic dysfunction mediated by the PERK-FoxO1 axis [64]. Specifically, FoxO1 drives glucose gene expression and inhibited by insulin, the deletion of Foxo1 in the liver can reduce excessive glucose production caused by general ablation of insulin receptors [65]. PERK is a key sensor of intracellular stress and has previously been shown to induce FoxO1 by phosphorylation on serine 298 [66]. These data indicate that TMAO may be central to the pathogenesis of metabolic syndrome.

4.2 Hypertension

Research conducted by Yang observed a significant decrease in microbial richness, diversity, and reproducibility in the spontaneously hypertensive rat [67], suggesting that the intestinal flora may be involved in the occurrence of hypertension, although the mechanism has not yet been fully elucidated. Meta-studies have shown that for every 5 μmol/L and 10 μmol/L increase in TMAO, the prevalence of hypertension will increase by 9% and 20% respectively, revealing a significant dose-dependent positive correlation between TMAO concentration and the risk of hypertension [68]. TMAO is an important metabolite of enteral malnutrition and may be involved in the pathogenesis of hypertension as a mediator.

A different study demonstrated that patients with hypertension have an increased abundance of the cutC gene in their intestines, which in turn produces more TMAO [69]. The integrity of the intestinal barrier is essential for maintaining host health and preventing inflammation and atherosclerotic processes. Kinga Jaworska confirmed that the permeability of the colon to TMA in hypertensive patients is increased and promotes its penetration into the circulatory system [70], representing a novel point of view that the high concentration of TMAO in cardiovascular diseases may depend upon the increased permeability of the colon to TMA, and changes in colon permeability rather than TMAO levels are the indicators of cardiovascular risk [70].

4.3 Chronic kidney disease (CKD)

The clearance of TMAO depends largely upon its excretion by the kidneys [71]. Serum TMAO concentrations substantially increase with deterioration of kidney function, and this effect is reversed by renal transplantation [72]. Meta-analyses of a total of 32 eligible clinical studies involving 42,062 participants showed that TMAO was strongly inversely correlated with glomerular filtration rate(GFR) and positively associated with the urine albumin-to-creatinine ratio, serum creatinine, and urine albumin excretion rates [73]. Jason R reported that the increase in TMAO concentration in CKD patients receiving coronary angiography was related to the load of coronary atherosclerosis and may be related to long-term mortality in CKD patients receiving coronary angiography [72]. Moreover, it was observed that TMAO predicts a poor overall survival rate in subjects with CKD [74, 75, 76].

TMAO is an independent predictor of CKD phase 3–5 mortality rate [77] and in animal models, TMAO levels significantly increase renal fibrosis compared with control (P < 0.05), while this result can be reversed by TMA lyase inhibitors [78]. This model was also demonstrated in ApoE KO mice, where the animals developed chronic kidney disease with elevated TMAO levels within 14 weeks of adenine administration, after using TMA inhibitors, and many markers of kidney injury (creatinine, cystatin C, FGF23, and urinary microalbumin) were significantly reduced [79]. This suggests that TMAO may represent a novel modifiable risk factor and therapeutic target for improving atherosclerosis in patients with CKD.

5. Is TMAO a reliable marker for atherosclerosis?

As discussed so far TMAO has a role in the pathological process of atherosclerosis, but can it also be used to clinically monitor the development and prognosis of patients with atherosclerosis.

5.1 TMAO predicts carotid atherosclerosis

TMAO is a significant predictor of carotid plaque formation which is more significant than gender, diastolic blood pressure, total cholesterol, or diabetes mellitus [80]. A study from southern Germany showed that higher TMAO levels can predict thickness of the carotid intima-media (cIMT) [81]. TMAO is also an independent predictor of new lesions as seen by MRI after carotid artery stenting (CAS) [82]. In patients with severe carotid stenosis (>70%) it was found that elevated plasma TMAO level were associated with the increased risk of new lesions on diffusion-weighted imaging(DWI) within 1–3 days of CAS1, and that a TMAO of 4.29 μmol/L can better predict ischemic brain damage which is secondary to CAS [82].

5.2 TMAO is a predictive marker of coronary plaque development

In patients with non-ST-segment elevation myocardial infarction (NSTEMI) and ST-segment elevation myocardial infarction (STEMI), TMAO was found to be independently related to the severity and prognosis of coronary atherosclerosis load [83, 84, 85]. TMAO is also significantly related to the severity of the culprit segment of calcification, including maximum calcification arc, maximum calcification thickness, and calcification length, and can be used as an indicator to the calcium load of the culprit plaque [86]. Studies have shown that repeated rupture or erosion of a plaque followed by healing, is related to the vulnerability of the plaque and a high level of inflammation, which helps to increase the high risk of coronary thrombosis [87, 88]. The study reported that in patients with STEMI, elevated TMAO levels indicated a higher SYNTAX score (response to coronary atherosclerosis load) and the presence of multivascular disease [89], and TMAO levels in patients with plaque rupture was significantly higher than those with no rupture [90]. While TMAO levels in patients with repaired plaques were significantly higher than those with unrepaired plaques, and can be used as a potential biomarker to predict healed plaque presence with a cutoff value of 2.9 μmol/L [91]. The author believes that TMAO levels in the circulation may reflect the vulnerability and progression of coronary plaques.

TMAO not only plays a role in the process of primary atherosclerosis, but also contributes to late stent thrombosis after revascularization. A recent study found that an area under the curve (AUC) value for plasma TMAO of 0.85 can distinguish patients with new atherosclerosis and old atherosclerosis in STEMI patients who underwent PCI surgery, by using OCT to evaluate the culprit plaque [92]. The above studies indicate that high TMAO levels can be used as a useful biomarker to distinguish between plaque instability and rupture and represents a promising diagnostic marker for the prediction of new atherosclerosis in a stent.

5.3 TMAO is an independent predictor of stroke development and prognosis

TMAO can cross the blood-brain barrier and cause oxidative stress, thus potentially leading to brain damage [93, 94]. Research has found that higher TMAO levels are associated with increased risk of a first stroke [95, 96, 97], and are closely related to a poor prognosis, such as post-stroke cognitive impairment (PSCI). When predicting moderate to severe stroke, a critical value for plasma TMAO levels has been found to be 4.95 μmol/L [98]. Other studies however, suggest an optimal value of 6.6 μmol/L [96]. PSCI is a common consequence of stroke, affecting approximately one-third of stroke survivors after one-year post stroke [99]. A study included 256 patients with ischemic stroke, suggesting that increases in plasma TMAO levels are related to poor cognitive function one year after ischemic stroke, and supports TMAO as a predictive biomarker of PSCI [100]. In patients with acute ischemic stroke, TMAO at a concentration of 5 μmol /L can predict early deterioration in neurological function [101], and can be used as an independent predictor of stroke severity and infarct volume in patients with acute ischemia [102, 103, 104]. Furthermore, TMAO is also associated with poor prognosis in patients with cerebral hemorrhage at 3 months [105]. Su H suggested that TMAO can promote an increase in reactive astrocytes and glial scar formation through the Smurf2/ALK5 axis, thus aggravating nervous system damage after ischemic stroke [106]. Therefore, the study of TMAO can provide important predictive insights into the development and prognosis in stroke patients.

5.4 TMAO predicts the risk of peripheral arterial disease (PAD)

Dr. Vichai Senthong first reported that elevated plasma TMAO level are an important prognostic marker for patients with PAD [107]. In these patients, the increase of TMAO levels was related to an increased risk of death by 2.7 times, after adjusting for traditional risk factors, such as inflammatory biomarkers and a history of coronary artery disease, where the highest quartile for TMAO can still predict 5-year mortality [107]. PAD is characterized by atherosclerotic stenosis of the lower extremity vessels, leading to ischemic muscle pain in the elderly. Compared to patients with intermittent claudication, patients with advanced chronic severe limb ischemia (CLI) showed higher serum carnitine and TMAO levels and poorer long-term survival [108], whereas PAD patients with TMAO >2.26 μmol/L exhibited higher risk of cardiovascular death [109]. To date potential mortality and pathophysiological predictors of PAD have not been clearly defined, this may be the reason why mortality and ischemic amputation rates in PAD patients are still very high [110]. These studies confirm the clinical prognostic value of TMAO levels in patients with PAD, and these findings may be used to develop a new prognostic indicator of risk stratification.

Taken together the experimental data on TMAO it might be suggested that TMAO is a potent atherogenic cytokine and increased risk of developing atherosclerotic diseases such as CAS, coronary atherosclerosis, stroke, and PAD (Table 1, Ref. [80, 81, 82, 83, 84, 85, 86, 89, 90, 91, 92, 95, 96, 97, 98, 99, 100, 102, 103, 104, 105, 107, 108, 109]).

Table 1.TMAO plays a role in atherosclerosis studies.
TMAO’s significance References
CAS TMAO is a significant independent predictor of carotid plaque burden. [80]
Higher TMAO levels predicted increased cIMT. [81]
Plasma level of TMAO can be a predictor of ischemic brain injury secondary to CAS. [82]
The optimal cutoff value of TMAO in patients with >70% carotid artery stenosis was 4.29 µmol/L. [82]
Coronary atherosclerosis High Plasma TMAO is associated with coronary atherosclerotic burden in patients with STIME or NSTEMI. [83],[84],[85],[89]
TMAO is positively correlated with the incidence of calcification in the culprit lesion segment. [86]
Plasma TMAO level is associated with plaque rupture. [85],[90],[92]
Plasma TMAO level is associated with healed culprit plaques. [91]
A cutoff value of TMAO was 2.9 µmol/L in patients with STEMI can be used as a potential biomarker to predict healed plaque presence. [91]
AUC value for plasma TMAO of 0.85 can distinguish patients with new atherosclerosis and old atherosclerosis in STEMI patients. [92]
Stroke Serum TMAO concentration exhibited higher risk of first Stroke in Chinese patients. [95],[96],[97]
TMAO level 4.95 (6.6) μmol/L has high sensitivity and specificity for moderate to severe stroke. [96],[98]
Higher TMAO levels correlate with worse neurological deficit . [96],[100]
TMAO is an independent predictor for cognitive impairment in post-stroke patients. [99]
TMAO is an independent predictor of functional outcome and mortality of patients with ischemic stroke. [102],[103],[104]
TMAO is also associated with poor prognosis in patients with cerebral hemorrhage at 3 months. [105]
PAD TMAO is associated with PAD severity and prognosis. [107],[108]
PAD patients with TMAO >2.26 µmol/L exhibited higher risk of cardiovascular death. [109]
6. Treatment

Choline and carnitine are the main sources of TMAO production related to intestinal microbiota, therefore, diet regulation is a reasonable and cost-effective intervention strategy. Studies have shown that a healthy diet (vegetarian or low calorie diet) significantly improves TMAO levels [111, 112, 113]. Interestingly, microwave cooking can reduce the content of L-carnitine in some seafood [114]. In addition, exercise can change the diversity and distribution of the human microbial community [115], and voluntary exercise for 8 weeks can inhibit the increase in plasma TMAO in obese mice induced by a high-fat diet and prevent heart dysfunction also [116]. Some plant extracts and fruits, such as oolong tea [117], hawthorn [118], resveratrol [119], and ginkgolide B [120], have been shown to reduce the levels of circulating TMAO, which is beneficial for the improvement of atherosclerosis.

The use of prebiotics and probiotics may also help to produce a positive impact on the composition of the intestinal flora. Thus an in-depth study of the CutC/D gene responsible for the synthesis of TMA lyase and the development of a strain that antagonizes the synthesis pathway of TMAO represents a promising avenue of research [121]. For example, the TMA blockers DMB and IMC can inhibit the activity of microbial choline TMA enzymes in the body, increase the excretion of neutral sterols in mouse feces in the form of fecal sterols (bacterial metabolite of cholesterol), and prevent the accumulation of liver cholesterol driven by the diet [122]. Akkermansia muciniphila plays a protective role by improving intestinal barrier function to resist atherosclerosis [123]. Fecal microbiota transplantation (FMT) can reduce fecal TMA content and the content of serum TMAO [124]. These strategies that have focused on gut microbiota and microbial metabolites provide promising new insights into the treatment of atherosclerosis and CVDs.

7. Summary and future prospects

Emerging data has established a direct link between diet, the intestinal flora metabolite TMAO, and atherosclerosis. TMAO is involved in the complex process of atherosclerosis and plays an important role in the pathogenesis of atherosclerosis caused by various risk factors such as diabetes, hypertension, and chronic kidney disease. In humans, it has been proven that TMAO is a reliable marker for predicting the development and prognosis of arterial vascular disease. Taken together this convincing evidence has emerged from preclinical studies suggest a strong relationship between TMAO and atherosclerosis. Further studies have focused on molecular mechanisms and the development of safe, effective, and feasible dietary strategies to use as an anti-atherosclerotic, involving TMAO reduction and this has led to potentially significant public health benefits. Innovative therapeutic approaches targeting gut microbiota and TMAO, including lifestyle modifications, TMA inhibitors, prebiotics, probiotics, and Chinese herbs, have shed new light on the great potential of targeting TMAO to elucidate the fundamental mechanisms underlying the disease. We propose to test TMAO levels clinically to fully assess its role as a potential marker for atherosclerosis.

Author contributions

GX and AY conceptualized the study and prepared the original draft wrote; GX and PL revised the manuscript; PL, LG and YW provided language help and writing assistance.

Ethics approval and consent to participate

Not applicable.

Acknowledgment

Thanks to all those who helped us during the writing of this manuscript, and the peer reviewers for their opinions and suggestions.

Funding

Tianjin Postgraduate Research and Innovation Project (2020YJSB196): Study on the Mechanism of Jiangzhi Ruanmai Recipe in Improving Atherosclerosis by Regulating Cholesterol Reverse Transport.

Conflict of interest

The authors declare no conflict of interest.

References
[1]
Aiello RJ, Bourassa PK, Lindsey S, Weng W, Natoli E, Rollins BJ, et al. Monocyte Chemoattractant Protein-1 Accelerates Atherosclerosis in Apolipoprotein E-Deficient Mice. Arteriosclerosis, Thrombosis, and Vascular Biology. 1999; 19: 1518–1525.
[2]
Chang C, Kao C. Current understanding of the gut microbiota shaping mechanisms. Journal of Biomedical Science. 2019; 26: 59.
[3]
van den Munckhof ICL, Kurilshikov A, Ter Horst R, Riksen NP, Joosten LAB, Zhernakova A, et al. Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: a systematic review of human studies. Obesity Reviews. 2018; 19: 1719–1734.
[4]
Drosos I, Tavridou A, Kolios G. New aspects on the metabolic role of intestinal microbiota in the development of atherosclerosis. Metabolism: Clinical and Experimental. 2015; 64: 476–481.
[5]
Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. The Journal of Biological Chemistry. 2015; 290: 5647–5660.
[6]
Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011; 472: 57–63.
[7]
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. Journal of the American Heart Association. 2017; 6: e004947.
[8]
Schiattarella GG, Sannino A, Toscano E, Giugliano G, Gargiulo G, Franzone A, et al. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. European Heart Journal. 2017; 38: 2948–2956.
[9]
Senthong V, Wang Z, Li XS, Fan Y, Wu Y, Tang WHW, et al. Intestinal Microbiota-Generated Metabolite Trimethylamine-N-Oxide and 5-Year Mortality Risk in Stable Coronary Artery Disease: the Contributory Role of Intestinal Microbiota in a COURAGE-Like Patient Cohort. Journal of the American Heart Association. 2016; 5: e002816.
[10]
Hochstrasser SR, Metzger K, Vincent AM, Becker C, Keller AKJ, Beck K, et al. Trimethylamine-N-oxide (TMAO) predicts short- and long-term mortality and poor neurological outcome in out-of-hospital cardiac arrest patients. Clinical Chemistry and Laboratory Medicine. 2020; 59: 393–402.
[11]
Heianza Y, Ma W, DiDonato JA, Sun Q, Rimm EB, Hu FB, et al. Long-Term Changes in Gut Microbial Metabolite Trimethylamine N-Oxide and Coronary Heart Disease Risk. Journal of the American College of Cardiology. 2020; 75: 763–772.
[12]
Gencer B, Li XS, Gurmu Y, Bonaca MP, Morrow DA, Cohen M, 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. Journal of the American Heart Association. 2020; 9: e015331.
[13]
Tang WHW, Kitai T, Hazen SL. Gut Microbiota in Cardiovascular Health and Disease. Circulation Research. 2017; 120: 1183–1196.
[14]
Seldin MM, Meng Y, Qi H, Zhu W, Wang Z, Hazen SL, et al. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-kappaB. Journal of the American Heart Association. 2016; 5: e2767.
[15]
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: 422-8.
[16]
Ding L, Chang M, Guo Y, Zhang L, Xue C, Yanagita T, et al. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids in Health and Disease. 2018; 17: 286.
[17]
Zeisel SH, Mar M, Howe JC, Holden JM. Concentrations of choline-containing compounds and betaine in common foods. The Journal of Nutrition. 2003; 133: 1302–1307.
[18]
Demarquoy J, Georges B, Rigault C, Royer M, Clairet A, Soty M, et al. Radioisotopic determination of l-carnitine content in foods commonly eaten in Western countries. Food Chemistry. 2004; 86: 137–142.
[19]
Cho CE, Taesuwan S, Malysheva OV, Bender E, Tulchinsky NF, Yan J, et al. 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. Molecular Nutrition & Food Research. 2017; 61.
[20]
Mitchell SM, Milan AM, Mitchell CJ, Gillies NA, D’Souza RF, Zeng N, et al. Protein Intake at Twice the RDA in Older Men Increases Circulatory Concentrations of the Microbiome Metabolite Trimethylamine-N-Oxide (TMAO). Nutrients. 2019; 11: 2207.
[21]
Spence JD. Trimethylamine N-oxide: not just red meat—egg yolk and renal function are also important. European Heart Journal. 2019; 40: 3498–3498.
[22]
Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature Medicine. 2013; 19: 576–585.
[23]
Taesuwan S, Cho CE, Malysheva OV, Bender E, King JH, Yan J, et al. The metabolic fate of isotopically labeled trimethylamine-N-oxide (TMAO) in humans. the Journal of Nutritional Biochemistry. 2017; 45: 77–82.
[24]
Chhibber-Goel J, Gaur A, Singhal V, Parakh N, Bhargava B, Sharma A. The complex metabolism of trimethylamine in humans: endogenous and exogenous sources. Expert Reviews in Molecular Medicine. 2016; 18: e8.
[25]
Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proceedings of the National Academy of Sciences of the United States of America. 2012; 109: 21307–21312.
[26]
Martínez-del Campo A, Bodea S, Hamer HA, Marks JA, Haiser HJ, Turnbaugh PJ, et al. Characterization and detection of a widely distributed gene cluster that predicts anaerobic choline utilization by human gut bacteria. MBio. 2015; 6: e00042-15.
[27]
Dalla VA, Gargari G, Taverniti V, Rondini G, Velardi I, Gambaro V, et al. Urinary TMAO Levels Are Associated with the Taxonomic Composition of the Gut Microbiota and with the Choline TMA-Lyase Gene (cutC) Harbored by Enterobacteriaceae. Nutrients. 2019; 12: 62.
[28]
Rath S, Heidrich B, Pieper DH, Vital M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome. 2017; 5: 54.
[29]
Romano KA, Vivas EI, Amador-Noguez D, Rey FE. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio. 2015; 6: e02481.
[30]
Koeth RA, Levison BS, Culley MK, Buffa JA, Wang Z, Gregory JC, et al. Γ-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metabolism. 2014; 20: 799–812.
[31]
Koeth RA, Lam-Galvez BR, Kirsop J, Wang Z, Levison BS, Gu X, et al. L-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. Journal of Clinical Investigation. 2019; 129: 373–387.
[32]
Gimbrone MA, García-Cardeña G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circulation Research. 2016; 118: 620–636.
[33]
Chou R, Chen C, Chen I, Huang H, Lu Y, Kuo C, et al. Trimethylamine N-Oxide, Circulating Endothelial Progenitor Cells, and Endothelial Function in Patients with Stable Angina. Scientific Reports. 2019; 9: 4249.
[34]
Ma G, Pan B, Chen Y, Guo C, Zhao M, Zheng L, et al. Trimethylamine N-oxide in atherogenesis: impairing endothelial self-repair capacity and enhancing monocyte adhesion. Bioscience Reports. 2017; 37: BSR20160244.
[35]
Chen H, Li J, Li N, Liu H, Tang J. Increased circulating trimethylamine N-oxide plays a contributory role in the development of endothelial dysfunction and hypertension in the RUPP rat model of preeclampsia. Hypertension in Pregnancy. 2019; 38: 96–104.
[36]
Brunt VE, Gioscia-Ryan RA, Casso AG, VanDongen NS, Ziemba BP, Sapinsley ZJ, et al. Trimethylamine-N-Oxide Promotes Age-Related Vascular Oxidative Stress and Endothelial Dysfunction in Mice and Healthy Humans. Hypertension. 2020; 76: 101–112.
[37]
Li T, Chen Y, Gua C, Li X. Elevated Circulating Trimethylamine N-Oxide Levels Contribute to Endothelial Dysfunction in Aged Rats through Vascular Inflammation and Oxidative Stress. Frontiers in Physiology. 2017; 8: 350.
[38]
Meng N, Xia M, Lu Y, Wang M, Boini KM, Li P, et al. Activation of NLRP3 inflammasomes in mouse hepatic stellate cells during Schistosoma J. infection. Oncotarget. 2016; 7: 39316–39331.
[39]
Boini KM, Hussain T, Li P, Koka S. Trimethylamine-N-Oxide Instigates NLRP3 Inflammasome Activation and Endothelial Dysfunction. Cellular Physiology and Biochemistry. 2017; 44: 152–162.
[40]
Sun X, Jiao X, Ma Y, Liu Y, Zhang L, He Y, et al. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochemical and Biophysical Research Communications. 2016; 481: 63–70.
[41]
Wu P, Chen J, Chen J, Tao J, Wu S, Xu G, et al. Trimethylamine N-oxide promotes apoE(-/-) mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway. Journal of Cellular Physiology. 2020; 235: 6582–6591.
[42]
Singh GB, Zhang Y, Boini KM, Koka S. High Mobility Group Box 1 Mediates TMAO-Induced Endothelial Dysfunction. International Journal of Molecular Sciences. 2019; 20: 3570.
[43]
Tang WHW, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, et al. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. Journal of the American College of Cardiology. 2014; 64: 1908–1914.
[44]
Sorci-Thomas MG, Thomas MJ. Microdomains, Inflammation, and Atherosclerosis. Circulation Research. 2016; 118: 679–691.
[45]
Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Org E, et al. Non-lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell. 2015; 163: 1585–1595.
[46]
Geng J, Yang C, Wang B, Zhang X, Hu T, Gu Y, et al. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomedicine & Pharmacotherapy. 2018; 97: 941–947.
[47]
Mohammadi A, Vahabzadeh Z, Jamalzadeh S, Khalili T. Trimethylamine-N-oxide, as a risk factor for atherosclerosis, induces stress in J774a.1 murine macrophages. Advances in Medical Sciences. 2018; 63: 57–63.
[48]
Koay YC, Chen Y, Wali JA, Luk AWS, Li M, Doma H, et al. Plasma levels of trimethylamine-N-oxide can be increased with ‘healthy’ and ‘unhealthy’ diets and do not correlate with the extent of atherosclerosis but with plaque instability. Cardiovascular Research. 2021; 117: 435–449.
[49]
Davì G, Patrono C. Platelet activation and atherothrombosis. The New England Journal of Medicine. 2007; 357: 2482–2494.
[50]
Zhu W, Wang Z, Tang W, Hazen SL. Gut Microbe-Generated Trimethylamine N-Oxide From Dietary Choline Is Prothrombotic in Subjects. Circulation. 2017; 135: 1671–1673.
[51]
Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell. 2016; 165: 111–124.
[52]
Grover SP, Mackman N. Tissue Factor: an Essential Mediator of Hemostasis and Trigger of Thrombosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2018; 38: 709–725.
[53]
Cheng X, Qiu X, Liu Y, Yuan C, Yang X. Trimethylamine N-oxide promotes tissue factor expression and activity in vascular endothelial cells: a new link between trimethylamine N-oxide and atherosclerotic thrombosis. Thrombosis Research. 2019; 177: 110–116.
[54]
Ma R, Fu W, Zhang J, Hu X, Yang J, Jiang H. TMAO: a potential mediator of clopidogrel resistance. Scientific Reports. 2021; 11: 6580.
[55]
Skye SM, Zhu W, Romano KA, Guo C, Wang Z, Jia X, et al. Microbial Transplantation with Human Gut Commensals Containing CutC is Sufficient to Transmit Enhanced Platelet Reactivity and Thrombosis Potential. Circulation Research. 2018; 123: 1164–1176.
[56]
Zhuang R, Ge X, Han L, Yu P, Gong X, Meng Q, et al. Gut microbe-generated metabolite trimethylamine N-oxide and the risk of diabetes: A systematic review and dose-response meta-analysis. Obesity Reviews. 2019; 20: 883–894.
[57]
Winther SA, Øllgaard JC, Tofte N, Tarnow L, Wang Z, Ahluwalia TS, et al. 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.
[58]
Schugar RC, Shih DM, Warrier M, Helsley RN, Burrows A, Ferguson D, et al. The TMAO-Producing Enzyme Flavin-Containing Monooxygenase 3 Regulates Obesity and the Beiging of White Adipose Tissue. Cell Reports. 2017; 19: 2451–2461.
[59]
Roy S, Yuzefpolskaya M, Nandakumar R, Colombo PC, Demmer RT. Plasma Trimethylamine-N-oxide and impaired glucose regulation: Results from The Oral Infections, Glucose Intolerance and Insulin Resistance Study (ORIGINS). PLoS ONE. 2020; 15: e227482.
[60]
Liu W, Wang C, Xia Y, Xia W, Liu G, Ren C, et al. Elevated plasma trimethylamine-N-oxide levels are associated with diabetic retinopathy. Acta Diabetologica. 2021; 58: 221–229.
[61]
Miao J, Ling AV, Manthena PV, Gearing ME, Graham MJ, Crooke RM, et al. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nature Communications. 2015; 6: 6498.
[62]
Chen CH, Lai JM, Chou TY, Chen CY, Su LJ, Lee YC, et al. VEGFA upregulates FLJ10540 and modulates migration and invasion of lung cancer via PI3K/AKT pathway. PLoS ONE. 2009; 4: e5052.
[63]
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. Journal of Bioscience and Bioengineering. 2014; 118: 476–481.
[64]
Chen S, Henderson A, Petriello MC, Romano KA, Gearing M, Miao J, et al. Trimethylamine N-Oxide Binds and Activates PERK to Promote Metabolic Dysfunction. Cell Metabolism. 2019; 30: 1141–1151.e5.
[65]
Matsumoto M, Pocai A, Rossetti L, Depinho RA, Accili D. Impaired regulation of hepatic glucose production in mice lacking the forkhead transcription factor Foxo1 in liver. Cell Metabolism. 2007; 6: 208–216.
[66]
Zhang W, Hietakangas V, Wee S, Lim SC, Gunaratne J, Cohen SM. ER stress potentiates insulin resistance through PERK-mediated FOXO phosphorylation. Genes & Development. 2013; 27: 441–449.
[67]
Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015; 65: 1331–1340.
[68]
Ge X, Zheng L, Zhuang R, Yu P, Xu Z, Liu G, et al. The Gut Microbial Metabolite Trimethylamine N-Oxide and Hypertension Risk: A Systematic Review and Dose-Response Meta-analysis. Advances in Nutrition. 2020; 11: 66–76.
[69]
Yan Q, Gu Y, Li X, Yang W, Jia L, Chen C, et al. Alterations of the Gut Microbiome in Hypertension. Frontiers in Cellular and Infection Microbiology. 2017; 7: 381.
[70]
Jaworska K, Huc T, Samborowska E, Dobrowolski L, Bielinska K, Gawlak M, et al. Hypertension in rats is associated with an increased permeability of the colon to TMA, a gut bacteria metabolite. PLoS ONE. 2017; 12: e189310.
[71]
Pelletier CC, Croyal M, Ene L, Aguesse A, Billon-Crossouard S, Krempf M, et al. Elevation of Trimethylamine-N-Oxide in Chronic Kidney Disease: Contribution of Decreased Glomerular Filtration Rate. Toxins. 2019; 11: 635.
[72]
Stubbs JR, House JA, Ocque AJ, Zhang S, Johnson C, Kimber C, et al. Serum Trimethylamine-N-Oxide is Elevated in CKD and Correlates with Coronary Atherosclerosis Burden. Journal of the American Society of Nephrology. 2016; 27: 305–313.
[73]
Zeng Y, Guo M, Fang X, Teng F, Tan X, Li X, et al. Gut Microbiota-Derived Trimethylamine N-Oxide and Kidney Function: a Systematic Review and Meta-Analysis. Advances in Nutrition. 2021; 12: 1286–1304.
[74]
Tan C, Wang H, Gao X, Xu R, Zeng X, Cui Z, et al. Dynamic Changes and Prognostic Value of Gut Microbiota-Dependent Trimethylamine-N-Oxide in Acute Ischemic Stroke. Frontiers in Neurology. 2020; 11: 29.
[75]
Ottiger M, Nickler M, Steuer C, Bernasconi L, Huber A, Christ-Crain M, et al. Gut, microbiota-dependent trimethylamine-N-oxide is associated with long-term all-cause mortality in patients with exacerbated chronic obstructive pulmonary disease. Nutrition. 2018; 45: 135–141.e1.
[76]
Tang WHW, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circulation Research. 2015; 116: 448–455.
[77]
Missailidis C, Hällqvist J, Qureshi AR, Barany P, Heimbürger O, Lindholm B, et al. Serum Trimethylamine-N-Oxide is Strongly Related to Renal Function and Predicts Outcome in Chronic Kidney Disease. PLoS ONE. 2016; 11: e0141738.
[78]
Organ CL, Li Z, Sharp TE, Polhemus DJ, Gupta N, Goodchild TT, et al. Nonlethal Inhibition of Gut Microbial Trimethylamine N‐oxide Production Improves Cardiac Function and Remodeling in a Murine Model of Heart Failure. Journal of the American Heart Association. 2020; 9: e016223.
[79]
Zhang W, Miikeda A, Zuckerman J, Jia X, Charugundla S, Zhou Z, et al. Inhibition of microbiota-dependent TMAO production attenuates chronic kidney disease in mice. Scientific Reports. 2021; 11: 518.
[80]
Bogiatzi C, Gloor G, Allen-Vercoe E, Reid G, Wong RG, Urquhart BL, et al. Metabolic products of the intestinal microbiome and extremes of atherosclerosis. Atherosclerosis. 2018; 273: 91–97.
[81]
Randrianarisoa E, Lehn-Stefan A, Wang X, Hoene M, Peter A, Heinzmann SS, et al. Relationship of Serum Trimethylamine N-Oxide (TMAO) Levels with early Atherosclerosis in Humans. Scientific Reports. 2016; 6: 26745.
[82]
Wu C, Li C, Zhao W, Xie N, Yan F, Lian Y, et al. Elevated trimethylamine N-oxide related to ischemic brain lesions after carotid artery stenting. Neurology. 2018; 90: e1283–e1290.
[83]
Bin Waleed K, Lu Y, Liu Q, Zeng F, Tu H, Wei Y, et al. Association of trimethylamine N-oxide with coronary atherosclerotic burden in patients with non-ST-segment elevation myocardial infarction. Medicine. 2020; 99: e20794.
[84]
Tan Y, Yang S, Chen R, Sheng Z, Zhou P, Liu C, et al. High Plasma Myeloperoxidase is Associated with Plaque Erosion in Patients with ST-Segment Elevation Myocardial Infarction. Journal of Cardiovascular Translational Research. 2020; 13: 908–915.
[85]
Tan Y, Sheng Z, Zhou P, Liu C, Zhao H, Song L, et al. Plasma Trimethylamine N-Oxide as a Novel Biomarker for Plaque Rupture in Patients with ST-Segment–Elevation Myocardial Infarction. Circulation: Cardiovascular Interventions. 2019; 12: e007281.
[86]
Li J, Tan Y, Zhou P, Liu C, Zhao H, Song L, et al. 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. Frontiers in Cardiovascular Medicine. 2021; 8: 628471.
[87]
Uemura S, Ishigami K, Soeda T, Okayama S, Sung JH, Nakagawa H, et al. Thin-cap fibroatheroma and microchannel findings in optical coherence tomography correlate with subsequent progression of coronary atheromatous plaques. European Heart Journal. 2012; 33: 78–85.
[88]
Shimokado A, Matsuo Y, Kubo T, Nishiguchi T, Taruya A, Teraguchi I, et al. In vivo optical coherence tomography imaging and histopathology of healed coronary plaques. Atherosclerosis. 2018; 275: 35–42.
[89]
Sheng Z, Tan Y, Liu C, Zhou P, Li J, Zhou J, et al. Relation of Circulating Trimethylamine N-Oxide with Coronary Atherosclerotic Burden in Patients with ST-segment Elevation Myocardial Infarction. The American Journal of Cardiology. 2019; 123: 894–898.
[90]
Fu Q, Zhao M, Wang D, Hu H, Guo C, Chen W, et al. Coronary Plaque Characterization Assessed by Optical Coherence Tomography and Plasma Trimethylamine-N-oxide Levels in Patients with Coronary Artery Disease. The American Journal of Cardiology. 2016; 118: 1311–1315.
[91]
Li J, Sheng Z, Tan Y, Zhou P, Liu C, Zhao H, et al. Association of plasma trimethylamine N-Oxide level with healed culprit plaques examined by optical coherence tomography in patients with ST-Segment elevation myocardial infarction. Nutrition, Metabolism and Cardiovascular Diseases. 2021; 31: 145–152.
[92]
Tan Y, Zhou J, Liu C, Zhou P, Sheng Z, Li J, et al. Association between Plasma Trimethylamine N-oxide and Neoatherosclerosis in Patients with very Late Stent Thrombosis. Canadian Journal of Cardiology. 2020; 36: 1252–1260.
[93]
Del Rio D, Zimetti F, Caffarra P, Tassotti M, Bernini F, Brighenti F, et al. The Gut Microbial Metabolite Trimethylamine-N-Oxide is Present in Human Cerebrospinal Fluid. Nutrients. 2017; 9: 1053.
[94]
Li D, Ke Y, Zhan R, Liu C, Zhao M, Zeng A, et al. Trimethylamine-N-oxide promotes brain aging and cognitive impairment in mice. Aging Cell. 2018; 17: e12768.
[95]
Nie J, Xie L, Zhao B, Li Y, Qiu B, Zhu F, et al. Serum Trimethylamine N-Oxide Concentration is Positively Associated with first Stroke in Hypertensive Patients. Stroke. 2018; 49: 2021–2028.
[96]
Rexidamu M, Li H, Jin H, Huang J. Serum levels of Trimethylamine-N-oxide in patients with ischemic stroke. Bioscience Reports. 2019; 39: BSR20190515.
[97]
Sun T, Zhang Y, Yin J, Peng X, Zhou L, Huang S, et al. Association of Gut Microbiota-Dependent Metabolite Trimethylamine N-Oxide with first Ischemic Stroke. Journal of Atherosclerosis and Thrombosis. 2020; 28: 320–328.
[98]
Wu C, Xue F, Lian Y, Zhang J, Wu D, Xie N, et al. Relationship between elevated plasma trimethylamine N-oxide levels and increased stroke injury. Neurology. 2020; 94: e667–e677.
[99]
Kirkevold M, Kildal BL, Bronken BA, Kvigne K, Martinsen R, Gabrielsen HE, et al. Promoting psychosocial well-being following stroke: study protocol for a randomized, controlled trial. BMC Psychol. 2018; 6 :12.
[100]
Zhu C, Li G, Lv Z, Li J, Wang X, Kang J, et al. Association of plasma trimethylamine-N-oxide levels with post-stroke cognitive impairment: a 1-year longitudinal study. Neurological Sciences. 2020; 41: 57–63.
[101]
Hou L, Zhang Y, Zheng D, Shi H, Zou C, Zhang H, et al. Increasing trimethylamine N-oxide levels as a predictor of early neurological deterioration in patients with acute ischemic stroke. Neurological Research. 2020; 42: 153–158.
[102]
Zhang J, Wang L, Cai J, Lei A, Liu C, Lin R, et al. Gut microbial metabolite TMAO portends prognosis in acute ischemic stroke. Journal of Neuroimmunology. 2021; 354: 577526.
[103]
Liang Z, Dong Z, Guo M, Shen Z, Yin D, Hu S, et al. Trimethylamine N-oxide as a risk marker for ischemic stroke in patients with atrial fibrillation. Journal of Biochemical and Molecular Toxicology. 2019; 33: e22246.
[104]
Zhai Q, Wang X, Chen C, Tang Y, Wang Y, Tian J, et al. Prognostic Value of Plasma Trimethylamine N-Oxide Levels in Patients with Acute Ischemic Stroke. Cellular and Molecular Neurobiology. 2019; 39: 1201–1206.
[105]
Zhai Q, Sun T, Sun C, Yan L, Wang X, Wang Y, et al. High plasma levels of trimethylamine N-oxide are associated with poor outcome in intracerebral hemorrhage patients. Neurological Sciences. 2021; 42: 1009–1016.
[106]
Su H, Fan S, Zhang L, Qi H. TMAO Aggregates Neurological Damage Following Ischemic Stroke by Promoting Reactive Astrocytosis and Glial Scar Formation via the Smurf2/ALK5 Axis. Frontiers in Cellular Neuroscience. 2021; 15: 569424.
[107]
Senthong V, Wang Z, Fan Y, Wu Y, Hazen SL, Tang WHW. Trimethylamine N-Oxide and Mortality Risk in Patients with Peripheral Artery Disease. Journal of the American Heart Association. 2016; 5: e004237.
[108]
Azab SM, Zamzam A, Syed MH, Abdin R, Qadura M, Britz-McKibbin P. Serum Metabolic Signatures of Chronic Limb-Threatening Ischemia in Patients with Peripheral Artery Disease. Journal of Clinical Medicine. 2020; 9: 1877.
[109]
Roncal C, Martínez-Aguilar E, Orbe J, Ravassa S, Fernandez-Montero A, Saenz-Pipaon G, et al. Trimethylamine-N-Oxide (TMAO) Predicts Cardiovascular Mortality in Peripheral Artery Disease. Scientific Reports. 2019; 9: 15580.
[110]
Fowkes FGR, Rudan D, Rudan I, Aboyans V, Denenberg JO, McDermott MM, et al. Comparison of global estimates of prevalence and risk factors for peripheral artery disease in 2000 and 2010: a systematic review and analysis. Lancet. 2013; 382: 1329–1340.
[111]
Erickson ML, Malin SK, Wang Z, Brown JM, Hazen SL, Kirwan JP. Effects of Lifestyle Intervention on Plasma Trimethylamine N-Oxide in Obese Adults. Nutrients. 2019; 11: 179.
[112]
Liu G, Li J, Li Y, Hu Y, Franke AA, Liang L, et al. Gut microbiota–derived metabolites and risk of coronary artery disease: a prospective study among us men and women. The American Journal of Clinical Nutrition. 2021; 114: 238–247.
[113]
Argyridou S, Davies MJ, Biddle GJH, Bernieh D, Suzuki T, Dawkins NP, et al. Evaluation of an 8-Week Vegan Diet on Plasma Trimethylamine-N-Oxide and Postchallenge Glucose in Adults with Dysglycemia or Obesity. The Journal of Nutrition. 2021; 151: 1844–1853.
[114]
Özogul Y, Kuley Boga E, Özogul F, Ayas D. L-Carnitine Contents in Seafoods Commonly Eaten in Middle Eastern Countries. Journal of Food Biochemistry. 2013; 37: 702–707.
[115]
Chen J, Guo Y, Gui Y, Xu D. Physical exercise, gut, gut microbiota, and atherosclerotic cardiovascular diseases. Lipids in Health and Disease. 2018; 17: 17.
[116]
Zhang H, Meng J, Yu H. Trimethylamine N-oxide Supplementation Abolishes the Cardioprotective Effects of Voluntary Exercise in Mice Fed a Western Diet. Frontiers in Physiology. 2017; 8: 944.
[117]
Chen P, Li S, Koh Y, Wu J, Yang M, Ho C, et al. Oolong Tea Extract and Citrus Peel Polymethoxyflavones Reduce Transformation of l-Carnitine to Trimethylamine-N-Oxide and Decrease Vascular Inflammation in l-Carnitine Feeding Mice. Journal of Agricultural and Food Chemistry. 2019; 67: 7869–7879.
[118]
He Z, Kwek E, Hao W, Zhu H, Liu J, Ma KY, et al. Hawthorn fruit extract reduced trimethylamine-N-oxide (TMAO)-exacerbated atherogenesis in mice via anti-inflammation and anti-oxidation. Nutrition & Metabolism. 2021; 18: 6.
[119]
Chen M, Yi L, Zhang Y, Zhou X, Ran L, Yang J, et al. Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by Regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota. MBio. 2016; 7: e02210–e02215.
[120]
Lv Z, Shan X, Tu Q, Wang J, Chen J, Yang Y. Ginkgolide B treatment regulated intestinal flora to improve high-fat diet induced atherosclerosis in ApoE−/− mice. Biomedicine & Pharmacotherapy. 2021; 134: 111100.
[121]
Din AU, Hassan A, Zhu Y, Yin T, Gregersen H, Wang G. Amelioration of TMAO through probiotics and its potential role in atherosclerosis. Applied Microbiology and Biotechnology. 2019; 103: 9217–9228.
[122]
Pathak P, Helsley RN, Brown AL, Buffa JA, Choucair I, Nemet I, et al. Small molecule inhibition of gut microbial choline trimethylamine lyase activity alters host cholesterol and bile acid metabolism. American Journal of Physiology-Heart and Circulatory Physiology. 2020; 318: H1474–H1486.
[123]
Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation. 2016; 133: 2434–2446.
[124]
Du D, Tang W, Zhou C, Sun X, Wei Z, Zhong J, et al. Fecal Microbiota Transplantation Is a Promising Method to Restore Gut Microbiota Dysbiosis and Relieve Neurological Deficits after Traumatic Brain Injury. Oxidative Medicine and Cellular Longevity. 2021; 2021: 5816837.
Share
Back to top