Next Article in Journal
Acute and Subacute Oral Toxicity of Mumefural, Bioactive Compound Derived from Processed Fruit of Prunus mume Sieb. et Zucc., in ICR Mice
Previous Article in Journal
The Effect of 4′-hydroxy-3,4,5-trimetoxystilbene, the Metabolite of Resveratrol Analogue DMU-212, on Growth, Cell Cycle and Apoptosis in DLD-1 and LOVO Colon Cancer Cell Lines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Trimethylamine N-Oxide in Relation to Cardiometabolic Health—Cause or Effect?

by
Christopher Papandreou
1,
Margret Moré
2 and
Aouatef Bellamine
3,*
1
Institut d’Investigació Sanitària Pere Virgili (IISPV), 43204 Reus, Spain
2
Analyze & Realize GmbH, 13467 Berlin, Germany
3
Lonza LLC, Morristown, NJ 07960, USA
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(5), 1330; https://doi.org/10.3390/nu12051330
Submission received: 14 April 2020 / Revised: 29 April 2020 / Accepted: 4 May 2020 / Published: 7 May 2020
(This article belongs to the Section Nutrition and Metabolism)

Abstract

:
Trimethylamine-N-oxide (TMAO) is generated in a microbial-mammalian co-metabolic pathway mainly from the digestion of meat-containing food and dietary quaternary amines such as phosphatidylcholine, choline, betaine, or L-carnitine. Fish intake provides a direct significant source of TMAO. Human observational studies previously reported a positive relationship between plasma TMAO concentrations and cardiometabolic diseases. Discrepancies and inconsistencies of recent investigations and previous studies questioned the role of TMAO in these diseases. Several animal studies reported neutral or even beneficial effects of TMAO or its precursors in cardiovascular disease model systems, supporting the clinically proven beneficial effects of its precursor, L-carnitine, or a sea-food rich diet (naturally containing TMAO) on cardiometabolic health. In this review, we summarize recent preclinical and epidemiological evidence on the effects of TMAO, in order to shed some light on the role of TMAO in cardiometabolic diseases, particularly as related to the microbiome.

1. Introduction

Cardiovascular diseases (CVD) are the leading cause of mortality and disability, especially in old age [1]. In 2015, the prevalence of type 2 diabetes (T2D) was 8.8% with increasing prevalence rates since 1980 [2] (4.7% in 1980 [3]). Coronary artery disease (CAD), myocardial infarction, stroke, heart failure (HF), atrial fibrillation (AF), and chronic kidney disease (CKD) are known to be closely associated with T2D [4,5], as well atherosclerosis [6], all of which are linked to inflammation [6,7].
Atherosclerosis and T2D can also lead to the development of kidney disease [8,9]. The prevalence of CKD is 5–15% in the adult population (13.6% in the US population [9]) and increases with age [10,11], making it one of the major contributors to the disease network around CVD and metabolic syndrome (Figure 1).
In recent years, there is a mounting body of evidence suggesting that the gut microbiota affects a wide range of systemic conditions, including CVD, T2D, and obesity [12,13], and these conditions also have reverse effects on the gut microbiota (Figure 1).
For instance, there are strong associations of atherosclerosis [14,15] and CKD [16] with dysbiosis. It has been reported that a disturbed proinflammatory microbiome will cause an impaired metabolism of cholesterol and lipids [17] and will lead to a reduction and changes in short-chain fatty acids (SCFAs) [18,19], generated by the gut microbiome. This is further supported by the fact that individuals with metabolic syndrome have an increased Firmicutes/Bacteriodetes ratio, as well as a reduced capacity to metabolize carbohydrates to SCFAs [20]. SCFAs in turn are known to protect from cardiovascular damage by maintaining a proper anti/pro-inflammatory balance [19], possibly by a regulatory T cell-dependent mechanism [21]. In addition, intestinal inflammatory responses (via FABP2-induced intestinal permeability) can aggravate atherosclerotic progress [22]. Dysbiosis also deteriorates CKD by making the lower intestines more permeable to microbiota-generated uremic toxins, which in turn will damage renal tissues [23]. In return, high urea concentrations resulting from CKD modify the gut microbiota to promoteuremic toxin-producing bacteria [23]. Therefore, some microbiota-derived products shift the homeostatic metabolic balance towards a low-grade chronic inflammation, a hallmark of metabolic syndrome [24].
Lately, the gut microbiota-dependent metabolite, trimethylamine N-oxide (TMAO), has received increasing attention for its association with CVD and T2D highlighting the importance of gut microbiome-metabolism in cardiometabolic health.
In diseased populations, elevated plasma TMAO levels have been associated with major adverse CVD events [25] and T2D [26,27]. Furthermore, elevated serum TMAO levels have been proposed as a biomarker of metabolic syndrome [28]. However, in these associations, TMAO has been measured among patients with already established diseases, and thus, it is not possible to assess the causality of increased TMAO levels in the etiopathology of these diseases. More recent prospective observational studies following otherwise healthy subjects but at high cardiovascular risk point to a role of high TMAO plasma levels in preventing the development of T2D, observations supported by animal mechanistic studies.
In addition to the rising number of original scientific literature on TMAO, TMAO has also been subject to extensive reviews, regarding its possible role in conditions such as CVD, T2D, or other aspects of cardiometabolic risk. However, current reviews critically evaluating the cause to effect relationship and the nature of TMAO’s association to cardiometabolic diseases are missing [8]. In the present review, we summarize the evidence from observational studies evaluating the association between TMAO and cardiometabolic diseases. We also discuss the possible mechanisms involved based on animal data and recent longitudinal trials in healthy subjects at risk of developing cardiometabolic diseases.

2. Where Does TMAO Come From?

TMAO is a small odorless molecule (molecular mass of 75.11 g/mol) formed by the oxidation of trimethylamine (TMA) by the hepatic Flavin-containing monooxygenases (e.g., FMO1-FMO5 in human [29]). TMA is generated in the colon by the gut microbiome from dietary quaternary amines (e.g., phosphatidylcholine/choline [30], L-carnitine [31], betaine, choline [32], dimethylglycine, and ergothioneine), if these are not fully absorbed during digestion within the small intestines. Generally, TMA is absorbed into the blood; alternatively, TMA can also be degraded to dimethylamine (DMA), methylamine, and ammonia within the colon (Figure 2).
However, the latter reactions take place to a lower extent: in rats, choline supplementation only resulted in a negligible increase in DMA concentration, with a much higher rise of TMA. Thus, non-absorbed choline was mostly converted to TMA, absorbed into the circulation, and oxidized to TMAO in the liver [32]. In a rare genetic disorder, the TMA → TMAO conversion is impaired, resulting in the accumulation of the volatile TMA and trimethylaminuria (fish odor syndrome) [33,34]. Metabolic retroconversion of TMAO to TMA by the gut microbiota has been demonstrated by Hoyles and colleagues [35]. In humans, TMAO and TMA are usually excreted in urine. Serum TMAO and TMA levels increased in patients with reduced renal function (haemodialysis patients) [36], showing the importance of renal clearance for TMAO/TMA level regulation.
In addition, TMAO itself has been shown to be directly derived from fish and sea food consumption and can be directly absorbed in the gut [40,41]. Therefore, plasma TMAO levels are influenced by TMA formation and its degradation as well as the secretion rate of TMA, DMA, and TMAO. Perhaps it would be more relevant to consider the TMA/TMAO ratio as a marker rather than just TMAO levels.
Other TMAO producers are described below.
L-carnitine is present mainly in red meat and dairy products [42]. The estimated daily intake in a western type of diet is 100–300 mg [43]; however, vegetarians have considerably lower L-carnitine intakes. L-carnitine can also be synthesized in the body from methionine and lysine but only 25% in the body of omnivores comes from endogenous synthesis [43]. Therefore, dietary supplementation with L-carnitine can contribute to dietary intake, particularly among vegetarians and people with chronic diseases associated with aging [44]. In addition, L-carnitine is recommended to be added to infant formulas because infants lack the ability to synthesize their own L-carnitine. An infant fed formula supplemented with L-carnitine normalized lipid levels prevented the development of metabolic and energy disorders [45].
The relative absorption of dietary L-carnitine is 54–87% by the small intestine, depending on the amount provided in a meal, but lower (14–18%) when more L-carnitine is ingested as a supplement (usually in larger amounts), pointing to a saturable transporter system [46]. The non-absorbed L-carnitine passes on to be fermented by the microbiome (Figure 2).
Independent from its role in producing TMAO [47,48], L-carnitine plays a crucial part in energy generation by its key role as a shuttle of medium and long chain fatty acids into mitochondria, where fatty acids are metabolized by β-oxidation to produce energy in all tissues, including the heart [49]. The healthy heart uses 50–70% of its energy from fatty acids [50]. Therefore, L-carnitine is essential for normal heart functioning [50,51]. In addition, L-carnitine supplementation reduces oxidative stress, inflammation, and necrosis in many tissues including the cardiac tissue [52]. Other roles of L-carnitine include calcium influx regulation, endothelial function, intracellular enzyme release, and membrane phospholipid composition [52]. L-carnitine supplementation was shown to normalize plasma lipid levels in patients with coronary heart disease [53]. In models of brain injury, L-carnitine improved energy metabolism, decreased oxidative stress, and prevented subsequent cell death [54], or provided tissue protection in ischemic stroke models [55]. In patients with acute myocardial infarction, L-carnitine improved survival by reducing infarct size, ventricular arrhythmias, left ventricular dilation, and heart failure (HF) incidence [56]. L-carnitine has also been associated with better insulin sensitivity in diabetics, insulin-mediated glucose uptake and oxidation in normoglycemic subjects [57], and lower risk of T2D in the PREDIMED (Prevención con Dieta Mediterránea) study [58]. The safety of L-carnitine has been demonstrated in many studies. There is no regulatory limitation on the dosage of L-carnitine by the European Food Safety Authority (EFSA), which considers L-carnitine well characterized [45]. The Spanish Agency for Food Safety and Nutrition limits L-carnitine or L-carnitine hydrochloride supplementation to 2000 mg per day and L-carnitine tartrate supplementation to 3000 mg per day [59].
Choline is found in beef/chicken liver, eggs, wheat germ, bacon, and soybeans among other foods [60]. The Food and Nutrition Board of the Institute of Medicine in the USA has established the daily adequate intake to be 550 mg for men and 425 mg for women [61]. The European Food Safety Authority defined a choline daily adequate intake of 400 mg for adults [62].
Choline is an essential molecule, which functions as a precursor for the synthesis of phospholipids, including phosphatidylcholine, a membrane building block and a precursor of the neurotransmitter acetylcholine. Thus, choline is particularly important for fetal brain development and maintenance of neurological function [63]. In adults, high intake of choline was associated with more efficient neural processing among overweight and obese individuals [64]. Furthermore, EFSA concluded a cause and effect relationship has been established between the consumption of choline and the maintenance of normal lipid metabolism, normal liver function, and normal homocysteine metabolism [65]. Phosphatidylcholine constitutes up to 40% of bile composition [66]. Free choline is re-absorbed within the small intestine [67], and only excess amounts are metabolized by the gut microbiome to produce TMA in the colon [32,39] (Figure 2). The single intake of up to 6 egg yolks (1 egg yolk equivalent to 119 mg choline) in 6 healthy volunteers resulted in high variability of post-prandial TMAO levels at 8 h (2–20 µM), returning to baseline values after 24 h [68]. In a study with 30 healthy males, fasting plasma TMAO concentrations were not affected after 4 weeks of ingestion of 400 mg/d choline, either via eggs (3 eggs/day) or choline supplementation [69]. Similarly, the intake of (phosphatidylcholine-rich) krill oil did not affect TMAO concentrations [70]. In a different study, consumption of up to 3 eggs/day for 4 weeks by 38 subjects beneficially increased high density lipoprotein cholesterol (HDL-c), reduced the low density lipoprotein cholesterol (LDL-c)/HDL-c ratio, and increased plasma choline, without changing fasting plasma TMAO concentrations [71]. Elevated circulating choline levels have been associated with CVD in some cross-sectional [72] and prospective studies [73]. The PREDIMED cohort reported positive associations between plasma choline and increased risk of major cardiovascular events [74]. In a recent cross-sectional study of 49 atrial fibrillation (AF) patients, plasma choline concentrations were higher than normal [75]. Results from 3 prospective cohort studies are also consistent with respect to the association between plasma choline and AF incidence [76]. A disruption of mitochondrial choline oxidation to betaine as part of mitochondrial dysfunction may precede the development of cardiovascular diseases. Oxidative stress and inflammation may impair betaine-homocysteinemethyltransferase activity, resulting in accumulation of circulating choline [77]. The relationship between circulating choline and T2D has been scarcely examined. Among diabetics a trend towards higher plasma choline was observed [78]; however, more recently, the PREDIMED study did not find any significant association [58].
Betaine is found in wheat bran, wheat germ, and spinach [60], and the usual intake is 100–400 mg/day [79]. Interestingly, increased plasma betaine levels were shown to be associated with lower risk of CVD [74]. However, recently, higher plasma concentrations of betaine were also associated with increased risk of AF [76]. Furthermore, two previous studies found that elevated plasma betaine concentrations were independently associated with HF risk [73,80]. Plasma betaine concentrations were reported to be reduced in insulin-resistant humans and were associated with insulin resistance [81] and a lower risk of T2D in a healthy Mediterranean population at high risk of developing CVD [58]. Even though betaine may be linked to TMAO production (Figure 2 and [37]), the single ingestion of 1760 mg betaine did not lead to an increase in urinary TMAO excretion in an experimental model [41]. Betaine appears to be a poor precursor for TMAO, as it has been shown to produce 100-fold less TMAO than choline [82].
Dimethylglycine is found in low levels in cereals, seeds, beans, and liver. Dimethylglycine is formed from betaine during the re-methylation of homocysteine to methionine by betaine-homocysteine methyltransferase (BHMT) [73] and is normally excreted in the urine or metabolized to sarcosine [83]. Dysregulation in dimethylglycine metabolism may lead to its accumulation in the plasma, which would inhibit BHMT activity, possibly causing an elevation in homocysteine concentrations [84]. Higher plasma concentrations of dimethylglycine were previously found to be associated with increased risk of acute myocardial infarction [85] and HF incidence in patients with CVD [73].
Ergothioneine is contained in mushrooms (bolete, oyster), kidney, liver, black beans, red beans, and oat bran [86]. It was found to have antioxidant and cytoprotectant properties [87] and was shown to be associated with a lower risk of cardiometabolic morbidity and mortality [88]. It is part of the biochemical network leading to TMA production, however its contribution to plasma TMA/TMAO levels requires further investigation [37].
Fish and seafood contain high amounts of TMAO, e.g., 311 mg per 100 g cod fillet [40]. TMAO has been shown to be directly absorbed by the intestine [37]. Similar to an acute supplementation of 2100 mg choline, 2970 mg L-carnitine, or 1670 mg TMAO by healthy volunteers, sea food consumption resulted in an increase in postprandial urinary excretion of TMA and TMAO after 24 h [41]. In a study with 40 healthy young men, consumption of meals producing TMAO provided directly from fish led to a significant increase in postprandial plasma TMA and TMAO levels, peaking at around 0.2 µM and 150 µM, respectively, 2 h post meals. However, egg or beef consumption, providing choline and L-carnitine, respectively, resulted only in negligible TMA/TMAO increases [40]. The difference between the studies could be due to different levels of L-carnitine and choline provided by supplementation or in the diet. TMAO has a plasma half-life of 4 h [40]. As is the case for plasma TMAO, urinary TMAO levels are also variable and observed in particular with high fish intake in humans [89]. In a human study with radiolabeled TMA or TMAO, 94.5% of the dose was excreted in the urine within 24 h, 4% of the dose was excreted in the faeces, and <1% in the breath [90]. In 20 Swedish men, high daily fish consumption elevated urinary TMA and TMAO levels to 47 (8.5–290) mmol TMAO/mol creatinine vs. 38 and 34 mmol TMAO/mol creatinine in non-fish consumers [91]. However, in most publications, only serum TMAO levels are considered as a marker.
Red meat contains both choline (138 mg in 100 g beefsteak) [92] and L-carnitine (49–144 mg in 100 g beefsteak) [93,94]. A diet high in red meat (258 mg L-carnitine and 573 mg choline per day), consumed for 4 weeks by 113 heathy omnivore volunteers, increased plasma TMAO levels from a baseline median level of 3.5 µM TMAO to 7 µM. Isotope tracing experiments revealed that TMAO was primarily generated from the L-carnitine; in addition, meat consumption led to a slight decrease in fractional renal TMAO excretion, possibly explaining in part the TMAO increase [95]. In a recent study, the Atkins diet, rich in red meat, increased plasma TMAO levels from 1.8 µM to 3.3 µM [96]. However, single consumption of 227 g of beef resulted in a 60-fold lower urinary TMA+TMAO excretion than 227 g of herring [41]. Furthermore, single consumption of 170 g of beefsteak only resulted in a negligible postprandial mean plasma TMAO increase, peaking 1 h after consumption [40].

3. Physiological Role/Effects of TMAO

In fish, TMAO functions as an osmolyte by supporting protein and nucleic acid stability by entropic and enthalpic mechanisms in the presence of high urea concentrations. This leads to maintaining cell volume under conditions of high osmotic urea and hydrostatic pressure stresses, which is especially important for the survival of the deep sea animals [97,98,99,100]. TMAO acts as a molecular chaperon and can also be used as such experimentally [101].
A mixture of osmolytes, including betaine and glycerophosphorylcholine, also counters the deleterious effects of highly concentrated urea and salt in mammalian kidneys [102]. If present at sufficient levels, TMAO and/or L-carnitine may support this process. L-carnitine (as well as betaine and erythritol) was also found to be an osmoprotectant and anti-inflammatory for human corneal epithelial cells [103].

3.1. TMAO Variability

Although the relationship between elevated TMAO and CVD/T2D is supported by several observational studies, TMAO concentrations were found to be highly variable. In a study including 100 healthy volunteers (median age: 63.0 years), plasma TMAO was measured at 2 time points one year apart. At both measurements TMAO varied within a range of 2–26 µM, although most values were within 2–8 µM. There was a greater within-person than between-person variability, making TMAO rather unsuitable as marker in epidemiological long-term studies [104]. A high plasma and urine TMAO variability was also reported in a study with overweight diabetic patients (median age 60 years), where TMAO concentrations as well as other osmolyte levels such as betaine were monitored four times at 6-month intervals over 2 years [105]. Thus, due to the observed intra-individual variability, increased TMAO may only serve as an indicator for associations with cardiometabolic diseases.

3.1.1. TMAO Variability with Age and Sex

Older subjects were found to have high levels of TMAO. In a study comparing 168 young, 118 middle aged and 141 elderly adults, mean TMAO levels were around 2.5 µM, 4.8 µM and 10 µM, respectively. However, inter-individual variability was also observed within these populations [106]. Animal testing reported similar observations in old mice [107].
Gender differences may play a role in the observed TMAO levels and has been reported between men and women. Indeed, differences in TMAO levels are possibly due to differences in food consumption patterns levels [108]. However, the gender effect was not reported by others [29]. Animal models reported effects of gender on TMAO levels related to differences in the FMO expression. This is discussed in detail in the following paragraph.

3.1.2. TMAO Variability through FMO

Some of the variability of TMAO in plasma levels was shown to be derived from inter-individual variability in FMO liver enzyme regulation [109]. FMO activity appears to vary with FMO isoform [110,111], as well as age [110,112]. Furthermore, menstrual cycle-dependent variations in TMA/TMAO occur in women, reflecting perhaps a hormonal-dependent FMO gene regulation, suggesting the possible function of TMA as a pheromone [34]. In addition, sex-specific differences in TMAO concentrations were reported in mice [110,113,114,115]. Thus, when both females and males are included, it is important to stratify the analysis according to sex, as female rats do have a significantly higher FMO3 expression.
In mice, FMO3 was identified as a central integrator of hepatic cholesterol and triacylglycerol metabolism, inflammation, and endoplasmic reticulum (ER) stress [116]. Knockdown of FMO3 in insulin-resistant mice prevented the development of hyperglycemia, hyperlipidemia and atherosclerosis [117], and reduced TMAO levels from 7 µM to 2.5 µM. A similar knockdown of FMO3 in LDL receptor knockout mice decreased TMAO levels only from 3.2 µM to 2.2 µM, while still affecting lipid and glucose metabolism [118]. This suggests that FMO3 may have a role in metabolic pathways and disease progression independently from TMAO [118]. Furthermore, it has been reported that FMO enzymes are involved in steroid hormone synthesis, important in metabolic disease etiology [119].
In a different study, FMO3-deletion protected choline supplemented LDL-receptor-/- mice from obesity, while reducing TMAO levels from 2.5 µM to 0.4 µM [120]. Interestingly, in various different FMO deficient mice (fmo1-/-, 2-/-, 4-/-, 5-/-), there was no correlation between atherosclerosis and either TMAO production or urinary TMAO concentration. The plasma total cholesterol concentration was negatively, but weakly, correlated with TMAO levels [110].
Variability in FMO3 expression was reported in patients with CKD, leading possibly to a variability in renal clearance of TMAO [121]. These effects could be the results of activation in FMOs elicited by octylamine and uremic serum [122]. Lower clearance in TMAO coincided with increased FMO3 expression in a CKD mouse model [123]. High plasma TMAO levels in CKD patients may contribute to the observed damage in blood vessels [122]. In pediatric CKD patients, plasma TMAO, DMA, and TMA levels were also increased, while the level of these compounds was decreased in the urine [124]. The microbiome was found to be altered in children with CKD where Cyanobacteria, Subdoligranulum, Faecalibacterium, Ruminococcus, and Akkermansia abundance were affected [124]. In a different study with CKD children, the Prevotella genus was reduced, while Lactobacillus and Bifidobacterium were increased [125,126]. More recently, Rath and colleagues reported the role of diet and age in the abundance of the TMA-producing bacteria and how diet and supplementation can modulate the abundance of the microbiota leading to the formation of TMA [127]. In particular, bacteria from Clostridium XIVa and Eubacterium sp., containing choline TMA-lyase genes, and Gammaproteobacteria, containing L-carnitine oxygenase genes, have been identified in the gut microbiota [128].

3.1.3. TMAO Variability by Diet

The influence of diet on TMAO levels has been examined in several studies. A diet with lower carbohydrate and high resistant starch content was associated with slightly higher plasma TMAO levels [129]. Interestingly, a paleolithic diet rich in whole grains also led to lower serum TMAO and abundance of TMA producer Hungatella [130]. In 115 healthy people at increased risk of developing colon cancer, a Mediterranean (moderate in fish) diet did not cause significant changes in the (fasting) TMAO concentrations after 6 months of supplementation [131]. Daily red meat consumption (providing 258 mg L-carnitine/day) [95] caused elevations in TMAO median levels of around 7 µM (interquartile range 4–11 µM). However, non-red meat, egg, or fish consumption in 271 subjects was not associated with elevated fasting TMAO, choline, or betaine concentrations, despite a slightly positive association between dairy food consumption and plasma TMAO concentrations [132]. Furthermore, in a 2-year study with 100 healthy volunteers, there were no associations between TMAO and the consumption of animal food (low fish-intake population); however, slightly higher TMAO levels (and lower levels of betaine and choline) were observed in prebiotics consumers [104].
Depending on the precursors, different chemical intermediates may be formed by the gut microbiome similarly to TMA (Figure 2). For instance, L-carnitine was found to be converted to gamma-butyrobetaine by several commensal bacteria; gamma-butyrobetaine in turn was found to be converted to TMA by a strain of the Clostridiales. The second conversion step took place at a low rate in omnivorous humans (n = 40) and at a very low rate in vegans/vegetarians (n = 32) [133], perhaps linking diet to microbiota differences and TMA formation. Previously, similar results were published showing that following a 250 mg L-carnitine challenge, omnivores had significantly higher plasma TMAO concentration, peaking at 14 µM 8 h post ingestion (n = 5) compared to vegans/vegetarians, slowly ascending to 1 µM 24 h after ingestion (n = 5) [94]. However, when larger populations are studied, fasting TMAO levels were about 2 µM in vegans/vegetarians (n = 26) and almost 3 µM in omnivores (n = 51) with the upper 95 percentiles of 4.5 µM in vegans/vegetarians and 7 µM in omnivores [94]. It is believed that vegetarians/vegans adapted to low-carnitine diets have a higher L-carnitine bioavailability than carnivores, because dietary L-carnitine is more efficiently absorbed [134] and therefore less available to the microbiome.
Altogether, dietary precursors would have short-term influence on postprandial TMAO levels but would not lead to lasting elevated fasting TMAO levels.

3.1.4. TMAO Variability by Microbiome Variability

As discussed, the microbiome is the central determinant for TMAO generation [135]. The TMAO precursor, TMA, is generated microbiologically within the intestine, and antibiotics were shown to suppress TMAO formation [94,115,136]. In human subjects (n = 5) ingestion of 250 mg radiolabeled L-carnitine together with a 227 g steak (providing an additional 180 mg L-carnitine [94]), resulted only in a modest TMAO increase in the plasma (2.5 µM TMAO 24 h post ingestion) and urine. If the L-carnitine challenge was repeated following microbial suppression using antibiotics, no TMAO could be detected. Three weeks later, the same L-carnitine challenge produced 12 µM labelled TMAO at 24 h from a (possibly still dysbiotic [137]) repopulated microbiome [94]. Gnotobiotic mouse studies showed that TMAO accumulates in the serum of mice colonized with TMA-producing species [138]. These results suggest that diet and in particular supplementation with TMA precursors can modulate the microbiome toward proliferation of TMA-producing bacteria.
In 40 healthy young men, the dietary precursors choline from eggs and L-carnitine from beef were converted to TMAO more efficiently in individuals with a higher Firmicutes to Bacteroidetes ratio and less gut microbiota diversity, demonstrating that TMAO production is a function of individual differences in the gut microbiome. Interestingly, baseline TMAO concentrations were 3.4 µM for both low (n = 15) and high (n = 11) TMAO producers [40], pointing again to individual differences in the microbiota.
An oral L-carnitine challenge test was designed to identify individuals with a TMAO-producer phenotype of their gut microbiota [139]. This also puts emphasis on the microbial composition of the microbiota, relevant for TMAO production and levels.
In a more pronounced and harmful way dysbiosis can also alter the microbiome. Dysbiosis is part of the disease network including cardiometabolic diseases (Figure 1). Dysbiosis in turn may alter TMAO levels, by either decreasing or increasing TMA producing strains within the microbiome. In some cases, TMA-producing bacteria are somehow reduced by dysbiosis [140,141,142]. In other cases, it has been shown that increased TMAO correlates with a dysbiotic microbiome [143,144]. Dysbiosis is known to be triggered by factors like an unhealthy diet, especially a high-animal fat diet [12]. However, several different factors can contribute to dysbiosis including the cardiometabolic disease network (Figure 1). Furthermore, dysbiosis contributes to the progression of CVDs by promoting major CVD risk factors: atherosclerosis and hypertension [12]. Dysbiosis has been shown to promote kidney disease, since the intestinal barrier becomes more permeable for microbially generated metabolites [145,146,147]. Relative abundance of Akkermansiamucinophilia in colon biopsies was shown to be inversely correlated with TMAO levels [131]. A. mucinophilia is a mucotroph bacterium and an indicator of a functioning intestinal mucus layer [126,148].
Altogether, the amount of microbial TMA formation largely depends on the composition of the intestinal flora as well as the state of the mucus layer, both of which can be influenced by several factors, including but not limited to diet and disease status (Figure 1).
Increases in TMAO levels resulting from TMA increases are plausible but provide no statement regarding any effects on health or disease by TMAO.
So far, only associations from observational studies have been described. In order to distinguish cause and effects, intervention and mechanistic studies are needed.

4. TMAO and Cardiometabolic Diseases

4.1. TMAO Levels in Humans Associated with CVD and T2D

Due to the associations of elevated TMAO levels with cardiovascular [136] or metabolic outcomes in diseased populations, TMAO has been used as a disease biomarker [28,38,149]. In this section the information is also summarized as a table (Table 1).
In a cross-sectional study, elevated plasma TMAO levels were found in 50 subjects with cardiovascular complications (heart attack, myocardial infarction, MI, stroke, or death), as compared to subjects (n = 50) without complications [115]. These results were confirmed in a cohort of 1876 subjects [115]. In 2595 subjects undergoing cardiac evaluation, plasma TMAO levels above the median of 4.6 µM predicted an increased CVD risk; elevated plasma dimethylglycine levels, a metabolite of choline, rose modestly but significantly from 4.29 to 4.5 µM with increased CVD risk prediction [94]. Within a population of 3903 subjects undergoing elective diagnostic coronary angiography, the median TMAO level was 3.7 µM (interquartile ranges 2.4–6.2 µM). High plasma choline and betaine levels were associated with higher risk of mortality, MI, or stroke with concomitant increase in TMAO in 3903 sequential stable subjects undergoing elective diagnostic coronary angiography [82]. In a prospective study of 622 hypertensive stroke patients and 622 matched controls, the serum TMAO level in the highest tertile was ≥3.19 μM/L, while in the lowest tertile it was <1.79 μM; the high TMAO levels were associated with increased risk of stroke [160].
A recent systematic review and meta-analysis of 19 prospective studies (n = 19,256, including 3315 incident CVD cases) concluded that elevated TMAO concentrations were associated with a 62% increased risk for all-cause mortality. However, individuals with elevated concentrations of TMAO precursors (L-carnitine, choline, or betaine) had only 1.3 to 1.4 times higher risk for major adverse cardiovascular disease events compared to those with low concentrations [169].
Another meta-analysis of 11 prospective studies with a total of 10,245 patients found that elevated circulating TMAO was associated with a 23% higher risk of cardiovascular events and 55% higher risk of all-cause mortality. Mean elevated TMAO levels ranged from 4.1 µM to 72.2 µM. Control subjects with the lowest risk had levels ranging from 2.3 to 32.2 µM [170]. When studies with missing control values and unusually high control values (27.5 and 32.2 µM) were omitted, TMAO levels [80,136,153,158] in the remaining 5799 patients ranged between 4.1 µM and 12.0 µM (mean 6.8 µM) for high-risk patients, and between 2.3 µM and 2.9 µM (mean 2.6 µM) for low-risk patients.
Recently, a prospective study investigated the relationship between 10-year changes in plasma TMAO levels and coronary heart disease incidence among 760 healthy women. Increases in TMAO levels during the follow-up were associated with higher coronary heart disease risk, and this relationship was strengthened by unhealthy dietary patterns and attenuated by healthy dietary patterns [171]. This points again to an effect of the microbiome independent from TMAO metabolism.
In a study with 435 patients undergoing elective cardiac risk factor evaluation, plasma TMAO concentrations were higher in subjects with T2D (around 4.9 µM) compared to subjects without T2D (3.3 µM TMAO), with high variability, however, between subjects [120]. In 353 CAD patients, increased plasma TMAO levels (median 5.5 µM; interquartile range 3.4 to 9.8 µM) were associated with a higher SYNTAX score (synergy between percutaneous coronary intervention with taxus and cardiac surgery; a measure of disease severity), but TMAO levels were not associated with subclinical myonecrosis [154].
In 283 subjects with a mean age of 66.7 years, T2D was associated with a higher plasma TMAO concentration (8.6 ±12.2 µM compared with 5.4 ±5.2 μM) [152]. In a case-control study with a cross-sectional design, a positive association between plasma TMAO concentrations and gestational diabetes mellitus was observed [172], where an inverse association was found in a recent prospective cohort [173]. Both studies were conducted among Chinese pregnant women. In a cross-sectional study with 137 adults, serum TMAO levels increased along with body mass index, visceral adiposity index, and fatty liver index; serum TMAO ≥8.74 µM was considered as predictive of metabolic syndrome [28]. In a recent prospective study with 300 T2D-free adults, a nonlinear association between baseline plasma TMAO and prevalent prediabetes was reported [174].
In 859 venous thromboembolism patients, TMAO levels showed a U-shaped association with mortality, with optimum levels around 4 µM [162]; however, TMAO was not associated with bleeding events in 81 patients with stable angina [175].
In a cross-sectional study with 271 healthy adults, plasma TMAO concentrations were positively associated with inflammation—participants in the top TMAO quartile (3.13 μM) had higher plasma concentrations of TNF-α, sTNF-R p55, and sTNF-R p75 than participants in the bottom quartile (2.08 μM) [132]. Converse results were found in a case-cohort study with development of 251 incident T2D cases in a random sample of 694 participants within the PREDIMED trial. These subjects were healthy and T2D free at baseline. After adjustment for recognized T2D risk factors, individuals with elevated plasma TMAO levels at baseline had a lower risk of T2D [58].
In another recent cross-sectional study using data collected from 1653 adults, higher plasma TMAO concentrations were associated with an adverse cardiometabolic risk profile and with a number of TMA-producing bacterial taxa (associations of plasma trimethylamine N-oxide, choline, carnitine, and betaine with inflammatory and cardiometabolic risk biomarkers and the fecal microbiome in the Multiethnic Cohort Adiposity Phenotype Study.) [176].
A recent study suggested an association between TMA rather than TMAO and cardiovascular diseases. In patients with severe aortic stenosis, TMAO levels were 5.5 µM (controls 3.6 µM); at the same time, TMA levels were 59.5 µM (controls 23.2 µM) [161]. TMA but not TMAO was also found to be associated with increased risk of gestational diabetes [173].
Altogether, one can conclude from the above literature that discrepancies are reported from different observational studies.
TMAO elevation may be just a feedback mechanism to counter disease progression in light of its role as a molecular chaperone supporting proper protein folding (see above) and other potential beneficial roles [177], as will be discussed later in this review. In addition, the microbiome involved in the metabolism of TMAO can play an independent role in disease progression, making TMAO a marker and not an inducer of disease.

4.2. TMAO in Patients with Compromised Renal Function

TMAO and TMA are known as microbiota-generated molecules, along with p-cresyl sulfate, indoxyl sulfate, and indole-3 acetic acid [161,178,179]. Absorbed from the intestine into the blood stream or metabolized by the liver, TMA and TMAO may accumulate if not excreted properly in patients with compromised renal function.
Along this line, a recent review suggested that elevated TMAO in the circulation could reflect T2D or atherosclerosis mediated renal dysfunction, which reduces TMAO excretion, thereby elevating TMAO plasma concentrations [8]. This suggests that TMAO may be a marker of disease, but not a causative factor.
It has been reported that diabetic CKD patients (n = 20) had an increase in TMA-producing bacteria leading to elevated plasma TMAO levels (1.516 µg/mL = 20.2 µM) compared to control subjects (0.183 µg/mL = 2.4 µM) [167]. In another study with 521 CKD patients, the median TMAO level was 7.9 µM (interquartile range, 5.2–12.4 μM), compared to a median of 3.4 µM TMAO in 3166 control subjects. In hemodialysis patients (with more severely impaired kidney function) TMAO levels reached 77 ± 26 µM, compared to 2 ± 1 µM in control subjects [163].
More studies are needed to fully understand the TMAO variability in this disease. It is possible that individual variability or disease severity in these patients cause variable TMAO levels. This is supported by the following observation: only a few patients with severe renal disorders (hemodialysis patients [163]; see Figure 1) have extremely high TMAO levels.

4.3. Cause to Effects

A recent bi-directional Mendelian randomization analysis examined the causal direction between gut microbiota-dependent TMAO, or its precursors, and cardiometabolic diseases such as risk of T2D, CAD, MI, stroke, AF, and CKD. Genome-wide association with TMAO, L-carnitine, choline, and betaine levels, and individual single nucleotide polymorphisms (SNP), allowed to the nature of these associations to be identified. Genetically predicted higher TMAO and L-carnitine were not associated with higher odds of T2D, AF, CAD, MI, stroke, and CKD after Bonferroni correction for multiple comparisons (p < 0.0005). However, higher choline was associated with an increased risk of T2D while higher betaine was associated with a decreased risk. In addition, it was reported that T2D and CKD were causally and directionally associated with higher TMAO levels, suggesting that these diseases are the cause of plasma TMAO elevation and supporting the feedback mechanism of TMAO increase as a result of disease [180]. This is supported by preventive animal studies described below.
For kidney disease, the lower excretion of TMAO by affected patients explains higher TMAO levels. In addition, CKD progression goes along with an increased gut permeability, which in turn sets free an overload of microbiota-generated compounds, including TMAO [178,181].
Altogether, the potential role of TMAO in cardiometabolic diseases, as previously proposed [94,115,169,170], requires further evaluation—as done below.

5. TMAO in Experimental Models

5.1. TMAO in Cell Culture

Controversial data have been reported in cell culture experiments. With high TMAO concentrations, inflammatory signals and cellular damage were observed. Human aortic endothelial cells had proinflammatory responses to extremely high levels of TMAO (200 µM), including the induction of COX-2, E-Sel, ICAM-1, and IL-6 and activation of mitogen-activated protein kinase and nuclear factor-κB (NF-κB) pathways [182].
In human platelets, the most pronounced changes in thrombin occurred with 100 µM TMAO, lower concentrations were less effective [183]. In cultured carotid artery endothelial cells, 30 µM TMAO increased caspase 1 activity, enhanced IL-1β production, and led to inflammasome formation [184]. Endothelial progenitor cells cultured with 200 and 500 µM TMAO promoted cellular inflammation, oxidative stress, and suppressed cellular functions, whereas 2 µM or 100 µM TMAO did not have such effects [175]. In a dose-response experiment on macrophages, TMAO up to 100 µM had no effect on inducing foam cell formation, the first steps in atherosclerosis development [185]. Furthermore, in isolated rat cardiomyocytes, 10 or 100 µM TMAO applied for 1 to 24 h did not affect cell viability, sarcomere length, intracellular reactive oxygen species (ROS), and mitochondrial membrane potential. Furthermore, the simultaneous treatment with TMAO and known cardiac insults, such as H2O2 or doxorubicin, did not enhance the treatment’s effects [186].
A recent study evaluated the effects of TMA and TMAO on rat cardiomyocytes. TMA at 1000 µM showed more adverse effects than TMAO at 100.000 µM. The concomitant treatment with TMA (1000 µM) and TMAO (100.000 µM) protected cardiomyocytes against the deleterious effects (cell shrinkage, detachment) of TMA [161]. These experiments have to be assessed with caution as all unfavorable effects were observed with TMAO concentrations well beyond the concentration range observed in humans affected by CVD, T2D, or other aspects of the disease network. It is unclear how TMAO mediates these effects. Recently, TMAO was found to bind to endoplasmic reticulum stress kinase PERK (EIF2AK3), selectively activating the PERK branch of the unfolded protein response. This induced the activation of the transcription factor FoxO1, which has been described to suppress lipogenesis [187].
Currently, reliable proof for any effects at physiological or slightly elevated levels (e.g., up to 12 µM) of TMAO as a direct cause or as a risk factor of cardiac damage at the cellular level is missing. On the contrary, TMAO may have beneficial effects, as described above.

5.2. TMAO in Animal Studies

The pieces of evidence for effects of TMAO or its precursors in animal experiments are summarized in Table 2 (unfavorable effects) and Table 3 (neutral or beneficial effects).
However, several factors including experimental design (intervention vs. prevention), diet, gender, and non-physiological high concentrations have led to contradictory results, similar to the culture experiments above. In addition, not all test systems are equally well suited to examine a possible role of TMAO in disease formation or prevention: several animal models may increase the frequency of disease symptoms by mutations or other distress to the animals in order to increase the occurrence of measurable events.
The majority of data were collected in the apoE−/− mouse model (apolipoprotein E-knockout). ApoE is a multifunctional protein with central roles in lipid metabolism; it transports lipids, including cholesterol, through the cerebrospinal fluid and plasma. Unlike humans, mice lack the cholesteryl ester transfer protein (CETP), which also transfers cholesterol ester from HDL-c to LDL-c particles. Normal mice have the majority of circulating cholesterol being carried by HDL particles, so they never develop atherosclerotic lesions on a chow diet; this is changed in the apoE−/− mouse model, where the ApoE protein is inactivated, which leads to enrichment in very low-density cholesterol (VLDL-c) [210]. Thus apoE−/− mice develop atherosclerotic lesions on a chow (plus fat) diet due to the resulting pro-atherogenic lipoprotein profile. Lesion formation collected in apoE−/− mice was previously considered to originate from increased oxidative stress [114]. Due the lack of CETP and other human-mouse differences, results obtained from apoE−/− should be interpreted carefully. A mouse strain with transgenes for two dyslipidemia-inducing mutations, the human apolipoprotein E-Leiden (APOE-Leiden), and human cholesteryl ester transfer protein (CETP) [211] may be more close to the human situation. Interestingly, when the E-Leiden/CETP mouse model is used, TMAO showed protective effects in preventing atherosclerosis development [185].
In a study of germ free apoE−/− mice on a high-fat Western-type diet, atherosclerosis developed, with no measurable difference to animals raised conventionally (with germs) [212]. However, if a similar experiment was done in germ free apoE−/− mice on regular chow, differences in atherosclerotic plaques between germ-free and conventionally reared apoE−/− mice became apparent. The latter group had fewer plaques due to a protective effect of the microbiota [213].
In some studies, a structural analogue of choline, 3,3-dimethyl-1-butanol (DMB), was used. DMB inhibits TMA formation from cultured microbes or physiologic polymicrobial cultures as well as distinct microbial TMA lyases concurrently with inhibition of foam cell formation and atherosclerotic lesion development. This lead to the conclusion that DMB can be used as a treatment for atherosclerosis [214]. In addition, DMB was shown to prevent hypertension in experimental animals [215].

5.2.1. Unfavorable Effects of TMAO in Animals

In several nonclinical settings (see Table 2), high TMAO levels (25 to 400 µM) were associated with CVD symptoms, e.g., atherosclerotic plaques. Control groups had fewer CVD symptoms and lower TMAO levels. However, in most of these studies, experimental animals were severely affected by disease, old age, and/or other factors. For instance, animals with partially ligated carotid artery/coronary ligation [184,191,193] are ill, in pain, and stressed from the operation; this procedure may severely perturb the system and induce dysbiosis. Interestingly, severe cardiovascular stress caused by coronary ligation increased TMAO levels up to 30 µM in rats; however, sham operations also caused slightly higher than baseline TMAO levels (10 µM), likely due to postoperative dysbiosis [193].
In most animal experiments, high TMAO (0.12–0.3% of chow/drinking water) or L-carnitine or choline supplementation (≥1%) resulted in plasma levels between 30 and 100 µM [94,115,165,182,191,193], in some cases up to 400 µM TMAO [94,192], and many of these studies were conducted in female mice, which express high FMO3 and give rise to high TMAO concentrations in plasma. It should be noted that 1.3% L-carnitine in the diet of a mouse approximately corresponds to 2080 mg L-carnitine/kg (see calculation in the footnote of Table 2), which is an excessive dose, corresponding to a human equivalent dose of 11,818 mg L-carnitine/day [185]. Acute high TMAO (achieving 91 µM TMAO) injection experiments increased thrombus formation in a carotid artery injury mouse model [183]. TMAO injection in LDL-receptor deficient mice (86 µM plasma concentration) induced increased levels of inflammatory markers and activated the mitogen-activated protein kinase, extracellular signal-related kinase, and nuclear factor-κB signaling cascade [182]. The relevance of these results for healthy humans, or even those affected by CVD or metabolic syndrome, remain limited.
In apoE−/− mice, chronic dietary L-carnitine supplementation (1.3%), corresponding to 2080 mg L-carnitine/kg, altered microbial composition and increased aortic root atherosclerotic lesions by 1.8-fold, while enhancing TMA/TMAO synthesis. However, this did not occur if intestinal microbiota were suppressed by antibiotics [94]. Similar effects (including atherosclerotic inflammation and foam cell formation) in the same mouse model were observed with a 1.0% choline supplementation [115]. Although significant, the difference in aortic lesion between supplemented vs. control was observed in only three mice, suggesting a possible type I error.
In a mouse study, high choline or TMAO supplementation (corresponding to 1600 mg choline/kg or 192 mg TMAO/kg) resulted in renal damage [165]. This study may explain a vicious cycle effect of further renal damage, once TMAO levels are extremely and unusually high.

5.2.2. Favorable or Neutral Effects of TMAO in Animals

When TMAO or its precursors are supplied in lower and recommended doses, protective effects on atherosclerosis hypertension and cholesterol are observed (Table 3), [185,202,206,208]. Protection against atherosclerosis was observed in mice supplemented with 352 mg/kg L-carnitine, corresponding to 2000 mg per day in humans. The resulting plasma TMAO levels were significantly elevated compared to baseline [185].
Moderate TMAO supplementation (approximately 6.7 mg TMAO/kg) also increased plasma TMAO levels by four- to five-fold while reducing diastolic dysfunction in the pressure-overloaded hearts of hypertensive rats [206].
Importantly, adverse effects on atherosclerosis or inflammation could not be observed in any of the studies detailed in Table 3, even where supplementation was relatively high, suggesting that other factors in the experimental design may interfere in the reported effects [207,209].
It should be noted that the animal models included in Table 3 are not severely compromised (e.g., impaired kidney function), as reported in Table 2, further emphasizing the difference in response to TMAO between relatively healthy and diseased populations. It is also possible that the adverse effects reported in Table 2 at levels up to 400 µM reflect non-specific effects.

5.2.3. Why TMAO Is a Marker but Not the Effector

As explained above, one of the dietary precursors of TMAO generation is L-carnitine. Furthermore, TMAO can become bioavailable as a result of consumption of fish and other sea foods (Figure 2). Evidence on the beneficial effects of L-carnitine and TMAO on human health is summarized below.
A recent review on L-carnitine and heart disease emphasized the role of exogenous L-carnitine as a preventive and/or therapeutic strategy against CVD, as well as associated dysfunctions including hypertension, hyperlipidemia, diabetic ketoacidosis, hyperglycemia, type 1 diabetes, insulin resistance, and obesity [52].
A systematic review and meta-analysis of 13 controlled trials with 3629 patients experiencing an acute myocardial infarction demonstrated that L-carnitine was associated with a 27% reduction in all-cause mortality, a 65% reduction in ventricular arrhythmias, and a 40% reduction in angina symptoms [216]. Although a more recent meta-analysis of 17 randomized controlled trials did not find significant differences in all-cause mortality with 1625 chronic HF patients compared to controls, L-carnitine was effective in improving clinical symptoms and cardiac functions and decreasing serum levels of heart insufficiency marker proteins [217]. Another meta-analysis of 5 studies revealed that L-carnitine was useful in treating patients with insulin resistance [218].
The following studies supplying L-carnitine (or a derivative) also evaluated TMAO levels in addition to cardiovascular effects.
In healthy, aged women, 24 weeks of 1500 mg/day L-carnitine Tartrate supplementation (n = 11) significantly increased plasma TMAO concentration (35 µM after 24 weeks), a 4.4-fold increase compared to the placebo group (n = 9; 8 µM) with no lipid profile changes or other markers of adverse cardiovascular events [47]. The lack of change in disease markers demonstrates that even a moderate elevation of TMAO in otherwise healthy individuals will not cause cardiometabolic dysfunctions. Furthermore, TMAO was shown to be an independent risk factor for disease and induced atherosclerosis in animal models without changing lipid profiles, so some may argue that a lack of change in lipid profiles and other markers in the face of elevations of TMAO is not necessarily indicative of a lack of cardiometabolic dysfunctions.
In 31 hemodialysis patients, 900 mg/day L-carnitine supplementation for 6 months increased total, free, and acyl L-carnitine, as well as plasma TMA and TMAO (from 222.5 µM to 548.4 µM, a factor 2.5; healthy control 174.3 µM—possibly method-derived high baseline values only allow a relative comparison). The supplementation decreased markers of vascular injury and oxidative stress such as intracellular adhesion molecule-1, vascular cell adhesion molecule-1, and malondialdehyde levels, thereby reducing advanced glycation end products and suggesting beneficial effects in these patients [48]. Therefore, there is accumulating evidence for beneficial cardiovascular effects of L-carnitine supplementation or a L-carnitine-rich diet.
Considering the above stated correlation between TMAO levels and cardiometabolic outcomes, a controversy also arises from the fact that the consumption of fish and seafood was previously shown to be associated with significant reduction in the risk of coronary heart disease-associated mortality [219]. In addition, 1–2 servings of fish per week were found to reduce the risk of coronary heart disease death by 36% and total mortality by 17% [220]. In 2 population-based cohort studies with 134,296 men and women, fish intake was inversely associated with risk of total and cause-specific mortality [221]. In a randomized controlled trial, a lean-seafood diet—compared to a non-seafood control—significantly improved fasting and the postprandial lipid profile [222]. Evidence on the potential effects of fish consumption on lipids arising from intervention trials indicates that consuming oily fish led to significant improvements in triglycerides and HDL-c levels [223].
With a Mediterranean diet (moderate amounts of fish), CVD incidence could be significantly reduced compared to a control diet [224]. Data did not support a significant association between basal plasma TMAO levels and increased risk of CVD in a Mediterranean population at high cardiovascular risk (PREDIMED-study) [74]. Interestingly, the PREDIMED study documented an inverse association between high plasma TMAO levels at baseline and incidence of T2D [58].

6. Summary and Conclusions

Given the complexity of cardiometabolic diseases and based on (1) the fact that observational trials cannot resolve the cause from the effects, (2) the recent PREDIMED data [58,74] measuring TMAO at baseline, and (3) the recent bi-directional Mendelian randomization analysis [180] examining the causal direction between TMAO and its precursors and the diseases, it can be concluded that TMAO increases in patients are the result (rather than the cause) of disease.
The apparent contradictions and the results from animal studies have to be interpreted in light of several factors. Animal studies, which apply excess amounts of TMAO or its precursors in distress-suffering animal models (e.g., coronary ligation models), are far from the normal situation and cannot explain a causal relationship between TMAO and disease development.
On the other hand, several animal experiments using lower doses of TMAO and its precursors did show protective effects.
The most convincing data in humans are derived from studies on fish consumption or intake of the dietary supplement L-carnitine. Indeed, the increased plasma TMAO levels observed in CVD/T2D may be a compensatory mechanism. Furthermore, the type of precursor for TMAO generation may also be important.
Increases in TMAO levels during CVD/T2D progression (beyond the observed wide natural intraindividual variability in TMAO levels) could be the result of disease-related dysbiosis. Another factor in such variability is related to the methodology itself. Indeed, most studies reported plasma TMAO only without considering TMA or urinary secretion. Perhaps it would be more relevant to consider the plasma TMA/TMAO ratio as a marker. It is also possible that TMAO elevation is a result of injuries caused by disease (e.g., atheroma lesions), which will induce TMAO up-regulation to promote the healing process. TMAO has been shown to be up-regulated in ischemic-injury models, as discussed above. These up-regulations may be mediated by FMO gene regulation, which in turn is modulated by many factors.
In conclusion, increased TMAO levels may be a compensatory mechanism in response to disease. Further research and intervention studies with relatively low levels of TMAO may be needed to confirm this theory.

7. Literature Search Methods

The PubMed database (www.ncbi.nlm.nih.gov) was searched for the term “Trimethylamine N-oxide” or “TMAO”. Of the total 1963 references initially identified from the literature search up to 11 November 2019, 179 hits and 67 hits were filtered out for “review” and “clinical trial”, respectively, as well as 334 hits for “rat or mouse” (Figure 3). Hits were screened for relevant publications, both for giving a literature overview and for finding nonclinical and clinical studies evaluating the hypotheses that TMAO is a cause or effect of cardiometabolic outcomes. Literature cited within articles was followed up to complement the search. Additional literature sources were added during the review phase up to 27 April 2020.

Author Contributions

Conceptualization, C.P. and A.B.; methodology, M.M.; validation C.P., A.B. and M.M., formal analysis, M.M.; investigation, C.P., A.B., and M.M.; resources, M.M. and A.B.; data curation, M.M.; writing—original draft preparation, C.P., A.B., and M.M.; writing—review and editing, C.P., M.M., and A.B.; visualization, C.P., M.M., and A.B.; supervision, A.B.; project administration, A.B. and M.M.; funding acquisition, A.B. and C.P. All authors have read and agreed to the published version of the manuscript.

Funding

CP is recipient of the Instituto de Salud Carlos III Miguel Servet fellowship (grant CP 19/00189). This research was funded in part by Lonza LLC.

Conflicts of Interest

C.P. has received an expense allowance from Lonza LLC for discussing the scope, writing, and editing the manuscript. M.M. is an independent consultant who received payment by the contract research organization analyze & realize GmbH, on behalf of Lonza LLC, for writing this manuscript. A.B. is an employee of Lonza LLC. This review has been funded by Lonza, a manufacturer of L-carnitine, one of the TMAO precursors.

References

  1. Lopez, A.D.; Mathers, C.D.; Ezzati, M.; Jamison, D.T.; Murray, C.J. Global and regional burden of disease and risk factors, 2001: Systematic analysis of population health data. Lancet 2006, 367, 1747–1757. [Google Scholar] [CrossRef]
  2. Saklayen, M.G. The Global Epidemic of the Metabolic Syndrome. Curr. Hypertens Rep. 2018, 20, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. WHO. Global Report on Diabetes. Online Publication. 2016. Available online: https://apps.who.int/iris/bitstream/handle/10665/204871/9789241565257_eng.pdf;jsessionid=BB2C89145E6EB31BD42C0A3A68D965DD?sequence=1 (accessed on 6 May 2020).
  4. Miura, S.I.; Shiga, Y.; Ike, A.; Iwata, A. Atherosclerotic Coronary Artery Disease in Patients With Cardiometabolic Syndrome. Cardiol. Res. 2019, 10, 69–73. [Google Scholar] [CrossRef] [PubMed]
  5. Adeva-Andany, M.M.; Martinez-Rodriguez, J.; Gonzalez-Lucan, M.; Fernandez-Fernandez, C.; Castro-Quintela, E. Insulin resistance is a cardiovascular risk factor in humans. Diabetes Metab. Syndr. 2019, 13, 1449–1455. [Google Scholar] [CrossRef] [PubMed]
  6. Taleb, S. Inflammation in atherosclerosis. Arch. Cardiovasc. Dis. 2016, 109, 708–715. [Google Scholar] [CrossRef] [PubMed]
  7. Reddy, P.; Lent-Schochet, D.; Ramakrishnan, N.; McLaughlin, M.; Jialal, I. Metabolic syndrome is an inflammatory disorder: A conspiracy between adipose tissue and phagocytes. Clin. Chim. Acta 2019, 496, 35–44. [Google Scholar] [CrossRef]
  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. Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; de Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef]
  10. Bruck, K.; Stel, V.S.; Gambaro, G.; Hallan, S.; Volzke, H.; Arnlov, J.; Kastarinen, M.; Guessous, I.; Vinhas, J.; Stengel, B.; et al. CKD Prevalence Varies across the European General Population. J. Am. Soc. Nephrol. 2016, 27, 2135–2147. [Google Scholar] [CrossRef]
  11. Saran, R.; Robinson, B.; Abbott, K.C.; Agodoa, L.Y.C.; Bhave, N.; Bragg-Gresham, J.; Balkrishnan, R.; Dietrich, X.; Eckard, A.; Eggers, P.W.; et al. US Renal Data System 2017 Annual Data Report: Epidemiology of Kidney Disease in the United States. Am. J. Kidney Dis. 2018, 71, A7. [Google Scholar] [CrossRef]
  12. Lau, K.; Srivatsav, V.; Rizwan, A.; Nashed, A.; Liu, R.; Shen, R.; Akhtar, M. Bridging the Gap between Gut Microbial Dysbiosis and Cardiovascular Diseases. Nutrients 2017, 9, 859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Miller, M.J. Risk factors for cardiovascular disease: A cautionary tale of diet-microbiome interactions. J. Am. Coll. Nutr. 2013, 32, 75–78. [Google Scholar] [CrossRef] [PubMed]
  14. Karlsson, F.H.; Fak, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Backhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012, 3, 1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Jonsson, A.L.; Backhed, F. Role of gut microbiota in atherosclerosis. Nat. Rev. Cardiol. 2017, 14, 79–87. [Google Scholar] [CrossRef]
  16. Nallu, A.; Sharma, S.; Ramezani, A.; Muralidharan, J.; Raj, D. Gut microbiome in chronic kidney disease: Challenges and opportunities. Transl. Res. 2017, 179, 24–37. [Google Scholar] [CrossRef] [Green Version]
  17. Schoeler, M.; Caesar, R. Dietary lipids, gut microbiota and lipid metabolism. Rev. Endocr. Metab. Disord. 2019, 20, 461–472. [Google Scholar] [CrossRef] [Green Version]
  18. Felizardo, R.J.F.; Watanabe, I.K.M.; Dardi, P.; Rossoni, L.V.; Camara, N.O.S. The interplay among gut microbiota, hypertension and kidney diseases: The role of short-chain fatty acids. Pharmacol. Res. 2019, 141, 366–377. [Google Scholar] [CrossRef]
  19. Ratajczak, W.; Ryl, A.; Mizerski, A.; Walczakiewicz, K.; Sipak, O.; Laszczynska, M. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol. 2019, 66, 1–12. [Google Scholar] [CrossRef] [Green Version]
  20. Santos-Marcos, J.A.; Perez-Jimenez, F.; Camargo, A. The role of diet and intestinal microbiota in the development of metabolic syndrome. J. Nutr. Biochem. 2019, 70, 1–27. [Google Scholar] [CrossRef]
  21. Bartolomaeus, H.; Balogh, A.; Yakoub, M.; Homann, S.; Marko, L.; Hoges, S.; Tsvetkov, D.; Krannich, A.; Wundersitz, S.; Avery, E.G.; et al. Short-Chain Fatty Acid Propionate Protects From Hypertensive Cardiovascular Damage. Circulation 2019, 139, 1407–1421. [Google Scholar] [CrossRef]
  22. Zhang, L.; Wang, F.; Wang, J.; Wang, Y.; Fang, Y. Intestinal fatty acid-binding protein mediates atherosclerotic progress through increasing intestinal inflammation and permeability. J. Cell Mol. Med. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Plata, C.; Cruz, C.; Cervantes, L.G.; Ramirez, V. The gut microbiota and its relationship with chronic kidney disease. Int. Urol. Nephrol. 2019. [Google Scholar] [CrossRef] [PubMed]
  24. Rajani, C.; Jia, W. Disruptions in gut microbial-host co-metabolism and the development of metabolic disorders. Clin. Sci. (London) 2018, 132, 791–811. [Google Scholar] [CrossRef] [PubMed]
  25. Tang, W.H.; Hazen, S.L. The contributory role of gut microbiota in cardiovascular disease. J. Clin. Invest. 2014, 124, 4204–4211. [Google Scholar] [CrossRef]
  26. Zhuang, R.; Ge, X.; Han, L.; Yu, P.; Gong, X.; Meng, Q.; Zhang, Y.; Fan, H.; Zheng, L.; Liu, Z.; et al. Gut microbe-generated metabolite trimethylamine N-oxide and the risk of diabetes: A systematic review and dose-response meta-analysis. Obes Rev. 2019, 20, 883–894. [Google Scholar] [CrossRef]
  27. Oellgaard, J.; Winther, S.A.; Hansen, T.S.; Rossing, P.; von Scholten, B.J. Trimethylamine N-oxide (TMAO) as a New Potential Therapeutic Target for Insulin Resistance and Cancer. Curr. Pharm. Des. 2017, 23, 3699–3712. [Google Scholar] [CrossRef]
  28. Barrea, L.; Annunziata, G.; Muscogiuri, G.; Di Somma, C.; Laudisio, D.; Maisto, M.; de Alteriis, G.; Tenore, G.C.; Colao, A.; Savastano, S. Trimethylamine-N-oxide (TMAO) as Novel Potential Biomarker of Early Predictors of Metabolic Syndrome. Nutrients 2018, 10, 1971. [Google Scholar] [CrossRef] [Green Version]
  29. Bennett, B.J.; de Aguiar Vallim, T.Q.; Wang, Z.; Shih, D.M.; Meng, Y.; Gregory, J.; Allayee, H.; Lee, R.; Graham, M.; Crooke, R.; et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metab. 2013, 17, 49–60. [Google Scholar] [CrossRef] [Green Version]
  30. al-Waiz, M.; Mikov, M.; Mitchell, S.C.; Smith, R.L. The exogenous origin of trimethylamine in the mouse. Metabolism 1992, 41, 135–136. [Google Scholar] [CrossRef]
  31. Rebouche, C.J.; Seim, H. Carnitine metabolism and its regulation in microorganisms and mammals. Annu Rev. Nutr. 1998, 18, 39–61. [Google Scholar] [CrossRef]
  32. Zeisel, S.H.; daCosta, K.A.; Youssef, M.; Hensey, S. Conversion of dietary choline to trimethylamine and dimethylamine in rats: Dose-response relationship. J. Nutr. 1989, 119, 800–804. [Google Scholar] [CrossRef] [PubMed]
  33. Treacy, E.P.; Akerman, B.R.; Chow, L.M.; Youil, R.; Bibeau, C.; Lin, J.; Bruce, A.G.; Knight, M.; Danks, D.M.; Cashman, J.R.; et al. Mutations of the flavin-containing monooxygenase gene (FMO3) cause trimethylaminuria, a defect in detoxication. Hum. Mol. Genet. 1998, 7, 839–845. [Google Scholar] [CrossRef]
  34. Mitchell, S.C.; Smith, R.L. Trimethylaminuria: The fish malodor syndrome. Drug Metab Dispos 2001, 29, 517–521. [Google Scholar] [PubMed]
  35. Hoyles, L.; Jimenez-Pranteda, M.L.; Chilloux, J.; Brial, F.; Myridakis, A.; Aranias, T.; Magnan, C.; Gibson, G.R.; Sanderson, J.D.; Nicholson, J.K.; et al. Metabolic retroconversion of trimethylamine N-oxide and the gut microbiota. Microbiome 2018, 6, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Bain, M.A.; Faull, R.; Fornasini, G.; Milne, R.W.; Evans, A.M. Accumulation of trimethylamine and trimethylamine-N-oxide in end-stage renal disease patients undergoing haemodialysis. Nephrol. Dial. Transplant. 2006, 21, 1300–1304. [Google Scholar] [CrossRef] [Green Version]
  37. Fennema, D.; Phillips, I.R.; Shephard, E.A. Trimethylamine and Trimethylamine N-Oxide, a Flavin-Containing Monooxygenase 3 (FMO3)-Mediated Host-Microbiome Metabolic Axis Implicated in Health and Disease. Drug Metab Dispos 2016, 44, 1839–1850. [Google Scholar] [CrossRef]
  38. Janeiro, M.H.; Ramirez, M.J.; Milagro, F.I.; Martinez, J.A.; Solas, M. Implication of Trimethylamine N-Oxide (TMAO) in Disease: Potential Biomarker or New Therapeutic Target. Nutrients 2018, 10, 1398. [Google Scholar] [CrossRef] [Green Version]
  39. Zeisel, S.H.; Wishnok, J.S.; Blusztajn, J.K. Formation of methylamines from ingested choline and lecithin. J. Pharmacol. Exp. Ther 1983, 225, 320–324. [Google Scholar]
  40. 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. [Google Scholar] [CrossRef]
  41. Zhang, A.Q.; Mitchell, S.C.; Smith, R.L. Dietary precursors of trimethylamine in man: A pilot study. Food Chem. Toxicol. 1999, 37, 515–520. [Google Scholar] [CrossRef]
  42. NIH. Carnitine: Fact Sheet for Health Professionals. 2017. Available online: https://ods.od.nih.gov/factsheets/Carnitine-HealthProfessional/#en6 (accessed on 14 October 2019).
  43. Feller, A.G.; Rudman, D. Role of carnitine in human nutrition. J. Nutr. 1988, 118, 541–547. [Google Scholar] [CrossRef] [PubMed]
  44. Rebouche, C.J. Carnitine function and requirements during the life cycle. FASEB J. 1992, 6, 3379–3386. [Google Scholar] [CrossRef] [PubMed]
  45. EFSA Panel on Dietetic Products, N.a.A.N. L-carnitine and contribution to normal lipid metabolism: Evaluation of a health claim pursuant to Article 13(5) of Regulation (EC) No 1924/2006. EFSA J. 2018, 16, 5137–5139. [Google Scholar] [CrossRef]
  46. Rebouche, C.J. Kinetics, pharmacokinetics, and regulation of L-carnitine and acetyl-L-carnitine metabolism. Ann. N. Y. Acad. Sci 2004, 1033, 30–41. [Google Scholar] [CrossRef] [PubMed]
  47. Samulak, J.J.; Sawicka, A.K.; Hartmane, D.; Grinberga, S.; Pugovics, O.; Lysiak-Szydlowska, W.; Olek, R.A. L-Carnitine Supplementation Increases Trimethylamine-N-Oxide but not Markers of Atherosclerosis in Healthy Aged Women. Ann. Nutr. Metab. 2019, 74, 11–17. [Google Scholar] [CrossRef] [PubMed]
  48. Fukami, K.; Yamagishi, S.; Sakai, K.; Kaida, Y.; Yokoro, M.; Ueda, S.; Wada, Y.; Takeuchi, M.; Shimizu, M.; Yamazaki, H.; et al. Oral L-carnitine supplementation increases trimethylamine-N-oxide but reduces markers of vascular injury in hemodialysis patients. J. Cardiovasc. Pharmacol. 2015, 65, 289–295. [Google Scholar] [CrossRef]
  49. Fritz, I.B.; Mc, E.B. Effects of carnitine on fatty-acid oxidation by muscle. Science 1959, 129, 334–335. [Google Scholar] [CrossRef]
  50. Longo, N.; Frigeni, M.; Pasquali, M. Carnitine transport and fatty acid oxidation. Biochim. Biophys Acta 2016, 1863, 2422–2435. [Google Scholar] [CrossRef]
  51. El-Hattab, A.W. Systemic Primary Carnitine Deficiency. In GeneReviews (R); Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Stephens, K., Amemiya, A., Eds.; GeneRviews: Seattle, WA, USA, 1993. [Google Scholar]
  52. Wang, Z.Y.; Liu, Y.Y.; Liu, G.H.; Lu, H.B.; Mao, C.Y. l-Carnitine and heart disease. Life Sci. 2018, 194, 88–97. [Google Scholar] [CrossRef]
  53. Lee, B.J.; Lin, J.S.; Lin, Y.C.; Lin, P.T. Effects of L-carnitine supplementation on lipid profiles in patients with coronary artery disease. Lipids Health Dis. 2016, 15, 107. [Google Scholar] [CrossRef] [Green Version]
  54. Ferreira, G.C.; McKenna, M.C. L-Carnitine and Acetyl-L-carnitine Roles and Neuroprotection in Developing Brain. Neurochem. Res. 2017, 42, 1661–1675. [Google Scholar] [CrossRef] [PubMed]
  55. Moghaddas, A.; Dashti-Khavidaki, S. L-Carnitine and Potential Protective Effects Against Ischemia-Reperfusion Injury in Noncardiac Organs: From Experimental Data to Potential Clinical Applications. J. Diet. Suppl. 2018, 15, 740–756. [Google Scholar] [CrossRef] [PubMed]
  56. Dinicolantonio, J.J.; Niazi, A.K.; McCarty, M.F.; Lavie, C.J.; Liberopoulos, E.; O’Keefe, J.H. L-carnitine for the treatment of acute myocardial infarction. Rev. Cardiovasc. Med. 2014, 15, 52–62. [Google Scholar] [PubMed]
  57. Mingrone, G.; Greco, A.V.; Capristo, E.; Benedetti, G.; Giancaterini, A.; De Gaetano, A.; Gasbarrini, G. L-carnitine improves glucose disposal in type 2 diabetic patients. J. Am. Coll Nutr. 1999, 18, 77–82. [Google Scholar] [CrossRef]
  58. Papandreou, C.; Bullo, M.; Zheng, Y.; Ruiz-Canela, M.; Yu, E.; Guasch-Ferre, M.; Toledo, E.; Clish, C.; Corella, D.; Estruch, R.; et al. Plasma trimethylamine-N-oxide and related metabolites are associated with type 2 diabetes risk in the Prevencion con Dieta Mediterranea (PREDIMED) trial. Am. J. Clin. Nutr. 2018, 108, 163–173. [Google Scholar] [CrossRef]
  59. Spanish Agency for Food Safety and Nutrition. Report of the Scientific Committee of the Spanish Agency for Food Safety and Nutrition (AESAN) on the Use Conditions for Certain Substances Other Than Vitamins, Minerals and Plants in Food Supplements. Available online: http://www.aecosan.msssi.gob.es/AECOSAN/docs/documentos/seguridad_alimentaria/evaluacion_riesgos/informes_cc_ingles/FOOD_SUPPLEMENTS.pdf (accessed on 6 May 2020).
  60. Zeisel, S.H.; Mar, M.H.; Howe, J.C.; Holden, J.M. Concentrations of choline-containing compounds and betaine in common foods. J. Nutr. 2003, 133, 1302–1307. [Google Scholar] [CrossRef]
  61. NIH. Choline: Fact Sheet for Health Professionals. 2019. Available online: https://ods.od.nih.gov/factsheets/Choline-HealthProfessional/ (accessed on 14 October 2019).
  62. EFSA Panel on Dietetic Products, N.a.A.N. Dietary Reference Values for choline. FEFSA J. 2016, 14, e04484. [Google Scholar] [CrossRef]
  63. Blusztajn, J.K.; Slack, B.E.; Mellott, T.J. Neuroprotective Actions of Dietary Choline. Nutrients 2017, 9, 815. [Google Scholar] [CrossRef] [Green Version]
  64. Edwards, C.G.; Walk, A.M.; Cannavale, C.N.; Flemming, I.R.; Thompson, S.V.; Reeser, G.R.; Holscher, H.D.; Khan, N.A. Dietary choline is related to neural efficiency during a selective attention task among middle-aged adults with overweight and obesity. Nutr. Neurosci. 2019, 1–10. [Google Scholar] [CrossRef]
  65. EFSA Panel on Dietetic Products, Nutrition and Allergies (NDA). Scientific Opinion on the substantiation of health claims related to choline and contribution to normal lipid metabolism (ID 3186), maintenance of normal liver function (ID 1501), contribution to normal homocysteine metabolism (ID 3090), maintenance of normal neurological function (ID 1502), contribution to normal cognitive function (ID 1502), and brain and neurological development (ID 1503) pursuant to Article 13(1) of Regulation (EC) No 1924/2006. EFSA J. 2011, 9, 2056. [Google Scholar] [CrossRef]
  66. Schmitz, M.G.; Renooij, W. Phospholipids from rat, human, and canine gastric mucosa. Composition and metabolism of molecular classes of phosphatidylcholine. Gastroenterology 1990, 99, 1292–1296. [Google Scholar] [CrossRef]
  67. Zeisel, S.H. Choline deficiency. J. Nutr. Biochem. 1990, 1, 332–349. [Google Scholar] [CrossRef]
  68. Miller, C.A.; Corbin, K.D.; da Costa, K.A.; Zhang, S.; Zhao, X.; Galanko, J.A.; Blevins, T.; Bennett, B.J.; O’Connor, A.; Zeisel, S.H. Effect of egg ingestion on trimethylamine-N-oxide production in humans: A randomized, controlled, dose-response study. Am. J. Clin. Nutr. 2014, 100, 778–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Lemos, B.S.; Medina-Vera, I.; Malysheva, O.V.; Caudill, M.A.; Fernandez, M.L. Effects of Egg Consumption and Choline Supplementation on Plasma Choline and Trimethylamine-N-Oxide in a Young Population. J. Am. Coll. Nutr. 2018, 1–8. [Google Scholar] [CrossRef] [PubMed]
  70. Berge, R.K.; Ramsvik, M.S.; Bohov, P.; Svardal, A.; Nordrehaug, J.E.; Rostrup, E.; Bruheim, I.; Bjorndal, B. Krill oil reduces plasma triacylglycerol level and improves related lipoprotein particle concentration, fatty acid composition and redox status in healthy young adults-a pilot study. Lipids Health Dis. 2015, 14, 163. [Google Scholar] [CrossRef] [Green Version]
  71. DiMarco, D.M.; Missimer, A.; Murillo, A.G.; Lemos, B.S.; Malysheva, O.V.; Caudill, M.A.; Blesso, C.N.; Fernandez, M.L. Intake of up to 3 Eggs/Day Increases HDL Cholesterol and Plasma Choline While Plasma Trimethylamine-N-oxide is Unchanged in a Healthy Population. Lipids 2017, 52, 255–263. [Google Scholar] [CrossRef]
  72. Konstantinova, S.V.; Tell, G.S.; Vollset, S.E.; Nygard, O.; Bleie, O.; Ueland, P.M. Divergent associations of plasma choline and betaine with components of metabolic syndrome in middle age and elderly men and women. J. Nutr. 2008, 138, 914–920. [Google Scholar] [CrossRef] [Green Version]
  73. Lever, M.; George, P.M.; Elmslie, J.L.; Atkinson, W.; Slow, S.; Molyneux, S.L.; Troughton, R.W.; Richards, A.M.; Frampton, C.M.; Chambers, S.T. Betaine and secondary events in an acute coronary syndrome cohort. PLoS ONE 2012, 7, e37883. [Google Scholar] [CrossRef]
  74. Guasch-Ferre, M.; Hu, F.B.; Ruiz-Canela, M.; Bullo, M.; Toledo, E.; Wang, D.D.; Corella, D.; Gomez-Gracia, E.; Fiol, M.; Estruch, R.; et al. Plasma Metabolites From Choline Pathway and Risk of Cardiovascular Disease in the PREDIMED (Prevention With Mediterranean Diet) Study. J. Am. Heart Assoc. 2017, 6. [Google Scholar] [CrossRef] [Green Version]
  75. Lai, S.; Hua, X.; Gao, R.; Zeng, L.; Song, J.; Liu, J.; Zhang, J. Combinational Biomarkers for Atrial Fibrillation Derived from Atrial Appendage and Plasma Metabolomics Analysis. Sci. Rep. 2018, 8, 16930. [Google Scholar] [CrossRef] [Green Version]
  76. Zuo, H.; Svingen, G.F.T.; Tell, G.S.; Ueland, P.M.; Vollset, S.E.; Pedersen, E.R.; Ulvik, A.; Meyer, K.; Nordrehaug, J.E.; Nilsen, D.W.T.; et al. Plasma Concentrations and Dietary Intakes of Choline and Betaine in Association With Atrial Fibrillation Risk: Results From 3 Prospective Cohorts With Different Health Profiles. J. Am. Heart Assoc. 2018, 7. [Google Scholar] [CrossRef] [PubMed]
  77. Martinez-Vega, R.; Garrido, F.; Partearroyo, T.; Cediel, R.; Zeisel, S.H.; Martinez-Alvarez, C.; Varela-Moreiras, G.; Varela-Nieto, I.; Pajares, M.A. Folic acid deficiency induces premature hearing loss through mechanisms involving cochlear oxidative stress and impairment of homocysteine metabolism. FASEB J. 2015, 29, 418–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Svingen, G.; Ueland, P.M.; Schartum-Hansen, H.; Pedersen, E.R.; Seifert, R.; Nygard, O.K. Metabolites in the choline oxidation pathway in relation to diabetes, glycemic control and insulin sensitivity among patients with angina pectoris. Atherosclerosis 2014, 235, e51. [Google Scholar] [CrossRef]
  79. Obeid, R. The metabolic burden of methyl donor deficiency with focus on the betaine homocysteine methyltransferase pathway. Nutrients 2013, 5, 3481–3495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. 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] [PubMed] [Green Version]
  81. Ejaz, A.; Martinez-Guino, L.; Goldfine, A.B.; Ribas-Aulinas, F.; De Nigris, V.; Ribo, S.; Gonzalez-Franquesa, A.; Garcia-Roves, P.M.; Li, E.; Dreyfuss, J.M.; et al. Dietary Betaine Supplementation Increases Fgf21 Levels to Improve Glucose Homeostasis and Reduce Hepatic Lipid Accumulation in Mice. Diabetes 2016, 65, 902–912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wang, Z.; Tang, W.H.; Buffa, J.A.; Fu, X.; Britt, E.B.; Koeth, R.A.; Levison, B.S.; Fan, Y.; Wu, Y.; Hazen, S.L. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur. Heart J. 2014, 35, 904–910. [Google Scholar] [CrossRef] [PubMed]
  83. Binzak, B.; Vockley, J.; Jenkins, R.; Vockley, J. Structure and analysis of the human dimethylglycine dehydrogenase gene. Mol. Genet. Metab. 2000, 69, 181–187. [Google Scholar] [CrossRef] [PubMed]
  84. Massy, Z. Potential strategies to normalize the levels of homocysteine in chronic renal failure patients. Kidney Int. 2003, 63, S134–S136. [Google Scholar] [CrossRef] [Green Version]
  85. Svingen, G.; Ueland, P.; Pedersen, E.; Schartum-Hansen, H.; Seifert, R.; Ebbing, M.; Løland, K.; Tell, G.; Nygård, O. Plasma Dimethylglycine and Risk of Incident Acute Myocardial Infarction in Patients With Stable Angina Pectoris. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2041–2048. [Google Scholar] [CrossRef] [Green Version]
  86. Ey, J.; Schomig, E.; Taubert, D. Dietary sources and antioxidant effects of ergothioneine. J. Agric. Food Chem. 2007, 55, 6466–6474. [Google Scholar] [CrossRef]
  87. Cheah, I.K.; Halliwell, B. Ergothioneine; antioxidant potential, physiological function and role in disease. Biochim. Biophys Acta 2012, 1822, 784–793. [Google Scholar] [CrossRef] [Green Version]
  88. Smith, E.; Ottoson, F.; Hellstrand, S.; Ericson, U.; Orho-Melander, M.; Fernandez, C.; Melander, O. Ergothioneine is associated with reduced mortality and decreased risk of cardiovascular disease. Heart 2019, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Yin, X.; Gibbons, H.; Rundle, M.; Frost, G.; McNulty, B.A.; Nugent, A.P.; Walton, J.; Flynn, A.; Brennan, L. The Relationship between Fish Intake and Urinary Trimethylamine-N-Oxide. Mol. Nutr. Food Res. 2020, 64, e1900799. [Google Scholar] [CrossRef] [PubMed]
  90. Al-Waiz, M.; Mitchell, S.C.; Idle, J.R.; Smith, R.L. The metabolism of 14C-labelled trimethylamine and its N-oxide in man. Xenobiotica 1987, 17, 551–558. [Google Scholar] [CrossRef] [PubMed]
  91. Svensson, B.G.; Akesson, B.; Nilsson, A.; Paulsson, K. Urinary excretion of methylamines in men with varying intake of fish from the Baltic Sea. J. Toxicol. Environ. Health 1994, 41, 411–420. [Google Scholar] [CrossRef] [PubMed]
  92. Zeisel, S.H.; Corbin, K.D. Present Knowledge in Nutrition, 10th ed.; Erdman, J., Jr., MacDonald, I., Zeisel, S., Eds.; Wiley-Blackwell: Washington, DC, USA, 2012; pp. 405–418. [Google Scholar]
  93. Rebouche, C. Carnitine. In Modern Nutrition in Health and Disease, 9th ed.; Shils, M., Olson, J., Shike, M., Ross, A., Eds.; Lippincott Williams and Wilkins: New York, NY, USA, 1999; pp. 505–512. [Google Scholar]
  94. 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] [PubMed] [Green Version]
  95. Wang, Z.; Bergeron, N.; Levison, B.S.; Li, X.S.; Chiu, S.; Jia, X.; Koeth, R.A.; Li, L.; Wu, Y.; Tang, W.H.W.; et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583–594. [Google Scholar] [CrossRef]
  96. Park, J.E.; Miller, M.; Rhyne, J.; Wang, Z.; Hazen, S.L. 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]
  97. Yancey, P.H. Organic osmolytes as compatible, metabolic and counteracting cytoprotectants in high osmolarity and other stresses. J. Exp. Biol. 2005, 208, 2819–2830. [Google Scholar] [CrossRef] [Green Version]
  98. Yancey, P.H.; Siebenaller, J.F. Co-evolution of proteins and solutions: Protein adaptation versus cytoprotective micromolecules and their roles in marine organisms. J. Exp. Biol. 2015, 218, 1880–1896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Treberg, J.R.; Driedzic, W.R. Elevated levels of trimethylamine oxide in deep-sea fish: Evidence for synthesis and intertissue physiological importance. J. Exp. Zool. 2002, 293, 39–45. [Google Scholar] [CrossRef] [PubMed]
  100. Ma, J.; Pazos, I.M.; Gai, F. Microscopic insights into the protein-stabilizing effect of trimethylamine N-oxide (TMAO). Proc. Natl. Acad. Sci. USA 2014, 111, 8476–8481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Bennion, B.J.; Daggett, V. Counteraction of urea-induced protein denaturation by trimethylamine N-oxide: A chemical chaperone at atomic resolution. Proc. Natl. Acad. Sci. USA 2004, 101, 6433–6438. [Google Scholar] [CrossRef] [Green Version]
  102. Jackson-Atogi, R.; Sinha, P.K.; Rosgen, J. Distinctive solvation patterns make renal osmolytes diverse. Biophys J. 2013, 105, 2166–2174. [Google Scholar] [CrossRef] [Green Version]
  103. Hua, X.; Su, Z.; Deng, R.; Lin, J.; Li, D.Q.; Pflugfelder, S.C. Effects of L-carnitine, erythritol and betaine on pro-inflammatory markers in primary human corneal epithelial cells exposed to hyperosmotic stress. Curr. Eye Res. 2015, 40, 657–667. [Google Scholar] [CrossRef]
  104. 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] [Green Version]
  105. McEntyre, C.J.; Lever, M.; Chambers, S.T.; George, P.M.; Slow, S.; Elmslie, J.L.; Florkowski, C.M.; Lunt, H.; Krebs, J.D. Variation of betaine, N,N-dimethylglycine, choline, glycerophosphorylcholine, taurine and trimethylamine-N-oxide in the plasma and urine of overweight people with type 2 diabetes over a two-year period. Ann. Clin. Biochem. 2015, 52, 352–360. [Google Scholar] [CrossRef] [Green Version]
  106. Li, D.; Ke, Y.; Zhan, R.; Liu, C.; Zhao, M.; Zeng, A.; Shi, X.; Ji, L.; Cheng, S.; Pan, B.; et al. Trimethylamine-N-oxide promotes brain aging and cognitive impairment in mice. Aging Cell 2018, 17, e12768. [Google Scholar] [CrossRef]
  107. Ke, Y.; Li, D.; Zhao, M.; Liu, C.; Liu, J.; Zeng, A.; Shi, X.; Cheng, S.; Pan, B.; Zheng, L.; et al. Gut flora-dependent metabolite Trimethylamine-N-oxide accelerates endothelial cell senescence and vascular aging through oxidative stress. Free Radic. Biol. Med. 2018, 116, 88–100. [Google Scholar] [CrossRef]
  108. Razavi, A.C.; Potts, K.S.; Kelly, T.N.; Bazzano, L.A. Sex, gut microbiome, and cardiovascular disease risk. Biol Sex. Differ. 2019, 10, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-Oxide: The Good, the Bad and the Unknown. Toxins (Basel) 2016, 8, 326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Veeravalli, S.; Karu, K.; Scott, F.; Fennema, D.; Phillips, I.R.; Shephard, E.A. Effect of Flavin-Containing Monooxygenase Genotype, Mouse Strain, and Gender on Trimethylamine N-oxide Production, Plasma Cholesterol Concentration, and an Index of Atherosclerosis. Drug Metab Dispos. 2018, 46, 20–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Turkanoglu Ozcelik, A.; Can Demirdogen, B.; Demirkaya, S.; Adali, O. Flavin containing monooxygenase 3 genetic polymorphisms Glu158Lys and Glu308Gly and their relation to ischemic stroke. Gene 2013, 521, 116–121. [Google Scholar] [CrossRef]
  112. Zane, N.R.; Chen, Y.; Wang, M.Z.; Thakker, D.R. Cytochrome P450 and flavin-containing monooxygenase families: Age-dependent differences in expression and functional activity. Pediatr. Res. 2018, 83, 527–535. [Google Scholar] [CrossRef]
  113. Falls, J.G.; Blake, B.L.; Cao, Y.; Levi, P.E.; Hodgson, E. Gender differences in hepatic expression of flavin-containing monooxygenase isoforms (FMO1, FMO3, and FMO5) in mice. J. Biochem. Toxicol. 1995, 10, 171–177. [Google Scholar] [CrossRef]
  114. Mayr, M.; Chung, Y.L.; Mayr, U.; Yin, X.; Ly, L.; Troy, H.; Fredericks, S.; Hu, Y.; Griffiths, J.R.; Xu, Q. Proteomic and metabolomic analyses of atherosclerotic vessels from apolipoprotein E-deficient mice reveal alterations in inflammation, oxidative stress, and energy metabolism. Arter. Thromb. Vasc. Biol. 2005, 25, 2135–2142. [Google Scholar] [CrossRef] [Green Version]
  115. 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] [Green Version]
  116. Warrier, M.; Shih, D.M.; Burrows, A.C.; Ferguson, D.; Gromovsky, A.D.; Brown, A.L.; Marshall, S.; McDaniel, A.; Schugar, R.C.; Wang, Z.; et al. The TMAO-Generating Enzyme Flavin Monooxygenase 3 Is a Central Regulator of Cholesterol Balance. Cell Rep. 2015, 10, 326–338. [Google Scholar] [CrossRef] [Green Version]
  117. Miao, J.; Ling, A.V.; Manthena, P.V.; Gearing, M.E.; Graham, M.J.; Crooke, R.M.; Croce, K.J.; Esquejo, R.M.; Clish, C.B.; Morbid Obesity Study, G.; et al. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nat. Commun. 2015, 6, 6498. [Google Scholar] [CrossRef]
  118. Shih, D.M.; Wang, Z.; Lee, R.; Meng, Y.; Che, N.; Charugundla, S.; Qi, H.; Wu, J.; Pan, C.; Brown, J.M.; et al. Flavin containing monooxygenase 3 exerts broad effects on glucose and lipid metabolism and atherosclerosis. J. Lipid Res. 2015, 56, 22–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Lee, M.Y.; Clark, J.E.; Williams, D.E. Induction of flavin-containing monooxygenase (FMO B) in rabbit lung and kidney by sex steroids and glucocorticoids. Arch. Biochem. Biophys 1993, 302, 332–336. [Google Scholar] [CrossRef] [PubMed]
  120. Schugar, R.C.; Shih, D.M.; Warrier, M.; Helsley, R.N.; Burrows, A.; Ferguson, D.; Brown, A.L.; Gromovsky, A.D.; Heine, M.; Chatterjee, A.; et al. The TMAO-Producing Enzyme Flavin-Containing Monooxygenase 3 Regulates Obesity and the Beiging of White Adipose Tissue. Cell Rep. 2017, 19, 2451–2461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Robinson-Cohen, C.; Newitt, R.; Shen, D.D.; Rettie, A.E.; Kestenbaum, B.R.; Himmelfarb, J.; Yeung, C.K. Association of FMO3 Variants and Trimethylamine N-Oxide Concentration, Disease Progression, and Mortality in CKD Patients. PLoS ONE 2016, 11, e0161074. [Google Scholar] [CrossRef]
  122. Prokopienko, A.J.; West, R.E., 3rd; Schrum, D.P.; Stubbs, J.R.; Leblond, F.A.; Pichette, V.; Nolin, T.D. Metabolic Activation of Flavin Monooxygenase-mediated Trimethylamine-N-Oxide Formation in Experimental Kidney Disease. Sci. Rep. 2019, 9, 15901. [Google Scholar] [CrossRef] [Green Version]
  123. Johnson, C.; Prokopienko, A.J.; West, R.E., 3rd; Nolin, T.D.; Stubbs, J.R. Decreased Kidney Function Is Associated with Enhanced Hepatic Flavin Monooxygenase Activity and Increased Circulating Trimethylamine N-Oxide Concentrations in Mice. Drug Metab. Dispos 2018, 46, 1304–1309. [Google Scholar] [CrossRef] [Green Version]
  124. Hsu, C.N.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Lu, P.C.; Tain, Y.L. Association of Trimethylamine, Trimethylamine N-oxide, and Dimethylamine with Cardiovascular Risk in Children with Chronic Kidney Disease. J. Clin. Med. 2020, 9, 336. [Google Scholar] [CrossRef] [Green Version]
  125. Hsu, C.N.; Lu, P.C.; Lo, M.H.; Lin, I.C.; Chang-Chien, G.P.; Lin, S.; Tain, Y.L. Gut Microbiota-Dependent Trimethylamine N-Oxide Pathway Associated with Cardiovascular Risk in Children with Early-Stage Chronic Kidney Disease. Int. J. Mol. Sci 2018, 19, 3699. [Google Scholar] [CrossRef] [Green Version]
  126. Swidsinski, A.; Loening-Baucke, V.; Schulz, S.; Manowsky, J.; Verstraelen, H.; Swidsinski, S. Functional anatomy of the colonic bioreactor: Impact of antibiotics and Saccharomyces boulardii on bacterial composition in human fecal cylinders. Syst. Appl. Microbiol. 2016, 39, 67–75. [Google Scholar] [CrossRef]
  127. Rath, S.; Rud, T.; Pieper, D.H.; Vital, M. Potential TMA-Producing Bacteria Are Ubiquitously Found in Mammalia. Front. Microbiol. 2019, 10, 2966. [Google Scholar] [CrossRef]
  128. Rath, S.; Heidrich, B.; Pieper, D.H.; Vital, M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 2017, 5, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Bergeron, N.; Williams, P.T.; Lamendella, R.; Faghihnia, N.; Grube, A.; Li, X.; Wang, Z.; Knight, R.; Jansson, J.K.; Hazen, S.L.; et al. Diets high in resistant starch increase plasma levels of trimethylamine-N-oxide, a gut microbiome metabolite associated with CVD risk. Br. J. Nutr. 2016, 116, 2020–2029. [Google Scholar] [CrossRef] [Green Version]
  130. Genoni, A.; Christophersen, C.T.; Lo, J.; Coghlan, M.; Boyce, M.C.; Bird, A.R.; Lyons-Wall, P.; Devine, A. Long-term Paleolithic diet is associated with lower resistant starch intake, different gut microbiota composition and increased serum TMAO concentrations. Eur. J. Nutr. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Griffin, L.E.; Djuric, Z.; Angiletta, C.J.; Mitchell, C.M.; Baugh, M.E.; Davy, K.P.; Neilson, A.P. A Mediterranean diet does not alter plasma trimethylamine N-oxide concentrations in healthy adults at risk for colon cancer. Food Funct. 2019, 10, 2138–2147. [Google Scholar] [CrossRef] [PubMed]
  132. Rohrmann, S.; Linseisen, J.; Allenspach, M.; von Eckardstein, A.; Muller, D. Plasma Concentrations of Trimethylamine-N-oxide Are Directly Associated with Dairy Food Consumption and Low-Grade Inflammation in a German Adult Population. J. Nutr. 2016, 146, 283–289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Koeth, R.A.; Lam-Galvez, B.R.; Kirsop, J.; Wang, Z.; Levison, B.S.; Gu, X.; Copeland, M.F.; Bartlett, D.; Cody, D.B.; Dai, H.J.; et al. l-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans. J. Clin. Invest. 2019, 129, 373–387. [Google Scholar] [CrossRef] [PubMed]
  134. Flanagan, J.L.; Simmons, P.A.; Vehige, J.; Willcox, M.D.; Garrett, Q. Role of carnitine in disease. Nutr. Metab. (London) 2010, 7, 30. [Google Scholar] [CrossRef] [Green Version]
  135. Warmbrunn, M.V.; Herrema, H.; Aron-Wisnewsky, J.; Soeters, M.R.; Van Raalte, D.H.; Nieuwdorp, M. Gut microbiota: A promising target against cardiometabolic diseases. Expert Rev. Endocrinol. Metab. 2020, 15, 13–27. [Google Scholar] [CrossRef] [Green Version]
  136. 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] [Green Version]
  137. More, M.I.; Swidsinski, A. Saccharomyces boulardii CNCM I-745 supports regeneration of the intestinal microbiota after diarrheic dysbiosis—A review. Clin. Exp. Gastroenterol. 2015, 8, 237–255. [Google Scholar] [CrossRef] [Green Version]
  138. Romano, K.A.; Vivas, E.I.; Amador-Noguez, D.; Rey, F.E. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. mBio 2015, 6, e02481. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Wu, W.K.; Chen, C.C.; Liu, P.Y.; Panyod, S.; Liao, B.Y.; Chen, P.C.; Kao, H.L.; Kuo, H.C.; Kuo, C.H.; Chiu, T.H.T.; et al. Identification of TMAO-producer phenotype and host-diet-gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut 2019, 68, 1439–1449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Xu, J.; Zhang, J.; Cai, S.; Dong, J.; Yang, J.Y.; Chen, Z. Metabonomics studies of intact hepatic and renal cortical tissues from diabetic db/db mice using high-resolution magic-angle spinning 1H NMR spectroscopy. Anal. Bioanal. Chem. 2009, 393, 1657–1668. [Google Scholar] [CrossRef] [PubMed]
  141. Wilson, A.; Teft, W.A.; Morse, B.L.; Choi, Y.H.; Woolsey, S.; DeGorter, M.K.; Hegele, R.A.; Tirona, R.G.; Kim, R.B. Trimethylamine-N-oxide: A Novel Biomarker for the Identification of Inflammatory Bowel Disease. Dig. Dis. Sci. 2015, 60, 3620–3630. [Google Scholar] [CrossRef]
  142. Yin, J.; Liao, S.X.; He, Y.; Wang, S.; Xia, G.H.; Liu, F.T.; Zhu, J.J.; You, C.; Chen, Q.; Zhou, L.; et al. Dysbiosis of Gut Microbiota With Reduced Trimethylamine-N-Oxide Level in Patients With Large-Artery Atherosclerotic Stroke or Transient Ischemic Attack. J. Am. Heart Assoc. 2015, 4. [Google Scholar] [CrossRef] [Green Version]
  143. Xu, K.Y.; Xia, G.H.; Lu, J.Q.; Chen, M.X.; Zhen, X.; Wang, S.; You, C.; Nie, J.; Zhou, H.W.; Yin, J. Impaired renal function and dysbiosis of gut microbiota contribute to increased trimethylamine-N-oxide in chronic kidney disease patients. Sci. Rep. 2017, 7, 1445. [Google Scholar] [CrossRef]
  144. Sitkin, S.I.; Tkachenko, E.I.; Vakhitov, T.Y. Metabolic Dysbiosis of the Gut Microbiota and Its Biomarkers. Eksp Klin. Gastroenterol. 2016, 12, 6–29. [Google Scholar]
  145. Ahmadmehrabi, S.; Tang, W.H.W. Gut microbiome and its role in cardiovascular diseases. Curr. Opin. Cardiol. 2017, 32, 761–766. [Google Scholar] [CrossRef]
  146. Cigarran Guldris, S.; Gonzalez Parra, E.; Cases Amenos, A. Gut microbiota in chronic kidney disease. Nefrologia 2017, 37, 9–19. [Google Scholar] [CrossRef] [Green Version]
  147. Jazani, N.H.; Savoj, J.; Lustgarten, M.; Lau, W.L.; Vaziri, N.D. Impact of Gut Dysbiosis on Neurohormonal Pathways in Chronic Kidney Disease. Diseases 2019, 7, 21. [Google Scholar] [CrossRef] [Green Version]
  148. Verhoog, S.; Taneri, P.E.; Roa Diaz, Z.M.; Marques-Vidal, P.; Troup, J.P.; Bally, L.; Franco, O.H.; Glisic, M.; Muka, T. Dietary Factors and Modulation of Bacteria Strains of Akkermansia muciniphila and Faecalibacterium prausnitzii: A Systematic Review. Nutrients 2019, 11, 1565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Chhibber-Goel, J.; Singhal, V.; Parakh, N.; Bhargava, B.; Sharma, A. The Metabolite Trimethylamine-N-Oxide is an Emergent Biomarker of Human Health. Curr Med. Chem. 2017, 24, 3942–3953. [Google Scholar] [CrossRef] [PubMed]
  150. Bae, S.; Ulrich, C.M.; Neuhouser, M.L.; Malysheva, O.; Bailey, L.B.; Xiao, L.; Brown, E.C.; Cushing-Haugen, K.L.; Zheng, Y.; Cheng, T.Y.; et al. Plasma choline metabolites and colorectal cancer risk in the Women’s Health Initiative Observational Study. Cancer Res. 2014, 74, 7442–7452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. 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] [Green Version]
  152. Obeid, R.; Awwad, H.M.; Rabagny, Y.; Graeber, S.; Herrmann, W.; Geisel, J. Plasma trimethylamine N-oxide concentration is associated with choline, phospholipids, and methyl metabolism. Am. J. Clin. Nutr. 2016, 103, 703–711. [Google Scholar] [CrossRef] [Green Version]
  153. Ottiger, M.; Nickler, M.; Steuer, C.; Odermatt, J.; Huber, A.; Christ-Crain, M.; Henzen, C.; Hoess, C.; Thomann, R.; Zimmerli, W.; et al. Trimethylamine-N-oxide (TMAO) predicts fatal outcomes in community-acquired pneumonia patients without evident coronary artery disease. Eur. J. Intern. Med. 2016, 36, 67–73. [Google Scholar] [CrossRef] [Green Version]
  154. Senthong, V.; Li, X.S.; Hudec, T.; Coughlin, J.; Wu, Y.; Levison, B.; Wang, Z.; Hazen, S.L.; Tang, W.H. Plasma Trimethylamine N-Oxide, a Gut Microbe-Generated Phosphatidylcholine Metabolite, Is Associated With Atherosclerotic Burden. J. Am. Coll. Cardiol. 2016, 67, 2620–2628. [Google Scholar] [CrossRef]
  155. 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. [Google Scholar] [CrossRef] [Green Version]
  156. 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. [Google Scholar] [CrossRef]
  157. Suzuki, T.; Heaney, L.M.; Bhandari, S.S.; Jones, D.J.; Ng, L.L. Trimethylamine N-oxide and prognosis in acute heart failure. Heart 2016, 102, 841–848. [Google Scholar] [CrossRef] [Green Version]
  158. Suzuki, T.; Heaney, L.M.; Jones, D.J.; Ng, L.L. Trimethylamine N-oxide and Risk Stratification after Acute Myocardial Infarction. Clin. Chem. 2017, 63, 420–428. [Google Scholar] [CrossRef] [PubMed]
  159. 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] [PubMed] [Green Version]
  160. Nie, J.; Xie, L.; Zhao, B.X.; Li, Y.; Qiu, B.; Zhu, F.; Li, G.F.; He, M.; Wang, Y.; Wang, B.; et al. Serum Trimethylamine N-Oxide Concentration Is Positively Associated With First Stroke in Hypertensive Patients. Stroke 2018, 49, 2021–2028. [Google Scholar] [CrossRef] [PubMed]
  161. Jaworska, K.; Hering, D.; Mosieniak, G.; Bielak-Zmijewska, A.; Pilz, M.; Konwerski, M.; Gasecka, A.; Kapłon-Cieślicka, A.; Filipiak, K.; Sikora, E.; et al. TMA, A Forgotten Uremic Toxin, but Not TMAO, Is Involved in Cardiovascular Pathology. Toxins (Basel) 2019, 11, 490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Reiner, M.F.; Muller, D.; Gobbato, S.; Stalder, O.; Limacher, A.; Bonetti, N.R.; Pasterk, L.; Mean, M.; Rodondi, N.; Aujesky, D.; et al. Gut microbiota-dependent trimethylamine-N-oxide (TMAO) shows a U-shaped association with mortality but not with recurrent venous thromboembolism. Thromb Res. 2019, 174, 40–47. [Google Scholar] [CrossRef]
  163. Hai, X.; Landeras, V.; Dobre, M.A.; DeOreo, P.; Meyer, T.W.; Hostetter, T.H. Mechanism of Prominent Trimethylamine Oxide (TMAO) Accumulation in Hemodialysis Patients. PLoS ONE 2015, 10, e0143731. [Google Scholar] [CrossRef]
  164. Kaysen, G.A.; Johansen, K.L.; Chertow, G.M.; Dalrymple, L.S.; Kornak, J.; Grimes, B.; Dwyer, T.; Chassy, A.W.; Fiehn, O. Associations of Trimethylamine N-Oxide With Nutritional and Inflammatory Biomarkers and Cardiovascular Outcomes in Patients New to Dialysis. J. Ren. Nutr. 2015, 25, 351–356. [Google Scholar] [CrossRef] [Green Version]
  165. 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] [Green Version]
  166. Missailidis, C.; Hallqvist, J.; Qureshi, A.R.; Barany, P.; Heimburger, 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] [Green Version]
  167. Al-Obaide, M.A.I.; Singh, R.; Datta, P.; Rewers-Felkins, K.A.; Salguero, M.V.; Al-Obaidi, I.; Kottapalli, K.R.; Vasylyeva, T.L. Gut Microbiota-Dependent Trimethylamine-N-oxide and Serum Biomarkers in Patients with T2DM and Advanced CKD. J. Clin. Med. 2017, 6, 86. [Google Scholar] [CrossRef] [Green Version]
  168. Shafi, T.; Powe, N.R.; Meyer, T.W.; Hwang, S.; Hai, X.; Melamed, M.L.; Banerjee, T.; Coresh, J.; Hostetter, T.H. Trimethylamine N-Oxide and Cardiovascular Events in Hemodialysis Patients. J. Am. Soc. Nephrol. 2017, 28, 321–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Heianza, Y.; Ma, W.; Manson, J.E.; Rexrode, K.M.; Qi, L. Gut Microbiota Metabolites and Risk of Major Adverse Cardiovascular Disease Events and Death: A Systematic Review and Meta-Analysis of Prospective Studies. J. Am. Heart Assoc. 2017, 6. [Google Scholar] [CrossRef] [PubMed]
  170. Qi, J.; You, T.; Li, J.; Pan, T.; Xiang, L.; Han, Y.; Zhu, L. Circulating trimethylamine N-oxide and the risk of cardiovascular diseases: A systematic review and meta-analysis of 11 prospective cohort studies. J. Cell Mol. Med. 2018, 22, 185–194. [Google Scholar] [CrossRef] [PubMed]
  171. 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] [PubMed]
  172. Li, P.; Zhong, C.; Li, S.; Sun, T.; Huang, H.; Chen, X.; Zhu, Y.; Hu, X.; Peng, X.; Zhang, X.; et al. Plasma concentration of trimethylamine-N-oxide and risk of gestational diabetes mellitus. Am. J. Clin. Nutr. 2018, 108, 603–610. [Google Scholar] [CrossRef] [PubMed]
  173. Huo, X.; Li, J.; Cao, Y.-F.; Li, S.-N.; Shao, P.; Leng, J.; Li, W.; Liu, J.; Yang, K.; Ma, R.; et al. Trimethylamine N-Oxide Metabolites in Early Pregnancy and Risk of Gestational Diabetes: A Nested Case-Control Study. J. Clin. Endocrinol. Metab. 2019, 104, 5529–5539. [Google Scholar] [CrossRef] [PubMed]
  174. Roy, S.; Yuzefpolskaya, M.; Nandakumar, R.; Colombo, P.C.; Demmer, R.T. Plasma Trimethylamine-N-oxide and impaired glucose regulation: Results from The Oral Infections, Glucose Intolerance and Insulin Resistance Study (ORIGINS). PLoS ONE 2020, 15, e0227482. [Google Scholar] [CrossRef]
  175. 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]
  176. Fu, B.C.; Hullar, M.A.J.; Randolph, T.W.; Franke, A.A.; Monroe, K.R.; Cheng, I.; Wilkens, L.R.; Shepherd, J.A.; Madeleine, M.M.; Le Marchand, L.; et al. Associations of plasma trimethylamine N-oxide, choline, carnitine, and betaine with inflammatory and cardiometabolic risk biomarkers and the fecal microbiome in the Multiethnic Cohort Adiposity Phenotype Study. Am. J. Clin. Nutr. 2020. [Google Scholar] [CrossRef]
  177. Shepshelovich, J.; Goldstein-Magal, L.; Globerson, A.; Yen, P.M.; Rotman-Pikielny, P.; Hirschberg, K. Protein synthesis inhibitors and the chemical chaperone TMAO reverse endoplasmic reticulum perturbation induced by overexpression of the iodide transporter pendrin. J. Cell Sci. 2005, 118, 1577–1586. [Google Scholar] [CrossRef] [Green Version]
  178. Castillo-Rodriguez, E.; Fernandez-Prado, R.; Esteras, R.; Perez-Gomez, M.V.; Gracia-Iguacel, C.; Fernandez-Fernandez, B.; Kanbay, M.; Tejedor, A.; Lazaro, A.; Ruiz-Ortega, M.; et al. Impact of Altered Intestinal Microbiota on Chronic Kidney Disease Progression. Toxins (Basel) 2018, 10, 300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Fernandez-Prado, R.; Esteras, R.; Perez-Gomez, M.V.; Gracia-Iguacel, C.; Gonzalez-Parra, E.; Sanz, A.B.; Ortiz, A.; Sanchez-Nino, M.D. Nutrients Turned into Toxins: Microbiota Modulation of Nutrient Properties in Chronic Kidney Disease. Nutrients 2017, 9, 489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Jia, J.; Dou, P.; Gao, M.; Kong, X.; Li, C.; Liu, Z.; Huang, T. Assessment of Causal Direction Between Gut Microbiota-Dependent Metabolites and Cardiometabolic Health: Abi-Directional Mendelian Randomisation Analysis. Diabetes 2019, 68, 1747–1755. [Google Scholar] [CrossRef]
  181. Mair, R.D.; Sirich, T.L.; Meyer, T.W. Uremic Toxin Clearance and Cardiovascular Toxicities. Toxins (Basel) 2018, 10, 226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-kappaB. J. Am. Heart Assoc. 2016, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M.; et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Boini, K.M.; Hussain, T.; Li, P.L.; Koka, S. Trimethylamine-N-Oxide Instigates NLRP3 Inflammasome Activation and Endothelial Dysfunction. Cell Physiol. Biochem. 2017, 44, 152–162. [Google Scholar] [CrossRef]
  185. 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] [Green Version]
  186. 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] [Green Version]
  187. Chen, S.; Henderson, A.; Petriello, M.C.; Romano, K.A.; Gearing, M.; Miao, J.; Schell, M.; Sandoval-Espinola, W.J.; Tao, J.; Sha, B.; et al. Trimethylamine N-Oxide Binds and Activates PERK to Promote Metabolic Dysfunction. Cell Metab. 2019, 30, 1141–1151.e1145. [Google Scholar] [CrossRef]
  188. Ufnal, M.; Jazwiec, R.; Dadlez, M.; Drapala, A.; Sikora, M.; Skrzypecki, J. Trimethylamine-N-oxide: A carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can. J. Cardiol. 2014, 30, 1700–1705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Li, T.; Chen, Y.; Gua, C.; Li, X. Elevated Circulating Trimethylamine N-Oxide Levels Contribute to Endothelial Dysfunction in Aged Rats through Vascular Inflammation and Oxidative Stress. Front. Physiol. 2017, 8, 350. [Google Scholar] [CrossRef] [PubMed]
  190. Snell, K. Renal disease of the rat. In Pathology of Laboratory Rats and Mice; Cotchin, E., Roe, F.J., Eds.; Blackwell Scientific: Oxford, UK, 1967; pp. 105–147. [Google Scholar]
  191. Yang, W.; Zhang, S.; Zhu, J.; Jiang, H.; Jia, D.; Ou, T.; Qi, Z.; Zou, Y.; Qian, J.; Sun, A.; et al. Gut microbe-derived metabolite trimethylamine N-oxide accelerates fibroblast-myofibroblast differentiation and induces cardiac fibrosis. J. Mol. Cell Cardiol. 2019. [Google Scholar] [CrossRef] [PubMed]
  192. Chen, P.Y.; Li, S.; Koh, Y.C.; Wu, J.C.; Yang, M.J.; Ho, C.T.; Pan, M.H. 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. J. Agric. Food Chem. 2019, 67, 7869–7879. [Google Scholar] [CrossRef] [PubMed]
  193. Li, X.; Sun, Y.; Zhang, X.; Wang, J. Reductions in gut microbiotaderived metabolite trimethylamine Noxide in the circulation may ameliorate myocardial infarctioninduced heart failure in rats, possibly by inhibiting interleukin8 secretion. Mol. Med. Rep. 2019, 20, 779–786. [Google Scholar] [CrossRef]
  194. Chen, Y.; Weng, Z.; Liu, Q.; Shao, W.; Guo, W.; Chen, C.; Jiao, L.; Wang, Q.; Lu, Q.; Sun, H.; et al. FMO3 and its metabolite TMAO contribute to the formation of gallstones. Biochim. Biophys Acta Mol. Basis Dis. 2019, 1865, 2576–2585. [Google Scholar] [CrossRef]
  195. John Hopkins University. Animal Care and Use Comittee.-The Mouse. Available online: http://web.jhu.edu/animalcare/procedures/mouse.html (accessed on 15 August 2019).
  196. The Jackson Laboratory. Body Weight Information fo B6.129P2-APOETM1UNC/J (002052). Available online: https://www.jax.org/jax-mice-and-services/strain-data-sheet-pages/body-weight-chart-002052 (accessed on 25 August 2019).
  197. Zhao, Y.; Yang, N.; Gao, J.; Li, H.; Cai, W.; Zhang, X.; Ma, Y.; Niu, X.; Yang, G.; Zhou, X.; et al. The Effect of Different l-Carnitine Administration Routes on the Development of Atherosclerosis in ApoE Knockout Mice. Mol. Nutr. Food Res. 2018, 62. [Google Scholar] [CrossRef]
  198. Koh, Y.C.; Li, S.; Chen, P.Y.; Wu, J.C.; Kalyanam, N.; Ho, C.T.; Pan, M.H. Prevention of Vascular Inflammation by Pterostilbene via Trimethylamine-N-Oxide Reduction and Mechanism of Microbiota Regulation. Mol. Nutr. Food Res. 2019, e1900514. [Google Scholar] [CrossRef]
  199. Liu, M.; Han, Q.; Yang, J. Trimethylamine-N-oxide (TMAO) increased aquaporin-2 expression in spontaneously hypertensive rats. Clin. Exp. Hypertens 2019, 41, 312–322. [Google Scholar] [CrossRef]
  200. 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. Invest. 2019, 99, 346–357. [Google Scholar] [CrossRef]
  201. Sun, G.; Yin, Z.; Liu, N.; Bian, X.; Yu, R.; Su, X.; Zhang, B.; Wang, Y. Gut microbial metabolite TMAO contributes to renal dysfunction in a mouse model of diet-induced obesity. Biochem. Biophys Res. Commun. 2017, 493, 964–970. [Google Scholar] [CrossRef] [PubMed]
  202. Martin, J.C.; Canlet, C.; Delplanque, B.; Agnani, G.; Lairon, D.; Gottardi, G.; Bencharif, K.; Gripois, D.; Thaminy, A.; Paris, A. 1H NMR metabonomics can differentiate the early atherogenic effect of dairy products in hyperlipidemic hamsters. Atherosclerosis 2009, 206, 127–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. 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] [PubMed]
  204. Empl, M.T.; Kammeyer, P.; Ulrich, R.; Joseph, J.F.; Parr, M.K.; Willenberg, I.; Schebb, N.H.; Baumgartner, W.; Rohrdanz, E.; Steffen, C.; et al. The influence of chronic L-carnitine supplementation on the formation of preneoplastic and atherosclerotic lesions in the colon and aorta of male F344 rats. Arch. Toxicol. 2015, 89, 2079–2087. [Google Scholar] [CrossRef] [PubMed]
  205. Weinert, C.H.; Empl, M.T.; Kruger, R.; Frommherz, L.; Egert, B.; Steinberg, P.; Kulling, S.E. The influence of a chronic L-carnitine administration on the plasma metabolome of male Fischer 344 rats. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef]
  206. Huc, T.; Drapala, A.; Gawrys, M.; Konop, M.; Bielinska, K.; Zaorska, E.; Samborowska, E.; Wyczalkowska-Tomasik, A.; Paczek, L.; Dadlez, M.; et al. 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]
  207. Lindskog Jonsson, A.; Caesar, R.; Akrami, R.; Reinhardt, C.; Fak Hallenius, F.; Boren, J.; Backhed, F. Impact of Gut Microbiota and Diet on the Development of Atherosclerosis in Apoe(-/-) Mice. Arter. Thromb Vasc. Biol. 2018, 38, 2318–2326. [Google Scholar] [CrossRef]
  208. 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, 2450–2462. [Google Scholar] [CrossRef]
  209. Aldana-Hernandez, 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. 2019. [Google Scholar] [CrossRef]
  210. Jawien, J.; Nastalek, P.; Korbut, R. Mouse models of experimental atherosclerosis. J. Physiol. Pharmacol. 2004, 55, 503–517. [Google Scholar]
  211. Bennett, B.J.; Davis, R.C.; Civelek, M.; Orozco, L.; Wu, J.; Qi, H.; Pan, C.; Packard, R.R.; Eskin, E.; Yan, M.; et al. Genetic Architecture of Atherosclerosis in Mice: A Systems Genetics Analysis of Common Inbred Strains. PLoS Genet. 2015, 11, e1005711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Wright, S.D.; Burton, C.; Hernandez, M.; Hassing, H.; Montenegro, J.; Mundt, S.; Patel, S.; Card, D.J.; Hermanowski-Vosatka, A.; Bergstrom, J.D.; et al. Infectious agents are not necessary for murine atherogenesis. J. Exp. Med. 2000, 191, 1437–1442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Stepankova, R.; Tonar, Z.; Bartova, J.; Nedorost, L.; Rossman, P.; Poledne, R.; Schwarzer, M.; Tlaskalova-Hogenova, H. Absence of microbiota (germ-free conditions) accelerates the atherosclerosis in ApoE-deficient mice fed standard low cholesterol diet. J. Atheroscler Thromb. 2010, 17, 796–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. 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.; et al. Non-lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Hsu, C.N.; Chang-Chien, G.P.; Lin, S.; Hou, C.Y.; Tain, Y.L. Targeting on Gut Microbial Metabolite Trimethylamine-N-Oxide and Short-Chain Fatty Acid to Prevent Maternal High-Fructose-Diet-Induced Developmental Programming of Hypertension in Adult Male Offspring. Mol. Nutr. Food Res. 2019, 63, e1900073. [Google Scholar] [CrossRef] [PubMed]
  216. DiNicolantonio, J.J.; Lavie, C.J.; Fares, H.; Menezes, A.R.; O’Keefe, J.H. L-carnitine in the secondary prevention of cardiovascular disease: Systematic review and meta-analysis. Mayo. Clin. Proc. 2013, 88, 544–551. [Google Scholar] [CrossRef] [Green Version]
  217. Song, X.; Qu, H.; Yang, Z.; Rong, J.; Cai, W.; Zhou, H. Efficacy and Safety of L-Carnitine Treatment for Chronic Heart Failure: A Meta-Analysis of Randomized Controlled Trials. Biomed. Res. Int. 2017, 2017, 6274854. [Google Scholar] [CrossRef] [Green Version]
  218. Xu, Y.; Jiang, W.; Chen, G.; Zhu, W.; Ding, W.; Ge, Z.; Tan, Y.; Ma, T.; Cui, G. L-carnitine treatment of insulin resistance: A systematic review and meta-analysis. Adv. Clin. Exp. Med. 2017, 26, 333–338. [Google Scholar] [CrossRef]
  219. He, K.; Song, Y.; Daviglus, M.L.; Liu, K.; Van Horn, L.; Dyer, A.R.; Greenland, P. Accumulated evidence on fish consumption and coronary heart disease mortality: A meta-analysis of cohort studies. Circulation 2004, 109, 2705–2711. [Google Scholar] [CrossRef] [Green Version]
  220. Mozaffarian, D.; Rimm, E.B. Fish intake, contaminants, and human health: Evaluating the risks and the benefits. JAMA 2006, 296, 1885–1899. [Google Scholar] [CrossRef] [Green Version]
  221. Takata, Y.; Zhang, X.; Li, H.; Gao, Y.T.; Yang, G.; Gao, J.; Cai, H.; Xiang, Y.B.; Zheng, W.; Shu, X.O. Fish intake and risks of total and cause-specific mortality in 2 population-based cohort studies of 134,296 men and women. Am. J. Epidemiol. 2013, 178, 46–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Aadland, E.K.; Lavigne, C.; Graff, I.E.; Eng, O.; Paquette, M.; Holthe, A.; Mellgren, G.; Jacques, H.; Liaset, B. Lean-seafood intake reduces cardiovascular lipid risk factors in healthy subjects: Results from a randomized controlled trial with a crossover design. Am. J. Clin. Nutr. 2015, 102, 582–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Alhassan, A.; Young, J.; Lean, M.E.J.; Lara, J. Consumption of fish and vascular risk factors: A systematic review and meta-analysis of intervention studies. Atherosclerosis 2017, 266, 87–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Estruch, R.; Ros, E.; Salas-Salvado, J.; Covas, M.I.; Corella, D.; Aros, F.; Gomez-Gracia, E.; Ruiz-Gutierrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef]
Figure 1. Disease network around cardiovascular disease and metabolic syndrome. Trimethylamine N-oxide (TMAO) can originate either directly from fish consumption or indirectly from intake of dietary precursors (e.g., L-carnitine, choline, or betaine). Dysbiosis has a major influence on cardiometabolic diseases or disease factors. During cardiovascular disease/type 2 diabetes, TMAO levels were found to increase to 4–12 µM in patients, possibly resulting from a disturbed microbiome and/or a decreased intestinal barrier. In the kidney, TMAO is rapidly excreted via the urine. Increased TMAO levels, as observed in studies with patients, may signal a decreased renal function. Unfavorable contribution of TMAO may take place at extremely high concentrations in patients with severely impaired renal function, e.g., hemodialysis patients (dotted arrow). Moderately increased TMAO levels may be a compensatory mechanism in diseased populations. Arrows pointing in two directions: both affect each other.
Figure 1. Disease network around cardiovascular disease and metabolic syndrome. Trimethylamine N-oxide (TMAO) can originate either directly from fish consumption or indirectly from intake of dietary precursors (e.g., L-carnitine, choline, or betaine). Dysbiosis has a major influence on cardiometabolic diseases or disease factors. During cardiovascular disease/type 2 diabetes, TMAO levels were found to increase to 4–12 µM in patients, possibly resulting from a disturbed microbiome and/or a decreased intestinal barrier. In the kidney, TMAO is rapidly excreted via the urine. Increased TMAO levels, as observed in studies with patients, may signal a decreased renal function. Unfavorable contribution of TMAO may take place at extremely high concentrations in patients with severely impaired renal function, e.g., hemodialysis patients (dotted arrow). Moderately increased TMAO levels may be a compensatory mechanism in diseased populations. Arrows pointing in two directions: both affect each other.
Nutrients 12 01330 g001
Figure 2. Main biochemical conversions leading to trimethylamine (TMA) and TMAO. Modified from [37,38]. The compounds with an asterisk can originate from the diet. The conversions within the grey box (gut microbiome) are induced by the microbiota (mainly Firmicutes, Proteobacteria), and the resulting TMA is absorbed within the colon and converted to TMAO by liver FMOs (flavin monooxygenases). The possibly partial microbial degradation of TMA and TMAO (by methylotrophs and other occasional bacteria (Pseudomonas/Bacillus)) can result in the formation of formaldehyde within the colon; also, methylamines (including TMA/TMAO) could be substrates for the formation of nitrosamines [32,39]. If TMA is absorbed from the colon, 95% of it is converted to TMAO, which is then excreted in the urine [34]. Arrows pointing in two directions: reactions go in both directions.
Figure 2. Main biochemical conversions leading to trimethylamine (TMA) and TMAO. Modified from [37,38]. The compounds with an asterisk can originate from the diet. The conversions within the grey box (gut microbiome) are induced by the microbiota (mainly Firmicutes, Proteobacteria), and the resulting TMA is absorbed within the colon and converted to TMAO by liver FMOs (flavin monooxygenases). The possibly partial microbial degradation of TMA and TMAO (by methylotrophs and other occasional bacteria (Pseudomonas/Bacillus)) can result in the formation of formaldehyde within the colon; also, methylamines (including TMA/TMAO) could be substrates for the formation of nitrosamines [32,39]. If TMA is absorbed from the colon, 95% of it is converted to TMAO, which is then excreted in the urine [34]. Arrows pointing in two directions: reactions go in both directions.
Nutrients 12 01330 g002
Figure 3. Search method for finding literature on TMAO. A total of 1963 references (up to November 2019) on TMAO were further searched with the indicated search terms. Additional references were found by following up citations within the literature and during the review phase in April 2020.
Figure 3. Search method for finding literature on TMAO. A total of 1963 references (up to November 2019) on TMAO were further searched with the indicated search terms. Additional references were found by following up citations within the literature and during the review phase in April 2020.
Nutrients 12 01330 g003
Table 1. TMAO levels in patients with cardiovascular diseases and renal dysfunction.
Table 1. TMAO levels in patients with cardiovascular diseases and renal dysfunction.
First Author, Year and CitationNumber of SubjectsStudy PopulationMean Age (Years)TMAO Level Median (or Mean) (µM)TMAO Interquartile Range (µM)Increased Disease Severity with Higher TMAO Levels
Tang, 2013 [136]720Patients with stable heart failure undergoing cardiac evaluation63.05.03.0–8.5yes
Koeth 2013 [94]2595Patients undergoing elective cardiac evaluation, GeneBank study624.6--yes
Bae 2014 [150]835Postmenopausal women with colorectal cancer within the Women’s Health Initiative Observational Study664.02.9–6.0yes
835Postmenopausal women healthy controls673.82.6–5.7NA
Lever 2014 [80]79Coronary Disease Cohort Study (CDCS), participants with T2D74.07.54.4–12.1yes
396CDCS participants without T2D68.04.83.0–9.1yes
Wang 2014 [82]3903Patients undergoing elective diagnostic coronary angiography633.72.4–6.2yes
Tang 2015 [151]112Patients with chronic systolic HF57.05.83.6–12.1yes
Obeid 2016 [152]283Subjects participating in a diabetes case-control study or a vitamin-supplementation trial66.74.36 **Not indicatedyes
Ottiger 2016 [153]317Community-acquired pneumonia patients72.03.01.7–5.4Yes
Rohrmann 2016 [132]104Healthy men50(geometric mean 2.55(95% CI 2.17–2.99)Only TNFα but not CRP and IL-6
167Healthy women44(geometric mean 2.52)(95% CI 2.22–2.86)
Senthong 2016 [154]353Atherosclerotic CAD patients65.05.53.4–9.8yes
Senthong 2016 [155]2235Patients with stable CAD who underwent elective coronary angiography63.03.82.5–6.5yes
Senthong 2016 [156]821Patients with peripheral artery disease664.82.9–8yes
Suzuki 2016 [157]972Patients with acute HF78.05.63.4–10.5yes
Suzuki 2017 [158]1079Acute MI patients673.74.6–6.4yes
Schugar 2017 [120]102Patients with elective cardiac risk factor evaluation with T2D55.94.83.3–7.7yes
333Patients with elective cardiac risk factor evaluation without T2D49.73.22.2–5.1NA
Tang 2017 [159]1216Patients with T2D who underwent elective diagnostic coronary angiography64.44.42.8–7.7yes
300Apparently healthy controls53.63.62.3–5.7yes
Nie 2018 [160]622Hypertensive stroke patients62.22.51.6–4.0yes
622Matched controls62.22.31.4–3.7NA
Jaworska 2019 [161]19Cardiovascular patients qualified for aortic valve replacement74.55.5 ± 0.6--No
9Healthy control38.93.6 ± 0.4--NA
Reiner 2019 [162]859Venous thromboembolism patients75.0--2.28–6.57U-shaped optimum level at 4 µM
Hai 2015 [163]7Hemodialysis patientsNot indicated(mean 77 ± 26)Not indicatedNot indicated
6Control subjectsNot indicated(mean 2 ± 1)Not indicatedNA
Kaysen 2015 [164]235Patients new to hemodialysis61.84327.5–66.6no
NATwo commercially available pooled control samplesNot indicated(mean 1.41 ± 0.49)NANA
Tang 2015 [165]521CKD patients707.95.2–12.4yes
3166Control subjects623.42.3–5.3NA
Missailidis 2016 [166]56CDK 3‒4 patients4214.6 *5.6–71.2yes
55CDK 5 patients on hemodialysis starting renal replacement7473.5 *26.4–191.0
80Controls625.8 *3.1–13.3NA
Al-Obaide 2017 [167]20Diabetic CKD patients64.412.5
(mean 20.2)
9.9–22.9yes
20Healthy controls54.31.2
(mean 2.4)
0.68–4.5NA
Shafi 2017 [168]1232Hemodialysis patients, 35% Caucasians and 65% African American58White 87
Black 88
(total mean 101.9 ± 63.9)
63–120
62–125
Yes in Caucasians
No in African Americans
IQR, interquartile range; * high level in healthy control group; ** 98 T2D patients with a mean TMAO plasma level of 8.6 ± 12.2 µM, compared to 5.4 ± 5.2 μM among 185 T2D free subjects.
Table 2. Examples of preclinical evidence for TMAO as a cardiometabolic risk factor ***.
Table 2. Examples of preclinical evidence for TMAO as a cardiometabolic risk factor ***.
Literature SourceTest SystemTreatmentOutcomeModel and SupplementationDaily Dosage of TMAO or Precursors *Plasma TMAO Level *
(or Control Level)
Wang 2011 [115]Atherosclerosis-prone male and female mice (apoE−/−)Control or diet (around 0.08% choline) or diet with 0.5% or 1.0% additional choline or 0.12% TMAO; from weaning–20 weeks of age (16 weeks treatment)Choline supplementation groups resulted in increased TMAO levels, with correlation between plasma TMAO and atherosclerotic plaque size.
Female mice had higher FMO activity and higher TMAO levels upon supplementation, however plaques were observed similarly in both genders, introducing a confounding factor.
Parallel examination of plasma cholesterol, triglycerides, lipoproteins, glucose levels, and hepatic triglyceride content in the mice failed to show significant increases that could account for the enhanced atherosclerosis.
apoE−/− mice;
high choline/TMAO supplementation
800 or 1600 mg choline/kg **
or
192 mg TMAO/kg **
Males/females
w/0.5% choline:
13 µM/140 µM;
w/1% choline:
24 µM/215 µM
w/0.12% TMAO:
23 µM/70 µM
(5 µM/10 µM controls)
Atherosclerosis-prone mice (apoE−/−)
Gender not clearly indicated (Figure S7b)
Control or diet or diet with 1.0% additional choline or 0.12% or TMAO or 1% betaine; for at least 3 weeks
half of them +antibiotics, for 3 weeks
Mice supplemented with either choline, TMAO, or betaine showed enhanced levels of scavenger receptors CD36 and SR-A1 (which bind to lipoproteins), markers for activated macrophages.
Increase in scavenger receptors was inhibited by antibiotics.
Mice on the control diet showed modest foam cell formation; 1% choline supplemented diet resulted in enhanced lipid-laden macrophages (foam cells). Foam cell formation was inhibited by antibiotics.
High choline or betaine supplementation1600 mg choline/kg **
or
192 mg TMAO/kg **
or 1600 mg betaine/kg **
50 µM (4 h after challenge w/d9(trimethyl)-choline
(baseline 0 µM)
TMAO conc. after other supplementation not indicated
Koeth 2013 [94]Atherosclerosis-prone female mice (apoE−/−)1.3% L-carnitine in drinking water (or control water) +/− antibiotics; standard chow, for at least 4 weeksSignificant increase in plasma TMA and TMAO in the L-carnitine group without antibiotics; negligible concentrations in the other groups.
L-carnitine did not increase other pro-atherogenic biomarkers (plasma lipids, lipoproteins, glucose, or insulin levels) in the plasma.
1.8-fold increase of aortic root lesion size in the L-carnitine group without antibiotics compared to the control, other groups similar to control. The difference between supplemented group and control was only 3 mice.
apoE−/− mice;
high L-carnitine supplementation
2080 mg L-carnitine/kg **130 µM
(10 µM)
Atherosclerosis-prone female mice (apoE−/−)1.3% L-carnitine diet or 1.3% choline in diet; +/− antibiotics; standard chow,
for 8 weeks
TMA/TMAO production abolished with antibiotics.
TMAO was shown to inhibit reverse cholesterol transport in aortic cells.
apoE−/− mice;
high L-carnitine or choline supplementation
2080 mg L-carnitine/kg **
2080 mg choline/kg **
190 µM-w/carnitine
110 µM-w/choline
(10 µM)
Atherosclerosis-prone female mice (apoE−/−)Supplementation of diet with 0.12% TMAO for 4 weeksTMAO was shown to inhibit reverse cholesterol transport and reduce the expression of Cyp7a1 (enzyme involved in bile acid synthesis and cholesterol metabolism).apoE−/−;
TMAO supplementation
192 mg TMAO/kg **35 µM
(9 µM)
Ufnal 2014 [188]Rats
gender not relevant
Osmotic pump infusion with saline, TMAO, low-dose Angiotensin II, or bothTMAO did not affect blood pressure in normotensive animals. However, it prolonged the hypertensive effect of Angiotensin II.High TMAO/angiotensinOsmotic pump infusion with TMAOTMAO: 58 µM
(0.57 µM)
Seldin 2016 [182]Female LDLR(-/-) mice “Chow with 1.3% choline provided ad libitum in drinking water” or controlAortas of LDLR(-/-) mice fed a choline diet showed elevated inflammatory gene expression compared with controls.LDLr(-/-);
high choline supplementation
2080 mg choline/kg **55 µM
(9 µM)
Tang 2015 [165]Male miceDiet with 1.0% choline or 0.12% TMAO, or control for 6 weeks or 16 weeksElevated TMAO levels were associated with increases in tubulointerstitial fibrosis, collagen deposition and phosphorylation of Smad3 (regulator of the pro-fibrotic TGF-β/Smad3 signaling pathway). TMAO-fed and choline-fed mice experienced increased kidney injury marker-1.
After 16 weeks increased serum cystatin C levels compared to chow-fed mice were observed. Altogether, dose-dependent relationships were noted between plasma TMAO levels and monitored indices of renal histopathological and functional impairment.
High choline or TMAO diet1600 mg choline/kg **
or
192 mg TMAO/kg **
100 µM-w/choline
40 µM-w/TMAO
(5 µM control)
Zhu 2016 [183]Female miceDiet supplemented with either 0.12% TMAO or 1% choline or control; antibiotic control for 6 weeksCholine or TMAO supplementation led to increased TMAO levels. Ex vivo platelet aggregation (induced by ADP stimulation) was significantly increased in these groups; antibiotics suppressed this effect for the choline supplemented mice (not the TMAO supplemented mice).High choline supplementation192 mg TMAO/kg **
1600 mg choline/kg
Not indicated
Boini 2017 [184]Male mice with partially ligated carotid arteryOsmotic pump infusion with TMAO (dosage not apparent) or control for 2 weeks post ligationMice with partially ligated carotid artery and infused with TMAO for 2 weeks had increased inflammasome formation.Partially ligated carotid artery;
TMAO infusion
Osmotic pump infusion with TMAO (dosage not apparent)Not indicated
Li 2017 [189]Male Fischer 344 rats
(Most Fischer 344 rats older than 2 years exhibit small local areas of nephritis; less than 25% show severe nephritis [190])
Young or old (22 months) rats treated with 1% DMB in the drinking water for 8 weeks; controlsCompared with the young control group, the old control group had higher plasma TMAO levels. In both age groups DMB reduced TMAO levels.
Old aortae exhibited increased expression of proinflammatory cytokines and superoxide production and decreased expression of endothelial nitric-oxide synthase (eNOS), all of which were restored by DMB treatment.
Old age: causes renal problems with these type of rats [190]DMB treatment to inhibit TMAO formationOld rats: 14.3 µM
(reduced by DMB to 6.0 µM)
(young rats 6.4 µM; reduced by DMB to 3.9 µM)
Yang 2019 [191]Coronary ligation to induce myocardial infarction (MI), or sham operation in male miceMice were fed a control diet, high choline diet (1.2%) or/and DMB (choline analogue and inhibitor) diet or a TMAO diet (0.12%) starting 3 weeks before MI; treatment for one more week after MICardiac fibrosis increased with 0.12% TMAO or 0.24% TMAO, but not with 0.06% TMAO.
In the MI model, TMAO or choline supplementation led to decreased cardiac function and cardiac fibrosis. DMB inhibited the TMAO-effect.
Choline or TAMO treated MI animals transformed fibroblasts into myofibroblasts and activated the TGF-βRI/Smad2 pathway, indicating cardiac fibrosis.
MI, choline or high TMAO or choline supplementation1920 mg choline/kg or 192 mg TMAO/kg **Before MI:
42.4 µM w/TMAO
(8.8 µM control)
before MI:
91.6 µM w/choline
(6.1 µM control; 27.8 µM w/choline + DMB)
Chen 2019 [192]Female miceMice treated with 1.3% L-carnitine in drinking water (drinking volume and body mass provided), flavonoids (or oolong tea)Mice treated with L-carnitine significantly increased plasma TMAO levels compared to control. L-carnitine also increased inflammation markers.
TMAO was remarkedly reduced by flavonoids. Antibiotics strongly reduced TMAO production.
Inflammation markers (TNF-α, E-selectin, and VCAM-1) were reduced by flavonoids or oolong tea or antibiotics.
High L-carnitine → high TMAO 2941 mg L-carnitine in L-carnitine group; 2657 mg L-carnitine in flavonoid group; 2424 mg L-carnitine in antibiotics group400 µM w/L-carnitine
(26 µM with no diet; 322 µM with flavonoids; 13 µM with antibiotics)
Li 2019 [193]Male rats with coronary ligation to induce MI or sham operationMI and sham rats treated with either vehicle (tap water) or 1.0% DMB in tap water, for 8 weeksPlasma TMAO levels were elevated in vehicle-treated MI rats compared with vehicle-treated sham rats; plasma TMAO levels were reduced in DMB-treated MI rats.
Manifestations of MI-induced heart failure were improved in DMB-treated MI rats. Elevated plasma TMAO levels went along with increased proinflammatory IL-8 plasma level in MI groups. In sham rats, DMB treatment reduced plasma TMAO but did not alter other parameters.
Coronary ligation → myocardial infarctionDMB treatment to inhibit TMAO formation30 µM w/o DMB
(Sham: 12 µM w/o DMB)
(MI & DMB: 10 µM)
(Sham & DMB: 3 µM)
Chen 2019 [194]Male mice1% cholesterol diet with/without 1% cholinePlasma TMAO levels increased about 4-fold with diet + choline; increased mRNA of cholesterol uptake and secretion genes (Abcg5 and g8, Ldlr); no difference in bile acid composition.High cholesterol diet, high choline1600 mg choline/kg **7.7 µM
(1 µM)
Male mice1% cholesterol diet and supplemented
with low dose (0.12%) or high dose (0.3%) TMAO
No pathological differences in liver tissue; cholesterol concentration in gallbladder bile increased with TMAO, more apparent at high dose; increased mRNA of cholesterol uptake and secretion genes (Abcg5 and g8, Ldlr, Srb1).High cholesterol diet, TMAO supplementation192 mg TMAO/kg or 480 mg TMAO/kgNot indicated
Gallstone-susceptible AKR/J male mice (biliary cholesterol hypersecretion)Lithogenic diet supplemented with 0.3% TMAO or not supplemented (control).With TMAO, the incidence of gallstones rose to 70%, compared no gallstones in the control mice. TMAO also induced increased hepatic Abcg5 and g8 expression.Lithogenic diet;
gallstone-susceptible AKR/J mice (cholesterol hypersecretion); high TMAO supplementation
480 mg TMAO/kg23,3 µM
(1 µM lithogenic diet)
* Plasma TMAO levels consecutive to the indicated intervention. In the case of multiple dosing levels, it corresponds to the minimum dosage for unfavourable effects and means that indicated dosages resulted in significant, relevant effects. Several concentrations are approximations derived from graphical illustrations, e.g., bar graphs (numerical values were not always published). Some baseline values are rather high, possibly indicating alternate methodology (see also section on methodology). ** Calculated from dietary intake as follows: The average daily consumption of feed and water for an adult 25 g mouse is 3–5 g (averaged to 4 g) and 4 mL (= 4 g) respectively [195]; 0.12% TMAO = 1,2 mg/g; → average daily dosage with 4 g intake: 4.8 mg. Male adult apoE−/− mice [196] weight around 30 g. Female adult apoE−/− mice weight around 20 g. It needs to be assumed that males eat and drink more than females. Thus, taking an average mouse weight of 25 g will be a good approximation for both genders. → Daily dosing would be approximately 4.8 mg/25 g body mass (bm) = 4.8 mg/0.025 kg bm → 192 mg TMAO/kg bm. 1% choline = 10 mg/g; → average daily dosage with 4 g intake: 40 mg → daily dosing 40 mg/25 g bm= 40 mg/0.025 kg bm → 1600 mg choline/kg bm. 1.3% L-carnitine = 13 mg/g; → average daily dosage with 4 g intake: 52 mg → daily dosing 52 mg/25 g bm = 52 mg/0.025 kg bm → 2080 mg L-carnitine/kg bm. Other calculations: if 352 mg/kg L-carnitine is equivalent to 2000 mg/day in humans [185], 2080 mg/kg L-carnitine would be equivalent to 11818 mg/day in humans (factor 5.9). *** Literature examples were randomly selected in the sequence as discovered during the search. Further literature appears to be similar in presenting adverse effects coupled to distress of experimental animals and rather high TMAO concentrations, e.g., [197,198,199,200,201].
Table 3. Preclinical evidence for cardio-protective or neutral effects of TMAO.
Table 3. Preclinical evidence for cardio-protective or neutral effects of TMAO.
Literature SourceTest SystemTreatmentOutcomeDistress FactorsDaily Dosage of TMAO or Precursors *Plasma TMAO Level *
(or Control Level)
Mayr 2005 [114]Male and female apoE−/− and apoE+/+mice on normal chow dietProteomics and metabolomics
to identify protein and metabolite changes in vessels
No significant difference in TMAO concentration in the aortas of 18-month-old apoE−/− and ApoE+/+mice.
Lesion formation in apoE−/− mice due to increase in oxidative stress, not to increased TMAO.
apoE−/− micenoneFemales 0.06
Males 0.25
(units not stated, controls not stated)
Martin 2009 [202]Male hamstersHyperlipidemic diet (normal diet plus 100 g/kg fat for 5 weeks + 200 g/kg for 12 weeks; fat as anhydrous butter or cheese) or controls; (1)H NMR-based metabonomics VLDL lipids, cholesterol, and N-acetylglycoproteins had the best correlation to onset of atherosclerosis.
TMAO was found to be negatively associated with atherogenesis.
High-fat dietnoneAbsolute concentrations not determined (only relative ones; personal communication)
Gao 2014 [203]Male miceControl, diet with 25% fat +/− 0.2% TMAO for 4 weeksDietary TMAO increased fasting insulin levels and insulin resistance and exacerbated impaired glucose tolerance and MCP-1 mRNA (pro-inflammatory cytokine) in HFD-fed mice.
IL-10 mRNA (anti-inflammatory cytokine) in adipose tissue was (more than already by the high-fat diet) decreased by TMAO.
However, the increase in atherosclerosis associated with a high-fat diet was prevented by TMAO, suggesting a protective effect of TMAO with regard to atherosclerosis.
High-fat diet;
TMAO supplementation
320 mg TMAO/kg **17.5 µM
(normal chow 11.9 µM; high-fat chow 12 µM)
Shih 2015 [118]Male mice, transgenic FMO3 overexpressionTransgenic compared to control mice, supplemented with water containing 1.3% choline chloride for 6 weeksFMO3 overexpression caused a 75% increase in plasma TMAO levels and increased hepatic and plasma lipids.FMO3 overexpression; high choline supplementationFMO3 overexpression;
2080 mg choline/kg **
16 µM
(9 µM for control transgene)
Shih 2015 [29,118]Male hyperlipidemic
mouse “E3L Tg” with transgenic FMO3 overexpression
Transgenic compared to control mice; low-fat or high-fat/1% cholesterol chow for 16 weeksIncreased plasma TG, VLDL/IDL/LDL, and unesterified cholesterol with both diets, increased glucose and insulin levels, increased levels of TG, TC, and phosphatidylcholine in the VLDL plasma fractions with high-fat diet.
Small increase (20%) in atherosclerotic lesion size compared to knockdown mouse (see above); it is implausible to explain the effect with the slight change in TMAO levels.
Hyperlipidemic
mouse with transgenic FMO3 overexpression; high-fat/high-cholesterol chow
High-fat dietw/high-fat/cholesterol:
2.6 µM
(2.2 µM for control transgene) (difference = trend)
Collins 2016 [185]Male apoE−/− mice expressing human cholesteryl ester transfer protein (hCETEP)12 week treatment with L-carnitine (87 mg/kg and 352 mg/kg; equivalent to 500 and 2000 mg/day in humans);
(and/or methimazole =FMO inhibitor)
High doses of L-carnitine resulted in a significant increase in plasma L-carnitine and TMAO levels. Plasma lipid and lipoprotein levels did not change.
Aortic root analysis showed significant decrease in lesion size with L-carnitine treatment and high TMAO compared to the control.
Significantly lower levels of lesion were found with elevated plasma TMAO (>0.05 ppm) compared to the low plasma TMAO (<0.05 ppm). TMAO may be protective against atherosclerosis development.
apoE−/− mice (plus hCETEP)352 mg L-carnitine/kg0.2 ppm = 2.7 µM
(0.08 ppm = 1.07 µM)
Empl 2015 [204]Male Fischer 344 ratsDaily 0, 0.1, 0.2 or 0.5 g/L L-carnitine (0; 70; 141; 352 mg/kg) via drinking water for one yearL-carnitine did not cause any preneoplastic, atherosclerotic, or other lesions.None352 mg L-carnitine/kg (highest dosage)See below
Weinert 2017 [205]Male Fischer 344 rats from study [204]Daily 0, 0.1, 0.2 or 0.5 g/L L-carnitine (0; 70; 141; 352 mg/kg) via drinking water for one yearHigh dose L-carnitine resulted in tenfold higher plasma TMAO concentration compared to the control (25.0 μM).
Supplementation did not cause changes in the plasma metabolome.
None352 mg L-carnitine/kg (highest dosage)25.0 μM
(2.5 µM for control)
Huc 2018 [206]Male spontaneously hypertensive rats (SHR) with pressure-overloaded hearts, 2 age groupsTMAO 333 mg/L with drinking water, water controls and normotensive rat controls for 9 weeks; metabolic cage for 2 days at end of studyChronic, low-dose trimethylamine oxide (TMAO) treatment increased plasma TMAO by 4 to 5-fold and reduced plasma NH2-terminal pro-B-type natriuretic peptide and vasopressin, left ventricular end-diastolic pressure, and cardiac fibrosis. TMAO may be beneficial for reduction of hypertension.Hypertension37 (16 weeks)/32 mg (56 weeks) TMAO/kg (personal communication)16 weeks: 37.3 µM
(SHR control: 8.8 µM)
(normal control: 6.3 µM)
56 weeks: 40.9 µM
(SHR control: 8.1 µM)
(normal control: 5.2 µM)
Lindskog Jonsson 2018 [207]Male germ-free or conventionally raised apoE−/− miceWestern diet alone or supplemented with 1.2% choline for 12 weeksConventionally raised mice had smaller
aortic lesions and lower plasma cholesterol levels on a chow diet compared to a Western diet; choline supplementation increased plasma TMAO levels in conventionally raised mice but not in germ-free mice. However, choline supplementation did not affect the size of aortic lesions or plasma cholesterol levels; increased plasma TMAO levels observed in conventionally raised apoE−/− mice fed a choline-supplemented diet did not correlate with aortic lesion size. The microbiota was required for TMAO production from dietary choline, but this process could not be linked to increased atherosclerosis. Choline supplementation reduced body weight and epididymal fat weight in conventionally but not germ-free mice when fed Western diet.
Western diet/germ-free existence; apoE−/− mice1920 mg choline/kg **Conventionally raised:
Western diet +choline: 8 µM
Chow +choline: 21 µM
(Western diet: 0.5 µM
Chow: 1 µM)
No TMAO in germ-free mice
Zhao 2019 [208]Male rats with steatohepatitis induced by high-fat high-cholesterol diet16-wk high-fat high-cholesterol (HFHC) diet feeding; daily TMAO (120 mg/kg/day) by oral gavage for 8 weeksHepatic and serum levels of cholesterol were both decreased by TMAO treatment in rats on HFHC diet.
TMAO treatment also downregulated cholesterol influx-related Niemann-Pick C1-like 1 (intestinal cholesterol absorption transporter) and upregulated cholesterol efflux-related ABCG5/8 in the small intestine; thus, TMAO inhibits intestinal cholesterol absorption.
Gut microbiota analysis showed that TMAO could alter the gut microbial profile and restore the diversity of gut flora.
TMAO also ameliorated hepatic ER stress and cell death under cholesterol overload, thereby attenuating HFHC diet-induced steatohepatitis in rats.
Steatohepatitis induced by high-fat high-cholesterol diet120 mg TMAO/kgNot determined (personal communication)
Aldana-Hernandez 2019 [209]Male Ldlr-/- mice
and
male apoE−/− mice
40% high-fat diet for atherosclerosis induction; control (0.1% choline) or supplemented with
1% choline or
0.9% betaine or
0.2% TMAO;
for 8 or 16 weeks
control diet (0.1% choline) or diet for ≤28 weeks supplemented with
1% choline or
0.9% betaine or
0.12% TMAO
for 12 or 28 weeks
In LDLr-/- mice, dietary supplementation for 8 wk with choline or TMAO increased plasma TMAO concentrations by 1.6-and 4-fold, respectively. After 16 wk, there was a 2-fold increase in plasma TMAO after dietary TMAO supplementation.
In apoe-/- mice, dietary supplementation with choline, betaine, or TMAO for 12 wk did not increase plasma TMAO concentrations. However, choline and TMAO supplementation for 28 wk significantly increased plasma TMAO concentrations by 1.8-and 1.5-fold, respectively.
Atherosclerotic lesion size was not altered by any of the dietary interventions, irrespective of mouse model.
Ldlr-/- mice (40% high-fat diet)
apoE−/− mice
1600 mg choline/kg (1%) **
1440 mg betaine/kg (0.9%) **
320 mg TMAO/kg (high TMAO dosage of 0.2%) **
192 mg TMAO/kg (low TMAO dosage of 0.12%) **
LDLr-/- mice (8 weeks/16 weeks):
w/choline: 1.3 µM/1.8 µM
w/betaine: 0.9 µM/1.1 µM
w/TMAO: 2.5 µM/1.1 µM
(0.5-1.1 µM for controls)
apoE−/− mice (12 weeks/28 weeks):
w/choline: 1.3µM/1.6 µM
w/betaine: 0.8 µM/0.8 µM
w/TMAO: 0.9 µM/1.3 µM
(1.0 µM/0.8 µM for controls)
* Plasma TMAO level consecutive to the indicated intervention. In the case of multiple dosing levels, it corresponds to the maximum dosage for beneficial/neutral effects and means that they are dosages that result in significant, relevant effects. Several concentrations are approximations derived from graphical illustrations, e.g., bar graphs (numerical values were not always published). Some baseline values are rather high, possibly indicating alternate methodology (see also section on methodology). ** See footnote ** of Table 2 for calculation.

Share and Cite

MDPI and ACS Style

Papandreou, C.; Moré, M.; Bellamine, A. Trimethylamine N-Oxide in Relation to Cardiometabolic Health—Cause or Effect? Nutrients 2020, 12, 1330. https://doi.org/10.3390/nu12051330

AMA Style

Papandreou C, Moré M, Bellamine A. Trimethylamine N-Oxide in Relation to Cardiometabolic Health—Cause or Effect? Nutrients. 2020; 12(5):1330. https://doi.org/10.3390/nu12051330

Chicago/Turabian Style

Papandreou, Christopher, Margret Moré, and Aouatef Bellamine. 2020. "Trimethylamine N-Oxide in Relation to Cardiometabolic Health—Cause or Effect?" Nutrients 12, no. 5: 1330. https://doi.org/10.3390/nu12051330

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