Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3861 2023-02-09 09:57:26 |
2 layout Meta information modification 3861 2023-02-09 10:00:51 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tacconi, E.;  Palma, G.;  Biase, D.D.;  Luciano, A.;  Barbieri, M.;  Nigris, F.D.;  Bruzzese, F. Trimethylamine N-Oxide Production in Physiological and Pathological Conditions. Encyclopedia. Available online: https://encyclopedia.pub/entry/41024 (accessed on 06 July 2024).
Tacconi E,  Palma G,  Biase DD,  Luciano A,  Barbieri M,  Nigris FD, et al. Trimethylamine N-Oxide Production in Physiological and Pathological Conditions. Encyclopedia. Available at: https://encyclopedia.pub/entry/41024. Accessed July 06, 2024.
Tacconi, Edoardo, Giuseppe Palma, Davide De Biase, Antonio Luciano, Massimiliano Barbieri, Filomena De Nigris, Francesca Bruzzese. "Trimethylamine N-Oxide Production in Physiological and Pathological Conditions" Encyclopedia, https://encyclopedia.pub/entry/41024 (accessed July 06, 2024).
Tacconi, E.,  Palma, G.,  Biase, D.D.,  Luciano, A.,  Barbieri, M.,  Nigris, F.D., & Bruzzese, F. (2023, February 09). Trimethylamine N-Oxide Production in Physiological and Pathological Conditions. In Encyclopedia. https://encyclopedia.pub/entry/41024
Tacconi, Edoardo, et al. "Trimethylamine N-Oxide Production in Physiological and Pathological Conditions." Encyclopedia. Web. 09 February, 2023.
Trimethylamine N-Oxide Production in Physiological and Pathological Conditions
Edit

Trimethylamine N-oxide (TMAO) is an amino oxide, produced from the trimethylamine (TMA) through oxidation by some liver enzymes called flavin monooxygenases 1 and 3 (FMO1 and FMO3).

trimethylamine N-oxide (TMAO) trimethylamine (TMA) gut microbiota diet

1. Introduction

The interest in human microbiota and its modulation of interactions between food sources and some pathologies such as the metabolic syndrome, cardiovascular disease and some types of cancer has been growing in the scientific world. Even if there is a long way to go, our attention has been directed to a molecule involved in systemic inflammation, called trimethylamine N-Oxide (TMAO) [1][2][3]. TMAO is an amino oxide, produced from the trimethylamine (TMA) through oxidation by some liver enzymes called flavin monooxygenases 1 and 3 (FMO1 and FMO3). Three forms of the enzyme, FMO1 found in fetal liver, FMO3 found in adult liver, and genes clustered in the 1q23-q25 region encode FMO3. Flavin-containing monooxygenases are NADPH-dependent flavoenzymes that catalyze the oxidation of soft nucleophilic heteroatom centers in xenobiotics, such as pesticides and drugs. The human FMO3 enzyme catalyzes several types of reactions, including the N-oxygenation of primary, secondary, and tertiary amines [4]. TMA production is indirectly influenced by some specific compounds such as L-carnitine, choline and other isoforms, betaine, and lecithin, and directly from gamma-butyrobetaine [2][5][6]. These compounds are metabolized in the gut through interactions with some of microbiota bacteria by different enzymes. First, the major clusters of TMA production start from the mouth with Streptococcus sanguis and the genes CutC and CutD, that are necessary for Desulfovibrio and Desulfovibrio desulfuricans to convert choline in TMA [7]. Other genes such as CntA and CntB contained in Actinobacter and Serratia, promote the oxidoreductase enzymes from L-carnitine to TMA. Furthermore, YeaW and YeaX are involved in oxygenase and oxidoreductase enzymes for choline, betaine, L-carnitine and gamma-butyrobetaine. Bacteria from gamma-proteobacteria like Escherichia coli, Citrobacter, Klebsiella pneaumoniae, and Shigella, Achromobacter from the strain of Betaproteobacteria, Sporosarcina from Firmicutes, and Actinobacteria have orthologue and homologue enzymes such as CntA, CntB, YeaW and YeaX, which encode the gene that can convert all food compounds including choline, betaine, lecithin, gamma-butyrobetaine, ergothioneine and L-carnitine into TMA [7].
Choline, choline esters (e.g., phosphatidylcholine) and lecithin, all important and essential nutrients for the nervous system health [8][9], are converted in TMA by TMA lyase; also, choline can be oxidase in betaine by choline dehydrogenase and choline aldehyde dehydrogenase, which is converted to TMA by betaine oxidoreductase [10]. Furthermore, lecithin, which is a source of choline, can be re-converted to choline by phospholipase D and again converted to TMA [9].
L-carnitine is oxidized directly to TMA by the carnitine oxidoreductase, and indirectly from gamma-butyrobetaine [11]. L-carnitine can be converted by gamma-butyrobetaine hydroxylase in gamma-butyrobetaine, and eventually in TMA by TMA lyase [12]. When the conversion from TMA to TMAO is inhibited, it manifests a particular metabolic syndrome called trimethylaminuria, or “fish odor syndrome”, due to the accumulation of TMA molecules excreted in the urine, sweat and breath which smell like putrid fish [13]. This pathology occurs with a mutation in the gene encoding the liver enzyme FMO3 [14], single nucleotide polymorphism E158K and E308G, which has 10-fold higher specific activity to convert TMA in TMAO then the FMO1 [15]. In some patients, low choline food sources have been recommended [16], but a promising future therapy is to intervene on gut microbiota to modulate the production of TMA and TMAO [17]. Strong evidence correlates high levels of L-carnitine with high levels of TMAO and its potential pro-atherogenic role [18][19] and its role was confirmed by a meta-analysis showing that all causes of mortality increased by 7% per each 10 µmol/L increment of circulating TMAO [20]. Several studies reported a remarkable increment of TMAO achieved by supplementation of L-carnitine, but some of them observed alterations of cardiovascular disease (CVD) markers [21][22][23][24][25]. A diet rich in animal protein, those with high fat content, such as processed and unprocessed meats [26], containing compounds such as choline, carnitine, but even betaine and lecithin in plants usually consumed [27], can produce molecules of TMA by microbiota interaction which can be eventually converted in TMAO [28]. Another molecule in meats, from liver and kidneys, mushrooms, and several type of beans, which is directly involved in TMA production, is ergothioneine, which is converted in TMA through ergothionase enzyme [10]. The TMA molecules are produced directly or indirectly mainly from food compounds containing L-carnitine and choline, but also from betaine and lecithin, ergothioneine and gamma-butyrobetaine. TMAO production is not a clear consequence but can potentially be influenced by dysbiosis and individual polymorphisms in the expression of FMO3 in the liver [14]; as well as the intake of fish and other seafood in the diet [26], and a more systemic disease [29][30]. As well as genetic factors and environmental factors, recent evidence suggests that even the metabolites deriving from the microbiota can play a protective role or promote the onset of tumors. These bacteria produce toxic metabolites, such as secondary bile salt from primary bile salt, hydrogen sulfide, trimethylamine-N-oxide (TMAO) from choline, indoxyl sulfate from amino acid tryptophan, and many more which are likely to promote inflammation, and prolonged inflammation can develop into cancer [5][7][31][32]. The potential role of several gut bacteria metabolites may cause localized inflammation in normal tissue of colon and promote the genotoxicity of intestinal epithelial cells, determining dysplasia and finally, colorectal cancer (CRC) [1][33].

2. TMAO in Physiological Conditions

TMAO molecules are produced in the liver, enter in the blood stream, from where most of them are excreted with urine within 24 h [7], and some can be reconverted in TMA by TMAO reductase [29]. In the intestine, the TMA conversion starts from choline and its isoform with a specific glycyl-radical-enzyme (GRE), GRE choline TMA-lyase (cutC) and its precursor GRE activase (cutD) [34] and from L-carnitine and gamma-butyrobetaine with Rieske-type oxygenase/reductase (cntA/B) [11]. The production of TMAO mainly depends on its pre-substrate TMA and the expression of liver enzymes FMO1 and mostly FMO3 [28]. TMAO levels also depend on genetic factors such as the presence of the E158K and E308G polymorphism on FMO3 [14]; another gene, founded in mice, called Slc30a7, associated with a zinc transporter (ZNT7) [35], seems to be correlated with plasma TMAO levels [36]. Although, as some studies report, the genetic factors in human associated with TMAO plasma levels are more complex [37] and correlated with pathological environment, such as CVD and comorbidities [38][39][40]. Some food sources have free TMA molecules that are absorbed by enterocytes by passive diffusion, oxidized, and expelled with a 3:95 TMA:TMAO ratio through urine (95%), feces (4%) and breathing (1%) [7]. The FMO3 enzyme is expressed in lungs, adrenals, and aorta. There is a gender difference in TMA activity, which is greater in female than male rats [41]. In males, the predominant steroid, testosterone, is responsible for a lower expression of FMO3 in the liver, whereas high levels of estrogen seem to elevate it [41], but a cohort study underlines that males have more TMAO levels than females [42]. However, some studies do not confirm a hormonal influence nor detect any significant sex differences in the levels of circulating TMAO [43][44][45]. Many other factors potentially mediate the production of TMAO from TMA, e.g., age, pathological status (CVD, low-grade inflammation, diabetes, tumors, genetic polymorphisms, protein-specific transport; sedentary life and inadequate nutritional status that may lead to gut dysbiosis. The production of TMA in physiological condition occurs through the interactions of compounds and some strains of gut microbiota. An in vitro study observed that the production was mainly by Firmicutes and Proteobacteria phyla, but not Bacteroidetes [46]. Yet other studies reported that all the production of TMA from choline and L-carnitine resulted from Firmicutes, Proteobacteria phyla, and none from Bacteroidetes [34][47]. One week with an antibiotic treatment to suppress some strains in the human gut leads to a reduction of TMAO levels, even with an L-carnitine [30] and choline supplementation [48]. From these studies, one could conclude that the amount of TMAO circulating is not only modulated by the ingestion of choline and L-carnitine food sources, but also by gut strains, in particular “high TMAO producers” with a high Firmicutes/Bacteroidetes ratio (roughly 2:1) [49]. In some studies, it was reported that a healthy gut microbiota in the adult is the one that has a high Firmicutes percentage on Bacteroidetes or roughly equal relative percentage [50][51]. Sometimes in other studies, it was found that obese people have a higher level of Bacteroidetes than thin individuals and a decrease of this strain has been seen after a period of caloric restriction [52]. Other gut microbes who involved in the TMA production are Deferribacteraceae, Anaeroplasmataceae, Prevotellaceae (Bacteroidetes phylum abundant in subjects consuming mostly starchy carbohydrates and fiber [53]), and Enterobacteraceae [34][54]. Furthermore, an increased TMAO has been found in those who consume a large amount of seafood per day. Similar levels were also found after the consumption of fish rich in omega-3 polyunsaturated fatty acids (e.g., salmon) [55], which may be due to their high content of free TMAO molecules [56]. Half of free TMAO intake from food is absorbed and eliminated in urine, the other half may be converted in TMA by the enzyme TMAO reductase [10]. Whether TMAO levels are correlated with milk and dairy products remains controversial [44]. Another interesting point is the link between the athlete/sportive amateur’s gut microbiota and TMAO production. It is well known that physical activity influences the population of gut microorganisms in a positive way, but it is quite evident that TMAO levels are also increased after exercise and sports [57]. Maybe this is a consequence of an omnivorous diet or one medium/high in animal protein that most athletes and sportive amateurs follow. The use of some ergogenic types of supplements such as L-carnitine [18] or choline isoforms (e.g., choline bitartrate) [58] may also contribute to high TMAO levels. However, TMAOs levels are not high in individuals on lacto-ovo vegetarian diet (egg, yolks and whole dairy products included) or vegan diet [30]. Nowadays, many professional and amateur athletes are switching to vegetarian and vegan diets, which could be a reason why TMAO plasma levels are low in these subjects [59]. Most of the caloric intake of athletes comes from carbohydrates, mainly starchy foods, such as whole grain cereal and tubers, but even fruits rich in fibers and oligosaccharides [60] that influence the gut microbiota by increasing Bacteroidetes phylum such as Prevotellaceae family and Actinobacteria. These families are partially responsible of TMA derived compounds conversion, but more importantly, they produce short-chain fatty acids (SCFA) [47]. In some studies, Firmicutes appears to be high in athletes and it is the main phylum implicated in TMA conversion [34][46][61]. Wolyniec et al. investigated TMAO levels before and after amateur runners’ 10 km or 100 Km ultramarathon there was no significant change in the levels, but only an acute increment. The only significant change was a 3.9-fold increase of TMAOs in the fastest runners of the 100 km race. The authors speculated that TMAOs level may impair the runner’s performance [62]. Certainly, high intensity physical activity requires a massive turnover of enzymes and substrates, leading to a huge production of metabolites, e.g., many L-carnitine isotypes that may influence TMA and TMAO conversion [63]. The athlete/sportive amateurs’ gut microbiota frequently shows some controversial aspects. Some studies report claims that physical activity reduces Firmicutes but increases Bacteroidetes, possibly due to the influence of the diet rich in whole grain carbohydrates, fruits, and vegetables [53][64]; others report the opposite effects because Firmicutes seems to be high in high caloric intake as most of athletes do [65]. It is undeniably controversial that some extreme physical activity shows an increment of TMAO levels, which already know that are strongly associated with CVD, obesity and type II diabetes, low-grade inflammation, gastrointestinal cancer [42], but at the same time reduce all these pathologies [66][67][68][69][70][71].

3. TMAO in Pathological Conditions

3.1. TMAO in Atherosclerosis and Cardiovascular Disease

Numerous studies indicate that gut microbiota is involved in the pathogenesis and progression of various cardiovascular diseases (CVD), such as heart failure (HF). HF causes changes in the composition of the intestinal microflora, which may affect the circulating levels of TMAO in human body. Researchers suggested intestinal strains, from Firmicutes and Proteobacteria phila, which can produce TMA, such as: Anaerococcus hydrogenalis, Clostridium asparagiforme, Clostridium hathewayi, Clostridium sporogenes, Escherichia fergusonii, Proteus penneri, Providencia rettgeri, and Edwardsiella tarda. The strains of these bacteria show an increased proportion in patients with HF. This indicates that changes in intestinal microbiota may affect TMAO levels by regulating TMA synthesis in the intestines [72]. Direct and indirect active roles of TMAOs in atherosclerosis and CVD are well established [5][30][39][42], also their associations in obesity and diabetes mellitus [19][29][39][73][74] as well as in low-grade inflammation [3][75] that are often are comorbidities. All of these may eventually also develop cancer [1][33][76][77][78]. The high levels of circulating TMAO in blood are strongly correlated to cardiovascular events such as stroke, myocardial infarctions, peripheral artery disease, acute coronary syndrome, and atherosclerosis [38][48][79][80]. Indeed, TMAO levels are correlated with the size of aortic atherosclerotic plaque, and they play a pivotal role as a pro-atherogenic factor. TMAO participates actively in the early stage of atherosclerotic process by promoting the macrophages migration, contributing to foam cell formation in the arterial intima [30][81]. Furthermore, high TMAO levels lead to an accumulation of ox-LDL particles within the macrophages by upregulating CD36 and scavenger receptor SR-A1 (Scavenger Receptor A1). These scavenger receptors are responsible for the transformation of macrophages into foam cells [82][83][84][85]. At the same time, the high levels of TMAO increase the expression of inflammatory cytokines, such as TNF-alpha and IL-6, that promote the migration of macrophages and their accumulation in arterial intima [82][86]. TMAO molecules seem to be involved even in endothelial dysfunction, a prelude to cardiovascular disease [7][36][81][87]. Vascular endothelial damage was observed in mice fed with a choline-rich diet [88], and high TMAO levels were associated with an increased systemic inflammation, oxidative stress, and fewer circulating endothelial progenitor cells (EPCs) [89][90][91][92][93] EPCs with a “spindle-shape” morphology, can take up acetylated LDL particles (acLDL) [94], whereas high TMAO levels reduce this. The formation of AcLDL in healthy subjects remains controversial, but it is often used as a model of oxidized or glycated LDL taken up by scavenger receptors. The potential accumulation of oxidized LDL particles is a precursor of inflammation and CVD [95]. TMAO can directly and indirectly< activate inflammatory signals such as NF-kB in aortic endothelial cells, and thereby contribute to atherosclerogenesis [96]. Furthermore, TMAOs reduce the expression of anti-inflammatory cytokines such as IL-10 that can protect the endothelial tissue from damage and inflammation, block the activity of NF-kB [97], and inhibit the adhesion of monocytes on the endothelial cells by downregulating the expression of CD18 and CD62-L on immune competent cells [98]. Finally, TMAOs can increase oxidative stress and reduce the endothelial nitric oxide synthases (eNOS) [99]. Oxidative stress is one of the most powerful promoters of atherosclerosis [87][96], whereas eNOS is a protective factor of endothelial health [100][101][102]. TMAO may have direct or indirect effects on (i) promotion macrophages migration and foam cells forming in the arterial intima; (ii) the accumulation of ox-LDL in situ within the macrophages; (iii) the reduction of EPCs, IL-10, eNOS, with the consequent rise of NF-kB, oxidative stress, and the LDL particles level in blood. All these effects lead to endothelial damages and cardiovascular dysfunction [103].

3.2. TMAO in Chronic Kidney Disease

High TMAO levels were found in patients with a chronic kidney disease (CKD). In patients with CKD treated with hemodialysis, pre-dialysis TMAO plasma levels were 77 ± 26 µM/dL, whereas in control group 2 ± 1 µM/dL, and other studies confirm that TMAO plasma levels of CKD patients are up to 40 times greater than normal [7][73][104]. CKD is also associated with CVD or a low-grade inflammation. All these show high levels of circulating TMAOs [20]. Missailidis et al. suggest that after a renal transplantation, the levels of TMAO were normalized [105], indicating not only a crucial role of the kidney in the excretion of these molecules, but also in their production. Indeed, some authors report a strong link between TMAO circulating levels, events of CVD and an effect on kidney health [105][106][107], with a direct connection with a low renal function [42], because there is a transporter called organic cation transporter 2 (OCT2) in basolateral kidney membrane that is responsible for the uptake of TMAOs [108]. A recent revision of Dongsheng et al. [109] confirms that the mechanism by which TMAO may enhance renal damage and aggravate nephropathy has not been well established. High TMAO plasma and urine levels may have a negative impact on CKD, due to the activation genes expression in the kidney tissue, but the mechanism is not known yet.

3.3. TMAO and Type II Diabetes

Several studies associate high plasma levels of TMAO and T2D or prediabetes [19][29][39][73][74][110][111][112][113], but none has proven a direct cause–effect. In mice feed with high fat diets plus 0.2% of TMAO molecules, it seems to impair glucose tolerance by affecting gene expression on insulin pathways in the liver and by increasing mRNA levels of pro-inflammatory cytokines in adipose tissue [114]. A bidirectional Mendelian assessment reports that high TMAO levels in T2D patients is more like a consequence [115]. For now, only the overall microbiota disproportion found in obese patients with pre-diabetes or T2D can lead to high plasma levels of TMAO is concluded [74][110][111][112][113].

3.4. TMAO, Microbiota Homeostasis and Cancer

The latest literature proved how the gut microbiota plays an important role in gastrointestinal cancer (GIC) [116]. Nowadays, the human microbiota is considered an organ that communicates in synergy with the other apparatus and with many physiological influences. Therefore, its homeostasis, called eubiosis, must be protected. When the microbiota is compromised, or when the proportions of microorganisms in it are modified, a syndrome called dysbiosis occurs [116]. Approximately 90% of these microorganisms constituting the microbiota belong to Firmicutes, Bacteroidetes, Proteobacteria and Actinobacteria. Viruses, eukarya, fungi, blastocystis, amoebozoa and archea [117][118][119][120][121] represent the other components. A good and healthy microbiota is considered necessary to regulate the immune function, intestinal mucosal protection, vitamins production, correct digestion, and nutrients absorption [122]. The most important phyla correlating with a healthy gut and, consequently, overall health, are the Firmicutes and Cytophaga-Flavobacterium-Bacteroides (CFB) that are indeed from Bacteroidetes phyla [118]. Dysbiosis is not only a difference of the microorganisms’ proportion, but it is correlated with various pathologies with different nature such as being overweight and severe obesity, which may carry over even to cardiovascular events, insulin resistance and type II diabetes [74]. A microbiota disproportion was founded and confirmed in some mental pathologies [123][124], such as depression, that it is important to include in the context of other pathologies which affect the digestive apparatus, such as inflammatory bowel disease (IBD) [125], liver disease [126], leaky gut and the intestinal mucosal function [127][128], and gastrointestinal cancer [116][129][130]. Gastrointestinal cancer is one of the most common neoplasia all over the world [131], and lot of data show how the microbiota interacts with this pathology [116][132][133][134][135][136]. Obesity and low-grade inflammation are two main factors which can lead to cancer development [75], and there is a strong correlation within obesity, low-grade inflammation, dysbiosis and colorectal cancer (CRC) [129][132][136][137].
In patients with gastrointestinal cancers, the most abundant family of microorganisms are the Enterobacteriaceae that are situated in the small intestine. A low presence of Lactobacillaceae and Acidoaminococcaceae is typical of colon cancers and Bifidobacteriaceae in those of the rectum [138]. At the species level, Bacteroides fragilis seem to account for many colorectal cancers and Helicobacter pylori for gastric cancers [116][129][139][140]. Even Escherichia coli [141] and Enterococcus faecalis [142] may be involved in cancer development by DNA mutation genes. Some studies have shown that chemotherapy in patients with GIC leads to a modification/restoration of microbiota dysbiosis [143], a much greater richness in Lactobacillaceae has been found after therapy, compared to untreated controls [138]. Another genus correlated with CLC, Fusobacterium, showed a significative reduction after debulking surgery, but not chemotherapy [144]. At last, in a very recent study that compared the microbiota composition in obese patients with CLC, non-obese patients with CLC and a healthy control, shows enormous differences in diversity and richness of the gut species [145]. A reduction in richness was found in both CLC groups in comparison with the healthy group, but much more incisive was the decrease in diversity. The major phyla detected in healthy group was Bacteroidetes, with more than 50%, whereas it was below 30% in the CLC groups. Firmicutes was 39–43% in CLC groups and about 21% in the healthy one, and Fusobacterium was 9.1% and 1.2%, respectively. In conclusion, there is strong evidence for a reduction of species which produce SCFA (Butyricimonas, Roseburia, Blautia, Faecalibacterium, and Ruminococcus), and an increase of pathogenic and induced-cancer ones (Fusobacterium, Clostridium, Prevotella, Desulfovibrio, and Enterococcus) [145]. In this context, the TMAOs molecules may be implicated too. An increased level in TMAO concentration may be caused by diet, changes in the composition of intestinal microflora, gut dysbiosis or impairment of the gut–blood barrier. Studies on mice have shown that intestinal bacteria are essential to convert dietary compounds to TMA [146]. The production of TMA and TMAO can be almost completely suppressed using broad spectrum antibiotics, and after one month of withdrawal of antibiotics, the TMAO concentration returns to normal [147]. Indeed, some studies have associated the high levels of TMAO with high TNF-alpha, IL-6, C-reactive protein [44], pro-inflammatory cytokines, IL-1beta [96] and even as a coadjuvant of Helicobacter pylori to promote infection in the gastric epithelial cells, increasing the activity of IL-6 and chemokine ligands. This suggests a potential link between TMAO and gastric cancer via the inflammatory process. Consistent with this, Yue et al. also demonstrated that TMAO can trigger the activation of the nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome [148][149], which was suggested to be implicated in the growth and/or metastasis of a variety of cancers including head and neck cancer, oral cancer, lung cancer, prostate cancer, and colorectal cancer [150][151]. Association studies provided further support for a link between TMAO and inflammation. The serum level of TMAO was shown to be positively correlated with the level of certain pro-inflammatory mediators including tumor necrosis factor-alpha (TNF-α) and IL-6. Moreover, studies have also displayed that an enhanced level of TMAO prompted the initiation of NF-Kappa-B route and improved the expression of pro-inflammatory genes involving chemokines, adhesion molecules and inflammatory cytokines [152]. Several data show, indeed, how the TMAO levels are quite high in patients with colorectal cancer and even higher in obese subjects with the same type of cancer [145][153][154]. Moreover, the presence of high quantity of Firmicutes phylum, Prevotellaceae, Enterobacteraceae and Desulfovibrio that can increase the conversion of choline to TMA by the expression of the cutC gene [34], and low levels of Bacteroidetes phylum [145] in relation to healthy controls with neither cancer nor obesity, are very illuminating and confirm the role of TMAO. This would indicate reducing all foods containing choline, betaine, lecithin, gamma-butyrobetaine, ergothioneine and L-carnitine, to avoid a massive conversion to TMAO [1]. Conversion of these compounds in TMA not only depends on the food sources [4][24][29], but also on microbiota composition [42] which is compromised by pathological status [129][137][155]. It is now clear that many gastro-intestinal pathologies are related to bacteria families of Firmicutes phylum [49], which even includes healthy strains of gut microbiota [50][51]. TMAO is frequently used as a risk marker of diseases, but its usefulness as a standalone marker is limited due to the high intra individual variability that may reflect dietary changes from day to day, in particular the intake of meat or fish [156]. Oxidative stress might also be one of the factors linking TMAO and cancer. Recent studies showed that TMAO could be implicated in oxidative stress and increased circulating TMAO was shown to induce superoxide production, a reactive oxygen species (ROS) linked to oxidative stress. In an in vitro assay, TMAO was also shown to stimulate the production of ROS in cells [157][158][159].

References

  1. 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.
  2. Tang, W.H.W.; Hazen, S.L. Microbiome, trimethylamine N-oxide, and cardiometabolic disease. Transl. Res. 2017, 179, 108–115.
  3. Janeiro, M.H.; Ramírez, M.J.; Milagro, F.I.; Martínez, J.A.; Solas, M. Implication of trimethylamine n-oxide (TMAO) in disease: Potential biomarker or new therapeutic target. Nutrients 2018, 10, 1398.
  4. Bain, M.; Fornasini, G.; Evans, A. Trimethylamine: Metabolic, Pharmacokinetic and Safety Aspects. Curr. Drug Metab. 2005, 6, 227–240.
  5. Ahmadmehrabi, S.; Tang, W.H.W. Gut microbiome and its role in cardiovascular diseases. Curr. Opin. Cardiol. 2017, 32, 761–766.
  6. Jie, Z.; Xia, H.; Zhong, S.L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845.
  7. Zeisel, S.H.; Warrier, M. Trimethylamine N-Oxide, the Microbiome, and Heart and Kidney Disease. Annu. Rev. Nutr. 2017, 37, 157–181.
  8. Wiedeman, A.M.; Barr, S.I.; Green, T.J.; Xu, Z.; Innis, S.M.; Kitts, D.D. Dietary choline intake: Current state of knowledge across the life cycle. Nutrients 2018, 10, 1513.
  9. Zeisel, S.H.; Da Costa, K.A. Choline: An essential nutrient for public health. Nutr. Rev. 2009, 67, 615–623.
  10. 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.
  11. Zhu, Y.; Jameson, E.; Crosatti, M.; Schäfer, H.; Rajakumar, K.; Bugg, T.D.H.; Chen, Y. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc. Natl. Acad. Sci. USA 2014, 111, 4268–4273.
  12. Koeth, R.A.; Levison, B.S.; Culley, M.K.; Buffa, J.A.; Wang, Z.; Gregory, J.C.; Org, E.; Wu, Y.; Li, L.; Smith, J.D.; et al. γ-butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab. 2014, 20, 799–812.
  13. Akerman, B.R.; Lemass, H.; Chow, L.M.L.; Lambert, D.M.; Greenberg, C.; Bibeau, C.; Mamer, O.A.; Treacy, E.P. Trimethylaminuria is caused by mutations of the FMO3 gene in a North American cohort. Mol. Genet. Metab. 1999, 68, 24–31.
  14. Treacy, E.P.; Akerman, B.R.; Chow, L.M.L.; 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.
  15. Bennett, B.J.; Vallim, T.Q.D.A.; 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.
  16. Busby, M.G.; Fischer, L.; Da Costa, K.A.; Thompson, D.; Mar, M.H.; Zeisel, S.H. Choline- and betaine-defined diets for use in clinical research and for the management of trimethylaminuria. J. Am. Diet. Assoc. 2004, 104, 1836–1845.
  17. Brugère, J.F.; Borrel, G.; Gaci, N.; Tottey, W.; O’Toole, P.W.; Malpuech-Brugère, C. Archaebiotics: Proposed therapeutic use of archaea to prevent trimethylaminuria and cardiovascular disease. Gut Microbes 2013, 5, 5–10.
  18. Sawicka, A.K.; Renzi, G.; Olek, R.A. The bright and the dark sides of L-carnitine supplementation: A systematic review. J. Int. Soc. Sports Nutr. 2020, 17, 1–10.
  19. Heianza, Y.; Ma, W.; Manson, J.A.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, 4947.
  20. Schiattarella, G.G.; Sannino, A.; Toscano, E.; Giugliano, G.; Gargiulo, G.; Franzone, A.; Trimarco, B.; Esposito, G.; Perrino, C. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: A systematic review and dose-response meta-analysis. Eur. Heart J. 2017, 38, 2948–2956.
  21. 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.
  22. Olek, R.A.; Samulak, J.J.; Sawicka, A.K.; Hartmane, D.; Grinberga, S.; Pugovics, O.; Lysiak-Szydlowska, W. Increased Trimethylamine N-Oxide Is Not Associated with Oxidative Stress Markers in Healthy Aged Women. Oxid. Med. Cell. Longev. 2019, 2019, 6247169.
  23. Bordoni, L.; Sawicka, A.K.; Szarmach, A.; Winklewski, P.J.; Olek, R.A.; Gabbianelli, R. A pilot study on the effects of L-carnitine and trimethylamine-N-oxide on platelet mitochondrial dna methylation and CVD biomarkers in aged women. Int. J. Mol. Sci. 2020, 21, 1047.
  24. Fukami, K.; Yamagishi, S.I.; 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.
  25. Vallance, H.D.; Koochin, A.; Branov, J.; Rosen-Heath, A.; Bosdet, T.; Wang, Z.; Hazen, S.L.; Horvath, G. Marked elevation in plasma trimethylamine-N-oxide (TMAO) in patients with mitochondrial disorders treated with oral L-carnitine. Mol. Genet. Metab. Rep. 2018, 15, 130–133.
  26. Wallace, T.C.; Blusztajn, J.K.; Caudill, M.A.; Klatt, K.C.; Natker, E.; Zeisel, S.H.; Zelman, K.M. The underconsumed and underappreciated essential nutrient. Nutr. Today 2018, 53, 240–253.
  27. 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.
  28. Schugar, R.C.; Brown, J.M. Emerging roles of flavin monooxygenase 3 in cholesterol metabolism and atherosclerosis. Curr. Opin. Lipidol. 2015, 26, 426–431.
  29. Chhibber-Goel, J.; Gaur, A.; Singhal, V.; Parakh, N.; Bhargava, B.; Sharma, A. The complex metabolism of trimethylamine in humans: Endogenous and exogenous sources. Expert Rev. Mol. Med. 2016, 18, e8.
  30. 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.
  31. Tremaroli, V.; Bäckhed, F. Functional interactions between the gut microbiota and host metabolism. Nature 2012, 489, 242–249.
  32. Kamada, N.; Seo, S.U.; Chen, G.Y.; Núñez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 2013, 13, 321–335.
  33. Yang, S.; Dai, H.; Lu, Y.; Li, R.; Gao, C.; Pan, S. Trimethylamine N-Oxide Promotes Cell Proliferation and Angiogenesis in Colorectal Cancer. J. Immunol. Res. 2022, 2022, 7043856.
  34. Craciun, S.; Balskus, E.P. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc. Natl. Acad. Sci. USA 2012, 109, 21307–21312.
  35. Hartiala, J.; Bennett, B.J.; Tang, W.H.W.; Wang, Z.; Stewart, A.F.R.; Roberts, R.; McPherson, R.; Lusis, A.J.; Hazen, S.L.; Allayee, H. Comparative genome-wide association studies in mice and humans for trimethylamine N-Oxide, a proatherogenic metabolite of choline and L-carnitine. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1307–1313.
  36. 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.
  37. Aslibekyan, S.; Irvin, M.R.; Hidalgo, B.A.; Perry, R.T.; Jeyarajah, E.J.; Garcia, E.; Shalaurova, I.; Hopkins, P.N.; Province, M.A.; Tiwari, H.K.; et al. Genome- and CD4+ T-cell methylome-wide association study of circulating trimethylamine-N-oxide in the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN). J. Nutr. Intermed. Metab. 2017, 8, 1–7.
  38. Senthong, V.; Wang, Z.; Fan, Y.; Wu, Y.; Hazen, S.L.; Tang, W.H.W. Trimethylamine N-oxide and mortality risk in patients with peripheral artery disease. J. Am. Heart Assoc. 2016, 5, e4237.
  39. Randrianarisoa, E.; Lehn-Stefan, A.; Wang, X.; Hoene, M.; Peter, A.; Heinzmann, S.S.; Zhao, X.; Königsrainer, I.; Königsrainer, A.; Balletshofer, B.; et al. Relationship of serum trimethylamine N-oxide (TMAO) levels with early atherosclerosis in humans. Sci. Rep. 2016, 6, 26745.
  40. 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.
  41. Esposito, T.; Varriale, B.; D’Angelo, R.; Amato, A.; Sidoti, A. Regulation of flavin-containing mono-oxygenase (Fmo3) gene expression by steroids in mice and humans. Horm. Mol. Biol. Clin. Investig. 2014, 20, 99–109.
  42. Manor, O.; Zubair, N.; Conomos, M.P.; Xu, X.; Rohwer, J.E.; Krafft, C.E.; Lovejoy, J.C.; Magis, A.T. A Multi-omic Association Study of Trimethylamine N-Oxide. Cell Rep. 2018, 24, 935–946.
  43. Kühn, T.; Rohrmann, S.; Sookthai, D.; Johnson, T.; Katzke, V.; Kaaks, R.; Von Eckardstein, A.; Müller, D. Intra-individual variation of plasma trimethylamine-N-oxide (TMAO), betaine and choline over 1 year. Clin. Chem. Lab. Med. 2017, 55, 261–268.
  44. Rohrmann, S.; Linseisen, J.; Allenspach, M.; Von Eckardstein, A.; Müller, 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.
  45. Krüger, R.; Merz, B.; Rist, M.J.; Ferrario, P.G.; Bub, A.; Kulling, S.E.; Watzl, B. Associations of current diet with plasma and urine TMAO in the KarMeN study: Direct and indirect contributions. Mol. Nutr. Food Res. 2017, 61, 1700363.
  46. 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, e02481-14.
  47. Falony, G.; Vieira-Silva, S.; Raes, J. Microbiology Meets Big Data: The Case of Gut Microbiota-Derived Trimethylamine. Annu. Rev. Microbiol. 2015, 69, 305–321.
  48. Tang, W.H.W.; 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.
  49. 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, 324.
  50. Biagi, E.; Franceschi, C.; Rampelli, S.; Severgnini, M.; Ostan, R.; Turroni, S.; Consolandi, C.; Quercia, S.; Scurti, M.; Monti, D.; et al. Gut Microbiota and Extreme Longevity. Curr. Biol. 2016, 26, 1480–1485.
  51. Santoro, A.; Ostan, R.; Candela, M.; Biagi, E.; Brigidi, P.; Capri, M.; Franceschi, C. Gut microbiota changes in the extreme decades of human life: A focus on centenarians. Cell. Mol. Life Sci. 2018, 75, 129–148.
  52. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023.
  53. Ley, R.E. Gut microbiota in 2015: Prevotella in the gut: Choose carefully. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 69–70.
  54. Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-oxide: The good, the bad and the unknown. Toxins 2016, 8, 326.
  55. Landfald, B.; Valeur, J.; Berstad, A.; Raa, J. Microbial trimethylamine- N -oxide as a disease marker: Something fishy? Microb. Ecol. Health Dis. 2017, 28, 1327309.
  56. Cheung, W.; Keski-Rahkonen, P.; Assi, N.; Ferrari, P.; Freisling, H.; Rinaldi, S.; Slimani, N.; Zamora-Ros, R.; Rundle, M.; Frost, G.; et al. A metabolomic study of biomarkers of meat and fish intake. Am. J. Clin. Nutr. 2017, 105, 600–608.
  57. Barton, W.; Penney, N.C.; Cronin, O.; Garcia-Perez, I.; Molloy, M.G.; Holmes, E.; Shanahan, F.; Cotter, P.D.; O’Sullivan, O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 2018, 67, 625–633.
  58. Penry, J.T.; Manore, M.M. Choline: An important micronutrient for maximal endurance-exercise performance? Int. J. Sport Nutr. Exerc. Metab. 2008, 18, 191–203.
  59. Rogerson, D. Vegan diets: Practical advice for athletes and exercisers. J. Int. Soc. Sports Nutr. 2017, 14, 1–15.
  60. Hamaker, B.R.; Tuncil, Y.E. A perspective on the complexity of dietary fiber structures and their potential effect on the gut microbiota. J. Mol. Biol. 2014, 426, 3838–3850.
  61. Sharma, P.; Bhandari, C.; Kumar, S.; Sharma, B.; Bhadwal, P.; Agnihotri, N. Dietary fibers: A way to a healthy microbiome. In Diet, Microbiome and Health; Elsever: Amsterdam, The Netherlands, 2018; ISBN 9780128114407.
  62. Wołyniec, W.; Kasprowicz, K.; Giebułtowicz, J.; Korytowska, N.; Zorena, K.; Bartoszewicz, M.; Rita-Tkachenko, P.; Renke, M.; Ratkowski, W. Changes in water soluble uremic toxins and urinary acute kidney injury biomarkers after 10-and 100-km runs. Int. J. Environ. Res. Public Health 2019, 16, 4153.
  63. Schranner, D.; Kastenmüller, G.; Schönfelder, M.; Römisch-Margl, W.; Wackerhage, H. Metabolite Concentration Changes in Humans After a Bout of Exercise: A Systematic Review of Exercise Metabolomics Studies. Sport. Med.-Open 2020, 6, 1–17.
  64. Kerksick, C.M.; Wilborn, C.D.; Roberts, M.D.; Smith-Ryan, A.; Kleiner, S.M.; Jäger, R.; Collins, R.; Cooke, M.; Davis, J.N.; Galvan, E.; et al. ISSN exercise & sports nutrition review update: Research & recommendations. J. Int. Soc. Sports Nutr. 2018, 5, 38.
  65. Hughes, R.L. A Review of the Role of the Gut Microbiome in Personalized Sports Nutrition. Front. Nutr. 2020, 6, 191.
  66. Ekelund, U.; Ward, H.A.; Norat, T.; Luan, J.; May, A.M.; Weiderpass, E.; Sharp, S.J.; Overvad, K.; Østergaard, J.N.; Tjønneland, A.; et al. Physical activity and all-cause mortality across levels of overall and abdominal adiposity in European men and women: The European prospective investigation into cancer and nutrition study (EPIC). Am. J. Clin. Nutr. 2015, 101, 613–621.
  67. Kyu, H.H.; Bachman, V.F.; Alexander, L.T.; Mumford, J.E.; Afshin, A.; Estep, K.; Veerman, J.L.; Delwiche, K.; Iannarone, M.L.; Moyer, M.L.; et al. Physical activity and risk of breast cancer, colon cancer, diabetes, ischemic heart disease, and ischemic stroke events: Systematic review and dose-response meta-analysis for the Global Burden of Disease Study 2013. BMJ 2016, 354, i3857.
  68. Wolin, K.Y.; Yan, Y.; Colditz, G.A.; Lee, I.M. Physical activity and colon cancer prevention: A meta-analysis. Br. J. Cancer 2009, 100, 611–616.
  69. World Cancer Research Fund/American Institute for Cancer Research. Diet, Nutrition, Physical Activity and Cancer: A Global Perspective; World Cancer Research Fund/American Institute for Cancer Research: Washington, DC, USA, 2018; ISBN 9781912259465.
  70. Fuchs, R. Physical Activity and Health. In International Encyclopedia of the Social & Behavioral Sciences, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2015; ISBN 9780080970875.
  71. Saint-Maurice, P.F.; Coughlan, D.; Kelly, S.P.; Keadle, S.K.; Cook, M.B.; Carlson, S.A.; Fulton, J.E.; Matthews, C.E. Association of Leisure-Time Physical Activity Across the Adult Life Course with All-Cause and Cause-Specific Mortality. JAMA Netw. Open 2019, 2, e190355.
  72. Zhang, Y.; Wang, Y.; Ke, B.; Du, J. TMAO: How gut microbiota contributes to heart failure. Transl. Res. 2021, 228, 109–125.
  73. Mueller, D.M.; Allenspach, M.; Othman, A.; Saely, C.H.; Muendlein, A.; Vonbank, A.; Drexel, H.; von Eckardstein, A. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis 2015, 243, 638–644.
  74. Hansen, T.H.; Gøbel, R.J.; Hansen, T.; Pedersen, O. The gut microbiome in cardio-metabolic health. Genome Med. 2015, 7, 33.
  75. Wu, S.; Rhee, K.J.; Albesiano, E.; Rabizadeh, S.; Wu, X.; Yen, H.R.; Huso, D.L.; Brancati, F.L.; Wick, E.; McAllister, F.; et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat. Med. 2009, 15, 1016–1022.
  76. 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.D.; et al. Plasma choline metabolites and colorectal cancer risk in the women’s health initiative observational study. Cancer Res. 2014, 74, 7442–7452.
  77. Xu, R.; Wang, Q.Q.; Li, L. A genome-wide systems analysis reveals strong link between colorectal cancer and trimethylamine N-oxide (TMAO), a gut microbial metabolite of dietary meat and fat. BMC Genom. 2015, 16, S4.
  78. Chan, C.W.H.; Law, B.M.H.; Waye, M.M.Y.; Chan, J.Y.W.; Wei So, W.K.; Chow, K.M. Trimethylamine-N-oxide as one hypothetical link for the relationship between intestinal microbiota and cancer-Where we are and where shall we go? J. Cancer 2019, 10, 5874.
  79. Li, X.S.; Obeid, S.; Klingenberg, R.; Gencer, B.; Mach, F.; Räber, L.; Windecker, S.; Rodondi, N.; Nanchen, D.; Muller, O.; et al. Gutmicrobiota-dependent trimethylamine N-oxide in acute coronary syndromes: A prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur. Heart J. 2017, 38, 814–824.
  80. Senthong, V.; Wang, Z.; Li, X.S.; Fan, Y.; Wu, Y.; Tang, W.H.W.; Hazen, S.L. Intestinal microbiota-generated metabolite Trimethylamine-N-oxide and 5-year mortality risk in stable coronary artery disease: The contributory role of intestinal microbiota in a COURAGE-like patient cohort. J. Am. Heart Assoc. 2016, 5, e002816.
  81. 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.
  82. Chistiakov, D.A.; Melnichenko, A.A.; Myasoedova, V.A.; Grechko, A.V.; Orekhov, A.N. Mechanisms of foam cell formation in atherosclerosis. J. Mol. Med. 2017, 95, 1153–1165.
  83. Thon, M.P.; Hemmler, A.; Glinzer, A.; Mayr, M.; Wildgruber, M.; Zernecke-Madsen, A.; Gee, M.W. A multiphysics approach for modeling early atherosclerosis. Biomech. Model. Mechanobiol. 2018, 17, 617–644.
  84. Geng, J.; Yang, C.; Wang, B.; Zhang, X.; Hu, T.; Gu, Y.; Li, J. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomed. Pharmacother. 2018, 97, 941–947.
  85. Mohammadi, A.; Najar, A.G.; Yaghoobi, M.M.; Jahani, Y.; Vahabzadeh, Z. Trimethylamine-N-Oxide Treatment Induces Changes in the ATP-Binding Cassette Transporter A1 and Scavenger Receptor A1 in Murine Macrophage J774A.1 cells. Inflammation 2016, 39, 393–404.
  86. Zhu, Y.; Li, Q.; Jiang, H. Gut microbiota in atherosclerosis: Focus on trimethylamine N-oxide. Apmis 2020, 128, 353–366.
  87. Sun, X.; Jiao, X.; Ma, Y.; Liu, Y.; Zhang, L.; He, Y.; Chen, Y. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem. Biophys. Res. Commun. 2016, 481, 63–70.
  88. Ren, D.; Liu, Y.; Zhao, Y.; Yang, X. Hepatotoxicity and endothelial dysfunction induced by high choline diet and the protective effects of phloretin in mice. Food Chem. Toxicol. 2016, 94, 203–212.
  89. 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.
  90. Ingram, D.A.; Mead, L.E.; Tanaka, H.; Meade, V.; Fenoglio, A.; Mortell, K.; Pollok, K.; Ferkowicz, M.J.; Gilley, D.; Yoder, M.C. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood 2004, 104, 2752–2760.
  91. Basile, D.P.; Yoder, M.C. Circulating and Tissue Resident Endothelial Progenitor Cells. J. Cell. Physiol. 2014, 229, 10–16.
  92. Chopra, H.; Hung, M.K.; Kwong, D.L.; Zhang, C.F.; Pow, E.H.N. Insights into endothelial progenitor cells: Origin, classification, potentials, and prospects. Stem Cells Int. 2018, 2018, 9847015.
  93. Prater, D.N.; Case, J.; Ingram, D.A.; Yoder, M.C. Working hypothesis to redefine endothelial progenitor cells. Leukemia 2007, 21, 1141–1149.
  94. Hur, J.; Yoon, C.H.; Kim, H.S.; Choi, J.H.; Kang, H.J.; Hwang, K.K.; Oh, B.H.; Lee, M.M.; Park, Y.B. Characterization of Two Types of Endothelial Progenitor Cells and Their Different Contributions to Neovasculogenesis. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 288–293.
  95. Davignon, J.; Ganz, P. Role of endothelial dysfunction in atherosclerosis. Circulation 2004, 109, III27–III32.
  96. Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.F.; 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-κb. J. Am. Heart Assoc. 2016, 5, e002767.
  97. Chen, H.; Li, J.; Li, N.; Liu, H.; Tang, J. Increased circulating trimethylamine N-oxide plays a contributory role in the development of endothelial dysfunction and hypertension in the RUPP rat model of preeclampsia. Hypertens. Pregnancy 2019, 38, 96–104.
  98. Mostafa Mtairag, E.; Chollet-Martin, S.; Oudghiri, M.; Laquay, N.; Jacob, M.P.; Michel, J.B.; Feldman, L.J. Effects of interleukin-10 on monocyte/endothelial cell adhesion and MMP-9/TIMP-1 secretion. Cardiovasc. Res. 2001, 49, 882–890.
  99. 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.
  100. Herrera, M.D.; Mingorance, C.; Rodríguez-Rodríguez, R.; Alvarez de Sotomayor, M. Endothelial dysfunction and aging: An update. Ageing Res. Rev. 2010, 9, 142–152.
  101. Donato, A.J.; Morgan, R.G.; Walker, A.E.; Lesniewski, L.A. Cellular and molecular biology of aging endothelial cells. J. Mol. Cell. Cardiol. 2015, 89, 122–135.
  102. Seals, D.R.; Jablonski, K.L.; Donato, A.J. Aging and vascular endothelial function in humans. Clin. Sci. 2011, 120, 357–375.
  103. Ma, G.H.; Pan, B.; Chen, Y.; Guo, C.X.; Zhao, M.M.; Zheng, L.M.; Chen, B.X. Trimethylamine N-oxide in atherogenesis: Impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci. Rep. 2017, 37, BSR20160244.
  104. 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.
  105. Missailidis, C.; Hällqvist, J.; Qureshi, A.R.; Barany, P.; Heimbürger, O.; Lindholm, B.; Stenvinkel, P.; Bergman, P. Serum trimethylamine-N-Oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS ONE 2016, 11, e0141738.
  106. Kim, R.B.; Morse, B.L.; Djurdjev, O.; Tang, M.; Muirhead, N.; Barrett, B.; Holmes, D.T.; Madore, F.; Clase, C.M.; Rigatto, C.; et al. Advanced chronic kidney disease populations have elevated trimethylamine N-oxide levels associated with increased cardiovascular events. Kidney Int. 2016, 89, 1144–1152.
  107. Robbiano, L.; Mereto, E.; Corbu, C.; Brambilla, G. DNA damage induced by seven N-nitroso compounds in primary cultures of human and rat kidney cells. Mutat. Res. 1996, 368, 41–47.
  108. Teft, W.A.; Morse, B.L.; Leake, B.F.; Wilson, A.; Mansell, S.E.; Hegele, R.A.; Ho, R.H.; Kim, R.B. Identification and Characterization of Trimethylamine-N-oxide Uptake and Efflux Transporters. Mol. Pharm. 2017, 14, 310–318.
  109. Zixin, Y.; Lulu, C.; Xiangchang, Z.; Qing, F.; Binjie, Z.; Chunyang, L.; Tai, R.; Dongsheng, O. TMAO as a potential biomarker and therapeutic target for chronic kidney disease: A review. Front. Pharmacol. 2022, 12, 929262.
  110. Kalagi, N.A.; Thota, R.N.; Stojanovski, E.; Alburikan, K.A.; Garg, M.L. Association between Plasma Trimethylamine N-Oxide Levels and Type 2 Diabetes: A Case Control Study. Nutrients 2022, 14, 2093.
  111. 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.
  112. Tang, W.H.W.; 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.
  113. Dambrova, M.; Latkovskis, G.; Kuka, J.; Strele, I.; Konrade, I.; Grinberga, S.; Hartmane, D.; Pugovics, O.; Erglis, A.; Liepinsh, E. Diabetes is Associated with Higher Trimethylamine N-oxide Plasma Levels. Exp. Clin. Endocrinol. Diabetes 2016, 124, 251–256.
  114. 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.
  115. 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: A Bidirectional Mendelian Randomization Analysis. Diabetes 2019, 68, 1747–1755.
  116. Rea, D.; Coppola, G.; Palma, G.; Barbieri, A.; Luciano, A.; Del Prete, P.; Rossetti, S.; Berretta, M.; Facchini, G.; Perdonà, S.; et al. Microbiota effects on cancer: From risks to therapies. Oncotarget 2018, 9, 17915–17927.
  117. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533.
  118. Cani, P.D. Metabolism in 2013: The gut microbiota manages host metabolism. Nat. Rev. Endocrinol. 2014, 10, 74–76.
  119. Ianiro, G.; Bruno, G.; Lopetuso, L.; Beghella, F.; Laterza, L.; D’Aversa, F.; Gigante, G.; Cammarota, G.; Gasbarrini, A. Role of Yeasts in Healthy and Impaired Gut Microbiota: The Gut Mycome. Curr. Pharm. Des. 2014, 20, 4565–4569.
  120. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230.
  121. Columpsi, P.; Sacchi, P.; Zuccaro, V.; Cima, S.; Sarda, C.; Mariani, M.; Gori, A.; Bruno, R. Beyond the gut bacterial microbiota: The gut virome. J. Med. Virol. 2016, 88, 1467–1472.
  122. Mills, S.; Stanton, C.; Lane, J.A.; Smith, G.J.; Ross, R.P. Precision nutrition and the microbiome, part I: Current state of the science. Nutrients 2019, 11, 923.
  123. Cenit, M.C.; Sanz, Y.; Codoñer-Franch, P. Influence of gut microbiota on neuropsychiatric disorders. World J. Gastroenterol. 2017, 23, 5486–5498.
  124. Slyepchenko, A.; Maes, M.; Jacka, F.N.; Köhler, C.A.; Barichello, T.; McIntyre, R.S.; Berk, M.; Grande, I.; Foster, J.A.; Vieta, E.; et al. Gut Microbiota, Bacterial Translocation, and Interactions with Diet: Pathophysiological Links between Major Depressive Disorder and Non-Communicable Medical Comorbidities. Psychother. Psychosom. 2016, 86, 31–46.
  125. Hold, G.L.; Smith, M.; Grange, C.; Watt, E.R.; El-Omar, E.M.; Mukhopadhya, I. Role of the gut microbiota in inflammatory bowel disease pathogenesis: What have we learnt in the past 10 years? World J. Gastroenterol. 2014, 20, 1192–1210.
  126. Llorente, C.; Schnabl, B. The Gut Microbiota and Liver Disease. Cell. Mol. Gastroenterol. Hepatol. 2 2015, 1, 275–284.
  127. Vindigni, S.M.; Zisman, T.L.; Suskind, D.L.; Damman, C.J. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: A tripartite pathophysiological circuit with implications for new therapeutic directions. Therap. Adv. Gastroenterol. 2016, 9, 606–625.
  128. Fukui, H. Increased Intestinal Permeability and Decreased Barrier Function: Does It Really Influence the Risk of Inflammation? Inflamm. Intest. Dis. 2016, 1, 135–145.
  129. Meng, C.; Bai, C.; Brown, T.D.; Hood, L.E.; Tian, Q. Human Gut Microbiota and Gastrointestinal Cancer. Genom. Proteom. Bioinform. 2018, 16, 33–49.
  130. Mima, K.; Ogino, S.; Nakagawa, S.; Sawayama, H.; Kinoshita, K.; Krashima, R.; Ishimoto, T.; Imai, K.; Iwatsuki, M.; Hashimoto, D.; et al. The role of intestinal bacteria in the development and progression of gastrointestinal tract neoplasms. Surg. Oncol. 2017, 26, 368–376.
  131. Bhandari, A.; Woodhouse, M.; Gupta, S. Colorectal cancer is a leading cause of cancer incidence and mortality among adults younger than 50 years in the USA: A SEER-based analysis with comparison to other young-onset cancers. J. Investig. Med. 2017, 65, 311–315.
  132. Zackular, J.P.; Baxter, N.T.; Iverson, K.D.; Sadler, W.D.; Petrosino, J.F.; Chen, G.Y.; Schloss, P.D. The gut microbiome modulates colon tumorigenesis. MBio 2013, 4, e00692-13.
  133. Zackular, J.P.; Baxter, N.T.; Chen, G.Y.; Schloss, P.D. Manipulation of the Gut Microbiota Reveals Role in Colon Tumorigenesis. mSphere 2016, 1, e00001-15.
  134. Bullman, S.; Pedamallu, C.S.; Sicinska, E.; Clancy, T.E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T.; et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 1443–1448.
  135. Drewes, J.L.; Housseau, F.; Sears, C.L. Sporadic colorectal cancer: Microbial contributors to disease prevention, development and therapy. Br. J. Cancer 2016, 115, 273–280.
  136. Gagnière, J.; Raisch, J.; Veziant, J.; Barnich, N.; Bonnet, R.; Buc, E.; Bringer, M.A.; Pezet, D.; Bonnet, M. Gut microbiota imbalance and colorectal cancer. World J. Gastroenterol. 2016, 22, 501–518.
  137. Sun, J.; Kato, I. Gut microbiota, inflammation and colorectal cancer. Genes Dis. 2016, 3, 130–143.
  138. Youssef, O.; Lahti, L.; Kokkola, A.; Karla, T.; Tikkanen, M.; Ehsan, H.; Carpelan-Holmström, M.; Koskensalo, S.; Böhling, T.; Rautelin, H.; et al. Stool Microbiota Composition Differs in Patients with Stomach, Colon, and Rectal Neoplasms. Dig. Dis. Sci. 2018, 63, 2950–2958.
  139. Sears, C.L.; Garrett, W.S. Microbes, microbiota, and colon cancer. Cell Host Microbe 2014, 15, 317–328.
  140. Chen, J.; Domingue, J.C.; Sears, C.L. Microbiota dysbiosis in select human cancers: Evidence of association and causality. Semin. Immunol. 2017, 32, 25–34.
  141. Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123.
  142. Huycke, M.M.; Abrams, V.; Moore, D.R. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis 2002, 23, 529–536.
  143. Sze, M.A.; Baxter, N.T.; Ruffin, M.T.; Rogers, M.A.M.; Schloss, P.D. Normalization of the microbiota in patients after treatment for colonic lesions. Microbiome 2017, 5, 150.
  144. Deng, X.; Li, Z.; Li, G.; Li, B.; Jin, X.; Lyu, G. Comparison of microbiota in patients treated by surgery or chemotherapy by 16S rRNA sequencing reveals potential biomarkers for colorectal cancer therapy. Front. Microbiol. 2018, 9, 1607.
  145. Sánchez-Alcoholado, L.; Ordóñez, R.; Otero, A.; Plaza-Andrade, I.; Laborda-Illanes, A.; Medina, J.A.; Ramos-Molina, B.; Gómez-Millán, J.; Queipo-Ortuño, M.I. Gut Microbiota-Mediated Inflammation and Gut Permeability in Patients with Obesity and Colorectal Cancer. Int. J. Mol. Sci. 2020, 21, 6782.
  146. Liu, X.; Liu, H.; Yuan, C.; Zhang, Y.; Wang, W.; Hu, S.; Liu, L.; Wang, Y. Preoperative serum TMAO level is a new prognostic marker for colorectal cancer. Biomarkers Med. 2017, 11, 443–447.
  147. Chen, M.L.; Zhu, X.H.; Ran, L.; Lang, H.-D.; Yi, L.; Mi, M.-T. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J. Am. Heart Assoc. 2017, 6, e006347.
  148. Yue, C.; Yang, X.; Li, J.; Chen, X.; Zhao, X.; Chen, Y.; Wen, Y. Trimethylamine N-oxide prime NLRP3 inflammasome via inhibiting ATG16L1-induced autophagy in colonicepithelial cells. Biochem. Biophys. Res. Commun. 2017, 490, 541–551.
  149. Huang, C.-F.; Chen, L.; Li, Y.-C.; Wu, L.; Yu, G.-T.; Zhang, W.-F.; Sun, Z.-J. NLRP3 inflammasome activation promotes inflammation-induced carcinogenesis in head and neck squamous cell carcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 116.
  150. Wang, H.; Luo, Q.; Feng, X.; Zhang, R.; Li, J.; Chen, F. NLRP3 promotes tumor growth and metastasis in human oral squamous cell carcinoma. BMC Cancer 2018, 18, 500.
  151. He, Q.; Fu, Y.; Tian, D.; Yan, W. The contrasting roles of inflammasomes in cancer. Am. J. Cancer Res. 2018, 8, 566–583.
  152. Chen, J.; Pitmon, E.; Wnag, K. Micorbiome, inflammation and colorec5toal cancer. Semin. Immunol. 2017, 32, 43–53.
  153. Wu, D.; Cao, M.; Peng, J.; Li, N.; Yi, S.; Song, L.; Wang, X.; Zhang, M.; Zhao, J. The effect of trimethylamine N-oxide on Helicobacter pylori-induced changes of immunoinflammatory genes expression in gastric epithelial cells. Int. Immunopharmacol. 2017, 43, 172–178.
  154. Kumari, N.; Dwarakanath, B.S.; Das, A.; Bhatt, A.N. Role of interleukin-6 in cancer progression and therapeutic resistance. Tumor Biol. 2016, 37, 11553–11572.
  155. Tilg, H.; Adolph, T.E.; Gerner, R.R.; Moschen, A.R. The Intestinal Microbiota in Colorectal Cancer. Cancer Cell 2018, 33, 954–964.
  156. 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 Pt 3, 352–360.
  157. Reuter, S.; Gupta, S.C.; Chaturvedi, M.M.; Aggarwal, B.B. Oxidative stress, inflammation, and cancer: How are they linked? Free Radic. Biol. Med. 2010, 49, 1603–1616.
  158. Federico, A.; Morgillo, F.; Tuccillo, C.; Ciardiello, F.; Loguercio, C. Chronic inflammation and oxidative stress in human carcinogenesis. Int. J. Cancer 2007, 121, 2381–2386.
  159. Liu, Y.; Dai, M. Trimethylamine N-Oxide Generated by the Gut Microbiota Is Associated with Vascular Inflammation: New Insights into Atherosclerosis. Mediat. Inflamm. 2020, 2020, 4634172.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 328
Revisions: 2 times (View History)
Update Date: 14 Feb 2023
1000/1000
Video Production Service