Open access peer-reviewed chapter

Gut Microbial Metabolite Trimethylamine-N-Oxide and Its Role in Cardiovascular Diseases

Written By

Shubham Barik, Aritra Mukherjee, Angelina Job Kolady, Boda Karunakar and Tony Grace

Submitted: 18 June 2022 Reviewed: 09 September 2022 Published: 14 October 2022

DOI: 10.5772/intechopen.107976

From the Edited Volume

Novel Pathogenesis and Treatments for Cardiovascular Disease

Edited by David C. Gaze

Chapter metrics overview

98 Chapter Downloads

View Full Metrics

Abstract

Atherosclerosis (AS) is the common pathological underpinning of numerous cardiovascular illnesses (CVDs), and it is the leading cause of death worldwide. In recent years, researchers have begun to recognize the importance of gut microbiota in AS. Gut microbial dysbiosis has been reported to be connected with various CVDs. Moreover, dietary choline, betaine, and L-carnitine produce trimethylamine N-oxide (TMAO), a key gut microbe-dependent metabolite. Multiple studies have found a link between plasma TMAO levels and the likelihood of developing AS. The mechanism underlying this link, however, is still unknown. In this chapter, we discuss the TMAO-mediated mechanisms of atherosclerotic CVD from the perspectives of dietary patterns and gut microbial metabolism. Finally, we explain how TMAO has emerged as a novel therapeutic target for CVDs, as well as many treatment options for lowering TMAO levels that are currently being investigated, such as medications, dietary changes, probiotics, and so on.

Keywords

  • cardiovascular disease
  • gut microbiota
  • trimethylamine (TMA)
  • trimethylamine-N-oxide (TMAO)
  • diet
  • metabolism

1. Introduction

Cardiovascular disease or CVD is a term for disorders affecting the heart or blood vessels. Except in Africa, cardiovascular illnesses are the main cause of mortality globally, resulting in 17.9 million deaths (32.1%) in 2015, an increase from 12.3 million (25.8%) in 1990 [1]. CVD deaths are more widespread and have been growing in most developing countries, whereas rates in most developed countries have declined since the 1970s [2]. Coronary artery disease and stroke account for 80% of CVD deaths in males and 75% in females. The majority of cardiovascular disease affects older people. In the United States, 11% of adults between the ages of 20 y and 40 y have CVD, 37% between the ages of 40 y and 60 y, 71% between the ages of 60 y and 80 y, and 85% above the age of 80 y have CVD. In developed countries, the average age of death from coronary artery disease is over 80 y, while it is roughly 68 y in the developing world [2]. Diagnosis of diseases typically occurs seven to ten years earlier in men than in women.

The underlying processes differ according to the illness. Dietary risk factors are responsible for 53% of CVD fatalities [3]. Atherosclerosis is a common factor in coronary artery disease, stroke, and peripheral artery disease [4]. High blood pressure, smoking, diabetes mellitus, lack of exercise, obesity, high blood cholesterol, poor food, excessive alcohol consumption, and poor sleep, among other factors, may contribute to this [5]. High blood pressure contributes around 13% of CVD fatalities, whereas tobacco accounts for 9%, diabetes accounts for 6%, lack of exercise factors for 6%, and obesity accounts for 5%. Untreated strep throat can potentially cause rheumatic heart disease. Up to 90% of cardiovascular disease is thought to be preventable [6]. Lowering risk factors through good food, exercise, avoiding cigarette smoke, and limiting alcohol use are all part of CVD prevention. It also treats risk factors such as high blood pressure, lipids, and diabetes. In those with strep throat, antibiotics can lower the risk of rheumatic heart disease [7].

The microbiome plays a beneficial role in the homeostatic regulation of different body tissues of the host [8]. The overall relationship between humans and their microbiota can be described as a mutualistic symbiosis, also known as eubiosis [9]. This healthy balance of gut bacteria can be disrupted, leading to the onset of a variety of chronic diseases with an underlying inflammatory condition [10]. A large population of microbiota, predominantly bacteria, that populate the human gut have a symbiotic connection with the host, and imbalances in host-microbial interaction (dysbiosis) hamper these homeostatic systems that govern health and activate numerous pathways that contribute to advancing CVD risk factors [11]. Dysbiosis is related to intestinal inflammation and decreased gut barrier integrity, which raises circulating levels of bacterial structural components and microbial metabolites such as trimethylamine-N-oxide and short-chain fatty acids, which may aid in the development of CVDs [11].

Trimethylamine-N- oxide (TMAO) is a type of osmolyte found in the tissues of marine crustaceans and fish, where it prevents protein distortion and, therefore, the animal’s death [12]. The concentration of TMAO increases as the animal’s depth in the seas increases [13]. It is a protein stabilizer that counteracts the protein-destabilizing effects of pressure. In general, the bodies of animals living at great depths are adapted to high-pressure environments by having pressure-resistant biomolecules and small organic molecules present in their cells, known as piezolytes, of which TMAO is the most abundant. These piezolytes give the proteins the flexibility to function properly under great pressure [13, 14, 15]. However, more importantly, TMAO has emerged not only as an important metabolite in the human diet but also as a major cardiometabolic risk factor. It has been associated with many cardiovascular complications including foam cell formation [16], endothelial dysfunction [17], acute heart failure [18], infracted coronary artery [19], inflammation [20] and vascular aging [21].

Advertisement

2. Role of gut microbes in regulating cardiovascular health and disease

The gut microbiome has emerged as a critical factor in human health and disease [22, 23] and cardio-metabolic diseases are no exception. Obesity and insulin resistance are serious cardiometabolic risk factors [24, 25, 26, 27], and gut microbial composition is a major regulator of these conditions. Changes in fecal microbial community composition have been linked to the development of obesity and insulin resistance, and microbial transplantation has been shown to transmit increased adiposity in the host [28, 29, 30]. Disruptions to the microbiota early in life have since been identified to induce increased obesity [31]. Koren et al. [32] argued that microbiota could be associated with atherosclerosis since human atherosclerotic plaques were found to contain bacterial DNA, albeit it was unclear if the DNA came from live bacteria within the arterial wall. The initial research into a possible link between the gut microbiome and cardiovascular disease (CVD) focused on trimethylamine N-oxide (TMAO), a metaorganismal metabolite generated after ingestion of food substances plentiful in a Western diet (eg, carnitine, lecithin, choline) [16, 33, 34]. TMAO has swiftly established itself as a biomarker for human CVD risk as well as a promoter of atherothrombotic disease [35, 36]. In fact, a Western-style diet deficient in microbiota-accessible carbohydrates (MACs) may cause irreversible microbial diversity loss and the extinction of particular bacterial species in the digestive system [37]. As a result, the low intake of dietary fiber and increased levels of fat and sugar in our food, which are typical of a westernized lifestyle and diet, may contribute to the depletion of specific bacterial taxa, at least in part [38]. Fiber, fruit, legume and vegetable consumption is linked to an increased microbial richness in the gut microbiota [39, 40], and several recent epidemiological studies have found an inverse relationship between dietary fiber consumption and CVD risk variables [41, 42, 43, 44, 45]. Non -digestible carbohydrates present in dietary fiber are converted by intestinal bacteria into Short-Chain fatty acids(SCFAs) like acetate, propionate and butyrate [46, 47]. SCFAs have been shown to have a direct effect on renin release and vasomotor function, resulting in lower blood pressure [48, 49, 50]. Butyrate has been shown to have a potential adjuvant effect in the lowering of diastolic blood pressure by reducing inflammation in a recent controlled experiment [51]. Moreover, in early pregnancy, the presence of butyrate-producing bacteria was found to be inversely related to blood pressure and plasminogen activator inhibitor-1 levels [52].

2.1 Gut microbiota dysbiosis and implications in CVD risk

Most microbiome-related diseases have skyrocketed in the last century, implying that a change in lifestyle could disturb gut microbiota symbiosis by removing helpful, protective bacteria [53]. Patients with a variety of CVD risk factors, such as hypertension, dyslipidemia, insulin resistance, and other metabolic abnormalities, have been found to have variations in microbial composition [36, 37]. Dysbiosis of the gut microbiota can lead to chronic inflammation, which is a major contributor to obesity, cardiovascular disease, and notably atherosclerosis [3839]. In symptomatic atherosclerosis patients, metagenome research indicated a higher concentration of triglycerides and a lower level of high-density lipoprotein in the circulation, as well as an increased abundance of Collinsella and a decreased abundance of Roseburia and Eubacterium [54]. Jie et al. [55] discovered an elevated relative abundance of Enterobacteriaceae and Streptococcus spp. taxa in atherosclerotic CVD patients. In coronary artery disease patients, Emoto et al. discovered a distinct alteration in microbial composition, with a large increase in Lactobacillales (Firmicutes) and a decrease in Bacteroidetes [56]. In another study, patients with type 2 diabetes had a lower number of Firmicutes and a non-significant rise in Bacteroidetes and Proteobacteria [57]. Some cross-sectional studies have found evidence that high-protein and high-fat diets (associated with Western lifestyles) are linked to gut microbial populations characterized by the Bacteroides enterotype, while diets heavy in carbohydrates and simple sugars are linked to the Prevotella enterotype [58].

The metabolism-independent pathway and the metabolism-dependent pathway are two key pathways via which gut dysbiosis can contribute to the development and progression of atherosclerosis [59]. In the metabolism -independent pathways, bacterial components located on the outer membrane of Gram-negative bacteria, such as lipopolysaccharides (LPS), can encourage the production of foam cells, which are a primary component of atherosclerotic plaque [60]. To prevent the accumulation of excess cholesterol in peripheral tissues, the body has internal homeostatic systems in place, such as reverse cholesterol transport (RCT). Excess cholesterol is transported to the liver and transformed into bile acids through the RCT process [61, 62, 63]. By producing metabolic endotoxemia, gut dysbiosis can overload systems like RCT and encourage the development of foam cells [64, 65, 66]. Metabolic endotoxemia is a condition marked by a high level of LPS in the bloodstream [67]. The presence of Bifidobacteria, which typically enhance intestinal barrier function and inhibit bacterial translocation, is reduced in high-fat (HF) diet-induced dysbiosis [60].

Advertisement

3. Synthesis and metabolism of gut microbial metabolite TMA and TMAO

3.1 Production of trimethylamine by gut bacteria

Trimethylamine (TMA) is the source of TMAO in humans. TMA is derived either directly from meals high in TMA, such seafood, [68, 69] or indirectly from the bacterial metabolism of dietary choline and choline-containing substances in the colon, such as phosphatidylcholine [16], betaine [70], and dietary L-carnitine [33, 71]. The ability of different gut microbes to produce TMA from food precursors varies. This is because it is produced in the gut via a variety of microbial mechanisms (Figure 1). As a result, the composition of an individual’s microbiota influences the magnitude of TMA production. It’s worth noting that the genes essential for TMA formation are found in just a small percentage of the microorganisms in the intestine (less than 1%) [72]. TMA formation appears to be possible even at extremely low concentrations of these microorganisms, highlighting the importance of the gut microbiota in this context [73]. TMA and TMAO levels have been linked to increased activity in bacteria belonging to the phylum Firmicutes and Proteobacteria, which are known producers of this metabolite. Furthermore, because Bacteroidetes are unable to make TMA [74], TMA and TMAO levels have been connected to an enhanced Firmicutes/Bacteroidetes ratio, with higher levels of Firmicutes and lower levels of Bacteroidetes [57, 58]. The genes coding for the glycyl radical enzyme choline TMA-lyase (CutC) and its related radical S-adenosyl-L-methionine (SAM)(CutD) activating protein were discovered in the Choline Utilization (cut) gene cluster in gut bacteria, which is responsible for the anaerobic breakdown of choline into TMA [75]. A two-component CntA/CntB oxygenase/reductase system capable of cleaving L-carnitine into TMA and malic semialdehyde is another microbial metabolic route that generates TMA from L-carnitine [76]. The yeaW/X gene products (YeaW/X TMA lyase) are a closely similar bacterial lyase. Choline, betaine, L-carnitine, and -butyrobetaine can all be converted to TMA by this promiscuous lyase [74, 77]. Aside from these TMA-generating processes from dietary trimethylamines, some gut microbes like E.coli have also been found to have another pathway that converts TMAO to TMA via the activity of a torA-like gene product which acts as a reductase [78, 79].

Figure 1.

Chemical formulae of TMA and TMAO’s principal dietary precursors. The key metabolic pathways for the synthesis of TMA by the gut microbiota and endogenous enzymes, as well as the conversion of TMA to TMAO by hepatic FMOs, are depicted in this diagram.

3.2 Conversion of TMA into TMAO and its regulation

TMA generated from a choline -rich diet through various metabolic pathways is absorbed from the gut into the hepatic portal circulation and oxidized by the enzymes flavin-dependent monooxygenase isoforms 1 and 3 (FMO1 and FMO3) in the liver to create Trimethylamine-N-oxide(TMAO) (Figure 2) [80]. TMAO is excreted out of the body, usually through urine [81]. Sweat, feces (4%), exhaled air (less than 1%), and other body secretions are some of the other ways TMAO is excreted [82]. TMAO can be metabolized to DMA(Dimehtylamine), formaldehyde, ammonia, and methane by methanogenic bacteria that carry the TMAO demethylase enzyme [83]. Furthermore, it has been demonstrated that TMAO derived from food can be absorbed directly in the gut [84]. As a result, plasma TMAO levels are regulated by TMA synthesis and degradation, as well as the rate at which TMA, and TMAO are secreted [85].

Figure 2.

Gut flora mediated synthesis of TMA and hepatic conversion to TMAO.

3.3 Dietary precursors of TMAO and the relationship between TMAO levels and dietary habits

As discussed in Section 3.1, seafood is a rich source of dietary TMA/TMAO and various dietary precursors like L-carnitine, choline, ergothioneine and betaine (Figure 1) equally contribute to the generation of TMAO in the body. Free TMAO present in seafood is not metabolized by gut microbiota and is directly absorbed into the systemic circulation [86]. L-carnitine is present in high concentrations in meals derived from animals (meat and dairy products), and in smaller amounts in grains and vegetables [87]. The most common sources of choline in the diet are eggs and liver, followed by meats and fish, whole grain, cereal, vegetables, fruits, milk, fats, and oils [88]. One of the most important sources of betaine is cereal-based foods [89]. Betaine can also be found in spinach, beets, crabs, and finfish [90]. Dietary sources are the only way to get ergothioneine. Ergothioneine is found in only a few foods, with the largest quantities found in boletus and oyster mushrooms, as well as to a lesser level in chicken and pork liver and kidney, oat bran, and black and red beans [91]. As discussed in Section 2, a westernized lifestyle and diet full of junk fatty foods and refined sugar, devoid of fiber and important nutrients, predisposes one to increased CVD risk and other chronic diseases. Plasma TMAO levels have been observed to rise when people eat Western-style or high-fat diets [92, 93, 94]. However, conversely, epidemiological studies have linked the Mediterranean diet to a lower risk of cardiovascular disease (CVD) [95]. A typical Mediterranean diet is defined by plant based foods (vegetables, fruits, nuts,), olive oil based fats and moderate to low amounts of seafood, eggs and meats [96, 97]. This makes this type of diet high in fiber and low in choline -rich food. The importance of fiber- rich foods has already been mentioned in Section 2. High dietary fiber consumption, followed by gut microbiota-mediated fermentation, appears to reduce TMAO levels in experiments on animal models and clinical medicine [98].

Advertisement

4. Role of TMAO in increasing cardiovascular disease risk

A choline-rich diet puts a person at risk of increased TMAO levels [16], which is directly correlated to an increased CVD risk [99]. Angiographic markers of coronary artery atherosclerotic burden and cardiac risks have strong relationships with systemic TMAO levels, and higher levels of TMAO in the blood are linked to an increased risk of incident cardiovascular events such as myocardial infarction, recurrent stroke, and even cardiovascular death [55, 100]. Gut microbes play a role in modifying platelet reactivity and generating a pro-thrombotic phenotype in vivo by producing TMAO (Figure 3) [101]. Zhu et al., has shown that direct exposure of platelets to TMAO, caused activation of the platelets by the release of intracellular calcium. This modulates the platelet hyper-responsiveness and the potential of thrombosis and causes thrombosis and atherosclerosis [101]. Rebecca et al., states that knockdown of FMOs can protect mice from obesity, which is a major cause for cardiovascular diseases [102, 103]. Increased amount of TMAO, obtained from the diet, causes monocytes to enter the subendothelial space and differentiate into colony -stimulating factors when they encounter the growth factors. These form large cells known as dendritic cells and macrophages which possess high expression of SR-A1 and CD36 [104]. These cells take up oxidized, low- density lipid particles to create foam cells that are irregular in the uptake of cholesterol with fatty acids and ester bonds, thus stimulating atherosclerosis [105]. It is suggested that CD36/MAPK/JNK pathways play a vital role in the formation of foam cells [106]. Research studies show that apoe−/− mice fed with choline diet for 8 weeks, gradually exhibited an increase in TMAO, which further recruited macrophages and pro – inflammatory cytokines [107]. Another study by Boini et al. indicates the link between TMAO and inflammation, where TMAO induces NLRP3 inflammasome formation and causes other immune responses [108]. An imbalance of cholesterol transport is observed in individuals with high TMAO, and studies show that mice with administered TMAO inhibited the synthesis of hepatic bile acid by downregulating the expression of Cyp7a1, which promoted atherosclerosis [109]. The activation of oxidative stress pathways following exposure to TMAO, which triggers inflammatory cytokines, is the molecular basis for increasing cardiovascular illnesses. It can also activate the p38 MAPK and NF-kappa beta signaling pathways, which enhances NLRP3 production in the inflammasome and promotes vascular calcification and endothelial cell damage [110]. High administration of TMAO causes oxidative stress, inflammation and suppressed cellular functions, while low levels exhibit a contrary response [111]. A recent study proved that patients with aortic stenosis, had their TMAO levels as 5.5 μM, when the control was 3.6 μM. TMA is also associated with cardiovascular diseases as the levels of TMA in these patients were 59.5 μM and the control was 23.2 μM [112]. Thus, TMAO is considered to be an independent risk factor for cardiovascular diseases.

Figure 3.

TMAO- mediated platelet hyper-responsiveness and increased thrombosis risk.

Advertisement

5. Targeting the TMA/TMAO pathway as a therapeutic strategy to combat CVD risk-current research and future directions

The gut microbiome is a growing area of research in metabolic health and its link to CVD risk. The development of high-throughput metagenomic tools has aided a new understanding of the gut microbiome’s role in CVD risk [113]. The gut microbiome can be targeted to modify TMAO synthesis, according to recent fecal microbial transplant research [114] and as a result TMA/TMAO levels can be regulated. Based on research by Maisto et al., in healthy subjects, grape pomace polyphenolic extract has been found to lower serum levels of TMAO [115]. Resveratrol (RSV) reduces TMAO- induced atherosclerosis by lowering TMAO levels and enhancing hepatic bile acid synthesis through gut microbiota remodeling [116]. Antimicrobial phytochemicals, such as allicin, a dietary dosage derived from garlic, effectively neutralize the metabolic ability of gut microbiota to produce TMAO- induced by L-carnitine intake [117]. Luhong granules, a complex blend of herbs, flowers, animal parts, seeds, and roots, prolong ventricular remodeling after myocardial infarction by lowering TMAO and LPS levels in the bloodstream by increasing the gut microflora and intestinal barrier function [118]. A single oral dosage of a cutC/D inhibitor lowers plasma TMAO levels for up to three days and reverses diet-induced platelet reactivity and thrombus formation as studied in animal models, with no toxicity or increased bleeding risk [119]. In experiments with mice models, Lactobacillus plantarum ZDY04 significantly reduced serum TMAO and cecal TMA levels in mice by modulating the relative abundance of specific bacterial species, including Bacteroids and significantly inhibiting the development of TMAO-induced atherosclerosis in choline fed mice [120]. In high-fat diet-induced obese mice, capsanthin extract prevents obesity, lowers serum TMAO levels, and modifies the gut microbiota composition by decreasing serum triglycerides, total cholesterol, and TMAO levels and markedly increasing microbial diversity [121]. The ability of several oral probiotics to modify circulating TMAO levels in different cohorts, including healthy participants and patients with a CVD-related disease, has been investigated [122, 123, 124, 125]. None of them, however, appeared to have a significant effect on TMAO levels in the treatment groups as compared to the placebo groups. In another study, TMA-degrading microorganisms were used by Qiu et al. (2017) to investigate another promising technique for lowering TMA levels in the gut where oral administration of a TMA-metabolizing strain (Enterobacter aerogenes ZDY01) reduced TMA in the cecum and TMAO in the serum, as well as changing the microbial community composition in mice, according to their findings [126]. In human studies, changes in urine TMAO levels have been discovered in untargeted metabolomics investigations following supplementation with Origanum dictamus tea and Curcuma longa extract [127, 128].

Advertisement

6. Conclusions

Diet has been shown to have an important role in the formation of TMAO because it offers the nutritional precursors needed to create TMA and TMAO. There is a positive correlation between circulating TMAO levels and the consumption of food rich in dietary precursors of TMAO like seafood, meat, eggs etc. Targeting the TMA/TMAO metabolism has emerged as a promising tool for cardiovascular disease prevention and treatment in recent years. Targeting the microbiota and host metabolic systems implicated in TMA and TMAO production shows potential for future intervention. Animal models have largely established the capacity of specific diets, food ingredients, and phytochemicals found in herbs to reduce circulation of TMAO levels. The link between changes in TMAO levels and gut microbiota has only been shown in a few cases, and the exact processes behind the impacts of the dietary items under investigation are yet unknown. More importantly, there are few studies that suggest that lowering circulating TMAO levels has a favorable effect in humans. Because the majority of the studies have been conducted on animal models, the results are difficult to apply to humans. Future research in this area should address conventional microbial research obstacles as well as those more specific to the study of TMA/TMAO metabolism, such as the substantial intra-individual variability of plasma TMAO levels observed in some humans. With the advancement and availability of next-generation sequencing and other omics technologies, a change from studies focusing on defining microbial community composition to more function-oriented research on the gut microbiota is envisaged. Bioinformatic approaches, shotgun metagenomics, meta-transcriptomics, meta-proteomics, and metabolomics, are all expected to be crucial in unraveling the intricate relationships between nutrition, microbial metabolism, and host health.

Advertisement

Acknowledgments

All images in this manuscript were made using BioRender.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. GBD 2013 mortality and cause of death collaborators. Global, regional, and national age–sex specific all-cause and cause-specific mortality for 240 causes of death, 1990-2013: A systematic analysis for the global burden of disease study 2013. Lancet. 2015;385(9963):117-171
  2. 2. Moran AE et al. Temporal trends in ischemic heart disease mortality in 21 world regions, 1980 to 2010: The global burden of disease 2010 study. Circulation. 2014;129(14):1483-1492
  3. 3. Petersen KS, Kris-Etherton PM. Diet quality assessment and the relationship between diet quality and cardiovascular disease risk. Nutrients. 2021;13(12):4305
  4. 4. Criqui MH, Denenberg JO. The generalized nature of atherosclerosis: How peripheral arterial disease may predict adverse events from coronary artery disease. Vascular Medicine. 1998;3(3):241-245
  5. 5. deGoma EM, Knowles JW, Angeli F, Budoff MJ, Rader DJ. The evolution and refinement of traditional risk factors for cardiovascular disease. Cardiology in Review. 2012;20(3):118-129
  6. 6. McGill HC, McMahan CA, Gidding SS. Preventing heart disease in the 21st century. Circulation. 2008;117(9):1216-1227
  7. 7. Spinks A, Glasziou PP, Del Mar CB. Antibiotics for treatment of sore throat in children and adults. Cochrane Database of Systematic Reviews. 2021;12, no. 12:CD000023
  8. 8. Schroeder BO, Bäckhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nature Medicine. 2016;22(10):1079-1089
  9. 9. Walter J, Britton RA, Roos S. Host-microbial symbiosis in the vertebrate gastrointestinal tract and the lactobacillus reuteri paradigm. Proceedings of the National Academy of Sciences. 2011;108(Supplement 1):4645-4652
  10. 10. Hand TW, Vujkovic-Cvijin I, Ridaura VK, Belkaid Y. Linking the microbiota, chronic disease, and the immune system. Trends in Endocrinology and Metabolism. 2016;27(12):831-843
  11. 11. Novakovic M et al. Role of gut microbiota in cardiovascular diseases. World Journal of Cardiology. 2020;12(4):110-122
  12. 12. Yancey PH, Clark ME, Hand SC, Bowlus RD, Somero GN. Living with water stress: Evolution of Osmolyte systems. Science (80-.). 1982;217(4566):1214-1222
  13. 13. Linley TD, Gerringer ME, Yancey PH, Drazen JC, Weinstock CL, Jamieson AJ. Fishes of the hadal zone including new species, in situ observations and depth records of Liparidae. Deep Sea Research part I: Oceanographic Research Papers. 2016;114:99-110
  14. 14. Yancey PH. Organic osmolytes as compatible, metabolic and counteracting cytoprotectants in high osmolarity and other stresses. The Journal of Experimental Biology. 2005;208(15):2819-2830
  15. 15. Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-oxide: The good, the bad and the unknown. Toxins (Basel). 8 Nov 2016;8(11):326. DOI: 10.3390/toxins8110326
  16. 16. Wang Z et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57-63
  17. 17. Sun X et al. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochemical and Biophysical Research Communications. 2016;481(1-2):63-70
  18. 18. Suzuki T, Heaney LM, Bhandari SS, Jones DJL, Ng LL. Trimethylamine N-oxide and prognosis in acute heart failure. Heart. 2016;102(11):841-848
  19. 19. Mafune A et al. Associations among serum trimethylamine-N-oxide (TMAO) levels, kidney function and infarcted coronary artery number in patients undergoing cardiovascular surgery: A cross-sectional study. Clinical and Experimental Nephrology. 2016;20(5):731-739
  20. 20. Chen ML, Zhu XH, Ran L, Lang HD, Yi L, Mi MT. Trimethylamine-N-oxide induces vascular inflammation by activating the NLRP3 Inflammasome through the SIRT3-SOD2-mtROS signaling pathway. Journal of the American Heart Association. 4 Sep 2017;6(9):e006347. DOI: 10.1161/JAHA.117.006347. Erratum in: J Am Heart Assoc.
  21. 21. Li D et al. Trimethylamine-N-oxide promotes brain aging and cognitive impairment in mice. Aging Cell. 2018;17(4):e12768
  22. 22. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: An integrative view. Cell. 2012;148(6):1258-1270
  23. 23. Wang J, Jia H. Metagenome-wide association studies: Fine-mining the microbiome. Nature Reviews. Microbiology. 2016;14(8):508-522
  24. 24. McFarlane SI, Banerji M, Sowers JR. Insulin resistance and cardiovascular disease. The Journal of Clinical Endocrinology and Metabolism. 2001;86(2):713-718
  25. 25. Reaven G, Abbasi F, McLaughlin T. Obesity, insulin resistance, and cardiovascular disease. Recent Progress in Hormone Research. 2004;59:207-224
  26. 26. Bonow RO, Eckel RH. Diet, obesity, and cardiovascular risk. The New England Journal of Medicine. 2003;348(21):2057-2133
  27. 27. Sharma AM. Obesity and cardiovascular risk. Growth Hormone & IGF Research. 2003;13:S10-S17
  28. 28. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proceedings of the National Academy of Sciences. 2005;102(31):11070-11075
  29. 29. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027-1031
  30. 30. Bäckhed F et al. The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences. 2004;101(44):15718-15723
  31. 31. Cho I et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature. 2012;488(7413):621-626
  32. 32. Koren O et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proceedings of the National Academy of Sciences. 2011;108(Supplement 1):4592-4598
  33. 33. Koeth RA et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature Medicine. 2013;19(5):576-585
  34. 34. Tang WHW et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. The New England Journal of Medicine. 2013;368(17):1575-1584
  35. 35. Guasti L et al. TMAO as a biomarker of cardiovascular events: A systematic review and meta-analysis. Internal and Emergency Medicine. 2021;16(1):201-207
  36. 36. Randrianarisoa E et al. Relationship of serum trimethylamine N-oxide (TMAO) levels with early atherosclerosis in humans. Scientific Reports. 2016;6(1):1-9
  37. 37. Sonnenburg ED, Smits SA, Tikhonov M, Higginbottom SK, Wingreen NS, Sonnenburg JL. Diet-induced extinctions in the gut microbiota compound over generations. Nature. 2016;529(7585):212-215
  38. 38. Sonnenburg ED, Sonnenburg JL. Starving our microbial self: The deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metabolism. 2014;20(5):779-786
  39. 39. Bourquin LD, Titgemeyer EC, Fahey GC Jr. Fermentation of various dietary fiber sources by human fecal bacteria. Nutrition Research. 1996;16(7):1119-1131
  40. 40. Bourquin LD, Titgemeyer EC, Fahey GC Jr. Vegetable fiber fermentation by human fecal bacteria: Cell wall polysaccharide disappearance and short-chain fatty acid production during in vitro fermentation and water-holding capacity of unfermented residues. The Journal of Nutrition. 1993;123(5):860-869
  41. 41. Liu L, Wang S, Liu J. Fiber consumption and all-cause, cardiovascular, and cancer mortalities: A systematic review and meta-analysis of cohort studies. Molecular Nutrition & Food Research. 2015;59(1):139-146
  42. 42. Micha R et al. Etiologic effects and optimal intakes of foods and nutrients for risk of cardiovascular diseases and diabetes: Systematic reviews and meta-analyses from the nutrition and chronic diseases expert group (NutriCoDE). PLoS One. 2017;12(4):e0175149
  43. 43. McRae MP. Dietary fiber is beneficial for the prevention of cardiovascular disease: An umbrella review of meta-analyses. Journal of Chiropractic Medicine. 2017;16(4):289-299
  44. 44. Kim Y, Je Y. Dietary fibre intake and mortality from cardiovascular disease and all cancers: A meta-analysis of prospective cohort studies. Archives of Cardiovascular Diseases. 2016;109(1):39-54
  45. 45. Hajishafiee M, Saneei P, Benisi-Kohansal S, Esmaillzadeh A. Cereal fibre intake and risk of mortality from all causes, CVD, cancer and inflammatory diseases: A systematic review and meta-analysis of prospective cohort studies. The British Journal of Nutrition. 2016;116(2):343-352
  46. 46. Duncan SH, Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE. Reduced dietary intake of carbohydrates by obese subjects results in decreased concentrations of butyrate and butyrate-producing bacteria in feces. Applied and Environmental Microbiology. 2007;73(4):1073-1078
  47. 47. Cummings J, Pomare EW, Branch WJ, Naylor CP, MacFarlane G. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28(10):1221-1227
  48. 48. Pluznick JL et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proceedings of the National Academy of Sciences. 2013;110(11):4410-4415
  49. 49. Marques FZ et al. High-fiber diet and acetate supplementation change the gut microbiota and prevent the development of hypertension and heart failure in hypertensive mice. Circulation. 2017;135(10):964-977
  50. 50. Natarajan N et al. Microbial short chain fatty acid metabolites lower blood pressure via endothelial G protein-coupled receptor 41. Physiological Genomics. 2016;48(11):826-834
  51. 51. Roshanravan N et al. Effect of butyrate and inulin supplementation on glycemic status, lipid profile and glucagon-like peptide 1 level in patients with type 2 diabetes: A randomized double-blind, placebo-controlled trial. Hormone and Metabolic Research. 2017;49(11):886-891
  52. 52. Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M, Dekker Nitert M. Increased systolic and diastolic blood pressure is associated with altered gut microbiota composition and butyrate production in early pregnancy. Hypertension. 2016;68(4):974-981
  53. 53. Logan AC, Jacka FN, Prescott SL. Immune-microbiota interactions: Dysbiosis as a global health issue. Current Allergy and Asthma Reports. 2016;16(2):1-9
  54. 54. Karlsson FH et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nature Communications. 2012;3(1):1-8
  55. 55. Jie Z et al. The gut microbiome in atherosclerotic cardiovascular disease. Nature Communications. 2017;8(1):845
  56. 56. Emoto T, Yamashita T, Sasaki N, Hirota Y, Hayashi T, So A, et al. Analysis of Gut Microbiota in Coronary Artery Disease Patients: a Possible Link between Gut Microbiota and Coronary Artery Disease. Journal of Atherosclerosis and Thrombosis. 1 Aug 2016;23(8):908-921. DOI: 10.5551/jat.32672
  57. 57. Wong JMW. Gut microbiota and cardiometabolic outcomes: Influence of dietary patterns and their associated components. The American Journal of Clinical Nutrition. 2014;100(suppl_1):369S-377S
  58. 58. Wu GD et al. Linking long-term dietary patterns with gut microbial Enterotypes. Science (80-.). 2011;334(6052):105-108
  59. 59. Brown JM, Hazen SL. The gut microbial endocrine organ: Bacterially derived signals driving cardiometabolic diseases. Annual Review of Medicine. 2015;66:343-359
  60. 60. Manco M, Putignani L, Bottazzo GF. Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocrine Reviews. 2010;31(6):817-844
  61. 61. Ohashi R, Mu H, Wang X, Yao Q , Chen C. Reverse cholesterol transport and cholesterol efflux in atherosclerosis. QJM. 2005;98(12):845-856
  62. 62. Spady DK. Reverse cholesterol transport and atherosclerosis regression. Circulation. 1999;100(6) Am Heart Assoc:576-578
  63. 63. Annema W, Tietge UJF. Regulation of reverse cholesterol transport-a comprehensive appraisal of available animal studies. Nutrition & Metabolism (London). 2012;9(1):1-18
  64. 64. Cuchel M, Rader DJ. Macrophage reverse cholesterol transport: Key to the regression of atherosclerosis? Circulation. 2006;113(21):2548-2555
  65. 65. Lo Sasso G et al. Intestinal specific LXR activation stimulates reverse cholesterol transport and protects from atherosclerosis. Cell Metabolism. 2010;12(2):187-193
  66. 66. Castrillo A et al. Crosstalk between LXR and toll-like receptor signaling mediates bacterial and viral antagonism of cholesterol metabolism. Molecular Cell. 2003;12(4):805-816
  67. 67. Mohammad S, Thiemermann C. Role of metabolic endotoxemia in systemic inflammation and potential interventions. Frontiers in Immunology. 11 Jan 2021;11:594150. DOI: 10.3389/fimmu.2020.594150
  68. 68. Seibel BA, Walsh PJ. Trimethylamine oxide accumulation in marine animals: Relationship to acylglycerol storagej. The Journal of Experimental Biology. 2002;205(3):297-306
  69. 69. Zeisel SH, DaCosta K-A. Increase in human exposure to methylamine precursors of N-nitrosamines after eating fish. Cancer Research. 1986;46(12 Part 1):6136-6138
  70. 70. Day-Walsh P et al. The use of an in-vitro batch fermentation (human colon) model for investigating mechanisms of TMA production from choline, l-carnitine and related precursors by the human gut microbiota. European Journal of Nutrition. 2021;60(7):3987-3999
  71. 71. Rajakovich LJ, Fu B, Bollenbach M, Balskus EP. Elucidation of an anaerobic pathway for metabolism of l-carnitine–derived γ-butyrobetaine to trimethylamine in human gut bacteria. Proceedings of the National Academy of Sciences. 2021;118(32):e2101498118
  72. 72. Rath S, Heidrich B, Pieper DH, Vital M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome. 2017;5(1):1-14
  73. 73. Romano KA, Vivas EI, Amador-Noguez D, Rey FE. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio. 2015;6(2):e02481-e02414
  74. 74. Falony G, Vieira-Silva S, Raes J. Microbiology meets big data: The case of gut microbiota–derived trimethylamine. Annual Review of Microbiology. 2015;69:305-321
  75. 75. Smaranda C, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proceedings of the National Academy of Sciences. 2012;109(52):21307-21312
  76. 76. Yijun Z et al. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proceedings of the National Academy of Sciences. 2014;111(11):4268-4273
  77. 77. Wang Z et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell. 2015;163(7):1585-1595
  78. 78. McCrindle SL, Kappler U, McEwan AG. Microbial dimethylsulfoxide and trimethylamine-N-oxide respiration. Advances in Microbial Physiology. 2005;50:147-198. DOI: 10.1016/S0065-2911(05)50004-3
  79. 79. Méjean V, Lobbi-Nivol C, Lepelletier M, Giordano G, Chippaux M, Pascal M. TMAO anaerobic respiration in Escherichia coli: Involvement of the tor operon. Molecular Microbiology. 1994;11(6):1169-1179
  80. 80. Bennett BJ et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metabolism. 2013;17(1):49-60
  81. 81. Yu D et al. Urinary levels of trimethylamine-N-oxide and incident coronary heart disease: A prospective investigation among urban Chinese adults. Journal of the American Heart Association. 2019;8(1):e010606
  82. 82. Papandreou C, Moré M, Bellamine A. Trimethylamine N-oxide in relation to cardiometabolic health—Cause or effect? Nutrients. 2020;12(5):1330
  83. 83. Chhibber-Goel J, Gaur A, Singhal V, Parakh N, Bhargava B, Sharma A. The complex metabolism of trimethylamine in humans: Endogenous and exogenous sources. Expert Reviews in Molecular Medicine. 29 Apr 2016;18:e8. DOI: 10.1017/erm.2016.6. Erratum in: Expert Rev Mol Med. 2016 Nov 23;18:e19.
  84. 84. Zhang AQ , Mitchell SC, Smith RL. Dietary precursors of trimethylamine in man: A pilot study. Food and Chemical Toxicology. 1999;37(5):515-520
  85. 85. Gessner A, di Giuseppe R, Koch M, Fromm MF, Lieb W, Maas R. Trimethylamine-N-oxide (TMAO) determined by LC-MS/MS: Distribution and correlates in the population-based PopGen cohort. Clinical Chemistry and Laboratory Medicine. 2020;58(5):733-740
  86. 86. Canyelles M, Tondo M, Cedó L, Farràs M, Escolà-Gil JC, Blanco-Vaca F. Trimethylamine N-oxide: A link among diet, gut microbiota, gene regulation of liver and intestine cholesterol homeostasis and HDL function. International Journal of Molecular Sciences. 2018;19(10):3228
  87. 87. Steiber A, Kerner J, Hoppel CL. Carnitine: A nutritional, biosynthetic, and functional perspective. Molecular Aspects of Medicine. 2004;25(5):455-473
  88. 88. Patterson KY et al. USDA Database for the Choline Content of Common Foods, Release Two. Center, ARS, USDA: Nutr. Data Lab. Beltsv. Hum. Nutr. Res; 2008
  89. 89. Filipčev B, Kojić J, Krulj J, Bodroža-Solarov M, Ilić N. Betaine in cereal grains and grain-based products. Foods. 2018 Mar 29;7(4):49. DOI: 10.3390/foods7040049
  90. 90. Zeisel SH, Mar M-H, Howe JC, Holden JM. Concentrations of choline-containing compounds and betaine in common foods. The Journal of Nutrition. 2003;133(5):1302-1307
  91. 91. Ey J, Schömig E, Taubert D. Dietary sources and antioxidant effects of Ergothioneine. Journal of Agricultural and Food Chemistry. 2007;55(16):6466-6474
  92. 92. Malinowska AM, Szwengiel A, Chmurzynska A. Dietary, anthropometric, and biochemical factors influencing plasma choline, carnitine, trimethylamine, and trimethylamine-N-oxide concentrations. International Journal of Food Sciences and Nutrition. 2017;68(4):488-495
  93. 93. Chen K, Zheng X, Feng M, Li D, Zhang H. Gut microbiota-dependent metabolite trimethylamine N-oxide contributes to cardiac dysfunction in Western diet-induced obese mice. Frontiers in Physiology. 21 Mar 2017;8:139. DOI: 10.3389/fphys.2017.00139
  94. 94. Boutagy NE et al. Short-term high-fat diet increases postprandial trimethylamine-N-oxide in humans. Nutrition Research. 2015;35(10):858-864
  95. 95. Serra-Majem L, Román-Viñas B, Sanchez-Villegas A, Guasch-Ferré M, Corella D, La Vecchia C. Benefits of the Mediterranean diet: Epidemiological and molecular aspects. Molecular Aspects of Medicine. 2019;67:1-55
  96. 96. Willett WC et al. Mediterranean diet pyramid: A cultural model for healthy eating. The American Journal of Clinical Nutrition. 1995;61(6):1402S-1406S
  97. 97. Bach-Faig A et al. Mediterranean diet pyramid today. Science and cultural updates. Public Health Nutrition. 2011;14(12A):2274-2284
  98. 98. Li Q, Wu T, Liu R, Zhang M, Wang R. Soluble dietary fiber reduces trimethylamine metabolism via gut microbiota and Co-regulates host AMPK pathways. Molecular Nutrition & Food Research. Dec 2017;61(12). DOI: 10.1002/mnfr.201700473
  99. 99. Haghikia A et al. Gut microbiota–dependent trimethylamine N-oxide predicts risk of cardiovascular events in patients with stroke and is related to proinflammatory monocytes. Arteriosclerosis, Thrombosis, and Vascular Biology. 2018;38(9):2225-2235
  100. 100. Hoyles L et al. Metabolic retroconversion of trimethylamine N-oxide and the gut microbiota. Microbiome. 2018;6(1):1-14
  101. 101. Zhu W et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 2016;165(1):111-124
  102. 102. Schugar RC et al. The TMAO-producing enzyme flavin-containing monooxygenase 3 regulates obesity and the beiging of white adipose tissue. Cell Reports. 2017;19(12):2451-2461
  103. 103. Liu Y, Dai M. Trimethylamine N-oxide generated by the gut microbiota is associated with vascular inflammation: New insights into atherosclerosis. Mediators of Inflammation. 17 Feb 2020;2020:4634172. DOI: 10.1155/2020/4634172
  104. 104. Yang S et al. Gut microbiota-dependent marker TMAO in promoting cardiovascular disease: Inflammation mechanism, clinical prognostic, and potential as a therapeutic target. Frontiers in Pharmacology. 2019;10:1360
  105. 105. Wu K et al. The gut microbial metabolite trimethylamine N-oxide aggravates GVHD by inducing M1 macrophage polarization in mice. Blood. 2020;136(4):501-515
  106. 106. Geng J et al. Trimethylamine N-oxide promotes atherosclerosis via CD36-dependent MAPK/JNK pathway. Biomedicine & Pharmacotherapy. 2018;97:941-947
  107. 107. Lindskog Jonsson A et al. Impact of gut microbiota and diet on the development of atherosclerosis in Apoe−/− mice. Arteriosclerosis, Thrombosis, and Vascular Biology. 2018;38(10):2318-2326
  108. 108. Boini KM, Hussain T, Li P-L, Koka SS. Trimethylamine-N-oxide instigates NLRP3 inflammasome activation and endothelial dysfunction. Cellular Physiology and Biochemistry. 2017;44(1):152-162
  109. 109. Ding L et al. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids in Health and Disease. 2018;17(1):1-8
  110. 110. Wang B, Qiu J, Lian J, Yang X, Zhou J. Gut Metabolite Trimethylamine-N-Oxide in Atherosclerosis: From Mechanism to Therapy. Frontiers in Cardiovascular Medicine. 23 Nov 2021;8:723886. DOI: 10.3389/fcvm.2021.723886
  111. 111. Chou R-H et al. Trimethylamine N-oxide, circulating endothelial progenitor cells, and endothelial function in patients with stable angina. Scientific Reports. 2019;9(1):1-10
  112. 112. Jaworska K et al. TMA, a forgotten uremic toxin, but not TMAO, is involved in cardiovascular pathology. Toxins (Basel). 2019;11(9):490
  113. 113. Kelly TN et al. Gut microbiome associates with lifetime cardiovascular disease risk profile among Bogalusa heart study participants. Circulation Research. 2016;119(8):956-964
  114. 114. Gregory JC et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. The Journal of Biological Chemistry. 2015;290(9):5647-5660
  115. 115. Annunziata G et al. Effects of grape pomace polyphenolic extract (Taurisolo®) in reducing TMAO serum levels in humans: Preliminary results from a randomized, placebo-controlled, cross-over study. Nutrients. 2019;11(1):139
  116. 116. Chen M et al. Atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. MBio. 2016;7:E02210-E02215
  117. 117. Wu W-K, Panyod S, Ho C-T, Kuo C-H, Wu M-S, Sheen L-Y. Dietary allicin reduces transformation of L-carnitine to TMAO through impact on gut microbiota. Journal of Functional Foods. 2015;15:408-417
  118. 118. Yang T, Qu H, Song X, Liu Q, Yang X, Xu J, et al. Luhong granules prevent ventricular remodelling after myocardial infarction by reducing the metabolites TMAO and LPS of the intestinal flora. Evidence-Based Complementary and Alternative Medicine. 16 Nov 2019;2019:8937427. DOI: 10.1155/2019/8937427. PMID: 31827566; PMCID: PMC6885292
  119. 119. Roberts AB et al. Development of a gut microbe–targeted nonlethal therapeutic to inhibit thrombosis potential. Nature Medicine. 2018;24(9):1407-1417
  120. 120. Qiu L, Tao X, Xiong H, Yu J, Wei H. Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice. Food & Function. 2018;9(8):4299-4309
  121. 121. Wu T et al. Capsanthin extract prevents obesity, reduces serum TMAO levels and modulates the gut microbiota composition in high-fat-diet induced obese C57BL/6J mice. Food Research International. 2020;128:108774
  122. 122. Montrucchio C et al. Serum trimethylamine-N-oxide concentrations in people living with HIV and the effect of probiotic supplementation. International Journal of Antimicrobial Agents. 2020;55(4):105908
  123. 123. Borges NA et al. Effects of probiotic supplementation on trimethylamine-N-oxide plasma levels in hemodialysis patients: A pilot study. Probiotics Antimicrobial Proteins. 2019;11(2):648-654
  124. 124. Tripolt NJ, Leber B, Triebl A, Köfeler H, Stadlbauer V, Sourij H. Effect of lactobacillus casei Shirota supplementation on trimethylamine-N-oxide levels in patients with metabolic syndrome: An open-label, randomized study. Atherosclerosis. 2015;242(1):141-144
  125. 125. Boutagy NE et al. Probiotic supplementation and trimethylamine-N-oxide production following a high-fat diet. Obesity. 2015;23(12):2357-2363
  126. 126. Qiu L, Yang D, Tao X, Yu J, Xiong H, Wei H. Enterobacter aerogenes ZDY01 attenuates choline-induced trimethylamine N-oxide levels by remodeling gut microbiota in mice. Journal of Microbiology and Biotechnology. 2017;27(8):1491-1499
  127. 127. Takis PG, Oraiopoulou M-E, Konidaris C, Troganis AN. 1H-NMR based metabolomics study for the detection of the human urine metabolic profile effects of Origanum dictamnus tea ingestion. Food & Function. 2016;7(9):4104-4115
  128. 128. Dall’Acqua S et al. New findings on the in vivo antioxidant activity of Curcuma longa extract by an integrated 1H NMR and HPLC–MS metabolomic approach. Fitoterapia. 2016;109:125-131

Written By

Shubham Barik, Aritra Mukherjee, Angelina Job Kolady, Boda Karunakar and Tony Grace

Submitted: 18 June 2022 Reviewed: 09 September 2022 Published: 14 October 2022