Skip to main content

Trimethylamine N-oxide: a meta-organismal axis linking the gut and fibrosis

Abstract

Background

Tissue fibrosis is a common pathway to failure in many organ systems and is the cellular and molecular driver of myriad chronic diseases that are incompletely understood and lack effective treatment. Recent studies suggest that gut microbe-dependent metabolites might be involved in the initiation and progression of fibrosis in multiple organ systems.

Main body of the manuscript

In a meta-organismal pathway that begins in the gut, gut microbiota convert dietary precursors such as choline, phosphatidylcholine, and L-carnitine into trimethylamine (TMA), which is absorbed and subsequently converted to trimethylamine N-oxide (TMAO) via the host enzyme flavin-containing monooxygenase 3 (FMO3) in the liver. Chronic exposure to elevated TMAO appears to be associated with vascular injury and enhanced fibrosis propensity in diverse conditions, including chronic kidney disease, heart failure, metabolic dysfunction-associated steatotic liver disease, and systemic sclerosis.

Conclusion

Despite the high prevalence of fibrosis, little is known to date about the role of gut dysbiosis and of microbe-dependent metabolites in its pathogenesis. This review summarizes recent important advances in the understanding of the complex metabolism and functional role of TMAO in pathologic fibrosis and highlights unanswered questions.

Background

Fibrosis is a pathologic process characterized by excessive deposition of extracellular matrix (ECM; all abbreviations are shown in Table 1) in response to injury, which leads to organ dysfunction and failure. However, formation of fibrotic tissue is also fundamental for tissue response to injury. After injury, fibroblasts become activated, increase their contractility, secrete inflammatory mediators, and synthesize ECM such as collagen or fibronectin, which then results in normal wound healing and repair. When there is severe injury, chronic inflammation, or dysregulation of the wound healing response, ECM deposition becomes excessive, leading to abnormal tissue architecture and organ dysfunction or failure. Fibrosis can affect any organ system and is ultimately responsible for up to 45% of all deaths in industrialized nations (Henderson et al. 2020).

Table 1 Abbreviations are presented in alphabetical order

The fibrogenic response involves many cell types and molecular pathways. Myofibroblasts and fibroblasts, responsible for homeostasis of the ECM (LeBleu et al. 2013; Caam et al. 2018), are considered key effectors in fibrosis (LeBleu et al. 2013; Hinz et al. 2012). Myofibroblasts possess microfilaments that consist of alpha-smooth muscle actin, which allows them to contract (Caam et al. 2018) and transmit contractile forces to the surrounding ECM through specialized focal adhesions containing transmembrane integrins (Duscher et al. 2014). Mechanical forces promote a pro-fibrotic environment via fibroblast secretion of inflammatory mediators and recruitment of inflammatory cells. (Wong et al. 2011) Pathological myofibroblasts originate from a variety of lineages local resident fibroblasts, including bone marrow-derived inflammatory cells, circulating and/or resident mesenchymal stromal stem cells, preadipocytes, vascular mural cells (pericytes) and endothelial or epithelial cells (through endothelial or epithelial-mesenchymal transition). (LeBleu et al. 2013; Rosenbloom et al. 2017)

There is accumulating evidence that microbiota—including those resident in the oral cavity, (Bai et al. 2022) lung, (O’Dwyer et al. 2019) and gut (Xu et al. 2023)—exert remote effects on the molecular pathways governing tissue fibrosis throughout the body. However, the exact mechanisms of these interactions are not yet known. Gut microbiota produce trimethylamine (TMA), which is subsequently converted in the host liver and other tissues to trimethylamine N-oxide (TMAO), a multifunctional molecule that has profibrotic activity. This review summarizes the current state of knowledge of the role and mechanism of the meta-organismal microbe-TMA-TMAO axis in fibrosis and highlights potential therapeutic targets in the prevention or control of pathological fibrosis.

TMAO metabolism

Animal-based diets are abundant in nutrients such as choline, phosphatidylcholine, betaine, and L-carnitine, which undergo conversion by gut microbes into TMA. (Tang et al. 2015a, b) In mammals, the gut bacterial genes encoding enzymes that catalyze TMA production include choline-TMA lyase (CutC), carnitine monooxygenase (CntA/B), and glycine betaine reductase (GrdH) (Rath et al. 2019) (Fig. 1). The TMAO reductase pathway, which is responsible for the retroconversion of TMAO to TMA, is catalyzed by TorA (Mejean et al. 1994) (Fig. 1). The TMAO reductase pathway is the most prevalent in the human gut, with Proteobacteria (mainly Klebsiella and Escherichia) contributing most of the TMAO reductase sequences, (Jameson et al. 2016) while CutC and GrdH are associated with Firmicutes. In a metagenomic analysis of 50 human fecal samples, CutC amplicons were found in all individuals but only 26% had CntA. (Rath et al. 2019) Germ-free and antibiotic-treated animal models have been shown to lack the capacity for TMAO production, thus providing evidence for the role of the microbiome in the metabolism of carnitine to TMA and TMAO. (Tang and Hazen 2017; Yap et al. 2008)

Fig. 1
figure 1

Pathways of trimethylamine (TMA) formation by gut microbiota and cycling of TMA to trimethylamine N-oxide (TMAO). GrdH, glycine betaine reductase. CntA/B, carnitine monooxygenase. CutC, choline-TMA lyase. FMO, flavin-containing monooxygenase. TorA, trimethylamine N-oxide reductase

Once TMA enters the host portal circulation, it is converted to TMAO, with flavin-containing monooxygenase 3 (FMO3) responsible for metabolizing the rate-limiting step. (Krueger and Williams 2005) FMO3 is predominantly expressed in the liver. However, it is also expressed in non-hepatic tissues including the lung, kidney, and brain, and we recently found, the skin. (Dolphin et al. 1996; Zhang and Cashman 2006) Systemic levels of TMA are low. (Al-Waiz et al. 1987) In humans, the capacity for TMA and TMAO production is increased in carnivores and omnivores compared to vegetarians, which is attributable to reduced or absent levels of CntA/B and CutC. (Rath et al. 2019) Approximately 50% of consumed TMAO passes through the body and is excreted in the urine. TMAO reductase can convert the remaining dietary TMAO into TMA. (Kwan and Barrett 1983) TMAO can also be consumed from dietary sources and be retroconverted to TMA. (Loo et al. 2022) Thus, systemic levels of TMAO are governed by several factors, including the diet, (Boutagy et al. 2015; Wang et al. 2019) composition and function of gut microbiota, (Rath et al. 2017) medication use, (Milks et al. 2018) FMO3 expression and activity, (Bennett et al. 2013) and renal excretion. (Janeiro et al. 2018) Due to redundant and overlapping regulatory pathways and exposure variables, plasma TMAO has high inter- and intra-individual variability. (Papandreou et al. 2020)

Fibrotic conditions and TMAO

A wide spectrum of human diseases is characterized by chronic inflammation leading to organ or tissue fibrosis. (Rosenbloom et al. 2017) As summarized below, TMAO has been implicated in kidney, heart, liver, and skin fibrosis. Organ-specific pathways for the pro-fibrotic effects of TMAO are shown in Fig. 2.

Fig. 2
figure 2

Overview of proposed mechanisms of TMAO-mediated fibrosis in the kidney, heart, liver, and systemic sclerosis. Details of mechanisms are provided in the text

Kidney

High levels of circulating TMAO are associated with concurrent adverse cardiovascular disease events including heart attack, stroke, and mortality in people with chronic kidney disease (CKD). (Kim et al. 2016; Missailidis et al. 2016; Tang et al. 2015a, b) In one prospective cohort study of 521 patients with CKD, TMAO was associated with a 2.8-fold dose-dependent increase in 5-year all-cause mortality. (Tang et al. 2015a, b) TMAO is also associated with the development of CKD. In a prospective cohort study of 1,434 people with normal renal function, patients who later developed CKD were found to have a 33% higher level of plasma TMAO compared to those who did not. (Rhee et al. 2013) In mouse and rat studies, dietary supplementation with choline or TMAO led to elevated levels of plasma TMAO, which is associated with both spontaneous (Tang et al. 2015a, b) and induced (Fang et al. 2021) kidney fibrosis and renal functional impairment.

Numerous animal studies have investigated inhibition of gut microbial TMA generation as an intervention to preserve renal function. In mice, oral administration of iodomethylcholine (IMC), a novel choline TMA-lyase inhibitor, markedly suppressed plasma TMAO levels, significantly reduced the decline in renal function, and reduced renal fibrosis in an isoproterenol model of CKD. (Gupta et al. 2020) In a separate study, investigators used methylcholine, an inhibitor of TMA lyase that functions similarly to IMC, and found attenuation of adenine-induced markers of CKD, including renal cortical scarring and collagen deposition following methylcholine treatment. Additionally, methylcholine supplementation significantly decreased expression of renal inflammatory and pro-fibrotic genes induced by adenine supplementation. (Zhang et al. 2021) Other studies have investigated inhibition of TMA production using 3,3-dimethyl-1-butanol (DMB), another TMA lyase inhibitor. (Sun et al. 2017; Zou et al. 2021) Oral DMB reduced plasma TMAO levels and prevented both renal and cardiac injury in rats with cardiorenal syndrome following induction of myocardial infarction and kidney damage. TMAO-associated increases in expression of pro-fibrotic markers TGF-ß1 and collagen I and pro-inflammatory cytokines interleukin (IL)-6 and IL-8 were attenuated by DMB. (Zou et al. 2021)

There are multiple potential mechanisms for the profibrotic role of TMAO in the kidney. SMAD3 is a member of the SMAD family of transcriptional regulators downstream of TGF-ß signaling, (Zimmerman and Padgett 2000; Kretzschmar and Massague 1998) a pathway that regulates proliferation, differentiation, and apoptosis, and tissue fibrosis. (Massague 2012; Bottinger 2007) SMAD3 phosphorylation is an important regulatory step in renal fibrosis in CKD. (Qu et al. 2014) In mice fed diets supplemented with choline and TMAO, there was increased phosphorylation of SMAD3 in the kidney. (Tang et al. 2015a, b) Mice with high fat diet-induced obesity had elevated plasma TMAO levels, increased renal fibrosis, and increased SMAD3 phosphorylation, all of which were reversed with DMB treatment. (Sun et al. 2017)

The nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome is another pathway that may mediate TMAO-induced renal fibrosis. In rat models of diabetic kidney disease, dietary supplementation with TMAO and choline were found to increase activation of NLRP3 inflammasome, leading to increased IL-1β and IL-18 secretion, renal inflammation, oxidative stress, and reactive oxygen species (ROS) formation. (Fang et al. 2021) In human renal fibroblasts, TMAO stimulation led to increased cell proliferation and NLRP-3 and caspase-1 protein expression in these cells. Knocking out NLRP-3 and caspase-1 prevented TMAO-dependent cell proliferation. Furthermore, inhibitors of protein kinase R-like endoplasmic reticulum kinase (PERK), protein kinase B (Akt), and mammalian target of rapamycin (mTOR) each attenuated renal fibroblast proliferation and collagen production, suggesting the mechanistic role for each in mediating the effect of TMAO on renal fibroblast proliferation. (Kapetanaki et al. 2021) Finally, TMAO enhanced TNF-α mediated renal inflammation by inducing the release of cytokines, chemokines, inflammatory mediators, and growth mediators from renal fibroblasts. (Stefania et al. 2024) Based on these findings and prior studies that have demonstrated the association between NLRP3 inflammasome, Akt, mTOR, and TMAO, (Artlett 2012; Artlett and Thacker 2015; Boini et al. 2017; Zhao et al. 2020; Gallego et al. 2020; Li et al. 2018a, b) the mechanisms underlying the effect of TMAO on renal fibrosis include TMAO binding to and activating PERK, which triggers the Akt/mTOR pathway, which, in turn, promotes cell proliferation and collagen production or regulates NLRP3 and caspase-1 to indirectly promote cell proliferation. Activated PERK may also directly regulate NLRP3 through NF-κB activation. (Kapetanaki et al. 2021)

Finally, progression of CKD and augmentation of kidney fibrosis by TMAO may involve two additional mechanisms: (1) upregulation of SMAD3/transforming growth factor-beta (TGF-ß) signaling and (2) generation of reactive oxygen species (ROS). The relationship between TGF-ß signaling and ROS has been well-documented in cancer biology, with studies suggesting that ROS activates latent TGF-ß to its active form. (Chung et al. 2021) Whether similar links are implicated in renal fibrosis is an area of intriguing future research.

Heart

Myocardial fibrosis, or the deposition of excess ECM in the cardiac interstitium, can be both reparative and pathologic. For example, after myocardial infarction, reparative fibrosis preserves the structural integrity of the heart. (Frangogiannis and Kovacic 2020) However, interstitial fibrosis secondary to systemic hypertension can lead to increased myocardial stiffness and diastolic dysfunction. (Diez et al. 2020) Gut microbial metabolites, including TMAO, (Guo et al. 2020; Troseid et al. 2015; Krack et al. 2005; Anker et al. 1997; Dong et al. 2021) and their precursors such as betaine and choline (Papandreou et al. 2021) have been associated with increased cardiovascular disease including heart failure (Guo et al. 2020; Troseid et al. 2015; Krack et al. 2005; Anker et al. 1997; Dong et al. 2021) and atrial fibrillation. (Papandreou et al. 2021)

Elevated systemic TMAO is associated with increased risk of major adverse cardiovascular events and mortality. (Heianza et al. 2017; Tang et al. 2013) This association persists in patients with heart failure. One meta-analysis involving nearly 7,000 patients with heart failure reported an increase in all-cause death, recurrent myocardial infarctions, and rehospitalizations with increasing systemic TMAO independent of renal function, (Li et al. 2020) and this has been corroborated in multiple other large studies. (Stubbs et al. 2019; Zhou et al. 2020; Li et al. 2022)

Numerous prospective cohort studies have also found elevated plasma TMAO levels to be a predictor of cardiovascular death in patients with acute (Schuett et al. 2017; Israr et al. 2021; Kinugasa et al. 2021; Suzuki et al. 2016) and chronic heart failure, (Zhou et al. 2020; Wei et al. 2022) peripheral artery disease, (Roncal et al. 2019) and patients with end-stage renal disease on hemodialysis. (Zhang et al. 2020) Of these studies, one prospective cohort study involving 2,490 patients with heart failure with reduced ejection fraction (HFrEF) or heart failure with preserved ejection fraction (HFpEF) found plasma TMAO levels to have a better predictive value than N-terminal pro-brain natriuretic peptide (NT-proBNP), an established risk marker in heart failure without direct connections to TMAO, for mortality and cardiovascular mortality in HFrEF patients. (Schuett et al. 2017) Another prospective cohort study with 196 participants found plasma TMAO levels to be helpful for risk stratification of patients with HFpEF, especially when used in conjunction with NT-proBNP. (Salzano et al. 2020) A study of patients with coronary artery disease found an association between plasma TMAO levels, NT-proBNP, and prognosis, risk stratification, and length of hospitalization. (Qiu et al. 2022) Of note, a prospective cohort study involving Polish patients with cardiovascular disease found a statistically significant but clinically non-significant association between circulating TMAO and 5-year mortality. Furthermore, a recent large study of patients with type 2 diabetes mellitus found no significant associations between TMAO, choline, or TMA with heart failure requiring hospitalization, cardiovascular death, or all-cause death. (Wargny et al. 2022) Thus, while most studies demonstrate the clinical utility of using TMAO to establish cardiovascular disease prognosis and outcome, these latter studies suggest the need for further research to elucidate the precise role of TMAO on the mortality of patients with cardiovascular disease. (Konieczny et al. 2022)

An association between cardiovascular complications, including cardiac fibrosis, and elevated TMAO in animal models has been demonstrated by multiple investigators. (Zou et al. 2021; Li et al. 2017, 2018a, b, 2019; Organ et al. 2016; Zhang et al. 2017; Yang et al. 2019a, b; Wang et al. 2020; Strilakou et al. 2016; Shuai et al. 2020; Nanto-Hara et al. 2020; Chen et al. 2017) One study in mice found that transverse aortic constriction-induced heart failure was significantly worse in mice fed diets supplemented with choline or TMAO. (Organ et al. 2016) In other studies, mice on diets supplemented with TMAO or choline exhibited significantly more surgical and doxorubicin-induced cardiac fibrosis than those on the control diet, (Yang et al. 2019a, b; Li et al. 2019) and inhibition of TMA lyase using DMB prevented choline-related cardiac fibrosis. (Yang et al. 2019a, b) High choline diets also exacerbated myocardial fibrosis and cardiac dysfunction in a mouse model of heart failure with HFpEF. (Shuai et al. 2020) Finally, in a mouse study on the cardioprotective effects of voluntary exercise on myocardial inflammation and fibrosis, the benefits of exercise were abrogated by TMAO supplementation. (Zhang et al. 2017)

Targeting circulating TMAO levels to decrease cardiac fibrosis is an active area of investigation with mixed results to date. Treatment with DMB prevented susceptibility to ventricular arrhythmia and adverse cardiac structural remodeling in mice with overload-induced heart failure from aortic banding surgery (Wang et al. 2020) and ameliorated cardiac diastolic dysfunction, myocardial fibrosis and inflammation in a mouse model of uninephrectomy and aldosterone-induced HFpEF, (Wang et al. 2020; Shuai et al. 2020) but these mice were not given a high choline or TMAO diet. On the other hand, in mice with Western diet-induced obesity, DMB reduced plasma TMAO levels but did not alter other parameters such as cardiac inflammation, fibrosis, and dysfunction. (Chen et al. 2017) One study explored the prospect of altering microbe-produced TMAO using a guanylate cyclase C (GC-C) agonist, linaclotide, and found that linaclotide reduced TMAO and ameliorated both renal and cardiac fibrosis in a mouse model of adenine-induced CKD. (Nanto-Hara et al. 2020) As receptor guanylyl cyclase C (GC-C) signaling in the gut epithelium has been implicated as an important factor in host defense against pathogenic bacteria, (Amarachintha et al. 2018) the study proposed that the increase in GC-C signaling from linaclotide reduced the bacterial species that generate TMA. In rats subjected to myocardial infarction, Luhong granules—a multi-medicinal herb combination—reduced both TMAO and lipopolysaccharide levels through gut microbial modification and reduced intestinal pathology, which ultimately decreased adverse ventricular remodeling. (Yang et al. 2019a, b)

While numerous studies have supported the positive association between cardiovascular disease or cardiac fibrosis with elevated TMAO levels, there are other studies, both in preclinical models and in human populations, that have not found such as association. In a study using pressure-overloaded hearts in hypertensive rats, it was shown that chronic treatment with low dose TMAO reduced cardiac fibrosis. (Huc et al. 2018) Another study found that a choline-deficient diet promoted fibrosis in rats (Strilakou et al. 2016). In a rat model of spontaneously hypertensive heart failure, higher TMAO levels reduced mortality and were associated with diuretic, natriuretic, and hypotensive effects. (Gawrys-Kopczynska et al. 2020) In fact, some studies have found that low levels of TMAO may have positive effects on the heart. (Huc et al. 2018; Gawrys-Kopczynska et al. 2020; Strilakou et al. 2013) For example, one study that explored the effects of dietary TMAO supplementation in spontaneously hypertensive rats found that a 4-5-fold increase in plasma TMAO levels was associated with reduced plasma levels of NT-proBNP and vasopressin and lower left ventricular end-diastolic pressure and cardiac fibrosis—all indications of improved diastolic dysfunction. (Huc et al. 2018) Collectively, these results suggest a context-dependent causative role of TMAO in cardiac fibrosis.

The known profibrotic mechanisms of TMAO in the myocardium appear to be similar to the mechanisms in the kidney and involve TGF-β and or the NLRP3 inflammasome. TMAO treatment of primary mouse cardiac fibroblasts was found to induce a dose-dependent increase in proliferation, migration, collagen secretion, and expression of profibrotic factors, TGF-β and phosphorylated SMAD3. (Li et al. 2019) Mouse cardiac fibroblasts treated with TMAO also exhibited increased NLRP3 inflammasome activation, while silencing RNA (siRNA)-mediated knockdown of NLRP3 in cardiac fibroblasts blunted TMAO-induced cell proliferation as well as TGF-β and collagen expression. (Li et al. 2019) TMAO has also been shown to transform atrial fibroblasts into myofibroblasts through the activation of the Wnt2a/β-catenin signaling pathway. (Yang et al. 2019a, b)

Systemic sclerosis

Systemic sclerosis (SSc) is a complex chronic autoimmune disease characterized by inflammation, vascular injury and fibrosis that synchronously affect virtually every organ system. (Allanore et al. 2015; Denton and Khanna 2017) While the precise etiology of SSc remains obscure, both genetic and environmental factors contribute. (Allanore et al. 2015) Several lines of evidence implicate gut dysbiosis in the pathogenesis of fibrosis characteristic of SSc. (Volkmann 2017; Patrone et al. 2017; Johnson et al. 2019) Multiple bacterial taxa that are enriched in the SSc gut microbiome (e.g. Ruminococcus) (Volkmann 2017) are known to be high TMA producers. (Wu et al. 2018) Expression of FMO3 is significantly up-regulated in both explanted skin fibroblasts, and in skin biopsies, from patients with SSc (Chadli et al. 2019; Kim et al. 2022; Skaug et al. 2020) Moreover, in a recent study, SSc patients with interstitial lung disease (ILD) and esophageal dysmotility had higher plasma TMAO levels than non-SSc controls. (Stec et al. 2023)

The endoplasmic reticulum stress kinase PERK (protein kinase R [PRK]-like endoplasmic reticulum kinase, EIF2AK3) has been identified as a potential receptor for TMAO. (Chen et al. 2019) Microarray analysis on rat hepatocytes treated in the presence or absence of pathophysiologic levels of TMAO showed that TMAO selectively induced the PERK pathway of the unfolded protein response (UPR) without upregulation of the inositol-requiring enzyme type 1 (IRE1) or activating transcription factor 6 (ATF6) pathways. In vivo studies in mice demonstrated that dietary supplementation of TMAO increased phosphorylation of PERK and increased expression of the transcription factor Foxo1, a key driver of metabolic syndrome, readouts which were blocked by co-administration of the PERK inhibitor GSK2656157 or liver-specific knock-out of PERK. Furthermore, direct interaction between TMAO and PERK was confirmed by isotope labeled TMAO binding to immunoprecipitated PERK. The study further identified the luminal domain of PERK as a specific domain involved in the TMAO-PERK interaction. (Chen et al. 2019) Other investigators found that TMAO leads to a dose-dependent increase in PERK activation in human fibroblasts in vitro and increased PERK phosphorylation and Foxo1 expression in human microvascular endothelial cells. The profibrotic effects of TMAO were blocked by PERK inhibition using the PERK inhibitor GSK2606414 and by siRNA silencing. (Kim et al. 2022) However, PERK binding and activation by TMAO may be cell-specific, as PERK activation was not detected in TMAO-treated kidney epithelial cells, (Zhang et al. 2021) suggesting that further investigation of the cell-specific mechanisms of TMAO activity is warranted.

When exposed to TMAO in vitro, skin fibroblasts, vascular endothelial cells, and adipocytic progenitor cells are reprogrammed into myofibroblasts, a transition mediated via PERK. (Kim et al. 2022) Another study showed that TMAO generated from the aorta and liver phosphorylates 12 cytosolic kinases via PERK, leading to endoplasmic reticulum stress, mitochondrial stress and metabolic reprogramming to establish trained immunity in aortic endothelial cells. (Saaoud et al. 2023) These studies suggest that exposure of fibroblasts, endothelial cells and other mesenchymal cells to high levels of TMAO may be sufficient to elicit a fibrotic response, including endothelial-mesenchymal transition. However, more studies are warranted in order to fully establish the pathogenic role, and mechanism of action, of TMAO in the vascular and fibrotic pathology characteristic of SSc.

Liver

Metabolic dysfunction-associated steatotic liver disease (MASLD), the pathologic accumulation of hepatocellular lipid, is a steadily growing global health issue that parallels the rise in patients with obesity and metabolic syndrome. (Wong et al. 2023) A subset of patients with MASLD develop metabolic dysfunction-associated steatohepatitis (MASH), characterized by varying degrees of inflammation and fibrosis in the liver. MASH can further predispose a patient to cirrhosis or hepatocellular carcinoma. (Teng et al. 2023; Huang et al. 2022)

MASLD pathogenesis is multifactorial (i.e., “multi-hit hypothesis”) and includes genetic and epigenetic factors, hormonal factors, involvement of adipose tissue, nutritional factors, and dysbiosis. (Teng et al. 2023; Huang et al. 2022; Buzzetti et al. 2016) Choline and choline/methionine-deficient diets are established means of inducing MASLD in animal models, (Buzzetti et al. 2016) with proposed mechanisms involving (1) endoplasmic reticulum stress, (2) triglyceride aggregation and deposition, (3) NLRP3 inflammasome activation, and (4) gut dysbiosis. (Vallianou et al. 2024) While MASLD induction by a choline-deficient diet in animal models might imply that low TMAO induces MASLD, studies across humans and cellular and animal models suggest a more complex relationship.

Numerous case control and cohort studies have shown a positive association between high TMAO levels and MASLD (Chen et al. 2016a, b; Shi et al. 2022; Zhao et al. 2019) and primary liver cancer. (Liu et al. 2018) In a case-control and cross-sectional analysis of 100 patients with or without MASLD, high circulating level of TMAO was found to be associated with greater severity of MASLD. (Chen et al. 2016a, b) In a prospective study of over 100 women with or without morbid obesity and with or without MASLD, circulating TMAO level was found to be significantly elevated in patients with MASLD. (Aragones et al. 2020) More nuanced findings have been observed in patients with more advanced liver pathologies, including cirrhosis. In a study of approximately 120 patients with cirrhosis and 5,000 healthy people in the PREVEND cohort study, severity of cirrhosis was found to be associated with high levels of betaine, a precursor of TMAO. Interestingly, in the patients with cirrhosis, TMAO levels increased after liver transplantation and were higher than pre-transplantation levels. (Berg et al. 2023) In a separate study of patients with cirrhosis, elevated circulating TMAO was a marker for hepatic encephalopathy. (Jimenez et al. 2010) Finally, in a case-control study of patients with MASLD and healthy controls, serum TMAO levels correlated with serum total bile acids and hepatic mRNA expression of cholesterol 7 alpha hydroxylase (CYP7A1). (Tan et al. 2019) The same study reported that mice fed a high-fat diet and treated with TMAO demonstrated impaired liver function, hepatic triglyceride accumulation and lipogenesis, and altered hepatic bile acid composition toward farnesoid X receptor-antagonistic bile acid species, while knockdown of CYP7A1 in hepatocytes blocked the effects of TMAO-induced lipogenesis, suggesting that TMAO aggravates liver steatosis by suppressing bile acid-mediated hepatic farnesoid X receptor signaling. (Tan et al. 2019)

The mechanisms by which TMAO causes MASLD in humans are varied. TMAO is related to risk factors such as diabetes mellitus and obesity. (Dehghan et al. 2020; Zhuang et al. 2019) In addition, in vitro, high fat and TMA exposure stimulates FMO gene expression and TMAO. (Shi et al. 2022) Furthermore, TMAO reduces bile acid production by suppressing CYP7A1 and CYP27A116 and restricts bile acid enterohepatic circulation by repressing the organic anion transporter and the expression of the multidrug resistance protein family. (Koeth et al. 2013; Makishima et al. 1999; Ding et al. 2018)

Animal studies suggest that the relationship between TMAO and liver fibrosis is likely complex and non-linear. Rats exposed to chronically high doses TMAO were found to have increased oxidative stress and inflammation in the liver, but there were no structural changes or evidence of increased fibrosis. (Florea et al. 2024) However, in mouse models of MASH and acute liver fibrosis, dietary TMAO supplementation was associated with decreased liver fibrosis, improved liver function, and greater gut microbiome diversity. (Zhou et al. 2022) Furthermore, ATP1B1, an astrocyte-specific isoform of the Na+/+-ATPase transmembrane pump, is upregulated in the liver endothelial cells of mice with liver fibrosis. Endothelial-specific knockout of ATP1B1 resulted in decreased expression of profibrotic factors such as C-X-C motif chemokine ligands 10 and 1 and connective tissue growth factor, while administration of a selective ATP1B1 inhibitor decreased liver fibrosis. In fibrotic mouse livers, treatment with TMAO reduced ATP1B1 protein expression. (Zhou et al. 2022) These findings suggest that TMAO may mitigate liver fibrosis through attenuation of ATP1B1 expression and consequent improvement of endothelial integrity, a potential mechanism that highlights the organ-specific relationship between TMAO and fibrosis. (Zhou et al. 2022)

In vitro data suggest alternative mechanisms for TMAO and liver fibrosis. TMAO induces release of hepatocyte-derived exosomes from mouse-derived hepatocytes, leading to endothelial uptake and downstream inflammation, apoptosis, and endothelial dysfunction via NF-κB pathway activation. (Liu et al. 2022) In a fatty liver cell model, TMAO induced gene expression of microRNAs miRNA-34a and miRNA-122, both of which are biomarkers of MASLD in humans. (Bahramirad et al. 2024) In a separate study, incubation of cells in the fatty liver cell model with TMAO led to increased lipid deposition and elevation in expression of liver fibrosis-related genes including keratin17. (Nian et al. 2023) Knockdown of keratin17 with siRNA resulted in attenuation of the observed lipid deposition and liver fibrosis. (Nian et al. 2023)

These data demonstrate a complex role for TMAO in both the development and mitigation of liver fibrosis. The variable effects of TMAO may be due cell type- and stage-specific. Further studies are needed to uncover the precise pathophysiologic relationship between TMAO and liver fibrotic diseases.

Therapeutic strategies

Inherent to the evidence supporting a direct causal link between TMAO and fibrosis in multiple organ systems is a presumption that modulation of TMAO could be a therapeutic strategy for preventing and treating fibrosis. As shown in Fig. 3, this could be achieved in several ways: dietary modification, modulation of the gut microbiome, or targeted approaches to selectively reduce TMA levels by reducing the conversion of dietary precursors into TMA.

Fig. 3
figure 3

Therapeutic strategies involving TMAO include dietary interventions (e.g., reducing choline, L-carnitine, or betaine-containing compounds, dietary supplements, lifestyle modification), probiotics that alter TMA and TMAO levels by changing the gut microbial community, 3,3-dimethyl-1-butanol (DMB) or other CutC/D inhibitors that suppress microbial production of TMA, and inhibitors of FMO3

Dietary interventions can be effective in lowering TMAO levels in humans. As an example, avoidance of red meat consumption was shown to reduce plasma TMAO levels within 4 weeks. (Wang et al. 2019) A red meat-rich diet increased TMAO levels through an increased supply of the TMA precursor carnitine, which elicited changes in the functional output of TMA by the gut microbial community and also lowered the renal excretion of TMAO. Reduced excretion of TMAO was reversed by a white meat and a non-meat diet. (Wang et al. 2019)

Another approach to reducing TMAO levels is to modulate the composition of the gut microbiota. Broad-spectrum antibiotics suppress production of TMA and TMAO, which, however, recover after the withdrawal of antibiotics. This strategy for lowering systemic TMAO levels has limited clinical potential since antibiotics broadly disrupt gut microbial ecology (Jernberg et al. 2007) and can lead to off-target effects on a myriad of host physiologic processes (Langdon et al. 2016) and antibiotic resistance. Probiotics (live microorganisms) or prebiotics (non-microbial substances that alter microbial community structure) also represent a potential strategy by altering the composition and the gut microbial community structure. Rats fed a high-fat diet and the probiotic Enterococcus faecium WEFA23 had reduced production of TMAO, (Huang et al. 2018) but the propensity for tissue fibrosis was not examined. Resveratrol, a natural polyphenol, was shown to decrease TMA production via gut microbiota remodeling in mice. On the other hand, treatment with resveratrol increased hepatic expression and activity of FMO3 and increased plasma TMAO levels following a single dose of TMA. (Chen et al. 2016a, b) Linaclotide, a GC-C agonist approved by the U.S. Food and Drug Administration for the treatment of constipation in irritable bowel syndrome, decreased the plasma levels of TMAO by modifying gut microbiota and reduced renal and cardiac fibrosis in choline-fed mice. (Nanto-Hara et al. 2020) Finally, transplantation of gut microbiota to regulate the proportions of specific high TMA-producing taxa is effective at reducing TMAO in mice. (Gregory et al. 2015)

Interfering with the ability of the host to convert microbial-derived TMA into TMAO would also lead to a decrease in circulating TMAO levels. Indeed, suppression of FMO3 has been shown to lower circulating TMAO levels and reduce diet-enhanced atherosclerosis in animal models. (Zhang et al. 2017; Miao et al. 2015) However, suppression of FMO3 has likely little clinical potential as inhibition of this enzyme has been shown to cause hepatic inflammation. (Warrier et al. 2015) Humans with inherited deficiency in FMO3 experience trimethylaminuria that is caused by the accumulation of TMA, resulting in a strong body odor. (Cashman et al. 2003)

Targeting TMA lyase activity in gut microbiota is another exciting strategy that appears to have potential as a clinical tool. Unlike antibiotics, small molecule inhibitors of TMA lyase like DMB and IMC are non-lethal to microbiota and likely exert less selective pressure for the development of resistance. DMB, a structural analogue of choline, was shown to inhibit microbial TMA lyases and reduce the level of TMAO in mice fed a high choline or carnitine diet. Treatment with DMB promoted reductions in the proportions of some microbial taxa that are associated with plasma TMA and TMAO levels. (Tang et al. 2015a, b) As discussed in previous sections, IMC has been shown to markedly suppress the production of TMA and lower plasma TMAO levels in animal models, (Gupta et al. 2020; Organ et al. 2016; Yang et al. 2019a, b; Roberts et al. 2018) suggesting that inhibition of TMA lyase activity can have a meaningful impact on host pathology. Taken together, these studies indicate strategies that inhibit TMA lyase activity and therefore TMA generation may serve as a therapeutic approach for the treatment of fibrotic diseases.

Conclusions

Gut microbiota generate a plethora of biologically active metabolites that can exert a powerful direct or indirect influence on many aspects of host homeostasis and pathophysiology. Accumulating evidence support a strong link between elevated circulating TMAO levels and fibrosis in the heart, kidney, liver, and skin in diverse diseases. Thus, the nutrient-microbe-host meta-organismal framework linking diet and microbial metabolism to fibrosis represents a targetable pathway for therapeutics, and it will be interesting to see if selectively inhibiting microbial TMA lyase translates to a decreased propensity of fibrosis. The recent discovery of the first selective inhibitors of microbial TMA production (Tang et al. 2015a, b; Roberts et al. 2018) now opens the door for limiting TMAO exposure by “non-lethal drugging” of the gut microbiome (Brown and Hazen 2017) to reduce TMAO levels and prevent, slow, or reverse fibrosis.

Data availability

Not applicable.

References

  • Al-Waiz M, Mitchell SC, Idle JR, Smith RL. The metabolism of 14 C-labelled trimethylamine and its N-oxide in man. Xenobiotica. 1987;17(5):551–8.

    Article  CAS  PubMed  Google Scholar 

  • Allanore Y, Simms R, Distler O, Trojanowska M, Pope J, Denton CP, et al. Systemic sclerosis. Nat Reviews Disease Primers. 2015;1:15002.

    Article  PubMed  Google Scholar 

  • Amarachintha S, Harmel-Laws E, Steinbrecher KA. Guanylate cyclase C reduces invasion of intestinal epithelial cells by bacterial pathogens. Sci Rep. 2018;8(1):1521.

    Article  PubMed  PubMed Central  Google Scholar 

  • Anker SD, Egerer KR, Volk HD, Kox WJ, Poole-Wilson PA, Coats AJ. Elevated soluble CD14 receptors and altered cytokines in chronic heart failure. Am J Cardiol. 1997;79(10):1426–30.

    Article  CAS  PubMed  Google Scholar 

  • Aragones G, Colom-Pellicer M, Aguilar C, Guiu-Jurado E, Martinez S, Sabench F, et al. Circulating microbiota-derived metabolites: a liquid biopsy? Int J Obes. 2020;44(4):875–85.

    Article  CAS  Google Scholar 

  • Artlett CM. The role of the NLRP3 inflammasome in fibrosis. Open Rheumatol J. 2012;6:80–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Artlett CM, Thacker JD. Molecular activation of the NLRP3 inflammasome in fibrosis: common threads linking divergent fibrogenic diseases. Antioxid Redox Signal. 2015;22(13):1162–75.

    Article  CAS  PubMed  Google Scholar 

  • Bahramirad Z, Moloudi MR, Moradzad M, Abdollahi A, Vahabzadeh Z. Trimethylamine-N-oxide, a new risk factor for non-alcoholic fatty liver Disease Changes the expression of miRNA-34a, and miRNA-122 in the fatty liver cell model. Biochem Genet. 2024.

  • Bai L, Wang YL, Chen YL, Li HX, Zhu SW, Liu Y, et al. The combination of experimental periodontitis and oral microbiota from periodontitis patients aggravates liver fibrosis in mice. J Clin Periodontol. 2022;49(10):1067–78.

    Article  CAS  PubMed  Google Scholar 

  • Bennett BJ, de Aguiar Vallim TQ, Wang Z, Shih DM, Meng Y, Gregory J, et al. Trimethylamine-N-oxide, a metabolite associated with atherosclerosis, exhibits complex genetic and dietary regulation. Cell Metabol. 2013;17(1):49–60.

    Article  CAS  Google Scholar 

  • Boini KM, Hussain T, Li PL, Koka S. Trimethylamine-N-Oxide instigates NLRP3 inflammasome activation and endothelial dysfunction. Cell Physiol Biochemistry: Int J Experimental Cell Physiol Biochem Pharmacol. 2017;44(1):152–62.

    Article  Google Scholar 

  • Bottinger EP. TGF-beta in renal injury and disease. Semin Nephrol. 2007;27(3):309–20.

    Article  CAS  PubMed  Google Scholar 

  • Boutagy NE, Neilson AP, Osterberg KL, Smithson AT, Englund TR, Davy BM, et al. Probiotic supplementation and trimethylamine-N-oxide production following a high-fat diet. Obes (Silver Spring). 2015;23(12):2357–63.

    Article  CAS  Google Scholar 

  • Brown JM, Hazen SL. Targeting of microbe-derived metabolites to improve Human Health: the Next Frontier for Drug Discovery. J Biol Chem. 2017.

  • Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metab Clin Exp. 2016;65(8):1038–48.

    Article  CAS  PubMed  Google Scholar 

  • Cashman JR, Camp K, Fakharzadeh SS, Fennessey PV, Hines RN, Mamer OA, et al. Biochemical and clinical aspects of the human flavin-containing monooxygenase form 3 (FMO3) related to trimethylaminuria. Curr Drug Metab. 2003;4(2):151–70.

    Article  CAS  PubMed  Google Scholar 

  • Chadli L, Sotthewes B, Li K, Andersen SN, Cahir-McFarland E, Cheung M, et al. Identification of regulators of the myofibroblast phenotype of primary dermal fibroblasts from early diffuse systemic sclerosis patients. Sci Rep. 2019;9(1):4521.

    Article  PubMed  PubMed Central  Google Scholar 

  • Chen YM, Liu Y, Zhou RF, Chen XL, Wang C, Tan XY, et al. Associations of gut-flora-dependent metabolite trimethylamine-N-oxide, betaine and choline with non-alcoholic fatty liver disease in adults. Sci Rep. 2016a;6:19076.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chen ML, Yi L, Zhang Y, Zhou X, Ran L, Yang J, et al. Resveratrol attenuates Trimethylamine-N-Oxide (TMAO)-Induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. mBio. 2016b;7(2):e02210–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Chen K, Zheng X, Feng M, Li D, Zhang H. Gut microbiota-dependent metabolite trimethylamine N-Oxide contributes to Cardiac Dysfunction in Western Diet-Induced obese mice. Front Physiol. 2017;8:139.

    Article  PubMed  PubMed Central  Google Scholar 

  • Chen S, Henderson A, Petriello MC, Romano KA, Gearing M, Miao J, et al. Trimethylamine N-Oxide binds and activates PERK to promote metabolic dysfunction. Cell Metabol. 2019;30(6):1141–51. e5.

    Article  CAS  Google Scholar 

  • Chung J, Huda MN, Shin Y, Han S, Akter S, Kang I et al. Correlation between oxidative stress and transforming growth factor-Beta in cancers. Int J Mol Sci. 2021;22(24).

  • Dehghan P, Farhangi MA, Nikniaz L, Nikniaz Z, Asghari-Jafarabadi M. Gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) potentially increases the risk of obesity in adults: an exploratory systematic review and dose-response meta- analysis. Obes Rev. 2020;21(5):e12993.

    Article  CAS  PubMed  Google Scholar 

  • Denton CP, Khanna D. Systemic sclerosis. Lancet. 2017;390(10103):1685–99.

    Article  PubMed  Google Scholar 

  • Diez J, Gonzalez A, Kovacic JC. Myocardial interstitial fibrosis in Nonischemic Heart Disease, Part 3/4: JACC Focus Seminar. J Am Coll Cardiol. 2020;75(17):2204–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Ding L, Chang M, Guo Y, Zhang L, Xue C, Yanagita T, et al. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids Health Dis. 2018;17(1):286.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Dolphin CT, Cullingford TE, Shephard EA, Smith RL, Phillips IR. Differential developmental and tissue-specific regulation of expression of the genes encoding three members of the flavin-containing monooxygenase family of man, FMO1, FMO3 and FM04. Eur J Biochem. 1996;235(3):683–9.

    Article  CAS  PubMed  Google Scholar 

  • Dong Z, Zheng S, Shen Z, Luo Y, Hai X. Trimethylamine N-Oxide is Associated with Heart failure risk in patients with preserved ejection fraction. Lab Med. 2021;52(4):346–51.

    Article  PubMed  Google Scholar 

  • Duscher D, Maan ZN, Wong VW, Rennert RC, Januszyk M, Rodrigues M, et al. Mechanotransduction and fibrosis. J Biomech. 2014;47(9):1997–2005.

    Article  PubMed  PubMed Central  Google Scholar 

  • Fang Q, Zheng B, Liu N, Liu J, Liu W, Huang X, et al. Trimethylamine N-Oxide exacerbates renal inflammation and fibrosis in rats with Diabetic kidney disease. Front Physiol. 2021;12:682482.

    Article  PubMed  PubMed Central  Google Scholar 

  • Florea CM, Rosu R, Moldovan R, Vlase L, Toma V, Decea N, et al. The impact of chronic trimethylamine N-oxide administration on liver oxidative stress, inflammation, and fibrosis. Food Chem Toxicology: Int J Published Br Industrial Biol Res Association. 2024;184:114429.

    Article  CAS  Google Scholar 

  • Frangogiannis NG, Kovacic JC. Extracellular Matrix in Ischemic Heart Disease, Part 4/4: JACC Focus Seminar. J Am Coll Cardiol. 2020;75(17):2219–35.

    Article  PubMed  PubMed Central  Google Scholar 

  • Gallego P, Castejon-Vega B, Del Campo JA, Cordero MD. The absence of NLRP3-inflammasome modulates hepatic fibrosis progression, lipid metabolism, and inflammation in KO NLRP3 mice during aging. Cells. 2020;9(10).

  • Gawrys-Kopczynska M, Konop M, Maksymiuk K, Kraszewska K, Derzsi L, Sozanski K et al. TMAO, a seafood-derived molecule, produces diuresis and reduces mortality in heart failure rats. eLife. 2020;9.

  • Gregory JC, Buffa JA, Org E, Wang Z, Levison BS, Zhu W, et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem. 2015;290(9):5647–60.

    Article  CAS  PubMed  Google Scholar 

  • Guo F, Qiu X, Tan Z, Li Z, Ouyang D. Plasma trimethylamine n-oxide is associated with renal function in patients with heart failure with preserved ejection fraction. BMC Cardiovasc Disord. 2020;20(1):394.

    Article  PubMed  PubMed Central  Google Scholar 

  • Gupta N, Buffa JA, Roberts AB, Sangwan N, Skye SM, Li L et al. Targeted inhibition of gut microbial trimethylamine N-Oxide production reduces renal Tubulointerstitial Fibrosis and Functional Impairment in a murine model of chronic kidney disease. Arteriosclerosis, thrombosis, and vascular biology. 2020;40(5):1239–55.

  • Heianza Y, Ma W, Manson JE, Rexrode KM, Qi L. Gut microbiota metabolites and risk of major adverse Cardiovascular Disease events and death: a systematic review and Meta-analysis of prospective studies. J Am Heart Association. 2017;6(7).

  • Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature. 2020;587(7835):555–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Hinz B, Phan SH, Thannickal VJ, Prunotto M, Desmouliere A, Varga J, et al. Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am J Pathol. 2012;180(4):1340–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Huang F, Zhang F, Xu D, Zhang Z, Xu F, Tao X, et al. Enterococcus faecium WEFA23 from infants lessens high-fat-diet-induced hyperlipidemia via cholesterol 7-alpha-hydroxylase gene by altering the composition of gut microbiota in rats. J Dairy Sci. 2018;101(9):7757–67.

    Article  CAS  PubMed  Google Scholar 

  • Huang DQ, Singal AG, Kono Y, Tan DJH, El-Serag HB, Loomba R. Changing global epidemiology of liver cancer from 2010 to 2019: NASH is the fastest growing cause of liver cancer. Cell Metabol. 2022;34(7):969–77. e2.

    Article  CAS  Google Scholar 

  • Huc T, Drapala A, Gawrys M, Konop M, Bielinska K, Zaorska E, et al. Chronic, low-dose TMAO treatment reduces diastolic dysfunction and heart fibrosis in hypertensive rats. Am J Physiol Heart Circ Physiol. 2018;315(6):H1805–20.

    Article  CAS  PubMed  Google Scholar 

  • Israr MZ, Bernieh D, Salzano A, Cassambai S, Yazaki Y, Heaney LM, et al. Association of gut-related metabolites with outcome in acute heart failure. Am Heart J. 2021;234:71–80.

    Article  CAS  PubMed  Google Scholar 

  • Jameson E, Doxey AC, Airs R, Purdy KJ, Murrell JC, Chen Y. Metagenomic data-mining reveals contrasting microbial populations responsible for trimethylamine formation in human gut and marine ecosystems. Microb Genom. 2016;2(9):e000080.

    PubMed  PubMed Central  Google Scholar 

  • Janeiro MH, Ramirez MJ, Milagro FI, Martinez JA, Solas M. Implication of trimethylamine N-Oxide (TMAO) in Disease: potential biomarker or New Therapeutic Target. Nutrients. 2018;10(10).

  • Jernberg C, Lofmark S, Edlund C, Jansson JK. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J. 2007;1(1):56–66.

    Article  CAS  PubMed  Google Scholar 

  • Jimenez B, Montoliu C, MacIntyre DA, Serra MA, Wassel A, Jover M, et al. Serum metabolic signature of minimal hepatic encephalopathy by (1)H-nuclear magnetic resonance. J Proteome Res. 2010;9(10):5180–7.

    Article  CAS  PubMed  Google Scholar 

  • Johnson ME, Franks JM, Cai G, Mehta BK, Wood TA, Archambault K, et al. Microbiome dysbiosis is associated with disease duration and increased inflammatory gene expression in systemic sclerosis skin. Arthritis Res Therapy. 2019;21(1):49.

    Article  Google Scholar 

  • Kapetanaki S, Kumawat AK, Persson K, Demirel I. The Fibrotic effects of TMAO on Human renal fibroblasts is mediated by NLRP3, Caspase-1 and the PERK/Akt/mTOR pathway. Int J Mol Sci. 2021;22(21).

  • Kim RB, Morse BL, Djurdjev O, Tang M, Muirhead N, Barrett B, et al. Advanced chronic kidney disease populations have elevated trimethylamine N-oxide levels associated with increased cardiovascular events. Kidney Int. 2016;89(5):1144–52.

    Article  CAS  PubMed  Google Scholar 

  • Kim SJ, Bale S, Verma P, Wan Q, Ma F, Gudjonsson JE, et al. Gut microbe-derived metabolite trimethylamine N-oxide activates PERK to drive fibrogenic mesenchymal differentiation. iScience. 2022;25(7):104669.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Kinugasa Y, Nakamura K, Kamitani H, Hirai M, Yanagihara K, Kato M, et al. Trimethylamine N-oxide and outcomes in patients hospitalized with acute heart failure and preserved ejection fraction. ESC Heart Fail. 2021;8(3):2103–10.

    Article  PubMed  PubMed Central  Google Scholar 

  • Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19(5):576–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Konieczny R, Zurawska-Plaksej E, Kaaz K, Czapor-Irzabek H, Bombala W, Mysiak A, et al. All-cause mortality and trimethylamine N-Oxide levels in patients with Cardiovascular Disease. Cardiology. 2022;147(4):443–52.

    Article  CAS  PubMed  Google Scholar 

  • Krack A, Sharma R, Figulla HR, Anker SD. The importance of the gastrointestinal system in the pathogenesis of heart failure. Eur Heart J. 2005;26(22):2368–74.

    Article  CAS  PubMed  Google Scholar 

  • Kretzschmar M, Massague J. SMADs: mediators and regulators of TGF-beta signaling. Curr Opin Genet Dev. 1998;8(1):103–11.

    Article  CAS  PubMed  Google Scholar 

  • Krueger SK, Williams DE. Mammalian flavin-containing monooxygenases: structure/function, genetic polymorphisms and role in drug metabolism. Pharmacol Ther. 2005;106(3):357–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Kwan HS, Barrett EL. Purification and properties of trimethylamine oxide reductase from Salmonella typhimurium. J Bacteriol. 1983;155(3):1455–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med. 2016;8(1):39.

    Article  PubMed  PubMed Central  Google Scholar 

  • LeBleu VS, Taduri G, O’Connell J, Teng Y, Cooke VG, Woda C, et al. Origin and function of myofibroblasts in kidney fibrosis. Nat Med. 2013;19(8):1047–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Li CX, Talele NP, Boo S, Koehler A, Knee-Walden E, Balestrini JL, et al. MicroRNA-21 preserves the fibrotic mechanical memory of mesenchymal stem cells. Nat Mater. 2017;16(3):379–89.

    Article  CAS  PubMed  Google Scholar 

  • Li X, Zhang X, Pan Y, Shi G, Ren J, Fan H, et al. mTOR regulates NLRP3 inflammasome activation via reactive oxygen species in murine lupus. Acta Biochim Biophys Sin (Shanghai). 2018a;50(9):888–96.

    Article  CAS  PubMed  Google Scholar 

  • Li Z, Wu Z, Yan J, Liu H, Liu Q, Deng Y et al. Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Laboratory investigation; a journal of technical methods and pathology. 2018b.

  • Li X, Geng J, Zhao J, Ni Q, Zhao C, Zheng Y, et al. Trimethylamine N-Oxide exacerbates Cardiac Fibrosis via activating the NLRP3 inflammasome. Front Physiol. 2019;10:866.

    Article  PubMed  PubMed Central  Google Scholar 

  • Li W, Huang A, Zhu H, Liu X, Huang X, Huang Y, et al. Gut microbiota-derived trimethylamine N-oxide is associated with poor prognosis in patients with heart failure. Med J Aust. 2020;213(8):374–9.

    Article  PubMed  Google Scholar 

  • Li N, Zhou J, Wang Y, Chen R, Li J, Zhao X, et al. Association between trimethylamine N-oxide and prognosis of patients with acute myocardial infarction and heart failure. ESC Heart Fail. 2022;9(6):3846–57.

    Article  PubMed  PubMed Central  Google Scholar 

  • Liu ZY, Tan XY, Li QJ, Liao GC, Fang AP, Zhang DM, et al. Trimethylamine N-oxide, a gut microbiota-dependent metabolite of choline, is positively associated with the risk of primary liver cancer: a case-control study. Nutr Metabolism. 2018;15:81.

    Article  CAS  Google Scholar 

  • Liu X, Tu J, Zhou Z, Huang B, Zhou J, Chen J. TMAO-Activated hepatocyte-derived exosomes are widely distributed in mice with different patterns and promote vascular inflammation. Cardiol Res Pract. 2022;2022:5166302.

    Article  PubMed  PubMed Central  Google Scholar 

  • Loo RL, Chan Q, Nicholson JK, Holmes E. Balancing the equation: a natural history of trimethylamine and Trimethylamine-N-oxide. J Proteome Res. 2022;21(3):560–89.

    Article  CAS  PubMed  Google Scholar 

  • Makishima M, Okamoto AY, Repa JJ, Tu H, Learned RM, Luk A, et al. Identification of a nuclear receptor for bile acids. Science. 1999;284(5418):1362–5.

    Article  CAS  PubMed  Google Scholar 

  • Massague J. TGFbeta signalling in context. Nat Rev Mol Cell Biol. 2012;13(10):616–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Mejean V, Iobbi-Nivol C, Lepelletier M, Giordano G, Chippaux M, Pascal MC. TMAO anaerobic respiration in Escherichia coli: involvement of the tor operon. Mol Microbiol. 1994;11(6):1169–79.

    Article  CAS  PubMed  Google Scholar 

  • Miao J, Ling AV, Manthena PV, Gearing ME, Graham MJ, Crooke RM, et al. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nat Commun. 2015;6:6498.

    Article  CAS  PubMed  Google Scholar 

  • Milks MW, Velez MR, Mehta N, Ishola A, Van Houten T, Yildiz VO, et al. Usefulness of integrating heart failure risk factors into impairment of global longitudinal strain to Predict Anthracycline-Related Cardiac Dysfunction. Am J Cardiol. 2018;121(7):867–73.

    Article  CAS  PubMed  Google Scholar 

  • Missailidis C, Hallqvist J, Qureshi AR, Barany P, Heimburger O, Lindholm B, et al. Serum Trimethylamine-N-Oxide is strongly related to renal function and predicts outcome in chronic kidney disease. PLoS ONE. 2016;11(1):e0141738.

    Article  PubMed  PubMed Central  Google Scholar 

  • Nanto-Hara F, Kanemitsu Y, Fukuda S, Kikuchi K, Asaji K, Saigusa D, et al. The guanylate cyclase C agonist linaclotide ameliorates the gut-cardio-renal axis in an adenine-induced mouse model of chronic kidney disease. Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association -. Eur Ren Association. 2020;35(2):250–64.

    CAS  Google Scholar 

  • Nian F, Zhu C, Jin N, Xia Q, Wu L, Lu X. Gut microbiota metabolite TMAO promoted lipid deposition and fibrosis process via KRT17 in fatty liver cells in vitro. Biochem Biophys Res Commun. 2023;669:134–42.

    Article  CAS  PubMed  Google Scholar 

  • O’Dwyer DN, Ashley SL, Gurczynski SJ, Xia M, Wilke C, Falkowski NR, et al. Lung microbiota contribute to pulmonary inflammation and Disease Progression in Pulmonary Fibrosis. Am J Respir Crit Care Med. 2019;199(9):1127–38.

    Article  PubMed  PubMed Central  Google Scholar 

  • Organ CL, Otsuka H, Bhushan S, Wang Z, Bradley J, Trivedi R, et al. Choline Diet and its gut microbe-derived metabolite, trimethylamine N-Oxide, exacerbate pressure overload-Induced Heart failure. Circulation Heart Fail. 2016;9(1):e002314.

    Article  CAS  Google Scholar 

  • Papandreou C, More M, Bellamine A. Trimethylamine N-Oxide in relation to Cardiometabolic Health-cause or Effect? Nutrients. 2020;12(5).

  • Papandreou C, Bullo M, Hernandez-Alonso P, Ruiz-Canela M, Li J, Guasch-Ferre M, et al. Choline Metabolism and Risk of Atrial Fibrillation and Heart failure in the PREDIMED Study. Clin Chem. 2021;67(1):288–97.

    Article  PubMed  Google Scholar 

  • Patrone V, Puglisi E, Cardinali M, Schnitzler TS, Svegliati S, Festa A, et al. Gut microbiota profile in systemic sclerosis patients with and without clinical evidence of gastrointestinal involvement. Sci Rep. 2017;7(1):14874.

    Article  PubMed  PubMed Central  Google Scholar 

  • Qiu WD, Xiao XJ, Xia S, Gao ZP, Li LW. [Predictive value of plasma TMAO combined with NT-proBNP on the prognosis and length of hospitalization of patients with ischemic heart failure]. Zhonghua Xin xue guan bing za zhi. 2022;50(7):684–9.

    CAS  PubMed  Google Scholar 

  • Qu X, Li X, Zheng Y, Ren Y, Puelles VG, Caruana G, et al. Regulation of renal fibrosis by Smad3 Thr388 phosphorylation. Am J Pathol. 2014;184(4):944–52.

    Article  CAS  PubMed  Google Scholar 

  • Rath S, Heidrich B, Pieper DH, Vital M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome. 2017;5(1):54.

    Article  PubMed  PubMed Central  Google Scholar 

  • Rath S, Rud T, Pieper DH, Vital M. Potential TMA-Producing Bacteria are ubiquitously found in Mammalia. Front Microbiol. 2019;10:2966.

    Article  PubMed  Google Scholar 

  • Rhee EP, Clish CB, Ghorbani A, Larson MG, Elmariah S, McCabe E, et al. A combined epidemiologic and metabolomic approach improves CKD prediction. J Am Soc Nephrology: JASN. 2013;24(8):1330–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Roberts AB, Gu X, Buffa JA, Hurd AG, Wang Z, Zhu W, et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med. 2018;24(9):1407–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Roncal C, Martinez-Aguilar E, Orbe J, Ravassa S, Fernandez-Montero A, Saenz-Pipaon G, et al. Trimethylamine-N-Oxide (TMAO) predicts Cardiovascular Mortality in Peripheral Artery Disease. Sci Rep. 2019;9(1):15580.

    Article  PubMed  PubMed Central  Google Scholar 

  • Rosenbloom J, Macarak E, Piera-Velazquez S, Jimenez SA. Human Fibrotic diseases: current challenges in Fibrosis Research. Methods Mol Biol. 2017;1627:1–23.

    Article  CAS  PubMed  Google Scholar 

  • Saaoud F, Liu L, Xu K, Cueto R, Shao Y, Lu Y et al. Aorta- and liver-generated TMAO enhances trained immunity for increased inflammation via ER stress/mitochondrial ROS/glycolysis pathways. JCI Insight. 2023;8(1).

  • Salzano A, Israr MZ, Yazaki Y, Heaney LM, Kanagala P, Singh A, et al. Combined use of trimethylamine N-oxide with BNP for risk stratification in heart failure with preserved ejection fraction: findings from the DIAMONDHFpEF study. Eur J Prev Cardiol. 2020;27(19):2159–62.

    Article  PubMed  Google Scholar 

  • Schuett K, Kleber ME, Scharnagl H, Lorkowski S, Marz W, Niessner A, et al. Trimethylamine-N-oxide and heart failure with reduced Versus Preserved Ejection Fraction. J Am Coll Cardiol. 2017;70(25):3202–4.

    Article  PubMed  Google Scholar 

  • Shi C, Pei M, Wang Y, Chen Q, Cao P, Zhang L, et al. Changes of flavin-containing monooxygenases and trimethylamine-N-oxide may be involved in the promotion of non-alcoholic fatty liver disease by intestinal microbiota metabolite trimethylamine. Biochem Biophys Res Commun. 2022;594:1–7.

    Article  CAS  PubMed  Google Scholar 

  • Shuai W, Wen J, Li X, Wang D, Li Y, Xiang J. High-choline Diet exacerbates Cardiac Dysfunction, Fibrosis, and inflammation in a mouse model of heart failure with preserved ejection fraction. J Card Fail. 2020;26(8):694–702.

    Article  PubMed  Google Scholar 

  • Skaug B, Khanna D, Swindell WR, Hinchcliff ME, Frech TM, Steen VD, et al. Global skin gene expression analysis of early diffuse cutaneous systemic sclerosis shows a prominent innate and adaptive inflammatory profile. Ann Rheum Dis. 2020;79(3):379–86.

    Article  CAS  PubMed  Google Scholar 

  • Stec A, Maciejewska M, Paralusz-Stec K, Michalska M, Giebultowicz J, Rudnicka L, et al. The Gut Microbial Metabolite Trimethylamine N-Oxide is linked to specific complications of systemic sclerosis. J Inflamm Res. 2023;16:1895–904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Stefania K, Ashok KK, Geena PV, Katarina P, Isak D. TMAO enhances TNF-alpha mediated fibrosis and release of inflammatory mediators from renal fibroblasts. Sci Rep. 2024;14(1):9070.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Strilakou AA, Lazaris AC, Perelas AI, Mourouzis IS, Douzis I, Karkalousos PL, et al. Heart dysfunction induced by choline-deficiency in adult rats: the protective role of L-carnitine. Eur J Pharmacol. 2013;709(1–3):20–7.

    Article  CAS  PubMed  Google Scholar 

  • Strilakou A, Perelas A, Lazaris A, Papavdi A, Karkalousos P, Giannopoulou I, et al. Immunohistochemical determination of the extracellular matrix modulation in a rat model of choline-deprived myocardium: the effects of carnitine. Fundam Clin Pharmacol. 2016;30(1):47–57.

    Article  CAS  PubMed  Google Scholar 

  • Stubbs JR, Stedman MR, Liu S, Long J, Franchetti Y, West RE 3, et al. Trimethylamine N-Oxide and Cardiovascular outcomes in patients with ESKD receiving maintenance hemodialysis. Clin J Am Soc Nephrology: CJASN. 2019;14(2):261–7.

    Article  CAS  Google Scholar 

  • Sun G, Yin Z, Liu N, Bian X, Yu R, Su X, et al. Gut microbial metabolite TMAO contributes to renal dysfunction in a mouse model of diet-induced obesity. Biochem Biophys Res Commun. 2017;493(2):964–70.

    Article  CAS  PubMed  Google Scholar 

  • Suzuki T, Heaney LM, Bhandari SS, Jones DJ, Ng LL. Trimethylamine N-oxide and prognosis in acute heart failure. Heart. 2016;102(11):841–8.

    Article  CAS  PubMed  Google Scholar 

  • Tan X, Liu Y, Long J, Chen S, Liao G, Wu S, et al. Trimethylamine N-Oxide aggravates liver steatosis through modulation of bile acid metabolism and inhibition of farnesoid X receptor signaling in nonalcoholic fatty liver disease. Mol Nutr Food Res. 2019;63(17):e1900257.

    Article  PubMed  Google Scholar 

  • Tang WH, Hazen SL. Microbiome, trimethylamine N-oxide, and cardiometabolic disease. Translational Research: J Lab Clin Med. 2017;179:108–15.

    Article  CAS  Google Scholar 

  • Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368(17):1575–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Tang WH, Wang Z, Shrestha K, Borowski AG, Wu Y, Troughton RW, et al. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J Card Fail. 2015a;21(2):91–6.

    Article  CAS  PubMed  Google Scholar 

  • Tang WH, Wang Z, Kennedy DJ, Wu Y, Buffa JA, Agatisa-Boyle B, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circul Res. 2015b;116(3):448–55.

    Article  CAS  Google Scholar 

  • Teng ML, Ng CH, Huang DQ, Chan KE, Tan DJ, Lim WH, et al. Global incidence and prevalence of nonalcoholic fatty liver disease. Clin Mol Hepatol. 2023;29(Suppl):S32–42.

    Article  PubMed  Google Scholar 

  • Troseid M, Ueland T, Hov JR, Svardal A, Gregersen I, Dahl CP, et al. Microbiota-dependent metabolite trimethylamine-N-oxide is associated with disease severity and survival of patients with chronic heart failure. J Intern Med. 2015;277(6):717–26.

    Article  CAS  PubMed  Google Scholar 

  • Vallianou NG, Kounatidis D, Psallida S, Panagopoulos F, Stratigou T, Geladari E, et al. The interplay between Dietary Choline and Cardiometabolic disorders: a review of current evidence. Curr Nutr Rep. 2024;13(2):152–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • van Caam A, Vonk M, van den Hoogen F, van Lent P, van der Kraan P. Unraveling SSc pathophysiology; the myofibroblast. Front Immunol. 2018;9:2452.

    Article  PubMed  PubMed Central  Google Scholar 

  • van den Berg EH, Flores-Guerrero JL, Garcia E, Connelly MA, de Meijer VE, TransplantLines I, et al. High plasma levels of betaine, a trimethylamine N-Oxide-related metabolite, are associated with the severity of cirrhosis. Liver Int. 2023;43(2):424–33.

    Article  PubMed  Google Scholar 

  • Volkmann ER. Intestinal microbiome in scleroderma: recent progress. Current opinion in rheumatology. 2017.

  • Wang Z, Bergeron N, Levison BS, Li XS, Chiu S, Jia X, et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur Heart J. 2019;40(7):583–94.

    Article  CAS  PubMed  Google Scholar 

  • Wang G, Kong B, Shuai W, Fu H, Jiang X, Huang H. 3,3-Dimethyl-1-butanol attenuates cardiac remodeling in pressure-overload-induced heart failure mice. J Nutr Biochem. 2020;78:108341.

    Article  CAS  PubMed  Google Scholar 

  • Wargny M, Croyal M, Ragot S, Gand E, Jacobi D, Trochu JN, et al. Nutritional biomarkers and heart failure requiring hospitalization in patients with type 2 diabetes: the SURDIAGENE cohort. Cardiovasc Diabetol. 2022;21(1):101.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Warrier M, Shih DM, Burrows AC, Ferguson D, Gromovsky AD, Brown AL, et al. The TMAO-Generating enzyme Flavin Monooxygenase 3 is a Central Regulator of cholesterol balance. Cell Rep. 2015;10(3):326–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Wei H, Zhao M, Huang M, Li C, Gao J, Yu T, et al. FMO3-TMAO axis modulates the clinical outcome in chronic heart-failure patients with reduced ejection fraction: evidence from an Asian population. Front Med. 2022;16(2):295–305.

    Article  PubMed  Google Scholar 

  • Wong VW, Akaishi S, Longaker MT, Gurtner GC. Pushing back: wound mechanotransduction in repair and regeneration. J Invest Dermatol. 2011;131(11):2186–96.

    Article  CAS  PubMed  Google Scholar 

  • Wong VW, Ekstedt M, Wong GL, Hagstrom H. Changing epidemiology, global trends and implications for outcomes of NAFLD. J Hepatol. 2023;79(3):842–52.

    Article  PubMed  Google Scholar 

  • Wu WK, Chen CC, Liu PY, Panyod S, Liao BY, Chen PC et al. Identification of TMAO-producer phenotype and host-diet-gut dysbiosis by carnitine challenge test in human and germ-free mice. Gut. 2018.

  • Xu H, Yang F, Bao Z. Gut microbiota and myocardial fibrosis. Eur J Pharmacol. 2023;940:175355.

    Article  CAS  PubMed  Google Scholar 

  • Yang W, Zhang S, Zhu J, Jiang H, Jia D, Ou T, et al. Gut microbe-derived metabolite trimethylamine N-oxide accelerates fibroblast-myofibroblast differentiation and induces cardiac fibrosis. J Mol Cell Cardiol. 2019a;134:119–30.

    Article  CAS  PubMed  Google Scholar 

  • 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. Evid Based Complement Alternat Med. 2019b;2019:8937427.

    Article  PubMed  PubMed Central  Google Scholar 

  • Yap IK, Li JV, Saric J, Martin FP, Davies H, Wang Y, et al. Metabonomic and microbiological analysis of the dynamic effect of Vancomycin-induced gut microbiota modification in the mouse. J Proteome Res. 2008;7(9):3718–28.

    Article  CAS  PubMed  Google Scholar 

  • Zhang J, Cashman JR. Quantitative analysis of FMO gene mRNA levels in human tissues. Drug Metab Dispos. 2006;34(1):19–26.

    Article  PubMed  Google Scholar 

  • Zhang H, Meng J, Yu H. Trimethylamine N-oxide supplementation abolishes the cardioprotective effects of Voluntary Exercise in mice Fed a Western Diet. Front Physiol. 2017;8:944.

    Article  PubMed  PubMed Central  Google Scholar 

  • Zhang P, Zou JZ, Chen J, Tan X, Xiang FF, Shen B, et al. Association of trimethylamine N-Oxide with cardiovascular and all-cause mortality in hemodialysis patients. Ren Fail. 2020;42(1):1004–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhang W, Miikeda A, Zuckerman J, Jia X, Charugundla S, Zhou Z, et al. Inhibition of microbiota-dependent TMAO production attenuates chronic kidney disease in mice. Sci Rep. 2021;11(1):518.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhao ZH, Xin FZ, Zhou D, Xue YQ, Liu XL, Yang RX, et al. Trimethylamine N-oxide attenuates high-fat high-cholesterol diet-induced steatohepatitis by reducing hepatic cholesterol overload in rats. World J Gastroenterology: WJG. 2019;25(20):2450–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhao W, Shi CS, Harrison K, Hwang IY, Nabar NR, Wang M, et al. AKT regulates NLRP3 inflammasome activation by phosphorylating NLRP3 serine 5. J Immunol. 2020;205(8):2255–64.

    Article  CAS  PubMed  Google Scholar 

  • Zhou X, Jin M, Liu L, Yu Z, Lu X, Zhang H. Trimethylamine N-oxide and cardiovascular outcomes in patients with chronic heart failure after myocardial infarction. ESC Heart Fail. 2020;7(1):188–93.

    PubMed  PubMed Central  Google Scholar 

  • Zhou D, Zhang J, Xiao C, Mo C, Ding BS. Trimethylamine-N-oxide (TMAO) mediates the crosstalk between the gut microbiota and hepatic vascular niche to alleviate liver fibrosis in nonalcoholic steatohepatitis. Front Immunol. 2022;13:964477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Zhuang R, Ge X, Han L, Yu P, Gong X, Meng Q, et al. Gut microbe-generated metabolite trimethylamine N-oxide and the risk of diabetes: a systematic review and dose-response meta-analysis. Obes Rev. 2019;20(6):883–94.

    Article  CAS  PubMed  Google Scholar 

  • Zimmerman CM, Padgett RW. Transforming growth factor beta signaling mediators and modulators. Gene. 2000;249(1–2):17–30.

    Article  CAS  PubMed  Google Scholar 

  • Zou D, Li Y, Sun G. Attenuation of circulating trimethylamine N-Oxide prevents the progression of Cardiac and Renal Dysfunction in a rat model of chronic Cardiorenal Syndrome. Front Pharmacol. 2021;12:751380.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Figs. 1 and 2, and 3 were created with BioRender.com.

Funding

This work was supported by an American Heart Association Postdoctoral Fellowship (to TS), the National Institute of Arthritis and Musculoskeletal and Skin Diseases (R33AR076821 and R61AR076821 to JV and KH), the National Institute of Heart, Lung, and Blood Institute (R01HL153306 to KH), and the National Institute on Aging (R21AG081706 to KH). The funders did not have any role in the design of the study, data collection, analysis, or interpretation, or in writing the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

JJ, EC, TS, and PV did the literature search and were major contributors to writing the original manuscript. JJ and KH created figures. All authors read, revised, and approved the final manuscript.

Corresponding author

Correspondence to Karen J. Ho.

Ethics declarations

Ethical approval and consent to participate

Not applicable (review paper which does not involve human subjects).

Consent for publication

Not applicable (manuscript does not include personal data).

Competing interests

None.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jang, J.W., Capaldi, E., Smith, T. et al. Trimethylamine N-oxide: a meta-organismal axis linking the gut and fibrosis. Mol Med 30, 128 (2024). https://doi.org/10.1186/s10020-024-00895-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s10020-024-00895-8

Keywords