Skip to main content
Cardiovascular Research logoLink to Cardiovascular Research
. 2022 Apr 14;118(16):3171–3182. doi: 10.1093/cvr/cvac057

Mechanisms, therapeutic implications, and methodological challenges of gut microbiota and cardiovascular diseases: a position paper by the ESC Working Group on Coronary Pathophysiology and Microcirculation

Dimitris Tousoulis 1,, Tomasz Guzik 2, Teresa Padro 3, Dirk J Duncker 4, Giuseppe De Luca 5, Etto Eringa 6, Marija Vavlukis 7, Alexios S Antonopoulos 8, Themistoklis Katsimichas 9, Edina Cenko 10, Ana Djordjevic-Dikic 11, Ingrid Fleming 12, Olivia Manfrini 13, Danijela Trifunovic 14, Charalambos Antoniades 15, Filippo Crea 16,2
PMCID: PMC11023489  PMID: 35420126

Abstract

The human gut microbiota is the microbial ecosystem in the small and large intestines of humans. It has been naturally preserved and evolved to play an important role in the function of the gastrointestinal tract and the physiology of its host, protecting from pathogen colonization, and participating in vitamin synthesis, the functions of the immune system, as well as glucose homeostasis and lipid metabolism, among others. Mounting evidence from animal and human studies indicates that the composition and metabolic profiles of the gut microbiota are linked to the pathogenesis of cardiovascular disease, particularly arterial hypertension, atherosclerosis, and heart failure. In this review article, we provide an overview of the function of the human gut microbiota, summarize, and critically address the evidence linking compositional and functional alterations of the gut microbiota with atherosclerosis and coronary artery disease and discuss the potential of strategies for therapeutically targeting the gut microbiota through various interventions.

Keywords: Coronary artery disease, Atherosclerosis, Gut microbiota, Trimethylamine N-oxide, Short-chain fatty acids

Graphical Abstract

Graphical Abstract.

Graphical Abstract

The pathophysiology of gut microbial associations with atherosclerosis.

The figure depicts some of the key players at the organ, cellular, and molecular level, regarding the links between the gut microbiota and atherosclerosis. Diet has a profound effect on the gut microbiota, as it provides energy sources for bacterial fermentation. Undigested fibre is processed by bacteria-producing short-chain fatty acid (SCFA). Lower levels of SCFA have been linked to host inflammation, which aggravates atherosclerosis. Multiple bacterial species also produce trimethylamine (TMA) from choline, itself derived from dietary phosphatidylcholine. TMA is absorbed and metabolized to trimethylamine N-oxide in the liver and has been linked to the atherosclerotic process. Disruption of intestinal permeability may lead to bacterial lipopolysaccharide leaking into the bloodstream, initiating Toll-like receptor–mediated chronic systemic inflammation, and aggravating atherosclerosis. See text for details. Black arrows: Microbiota-related metabolites and host responses. Red arrows: Suggested proatherogenic effects.

1. Introduction

Cardiovascular disease (CVD), particularly coronary artery disease (CAD), still represents the leading cause of death worldwide1 despite effective progress in therapeutic interventions, such as early revascularization after acute coronary syndromes, lowering blood cholesterol levels, and inhibiting the renin-angiotensin-aldosterone system. Therefore, intense effort has been put into exploring and identifying new, therapeutically exploitable risk factors for atherosclerosis, to potentiate primary and secondary CAD prevention. Growing awareness of the influence of the human gut microbiota (GM) in the physiology of its host has led to the suggestion that it could contribute to the occurrence and development of atherosclerosis.2 Indeed, this microbial community living in the human intestinal tract can produce numerous metabolites that can enter systemic circulation and affect host health.3 Both metabolism-dependent and independent pathways have been proposed to explain the impact of the GM on atherogenesis.4 For example, the GM may exert pro-atherogenic effects via the synthesis of metabolites such as trimethylamine (TMA).5 Moreover, bacterial components such as lipopolysaccharides (LPS), found on the outer membrane of Gram-negative bacteria, can cross the host intestinal barrier, leak into the systemic circulation, and under certain circumstances contribute to low-grade chronic systemic inflammation, which by itself is a proatherogenic state.6

In this review, we briefly summarize existing knowledge on the GM and the tools developed for its compositional analysis, and we critically review the links between the GM and CAD. A glossary of terms is provided in Table 1.

Table 1.

Glossary of terms

Term Definition
Amplicon sequencing The identification and amplification of a certain genetic sequence, e.g. regions of the 16S ribosomal RNA gene of the microorganisms in a human stool sample.
Alpha diversity A term referring to individual characteristics of a microbial community (e.g. the number of different species).
Beta diversity A concept of direct comparison of different microbial communities by a certain metric.
Commensalism Co-existence of host and hosted organism, where only one of the two benefits.
Dysbiosis A change in microbial composition, relative to an arbitrarily defined healthy state.
Metagenomics The genetic analysis and direct identification of all genomes in a sample of matter (e.g. a stool sample).
Microbiome All the genomes of all the microorganisms in a specific environment.
Microbiota The community of microorganisms living in a specific environment.
Mutualism A beneficial relationship for both the host and the hosted organism.
NGS Modern DNA sequencing techniques.
Prebiotics Indigestible food compounds that are fermented by and stimulate growth/activity of beneficial intestinal microorganisms (e.g. components of dietary fibre).
Probiotics An accepted definition is ‘live microorganisms which when administered in adequate amounts confer a health benefit on the host’.
Synbiotics The official definition is ‘a mixture comprising live microorganisms and substrate(s) selectively utilized by host microorganisms that confers a health benefit on the host’.

2. Methods of microbial analysis

Analysing the microbiome of diverse species and environments using next generation sequencing (NGS) techniques has significantly enhanced our understanding of metabolic, physiological, and ecological roles of environmental microorganisms (Table 2). The elucidation of the microbiome is not straightforward and is concerned with experimental conditions (i.e. sampling issues, sequencing errors, and genomic repeats) and computationally intensive and cumbersome downstream analysis (i.e. quality control, assembly, binning, and statistical analyses). Thorough reviews on the best practices and pitfalls associated with microbial analysis have been previously published.7,8

Table 2.

Methods of microbial analysis

Method Advantages Disadvantages
Amplicon sequencing (marker gene analysis)
  • Simple and relatively cheap

  • Large, publicly available datasets

  • Provides genus-level resolution at most

  • No metabolic information (predictive algorithms required on top of analysis)

  • No discrimination between live and dead organisms

  • Multiple technical issues involved (choice of primers, amplification bias, other)

Metagenomics (metagenome analysis)
  • Species- and strain- level resolution

  • Direct inferement of the relative abundance of metabolic genes

  • No PCR bias

  • Expensive and complex

  • Sensitive to contamination

  • No discrimination between live and dead organisms

Metatranscriptomics (metatranscpriptome analysis)
  • Allows estimation of actively transcribing organisms

  • Direct evaluation of microbial activity

  • Expensive and complex

  • Sensitive to contamination

  • Data skewed towards organisms with high transcription rates

PCR, polymerase chain reaction.

Gut microbial genetic analysis is generally performed by either amplicon sequencing or metagenomics.7 The most widely used target gene for bacterial identification in amplicon sequencing is the 16S rRNA gene, which encodes the 16S RNA component of the 30S small subunit of the prokaryotic ribosome. This method is relatively inexpensive and easily implemented, yielding genus-level taxonomic and relative abundance information on the GM sample.

Metagenomics is the direct genetic analysis of all genomes obtained from a given sample, without the need for cell cultures.9,10 This approach comprehensively catalogues all microbial genes, allowing the detection of genes that can provide information on molecular function and the metabolic profiling of a microbial community.7 Although this technology is more expensive and computationally challenging, it provides information on genetic diversity and potential microbial functions rather than simply on the taxonomic diversity of a community.9,11,12

Further options for GM analysis include metatranscriptomics, which uses RNA sequencing to profile transcription in microbiomes, providing information on gene expression and the active functional output of the microbiome. Sequencing microbial RNA provides better insight into the functional activity of a microbial community, though it is biased towards organisms with higher rates of transcription.13 Data from metagenomics and metatranscriptomics can be combined and compared by various bioinformatics tools.7 The exploration of additional -omics data, such as meta-proteomics and metabolomics, can also be performed. Newer developments are constantly under way, and long-read sequencing, a promising technique assessing genetic sequences of thousands of base pairs,14 has already been applied to canine faecal samples.15

3. Statistical analysis of microbial data

The two principal components of sample comparison in gut microbial research are alpha and beta diversity. Alpha diversity, often described as within-sample diversity, pertains to specific characteristics of a given microbial community, such as the number of different species. Biodiversity indices like the Shannon index can be calculated to average alpha diversity variation and compare it between groups of samples. Beta diversity, often described as between-sample diversity, refers to the direct comparison of different microbial communities. Similarity measures between samples are calculated, and the resulting data matrix is processed by multivariate mathematical tools, most popular of which are ANalysis Of Similarities (ANOSIM) and Permutational multivariate analysis of variance (PERMANOVA).16,17 These tools test whether microbial variation between a priori selected groups of samples is greater than variation within each group, yielding a decision for statistical significance. A framework for the analysis of multivariate ecological data has been thoroughly described.18

Differential abundance analysis between samples is demanding, with key issues being that relative abundance data are compositional (i.e. fractions or percentages, which when added must equal 1 or 100%), sparse, and non-normally distributed. Moreover, false-positive results are inevitable after hundreds of individual comparisons and must be controlled for. Analysis of compositions of microbiome (ANCOM) and its evolutions is one of the methods employed in this analysis.19 A recommended approach is to use the isometric log ratio transformation to render standard statistical tests valid in differential abundance testing.7 These issues also affect correlational analysis, necessitating specially designed statistical software, such as SParse InversE Covariance Estimation for Ecological Association Inference (SPIEC-EASI).20

A wide variety of bioinformatics software tools have been used to classify microbiota based on sequencing data. A study comparing 11 software tools for interpreting shotgun metagenomics found that they yielded different conclusions, highlighting the need to improve the accuracy of results by combining existing bioinformatics tools with different classification principles, thus controlling each software tool’s specific limitations and strengths.21 Machine learning algorithms may provide a solution, as these have been utilized to predict sample origin (e.g. patient vs. control subject) from microbiome data, among other predictions, such as future states of disease.7

4. Physiology and pathophysiology of the GM

4.1. Physiology of the microbial ecosystem in the human gut

The human GM has been estimated to comprise approximately 100 trillion microorganisms, including archaea, fungi, and viruses, but predominantly anaerobic bacteria belonging to several major bacterial phyla.22 Among a multitude of functions, the GM, with its commensal, symbiotic, and mutualistic bacteria, plays a vital role in the dietary nutrient and xenobiotic metabolism of its host, participates in the maintenance of the physiology and structural integrity of the gut mucosa, as well as in bile salt metabolism, vitamin synthesis, and immunomodulation and defence against pathogens.23–25 Importantly, several bacterial species produce short-chain fatty acids (SCFA), which exert beneficial effects on humans, with butyrate being a strong anti-inflammatory molecule and the main energy source for enterocytes.26 The GM has also been shown to metabolize drugs of cardiovascular interest, including digoxin,27 while microbial metabolites correlate with the response to statins.28 Indeed, many mechanisms have been proposed by which the GM can affect the bioavailability and actions of various cardiovascular drugs and how these drugs may affect the GM, although, to date, sufficient knowledge of these processes is still lacking.29 Even though the human GM remains relatively stable throughout a person’s adult life, compositional intra- and inter-individual variability are considerable and can be influenced by host genetics, geographic origin, age, early life antibiotic use, as well as several other factors, most importantly dietary habits.30–36 Population-level studies have attempted to explain the variation that characterizes the human GM. A large metagenomics study from the Netherlands associated part of the variation with a total of 126 factors, both environmental and host-releated.37 Most of these factors, however, had a weak effect on microbial composition and diversity. A combination of the database of this study with data from a Belgian population described a core GM shared by 95% of all subjects, yielded further insights, and provided evidence that medication received by the host explains a significant part of the total microbial variation.38 Surprisingly, the core GM consisted of less than 20 genera, underscoring the enormous inter-individual variation.

Host genetics can affect the composition of the GM, and colonization experiments have indicated that some host-microbiota metabolic interactions are transferrable traits. For example, genetic aberration leading to absence of the bile acid-sensing Farnesoid X nuclear receptor is responsible for GM-dependent, diet-induced obesity in germ-free mice.39 Likewise, early-life, transient antibiotic therapy may provoke significant changes in GM composition, depending on the composition of the microbial community and the antimicrobial spectrum of the antibiotics used, with prolonged consequences for the host.40–42 However, a recent randomized controlled trial in preterm infants contradicts this hypothesis.43

Host diet is considered to be one of the primary determinants of GM composition.31,36 Western diet has been associated with decreased gut microbial diversity, compared to non-Western dietary habits.31 The beneficial effects of the Mediterranean diet are partly attributed to the fact that consumption of fruits, vegetables, and legumes, rich in fibre that is processed by bacteria, has been associated with increased SCFA levels. High fibre diets have been proposed to be associated with the largest benefit, due to their association with higher levels of SCFA and increased bacterial diversity.31

4.2. The pathophysiological basis of gut microbial associations with CAD

Compositional and functional changes of the GM relative to an–arbitrary–healthy state constitute what has been called dysbiosis and can generate CAD-related risk factors, in what has come to be known as the ‘gut-heart axis’. An overview of the pathophysiology will be provided here. The evidence from the relevant animal and human studies are presented in the next section.

Interactions between the GM and components of the cardiovascular system are mainly mediated by bacterial metabolites absorbed by the gut, bacterial molecular signals that can affect host cellular functions, and GM-derived compounds that leak into the systemic circulation (Table 3).44,45 GM-mediated systemic inflammation may be the main driver of the influence that gut bacteria exert on atherosclerosis and subsequent CAD.45,46 Inflammatory pathways are partly mediated by Toll-like receptor (TLR) activity, but although convincing data causally linking inflammation with atherosclerosis exist in rodents, the evidence in humans is scarce.45

Table 3.

Pathophysiological links between the gut microbiota and CAD

Process Molecules
Infections cause inflammation that may affect atherosclerotic plaque stability. Cytokines, including tumour necrosis factor-α (TNF-α).
Lipopolysaccharides leaking into the bloodstream trigger low-grade systemic inflammation. Lipopolysaccharides, TLRs.
Bacterial metabolites may affect proatherogenic celullar functions through foam cell formation and accumulation. Trimethylamine N-Oxide (TMAO).
Reduced production of anti-inflammatory short-chain fatty acids or indole derivatives may promote systemic inflammation or metabolic dysregulation. Propionate, acetate, butyrate, tryptophan, indole, indole derivatives.
GM-mediated aberrations in lipid metabolism might influence atherosclerosis. Bile acids, cholesterol.

Metabolites produced by the GM have emerged as pivotal regulators of signalling pathways directly involved in atherosclerosis and arterial thrombosis. TMAO, produced by the liver from gut microbial TMA, has received considerable attention as a potential key player in the ‘gut-heart axis’, mechanistically linked to atherosclerosis progression by potentially promoting foam cell production and accumulation.5 Tryptophan metabolism by gut bacteria has been linked to CVD, through GM-mediated production of indole derivatives. These molecules are mostly beneficial, stimulating production of IL-22 in the gut and possibly strengthening the epithelial barrier.47,48 A reduced capacity of the GM to metabolize tryptophan has been associated with host metabolic dysregulation.49 Certain conditions, including obesity, inhibit GM-mediated production of indole metabolites, by enhancing tryptophan metabolism towards kynurenine, through increased activity of indoleamine 2,3-dioxygenase (IDO).50 Moreover, SCFAs are very important molecules with multiple links to human physiological functions. In general, they are considered beneficial metabolites, associated, among others, with energy production that benefits the host, lipid regulation, immune system modulation, and cardioprotective effects such as beneficial effects of against hypertension-induced cardiac injury and vascular remodelling and atheroprotective properties;51–56 these are mediated either directly or via signalling pathways involving G protein-coupled receptors, such as GPR41and GPR43, and the peroxisome proliferator-activated receptors. Lower levels of SCFA are thus likely implicated in aberrations of host physiology.

Other studies have also highlighted the modulation of host lipid metabolism by the GM.57–59 Existing evidence links this modulation not only to SCFA but to bile acid signalling as well,60 through the bile acid receptor, alternatively known as the Farnesoid X receptor. Bile acid deconjugation in the intestines is known to be mediated by gut bacteria, which also generate secondary bile acids.

The impact of the GM on CAD is not likely restricted to metabolic profiles, as studies have revealed the presence of bacterial DNA in atherosclerotic plaques, implying that gut bacteria could translocate to such lesions to potentially influence plaque inflammatory status and stability.61,62

Finally, gut microbial components can trigger low-grade inflammation in humans.63–65 LPS from Gram-negative bacteria can leak into the circulation via a disrupted or even intact intestinal barrier, induce inflammation, and contribute to atherosclerosis. The evidence linking LPS to atherosclerosis has been accumulating for more than two decades, highlighting the detrimental role of endotoxins to human health.6,66

5. Animal and human studies on the association between GM and CAD

5.1. Animal studies

Studies in mice have revealed that genetically linked or diet-induced obesity is accompanied by changes in GM composition as well as by induction of systemic inflammation and vascular dysfunction67,68 that could be prevented by intermittent fasting.67 Dysregulation of GM-related microRNA functions has been shown to induce obesity in mice and inflammation in white adipose tissue.69 Similarly, in swine treated with deoxycorticosterone acetate and fed a diet high in fat, salt, and sugar, the diversity of the GM was reduced and its composition shifted towards bacteria associated with host inflammation that was linked with increased circulating levels of tumour necrosis factor-α.70 Such studies reveal the links between GM aberrations and host inflammation, a powerful driver of atherosclerosis and, ultimately, CAD.

To investigate causality between gut dysbiosis and CAD, experimental studies have explored the effects of faecal microbiota transplantation to mice from animals that received a high caloric diet71 or from patients with CAD.72 Such interventions led to alterations in glucose and bile acid metabolism, immune activation, and vascular dysfunction.71,72 In LDL-receptor deficient mice, the presence of GM (as opposed to germ-free counterparts) promoted atherothrombosis in a model of ultrasound-induced plaque rupture.73 Similarly, microbiota with pro-inflammatory potential transplanted to germ-depleted, LDL-receptor deficient mice accelerated the atherosclerotic process and reduced SCFA concentrations.74 Such studies imply that the GM and its dysbiotic states may contribute to CVD, by promoting inflammation and atherosclerosis.

Importantly, in rats suffering a mechanically induced myocardial infarction, alterations in the GM were paralleled by impairment of the intestinal barrier.75 These observations suggest that not only intestinal dysbiosis might lead to CAD, but that in turn CAD could perpetuate intestinal dysbiosis, thereby creating a vicious cycle.

Regarding GM-mediated bile salt metabolism and its role in atherosclerosis, FXR-deficient mice have been shown to develop hypercholesterolaemia.76  Apoe and Fxr double-knockout mice develop larger atherosclerotic lesions than ApoE−/− mice.77 However, evidence in humans for a role of bile salt metabolism in atherosclerosis is limited.

In a landmark study, TMAO was causally linked to atherosclerosis in rodents.5 Susceptibility to atherosclerosis in mice can even be transmitted from high TMAO-producing, atherosclerosis-prone mice to low TMAO-producing, atherosclerosis-resistant mice via the faecal microbiota.78 GM-derived TMA can also directly contribute to platelet hyper-reactivity in a number of platelet agonists to enhance a prothrombotic potential.79

Finally, in an elegant study, inhibition or deletion of IDO1 in mice fed with a high fat diet resulted in significant differences in gut microbial composition and shifted host tryptophan metabolism away from the kynurenine pathway and towards GM-mediated generation of indole derivatives, resulting in the production of anti-inflammatory IL-22 and decreased endotoxemia.50

5.2. Human studies

A growing body of clinical evidence also indicates an important role of the GM in CAD (Table 4). Although earlier characterization of atherosclerotic plaque microbial communities had been attempted,61,80 Koren et al.62 were the first to examine both the GM and plaque microbial colonies in atherosclerotic patients. They found that microbial colonies of atherosclerotic plaques and gut bacterial communities have distinct compositional differences. They also reported that the GM of atherosclerotic patients was not significantly different from that of controls, although it is unclear how detailed this comparison was. Other studies, however, have found significant differences in beta diversity or individual taxa between patients and controls.81–88 Moreover, metabolic features of the GM have been shown to differ between atherosclerotic patients and controls.81,84 Metabolic alterations of the GM in individuals with atherosclerosis suggest a microbiota with a higher inflammatory potential.84 Studies have demonstrated a decreased abundance of microbes with capacity for producing butyrate and increased circulating levels of TMAO in atherosclerotic patients.84,86

Table 4.

Human studies on the association of the gut microbiota with atherosclerosis and CAD

Study Location Sample size Age (y) patients/controls Sex (M, %) patients/controls Analysis approach Major findings
Koren et al.62 Single-centre (Sweden) 15 patients/15 controls 65.7 ± 0.5/70.5 ± 0.5 80%/80% 16S rRNA sequencing of stool and atherosclerotic plaque samples Plaque and gut communities different in beta diversity but several phylotypes common in the same individual. Patient and control group gut microbiota similar.
Karlsson et al.81 Single-centre (Sweden) 12 patients/13 controls 67.6 ± 8.6/70.5 ± 0.5 75%/77% Metagenomics in stool samples Groups different in beta diversity. Collinsella, Roseburia, and Eubacterium genera statistically different in relative abundance between groups.
Yin et al.83 Single-centre (China) 322 patients/231 controls 61 ± 19/56 ± 11 68.3%/56.3% 16S rRNA sequencing of stool samples, liquid chromatography/mass spectrometry for TMAO measurement Alpha diversity bigger in patients, beta diversity different between two subgroups of age- and sex-matched pairs. TMAO levels lower in patients than in controls.
Emoto et al.82 Single-centre (Japan) 39 CAD patients/30 subjects with CVRF 61.1 ± 9.4/62.1 ± 6.4 85%/77% T-RFLP in stool samples Increased Lactobacillales and decreased Bacteroides and Prevotella in CAD patients.
Jie et al.84 Single-centre (China) 218 ACVD patients/187 controls 60.9 ± 9.8/60.2 ± 9.8 76%/41% metagenome-wide association study in stool samples. The ACVD gut microbiome deviates from the healthy status by increased abundance of Enterobacteriaceae and Streptococcus spp.
Cui et al.85 Multi-centre (China) 29 CHD in-hospital patients/35 HV 68.3 ± 9.5/66.1 ± 11.4 51%/66% 16S rRNA sequencing of stool samples Significant decrease in Bacteroidetes and increase in Firmicutes in faecal samples of CHD patients.
Zhu et al.86 Single-centre (China) 70 CAD patients/98 HV 63.3/60.1 43%/42% 16S rRNA sequencing of stool samples Escherichia, Shigella and Enterococcus were enriched in the CAD group, while Faecalibacterium, Subdoligranulum, Roseburia, and Eubacterium rectale were decreased with CAD.
Liu et al.87 Single-centre (China) 161 CAD patients/40 HV 62 [54–70]/55 [49–62] 73%/43% 16S rRNA sequencing of stool samples and serum metabolomics 29 metabolite modules that were separately classified as being positively or negatively correlated with CAD phenotypes, and the bacterial co-abundance group with characteristic changes at different stages of CAD was represented by Roseburia, Klebsiella, Clostridium IV, and Ruminococcaceae.
Toya et al.88 Single-centre (USA) 213 CAD patients/53 BMI-matched controls  64.1 ± 8.6/61.6 ± 10.0 60%/56% 16S rRNA sequencing of stool samples Decreased richness and evenness of gut microbiome in CAD patients. Decreased abundance of Lachnospiraceae NK4B4 group and Ruminococcus gauvreauii and increased abundance of R. gnavus in CAD patients.

ACVD, atherosclerotic cardiovascular disease; BMI: body mass index; CHD, coronary heart disease; CVRF, cardiovascular risk factor; HV, healthy volunteer; T-RFLP, terminal restriction fragment length polymorphism; n.i.: not indicated.

As previously mentioned,6,64,65 endotoxins are known causative factors of low-grade systemic inflammation, a crucial step in the pathway of atherosclerosis. In an important prospective study, patients who smoked and had LPS levels above the 90th percentile faced a three-fold increase of the risk for incident atherosclerosis, independently of vascular risk factors.6 In high-risk patients for CVD, increased circulating markers of gut-related inflammation, including LPS-binding protein, carried a two-fold increased risk of adverse cardiovascular outcomes.89 Circulating levels of LPS have also been associated with a higher risk of major adverse cardiovascular events in a large cohort of patients with atrial fibrillation.90

In a seminal study, Wang et al.5 reported that intestinal microbes participate in phosphatidylcholine metabolism to produce TMA, which is converted to TMAO by the liver, and that TMAO levels predicted major adverse cardiovascular events over a 3-year follow-up. Much research on TMAO followed that pioneering work. Two recent meta-analyses made a systematic evaluation of the relationship between TMAO plasma levels, mortality, and major adverse cardio- and cerebrovascular events.91,92 These studies suggested a direct and concentration-dependent association between TMAO levels and all-cause mortality, regardless of conventional risk factors. Fasting plasma TMAO levels were an independent predictor of a high atherosclerotic burden, as estimated with the SYNTAX score in patients with CAD, and of subclinical myocardial injury as quantified by high sensitivity cardiac troponin T (hs-cTnT).93 Further, a study in patients presenting with an acute coronary syndrome showed that rapid quantification of trimethyllysine and TMAO at presentation may provide added prognostic value for identifying patients at risk for either short- or long-term adverse cardiovascular events, including in patients with negative hs-cTnT levels at baseline.94 However, many other studies have failed to show an association of TMAO with CAD or CVD in general.3,95–99 Most importantly, a Mendelian randomization analysis has not shown any relation.100

6. Therapeutic options

6.1. Probiotics

Probiotics are ingestible microorganisms that reach the intestinal lumen, where they can play functional roles in host physiology.101 The oral administration of adequate amounts of probiotics has been reported to provide cardiovascular benefit.102 Potential mechanisms include the strengthening of gut epithelial tight junctions to reduce LPS leakage and the induction of bile acid deconjugation; this will increase bile acid excretion and force the host to use more cholesterol to counter this effect.

A summary of studies on the antiatherogenic properties of probiotics can be found elsewhere.103 As examples, oral administration of the probiotic Lactobacillus rhamnosus GR-1 ameliorated cardiac remodelling and pump failure in rats with an acute myocardial infarction produced by a permanent coronary artery occlusion.104 Moreover, treating ApoE-deficient mice with Bacteroides vulgatus and B. dorei, two species that may have a lower abundance in CAD patients, inhibited the formation of atherosclerotic plaques.105 Consumption of L. acidophilus preparations may have a greater effect in lowering cholesterol than other probiotics.106  Lactobacillus plantarum ZDY04 significantly reduced plasma TMAO in mice, possibly via remodelling of the GM.107

Although preliminary, these studies highlight the therapeutic potential of some probiotic products. However, most positive health claims for probiotics rely on poor evidence, and there are as yet no strong data that probiotic use can prevent CVD. Evidence in humans is scarce and strain-specific effects remain largely unclear.108

6.2. Prebiotics

Prebiotics are defined as indigestible food components, such as inulins, that promote the growth and/or activity of specific microorganisms within the GM. Conceivably, enhancing the metabolic potential of beneficial bacteria will confer some benefit to host physiology. A summary of animal and human studies of prebiotics can be found elsewhere.109 The enhanced production of SCFA and a strengthening of gut epithelial tight junctions are among the potential mechanisms of benefit, although the evidence is less clear in humans. Notably, bacterial metabolite-based interventions (‘postbiotics’) have also been described in the context of intestinal disease, holding promise for broader use.110

6.3. Synbiotics

Synbiotics are ingestible combinations of probiotics and prebiotics (see definition in the Glossary). A few studies have shown some benefit of synbiotic use in cardiovascular disorders. A summary of human trials can be found in Sáez-Lara et al.111 As examples, in diabetic patients with CAD, a 12-week intervention with a synbiotic mixture improved glycemic status and HDL levels, but did not alter other cardiovascular risk factors.112 Similar studies aimed at reducing hyperglycemia in diabetes had variable results.113 In a small randomized, placebo-controlled human trial, the 12-week use of a synbiotic showed a modest reduction in both total cholesterol and low density lipoprotein.114 Another randomized controlled trial has also shown benefits in lipid metabolism.115 However, the definition of the administered compounds as synbiotics in some trials has been challenged.116 Although such results are promising, the most effective synbiotic mixture for reduction of cardiovascular risk has yet to be identified.

6.4. Antibiotics

Given the nature of the subject and the abundance of antimicrobial compounds in the medical arsenal, the use of antibiotics has been tested in the context of CAD. However, all relevant trials in the field failed to show any benefit,117 and the detrimental effect of broad-spectrum antibiotics to beneficial gut bacteria severely limits this approach.

6.5. Dietary interventions

The relationship between diet and human health is well established. A diet characterized by a very low consumption of fibres and a high intake of red meat (or animal proteins), saturated fats, and simple sugars, such as that of Western industrialized societies, has been associated with a high risk of CVD.118,119 On the contrary, a nutritional regime characterized by a high consumption of cereals, legumes, nuts, vegetables and fruits, consumption of fish, white meat and eggs, and a low intake of wine, like that of some Mediterranean countries, has been related to a low risk of CVD.120–124

As mentioned above, diet has a fast and considerable effect on the composition of the GM.36 A summary of the effects that dietary interventions have on the human GM can be found in the studies by Santos-Marcos et al.109 and Gerdes et al.125 A dietary pattern rich in fibres may lead to beneficial compositional changes of the GM and higher production of SCFA and may partly explain the benefit derived from the Mediterranean diet.126,127 Butyrate in particular has strong anti-inflammatory properties and is considered highly beneficial for human health, providing the main energy source for colonocytes, as already mentioned. Low fibre intake leads not only to a reduction in GM composition and diversity but may also lead to a reduction in the production of SCFA.128 At least in rodents, diet rich in saturated fat can increase gut permeability and result in local increase of inflammatory cytokines.129 Thus, targeting the interactions between the host and gut microorganisms through change of dietary patterns may lead to prevention of CVD.

6.6. Additional interventions that modulate GM–host interactions

Multiple interventions aimed at skewing the profile of the GM towards a more host-beneficial state have been tested. These range from untargeted approaches, such as physical exercise and faecal transplantation, to targeted approaches that include bacterial engineering and drugs affecting bacterial metabolism.3 Most of these interventions remain far from reaching clinical practice.

Increased physical activity in humans has been linked to an enhanced bacterial potential for production of SCFA.130 The influence of exercise has been reviewed elsewhere.125 Faecal transplantation to human recipients, although of medical value in certain pathological conditions like Clostridioides difficile infection,131 currently lacks robust evidence for a connection to cardiovascular health.3 There are however hints of a benefit, as one small study showed that the transfer of GM from lean donors to subjects with metabolic syndrome improved insulin sensitivity and increased GM diversity.132 Moreover, in animal models, genetically modified microorganisms may have a therapeutic effect regarding the spectrum of CVD.133

Given the numerous studies mentioned above that link elevated TMAO levels with CVD, efforts have been made to inhibit TMAO production.134–137 Non-lethal (to microbes) small molecule drugs have been developed to inhibit bacterial trimethylamine lyase systems and have been experimentally tested in animal models, with promising results.

Finally, preliminary approaches in the field of nanomedicine have been conceptualized for future use, to modulate the GM composition, e.g. by using nanoparticles to deliver to the host specific microorganisms associated with favourable metabolic profiles.30

7. Critical interpretation of published data and methodological challenges

Understanding the role of the GM in cardiovascular risk and CAD has several diagnostic as well as therapeutic implications. Efforts to explore the GM and develop significant clinical applications depend on accurate analyses of microbial communities. Unfortunately, GM studies are confounded by the complexity of microbiome measurements11 and are very heterogenous in study design, employed methods, sampling, preservation, measured parameters, and study populations. Such differences limit the ability to reproduce and compare study results, as well as to extrapolate findings to other patient populations. Most importantly, there is yet no universally accepted consensus on what constitutes a healthy GM, and interindividual variability is enormous. Hence, the presence of control subjects in any given study is an a priori necessity.

Studies on GM are also often underpowered to capture the substantial variation in the gut microbiome, lack positive and negative controls, and parallel plasma and/or serum samples, making it difficult to translate the functional alterations of the GM to a final impact on health that is reflected in circulating, microbiota-dependent metabolites.11,12

Moreover, in most studies, the composition of the GM is typically determined by amplicon sequencing (marker gene analysis), but this approach only yields genus level taxonomic profiles,9 lacking any data on bacterial metabolic genes, and is no longer the method of choice.11 Various procedural steps in the microbial analysis pipeline of such studies can also be responsible for divergent results between them, as there is no detailed consensus on how to analyse the gut microbiome. A recent study comparing six different approaches reported that DNA extraction methods had the highest impact on observed microbiome variability.138 Similarly, a study comparing the European Metagenomics of the Human Intestinal Tract and the American Human Microbiome Project reported significant differences in distribution of bacterial taxa depending on the DNA extraction method.139 These results emphasize the risks of comparing data across studies that apply different methodologies and reinforce the need for well-designed longitudinal studies and randomized clinical trials that provide parallel microbiota and plasma/serum samples and are controlled for critical covariates.

Indeed, many studies on the human GM lack the incorporation or statistical handling of key covariates, such as sex, age, body mass index, diet, lifestyle, ethnicity, geographical region, comorbidities, and medication, that could contribute to CVD development and progression, as well as affect the composition of the GM itself.12,34,35,140–142 Of note, the use of commonly prescribed drugs such as metformin or proton pump inhibitors leads to changes in the GM,140,143,144 and their contribution to compositional variance must be disentangled and accounted for.

Furthermore, the differences between gut microbial communities in humans and rodents hinder the extrapolation of animal study results to human subjects.145 Thus, while the analysis of gut microbial composition is a powerful research tool, it remains a complicated process, being relatively far from the point of clinical implementation.

The use of GM metabolites as biomarkers to identify high- and low-risk populations can be more easily implemented.146 This may be applicable to widely studied markers such as TMAO, uremic toxins (p-cresol sulphate; indoxyl sulphate—which are particularly significant in the context of chronic kidney disease), bile acids and SCFAs, as well as LPS and other bacterial wall constituents.147,148 Nevertheless, integrating multi-omics data is inherently difficult and identifying a metabolite from the microbiome is particularly challenging, as is identifying which microorganism or group of microorganisms is linked to alterations of the levels of a particular metabolite.

Interest in TMAO as a biomarker seems justified given that in healthy subjects, it was associated with a risk of myocardial infarction, stroke and death over a 3-year follow-up.149 Also, increased TMAO predicted outcomes even in the presence of other cardiovascular syndromes, such as peripheral artery disease,150 and heart failure.151,152 TMAO also has prothrombotic and proatherogenic effects,5,153 that are not avoided by the platelet aggregation inhibitor ticagrelor.154 However, TMAO as a biomarker remains controversial and debated,95–99 as conflicting results exist and clear-cut causality in humans has not been proven.

Lastly, as described in the related sections, a number of studies have focused on discerning the effects of GM modulation on cardiovascular health. Although promising evidence exists from research on probiotics and related compounds, this field is still far away from clinical implementation, and more, large, randomized controlled trials are required.

8. Conclusion

In a famous philosophical assay published in 1862, Ludwig Feuerbach wrote that we are what we eat. This extreme statement has since been challenged by many other philosophers. While what we eat certainly influences health, it is also not entirely correct, as we are also influenced by GM-dependent interpretation of our diet, which in turn affects various aspects of human physiology and plays a role in different human diseases, spanning from Parkinson’s disease to cancer. Ample evidence suggests a link between the GM and CAD, and while experimental investigations and association studies in human patients suggest that the link may be causal, interventional studies that alter the composition or function of the GM to influence the risk of disease are still lacking. Nevertheless, the information already available indicates that markers of gut bacterial dysbiosis may improve the risk stratification for CAD. The interest in this new human ‘organ’ and our ‘second genome’ is escalating fast.

Contributor Information

Dimitris Tousoulis, 1st Cardiology Department, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, 11527 Athens, Greece.

Tomasz Guzik, Institute of Cardiovascular Medical Sciences, BHF Glasgow Cardiovascular Research Centre, UK.

Teresa Padro, Sant Pau Institute for Biomedical Research, Barcelona, Spain.

Dirk J Duncker, Department of Cardiology, Thorax Center, Erasmus MC, Rotterdam, the Netherlands.

Giuseppe De Luca, Division of Cardiology, Eastern Piedmont University, Novara, Italy.

Etto Eringa, Institute of Cardiovascular Research, Amsterdam University Medical Center, Amsterdam, the Netherlands.

Marija Vavlukis, University Ss. Cyril and Methodius, Skopje, North Macedonia.

Alexios S Antonopoulos, 1st Cardiology Department, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, 11527 Athens, Greece.

Themistoklis Katsimichas, 1st Cardiology Department, National and Kapodistrian University of Athens, Vas. Sofias Avenue 114, 11527 Athens, Greece.

Edina Cenko, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.

Ana Djordjevic-Dikic, University Clinical Center of Serbia, Belgrade, Serbia.

Ingrid Fleming, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany.

Olivia Manfrini, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy.

Danijela Trifunovic, Cardiology Department, University of Belgrade, Belgrade, Serbia.

Charalambos Antoniades, RDM Division of Cardiovascular Medicine, University of Oxford, Oxford, UK.

Filippo Crea, Department of Cardiology and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy.

References

  • 1. Virani  SS, Alonso  A, Aparicio  HJ, Benjamin  EJ, Bittencourt  MS, Callaway  CW, Carson  AP, Chamberlain  AM, Cheng  S, Delling  FN, Elkind  MSV, Evenson  KR, Ferguson  JF, Gupta  DK, Khan  SS, Kissela  BM, Knutson  KL, Lee  CD, Lewis  TT, Liu  J, Loop  MS, Lutsey  PL, Ma  J, Mackey  J, Martin  SS, Matchar  DB, Mussolino  ME, Navaneethan  SD, Perak  AM, Roth  GA, Samad  Z, Satou  GM, Schroeder  EB, Shah  SH, Shay  CM, Stokes  A, Van Wagner  LB, Wang  NY, Tsao  CW; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee . Heart disease and stroke statistics-2021 update: a report from the American Heart Association. Circulation  2021;143:254–743. [DOI] [PubMed] [Google Scholar]
  • 2. Witkowski  M, Weeks  TL, Hazen  SL. Gut microbiota and cardiovascular disease. Circ Res  2020;127:553–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Fan  Y, Pedersen  O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol  2021;19:55–71. [DOI] [PubMed] [Google Scholar]
  • 4. Brown  JM, Hazen  SL. The gut microbial endocrine organ: bacterially derived signals driving cardiometabolic diseases. Annu Rev Med  2015;66:343–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Wang  Z, Klipfell  E, Bennett  BJ, Koeth  R, Levison  BS, Dugar  B, Feldstein  AE, Britt  EB, Fu  X, Chung  YM, Wu  Y, Schauer  P, Smith  JD, Allayee  H, Tang  WH, DiDonato  JA, Lusis  AJ, Hazen  SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature  2011;472:57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Wiedermann  CJ, Kiechl  S, Dunzendorfer  S, Schratzberger  P, Egger  G, Oberhollenzer  F, Willeit  J. Association of endotoxemia with carotid atherosclerosis and cardiovascular disease: prospective results from the Bruneck study. J Am Coll Cardiol  1999;34:1975–1981. [DOI] [PubMed] [Google Scholar]
  • 7. Knight  R, Vrbanac  A, Taylor  BC, Aksenov  A, Callewaert  C, Debelius  J, Gonzalez  A, Kosciolek  T, McCall  LI, McDonald  D, Melnik  AV, Morton  JT, Navas  J, Quinn  RA, Sanders  JG, Swafford  AD, Thompson  LR, Tripathi  A, Xu  ZZ, Zaneveld  JR, Zhu  Q, Caporaso  JG, Dorrestein  PC. Best practices for analysing microbiomes. Nat Rev Microbiol  2018;16:410–422. [DOI] [PubMed] [Google Scholar]
  • 8. Bharti  R, Grimm  DG. Current challenges and best-practice protocols for microbiome analysis. Brief Bioinform  2021;22:178–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Brooks  JP, Edwards  DJ, Harwich  MD  Jr, Rivera  MC, Fettweis  JM, Serrano  MG, Reris  RA, Sheth  NU, Huang  B, Girerd  P, Strauss  JF  III, Jefferson  KK, Buck  GA.  The truth about metagenomics: quantifying and counteracting bias in 16S rRNA studies. BMC Microbiol  2015;15:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Quince  C, Walker  AW, Simpson  JT, Loman  NJ, Segata  N. Shotgun metagenomics, from sampling to analysis. Nat Biotechnol  2017;35:833–844. [DOI] [PubMed] [Google Scholar]
  • 11. Schloss  PD. Identifying and overcoming threats to reproducibility, replicability, robustness, and generalizability in microbiome research. mBio  2018;9:00525-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Trøseid  M, Andersen  G, Broch  K, Hov  JR. The gut microbiome in coronary artery disease and heart failure: current knowledge and future directions. EBioMedicine  2020;52:102649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Bashiardes  S, Zilberman-Schapira  G, Elinav  E. Use of metatranscriptomics in microbiome research. Bioinform Biol Insights  2016;10:19–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Amarasinghe  SL, Su  S, Dong  X, Zappia  L, Ritchie  ME, Gouil  Q. Opportunities and challenges in long-read sequencing data analysis. Genome Biol  2020;21:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Cuscó  A, Pérez  D, Viñes  J, Fàbregas  N, Francino  O. Long-read metagenomics retrieves complete single-contig bacterial genomes from canine feces. BMC Genomics  2021;22:330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Clarke  K. Non-parametric multivariate analyses of changes in community structure. Austral Ecol  1993;18:117–143. [Google Scholar]
  • 17. Anderson  MJ. A new method for non-parametric multivariate analysis of variance. Austral Ecol  2001;26:32–46. [Google Scholar]
  • 18. Clarke  KR, Gorlet  RN, Somerfield  PJ, Warwick  RM. Change in marine communities: an approach to statistical analysis and interpretation. 3rd ed. Plymouth: PRIMER-E; 2014. [Google Scholar]
  • 19. Mandal  S, Van Treuren  W, White  RA, Eggesbø  M, Knight  R, Peddada  SD. Analysis of composition of microbiomes: a novel method for studying microbial composition. Microb Ecol Health Dis  2015;26:27663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Kurtz  ZD, Müller  CL, Miraldi  ER, Littman  DR, Blaser  MJ, Bonneau  RA. Sparse and compositionally robust inference of microbial ecological networks. PLoS Comput Biol  2015;11:e1004226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. McIntyre  ABR, Ounit  R, Afshinnekoo  E, Prill  RJ, Hénaff  E, Alexander  N, Minot  SS, Danko  D, Foox  J, Ahsanuddin  S, Tighe  S, Hasan  NA, Subramanian  P, Moffat  K, Levy  S, Lonardi  S, Greenfield  N, Colwell  RR, Rosen  GL, Mason  CE. Comprehensive benchmarking and ensemble approaches for metagenomic classifiers. Genome Biol  2017;18:182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Eckburg  PB, Bik  EM, Bernstein  CN, Purdom  E, Dethlefsen  L, Sargent  M, Gill  SR, Nelson  KE, Relman  DA. Diversity of the human intestinal microbial flora. Science  2005;308:1635–1638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Clemente  JC, Ursell  LK, Parfrey  LW, Knight  R. The impact of the gut microbiota on human health: an integrative view. Cell  2012;148:1258–1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Nicholson  JK, Holmes  E, Kinross  J, Burcelin  R, Gibson  G, Jia  W, Pettersson  S. Host gut microbiota metabolic interactions. Science  2012;336:1262–1267. [DOI] [PubMed] [Google Scholar]
  • 25. Guarner  F, Malagelada  JR. Gut flora in health and disease. Lancet  2003;361:512–519. [DOI] [PubMed] [Google Scholar]
  • 26. Salvi  PS, Cowles  RA. Butyrate and the intestinal epithelium: modulation of proliferation and inflammation in homeostasis and disease. Cells  2021;10:1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Lindenbaum  J, Rund  DG, Butler  VP  Jr, Tse-Eng  D, Saha  JR. Inactivation of digoxin by the gut flora: reversal by antibiotic therapy. N Engl J Med  1981;305:789–794. [DOI] [PubMed] [Google Scholar]
  • 28. Kaddurah-Daouk  R, Baillie  RA, Zhu  H, Zeng  ZB, Wiest  MM, Nguyen  UT, Wojnoonski  K, Watkins  SM, Trupp  M, Krauss  RM. Enteric microbiome metabolites correlate with response to simvastatin treatment. PLoS One  2011;6:e25482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Tuteja  S, Ferguson  JF. Gut microbiome and response to cardiovascular drugs. Circ Genom Precis Med  2019;12:421–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kazemian  N, Mahmoudi  M, Halperin  F, Wu  JC, Pakpour  S. Gut microbiota and cardiovascular disease: opportunities and challenges. Microbiome  2020;8:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Hasan  N, Yang  H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ  2019;7:e7502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Sears  CL. A dynamic partnership: celebrating our gut flora. Anaerobe  2005;11:247–251. [DOI] [PubMed] [Google Scholar]
  • 33. Qin  J, Li  R, Raes  J, Arumugam  M, Burgdorf  KS, Manichanh  C, Nielsen  T, Pons  N, Levenez  F, Yamada  T, Mende  DR, Li  J, Xu  J, Li  S, Li  D, Cao  J, Wang  B, Liang  H, Zheng  H, Xie  Y, Tap  J, Lepage  P, Bertalan  M, Batto  JM, Hansen  T, Le Paslier  D, Linneberg  A, Nielsen  HB, Pelletier  E, Renault  P, Sicheritz-Ponten  T, Turner  K, Zhu  H, Yu  C, Li  S, Jian  M, Zhou  Y, Li  Y, Zhang  X, Li  S, Qin  N, Yang  H, Wang  J, Brunak  S, Doré  J, Guarner  F, Kristiansen  K, Pedersen  O, Parkhill  J, Weissenbach  J, MetaHIT Consortium, Bork  P, Ehrlich  SD, Wang  J.  A human gut microbial gene catalogue established by metagenomic sequencing. Nature  2010;464:59–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Turnbaugh  PJ, Ridaura  VK, Faith  JJ, Rey  FE, Knight  R, Gordon  JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med  2009;1:6ra14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Yatsunenko  T, Rey  FE, Manary  MJ, Trehan  I, Dominguez-Bello  MG, Contreras  M, Magris  M, Hidalgo  G, Baldassano  RN, Anokhin  AP, Heath  AC, Warner  B, Reeder  J, Kuczynski  J, Caporaso  JG, Lozupone  CA, Lauber  C, Clemente  JC, Knights  D, Knight  R, Gordon  JI. Human gut microbiome viewed across age and geography. Nature  2012;486:222–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. David  LA, Maurice  CF, Carmody  RN, Gootenberg  DB, Button  JE, Wolfe  BE, Ling  AV, Devlin  AS, Varma  Y, Fischbach  MA, Biddinger  SB, Dutton  RJ, Turnbaugh  PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature  2014;505:559–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Zhernakova  A, Kurilshikov  A, Bonder  MJ, Tigchelaar  EF, Schirmer  M, Vatanen  T, Mujagic  Z, Vila  AV, Falony  G, Vieira-Silva  S, Wang  J, Imhann  F, Brandsma  E, Jankipersadsing  SA, Joossens  M, Cenit  MC, Deelen  P, Swertz  MA; LifeLines cohort study, Weersma  RK, Feskens  EJ, Netea  MG, Gevers  D, Jonkers  D, Franke  L, Aulchenko  YS, Huttenhower  C, Raes  J, Hofker  MH, Xavier  RJ, Wijmenga  C, Fu  J.  Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science  2016;352:565–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Falony  G, Joossens  M, Vieira-Silva  S, Wang  J, Darzi  Y, Faust  K, Kurilshikov  A, Bonder  MJ, Valles-Colomer  M, Vandeputte  D, Tito  RY, Chaffron  S, Rymenans  L, Verspecht  C, De Sutter  L, Lima-Mendez  G, D'hoe  K, Jonckheere  K, Homola  D, Garcia  R, Tigchelaar  EF, Eeckhaudt  L, Fu  J, Henckaerts  L, Zhernakova  A, Wijmenga  C, Raes  J. Population-level analysis of gut microbiome variation. Science  2016;352:560–564. [DOI] [PubMed] [Google Scholar]
  • 39. Parséus  A, Sommer  N, Sommer  F, Caesar  R, Molinaro  A, Ståhlman  M, Greiner  TU, Perkins  R, Bäckhed  F. Microbiota-induced obesity requires Farnesoid X receptor. Gut  2017;66:429–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Ianiro  G, Tilg  H, Gasbarrini  A. Antibiotics as deep modulators of gut microbiota: between good and evil. Gut  2016;65:1906–1915. [DOI] [PubMed] [Google Scholar]
  • 41. Blaser  MJ, Falkow  S. What are the consequences of the disappearing human microbiota?  Nat Rev Microbiol  2009;7:887–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Tanaka  S, Kobayashi  T, Songjinda  P, Tateyama  A, Tsubouchi  M, Kiyohara  C, Shirakawa  T, Sonomoto  K, Nakayama  J. Influence of antibiotic exposure in the early postnatal period on the development of intestinal microbiota. FEMS Immunol Med Microbiol  2009;56:80–87. [DOI] [PubMed] [Google Scholar]
  • 43. Russell  JT, Lauren Ruoss  J, de la Cruz  D, Li  N, Bazacliu  C, Patton  L, McKinley  KL, Garrett  TJ, Polin  RA, Triplett  EW, Neu  J. Antibiotics and the developing intestinal microbiome, metabolome and inflammatory environment in a randomized trial of preterm infants. Sci Rep  2021;11:1943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Katsimichas  T, Antonopoulos  AS, Katsimichas  A, Ohtani  T, Sakata  Y, Tousoulis  D. The intestinal microbiota and cardiovascular disease. Cardiovasc Res  2019;115:1471–1486. [DOI] [PubMed] [Google Scholar]
  • 45. Jonsson  AL, Bäckhed  F. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol  2017;14:79–87. [DOI] [PubMed] [Google Scholar]
  • 46. Sanchez-Rodriguez  E, Egea-Zorrilla  A, Plaza-Díaz  J, Aragón-Vela  J, Muñoz-Quezada  S, Tercedor-Sánchez  L, Abadia-Molina  F. The gut microbiota and its implication in the development of atherosclerosis and related cardiovascular diseases. Nutrients  2020;12:605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Melhem  NJ, Taleb  S. Tryptophan: from diet to cardiovascular diseases. Int J Mol Sci  2021;22:9904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Taleb  S. Tryptophan dietary impacts gut barrier and metabolic diseases. Front Immunol  2019;10:2113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Natividad  JM, Agus  A, Planchais  J, Lamas  B, Jarry  AC, Martin  R, Michel  ML, Chong-Nguyen  C, Roussel  R, Straube  M, Jegou  S, McQuitty  C, Le Gall  M, da Costa  G, Lecornet  E, Michaudel  C, Modoux  M, Glodt  J, Bridonneau  C, Sovran  B, Dupraz  L, Bado  A, Richard  ML, Langella  P, Hansel  B, Launay  JM, Xavier  RJ, Duboc  H, Sokol  H. Impaired aryl hydrocarbon receptor ligand production by the gut microbiota is a key factor in metabolic syndrome. Cell Metab  2018;28:737–749.e4. [DOI] [PubMed] [Google Scholar]
  • 50. Laurans  L, Venteclef  N, Haddad  Y, Chajadine  M, Alzaid  F, Metghalchi  S, Sovran  B, Denis  RGP, Dairou  J, Cardellini  M, Moreno-Navarrete  JM, Straub  M, Jegou  S, McQuitty  C, Viel  T, Esposito  B, Tavitian  B, Callebert  J, Luquet  SH, Federici  M, Fernandez-Real  JM, Burcelin  R, Launay  JM, Tedgui  A, Mallat  Z, Sokol  H, Taleb  S. Genetic deficiency of indoleamine 2,3-dioxygenase promotes gut microbiota-mediated metabolic health. Nat Med  2018;24:1113–1120. [DOI] [PubMed] [Google Scholar]
  • 51. Kimura  I, Ozawa  K, Inoue  D, Imamura  T, Kimura  K, Maeda  T, Terasawa  K, Kashihara  D, Hirano  K, Tani  T, Takahashi  T, Miyauchi  S, Shioi  G, Inoue  H, Tsujimoto  G. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nat Commun  2013;4:1829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Brown  AJ, Goldsworthy  SM, Barnes  AA, Eilert  MM, Tcheang  L, Daniels  D, Muir  AI, Wigglesworth  MJ, Kinghorn  I, Fraser  NJ, Pike  NB, Strum  JC, Steplewski  KM, Murdock  PR, Holder  JC, Marshall  FH, Szekeres  PG, Wilson  S, Ignar  DM, Foord  SM, Wise  A, Dowell  SJ. The orphan G protein-coupled receptors GPR41 and GPR43 are activated by propionate and other short chain carboxylic acids. J Biol Chem  2003;278:11312–11319. [DOI] [PubMed] [Google Scholar]
  • 53. Canfora  EE, Jocken  JW, Blaak  EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol  2015;11:577–591. [DOI] [PubMed] [Google Scholar]
  • 54. Furusawa  Y, Obata  Y, Fukuda  S, Endo  TA, Nakato  G, Takahashi  D, Nakanishi  Y, Uetake  C, Kato  K, Kato  T, Takahashi  M, Fukuda  NN, Murakami  S, Miyauchi  E, Hino  S, Atarashi  K, Onawa  S, Fujimura  Y, Lockett  T, Clarke  JM, Topping  DL, Tomita  M, Hori  S, Ohara  O, Morita  T, Koseki  H, Kikuchi  J, Honda  K, Hase  K, Ohno  H, Furusawa  Y, Obata  Y, Fukuda  S, Endo  TA, Nakato  G, Takahashi  D, Nakanishi  Y, Uetake  C, Kato  K, Kato  T, Takahashi  M, Fukuda  NN, Murakami  S, Miyauchi  E, Hino  S, Atarashi  K, Onawa  S, Fujimura  Y, Lockett  T, Clarke  JM, Topping  DL, Tomita  M, Hori  S, Ohara  O, Morita  T, Koseki  H, Kikuchi  J, Honda  K, Hase  K, Ohno  H. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature  2013;504:446–450. [DOI] [PubMed] [Google Scholar]
  • 55. Bartolomaeus  H, Balogh  A, Yakoub  M, Homann  S, Markó  L, Höges  S, Tsvetkov  D, Krannich  A, Wundersitz  S, Avery  EG, Haase  N, Kräker  K, Hering  L, Maase  M, Kusche-Vihrog  K, Grandoch  M, Fielitz  J, Kempa  S, Gollasch  M, Zhumadilov  Z, Kozhakhmetov  S, Kushugulova  A, Eckardt  KU, Dechend  R, Rump  LC, Forslund  SK, Müller  DN, Stegbauer  J, Wilck  N. Short-chain fatty acid propionate protects from hypertensive cardiovascular damage. Circulation  2019;139:1407–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Haghikia  A, Zimmermann  F, Schumann  P, Jasina  A, Roessler  J, Schmidt  D, Heinze  P, Kaisler  J, Nageswaran  V, Aigner  A, Ceglarek  U, Cineus  R, Hegazy  AN, van der Vorst  EPC, Döring  Y, Strauch  CM, Nemet  I, Tremaroli  V, Dwibedi  C, Kränkel  N, Leistner  DM, Heimesaat  MM, Bereswill  S, Rauch  G, Seeland  U, Soehnlein  O, Müller  DN, Gold  R, Bäckhed  F, Hazen  SL, Haghikia  A, Landmesser  U. Propionate attenuates atherosclerosis by immune-dependent regulation of intestinal cholesterol metabolism. Eur Heart J  2022;43:518–533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Velagapudi  VR, Hezaveh  R, Reigstad  CS, Gopalacharyulu  P, Yetukuri  L, Islam  S, Felin  J, Perkins  R, Borén  J, Orešič  M, Bäckhed  F. The gut microbiota modulates host energy and lipid metabolism in mice. J Lipid Res  2010;51:1101–1112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Le Chatelier  E, Nielsen  T, Qin  J, Prifti  E, Hildebrand  F, Falony  G, Almeida  M, Arumugam  M, Batto  JM, Kennedy  S, Leonard  P, Li  J, Burgdorf  K, Grarup  N, Jørgensen  T, Brandslund  I, Nielsen  HB, Juncker  AS, Bertalan  M, Levenez  F, Pons  N, Rasmussen  S, Sunagawa  S, Tap  J, Tims  S, Zoetendal  EG, Brunak  S, Clément  K, Doré  J, Kleerebezem  M, Kristiansen  K, Renault  P, Sicheritz-Ponten  T, de Vos  WM, Zucker  JD, Raes  J, Hansen  T, MetaHIT consortium, Bork  P, Wang  J, Ehrlich  SD, Pedersen  O.  Richness of human gut microbiome correlates with metabolic markers. Nature  2013;500:541–546. [DOI] [PubMed] [Google Scholar]
  • 59. Fu  J, Bonder  MJ, Cenit  MC, Tigchelaar  EF, Maatman  A, Dekens  JA, Brandsma  E, Marczynska  J, Imhann  F, Weersma  RK, Franke  L, Poon  TW, Xavier  RJ, Gevers  D, Hofker  MH, Wijmenga  C, Zhernakova  A. The gut microbiome contributes to a substantial proportion of the variation in blood lipids. Circ Res  2015;117:817–824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Wahlstrom  A, Sayin  SI, Marschall  HU, Backhed  F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab  2016;24:41–50. [DOI] [PubMed] [Google Scholar]
  • 61. Ott  SJ, El Mokhtari  NE, Musfeldt  M, Hellmig  S, Freitag  S, Rehman  A, Kühbacher  T, Nikolaus  S, Namsolleck  P, Blaut  M, Hampe  J, Sahly  H, Reinecke  A, Haake  N, Günther  R, Krüger  D, Lins  M, Herrmann  G, Fölsch  UR, Simon  R, Schreiber  S. Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation  2006;113:929–937. [DOI] [PubMed] [Google Scholar]
  • 62. Koren  O, Spor  A, Felin  J, Fåk  F, Stombaugh  J, Tremaroli  V, Behre  CJ, Knight  R, Fagerberg  B, Ley  RE, Bäckhed  F. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci USA  2011;108(Suppl. 1):4592–4598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. van den Munckhof  ICL, Kurilshikov  A, Ter Horst  R, Riksen  NP, Joosten  LAB, Zhernakova  A, Fu  J, Keating  ST, Netea  MG, de Graaf  J, Rutten  JHW. Role of gut microbiota in chronic low-grade inflammation as potential driver for atherosclerotic cardiovascular disease: a systematic review of human studies. Obes Rev  2018;19:1719–1734. [DOI] [PubMed] [Google Scholar]
  • 64. Laugerette  F, Vors  C, Géloën  A, Chauvin  MA, Soulage  C, Lambert-Porcheron  S, Peretti  N, Alligier  M, Burcelin  R, Laville  M, Vidal  H, Michalski  MC. Emulsified lipids increase endotoxemia: possible role in early postprandial low-grade inflammation. J Nutr Biochem  2011;22:53–59. [DOI] [PubMed] [Google Scholar]
  • 65. Erridge  C, Attina  T, Spickett  CM, Webb  DJ. A high-fat meal induces low-grade endotoxemia: evidence of a novel mechanism of postprandial inflammation. Am J Clin Nutr  2007;86:1286–1292. [DOI] [PubMed] [Google Scholar]
  • 66. Stoll  LL, Denning  GM, Weintraub  NL. Potential role of endotoxin as a proinflammatory mediator of atherosclerosis. Arterioscler Thromb Vasc Biol  2004;24:2227–2236. [DOI] [PubMed] [Google Scholar]
  • 67. Beli  E, Yan  Y, Moldovan  L, Vieira  CP, Gao  R, Duan  Y, Prasad  R, Bhatwadekar  A, White  FA, Townsend  SD, Chan  L, Ryan  CN, Morton  D, Moldovan  EG, Chu  FI, Oudit  GY, Derendorf  H, Adorini  L, Wang  XX, Evans-Molina  C, Mirmira  RG, Boulton  ME, Yoder  MC, Li  Q, Levi  M, Busik  JV, Grant  MB. Restructuring of the gut microbiome by intermittent fasting prevents retinopathy and prolongs survival in db/db mice. Diabetes  2018;67:1867–1879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Kain  V, Van Der Pol  W, Mariappan  N, Ahmad  A, Eipers  P, Gibson  DL, Gladine  C, Vigor  C, Durand  T, Morrow  C, Halade  GV. Obesogenic diet in aging mice disrupts gut microbe composition and alters neutrophil:lymphocyte ratio, leading to inflamed milieu in acute heart failure. FASEB J  2019;33:6456–6469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Virtue  AT, McCright  SJ, Wright  JM, Jimenez  MT, Mowel  WK, Kotzin  JJ, Joannas  L, Basavappa  MG, Spencer  SP, Clark  ML, Eisennagel  SH, Williams  A, Levy  M, Manne  S, Henrickson  SE, Wherry  EJ, Thaiss  CA, Elinav  E, Henao-Mejia  J. The gut microbiota regulates white adipose tissue inflammation and obesity via a family of microRNAs. Sci Transl Med  2019;11:eaav1892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. O'Donovan  AN, Herisson  FM, Fouhy  F, Ryan  PM, Whelan  D, Johnson  CN, Cluzel  G, Ross  RP, Stanton  C, Caplice  NM. Gut microbiome of a porcine model of metabolic syndrome and HF-pEF. Am J Physiol Heart Circ Physiol  2020;318:H590–H603. [DOI] [PubMed] [Google Scholar]
  • 71. Zoll  J, Read  MN, Heywood  SE, Estevez  E, Marshall  JPS, Kammoun  HL, Allen  TL, Holmes  AJ, Febbraio  MA, Henstridge  DC. Fecal microbiota transplantation from high caloric-fed donors alters glucose metabolism in recipient mice, independently of adiposity or exercise status. Am J Physiol Endocrinol Metab  2020;319:E203–E216. [DOI] [PubMed] [Google Scholar]
  • 72. Liu  H, Tian  R, Wang  H, Feng  S, Li  H, Xiao  Y, Luan  X, Zhang  Z, Shi  N, Niu  H, Zhang  S. Gut microbiota from coronary artery disease patients contributes to vascular dysfunction in mice by regulating bile acid metabolism and immune activation. J Transl Med  2020;18:382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Kiouptsi  K, Jäckel  S, Pontarollo  G, Grill  A, Kuijpers  MJE, Wilms  E, Weber  C, Sommer  F, Nagy  M, Neideck  C, Jansen  Y, Ascher  S, Formes  H, Karwot  C, Bayer  F, Kollar  B, Subramaniam  S, Molitor  M, Wenzel  P, Rosenstiel  P, Todorov  H, Gerber  S, Walter  U, Jurk  K, Heemskerk  JWM, van der Vorst  EPC, Döring  Y, Reinhardt  C. The microbiota promotes arterial thrombosis in low-density lipoprotein receptor-deficient mice. mBio  2019;10:02298-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Brandsma  E, Kloosterhuis  NJ, Koster  M, Dekker  DC, Gijbels  MJJ, van der Velden  S, Ríos-Morales  M, van Faassen  MJR, Loreti  MG, de Bruin  A, Fu  J, Kuipers  F, Bakker  BM, Westerterp  M, de Winther  MPJ, Hofker  MH, van de Sluis  B, Koonen  DPY. A proinflammatory gut microbiota increases systemic inflammation and accelerates atherosclerosis. Circ Res  2019;124:94–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Wu  ZX, Li  SF, Chen  H, Song  JX, Gao  YF, Zhang  F, Cao  CF. The changes of gut microbiota after acute myocardial infarction in rats. PLoS One  2017;12:e0180717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Lambert  G, Amar  MJ, Guo  G, Brewer  HB  Jr, Gonzalez  FJ, Sinal  CJ. The Farnesoid X-receptor is an essential regulator of cholesterol homeostasis. J Biol Chem  2003;278:2563–2570. [DOI] [PubMed] [Google Scholar]
  • 77. Hanniman  EA, Lambert  G, McCarthy  TC, Sinal  CJ. Loss of functional Farnesoid X receptor increases atherosclerotic lesions in apolipoprotein E-deficient mice. J Lipid Res  2005;46:2595–2604. [DOI] [PubMed] [Google Scholar]
  • 78. Gregory  JC, Buffa  JA, Org  E, Wang  Z, Levison  BS, Zhu  W, Wagner  MA, Bennett  BJ, Li  L, DiDonato  JA, Lusis  AJ, Hazen  SL. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem  2015;290:5647–5660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Zhu  W, Gregory  JC, Org  E, Buffa  JA, Gupta  N, Wang  Z, Li  L, Fu  X, Wu  Y, Mehrabian  M, Sartor  RB, McIntyre  TM, Silverstein  RL, Tang  WHW, DiDonato  JA, Brown  JM, Lusis  AJ, Hazen  SL. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell  2016;165:111–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Latsios  G, Saetta  A, Michalopoulos  NV, Agapitos  E, Patsouris  E. Detection of Cytomegalovirus, Helicobacter pylori and Chlamydia pneumoniae DNA in carotid atherosclerotic plaques by the polymerase chain reaction. Acta Cardiol  2004;59:652–657. [DOI] [PubMed] [Google Scholar]
  • 81. Karlsson  FH, Fåk  F, Nookaew  I, Tremaroli  V, Fagerberg  B, Petranovic  D, Bäckhed  F, Nielsen  J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun  2012;3:1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Emoto  T, Yamashita  T, Sasaki  N, Hirota  Y, Hayashi  T, So  A, Kasahara  K, Yodoi  K, Matsumoto  T, Mizoguchi  T, Ogawa  W, Hirata  K. Analysis of gut microbiota in coronary artery disease patients: a possible link between gut microbiota and coronary artery disease. J Atheroscler Thromb  2016;23:908–921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Yin  J, Liao  SX, He  Y, Wang  S, Xia  GH, Liu  FT, Zhu  JJ, You  C, Chen  Q, Zhou  L, Pan  SY, Zhou  HW. Dysbiosis of gut microbiota with reduced trimethylamine-N-oxide level in patients with large-artery atherosclerotic stroke or transient ischemic attack. J Am Heart Assoc  2015;4:pii:e002699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Jie  Z, Xia  H, Zhong  SL, Feng  Q, Li  S, Liang  S, Zhong  H, Liu  Z, Gao  Y, Zhao  H, Zhang  D, Su  Z, Fang  Z, Lan  Z, Li  J, Xiao  L, Li  J, Li  R, Li  X, Li  F, Ren  H, Huang  Y, Peng  Y, Li  G, Wen  B, Dong  B, Chen  JY, Geng  QS, Zhang  ZW, Yang  H, Wang  J, Wang  J, Zhang  X, Madsen  L, Brix  S, Ning  G, Xu  X, Liu  X, Hou  Y, Jia  H, He  K, Kristiansen  K. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun  2017;8:845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Cui  L, Zhao  T, Hu  H, Zhang  W, Hua  X. Association study of gut flora in coronary heart disease through high-throughput sequencing. Biomed Res Int  2017;2017:3796359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Zhu  Q, Gao  R, Zhang  Y, Pan  D, Zhu  Y, Zhang  X, Yang  R, Jiang  R, Xu  Y, Qin  H. Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol Genomics  2018;50:893–903. [DOI] [PubMed] [Google Scholar]
  • 87. Liu  H, Chen  X, Hu  X, Niu  H, Tian  R, Wang  H, Pang  H, Jiang  L, Qiu  B, Chen  X, Zhang  Y, Ma  Y, Tang  S, Li  H, Feng  S, Zhang  S, Zhang  C. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome  2019;7:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Toya  T, Corban  MT, Marrietta  E, Horwath  IE, Lerman  LO, Murray  JA, Lerman  A. Coronary artery disease is associated with an altered gut microbiome composition. PLoS One  2020;15:e0227147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Awoyemi  A, Trøseid  M, Arnesen  H, Solheim  S, Seljeflot  I. Effects of dietary intervention and n-3 PUFA supplementation on markers of gut-related inflammation and their association with cardiovascular events in a high-risk population. Atherosclerosis  2019;286:53–59. [DOI] [PubMed] [Google Scholar]
  • 90. Pastori  D, Carnevale  R, Nocella  C, Novo  M, Santulli  M, Cammisotto  V, Menichelli  D, Pignatelli  P, Violi  F. Gut-derived serum lipopolysaccharide is associated with enhanced risk of major adverse cardiovascular events in atrial fibrillation: Effect of adherence to Mediterranean diet. J Am Heart Assoc  2017;6:e005784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Schiattarella  GG, Sannino  A, Toscano  E, Giugliano  G, Gargiulo  G, Franzone  A, Trimarco  B, Esposito  G, Perrino  C. Gut microbe-generated metabolite trimethylamine-N-oxide as cardiovascular risk biomarker: a systematic review and dose-response meta-analysis. Eur Heart J  2017;38:2948–2956. [DOI] [PubMed] [Google Scholar]
  • 92. 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 Assoc  2017;6:e004947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Senthong  V, Li  XS, Hudec  T, Coughlin  J, Wu  Y, Levison  B, Wang  Z, Hazen  SL, Tang  WH. Plasma trimethylamine N-oxide, a gut microbe-generated phosphatidylcholine metabolite, is associated with atherosclerotic burden. J Am Coll Cardiol  2016;67:2620–2628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Li  XS, Obeid  S, Wang  Z, Hazen  BJ, Li  L, Wu  Y, Hurd  AG, Gu  X, Pratt  A, Levison  BS, Chung  YM, Nissen  SE, Tang  WHW, Mach  F, Räber  L, Nanchen  D, Matter  CM, Lüscher  TF, Hazen  SL. Trimethyllysine, a trimethylamine N-oxide precursor, provides near- and long-term prognostic value in patients presenting with acute coronary syndromes. Eur Heart J  2019;40:2700–2709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Mueller  DM, Allenspach  M, Othman  A, Saely  CH, Muendlein  A, Vonbank  A, Drexel  H, von Eckardstein  A. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis  2015;243:638–644. [DOI] [PubMed] [Google Scholar]
  • 96. Aldana-Hernández  P, Leonard  KA, Zhao  YY, Curtis  JM, Field  CJ, Jacobs  RL. Dietary choline or trimethylamine N-oxide supplementation does not influence atherosclerosis development in Ldlr−/− and Apoe−/− male mice. J Nutr  2020;150:249–255. [DOI] [PubMed] [Google Scholar]
  • 97. Skagen  K, Trøseid  M, Ueland  T, Holm  S, Abbas  A, Gregersen  I, Kummen  M, Bjerkeli  V, Reier-Nilsen  F, Russell  D, Svardal  A, Karlsen  TH, Aukrust  P, Berge  RK, Hov  JE, Halvorsen  B, Skjelland  M. The Carnitine-butyrobetaine-trimethylamine-N-oxide pathway and its association with cardiovascular mortality in patients with carotid atherosclerosis. Atherosclerosis  2016;247:64–69. [DOI] [PubMed] [Google Scholar]
  • 98. Meyer  KA, Benton  TZ, Bennett  BJ, Jacobs  DR  Jr, Lloyd-Jones  DM, Gross  MD, Carr  JJ, Gordon-Larsen  P, Zeisel  SH. Microbiota-dependent metabolite trimethylamine N-oxide and coronary artery calcium in the coronary artery risk development in young adults study (CARDIA). J Am Heart Assoc  2016;5:e003970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Papandreou  C, Moré  M, Bellamine  A. Trimethylamine N-oxide in relation to cardiometabolic health-cause or effect?  Nutrients  2020;12:1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Jia  J, Dou  P, Gao  M, Kong  X, Li  C, Liu  Z, Huang  T. Assessment of causal direction between gut microbiota-dependent metabolites and cardiometabolic health: A bidirectional Mendelian randomization analysis. Diabetes  2019;68:1747–1755. [DOI] [PubMed] [Google Scholar]
  • 101. Sánchez  B, Delgado  S, Blanco-Míguez  A, Lourenço  A, Gueimonde  M, Margolles  A. Probiotics, gut microbiota, and their influence on host health and disease. Mol Nutr Food Res  2017;61:1600240. [DOI] [PubMed] [Google Scholar]
  • 102. Thushara  RM, Gangadaran  S, Solati  Z, Moghadasian  MH. Cardiovascular benefits of probiotics: a review of experimental and clinical studies. Food Funct  2016;7:632–642. [DOI] [PubMed] [Google Scholar]
  • 103. O’Morain  VL, Ramji  DP. The potential of probiotics in the prevention and treatment of atherosclerosis. Mol Nutr Food Res  2020;64:e1900797. [DOI] [PubMed] [Google Scholar]
  • 104. Gan  XT, Ettinger  G, Huang  CX, Burton  JP, Haist  JV, Rajapurohitam  V, Sidaway  JE, Martin  G, Gloor  GB, Swann  JR, Reid  G, Karmazyn  M. Probiotic administration attenuates myocardial hypertrophy and heart failure after myocardial infarction in the rat. Circ Heart Fail  2014;7:491–499. [DOI] [PubMed] [Google Scholar]
  • 105. Yoshida  N, Emoto  T, Yamashita  T, Watanabe  H, Hayashi  T, Tabata  T, Hoshi  N, Hatano  N, Ozawa  G, Sasaki  N, Mizoguchi  T, Amin  HZ, Hirota  Y, Ogawa  W, Yamada  T, Hirata  KI. Bacteroides vulgatus and Bacteroides dorei reduce gut microbial lipopolysaccharide production and inhibit atherosclerosis. Circulation  2018;138:2486–2498. [DOI] [PubMed] [Google Scholar]
  • 106. Rondanelli  M, Faliva  MA, Perna  S, Giacosa  A, Peroni  G, Castellazzi  AM. Using probiotics in clinical practice: where are we now? A review of existing meta-analyses. Gut Microbes  2017;8:521–543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. 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 Funct  2018;9:4299–4309. [DOI] [PubMed] [Google Scholar]
  • 108. Quigley  EM. Prebiotics and probiotics in digestive health. Clin Gastroenterol Hepatol  2019;17:333–344. [DOI] [PubMed] [Google Scholar]
  • 109. Santos-Marcos  JA, Perez-Jimenez  F, Camargo  A. The role of diet and intestinal microbiota in the development of metabolic syndrome. J Nutr Biochem  2019;70:1–27. [DOI] [PubMed] [Google Scholar]
  • 110. Levy  M, Kolodziejczyk  AA, Thaiss  CA, Elinav  E. Dysbiosis and the immune system. Nat Rev Immunol  2017;17:219–232. [DOI] [PubMed] [Google Scholar]
  • 111. Sáez-Lara  MJ, Robles-Sanchez  C, Ruiz-Ojeda  FJ, Plaza-Diaz  J, Gil  A. Effects of probiotics and synbiotics on obesity, insulin resistance syndrome, type 2 diabetes and non-alcoholic fatty liver disease: A review of human clinical trials. Int J Mol Sci  2016;17:928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Tajabadi-Ebrahimi  M, Sharifi  N, Farrokhian  A, Raygan  F, Karamali  F, Razzaghi  R, Taheri  S, Asemi  Z. A randomized controlled clinical trial investigating the effect of synbiotic administration on markers of insulin metabolism and lipid profiles in overweight type 2 diabetic patients with coronary heart disease. Exp Clin Endocrinol Diabetes  2017;125:21–27. [DOI] [PubMed] [Google Scholar]
  • 113. Nikbakht  E, Khalesi  S, Singh  I, Williams  LT, West  NP, Colson  N. Effect of probiotics and synbiotics on blood glucose: a systematic review and meta-analysis of controlled trials. Eur J Nutr  2018;57:95–106. [DOI] [PubMed] [Google Scholar]
  • 114. Ooi  LG, Ahmad  R, Yuen  KH, Liong  MT. Lactobacillus gasseri [corrected] CHO-220 and inulin reduced plasma total cholesterol and low-density lipoprotein cholesterol via alteration of lipid transporters. J Dairy Sci  2010;93:5048–5058. [DOI] [PubMed] [Google Scholar]
  • 115. Shakeri  H, Hadaegh  H, Abedi  F, Tajabadi-Ebrahimi  M, Mazroii  N, Ghandi  Y, Asemi  Z. Consumption of synbiotic bread decreases triacylglycerol and VLDL levels while increasing HDL levels in serum from patients with type-2 diabetes. Lipids  2014;49:695–701. [DOI] [PubMed] [Google Scholar]
  • 116. Swanson  KS, Gibson  GR, Hutkins  R, Reimer  RA, Reid  G, Verbeke  K, Scott  KP, Holscher  HD, Azad  MB, Delzenne  NM, Sanders  ME. The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of Synbiotics. Nat Rev Gastroenterol Hepatol  2020;17:687–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Taylor-Robinson  D, Boman  J. The failure of antibiotics to prevent heart attacks. BMJ  2005;331:361–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Kromhout  D, Keys  A, Aravanis  C, Buzina  R, Fidanza  F, Giampaoli  S, Jansen  A, Menotti  A, Nedeljkovic  S, Pekkarinen  M. Food consumption patterns in the 1960s in seven countries. Am J Clin Nutr  1989;49:889–894. [DOI] [PubMed] [Google Scholar]
  • 119. Liu  S, Willett  WC, Stampfer  MJ, Hu  FB, Franz  M, Sampson  L, Hennekens  CH, Manson  JE. A prospective study of dietary glycemic load, carbohydrate intake, and risk of coronary heart disease in US women. Am J Clin Nutr  2000;71:1455–1461. [DOI] [PubMed] [Google Scholar]
  • 120. Menotti  A, Puddu  PE. How the Seven Countries Study contributed to the definition and development of the Mediterranean diet concept: a 50-year journey. Nutr Metab Cardiovasc Dis  2015;25:245–252. [DOI] [PubMed] [Google Scholar]
  • 121. Davis  CR, Hodgson  JM, Woodman  R, Bryan  J, Wilson  C, Murphy  KJ. A Mediterranean diet lowers blood pressure and improves endothelial function: results from the MedLey randomized intervention trial. Am J Clin Nutr  2017;105:1305–1313. [DOI] [PubMed] [Google Scholar]
  • 122. de Lorgeril  M, Salen  P, Martin  JL, Monjaud  I, Delaye  J, Mamelle  N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: final report of the Lyon Diet Heart Study. Circulation  1999;99:779–785. [DOI] [PubMed] [Google Scholar]
  • 123. Mattioli  AV, Palmiero  P, Manfrini  O, Puddu  PE, Nodari  S, Dei Cas  A, Mercuro  G, Scrutinio  D, Palermo  P, Sciomer  S, Di Francesco  S, Novo  G, Novo  S, Pedretti  RFE, Zito  A, Parati  G, Pedrinelli  R, Farinetti  A, Maiello  M, Moscucci  F, Tenaglia  RL, Sucato  V, Triggiani  M, Cugusi  L, Scicchitano  P, Saba  PS, Ciccone  MM. Mediterranean diet impact on cardiovascular diseases: a narrative review. J Cardiovasc Med (Hagerstown)  2017;18:925–935. [DOI] [PubMed] [Google Scholar]
  • 124. Estruch  R, Ros  E, Salas-Salvadó  J, Covas  MI, Corella  D, Arós  F, Gómez-Gracia  E, Ruiz-Gutiérrez  V, Fiol  M, Lapetra  J, Lamuela-Raventos  RM, Serra-Majem  L, Pintó  X, Basora  J, Muñoz  MA, Sorlí  JV, Martínez  JA, Fitó  M, Gea  A, Hernán  MA, Martínez-González  MA; PREDIMED Study Investigators . Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. N Engl J Med  2018;378:e34. [DOI] [PubMed] [Google Scholar]
  • 125. Gerdes  V, Gueimonde  M, Pajunen  L, Nieuwdorp  M, Laitinen  K. How strong is the evidence that gut microbiota composition can be influenced by lifestyle interventions in a cardio-protective way?  Atherosclerosis  2020;311:124–142. [DOI] [PubMed] [Google Scholar]
  • 126. Krznarić  Ž, Vranešić Bender  D, Meštrović  T. The Mediterranean diet and its association with selected gut bacteria. Curr Opin Clin Nutr Metab Care  2019;22:401–406. [DOI] [PubMed] [Google Scholar]
  • 127. Garcia-Mantrana  I, Selma-Royo  M, Alcantara  C, Collado  MC. Shifts on gut microbiota associated to Mediterranean diet adherence and specific dietary intakes on general adult population. Front Microbiol  2018;9:890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. O’Keefe  SJ, Li  JV, Lahti  L, Ou  J, Carbonero  F, Mohammed  K, Posma  JM, Kinross  J, Wahl  E, Ruder  E, Vipperla  K, Naidoo  V, Mtshali  L, Tims  S, Puylaert  PG, DeLany  J, Krasinskas  A, Benefiel  AC, Kaseb  HO, Newton  K, Nicholson  JK, de Vos  WM, Gaskins  HR, Zoetendal  EG. Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun  2015;6:6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Lam  YY, Ha  CW, Campbell  CR, Mitchell  AJ, Dinudom  A, Oscarsson  J, Cook  DI, Hunt  NH, Caterson  ID, Holmes  AJ, Storlien  LH. Increased gut permeability and microbiota change associate with mesenteric fat inflammation and metabolic dysfunction in diet-induced obese mice. PLoS One  2012;7:e34233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Allen  JM, Mailing  LJ, Niemiro  GM, Moore  R, Cook  MD, White  BA, Holscher  HD, Woods  JA. Exercise alters gut microbiota composition and function in lean and obese humans. Med Sci Sports Exerc  2018;50:747–757. [DOI] [PubMed] [Google Scholar]
  • 131. Moayyedi  P, Yuan  Y, Baharith  H, Ford  AC. Faecal microbiota transplantation for Clostridium difficile-associated diarrhoea: a systematic review of randomised controlled trials. Med J Aust  2017;207:166–172. [DOI] [PubMed] [Google Scholar]
  • 132. Vrieze  A, Van Nood  E, Holleman  F, Salojärvi  J, Kootte  RS, Bartelsman  JF, Dallinga-Thie  GM, Ackermans  MT, Serlie  MJ, Oozeer  R, Derrien  M, Druesne  A, Van Hylckama Vlieg  JE, Bloks  VW, Groen  AK, Heilig  HG, Zoetendal  EG, Stroes  ES, de Vos  WM, Hoekstra  JB, Nieuwdorp  M. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology  2012;143:913–916.e7. [DOI] [PubMed] [Google Scholar]
  • 133. Duan  FF, Liu  JH, March  JC. Engineered commensal bacteria reprogram intestinal cells into glucose-responsive insulin-secreting cells for the treatment of diabetes. Diabetes  2015;64:1794–1803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Organ  CL, Li  Z, Sharp  TE  III, Polhemus  DJ, Gupta  N, Goodchild  TT, Tang  WHW, Hazen  SL, Lefer  DJ.  Nonlethal inhibition of gut microbial trimethylamine N-oxide production improves cardiac function and remodeling in a murine model of heart failure. J Am Heart Assoc  2020;9:e016223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Roberts  AB, Gu  X, Buffa  JA, Hurd  AG, Wang  Z, Zhu  W, Gupta  N, Skye  SM, Cody  DB, Levison  BS, Barrington  WT, Russell  MW, Reed  JM, Duzan  A, Lang  JM, Fu  X, Li  L, Myers  AJ, Rachakonda  S, DiDonato  JA, Brown  JM, Gogonea  V, Lusis  AJ, Garcia-Garcia  JC, Hazen  SL. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med  2018;24:1407–1417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Wang  Z, Roberts  AB, Buffa  JA, Levison  BS, Zhu  W, Org  E, Gu  X, Huang  Y, Zamanian-Daryoush  M, Culley  MK, DiDonato  AJ, Fu  X, Hazen  JE, Krajcik  D, DiDonato  JA, Lusis  AJ, Hazen  SL. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell  2015;163:1585–1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Steinke  I, Ghanei  N, Govindarajulu  M, Yoo  S, Zhong  J, Amin  RH. Drug discovery and development of novel therapeutics for inhibiting TMAO in models of atherosclerosis and diabetes. Front Physiol  2020;11:567899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Bartolomaeus  TUP, Birkner  T, Bartolomaeus  H, Löber  U, Avery  EG, Mähler  A, Weber  D, Kochlik  B, Balogh  A, Wilck  N, Boschmann  M, Müller  DN, Markó  L, Forslund  SK. Quantifying technical confounders in microbiome studies. Cardiovasc Res  2021;117:863–875. [DOI] [PubMed] [Google Scholar]
  • 139. Wesolowska-Andersen  A, Bahl  MI, Carvalho  V, Kristiansen  K, Sicheritz-Pontén  T, Gupta  R, Licht  TR. Choice of bacterial DNA extraction method from fecal material influences community structure as evaluated by metagenomic analysis. Microbiome  2014;2:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Forslund  K, Hildebrand  F, Nielsen  T, Falony  G, Le Chatelier  E, Sunagawa  S, Prifti  E, Vieira-Silva  S, Gudmundsdottir  V, Pedersen  HK, Arumugam  M, Kristiansen  K, Voigt  AY, Vestergaard  H, Hercog  R, Costea  PI, Kultima  JR, Li  J, Jørgensen  T, Levenez  F, Dore  J, Nielsen  HB, Brunak  S, Raes  J, Hansen  T, Wang  J, Ehrlich  SD, Bork  P, Pedersen  O. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature  2015;528:262–266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Borgo  F, Garbossa  S, Riva  A, Severgnini  M, Luigiano  C, Benetti  A, Pontiroli  AE, Morace  G, Borghi  E. Body mass index and sex affect diverse microbial niches within the gut. Front Microbiol  2018;9:213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Li  J, Jia  H, Cai  X, Zhong  H, Feng  Q, Sunagawa  S, Arumugam  M, Kultima  JR, Prifti  E, Nielsen  T, Juncker  AS, Manichanh  C, Chen  B, Zhang  W, Levenez  F, Wang  J, Xu  X, Xiao  L, Liang  S, Zhang  D, Zhang  Z, Chen  W, Zhao  H, Al-Aama  JY, Edris  S, Yang  H, Wang  J, Hansen  T, Nielsen  HB, Brunak  S, Kristiansen  K, Guarner  F, Pedersen  O, Doré  J, Ehrlich  SD, Bork  P, Wang  J. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol  2014;32:834–841. [DOI] [PubMed] [Google Scholar]
  • 143. Weitsman  S, Celly  S, Leite  G, Mathur  R, Sedighi  R, Barlow  GM, Morales  W, Sanchez  M, Parodi  G, Villanueva-Millan  MJ, Rezaie  A, Pimentel  M. Effects of proton pump inhibitors on the small bowel and stool microbiomes. Dig Dis Sci  2022;67:224–232. [DOI] [PubMed] [Google Scholar]
  • 144. Imhann  F, Bonder  MJ, Vich Vila  A, Fu  J, Mujagic  Z, Vork  L, Tigchelaar  EF, Jankipersadsing  SA, Cenit  MC, Harmsen  HJ, Dijkstra  G, Franke  L, Xavier  RJ, Jonkers  D, Wijmenga  C, Weersma  RK, Zhernakova  A. Proton pump inhibitors affect the gut microbiome. Gut  2016;65:740–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Nguyen  TL, Vieira-Silva  S, Liston  A, Raes  J. How informative is the mouse for human gut microbiota research?  Dis Model Mech  2015;8:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Patel  J. The gut microbiome: a novel cardio-metabolic target?  Cardiovasc Res  2019;115:82–84. [DOI] [PubMed] [Google Scholar]
  • 147. Chen  YY, Chen  DQ, Chen  L, Liu  JR, Vaziri  ND, Guo  Y, Zhao  YY. Microbiome-metabolome reveals the contribution of gut-kidney axis on kidney disease. J Transl Med  2019;17:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Tang  WH, Kitai  T, Hazen  SL. Gut microbiota in cardiovascular health and disease. Circ Res  2017;120:1183–1196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Tang  WH, Wang  Z, Levison  BS, Koeth  RA, Britt  EB, Fu  X, Wu  Y, Hazen  SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med  2013;368:1575–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Senthong  V, Wang  Z, Fan  Y, Wu  Y, Hazen  SL, Tang  WH. Trimethylamine N-oxide and mortality risk in patients with peripheral artery disease. J Am Heart Assoc  2016;5:e004237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Tang  WH, Wang  Z, Fan  Y, Levison  B, Hazen  JE, Donahue  LM, Wu  Y, Hazen  SL. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol  2014;64:1908–1914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Li  W, Huang  A, Zhu  H, Liu  X, Huang  X, Huang  Y, Cai  X, Lu  J, Huang  Y. Gut microbiota-derived trimethylamine N-oxide is associated with poor prognosis in patients with heart failure. Med J Aust  2020;213:374–379. [DOI] [PubMed] [Google Scholar]
  • 153. Zhu  W, Wang  Z, Tang  WHW, Hazen  SL. Gut microbe-generated trimethylamine N-oxide from dietary choline is prothrombotic in subjects. Circulation  2017;135:1671–1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Gencer  B, Li  XS, Gurmu  Y, Bonaca  MP, Morrow  DA, Cohen  M, Bhatt  DL, Steg  PG, Storey  RF, Johanson  P, Wang  Z, Hazen  SL, Sabatine  MS. Gut microbiota-dependent trimethylamine N-oxide and cardiovascular outcomes in patients with prior myocardial infarction: a nested case control study from the PEGASUS-TIMI 54 trial. J Am Heart Assoc  2020;9:e015331. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cardiovascular Research are provided here courtesy of Oxford University Press

RESOURCES