Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 May 1.
Published in final edited form as: Curr Opin Cardiol. 2020 May;35(3):207–218. doi: 10.1097/HCO.0000000000000720

Gut microbiome and cardiovascular disease

Yongzhong Zhao 1, Zeneng Wang 1,2
PMCID: PMC7442318  NIHMSID: NIHMS1601803  PMID: 32068612

Abstract

Purpose of the review

This review aims to highlight the association between gut microbiome and cardiovascular disease (CVD) with emphasis on the possible molecular mechanisms by which how gut microbiome contributes to CVD.

Recent findings

Increasingly, the roles of gut microbiome in cardiovascular health and disease have gained much attention. Most of the investigations focus on how the gut dysbiosis contributes to CVD risk factors and which gut microbial derived metabolites mediate such effects.

Summary

In this review, we discuss the molecular mechanisms of gut microbiome contributing to CVD, which include gut microbes translocalization to aortic artery due to gut barrier defect to initiate inflammation and microbial derived metabolites inducing inflammation signaling pathway and renal insufficiency. Specifically, we categorize beneficial and deleterious microbial derived metabolites in cardiovascular health. We also summarize recent findings in the gut microbiome modulation of drug efficacy in treatment of CVD and the microbiome mechanisms by which how physical exercise ameliorates cardiovascular health. Gut microbiome has become an essential component of cardiovascular research and a crucial consideration factor in cardiovascular health and disease.

Keywords: Cardiovascular disease (CVD), gut microbiome, microbial metabolites, drug efficacy, physical exercise

INTRODUCTION

Taxonomically well-structured and fine-functional human gut microbiome is essential to human health, such as helping digestion of dietary polysaccharides that enzymes of the host cannot breakdown [1], inducing and training the host immune system [2], producing vitamins (B, K) [3*,4] and maintaining intestinal barrier [5] as well. In healthy humans, commensal and potentially pathogenic bacteria are in a homeostatic balance [6]. Gut microbiome dysbiosis, the condition of dysregulated and disrupted intestinal bacterial homeostasis, is associated with an array of complex diseases such as inflammatory bowel disease (IBD), obesity, type 1 and type 2 diabetes, cardiovascular disease (CVD), autism, amyotrophic lateral sclerosis, Parkinson’s disease, Huntington’s disease, rheumatic disease and certain gastrointestinal cancers [6-8, 9*, 10*, 11, 12*]. Remarkably, roles of gut microbiome in CVD have gained much attention, given that CVD are the leading cause of mortality and morbidity. Differences in gut microbiota community between patients with CVD and healthy controls have been investigated in several groups worldwide and Table 1. lists some gut microbiota community shifts with different CVD phenotypes.

Table 1.

Gut microbiome and cardiovascular disease.

Cardiovascular disease phenotype Microbiota community shift References
hypertension Prevotella and Klebsiella [13]
heart failure Faecalibacterium prausnitziiRuminococcus gnavus [14]
coronary artery disease Lactobacillales, Escherichia-Shigella and EnterococcusFaecalibacterium, Subdoligranulum, Roseburia and Eubacterium rectale, Bacteroidetes↓ [15,16]
ischemic stroke Atopobium cluster and Lactobacillus ruminisLactobacillus sakei [17]
atrial fibrillation Ruminococcus, Streptococcus and EnterococcusFaecalibacterium, Alistipes, Oscillibacter and Bilophila [18]
atherosclerotic cardiovascular disease Enterobacteriaceae:Escherichia coli, Klebsiella spp., Enterobacter aerogenes, Streptococcus spp.,Lactobacillus salivarius, Solobacterium moorei, Atopobium parvulum, Ruminococcus gnavus, and Eggerthella lentaRoseburia intestinalis, Faecalibacterium cf. prausnitzii, Bacteroides spp., Prevotella copri, andAlistipes shahii [19]

Supplementing to those insightful reviews on gut microbiome in CVD [20**, 21**, 22, 23**, 24, 25], here we focus on mechanisms by which and how gut microbiome shapes cardiovascular health and disease, underscoring gut microbiota derived metabolites, the modulatory effect of gut microbiome to cardiovascular drug efficacy and toxicity, the beneficial role of physical exercises in cardiovascular health via modulating the taxonomic composition and function of the human gut microbiome. Uncovering gut microbiome-CVD mechanisms and translating such knowledge into clinical practice are anticipated as primary priority in CVD research. Individual human gut microbiome, drug targets alongside companion and complementary diagnostics, are keys to precision medicine.

Gut barrier defect provides a pathway for gut microbes to inhabit the aortic artery

Gut barrier, comprised of several layers, including the physical barrier composed of gut microbiota, mucus, epithelial cells and the innate and adaptive immune cells [26], plays an important role in health and disease. Gut barrier can prevent bacterium entering circulatory system and the defects have been shown to be associated with gastrointestinal disease (e.g. celiac disease (CeD), inflammatory bowel disease (IBD), colon carcinoma), chronic liver disease, type 1 diabetes, obesity and food allergies [27,28]. Bacterium can be detected in human atherosclerotic plaque of the patients with periodontal disease [29]. However, some bacterium species in atherosclerotic plaque cannot be found in mouth, but can be found in feces, suggesting that gut microbes can also contribute to the atherosclerotic plaque microbial diversity [30]. Gut bacteria enter circulatory system due to gut barrier defect, leading to increased intestinal permeability thereby favoring translocation of gut microbes [31].

A study on 28 patients undergone the carotid endarterectomy by 16S rRNA gene sequencing confirmed that the most abundant bacterium is Proteobacteria alongside three other main phyla, Actinobacteria, Bacteroidetes and Firmicutes found in aortic plaque [32], indicating a large difference from the gut and could be due to environmental discrimination between gut and blood. Bacterium infection can directly drive atherosclerosis. Using B6 Apoeshl mouse model with oral infection of H. cinaedi, the investigators found that H. cinaedi can induce advanced atherosclerotic lesion development by altered expression of cholesterol receptors or transporters and by increased proinflammatory cytokines’ expression. These molecular events result in macrophage and neutrophil accumulation, leading to foam cell formation in atherosclerotic lesions [33].

The gut barrier dysfunction and the subsequent increase of intestinal permeability facilitates the translocation of lipopolysaccharide (LPS), also called as endotoxin, from gut lumen to circulatory blood system. LPS is the outer membrane component of Gram negative bacterium, which protects bacterium from the entry of many noxious compounds [34, 35]. Gram-negative bacteria produce outer membrane vesicles (OMVs) that contain LPS [36]. LPS binds TLR4 to activate NF-κB signaling, leading to overproduction of proinflammatory cytokines and adhesion molecules, therefore resulting in sepsis and atherosclerosis [31, 37]. Besides LPS/TLR4/NF-κB signaling pathway, LPS can also be internalized to cytosol through endocytosis and released into the cytosol, which activates caspase-11, leading to further activation of the NLRP3 inflammasome [38]. The activation of NLRP3 inflammasome leads to caspase-1 activation and IL-1β and IL-18 secretion, which is a key step in the inflammatory process of atherosclerosis [39, 40].

On the other hand, CVD can also induce gut barrier defect, which further exaggerates CVD. Thus, a vicious pathophysiology loop can emerge. An aortic dissection happened in the aorta is characterized by a tear in the inner layer of artery wall, allowing blood to enter into the wall, creating a new passage for blood, known as the “false lumen” [41]. Aortic dissection induces intestinal ischemia and intestinal epithelial barrier dysfunction, thereby leading to the translocation of gut bacteria to the bowel wall and bloodstream, further leading to septic shock [42].

Dietary fiber shows benefit to human health after fermentation by gut microbiota, e.g., the product butyric acid with a function of improving gut barrier [4345]. Consistently, mice deprived of dietary fiber showed greater epithelial access and lethal colitis [46]. Some nutraceuticals, such as resveratrol, berberine, have been used for clinical trial to treat CVD [47], as preclinical models had shown that these nutraceuticals can improve gut barrier both in vitro cell culture and in vivo animal models[4850]. Meanwhile, some probiotics, such as Akkermansia muciniphila, can improve gut barrier by modulation of mucus layer thickness [51, 52*], which is consistent to its protective effect against atherosclerosis [53].

Gut microbiota derived metabolites and cardiovascular health and disease

In health individuals, gut barrier defects happen only occasionally. Thus, in most cases, gut microbiota leading to CVD is mediated by metabolites. Over the past decades a lot of gut microbial metabolites have received increasing attention. We summarized gut microbiota derived metabolites, related bacteria, target cells of the host, CVD type, putative molecular mechanisms and reference as shown in Table 2.

Table 2.

Gut microbiota derived metabolites and cardiovascular health and disease

Metabolite Substrate Gene/enzyme Bacteria Biological Effects Host target Disease Ref.
esculetin esculin NA Enterococcus faecalis, Selenomonas ruminantium 2358, et al. ↑NO aortic endothelial cell atherosclerosis 56-57
protocatechuic acid anthocyanin NA NA ↓miR-10b, ↑ABCA1 and ABCG1 macrophage atherosclerosis 58-61
urolithin ellagitannin NA Gordonibacter urolithinfaciens sp. nov. and Bifidobacterium seudocatenulatum INIA P815 Inhibiting endothelial cell migration, ↓ chemokine ligand 2 and IL-8, ↑ eNOS endothelial cell atherosclerosis 62-69
enterolactone phytolignan NA NA ↓lipid peroxidation NA heart disease- and CVD-related mortality 70-71
TMAO phosphatidylcho-line, choline, carnitine, buytrobetaine, betaine, trimethyllysine, valerobetaine, ergothioneine cutC/D, yeaW/X, Grdh, egtA, egtB, egtC, egtD, egtE, torA Diverse ↑CD36, SR-A1 ↓Cyp7a1 and Cyp27a1,
↑MAPK, NFκB, NLRP3 signaling
↑Ca2+ release
macrophage, endothelial cell, platelet atherosclerosis, thrombosis 80-110
indoxyl sulfate tryptophan NA Lactobacillus, Bifidobacterium longum, Bacteroides fragilis, Parabacteroides distasonis, Clostridium bartlettii and Eubacterium hallii ↑oxidative stress,
↓nitric oxide,
↑TNFα
monocyte, vascular endothelial cells adverse cardiac event 114-124
p-cresyl sulfate tyrosine Tyrosine phenol-lyase (EC 4.1.99.2.),tyrosine transaminase (EC 2.6.1.5.) or by aromatic-amino-acid transaminase (EC 2.6.1.57.), phenylalanine dehydrogenase (EC 1.4.1.20.),p-hydroxyphenylpyruvate oxidase,p-hydroxyphenylacetate decarboxylase Bacteroidaceae, Bifidobacteriaceae, Clostridiaceae, Enterobacteriaceae, Enterococcaceae, Eubacteriaceae, Fusobacteriaceae, Lachnospiraceae, Lactobacillaceae, Porphyromonadaceae, Staphylococcaceae, Ruminococcaceae, Veillonellaceae ↑ NADPH oxidase,
↑ROS,
cardiomyocyte cardiovascular event and all-cause mortality in elderly hemodialysis patients 126-128
phenylacetylglutamine phenylalanine amino transferase (Aat), PorA, Clostridium sporogenes, NA NA Cardiac event 133

NA, not available; CVD, cardiovascular disease. ↑ ↓ increases or decreases compared with control, respectively.

Some beneficial gut microbial derived metabolites

Gut microbiota can degrade some macromolecules in diet to improve cardiovascular health. As mentioned above, the dietary fiber degraded can produce butyric acid, which can maintain gut barrier and inhibit cholesterol absorption and prevent atherosclerosis [54,55].

Esculin, the glucoside of esculetin, can be hydrolyzed by gut microbes to release free esculetin [56]. Esculetin can significantly inhibit hydrogen peroxide- and Ang-II-induced cell death in human aortic endothelial cells by enhancing NO production via AMPK-mediated eNOS phosphorylation [57].

Anthocyannin has a potential effect as an antiplatelet agent that subsequently can prevent thrombosis and CVD [58]. An earlier study suggests that protocatechuic acid, one gut microbiota metabolite of anthocyanin, decreases miR-10b expression in macrophage, therefore increases ABCA1 and ABCG1 expression and enhances cholesterol reverse transport, leading to attenuation of atherosclerosis [59]. Anthocyannin is a polyphenol compound, which acts as anti-oxidant and can arrest free radicals in human body [60, 61].

Besides anthocyanin, another natural polyphenol, ellagitannin abundant in some fruits, nuts, tea and seeds such as pomegranates, berries and walnuts, shows some cardiovascular benefit [6265]. However, ellagitannin has a very low bioavailability, and most of the intake ellagitanin from diet cannot reach circulatory system and gut microbes can metabolize ellagitanin as urolithin A or B, which can be absorbed into circulatory system [64,66]. The gut bacteria which can produce urolithin were isolated from human fecal samples, including Gordonibacter urolithinfaciens sp. nov. and Bifidobacterium pseudocatenulatum INIA P815 [65, 67]. Urolithin A can inhibit endothelial cell migration and decrease the expression of chemokine (C-C motif) ligand 2 and interleukin-8, therefore ameliorate TNFα-induced inflammation and associated molecular markers in human aortic endothelial cells [68], urolithin B-glucuronide can activate eNOS expression, which is considered as an effective strategy for CVD prevention [69].

Enterolactone, a gut microbiota derived metabolite of phytolignans, is a polyphenol compound acting as anti-oxidant, and the low serum concentration of enterolactone is associated with enhanced in vivo lipid peroxidation and increased coronary heart disease- and CVD-related mortality [70, 71].

In addition, gut microbiome modulates host bile acid profile by deconjugation, dehydroxylation and epimerization [7274], which further affect absorption of cholesterol and triglyceride in small intestine leading to decreased blood cholesterol and LDL [73]. Bile acid receptors, FXR and TGR5, mediate bile acids’ effect in increasing reverse cholesterol efflux and further decreasing foam cell formation and atherosclerosis [7577]. The deconjugation of bile acids in intestine is catalyzed by bile salt hydrolase (BSH) in microbes and the probiotic bacterium expressing BSH shows potential to treat and prevent atherosclerosis [78, 79].

On the other hand, some gut microbial derived metabolites are mechanistically linked to CVD, including trimethylamine N-oxide (TMAO), aromatic amino acid metabolites, p-cresyl sulfate and indoxylsulfate.

TMAO

TMAO is the gut microbiota derived metabolite of phosphatidylcholine, choline, carnitine, γ-buytrobetaine, betaine, trimethyllysine, valerobetaine and ergothioneine as well [80-83, 84*, 85, 86*, 87*, 88]. There are two steps for the biosynthesis of TMAO: the first step is cleavage of precursors with structural moiety containing trimethylamine (TMA) group to form TMA, which is catalyzed by enzymes in gut microbes; the second step is the oxidation of TMA to TMAO by hepatic flavin monooxygenase. The two steps constitute a metaorganismal pathway of TMAO biosynthesis [24]. Several bacterium enzymes involved in the first step were identified, such as choline TMA lyase (CutC/D), carnitine Rieske-type oxygenase/reductase (CntAB), YeaW/X, betaine reductase and ergothionase [83, 8992]. In human gut, TMAO reductase is widely distributed in bacterium and can reduce TMAO as TMA [93]. C57BL/6J ApoE−/− mouse is an atherosclerosis-prone mouse model, which develops atherosclerosis similar to humans. The mice fed TMAO supplemented chow diet shows enhanced atherosclerotic plaque when compared with control chow diet, and choline supplemented chow diet enhanced atherosclerosis which is dependent on gut microbiota, whereas the deprivation of gut microbiota by oral supplementation of broad spectrum of antibiotics can attenuate choline promoting atherosclerosis [80]. The fecal microbiota transplant mice model confirmed that microbes from mice tending to develop atherosclerosis can make germ free mice recipient develop larger atherosclerotic plaque compared with atherosclerosis-resistant mice [94]. The other precursors, such as carnitine, γ-butyrobetaine, also show enhanced atherosclerosis which is mediated by gut microbial production of TMAO [82, 83]. Besides atherosclerosis, TMAO can also promote thrombosis [95].

TMAO is mechanistically linked to atherosclerotic CVD and thrombosis through multiple mechanisms. First TMAO enhances endogenous macrophage expression of scavenger receptors, CD36 and SR-A1, leading to uptake of modified LDL to develop foam cells [80]. Second TMAO inhibits expression of the two key bile acid synthetic enzymes, Cyp7a1 and Cyp27a1, and multiple bile acid transporters (Oatp1, Oatp4, Mrp2, and Ntcp) in the liver, therefore decreasing bile acid pool size and subsequent cholesterol excretion [82]. Third TMAO can activate MAPK, NFκB and ROS-TXNIP-NLRP3 inflammasome signaling and promotes recruitment of activated leukocytes to endothelial cells [96, 97]. TMAO also elicits intracellular Ca2+ release and activates platelet aggregation, therefore causing thrombosis [95].

Targeting gut microbial metaorganismal pathway of TMAO biosynthesis either by administration of choline TMA lyase inhibitor or by peritoneal injection of anti-sense flavin monooxgenase 3 oligonucleotides shows attenuation of atherosclerosis and thrombosis [98, 99*, 100]. Some methanogenic archaea can consume trimethylamine [101], and the colonization with methanogenic archaea lowers circulating TMAO, indicating a promising way to attenuate atherosclerosis [102*].

In humans, higher levels of circulatory TMAO can track future risk for major adverse cardiac events [81]. Patients with stable heart failure (HF) have significantly higher plasma levels of TMAO than human subjects without HF and TMAO concentrations show significant positive correlation to B-type natriuretic peptide levels [103, 104]. The causality of TMAO and HF has been confirmed by surgical transverse aortic constriction and coronary ligation animal models, which indicates that TMAO increased HF susceptibility and reducing circulating TMAO ameliorates the development of chronic HF [105*]. The association between TMAO and CVD prevalence and cardiac event has been confirmed by other different groups worldwide [106, 107*, 108, 109*, 110*]

TMAO was initially reported as a chemical chaperone and it can stabilize protein conformation by acting as a surfactant for the heterogeneous surfaces of folded proteins [111, 112] TMAO is abundant in marine fish, which acts as cryo-protectant. TMAO demethylase (TMAOase) in the muscle can catalyze the degradation of TMAO and one product is formaldehyde during fish storage, which constitutes another reason of fish spoilage [113].

Indoxyl sulfate

Indoxyl sulfate is a gut microbial derived metabolite of tryptophan [114, 115]. The metaorganismal biosynthesis of indoxyl sulfate includes microbial cleavage of tryptophan to indole and further oxidized to indoxyl and eventually conjugated as indoxyl sulfate in liver, indoxyl and indoxyl sulfate, which can be excreted to urine [116]. The microbial enzyme, tryptophanase, responsible for cleavage of tryptophan to indole, has been found in Lactobacillus, Bifidobacterium longum, Bacteroides fragilis, Parabacteroides distasonis, Clostridium bartlettii and E. hallii [117].

Plasma indoxyl sulfate was associated with first heart failure event in patients on hemodialysis and predicts major adverse cardiac events in patients with chronic kidney disease [118, 119*]. Indoxyl sulfate is mechanistically linked to CVD through multiple mechanisms. Indoxyl sulfate can induce human umbilical vein endothelial cells (HUVEC) oxidative stress, causing endothelial dysfunction including inhibition of proliferation and nitric oxide production and the anti-oxidant pre-treatment can ameliorate the inhibitory effect [120]. Indoxyl sulfate can also stimulate monocyte to release TNFα through the aryl hydrocarbon receptor (AhR), which further stimulates human vascular endothelial cells to produce CX3CL1, recruiting CD4(+)CD28(−)T cells, which exhibits cytotoxic capability and induces apoptosis in HUVECs, leading to vascular endothelial cell damage [121]. Indoxyl sulfate is also regarded as pro-thrombotic agent. It enhances platelet activities, including causing elevated response to collagen and thrombin and increasing platelet-derived microparticles and platelet-monocyte aggregates [122]. In addition, indoxyl sulfate impairs oxygen sensing in erythropoietin (EPO)-producing cells, thereby suppressing EPO production and resulting in anemia [123, 124].

P-cresyl sulfate

P-cresyl sulfate (PCS) is significantly higher in patients with HF and predicts future risk for a composite event of death or HF-related re-hospitalization [125]. PCS is a gut microbiota derived metabolite of tyrosine, which was processed by at least 4 different enzymes with 4 steps: the first step to the third step are carried out in gut microbes to form intermediates, 4-hydroxyphenylpyruvate, 4-hydroxyphenylacetate and p-cresol; and the last step is to form PCS in gut mucosa or liver [126]. PCS predicts cardiovascular event and all-cause mortality in elderly hemodialysis patients [127]. PCS induces NADPH oxidase activity and reactive oxygen species production contributing to direct cytotoxicity to cardiomyocytes, facilitating cardiac apoptosis and resulting in diastolic dysfunction [128], which is similar to indoxyl sulfate.

Phenylacetylglutamine

Phenylacetylglutamine (PAG) is excreted as a nitrogen waste, which can replace urea in patients lacking carbamyl phosphate synthetase [129]. PAG is a major nitrogenous metabolite that accumulates in uremia [130]. It is a gut microbiota and host co-metabolite of phenylalanine. Aminotransferase and pyruvate: ferredoxin oxidoreductase A (PorA) in bacterium were involved in the conversion from phenylalanine to phenylacetic acid and the activation of phenylacetic acid to form phenylacetyl-CoA and ligate to glutamine are carried out in human liver and kidney [131, 132]. Clostridium sporogenes expresses aminotransferase and PorA [130]. In patients with chronic kidney disease, high serum PAG level is associated with overall mortality and CVD [133].

More gut microbiota derived metabolites were summarized in reference [134], but whether they are involved in CVD pathogenesis or show beneficial effects on cardiovascular health need further investigation.

Gut microbiota derived metabolites contributing to CVD is related to renal insufficiency

TMAO, indoxylsulfate, PCS and PAG are uremic toxins. The elevated levels in circulatory blood is not only dependent on gut microbiome, diet, but also related to renal insufficiency. In non-chronic kidney disease patients, the kidney can excrete those uremic toxins in time without accumulation through tubular secretion [135]. For TMAO, if the fractional renal excretion (%) calculated is based on creatinine, c-mannosyltryptophan, pseudouridine or symmetric dimethylarginine as a surrogate for renal function, it can be higher than 100% [136*], which suggests that TMAO can be easily cleared off. However, in animal models, elevated dietary choline or TMAO directly led to progressive renal tubulointerstitial fibrosis and dysfunction by activating fibrotic TGF-β/Smad3 signaling pathway [137]. So TMAO exacerbates chronic kidney disease progression, which further impairs renal clearance of TMAO. Indoxylsulfate and PCS are protein bound uremic toxins, which are non-dialyzable [138*, 139**]. The kidney plays an important role in mediating the effect of gut microbiota derived metabolites on CVD progression.

Gut microbiome modulates the efficacy of drugs in the treatment of CVD

Gut microbiome can modulate drug efficacy and toxicity and inhibit its metabolism via direct biochemical reactions, such as acetylation, deacylation, decarboxylation, dehydroxylation, demethylation, dehalogenation, deconjugation and β-glucuronidation and indirect pathways through competition for host transporters and enzymes, modulation of host receptor signaling, altering host gene expression and gastrointestinal tract environment [140, 141*].

Statin is a widely used drug to decrease LDL cholesterol. Some patients after statin medication show decreased LDL cholesterol and other patients show no effect [142]. By comparison of gut microbiota from 202 hyperlipidemic patients with statin sensitive (SS) response and statin resistant (SR) response in East China, the investigators found that the SS group shows increased proportion of genera Lactobacillus, Eubacterium, Faecalibacterium, and Bifidobacterium and decreased proportion of genus Clostridium compared to Group SR group [143*], which suggests gut microbiota community may affect the statin efficacy. The bacterium community enriched in SS group contributing to statin sensitiveness may be related to elevated BSH, which hydrolyzes conjugated bile acid and the free bile acid will not be absorbed to circulatory blood leading to more cholesterol metabolism to bile acid [72, 144].

Monacolin K is a natural statin in some food, such as oyster mushrooms, red yeast rice, and Puerh tea [145], but it has no bioactivity until metabolized to beta-hydroxy acid form (MKA) under alkaline pH. Gut microbiome can catabolize MKA to lose bioactivity [146*].

Digoxin, a drug used for the treatment of atrial fibrillation, atrial flutter, and heart failure, can be inactivated by Eggerthella lenta by metabolism to dihydrodigoxin within gastrointestinal tract [147]. A cytochrome-encoding operon, termed the cardiac glycoside reductase, can be activated by digoxin and inhibited by arginine in some E lenta strains [148].

On the other hand, drug can also modulate gut microbiome community and affects the gut microbial derived metabolite production. Metformin is a widely prescribed drug to treat multiple diseases such as diabetes, cancer, CVD, Alzheimer’s disease, obesity and non-alcoholic fatty liver disease [149]. Metformin reduces cholesterol synthesis in macrophage and increase cholesterol efflux by up-regulating FGF21 expression [150, 151]. A study using 18 healthy individuals taking metformin showed that gut microbiome shift in one day with reduction of inner diversity of gut microbiota and an increase in relative abundance of common gut opportunistic pathogen Escherichia-Shigella spp, which are related to the severity of gastrointestinal side effect [152*]. Atorvastatin and rosuvastatin were investigated in an aged mouse model of high-fat diet-induced obesity and fecal microbiota transplantation with fecal material collected from rosuvastatin-treated mouse groups showed improved hyperglycemia [153*]. Aspirin, a drug widely used for antipyretic, analgesic, anti-inflammation and anti-coagulation, also shows gut bacteria discrimination from no medication in four bacteria taxa, Prevotella spp, Bacteroides spp, family Ruminococcaceae, and Barnesiella spp [154].

Physical exercise modulates gut microbiome community beneficial to cardiovascular health

Physical exercise can improve our health and reduce CVD risk by increasing circulatory high density lipoprotein and endothelial nitric oxide production and attenuation of oxidative damage as well [155*]. Intriguingly, physical exercise can also modulate gut microbiome community, showing beneficial effect to cardiovascular health. Physical exercise can increase microbiome richness and Bacteriodetes/Firmicutes ratio, which leads to increased short chain fatty acids production and release of glucogen-like peptide therefore improving insulin sensitivity and decreased lipopolysaccharide (LPS) production as well [156*]. The gut microbiota from exercise mice transplanted to germ free mice shows some benefit to attenuate response to chemical colitis by dextran sodium sulfate compared to sedentary mice [157].

LPS can cause vascular inflammatory responses including lipid accumulation, induced expression of interleukin (IL)-6, IL-8, monocyte chemoattractant protein 1, endothelial cell adhesion molecules, intercellular adhesion molecular-1 and vascular cell adhesion molecule-1 in human coronary artery endothelial cells (HCAECs) via TLR4-NF-κB pathway [158, 159]. LPS is a component of Gram negative bacterium outer membrane and its accumulation due to gut barrier dysfunction induces series inflammatory reaction leading to sepsis [160, 161]. Exercise can improve gut microbiota profiles, enhance the number of beneficial microbial species and reduce endotoxemia [162*, 163].

Exercise can increase Clostridiales, Roseburia, Lachnospiraceae, Erysipelotrichaceae, Ruminococcaceae and Eubacteriaceae abundance and those taxa are butyrate producers [164*, 165]. Butyrate can lower artery blood pressure by suppressing the prorenin receptor-mediated intrarenal renin-angiotensin system and is inversely correlated with inflammatory markers and serum endotoxin [166*, 167]. In addition, butyrate can maintain gut barrier to prevent endotoxin entering circulatory blood system [168*, 169*].

CONCLUSIONS

Gut microbiome, as an endocrine organ, affects multi-organ health. Gut microbiome can produce short chain fatty acids, modulating immune-response, improving insulin sensitivity and decreasing LPS level, which maintains human organism under a good condition. On the other hand, dysbiosis and unhealthy diet intake lead to gut barrier dysfunction, LPS and uremic toxin accumulation, which speeds up aortic endothelial cell inflammation, atherosclerosis and thrombosis. Some other gut microbiota derived metabolites, TMAO, indoxyl sulfate and p-cresyl sulfate, show clinical relevance of CVD and are mechanistically linked to atherosclerosis, thrombosis and heart failure. Physical exercise can modulate gut microbiome diversity, increase butyrate bacterium producer taxa abundance and attenuate oxidative damage, therefore improving cardiovascular health.

KEY POINTS.

  • Increasing evidence has shown that dysbiosis contributes to CVD.

  • Gut microbes translocation to aortic artery directly initiates inflammation.

  • Gut microbiota derived metabolites mediate the effects of gut microbiome contributing to cardiovascular health and disease.

  • Gut microbiome modulates the drug efficacy in the treatment of CVD.

  • Physical exercise can modulate gut microbiome to ameliorate cardiovascular health.

Acknowledgements

Z.W. is supported by grants from the National Institutes of Health and the Office of Dietary Supplements (R01HL130819).

Footnotes

Conflict of interest

Z.W. is named as co-inventor on pending and issued patents held by the Cleveland Clinic relating to cardiovascular diagnostics and therapeutics, and have the right to receive royalty payment for inventions or discoveries related to cardiovascular diagnostics or therapeutics from Cleveland Heart Lab or Procter & Gamble. Y.Z. declares no conflict of interest.

References

  • 1.El Kaoutari A, Armougom F, Raoult D, Henrissat B. Gut microbiota and digestion of polysaccharides. Med Sci (Paris). 2014;30:259–65. [DOI] [PubMed] [Google Scholar]
  • 2.Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.McCann A, Jeffery IB, Ouliass B, et al. Exploratory analysis of covariation of microbiota-derived vitamin K and cognition in older adults. Am J Clin Nutr. 2019;110:1404–1415.* Vitamin K in the form of menaquinone (MK) is synthesized by gut microbes. The total concentrations of MK did not covary with cognition, but certain MK isoforms, particularly the longer chains, are positively associated with cognition.
  • 4.LeBlanc JG, Milani C, de Giori GS, et al. Bacteria as vitamin suppliers to their host: a gut microbiota perspective. Curr Opin Biotechnol. 2013;24:160–8. [DOI] [PubMed] [Google Scholar]
  • 5.Malago JJ. Contribution of microbiota to the intestinal physicochemical barrier. Benef Microbes. 2015;6:295–311. [DOI] [PubMed] [Google Scholar]
  • 6.DeGruttola AK, Low D, Mizoguchi A, Mizoguchi E. Current understanding of dysbiosis in disease in human and animal models. Inflamm Bowel Dis. 2016;22:1137–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rowin J, Xia Y, Jung B, Sun J. Gut inflammation and dysbiosis in human motor neuron disease. Physiol Rep. 2017;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Scher JU. Intestinal dysbiosis and potential consequences of microbiome-altering antibiotic use in the pathogenesis of human rheumatic disease. J Rheumatol. 2015;42:355–7. [DOI] [PubMed] [Google Scholar]
  • 9.Hasuike Y, Endo T, Koroyasu M, et al. Bile acid abnormality induced by intestinal dysbiosis might explain lipid metabolism in Parkinson’s disease. Med Hypotheses. 2019;134:109436.* Intestinal dysbiosis leads to bacterium overgrowth, which breaks down bile acid. Lipid and vitamin D absorption are dependent on bile acid, so bacterium overgrowth causes low triglyceride and vitamin D, leading to patients losing weight and osteoporosis and fractures, which explains the causality effect of intestinal dysbiosis on Parkinson’s disease.
  • 10.Kong G, Cao KL, Judd LM, et al. Microbiome profiling reveals gut dysbiosis in a transgenic mouse model of Huntington’s disease. Neurobiol Dis. 2018:104268.* Huntington’s disease (HD) is a progressive neurodegenerative disorder. By comparing gut microbiome between R6/1 transgenic mouse model of HD and wild-type (WT) mice, authors found the gut microbiome dysregulation in HD, characterized by an increase in Bacteriodetes and a proportional decrease in Firmicutes.
  • 11.Pistollato F, Sumalla Cano S, Elio I, et al. Role of gut microbiota and nutrients in amyloid formation and pathogenesis of Alzheimer disease. Nutr Rev. 2016;74:624–34. [DOI] [PubMed] [Google Scholar]
  • 12.Wang X, Sun G, Feng T, et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019;29:787–803.* Gut dysbiosis promoted neuroinflammation in Alzheimer’s disease (AD) progression through peripheral accumulation of phenylalanine and isoleucine, stimulating the differentiation and proliferation of proinflammatory T helper 1 (Th1) cells and further M1 microglia activation, contributing to AD-associated neuroinflammation. The drug, GV-971, a sodium oligomannate, suppresses gut dysbiosis and the associated phenylalanine/isoleucine accumulation, therefore shaping neuroinflammation to inhibit AD progression.
  • 13.Li J, Zhao F, Wang Y, Chen J, et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome. 2017;5:14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Cui X, Ye L, Li J, et al. Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic heart failure patients. Sci Rep. 2018;8:635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhu Q, Gao R, Zhang Y, et al. Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol Genomics. 2018;50:893–903.* This paper reports the difference in gut microbiota community between healthy controls and CAD patients by 16S rRNA gene sequencing and found some signatures of CAD patients.
  • 16.Yamashita T, Emoto T, Sasaki N, Hirata KI. Gut Microbiota and Coronary Artery Disease. Int Heart J. 2016;57:663–71. [DOI] [PubMed] [Google Scholar]
  • 17.Yamashiro K, Tanaka R, Urabe T, et al. Gut dysbiosis is associated with metabolism and systemic inflammation in patients with ischemic stroke. PLoS One. 2017;12:e0171521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zuo K, Li J, Li K, et al. Disordered gut microbiota and alterations in metabolic patterns are associated with atrial fibrillation. Gigascience. 2019;8(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Jie Z, Xia H, Zhong SL, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun. 2017;8:845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ahmad AF, Dwivedi G, O’Gara F, et al. The gut microbiome and cardiovascular disease: current knowledge and clinical potential. Am J Physiol Heart Circ Physiol. 2019;317:923–38.** This is a review paper, which focus on the role of the gut microbiome and associated functional components in the development and progression of atherosclerosis. Different CVD phenotypes were introduced and the association with gut microbiome were reviewed. Finally the review paper discussed the potential treatments of atherosclerosis and CVD by alteration of gut microbiome.
  • 21.Steffen HM, Demir M. Gut Microbiome and Cardiovascular Disease. Dtsch Med Wochenschr. 2019;144:957–63.** This review paper discussed the role of the gut microbiome in regulating blood pressure, vascular function and its possible contribution to atherosclerosis and heart failure based on the available data.
  • 22.Lezutekong JN, Nikhanj A, Oudit GY. Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in cardiovascular disease. Clin Sci (Lond). 2018;132:901–4. [DOI] [PubMed] [Google Scholar]
  • 23.Peng J, Xiao X, Hu M, Zhang X. Interaction between gut microbiome and cardiovascular disease. Life Sci. 2018;214:153–7.** This review paper summarized the gut microbiota derived metabolite, mainly focused on trimethylamine N-oxide (TMAO), in the pathogenesis of cardiovascular disease and some microbiome discrimination was mentioned in patients with hypertension. Targeting gut microbiome and gut microbiota metabolite was suggested to be a novel and attractive field in the treatment of CVD.
  • 24.Wang Z, Zhao Y. Gut microbiota derived metabolites in cardiovascular health and disease. Protein Cell. 2018;9:416–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Tang WH, Hazen SL. The contributory role of gut microbiota in cardiovascular disease. J Clin Invest. 2014;124:4204–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Viggiano D, Ianiro G, Vanella G, et al. Gut barrier in health and disease: focus on childhood. Eur Rev Med Pharmacol Sci. 2015;19:1077–85. [PubMed] [Google Scholar]
  • 27.Vancamelbeke M, Vermeire S. The intestinal barrier: a fundamental role in health and disease. Expert Rev Gastroenterol Hepatol. 2017;11:821–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Konig J, Wells J, Cani PD, et al. Human Intestinal Barrier Function in Health and Disease. Clin Transl Gastroenterol. 2016;7:e196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Calandrini CA, Ribeiro AC, Gonnelli AC, et al. Microbial composition of atherosclerotic plaques. Oral Dis. 2014;20:e128–34. [DOI] [PubMed] [Google Scholar]
  • 30.Koren O, Spor A, Felin J, et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci U S A. 2011;108 Suppl 1:4592–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Neves AL, Coelho J, Couto L, et al. Metabolic endotoxemia: a molecular link between obesity and cardiovascular risk. J Mol Endocrinol. 2013;51:R51–64. [DOI] [PubMed] [Google Scholar]
  • 32.Ziganshina EE, Sharifullina DM, Lozhkin AP, et al. Bacterial Communities Associated with Atherosclerotic Plaques from Russian Individuals with Atherosclerosis. PLoS One. 2016;11:e0164836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Khan S, Rahman HN, Okamoto T, et al. Promotion of atherosclerosis by Helicobacter cinaedi infection that involves macrophage-driven proinflammatory responses. Sci Rep. 2014;4:4680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Morrison DC, Ryan JL. Endotoxins and disease mechanisms. Annu Rev Med. 1987;38:417–32. [DOI] [PubMed] [Google Scholar]
  • 35.Sperandeo P, Martorana AM, Polissi A. Lipopolysaccharide Biosynthesis and Transport to the Outer Membrane of Gram-Negative Bacteria. Subcell Biochem. 2019;92:9–37.* This review paper summarized lipopolysaccharide structure, functions and biosynthetic pathway and then discussed how it is transported and assembled to the cell surface.
  • 36.Kulp A, Kuehn MJ. Biological functions and biogenesis of secreted bacterial outer membrane vesicles. Annu Rev Microbiol. 2010;64:163–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Solov’eva T, Davydova V, Krasikova I, Yermak I. Marine compounds with therapeutic potential in gram-negative sepsis. Mar Drugs. 2013;11:2216–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Vanaja SK, Russo AJ, Behl B, et al. Bacterial Outer Membrane Vesicles Mediate Cytosolic Localization of LPS and Caspase-11 Activation. Cell. 2016;165:1106–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Li WL, Hua LG, Qu P, et al. NLRP3 inflammasome: a novel link between lipoproteins and atherosclerosis. Arch Med Sci. 2016;12:950–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Baldrighi M, Mallat Z, Li X. NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis. 2017;267:127–38. [DOI] [PubMed] [Google Scholar]
  • 41.Juang D, Braverman AC, Eagle K. Cardiology patient pages. Aortic dissection. Circulation. 2008;118:e507–10. [DOI] [PubMed] [Google Scholar]
  • 42.Feng J, Liu R, Ma S, et al. Aortic Dissection Presenting as Septic Shock: A Case Report and Literature Review. Case Rep Emerg Med. 2018;2018:9706290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Pham VT, Seifert N, Richard N, et al. The effects of fermentation products of prebiotic fibres on gut barrier and immune functions in vitro. PeerJ. 2018;6:e5288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hu ED, Chen DZ, Wu JL, Lu FB, Chen L, Zheng MH, et al. High fiber dietary and sodium butyrate attenuate experimental autoimmune hepatitis through regulation of immune regulatory cells and intestinal barrier. Cell Immunol. 2018;328:24–32. [DOI] [PubMed] [Google Scholar]
  • 45.Andoh A, Bamba T, Sasaki M. Physiological and anti-inflammatory roles of dietary fiber and butyrate in intestinal functions. JPEN J Parenter Enteral Nutr. 1999;23:S70–3. [DOI] [PubMed] [Google Scholar]
  • 46.Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, et al. A Dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167:1339–53 e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Moss JW, Ramji DP. Nutraceutical therapies for atherosclerosis. Nat Rev Cardiol. 2016;13:513–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Jha RK, Yong MQ, Chen SH. The protective effect of resveratrol on the intestinal mucosal barrier in rats with severe acute pancreatitis. Med Sci Monit. 2008;14:BR14–9. [PubMed] [Google Scholar]
  • 49.Wang N, Han Q, Wang G, Ma WP, Wang J, Wu WX, et al. Resveratrol Protects Oxidative Stress-Induced Intestinal Epithelial Barrier Dysfunction by Upregulating Heme Oxygenase-1 Expression. Dig Dis Sci. 2016;61:2522–34. [DOI] [PubMed] [Google Scholar]
  • 50.Shan CY, Yang JH, Kong Y, Wang XY, Zheng MY, Xu YG, et al. Alteration of the intestinal barrier and GLP2 secretion in Berberine-treated type 2 diabetic rats. J Endocrinol. 2013;218:255–62. [DOI] [PubMed] [Google Scholar]
  • 51.Ottman N, Reunanen J, Meijerink M, Pietila TE, Kainulainen V, Klievink J, et al. Pili-like proteins of Akkermansia muciniphila modulate host immune responses and gut barrier function. PLoS One. 2017;12:e0173004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Zhao F, Zhou G, Liu X, Song S, Xu X, Hooiveld G, et al. Dietary protein sources differentially affect the growth of Akkermansia muciniphila and maintenance of the gut Mucus barrier in mice. Mol Nutr Food Res. 2019:e1900589.* This paper highlights the importance of Akkermansia muciniphila (AKK) in maintaining gut barrier, and the intake of soy-based protein can decrease AKK abundance compared with chicken based protein.
  • 53.Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia Muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe−/− Mice. Circulation. 2016;133:2434–46. [DOI] [PubMed] [Google Scholar]
  • 54.Bultman SJ. Bacterial butyrate prevents atherosclerosis. Nat Microbiol. 2018;3:1332–3. [DOI] [PubMed] [Google Scholar]
  • 55.Chen Y, Xu C, Huang R, Song J, Li D, Xia M. Butyrate from pectin fermentation inhibits intestinal cholesterol absorption and attenuates atherosclerosis in apolipoprotein E-deficient mice. J Nutr Biochem. 2018;56:175–82. [DOI] [PubMed] [Google Scholar]
  • 56.Duncan SH, Flint HJ, Stewart CS. Inhibitory activity of gut bacteria against Escherichia coli O157 mediated by dietary plant metabolites. FEMS Microbiol Lett. 1998;164:283–8. [DOI] [PubMed] [Google Scholar]
  • 57.Karnewar S, Vasamsetti SB, Gopoju R, et al. Mitochondria-targeted esculetin alleviates mitochondrial dysfunction by AMPK-mediated nitric oxide and SIRT3 regulation in endothelial cells: potential implications in atherosclerosis. Sci Rep. 2016;6:24108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gaiz AA, Mosawy S, Colson N, Singh I. Potential of Anthocyanin to Prevent Cardiovascular Disease in Diabetes. Altern Ther Health Med. 2018;24:40–7. [PubMed] [Google Scholar]
  • 59.Wang D, Xia M, Yan X, et al. Gut microbiota metabolism of anthocyanin promotes reverse cholesterol transport in mice via repressing miRNA-10b. Circ Res. 2012;111:967–81. [DOI] [PubMed] [Google Scholar]
  • 60.Alafiatayo AA, Syahida A, Mahmood M. Total anti-oxidant capacity, flavonoid, phenolic acid and polyphenol content in ten selected species of Zingiberaceae rhizomes. Afr J Tradit Complement Altern Med. 2014;11:7–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Varga E, Domokos E, Fogarasi E, et al. Polyphenolic compounds analysis and antioxidant activity in fruits of Prunus spinosa L. Acta Pharm Hung. 2017;87:19–25. [PubMed] [Google Scholar]
  • 62.Heber D, Seeram NP, Wyatt H, et al. Safety and antioxidant activity of a pomegranate ellagitannin-enriched polyphenol dietary supplement in overweight individuals with increased waist size. J Agric Food Chem. 2007;55:10050–4. [DOI] [PubMed] [Google Scholar]
  • 63.Bakkalbasi E, Mentes O, Artik N. Food ellagitannins-occurrence, effects of processing and storage. Crit Rev Food Sci Nutr. 2009;49:283–98. [DOI] [PubMed] [Google Scholar]
  • 64.Larrosa M, Garcia-Conesa MT, Espin JC, Tomas-Barberan FA. Ellagitannins, ellagic acid and vascular health. Mol Aspects Med. 2010;31:513–39. [DOI] [PubMed] [Google Scholar]
  • 65.Gaya P, Peiroten A, Median M, Alvarez I, Landete JM. Bifidobacterium pseudocatenulatum INIA P815: The first bacterium able to produce urolithins A and B from ellagic acid. J Func Food. 2018;45:95–9. [Google Scholar]
  • 66.Kiss AK, Piwowarski JP. Ellagitannins, Gallotannins and their Metabolites- The Contribution to the Anti-Inflammatory Effect of Food Products and Medicinal Plants. Curr Med Chem. 2018;25:4946–67. [DOI] [PubMed] [Google Scholar]
  • 67.Selma MV, Tomas-Barberan FA, Beltran D, et al. Gordonibacter urolithinfaciens sp. nov., a urolithin-producing bacterium isolated from the human gut. Int J Syst Evol Microbiol. 2014;64:2346–52. [DOI] [PubMed] [Google Scholar]
  • 68.Gimenez-Bastida JA, Gonzalez-Sarrias A, Larrosa M, et al. Ellagitannin metabolites, urolithin A glucuronide and its aglycone urolithin A, ameliorate TNF-alpha-induced inflammation and associated molecular markers in human aortic endothelial cells. Mol Nutr Food Res. 2012;56:784–96. [DOI] [PubMed] [Google Scholar]
  • 69.Spigoni V, Mena P, Cito M, et al. Effects on Nitric Oxide Production of Urolithins, Gut-Derived Ellagitannin Metabolites, in Human Aortic Endothelial Cells. Molecules. 2016;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Vanharanta M, Voutilainen S, Nurmi T, et al. Association between low serum enterolactone and increased plasma F2-isoprostanes, a measure of lipid peroxidation. Atherosclerosis. 2002;160:465–9. [DOI] [PubMed] [Google Scholar]
  • 71.Vanharanta M, Voutilainen S, Rissanen TH, et al. Risk of cardiovascular disease-related and all-cause death according to serum concentrations of enterolactone: Kuopio Ischaemic Heart Disease Risk Factor Study. Arch Intern Med. 2003;163:1099–104. [DOI] [PubMed] [Google Scholar]
  • 72.Kumar RS, Brannigan JA, Prabhune AA, et al. Structural and functional analysis of a conjugated bile salt hydrolase from Bifidobacterium longum reveals an evolutionary relationship with penicillin V acylase. J Biol Chem. 2006;281:32516–25. [DOI] [PubMed] [Google Scholar]
  • 73.Ridlon JM, Harris SC, Bhowmik S, et al. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes. 2016;7:22–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Dawson PA, Karpen SJ. Intestinal transport and metabolism of bile acids. J Lipid Res. 2015;56:1085–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Zhang Y, Yin L, Anderson J, et al. Identification of novel pathways that control farnesoid X receptor-mediated hypocholesterolemia. J Biol Chem. 2010;285:3035–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Pols TW, Nomura M, Harach T, et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. 2011;14:747–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hartman HB, Gardell SJ, Petucci CJ, et al. Activation of farnesoid X receptor prevents atherosclerotic lesion formation in LDLR−/− and apoE−/− mice. J Lipid Res. 2009;50:1090–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Patel AK, Singhania RR, Pandey A, Chincholkar SB. Probiotic bile salt hydrolase: current developments and perspectives. Appl Biochem Biotechnol. 2010;162:166–80. [DOI] [PubMed] [Google Scholar]
  • 79.Jones ML, Tomaro-Duchesneau C, Martoni CJ, Prakash S. Cholesterol lowering with bile salt hydrolase-active probiotic bacteria, mechanism of action, clinical evidence, and future direction for heart health applications. Expert Opin Biol Ther. 2013;13:631–42. [DOI] [PubMed] [Google Scholar]
  • 80.Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tang WH, Wang Z, Levison BS, et al. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Koeth RA, Wang Z, Levison BS, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Koeth RA, Levison BS, Culley MK, et al. gamma-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab. 2014;20:799–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Li XS, Obeid S, Wang Z, et al. Trimethyllysine, a trimethylamine N-oxide precursor, provides near- and long-term prognostic value in patients presenting with acute coronary syndromes. Eur Heart J. 2019;40:2700–9.* Trimethyllysine, a trimethylamine N-oxide precursor, in patients presenting with acute coronary syndromes, can predict 30-day risk and 1-year incident risk for major adverse cardiac event.
  • 85.Wang Z, Tang WH, Buffa JA, et al. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur Heart J. 2014;35:904–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Servillo L, D’Onofrio N, Giovane A, et al. Ruminant meat and milk contain delta-valerobetaine, another precursor of trimethylamine N-oxide (TMAO) like gamma-butyrobetaine. Food Chem. 2018;260:193–9.*This manuscript introduced a new trimethylamine N-oxide precursor, delta-valerobetaine, which is abundant in ruminant meat and milk. Meanwhile it can also be produced by metabolism of trimthyllysine by gut microbes and the enzymatic pathways were figured out.
  • 87.Halliwell B, Cheah IK, Tang RMY. Ergothioneine - a diet-derived antioxidant with therapeutic potential. FEBS Lett. 2018;592:3357–66.* Ergothioneine is a chemical nutrient containing trimethylamine group in structural formula, which is synthesized in fungi and bacterium, and it can be used as anti-oxidant. In humans, a transporter, OCTN1, is involved in the nutrient transport and the low concentrations in vivo is related to some diseases.
  • 88.Baldridge RC, Lewis HB. Diet and the ergothioneine content of blood. J Biol Chem. 1953;202:169–76. [PubMed] [Google Scholar]
  • 89.Craciun S, Marks JA, Balskus EP. Characterization of choline trimethylamine-lyase expands the chemistry of glycyl radical enzymes. ACS Chem Biol. 2014;9:1408–13. [DOI] [PubMed] [Google Scholar]
  • 90.Zhu Y, Jameson E, Crosatti M, et al. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc Natl Acad Sci U S A. 2014;111:4268–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Muramatsu H, Matsuo H, Okada N, et al. Characterization of ergothionase from Burkholderia sp. HME13 and its application to enzymatic quantification of ergothioneine. Appl Microbiol Biotechnol. 2013;97:5389–400. [DOI] [PubMed] [Google Scholar]
  • 92.Naumann E, Hippe H, Gottschalk G. Betaine: New Oxidant in the Stickland Reaction and Methanogenesis from Betaine and l-Alanine by a Clostridium sporogenes-Methanosarcina barkeri Coculture. Appl Environ Microbiol. 1983;45:474–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Jameson E, Doxey AC, Airs R, et al. Metagenomic data-mining reveals contrasting microbial populations responsible for trimethylamine formation in human gut and marine ecosystems. Microb Genom. 2016;2:e000080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gregory JC, Buffa JA, Org E, et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem. 2015;290:5647–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zhu W, Gregory JC, Org E, et al. Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk. Cell. 2016;165:111–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Seldin MM, Meng Y, Qi H, et al. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-kappaB. J Am Heart Assoc. 2016;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sun X, Jiao X, Ma Y, et al. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem Biophys Res Commun. 2016;481:63–70. [DOI] [PubMed] [Google Scholar]
  • 98.Wang Z, Roberts AB, Buffa JA, et al. Non-lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell. 2015;163:1585–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Roberts AB, Gu X, Buffa JA, et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med. 2018;24:1407–17.This paper reported a series of choline analogues, fluromethylcholine, chloromethylcholine, bromomethylcholine and iodomethylcholime, which can inhibit choline trimethylamine lyase efficiently and subsequently inhibit choline promoting platelet aggregation and thrombosis. These choline analogues do not show inhibitory effect on bacterium growth nor any adverse effect on animal liver and kidney function, so they are non-lethal inhibitors.
  • 100.Shih DM, Wang Z, Lee R, et al. Flavin containing monooxygenase 3 exerts broad effects on glucose and lipid metabolism and atherosclerosis. J Lipid Res. 2015;56:22–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Colby J, Zatman LJ. Trimethylamine metabolism in obligate and facultative methylotrophs. Biochem J. 1973;132:101–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ramezani A, Nolin TD, Barrows IR, et al. Gut colonization with methanogenic Archaea lowers plasma trimethylamine N-oxide concentrations in Apolipoprotein e−/− Mice. Sci Rep. 2018;8:14752.This paper reported that gut colonization with methanogenic Archaea which can consume trimethylamine, therefore decreasing plasma trimethylamine N-oxide and further decreasing atherosclerosis.
  • 103.Tang WH, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, et al. 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(18):1908–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Organ CL, Otsuka H, Bhushan S, Wang Z, Bradley J, Trivedi R, et al. Choline Diet and Its Gut Microbe-Derived Metabolite, Trimethylamine N-Oxide, Exacerbate Pressure Overload-Induced Heart Failure. Circ Heart Fail. 2016;9(1):e002314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Li X, Sun Y, Zhang X, Wang J. Reductions in gut microbiotaderived metabolite trimethylamine Noxide in the circulation may ameliorate myocardial infarctioninduced heart failure in rats, possibly by inhibiting interleukin8 secretion. Mol Med Rep. 2019;20(1):779–86.* This paper reports that reducing circulatory TMAO by using choline TMA lyase inhibitor, 3,3-dimethyl-1-butanol, can ameliorate the development of chronic HF following MI in rats, potentially by inhibiting IL8 secretion.
  • 106.Mente A, Chalcraft K, Ak H, et al. The Relationship between trimethylamine-N-oxide and prevalent cardiovascular disease in a multiethnic population living in Canada. Can J Cardiol. 2015;31:1189–94. [DOI] [PubMed] [Google Scholar]
  • 107.Hsu CN, Lu PC, Lo MH, et al. Gut Microbiota-Dependent Trimethylamine N-Oxide Pathway Associated with Cardiovascular Risk in Children with Early-Stage Chronic Kidney Disease. Int J Mol Sci. 2018;19.* This paper reported that about two-thirds of chronic kidney disease (CKD) children had blood pressure abnormalities. Children with CKD stage 2–3 had lower urinary TMAO than those with stage 1. Urinary TMAO level was positively correlated with the abundances of genera Bifidobacterium and CKD children with abnormal ambulatory blood-pressure monitoring (ABPM) had a lower abundance of the Prevotella genus than those with normal ABPM.
  • 108.Kim RB, Morse BL, Djurdjev O, et al. Advanced chronic kidney disease populations have elevated trimethylamine N-oxide levels associated with increased cardiovascular events. Kidney Int. 2016;89:1144–52. [DOI] [PubMed] [Google Scholar]
  • 109.Roncal C, Martinez-Aguilar E, Orbe J, et al. Trimethylamine-N-oxide (TMAO) predicts cardiovascular mrtality in peripheral artery disease. Sci Rep. 2019;9:15580.*This paper highlighted the association between circulating trimethylamine N-oxide (TMAO) and peripheryl artery disease (PAD) severity and prognosis. The higher plasma TMAO will lead to higher risk for cardiovascular mortality in PAD patients.
  • 110.Zheng L, Zheng J, Xie Y, et al. Serum gut microbe-dependent trimethylamine N-oxide improves the prediction of future cardiovascular disease in a community-based general population. Atherosclerosis. 2019;280:126–31.* This paper highlighted the association of circulating trimethylamine N-oxide (TMAO) and stroke severity and the patients with stroke have a significant higher levels of TMAO than healthy controls and plasma TMAO levels were increased with the severity of stroke.
  • 111.Liao YT, Manson AC, DeLyser MR, et al. Trimethylamine N-oxide stabilizes proteins via a distinct mechanism compared with betaine and glycine. Proc Natl Acad Sci U S A. 2017;114:2479–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Bennion BJ, Daggett V. Counteraction of urea-induced protein denaturation by trimethylamine N-oxide: a chemical chaperone at atomic resolution. Proc Natl Acad Sci U S A. 2004;101:6433–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Takeuchi K, Hatanaka A, Kimura M, et al. Aspolin, a novel extremely aspartic acid-rich protein in fish muscle, promotes iron-mediated demethylation of trimethylamine-N-oxide. J Biol Chem. 2003;278:47416–22. [DOI] [PubMed] [Google Scholar]
  • 114.Bryan GT. Urinary excretion of indoxyl sulfate (indican) by human subjects ingesting a semisynthetic diet containing variable quantities of L-tryptophan. Am J Clin Nutr. 1966;19:113–9. [DOI] [PubMed] [Google Scholar]
  • 115.Bryan GT. Quantitative studies on the urinary excretion of indoxyl sulfate (indican) in man following administration of L-tryptophan and acetyl-L-tryptophan. Am J Clin Nutr. 1966;19:105–12. [DOI] [PubMed] [Google Scholar]
  • 116.Banoglu E, Jha GG, King RS. Hepatic microsomal metabolism of indole to indoxyl, a precursor of indoxyl sulfate. Eur J Drug Metab Pharmacokinet. 2001;26:235–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhang LS, Davies SS. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 2016;8:46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Cao XS, Chen J, Zou JZ, Zhong YH, Teng J, Ji J, et al. Association of indoxyl sulfate with heart failure among patients on hemodialysis. Clin J Am Soc Nephrol. 2015;10:111–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Fan PC, Chang JC, Lin CN, et al. Serum indoxyl sulfate predicts adverse cardiovascular events in patients with chronic kidney disease. J Formos Med Assoc. 2019;118:1099–106.* This study measured serum indoxyl sulfate by LC/MS and using multivariable regression analysis and found that indoxylsulfate in patients with CKD can independently predict the risk of major adverse CV events (MACEs) with a AUC in ROC of 0.708 (95% confidence interval: 0.618–0.798)
  • 120.Yu M, Kim YJ, Kang DH. Indoxyl sulfate-induced endothelial dysfunction in patients with chronic kidney disease via an induction of oxidative stress. Clin J Am Soc Nephrol. 2011;6:30–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kim HY, Yoo TH, Hwang Y, et al. Indoxyl sulfate (IS)-mediated immune dysfunction provokes endothelial damage in patients with end-stage renal disease (ESRD). Sci Rep. 2017;7:3057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Yang K, Du C, Wang X, et al. Indoxyl sulfate induces platelet hyperactivity and contributes to chronic kidney disease-associated thrombosis in mice. Blood. 2017;129:2667–79. [DOI] [PubMed] [Google Scholar]
  • 123.Chiang CK, Tanaka T, Inagi R, et al. Indoxyl sulfate, a representative uremic toxin, suppresses erythropoietin production in a HIF-dependent manner. Lab Invest. 2011;91:1564–71. [DOI] [PubMed] [Google Scholar]
  • 124.Wu CJ, Chen CY, Lai TS, et al. The role of indoxyl sulfate in renal anemia in patients with chronic kidney disease. Oncotarget. 2017;8:83030–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wang CH, Cheng ML, Liu MH, Shiao MS, Hsu KH, Huang YY, et al. Increased p-cresyl sulfate level is independently associated with poor outcomes in patients with heart failure. Heart Vessels. 2016;31(7):1100–8. [DOI] [PubMed] [Google Scholar]
  • 126.Gryp T, Vanholder R, Vaneechoutte M, Glorieux G. p-Cresyl Sulfate. Toxins (Basel). 2017;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Lin CJ, Chuang CK, Jayakumar T, et al. Serum p-cresyl sulfate predicts cardiovascular disease and mortality in elderly hemodialysis patients. Arch Med Sci. 2013;9:662–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Han H, Zhu J, Zhu Z, et al. p-Cresyl sulfate aggravates cardiac dysfunction associated with chronic kidney disease by enhancing apoptosis of cardiomyocytes. J Am Heart Assoc. 2015;4:e001852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Brusilow SW. Phenylacetylglutamine may replace urea as a vehicle for waste nitrogen excretion. Pediatr Res. 1991;29:147–50. [DOI] [PubMed] [Google Scholar]
  • 130.Zimmerman L, Egestad B, Jornvall H, Bergstrom J. Identification and determination of phenylacetylglutamine, a major nitrogenous metabolite in plasma of uremic patients. Clin Nephrol. 1989;32:124–8. [PubMed] [Google Scholar]
  • 131.Dodd D, Spitzer MH, Van Treuren W, et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature. 2017;551:648–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Moldave K, Meister A. Synthesis of phenylacetylglutamine by human tissue. J Biol Chem. 1957;229:463–76. [PubMed] [Google Scholar]
  • 133.Poesen R, Claes K, Evenepoel P, et al. Microbiota-Derived Phenylacetylglutamine Associates with Overall Mortality and Cardiovascular Disease in Patients with CKD. J Am Soc Nephrol. 2016;27:3479–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–7. [DOI] [PubMed] [Google Scholar]
  • 135.Mair RD, Sirich TL, Meyer TW. Uremic Toxin Clearance and Cardiovascular Toxicities. Toxins (Basel). 2018;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wang Z, Bergeron N, Levison BS, et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur Heart J. 2019;40:583–94.* This study compared circulating and urinary TMAO among 113 healthy human subjects after random cross consumption of three different protein source diets, red-meat, white-meat and non-meat for one month. Results indicated that red meat consumption can significantly increase plasma TMAO and decrease fractional renal excretion of TMAO compared with non-meat or white meat.
  • 137.Tang WH, Wang Z, Kennedy DJ, et al. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ Res. 2015;116:448–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Paroni R, Casati S, Dei Cas M, et al. Unambiguous Characterization of p-Cresyl Sulfate, a Protein-Bound Uremic Toxin, as Biomarker of Heart and Kidney Disease. Molecules. 2019;24.* This study reported that when measuring p-cresyl sulfate, another isomer 2-hydroxy-5-methylbenzenesulfonic acid, which shows the same fragmentation pattern by mass spectrometry, can also be present, which is easy to get confused with p-cresyl sulfate and the two compounds were synthesized from different pathways and they can be distinguished from each other by NMR.
  • 139.Savira F, Magaye R, Hua Y, et al. Molecular mechanisms of protein-bound uremic toxin-mediated cardiac, renal and vascular effects: underpinning intracellular targets for cardiorenal syndrome therapy. Toxicol Lett. 2019;308:34–49.** This review paper highlighted the roles of protein bound uremic toxins in cardiorenal syndrome and the molecular mechanisms, which includes oxidative stress, fibrosis and collagen biosynthesis as well.
  • 140.Wilson ID, Nicholson JK. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl Res. 2017;179:204–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Tuteja S, Ferguson JF. Gut Microbiome and Response to Cardiovascular Drugs. Circ Genom Precis Med. 2019;12:421–9.* This paper highlighted the involvement of gut microbiome modulation of the efficacy of drugs on anti-cardiovascular disease. Gut microbes can metabolize the drugs and affect their absorption. The efficacy of drugs was suggested to meet the personalized gut microbiome.
  • 142.Ridker PM, Mora S, Rose L, Group JTS. Percent reduction in LDL cholesterol following high-intensity statin therapy: potential implications for guidelines and for the prescription of emerging lipid-lowering agents. Eur Heart J. 2016;37:1373–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Sun B, Li L, Zhou X. Comparative analysis of the gut microbiota in distinct statin response patients in East China. J Microbiol. 2018;56:886–92.* This study suggested that statin sensitive medication and statin resistant medication are related to individual gut microbiome structure. The former group of patients show increased genera Lactobacillus, Eubacterium, Faecalibacterium and Bifidobacterium and decreased genus Clostridium compared to Group statin resistance.
  • 144.Jones ML, Martoni CJ, Prakash S. Cholesterol lowering and inhibition of sterol absorption by Lactobacillus reuteri NCIMB 30242: a randomized controlled trial. Eur J Clin Nutr. 2012;66:1234–41. [DOI] [PubMed] [Google Scholar]
  • 145.Svoboda P, Sander D, Plachka K, Novakova L. Development of matrix effect-free MISPE-UHPLC-MS/MS method for determination of lovastatin in Pu-erh tea, oyster mushroom, and red yeast rice. J Pharm Biomed Anal. 2017;140:367–76. [DOI] [PubMed] [Google Scholar]
  • 146.Beltran D, Frutos-Lison MD, Espin JC, Garcia-Villalba R. Re-examining the role of the gut microbiota in the conversion of the lipid-lowering statin monacolin K (lovastatin) into its active beta-hydroxy acid metabolite. Food Funct. 2019;10:1787–91.* It was reported previously that gut microbiome can activate monacolin K (MK, lovastatin), a naturally occurring statin, to its active β-hydroxy acid form (MKA). Now in this new study, authors reported that MK can spontaneously be converted to MKA. The real role of gut microbiome is to catabolize MKA, therefore hampering its lipid-lowering effect.
  • 147.Saha JR, Butler VP Jr.,Neu HC, Lindenbaum J. Digoxin-inactivating bacteria: identification in human gut flora. Science. 1983;220:325–7. [DOI] [PubMed] [Google Scholar]
  • 148.Haiser HJ, Gootenberg DB, Chatman K, et al. Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science. 2013;341:295–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Maniar K, Moideen A, Mittal A, et al. A story of metformin-butyrate synergism to control various pathological conditions as a consequence of gut microbiome modification: Genesis of a wonder drug? Pharmacol Res. 2017;117:103–28. [DOI] [PubMed] [Google Scholar]
  • 150.Koren-Gluzer M, Aviram M, Hayek T. Metformin inhibits macrophage cholesterol biosynthesis rate: possible role for metformin-induced oxidative stress. Biochem Biophys Res Commun. 2013;439:396–400. [DOI] [PubMed] [Google Scholar]
  • 151.Luo F, Guo Y, Ruan G, Li X. Metformin promotes cholesterol efflux in macrophages by up-regulating FGF21 expression: a novel anti-atherosclerotic mechanism. Lipids Health Dis. 2016;15:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Elbere I, Kalnina I, Silamikelis I, et al. Association of metformin administration with gut microbiome dysbiosis in healthy volunteers. PLoS One. 2018;13:e0204317.* Metformin increases the abundance of opportunistic pathogens and further triggers the occurrence of side effects associated with the observed dysbiosis.
  • 153.Kim J, Lee H, An J, et al. Alterations in gut microbiota by statin therapy and possible intermediate effects on hyperglycemia and hyperlipidemia. Front Microbiol. 2019;10:1947.* Statin modulates gut microbiome and mediates the effect of metabolic improvement and inflammatory response, which is confirmed by fecal transplant.
  • 154.Rogers MAM, Aronoff DM. The influence of non-steroidal anti-inflammatory drugs on the gut microbiome. Clin Microbiol Infect. 2016;22:178 e1–e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Tian D, Meng J. Exercise for prevention and relief of cardiovascular disease: prognoses, mechanisms, and approaches. Oxid Med Cell Longev. 2019;2019:3756750.* Physical activity can improve insulin sensitivity, alleviate plasma dyslipidemia, normalize elevated blood pressure, decrease blood viscosity, promote endothelial nitric oxide production, and improve leptin sensitivity to protect the heart and vessels.
  • 156.Sohail MU, Yassine HM, Sohail A, Al Thani AA. Impact of physical exercise on gut microbiome, inflammation, and the pathobiology of metabolic disorders. Rev Diabet Stud. 2019;15:35–48.* Exercise-induced microbial changes affect the host’s immune pathways and improve energy homeostasis. Microbes release certain neuroendocrine and immune-modulatory factors that may lower inflammatory and oxidative stress and relieve patients suffering from metabolic disorders.
  • 157.Allen JM, Mailing LJ, Cohrs J, et al. Exercise training-induced modification of the gut microbiota persists after microbiota colonization and attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes. 2018;9:115–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Wei Z, Sun X, Xu Q, et al. TAK-242 suppresses lipopolysaccharide-induced inflammation in human coronary artery endothelial cells. Pharmazie. 2016;71:583–7. [DOI] [PubMed] [Google Scholar]
  • 159.Wang J, Si Y, Wu C, et al. Lipopolysaccharide promotes lipid accumulation in human adventitial fibroblasts via TLR4-NF-kappaB pathway. Lipids Health Dis. 2012;11:139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Fukui H Increased intestinal permeability and decreased barrier function: does it really influence the risk of inflammation? Inflamm Intest Dis. 2016;1:135–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Opal SM. Endotoxins and other sepsis triggers. Contrib Nephrol. 2010;167:14–24. [DOI] [PubMed] [Google Scholar]
  • 162.Motiani KK, Collado MC, Eskelinen JJ, et al. Exercise training modulates gut microbiota profile and improves endotoxemia. Med Sci Sports Exerc. 2020;52:94–104.Exercise training modulates gut microbiota profile with increased Bacteroidetes phylum and decreased Firmicutes/Bacteroidetes ratio, and decreased Clostridium and Blautia genera. Excise can decrease jejunal fatty acid uptake. Exercise improves whole-body insulin sensitivity reduces endotoxemia.
  • 163.Monda V, Villano I, Messina A, et al. Exercise modifies the gut microbiota with positive health effects. Oxid Med Cell Longev. 2017;2017:3831972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Nagano T, Yano H. Effect of dietary cellulose nanofiber and exercise on obesity and gut microbiota in mice fed a high-fat-diet. Biosci Biotechnol Biochem. 2019:1–8.* Cellulose nanofiber (CN) consumption with exercise can ameliorate obesity. Exercise can shift gut microbiome and increase Ruminococcaceae abundance, whereas exercise and CN intake together increased Eubacteriaceae abundance. These two taxa are butyrate producers, which explained the mechanism on exercise attenuation of obesity.
  • 165.Estaki M, Pither J, Baumeister P, et al. Cardiorespiratory fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions. Microbiome. 2016;4:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Juanola O, Ferrusquia-Acosta J, Garcia-Villalba R, et al. Circulating levels of butyrate are inversely related to portal hypertension, endotoxemia, and systemic inflammation in patients with cirrhosis. FASEB J. 2019;33:11595–605.* Short-chain fatty acids (SCFAs) are gut microbiota-derived products that participate in maintaining the gut barrier integrity and host’s immune response. SCFAs are inversely associated with disease severity. Butyric acid inversely correlated with inflammatory markers and serum endotoxin. A global reduction in the blood levels of SCFA in patients with cirrhosis is associated with a more advanced liver disease.
  • 167.Wang L, Zhu Q, Lu A, et al. Sodium butyrate suppresses angiotensin II-induced hypertension by inhibition of renal (pro)renin receptor and intrarenal renin-angiotensin system. J Hypertens. 2017;35:1899–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Kaden-Volynets V, Gunther C, Zimmermann J, et al. Deletion of the Casp8 gene in mice results in ileocolitis, gut barrier dysfunction, and malassimilation, which can be partially attenuated by inulin or sodium butyrate. Am J Physiol Gastrointest Liver Physiol. 2019;317:G493–G507.* Caspase-8 is very important to maintain gut barrier and liver function. Deletion of the caspase-8 gene results not only in ileocolitis but also in gut barrier dysfunction, liver steatosis, and malassimilation, which can be partially attenuated by oral inulin or sodium butyrate.
  • 169.Ye J, Lv L, Wu W, et al. Butyrate protects mice against methionine-choline-deficient diet-induced non-alcoholic steatohepatitis by improving gut barrier function, attenuating inflammation and reducing endotoxin levels. Front Microbiol. 2018;9:1967.* Butyrate modified gut microbiota and fecal metabolites, which were strongly correlated with the alleviation of hepatic injury, fibrosis progression, inflammation, and lipid metabolism and intestinal barrier dysfunction.

RESOURCES