Next Article in Journal
Chitosan Composites with Bacterial Cellulose Nanofibers Doped with Nanosized Cerium Oxide: Characterization and Cytocompatibility Evaluation
Next Article in Special Issue
Microbiome Composition in Microscopic Colitis: A Systematic Review
Previous Article in Journal
Heterologous Expression and Bioactivity Determination of Monochamus alternatus Antibacterial Peptide Gene in Komagataella phaffii (Pichia pastoris)
Previous Article in Special Issue
Gut Microbiota Deficiency Exacerbates Liver Injury in Bile Duct Ligated Mice via Inflammation and Lipid Metabolism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of the Gut Microbiome in the Development of Atherosclerotic Cardiovascular Disease

1
Internal Medicine Department, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada
2
Cardiology Department, Faculty of Medicine, University of Montreal, Montréal, QC H3T 1J4, Canada
3
Centre de Biomédecine, CIUSSS-NÎM/Hôpital du Sacré-Cœur, Montréal, QC H4J 1C5, Canada
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5420; https://doi.org/10.3390/ijms24065420
Submission received: 12 February 2023 / Revised: 6 March 2023 / Accepted: 9 March 2023 / Published: 12 March 2023
(This article belongs to the Special Issue Gut Microbiota in Disease and Health 2.0)

Abstract

:
Atherosclerotic cardiovascular disease (ASCVD) is the primary cause of death globally, with nine million deaths directly attributable to ischemic heart diseases in 2020. Since the last few decades, great effort has been put toward primary and secondary prevention strategies through identification and treatment of major cardiovascular risk factors, including hypertension, diabetes, dyslipidemia, smoking, and a sedentary lifestyle. Once labelled “the forgotten organ”, the gut microbiota has recently been rediscovered and has been found to play key functions in the incidence of ASCVD both directly by contributing to the development of atherosclerosis and indirectly by playing a part in the occurrence of fundamental cardiovascular risk factors. Essential gut metabolites, such as trimethylamine N-oxide (TMAO), secondary bile acids, lipopolysaccharides (LPS), and short-chain fatty acids (SCFAs), have been associated with the extent of ischemic heart diseases. This paper reviews the latest data on the impact of the gut microbiome in the incidence of ASCVD.

1. Introduction

Despite major advances in prevention and treatment strategies, atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of morbidity and mortality all around the world [1]. This alarming statistic brought to light the complex etiology of atherosclerosis, which has been recognized as not being solely induced by conventional risk factors, such as hypertension, diabetes, dyslipidemia, male sex, and smoking. In 2000, Haraszthy et al. proposed for the first time that the gut microbiota was associated with the occurrence of ASCVD, after finding DNA from multiple bacteria species in a plaque of cholesterol [2].
Trillions of micro-organisms weighing about 1.5 kg inhabit the gut, carrying out key functions that the rest of the human body is incapable of performing [3]. These micro-organisms have combined genomes (the microbiome) that exceed the human genome by many times [4,5,6]. The gut microbiota is dominated by anaerobic bacteria, with Firmicutes (Gram-positive) and Bacteroidetes (Gram-negative) composing more than 90% of intestinal bacterial species [7].
This paper reviews the latest available information on the role of the gut microbiome in the incidence and progression of ASCVD.

2. Metabolic Pathways

Major metabolites have been identified and linked to the development of cardiovascular diseases, including but not limited to trimethylamine N-oxide (TMAO), secondary bile acids, lipopolysaccharides (LPS), short-chain fatty acids (SCFAs), and phenylacetylglutamine (PAGln). Figure 1 illustrates major gut metabolic pathways leading to ASCVD.
First, TMAO directly contributes to the pathogenesis and extent of ASCVD. The correlation between these two entities was first reported by Wang et al. in 2011 [8]. TMAO goes through several enzyme modifications before being transformed into its final active form. In fact, the first step requires the intestinal conversion of one of three metabolites found in the food, namely L-carnitine, choline, and betaine, into TMA by the enzyme TMA lyase, which is derived from the Firmicutes species found in the gut microbiota [9]. These three nutrients are naturally found in foods, such as eggs, red meat, and fish. After their conversion into TMA, this amine is absorbed into the bloodstream before being transported to the liver, where it is transformed into TMAO by the enzyme flavin-dependent mono oxygenase 3 (FMO3) [10]. In physiologic states, the kidney excretes in the urine close to 95% of TMA oxidized into TMAO [11]. Hence, changes to any component along this metabolic pathway, from food ingestion to hepatorenal function together with the liver FMO3 activity, could lead to increased levels of TMAO with associated complications, such as ASCVD [12].
Primary bile acids are synthesized in the liver from cholesterol and are conjugated with glycine, subsequently leading to the formation of cholic acid and chenodeoxycholic acid. Then, primary bile acids are transported in the gut, in which the microbiota contributes to their deconjugation to form secondary bile acids in the distal ileum [13]. Secondary bile acids permit absorption of lipid nutrients and fat-soluble vitamins [14]. They are also involved in the activation of two key receptors, namely farnesoid X receptor (FXR) and takeda G protein-coupled receptor 5 (TGR5). These receptors modulate glucose and cholesterol metabolism. In fact, TGR5 leads to an increased secretion of glucagon-like peptide 1 (GLP-1), which contributes to an improved glucose tolerance [15,16]. TGR5 is also thought to possess anti-inflammatory properties by inhibiting nuclear factor-κB (NF-κB), thus decreasing the production of pro-inflammatory cytokines [17]. In another trial, simultaneous inhibition of both FXR and TGR5 exacerbated atherosclerotic formation, thereby highlighting their benefits in disease control [18]. Their anti-inflammatory and anti-atherogenic properties arise from suppression of tumor necrosis factor-α (TNF-α) and NF-κB signaling pathways in addition to a decreased secretion of pro-inflammatory cytokines [19]. Therefore, secondary bile acids activate two key receptors involved in the inhibition of major atherosclerotic pathways.
Similar to TMAO, LPS are endotoxins found on the outer membrane of Gram-negative bacteria and are involved in the pathogenesis of ASCVD. LPS are recognized by the innate immune system by toll-like receptor 4 (TLR4), which is a subtype of pattern recognition receptors (PRR) [20]. Upon recognition of LPS, TLR4 induces a pro-inflammatory state with an increased production and secretion of cytokines and chemokines [21]. LPS are also identified by other receptors, such as LPS-binding protein (LBP), myeloid differentiation protein 2 (MD-2), and cluster of differentiation 14 (CD14) [22]. These receptors, which are mainly expressed on macrophages, activate and enhance several protein kinases, such as IL-1 receptor-associated kinase (IRAK-1) and myeloid differentiation factor 88 (MyD88). NF-κB is subsequently activated, which, along with LPS, stimulate numerous pro-atherosclerotic inflammatory pathways [23,24,25]. In fact, LPS induce endothelial dysfunction, increase oxidative stress through production of reactive oxygen species (ROS), and produce several pro-inflammatory cytokines, such as TNF-α, interleukin-1 (IL-1), interleukin-6 (IL-6), and interleukin-8 (IL-8) [26,27,28]. Therefore, LPS contribute to ASCVD by promoting inflammation through various pathways.
In contrast to TMAO and LPS, SCFAs are protective against the occurrence of atherosclerosis and are the result of the ingestion and digestion of complex carbohydrates by numerous gut bacteria, including Anaerostipes butyraticus, Faecalibacterium prausnitzii, and Roseburia intestinalis [29,30]. The most frequent SCFAs produced are acetate, butyrate, and propionate [31]. These nutrients serve many roles, but their primary function is to modulate the host immune system through increased production of regulatory T cells and suppression of histone deacetylases (HDACs) [32,33]. By inhibiting HDACs, SCFAs inhibit inflammatory pathways owing to a decrease in NF-κB activation together with a reduced production of pro-inflammatory cytokines [34]. Other functions attributed to SCFAs include enhanced intestinal barrier stability and protection against pathogen invasion [32,33]. Thus, SCFAs protect against atherosclerosis by modulating inflammatory pathways.
Phenylacetylglutamine (PAGln) is a metabolite that was recently discovered and was shown to be positively associated with the development of cardiovascular diseases [35]. PAGln is derived from a simple amino acid, phenylalanine, that undergoes a series of alterations before arriving to its active metabolite. In fact, Nemet et al. have demonstrated that the microbial porA gene permits the transformation of phenylalanine into phenylacetic acid, with subsequent hepatic metabolization of phenylacetic acid into PAGln [36]. PAGln was first reported to be positively correlated with ASCVD and overall mortality in patients suffering from chronic kidney disease (CKD) [37]. Several pathophysiologic mechanisms were hypothesized to explain this association. As a matter of fact, PAGln was shown to increase platelets’ activation and responsiveness, resulting in increased thrombosis potential leading to ASCVD. [38] PAGln also transmits cellular events via G-protein coupled receptors, specifically the α2A, α2B, and β2 adrenergic receptors [36]. Interestingly, carvedilol, a commonly used β-blocker in clinical practice, was shown to inhibit these prothrombotic effects [36]. Thus, PAGln is involved in the occurrence of ASCVD through an accelerated rate of thrombus generation and vessel occlusion, potentially giving rise to acute myocardial infarction.

3. Gut Microbiome and Hypertension

Arterial hypertension is a well-recognized and major risk factor for the development of ASCVD [39,40,41]. Leading medical communities of cardiology recommend blood pressure control both pharmacologically and non-pharmacologically as part of cardiovascular diseases’ primary and secondary preventions [42,43,44]. Even though the exact cause of essential hypertension remains unclear, many risk factors are thought to contribute to its development, including advanced age, positive family history, obesity, a high-sodium diet, and a sedentary lifestyle [42,43,44].
Recently, the gut microbiota has been found to play a role in the development of hypertension [45,46,47,48,49,50,51,52,53,54]. Li et al. have demonstrated that high blood pressure was transferrable through fecal transplantation from hypertensive subjects to germ-free mice, thus confirming the implication of the intestinal microbiome [47]. Additionally, compared to healthy controls, pre-hypertensive and hypertensive individuals have decreased microbial richness and variety with an overgrowth of specific bacteria, namely Prevotella and Klebsiella [47]. A decrease in microbial richness constitutes an alteration in gut microbiome, thus defining dysbiosis. Dysbiosis is thought to induce low-grade inflammation, which in turn can provoke hypertension when the inflammation is persistent [55,56]. Additionally, a reduction in Lactobacillus abundance can induce higher blood pressure values in both mice and humans when compared to healthy controls [57].
Yang et al. recently proved that the Firmicutes on Bacteroidetes ratio was increased in spontaneously hypertensive rats, in angiotensin II-induced hypertensive rats, and in a small group of humans with hypertension [51]. It is noteworthy to note that by normalizing this ratio with the administration of minocycline, blood pressure of spontaneously and induced-hypertensive rats also normalizes [51]. Additionally, fasting for a five-day period seems to induce a modification in the gut microbiota, subsequently reducing blood pressure in hypertensive patients [58].
The gut microbiota produces various metabolites with different effects on blood pressure regulation [59]. Beneficial metabolites include SCFAs and vitamins. Acetate, propionate, and butyrate account for 80% of the total SCFAs produced [60]. SCFAs are thought to be beneficial in blood pressure reduction through mainly their vasorelaxant and anti-inflammatory effects [61]. Indeed, Bartolomaeus et al. demonstrated that the administration of propionate in mouse models was associated with a better control of high blood pressure together with a decrease in vascular inflammation and cardiac damage [62]. In another mouse model, acetate was shown to be highly effective in improving cardiac function by reducing left ventricular wall thickness and body weight in addition to a decrease in systemic blood pressure [63].
By contrast, TMAO, another metabolite produced by the gut microbiome, is positively associated with hypertension [64]. TMAO has a proatherogenic and prothrombotic effect [65,66]. This toxic metabolite is thought to induce hypertension through prolongation of the hypertensive effect of angiotensin II and facilitation of angiotensin II-induced vasoconstriction [67,68]. TMAO also enhances stiffening of the large arteries, namely the aorta and carotid arteries, which amplifies the risk of ASCVD both directly and indirectly through increased systolic blood pressure [69].
Thus, the gut microbiome fabricates different metabolites with various effects on blood pressure. SCFAs improve its control while TMAO is deleterious.

4. Gut Microbiome and Diabetes

In 2019, diabetes was estimated to affect around 463 million people worldwide [70]. That alarming number is expected to rise to 700 million by 2045 [70]. Obesity contributes to the development of type 2 diabetes (T2D) through many pathophysiologic mechanisms, mainly insulin resistance [71]. Diabetes results in microvascular and macrovascular complications, with cardiovascular disease being the most common cause of morbidity and mortality among people suffering from diabetes [72]. Like hypertension, the exact etiology of diabetes remains unclear but many risk factors have been identified, including a positive family history, advanced age, obesity, hypertension, and a history of cardiovascular disease [73,74].
In 2004, Backhed et al. suggested for the first time that the gut microbiota could be linked to the development of T2D by inducing alterations in glucose metabolism [75]. Many studies have reported that obesity and alterations in glucose metabolism were associated with an altered ratio between the two most common bacteria composing the intestine, with increased Bacteroidetes and decreased Firmicutes levels [76,77,78]. Thereafter, experiments using metagenomic sequencing in human volunteers established that people with T2D have a dysbiotic gut microbiota [7,79]. Both trials reported that people with diabetes had less butyrate-producing bacteria. Butyrate is one of the three main SCFAs and is thought to possess an advantageous effect on insulin sensitivity and energy balance [80]. Several studies have followed and have reported that gut microbiome dysbiosis contributes to a less favorable course of T2D by inducing a rapid progression of insulin resistance [81,82,83,84,85].
More than 80% of patients with T2D are overweight [86]. The underlying pathophysiological mechanism linking these two conditions is insulin resistance induced by obesity [87]. Numerous studies from animal models have demonstrated that the gut microbiota is implicated in the development of obesity [75,76,77,88,89]. Several trials have provided an explanation, with one interesting experiment reporting that low bacterial variety in the microbiome is associated with insulin resistance, fatty liver, low-grade inflammation, and obesity when compared to high bacterial diversity [90]. In another experimentation, scientists isolated the microbiota of obese animals and transplanted it into germ-free animals; obesity developed after 14 days [75]. Other trials have focused on the potential role of SCFAs and have found that mice suffering from diabetes exhibit lower levels of butyrate-producing bacteria, such as Fecalibacterium prausnitzii, Eubacterium rectale, and Roseburia intestinalis, when compared to healthy controls [91,92]. Thus, SCFAs, particularly butyrate, are beneficial metabolites that seem to protect against the incidence of diabetes.
In addition, LPS have been found to be early triggers of obesity by inducing an inflammatory state through secretion of cytokines and chemokines [93]. In healthy individuals, the ingestion of a high-fat meal leads to a transitory increase in plasma LPS levels while in patients suffering from obesity and insulin resistance, LPS levels were found to be chronically elevated, thus contributing to the development of T2D [94,95].
Furthermore, recent animal studies have suggested that elevated levels of circulating TMAO are associated with an increased risk of developing T2D, mainly through impaired glucose tolerance, insulin resistance, and oxidative stress [96,97]. Chronic high levels of TMAO are also linked with an increased risk of obesity via secretion of inflammatory cytokines, thus contributing to the occurrence of T2D [98]. A recently published meta-analysis confirmed previous findings and suggested a positive association between T2D and TMAO levels in a dose-dependent manner [99].
Gut microbiota is strongly linked with both microvascular and macrovascular diabetic complications [100,101,102,103]. Indeed, patients with end-stage diabetic nephropathy were found to have an abundance of Haemophilus and Lachnospiraceae bacteria when compared to earlier stages [104]. Furthermore, Pasteurellaceae bacteria are significantly lower in patients with diabetic retinopathy as compared to patients without this complication [105]. Plasma TMAO levels are also significantly increased in individuals with diabetic retinopathy, and its levels are associated with the incidence of this microvascular complication [106,107]. These data highlight the importance of the intestinal microbiome and suggest that dysbiosis could play an important role in the development of diabetic complications.

5. Gut Microbiome and Dyslipidemia

Dyslipidemia is one of the major risk factors for both the occurrence and progression of cardiac diseases [108]. In recent decades, primary and secondary prevention strategies were implemented to decrease cholesterol levels. Despite major improvements, dyslipidemia still affects around 12% of adults in the United States [109]. Some of the identified risk factors for increased cholesterol levels are obesity, lack of physical activity, smoking, an unhealthy diet, and diabetes [110,111,112]. Uncontrolled diabetes is one of the most common conditions contributing to dyslipidemia through insulin resistance, therefore leading to hyperinsulinemia. Elevated insulin concentrations contribute to increases in both low-density lipoprotein-cholesterol (LDL-C) and triglycerides levels in contrast to fewer high-density lipoprotein-cholesterol (HDL-C) particles [113,114,115].
Gut microbiota has been shown to be involved in the occurrence of hyperlipidemia [116]. In fact, a recent study reported that people with dyslipidemia exhibit lower levels of fecal butyrate, acetate, and propionate when compared to healthy controls [117]. These metabolites represent the main SCFAs and are produced by a variety of gut bacteria, such as Bifidobacterium, Lactobacillus, Faecalibacterium prausnitzii, and Roseburia [118]. Indeed, they are thought to protect against obesity and diabetes by improving lipid and glucose homeostasis as well as glucose tolerance [80,119,120,121].
Additionally, in contrast to a lower abundance of SCFAs, patients with high levels of cholesterol excrete feces with a higher quantity of LPS-producing bacteria, such as Escherichia coli and Enterobacter cloacae [118]. LPS compose the cell walls of Gram-negative bacteria and are responsible for the release of pro-inflammatory cytokines [122]. An overproduction of these cytokines leads to increased circulating levels of nitric oxide, subsequently triggering a global activation of inflammatory reactions resulting in cardiac, renal, hepatic, and pulmonary failures [123,124,125].
Furthermore, patients with dyslipidemia tend to exhibit high levels of TMAO, which reduce levels of HDL-C, therefore increasing the risk of ischemic heart disease [126,127]. TMAO was also shown to reduce the expression of cytochrome P450 family 7, subfamily A member 1 (CYP7A1), which is a key enzyme in cholesterol and bile acid metabolism, in addition to inhibiting cholesterol transport, thus inducing cholesterol accumulation in cells [128].
Finally, the gut microbiome is involved in the production of secondary bile acids, which were shown to be protective against the development of dyslipidemia [129,130]. In fact, these metabolites modulate glucose and cholesterol metabolism through the activation of two key receptors, specifically FXR and TGR5 [131,132]. Several studies have established that a deficiency in any one of these receptors, particularly FXR, leads to dyslipidemia, with increased triglycerides and non-HDL-C levels [133,134,135]. In contrast, the activation of FXR by secondary bile acids increases the activity and expression of LDL receptors in addition to an inhibition of the activity of proprotein convertase subtilisin/kexin type 9 (PCSK9) [136,137,138]. Thus, FXR activation by the gut microbiome could lower LDL-C levels and contribute to a better control of dyslipidemia.
Altogether, the previous data suggest that SCFAs and secondary bile acids are protective against the incidence of dyslipidemia while other metabolites, such as LPS and TMAO, are detrimental, contributing to an increase in cholesterol levels.

6. Gut Microbiome and Atherosclerotic Cardiovascular Disease

Even though there has been substantial improvement in cardiovascular disease outcomes in the past few decades, ASCVD remains the leading cause of death around the world [139,140,141]. Insufficient prevention strategies and uncontrolled risk factors are the reasons why cardiac diseases still top the list [139,140]. Uncontrolled dyslipidemia, persistent inflammation, and high levels of oxidative stress greatly contribute to atherosclerosis [142].
Traditionally, ASCVD prevention strategies focused solely on lifestyle modifications, such as eating a healthy diet and doing exercise, in addition to taking beneficial medications, such as aspirin and beta-blockers [143,144,145]. The gut microbiota was neglected until the scientific community realized its importance in playing key functions in the body, thus labeling it “the forgotten organ” [146]. At the beginning of the millennium, a relationship between the microbiota and atherosclerosis was demonstrated for the first time, after multiple studies reported the presence of DNA of numerous bacterial species in a plaque of cholesterol [2,147]. Another trial suggested the presence of dysbiosis in individuals suffering from atherosclerosis, with an abundance of Actinobacteria in their intestine as compared to a large quantity of butyrate-producing bacteria in healthy controls [148].
Recent studies in mice have suggested an important role of the gut microbiota in converting dietary phosphatidylcholine to TMA, which is then oxidized in the liver to TMAO [8,149]. TMAO is a pro-atherosclerotic molecule, with patients suffering from ASCVD displaying significantly higher levels of TMAO as compared with healthy individuals [149]. Several experiments have demonstrated that TMAO was involved in all steps leading to atherogenesis, specifically foam cells’ formations, endothelial dysfunction, thrombus generation, and plaque instability leading, ultimately, to plaque rupture and acute coronary syndrome [8,65,66,150].
Five mechanisms linking TMAO to ASCVD have been recently proposed. Figure 2 summarizes these mechanisms. The first pro-atherosclerotic mechanism is an increased migration of macrophages and an augmented formation of foam cells in cholesterol plaques [151,152]. In a trial involving mice supplemented with either choline or TMAO, Park et al. found that scavenger receptor-A (SR-A) and CD36, two macrophage receptors associated with atherosclerosis, were both increased when compared to control mice [153].
The second metabolic pathway states that TMAO impacts cholesterol metabolism by inhibiting the reverse cholesterol transport (RCT) system and diminishing cholesterol excretion through the biliary system [66]. The RCT system helps maintain cholesterol homeostasis by transporting cholesterol from peripheral tissues to the liver for biliary excretion [154]. A recently published study demonstrated a 35% decrease in the RCT system in mice fed with a diet containing TMAO when compared with healthy controls [66]. TMAO also increases cholesterol levels by downregulating two cytochromes, namely CYP7A1 and CYP27A1. Downregulation of these enzymes leads to decreased bile acid secretion, which reduces cholesterol excretion, thereby contributing to accelerated atherosclerosis [155,156].
The third likely pathway is TMAO-induced endothelial dysfunction [157]. Indeed, TMAO induces vascular inflammation by increasing recruitment of leucocytes to endothelial cells through a G-protein-coupled receptor (GPCR) pathway [158]. TMAO also causes inflammation through mitogen-activated protein kinase (MAPK) and NF-κB signaling pathways [158]. Finally, protein kinase C is a known mediator of endothelial dysfunction, and its activity was found to be significantly increased in response to a diet enhanced with TMAO [159,160].
The fourth pathway involves an increase in oxidative stress. Recent trials have demonstrated that TMAO activates the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome in endothelial cells. Activation of the NLRP3 inflammasome is involved in the production of ROS through activation of the mitochondrial reactive oxygen species signaling pathway [161,162]. Oxidative stress causes cell damage and is involved in the pathogenesis of multiple diseases, including ASCVD [163].
The fifth and last identified pathway mediates TMAO-associated atherosclerosis through suppression of endothelial progenitor cells’ (EPC) production [164]. Several trials have demonstrated that decreased levels of EPCs contribute to endothelial dysfunction, since EPCs are known for their role in repairing and regenerating damaged endothelium following vascular injury [165,166,167]. Chou et al. have found that TMAO levels were proportional to plasmatic inflammatory markers, specifically high-sensitivity C-reactive protein (hsCRP), IL-6, and TNF-α [164]. By contrast, TMAO levels are inversely proportional to EPC levels, thus leading to impaired endothelial function [164].
Other than TMAO, LPS are other pro-atherosclerotic metabolites released by Gram-negative bacteria [168]. In healthy individuals, butyrate is secreted by the gut microbiota in a sufficient amount to maintain the intestinal barrier [169]. In atherosclerosis, gut microbiome dysbiosis results in a reduced number of butyrate-producing bacteria, subsequently leading to increased intestinal permeability and increased LPS levels [170,171]. LPS activate numerous inflammatory pathways that contribute to the occurrence of atherosclerosis. Indeed, LPS induce the generation of ROS by activating nicotinamide adenine dinucleotide phosphate (NADPH) oxidase [172]. NADPH oxidase produces ROS and induces the production of pro-inflammatory cytokines, such as TNF-α, IL-6, and IL-8 [173]. Furthermore, LPS provoke expression of inflammatory mediators, resulting in increased infiltration of inflammatory cells in cholesterol plaques [174]. These inflammatory cells include neutrophils, monocytes, selectins, and integrins, and are involved in the progression of atherosclerosis [175]. Thus, LPS directly contribute to the development and progression of atherosclerosis.
Moreover, the gut microbiome produces secondary bile acids, which are involved in the activation of two key receptors, the membranous TGR5 and the nuclear FXR [130]. Their activation is associated with a slowed progression of atherosclerosis through an inhibition of NF-κB activity, resulting in a decreased production of pro-inflammatory cytokines, as well as inhibited LDL uptake via a lowered expression of CD36 [130]. In contrast, the absence of FXR in mouse models was demonstrated to be linked with a decreased survival rate owing to more severe atherosclerosis with increased atherosclerotic plaque burden [176].
Likewise, SCFAs are thought to be beneficial on ASCVD by inhibiting various inflammatory mechanisms thought to induce atherosclerosis. SCFAs are produced by the gut microbiota through fermentation of dietary fibers [60,177]. Ingestion of a high-fiber diet contributes to an improved glycemic control and weight loss as well as increased blood concentrations of SCFAs [178,179,180]. SCFAs, particularly butyrate, were recently shown to suppress atherosclerotic lesions in mice supplemented with a high-fiber diet [181,182]. Butyrate is thought to increase plaque stability by decreasing ROS and nitric oxide release from macrophages as well as reducing production of known inflammatory molecules, such as chemotaxis protein-1, vascular cell adhesion molecule-1, and matrix metalloproteinase-2 [181,182].
A recently published trial demonstrated that a dysbiotic microbiota is positively associated with an increase in the size of acute myocardial infarction in rats [183]. Probiotics were shown to attenuate the infarct size observed in the dysbiotic group suggesting that microbiota is an important component of ischemic damage. In addition to an increase in infarct size, other noteworthy findings include a higher plasma LPS concentration secondary to increased gut permeability together with an increased Firmicutes to Bacteroidetes ratio.
Thus, dysbiosis contributes to the development of atherosclerosis through increases in TMAO and LPS levels while secondary bile acids and SCFAs are protective. Figure 3 illustrates the implication of the gut microbiome in the occurrence of ASCVD.

7. Conclusions

In the last few decades, major advances have been made in the understanding of physiological and pathological functions of the gut microbiota. In the cardiovascular field, there is no doubt nowadays that the microbiome plays a crucial role in the development of ASCVD. The microbiome is directly involved in all steps leading to atherogenesis, including all major cardiovascular risk factors, specifically hypertension, obesity, diabetes, and dyslipidemia. In the upcoming years, the challenge will be to transition from theoretical understanding to clinical practice as major pathophysiologic mechanisms linking the gut microbiota to ASCVD have been elucidated. In the near future, can an in-depth analysis of the gut microbiota be used as a cardiovascular risk marker for which the use of probiotics might prove beneficial when used adequately?

Author Contributions

Writing, review, and editing: A.A.S., M.P. and G.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The figures were made using Biorender.com (accessed on 5 March 2023).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Khan, M.A.; Hashim, M.J.; Mustafa, H.; Baniyas, M.Y.; Al Suwaidi, S.K.B.M.; AlKatheeri, R.; Alblooshi, F.M.K.; Almatrooshi, M.E.A.H.; Alzaabi, M.E.H.; Al Darmaki, R.S.; et al. Global Epidemiology of Ischemic Heart Disease: Results from the Global Burden of Disease Study. Cureus 2020, 12, e9349. [Google Scholar] [CrossRef]
  2. Haraszthy, V.I.; Zambon, J.J.; Trevisan, M.; Zeid, M.; Genco, R.J. Identification of periodontal pathogens in atheromatous plaques. J. Periodontol. 2000, 71, 1554–1560. [Google Scholar] [CrossRef] [PubMed]
  3. Allin, K.H.; Nielsen, T.; Pedersen, O. Mechanisms in endocrinology: Gut microbiota in patients with type 2 diabetes mellitus. Eur. J. Endocrinol. 2015, 172, R167–R177. [Google Scholar] [CrossRef] [Green Version]
  4. Lepage, P.; Leclerc, M.C.; Joossens, M.; Mondot, S.; Blottière, H.M.; Raes, J.; Ehrlich, D.; Doré, J. A metagenomic insight into our gut’s microbiome. Gut 2013, 62, 146–158. [Google Scholar] [CrossRef]
  5. Flint, H.J.; Scott, K.P.; Louis, P.; Duncan, S.H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 577–589. [Google Scholar] [CrossRef] [PubMed]
  6. Shanahan, F. The gut microbiota—A clinical perspective on lessons learned. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 609–614. [Google Scholar] [CrossRef]
  7. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [Green Version]
  9. Spencer, M.D.; Hamp, T.J.; Reid, R.W.; Fischer, L.M.; Zeisel, S.H.; Fodor, A.A. Association between composition of the human gastrointestinal microbiome and development of fatty liver with choline deficiency. Gastroenterology 2011, 140, 976–986. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Krueger, S.K.; Williams, D.E. Mammalian flavin-containing monooxygenases: Structure/function, genetic polymorphisms and role in drug metabolism. Pharmacol. Ther. 2005, 106, 357–387. [Google Scholar] [CrossRef] [Green Version]
  11. Fennema, D.; Phillips, I.R.; Shephard, E.A. Trimethylamine and Trimethylamine N-Oxide, a Flavin-Containing Monooxygenase 3 (FMO3)-Mediated Host-Microbiome Metabolic Axis Implicated in Health and Disease. Drug. Metab. Dispos. 2016, 44, 1839–1850. [Google Scholar] [CrossRef] [Green Version]
  12. Gatarek, P.; Kaluzna-Czaplinska, J. Trimethylamine N-oxide (TMAO) in human health. EXCLI J. 2021, 20, 301–319. [Google Scholar] [PubMed]
  13. Chiang, J.Y. Bile acid metabolism and signaling. Compr. Physiol. 2013, 3, 1191–1212. [Google Scholar] [PubMed] [Green Version]
  14. Porez, G.; Prawitt, J.; Gross, B.; Staels, B. Bile acid receptors as targets for the treatment of dyslipidemia and cardiovascular disease. J. Lipid. Res. 2012, 53, 1723–1737. [Google Scholar] [CrossRef] [Green Version]
  15. Watanabe, M.; Houten, S.M.; Mataki, C.; Christoffolete, M.A.; Kim, B.W.; Sato, H.; Messaddeq, N.; Harney, J.W.; Ezaki, O.; Kodama, T.; et al. Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation. Nature 2006, 439, 484–489. [Google Scholar] [CrossRef]
  16. Thomas, C.; Gioiello, A.; Noriega, L.; Strehle, A.; Oury, J.; Rizzo, G.; Macchiarulo, A.; Yamamoto, H.; Mataki, C.; Pruzanski, M.; et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell. Metab. 2009, 10, 167–177. [Google Scholar] [CrossRef] [Green Version]
  17. Yoo, J.Y.; Sniffen, S.; McGill Percy, K.C.; Pallaval, V.B.; Chidipi, B. Gut Dysbiosis and Immune System in Atherosclerotic Cardiovascular Disease (ACVD). Microorganisms 2022, 10, 108. [Google Scholar] [CrossRef]
  18. Miyazaki-Anzai, S.; Masuda, M.; Kohno, S.; Levi, M.; Shiozaki, Y.; Keenan, A.L.; Miyazaki, M. Simultaneous inhibition of FXR and TGR5 exacerbates atherosclerotic formation. J. Lipid. Res. 2018, 59, 1709–1713. [Google Scholar] [CrossRef] [Green Version]
  19. Hu, Y.B.; Liu, X.Y.; Zhan, W. Farnesoid. X receptor. agonist. INT-767 attenuates liver steatosis and inflammation in rat model of nonalcoholic steatohepatitis. Drug. Des. Devel. Ther. 2018, 12, 2213–2221. [Google Scholar] [PubMed] [Green Version]
  20. Mullen, L.M.; Chamberlain, G.; Sacre, S. Pattern recognition receptors as potential therapeutic targets in inflammatory rheumatic disease. Arthritis. Res. Ther. 2015, 17, 122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Guha, M.; Mackman, N. LPS induction of gene expression in human monocytes. Cell. Signal. 2001, 13, 85–94. [Google Scholar] [CrossRef]
  22. Gorabi, A.M.; Kiaie, N.; Khosrojerdi, A.; Jamialahmadi, T.; Al-Rasadi, K.; Johnston, T.P.; Sahebkar, A. Implications for the role of lipopolysaccharide in the development of atherosclerosis. Trends. Cardiovasc. Med. 2022, 32, 525–533. [Google Scholar] [CrossRef]
  23. Watts, C.; West, M.A.; Zaru, R. TLR signalling regulated antigen presentation in dendritic cells. Curr. Opin. Immunol. 2010, 22, 124–130. [Google Scholar] [CrossRef]
  24. Karnati, H.K.; Pasupuleti, S.R.; Kandi, R.; Undi, R.B.; Sahu, I.; Kannaki, T.R.; Subbiah, M.; Gutti, R.K. TLR-4 signalling pathway: MyD88 independent pathway up-regulation in chicken breeds upon LPS treatment. Vet. Res. Commun. 2015, 39, 73–78. [Google Scholar] [CrossRef]
  25. Chen, T.; Huang, W.; Qian, J.; Luo, W.; Shan, P.; Cai, Y.; Lin, K.; Wu, G.; Liang, G. Macrophage-derived myeloid differentiation protein 2 plays an essential role in ox-LDL-induced inflammation and atherosclerosis. EBioMedicine 2020, 53, 102706. [Google Scholar] [CrossRef]
  26. Griendling, K.K.; Sorescu, D.; Ushio-Fukai, M. NAD(P)H oxidase: Role in cardiovascular biology and disease. Circ. Res. 2000, 86, 494–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Sweet, M.J.; Hume, D.A. Endotoxin signal transduction in macrophages. J. Leukoc. Biol. 1996, 60, 8–26. [Google Scholar] [CrossRef] [PubMed]
  28. Diks, S.H.; van Deventer, S.J.; Peppelenbosch, M.P. Lipopolysaccharide recognition, internalisation, signalling and other cellular effects. J. Endotoxin. Res. 2001, 7, 335–348. [Google Scholar] [CrossRef] [PubMed]
  29. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory. Bowel. Dis. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  30. Flint, H.J.; Scott, K.P.; Duncan, S.H.; Louis, P.; Forano, E. Microbial degradation of complex carbohydrates in the gut. Gut. Microbes. 2012, 3, 289–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Chambers, E.S.; Preston, T.; Frost, G.; Morrison, D.J. Role of Gut Microbiota-Generated Short-Chain Fatty Acids in Metabolic and Cardiovascular Health. Curr. Nutr. Rep. 2018, 7, 198–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [Green Version]
  33. Zheng, X.X.; Zhou, T.; Wang, X.A.; Tong, X.H.; Ding, J.W. Histone deacetylases and atherosclerosis. Atherosclerosis. 2015, 240, 355–366. [Google Scholar] [CrossRef] [PubMed]
  34. Usami, M.; Kishimoto, K.; Ohata, A.; Miyoshi, M.; Aoyama, M.; Fueda, Y.; Kotani, J. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr. Res. 2008, 28, 321–328. [Google Scholar] [CrossRef] [PubMed]
  35. Ottosson, F.; Brunkwall, L.; Smith, E.; Orho-Melander, M.; Nilsson, P.M.; Fernandez, C.; Melander, O. The gut microbiota-related metabolite phenylacetylglutamine associates with increased risk of incident coronary artery disease. J. Hypertens. 2020, 38, 2427–2434. [Google Scholar] [CrossRef]
  36. Nemet, I.; Saha, P.P.; Gupta, N.; Zhu, W.; Romano, K.A.; Skye, S.M.; Cajka, T.; Mohan, M.L.; Li, L.; Wu, Y.; et al. A Cardiovascular Disease-Linked Gut Microbial Metabolite Acts via Adrenergic Receptors. Cell 2020, 180, 862–877e22. [Google Scholar] [CrossRef]
  37. Poesen, R.; Claes, K.; Evenepoel, P.; de Loor, H.; Augustijns, P.; Kuypers, D.; Meijers, B. Microbiota-Derived Phenylacetylglutamine Associates with Overall Mortality and Cardiovascular Disease in Patients with CKD. J. Am. Soc. Nephrol. 2016, 27, 3479–3487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Liu, Y.; Liu, S.; Zhao, Z.; Song, X.; Qu, H.; Liu, H. Phenylacetylglutamine is associated with the degree of coronary atherosclerotic severity assessed by coronary computed tomographic angiography in patients with suspected coronary artery disease. Atherosclerosis 2021, 333, 75–82. [Google Scholar] [CrossRef] [PubMed]
  39. Camen, S.; Csengeri, D.; Geelhoed, B.; Niiranen, T.; Gianfagna, F.; Vishram-Nielsen, J.K.; Costanzo, S.; Söderberg, S.; Vartiainen, E.; Börschel, C.S.; et al. Risk Factors, Subsequent Disease Onset, and Prognostic Impact of Myocardial Infarction and Atrial Fibrillation. J. Am. Heart. Assoc. 2022, 11, e024299. [Google Scholar] [CrossRef]
  40. Hajar, R. Risk Factors for Coronary Artery Disease: Historical Perspectives. Heart Views 2017, 18, 109–114. [Google Scholar] [CrossRef]
  41. Wang, Y.; Li, J.; Zheng, X.; Jiang, Z.; Hu, S.; Wadhera, R.K.; Bai, X.; Lu, J.; Wang, Q.; Li, Y.; et al. Risk Factors Associated With Major Cardiovascular Events 1 Year After Acute Myocardial Infarction. JAMA Netw. Open. 2018, 1, e181079. [Google Scholar]
  42. Rabi, D.M.; McBrien, K.A.; Sapir-Pichhadze, R.; Nakhla, M.; Ahmed, S.B.; Dumanski, S.M.; Butalia, S.; Leung, A.A.; Harris, K.C.; Cloutier, L.; et al. Hypertension Canada’s 2020 Comprehensive Guidelines for the Prevention, Diagnosis, Risk Assessment, and Treatment of Hypertension in Adults and Children. Can. J. Cardiol. 2020, 36, 596–624. [Google Scholar] [CrossRef]
  43. Whelton, P.K.; Carey, R.M.; Aronow, W.S.; Casey, D.E., Jr.; Collins, K.J.; Dennison Himmelfarb, C.; DePalma, S.M.; Gidding, S.; Jamerson, K.A.; Jones, D.W.; et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults: Executive Summary: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Hypertension 2018, 71, 1269–1324. [Google Scholar]
  44. Williams, B.; Mancia, G.; Spiering, W.; Agabiti Rosei, E.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur. Heart J. 2018, 39, 3021–3104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Dan, X.; Mushi, Z.; Baili, W.; Han, L.; Enqi, W.; Huanhu, Z.; Shuchun, L. Differential analysis of hypertension-associated intestinal microbiota. Int. J. Med. Sci. 2019, 16, 872–881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. de la Cuesta-Zuluaga, J.; Mueller, N.T.; Alvarez-Quintero, R.; Velásquez-Mejía, E.P.; Sierra, J.A.; Corrales-Agudelo, V.; Carmona, J.A.; Abad, J.M.; Escobar, J.S. Higher fecal short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity, hypertension and cardiometabolic disease risk fac- tors. Nutrients 2018, 11, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Sun, S.; Lulla, A.; Sioda, M.; Winglee, K.; Wu, M.C.; Jacobs, D.R., Jr.; Shikany, J.M.; Lloyd-Jones, D.M.; Launer, L.J.; Fodor, A.A.; et al. Gut microbiota com- position and blood pressure. Hypertension 2019, 73, 998–1006. [Google Scholar] [CrossRef]
  49. Verhaar, B.J.H.; Collard, D.; Prodan, A.; Levels, J.H.M.; Zwinderman, A.H.; Bäckhed, F.; Vogt, L.; Peters, M.J.L.; Muller, M.; Nieuwdorp, M.; et al. Associations between gut microbiota, faecal short-chain fatty acids, and blood pressure across ethnic groups: The HELIUS study. Eur. Heart J. 2020, 41, 4259–4267. [Google Scholar] [CrossRef]
  50. Yan, Q.; Gu, Y.; Li, X.; Yang, W.; Jia, L.; Chen, C.; Han, X.; Huang, Y.; Zhao, L.; Li, P.; et al. Alterations of the gut microbiome in hypertension. Front. Cell. Infect. Microbiol. 2017, 7, 381. [Google Scholar] [CrossRef] [Green Version]
  51. Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal, J.M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J.; et al. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kim, S.; Goel, R.; Kumar, A.; Qi, Y.; Lobaton, G.; Hosaka, K.; Mohammed, M.; Handberg, E.M.; Richards, E.M.; Pepine, C.J.; et al. Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. Clin. Sci. 2018, 132, 701–718. [Google Scholar] [CrossRef] [Green Version]
  53. Huart, J.; Leenders, J.; Taminiau, B.; Descy, J.; Saint-Remy, A.; Daube, G.; Krzesinski, J.M.; Melin, P.; de Tullio, P.; Jouret, F. Gut microbiota and fecal levels of short-chain fatty acids differ upon 24-hour blood pressure levels in men. Hypertension 2019, 74, 1005–1013. [Google Scholar] [CrossRef]
  54. Jackson, M.A.; Verdi, S.; Maxan, M.E.; Shin, C.M.; Zierer, J.; Bowyer, R.C.E.; Martin, T.; Williams, F.M.K.; Menni, C.; Bell, J.T.; et al. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat. Commun. 2018, 9, 2655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Schiffrin, E.L. Immune mechanisms in hypertension and vascular injury. Clin. Sci. 2014, 126, 267–274. [Google Scholar] [CrossRef]
  56. Cotillard, A.; Kennedy, S.P.; Kong, L.C.; Prifti, E.; Pons, N.; Le Chatelier, E.; Almeida, M.; Quinquis, B.; Levenez, F.; Galleron, N.; et al. Dietary intervention impact on gut microbial gene richness. Nature 2013, 500, 585–858. [Google Scholar] [CrossRef] [PubMed]
  57. Wilck, N.; Matus, M.G.; Kearney, S.M.; Olesen, S.W.; Forslund, K.; Bartolomaeus, H.; Haase, S.; Mähler, A.; Balogh, A.; Markó, L.; et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature 2017, 551, 585–589. [Google Scholar] [CrossRef] [PubMed]
  58. Maifeld, A.; Bartolomaeus, H.; Löber, U.; Avery, E.G.; Steckhan, N.; Markó, L.; Wilck, N.; Hamad, I.; Šušnjar, U.; Mähler, A.; et al. Fasting alters the gut microbiome reducing blood pressure and body weight in metabolic syndrome patients. Nat. Commun. 2021, 12, 1970. [Google Scholar] [CrossRef]
  59. Wikoff, W.R.; Anfora, A.T.; Liu, J.; Schultz, P.G.; Lesley, S.A.; Peters, E.C.; Siuzdak, G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. USA 2009, 106, 3698–3703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Cummings, J.H.; Pomare, E.W.; Branch, W.J.; Naylor, C.P.; Macfarlane, G.T. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987, 28, 1221–1227. [Google Scholar] [CrossRef] [Green Version]
  61. Marques, F.Z.; Mackay, C.R.; Kaye, D.M. Beyond gut feelings: How the gut microbiota regulates blood pressure. Nat. Rev. Cardiol. 2018, 15, 20–32. [Google Scholar] [CrossRef]
  62. Bartolomaeus, H.; Balogh, A.; Yakoub, M.; Homann, S.; Markó, L.; Höges, S.; Tsvetkov, D.; Krannich, A.; Wundersitz, S.; Avery, E.G.; et al. Short-Chain Fatty Acid Propionate Protects From Hypertensive Cardiovascular Damage. Circulation 2019, 139, 1407–1421. [Google Scholar] [CrossRef] [PubMed]
  63. Marques, F.Z.; Nelson, E.; Chu, P.Y.; Horlock, D.; Fiedler, A.; Ziemann, M.; Tan, J.K.; Kuruppu, S.; Rajapakse, N.W.; El-Osta, A.; et al. High-Fiber Diet and Acetate Supplementation Change the Gut Microbiota and Prevent the Development of Hypertension and Heart Failure in Hypertensive Mice. Circulation 2017, 135, 964–977. [Google Scholar] [CrossRef]
  64. Ge, X.; Zheng, L.; Zhuang, R.; Yu, P.; Xu, Z.; Liu, G.; Xi, X.; Zhou, X.; Fan, H. The gut microbial metabolite trimethylamine N-oxide and hypertension risk: A systematic review and dose-response meta-analysis. Adv. Nutr. 2020, 11, 66–76. [Google Scholar] [CrossRef] [PubMed]
  65. Zhu, W.; Gregory, J.C.; Org, E.; Buffa, J.A.; Gupta, N.; Wang, Z.; Li, L.; Fu, X.; Wu, Y.; Mehrabian, M.; et al. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016, 165, 111–124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Jiang, S.; Shui, Y.; Cui, Y.; Tang, C.; Wang, X.; Qiu, X.; Hu, W.; Fei, L.; Li, Y.; Zhang, S.; et al. Gut microbiota dependent trimethylamine N-oxide aggravates angiotensin II-induced hypertension. Redox. Biol. 2021, 46, 102115. [Google Scholar] [CrossRef]
  68. Ufnal, M.; Jazwiec, R.; Dadlez, M.; Drapala, A.; Sikora, M.; Skrzypecki, J. Trimethylamine-N-oxide: A carnitine-derived metabolite that prolongs the hypertensive effect of angiotensin II in rats. Can. J. Cardiol. 2014, 30, 1700–1705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Brunt, V.E.; Gioscia-Ryan, R.A.; Richey, J.J.; Zigler, M.C.; Cuevas, L.M.; Gonzalez, A.; Vázquez-Baeza, Y.; Battson, M.L.; Smithson, A.T.; Gilley, A.D.; et al. Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice. J. Physiol. 2019, 597, 2361–2378. [Google Scholar] [CrossRef] [Green Version]
  70. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045 Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes. Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [Green Version]
  71. Shuldiner, A.R.; Yang, R.; Gong, D.W. Resistin, obesity, and insulin resistance – the emerging role of the adipocyte as an endocrine organ. N. Engl. J. Med. 2001, 345, 1345–1346. [Google Scholar] [CrossRef] [PubMed]
  72. Matheus, A.S.; Tannus, L.R.; Cobas, R.A.; Palma, C.C.; Negrato, C.A.; Gomes, M.B. Impact of diabetes on cardiovascular disease: An update. Int. J. Hypertens. 2013, 2013, 653789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Fletcher, B.; Gulanick, M.; Lamendola, C. Risk factors for type 2 diabetes mellitus. J. Cardiovasc. Nurs. 2002, 16, 17–23. [Google Scholar] [CrossRef]
  74. Grarup, N.; Sandholt, C.H.; Hansen, T.; Pedersen, O. Genetic susceptibility to type 2 diabetes and obesity: From genome-wide association studies to rare variants and beyond. Diabetologia 2014, 57, 1528–1541. [Google Scholar] [CrossRef] [PubMed]
  75. Backhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut microbiota as an environmental factor that regulates fat storage. PNAS 2004, 101, 15718–15723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
  77. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  78. Chávez-Carbajal, A.; Pizano-Zárate, M.L.; Hernández-Quiroz, F.; Ortiz-Luna, G.F.; Morales-Hernández, R.M.; De Sales-Millán, A.; Hernández-Trejo, M.; García-Vite, A.; Beltrán-Lagunes, L.; Hoyo-Vadillo, C.; et al. Characterization of the Gut Microbiota of Individuals at Different T2D Stages Reveals a Complex Relationship with the Host. Microorganisms 2020, 8, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103. [Google Scholar] [CrossRef] [PubMed]
  80. Gao, Z.; Yin, J.; Zhang, J.; Ward, R.E.; Martin, R.J.; Lefevre, M.; Cefalu, W.T.; Ye, J. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes 2009, 58, 1509–1517. [Google Scholar] [CrossRef] [Green Version]
  81. Kuitunen, M.; Kukkonen, K.; Juntunen-Backman, K.; Korpela, R.; Poussa, T.; Tuure, T.; Haahtela, T.; Savilahti, E. Probiotics prevent IgE-associated allergy until age 5 years in cesarean-delivered children but not in the total cohort. J. Allergy. Clin. Immunol. 2009, 123, 335–341. [Google Scholar] [CrossRef]
  82. McLoughlin, R.M.; Mills, K.H. Influence of gastrointestinal commensal bacteria on the immune responses that mediate allergy and asthma. J. Allergy. Clin. Immunol. 2011, 127, 1097–1107. [Google Scholar] [CrossRef] [PubMed]
  83. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J.; et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Musso, G.; Gambino, R.; Cassader, M. Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes. Annu. Rev. Med. 2011, 62, 361–380. [Google Scholar] [CrossRef]
  85. Sharma, S.; Tripathi, P. Gut microbiome and type 2 diabetes: Where we are and where to go? J. Nutr. Biochem. 2019, 63, 101–108. [Google Scholar] [CrossRef]
  86. Tilg, H.; Moschen, A.R. Microbiota and diabetes: An evolving relationship. Gut 2014, 63, 1513–1521. [Google Scholar] [CrossRef] [PubMed]
  87. Tilg, H.; Moschen, A.R. Inflammatory mechanisms in the regulation of insulin resistance. Mol. Med. 2008, 14, 222–231. [Google Scholar] [CrossRef] [PubMed]
  88. Ley, R.E.; Backhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [Green Version]
  89. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef]
  91. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef]
  92. Wang, L.; Li, C.; Huang, Q.; Fu, X. Polysaccharide from Rosa roxburghii Tratt Fruit Attenuates Hyperglycemia and Hyperlipidemia and Regulates Colon Microbiota in Diabetic db/db Mice. J. Agric. Food Chem. 2020, 68, 147–159. [Google Scholar] [CrossRef]
  93. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772. [Google Scholar] [CrossRef] [Green Version]
  94. Erridge, C.; Attina, T.; Spickett, C.M.; Webb, D.J. A high-fat meal induces low-grade endotoxemia: Evidence of a novel mechanism of postprandial inflammation. Am. J. Clin. Nutr. 2007, 86, 1286–1292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Pussinen, P.J.; Havulinna, A.S.; Lehto, M.; Sundvall, J.; Salomaa, V. Endotoxemia is associated with an increased risk of incident diabetes. Diabetes Care 2011, 34, 392–397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Chen, S.; Henderson, A.; Petriello, M.C.; Romano, K.A.; Gearing, M.; Miao, J.; Schell, M.; Sandoval-Espinola, W.J.; Tao, J.; Sha, B.; et al. Trimethylamine N-Oxide Binds and Activates PERK to Promote Metabolic Dysfunction. Cell. Metab. 2019, 30, 1141–1151.e5. [Google Scholar] [CrossRef]
  97. Naghipour, S.; Cox, A.J.; Peart, J.N.; Du Toit, E.F.; Headrick, J.P. Trimethylamine N-oxide: Heart of the microbiota-CVD nexus? Nutr. Res. Rev. 2021, 34, 125–146. [Google Scholar] [CrossRef] [PubMed]
  98. Dehghan, P.; Farhangi, M.A.; Nikniaz, L.; Nikniaz, Z.; Asghari-Jafarabadi, M. Gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) potentially increases the risk of obesity in adults: An exploratory systematic review and dose-response meta- analysis. Obes. Rev. 2020, 21, e12993. [Google Scholar] [CrossRef]
  99. Zhuang, R.; Ge, X.; Han, L.; Yu, P.; Gong, X.; Meng, Q.; Zhang, Y.; Fan, H.; Zheng, L.; Liu, Z.; et al. Gut microbe-generated metabolite trimethylamine N-oxide and the risk of diabetes: A systematic review and dose-response meta-analysis. Obes. Rev. 2019, 20, 883–894. [Google Scholar] [CrossRef] [PubMed]
  100. Salguero, M.V.; Al-Obaide, M.A.I.; Singh, R.; Siepmann, T.; Vasylyeva, T.L. Dysbiosis of Gram-negative gut microbiota and the associated serum lipopolysaccharide exacerbates inflammation in type 2 diabetic patients with chronic kidney disease. Exp. Ther. Med. 2019, 18, 3461–3469. [Google Scholar] [CrossRef] [Green Version]
  101. Jayasudha, R.; Das, T.; Kalyana Chakravarthy, S.; Sai Prashanthi, G.; Bhargava, A.; Tyagi, M.; Rani, P.K.; Pappuru, R.R.; Shivaji, S. Gut mycobiomes are altered in people with type 2 Diabetes Mellitus and Diabetic Retinopathy. PLoS ONE. 2020, 15, e0243077. [Google Scholar] [CrossRef]
  102. Xie, J.; Song, W.; Liang, X.; Zhang, Q.; Shi, Y.; Liu, W.; Shi, X. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed. Pharmacother. 2020, 127, 110147. [Google Scholar] [CrossRef] [PubMed]
  103. Zhang, Y.; Lu, S.; Yang, Y.; Wang, Z.; Wang, B.; Zhang, B.; Yu, J.; Lu, W.; Pan, M.; Zhao, J.; et al. The diversity of gut microbiota in type 2 diabetes with or without cognitive impairment. Aging Clin. Exp. Res. 2021, 33, 589–601. [Google Scholar]
  104. Du, X.; Liu, J.; Xue, Y.; Kong, X.; Lv, C.; Li, Z.; Huang, Y.; Wang, B. Alteration of gut microbial profile in patients with diabetic nephropathy. Endocrine 2021, 73, 71–84. [Google Scholar] [CrossRef]
  105. Huang, Y.; Wang, Z.; Ma, H.; Ji, S.; Chen, Z.; Cui, Z.; Chen, J.; Tang, S. Dysbiosis and Implication of the Gut Microbiota in Diabetic Retinopathy. Front. Cell. Infect. Microbiol. 2021, 11, 646348. [Google Scholar] [CrossRef] [PubMed]
  106. Liu, W.; Wang, C.; Xia, Y.; Xia, W.; Liu, G.; Ren, C.; Gu, Y.; Li, X.; Lu, P. Elevated plasma trimethylamine-N-oxide levels are associated with diabetic retinopathy. Acta Diabetol. 2021, 58, 221–229. [Google Scholar] [CrossRef] [PubMed]
  107. Yakar, B.; Onalan, E.; Kaymaz, T.; Donder, E.; Gursu, M.F. The role of trimethylamine-N-oxide level in the diagnosis of diabetic retinopathy and the differential diagnosis of diabetic and nondiabetic retinopathy. Arq. Bras. Oftalmol. 2022. [Google Scholar] [CrossRef]
  108. Pol, T.; Held, C.; Westerbergh, J.; Lindbäck, J.; Alexander, J.H.; Alings, M.; Erol, C.; Goto, S.; Halvorsen, S.; Huber, K.; et al. Dyslipidemia and risk of cardiovascular events in patients with atrial fibrillation treated with oral anticoagulation therapy: Insights from the ARISTOTLE (Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation) Trial. J. Am. Heart Assoc. 2018, 7, e007444. [Google Scholar] [CrossRef] [Green Version]
  109. Carroll, M.D.; Fryar, C.D.; Nguyen, D.T. Total and high-density lipoprotein cholesterol in adults: United States, 2015-2016. NCHS Data Brief. 2017, 290, 1–8. [Google Scholar]
  110. Facchini, F.S.; Hollenbeck, C.B.; Jeppesen, J.; Chen, Y.D.; Reaven, G.M. Insulin resistance and cigarette smoking. Lancet 1992, 339, 1128–1130. [Google Scholar] [CrossRef]
  111. Criqui, M.H.; Cowan, L.D.; Tyroler, H.A.; Bangdiwala, S.; Heiss, G.; Wallace, R.B.; Cohn, R. Lipoproteins as mediators for the effects of alcohol consumption and cigarette smoking on cardiovascular mortality: Results from the Lipid Research Clinics Follow-up Study. Am. J. Epidemiol. 1987, 126, 629–637. [Google Scholar] [CrossRef] [PubMed]
  112. Hubert, H.B.; Feinleib, M.; McNamara, P.M.; Castelli, W.P. Obesity as an independent risk factor for cardiovascular disease: A 26-year follow-up of participants in the Framingham Heart Study. Circulation 1983, 67, 968–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zavaroni, I.; Dall’Aglio, E.; Alpi, O.; Bruschi, F.; Bonora, E.; Pezzarossa, A.; Butturini, U. Evidence for an independent relationship between plasma insulin and concentration of high-density lipoprotein cholesterol and triglyceride. Atherosclerosis 1985, 55, 259–266. [Google Scholar] [CrossRef] [PubMed]
  114. Garg, A.; Grundy, S.M. Nicotinic acid as therapy for dyslipidemia in non-insulin-dependent diabetes mellitus. JAMA 1990, 264, 723–726. [Google Scholar] [CrossRef]
  115. Howard, B.V. Insulin resistance and lipid metabolism. Am. J. Cardiol. 1999, 84, 28J–32J. [Google Scholar] [CrossRef]
  116. He, K.; Hu, Y.; Ma, H.; Zou, Z.; Xiao, Y.; Yang, Y.; Feng, M.; Li, X.; Ye, X. Rhizoma Coptidis alkaloids alleviate hyperlipidemia in B6 mice by modulating gut microbiota and bile acid pathways. Biochim. Biophys. Acta 2016, 1862, 1696–1709. [Google Scholar] [CrossRef] [PubMed]
  117. Gargari, G.; Deon, V.; Taverniti, V.; Gardana, C.; Denina, M.; Riso, P.; Guardamagna, O.; Guglielmetti, S. Evidence of dysbiosis in the intestinal microbial ecosystem of children and adolescents with primary hyperlipidemia and the potential role of regular hazelnut intake. FEMS. Microbiol. Ecol. 2018, 94, fiy045. [Google Scholar] [CrossRef]
  118. Moreno-Indias, I.; Sánchez-Alcoholado, L.; Pérez-Martínez, P.; Andrés-Lacueva, C.; Cardona, F.; Tinahones, F.; Queipo-Ortuño, M.I. Red wine polyphenols modulate fecal microbiota and reduce markers of the metabolic syndrome in obese patients. Food Funct. 2016, 7, 1775–1787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Yamashita, H.; Fujisawa, K.; Ito, E.; Idei, S.; Kawaguchi, N.; Kimoto, M.; Hiemori, M.; Tsuji, H. Improvement of obesity and glucose tolerance by acetate in Type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Biosci. Biotechnol. Biochem. 2007, 71, 1236–1243. [Google Scholar] [CrossRef]
  120. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [Green Version]
  121. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Tucureanu, M.M.; Rebleanu, D.; Constantinescu, C.A.; Deleanu, M.; Voicu, G.; Butoi, E.; Calin, M.; Manduteanu, I. Lipopolysaccharide-induced inflammation in monocytes/macrophages is blocked by liposomal delivery of Gi-protein inhibitor. Int. J. Nanomed. 2017, 13, 63–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Feihl, F.; Waeber, B.; Liaudet, L. Is nitric oxide overproduction the target of choice for the management of septic shock? Pharmacol. Ther. 2001, 91, 179–213. [Google Scholar]
  124. Weigand, M.A.; Hörner, C.; Bardenheuer, H.J.; Bouchon, A. The systemic inflammatory response syndrome. Best. Pract. Res. Clin. Anaesthesiol. 2004, 18, 455–475. [Google Scholar] [CrossRef]
  125. Wang, H.; Xu, T.; Lewin, M.R. Future possibilities for the treatment of septic shock with herbal components. Am. J. Emerg. Med. 2009, 27, 107–112. [Google Scholar] [CrossRef] [PubMed]
  126. Agerholm-Larsen, B.; Nordestgaard, B.G.; Steffensen, R.; Jensen, G.; Tybjaerg-Hansen, A. Elevated HDL cholesterol is a risk factor for ischemic heart disease in white women when caused by a common mutation in the cholesteryl ester transfer protein gene. Circulation 2000, 101, 1907–1912. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Qi, J.; You, T.; Li, J.; Pan, T.; Xiang, L.; Han, Y.; Zhu, L. Circulating trimethylamine N-oxide and the risk of cardiovascular diseases: A systematic review and meta-analysis of 11 prospective cohort studies. J. Cell. Mol. Med. 2018, 22, 185–194. [Google Scholar] [CrossRef]
  128. Pathak, P.; Helsley, R.N.; Brown, A.L.; Buffa, J.A.; Choucair, I.; Nemet, I.; Gogonea, C.B.; Gogonea, V.; Wang, Z.; Garcia-Garcia, J.C.; et al. Small molecule inhibition of gut microbial choline trimethylamine lyase activity alters host cholesterol and bile acid metabolism. Am. J. Physiol. Heart. Circ. Physiol. 2020, 318, H1474–H1486. [Google Scholar] [CrossRef]
  129. Dabke, K.; Hendrick, G.; Devkota, S. The gut microbiome and metabolic syndrome. J. Clin. Investig. 2019, 129, 4050–4057. [Google Scholar] [CrossRef]
  130. Matey-Hernandez, M.L.; Williams, F.M.K.; Potter, T.; Valdes, A.M.; Spector, T.D.; Menni, C. Genetic and microbiome influence on lipid metabolism and dyslipidemia. Physiol. Genom. 2018, 50, 117–126. [Google Scholar] [CrossRef] [Green Version]
  131. Kaska, L.; Sledzinski, T.; Chomiczewska, A.; Dettlaff-Pokora, A.; Swierczynski, J. Improved glucose metabolism following bariatric surgery is associated with increased circulating bile acid concentrations and remodeling of the gut microbiome. World J. Gastroenterol. 2016, 22, 8698–8719. [Google Scholar] [CrossRef] [PubMed]
  132. Ryan, K.K.; Tremaroli, V.; Clemmensen, C.; Kovatcheva-Datchary, P.; Myronovych, A.; Karns, R.; Wilson-Pérez, H.E.; Sandoval, D.A.; Kohli, R.; Bäckhed, F.; et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature 2014, 509, 183–188. [Google Scholar] [CrossRef] [Green Version]
  133. Lambert, G.; Amar, M.J.; Guo, G.; Brewer, H.B.; Gonzalez, F.J., Jr.; Sinal, C.J. The farnesoid X-receptor is an essential regulator of cholesterol homeostasis. J. Biol. Chem. 2003, 278, 2563–2570. [Google Scholar] [CrossRef] [Green Version]
  134. Sinal, C.J.; Tohkin, M.; Miyata, M.; Ward, J.M.; Lambert, G.; Gonzalez, F.J. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 2000, 102, 731–744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Cariou, B.; van Harmelen, K.; Duran-Sandoval, D.; van Dijk, T.H.; Grefhorst, A.; Abdelkarim, M.; Caron, S.; Torpier, G.; Fruchart, J.C.; Gonzalez, F.J.; et al. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J. Biol. Chem. 2006, 281, 11039–11049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Taniguchi, T.; Chen, J.; Cooper, A.D. Regulation of cholesterol 7 alpha-hydroxylase gene expression in Hep-G2 cells. Effect of serum, bile salts, and coordinate and noncoordinate regulation with other sterol-responsive genes. J. Biol. Chem. 1994, 269, 10071–10078. [Google Scholar] [CrossRef] [PubMed]
  137. Nakahara, M.; Fujii, H.; Maloney, P.R.; Shimizu, M.; Sato, R. Bile acids enhance low density lipoprotein receptor gene expression via a MAPK cascade-mediated stabilization of mRNA. J. Biol. Chem. 2002, 277, 37229–37234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Langhi, C.; Le May, C.; Kourimate, S.; Caron, S.; Staels, B.; Krempf, M.; Costet, P.; Cariou, B. Activation of the farnesoid X receptor represses PCSK9 expression in human hepatocytes. FEBS. Lett. 2008, 582, 949–955. [Google Scholar] [CrossRef] [Green Version]
  139. Weir, H.K.; Anderson, R.N.; Coleman King, S.M.; Soman, A.; Thompson, T.D.; Hong, Y.; Moller, B.; Leadbetter, S. Heart disease and cancer deaths-trends and projections in the United States, 1969-2020. Prev. Chronic. Dis. 2016, 13, E157. [Google Scholar]
  140. Johnson, N.B.; Hayes, L.D.; Brown, K.; Hoo, E.C.; Ethier, K.A.; Centers for Disease Control and Prevention (CDC). CDC National Health Report: Leading causes of morbidity and mortality and associated behavioral risk and protective factors-United States, 2005–2013. MMWR Suppl. 2014, 63, 3–27. [Google Scholar]
  141. Xu, J.; Murphy, S.L.; Kochanek, K.D.; Arias, E. Mortality in the United States, 2015. NCHS Data Brief. 2016, 267, 1–8. [Google Scholar]
  142. Rader, D.J.; Daugherty, A. Translating molecular discoveries into new therapies for atherosclerosis. Nature 2008, 451, 904–913. [Google Scholar] [CrossRef]
  143. Mancini, G.B.; Gosselin, G.; Chow, B.; Stone, J.; Yvorchuk, K.J.; Abramson, B.L.; Cartier, R.; Huckell, V.; Tardif, J.C.; Connelly, K.; et al. Canadian Cardiovascular Society guidelines for the diagnosis and management of stable ischemic heart disease. Can. J. Cardiol. 2014, 30, 837–849. [Google Scholar] [CrossRef] [PubMed]
  144. Collet, J.P.; Thiele, H.; Barbato, E.; Barthélémy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T.; et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur. Heart J. 2021, 42, 1289–1367. [Google Scholar] [CrossRef]
  145. Gulati, M.; Levy, P.D.; Mukherjee, D.; Amsterdam, E.; Bhatt, D.L.; Birtcher, K.K.; Blankstein, R.; Boyd, J.; Bullock-Palmer, R.P.; Conejo, T.; et al. 2021 AHA/ACC/ASE/CHEST/SAEM/SCCT/SCMR Guideline for the Evaluation and Diagnosis of Chest Pain: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines Circulation. J. Am. Coll. Cardiol. 2021, 144, e368–e454. [Google Scholar]
  146. O’Hara, A.M.; Shanahan, F. The gut flora as a forgotten organ. EMBO. Rep. 2006, 7, 688–693. [Google Scholar] [CrossRef] [Green Version]
  147. Ott, S.J.; El Mokhtari, N.E.; Musfeldt, M.; Hellmig, S.; Freitag, S.; Rehman, A.; Kühbacher, T.; Nikolaus, S.; Namsolleck, P.; Blaut, M.; et al. Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation 2006, 113, 929–937. [Google Scholar] [CrossRef] [Green Version]
  148. Karlsson, F.H.; Fak, 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Fu, Q.; Zhao, M.; Wang, D.; Hu, H.; Guo, C.; Chen, W.; Li, Q.; Zheng, L.; Chen, B. Coronary Plaque Characterization Assessed by Optical Coherence Tomography and Plasma Trimethylamine-N-oxide Levels in Patients With Coronary Artery Disease. Am. J. Cardiol. 2016, 118, 1311–1315. [Google Scholar] [CrossRef]
  151. Zeisel, S.H.; Warrier, M. Trimethylamine N-Oxide, the Microbiome, and Heart and Kidney Disease. Annu. Rev. Nutr. 2017, 37, 157–181. [Google Scholar] [CrossRef]
  152. Wolf, D.; Ley, K. Immunity and Inflammation in Atherosclerosis. Circ. Res. 2019, 124, 315–327. [Google Scholar] [CrossRef]
  153. Park, Y.M. CD36, a scavenger receptor implicated in atherosclerosis. Exp. Mol. Med. 2014, 46, e99. [Google Scholar] [CrossRef] [Green Version]
  154. Ohashi, R.; Mu, H.; Wang, X.; Yao, Q.; Chen, C. Reverse cholesterol transport and cholesterol efflux in atherosclerosis. QJM. 2005, 98, 845–856. [Google Scholar] [CrossRef]
  155. Charach, G.; Rabinovich, A.; Argov, O.; Weintraub, M.; Rabinovich, P. The role of bile acid excretion in atherosclerotic coronary artery disease. Int. J. Vasc. Med. 2012, 2012, 949672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Lu, Y.; Feskens, E.J.; Boer, J.M.; Müller, M. The potential influence of genetic variants in genes along bile acid and bile metabolic pathway on blood cholesterol levels in the population. Atherosclerosis 2010, 210, 14–27. [Google Scholar] [CrossRef]
  157. Zheng, Y.; He, J.Q. Pathogenic Mechanisms of Trimethylamine N-Oxide-induced Atherosclerosis and Cardiomyopathy. Curr. Vasc. Pharmacol. 2022, 20, 29–36. [Google Scholar] [CrossRef] [PubMed]
  158. Seldin, M.M.; Meng, Y.; Qi, H.; Zhu, W.; Wang, Z.; Hazen, S.L.; Lusis, A.J.; Shih, D.M. Trimethylamine N-Oxide Promotes Vascular Inflammation Through Signaling of Mitogen-Activated Protein Kinase and Nuclear Factor-κB. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Ma, G.; Pan, B.; Chen, Y.; Guo, C.; Zhao, M.; Zheng, L.; Chen, B. Trimethylamine N-oxide in atherogenesis: Impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci. Rep. 2017, 37, BSR20160244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Durpès, M.C.; Morin, C.; Paquin-Veillet, J.; Beland, R.; Paré, M.; Guimond, M.O.; Rekhter, M.; King, G.L.; Geraldes, P. PKC-β activation inhibits IL-18-binding protein causing endothelial dysfunction and diabetic atherosclerosis. Cardiovasc. Res. 2015, 106, 303–313. [Google Scholar] [CrossRef]
  161. Zhou, X.; Chen, M.; Zeng, X.; Yang, J.; Deng, H.; Yi, L.; Mi, M.T. Resveratrol regulates mitochondrial reactive oxygen species homeostasis through Sirt3 signaling pathway in human vascular endothelial cells. Cell. Death. Dis. 2014, 5, e1576. [Google Scholar] [CrossRef] [Green Version]
  162. Chen, M.L.; Zhu, X.H.; Ran, L.; Lang, H.D.; Yi, L.; Mi, M.T. Trimethylamine-N-Oxide Induces Vascular Inflammation by Activating the NLRP3 Inflammasome Through the SIRT3-SOD2-mtROS Signaling Pathway. J. Am. Heart Assoc. 2017, 6, e006347. [Google Scholar] [CrossRef]
  163. Ray, P.D.; Huang, B.W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell. Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef] [Green Version]
  164. Chou, R.H.; Chen, C.Y.; Chen, I.C.; Huang, H.L.; Lu, Y.W.; Kuo, C.S.; Chang, C.C.; Huang, P.H.; Chen, J.W.; Lin, S.J. Trimethylamine N-Oxide, Circulating Endothelial Progenitor Cells, and Endothelial Function in Patients with Stable Angina. Sci. Rep. 2019, 9, 4249. [Google Scholar] [CrossRef] [Green Version]
  165. Toya, T.; Ozcan, I.; Corban, M.T.; Sara, J.D.; Marietta, E.V.; Ahmad, A.; Horwath, I.E.; Loeffler, D.L.; Murray, J.A.; Lerman, L.O.; et al. Compositional change of gut microbiome and osteocalcin expressing endothelial progenitor cells in patients with coronary artery disease. PLoS ONE 2021, 16, e0249187. [Google Scholar] [CrossRef]
  166. Michowitz, Y.; Goldstein, E.; Wexler, D.; Sheps, D.; Keren, G.; George, J. Circulating endothelial progenitor cells and clinical outcome in patients with congestive heart failure. Heart 2007, 93, 1046–1050. [Google Scholar] [CrossRef] [Green Version]
  167. Rauscher, F.M.; Goldschmidt-Clermont, P.J.; Davis, B.H.; Wang, T.; Gregg, D.; Ramaswami, P.; Pippen, A.M.; Annex, B.H.; Dong, C.; Taylor, D.A. Aging, progenitor cell exhaustion, and atherosclerosis. Circulation. 2003, 108, 457–463. [Google Scholar] [CrossRef] [Green Version]
  168. Shen, X.; Li, L.; Sun, Z.; Zang, G.; Zhang, L.; Shao, C.; Wang, Z. Gut Microbiota and Atherosclerosis-Focusing on the Plaque Stability. Front. Cardiovasc. Med. 2021, 8, 668532. [Google Scholar] [CrossRef]
  169. Peng, L.; Li, Z.R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef] [Green Version]
  170. Moreira, A.P.B.; Texeira, T.F.S.; Ferreira, A.B.; Peluzio, M.; Peluzio, M.D.C.G.; Alfenas, R.D.C.G. Influence of a high-fat diet on gut microbiota, intestinal permeability and metabolic endotoxaemia. Br. J. Nutr. 2012, 108, 801–809. [Google Scholar] [CrossRef]
  171. Sturm, A.; Dignass, A.U. Epithelial restitution and wound healing in inflammatory bowel disease. World, J. Gastroenterol. 2008, 14, 348–353. [Google Scholar]
  172. Ulevitch, R.J.; Tobias, P.S. Receptor-dependent mechanisms of cell stimulation by bacterial endotoxin. Annu. Rev. Immunol. 1995, 13, 437–457. [Google Scholar] [CrossRef]
  173. Marumo, T.; Schini-Kerth, V.B.; Fisslthaler, B.; Busse, R. Platelet-derived growth factor-stimulated superoxide anion production modulates activation of transcription factor NF-kappaB and expression of monocyte chemoattractant protein 1 in human aortic smooth muscle cells. Circulation. 1997, 96, 2361–2367. [Google Scholar] [CrossRef]
  174. Sawa, Y.; Ueki, T.; Hata, M.; Iwasawa, K.; Tsuruga, E.; Kojima, H.; Ishikawa, H.; Yoshida, S. LPS-induced IL-6, IL-8, VCAM-1, and ICAM-1 expression in human lymphatic endothelium. J. Histochem. Cytochem. 2008, 56, 97–109. [Google Scholar] [CrossRef] [Green Version]
  175. Kim, S.J.; Park, J.H.; Kim, K.H.; Lee, W.R.; Pak, S.C.; Han, S.M.; Park, K.K. The Protective Effect of Apamin on LPS/Fat-Induced Atherosclerotic Mice. Evid. Based Complement. Alternat. Med. 2012, 2012, 305454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Hanniman, E.A.; Lambert, G.; McCarthy, T.C.; Sinal, C.J. Loss of functional farnesoid X receptor increases atherosclerotic lesions in apolipoprotein E-deficient mice. J. Lipid. Res. 2005, 46, 2595–2604. [Google Scholar] [CrossRef] [Green Version]
  177. Macfarlane, G.T.; Macfarlane, S. Bacteria, colonic fermentation, and gastrointestinal health. J. AOAC. Int. 2012, 95, 50–60. [Google Scholar] [CrossRef]
  178. Trompette, A.; Gollwitzer, E.S.; Yadava, K.; Sichelstiel, A.K.; Sprenger, N.; Ngom-Bru, C.; Blanchard, C.; Junt, T.; Nicod, L.P.; Harris, N.L.; et al. Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis. Nat. Med. 2014, 20, 159–166. [Google Scholar] [CrossRef] [PubMed]
  179. Howarth, N.C.; Saltzman, E.; Roberts, S.B. Dietary fiber and weight regulation. Nutr. Rev. 2001, 59, 129–139. [Google Scholar] [CrossRef]
  180. Silva, F.M.; Kramer, C.K.; de Almeida, J.C.; Steemburgo, T.; Gross, J.L.; Azevedo, M.J. Fiber intake and glycemic control in patients with type 2 diabetes mellitus: A systematic review with meta-analysis of randomized controlled trials. Nutr. Rev. 2013, 71, 790–801. [Google Scholar] [CrossRef] [PubMed]
  181. Aguilar, E.C.; Leonel, A.J.; Teixeira, L.G.; Silva, A.R.; Silva, J.F.; Pelaez, J.M.; Capettini, L.S.; Lemos, V.S.; Santos, R.A.; Alvarez-Leite, J.I. Butyrate impairs atherogenesis by reducing plaque inflammation and vulnerability and decreasing NFκB activation. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 606–613. [Google Scholar] [CrossRef]
  182. Aguilar, E.C.; Santos, L.C.; Leonel, A.J.; de Oliveira, J.S.; Santos, E.A.; Navia-Pelaez, J.M.; da Silva, J.F.; Mendes, B.P.; Capettini, L.S.; Teixeira, L.G.; et al. Oral butyrate reduces oxidative stress in atherosclerotic lesion sites by a mechanism involving NADPH oxidase down-regulation in endothelial cells. J. Nutr. Biochem. 2016, 34, 99–105. [Google Scholar] [CrossRef] [PubMed]
  183. Gagné, M.A.; Barbeau, C.; Frégeau, G.; Gilbert, K.; Mathieu, O.; Auger, J.; Tompkins, T.A.; Charbonney, E.; Godbout, R.; Rousseau, G. Dysbiotic microbiota contributes to the extent of acute myocardial infarction in rats. Sci. Rep. 2022, 12, 16517. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Major metabolic pathways involving the gut microbiome and leading to the development and progression of atherosclerotic cardiovascular disease. FXR: farnesoid X receptor, Gln: glutamine, IL-1: interleukin-1, IL-6: interleukin-6, IL-8: interleukin-8, LPS: lipopolysaccharides, NF-κB: nuclear factor-κB, SCFAs: short-chain fatty acids, TGR5: takeda G protein-coupled receptor 5, TLR: toll-like receptor, TMA: trimethylamine, TMAO: trimethylamine N-oxide, TNF-α: tumor necrosis factor-α.
Figure 1. Major metabolic pathways involving the gut microbiome and leading to the development and progression of atherosclerotic cardiovascular disease. FXR: farnesoid X receptor, Gln: glutamine, IL-1: interleukin-1, IL-6: interleukin-6, IL-8: interleukin-8, LPS: lipopolysaccharides, NF-κB: nuclear factor-κB, SCFAs: short-chain fatty acids, TGR5: takeda G protein-coupled receptor 5, TLR: toll-like receptor, TMA: trimethylamine, TMAO: trimethylamine N-oxide, TNF-α: tumor necrosis factor-α.
Ijms 24 05420 g001
Figure 2. Proposed mechanisms linking trimethylamine N-oxide to atherosclerotic cardiovascular disease. HDL-C: high-density lipoprotein-cholesterol, RCT: reverse cholesterol transport, ROS: reactive oxygen species, TMAO: trimethylamine N-oxide.
Figure 2. Proposed mechanisms linking trimethylamine N-oxide to atherosclerotic cardiovascular disease. HDL-C: high-density lipoprotein-cholesterol, RCT: reverse cholesterol transport, ROS: reactive oxygen species, TMAO: trimethylamine N-oxide.
Ijms 24 05420 g002
Figure 3. Role of the gut microbiome in the incidence of atherosclerotic cardiovascular disease. A low-fiber diet is associated with a decreased production of the short-chain fatty acid butyrate, subsequently aggravating dysbiosis as well as sustaining local and systemic inflammation through leakage of bacterial toxins, notably LPS. A modern western diet rich in red meat promotes bacterial production of TMA, which is then oxidized to the pro-atherosclerotic metabolite TMAO in the liver. FMO3: flavin-containing monooxygenase 3, LPS: lipopolysaccharides, TMA: trimethylamine, TMAO: trimethylamine N-oxide.
Figure 3. Role of the gut microbiome in the incidence of atherosclerotic cardiovascular disease. A low-fiber diet is associated with a decreased production of the short-chain fatty acid butyrate, subsequently aggravating dysbiosis as well as sustaining local and systemic inflammation through leakage of bacterial toxins, notably LPS. A modern western diet rich in red meat promotes bacterial production of TMA, which is then oxidized to the pro-atherosclerotic metabolite TMAO in the liver. FMO3: flavin-containing monooxygenase 3, LPS: lipopolysaccharides, TMA: trimethylamine, TMAO: trimethylamine N-oxide.
Ijms 24 05420 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Al Samarraie, A.; Pichette, M.; Rousseau, G. Role of the Gut Microbiome in the Development of Atherosclerotic Cardiovascular Disease. Int. J. Mol. Sci. 2023, 24, 5420. https://doi.org/10.3390/ijms24065420

AMA Style

Al Samarraie A, Pichette M, Rousseau G. Role of the Gut Microbiome in the Development of Atherosclerotic Cardiovascular Disease. International Journal of Molecular Sciences. 2023; 24(6):5420. https://doi.org/10.3390/ijms24065420

Chicago/Turabian Style

Al Samarraie, Ahmad, Maxime Pichette, and Guy Rousseau. 2023. "Role of the Gut Microbiome in the Development of Atherosclerotic Cardiovascular Disease" International Journal of Molecular Sciences 24, no. 6: 5420. https://doi.org/10.3390/ijms24065420

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop