Next Article in Journal
Self-Locking in Collapsed Carbon Nanotube Stacks via Molecular Dynamics
Previous Article in Journal
Toxicological Evaluation of Kaempferol and Linearolactone as Treatments for Amoebic Liver Abscess Development in Mesocricetus auratus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation

Department of Microbiology, Stellenbosch University, Stellenbosch 7600, South Africa
Int. J. Mol. Sci. 2024, 25(19), 10634; https://doi.org/10.3390/ijms251910634
Submission received: 3 September 2024 / Revised: 26 September 2024 / Accepted: 1 October 2024 / Published: 2 October 2024
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Cardiovascular disease (CVD) may be inherited, as recently shown with the identification of single nucleotide polymorphisms (SNPs or “snips”) on a 250 kb DNA fragment that encodes 92 proteins associated with CVD. CVD is also triggered by microbial dysbiosis, microbial metabolites, metabolic disorders, and inflammatory intestinal epithelial cells (IECs). The epithelial cellular adhesion molecule (Ep-CAM) and trefoil factor 3 (TFF3) peptide keeps the gut wall intact and healthy. Variations in Ep-CAM levels are directly linked to changes in the gut microbiome. Leptin, plasminogen activator inhibitor 1 (PAI1), and alpha-1 acid glycoprotein 1 (AGP1) are associated with obesity and may be used as biomarkers. Although contactin 1 (CNTN1) is also associated with obesity and adiposity, it regulates the bacterial metabolism of tryptophan (Trp) and thus appetite. A decrease in CNTN1 may serve as an early warning of CVD. Short-chain fatty acids (SCFAs) produced by gut microbiota inhibit pro-inflammatory cytokines and damage vascular integrity. Trimethylamine N-oxide (TMAO), produced by gut microbiota, activates inflammatory Nod-like receptors (NLRs) such as Nod-like receptor protein 3 (NLRP3), which increase platelet formation. Mutations in the elastin gene (ELN) cause supra valvular aortic stenosis (SVAS), defined as the thickening of the arterial wall. Many of the genes expressed by human cells are regulated by gut microbiota. The identification of new molecular markers is crucial for the prevention of CVD and the development of new therapeutic strategies. This review summarizes the causes of CVD and identifies possible CVD markers.

1. Introduction

Cardiovascular disease (CVD) is mostly associated with an unhealthy diet that leads to obesity, coronary sickness, stroke, and diabetes, but may also be genetically inherited. However, recent studies have shown that CVD is a complex multifactorial disease that involves metabolic disorders and inflammatory reactions of various organs, including the liver, intestine, and central nervous system (CNS). CVD should thus be defined as a ‘systemic disease’ rather than a single-organ disease. Genomic and metagenomic studies group CVD in the same category as other systemic diseases such as type 2 diabetes [1], inflammatory bowel disease (IBD) [2,3], and certain cancers [4]. Most studies were conducted in isolation, focusing on a specific organ or a group of gut microbiota. Few studies addressed the molecular changes in plasma that develop during interactions between gut microbiota, organs, and the human genome.
The vast majority of human microbiota (97%) is found in the gastrointestinal tract (GIT), largely in the colon [5,6]. More than 90% of the human gut microbiome, close to 2800 species, belong to the phyla Proteobacteria, Firmicutes, Actinobacteria, and Bacteroidetes [7,8,9,10,11,12,13]. The remaining 10% of the gut microbiome comprises Fusobacteria and Verrucomicrobia [14]. Despite numerous studies, 60–80% of gut bacteria have not been cultured. Their phenotypic properties remain unknown, including their role in the GIT, gut homeostasis, regulation of the immune system, maintenance of intestinal epithelial cells (IECs), regulation of neurological and endocrine functions, and CVD [15,16]. Neurotransmitters such as γ-aminobutyric acid (GABA), norepinephrine (NE), serotonin (5-HT), dopamine (DA), short-chain fatty acids (SCFAs) [17], glutamate (Glu), tryptophan (Trp) [18], histamine (His) [19], and secondary bile acids [20] produced by gut bacteria reach the CNS. In turn, signals from the brain communicate with enterochromaffin cells (ECs) and enteroendocrine cells (EECs) in the gut wall and the mucosal immune system [21]. Communication between the gut and the brain (gut–brain axis, GBA) improves the integrity of the gut wall, reduces peripheral inflammation, and inhibits the release of pro-inflammatory cytokines [22]. Signals generated by the hypothalamus, in response to metabolites produced by gut microbiota, reach the pituitary and adrenal glands and communicate with EECs via the hypothalamic–pituitary–adrenal axis (HPA) [23]. The role gut microbiota play in CVD is not well understood.
A genetic study by Zhernakova et al. [24] shed some light on the association between gut microbiota and CVD. The authors performed a protein quantitative trait loci (pQTLs) analysis by testing the single nucleotide polymorphisms (SNPs or “snips”) located within a 250 kb DNA fragment encoding 92 CVD-related proteins. Mapping pQTLs far from the cognate gene (a gene coding for an essential protein constantly produced) or on a different chromosome (trans-pQTLs) yielded 85 disease-associated proteins. Of these, 19 cis-pQTLs (SNPs located within 1 Mb upstream or downstream of the corresponding protein-coding gene’s transcription start site), and 74 trans-pQTLs were associated with complex traits and diseases. At least 10 cis- and 7 trans-regulated proteins were associated with CVD. The analyses of 422 putative CVD-associated SNPs revealed 14 pQTLs, each encoding a protein that may be involved in regulating CVD genes [24]. An example is the trans-pQTL gene KLKB1, which codes for prekallikrein (a serine protease) in the liver [25] (Figure 1). Factor XII, a protease that is mainly produced in the liver, converts prekallikrein to plasma kallikrein (a subgroup of serine proteases found in human urine) that has hypotensive properties and produces more factor XII [26] (Figure 1). Plasma kallikrein and factor XII initiate the early stages of coagulation and blood clotting [26]. The interaction between plasma kallikrein and factor XII also initiates a series of chemical reactions that lead to the release of the nanopeptide bradykinin [27] (Figure 1). The latter increases the permeability of blood vessel walls, leading to leakage and inflammation [26]. Several plasma proteins are linked to the gut microbiome, e.g., plasminogen activator inhibitor (PAI)-1, that decreases fibrinolysis and promotes clot formation; urokinase plasminogen activator (uPA), a serine protease that converts inactive plasminogen into active plasmin; epidermal growth factor receptor (EGFR), a protein involved in cell signaling pathways, cell division, and survival; paraoxonases (PONs), especially PON1, which protects LDL and outer cell membranes against harmful oxidative modifications; tumor necrosis factor receptor 2 (TNF-R2), which has pro- and anti-inflammatory properties protecting oligodendrocytes, cardiomyocytes, and keratinocytes; and insulin-like growth factor (IGF), a hormone that controls the production of growth hormones (GHs) and promotes the growth of bones and tissues [24]. Although an individual’s genetic makeup contributes most strongly to immune-stimulating proteins, the gut microbiome takes the lead in the production of proteins involved in metabolism and obesity-related inflammation [24]. Based on these findings, intestinal health, lipid oxidation, and the CNS play a critical role in CVD. Much more research is required to fully understand the interactions between host (human) genetics and the gut microbiome.
This review focuses on the different factors involved in CVD. The review does not intend to cover all of the aspects in depth, but instead provide the reader with a broad overview of how inflammation may lead to CVD, the influence of eating disorders, and the role gut microbiota and short-chain fatty acids play in inflammation. The possible markers that may be used to detect CVD are pointed out.

2. The Role of Epithelial Cellular Adhesion Molecule (Ep-CAM) and Trefoil Factor 3 (TFF3) in Inflammation and Cardiovascular Disease (CVD)

Two proteins attracting much attention in CVD research are Ep-CAM (epithelial cellular adhesion molecule) and the small peptide trefoil factor 3 (TFF3) secreted by intestinal goblet cells. The function of Ep-CAM is to keep intestinal epithelial cells (IECs) in close contact by acting as a “sticking agent” that also prevents mucosal infections. Ep-CAM-deficient mice developed increased intestinal permeability accompanied by a decrease in ion transport, both associated with CVD [28]. The study of Zhernakova et al. [24] provided evidence for an interplay between genetics, the gut microbiome, and Ep-CAM plasma levels. As much as 26.5% of the variation in Ep-CAM levels is associated with changes in gut microbiota patterns and metabolic processes [24]. Only 7% of the changes in Ep-CAM levels are linked to two trans-pQTL loci [24]. The trans-pQTL locus FUT2 is also associated with changes in plasma vitamin B12 levels, microbial infections, and Crohn’s disease (CD). Changes in Ep-CAM levels affect the impact of FUT2 on butyrate-producing Blautia spp. [24], which has a significant correlation with obesity, diabetes, cancer, and various inflammatory diseases [29]. TFF3 plays a role in repairing the intestinal mucosa [30]. However, more recent research has linked TFF3 to cancer, colitis, gastric ulcers, diabetes mellitus, non-alcoholic fatty liver disease (NAFLD), and abnormalities of the CNS [30]. Despite these observations, the biological functions and specific modes of action of TFF3 in the respective tissues have not been elucidated (for further information, the reader is referred to Yang et al. [30]). Further research on the molecular function of TFF3 in specific diseases may lead to the development of novel therapeutic drugs.

3. Obesity, Proteins Contactin 1 (CNTN1), Notch 3, and Elastin

Obesity, which can be defined as the accumulation of lipids, is often associated with elderly people, insulin resistance, hyperlipidemia, hypertension, metabolic syndrome (MetS), and chronic low-grade inflammations [31]. Of these symptoms, obesity, and MetS are directly linked to increased CVD [32]. Other factors contributing to the risk of developing CVD are excessive alcohol consumption, smoking, exposure to pollutants and carcinogens, inactivity, depression, persistent anxiety, and hereditary and ethnic background [33]. Visceral adipose tissue (VAT), subcutaneous adipose tissue (SAT), pericardial fat, and intrathoracic fat are all associated with metabolic risk, with VAT being the strongest associated with cardiometabolic risk and CVD [32]. The authors identified four biomarkers (leptin, plasminogen activator inhibitor 1 (PAI1), alpha-1 acid glycoprotein 1 (AGP1), and CNTN1) for obesity and adiposity. All except CNTN1 were positively associated with obesity and adiposity [32]. The role of AGP1 and CNTN1 in cardiometabolic disease is ill-defined [32]. The CNTN1 gene (CNTN1) encodes an unknown regulator with a key function in the CNS. There is a clear association between CNTN1 and the bacterial metabolism of Trp [24]. Trp is important in appetite regulation and is linked to the CNS via the GBA [34,35]. The decrease in plasma concentrations of CNTN1 in obese individuals may be used as a marker for the early detection of CVD [36].
Diseases such as hypertension, atherosclerosis, vascular stenosis, and dilation/thinning of the vascular wall that causes aneurysms are characterized by changes in the proliferation and differentiation of vascular smooth muscle cells (VSMCs) [37]. Notch 3 protein, encoded by the gene NOTCH3, plays a pivotal role in maintaining VSMCs surrounding blood vessels and gene transcription regulation [37]. The one domain of Notch 3 remains inside the cell, the middle domain spans across the cell membrane, and the extracellular domain protrudes from the cell to attach to other proteins (ligands) [38]. Upon binding to ligands, the intracellular domain detaches from the rest of the protein, now defined as the Notch intracellular domain (NICD), enters the cell nucleus, and regulates gene transcriptions [38]. NOTCH3 plays an important role in intracellular communication, neural development, and binding of epidermal growth factor (EGF) to its receptor, EGFR. EGF and tumor growth factor α (TGFα) stimulate the division of multiple cell types in the gastrointestinal tract and enhance mucosal healing and angiogenesis [39].
Elastin, one of the most abundant proteins in the human body and a major component of the extracellular matrix, constitutes a large part of blood vessel walls. Mutations in the elastin gene (ELN) cause supra valvular aortic stenosis (SVAS), defined as the thickening of the arterial wall. This leads to increased stenosis of arteries, including coronary arteries [40]. Although deficiencies or the degradation of elastin do not cause atherosclerosis, it has an indirect effect by allowing for the formation of lipid plaques in arteries [41].

4. Lipoproteins and Paraoxonases (PONS)

The oxidation of low-density lipoprotein (LDL or “bad” cholesterol) increases the risk of developing CVD (https://www.cdc.gov/cholesterol/about/ldl-and-hdl-cholesterol-and-triglycerides.html; accessed on 15 August 2024). Oxidation is executed by paraoxonases (PONS) attached to high-density lipoprotein (HDL or “good” cholesterol) [42], as displayed in Figure 2. PON1 and PON3 are associated with HDL. PON1 oxidizes lipid peroxides to prevent their accumulation on LDL (Figure 2). PON1 regulates reverse cholesterol transport and has antioxidative, anti-inflammatory, antiapoptotic, vasodilative, and antithrombotic activities [43]. Studies performed on transgenic PON1 knockout mice have shown that PON1 prevents atherogenesis [42]. The expression of PON1 is downregulated by reactive oxygen species (ROS) [44]. Oxidized LDL (OX-LDL) triggers macrophages to stimulate anti-inflammatory activity and form foam cells with lipids accumulated in their cytoplasm (Figure 2). Cytokines from adipose tissue contribute to plaque formation in the vascular endothelium (Figure 2). HDL protects LDL against oxidative modification, as observed by less LDL lipid peroxidation in the presence of HDL [42] (Figure 2). PON1, PON2, and PON3 are structurally similar but differ in activity. PON1 is a lactonase and ester hydrolase that degrades thiolactones, xenobiotics, e.g., organophosphate esters, unsaturated aliphatic esters, aromatic carboxylic esters, and carbamates [45]. PON2 and PON3 do not degrade xenobiotics, but have strong lactonase activity [46]. PON2 has an antiatherogenic effect, attributed to the lowering of ROS and apoptosis (Figure 2), and protects mitochondria against oxidative stress [47]. PON3 interacts with apolipoprotein A-1 (apoA1)-HDL similar to that described for PON1, and is strongly associated with microbial diversity and their metabolic pathways, especially the biosynthesis of antioxidant vitamin B1 [46,48]. PON3 is the least studied, and further research is required to determine its exact physiological function [48].

5. Gut Microbiota and Inflammation

The gut microbiome of healthy individuals is composed of Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria. However, in individuals diagnosed with CVD, Firmicutes and Bacteroidetes account for approximately 90% of the gut microbiome [49] and they have a higher tendency to produce lipopolysaccharides (LPS) [50]. Hexa-acylated LPS induces inflammation, whereas penta-acylated LPS does not [51]. The binding of microbial LPS and other microbial cell components to Toll-like receptors (TLRs) triggers many inflammatory factors that may lead to CVD. LPS bind to Toll-like receptor 4 (TLR4), bacterial flagella to TLR5, and peptidoglycan to TLR2 [52]. LPS induces inflammation and immune responses that increase intestinal permeability [49]. LPS also promotes the expression of Nod-like receptors (NLRs), especially NLRP3 with its Pyrin domain-containing protein 3 [53]. Upon binding to NLRP3, NLRs activate the nuclear factor kappa-B (NF-κB) or mitogen-activated protein kinase (MAPK) pathways to induce the expression of cytokines and chemokines. This causes a chain reaction inducing an immune inflammatory response [54] leading to CVDs [55,56]. Activation of the TLR4-pathway by LPS leads to the activation of nicotinamide adenine dinucleotide phosphate (NADPH)/ ROS/endothelial nitric oxide synthase (eNOS) and MAPK/NF-κB pathways, leading to endothelial dysfunction and vascular inflammation [57]. A high-fat diet could result in decreased intestinal levels of Gram-positive bifidobacteria and increased LPS-containing gut microbiota, which further leads to obesity, the main risk factor for CVDs [58]. Oxidative stress caused by dysbiosis of gut microbiota promotes the oxidation of LDL to OX-LDL, which inhibits the expression of Endothelial nitric oxide synthase (eNOS), vasoconstriction, and hypertension [59]. Peptidoglycan from bacterial cell walls binds to NOD1 and NOD2, which leads to inflammatory responses and atherosclerosis [56].
Patients with dysbiosis and CVD often develop insulin resistance, lipogenesis, fat accumulation, mitochondrial dysfunction, and systemic or local inflammation [60,61]. These abnormalities lead to altering interactions between gut bacteria and IECs, including the activation of several signaling pathways that regulate host pathophysiological processes such as energy metabolism, local and systemic inflammation, and oxidative stress [62]. Apart from CVD, inflammation may lead to chronic kidney disease (CKD) and angiotensin II-directed hypertension [63,64]. Most research on atherosclerosis and coronary artery disease (CAD) does not differentiate between chronic and acute events. Research on the role gut microbiota plays in acute coronary syndromes needs to be prioritized.

6. Short-Chain Fatty Acids and Bile Acids

In many cases, microbial dysbiosis leads to a decrease in short-chain fatty acids (SCFAs) and the disruption of glucose metabolism. Most SCFAs are produced in the colon by Bifidobacterium, Lactobacillus, Lachnospiraceae, Blautia, Coprococcus, Roseburia, Faecalibacterium, Clostridium, and Eubacterium [65,66]. SCFAs adhere to G-protein-coupled receptors (GPCRs), referred to as free fatty acid receptors (FFARs), and regulate histone deacetylase (HDAC) that deacetylates histones and prevents gene expression. G-protein receptor 43 (GPR43/FFAR2) and GPR41 (FFAR3) are located on the surface of IECs [67], neurons of the enteric nervous system (ENS), portal nerve, and sensory ganglia [68,69]. Since GPR41 regulates host energy and promotes glucose and lipid metabolism, a decrease in SCFAs leads to less activated GPR41 and an accumulation of carbohydrates [70].
SCFAs produced in the GIT play an essential role in promoting the secretion of glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) [71]. GLP-1 increases the secretion of insulin [72], and PYY reduces food intake, inhibits intestinal motility, and stimulates bowel movement [70]. Butyrate and propionate, two of the best-studied SCFAs, inhibit tumor necrosis factor (TNF) and NF-κB signaling pathways [73]. Studies conducted on mice have shown that butyrate controls significant components of the tight junction complex in IECs by acting on NLRs [74]. NLRs are essential in the modulation of inflammatory responses [75]. In mice prone to steatohepatitis, butyrate repaired damage to the intestinal mucosa, upregulated zonulin, and lowered endotoxin levels [76].
Butyrate, and to a lesser extent propionate and acetate, engage with GPR41, GPR43, and GPR109a, leading to the activation of MAPK/extracellular signal-regulated kinase (ERK) and p38MAPK signaling pathways, and the generation of extrathymic forkhead box P3 (FOXP3)+ Treg cells. This limits inflammation in the GIT and regulates physiological processes in colonocytes [77,78,79]. The engagement of SCFAs with GPR41 promotes the production of interleukin (IL)-22 in CD4+ T cells [80]. Butyrate coupled to GPR43 stimulates dendritic cell maturation and induces the activity of retinaldehyde dehydrogenase (RALDH) [81]. The absorption of butyrate to GPR109a enhances retinoic acid production and promotes B cell and IgA responses [82]. SCFAs also signal Th1 cells to activate signal transducer and activator of transcription 3 (STAT3) and mammalian target of rapamycin (mTOR) pathways to produce anti-inflammatory cytokine IL-10 [83]. The role of SCFAs in regulating cytokine production, specifically IL-10 and IL-18, and their role in anti-inflammatory responses are indispensable [78,84]. Additional health-benefitting properties of SCFAs include the control of glucose and cholesterol metabolism [85]. In volunteers with type 2 diabetes, dietary-driven microbial production of SCFAs correlated with clinical improvement in blood glucose control, an increase in GLP-1, and a reduction in acetylated hemoglobin [86].
An increase in mucin-degrading bacteria was noted when gnotobiotic mice were fed a low-fiber diet. This resulted in a reduction in mucus thickness and led to barrier dysfunctions [87,88]. A decrease in SCFA production may lead to serious health issues, including a progressive and irreversible reduction in bacterial diversity over successive generations [89,90]. The diminished ability to degrade fibers from plant material may be rectified by replacing the lost bacterial species. One option is to enrich diets with fermented foods [91]. However, it is important to keep in mind that food–microbe interactions differ between individuals on the same diet and are thus highly personalized [92]. Butyrate acts as a helper in lowering diastolic blood pressure (DPB) [93] and reduces the risk of developing CVD [94].
High-fiber diets consisting of whole grains such as millet, maize, brown rice, sorghum, soya, tapioca, nuts, beans, fibrous vegetables, cashews, watermelon, pineapples, and apples are popular in sub-Saharan countries [95]. These diets support gut microbiota that produce glycan-degrading carbohydrate-active enzymes [91]. The degradation of plant fibers in the colon to SCFAs such as butyrate, propionate, and acetate is important to keep IECs and mucosal layers in a healthy state, ensure proper barrier functions [96], and prevent bacterial translocation [97]. Fructan and galactooligosaccharide (GOS)-rich diets stimulate the growth of Bifidobacterium and Lactobacillus [98]. Members of these genera are known for the production of SCFAs and their probiotic properties [99,100]. Cereals stimulate the proliferation of Bacteroides spp. [101]. According to McDonald et al. [102], the gut microbiome of individuals who consumed more than 30 plant types weekly is dominated by SCFA producers such as Facealibacterium prausnitzii and Oscillospira spp. The growth of these species is stimulated by acetate-producing Bifidobacterium and Akkermansia [103]. The composition of a diet determines the type and concentration of SCFAs produced, as shown in a study conducted on ethnic groups in Ghana, Nigeria, Benin, Burkina Faso, Uganda, Kenya, Ethiopia, and South Africa [104]. Findings from this study showed that genera of the order Lactobacillales were dominant in fermented cereal and cassava fermentations. Alcoholic beverages contained species of Lactobacillus, Zymomonas, and Bacillus. Diets containing a variety of ingredients support a greater diversity of gut microbiota, especially the alpha group, reviewed by McDonald et al. [102] and Heiman et al. [105].
Bile acids (BAs), produced as a result of microbial metabolism, prevent CVDs by regulating vascular tension and altering ion exchange through cardiomyocyte membranes [106]. Stimulation of the conversion of BAs to free BAs, and secondary BAs by bile salt hydrolase (BSH) and cholesterol 7-alpha hydroxy-lase (CYP7A1) [107], may thus be an alternative way to treat CVDs. Different types of BAs may have opposite effects on Farnesoid receptors (FXRs), which suggests that the regulation of atherosclerosis may rely on much more comprehensive interactions. G protein-coupled receptors (GPCRs) function as cell surface receptors for BAs. Bile acid coupling GPCR1 (GPBAR1), also known as GPCR19, a membrane-type receptor for bile acids (M-BAR) or Takeda G protein-coupled receptor 5 (TGR5), reduces atherosclerotic plaque formation, and thus also plague inflammation, and inhibits phagocytosis of OX-LDL by macrophages [108,109]. Obeticholic acid (OCA or INT-747), a BA-derived agonist, activates FXR and stimulates TGR5 [110]. INT-767 may thus have beneficial therapeutic properties and may reduce atherosclerotic plaques.
Taurocholate alters the mobilization of calcium ions (Ca2+) and may lead to irregular contractions of cardiac muscle cells (cardiomyocytes) [32]. Ursodeoxycholic acid (UDCA) prevents re-entrant arrhythmias, causing atrial fibrillation, atrioventricular nodal reentrant tachycardia, atrioventricular reciprocating tachycardia, and ventricular tachycardia [111]. Gut microbiota and their metabolites (especially secondary BAs) play crucial roles in hypertension, atherosclerosis, unstable angina, and heart failure.

7. Trimethylamine N-oxide

Trimethylamine N-oxide (TMAO), produced by gut microbiota from choline, phosphatidylcholine, and l-carnitine, causes vascular endothelial damage by activating nitric oxide dismutase (NOD), leucine-rich repeats (LRR) and pyrin domain-containing NLRP3 inflammatory bodies, increases the release of intracellular calcium ions which activates platelet formation [112], and promotes thrombosis [113] and atherosclerosis, thereby increasing the risk of CVDs [114,115]. TMAO also inhibits the sirtuin 3-superoxide dismutase 2-mitochondrial ROS pathway and the ROS-thioredoxin interactive protein axis [116] and activates the protein kinase C/NF-κB (canonical NF-κB)/vascular cell adhesion molecule-1 pathway, all of which promote atherosclerosis [117] (Figure 3). Furthermore, TMAO inhibits the synthesis of BAs and accelerates the formation of aortic lesions by activating FXRs and small heterodimers [118] (Figure 3). TMAO also upregulates CD36 expression and an ATP-binding cassette transporter A1 in macrophages, resulting in the accumulation of cholesterol [119]. From these findings, TMAO induces systemic inflammation that contributes to the progression of CAD. Exosomes secreted by TMAO-stimulated hepatocytes increase the secretion of TNF-α and IL-6, thus promoting inflammation [120]. TMAO can thus be used as a reporter of atherosclerosis, but also as a target for the prevention and treatment of the disease.
A study that involved 2490 patients with chronic heart failure over almost 10 years showed that elevated levels of TMAO were responsible for heart failure with reduced ejection fraction (HFrEF), a condition ascribed to a 40% or less ejection from the left ventricle, accompanied by progressive left ventricular dilatation and adverse changes in the heart’s size and shape [121]. Flavin-containing monooxygenase 3 (FMO3), produced in the liver, catalyzes the formation of TMAO from gut microbe-derived TMA (Figure 3). The inability to oxidize TMA to TMAO present in humans as trimethylaminuria or “fish odor syndrome”, a rare metabolic disease associated with the accumulation of TMA in a patient’s urine, sweat, and breath. Early studies [122,123,124] have shown that primary trimethylaminuria is linked to mutations in the gene encoding FMO3. It is interesting to note that in cases of potential heart failure, gut microbiota produced more N,N,N trimethyl-5-aminovaleric acid (TMAVA) from the TMAO precursor trimethyl lysine (TML) [125]. Coherent with this, a decrease in the oxidation of fatty acids increases the risk of cardiac hypertrophy and mortality.
Patients suffering from atrial fibrillation (AF) have higher levels of TMAO-producing gut microbiota [126]. The reason why AF patients have higher serum TMAO levels is unknown. We do know that TMAO leads to the instability of the electrophysiology in arteries [127]. TMAO also regulates autonomic nerve conduction through the ganglion plexus and enhances the activation of the NF-κB-mediated inflammatory signaling pathways that induce AF [128] (Figure 3). The precursors of TMAO (choline, betaine, and dimethylglycine) may play an important role. Nevertheless, compelling evidence of changes in gut microbiota and the production of their metabolites points to the development of CVDs [129,130]. LPS can stimulate AF by increasing the activity of the NLRP3 inflammasome (Figure 3). Animal studies [131] have shown that LPS also increases the expression of connexin 43 in cardiomyocytes and the expression of L-type Ca2+ channel protein. This decreased myocardium infractions and increased AF. AF may thus be treated by reducing serum levels of LPS. Patients suffering from AF have elevated numbers of Rumenococcus, Streptococcus, Enterococcus, and less Faecali bacterium, Altococcus, Oscillobacter, and Biliophilus [132].

8. Conclusions

Gut microbiota is involved in CVDs, especially through inflammatory responses, changes in gut barrier integrity, and metabolic processes. CVD, likewise, affects the structure and function of the gut microbiome. In the future, metagenomics and metabolomics will play an important role in studying the relationship between gut microbiota and CVDs. Although the epithelial cellular adhesion molecule (Ep-CAM), trefoil factor 3 (TFF3) peptide, leptin, plasminogen activator inhibitor 1 (PAI1), alpha-1 acid glycoprotein 1 (AGP1), contactin 1 (CNTN1), certain short-chain fatty acids (SCFAs), trimethylamine N-oxide (TMAO), Nod-like receptor NLRP3, elastin, and TMAO are possible biomarkers for CVD, gut microbiota and their metabolites should also be explored. The identification of certain immune responses to CVDs must be studied in-depth. The two-way relationship between gut microbiota and CVD is more complex and involves multiple facets of immune regulation, inflammatory responses, gastrointestinal barrier integrity, and metabolic pathways. Therapeutic interventions concerning dysbiosis of the gut microbiota could be an innovative approach to improve the clinical course of CVDs and heart failure.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. 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] [PubMed]
  2. Knights, D.; Silverberg, M.S.; Weersma, R.K.; Gevers, D.; Dijkstra, G.; Huang, H.; Tyler, A.D.; van Sommeren, S.; Imhann, F.; Stempak, J.M.; et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014, 6, 107. [Google Scholar] [CrossRef] [PubMed]
  3. Imhann, F.; Vila, A.V.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119. [Google Scholar] [CrossRef]
  4. Nakatsu, G.; Li, X.; Zhou, H.; Sheng, J.; Wong, S.H.; Wu, W.K.K.; Ng, S.C.; Tsoi, H.; Dong, Y.; Zhang, N.; et al. Gut mucosal microbiome across stages of colorectal carcinogenesis. Nat. Commun. 2015, 6, 8727. [Google Scholar] [CrossRef]
  5. Sender, R.; Fuchs, S.; Milo, R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
  6. Sender, R.; Fuchs, S.; Milo, R. Are we really vastly outnumbered? Revisiting the ratio of bacterial to host cells in humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef] [PubMed]
  7. Huttenhower, C.; Gevers, D.; Knight, R.; Abubucker, S.; Badger, J.H.; Chinwalla, A.T. Human microbiome project consortium: Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar]
  8. Nelson, W.C.; Weinstock, G.M.; Highlander, S.K.; Worley, K.C.; Creasy, H.H.; Wortman, J.R.; Rusch, D.B.; Mitreva, M.; Sodergren, E.; Chinwalla, A.T.; et al. The human microbiome jumpstart reference strains consortium. A catalog of reference genomes from the human microbiome. Science 2010, 328, 994–999. [Google Scholar]
  9. 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]
  10. Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef]
  11. Hugon, P.; Dufour, J.-C.; Colson, P.; Fournier, P.-E.; Sallah, K.; Raoult, D. A comprehensive repertoire of prokaryotic species; identified in human beings. Lancet Infect. Dis. 2015, 15, 1211–1219. [Google Scholar] [CrossRef] [PubMed]
  12. Bilen, M.; Dufour, J.-C.; Lagier, J.-C.; Cadoret, F.; Daoud, Z.; Dubourg, G.; Raoult, D. The contribution of culturomics to the repertoire of isolated human bacterial and archaeal species. Microbiome 2018, 6, 94. [Google Scholar] [CrossRef] [PubMed]
  13. Giovannini, M.G.; Lana, D.; Traini, C.; Vannucchi, M.G. The microbiota–gut–brain axis and Alzheimer disease. From dysbiosis to neurodegeneration: Focus on the central nervous system glial cells. J. Clin. Med. 2021, 10, 2358. [Google Scholar] [CrossRef]
  14. Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef]
  15. Bauer, T.M.; Schwacha, H.; Steinbrückner, B.; E Brinkmann, F.; Ditzen, A.K.; Kist, M.; E Blum, F. Diagnosis of small intestinal bacterial overgrowth in patients with cirrhosis of the liver: Poor performance of the glucose breath hydrogen test. J. Hepatol. 2000, 33, 382–386. [Google Scholar] [CrossRef]
  16. Simren, M.; Stotzer, P.O. Use and abuse of hydrogen breath tests. Gut 2006, 55, 297–303. [Google Scholar] [CrossRef]
  17. Van De Wouw, M.; Boehme, M.; Lyte, J.M.; Wiley, N.; Strain, C.; O’Sullivan, O.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Short-chain fatty acids: Microbial metabolites that alleviate stress-induced brain-gut axis alterations. J. Physiol. 2018, 596, 4923–4944. [Google Scholar] [CrossRef] [PubMed]
  18. Agus, A.; Planchais, J.; Sokol, H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 2018, 23, 716–724. [Google Scholar] [CrossRef]
  19. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  20. MahmoudianDehkordi, S.; Arnold, M.; Nho, K.; Ahmad, S.; Jia, W.; Xie, G.; Louie, G.; Kueider-Paisley, A.; Moseley, M.A.; Thompson, J.W.; et al. Altered bile acid profile associates with cognitive impairment in Alzheimer’s disease—An emerging role for gut microbiome. Alzheimer’s Dement 2019, 15, 76–92. [Google Scholar] [CrossRef]
  21. Baj, A.; Moro, E.; Bistoletti, M.; Orlandi, V.; Crema, F.; Giaroni, C. Glutamatergic signaling along the microbiota-gut-brain axis. Int. J. Mol. Sci. 2019, 20, 1482. [Google Scholar] [CrossRef] [PubMed]
  22. Tracey, K.J.; Chavan, S.S. Nerve Stimulation for Treatment of Diseases and Disorders. International Patent PCT/US2016/018573, 19 February 2016. [Google Scholar]
  23. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef] [PubMed]
  24. Zhernakova, D.V.; Le, T.H.; Kurilshikov, A.; Atanasovska, B.; Bonder, M.J.; Sanna, S.; Claringbould, A.; Võsa, U.; Deelen, P.; Franke, L.; et al. Individual variations in cardiovascular disease-related protein levels are driven by genetics and gut microbiome. Nat. Genet. 2018, 50, 1524–1532. [Google Scholar] [CrossRef]
  25. Bjorkqvist, J.; Jamsa, A.; Renne, T. Plasma kallikrein: The bradykinin-producing enzyme. Thromb. Haemost. 2013, 110, 399–407. [Google Scholar] [CrossRef] [PubMed]
  26. Stavrou, E.; Schmaier, A.H. Factor XII: What does it contribute to our understanding of the physiology and pathophysiology of hemostasis & thrombosis. Thromb. Res. 2010, 125, 210–215. [Google Scholar]
  27. Hofman, Z.; de Maat, S.; Hack, C.E.; Maas, C. Product of the coagulation system. Clin. Rev. Allergy Immunol. 2016, 51, 152–161. [Google Scholar] [CrossRef] [PubMed]
  28. Kozan, P.A.; McGeough, M.D.; Peña, C.A.; Mueller, J.L.; Barrett, K.E.; Marchelletta, R.R.; Sivagnanam, M. Mutation of EpCAM leads to intestinal barrier and ion transport dysfunction. J. Mol. Med. 2015, 93, 535–545. [Google Scholar] [CrossRef]
  29. Liu, X.; Mao, B.; Gu, J.; Wu, J.; Cui, S.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Blautia—A new functional genus with potential probiotic properties? Gut Microbes 2021, 13, 1875796. [Google Scholar] [CrossRef]
  30. Yang, Y.; Lin, Z.; Lin, Q.; Bei, W.; Guo, J. Pathological and therapeutic roles of bioactive peptide trefoil factor 3 in diverse diseases: Recent progress and perspective. Cell Death Dis. 2022, 13, 62. [Google Scholar] [CrossRef]
  31. Trim, W.; Turner, J.E.; Thompson, D. Parallels in Immunometabolic adipose tissue dysfunction with ageing and obesity. Front. Immunol. 2018, 9, 169. [Google Scholar] [CrossRef]
  32. Lau, E.S.; Paniagua, S.M.; Zarbafian, S.; Hoffman, U.; Long, M.T.; Hwang, S.; Courchesne, P.; Yao, C.; Ma, J.; Larson, M.G.; et al. Cardiovascular biomarkers of obesity and overlap with cardiometabolic dysfunction. J. Am. Heart. Assoc. 2021, 10, e020215. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, Z.; Lv, T.; Wang, X.; Wu, M.; Zhang, R.; Yang, X.; Fu, Y.; Liu, Z. Role of the microbiota–gut–heart axis between bile acids and cardiovascular disease. Biomed. Pharmacother. 2024, 174, 116567. [Google Scholar] [CrossRef] [PubMed]
  34. Dicks, L.M.T. Gut bacteria and neurotransmitters. Microorganisms 2022, 10, 1838. [Google Scholar] [CrossRef]
  35. Dicks, L.M.T. How does quorum sensing of intestinal bacteria affect our health and mental status? Microorganisms 2022, 10, 1969. [Google Scholar] [CrossRef]
  36. Yin, X.; Subramanian, S.; Hwang, S.-J.; O’Donnell, C.J.; Fox, C.S.; Courchesne, P.; Muntendam, P.; Gordon, N.; Adourian, A.; Juhasz, P.; et al. Protein biomarkers of new-onset cardiovascular disease: Prospective study from the systems approach to biomarker research in cardiovascular disease initiative. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 939–945. [Google Scholar] [CrossRef]
  37. Malka, K.; Liaw, L. NOTCH3 as a modulator of vascular disease: A target in elastin deficiency and arterial pathologies. J. Clin. Investig. 2022, 132, e157007. [Google Scholar] [CrossRef]
  38. Bray, S.J. Notch Signalling: A simple pathway becomes complex. Nat. Rev. Mol. Cell Biol 2006, 7, 678–689. [Google Scholar] [CrossRef]
  39. Rascher-Eggstein, G.; Liebner, S.; Wolburg, H. The Blood–Brain Barrier in the Human Glioma. In Blood-Spinal Cord and Brain Barriers in Health and Disease; Sharma, H.S., Westman, J., Eds.; Academic Press: Academic Press, San Diego, 2004; pp. 561–576. ISBN 9780126390117. [Google Scholar]
  40. Merla, G.; Brunetti-Pierri, N.; Piccolo, P.; Micale, L.; Loviglio, M.N. Supravalvular aortic stenosis: Elastin arteriopathy. Circ. Cardiovasc. Genet. 2012, 5, 692–696. [Google Scholar] [CrossRef] [PubMed]
  41. Cocciolone, A.J.; Hawes, J.Z.; Staiculescu, M.C.; Johnson, E.O.; Murshed, M.; Wagenseil, J.E. Elastin, arterial mechanics, and cardiovascular disease. Am. J. Physiol. Heart Circ. Physiol. 2018, 315, H189–H205. [Google Scholar] [CrossRef]
  42. Durrington, P.N.; Mackness, B.; Mackness, M.I. Paraoxonase and atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2001, 21, 473–480. [Google Scholar] [CrossRef]
  43. Khattib, A.; Musa, S.; Halabi, M.; Hayek, T.; Khatib, S. Lyso-DGTS. Lipid derivatives enhance PON1 activities and prevent oxidation of LDL: A structure-activity relationship study. Antioxidants 2022, 11, 2058. [Google Scholar] [CrossRef] [PubMed]
  44. Ng, C.J.; Shih, D.M.; Hama, S.Y.; Villa, N.; Navab, M.; Reddy, S.T. The paraoxonase gene family and atherosclerosis. Free. Radic. Biol. Med. 2005, 38, 153–163. [Google Scholar] [CrossRef] [PubMed]
  45. Billecke, S.; Draganov, D.; Counsell, R.; Stetson, P.; Watson, C.; Hsu, C.; La Du, B.N. Human serum paraoxonase (PON1) isozymes Q and R hydrolyze lactones and cyclic carbonate esters. Drug Metab. Dispos. 2000, 28, 1335–1342. [Google Scholar] [PubMed]
  46. Draganov, D.I.; Teiber, J.F.; Speelman, A.; Osawa, Y.; Sunahara, R.; La Du, B.N. Human paraoxonases (PON1, PON2, and PON3) are lactonases with overlapping and distinct substrate specificities. J. Lip. Res. 2005, 46, 1239–1247. [Google Scholar] [CrossRef]
  47. Devarajan, A.; Bourquard, N.; Hama, S.; Navab, M.; Grijalva, V.R.; Morvardi, S.; Clarke, C.F.; Vergnes, L.; Reue, K.; Teiber, J.F.; et al. Paraoxonase 2 deficiency alters mitochondrial function and exacerbates the development of atherosclerosis. Antioxid. Redox. Signal. 2011, 14, 341–351. [Google Scholar] [CrossRef]
  48. Priyanka, K.; Singh, S.; Gill, K. Paraoxonase 3: Structure and its role in pathophysiology of coronary artery disease. Biomolecules 2019, 9, 817. [Google Scholar] [CrossRef]
  49. Wang, L.; Wang, S.; Zhang, Q.; He, C.; Fu, C.; Wei, Q. The role of the gut microbiota in health and cardiovascular diseases. Mol. Biomed. 2022, 3, 30. [Google Scholar] [CrossRef]
  50. Trøseid, M.; Nestvold, T.K.; Rudi, K.; Thoresen, H.; Nielsen, E.W.; Lappegård, K.T. Plasma lipopolysaccharide is closely associated with glycemic control and abdominal obesity. Evidence from bariatric surgery. Diabetes Care 2013, 36, 3627–3632. [Google Scholar] [CrossRef]
  51. Brandsma, E.; Kloosterhuis, N.J.; Koster, M.; Dekker, D.C.; Gijbels, M.J.; van der Velden, S.; Ríos-Morales, M.; van Faassen, M.J.; Loreti, M.G.; de Bruin, A.; et al. A proinflammatory gut microbiota increases systemic inflammation and accelerates atherosclerosis. Circ. Res. 2019, 124, 94–100. [Google Scholar] [CrossRef]
  52. An, H.; Qian, C.; Cao, X. Regulation of Toll-like receptor signaling in the innate immunity. Sci. China Life Sci. 2010, 53, 34–43. [Google Scholar] [CrossRef]
  53. Kienes, I.; Weidl, T.; Mirza, N.; Chamaillard, M.; Kufer, T.A. Role of NLRs in the regulation of type I interferon signaling, host defense and tolerance to inflammation. Int. J. Mol. Sci. 2021, 22, 1301. [Google Scholar] [CrossRef] [PubMed]
  54. Jaén, R.I.; Val-Blasco, A.; Prieto, P.; Gil-Fernández, M.; Smani, T.; López-Sendón, J.L.; Delgado, C.; Boscá, L.; Fernández-Velasco, M. Innate immune receptors, key actors in cardiovascular diseases. JACC Basic Transl. Sci. 2020, 5, 735–749. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, H.-Q.; Zhang, X.-Y.; Edfeldt, K.; Nijhuis, M.O.; Idborg, H.; Bäck, M.; Roy, J.; Hedin, U.; Jakobsson, P.-J.; Laman, J.D.; et al. NOD2-mediated innate immune signaling regulates the eicosanoids in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2193–2201. [Google Scholar] [CrossRef] [PubMed]
  56. Moreno, L.; Gatheral, T. Therapeutic targeting of NOD1 receptors. Br. J. Pharmacol. 2013, 170, 475–485. [Google Scholar] [CrossRef] [PubMed]
  57. Grylls, A.; Seidler, K.; Neil, J. Link between microbiota and hypertension: Focus on LPS/TLR4 pathway in endothelial dysfunction and vascular inflammation, and therapeutic implication of probiotics. Biomed. Pharmacother. 2021, 137, 111334. [Google Scholar] [CrossRef]
  58. Zhi, C.; Huang, J.; Wang, J.; Cao, H.; Bai, Y.; Guo, J.; Su, Z. Connection between gut microbiome and the development of obesity. Eur. J. Clin. Microbiol. Infect. Dis. 2019, 38, 1987–1998. [Google Scholar] [CrossRef]
  59. Subah, P.C. Estrogen protection, oxidized LDL, endothelial dysfunction and vasorelaxation in cardiovascular disease: New insights into a complex issue. Cardiovasc. Res. 2007, 73, 6–7. [Google Scholar]
  60. Rath, E.; Moschetta, A.; Haller, D. Mitochondrial function—Gatekeeper of intestinal epithelial cell homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2018, 158, 497–516. [Google Scholar] [CrossRef]
  61. Tang, W.W.; Wang, Z.; Shrestha, K.; Borowski, A.G.; Wu, Y.; Troughton, R.W.; Klein, A.L.; Hazen, S.L. Intestinal microbiota-dependent phosphatidylcholine metabolites, diastolic dysfunction, and adverse clinical outcomes in chronic systolic heart failure. J. Card. Fail. 2015, 21, 91–96. [Google Scholar] [CrossRef]
  62. Campbell, E.L.; Colgan, S.P. Control and dysregulation of redox signalling in the gastrointestinal tract. Nat. Rev. Gastroenterol. Hepatol. 2018, 16, 106–120. [Google Scholar] [CrossRef]
  63. Kossmann, S.; Schwenk, M.; Hausding, M.; Karbach, S.H.; Schmidgen, M.I.; Brandt, M.; Knorr, M.; Hu, H.; Kröller-Schön, S.; Schönfelder, T.; et al. Angiotensin Ii-induced vascular dysfunction depends on interferon-g- driven immune cell recruitment and mutual activation of monocytes and Nk-cells. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 1313–1319. [Google Scholar] [CrossRef] [PubMed]
  64. Karbach, S.H.; Schönfelder, T.; Brandão, I.; Wilms, E.; Hörmann, N.; Jäckel, S.; Schüler, R.; Finger, S.; Knorr, M.; Lagrange, J.; et al. Gut Microbiota Promote Angiotensin II-Induced Arterial Hypertension and Vascular Dysfunction. J. Am. Heart. Assoc. 2016, 5, e003698. [Google Scholar] [CrossRef] [PubMed]
  65. 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 diseases. Front. Immunol. 2019, 10, 277. [Google Scholar]
  66. Cushing, K.; Alvarado, D.M.; Ciorba, M.A. Butyrate and mucosal inflammation: New scientific evidence supports clinical observation. Clin. Transl. Gastroenterol. 2015, 6, e108. [Google Scholar] [CrossRef]
  67. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
  68. De Vadder, F.; Grasset, E.; Holm, L.M.; Karsenty, G.; MacPherson, A.J.; Olofsson, L.E.; Bäckhed, F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc. Natl. Acad. Sci. USA 2018, 115, 6458–6463. [Google Scholar] [CrossRef]
  69. Egerod, K.L.; Petersen, N.; Timshel, P.N.; Rekling, J.C.; Wang, Y.; Liu, Q.; Schwartz, T.W.; Gautron, L. Profiling of G protein-coupled receptors in vagal afferents reveals novel gut-to-brain sensing mechanisms. Mol. Metab. 2018, 12, 62–75. [Google Scholar] [CrossRef]
  70. Samuel, B.S.; Shaito, A.; Motoike, T.; Rey, F.E.; Backhed, F.; Manchester, J.K.; Hammer, R.E.; Williams, S.C.; Crowley, J.; Yanagisawa, M.; et al. Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, gpr41. Proc. Natl. Acad. Sci. USA 2008, 105, 16767–16772. [Google Scholar] [CrossRef]
  71. Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. 2015, 39, 424–429. [Google Scholar] [CrossRef]
  72. Zhou, J.; Martin, R.J.; Tulley, R.T.; Raggio, A.M.; McCutcheon, K.L.; Shen, L.; Danna, S.C.; Tripathy, S.; Hegsted, M.; Keenan, M.J. Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E1160–E1166. [Google Scholar] [CrossRef]
  73. Liu, T.; Li, J.; Liu, Y.; Xiao, N.; Suo, H.; Xie, K.; Yang, C.; Wu, C. Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-κB pathway in RAW264.7 cells. Inflammation 2012, 35, 1676–1684. [Google Scholar] [CrossRef] [PubMed]
  74. Takeuchi, O.; Akira, S. Pattern Recognition Receptors and Inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed]
  75. Cheng, D.; Xu, J.-H.; Li, J.-Y.; Wang, S.-Y.; Wu, T.-F.; Chen, Q.-K.; Yu, T. Butyrate Ameliorated-NLRC3 Protects the Intestinal Barrier in a GPR43-Dependent Manner. Exp. Cell Res. 2018, 368, 101–110. [Google Scholar] [CrossRef]
  76. Tong, L.-C.; Wang, Y.; Wang, Z.-B.; Liu, W.-Y.; Sun, S.; Li, L.; Su, D.-F.; Zhang, L.-C. Propionate Ameliorates Dextran Sodium Sulfate-Induced Colitis by Improving Intestinal Barrier Function and Reducing Inflammation and Oxidative Stress. Front. Pharmacol. 2016, 7, 253. [Google Scholar] [CrossRef]
  77. 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]
  78. Campos-Perez, W.; Martinez-Lopez, E. Effects of short chain fatty acids on metabolic and inflammatory processes in human health. Biochim Biophys Acta Mol Cell Biol Lipids 2021, 1866, 158900. [Google Scholar] [CrossRef] [PubMed]
  79. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly-Y, M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, shortchain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef]
  80. Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
  81. Tan, J.; McKenzie, C.; Vuillermin, P.J.; Goverse, G.; Vinuesa, C.G.; Mebius, R.E.; Macia, L.; Mackay, C.R. Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways. Cell Rep. 2016, 15, 2809–2824. [Google Scholar] [CrossRef]
  82. Wu, W.; Sun, M.; Chen, F.; Cao, A.; Liu, H.; Zhao, Y.; Huang, X.; Xiao, Y.; Yao, S.; Zhao, Q.; et al. Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal. Immunol. 2017, 10, 946–956. [Google Scholar] [CrossRef]
  83. Sun, M.; Wu, W.; Chen, L.; Yang, W.; Huang, X.; Ma, C.; Chen, F.; Xiao, Y.; Zhao, Y.; Ma, C.; et al. Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat. Commun. 2018, 9, 3555. [Google Scholar] [CrossRef] [PubMed]
  84. Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef]
  85. Armet, A.M.; Deehan, E.C.; Thöne, J.V.; Hewko, S.J.; Walter, J. The effect of isolated and synthetic dietary fibers on markers of metabolic diseases in human intervention studies: A systematic review. Adv. Nutr. 2020, 11, 420–438. [Google Scholar] [CrossRef] [PubMed]
  86. Zhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 2018, 359, 1151–1156. [Google Scholar] [CrossRef] [PubMed]
  87. Desai, M.S.; Seekatz, A.M.; Koropatkin, N.M.; Kamada, N.; Hickey, C.A.; Wolter, M.; Pudlo, N.A.; Kitamoto, S.; Terrapon, N.; Muller, A.; et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 2016, 167, 1339–1353.e21. [Google Scholar] [CrossRef]
  88. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.-I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary prevention of cardiovascular disease with a Mediterranean diet supplemented with extra-virgin olive oil or nuts. New Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef]
  89. Vangay, P.; Johnson, A.J.; Ward, T.L.; Al-Ghalith, G.A.; Shields-Cutler, R.R.; Hillmann, B.M.; Lucas, S.K.; Beura, L.K.; Thompson, E.A.; Till, L.M.; et al. US immigration westernizes the human gut microbiome. Cell 2018, 175, 962–972. [Google Scholar] [CrossRef]
  90. Sonnenburg, E.D.; Smits, S.A.; Tikhonov, M.; Higginbottom, S.K.; Wingreen, N.S.; Sonnenburg, J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016, 529, 212–215. [Google Scholar] [CrossRef] [PubMed]
  91. Wastyk, H.C.; Fragiadakis, G.K.; Perelman, D.; Dahan, D.; Merrill, B.D.; Yu, F.B.; Topf, M.; Gonzalez, C.G.; Van Treuren, W.; Han, S.; et al. Gut-microbiota-targeted diets modulate human immune status. Cell 2021, 184, 4137–4153.e14. [Google Scholar] [CrossRef]
  92. Johnson, A.J.; Vangay, P.; Al-Ghalith, G.A.; Hillmann, B.M.; Ward, T.L.; Shields-Cutler, R.R.; Kim, A.D.; Shmagel, A.K.; Syed, A.N.; Walter, J.; et al. Daily sampling reveals personalized diet-microbiome associations in humans. Cell Host Microbe 2019, 25, 789–802. [Google Scholar] [CrossRef]
  93. Roshanravan, N.; Mahdavi, R.; Alizadeh, E.; Jafarabadi, M.A.; Hedayati, M.; Ghavami, A.; Alipour, S.; Alamdari, N.M.; Barati, M.; Ostadrahimi, A. Effect of butyrate and inulin supplementation on glycemic status, lipid profile and glucagon-like peptide 1 level in patients with type 2 diabetes: A randomized double-blind, placebo-controlled trial. Horm. Metab. Res. 2017, 49, 886–891. [Google Scholar] [CrossRef] [PubMed]
  94. Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Nitert, M.D. Increased systolic and diastolic blood pressure is associated with altered gut microbiota composition and butyrate production in early pregnancy. Hypertension 2016, 68, 974–981. [Google Scholar] [CrossRef] [PubMed]
  95. Isibor, P.; Akinduti, P.; Aworunse, O.; Oyewale, J.; Oshamika, O.; Ugboko, H.; Taiwo, O.; Ahuekwe, E.; Obafemi, Y.; Onibokun, E.; et al. Significance of African diets in biotherapeutic modulation of the gut microbiome. Bioinform. Biol. Insights 2021, 15, 11779322211012697. [Google Scholar] [CrossRef]
  96. Statovci, D.; Aguilera, M.; MacSharry, J.; Melgar, S. The impact of Western diet and nutrients on the microbiota and immune response at mucosal interfaces. Front. Immunol. 2017, 8, 838. [Google Scholar] [CrossRef]
  97. Wrzosek, L.; Miquel, S.; Noordine, M.-L.; Bouet, S.; Chevalier-Curt, M.J.; Robert, V.; Philippe, C.; Bridonneau, C.; Cherbuy, C.; Robbe-Masselot, C.; et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol. 2013, 11, 61. [Google Scholar] [CrossRef]
  98. So, D.; Whelan, K.; Rossi, M.; Morrison, M.; Holtmann, G.; Kelly, J.T.; Shanahan, E.R.; Staudacher, H.M.; Campbell, K.L. Dietary fiber intervention on gut microbiota composition in healthy adults: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2018, 107, 965–983. [Google Scholar] [CrossRef]
  99. Dicks, L.M.T.; Grobbelaar, M.J. Double-barrel shotgun: Probiotic lactic acid bacteria with antiviral properties modified to serve as vaccines. Microorganisms 2021, 9, 1565. [Google Scholar] [CrossRef]
  100. Dicks, L.M.T.; Geldenhuys, J.; Mikkelsen, L.S.; Brandsborg, E.; Marcotte, H. Our gut microbiota: A long walk to homeostasis. Benef. Microbes 2018, 9, 3–19. [Google Scholar] [CrossRef] [PubMed]
  101. Chen, T.; Long, W.; Zhang, C.; Liu, S.; Zhao, L.; Hamaker, B.R. Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota. Sci. Rep. 2017, 7, 2594. [Google Scholar] [CrossRef]
  102. McDonald, D.; Hyde, E.; Debelius, J.W.; Morton, J.T.; Gonzalez, A.; Ackermann, G.; Aksenov, A.A.; Behsaz, B.; Brennan, C.; Chen, Y.; et al. American gut: An open platform for citizen science microbiome research. mSystems 2018, 3, e00031-18. [Google Scholar] [CrossRef]
  103. Blaak, E.E.; Canfora, E.E.; Theis, S.; Frost, G.; Groen, A.K.; Mithieux, G.; Nauta, A.; Scott, K.; Stahl, B.; Van Harsselaar, J.; et al. Short chain fatty acids in human gut and metabolic health. Benef. Microbes 2020, 11, 411–455. [Google Scholar] [CrossRef] [PubMed]
  104. Diaz, M.; Kellingray, L.; Akinyemi, N.; Adefiranye, O.O.; Olaonipekun, A.B.; Bayili, G.R.; Ibezim, J.; du Plessis, A.S.; Houngbédji, M.; Kamya, D.; et al. Comparison of the microbial composition of African fermented foods using amplicon sequencing. Sci. Rep. 2019, 9, 13863. [Google Scholar] [CrossRef] [PubMed]
  105. Heiman, M.L.; Greenway, F.L. A healthy gastrointestinal microbiome is dependent on dietary diversity. Mol. Metab. 2016, 5, 317–320. [Google Scholar] [CrossRef] [PubMed]
  106. Guizoni, D.M.; Vettorazzi, J.F.; Carneiro, E.M.; Davel, A.P. Modulation of endothelium-derived nitric oxide production and activity by taurine and taurine-conjugated bile acids. Nitric Oxide 2020, 94, 48–53. [Google Scholar] [CrossRef]
  107. Callender, C.; Attaye, I.; Nieuwdorp, M. The interaction between the gut microbiome and bile acids in cardiometabolic diseases. Metabolites 2022, 12, 65. [Google Scholar] [CrossRef]
  108. Pols, T.W.; Nomura, M.; Harach, T.; Sasso, G.L.; Oosterveer, M.H.; Thomas, C.; Rizzo, G.; Gioiello, A.; Adorini, L.; Pellicciari, R.; et al. TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading. Cell Metab. 2011, 14, 747–757. [Google Scholar] [CrossRef]
  109. Pols, T.W. TGR5 in inflammation and cardiovascular disease. Biochem. Soc. Trans. 2014, 42, 244–249. [Google Scholar] [CrossRef]
  110. Baghdasaryan, A.; Claudel, T.; Gumhold, J.; Silbert, D.; Adorini, L.; Roda, A.; Vecchiotti, S.; Gonzalez, F.J.; Schoonjans, K.; Strazzabosco, M.; et al. Dual farnesoid X receptor/TGR5 agonist INT-767 reduces liver injury in the Mdr2-/- (Abcb4-/-) mouse cholangiopathy model by promoting biliary HCO⁻₃ output. Hepatology 2011, 54, 1303–1312. [Google Scholar] [CrossRef]
  111. Kaplan, J.; Kanwal, A.; Ahmed, I.; Lala, V. Reentrant Arrhythmias. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. Available online: https://www.ncbi.nlm.nih.gov/books/NBK557775/ (accessed on 2 August 2024).
  112. 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]
  113. 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]
  114. Katsimichas, T.; Antonopoulos, A.S.; Katsimichas, A.; Ohtani, T.; Sakata, Y.; Tousoulis, D. The intestinal microbiota and cardiovascular disease. Cardiovasc. Res. 2019, 115, 1471–1486. [Google Scholar] [CrossRef] [PubMed]
  115. Xie, G.; Yan, A.; Lin, P.; Wang, Y.; Guo, L. Trimethylamine N-oxide-a marker for atherosclerotic vascular disease. Rev. Cardiovasc. Med. 2021, 22, 787–797. [Google Scholar] [CrossRef] [PubMed]
  116. 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, e003698. [Google Scholar] [CrossRef] [PubMed]
  117. 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]
  118. Ding, L.; Chang, M.; Guo, Y.; Zhang, L.; Xue, C.; Yanagita, T.; Zhang, T.; Wang, Y. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids Health Dis. 2018, 17, 286. [Google Scholar] [CrossRef]
  119. Li, Y.; Zhang, L.; Ren, P.; Yang, Y.; Li, S.; Qin, X.; Zhang, M.; Zhou, M.; Liu, W. Qing-xue-xiao-zhi formula attenuates atherosclerosis by inhibiting macrophage lipid accumulation and inflammatory response via TLR4/myd88/NF-κB pathway regulation. Phytomedicine 2021, 93, 153812. [Google Scholar] [CrossRef]
  120. Liu, X.; Shao, Y.; Tu, J.; Sun, J.; Li, L.; Tao, J.; Chen, J. Trimethylamine-N-oxide stimulated hepatocyte-derived exosomes promote inflammation and endothelial dysfunction through nuclear factor-kappa b signaling. Ann. Transl. Med. 2021, 9, 1670. [Google Scholar] [CrossRef]
  121. Schuett, K.; Kleber, M.E.; Scharnagl, H.; Lorkowski, S.; März, W.; Niessner, A.; Marx, N.; Meinitzer, A. Trimethylamine-N-oxide and heart failure with reduced versus preserved ejection fraction. J. Am. Coll. Cardiol. 2017, 70, 3202–3204. [Google Scholar] [CrossRef]
  122. Akerman, B.; Lemass, H.; Chow, L.; Lambert, D.; Greenberg, C.; Bibeau, C.; Mamer, O.; Treacy, E. Trimethylaminuria is caused by mutations of the FMO3 gene in a North American cohort. Mol. Genet. Metab. 1999, 68, 24–31. [Google Scholar] [CrossRef]
  123. Cashman, J.R.; Bi, Y.-A.; Lin, J.; Youil, R.; Knight, M.; Forrest, S.; Treacy, E. Human flavin-containing monooxygenase form 3: cDNA expression of the enzymes containing amino acid substitutions observed in individuals with trimethylaminuria. Chem. Res. Toxicol. 1997, 10, 837–841. [Google Scholar] [CrossRef]
  124. Treacy, E.P.; Akerman, B.R.; Chow, L.M.L.; Youil, R.; Lin, C.B.; Bruce, A.G.; Knight, M.; Danks, D.M.; Cashman, J.R.; Forrest, S.M. Mutations of the flavin-containing monooxygenase gene (FMO3) cause trimethylaminuria, a defect in detoxication. Hum. Mol. Genet. 1998, 7, 839–845. [Google Scholar] [CrossRef] [PubMed]
  125. Zhao, M.; Wei, H.; Li, C.; Zhan, R.; Liu, C.; Gao, J.; Yi, Y.; Cui, X.; Shan, W.; Ji, L.; et al. Gut microbiota production of trimethyl-5-aminovaleric acid reduces fatty acid oxidation and accelerates cardiac hypertrophy. Nat. Commun. 2022, 13, 1757. [Google Scholar] [CrossRef] [PubMed]
  126. Zuo, K.; Liu, X.; Wang, P.; Jiao, J.; Han, C.; Liu, Z.; Yin, X.; Li, J.; Yang, X. Metagenomic data mining reveals enrichment of trimethylamine-N-oxide synthesis in gut microbiome in atrial fibrillation patients. BMC Genom. 2020, 21, 526. [Google Scholar] [CrossRef] [PubMed]
  127. Yu, L.; Meng, G.; Huang, B.; Zhou, X.; Stavrakis, S.; Wang, M.; Li, X.; Zhou, L.; Wang, Y.; Wang, M.; et al. A potential relationship between gut microbes and atrial fibrillation: Trimethylamine N-oxide, a gut microbe-derived metabolite, facilitates the progression of atrial fibrillation. Int. J. Cardiol. 2018, 255, 92–98. [Google Scholar] [CrossRef] [PubMed]
  128. Hou, Y.; Scherlag, B.J.; Lin, J.; Zhang, Y.; Lu, Z.; Truong, K.; Patterson, E.; Lazzara, R.; Jackman, W.M.; Po, S.S. Ganglionated plexi modulate extrinsic cardiac autonomic nerve input: Effects on sinus rate, atrioventricular conduction, refractoriness, and inducibility of atrial fibrillation. J. Am. Coll. Cardiol. 2007, 50, 61–68. [Google Scholar] [CrossRef] [PubMed]
  129. Cui, X.; Ye, L.; Li, J.; Jin, L.; Wang, W.; Li, S.; Bao, M.; Wu, S.; Li, L.; Geng, B.; et al. Metagenomic and metabolomic analyses unveil dysbiosis of gut microbiota in chronic heart failure patients. Sci. Rep. 2018, 8, 635. [Google Scholar] [CrossRef]
  130. 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, 862–877. [Google Scholar] [CrossRef]
  131. Chen, Y.-Y.; Sun, Z.-W.; Jiang, J.-P.; Kang, X.-D.; Wang, L.-L.; Shen, Y.-L.; Xie, X.-D.; Zheng, L.-R. A-adrenoceptor-mediated enhanced inducibility of atrial fibrillation in a canine system inflammation model. Mol. Med. Rep. 2017, 15, 3767–3774. [Google Scholar] [CrossRef]
  132. Zuo, K.; Li, J.; Li, K.; Hu, C.; Gao, Y.; Chen, M.; Hu, R.; Liu, Y.; Chi, H.; Wang, H.; et al. Disordered gut microbiota and alterations in metabolic patterns are associated with atrial fibrillation. Gigascience 2019, 8, giz058. [Google Scholar] [CrossRef]
Figure 1. A basic explanation for the role of plasma kallikrein in inflammation and blot clotting. Prekallikrein is encoded by the trans-pQTL gene KLKB1 in the liver. Factor XII converts prekallikrein to plasma kallikrein and leads to the production of more factor XI. The early stages of blood clotting and the formation of bradykinin are initiated by a combined reaction of plasma kallikrein and factor XII. Bradykinin increases the permeability of blood vessel walls, leading to leakage and inflammation. Trans-pQTL = trans-protein quantitative trait loci. Created using Biorender.com (accessed on 22 August 2024).
Figure 1. A basic explanation for the role of plasma kallikrein in inflammation and blot clotting. Prekallikrein is encoded by the trans-pQTL gene KLKB1 in the liver. Factor XII converts prekallikrein to plasma kallikrein and leads to the production of more factor XI. The early stages of blood clotting and the formation of bradykinin are initiated by a combined reaction of plasma kallikrein and factor XII. Bradykinin increases the permeability of blood vessel walls, leading to leakage and inflammation. Trans-pQTL = trans-protein quantitative trait loci. Created using Biorender.com (accessed on 22 August 2024).
Ijms 25 10634 g001
Figure 2. A schematic representation explaining the role paraoxonases (PON1, PON2, and PON3) play in cardiovascular disease (CVD). PON1 and PON3 are attached to high-density lipoprotein (HDL) and oxidize lipid peroxides to prevent their accumulation on low-density lipoprotein (LDL). Oxidized LDL (OX-LDL) triggers macrophages to an anti-inflammatory state and forms foam cells. Foam cells and the release of pro-inflammatory cytokines from adipose tissue lead to plaque formation. Plague formation may also be triggered by hyperglycemia. The HDL-PON complex prevents the aggregation of platelets. PON2 represses the formation of reactive oxygen species (ROS) and has an antiatherogenic effect. ATP-binding cassette transporter A1 (ABCA1) mediates the cellular efflux of phospholipids and cholesterol to lipid-poor apolipoprotein A1 (apoA1)-HDL and plays a significant role in the metabolism of HDL. Blue dots in adipose tissue represent cytokines, lighter circles in the foam cell represent the accumulation of triglycerides, the yellow area in the artery represents hyperglycemia and atherosclerosis, LRP1 = low-density lipoprotein receptor-related protein 1, PAI1 = plasminogen activator inhibitor 1, ABCA1 = ATP-binding cassette transporter A1, SR = scavenger receptor. Created using Biorender.com (accessed on 20 August 2024).
Figure 2. A schematic representation explaining the role paraoxonases (PON1, PON2, and PON3) play in cardiovascular disease (CVD). PON1 and PON3 are attached to high-density lipoprotein (HDL) and oxidize lipid peroxides to prevent their accumulation on low-density lipoprotein (LDL). Oxidized LDL (OX-LDL) triggers macrophages to an anti-inflammatory state and forms foam cells. Foam cells and the release of pro-inflammatory cytokines from adipose tissue lead to plaque formation. Plague formation may also be triggered by hyperglycemia. The HDL-PON complex prevents the aggregation of platelets. PON2 represses the formation of reactive oxygen species (ROS) and has an antiatherogenic effect. ATP-binding cassette transporter A1 (ABCA1) mediates the cellular efflux of phospholipids and cholesterol to lipid-poor apolipoprotein A1 (apoA1)-HDL and plays a significant role in the metabolism of HDL. Blue dots in adipose tissue represent cytokines, lighter circles in the foam cell represent the accumulation of triglycerides, the yellow area in the artery represents hyperglycemia and atherosclerosis, LRP1 = low-density lipoprotein receptor-related protein 1, PAI1 = plasminogen activator inhibitor 1, ABCA1 = ATP-binding cassette transporter A1, SR = scavenger receptor. Created using Biorender.com (accessed on 20 August 2024).
Ijms 25 10634 g002
Figure 3. The link between gut microbiota and atherosclerosis. Gut microbiota produces trimethylamine (TMA) from choline, derived from dietary phosphatidylcholine. Choline is microbially converted to TMA and then oxidized to the pro-atherogenic metabolite trimethylamine N-oxide (TMAO). TMAO may contribute to atherosclerosis by interference with cholesterol transportation, foam cell formation, and platelet aggregation. Platelet aggregation leads to atherosclerosis. Disruption of intestinal permeability by damaged intestinal epithelial cells (IECs) results in the leakage of bacterial toxins such as lipopolysaccharides (LPS) formed by microbiota, and cell wall peptidoglycan into the bloodstream. These toxins react with Toll-like receptors (TLRs), leading to systemic inflammation and the aggravation of atherosclerosis. A diet with less dietary fiber decreases the microbial production of short-chain fatty acids (SCFAs) such as butyrate. Butyrate is the main energy source for colonocytes and has an immunomodulatory effect on gut mucosa. Dotted lines denote the effect of diets on IECs and inflammation, lighter circles in the foam cell represent the accumulation of triglycerides, BAs = bile acids, ROS = reactive oxygen species, C/NF-κB = canonical nuclear factor kappa-B, FXRs = Farnesoid receptors, NLRP3 = Nod-like receptor protein 3, FMO3 = Flavin-containing monooxygenase 3. Created using Biorender.com (accessed on 20 August 2024).
Figure 3. The link between gut microbiota and atherosclerosis. Gut microbiota produces trimethylamine (TMA) from choline, derived from dietary phosphatidylcholine. Choline is microbially converted to TMA and then oxidized to the pro-atherogenic metabolite trimethylamine N-oxide (TMAO). TMAO may contribute to atherosclerosis by interference with cholesterol transportation, foam cell formation, and platelet aggregation. Platelet aggregation leads to atherosclerosis. Disruption of intestinal permeability by damaged intestinal epithelial cells (IECs) results in the leakage of bacterial toxins such as lipopolysaccharides (LPS) formed by microbiota, and cell wall peptidoglycan into the bloodstream. These toxins react with Toll-like receptors (TLRs), leading to systemic inflammation and the aggravation of atherosclerosis. A diet with less dietary fiber decreases the microbial production of short-chain fatty acids (SCFAs) such as butyrate. Butyrate is the main energy source for colonocytes and has an immunomodulatory effect on gut mucosa. Dotted lines denote the effect of diets on IECs and inflammation, lighter circles in the foam cell represent the accumulation of triglycerides, BAs = bile acids, ROS = reactive oxygen species, C/NF-κB = canonical nuclear factor kappa-B, FXRs = Farnesoid receptors, NLRP3 = Nod-like receptor protein 3, FMO3 = Flavin-containing monooxygenase 3. Created using Biorender.com (accessed on 20 August 2024).
Ijms 25 10634 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

Dicks, L.M.T. Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation. Int. J. Mol. Sci. 2024, 25, 10634. https://doi.org/10.3390/ijms251910634

AMA Style

Dicks LMT. Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation. International Journal of Molecular Sciences. 2024; 25(19):10634. https://doi.org/10.3390/ijms251910634

Chicago/Turabian Style

Dicks, Leon M. T. 2024. "Cardiovascular Disease May Be Triggered by Gut Microbiota, Microbial Metabolites, Gut Wall Reactions, and Inflammation" International Journal of Molecular Sciences 25, no. 19: 10634. https://doi.org/10.3390/ijms251910634

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