Next Article in Journal
Adeno-Associated Viral Gene Delivery of Wild-Type Human Tau Induces Progressive Hyperphosphorylation and Neuronal Cell Death in the Hippocampi of Middle-Aged Rats
Previous Article in Journal
Systematic Comparison of Temperature Effects on Antibody Performance via Automated Image Analysis: A Key for Primary Ciliary Dyskinesia Diagnostic
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbial Metabolites and Cardiovascular Dysfunction: A New Era of Diagnostics and Therapy

Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
Cells 2025, 14(16), 1237; https://doi.org/10.3390/cells14161237
Submission received: 1 July 2025 / Revised: 30 July 2025 / Accepted: 8 August 2025 / Published: 11 August 2025

Abstract

Cardiovascular diseases (CVDs) pose a significant threat to human life and mortality worldwide, encompassing a variety of conditions that affect the heart and blood vessels. These diseases are influenced by both genetic and environmental factors, which play a critical role in their development. Recent research has highlighted the importance of gut microbes—the diverse community of bacteria in the gastrointestinal tract—that function as a “super organ” within the human body. These microbes have a remarkable impact on metabolic pathways and are increasingly recognized for their role in serious conditions like CVDs. They contribute to metabolic regulation, provide essential nutrients and vitamins, and help protect against diseases. Various internal and external factors influence the dynamic relationship between the human host and gut microbiota, thereby regulating overall metabolism. This review explores the complex connection between gut microbiota and microbial metabolites—such as short-chain fatty acids (SCFAs), bile acids (BAs), and trimethylamine N-oxide (TMAO)—and their potential influence on the development and progression of CVDs. We also examine the interaction between dietary interventions and gut microbes in the context of conditions including atherosclerosis, obesity, type 2 diabetes, heart failure, hypertension, atrial fibrillation, and myocardial infarction. Gaining a deeper understanding of the gut microbiota’s role in maintaining physiological balance creates exciting possibilities for identifying novel diagnostic biomarkers and therapeutic targets for treating CVDs. This knowledge offers hope for early disease prediction, improved clinical management, and innovative treatments.

1. Introduction

Globally, CVDs are major contributors to morbidity, disability, and mortality, with rising incidences in the recent years. Therefore, they pose a major concern for all developing countries regarding health and socioeconomic challenges. Gut microbes and their metabolites might have therapeutic potential for CVDs and that needs to be connected. The common risk factors for the development of cardiovascular diseases include obesity, hypertension, and type 2 diabetes (T2D) [1]. All three factors are lifestyle and environmental, and might be prevented by monitoring diets, maintaining gut microbes, and engaging in physical activities. Humans have a plethora of microbiotas, which are present throughout the body, including the skin, mouth, stomach, and intestines, in a mutualistic relationship [2].
There is a wide diversity of bacteria in our foregut and midgut, which is regulated by our dietary habits. An equilibrium of different types of bacteria generally benefits human health, while disequilibrium may lead to more severe health problems [3,4,5,6,7,8,9,10,11]. The commonly found species of bacteria in the human gut include Firmicutes, Actinobacteria, Bacteroidetes, Verrucomicrobia, and Proteobacteria [12,13,14,15,16].
The digestion of food is regulated by the microbial community. It helps in converting complex compounds into simpler ones through various metabolites and helpful enzymes [17]. They benefit humans by aiding in synthesizing vital components such as vitamins B and K, fermenting different dietary products like short-chain fatty acids (SCFAs) and secondary bile acids (BAs), and aiding in the metabolism of bile, xenobiotics, and sterol compounds [18,19,20]. The gut microbiome is central in modulating gut function and synthesizing, controlling, and suppressing essential metabolic enzymes closely associated with various health and disease mechanisms [21,22,23]. Hence, it would be right to consider gut microbiota as body’s “super organ” because it is closely connected to our overall health. With aging, the balance of intestinal microbes and human metabolism are affected. Multiple studies utilizing metagenomics, metabolomics, and metalloenzymes have contributed to our understanding of the gene sequences of gut microbial communities [24,25,26,27]. It shows that human health depends on gut microbes, and that microbes involved in the pathophysiology and metabolism pathways where here it plays a role in human health development through nutrients process. Numerous studies have exhibited the vital role of gut bacteria in regulating different body parts, leading to terms such as the gut–liver axis, gut–brain axis, and gut–kidney axis being coined by other research groups [28,29,30,31,32]. In addition to the positive role played in the maintenance and regulation of various metabolic processes, the gut microbes are intricately involved in the development of many diseases, such as colorectal cancer, cerebral ischemia–reperfusion injury, diabetes mellitus type 1 and 2, metabolic syndrome, and CVDs [33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58]. They have the potential to impact the function of various metabolic pathways in the body’s organs; thus, their influence is extended beyond the development of the disease (Figure 1). Gut dysbiosis plays a significant role in development of CVDs such as heart failure and myocardial infarction [59,60,61,62,63,64,65,66,67,68]. For example, Desulfovibrio desulfuricans is involved in TMA/trimethylamine production, while Firmicutes and Proteobacteria are linked to choline consumption [63,69]. Metabolites like lipopolysaccharide (LPS), TMAO, SCFAs, secondary BAs, and uremic toxins such as p-cresol sulfate and indoxyl sulfate may also play a role in cardiovascular function [63,69,70,71,72,73,74,75,76,77]. This review emphasizes our present comprehension, presenting noteworthy proof of the intimate connection between human intestinal microorganisms and cardiovascular conditions. It also aims to assess potential therapeutic approaches that can influence gut flora’s diversity, function, and metabolites.

2. Key Gut Microbial Metabolites and CVDs

The gut microbiota’s metabolites have been linked to various human metabolic processes. Some of these metabolites have been found to influence the development of different types of cardiovascular diseases, as outlined in Table 1.

2.1. Short-Chain Fatty Acids

The fermentation of dietary fibers, primarily polysaccharides, in the human body by gut microbes is essential in digesting complex carbohydrates since the human body’s physiological conditions do not facilitate this process [78]. The composition of gut microbiota significantly influences fiber fermentation, resulting in glycolysis that breaks down glucose into pyruvate, which produces short-chain fatty acids (SCFAs) [79,80]. The most common SCFAs in the human body are acetate, propionate, butyrate, and isovalerate [81,82]. Acetate is vital in gluconeogenesis and reduces appetite through a central homeostatic mechanism. Additionally, acetate is essential in synthesizing low-density lipoprotein (LDL) cholesterol. Propionate has been reported to affect intestinal gluconeogenesis, while butyrate is an energy source for intestinal epithelial cells, contributing to colonic homeostasis care [83,84]. Furthermore, SCFAs are potentially involved in immunomodulatory action, regulating anti-inflammatory responses, producing mucus in the gastrointestinal tract, preventing intestinal inflammation, and maintaining intestinal barrier integrity [79,84].
SCFAs have the potential to modulate microbiota that in turn regulates the cardiovascular diseases (CVDs). Gut microbiota, which produce sodium butyrate and propionate, relate to the function of the prorenin receptor-mediated intrarenal renin–angiotensin system. According to a study, a high-fiber diet and acetate supplements help balance blood pressure and cardiac fibrosis by improving gut dysbiosis and helps in the maintenance of Bacteroides acidifaciens. Bacteria that produce propionate and butyrate (such as Roseburia intestinalis) protect from the risk of hypertensive cardiovascular damage and aortic atherosclerotic lesions [85,86,87,88]. SCFAs are known to interact with immune and endocrine cell receptors in the nervous system, kidneys, and blood vessels; for example, SCFAs regulate blood pressure with the help of Olfr78 (olfactory receptor 78) and GPR41 (G-protein-coupled receptor 41) in small resistance vessels [88,89], and both receptors are essential for maintaining blood pressure regulation under healthy conditions. GPR41 acts as a hypotensive protein to dilate resistance vessels in an endothelium-dependent manner, while Olfr78 functions as a hypersensitive protein [89,90]. The direct activation of GPR1 has been reported to impact inflammation and enteroendocrine regulation.
It has been reported that microbiota-based SCFAs are immunomodulatory in attenuating oxidative stress and maintaining the functional system [91]. Butyrate produced by gut microbiota has been shown to slow atherosclerosis progression due to its anti-inflammatory function. Butyrate also regulates luminal gut oxygen levels by activating peroxisome proliferator-activated receptor gamma in colonocytes. There is a role of acetate and butyrate in improving rat aortic endothelial dysfunction by increasing the bioavailability of nitric oxide, which occurs due to the activation of GPR41/43 for butyrate only [90]. Other GPCRs involved are GPR43 and GPR109A, or histone deacetylases that act via Tregs (immune-suppressive regulatory T cells), interleukin-18 secretion in the intestinal epithelium, and interleukin 10-producing T cells. GPR109A helps in the prevention of colon cancer initiation. SCFAs and microbiota have beneficial effects in preventing cardiac hypertrophy, fibrosis, vascular dysfunction, obesity, and hypertension [86]. Based on a study involving TLR5-knockout mice, it was reported that short-chain fatty acids (SCFAs) have been linked to metabolic syndrome [92]. The dysbiosis of gut microbiota leads to excessive and prolonged SCFA production, which triggers metabolic syndrome [93,94]. TLR5 is a receptor for flagellin, which plays a role in maintaining microbiota balance. This research indicates that the presence of Phascolarcto bacterium, Veillonellaceae, and Proteus mirabilis is strongly associated with obesity, and their alteration leads to the increased production of acetate and propionate [95]. It might be concluded that the different gut microbiota have an essential role in the production of SCFAs that regulate metabolic syndrome, in turn, affecting CVDs.

2.2. Bile Acids

In humans, BAs are hydroxylated and saturated steroids. Bile acids have multiple functions, such as microbial activity, acting as signaling molecules, and stimulating metabolism. Primary bile acids (found in the liver), such as cholic acid (CA) and chenodeoxycholic acid (CDCA), are synthesized from cholesterol in a multistep process in the liver. Several catalytic enzymes, including cholesterol 7a-hydroxylase (CYP7A1), sterol-27-hydroxylase (CYP27A1), and oxysterol 7a-hydroxylase (CYP7B1), are involved in this process and play a crucial role in the emulsification, digestion, and absorption of fat-soluble molecules, dietary fats, and vitamins [96]. The expression of catalytic enzymes bile salt hydrolases (BSHs) is controlled by the human intestinal commensal microbes, e.g., the Gram-positive Bifidobacterium, Clostridium, Enterococcus, and Lactobacillus and the Gram-negative Bacteroides. These gut microbiota in the intestine convert the primary BAs into secondary BAs with the help of bacterial salt hydrolase activity (which removes -OH) [97,98]. The 7-dehydoxylase enzyme, sourced from Clostridium and Eubacterium, is vital for converting primary BAs into secondary BAs. In the small intestine, conjugated BAs are deconjugated via microbial hydrolases, reducing resorption and improving cholesterol exclusion. Microbially metabolized bile acids in the blood regulate host metabolism, which can lead to cardiovascular diseases. BA modification by intestinal microbes occurs before returning to the liver (re-conjugation) and to the circulation. The FXR (Farnesoid X-activated receptor) bile acid receptor is a critical sensor [99]. FXR activation inhibits atherosclerosis injuries in atherosclerosis-prone mice. Deletion of FXR in ApoE-/- mice initiates plaque buildup and increases the severity of lipid metabolism [99,100,101]. However, double-mutation (FXR/ApoE or FXR/LDL) mice showed a reduction in plaque buildup and plasma low-density lipoprotein [102]. The TGR5 (Takeda G-protein-coupled receptor), a BA receptor, activated by secondary BAs, causes decreased intraplaque inflammation, reduced plaque macrophage content, and decreased lipid loading to diminish vascular lesion buildup. Many studies have reported on the mechanism of bile acids in cardiovascular disease pathogenesis [103].
The risk of atherosclerosis or plaque formation increases if the cholesterol circulation level decreases due to decreased primary and secondary bile acid levels. In chronic heart failure patients, the level of primary BAs was reduced while the level of secondary BAs increased. Microbiome-based bile acids can potentially cause atherosclerosis and myocardial infarction (MI) through different types of bile acid receptors by reducing bile acid synthesis and increasing plasma LDL cholesterol [104,105].

2.3. Trimethylamine (TMA)

The human gut microbes initiate TMAO co-metabolite production via TMA hepatic oxidation with the help of flavin monooxygenase enzymes (FMOs, mainly FMO3) [106]. Initially, TMAO was generated within the bacterial metabolism of dietary choline [107], L-carnitine [108], and phosphatidylcholine [106]. It is an organic compound metabolized by intestinal microbes into TMA in the blood. CVD risk is higher when the hematic concentrations of TMAO are increased [108]. In vivo, its high concentration relates to the impairment of calcium signaling pathways, finally stimulating prothrombotic phenotype and platelet hyper-reactivity [109]. It was reported that, in mice, a microbial composition alteration due to chronic dietary L-carnitine increases TMA and TMAO production and ultimately initiates atherosclerosis plaque enhancement. However, when the supplement diet reduces and alters intestinal microbiota composition, it inhibits TMAO production and decreases atherosclerosis lesions [108,110,111]. In the human gut, seven types of microbiotas, such as Anaerococcus hydrogenates, Clostridium hathewayi, Clostridium sporogenes, Clostridium asparagiforme, Escherichia fergusonii, Providencia rettgeri, and Proteus penneri, are observed to regulate the TMA lyase Cut C/D [107,112,113]. It consists of functional microbial genes that are accountable for choline-related TMA transformation. Primarily, gut bacteria regulate TMAO production through different mechanisms like foam cell formation (HSP70 or HSP60, SR-A1, ox-LDL), inflammation (IL-6, COX-2 (cyclooxygenase 2) [108,111,114], intracellular adhesion molecule 1, MAPK, NF-iB), lipid metabolism (RCT (reverse cholesterol transport)), and platelet hyper-reactivity and thrombosis (ADP (adenosine diphosphate), collagen, and thrombin) [115]. Based on different in vivo and in vitro experiments, it has been reported that TMAO has a significant potential to affect the inflammation of aortic endothelial cells, oxidative stress, and inflammatory response, which increases the CVD risk. Choline or TMAO in animal studies shows the connection between gut microbiota and TMAO, which causes CVDs. Studies also indicate a rational relation between plasma TMAO and CVD possibility. For the diagnosis of and the prediction of the possibility of different CVDs like MI (myocardial infarction), stroke, HF (heart failure), and peripheral artery disease, the high value of circulating TMAO should be used as a biomarker. A reduced use of carnitine, choline, and lecithin-rich diets prevents the onset of CVDs. Some natural compounds like DMB (3,3-dimethyl-1-butanol), present widely in red wines, vinegar, and some grape seed oils, prevent the TMA lyase activity of microbial choline [116,117]. Its effects are also observed in the atherosclerotic lesions of Apoe-/- mice [106]. Other TMA lyase inhibitors (like choline) that reduce the plasma TMAO levels are bromomethylcholine, chloromethylcholine, FMC (fluromethylcholine), and IMC (iodomethylcholine). This means, significance of healthy gut microbiota in regulating TMA/TMAO is important for the regulation of good health.

2.4. Other Metabolites

Microbial metabolites derived from aromatic amino acids (AAAs) impact cardiovascular health. These aromatic amino acids are added to diets primarily through dietary proteins found in chicken, fish, pork, beef, and goat. It has been reported that the metabolites, such as phenylacetylglutamine (PAG) and others, have been closely linked to CVDs through various biochemical pathways, thus highlighting the critical role played by gut microbiota in human health as well as disease [118,119,120,121]. Notably, research has indicated that microorganisms in the human gut, specifically Clostridium sporogenes, are responsible for producing metabolites from AAAs [118].
The studies have identified microbial metabolites such as PAG and their potential association with CVDs via the GPCR signaling pathways [119]. Similarly, reduced levels of microbe-derived tryptophan metabolites in the blood plasma of individuals with atherosclerosis have been documented, indicating a possible link to cardiovascular health outcomes [122]. In the rodent system, it has been reported that there is an involvement of tyrosine and phenylalanine microbial metabolites in myocardial infarction. The other intestinal microbe-related metabolites such as indoxyl sulfate (IS) and p-cresol sulfate (PCS) have shown therapeutic potential in diagnosing the onset of CVDs. However, there are conflicting findings regarding their direct connection to cardiovascular health [123,124,125,126].
In the realm of human health, the impact of plant-derived polyphenols on overall well-being has gained significant attention. These organic molecules, found in foods like tea, coffee, apples, onions, cocoa, and citrus fruits, are recognized for their potential in preventing T2D and CVDs. The absorption, circulation, and transportation of polyphenols within the body are influenced by gut microbiota, underscoring the intricate interplay between diet, gut microbiota, and human health. Notable subclasses of dietary polyphenols, including anthocyanins, flavanols, flavanones, and hydroxycinnamates, have been the focus of numerous research due to their potential health benefits and impact on disease prevention [127,128].

3. Gut Microbial Dysbiosis and CVDs

The human gut, a bustling hub of non-pathogenic microorganisms, is a significant regulator of a wide range of CVDs. These encompass hypertension, T2D, obesity, atherosclerosis, and heart failure (Figure 2). A multitude of studies have explored the complex relationship between gut microbial dysbiosis and these specific cardiovascular conditions, unveiling potential mechanisms and implications for treatment and prevention strategies.

3.1. Hypertension

Dysregulated blood pressure is a global disease and a significant cause of cardiovascular disease. The changes induced due to hypertension initiate changes in the intestinal environment that induce the dysbiosis of microbial communities residing in the gut. The dysbiosis of gut bacteria and related metabolites, due to the increased Firmicutes/Bacteroidetes ratio, the abundance of Klebsiella spp., Parabacteroides merdae, and Streptococcus spp., the diminished population of acetate/butyrate metabolite bacteria, SCFAs, and TMAO are well connected with the seriousness of hypertension [129,130]. A disruption of the intestinal barrier along with the elevation of harmful gut microbiota result in chronic inflammation in hypertensive patients. These harmful bacterial genera increase the pro-inflammatory signals and reduce the immunity of the intestine in hypertensive patients.
Mell et al. (2015) [129]. present a significant difference in microbiota in salt-sensitive and salt-resistant strains in Dahl rats [129]. It was observed that the angiotensin-II-infused germ-free mice, angiotensin II-induced vascular dysfunction, and hypertension are regulated by mice gut bacteria [131]. An increased level of Odoribacter (butyrate-producing genus) relates to heavy-weight hypotensive women [132]. Using the metagenomics data based upon the different fecal samples of healthy, prehypertensive, and hypertensive individuals suggested the role of gut microbiota in hypertension pathogenesis. The elevation in the Klebsiella and Prevotella bacterial population in prehypertensive and hypertensive individuals was reported [133]. Numerous studies in mice, rats, and germ–free rats have reported that the unbalanced composition of gut bacteria and inflammation due to a dysfunctional sympathetic nervous system–gut interaction [134,135,136]. Hypertension pathogenesis is changed with the alteration in the relevant gut flora, which shows that intestinal microbiota may have a substantial role in hypertension treatment by using potential probiotics, FMT (fecal transplant), and antibiotics.

3.2. Heart Failure (HF)

HF is a disease in which heart blood pumping efficiency is reduced and does not supply sufficient blood and oxygen as per the body’s needs. Many factors contribute to HF, and gut microbiota is one of them. It involves regulating the immune and inflammatory responses that favor the HF pathophysiological mechanisms [137]. Gut dysbiosis involves reduced cardiac efficiency and raised systemic congestion that leads to ischemia and edema of the intestine mucosa, improves bacterial translocation, and increases circulating lipopolysaccharide levels (endotoxins), which play a role in the underlying inflammation in HF patients [138,139,140]. Moreover, Clostridium difficile infection is reported in the HF patients. Furthermore, the sequencing data of microbial diversity of fecal samples of HF patients showed a diminution of many butyrate-producing bacteria and Dorea longicatena and Eubacterium rectale. However, pathogenic Campylobacter, Shigella, and Salmonella bacteria are abundant [141]. In addition, based on metagenomics and metabolomics data, it is shown that some HF patients reported a significant decrease in Blautia, Collinsella, and Faecalibacterium prausnitzii [137,142]. Paisini et al. (2016) [141] study shows that chronic heart failure patients have higher intestinal permeability and a higher number of gut bacteria (in feces) than control individuals [141]. Different animal studies have demonstrated the richness of different types of fecal flora in HF animals. This indicates that HF may cause the dysbiosis of gut microflora that supports the progression of the disease [143]. TMAO is a microbiota-associated metabolite, and few studies have demonstrated that increased levels of circulating TMAO are predictors of acute and chronic heart patients compared to healthy controls [144]. This metabolite is also associated with other CVDs like left ventricular dilatation, myocardial fibrosis, ventricular remodeling, and cardiac hypertrophy [145].

3.3. Acute Myocardial Infraction (AMI)

Arterial blockage is a major health issue. Based on this, coronary artery disease is divided into stable coronary artery disease (CAD) and acute coronary syndromes (ACS) [146]. The leading cause of CAD is a two-sided, blood supply–demand event mismatch association with myocardial ischemia. In contrast, ACS clinically shows AMI, unstable angina pectoris (UAP), or sudden cardiac death (SCD) [147]. After outstanding achievements in medical fields, the early prediction of AMI is still a big challenge. Many diagnostic biomarkers are available to diagnose AMI, but reliable and effective techniques are still unavailable. Therefore, more research is needed for an accurate biomarker to diagnose clinical AMI. The primary biomarkers for AMI are cTnI (cardiac troponins I) and cTnT (cardiac troponins T). The main limitation of cTnI is that it is usually high in both sCDA and MI patients [148,149]. Different studies have demonstrated that, in AMI patients, exosomal miR-1, mir-4507, mir-1915-3p, and miR-3656, CK (creatine kinase) and CK-MB (creatinine kinase myocardial band) are drastically higher when compared with sCAD and healthy persons. However, their forecast precision is not satisfactory [150,151,152]. Therefore, more biomarkers with high sensitivity and specificity for AMI are needed. Numerous animal studies demonstrated intestinal microflora’s role and related metabolites in AMI occurrence. Different bacterial families are significantly higher in AMI rats than the SHAM controls, such as Dethiosulfo vibrionaceae, Eubacteriaceae, Lachnospiraceae, and Syntrophomonadaceae and the genera Tissierella and Soehngenia. In comparison with healthy individuals, patients with AMI harbor plenty of Acidaminococcus, Butyricimonas, Desulfovibrio, and Megasphaera [153,154,155,156,157,158,159,160,161,162,163,164]. The study of Liu et al. (2019) [155]. showed that the typical changes in bacterial co-abundance group (CAG) in coronary artery disease (CAD) were dominated by Roseburia, Klebsiella, Clostridium IV, and Ruminococcaceae [155]. In contrast, a recent study reported that Alistipes, Streptococcus, Ruminococcus, and Lactobacillus are efficient in differentiating AMI from sCAD, and their prognostic competence has been confirmed in an independent cohort of CAD patients [156]. Meanwhile, a high level of TMAO in blood plasma and proatherogenic and prothrombotic bacterial metabolites relate to different adverse symptoms in ACS patients. Furthermore, the tryptophan metabolites of the kynurenin pathway have been linked to a higher likelihood of AMI in patients with suspected sCAD [157,158]. Based on these studies, intestinal microflora strongly correlates with CAD patients, altering the blood metabolite content and composition. Hence, intestinal microflora and serum metabolites have been identified as potential diagnostic markers of AMI.

3.4. Atherosclerosis

It is a chronic inflammatory disease leading to the initial endothelial injury or dysfunction of vascular cells and the development of plaque in the arteries due to the buildup of LDL bits. Different types of cells, like endothelial cells, macrophages, foam cells, and leukocytes, accumulated lipids like cholesterol and triglycerides which involved in a plaque formation. Atherosclerosis risk is generally high in individuals with hypertension, obesity, diabetes mellitus, and hypercholesterolemia [159,160]. Ott et al. (2006) [161] reported Staphylococcus species, Proteus vulgaris, Klebsiella pneumoniae, and Streptococcus species in atherosclerosis plaques and the gut of the same individual. These results recognized the possibility of the presence of gut microbiota in the process impacting plaque formation and stability and causing atherosclerosis [161]. Atherosclerosis is a common chronic inflammatory CVD that affects the coronary and peripheral arteries. Gut microbiota and its metabolites play a crucial role in the development of CVDs. Increased Firmicutes/Bacteroidota (F/B) ratio in gut is an indicator of CVDs.
Furthermore, the presence of oral microbiota is also reported in the atherosclerotic plaque in humans [162]. Karlsson et al. (2012) [163] used metagenomic sequencing to study the composition of gut bacteria in stool samples and reported the dysbiosis of gut microflora in patients with stable plaques versus unstable plaques. They found a diminished level of butyrate-producing bacterial genus Roseburiam in the unstable plaque patients, and symptomatic atherosclerotic patients harbor a greater abundance of Collinsella compared with healthy controls [163]. Gut metagenomes, coding genes for pro-inflammatory peptidoglycan synthesis are abundant, and the phytoene dehydrogenase level is low in patient. The gut microbiome of the patients shows a low production of anti-inflammatory serum levels of β-carotene [43]. Other studies on humans show that the gut microbiome is possibly involved in the pathogenesis of atherosclerosis through its role in lipid metabolism. Middle-aged men with improper total cholesterol and LDL-C values harbor more Prevotella and lower levels of Clostridium and Faecalibacterium [164]. However, other metagenome-wide association studies reported a lesser number of Bacteroides and Prevotella and the enrichment of Enterobacteriaceae (including Escherichia coli, Enterobacter aerogenes, and Klebsiella spp.) and Streptococcus spp. in atherosclerotic cardiovascular disease patients [165]. There is a relationship between various gut microbiome metabolite levels, such as TMAO, BA, and SCFA, and the pathogenesis of CVD. Different factors and CVD symptoms are involved in microbial dysbiosis, such as the mutation of TMAO (toxic metabolites) and pathogenic bacteria [165,166,167,168]. To understand the specific gut microbiome biomarkers of atherosclerosis development and their mechanism, more investigation is needed.

3.5. Atrial Fibrillation (AF)

AF is not just a local issue but a global one, affecting people worldwide. It is one of the most ubiquitous and extensive cardiac arrhythmias in clinics. The fast and uneven atrial beating represents an abnormal heart rhythm [169]. Ischemic stroke (cerebral infarction) and cardiac insufficiency risk factors are increased in the AF. Based on clinical and pathological systems of AF, AF decreases the end-diastolic volume and causes fast atrioventricular desynchrony, a fast ventricular rate, and a loss of atrial systolic pump function, leading to a decline in cardiac systolic function and heart failure. Furthermore, in ischemic stroke patients, AF enhances the mortality, recurrence rate, and infirmity rate [170,171]. It is a global disease, and due to its infirmity and morbidity rate, AF generates a serious issue for society and affects the excellence of human life. Therefore, the prevention of AF is needed. The human gut microflora plays a vital role in the onset of AF. Zuo et al.’s (2019) [172] study showed that the composition of AF patients’ gut flora significantly differs from that of normal individuals. In AF patients, there is an abundance of Enterococcus, Ruminococcus, and Streptococcus. However, decreased levels of Alistipes, Oscillibacter, Bilophila, and Faecalibacteriumare were reported [172]. Moreover, another study based on metagenomics and metabolomics reported the short and long psAF (persistent AF) and highlighted the relationship of gut bacteria with psAF maintenance. They found significantly different types of gut bacteria in both short- and long-term psAF patients, with an elevated microbial dysbiosis, diverse composition, and inconsistent structure. They also observed modifications in related bacterial metabolites and disorders in gut microbial function [173]. LPS (lipopolysaccharide) produced from bacteria reacts as an endotoxin in human blood by stimulating pro-inflammatory cytokines that reduce the L-type Ca2+ channel expression and shorten ERP (effective refractory period). ERP shortening is also observed due to the other bacterial metabolites like choline that activate the acetylcholine-dependent potassium channels at high concentrations. LPS is also connected with different CVDs like atherosclerosis, heart failure, and cardiac arrhythmia (mainly AF) [174,175,176]. Altogether, it represents an association of AF with differential gut microbial flora and their related metabolite pattern. The alteration in gut microbiota may be used as a biomarker for AF disease.

3.6. Obesity and Type 2 Diabetes Mellitus (T2DM)

Obesity is a global issue that occurs due to the lower consumption of energy in comparison with intake. The risk of T2DM-type multifactorial disease is increased in the case of obesity. Metabolites have enough competence to be a factor in the host–microbiota relations. Researchers have used a combined metagenomic and metabolomic methodology to explore the associations between the alterations in the gut microbial flora and the onset of metabolic malformations in obesity and diabetes [177]. The most consistent phylum associated with obesity is Proteobacteria, and the Firmicutes/Bacteroides ratio usually increases in obese individuals [178]. Due to restricted diets like low-calorie, low-fat, or low- carbohydrate, in individuals aiming for weight loss, the Firmicutes and Bacteroidetes ratio is reduced [178,179]. Duodenal microbiomes, such as Bifidobacterium and Lactobacillus, were also identified in obese persons, and their observed diversity was different when compared with that of the overweight person, which indicates their significant therapeutic potential. Metagenomics data support the connection between the alteration in gut bacterial composition in obesity and T2DM. In T2DM, butyrate-producing bacteria diminished and enhanced the Lactobacillus spp. [180,181,182,183,184]. Another study also suggested that glucose tolerance impairment is associated with T2DM; vancomycin decreases the butyrate-producing bacteria in metabolic syndrome patients and diminishes insulin sensitivity [181,182,183,184,185]. Hosomi et al. (2022) [186] reported that the Blautia genus, mainly B. wexlerae, is inversely associated with obesity and T2DM. The impacts of these bacteria are interrelated with unique regulatory pathways. It helps in the production of acetylcholine, S-adenosylmethionine, and L-ornithine (amino acid metabolism), the accumulation of amylopectin (carbohydrate metabolism), and production of succinate, lactate, and acetate [186]. Other researchers found many bacterial taxa, including Acidaminococcales, Bacteroides plebeius, and Phascolarctobacterium sp. CAG207 bacterial species significantly differ in obese patients with T2DM and healthy controls [187]. T2DM individuals have lower levels of hydrogen sulfide. Sulfate-reducing bacteria, such as Desulfovibrio and Desulfomonas, produce hydrogen sulfide, a product of protein fermentation that affects the basal hepatic glucose levels [188].
Cardiovascular insulin resistance (CVIR) refers to the reduced capability of cardiovascular tissues, specifically vascular smooth muscle cells, endothelial cells, and cardiomyocytes, to respond to insulin. This dysfunction occurs in two ways: “direct and indirect”. Direct dysfunction happens within the heart or vasculature itself; however, indirect dysfunction results from systemic insulin resistance in liver, muscle, and adipose tissues. One molecular paradox is PI3K-Akt that promotes nitric oxide production, vasodilation, anti-inflammatory effects [189,190].

4. Gut Microbiota and Therapeutic Potential

Based on numerous studies, vast evidence indicates and supports gut microbiota and its metabolites as a critical factor in the onset of cardiovascular diseases (Table 2).
Thus, gut bacteria might be used as a biomarker and an ideal target for disease prevention, diagnosis, and treatment. A key strategy in maintaining the composition of these bacteria in the gut is dietary intervention. By adopting heart-healthy diets rich in fiber and green vegetables, individuals can take control of their health and influence their gut microbiota. These diets should also include fruits, legumes, and unsaturated fats. Dietary fibers are particularly helpful in producing SCFAs and maintaining the composition of beneficial gut microbes [191]. Antibiotics also play a role in modulating gut microflora to prevent CVDs. In one study, chronic and resistant hypertensive patients were cured after antibiotic treatment [192].
Additionally, after using vancomycin, the myocardial infarct area was reduced, and post-ischemic recovery improved with a reduction in different intestinal microbial groups. Recently, it has been reported that imidazole propionate (ImP), produced by microorganisms, might be explored as an atherosclerosis target [193]. Dietary fiber food like chickpeas and whole-grain oats enhances the bacterial dysbiosis by altering the relative abundance of Bacteroides and Lactobacillus. Chickpeas help maintain gut bacterial composition and prevent hyperglycemia, whereas whole-grain oats control insulin sensitivity and cholesterol levels [194,195]. Prebiotics are nonmicrobial bodies supplied to stimulate a promising effect on microbial population structure and function. They primarily include dietary fibers, non-digestible molecules, or plant-based supplement food containing oligosaccharides and polysaccharides, which help increase friendly gut microbes. Numerous studies have reported that prebiotics impact host metabolism and have beneficial anti-CVD effects through lipid control, glycemic control, reduced endotoxemia and inflammation, and maintained blood pressure [196,197]. Oligofructose supplementation helps obese patients lose weight and improve glucose tolerance [198,199,200,201,202]. On the other hand, probiotics include beneficial microbes determined to establish the usual gut microbiome equilibrium. One meta-analysis study reported that probiotic treatment may help lower the total cholesterol as well as the low-density lipoprotein (LDL) cholesterol levels, improve blood pressure, and modulate inflammatory cytokines. The most significant probiotic species are Bifidobacterium spp. and Lactobacillus spp., and at the same time, the most common probiotics are yogurt, kefir, sauerkraut, and kimchi in human diets. As with prebiotics, probiotics also help improve CVDs, providing reassurance and confidence in their use. Lactobacillus plantarum helps in the improvement of endothelium-dependent vasodilation [203], Lactobacillus fermentum CECT5716 treatment improves endothelial function and controls vascular oxidative stress [204], and yogurt helps in blood glucose and antioxidant level maintenance [205]. Bifidobacterium longum BB536 can potentially diminish the total cholesterol and liver lipid deposition levels and adipocyte size [206]. Akkermansia muciniphila reduces atherosclerotic lesions [207]. Other probiotics are Christensenella minuta, which protects from obesity, and Lactobacillus reuteri, which improves insulin secretion [208,209]. Symbiotic is a blend of prebiotics and probiotics designed to promote the growth as well as the survival of beneficial microorganisms. Numerous studies on animals like chickens and mice reported the benefits of symbiotics for improving gut microbes’ composition, immune functions, and modifications in body weight and metabolic syndrome [210,211,212]. Another therapeutic approach is FMT (fecal microbiota transplantation), in which a patient’s intestinal microbes are replaced by beneficial microbes from healthy subjects to treat gastrointestinal diseases [213]. There are two types of transfers: one is autologous transfer, in which the same individual’s feces microbiota is used, and another is allogenic transfer, in which other healthy individual feces microbiota is transferred. FMT is more beneficial in humans with Clostridium difficile antibiotic-resistant disease. It is currently tested against cardiometabolic disorders [214]. The limitation of FMT is that it is associated with risks, including the possibility of transferring endotoxins or infectious agents that may cause new complications. Thus, more research is warranted to guarantee the effectiveness and safety of FMT.

5. Conclusions and Future Perspectives

Human diets modify human health, and numerous animal and human studies support communalism bacteria. Recent metagenomics and metabolomics approaches, next-generation high-throughput sequencing, and bioinformatic tools help to uncover the different untapped gene pools of gut microbes, metabolites, and their effects on human health that were not reported earlier. Moreover, it is important to characterize the bacterial metabolites that play a role in the host’s physiological modifications to treat cardiometabolic disorders. Numerous microbial pathways are involved directly or indirectly in the stimulation of CVDs. Different host pathways attract microbes and help alter the composition of microbial metabolites that may cause CVDs. More attention is needed to understand how gut microbes convert dietary and endogenous molecules into metabolites and their connections with different tissues and organs of the host. It is also essential to gather some knowledge about the roles of microbial protein interactions, modifications, and post-translational modifications and their impacts and connections with host proteins related to CVDs. Alterations in intestinal microbe structures and functions through dietary interventions, prebiotics, probiotics, symbiotics, antibiotics, and FMT have the potential to alter host metabolism as needed. In the early diagnosis or as therapeutic targets of CVDs, gut microbiota shows their potential. Future research should focus on finding the exact bacteria linked to each type of cardiovascular disease and exploring the substances these bacteria produce that may affect the disease. This could help in the identification of new metabolomic biomarkers of diseases and further our understanding of the role of gut microbiota in CVDs.

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.

Acknowledgments

I am grateful to Santosh Kumar, Department of Internal Medicine, University of Iowa, USA, for his help in this manuscript. Khusboo Sharma, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India, helped us improve the text and references. Rakesh Roshan, Department of Zoology, Shivaji College, University of Delhi, New Delhi, India, helped us to improve the language of the manuscript, figures, and tables. Manish Sharma, Department of Zoology, University of Allahabad, Prayagraj, Uttar Pradesh, India, helped us with manuscript editing.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Ginsberg, H.N.; MacCallum, P.R. The obesity, metabolic syndrome, and type 2 diabetes mellitus pandemic: Part I. Increased cardiovascular disease risk and the importance of atherogenic dyslipidemia in persons with the metabolic syndrome and type 2 diabetes mellitus. J. CardioMetab. Syndr. 2009, 4, 113–119. [Google Scholar] [CrossRef]
  2. Ursell, L.K.; Metcalf, J.L.; Parfrey, L.W.; Knight, R. Defining the human microbiome. Nutr. Rev. 2012, 70 (Suppl. 1), S38–S44. [Google Scholar] [CrossRef]
  3. Lee, J.K.-F.; Hern Tan, L.T.; Ramadas, A.; Ab Mutalib, N.-S.; Lee, L.-H. Exploring the Role of Gut Bacteria in Health and Disease in Preterm Neonates. Int. J. Environ. Res. Public Health 2020, 17, 6963. [Google Scholar] [CrossRef]
  4. Rosen, C.E.; Palm, N.W. Functional Classification of the Gut Microbiota: The Key to Cracking the Microbiota Composition Code: Functional classifications of the gut microbiota reveal previously hidden contributions of indigenous gut bacteria to human health and disease. Bioessays 2017, 39, 1700032. [Google Scholar] [CrossRef] [PubMed]
  5. Long, S.L.; Gahan, C.G.M.; Joyce, S.A. Interactions between gut bacteria and bile in health and disease. Mol. Asp. Med. 2017, 56, 54–65. [Google Scholar] [CrossRef] [PubMed]
  6. Ross, F.C.; Patangia, D.; Grimaud, G.; Lavelle, A.; Dempsey, E.M.; Ross, R.P.; Stanton, C. The interplay between diet and the gut microbiome: Implications for health and disease. Nat. Rev. Microbiol. 2024, 22, 671–686. [Google Scholar] [CrossRef] [PubMed]
  7. Kant, R.; de Vos, W.M.; Palva, A.; Satokari, R. Immunostimulatory CpG motifs in the genomes of gut bacteria and their role in human health and disease. J. Med. Microbiol. 2014, 63 Pt 2, 293–308. [Google Scholar] [CrossRef]
  8. Boleij, A.; Tjalsma, H. Gut bacteria in health and disease: A survey on the interface between intestinal microbiology and colorectal cancer. Biol. Rev. Camb. Philos. Soc. 2012, 87, 701–730. [Google Scholar] [CrossRef]
  9. Barbara, G.; Stanghellini, V.; Brandi, G.; Cremon, C.; Di Nardo, G.; De Giorgio, R.; Corinaldesi, R. Interactions between commensal bacteria and gut sensorimotor function in health and disease. Am. J. Gastroenterol. 2005, 100, 2560–2568. [Google Scholar] [CrossRef]
  10. Stagg, A.J.; Hart, A.L.; Knight, S.C.; Kamm, M.A. Microbial-gut interactions in health and disease. Interactions between dendritic cells and bacteria in the regulation of intestinal immunity. Best Pract. Res. Clin. Gastroenterol. 2004, 18, 255–270. [Google Scholar] [CrossRef]
  11. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  12. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef]
  13. Li, J.; Si, H.; Du, H.; Guo, H.; Dai, H.; Xu, S.; Wan, J. Comparison of gut microbiota structure and actinobacteria abundances in healthy young adults and elderly subjects: A pilot study. BMC Microbiol. 2021, 21, 13. [Google Scholar] [CrossRef] [PubMed]
  14. Fujio-Vejar, S.; Vasquez, Y.; Morales, P.; Magne, F.; Vera-Wolf, P.; Ugalde, J.A.; Navarrete, P.; Gotteland, M. The Gut Microbiota of Healthy Chilean Subjects Reveals a High Abundance of the Phylum Verrucomicrobia. Front. Microbiol. 2017, 8, 1221. [Google Scholar] [CrossRef] [PubMed]
  15. Stojanov, S.; Berlec, A.; Štrukelj, B. The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio in the Treatment of Obesity and Inflammatory Bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef] [PubMed]
  16. Vaiserman, A.; Romanenko, M.; Piven, L.; Moseiko, V.; Lushchak, O.; Kryzhanovska, N.; Guryanov, V.; Koliada, A. Differences in the gut Firmicutes to Bacteroidetes ratio across age groups in healthy Ukrainian population. BMC Microbiol. 2020, 20, 221. [Google Scholar] [CrossRef]
  17. Chen, Y.; Zhou, J.; Wang, L. Role and Mechanism of Gut Microbiota in Human Disease. Front. Cell. Infect. Microbiol. 2021, 11, 625913. [Google Scholar] [CrossRef]
  18. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef]
  19. Oliphant, K.; Allen-Vercoe, E. Macronutrient metabolism by the human gut microbiome: Major fermentation by-products and their impact on host health. Microbiome 2019, 7, 91. [Google Scholar] [CrossRef]
  20. Koppel, N.; Rekdal, V.M.; Balskus, E.P. Chemical transformation of xenobiotics by the human gut microbiota. Science 2017, 356, eaag2770. [Google Scholar] [CrossRef]
  21. Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef] [PubMed]
  22. Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef]
  23. Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  24. Claesson, M.J.; Wang, Q.; O’Sullivan, O.; Greene-Diniz, R.; Cole, J.R.; Ross, R.; O’Toole, P.W. Comparison of two next-generation sequencing technologies for resolving highly complex microbiota composition using tandem variable 16S rRNA gene regions. Nucleic Acids Res. 2010, 38, e200. [Google Scholar] [CrossRef] [PubMed]
  25. Bartram, A.K.; Lynch, M.D.J.; Stearns, J.C.; Moreno-Hagelsieb, G.; Neufeld, J.D. Generation of multimillion-sequence 16S rRNA gene libraries from complex microbial communities by assembling paired-end illumina reads. Appl. Environ. Microbiol. 2011, 77, 3846–3852. [Google Scholar] [CrossRef]
  26. Nelson, M.C.; Morrison, H.G.; Benjamino, J.; Grim, S.L.; Graf, J.; Heimesaat, M.M. Analysis, optimization and verification of Illumina-generated 16S rRNA gene amplicon surveys. PLoS ONE 2014, 9, e94249. [Google Scholar] [CrossRef]
  27. Razali, H.; O’cOnnor, E.; Drews, A.; Burke, T.; Westerdahl, H. A quantitative and qualitative comparison of illumina MiSeq and 454 amplicon sequencing for genotyping the highly polymorphic major histocompatibility complex (MHC) in a non-model species. BMC Res. Notes 2017, 10, 346. [Google Scholar] [CrossRef]
  28. Forkosh, E.; Ilan, Y. The heart-gut axis: New target for atherosclerosis and congestive heart failure therapy. Open Heart 2019, 6, e000993. [Google Scholar] [CrossRef]
  29. Kamo, T.; Akazawa, H.; Suzuki, J.-I.; Komuro, I. Novel Concept of a Heart-Gut Axis in the Pathophysiology of Heart Failure. Korean Circ. J. 2017, 47, 663–669. [Google Scholar] [CrossRef]
  30. Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef]
  31. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  32. Stavropoulou, E.; Kantartzi, K.; Tsigalou, C.; Konstantinidis, T.; Romanidou, G.; Voidarou, C.; Bezirtzoglou, E. Focus on the Gut-Kidney Axis in Health and Disease. Front. Med. 2021, 7, 620102. [Google Scholar] [CrossRef]
  33. Shi, L.; Xu, Y.; Feng, M. Role of Gut Microbiome in Immune Regulation and Immune Checkpoint Therapy of Colorectal Cancer. Dig. Dis. Sci. 2023, 68, 370–379. [Google Scholar] [CrossRef]
  34. Siddiqui, R.; Boghossian, A.; Alharbi, A.M.; Alfahemi, H.; Khan, N.A. The Pivotal Role of the Gut Microbiome in Colorectal Cancer. Biology 2022, 11, 1642. [Google Scholar] [CrossRef]
  35. Rebersek, M. Gut microbiome and its role in colorectal cancer. BMC Cancer 2021, 21, 1325. [Google Scholar] [CrossRef]
  36. Chen, F.; Dai, X.; Zhou, C.-C.; Li, K.-X.; Zhang, Y.-J.; Lou, X.-Y.; Zhu, Y.-M.; Sun, Y.-L.; Peng, B.-X.; Cui, W. Integrated analysis of the faecal metagenome and serum metabolome reveals the role of gut microbiome-associated metabolites in the detection of colorectal cancer and adenoma. Gut 2022, 71, 1315–1325. [Google Scholar] [CrossRef]
  37. Genua, F.; Raghunathan, V.; Jenab, M.; Gallagher, W.M.; Hughes, D.J. The Role of Gut Barrier Dysfunction and Microbiome Dysbiosis in Colorectal Cancer Development. Front. Oncol. 2021, 11, 626349. [Google Scholar] [CrossRef] [PubMed]
  38. Huang, Q.-Y.; Yao, F.; Zhou, C.-R.; Huang, X.-Y.; Wang, Q.; Long, H.; Wu, Q.-M. Role of gut microbiome in regulating the effectiveness of metformin in reducing colorectal cancer in type 2 diabetes. World J. Clin. Cases 2020, 8, 6213–6228. [Google Scholar] [CrossRef] [PubMed]
  39. Leonard, M.M.; Valitutti, F.; Karathia, H.; Pujolassos, M.; Kenyon, V.; Fanelli, B.; Troisi, J.; Subramanian, P.; Camhi, S.; Colucci, A.; et al. Microbiome signatures of progression toward celiac disease onset in at-risk children in a longitudinal prospective cohort study. Proc. Natl. Acad. Sci. USA 2021, 118, e2020322118. [Google Scholar] [CrossRef] [PubMed]
  40. Kumar, J.; Kumar, M.; Pandey, R.; Chauhan, N.S. Physiopathology and Management of Gluten-Induced Celiac Disease. J. Food Sci. 2017, 82, 270–277. [Google Scholar] [CrossRef]
  41. Wang, H.; Ren, S.; Lv, H.; Cao, L. Gut microbiota from mice with cerebral ischemia-reperfusion injury affects the brain in healthy mice. Aging 2021, 13, 10058–10074. [Google Scholar] [CrossRef]
  42. Hu, W.; Kong, X.; Wang, H.; Li, Y.; Luo, Y. Ischemic stroke and intestinal flora: An insight into brain–gut axis. Eur. J. Med. Res. 2022, 27, 73. [Google Scholar] [CrossRef]
  43. Qiu, P.; Ishimoto, T.; Fu, L.; Zhang, J.; Zhang, Z.; Liu, Y. The Gut Microbiota in Inflammatory Bowel Disease. Front. Cell. Infect. Microbiol. 2022, 12, 733992. [Google Scholar] [CrossRef]
  44. Lloyd-Price, J.; Arze, C.; Ananthakrishnan, A.N.; Schirmer, M.; Avila-Pacheco, J.; Poon, T.W.; Andrews, E.; Ajami, N.J.; Bonham, K.S.; Brislawn, C.J.; et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 2019, 569, 655–662. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, B.-N.; Liu, X.-T.; Liang, Z.-H.; Wang, J.-H. Gut microbiota in obesity. World J. Gastroenterol. 2021, 27, 3837–3850. [Google Scholar] [CrossRef] [PubMed]
  46. Duan, M.; Wang, Y.; Zhang, Q.; Zou, R.; Guo, M.; Zheng, H.; Ling, Z. Characteristics of gut microbiota in people with obesity. PLoS ONE 2021, 16, e0255446. [Google Scholar] [CrossRef] [PubMed]
  47. Jin, J.; Cheng, R.; Ren, Y.; Shen, X.; Wang, J.; Xue, Y.; Zhang, H.; Jia, X.; Li, T.; He, F.; et al. Distinctive Gut Microbiota in Patients with Overweight and Obesity with Dyslipidemia and its Responses to Long-term Orlistat and Ezetimibe Intervention: A Randomized Controlled Open-label Trial. Front. Pharmacol. 2021, 12, 732541. [Google Scholar] [CrossRef]
  48. Zhou, Z.; Sun, B.; Yu, D.; Zhu, C. Gut Microbiota: An Important Player in Type 2 Diabetes Mellitus. Front. Cell. Infect. Microbiol. 2022, 12, 834485. [Google Scholar] [CrossRef]
  49. Zhou, H.; Sun, L.; Zhang, S.; Zhao, X.; Gang, X.; Wang, G. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front. Endocrinol. 2020, 11, 125. [Google Scholar] [CrossRef]
  50. Jamshidi, P.; Hasanzadeh, S.; Tahvildari, A.; Farsi, Y.; Arbabi, M.; Mota, J.F.; Sechi, L.A.; Nasiri, M.J. Is there any association between gut microbiota and type 1 diabetes? A systematic review. Gut Pathog. 2019, 11, 49. [Google Scholar] [CrossRef]
  51. Girdhar, K.; Huang, Q.; Chow, I.-T.; Vatanen, T.; Brady, C.; Raisingani, A.; Autissier, P.; Atkinson, M.A.; Kwok, W.W.; Kahn, C.R.; et al. A gut microbial peptide and molecular mimicry in the pathogenesis of type 1 diabetes. Proc. Natl. Acad. Sci. USA 2022, 119, e2120028119. [Google Scholar] [CrossRef]
  52. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef] [PubMed]
  53. Romano, S.; Savva, G.M.; Bedarf, J.R.; Charles, I.G.; Hildebrand, F.; Narbad, A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. npj Park. Dis. 2021, 7, 27. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, N.Y.; Suk, K.T. The Role of the Gut Microbiome in Liver Cirrhosis Treatment. Int. J. Mol. Sci. 2020, 22, 199. [Google Scholar] [CrossRef] [PubMed]
  55. Jones, R.M.; Neish, A.S. Gut Microbiota in Intestinal and Liver Disease. Annu. Rev. Pathol. Mech. Dis. 2021, 16, 251–275. [Google Scholar] [CrossRef]
  56. Li, Y.; Liu, T.; Yan, C.; Xie, R.; Guo, Z.; Wang, S.; Zhang, Y.; Li, Z.; Wang, B.; Cao, H. Diammonium Glycyrrhizinate Protects against Nonalcoholic Fatty Liver Disease in Mice through Modulation of Gut Microbiota and Restoration of Intestinal Barrier. Mol. Pharm. 2018, 15, 3860–3870. [Google Scholar] [CrossRef]
  57. Rahman, M.; Islam, F.; -Or-Rashid, H.; Al Mamun, A.; Rahaman, S.; Islam, M.; Meem, A.F.K.; Sutradhar, P.R.; Mitra, S.; Mimi, A.A.; et al. The Gut Microbiota (Microbiome) in Cardiovascular Disease and Its Therapeutic Regulation. Front. Cell. Infect. Microbiol. 2022, 12, 903570. [Google Scholar] [CrossRef]
  58. Jonsson, A.L.; Backhed, F. Role of gut microbiota in atherosclerosis. Nat. Rev. Cardiol. 2017, 14, 79–87. [Google Scholar] [CrossRef]
  59. Liu, C.; Li, Z.; Song, Z.; Fan, X.; Shao, H.; Schönke, M.; Boon, M.R.; Rensen, P.C.; Wang, Y. Choline and butyrate beneficially modulate the gut microbiome without affecting atherosclerosis in APOE*3-Leiden.CETP mice. Atherosclerosis 2022, 362, 47–55. [Google Scholar] [CrossRef]
  60. O’Donnell, J.A.; Zheng, T.; Meric, G.; Marques, F.Z. The gut microbiome and hypertension. Nat. Rev. Nephrol. 2023, 19, 153–167. [Google Scholar] [CrossRef]
  61. Silveira-Nunes, G.; Durso, D.F.; De Oliviera, L.R.A., Jr.; Cunha, E.H.M.; Maioli, T.U.; Vieira, A.T.; Speziali, E.; Corrêa-Oliveira, R.; Martins-Filho, O.A.; Teixeira-Carvalho, A.; et al. Hypertension Is Associated With Intestinal Microbiota Dysbiosis and Inflammation in a Brazilian Population. Front. Pharmacol. 2020, 11, 258. [Google Scholar] [CrossRef]
  62. Wang, Z.; Zhao, Y. Gut microbiota derived metabolites in cardiovascular health and disease. Protein Cell 2018, 9, 416–431. [Google Scholar] [CrossRef] [PubMed]
  63. Tang, W.W. Dysregulated amino acid metabolism in heart failure: Role of gut microbiome. Curr. Opin. Clin. Nutr. Metab. Care 2022, 26, 195–200. [Google Scholar] [CrossRef] [PubMed]
  64. Li, L.; Zhong, S.-J.; Hu, S.-Y.; Cheng, B.; Qiu, H.; Hu, Z.-X. Changes of gut microbiome composition and metabolites associated with hypertensive heart failure rats. BMC Microbiol. 2021, 21, 141. [Google Scholar] [CrossRef] [PubMed]
  65. Yoshihisa, A. Altered Gut Flora and Gut Microbiome-Derived Metabolites in Heart Failure Patients in the Compensated and Decompensated Phases. Circ. J. 2018, 83, 30–31. [Google Scholar] [CrossRef]
  66. Liu, C.; Sun, Z.; Shali, S.; Mei, Z.; Chang, S.; Mo, H.; Xu, L.; Pu, Y.; Guan, H.; Chen, G.-C.; et al. The gut microbiome and microbial metabolites in acute myocardial infarction. J. Genet. Genom. 2022, 49, 569–578. [Google Scholar] [CrossRef]
  67. Gawalko, M.; Agbaedeng, T.A.; Saljic, A.; Müller, D.N.; Wilck, N.; Schnabel, R.; Penders, J.; Rienstra, M.; van Gelder, I.; Jespersen, T.; et al. Gut microbiota, dysbiosis and atrial fibrillation. Arrhythmogenic mechanisms and potential clinical im-plications. Cardiovasc. Res. 2022, 118, 2415–2427. [Google Scholar] [CrossRef]
  68. Zuo, K.; Yin, X.; Li, K.; Zhang, J.; Wang, P.; Jiao, J.; Liu, Z.; Liu, X.; Liu, J.; Li, J.; et al. Different Types of Atrial Fibrillation Share Patterns of Gut Microbiota Dysbiosis. mSphere 2020, 5, e00071-20. [Google Scholar] [CrossRef]
  69. Mohr, A.E.; Crawford, M.; Jasbi, P.; Fessler, S.; Sweazea, K.L. Lipopolysaccharide and the gut microbiota: Considering structural variation. FEBS Lett. 2022, 596, 849–875. [Google Scholar] [CrossRef]
  70. Li, D.; Lu, Y.; Yuan, S.; Cai, X.; He, Y.; Chen, J.; Wu, Q.; He, D.; Fang, A.; Bo, Y.; et al. Gut microbiota–derived metabolite trimethylamine-N-oxide and multiple health outcomes: An umbrella review and updated meta-analysis. Am. J. Clin. Nutr. 2022, 116, 230–243. [Google Scholar] [CrossRef]
  71. Ohira, H.; Tsutsui, W.; Fujioka, Y. Are Short Chain Fatty Acids in Gut Microbiota Defensive Players for Inflammation and Atherosclerosis? J. Atheroscler. Thromb. 2017, 24, 660–672. [Google Scholar] [CrossRef]
  72. Yan, J.; Pan, Y.; Shao, W.; Wang, C.; Wang, R.; He, Y.; Zhang, M.; Wang, Y.; Li, T.; Wang, Z.; et al. Beneficial effect of the short-chain fatty acid propionate on vascular calcification through intestinal microbiota remodelling. Microbiome 2022, 10, 195. [Google Scholar] [CrossRef]
  73. Swann, J.R.; Want, E.J.; Geier, F.M.; Spagou, K.; Wilson, I.D.; Sidaway, J.E.; Nicholson, J.K.; Holmes, E. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. 1), 4523–4530. [Google Scholar] [CrossRef] [PubMed]
  74. Nguyen, C.C.; Duboc, D.; Rainteau, D.; Sokol, H.; Humbert, L.; Seksik, P.; Bellino, A.; Abdoul, H.; Bouazza, N.; Treluyer, J.-M.; et al. Circulating bile acids concentration is predictive of coronary artery disease in human. Sci. Rep. 2021, 11, 22661. [Google Scholar] [CrossRef] [PubMed]
  75. Bao, M.; Zhang, P.; Guo, S.; Zou, J.; Ji, J.; Ding, X.; Yu, X. Altered gut microbiota and gut-derived p-cresyl sulfate serum levels in peritoneal dialysis patients. Front. Cell. Infect. Microbiol. 2022, 12, 639624. [Google Scholar] [CrossRef] [PubMed]
  76. Han, H.; Zhu, J.; Zhu, Z.; Ni, J.; Du, R.; Dai, Y.; Chen, Y.; Wu, Z.; Lu, L.; Zhang, R. p-Cresyl sulfate aggravates cardiac dysfunction associated with chronic kidney disease by enhancing apoptosis of cardiomyocytes. J. Am. Heart Assoc. 2015, 4, e001852. [Google Scholar] [CrossRef]
  77. Lin, X.; Liang, W.; Li, L.; Xiong, Q.; He, S.; Zhao, J.; Guo, X.; Xiang, S.; Zhang, P.; Wang, H.; et al. The Accumulation of Gut Microbiome–derived Indoxyl Sulfate and P-Cresyl Sulfate in Patients With End-stage Renal Disease. J. Ren. Nutr. 2022, 32, 578–586. [Google Scholar] [CrossRef]
  78. 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]
  79. Hess, M.; Sczyrba, A.; Egan, R.; Kim, T.-W.; Chokhawala, H.; Schroth, G.; Luo, S.; Clark, D.S.; Chen, F.; Zhang, T.; et al. Metagenomic discovery of biomass-degrading genes and genomes from cow rumen. Science 2011, 331, 463–467. [Google Scholar] [CrossRef]
  80. Tasse, L.; Bercovici, J.; Pizzut-Serin, S.; Robe, P.; Tap, J.; Klopp, C.; Cantarel, B.L.; Coutinho, P.M.; Henrissat, B.; Leclerc, M.; et al. Functional metagenomics to mine the human gut microbiome for dietary fiber catabolic enzymes. Genome Res. 2010, 20, 1605–1612. [Google Scholar] [CrossRef]
  81. Clausen, M.R.; Mortensen, P.B. Kinetic studies on colonocyte metabolism of short chain fatty acids and glucose in ulcerative colitis. Gut 1995, 37, 684–689. [Google Scholar] [CrossRef]
  82. Ritzhaupt, A.; Ellis, A.; Hosie, K.B.; Shirazi-Beechey, S.P. The characterization of butyrate transport across pig and human colonic luminal membrane. J. Physiol. 1998, 507 Pt 3, 819–830. [Google Scholar] [CrossRef]
  83. Frost, G.; Sleeth, M.L.; Sahuri-Arisoylu, M.; Lizarbe, B.; Cerdan, S.; Brody, L.; Anastasovska, J.; Ghourab, S.; Hankir, M.; Zhang, S.; et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat. Commun. 2014, 5, 3611. [Google Scholar] [CrossRef]
  84. Mascolo, N.; Rajendran, V.M.; Binder, H.J. Mechanism of short-chain fatty acid uptake by apical membrane vesicles of rat distal colon. Gastroenterology 1991, 101, 331–338. [Google Scholar] [CrossRef]
  85. 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]
  86. 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]
  87. Kasahara, K.; Krautkramer, K.A.; Org, E.; Romano, K.A.; Kerby, R.L.; Vivas, E.I.; Mehrabian, M.; Denu, J.M.; Bäckhed, F.; Lusis, A.J.; et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 2018, 3, 1461–1471. [Google Scholar] [CrossRef] [PubMed]
  88. Natarajan, A.; Nadarajah, V.; Felsovalyi, K.; Wang, W.; Jeyachandran, V.R.; Wasson, R.A.; Cardozo, T.; Bracken, C.; Krogsgaard, M. Structural Model of the Extracellular Assembly of the TCR-CD3 Complex. Cell Rep. 2016, 14, 2833–2845. [Google Scholar] [CrossRef] [PubMed]
  89. Pluznick, J.L.; Protzko, R.J.; Gevorgyan, H.; Peterlin, Z.; Sipos, A.; Han, J.; Brunet, I.; Wan, L.X.; Rey, F.; Wang, T.; et al. Olfactory receptor responding to gut microbiota derived signals plays a role in renin secretion and blood pressure regulation. Proc. Natl. Acad. Sci. USA 2013, 110, 4410–4415. [Google Scholar] [CrossRef]
  90. Natarajan, N.; Hori, D.; Flavahan, S.; Steppan, J.; Flavahan, N.A.; Berkowitz, D.E.; Pluznick, J.L. Microbial short chain fatty acid metabolites lower blood pressure via endothelial G protein-coupled receptor 41. Physiol. Genom. 2016, 48, 826–834. [Google Scholar] [CrossRef]
  91. Aguilar, E.; Leonel, A.; Teixeira, L.; Silva, A.; Silva, J.; Pelaez, J.; Capettini, L.; Lemos, V.; Santos, R.; Alvarez-Leite, J. Butyrate impairs atherogenesis by reducing plaque inflammation and vulnerability and decreasing NFκB activation. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 606–613. [Google Scholar] [CrossRef]
  92. Singh, V.; Chassaing, B.; Zhang, L.; Yeoh, B.S.; Xiao, X.; Kumar, M.; Baker, M.T.; Cai, J.; Walker, R.; Borkowski, K.; et al. Microbiota-Dependent Hepatic Lipogenesis Mediated by Stearoyl CoA Desaturase 1 (SCD1) Promotes Metabolic Syndrome in TLR5-Deficient Mice. Cell Metab. 2015, 22, 983–996. [Google Scholar] [CrossRef]
  93. Sanna, S.; Van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vila, A.V.; Võsa, U.; Mujagic, Z.; Masclee, A.A.M.; Jonkers, D.M.A.E.; Oosting, M.; et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 2019, 51, 600–605. [Google Scholar] [CrossRef]
  94. Tirosh, A.; Calay, E.S.; Tuncman, G.; Claiborn, K.C.; Inouye, K.E.; Eguchi, K.; Alcala, M.; Rathaus, M.; Hollander, K.S.; Ron, I.; et al. The short-chain fatty acid propionate increases glucagon and FABP4 production, impairing insulin action in mice and humans. Sci. Transl. Med. 2019, 11, eaav0120. [Google Scholar] [CrossRef]
  95. LeComte, V.; Kaakoush, N.O.; Maloney, C.A.; Raipuria, M.; Huinao, K.D.; Mitchell, H.M.; Morris, M.J. Changes in gut microbiota in rats fed a high fat diet correlate with obesity-associated metabolic parameters. PLoS ONE 2015, 10, e0126931. [Google Scholar] [CrossRef] [PubMed]
  96. Sayin, S.I.; Wahlström, A.; Felin, J.; Jäntti, S.; Marschall, H.-U.; Bamberg, K.; Angelin, B.; Hyötyläinen, T.; Orešič, M.; Bäckhed, F. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 2013, 17, 225–235. [Google Scholar] [CrossRef] [PubMed]
  97. Yao, L.; Seaton, S.C.; Ndousse-Fetter, S.; Adhikari, A.A.; DiBenedetto, N.; Mina, A.I.; Banks, A.S.; Bry, L.; Devlin, A.S. A selective gut bacterial bile salt hydrolase alters host metabolism. eLife 2018, 7, e37182. [Google Scholar] [CrossRef] [PubMed]
  98. Ridlon, J.M.; Kang, D.J.; Hylemon, P.B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 2006, 47, 241–259. [Google Scholar] [CrossRef]
  99. Hartman, H.B.; Gardell, S.J.; Petucci, C.J.; Wang, S.; Krueger, J.A.; Evans, M.J. Activation of farnesoid X receptor prevents atherosclerotic lesion formation in LDLR−/− and apoE−/− mice. J. Lipid Res. 2009, 50, 1090–1100. [Google Scholar] [CrossRef]
  100. 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]
  101. Guo, G.L.; Santamarina-Fojo, S.; Akiyama, T.E.; Amar, M.J.; Paigen, B.J.; Brewer, B.; Gonzalez, F.J. Effects of FXR in foam-cell formation and atherosclerosis development. Biochim. Biophys. Acta (BBA)-Mol. Cell Biol. Lipids 2006, 1761, 1401–1409. [Google Scholar] [CrossRef]
  102. Zhang, Y.; Wang, X.; Vales, C.; Lee, F.Y.; Lee, H.; Lusis, A.J.; Edwards, P.A. FXR deficiency causes reduced atherosclerosis in Ldlr−/− mice. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 2316–2321. [Google Scholar] [CrossRef] [PubMed]
  103. 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] [PubMed]
  104. Mayerhofer, C.C.; Ueland, T.; Broch, K.; Vincent, R.P.; Cross, G.F.; Dahl, C.P.; Aukrust, P.; Gullestad, L.; Hov, J.R.; Trøseid, M. Increased Secondary/Primary Bile Acid Ratio in Chronic Heart Failure. J. Card. Fail. 2017, 23, 666–671. [Google Scholar] [CrossRef] [PubMed]
  105. Li, W.; Shu, S.; Cheng, L.; Hao, X.; Wang, L.; Wu, Y.; Yuan, Z.; Zhou, J. Fasting serum total bile acid level is associated with coronary artery disease, myocardial infarction and severity of coronary lesions. Atherosclerosis 2020, 292, 193–200. [Google Scholar] [CrossRef]
  106. 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]
  107. Romano, K.A.; Vivas, E.I.; Amador-Noguez, D.; Rey, F.E.; Blaser, M.J. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. mBio 2015, 6, e02481. [Google Scholar] [CrossRef]
  108. 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]
  109. Li, X.S.; Obeid, S.; Klingenberg, R.; Gencer, B.; Mach, F.; Räber, L.; Windecker, S.; Rodondi, N.; Nanchen, D.; Muller, O.; et al. Gut microbiota-dependent trimethylamine N-oxide in acute coronary syndromes: A prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur. Heart J. 2017, 38, 814–824. [Google Scholar] [CrossRef]
  110. 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]
  111. 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-kappaB. J. Am. Heart Assoc. 2016, 5, e002767. [Google Scholar] [CrossRef]
  112. Zhu, Y.; Jameson, E.; Crosatti, M.; Schäfer, H.; Rajakumar, K.; Bugg, T.D.H.; Chen, Y. Carnitine metabolism to trimethylamine by an unusual Rieske-type oxygenase from human microbiota. Proc. Natl. Acad. Sci. USA 2014, 111, 4268–4273. [Google Scholar] [CrossRef] [PubMed]
  113. Rath, S.; Heidrich, B.; Pieper, D.H.; Vital, M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 2017, 5, 54. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, K.; Zheng, X.; Feng, M.; Li, D.; Zhang, H. Gut Microbiota-Dependent Metabolite Trimethylamine N-Oxide Contributes to Cardiac Dysfunction in Western Diet-Induced Obese Mice. Front. Physiol. 2017, 8, 139. [Google Scholar] [CrossRef] [PubMed]
  115. Aldana-Hernández, P.; Leonard, K.-A.; Zhao, Y.-Y.; Curtis, J.M.; Field, C.J.; Jacobs, R.L. Dietary Choline or Trimethylamine N-oxide Supplementation Does Not Influence Atherosclerosis Development in Ldlr−/− and Apoe−/− Male Mice. J. Nutr. 2020, 150, 249–255. [Google Scholar] [CrossRef]
  116. Wang, Z.; Bergeron, N.; Levison, B.S.; Li, X.S.; Chiu, S.; Jia, X.; Koeth, R.A.; Li, L.; Wu, Y.; Tang, W.H.W.; et al. Impact of chronic dietary red meat, white meat, or non-meat protein on trimethylamine N-oxide metabolism and renal excretion in healthy men and women. Eur. Heart J. 2019, 40, 583–594. [Google Scholar] [CrossRef]
  117. Wang, Z.; Roberts, A.B.; Buffa, J.A.; Levison, B.S.; Zhu, W.; Org, E.; Gu, X.; Huang, Y.; Zamanian-Daryoush, M.; Culley, M.K.; et al. Non-lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef]
  118. Tessari, P.; Lante, A.; Mosca, G. Essential amino acids: Master regulators of nutrition and environmental footprint? Sci. Rep. 2016, 6, 26074. [Google Scholar] [CrossRef]
  119. 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.e22. [Google Scholar] [CrossRef]
  120. Yu, F.; Feng, X.; Li, X.; Luo, Y.; Wei, M.; Zhao, T.; Xia, J. Gut-Derived Metabolite Phenylacetylglutamine and White Matter Hyperintensities in Patients With Acute Ischemic Stroke. Front. Aging Neurosci. 2021, 13, 675158. [Google Scholar] [CrossRef]
  121. 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] [PubMed]
  122. Cason, C.A.; Dolan, K.T.; Sharma, G.; Tao, M.; Kulkarni, R.; Helenowski, I.B.; Doane, B.M.; Avram, M.J.; McDermott, M.M.; Chang, E.B.; et al. Plasma microbiome-modulated indole- and phenyl-derived metabolites associate with advanced atherosclerosis and postoperative outcomes. J. Vasc. Surg. 2018, 68, 1552–1562.e7. [Google Scholar] [CrossRef] [PubMed]
  123. Lam, V.; Su, J.; Hsu, A.; Gross, G.J.; Salzman, N.H.; Baker, J.E.; Calvert, J. Intestinal Microbial Metabolites Are Linked to Severity of Myocardial Infarction in Rats. PLoS ONE 2016, 11, e0160840. [Google Scholar] [CrossRef] [PubMed]
  124. Lin, C.-J.; Liu, H.-L.; Pan, C.-F.; Chuang, C.-K.; Jayakumar, T.; Wang, T.-J.; Chen, H.-H.; Wu, C.-J. Indoxyl sulfate predicts cardiovascular disease and renal function deterioration in advanced chronic kidney disease. Arch. Med. Res. 2012, 43, 451–456. [Google Scholar] [CrossRef]
  125. Glorieux, G.; Vanholder, R.; Van Biesen, W.; Pletinck, A.; Schepers, E.; Neirynck, N.; Speeckaert, M.; De Bacquer, D.; Verbeke, F. Free p-cresyl sulfate shows the highest association with cardiovascular outcome in chronic kidney disease. Nephrol. Dial. Transplant. 2021, 36, 998–1005. [Google Scholar] [CrossRef]
  126. Shafi, T.; Sirich, T.L.; Meyer, T.W.; Hostetter, T.H.; Plummer, N.S.; Hwang, S.; Melamed, M.L.; Banerjee, T.; Coresh, J.; Powe, N.R. Results of the HEMO Study suggest that p-cresol sulfate and indoxyl sulfate are not associated with cardiovascular outcomes. Kidney Int. 2017, 92, 1484–1492. [Google Scholar] [CrossRef]
  127. Ross, F.C.; Mayer, D.E.; Horn, J.; Cryan, J.F.; Del Rio, D.; Randolph, E.; Gill, C.I.R.; Gupta, A.; Ross, R.P.; Stanton, C.; et al. Potential of dietary polyphenols for protection from age-related decline and neurodegeneration: A role for gut microbiota? Nutr. Neurosci. 2024, 27, 1058–1076. [Google Scholar] [CrossRef]
  128. Ottaviani, J.I.; Borges, G.; Momma, T.Y.; Spencer, J.P.E.; Keen, C.L.; Crozier, A.; Schroeter, H. The metabolome of [2-(14)C](-)-epicatechin in humans: Implications for the assessment of efficacy, safety, and mechanisms of action of polyphenolic bioactives. Sci. Rep. 2016, 6, 29034. [Google Scholar] [CrossRef]
  129. Mell, B.; Jala, V.R.; Mathew, A.V.; Byun, J.; Waghulde, H.; Zhang, Y.; Haribabu, B.; Vijay-Kumar, M.; Pennathur, S.; Joe, B. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiol. Genom. 2015, 47, 187–197. [Google Scholar] [CrossRef]
  130. 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]
  131. 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]
  132. 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]
  133. 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]
  134. Santisteban, M.M.; Qi, Y.; Zubcevic, J.; Kim, S.; Yang, T.; Shenoy, V.; Cole-Jeffrey, C.T.; Lobaton, G.O.; Stewart, D.C.; Rubiano, A.; et al. Hypertension-Linked Pathophysiological Alterations in the Gut. Circ. Res. 2017, 120, 312–323. [Google Scholar] [CrossRef] [PubMed]
  135. Adnan, S.; Nelson, J.W.; Ajami, N.J.; Venna, V.R.; Petrosino, J.F.; Bryan, R.M.; Durgan, D.J. Alterations in the gut microbiota can elicit hypertension in rats. Physiol. Genom. 2017, 49, 96–104. [Google Scholar] [CrossRef] [PubMed]
  136. Honour, J. The possible involvement of intestinal bacteria in steroidal hypertension. Endocrinology 1982, 110, 285–287. [Google Scholar] [CrossRef]
  137. Luedde, M.; Winkler, T.; Heinsen, F.-A.; Rühlemann, M.C.; Spehlmann, M.E.; Bajrovic, A.; Lieb, W.; Franke, A.; Ott, S.J.; Frey, N. Heart failure is associated with depletion of core intestinal microbiota. ESC Heart Fail. 2017, 4, 282–290. [Google Scholar] [CrossRef]
  138. Sandek, A.; Bauditz, J.; Swidsinski, A.; Buhner, S.; Weber-Eibel, J.; von Haehling, S.; Schroedl, W.; Karhausen, T.; Doehner, W.; Rauchhaus, M.; et al. Altered intestinal function in patients with chronic heart failure. J. Am. Coll. Cardiol. 2007, 50, 1561–1569. [Google Scholar] [CrossRef]
  139. Sandek, A.; Bjarnason, I.; Volk, H.-D.; Crane, R.; Meddings, J.B.; Niebauer, J.; Kalra, P.R.; Buhner, S.; Herrmann, R.; Springer, J.; et al. Studies on bacterial endotoxin and intestinal absorption function in patients with chronic heart failure. Int. J. Cardiol. 2012, 157, 80–85. [Google Scholar] [CrossRef]
  140. Krack, A.; Richartz, B.M.; Gastmann, A.; Greim, K.; Lotze, U.; Anker, S.D.; Figulla, H.R. Studies on intragastric PCO2 at rest and during exercise as a marker of intestinal perfusion in patients with chronic heart failure. Eur. J. Heart Fail. 2004, 6, 403–407. [Google Scholar] [CrossRef]
  141. Pasini, E.; Aquilani, R.; Testa, C.; Baiardi, P.; Angioletti, S.; Boschi, F.; Verri, M.; Dioguardi, F. Pathogenic Gut Flora in Patients With Chronic Heart Failure. JACC Heart Fail. 2016, 4, 220–227. [Google Scholar] [CrossRef]
  142. 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]
  143. Campbell, R.B.P.; Duffourc, M.M.; Schoborg, R.V.; Xu, Y.; Liu, X.; KenKnight, B.H.; Beaumont, E. Aberrant fecal flora observed in guinea pigs with pressure overload is mitigated in animals receiving vagus nerve stimulation therapy. Am. J. Physiol.-Gastrointest. Liver Physiol. 2016, 311, G754–G762. [Google Scholar] [CrossRef]
  144. Tang, W.H.W.; Wang, Z.; Wu, Y.; Fan, Y.; Koeth, R.A.; Hazen, S. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: Refining the gut hypothesis. J. Am. Coll. Cardiol. 2013, 61, E750. [Google Scholar] [CrossRef]
  145. Li, Z.; Wu, Z.; Yan, J.; Liu, H.; Liu, Q.; Deng, Y.; Ou, C.; Chen, M. Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Lab. Investig. 2019, 99, 346–357. [Google Scholar] [CrossRef] [PubMed]
  146. Piepoli, M.F.; Hoes, A.W.; Agewall, S.; Albus, C.; Brotons, C.; Catapano, A.L.; Cooney, M.-T.; Corrà, U.; Cosyns, B.; Deaton, C.; et al. 2016 European Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2016, 37, 2315–2381. [Google Scholar] [CrossRef] [PubMed]
  147. Ahmadi, A.; Argulian, E.; Leipsic, J.; Newby, D.E.; Narula, J. From Subclinical Atherosclerosis to Plaque Progression and Acute Coronary Events: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 74, 1608–1617. [Google Scholar] [CrossRef] [PubMed]
  148. Farmakis, D.; Mueller, C.; Apple, F.S. High-sensitivity cardiac troponin assays for cardiovascular risk stratification in the general population. Eur. Heart J. 2020, 41, 4050–4056. [Google Scholar] [CrossRef]
  149. Jeremias, A.; Kleiman, N.S.; Nassif, D.; Hsieh, W.-H.; Pencina, M.; Maresh, K.; Parikh, M.; Cutlip, D.E.; Waksman, R.; Goldberg, S.; et al. Prevalence and prognostic significance of preprocedural cardiac troponin elevation among patients with stable coronary artery disease undergoing percutaneous coronary intervention: Results from the evaluation of drug eluting stents and ischemic events registry. Circulation 2008, 118, 632–638. [Google Scholar]
  150. Sullivan, J.F.O.; Neylon, A.; McGorrian, C.; Blake, G.J. miRNA-93-5p and other miRNAs as predictors of coronary artery disease and STEMI. Int. J. Cardiol. 2016, 224, 310–316. [Google Scholar] [CrossRef]
  151. Su, J.; Li, J.; Yu, Q.; Wang, J.; Li, X.; Yang, J.; Xu, J.; Liu, Y.; Xu, Z.; Ji, L.; et al. Exosomal miRNAs as potential biomarkers for acute myocardial infarction. IUBMB Life 2020, 72, 384–400. [Google Scholar] [CrossRef]
  152. Ren, X.; Ellis, B.W.; Ronan, G.; Blood, S.R.; DeShetler, C.; Senapati, S.; March, K.L.; Handberg, E.; Anderson, D.; Pepine, C.; et al. A multiplexed ion-exchange membrane-based miRNA (MIX.miR) detection platform for rapid diagnosis of myocardial infarction. Lab Chip 2021, 21, 3876–3887. [Google Scholar] [CrossRef]
  153. Wu, Z.-X.; Li, S.-F.; Chen, H.; Song, J.-X.; Gao, Y.-F.; Zhang, F.; Cao, C.-F.; Ahuja, S.K. The changes of gut microbiota after acute myocardial infarction in rats. PLoS ONE 2017, 12, e0180717. [Google Scholar] [CrossRef]
  154. Han, Y.; Gong, Z.; Sun, G.; Xu, J.; Qi, C.; Sun, W.; Jiang, H.; Cao, P.; Ju, H. Dysbiosis of Gut Microbiota in Patients With Acute Myocardial Infarction. Front. Microbiol. 2021, 12, 680101. [Google Scholar] [CrossRef]
  155. Liu, H.; Chen, X.; Hu, X.; Niu, H.; Tian, R.; Wang, H.; Pang, H.; Jiang, L.; Qiu, B.; Chen, X.; et al. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome 2019, 7, 68. [Google Scholar] [CrossRef] [PubMed]
  156. Dong, C.; Yang, Y.; Wang, Y.; Hu, X.; Wang, Q.; Gao, F.; Sun, S.; Liu, Q.; Li, L.; Liu, J.; et al. Gut microbiota combined with metabolites reveals unique features of acute myocardial infarction patients different from stable coronary artery disease. J. Adv. Res. 2022, 46, 101–112. [Google Scholar] [CrossRef]
  157. Pedersen, E.R.; Tuseth, N.; Eussen, S.J.; Ueland, P.M.; Strand, E.; Svingen, G.F.T.; Midttun, Ø.; Meyer, K.; Mellgren, G.; Ulvik, A.; et al. Associations of plasma kynurenines with risk of acute myocardial infarction in patients with stable angina pectoris. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 455–462. [Google Scholar] [CrossRef] [PubMed]
  158. Zhang, L.; Wei, T.-T.; Li, Y.; Li, J.; Fan, Y.; Huang, F.-Q.; Cai, Y.-Y.; Ma, G.; Liu, J.-F.; Chen, Q.-Q.; et al. Functional Metabolomics Characterizes a Key Role for N -Acetylneuraminic Acid in Coronary Artery Diseases. Circulation 2018, 137, 1374–1390. [Google Scholar] [CrossRef] [PubMed]
  159. Davignon, J.; Ganz, P. Role of endothelial dysfunction in atherosclerosis. Circulation 2004, 109, III27–III32. [Google Scholar] [CrossRef]
  160. Libby, P.; Ridker, P.M.; Maseri, A. Inflammation and atherosclerosis. Circulation 2002, 105, 1135–1143. [Google Scholar] [CrossRef]
  161. 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]
  162. Yang, L.; Yang, J.; Zhang, T.; Xie, X.; Wu, Q.; Sui, Y. Gut microbiota: A novel strategy affecting atherosclerosis. Microbiol. Spectr. 2025, 13, 8. [Google Scholar] [CrossRef]
  163. Karlsson, F.H.; Fåk, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Bäckhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012, 3, 1245. [Google Scholar] [CrossRef] [PubMed]
  164. Gózd-Barszczewska, A.; Kozioł-Montewka, M.; Barszczewski, P.; Młodzińska, A.; Humińska, K. Gut microbiome as a biomarker of cardiometabolic disorders. Ann. Agric. Environ. Med. 2017, 24, 416–422. [Google Scholar] [CrossRef] [PubMed]
  165. Jie, Z.; Xia, H.; Zhong, S.-L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845. [Google Scholar] [CrossRef]
  166. Kitai, T.; Tang, W.W. Gut microbiota in cardiovascular disease and heart failure. Clin. Sci. 2018, 132, 85–91. [Google Scholar] [CrossRef]
  167. Tang, W.W.; Kitai, T.; Hazen, S.L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017, 120, 1183–1196. [Google Scholar] [CrossRef] [PubMed]
  168. Zabell, A.; Tang, W.H.W. Targeting the Microbiome in Heart Failure. Curr. Treat. Options Cardiovasc. Med. 2017, 19, 27. [Google Scholar] [CrossRef]
  169. Lindberg, T.; Wimo, A.; Elmståhl, S.; Qiu, C.; Bohman, D.M.; Berglund, J.S. Prevalence and Incidence of Atrial Fibrillation and Other Arrhythmias in the General Older Population: Findings From the Swedish National Study on Aging and Care. Gerontol. Geriatr. Med. 2019, 5, 2333721419859687. [Google Scholar] [CrossRef]
  170. Kotecha, D.; Piccini, J.P. Atrial fibrillation in heart failure: What should we do? Eur. Heart J. 2015, 36, 3250–3257. [Google Scholar] [CrossRef]
  171. Kaplan, R.M.; Koehler, J.; Ziegler, P.D.; Sarkar, S.; Zweibel, S.; Passman, R.S. Stroke Risk as a Function of Atrial Fibrillation Duration and CHA2DS2-VASc Score. Circulation 2019, 140, 1639–1646. [Google Scholar] [CrossRef]
  172. 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]
  173. Zuo, K.; Li, J.; Wang, P.; Liu, Y.; Liu, Z.; Yin, X.; Liu, X.; Yang, X.; Chia, N. Duration of Persistent Atrial Fibrillation Is Associated with Alterations in Human Gut Microbiota and Metabolic Phenotypes. mSystems 2019, 4, e00422-19. [Google Scholar] [CrossRef]
  174. Okazaki, R.; Iwasaki, Y.-K.; Miyauchi, Y.; Hirayama, Y.; Kobayashi, Y.; Katoh, T.; Mizuno, K.; Sekiguchi, A.; Yamashita, T. Lipopolysaccharide induces atrial arrhythmogenesis via down-regulation of L-type Ca2+ channel genes in rats. Int. Heart J. 2009, 50, 353–363. [Google Scholar] [CrossRef]
  175. Pastori, D.; Carnevale, R.; Nocella, C.; Novo, M.; Santulli, M.; Cammisotto, V.; Menichelli, D.; Pignatelli, P.; Violi, F. Gut-Derived Serum Lipopolysaccharide is Associated With Enhanced Risk of Major Adverse Cardiovascular Events in Atrial Fibrillation: Effect of Adherence to Mediterranean Diet. J. Am. Heart Assoc. 2017, 6, e005784. [Google Scholar] [CrossRef]
  176. Navarro-Polanco, R.A.; Aréchiga-Figueroa, I.A.; Salazar-Fajardo, P.D.; Benavides-Haro, D.E.; Rodríguez-Elías, J.C.; Sachse, F.B.; Tristani-Firouzi, M.; Sánchez-Chapula, J.A.; Moreno-Galindo, E.G. Voltage sensitivity of M2 muscarinic receptors underlies the delayed rectifier-like activation of ACh-gated K(+) current by choline in feline atrial myocytes. J. Physiol. 2013, 591, 4273–4286. [Google Scholar] [CrossRef] [PubMed]
  177. Safari-Alighiarloo, N.; Emami, Z.; Rezaei-Tavirani, M.; Alaei-Shahmiri, F.; Razavi, S. Gut Microbiota and Their Associated Metabolites in Diabetes: A Cross Talk Between Host and Microbes—A Review. Metab. Syndr. Relat. Disord. 2023, 21, 3–15. [Google Scholar] [CrossRef] [PubMed]
  178. Saad, M.J.A.; Santos, A. The Microbiota and Evolution of Obesity. Endocr. Rev. 2025, 46, 300–316. [Google Scholar] [CrossRef] [PubMed]
  179. 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]
  180. Ley, R.E.; Bäckhed, 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]
  181. Leite, G.; Barlow, G.M.; Rashid, M.; Hosseini, A.; Cohrs, D.; Parodi, G.; Morales, W.; Weitsman, S.; Rezaie, A.; Pimentel, M.; et al. Characterization of the Small Bowel Microbiome Reveals Different Profiles in Human Subjects Who Are Overweight or Have Obesity. Am. J. Gastroenterol. 2024, 119, 1141–1153. [Google Scholar] [CrossRef] [PubMed]
  182. 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]
  183. 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]
  184. 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]
  185. Larsen, N.; Vogensen, F.K.; Van Den Berg, F.W.J.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sørensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef]
  186. Hosomi, K.; Saito, M.; Park, J.; Murakami, H.; Shibata, N.; Ando, M.; Nagatake, T.; Konishi, K.; Ohno, H.; Tanisawa, K.; et al. Oral administration of Blautia wexlerae ameliorates obesity and type 2 diabetes via metabolic remodeling of the gut microbiota. Nat. Commun. 2022, 13, 4477. [Google Scholar] [CrossRef]
  187. Wang, T.-Y.; Zhang, X.-Q.; Chen, A.-L.; Zhang, J.; Lv, B.-H.; Ma, M.-H.; Lian, J.; Wu, Y.-X.; Zhou, Y.-T.; Ma, C.-C.; et al. A comparative study of microbial community and functions of type 2 diabetes mellitus patients with obesity and healthy people. Appl. Microbiol. Biotechnol. 2020, 104, 7143–7153. [Google Scholar] [CrossRef]
  188. Suzuki, K.; Sagara, M.; Aoki, C.; Tanaka, S.; Aso, Y. Clinical implication of plasma hydrogen sulfide levels in Japanese patients with type 2 diabetes. Intern. Med. 2017, 56, 17–21. [Google Scholar] [CrossRef]
  189. Kosmas, C.E.; Bousvarou, M.D.; Kostara, C.E.; Papakonstantinou, E.J.; Salamou, E.; Guzman, E. Insulin resistance and cardiovascular disease. J. Int. Med. Res. 2023, 51, 03000605231164548. [Google Scholar] [CrossRef]
  190. Cianciulli, A.; Calvello, R.; Porro, C.; Trotta, T.; Salvatore, R.; Panaro, M.A. PI3k/Akt signalling pathway plays a crucial role in the anti-inflammatory effects of curcumin in LPS-activated microglia. Int. Immunopharmacol. 2016, 36, 282–290. [Google Scholar] [CrossRef]
  191. Makki, K.; Deehan, E.C.; Walter, J.; Bäckhed, F. The Impact of Dietary Fiber on Gut Microbiota in Host Health and Disease. Cell Host Microbe 2018, 23, 705–715. [Google Scholar] [CrossRef]
  192. Qi, Y.; Aranda, J.M.; Rodriguez, V.; Raizada, M.K.; Pepine, C.J. Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension—A case report. Int. J. Cardiol. 2015, 201, 157–158. [Google Scholar] [CrossRef]
  193. Mastrangelo, A.; Robles-Vera, I.; Mañanes, D.; Galán, M.; Femenía-Muiña, M.; Redondo-Urzainqui, A.; Barrero-Rodríguez, R.; Papaioannou, E.; Amores-Iniesta, J.; Devesa, A.; et al. Imidazole propionate is a driver and therapeutic target in atherosclerosis. Nature 2025, 1–8. [Google Scholar] [CrossRef]
  194. Han, J.; Zhang, R.; Muheyati, D.; Lv, M.X.; Aikebaier, W.; Peng, B.X. The Effect of Chickpea Dietary Fiber on Lipid Metabolism and Gut Microbiota in High-Fat Diet-Induced Hyperlipidemia in Rats. J. Med. Food 2021, 24, 124–134. [Google Scholar] [CrossRef] [PubMed]
  195. Zhou, A.L.; Hergert, N.; Rompato, G.; Lefevre, M. Whole grain oats improve insulin sensitivity and plasma cholesterol profile and modify gut microbiota composition in C57BL/6J mice. J. Nutr. 2015, 145, 222–230. [Google Scholar] [CrossRef] [PubMed]
  196. Parnell, J.A.; Reimer, R.A. Effect of prebiotic fibre supplementation on hepatic gene expression and serum lipids: A dose–response study in JCR:LA-cp rats. Br. J. Nutr. 2010, 103, 1577–1584. [Google Scholar] [CrossRef] [PubMed]
  197. Rault-Nania, M.-H.; Gueux, E.; Demougeot, C.; Demigné, C.; Rock, E.; Mazur, A. Inulin attenuates atherosclerosis in apolipoprotein E-deficient mice. Br. J. Nutr. 2006, 96, 840–844. [Google Scholar] [CrossRef]
  198. Tiwari, U.; Cummins, E. Meta-analysis of the effect of beta-glucan intake on blood cholesterol and glucose levels. Nutrition 2011, 27, 1008–1016. [Google Scholar] [CrossRef]
  199. Cani, P.D.; Neyrinck, A.M.; Fava, F.; Knauf, C.; Burcelin, R.G.; Tuohy, K.M.; Gibson, G.R.; Delzenne, N.M. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007, 50, 2374–2383. [Google Scholar] [CrossRef]
  200. Kaye, D.M.; Shihata, W.A.; Jama, H.A.; Tsyganov, K.; Ziemann, M.; Kiriazis, H.; Horlock, D.; Vijay, A.; Giam, B.; Vinh, A.; et al. Deficiency of Prebiotic Fiber and Insufficient Signaling Through Gut Metabolite-Sensing Receptors Leads to Cardiovascular Disease. Circulation 2020, 141, 1393–1403. [Google Scholar] [CrossRef]
  201. Robertson, M.D.; Bickerton, A.S.; Dennis, A.L.; Vidal, H.; Frayn, K.N. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am. J. Clin. Nutr. 2005, 82, 559–567. [Google Scholar] [CrossRef]
  202. Parnell, J.A.; Reimer, R.A. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am. J. Clin. Nutr. 2009, 89, 1751–1759. [Google Scholar] [CrossRef]
  203. Malik, M.; Suboc, T.M.; Tyagi, S.; Salzman, N.; Wang, J.; Ying, R.; Tanner, M.J.; Kakarla, M.; Baker, J.E.; Widlansky, M.E. Lactobacillus plantarum 299v Supplementation Improves Vascular Endothelial Function and Reduces Inflammatory Biomarkers in Men with Stable Coronary Artery Disease. Circ. Res. 2018, 123, 1091–1102. [Google Scholar] [CrossRef]
  204. Robles-Vera, I.; Toral, M.; de la Visitación, N.; Sánchez, M.; Romero, M.; Olivares, M.; Jiménez, R.; Duarte, J. The Probiotic Lactobacillus fermentum Prevents Dysbiosis and Vascular Oxidative Stress in Rats with Hypertension Induced by Chronic Nitric Oxide Blockade. Mol. Nutr. Food Res. 2018, 62, e1800298. [Google Scholar] [CrossRef] [PubMed]
  205. Ejtahed, H.S.; Mohtadi-Nia, J.; Homayouni-Rad, A.; Niafar, M.; Asghari-Jafarabadi, M.; Mofid, V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 2012, 28, 539–543. [Google Scholar] [CrossRef] [PubMed]
  206. Al-Sheraji, S.; Amin, I.; Azlan, A.; Manap, M.; Hassan, F. Effects of Bifidobacterium longum BB536 on lipid profile and histopathological changes in hypercholesterolaemic rats. Benef. Microbes 2015, 6, 661–668. [Google Scholar] [CrossRef] [PubMed]
  207. Wan, Y.; Wang, F.; Yuan, J.; Li, J.; Jiang, D.; Zhang, J.; Li, H.; Wang, R.; Tang, J.; Huang, T.; et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: A 6-month randomised controlled-feeding trial. Gut 2019, 68, 1417–1429. [Google Scholar] [CrossRef]
  208. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human genetics shape the gut microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef]
  209. Simon, M.-C.; Strassburger, K.; Nowotny, B.; Kolb, H.; Nowotny, P.; Burkart, V.; Zivehe, F.; Hwang, J.-H.; Stehle, P.; Pacini, G.; et al. Intake of Lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: A proof of concept. Diabetes Care 2015, 38, 1827–1834. [Google Scholar] [CrossRef]
  210. Dibaji, S.M.; Seidavi, A.; Asadpour, L.; da Silva, F.M. Effect of a synbiotic on the intestinal microflora of chickens. J. Appl. Poult. Res. 2014, 23, 1–6. [Google Scholar] [CrossRef]
  211. Mookiah, S.; Sieo, C.C.; Ramasamy, K.; Abdullah, N.; Ho, Y.W. Effects of dietary prebiotics, probiotic and synbiotics on performance, caecal bacterial populations and caecal fermentation concentrations of broiler chickens. J. Sci. Food Agric. 2013, 94, 341–348. [Google Scholar] [CrossRef]
  212. Ke, X.; Walker, A.; Haange, S.-B.; Lagkouvardos, I.; Liu, Y.; Schmitt-Kopplin, P.; von Bergen, M.; Jehmlich, N.; He, X.; Clavel, T.; et al. Synbiotic-driven improvement of metabolic disturbances is associated with changes in the gut microbiome in diet-induced obese mice. Mol. Metab. 2019, 22, 96–109. [Google Scholar] [CrossRef]
  213. Colman, R.J.; Rubin, D.T. Fecal microbiota transplantation as therapy for inflammatory bowel disease: A systematic review and meta-analysis. J. Crohn’s Colitis 2014, 8, 1569–1581. [Google Scholar] [CrossRef]
  214. Brandt, L.J.; Aroniadis, O.C.; Mellow, M.; Kanatzar, A.; Kelly, C.; Park, T.; Stollman, N.; Rohlke, F.; Surawicz, C. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am. J. Gastroenterol. 2012, 107, 1079–1087. [Google Scholar] [CrossRef]
Figure 1. Dysbiosis of gut microbiota and related human diseases.
Figure 1. Dysbiosis of gut microbiota and related human diseases.
Cells 14 01237 g001
Figure 2. Gut microbiota and metabolite’s role in cardiovascular diseases.
Figure 2. Gut microbiota and metabolite’s role in cardiovascular diseases.
Cells 14 01237 g002
Table 1. Key gut microbiome-derived metabolites and their functions and roles in CVDs.
Table 1. Key gut microbiome-derived metabolites and their functions and roles in CVDs.
MetaboliteMicrobiotaExamplesFunctionsMechanismsRole in CVDs
SCFAsButyricimonas, Akkermansia, Bacteroides, Prevotella,
Enterolactone Anaerostipes, Blautia, Coprococcus, Eubacterium, Faecalibacterium, Marvinbryantia, Megasphaera,
Roseburia, Ruminococcus
Acetate, propionate, butyrateRegulate immune responses and inflammation, maintain gut barrier integrity, influence glucose and lipid metabolismAct via G-protein-coupled receptors (e.g., GPR41, GPR43) and inhibits histone deacetylases (HDACs)SCFAs increase atherosclerosis, heart failure, and hypertension
TMAOCollinsella, Escherichia,
Shigella, Enterococcus
Micrococcus, Mobiluncus
Clostridium, Staphylococcus, Sarcina, Campylobacter, Pseudomonas, Anaerococcus, Desulfovibrio, Edwardsiella,
Proteus, Providencia
Trimethylamine (TMA), oxidized in the liver to TMAOPromotes atherosclerosis, associated with cardiovascular disease (CVD) riskAlters cholesterol metabolism and enhances platelet aggregationTMAO increases atherosclerosis and heart failure
Aromatic Amino Acids (AAAs)Bacteroides, Bifidobacterium, Clostridium, Lactobacillus, Peptostreptococcus, Ruminococcus, RuminiclostridiumIndole, indole-3-acetic acid (IAA), indole-3-propionic acid (IPA), phenylacetylglutamineModulate mucosal immunity via aryl hydrocarbon receptor (AhR), influence gut-brain axis and neuroinflammationAct via G-protein-coupled receptorsRole of indoxyl sulfate in aortic calcification and increased carotid intima media thickness
Bile acids (BAs)Butyricimonas, Akkermansia, Bacteroides, Prevotella, EnterolactoneDeoxycholic acid (DCA), lithocholic acid (LCA)Regulate lipid and glucose metabolism, modulate signaling via FXR and TGR5 receptorsDysregulation linked to insulin resistance and NAFLDRole in myocardial fibrosis
Table 2. CVD-related key gut microbiota and related metabolites.
Table 2. CVD-related key gut microbiota and related metabolites.
DiseasesMicrobiotaMetabolites
AtherosclerosisEnterobacteriaceae, Ruminococcus gnavus, Eggerthella lenta, Roseburia intestinalis, Faecalibacterium cf., prausnitzii, Lactobacillus, Streptococcus, Clostridium subcluster, BacteroidesIncrease TMAO
Myocardial infarctionTissierella soehngenia genera, Lactobacillus plantarum 299v, Bacteroides fragilisTMAO and SCFAs
Heart failureFaecalibacterium prausnitzii, Bacteroides fragilis, Escherichia coli, Klebsiella pneumonia, Streptococcus viridans, Campylobacter, Candida, Shigella, Salmonella, Yersinia enterocoliticaIncrease TMAO
ArrhythmiaRuminococcus, Streptococcus, Enterococcus, Faecalibacterium, Alistipes, Oscillibacter, BilophilaRole of TMAO in arrythmia
HypertensionFirmicutes, Bacteroides, Prevotella, Erwinia, Corynebacteriac-eae, Anaerostipes, Lactobacillus murinus, Roseburia intestinalisIncrease SCFAs
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

Kumar, J. Microbial Metabolites and Cardiovascular Dysfunction: A New Era of Diagnostics and Therapy. Cells 2025, 14, 1237. https://doi.org/10.3390/cells14161237

AMA Style

Kumar J. Microbial Metabolites and Cardiovascular Dysfunction: A New Era of Diagnostics and Therapy. Cells. 2025; 14(16):1237. https://doi.org/10.3390/cells14161237

Chicago/Turabian Style

Kumar, Jitendra. 2025. "Microbial Metabolites and Cardiovascular Dysfunction: A New Era of Diagnostics and Therapy" Cells 14, no. 16: 1237. https://doi.org/10.3390/cells14161237

APA Style

Kumar, J. (2025). Microbial Metabolites and Cardiovascular Dysfunction: A New Era of Diagnostics and Therapy. Cells, 14(16), 1237. https://doi.org/10.3390/cells14161237

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