Open Access

Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review)

  • Authors:
    • Li Lin
    • Shaowei Xiang
    • Yuan Chen
    • Yan Liu
    • Dingwen Shen
    • Xiaoping Yu
    • Zhe Wu
    • Yanling Sun
    • Kequan Chen
    • Jia Luo
    • Guilai Wei
    • Zhiguo Wang
    • Zhifeng Ning
  • View Affiliations

  • Published online on: September 11, 2024     https://doi.org/10.3892/etm.2024.12716
  • Article Number: 427
  • Copyright: © Lin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The gut microbiota refers to the diverse bacterial community residing in the gastrointestinal tract. Recent data indicate a strong correlation between alterations in the gut microbiota composition and the onset of various diseases, notably cardiovascular disorders. Evidence suggests the gut‑cardiovascular axis signaling molecules released by the gut microbiota play a pivotal role in regulation. This review systematically delineates the association between dysbiosis of the gut microbiota and prevalent cardiovascular diseases, including atherosclerosis, hypertension, myocardial infarction and heart failure. Furthermore, it provides an overview of the putative pathogenic mechanisms by which dysbiosis in the gut microbiota contributes to the progression of cardiovascular ailments. The potential modulation of gut microbiota as a preventive strategy against cardiovascular diseases through dietary interventions, antibiotic therapies and probiotic supplementation is also explored and discussed within the present study.

1. Introduction

The human gut microbiota constitutes a highly intricate community comprised of ~500-1,000 bacterial species (1). Despite this diversity, a substantial discrepancy exists in the abundance of each bacterial type. Notably, a mere 30-40 species contribute to >99% of the total bacterial population, with the remaining species collectively constituting less than 1% (2). Crucially, the gut microbiota plays a pivotal role in essential physiological functions including immunity (3), metabolism (4), inflammation (5) and cell proliferation (6). These microorganisms not only interact extensively with the intestinal epithelium but also engage in communication with diverse organ systems. It is estimated that the cumulative weight of gut bacteria in an adult can range between 1.0-1.5 kg, housing an immense population of ~1014 bacteria (7). The intestinal genome contains millions of genes, 100 times more than the human genome and contains various gene clusters associated with metabolic processes, immune responses and multiple signaling pathways (8). Beyond their influence on body mass and metabolism, the gut microbiota is intricately linked to variety of diseases, including, but not limited to, inflammatory bowel disease, diabetes, obesity, gastrointestinal cancer, cirrhosis, hyperuricemia and autoimmune disorders (9-11). The significance of a healthy gut microbiota as a crucial determinant of overall health cannot be understated, as alterations in its composition strongly correlate with changes in an individual's health status. The National Institutes of Health (NIH) has undertaken a substantial commitment to human microbiome research, investing a substantial $15 billion since 2008. This extensive investment has facilitated comprehensive population studies focusing on microbial populations across various tissues such as the skin, respiratory tract, gastrointestinal tract, urinary tract and vaginal tract (12). Since the commencement of the NIH Human Microecology Program and the EU Intestinal Microecology Program, gut microbiota has attracted much attention and has become an international research hotspot. Now, a number of countries in the world have launched human microbiome projects, including Canada, Australia, France, North Korea and Ireland (13).

Advancements in molecular biology techniques have greatly expanded our understanding of gut microbiota. Techniques such as fluorescence in situ hybridization, polymerase chain reaction denaturing gradient gel electrophoresis, gene chips and sequencing have played a vital role. These techniques have enabled more detailed and accurate analyses of gut microbiota. Specifically, next-generation sequencing and macrogenomics have been revolutionary, notably advancing the study of the gut microbiome. These advances mark a significant leap forward in this field of research (14,15).

The morbidity and mortality of cardiovascular diseases have been increasing globally, posing a serious threat to human health. Consequently, there is a pressing need for early preventive measures and innovative treatment strategies for cardiovascular diseases. This has become a focal point of research in the field. Recent studies exploring the relationship between gut microbiota and various diseases have shed light on the crucial role these microbes play in human health (16-18). Recent investigations have highlighted a connection between gut microbiota and cardiovascular disease (19,20). Modulating gut microbiota is anticipated as a potential emerging therapeutic approach for managing cardiovascular conditions (21,22).

Due to of the multiple functions of the gut microbiota, scientists have focused on comprehensive studies of this intricate ecosystem. The traditional approach to study microorganisms is to analyze the physiological functions of individual strains or groups of bacteria using in vitro culture. However, numerous species within the gut microbiota can only thrive within the human body and are challenging to culture in vitro. Consequently, traditional methods face limitations when addressing the complexity of the gut microbiota. However, with the advancement of sequencing technology, especially the increasing maturity of macrogenomics, the composition and complexity of gut microbiota were clarified. This breakthrough enables a more profound exploration of the relationship between the gut microbiota and human health, overcoming the constraints of traditional methodologies.

The present review provided a systematic exploration of the link between imbalances in gut microbiota and various cardiovascular diseases, such as atherosclerosis, hypertension, heart failure and myocardial infarction. Drawing from recent clinical research and biological studies, it delineates the association between dysbiosis of gut microbiota and the onset, as well as the advancement, of cardiovascular diseases. Emphasizing this connection, the present review underscores the potential of modulating gut microbiota as a viable therapeutic avenue for managing cardiovascular conditions.

2. Methods

To generate the initial list of studies in this review, literature searches were conducted on PubMed using specific keywords such as ‘gut microbiome’ or ‘gut microbiota’ combined with terms in the category of cardiovascular diseases such as ‘Atherosclerosis’, ‘Hypertension’, ‘Heart failure’ and ‘Cardiac infarction’. These searches were performed to identify the most recent research papers published within the last decade (10 years). Papers published within the past 5 years were prioritized to ensure the inclusion of the most current findings in the present review. Additionally, the inclusion of the papers published in high impact journals and those with higher citation numbers were prioritized. Through these specific criteria, the literature collection process collected ~200 reviews and research papers. To ensure the relevance of the information presented in the present review, papers published within the last 10 years were excluded if they had subsequent updates. Research articles older than 10 years were included if they keep having recent citations or ongoing significance within the field. This selection process helped us compile a comprehensive and up-to-date set of literature for the present review.

3. Effects of gut microbiota on cardiovascular diseases

Composition of microbiota and linkage with cardiovascular diseases

The gut microbiota is composed of four main phyla: Firmicutes, Bacteroidetes, Actinobacteria and Proteobacteria. Saprophytes and Bacteroidetes constitute the majority of the intestinal community in healthy adults and the ratio of Firmicutes:Bacteroidetes is considered an indicator of the health of the gut microbiota (23,24). However, microbial composition varies from person to person and is dynamically sensitive to host factors and environmental parameters (25). Notably, increased abundance of host opportunistic pathogens (e.g., E. coli, Clostridium lambda, C. carinii and Eggerthella lenta) and decreased short-chain fatty acid (SCFA)-producing bacteria (e.g., Roseburia, Faecalibacterium and Eubacterium rectale) are both associated with an increased risk of cardiovascular diseases (26). Thus, both the type of microorganism and their relative abundance are potential risk factors that may alter susceptibility to developing cardiovascular diseases.

Atherosclerosis

Atherosclerosis is the most important and common group of atherosclerotic vascular diseases and the pathogenesis of this disease has been discussed in various theories (27), including the lipid infiltration theory, thrombosis theory, smooth muscle cell cloning theory, inflammation theory and infection theory. In recent years, most researchers have supported the endothelial injury response theory, suggesting that the main risk factors for the disease ultimately damage the intima and that the formation of atherosclerotic lesions is the result of the inflammatory response of the arteries to intimal damage (28). Inflammation plays a key role in the onset and progression of atherosclerosis (29). Currently, bacteria are considered to be a contributing factor to the disease.

In the intestine of patients with symptomatic atherosclerosis, there are changes in some genera, such as an increase in Corynebacterium, and in healthy controls, there were more Rhodobacter and Eubacterium. This difference suggests that changes in gut microbiota may be associated with atherosclerosis (30). In addition, some researchers have identified Aeromonas, Veronella and Streptococcus in atherosclerotic plaques in both animal and patient studies (31-33).

In an animal study, it was discovered that the gut microbiota's role in metabolizing and absorbing bile acids and cholesterol significantly influences blood cholesterol levels. These cholesterol levels, in turn, represent crucial factors contributing to the development of atherosclerosis (34). The mechanisms that affect cholesterol metabolism include the production of cholesterol oxidase, inhibition of hepatic lipid synthase activity, regulation of cholesterol redistribution in blood and liver and influence on bile salt hepato-intestinal circulation (35). In addition, the generation of gut microbiota-dependent trimethylamine N-oxide (TMAO) alters the main pathway of cholesterol removal in the body (36). TMAO is a substance that is metabolized in the liver after trimethylamine (TMA) is absorbed in the intestine (37). Trenteseaux et al (38) found that in mice, TMAO could cause intracellular accumulation of cholesterol by increasing the expression of proatherosclerotic CD36 and scavenger receptor A. TMAO also decreases Cyp7a1 expression, which is an important enzyme for bile acid synthesis, inhibits cholesterol transport and causes intracellular cholesterol accumulation and foam cell formation, thus becoming a risk factor for atherosclerosis (38).

Karlsson et al (30) compared the macrogenomes of gut microbiota in patients with stroke and healthy individuals and found significant differences. The macrogenome of patients with stroke had an abundance of peptidoglycan biosynthesis genes, which is the main component of the cell wall of Gram-positive bacteria, accounting for ~40-90% of the cell wall weight and supporting the structure of the cell wall (39). Peptidoglycan has pro-inflammatory properties and has been demonstrated to be an immune enhancer of the human immune system, stimulating the release of immunomodulatory substances such as TNF-α, IL-1 and IL-6 from mononuclear phagocytes and endothelial cells (40,41). By contrast, in healthy controls the gene encoding phytoene synthase is enriched, which is required for the synthesis of carotenoids (42), acting as antioxidants (43) and having anti-angina and anti-stroke effects (44), confirming that changes in human gut microbiota are associated with atherosclerosis and stroke.

In addition to correlation analyses, numerous studies are also investigating potential mechanisms that establish a connection between atherosclerosis and gut microbiota. An animal study showed that the ability of gut microbiota to reduce cholesterol levels (45). The mechanism involves: i) Co-precipitation; bacteria produce bile salt hydrolases, which break down conjugated bile salts into free bile salts. At a pH of 5.5, this process leads to the co-precipitation of cholesterol and bile acids, thereby reducing the ability of cholesterol entering the bloodstream (46); ii) Cholesterol uptake by bacteria; bacterial cells uptake cholesterol, representing the primary ways of cholesterol removal by these microorganisms. While the capacity for cholesterol uptake varies among bacterial strains, this process contributes to reducing serum cholesterol concentrations (47). These mechanisms associated with gut microbiota and atherosclerosis also suggest potential therapeutic targets for the prevention and treatment of atherosclerosis.

Hypertension

Hypertension is a progressive cardiovascular syndrome with both genetic and environmental causes, leading to changes in cardiac and vascular function and structure (48). Except for genetic causes and environmental factors, gut microbiota also contributes to the progression of hypertension (49). The first evidence suggesting the role of the gut microbiota in development of hypertension was the effect of antibiotic treatment on blood pressure observed in rats (50). In 2015, Yang et al (51) reported that dysbiosis of gut flora may be associated with hypertension with the finding of increasing of fecal microbiota variability in both rat model and patient. It has been reported that the microbial profiles of the pre-hypertensive and hypertensive populations were similar, with significant overgrowth of Prevotella and Klebsiella in both groups. These findings were established through experiments involving the transplantation of fecal microbiota from patients with hypertension into germ-free mice (52). The abundance of butyrate-producing Odoribacter spp. and butyrate production were inversely correlated with blood pressure levels in women at higher risk of developing pregnancy-induced hypertension and preeclampsia (53,54).

Antibiotics for hypertension are a new discovery in the cardiovascular field and are not yet well supported by clinical studies (55). An effective animal study conducted on rats demonstrated the potential use of broad-spectrum antibiotics, including vancomycin, rifampicin and levofloxacin, in aiding the treatment of resistant hypertension, suggesting indirect evidence that antibiotics can improve gut microbiota and initiate potential blood pressure regulatory mechanisms to decreasing blood pressure (56). However, the mechanism of antibiotic against hypotension has yet to be investigated.

The mechanisms by which gut microbiota affects hypertension are complex. Studies found that short chain fatty acids (SCFAs) produced by the gut microbiota regulate blood pressure through various receptors and pathways (57,58). Additional animal research conducted on rats indicated that the inhibition of intestinal sodium transporters might offer potential benefits in ameliorating cardiorenal damage associated with essential hypertension (59). Angiotensin-converting enzyme 2 (ACE2) plays a significant role in the pathophysiology of hypertension (60). It has been demonstrated that ACE2 serves as a regulator of gut microbiota homeostasis, inflammation and genetic susceptibility to colitis (61,62).

Heart failure

A growing number of studies support the role of the gut in the pathogenesis of heart failure (63,64). Under normal circumstances, the gut microbiota maintains a balanced state and performs a crucial role in supporting the integrity and functionality of the intestinal barrier (65). However, in patients with heart failure, the intestinal tract function is altered and dysbiosis of the gut microbiota occurs due to factors such as congestion of the visceral circulation and immune deficiency of the host (66). It has been shown that patients with chronic heart failure may have excessive growth of pathogenic gut microbiota and increased intestinal permeability, which is related to the severity of heart failure (67).

In addition to the clear link between TMAO and risk of atherosclerotic cardiovascular disease, TMAO levels have recently been associated with the development and poor prognosis of patients with heart failure (35). Previous studies have shown that circulating TMAO levels are higher in patients with heart failure compared with age- and sex-matched subjects without heart failure (68,69). In addition, the study observed significantly strong adverse prognostic values associated with elevated plasma TMAO levels in a group of stable patients with heart failure, which were incremental to traditional risk factors, cardiac and renal indices and systemic inflammatory markers (68). However, the mechanisms explaining the elevated TMAO levels in patients with heart failure remain to be elucidated.

Cardiac infarction

Compared to normal subjects, patients with myocardial infarction have high levels of phosphatidylcholine metabolites, which are plasma choline, TMAO and betaine, and these three substances are significantly associated with the development of cardiovascular diseases (70). In both patients and mice, their metabolism, which involves the dietary phosphatidylcholine pathway, relies on the active participation of gut microbiota (71). In addition, an association between gut microbiota and myocardial infarct severity has been reported in rats (71-73). The use of broad-spectrum antibiotics has been shown to affect the levels of analytes produced during leptin and aromatic amino acid catabolism, which are associated with a reduction in myocardial infarct size. In addition, in rodent model studies, delivery of Lactobacillus plantarum was correlated with a significant infarct size reduction following myocardial infarction relief and improvement in left ventricular function. Another study showed that administration of Lactobacillus rhamnosus GR-1 attenuated left ventricular hypertrophy and heart failure after myocardial infarction in rats (74). These observations not only demonstrate the relationship between gut microbiota and myocardial infarction, but also suggest that altering the composition of the flora can be a potential therapeutic strategy.

4. Effects of gut microbiota on metabolites related to cardiovascular diseases

Cholesterol

The gut microbiota can influence the regulation of cholesterol metabolism in the liver (75,76) and plays a role in altering bile acids, which can affect systemic cholesterol levels (77). Bile acids, catalyzed by the rate-limiting enzyme cholesterol 7-α-hydroxylase (78), are the main metabolites of cholesterol in the liver and contribute to the absorption of fats and lipophilic vitamins (79), as well as to the regulation of lipid, glucose and energy metabolism (80,81). Primary bile acids are conjugated with the amino acids taurine or glycine to form bile salts, which are secreted into the bile and stored in the gallbladder until they are released into the small intestine, where they emulsify fats and form micelles that are absorbed by enterocytes (79). In the intestine, primary bile acids such as cholic acid and chenodeoxycholic acid (CDCA) are deconjugated by the gut microbiota and bile salt hydrolase to form secondary bile acids including deoxycholic acid, lithodeoxycholic acid and ursodeoxycholic acid (82). The ratio of primary to secondary bile acids may be associated with the development of hypercholesterolemia and cardiovascular diseases. For example, one study (83) found that patients with heart failure had reduced plasma primary bile acids and a higher ratio of secondary to primary bile acids. Bile acids may also play a role in cardiovascular function by modulating channel conductance and calcium dynamics in sinoatrial and ventricular cardiomyocytes and by regulating vascular tone to reduce heart rate (83). Furthermore, it has been proposed that an unbalanced and unhealthy gut microbiota, which regulates the bile acid ratio, may lead to a decrease in secondary bile acids, which may increase primary bile acids, such as CDCA, activating farnesoid X receptor and downregulating bile acid secretion, thereby increasing cholesterol and cardiovascular diseases development. Therefore, the gut microbiota and the underlying mechanisms involved require further study.

It has long been known that certain gut microbiota has the ability to convert absorbable cholesterol into coprostanols, which are reduced non-absorbable sterols that are excreted in the feces (84-86). Coprostanol production in humans begins at 6 months after birth (87) and is sex-dependent, with young women possessing a higher production compared with young men (88). Furthermore, the conversion of microbial cholesterol to coprostanol in human populations is currently considered to be bimodal, with high converters converting almost 100% cholesterol and low converters converting less than one-third of the fecal neutral sterol content (89,90). To date, isolated cholesterol-reducing strains have been limited to the genera Eubacterium (E. coprostanoligenes) and Bacteroides (Bacteroides sp. strain D8) (91,92), but a number of remain to be discovered. In rabbit models with diet-induced hypercholesterolemia, oral administration of coplanar alkanoic bacteria caused a significant decrease in plasma cholesterol levels that lasted for >34 days after the last bacterial feeding (93). Clinical studies have extensively investigated cholesterol metabolism in the intestine (87,89,91,94,95). These studies have indicated an inverse relationship between human serum cholesterol levels and the ratio of human fecal coprostanol to cholesterol (96,97). However, these studies used very small sample sizes, limited variation in sample populations and lack of diverse population backgrounds, which reduces the generalizability of the studies. Attempts to isolate the specific microbial strains responsible for copolymer/cholesterol conversion in these studies were also unsuccessful, making it difficult to continue the mechanistic studies that followed. In addition, the genes or enzymes involved in the conversion of cholesterol to coplanar alcohols in the intestine remain unknown (98).

Short chain fatty acids (SCFAs)

SCFAs are metabolites derived from microbial activity during the fermentation of complex carbohydrates (99,100). These compounds exert influence on microbiota composition and gut motility, thereby affecting various host processes (101). The most abundant SCFAs are acetate, propionate and butyrate (100). Phylum Bacteroidetes members produce acetate and butyrate, while Phylum Siliques produces butyrate. SCFAs are also positively correlated with Alistipes putredinis, Bacteroides spp, Roseburia, Eubacterium rectale and Faecal prausnitzii (102). In addition, SCFAs play a pivotal role in preserving intestinal barrier integrity by modulating the expression of tight junction proteins (103). SCFAs can also reduce serum lipid levels by blocking cholesterol synthesis and transferring it to the liver. Therefore, they are considered as a protective factor in the progression of cardiovascular diseases. The presence of SCFA-producing bacteria is diminished due to the activation of G protein-coupled receptor 41(104) observed in certain cases of cardiovascular diseases (30,105). This reduction is also apparent in the dysbiosis of the gut microbiota observed in hypertensive patients (106). Therefore, their role in vivo and their targets need to be further examined.

TMAO

TMAO is identified as a risk factor contributing to the development of cardiovascular diseases (71,75). This compound originates from various sources, including red meat, eggs, fish and vegetables (107). Compounds such as choline, betaine, phosphatidylcholine, lecithin and L-carnitine (71,108,109) found in the diet are implicated in the production of TMAO. A clinical study revealed that elevated levels of TMAO are linked to an increased risk of death, nonfatal myocardial infarction, or stroke (110). An in vitro study revealed the ability of the gut microbiota to produce choline through the enzyme phospholipase D (111). TMA molecules, produced by microbiota, can enter the circulation of the host and subsequently reach hepatocytes. Within these hepatocytes, they undergo metabolism to form TMAO through the action of flavin-containing monooxygenases (FMO) encoded by the FMO gene. These enzymatic reactions occur predominantly in the liver, kidneys and other tissues (109). Elevated production of TMAO has been shown to affect blood lipids (112) and is associated with an increasing risk of cardiovascular diseases (71,108,113). In situations involving heightened intestinal permeability, TMAO exhibits associations with C-reactive protein and endothelial dysfunction. In addition, increased serum levels of LPS endotoxin (114) are also linked to TMAO. Moreover, TMAO has been observed to induce calcium release and increase platelet hyperreactivity (115), potentially influencing the progression of cardiovascular diseases.

The gut microbiota markedly influences the production of TMAO. Healthy individuals have a large number of TMAO-producing microorganisms, with a 2:1 ratio of Firmicutes to Bacteroidetes (116). TMAO production was found in 102 genomes covering 36 species, including Firmicutes, Aspergillus and Actinobacteria (107). It has been shown that Firmicutes and Proteobacter are associated with the production of TMA (114). One study found that eight species from Stachybotrys and Proteobacter consumed >60% of the choline used for TMA production (117). Acinetobacter, Sporotrichum, Prevotella (107) and Micrococcus (118) are intestinal species capable of producing higher TMAO than other species and they are associated with atherosclerotic cardiovascular diseases. Therefore, metabolites including choline, TMAO and betaine can help predict the development of cardiovascular diseases. As a potential therapeutic pathway, probiotic or pharmacological interventions can be used to inhibit or block specific metabolic pathways and reduce TMAO-producing microbes (119).

5. Potential therapeutics

Probiotics

The role of lactobacilli with probiotic effects such as immunomodulation, absorption promotion and disease prevention in the intervention of hyperlipidemia and hypertension is of interest. Previous animal studies have identified a variety of lactobacilli with antihypertensive effects, the main one being Lactobacillus swiss (120,121). Experiments in mice suggest that Lactobacillus rhamnosus can lower cholesterol and improve non-alcoholic fatty liver disease (122).

There have been reviews on the role of probiotics in regulating blood lipids (123,124). Entry of probiotic flora (mainly Bifidobacterium, Lactobacillus and Propionibacterium) into the intestine from dietary sources leads to an increase in SCFAs-producing flora and a decrease in the number of protein-producing flora, thus promoting an increase in the synthesis of SCFAs and butyrate and a decrease in protein synthesis, leading to changes in carbohydrate metabolism. Probiotics have been proven to regulate blood lipids, but their mechanism has not been determined yet. From a certain perspective, it is suggested that Lactobacillus and other probiotics facilitate the metabolism of cholesterol during their growth, consequently leading to a reduction in cholesterol levels (125). Additionally, the hypolipidemic effect attributed to lactic acid bacteria has been linked to the modulation of 3-hydroxy-3-methylglutaryl CoA reductase expression, which is the rate-limiting enzyme in cholesterol synthesis.

Probiotics and their fermented products (e.g., yogurt) have been shown in a number of studies to have significant antihypertensive effects. Khalesi et al (126) showed that blood pressure (both systolic and diastolic) was significantly lower in individuals whose intake was ≥1 billion probiotic colonies per day. This indirect evidence indicated that the gut microbiota plays a crucial role in maintaining stability in blood pressure. It suggests the mechanism underlying the hypotensive effect of probiotic products based on two primary aspects: i) The hypotensive effect of probiotic products is attributed to the hydrolysis of extracellular proteases and peptidases (such as carboxypeptidase and aminopeptidase). This process leads to the liberation of peptides from food proteins, such as milk protein, which exhibit hypotensive activity. These released peptides, such as ACE inhibitory peptide and opioid active peptide, contribute to the observed antihypertensive effects; ii) Certain probiotic organisms, particularly Lactobacillus, can transit to the intestinal tract as live bacteria. These organisms facilitate the intestinal absorption of specific minerals known to regulate blood pressure. Through this action, they demonstrate antihypertensive effects. Therefore, the application of probiotics to modify the composition of the gut microbiota is a potential therapeutic target for cardiovascular diseases.

Dietary intervention

Dietary interventions to correct intestinal dysbiosis could be a therapeutic strategy for cardiovascular disease. Indeed, a heart-healthy diet, abundant in vegetables and high in fiber, is regarded as advantageous for the cardiovascular system (127). Certainly, lifestyle can markedly impact bacterial transmission, particularly environmental sanitation practices and drinking water treatment methods. These aspects play a crucial role in influencing alterations observed in the gut microbiota. The Mediterranean diet is a combination of fish, beans, vegetables, fruits, nuts, olive oil and a moderate amount of red wine (128,129). This diet has become particularly popular in recent years and is considered to prevent cardiovascular disease and reduce cardiovascular mortality in both men and women (130,131). For most individuals, a Mediterranean-style diet is an effective and viable way to prevent heart disease and other health problems.

Antibiotics

The microbiota can be regulated with antibiotics and used to restore the microbiome and prevent cardiovascular disease. A study showed that oral vancomycin reduced infarct size and improved post-infarct cardiac function in rats (72). Furthermore, in an experimental mouse model, antibiotics reduced bacterial translocation, inflammation and myocardial injury (132). In one study, a 69-year-old patient with a long history (44 years) of hypertension and resistant hypertension showed reduced blood pressure after combination antibiotic therapy (133). Moreover, a two-month dietary intervention involving a broad-spectrum antibiotic cocktail successfully reversed vascular dysfunction induced by a Western diet in mice. This reversal represents a crucial preclinical step in preventing the progression of cardiovascular disease (134). In addition, a study on rats showed that vancomycin administration was associated with a reduction in myocardial infarct size and improved recovery of mechanical function after ischemia, while effectively reducing the total number and group of intestinal microbes (72).

Collectively, various clinical and animal studies have strongly indicated the involvement of gut microbiota composition in acute myocardial infarction. However, the specific direction of microbiota alterations and the potential metabolic or inflammatory pathways involved remain poorly understood.

6. Conclusions

The relevance of gut microbiota to disease is in its infancy and, similarly, the relevance to cardiovascular disease is still at an early stage and a number of key questions need to be explored. Increasing evidence indicates an association between the gut microbiome and the incidence of cardiovascular disease. Studies suggest that the microbiota interacts with the host through multiple pathways. Abnormal gut microbiota composition or microbial metabolites may be responsible for altered cardiovascular disease risk and its associated pathological changes (Fig. 1). Thus, the potential role of the gut microbiota could be utilized in the future to develop novel therapeutic strategies for the prevention and treatment of cardiovascular disease.

As a potential therapeutic strategy, the feasibility of gut microbiota transplantation has been demonstrated in short-term studies, but long-term trials are needed to evaluate the safety and efficacy. Modulation of gut microbiota as a potential therapeutic target for cardiovascular disease needs to be further evaluated and the therapeutic indications and specific strategies need to be further demonstrated.

Acknowledgements

Not applicable.

Funding

Funding: The present study was funded by China National Natural Science Funding (grant no. 81902937), Hubei University of Science and Technology ENT special project (grant no. 2020WG06) for SYL, Hubei University of Science and Technology ENT special project (No. 2) for NZF and (grant no. 2020XZ30) for SDW, Hubei province Key R and D plan (grant no. 2022BCE011) for NZF.

Availability of data and materials

Not applicable.

Authors' contributions

ZFN, LL, KQC and GLW conceived and supervised the present study. SWX, YC, YL and DWS prepared the first draft of the manuscript. XPY, ZWu, YLS, ZhiW, JL and ZFN reviewed the manuscript for important intellectual content. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Barreto HC and Gordo I: Intrahost evolution of the gut microbiota. Nat Rev Microbiol. 21:590–603. 2023.PubMed/NCBI View Article : Google Scholar

2 

Guarner F and Malagelada JR: Gut flora in health and disease. Lancet. 361:512–519. 2003.PubMed/NCBI View Article : Google Scholar

3 

Shayya NW, Foote MS, Langfeld LQ, Du K, Bandick R, Mousani S, Bereswill S and Heimesaat MM: Human microbiota associated IL-10-/- mice: A valuable enterocolitis model to dissect the interactions of Campylobacter jejuni with host immunity and gut microbiota. Eur J Microbiol Immunol (Bp). 12:107–122. 2023.PubMed/NCBI View Article : Google Scholar

4 

Couvillion SP, Danczak RE, Cao X, Yang Q, Keerthising TP, McClure RS, Bitounis D, Bunret MC, Fansler SJ, Richardson RE, et al: Graphene oxide exposure alters gut microbial community composition and metabolism in an in vitro human model. NanoImpact. 30(100463)2023.PubMed/NCBI View Article : Google Scholar

5 

Mishra SP, Wang B, Jain S, Ding J, Rejeski J, Furdui CM, Kitzman DW, Taraohder S, Brechot C, Kumar A and Yadav H: A mechanism by which gut microbiota elevates permeability and inflammation in obese/diabetic mice and human gut. Gut. 72:1848–1865. 2023.PubMed/NCBI View Article : Google Scholar

6 

Vallino L, Garavaglia B, Visciglia A, Amoruso A, Pane M, Ferraresi A and Lsidoro C: Cell-free Lactiplantibacillus plantarum OC01 supernatant suppresses IL-6-induced proliferation and invasion of human colorectal cancer cells: Effect on β-Catenin degradation and induction of autophagy. J Tradit Complement Med. 13:193–206. 2023.PubMed/NCBI View Article : Google Scholar

7 

Hooper LV and Gordon JI: Commensal host-bacterial relationships in the gut. Science. 292:1115–1118. 2001.PubMed/NCBI View Article : Google Scholar

8 

Dethlefsen L, McFall-Ngai M and Relman DA: An ecological and evolutionary perspective on human-microbe mutualism and disease. Nature. 449:811–818. 2007.PubMed/NCBI View Article : Google Scholar

9 

Maciel-Fiuza MF, Muller GC, Campos DMS, Costa PSS, Peruzzo J, Bonamigo RR, Veit T and Vianna FSL: Role of gut microbiota in infectious and inflammatory diseases. Front Microbiol. 14(1098386)2023.PubMed/NCBI View Article : Google Scholar

10 

Zhao J, Hu Y, Qian C, Hussain M, Liu S, Zhang A, He R and Sun P: The interaction between mushroom polysaccharides and gut microbiota and their effect on human health: A review. Biology (Basel). 12(122)2023.PubMed/NCBI View Article : Google Scholar

11 

Belvoncikova P, Splichalova P, Videnska P and Gardlik R: The human mycobiome: Colonization, composition and the role in health and disease. J Fungi. 8(1046)2022.PubMed/NCBI View Article : Google Scholar

12 

Ley RE, Lozupone CA, Hamady M, Knight R and Gordon JI: Worlds within worlds: Evolution of the vertebrate gut microbiota. Nat Rev Microbiol. 6:776–788. 2008.PubMed/NCBI View Article : Google Scholar

13 

Faust K, Sathirapongsasuti JF, Izard J, Segata N, Gevers D, Raes J and Huttenhower C: Microbial co-occurrence relationships in the human microbiome. PLoS Comput Biol. 8(e1002606)2012.PubMed/NCBI View Article : Google Scholar

14 

Kwa WT, Sundarajoo S, Toh KY and Lee J: Application of emerging technologies for gut microbiome research. Singapore Med J. 64:45–52. 2023.PubMed/NCBI View Article : Google Scholar

15 

Sauceda C, Bayne C, Sudqi K, Gonzalez A, Dulai PS, Knight R, Gonzalez DJ and Gonzalez C: Stool multi-omics for the study of host-microbe interactions in inflammatory bowel disease. Gut Microbes. 14(2154092)2022.PubMed/NCBI View Article : Google Scholar

16 

Minj J, Riordan J, Teets C, Fernholz-Hartman H, Tanggono A, Lee Y, Chauvin T, Carbonero F and Solverson P: Diet-induced rodent obesity is prevented and the fecal microbiome is improved with elderberry (Sambucus nigra ssp. canadensis) juice powder. J Agric Food Chem. 72:12555–12565. 2024.PubMed/NCBI View Article : Google Scholar

17 

Rochoń J, Kalinowski P, Szymanek-Majchrzak K and Grąt M: Role of gut-liver axis and glucagon-like peptide-1 receptor agonists in the treatment of metabolic dysfunction-associated fatty liver disease. World J Gastroenterol. 30:2964–2980. 2024.PubMed/NCBI View Article : Google Scholar

18 

Zhang Y, Zheng T, Ma D, Shi P, Zhang H, Li J and Sun Z: Probiotics Bifidobacterium lactis M8 and Lactobacillus rhamnosus M9 prevent high blood pressure via modulating the gut microbiota composition and host metabolic products. mSystems. 8(e0033123)2023.PubMed/NCBI View Article : Google Scholar

19 

Yan D, Si W, Zhou X, Yang M, Chen Y, Chang Y, Lu Y, Liu J, Wang K, Yan M, et al: Eucommia ulmoides bark extract reduces blood pressure and inflammation by regulating the gut microbiota and enriching the Parabacteroides strain in high-salt diet and N(omega)-nitro-L-arginine methyl ester induced mice. Front Microbiol. 18(967649)2022.PubMed/NCBI View Article : Google Scholar

20 

Mao Y, Kong C, Zang T, You L, Wang LS, Shen L and Ge JB: Impact of the gut microbiome on atherosclerosis. mLife. 3:167–175. 2024.PubMed/NCBI View Article : Google Scholar

21 

Glorieux G, Nigam SK, Vanholder R and Verbeke F: Role of the microbiome in gut-heart-kidney cross talk. Circ Res. 132:1064–1083. 2023.PubMed/NCBI View Article : Google Scholar

22 

Hijová E: Benefits of biotics for cardiovascular diseases. Int J Mol Sci. 24(6292)2023.PubMed/NCBI View Article : Google Scholar

23 

Liao L, Huang J, Zheng J, Ma X, Huang L and Xu W: Gut microbiota in Chinese and Japanese patients with cardiovascular diseases: A systematic review and meta-analysis. Ann Saudi Med. 43:105–114. 2023.PubMed/NCBI View Article : Google Scholar

24 

Zhang H, Li H, Pan B, Zhang S, Su X, Sun W, Zhang T, Zhang Z, Lv S and Cui H: Integrated 16S rRNA Sequencing and untargeted metabolomics analysis to reveal the protective mechanisms of polygonatum sibiricum polysaccharide on type 2 diabetes mellitus model rats. Curr Drug Metab. 10(1389200224666230406114012)2023.PubMed/NCBI View Article : Google Scholar

25 

Song Z, Song R, Liu Y, Wu Z and Zhang X: Effects of ultra-processed foods on the microbiota-gut-brain axis: The bread-and-butter issue. Food Res Int. 167(112730)2023.PubMed/NCBI View Article : Google Scholar

26 

Sajdel-Sulkowska EM: Neuropsychiatric ramifications of COVID-19: Short-chain fatty acid deficiency and disturbance of microbiota-gut-brain axis signaling. Biomed Res Int. 2021(7880448)2021.PubMed/NCBI View Article : Google Scholar

27 

Shantsila E, Kamphuisen PW and Lip GY: Circulating microparticles in cardiovascular disease: Implications for atherogenesis and atherothrombosis. J Thromb Haemost. 8:2358–2368. 2010.PubMed/NCBI View Article : Google Scholar

28 

Philippova M, Suter Y, Toggweiler S, Schoenenberger AW, Joshi MB, Kyriakakis E, Erne P and Resink TJ: T-cadherin is present on endothelial microparticles and is elevated in plasma in early atherosclerosis. Eur Heart J. 32:760–771. 2011.PubMed/NCBI View Article : Google Scholar

29 

Markin AM, Markina YV, Bogatyreva AI, Tolstik TV, Chakal DA, Breshenkov DG and Charchyan ER: The role of cytokines in cholesterol accumulation in cells and atherosclerosis progression. Int J Mol Sci. 24(6426)2023.PubMed/NCBI View Article : Google Scholar

30 

Karlsson FH, Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, Bäckhed F and Nielsen J: Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun. 3(1245)2012.PubMed/NCBI View Article : Google Scholar

31 

Khalili L, Centner AM and Salazar G: Effects of berries, phytochemicals, and probiotics on atherosclerosis through gut microbiota modification: A meta-analysis of animal studies. Int J Mol Sci. 24(3084)2023.PubMed/NCBI View Article : Google Scholar

32 

Kumar T, Dutta RR, Velagala VR, Ghosh B and Mudey A: Analyzing the complicated connection between intestinal microbiota and cardiovascular diseases. Cureus. 14(e28165)2022.PubMed/NCBI View Article : Google Scholar

33 

Shi X, Wu H, Liu Y, Huang H, Liu L, Yang Y, Jiang T, Zhou M and Dai M: Inhibiting vascular smooth muscle cell proliferation mediated by osteopontin via regulating gut microbial lipopolysaccharide: A novel mechanism for paeonol in atherosclerosis treatment. Front Pharmacol. 13(936677)2022.PubMed/NCBI View Article : Google Scholar

34 

Liu S, He F, Zheng T, Wan S, Chen S, Yang F, Xu X and Pei X: Ligustrum robustum alleviates atherosclerosis by decreasing serum TMAO, modulating gut microbiota, and decreasing bile acid and cholesterol absorption in mice. Mol Nutr Food Res. 65(e2100014)2021.PubMed/NCBI View Article : Google Scholar

35 

Zhen J, Zhou Z, He M, Han HX, Lv EH, Wen PB, Liu X, Wang YT, Cai XC, Tian JQ, et al: The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne). 14(1085041)2023.PubMed/NCBI View Article : Google Scholar

36 

Canyelles M, Tondo M, Cedó L, Farràs M, Escolà-Gil JC and Blanco-Vaca F: Trimethylamine N-Oxide: A link among diet, gut microbiota, gene regulation of liver and intestine cholesterol homeostasis and HDL function. Int J Mol Sci. 19(3228)2018.PubMed/NCBI View Article : Google Scholar

37 

Shi C, Pei M, Wang Y, Chen Q, Cao P, Zhang L, Guo J, Deng W, Wang L, Li X and Gong Z: Changes of flavin-containing monooxygenases and trimethylamine-N-oxide may be involved in the promotion of non-alcoholic fatty liver disease by intestinal microbiota metabolite trimethylamine. Biochem Biophys Res Commun. 594:1–7. 2022.PubMed/NCBI View Article : Google Scholar

38 

Trenteseaux C, Gaston AT, Aguesse A, Poupeau G, Coppet P, Andriantsitohaina R, Laschet J, Amarger V, Krempf M, Nobecourt-Dupuy E and Ouguerram K: Perinatal hypercholesterolemia exacerbates atherosclerosis lesions in offspring by altering metabolism of trimethylamine-n-oxide and bile acids. Arterioscler Thromb Vasc Biol. 37:2053–2063. 2017.PubMed/NCBI View Article : Google Scholar

39 

Schleifer KH and Kandler O: Peptidoglycan types of bacterial cell walls and their taxonomic implications. Bacteriol Rev. 36:407–477. 1972.PubMed/NCBI View Article : Google Scholar

40 

Popescu NI, Girton A, Burgett T, Lovelady K and Coggeshall KM: Monocyte procoagulant responses to anthrax peptidoglycan are reinforced by proinflammatory cytokine signaling. Blood Adv. 3:2436–2447. 2019.PubMed/NCBI View Article : Google Scholar

41 

Xie Y, Li Y, Cai X, Wang X and Li J: Interleukin-37 suppresses ICAM-1 expression in parallel with NF-κB down-regulation following TLR2 activation of human coronary artery endothelial cells. Int Immunopharmacol. 38:26–30. 2016.PubMed/NCBI View Article : Google Scholar

42 

Dong C, Zhang M, Song S, Wei F, Qin L, Fan P, Shi Y, Wang X and Wang R: A small subunit of Geranylgeranyl Diphosphate synthase functions as an active regulator of carotenoid synthesis in nicotiana tabacum. Int J Mol Sci. 24(992)2023.PubMed/NCBI View Article : Google Scholar

43 

Jo HE, Son SY and Lee CH: Comparison of metabolome and functional properties of three Korean cucumber cultivars. Front Plant Sci. 13(882120)2022.PubMed/NCBI View Article : Google Scholar

44 

Bocchini M, D'Amato R, Ciancaleoni S, Fontanella MC, Palmerini CA, Beone GM, Onofri A, Negri V, Marconi G, Albertini E and Businelli D: Soil Selenium (Se) biofortification changes the physiological, biochemical and epigenetic responses to water stress in Zea mays L. by inducing a higher drought tolerance. Front Plant Sci. 9(389)2018.PubMed/NCBI View Article : Google Scholar

45 

Wang Y, Zheng Y, Liu Y, Shan G, Zhang B, Cai Q, Lou J and Qu Y: The lipid-lowering effects of fenugreek gum, hawthorn pectin, and burdock inulin. Front Nutr. 10(1149094)2023.PubMed/NCBI View Article : Google Scholar

46 

Tahri K, Grill JP and Schneider F: Bifidobacteria strain behavior toward cholesterol: Coprecipitation with bile salts and assimilation. Curr Microbiol. 33:187–193. 1996.PubMed/NCBI View Article : Google Scholar

47 

Bordoni A, Amaretti A, Leonardi A, Boschetti E, Danesi F, Matteuzzi D, Roncaglia L, Raimondi S and Rossi M: Cholesterol-lowering probiotics: In vitro selection and in vivo testing of bifidobacteria. Appl Microbiol Biotechnol. 97:8273–8281. 2013.PubMed/NCBI View Article : Google Scholar

48 

Burnier M and Damianaki A: Hypertension as cardiovascular risk factor in chronic kidney disease. Circ Res. 132:1050–1063. 2023.PubMed/NCBI View Article : Google Scholar

49 

Mutengo KH, Masenga SK, Mweemba A, Mutale W and Kirabo A: Gut microbiota dependant trimethylamine N-oxide and hypertension. Front Physiol. 14(1075641)2023.PubMed/NCBI View Article : Google Scholar

50 

Honour JW, Borriello SP, Ganten U and Honour P: Antibiotics attenuate experimental hypertension in rats. J Endocrinol. 105:347–350. 1985.PubMed/NCBI View Article : Google Scholar

51 

Yang Z, Wang Q, Liu Y, Wang L, Ge Z, Li Z, Feng S and Wu C: Gut microbiota and hypertension: Association, mechanisms and treatment. Clin Exp Hypertens. 45(2195135)2023.PubMed/NCBI View Article : Google Scholar

52 

Li J, Zhao F, Wang Y, Chen J, Tao G, Tian G, Wu S, Liu W, Cui Q, Geng B, et al: Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome. 5(14)2017.PubMed/NCBI View Article : Google Scholar

53 

Jin J, Gao L, Zou X, Zhang Y, Zheng Z, Zhang X, Li J, Tian Z, Wang X, Gu J, et al: Gut dysbiosis promotes preeclampsia by regulating macrophages and trophoblasts. Circ Res. 131:492–506. 2022.PubMed/NCBI View Article : Google Scholar

54 

Gomez-Arango LF, Barrett HL, McIntyre HD, Callaway LK, Morrison M and Nitert MD: SPRING Trial Group. Increased systolic and diastolic blood pressure is associated with altered gut microbiota composition and butyrate production in early pregnancy. Hypertension. 68:974–981. 2016.PubMed/NCBI View Article : Google Scholar

55 

Lucas SE, Walton SL, Colafella KM, Mileto SJ, Lyras D and Denton KM: Antihypertensives and antibiotics: Impact on intestinal dysfunction and hypertension. Hypertension. 11:1393–1402. 2023.PubMed/NCBI View Article : Google Scholar

56 

Kyoung J and Yang T: Depletion of the gut microbiota enhances the blood pressure-lowering effect of captopril: Implication of the gut microbiota in resistant hypertension. Hypertens Res. 45:1505–1510. 2022.PubMed/NCBI View Article : Google Scholar

57 

Chen XF, Ren SC, Tang G, Wu C, Chen X and Tang XQ: Short-chain fatty acids in blood pressure, friend or foe. Chin Med J (Engl). 134:2393–2394. 2021.PubMed/NCBI View Article : Google Scholar

58 

Dinakis E, O'Donnell JA and Marques FZ: The gut-immune axis during hypertension and cardiovascular diseases. Acta Physiol (Oxf). 20(e14193)2024.PubMed/NCBI View Article : Google Scholar

59 

Spencer AG, Labonte ED, Rosenbaum DP, Plato CF, Carreras CW, Leadbetter MR, Kozuka K, Kohler J, Koo-McCoy S, He L, et al: Intestinal inhibition of the Na+/H+ exchanger 3 prevents cardiorenal damage in rats and inhibits Na+ uptake in humans. Sci Transl Med. 6(277ra36)2014.PubMed/NCBI View Article : Google Scholar

60 

Ohman KP, Karlberg BE, Nilsson OR and Wettre S: Captopril in primary hypertension. Effects related to the renin-angiotensin-aldosterone and kallikrein-kinin systems. Acta Med Scand Suppl. 646:98–105. 1981.PubMed/NCBI

61 

Andrade JM, de Farias Lelis D, Mafra V and Cota J: The angiotensin converting enzyme 2 (ACE2), gut microbiota, and cardiovascular health. Protein Pept Lett. 24:827–832. 2017.PubMed/NCBI View Article : Google Scholar

62 

Perlot T and Penninger JM: ACE2-from the renin-angiotensin system to gut microbiota and malnutrition. Microbes Infect. 15:866–873. 2013.PubMed/NCBI View Article : Google Scholar

63 

Zhang QL, Chen XH, Zhou SJ, Lei YQ, Huang JS, Chen Q and Cao H: Relationship between disorders of the intestinal microbiota and heart failure in infants with congenital heart disease. Front Cell Infect Microbiol. 13(1152348)2023.PubMed/NCBI View Article : Google Scholar

64 

Desai D, Desai A, Jamil A, Csendes D, Gutlapalli SD, Prakash K, Swarnakari KM, Bai M, Manoharan MP, Raja R and Khan S: Re-defining the gut heart axis: A systematic review of the literature on the role of gut microbial dysbiosis in patients with heart failure. Cureus. 15(e34902)2023.PubMed/NCBI View Article : Google Scholar

65 

Xu X, Hu H, Zeng H, Li B, Yin Q, Jiang Y, Zang L, Zhao C and Qian G: Sinisan ameliorates colonic injury induced by water immersion restraint stress by enhancing intestinal barrier function and the gut microbiota structure. Pharm Biol. 61:598–609. 2023.PubMed/NCBI View Article : Google Scholar

66 

Tousoulis D, Guzik T, Padro T, Duncker DJ, Luca GD, Eringa E, Vavlukis M, Antonopoulos AS, Katsimichas T, Cenko E, et al: Mechanisms, therapeutic implications, and methodological challenges of gut microbiota and cardiovascular diseases: A position paper by the ESC working group on coronary pathophysiology and microcirculation. Cardiovasc Res. 118:3171–3182. 2022.PubMed/NCBI View Article : Google Scholar

67 

Cui X, Su Y, Huang X, Chen J, Ma J, Liao P and He X: Combined analysis of plasma metabolome and intestinal microbiome sequencing to explore jiashen prescription and its potential role in changing intestine-heart axis and effect on chronic heart failure. Front Cardiovasc Med. 10(1147438)2023.PubMed/NCBI View Article : Google Scholar

68 

Zong X, Fan Q, Yang Q, Pan R, Zhuang L, Xi R, Zhang R and Tao R: Trimethyllysine, a trimethylamine N-oxide precursor, predicts the presence, severity, and prognosis of heart failure. Front Cardiovasc Med. 9(907997)2022.PubMed/NCBI View Article : Google Scholar

69 

Li X, Fan Z, Cui J, Li D, Lu J, Cui X, Xie L, Wu Y, Lin Q and Li Y: Trimethylamine N-Oxide in heart failure: A meta-analysis of prognostic value. Front Cardiovasc Med. 16(817396)2022.PubMed/NCBI View Article : Google Scholar

70 

Wang Z, Tang WH, Buffa JA, Fu X, Britt EB, Koeth RA, Levison BS, Fan Y, Wu Y and Hazen SL: Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur Heart J. 35:904–910. 2014.PubMed/NCBI View Article : Google Scholar

71 

Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein EB, Britt EB, Fu X, Chung YM, et al: Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 472:57–63. 2011.PubMed/NCBI View Article : Google Scholar

72 

Lam V, Su J, Koprowski S, Hsu A, Tweddell JS, Rafiee P, Gross GJ, Salzmman NH and Baker JE: Intestinal microbiota determine severity of myocardial infarction in rats. FASEB J. 26:1727–1735. 2012.PubMed/NCBI View Article : Google Scholar

73 

Lam V, Su J, Hsu A, Gross GJ, Salzman NH and Baker JE: Intestinal microbial metabolites are linked to severity of myocardial infarction in rats. PLoS One. 11(e0160840)2016.PubMed/NCBI View Article : Google Scholar

74 

Gan XT, Ettinger G, Huang CX, Burton JP, Haist JV, Rajapurohitam V, Sidaway JE, Martin G, Gloor GB, Swann JR, et al: Probiotic administration attenuates myocardial hypertrophy and heart failure after myocardial infarction in the rat. Circ Heart Fail. 7:491–499. 2014.PubMed/NCBI View Article : Google Scholar

75 

Tang WH and Hazen SL: The contributory role of gut microbiota in cardiovascular disease. J Clin Invest. 124:4204–4211. 2014.PubMed/NCBI View Article : Google Scholar

76 

Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF and Gordon JI: The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci USA. 101:15718–15723. 2004.PubMed/NCBI View Article : Google Scholar

77 

Jones BV, Begley M, Hill C, Gahan CG and Marchesi JR: Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome. Proc Natl Acad Sci USA. 105:13580–13585. 2008.PubMed/NCBI View Article : Google Scholar

78 

Chiang JY: Bile acids: Regulation of synthesis. J Lipid Res. 50:1955–1966. 2009.PubMed/NCBI View Article : Google Scholar

79 

Ferrell JM, Boehme S, Li F and Chiang JY: Cholesterol 7α-hydroxylase-deficient mice are protected from high-fat/high-cholesterol diet-induced metabolic disorders. J Lipid Res. 57:1144–1154. 2016.PubMed/NCBI View Article : Google Scholar

80 

Li T, Francl JM, Boehme S, Ochoa A, Zhang Y, Klaassen CD, Erickson SK and Chiang JY: Glucose and insulin induction of bile acid synthesis: Mechanisms and implication in diabetes and obesity. J Biol Chem. 287:1861–1873. 2012.PubMed/NCBI View Article : Google Scholar

81 

Broeders EP, Nascimento EB, Havekes B, Brans B, Roumans KH, Tailleux A, Schaart G, Kouach M, Charton J, Deprez B, et al: The bile acid chenodeoxycholic acid increases human brown adipose tissue activity. Cell Metab. 22:418–426. 2015.PubMed/NCBI View Article : Google Scholar

82 

Kumar PS, Mason MR, Brooker MR and O'Brien K: Pyrosequencing reveals unique microbial signatures associated with healthy and failing dental implants. J Clin Periodontol. 39:425–433. 2012.PubMed/NCBI View Article : Google Scholar

83 

Mayerhofer CCK, Ueland T, Broch K, Vincent RP, Cross GF, Dahl CP, Aukrust P, Gullestad L, Hov JR and Trøseid M: Increased Secondary/Primary bile acid ratio in chronic heart failure. J Card Fail. 23:666–671. 2017.PubMed/NCBI View Article : Google Scholar

84 

Dam H: The formation of coprosterol in the intestine: The action of intestinal bacteria on cholesterol. Biochem J. 28:820–825. 1934.PubMed/NCBI View Article : Google Scholar

85 

Lichtenstein AH: Intestinal cholesterol metabolism. Ann Med. 22:49–52. 1990.PubMed/NCBI View Article : Google Scholar

86 

Illman RJ, Storer GB and Topping DL: White wheat flour lowers plasma cholesterol and increases cecal steroids relative to whole wheat flour, wheat bran and wheat pollard in rats. J Nutr. 123:1094–1100. 1993.PubMed/NCBI View Article : Google Scholar

87 

Midtvedt AC and Midtvedt T: Conversion of cholesterol to coprostanol by the intestinal microflora during the first two years of human life. J Pediatr Gastroenterol Nutr. 17:161–168. 1993.PubMed/NCBI View Article : Google Scholar

88 

Benno P, Midtvedt K, Alam M, Collinder E, Norin E and Midtvedt T: Examination of intestinal conversion of cholesterol to coprostanol in 633 healthy subjects reveals an age- and sex-dependent pattern. Microbial Ecology in Health and Disease. 17:200–204. 2005.

89 

Midtvedt T, Lingaas E, Carlstedt-Duke B, Höverstad T, Midtvedt AC, Saxerholt H, Steinbakk M and Norin KE: Intestinal microbial conversion of cholesterol to coprostanol in man. Influence of antibiotics. APMIS. 98:839–844. 1990.PubMed/NCBI View Article : Google Scholar

90 

Veiga P, Juste C, Lepercq P, Saunier K, Béguet F and Gérard P: Correlation between faecal microbial community structure and cholesterol-to-coprostanol conversion in the human gut. FEMS Microbiol Lett. 242:81–86. 2005.PubMed/NCBI View Article : Google Scholar

91 

Gerard P, Lepercq P, Leclerc M, Gavini F, Raibaud P and Juste C: Bacteroides sp. strain D8, the first cholesterol-reducing bacterium isolated from human feces. Appl Environ Microbiol. 73:5742–5749. 2007.PubMed/NCBI View Article : Google Scholar

92 

Ren D, Li L, Schwabacher AW, Young JW and Beitz DC: Mechanism of cholesterol reduction to coprostanol by Eubacterium coprostanoligenes ATCC 51222. Steroids. 61:33–40. 1996.PubMed/NCBI View Article : Google Scholar

93 

Li L, Baumann CA, Meling DD, Sell JL and Beitz DC: Effect of orally administered Eubacterium Coprostanoligenes ATCC 51222 on plasma cholesterol concentration in laying hens. Poult Sci. 75:743–745. 1996.PubMed/NCBI View Article : Google Scholar

94 

Rosenfeld RS, Fukushima DK, Hellman L and Gallagher TF: The transformation of cholesterol to coprostanol. J Biol Chem. 211:301–311. 1954.PubMed/NCBI

95 

Antharam VC, McEwen DC, Garrett TJ, Dossey AT, Li EC, Kozlov AN, Mesbah Z and Wang GP: An integrated metabolomic and microbiome analysis identified specific gut microbiota associated with fecal cholesterol and coprostanol in clostridium difficile infection. PLoS One. 11(e0148824)2016.PubMed/NCBI View Article : Google Scholar

96 

Lye HS, Rusul G and Liong MT: Removal of cholesterol by lactobacilli via incorporation and conversion to coprostanol. J Dairy Sci. 93:1383–1392. 2010.PubMed/NCBI View Article : Google Scholar

97 

Tahri K, Grill JP and Schneider F: Involvement of trihydroxyconjugated bile salts in cholesterol assimilation by bifidobacteria. Curr Microbiol. 34:79–84. 1997.PubMed/NCBI View Article : Google Scholar

98 

Gérard P: Metabolism of cholesterol and bile acids by the gut microbiota. Pathogens. 3:14–24. 2013.PubMed/NCBI View Article : Google Scholar

99 

Nutting CW, Islam S and Daugirdas JT: Vasorelaxant effects of short chain fatty acid salts in rat caudal artery. Am J Physiol. 261:H561–H567. 1991.PubMed/NCBI View Article : Google Scholar

100 

Macfarlane GT and Macfarlane S: Fermentation in the human large intestine: Its physiologic consequences and the potential contribution of prebiotics. J Clin Gastroenterol. 45 (Suppl):S120–S127. 2011.PubMed/NCBI View Article : Google Scholar

101 

Musso G, Gambino R and Cassader M: Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes. Annu Rev Med. 62:361–380. 2011.PubMed/NCBI View Article : Google Scholar

102 

David LA, Maurice CF, Carmody RN, Gootenbreg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, et al: Diet rapidly and reproducibly alters the human gut microbiome. Nature. 505:559–563. 2014.PubMed/NCBI View Article : Google Scholar

103 

Krishnan S, Alden N and Lee K: Pathways and functions of gut microbiota metabolism impacting host physiology. Curr Opin Biotechnol. 36:137–145. 2015.PubMed/NCBI View Article : Google Scholar

104 

Pluznick J: A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes. 5:202–207. 2014.PubMed/NCBI View Article : Google Scholar

105 

Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh M, Gong M, Qi Y, Zubcevic J, et al: Gut dysbiosis is linked to hypertension. Hypertension. 65:1331–1340. 2015.PubMed/NCBI View Article : Google Scholar

106 

Jama HA, Snelson M, Schutte AE, Muir J and Marques FZ: Recommendations for the use of dietary fiber to improve blood pressure control. Hypertension. 81:1450–1459. 2024.PubMed/NCBI View Article : Google Scholar

107 

Falony G, Vieira-Silva S and Raes J: Microbiology meets big data: The case of gut microbiota-derived Trimethylamine. Annu Rev Microbiol. 69:305–321. 2015.PubMed/NCBI View Article : Google Scholar

108 

Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, et al: Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 19:576–585. 2013.PubMed/NCBI View Article : Google Scholar

109 

Zeisel SH and Warrier M: Trimethylamine N-oxide, the microbiome, and heart and kidney disease. Annu Rev Nutr. 37:157–181. 2017.PubMed/NCBI View Article : Google Scholar

110 

Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y and Hazen SL: Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 368:1575–1584. 2013.PubMed/NCBI View Article : Google Scholar

111 

Chittim CL, del Campo AM and Balskus EP: Gut bacterial phospholipase Ds support disease-associated metabolism by generating choline. Nat Microbiol. 4:155–163. 2019.PubMed/NCBI View Article : Google Scholar

112 

Allayee H and Hazen SL: Contribution of gut bacteria to lipid levels: Another metabolic role for microbes? Circ Res. 117:750–754. 2015.PubMed/NCBI View Article : Google Scholar

113 

Ghazalpour A, Cespedes I, Bennett BJ and Allayee H: Expanding role of gut microbiota in lipid metabolism. Curr Opin Lipidol. 27:141–147. 2016.PubMed/NCBI View Article : Google Scholar

114 

Al-Obaide MAI, Singh R, Datta P, Rewers-Felkins KA, Salguero MV, Al-Obaidi I, Kottapalli KR and Vasylyeva TL: Gut microbiota-dependent trimethylamine-N-oxide and serum biomarkers in patients with T2DM and advanced CKD. J Clin Med. 6(86)2017.PubMed/NCBI View Article : Google Scholar

115 

Zhu W, Gregory JC, Org E, Buffa JA, Gupte N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, et al: Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell. 165:111–124. 2016.PubMed/NCBI View Article : Google Scholar

116 

Ley RE, Turnbaugh PJ, Klein S and Gordon JI: Microbial ecology: Human gut microbes associated with obesity. Nature. 444:1022–1023. 2006.PubMed/NCBI View Article : Google Scholar

117 

Liu TX, Niu HT and Zhang SY: Intestinal microbiota metabolism and atherosclerosis. Chin Med J (Engl). 128:2805–2811. 2015.PubMed/NCBI View Article : Google Scholar

118 

Wang Z, Roberts AB, Buffa JA, Levison BS, Zhu W, Oeg E, Gu X, Huang Y, Zamanian-Daryoush M, Culley MK, et al: Non-lethal inhibition of gut microbial Trimethylamine production for the treatment of atherosclerosis. Cell. 163:1585–1595. 2015.PubMed/NCBI View Article : Google Scholar

119 

Martin FP, Wang Y, Sprenger N, Yap IKS, Lundsedt T, Lek P, Rezzi S, Ramadan Z, van Bladeren P, Fay LB, et al: Probiotic modulation of symbiotic gut microbial-host metabolic interactions in a humanized microbiome mouse model. Mol Syst Biol. 4(157)2008.PubMed/NCBI View Article : Google Scholar

120 

Nakamura Y, Masuda O and Takano T: Decrease of tissue angiotensin I-converting enzyme activity upon feeding sour milk in spontaneously hypertensive rats. Biosci Biotechnol Biochem. 60:488–489. 1996.PubMed/NCBI View Article : Google Scholar

121 

Nakamura Y, Yamamoto N, Sakai K and Takano T: Antihypertensive effect of sour milk and peptides isolated from it that are inhibitors to angiotensin I-converting enzyme. J Dairy Sci. 78:1253–1257. 1995.PubMed/NCBI View Article : Google Scholar

122 

Kim B, Park KY, Ji Y, Park S, Holzapfel W and Hyun CK: Protective effects of Lactobacillus rhamnosus GG against dyslipidemia in high-fat diet-induced obese mice. Biochem Biophys Res Commun. 473:530–536. 2016.PubMed/NCBI View Article : Google Scholar

123 

Derrien M and van Hylckama Vlieg JE: Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol. 23:354–366. 2015.PubMed/NCBI View Article : Google Scholar

124 

Deng M, Zhang S, Wu S, Jiang Q, Teng W, Luo T, Ouyang Y, Liu J and Gu B: Lactiplantibacillus plantarum N4 ameliorates lipid metabolism and gut microbiota structure in high fat diet-fed rats. Front Microbiol. 7(1390293)2024.PubMed/NCBI View Article : Google Scholar

125 

Ebel B, Lemetais G, Beney L, Cachon R, Sokol H, Langella P and Gervais P: Impact of probiotics on risk factors for cardiovascular diseases. A review. Crit Rev Food Sci Nutr. 54:175–189. 2014.PubMed/NCBI View Article : Google Scholar

126 

Khalesi S, Sun J, Buys N and Jayasinghe R: Effect of probiotics on blood pressure: A systematic review and meta-analysis of randomized, controlled trials. Hypertension. 64:897–903. 2014.PubMed/NCBI View Article : Google Scholar

127 

De Filippis F, Pellegrini N, Vannini L, Jeffery IB, Storia AL, Laghi L, Serrazanetti D, Cagno RD, Ferrocino I, Lazzi C, et al: High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut. 65:1812–1821. 2016.PubMed/NCBI View Article : Google Scholar

128 

Willett WC, Sacks F, Trichopoulou A, Drescher G, Ferro-Luzzi A, Helsing E and Trichopoulos D: Mediterranean diet pyramid: A cultural model for healthy eating. Am J Clin Nutr. 61:1402S–1406S. 1995.PubMed/NCBI View Article : Google Scholar

129 

Sofi F, Abbate R, Gensini GF and Casini A: Accruing evidence on benefits of adherence to the Mediterranean diet on health: An updated systematic review and meta-analysis. Am J Clin Nutr. 92:1189–1196. 2010.PubMed/NCBI View Article : Google Scholar

130 

Lopez-Garcia E, Rodriguez-Artalejo F, Li TY, Fung TT, Li S, Willett WC, Rimm EB and Hu FB: The Mediterranean-style dietary pattern and mortality among men and women with cardiovascular disease. Am J Clin Nutr. 99:172–180. 2014.PubMed/NCBI View Article : Google Scholar

131 

Cani PD, Delzenne NM, Amar J and Burcelin R: Role of gut microflora in the development of obesity and insulin resistance following high-fat diet feeding. Pathol Biol (Paris). 56:305–309. 2008.PubMed/NCBI View Article : Google Scholar

132 

Zhou X, Li J, Guo J, Geng B, Ji W, Zhao Q, Li J, Liu X, Liu J, Guo Z, et al: Gut-dependent microbial translocation induces inflammation and cardiovascular events after ST-elevation myocardial infarction. Microbiome. 6(66)2018.PubMed/NCBI View Article : Google Scholar

133 

Qi Y, Aranda JM, Rodriguez V, Raizada MK and Pepine CJ: Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension-A case report. Int J Cardiol. 201:157–158. 2015.PubMed/NCBI View Article : Google Scholar

134 

Battson ML, Lee DM, Jarrell DK, Hou S, Ecton KE, Weir TL and Gentile CL: Suppression of gut dysbiosis reverses Western diet-induced vascular dysfunction. Am J Physiol Endocrinol Metab. 314:E468–E477. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

November-2024
Volume 28 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin L, Xiang S, Chen Y, Liu Y, Shen D, Yu X, Wu Z, Sun Y, Chen K, Luo J, Luo J, et al: Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review). Exp Ther Med 28: 427, 2024.
APA
Lin, L., Xiang, S., Chen, Y., Liu, Y., Shen, D., Yu, X. ... Ning, Z. (2024). Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review). Experimental and Therapeutic Medicine, 28, 427. https://doi.org/10.3892/etm.2024.12716
MLA
Lin, L., Xiang, S., Chen, Y., Liu, Y., Shen, D., Yu, X., Wu, Z., Sun, Y., Chen, K., Luo, J., Wei, G., Wang, Z., Ning, Z."Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review)". Experimental and Therapeutic Medicine 28.5 (2024): 427.
Chicago
Lin, L., Xiang, S., Chen, Y., Liu, Y., Shen, D., Yu, X., Wu, Z., Sun, Y., Chen, K., Luo, J., Wei, G., Wang, Z., Ning, Z."Gut microbiota: Implications in pathogenesis and therapy to cardiovascular disease (Review)". Experimental and Therapeutic Medicine 28, no. 5 (2024): 427. https://doi.org/10.3892/etm.2024.12716