16,360
Views
8
CrossRef citations to date
0
Altmetric
Review Article

Gut microbiota and hypertension: association, mechanisms and treatment

, , , , , , & show all
Article: 2195135 | Received 30 Nov 2022, Accepted 20 Mar 2023, Published online: 30 Mar 2023

ABSTRACT

Objectives

Hypertension is one of the most important risk factors for cardio-cerebral vascular diseases, which brings a heavy economic burden to society and becomes a major public health problem. At present, the pathogenesis of hypertension is unclear. Increasing evidence has proven that the pathogenesis of hypertension is closely related to the dysbiosis of gut microbiota. We briefly reviewed relevant literature on gut microbiota and hypertension to summarize the relationship between gut microbiota and hypertension, linked the antihypertension effects of drugs with their modulation on gut microbiota, and discussed the potential mechanisms of various gut microbes and their active metabolites to alleviate hypertension, thus providing new research ideas for the development of antihypertension drugs.

Methods

The relevant literature was collected systematically from scientific database, including Elsevier, PubMed, Web of Science, China National Knowledge Infrastructure (CNKI), Baidu Scholar, as well as other literature sources, such as classic books of herbal medicine.

Results

Hypertension can lead to gut microbiota imbalance and gut barrier dysfunction, including increased harmful bacteria and hydrogen sulfide and lipopolysaccharide, decreased beneficial bacteria and short-chain fatty acids, decreased intestinal tight junction proteins and increased intestinal permeability. Gut microbiota imbalance is closely related to the occurrence and development of hypertension. At present, the main methods to regulate the gut microbiota include fecal microbiota transplantation, supplementation of probiotics, antibiotics, diet and exercise, antihypertensive drugs, and natural medicines.

Conclusions

Gut microbiota is closely related to hypertension. Investigating the correlation between gut microbiota and hypertension may help to reveal the pathogenesis of hypertension from the perspective of gut microbiota, which is of great significance for the prevention and treatment of hypertension.

Introduction

Hypertension is not only a global public health problem but also the most important risk factor for cardiovascular diseases, bringing a heavy economic burden to the society. The epidemiology of hypertension is characterized by high incidence, high disability rate, high mortality rate, and low awareness rate (Citation1) (Citation2). In 2021, about 330 million people are suffering from cardiovascular diseases in China, in which 245 million people are hypertensive patients (Citation3). As the most important risk factor, control of blood pressure has been deemed as an essential method to prevent the incidence of cardiovascular diseases. Currently, available drugs for the treatment of hypertension mainly include angiotensin-converting enzyme inhibitors, angiotensin II (Ang II) receptor antagonists, calcium channel blockers, β-blockers, diuretics, and so on. For patients with poor blood pressure control, it is often necessary to take a combination of multiple antihypertension drugs (Citation1). Despite the continuous development of antihypertension drugs and occurrence of new surgical methods, the control of hypertension is still far from satisfactory (Citation4). How to increase the efficiency of antiprevention drugs and delay the occurrence of hypertension-related cardiovascular diseases is a major challenge at present and even in the future.

Generally, hypertension is the result of superposition of many factors, comprising both genetic and environmental factors (Citation5). The classic pathogenesis of hypertension involves hyperactivity of the sympathetic nervous system, activation of the renin-angiotensin-aldosterone system, vascular endothelial dysfunction, insulin resistance, dysregulation of neurohumoral factors (Citation6,Citation7). Recent investigations have evoked that the gut microbiota is an essential bridge between various environmental factors and our eukaryotic body, playing key roles in the development and progress of multiple diseases (Citation8–10) and mediating the in vivo effects of various medicines (Citation11–14). Previous studies on hypertension mainly focus on peripheral vascular remodeling and central regulation of the body, while environmental factors, especially the internal ecosystem of the human body – gut microbiota, have not been paid enough attention. Increasing studies have demonstrated that the imbalance of gut microbiota plays a key role in the occurrence and development of hypertension (Citation15), and hypertension can also significantly impact the structure and composition of gut microbiota (Citation16,Citation17). Medical interventions by fecal microbiota transplantation (FMT) or supplement of specific gut microbes can effectively change animal’s blood pressure (Citation18,Citation19). Maintaining the homeostasis of gut microbiota contributes to the satisfactory management of blood pressure. Taking gut microbiota as the starting point, this paper reviews the relationship between gut microbiota and hypertension as well as the mechanisms through which gut microbiota regulates the development and progress of hypertension, so as to provide new insights for the development of antihypertension drugs.

Research history for the relationship between gut microbiota and hypertension

The research history of for the relationship between gut microbiota and hypertension is shown in . In 2013, a meta-analysis of 14 randomized controlled clinical trials involving 702 hypertensive patients showed that probiotic fermented milk could significantly reduce the systolic and diastolic blood pressure in hypertensive patients (Citation20). Afterward, Yang et al. (Citation21) reported that in spontaneously hypertensive rats (SHR) and in rats with the long-term injection of Ang II, the richness, diversity, and evenness of gut microbiota decreased significantly and the ratio of Firmicutes/Bacteroidetes (F/B) increased, revealing the imbalance of gut microbiota in hypertensive animals. In 2017, Li et al. (Citation15) reported that the richness, diversity, and gene number of gut microbiota in patients with prehypertension and hypertension were also lower than those in healthy subjects. Further investigations displayed that high-salt diet-induced hypertension involved in dysregulation of gut microbiota. Mell et al. (Citation22) first demonstrated that high salt diet increased blood pressure through modulation of gut microbiota. Wilck et al. (Citation19) found that a high-salt diet could significantly reduce intestinal Lactobacillus while supplementation of Lactobacillus could reduce blood pressure in salt-sensitive hypertensive mice, suggesting that Lactobacillus is closely related to the hypertension caused by a high-salt diet. Bartolomaeus et al. (Citation23) found that exogenous supplementation of gut microbial metabolite propionate could decrease blood pressure in hypertensive mice induced by Ang II infusion. In 2019, Sun et al. (Citation24) published the first population-based cohort study on the relationship between gut microbiota and hypertension, and found that the diversity of gut microbiota was negatively correlated with hypertension. There were significant differences in the structure and composition of gut microbiota in hypertensive patients with different cardiovascular risk stratifications, and some gut microbes were tightly related to the severity of hypertension (Citation25), and suggesting that gut microbiota may participate in the process of hypertension (Citation26). These studies suggest that gut microbiota is a key factor involved in the development and progress of hypertension.

Figure 1. Research history of gut microbiota and hypertension.

Figure 1. Research history of gut microbiota and hypertension.

Association of gut microbiota and hypertension

The changes of gut microbiota in hypertensive patients

In 2017, Li et al. (Citation15) analyzed the structure of gut microbiota in healthy people, prehypertensive and essential hypertension patients through metagenomics and metabonomics. The results showed that the richness, diversity, and gene number of gut microbiota in patients with hypertension were lower than those in healthy controls, and the gut microbiota in subjects with prehypertension was similar to that in hypertensive patients. Later, Sun et al. (Citation24) investigated the cross-sectional association between gut microbiota and blood pressure in the cohort of Coronary Artery Risk Development in Young Adults (CARDIA) study. They found that the gut microbial diversity was negatively correlated with hypertension. This is the first population-based cohort study on the relationship between gut microbiota and hypertension. Afterward, Liu et al. (Citation27) used reverse transcription-quantitative polymerase chain reaction to detect the quantities of gut microbes, which showed that the number of Bacteroides thetaiotaomicron and Bifidobacterium decreased while the abundance of Eubacterium rectale increased in patients with hypertension. Palmu et al. (Citation28) investigated the linkage between the gut microbiota and hypertension in a cohort of 6953 Finns study participants, which showed that hypertensive patients displayed significantly changed gut microbial genera, especially those belonging to Firmicutes. Yan et al. (Citation17) found that some opportunistic pathogens were higher in hypertensive patients, such as, Klebsiella, Streptococcus and Parabacteroides, while short-chain fatty acids (SCFAs) producers including Roseburia and Bacillus freundii were higher in the control group. In a metagenomics study, the composition and function of microbiota in hypertensive patients were significantly changed and the content of fecal butyrate largely decreased compared with those from normal controls (Citation16). Moreover, some gut bacteria are associated with the severity of hypertension and blood pressure fluctuation. The relative abundance of Alistipes finegoldii and Lactobacillus was positively correlated with lower blood pressure variability, while Clostridium and Prevotella with higher blood pressure variability (Citation25). Another clinical study confirmed that there were significant differences in gut microbial structure and relative abundance of special taxa in hypertensive patients with different cardiovascular risk stratifications. The abundance of probiotics decreased significantly with the increase of cardiovascular risk, which suggested that gut microorganisms may be involved in the process of hypertension (Citation26).

Changes of gut microbiota in hypertensive animal models

There are many animal models of hypertension, including spontaneous hypertension rat (SHR), Dahl salt-sensitive (DSS) rats, chronic Ang II infusion-induced hypertensive rats, stress hypertensive rats, and deoxycorticosterone acetate (DOCA)-induced sodium appetite mice. Analysis of fecal samples from SHR or chronic Ang II infusion-induced rats showed that both groups possessed similar dysbiosis of gut microbiota, that is, the ratio of F/B increased, the richness, diversity, and uniformity of microorganisms decreased, and the abundance of acetate- and butyrate-producing bacteria decreased significantly (Citation21). Comparison between the salt-sensitive and salt-tolerant rats showed that their gut microbiota was different, especially, the number of harmful bacteria in the salt-sensitive group increased significantly (Citation19). Wu et al. (Citation29) found that the richness and diversity of gut microbiota in stress hypertensive rats significantly decreased. Chronic stress resulted in the increase of blood pressure in normal rats but did not in antibiotics-treated rats, indicating that gut microbiota might play an important role in the occurrence of stress hypertension. Marque et al. (Citation30) fed high fiber diet and acetate diet to DOCA hypertensive mice respectively. Their results showed that a high-fiber diet could reduce blood pressure by increasing the level of intestinal Bacteroides and acetate concentration. In addition, in the rat model of apnea syndrome with hypertension, the number of lactic acid-producing bacteria increased, while the number of Eubacterium that could convert lactic acid to butyrate decreased exponentially (Citation31). Li et al. (Citation15) transplanted feces from hypertensive patients to aseptic mice and found that systolic blood pressure (SBP), diastolic blood pressure (DBP) and mean arterial pressure of the recipient mice were significantly higher than the healthy control, indicating that gut microbiota can not only regulate blood pressure in the host but also play a role in “hypertensive transmission.”

The relationship between gut barrier dysfunction and hypertension

Gut barrier dysfunction refers to the abnormal change of intestinal permeability and structural damage of intestinal mucosa caused by various reasons, which results in the translocation of bacteria and toxic products into the blood circulation and causes systemic inflammation (Citation32). Several studies have shown that there are obvious gut microbiota disorder and gut barrier dysfunction in patients with hypertension (Citation21,Citation33,Citation34). Gut barrier damage not only causes dysbiosis of gut microbiota, but also leads to an increase in intestinal permeability. Bacterial translocation caused by impaired gut barrier function results in systemic inflammation, which further leads to endothelial cell dysfunction and vascular sclerosis and eventually aggravates hypertension (Citation35). It has been demonstrated that the incidence of gut barrier dysfunction was higher in patients with hypertension than in those without hypertension, and gut barrier dysfunction was mainly manifested by small intestinal mucosal damage and bacterial endotoxin translocation (Citation36). Manner et al. surveyed patients with human immunodeficiency virus infection and found that endotoxin translocation caused by gut barrier dysfunction is a risk factor for hypertension, suggesting that gut barrier damage may be associated with the development of hypertension (Citation37). Kim et al. (Citation16) showed that intestinal fatty acid binding proteins, lipopolysaccharide (LPS), and intestinal-targeted pro-inflammatory T helper 17 (Th17) cells were significantly increased in patients with hypertension, indicating increased intestinal inflammation and permeability in hypertensive patients. Simultaneously, the levels of tight junction protein modulators in the intestinal epithelium were significantly increased, further supporting the speculation that hypertension is accompanied by gut barrier dysfunction. Furthermore, several researches discovered that SHR showed decreased intestinal mucosal thickness and blood flow, lessened glandular goblet cells, reduced intestinal villus height, decreased tight junction proteins, and increased intestinal permeability, implying that hypertension could lead to impaired intestinal barrier function (Citation33,Citation38).

Imbalance of gut microbiota aggravates hypertension

Gut microbiota imbalance refers to the decrease of gut microbiota diversity and the reduction of dominant genera and their beneficial metabolites, which deteriorates gut barrier function, host immune function and gut colonization resistance (Citation39). The normal composition of gut microbiota is essential to maintain the health of the body, and the imbalance of gut microbiota is closely related to the occurrence and progress of hypertension. The ratio of F/B is an important parameter to reflect the imbalance of gut microbiota. The state of intestinal flora imbalance, especially F/B, is related to the change in blood pressure. Studies have found that F/B is significantly increased in hypertensive rat models (Citation40), and the number of Gram-negative bacteria that destroyed the intestinal mucosal barrier increased in hypertensive patients, while the number of Bifidobacterium that protects the intestinal mucosal barrier decreased, which collectively aggravated the chronic inflammatory reaction (Citation19). Robles-Vera et al. (Citation41) found that the number of inflammatory cells in the serum and intestinal tissue of hypertensive patients increased significantly, while the number of Bifidobacterium decreased. Bifidobacterium is a probiotic and participates in the regulation of body immunity. Yan et al. (Citation42) found that the average levels of serum C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) in hypertensive elder patients were significantly higher than the healthy elder controls. Furthermore, they found that Bifidobacterium and Lactobacillus were negatively correlated with CRP, IL-6, and TNF-α, while Escherichia coli and Enterococcus were positively correlated with these inflammatory cytokines. Wilck et al. (Citation19) found that Lactobacillus decreased in the intestines of mice fed with a high-salt diet, which may lead to the increase of Th17 cells and deterioration of salt-sensitive hypertension via driving autoimmunity. In addition, gut microbiota imbalance can destroy intestinal barrier function, increase intestinal mucosal permeability, stimulate bacterial translocation, increase serum level of endotoxin, and cause an inflammatory reaction, which ultimately leads to endothelial dysfunction and hypertension (Citation35). Li et al. (Citation15) transplanted fecal samples from hypertensive patients to germ-free mice and found that hypertension could be transmitted by gut microbiota, confirming that gut microbiota is an important pathogenic factor of hypertension. Both gut microbiota balance and gut barrier dysfunction play an important role in the development of hypertension. Controlling blood pressure by maintenance of gut microbiota homeostasis provides a novel way to prevent and treat hypertension. The relationship between gut microbiota and hypertension is shown in .

Figure 2. The relationship between gut microbiota and hypertension. In healthy individuals, the gut microbiota is in a dynamic balance, including homeostasis of gut microbiota and normal gut barrier function. Patients with hypertension have obvious gut microbiota imbalance and gut barrier dysfunction, including increased harmful bacteria and hydrogen sulfide and LPS, decreased beneficial bacteria and short-chain fatty acids, decreased intestinal tight junction proteins and increased intestinal permeability.

Figure 2. The relationship between gut microbiota and hypertension. In healthy individuals, the gut microbiota is in a dynamic balance, including homeostasis of gut microbiota and normal gut barrier function. Patients with hypertension have obvious gut microbiota imbalance and gut barrier dysfunction, including increased harmful bacteria and hydrogen sulfide and LPS, decreased beneficial bacteria and short-chain fatty acids, decreased intestinal tight junction proteins and increased intestinal permeability.

The mechanism of gut microbiota to regulate blood pressure

Gut microbiota participates in the occurrence and development of hypertension in various ways. Existing evidence has proven that gut barrier function, gut microbiota structure, and gut microbial metabolites are key factors involved in the occurrence and development of hypertension, .

Figure 3. Relevant mechanisms of gut microbiota to regulate blood pressure. The gut microbiota can regulate blood pressure through multiple pathways. When the gut barrier function is normal, the permeability of the intestine is low, which can effectively inhibit the leakage of intestinal pathogens and enterotoxins into the body and reduce the inflammatory damage to intestinal blood vessels, thereby maintaining normal blood pressure. When the gut barrier function is impaired, intestinal permeability increases and harmful gut microbial metabolites pass through the gut barrier and enter the blood circulation to increase blood pressure. When the gut microbiota is imbalanced, a variety of pro-inflammatory factors will be over-produced, which will lead to an increase in intestinal permeability and damage the gut barrier function. At the same time, the level of enteric pathogenic bacteria and enterotoxin LPS increases, both of which will enter the blood circulation, trigger a chronic inflammatory response and damage to vascular endothelial function, thus promoting hypertension via reducing vasodilator factors and enhancing vasoconstrictor factors. Some metabolites of gut microbiota (SCFAs, BAs, H2S) can decrease blood pressure by dilating peripheral blood vessels, maintaining vascular endothelial function, improving insulin sensitivity, lowering blood lipids, reducing inflammatory response, decreasing heart rate, inhibiting the sympathetic nervous system, and protecting kidney function, while other metabolites (TMAO, LPS) can increase blood pressure by constricting blood vessels, increasing inflammatory response, damaging vascular endothelial function, and exacerbating AS.

Figure 3. Relevant mechanisms of gut microbiota to regulate blood pressure. The gut microbiota can regulate blood pressure through multiple pathways. When the gut barrier function is normal, the permeability of the intestine is low, which can effectively inhibit the leakage of intestinal pathogens and enterotoxins into the body and reduce the inflammatory damage to intestinal blood vessels, thereby maintaining normal blood pressure. When the gut barrier function is impaired, intestinal permeability increases and harmful gut microbial metabolites pass through the gut barrier and enter the blood circulation to increase blood pressure. When the gut microbiota is imbalanced, a variety of pro-inflammatory factors will be over-produced, which will lead to an increase in intestinal permeability and damage the gut barrier function. At the same time, the level of enteric pathogenic bacteria and enterotoxin LPS increases, both of which will enter the blood circulation, trigger a chronic inflammatory response and damage to vascular endothelial function, thus promoting hypertension via reducing vasodilator factors and enhancing vasoconstrictor factors. Some metabolites of gut microbiota (SCFAs, BAs, H2S) can decrease blood pressure by dilating peripheral blood vessels, maintaining vascular endothelial function, improving insulin sensitivity, lowering blood lipids, reducing inflammatory response, decreasing heart rate, inhibiting the sympathetic nervous system, and protecting kidney function, while other metabolites (TMAO, LPS) can increase blood pressure by constricting blood vessels, increasing inflammatory response, damaging vascular endothelial function, and exacerbating AS.

Gut barrier function and the regulation of blood pressure

The gut barrier plays a key role in the interaction between the host and the external environment. When the gut barrier function is normal, the intestinal permeability is low, which can effectively inhibit the leakage of intestinal pathogens, enteroendotoxins, and other substances into the body, thus reducing acute and chronic inflammation in all organs (Citation43,Citation44), and to some extent reducing the inflammatory damage to blood vessels and lowering blood pressure. The imbalance of gut microbiota may lead to gut barrier dysfunction and increase intestinal permeability, which stimulates intestinal pathogenic bacteria and LPS entering the blood stream and results in systemic inflammation. These changes will further lead to endothelial cell dysfunction and vascular sclerosis, thus causing or aggravating hypertension (Citation35). In addition, gut barrier dysfunction may affect the growth of probiotics, resulting in the imbalance of gut microbiota and increasing intestinal pathogenic bacteria and LPS (Citation45). These harmful substances will enter the blood circulation through the mesentery, which further aggravates hypertension.

Gut microbiota structure and the regulation of blood pressure

Gut microbiota plays an important role in maintaining the stability of blood pressure, and the structural changes in gut microbiota is closely related to the occurrence of hypertension (Citation46). Previous studies have shown that the structure of gut microbiota is imbalanced, and the number of harmful bacteria Proteobacteria significantly increased in hypertensive patients, which are closely related to intestinal inflammation and immune disorders (Citation33,Citation47). The mechanisms of gut microbiota imbalance to elicit hypertension involve several aspects. First, the imbalanced gut microbiota is usually manifested by reduced probiotics and increased harmful bacteria, which promotes inflammation and leads to abnormal expression of tight junction proteins, zonula occludin-1 (ZO-1) and occluding, in the intestinal mucosa, thus increasing the intestinal permeability and impairing gut barrier function (Citation48). At the same time, the level of intestinal pathogenic bacteria and enterotoxin increases, and these substances enter the blood circulation through the mesentery, thus triggering a chronic inflammation and vascular endothelial damage, and resulting in decreased vasodilator factors and increased vasoconstrictor factors. The increased peripheral resistance eventually leads to increased blood pressure (Citation49). Second, gut microbiota can regulate blood pressure through the inflammatory response. Animal experiments showed that a high-salt diet could result in a significant reduction in intestinal lactic acid bacteria and an increase in CD4+, IL-17A+, and Th17 cells in the intestinal immune system, which caused salt-sensitive hypertension as in humans (Citation19). Transplantation of feces from conventional high-salt-fed mice to germ-free mice elicited obvious inflammation and increased blood pressure, suggesting that gut microbiota is involved in the formation of inflammation and hypertension (Citation50). Furthermore, gut microbiota can stimulate intestinal chromaffin cells to produce serotonin, dopamine, and norepinephrine, which also affects blood pressure levels (Citation49,Citation51). Additionally, gut microbiota can also regulate blood pressure by modulation of steroid hormone levels (Citation52).

Gut microbial metabolites and the regulation of blood pressure

The gut microbiota produces many metabolites, such as trimethylamine-N-oxide (TMAO), SCFAs, corticosterone, hydrogen sulfide (H2S), choline, bile acids (BAs), indole sulfate, LPS, etc., among which SCFAs, TMAO, BAs, H2S, and LPS are closely related to the development of hypertension.

The mechanisms of SCFAs to regulate blood pressure

SCFAs, mainly comprised of butyrate, acetate, and propionate, are an important class of gut microbial metabolites, which are mainly produced by colonic bacteria through fermenting indigestible polysaccharides (fibers). Clinical investigations have shown that although the fecal levels of SCFAs in hypertensive people are significantly higher than that in normal controls (Citation53), the serum levels of SCFAs are negatively correlated with blood pressure (Citation54). Researches have demonstrated that SCFAs can directly dilate blood vessels to decrease blood pressure. In vitro experiments showed that propionate could dilate human colonic resistance arteries (Citation55). Injection of 10 mmol/L propionates into the tail vein of wild mice can reduce blood pressure by about 20 mmHg within 1–2 minutes and the effect is dose-dependent (Citation56). In vivo experiments have shown that supplementing butyrate or acetate in drinking water could prevent the increase of blood pressure in SHR (Citation57), and exogenous propionate supplementation could effectively prevent hypertension in Ang II-induced hypertensive rats (Citation23).

Investigations have shown that SCFAs can regulate blood pressure via activation of G protein-coupled receptors (GPCRs). SCFAs can interact with at least four GPCRs to regulate blood pressure (Citation58), including G protein-coupled receptor 41 (GPR41), G protein-coupled receptor 43 (GPR43), G protein-coupled receptor 109A (GPR109A) and olfactory receptor 78 (Olfr78). Animal experiments showed that exogenous propionate supplementation effectively reduced blood pressure in Ang II-induced hypertensive rats, and this effect was related to the activation of GPR41 in the vascular endothelium by propionate (Citation23). GPR41 and GPR43 are receptors of SCFAs, and propionate is the most potent agonist. Their interaction promoted the release of intracellular Ca2+ (Citation59) and thereby reduced blood pressure. Butyrate is another key SCFA with antihypertension activity. Treatment with butyrate effectively improves proteinuria and lowers blood pressure by protecting podocytes on the glomerular basement membrane and reducing glomerular sclerosis and tissue inflammation, and this effect depends on the expression of GPR109A level (Citation60). Kaye et al showed that lack of prebiotic fiber predisposed mice to hypertension in the presence of a mild hypertensive stimulus, while reintroduction of SCFAs to fiber-depleted mice had protective effects on the development of hypertension, cardiac hypertrophy, and fibrosis via the cognate SCFA receptors GPR43/GPR109A (Citation61). Another study has shown that propionate can regulate the secretion level of renin in the blood and modulate blood pressure through Olfr78 (Citation62). In addition, SCFAs receptors Olfr78, GPR43, and GPR41 were also found to be present in the sympathetic ganglia. SFCAs can directly modulate the sympathetic nervous system by acting on receptors expressed on the sympathetic ganglia (Citation63) and affects the neurofeedback of the gut through receptors expressed on the vagus nerve, thereby participating in the neuromodulation mechanism of blood pressure (Citation51). Together, SCFAs, a key class of gut microbial metabolites, can regulate blood pressure by activating special GPCRs.

Modulation of immune-inflammatory response is another route through which SCFAs regulate blood pressure. Animal experiments have shown that SCFAs promote GPR43-mediated IL-10 increase, thereby reducing intestinal inflammation (Citation64). Supplementation of SCFAs can also regulate blood pressure through modulation of Th17, regulatory T cells (Treg), etc. In AngII-induced hypertension rats, exogenous propionate exerts antihypertensive, anti-inflammatory, and anti-arteriosclerotic effects, which may be related to the immune-inflammatory response mediated by Th17, Treg, and IL-10. Depletion of intrasplenic Tregs by intraperitoneal injection of antibodies substantially abolished the cardioprotective effect of propionate, indicating a critical role of Tregs in propionate-elicited antihypertensive effect (Citation23). Robles-Vera et al (Citation57) administrated SHR with probiotics Bifidobacterium breve and Lactobacillus fermentum and gut microbial metabolites butyrate and acetate. They found that Bifidobacterium breve and Lactobacillus fermentum reduced blood pressure via promoting butyrate-producing bacteria, and the hypotensive effects of butyrate and acetate might be achieved by restoring the balance of Th17/Treg in mesenteric lymph nodes and normalizing endotoxin (Citation57). Supplementation with SCFAs-producing Lactobacillus fermentum could reduce serum Th17 and blood pressure in both high-salt-fed mice and humans (Citation19). Compared with healthy pregnant women, the serum levels of Th1 and Th17 were significantly increased while Treg was decreased in pregnant women with eclampsia, which was accompanied by abnormal levels of inflammatory mediators IL-8, TNF-α, and IL-17 (Citation65). Transplantation of fecal microbes from pregnant women with preeclampsia to pre-pregnancy mice led to Th17/Treg imbalance and increased inflammatory factors in pregnant mice, which in turn elevated blood pressure (Citation66). Exogenous supplementation of SCFAs can alleviate Th17/Treg-mediated inflammatory responses (Citation67,Citation68). Together, the immune-inflammatory response plays an important role in the occurrence and development of hypertension, and SCFAs can regulate blood pressure BP by modulating the levels of immune cells and inflammatory factors.

The mechanisms of TMAO to regulate blood pressure

TMAO is a gut microbial metabolite of dietary substance containing choline or trimethylamine (trimethylamine, TMA), such as phosphatidylcholine, betaine, L-carnitine, trimethylglycine, etc. The TMA lyase (such as choline TMA lyase CutC/D, betaine reductase grade, L carnitine monooxygenase CntA/B and yeaW/X) contained in specific gut microorganisms can convert above components into TMA, which is then absorbed by the host and transformed into TMAO by flavin monooxygenase 3 (FMO3) in the liver (Citation69,Citation70). TMAO enters the systemic circulation and affects hypertension by altering lipid metabolism, platelet activity, obesity status, and the development of atherosclerosis (AS) (Citation71). A meta-analysis showed that plasma TMAO levels were associated with the prevalence of hypertension in a dose-dependent manner (Citation72). The specific mechanism of TMAO to raise blood pressure has not yet been fully elucidated. Studies have shown that TMAO enhances angiotensin II-induced vasoconstriction and promotes angiotensin II-induced hypertension, which is related to protein kinase R (PKR)-like endoplasmic reticulum kinase, PERK)]/reactive oxygen species (ROS)/Ca2+/calmodulin-dependent protein kinase II (CaMKII)/phospholipase Cβ3 (PLCβ3) axis (Citation73). Besides, elevated TMAO can stimulate the release of vasopressin (AVP), up-regulate the expression of aquaporin-2 (AQP-2) in the apical membrane of chief cells of the renal collecting duct, and increase sodium-water retention. In other words, TMAO can increase blood pressure through the “TMAO-AVP-AQP-2 axis” (Citation74). Other studies have shown that TMAO can also increase blood pressure through inducing vascular endothelial dysfunction, which is achieved by promoting oxidative stress and the production of inflammatory mediators in vascular endothelial cells, and inhibiting nitric oxide (NO) synthase-induced NO production (Citation75). However, the specific mechanism of TMAO to increase blood pressure needs more investigations in the future.

The mechanism of BAs to regulate blood pressure

BAs are the main components of bile, and their synthesis and metabolism involve the participation of gut microbes (Citation76). As endocrine-like signaling molecules, BAs regulate lipid metabolism, accelerate energy consumption, maintain gut microbiota homeostasis and protect the intestinal barrier, thus inhibiting inflammation and preventing arteriosclerosis (AS) (Citation77). Under imbalanced gut microbiota conditions, various pathological factors impact the homeostasis of BAs, thereby leading to the occurrence and development of multiple diseases including hypertension. BAs may affect the occurrence and development of hypertension through cardiovascular, renal and TMAO mechanisms.

The most important feature of endothelial dysfunction is the impair of vasomotor regulation. BA receptors such as nuclear receptor farnesoid X receptor (FXR) and membrane receptor G protein-coupled bile acid receptor-1 (GPBAR1) play important roles in regulating endothelial function (Citation78,Citation79). As endogenous vasodilators, BAs promote the production of NO and inhibit the release of Endothelin-1 (ET-1), thus regulating vasomotion and blood pressure (Citation80). Renal-humor feedback is the main mechanism of long-term regulation of blood pressure. Abnormal renal function is a key factor in the pathogenesis of hypertension. In mice, deletion of FXR- or TGR5-related genes is associated with reduced expression of AQP-2 and impaired urine concentration (Citation81), which impacts the development of hypertension. FXR and TGR5 are endogenous renal receptors. BAs can regulate renal pathophysiology by activating FXR, TGR5, and genes involved in inflammation and renal fibrosis (Citation82). Furthermore, BA metabolism is closely related to the TMAO pathway. FXR regulates the activity of FMO3 (Citation83). After entering the liver through the portal circulation, TMA will be oxidized into TMAO by liver FMO3. Afterward, TMAO enters the systemic circulation and promotes hypertension by altering lipid metabolism, platelet activity, and vascular promotion of AS (Citation71). BAs can regulate lipid metabolism by activating FXR (Citation84). Activation of TGR5 can reduce inflammation (Citation85), promote insulin secretion, and improve insulin resistance, which in turn regulates the occurrence of hypertension (Citation86).

The mechanism of H2S to regulate blood pressure

Like mammalian cells and tissues, gut microbes also produce H2S gas which participates in multiple physiological processes, including smooth muscle relaxation, oxidative regulation, and inflammation. H2S is an endogenous vasoactive factor that causes concentration-dependent vasodilation. Studies have shown that Olfr78 expression is up-regulated and GPR41 and GPR43 levels are down-regulated in Ang II-induced hypertensive mice, and exogenous H2S supplementation can partially normalize this change (Citation87). Tomasova et al. (Citation88) found that increasing H2S in the gut can reduce blood pressure, while reduced H2S production showed adverse effects on blood pressure. In addition, H2S can decrease blood pressure by reducing the synthesis and release of renin (Citation89,Citation90). H2S may also lower blood pressure by dilating peripheral blood vessels and lowering heart rate (Citation91). However, Huc et al found that the colon-derived H2S decreased arterial blood pressure but increased portal blood pressure (Citation89,Citation90). Therefore, the precise effect of H2S on blood pressure may be site-specific.

The mechanism of LPS to regulate blood pressure

Gut microbiota disturbance can cause increased secretion and release of intestinal LPS into the blood circulation, which then activates intestinal inflammatory response, damages intestinal mucosal function, and increases intestinal permeability. LPS is a component of Gram-negative bacteria such as Escherichia coli. In animal experiments, LPS can be used to induce vascular dysfunction (Citation92). The integrity of the intestinal barrier and the expression of tight junction proteins in hypertensive patients are damaged, which promotes LPS entering blood circulation to trigger inflammatory response and aggravate hypertension (Citation16,Citation93). Supplementation of probiotics can effectively ameliorate vasodilation, vascular inflammation and hypertension, and these effects were accompanied by decreased LPS levels (Citation94).

Management of hypertension by regulation of gut microbiota

Gut microbiota is closely related to the occurrence and development of hypertension. Therefore, exploring gut microbiota-based intervention methods may provide new strategies for the treatment of hypertension. At present, the main methods to regulate the gut microbiota include FMT, supplementation of probiotics, antibiotics, diet and exercise, antihypertensive drugs, and natural medicines ().

Figure 4. Interventions to regulate gut microbiota.

Figure 4. Interventions to regulate gut microbiota.

FMT

FMT refers to the transplantation of functional bacteria from the stool of a healthy host into the gastrointestinal tract of the patient to rebuild the gut microbiota and achieve the therapeutic purpose. In recent years, more and more attention has been paid to the therapeutic effect of FMT on hypertension. Studies have shown that transplanting feces from hypertensive rats into healthy rats resulted in typical hypertension, while transplanting feces from healthy rats into hypertensive rats can reduce blood pressure (Citation95). Kim et al. (Citation96) transplanted fecal bacteria from healthy mice fed with resveratrol into Ang II induced hypertensive mice and found that the systolic blood pressure (SBP) of hypertensive mice was reduced, which indicated that FMT may be a way to treat hypertension. At present, the mechanism of FMT to alleviate hypertension is still unclear. High-quality clinical trials are still needed to provide rigorous evidence for the effectiveness of FMT to treat hypertension.

Probiotics

Probiotics can change the diversity and structure of gut microbiota, regulate the production of gut microbial metabolites, and improve blood pressure. Clinical trials have found that supplementation of probiotics effectively reduces blood pressure in hypertensive patients. Khalesi et al. (Citation97) conducted a systematic review on the effectiveness of probiotics to control blood pressure. Compared with the control group, intake of probiotics significantly reduced systolic and diastolic blood pressure, and probiotics combination showed more obvious antihypertensive effects than a single one. A meta-analysis based on 14 randomized placebo-controlled clinical trials showed that hypertensive patients received probiotic fermented milk showed a mean decrease in SBP of 3.10 mmHg and DBP of 1.09 mmHg, suggesting that probiotics can effectively reduce systolic and diastolic blood pressure (Citation20). A study in elderly people showed that consumption of dairy products containing Lactobacillus casei fermentation at least 3 times per week can reduce the risk of hypertension (Citation98). In addition, Lactobacillus and Bifidobacterium can exert antihypertensive effects via regulating the renin-angiotensin system (RAS) by producing angiotensin-converting enzyme (ACE) inhibitory peptides, SCFAs, conjugated linoleic acid and gamma-aminobutyric acid (GABA) (Citation99,Citation100). Sipola et al. (Citation101) reported that yogurt fermented by Lactobacillus LBK-16 H contained ACE inhibitory peptides which could effectively delay the progression of hypertension in SHR. Mizushim et al. (Citation102) showed that daily intake of 160 g of Lactobacillus-fermented yogurt for 4 weeks resulted in a decrease in SBP of 5.2 mmHg and DBP of 2.0 mmHg. Inoue et al. (Citation103) reported that ingest of Lactobacillus-fermented yogurt for 12 weeks decreased SBP by (17.4 ± 4.3) mmHg and decreased DBP by (7.5 ± 5.7) mmHg in patients with mild hypertension. Overall, probiotics is promising to treat hypertension, but more studies are needed to fully understand their mechanisms to control blood pressure.

Antibiotics

The effectiveness of antibiotics to treat hypertension remains controversial and individual difference should be noted. Yang et al. (Citation21) found that high blood pressure is associated with gut microbiota dysbiosis and treatment with minocycline could rebalance the dysbiotic gut microbiota and attenuate hypertension. Galla et al. (Citation104) used amoxicillin to change the gut microbiota of young genetically hypertensive rats and pregnant and lactating rats. Amoxicillin remodeled the structure of gut microbiota and reduced the ratio of F/B, which eventually decreased blood pressure. In young rats treated with amoxicillin, the blood pressure-lowering effect persisted even after antibiotic discontinuation. These results suggest that antibiotics can decrease blood pressure through remodeling gut microbiota. In an experiment on drug interaction between amlodipine and antibiotics (ampicillin), gut microbiota participated in the metabolism of amlodipine, and the intake of antibiotics may improve the bioavailability of amlodipine by inhibiting gut microbes, thus improving its antihypertensive efficacy (Citation105). It has been reported that the combination of antibiotics to inhibit the growth of some gut bacteria can reduce blood pressure in the treatment of refractory hypertension (Citation106). However, some studies have revealed that the antihypertensive effects of antibiotics are not the same on different animal models. Galla et al reported that oral administration of antibiotics increased systolic blood pressure of the Dahl salt-sensitive rat, while minocycline and vancomycin, but not neomycin, lowered systolic blood pressure in the SHR (Citation107). At present, the clinical research on the antihypertensive effect of antibiotics is relatively insufficient. The feasibility of antibiotics for the treatment of hypertension needs further investigation because of the high adverse effects of antibiotics to human beings.

Diet and exercise

There is increasing evidence that dietary nutrients are essential for the maintenance of gut microbiota homeostasis and host health (Citation108–110). A high-fiber diet attributes to healthy gut microbiota and exerts antihypertensive effect. Feeding with high-fiber diet to hypertensive mice effectively reduced blood pressure (Citation30). Ingest of high-fiber diet increases the amount of acetate and other SCFAs and modulates the structure and composition of gut microbiota, which is beneficial for blood pressure control. SCFAs are usually fermentation products of indigestible polysaccharides (fiber) by gut microbes. Dietary fiber can modulate gut microbiota and promote the production of SCFAs (acetic acid, propionic acid, butyric acid, etc.). Both systolic and diastolic blood pressure of hypertensive mice can be effectively decreased by supplementing high-fiber food and acetate (Citation56). Therefore, dietary supplementation of fiber and SCFAs can alleviate hypertension and maintain normal blood pressure. Marque et al. (Citation30) showed that a high-fiber diet could play an antihypertensive role by increasing the level of intestinal Bacteroides and the concentration of acetate. Exercise can also increase the diversity of gut microbiota, promote SCFAs production, and regulate blood pressure (Citation111). Compared with sedentary people, the levels of SCFAs in athletes are relatively increased (Citation112). Simultaneously, exercise can increase the diversity and richness of gut bacteria, enhance the levels of SCFAs andBAs, and improve inflammatory response (Citation112).

Drug intervention

Antihypertensive drugs

Losartan, captopril, benazepril, and amlodipine are common antihypertensive drugs. Robles-Vera et al (Citation113) evaluated the effect of the angiotensin receptor antagonist losartan on the gut microbiota of SHR and found that losartan significantly changed the structure of gut microbiota. To some extent, losartan can protect the vascular system and reduce blood pressure by regulating the intestinal immune system. Other studies have shown that angiotensin-converting enzyme inhibitor captopril can reduce blood pressure and restore the permeability of the intestinal barrier in SHR (Citation33). Xu et al (Citation114) assessed the impact of benazepril and amlodipine on gut microbiota in SHR and found that both drugs modulated the αand β diversity of gut microbiota and changed the composition at the phylum and genus levels. Gut microbiota is also involved in the metabolism of amlodipine, and combination with antibiotics improves the bioavailability of amlodipine and enhances its antihypertensive effect (Citation105).

Natural medicines

It has been shown that natural medicines can alleviate hypertension through modulation in different ways, such as regulating the ratio of probiotics to pathogenic bacteria, restoring the diversity of gut microbiota, improving intestinal barrier function, and regulating the metabolites of gut microbiota (Citation115). Ma et al (Citation116) showed that treatment with Huanglian Jiedu decoction for 6 weeks increased the diversity of gut microbiota, enhanced the abundance of Lactobacillus, and decreased Firmicutes. It is speculated that Huanglian Jiedu decoction may reduce blood pressure through regulating the structure of gut microbiota, promoting beneficial bacteria, and inhibiting harmful bacteria (Citation117). Wu et al (Citation118) found that Sanhuang Xiexin decoction could dilate blood vessels, protect vascular endothelial cells, and decrease blood pressure, which is accompanied by significantly increase of beneficial bacteria such as Lactobacillus. Administration of Radix Astragali-Radix Salviae Miltiorrhizae to SHR for 28 days effectively decreased the blood pressure and enhanced the diversity of gut microbiota. The increase of probiotics such as Lactobacillus and Bifidobacterium was closely related to the decrease of blood pressure (Citation119). Qi et al (Citation120) assessed the effect of Eucommia ulmoides-Tribuli Fructus (EUTF) on blood pressure and gut microbiota in SHR. They found that EUTF could reduce blood pressure and increase the diversity of gut microbiota, suggesting that the antihypertensive effect of EUTF is associated with the improvement of gut microbiota. Xiong et al (Citation121) found that Qinggan Yishen Qufeng could improve the intestinal permeability, increase the expression of intestinal tight junction protein, and reduce the intestinal damage caused by hypertensive lesions. Yu et al (Citation122) showed that the antihypertensive drug Zhengan Xifeng decoction could effectively reduce the F/B value and enhance the production of SCFAs, thereby maintaining the ecological balance and integrity of the intestinal tract. It has been well-documented that some natural products such as berberine (Citation123) and guneulsterone (Citation124) can regulate the gut microbiota⁃TMA pathway to reduce the TMAO levels in peripheral blood. Resveratrol can regulate BAs and down-regulate the farnesoid X receptor -fibroblast growth factor 15 (FXR-FGF15) pathway, which plays a role in preventing atherosclerosis in ApoE-/- mice (Citation125). Quercetin can reduce the production of LPS and alleviate inflammation, thus improving vascular remodeling and lowering blood pressure (Citation126). Han et al (Citation127) reported that Huangqi (Astragali Radix) plus Danshen (Salviae Miltiorrhizae Radix Et Rhizoma) could improve gut microbiota and enhance the production of beneficial metabolites, thus reducing blood pressure SHR.

Conclusion and perspectives

Gut microbiota and its metabolites are closely related to the occurrence and development of hypertension. Investigating the correlation between gut microbiota and hypertension may help to reveal the pathogenesis of hypertension from the perspective of gut microbiota, which is of great significance for the prevention and treatment of hypertension. Gut microbiota-targeted antihypertensive therapy is a promising strategy, providing great inspiration for solving the dilemma of current hypertension treatment. The study on the mechanisms of gut microbiota in the formation of hypertension not only aims to explore the etiology, but also provides a large number of new targets for clinical treatment, which is helpful to develop novel drugs to treat hypertension and choose proper antihypertensive drugs based on the specific gut microbial characteristics in individuals. Treatment of hypertension by regulating gut microbiota may be a new strategy that makes up for the deficiency of current antihypertensive therapies. The combination of a high-fiber diet, probiotics and prebiotics, fecal bacteria transplantation, natural medicines, and other gut microbiota-targeted therapies with conventional drugs may be a new approach to achieve ideal hypertension management.

Due to the complex etiology of hypertension and the diverse structure of gut microbiota, the current research on the relationship between gut microbiota and hypertension is still in the initial stage, and the research methods are not mature enough. In addition, the relationship between gut microbiota and hypertension is heavily affected by diet, exercise, drugs, psychology, and other factors. Therefore, future studies should pay special attention to the standardization of sample acquisition, processing and metagenomic data analysis. The establishment of a microecological characteristic diagnosis and treatment system for hypertension based on gut microbiota can provide a constructive scheme for the prevention and treatment of hypertension in the future. According to the specific function of microorganisms, new probiotics can be developed to promote human health. Gut microbiota is directly or indirectly involved in diet and drug metabolism. Therefore, it is helpful to realize the characteristic diagnosis and treatment model of hypertension as soon as possible by studying the targeted and specialized drug delivery of gut microbiota and mining new bacteria, new genes, and new functions.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was financially supported by the National Natural Science Foundation of China [81973217], Natural Science Foundation project of Inner Mongolia [2021ZD16], and The Central Government Guiding Special Funds for Development of Local Science and Technology [2021ZY0046]

References

  • Williams B, Mancia G, Spiering W, Agabiti Rosei E, Azizi M, Burnier M, Clement DL, Coca A, de Simone G, Dominiczak A, et al. ESC/ESH guidelines for the management of arterial hypertension. Eur Heart J. 2018;39(33):3021–15. doi:10.1093/eurheartj/ehy339.
  • Wu Y, Jin A, Xie G, Wang L, Liu K, Jia G, Liang X, Xu J. The 20 most important and most preventable health problems of China: a DelphI consultation of Chinese Experts. Am J Public Health. 2018;108(12):1592–98. doi:10.2105/ajph.2018.304684.
  • The Writing Committee of the Report on Cardiovascular Health and Diseases in China. Report on Cardiovascular health and diseases in China 2021: an updated summary. Chin Circ J. 2022;37:553–78. doi:10.3969/j.issn.1000-3614.2022.06.001.
  • Wang Z, Chen Z, Zhang L, Wang X, Hao G, Zhang Z, Shao L, Tian Y, Dong Y, Zheng C, et al. Status of Hypertension in China: results from the China Hypertension Survey, 2012-2015. Circulation. 2018;137(22):2344–56. doi:10.1161/CIRCULATIONAHA.117.032380.
  • Sekar D, Shilpa BR, Das AJ. Relevance of microRNA 21 in different Types of Hypertension. Curr Hypertens Rep. 2017;19(7):57. doi:10.1007/s11906-017-0752-z.
  • Pugh D, Dhaun N. Hypertension and Vascular inflammation another piece of the Genetic puzzle. Hypertension. 2021;77(1):190–92. doi:10.1161/Hypertensionaha.120.16420.
  • McMaster WG, Kirabo A, Madhur MS, Harrison DG. Inflammation, immunity, and Hypertensive end-organ damage. Circ Res. 2015;116(6):1022–33. doi:10.1161/Circresaha.116.303697.
  • Duan Y, Wu X, Yang Y, Gu L, Liu L, Yang Y, Zhou J, Wu C, Jin F. Marked shifts in gut microbial structure and neurotransmitter metabolism in fresh inmates revealed a close link between gut microbiota and mental health: a case-controlled study. Int J Clin Health Psychol. 2022;22(3):100323. doi:10.1016/j.ijchp.2022.100323.
  • 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. doi:10.1016/j.jare.2022.06.008.
  • Miyauchi E, Shimokawa C, Steimle A, Desai MS, Ohno H. The impact of the gut microbiome on extra-intestinal autoimmune diseases. Nat Rev Immunol. 2022;23(1):9–23. doi:10.1038/s41577-022-00727-y.
  • Wu C, Zhao Y, Zhang Y, Yang Y, Su W, Yang Y, Sun L, Zhang F, Yu J, Wang Y, et al. Gut microbiota specifically mediates the anti-hypercholesterolemic effect of berberine (BBR) and facilitates to predict BBR’s cholesterol-decreasing efficacy in patients. J Adv Res. 2022;37:197–208.
  • Zhang X, Yang Y, Zhang F, Yu J, Sun W, Wang R, Wu C. Traditional Chinese medicines differentially modulate the gut microbiota based on their nature (Yao-Xing). Phytomedicine. 2021;85:153496. doi:10.1016/j.phymed.2021.153496.
  • Yang YN, Wang QC, Xu W, Yu J, Zhang H, Wu C. The berberine-enriched gut commensal Blautia producta ameliorates high-fat diet (HFD)-induced hyperlipidemia and stimulates liver LDLR expression. Biomed Pharmacother. 2022;155:113749. doi:10.1016/j.biopha.2022.113749.
  • McCulloch JA, Davar D, Rodrigues RR, Badger JH, Fang JR, Cole AM, Balaji AK, Vetizou M, Prescott SM, Fernandes MR, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med. 2022;28(3):545–56. doi:10.1038/s41591-022-01698-2.
  • 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(1):14. doi:10.1186/s40168-016-0222-x.
  • Kim S, Goel R, Kumar A, Qi Y, Lobaton G, Hosaka K, Mohammed M, Handberg EM, Richards EM, Pepine CJ, et al. Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. Clin Sci Lond. 2018;132(6):701–18. doi:10.1042/CS20180087.
  • 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.
  • Adnan S, Nelson JW, Ajami NJ, Venna VR, Petrosino JF, Bryan RM Jr., Durgan DJ. Alterations in the gut microbiota can elicit hypertension in rats. Physiol Genomics. 2017;49(2):96–104. doi:10.1152/physiolgenomics.00081.2016.
  • Wilck N, Matus MG, Kearney SM, Olesen SW, Forslund K, Bartolomaeus H, Haase S, Mahler A, Balogh A, Marko L, et al. Salt-responsive gut commensal modulates TH17 axis and disease. Nature. 2017;551(7682):585–89. doi:10.1038/nature24628.
  • Dong JY, Szeto IMY, Makinen K, Gao QT, Wang JK, Qin LQ, Zhao YY. Effect of probiotic fermented milk on blood pressure: a meta-analysis of randomised controlled trials. Brit J Nutr. 2013;110(7):1188–94. doi:10.1017/S0007114513001712.
  • 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. 2015;65(6):1331–40. doi:10.1161/hypertensionaha.115.05315.
  • Mell B, Jala VR, Mathew AV, 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 Genomics. 2015;47(6):187–97. doi:10.1152/physiolgenomics.00136.2014.
  • Bartolomaeus H, Balogh A, Yakoub M, Homann S, Marko L, Hoges S, Tsvetkov D, Krannich A, Wundersitz S, Avery EG, et al. Short-Chain fatty acid propionate protects from Hypertensive Cardiovascular damage. Circulation. 2019;139(11):1407–21. doi:10.1161/CIRCULATIONAHA.118.036652.
  • Sun S, Lulla A, Sioda M, Winglee K, Wu MC, Jacobs DR Jr., Shikany JM, Lloyd-Jones DM, Launer LJ, Fodor AA, et al. Gut microbiota composition and blood pressure. Hypertension. 2019;73(5):998–1006. doi:10.1161/HYPERTENSIONAHA.118.12109.
  • Dinakis E, Nakai M, Gill P, Ribeiro R, Yiallourou S, Sata Y, Muir J, Carrington M, Head GA, Kaye DM, et al. Association between the gut microbiome and their metabolites with human blood pressure variability. Hypertension. 2022;79(8):1690–701. doi:10.1161/Hypertensionaha.122.19350.
  • Chen WY, Chen YP, Feng T, Hu XW, Zhou FY, Lu XQ, Zhang QX, Zhang XL, Chen P, Xiu JC. Analysis of gut microbiota diversity in people with different cardiovascular risk stratified of hypertension. J Pract Med. 2019;35:1415–20. doi:10.3969/j.issn.1006⁃5725.2019.09.001.
  • Liu J, An N, Ma C, Li X, Zhang J, Zhu W, Zhang Y, Li J. Correlation analysis of intestinal flora with hypertension. Exp Ther Med. 2018;16:2325–30. doi:10.3892/etm.2018.6500.
  • Palmu J, Salosensaari A, Havulinna AS, Cheng S, Inouye M, Jain M, Salido RA, Sanders K, Brennan C, Humphrey GC, et al. Association between the gut microbiota and blood pressure in a population Cohort of 6953 individuals. J Am Heart Assoc. 2020;9(15):e016641. doi:10.1161/jaha.120.016641.
  • Wu Q, Xu ZY, Liu LP, Zhang WY, Song SY. Role of gut microbiota in stress-induced hypertension in rats. Biotechnol Bulletin. 2020;36:83–90. doi:10.13560/j.cnki.biotech.bull.1985.2019-1215.
  • Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann M, Tan JK, Kuruppu S, Rajapakse NW, 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(10):964–77. doi:10.1161/CIRCULATIONAHA.116.024545.
  • Durgan DJ, Ganesh BP, Cope JL, Ajami NJ, Phillips SC, Petrosino JF, Hollister EB, Bryan RM Jr. Role of the gut microbiome in obstructive sleep Apnea–Induced Hypertension. Hypertension. 2016;67(2):469–74. doi:10.1161/HYPERTENSIONAHA.115.06672.
  • Druml W. Intestinal cross-talk: the gut as motor of multiple organ failure. Medizinische Klinik - Intensivmedizin und Notfallmedizin. 2018;113(6):470–77. doi:10.1007/s00063-018-0475-1.
  • Santisteban MM, Qi Y, Zubcevic J, Kim S, Yang T, Shenoy V, Cole-Jeffrey CT, Lobaton GO, Stewart DC, Rubiano A, et al. Hypertension-linked Pathophysiological alterations in the Gut. Circ Res. 2017;120(2):312–23. doi:10.1161/CIRCRESAHA.116.309006.
  • 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. 2018;6(1):66. doi:10.1186/s40168-018-0441-4.
  • Andersen K, Kesper MS, Marschner JA, Konrad L, Ryu M, Kumar Vr S, Kulkarni OP, Mulay SR, Romoli S, Demleitner J, et al. Intestinal Dysbiosis, Barrier Dysfunction, and Bacterial translocation account for CKD–Related systemic inflammation. J Am Soc Nephrol. 2017;28(1):76–83. doi:10.1681/ASN.2015111285.
  • Lin D. Assocation between hypertension and impairment of intestinal barrier function. Guangzhou (CN): Guangdong Pharmaceutical University. 2019.
  • Manner IW, Baekken M, Kvale D, Oektedalen O, Pedersen M, Nielsen SD, Nowak P, Os I, Troseid M. Markers of microbial translocation predict hypertension in HIV-infected individuals. HIV Med. 2013;14(6):354–61. doi:10.1111/hiv.12015.
  • Jaworska K, Huc T, Samborowska E, Dobrowolski L, Bielinska K, Gawlak M, Ufnal M, Chamaillard M. Hypertension in rats is associated with an increased permeability of the colon to TMA, a gut bacteria metabolite. Plos One. 2017;12(12):e0189310. doi:10.1371/journal.pone.0189310.
  • Yang ZH, Liu YX, Wang L, Lin SS, Dai XD, Yan HF, Ge Z, Ren QA, Wang H, Zhu F, et al. Traditional Chinese medicine against COVID-19: role of the gut microbiota. Biomed Pharmacother. 2022;149:112787. 10.1016/j.biopha.2022.112787
  • Karbach SH, Schonfelder T, Brandao I, Wilms E, Hormann N, Jackel S, Schuler 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(9). doi:10.1161/JAHA.116.003698.
  • Robles-Vera I, Toral M, de la Visitacion N, Sanchez M, Romero M, Olivares M, Jimenez 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(19):e1800298. doi:10.1002/mnfr.201800298.
  • Yan PL, Luo YJ, Liu HF, Chen Y, Ge MH. Changes of gut microbiota and its correlation with serum inflammatory indexes in elderly patients with hypertensio. Chin J Gerontology. 2020;40:3871–74. doi:10.3969/j.issn.1005-9202.2020.18.021.
  • Di Tommaso N, Gasbarrini A, Ponziani FR. Intestinal barrier in human health and disease. Int J Environ Res Public Health. 2021;18(23):12836. doi:10.3390/ijerph182312836.
  • Qiao J, Chen R, Wang M, Bai R, Cui X, Liu Y, Wu C, Chen C. Perturbation of gut microbiota plays an important role in micro/nanoplastics-induced gut barrier dysfunction. Nanoscale. 2021;13(19):8806–16. doi:10.1039/d1nr00038a.
  • Yang Z, Liu Y, Wang L, Lin S, Dai X, Yan H, Ge Z, Ren Q, Wang H, Zhu F, et al. Traditional Chinese medicine against COVID-19: role of the gut microbiota. Biomed Pharmacother. 2022;149:112787.
  • Galla S, Chakraborty S, Mell B, Vijay-Kumar M, Joe B. Microbiotal-Host Interactions and Hypertension. Physiology. 2017;32(3):224–33. doi:10.1152/physiol.00003.2017.
  • Mushtaq N, Hussain S, Zhang S, Yuan L, Li H, Ullah S, Wang Y, Xu J. Molecular characterization of alterations in the intestinal microbiota of patients with grade 3 hypertension. Int J Mol Med. 2019;44:513–22. doi:10.3892/ijmm.2019.4235.
  • Larabi A, Barnich N, Nguyen HTT. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy. 2020;16(1):38–51. doi:10.1080/15548627.2019.1635384.
  • Sircana A, De Michieli F, Parente R, Framarin L, Leone N, Berrutti M, Paschetta E, Bongiovanni D, Musso G. Gut microbiota, hypertension and chronic kidney disease: recent advances. Pharmacol res. 2019;144:390–408. doi:10.1016/j.phrs.2018.01.013.
  • Ferguson JF, Aden LA, Barbaro NR, Van Beusecum JP, Xiao L, Simmons AJ, Warden C, Pasic L, Himmel LE, Washington MK, et al. High dietary salt–induced DC activation underlies microbial dysbiosis-associated hypertension. JCI Insight. 2019;4(13):5. doi:10.1172/jci.insight.126241.
  • Zubcevic J, Richards EM, Yang T, Kim S, Sumners C, Pepine CJ, Raizada MK. Impaired autonomic nervous system-microbiome circuit in Hypertension. Circ Res. 2019;125(1):104–16. doi:10.1161/CIRCRESAHA.119.313965.
  • Yan X, Jin J, Su X, Yin X, Gao J, Wang X, Zhang S, Bu P, Wang M, Zhang Y, et al. Intestinal Flora modulates blood pressure by regulating the Synthesis of intestinal-derived Corticosterone in high salt-induced Hypertension. Circ Res. 2020;126(7):839–53. doi:10.1161/CIRCRESAHA.119.316394.
  • Huart J, Leenders J, Taminiau B, Descy J, Saint-Remy A, Daube G, Krzesinski J-M, Melin P, de Tullio P, Jouret F. Gut microbiota and fecal levels of short-Chain fatty acids differ upon 24-hour blood pressure levels in men. Hypertension. 2019;74(4):1005–13. doi:10.1161/hypertensionaha.118.12588.
  • Chen L, He FJ, Dong Y, Huang Y, Wang C, Harshfield GA, Zhu H. ModesT sodium reduction increases circulating short-Chain Fatty Acids in Untreated Hypertensives: a Randomized, Double-Blind, Placebo-Controlled Trial. Hypertension. 2020;76(1):73–79. doi:10.1161/HYPERTENSIONAHA.120.14800.
  • Mortensen FV, Nielsen H, Mulvany MJ, Hessov I. Short chain fatty acids dilate isolated human colonic resistance arteries. Gut. 1990;31(12):1391–94. doi:10.1136/gut.31.12.1391.
  • Pluznick JL, Protzko RJ, Gevorgyan H, Peterlin Z, Sipos A, Han J, Brunet I, Wan LX, 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 U S A. 2013;110(11):4410–15. doi:10.1073/pnas.1215927110.
  • Robles-Vera I, Toral M, de la Visitacion N, Sanchez M, Gomez-Guzman M, Romero M, Yang T, Izquierdo-Garcia JL, Jimenez R, Ruiz-Cabello J, et al. Probiotics Prevent Dysbiosis and the rise in blood pressure in genetic Hypertension: role of Short-Chain Fatty Acids. Mol Nutr Food Res. 2020;64(6):e1900616. doi:10.1002/mnfr.201900616.
  • Tan JK, McKenzie C, Marino E, Macia L, Mackay CR. Metabolite-sensing G protein–Coupled Receptors—Facilitators of diet-related immune regulation. Annu Rev Immunol. 2017;35(1):371–402. doi:10.1146/annurev-immunol-051116-052235.
  • Le Poul E, Loison C, Struyf S, Springael JY, Lannoy V, Decobecq ME, Brezillon S, Dupriez V, Vassart G, Van Damme J, et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278(28):25481–89. doi:10.1074/jbc.M301403200.
  • Felizardo RJF, de Almeida DC, Pereira RL, Watanabe IKM, Doimo NTS, Ribeiro WR, Cenedeze MA, Hiyane MI, Amano MT, Braga TT, et al. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms. Faseb J. 2019;33(11):11894–908. doi:10.1096/fj.201901080R.
  • Kaye DM, Shihata WA, Jama HA, 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(17):1393–403. doi:10.1161/circulationaha.119.043081.
  • Pluznick J. A novel SCFA receptor, the microbiota, and blood pressure regulation. Gut Microbes. 2014;5(2):202–07. doi:10.4161/gmic.27492.
  • Nohr MK, Egerod KL, Christiansen SH, Gille A, Offermanns S, Schwartz TW, Moller M. Expression of the short chain fatty acid receptor GPR41/FFAR3 in autonomic and somatic sensory ganglia. Neuroscience. 2015;290:126–37. doi:10.1016/j.neuroscience.2015.01.040.
  • Sun M, Wu W, Chen L, Yang W, Huang X, Ma C, Chen F, Xiao Y, Zhao Y, Ma C, et al. Microbiota-derived short-chain fatty acids promote Th1 cell IL-10 production to maintain intestinal homeostasis. Nat Commun. 2018;9(1):3555. doi:10.1038/s41467-018-05901-2.
  • Salazar Garcia MD, Mobley Y, Henson J, Davies M, Skariah A, Dambaeva S, Gilman-Sachs A, Beaman K, Lampley C, Kwak-Kim J. Early pregnancy immune biomarkers in peripheral blood may predict preeclampsia. J Reprod Immunol. 2018;125:25–31. doi:10.1016/j.jri.2017.10.048.
  • Chen X, Li P, Liu M, Zheng H, He Y, Chen M-X, Tang W, Yue X, Huang Y, Zhuang L, et al. Gut dysbiosis induces the development of pre-eclampsia through bacterial translocation. Gut. 2020;69(3):513–22. doi:10.1136/gutjnl-2019-319101.
  • Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–55. doi:10.1038/nature12726.
  • Liu YJ, Tang B, Wang FC, Tang L, Lei YY, Luo Y, Huang SJ, Yang M, Wu LY, Wang W, et al. Parthenolide ameliorates colon inflammation through regulating Treg/Th17 balance in a gut microbiota-dependent manner. Theranostics. 2020;10(12):5225–41. doi:10.7150/thno.43716.
  • Romano KA, Vivas EI, Amador-Noguez D, Rey FE, Blaser MJ. Intestinal microbiota composition modulates choline bioavailability from diet and accumulation of the proatherogenic metabolite trimethylamine-N-oxide. mBio. 2015;6(2):e02481. doi:10.1128/mBio.02481-14.
  • Liu Y, Dai M. Trimethylamine N-Oxide generated by the gut microbiota is associated with Vascular inflammation: new insights into Atherosclerosis. Mediators Inflamm. 2020;2020:4634172. doi:10.1155/2020/4634172.
  • Duttaroy AK. Role of gut microbiota and their metabolites on Atherosclerosis, Hypertension and Human Blood Platelet Function: a review. Nutrients. 2021;13(1):144. doi:10.3390/nu13010144.
  • Ge X, Zheng L, Zhuang R, Yu P, Xu Z, Liu G, Xi X, Zhou X, Fan H. The gut microbial metabolite Trimethylamine N-Oxide and Hypertension risk: a systematic review and dose–response meta-analysis. Adv Nutrition. 2020;11(1):66–76. doi:10.1093/advances/nmz064.
  • Jiang S, Shui Y, Cui Y, Tang C, Wang X, Qiu X, Hu W, Fei L, Li Y, Zhang S, et al. Gut microbiota dependent trimethylamine N-oxide aggravates angiotensin II–induced hypertension. Redox Biol. 2021;46:102115.
  • Liu M, Han Q, Yang J. Trimethylamine-N-oxide (TMAO) increased aquaporin-2 expression in spontaneously hypertensive rats. Clin Exp Hypertens. 2019;41(4):312–22. doi:10.1080/10641963.2018.1481420.
  • Chen H, Li J, Li N, Liu H, Tang J. Increased circulating trimethylamine N-oxide plays a contributory role in the development of endothelial dysfunction and hypertension in the RUPP rat model of preeclampsia. Hypertens Pregnancy. 2019;38(2):96–104. doi:10.1080/10641955.2019.1584630.
  • Yang Y, Wu C. Targeting gut microbial bile salt hydrolase (BSH) by diet supplements: new insights into dietary modulation of human health. Food Funct. 2022;13(14):7409–22. doi:10.1039/d2fo01252a.
  • Brown JM, Hazen SL. Microbial modulation of cardiovascular disease. Nat Rev Microbiol. 2018;16(3):171–81. doi:10.1038/nrmicro.2017.149.
  • Schwabl P, Hambruch E, Seeland BA, Hayden H, Wagner M, Garnys L, Strobel B, Schubert T-L, Riedl F, Mitteregger D, et al. The FXR agonist PX20606 ameliorates portal hypertension by targeting vascular remodelling and sinusoidal dysfunction. J Hepatol. 2017;66(4):724–33. doi:10.1016/j.jhep.2016.12.005.
  • Kida T, Omori K, Hori M, Ozaki H, Murata T. Stimulation of G protein–coupled bile acid receptor enhances Vascular Endothelial Barrier function via activation of protein Kinase a and Rac1. J Pharmacol Exp Ther. 2014;348(1):125–30. doi:10.1124/jpet.113.209288.
  • Fiorucci S, Zampella A, Cirino G, Bucci M, Distrutti E. Decoding the vasoregulatory activities of bile acid-activated receptors in systemic and portal circulation: role of gaseous mediators. Am J Physiol Heart Circ Physiol. 2017;312(1):H21–32. doi:10.1152/ajpheart.00577.2016.
  • Li S, Li C, Wang W. Bile acid signaling in renal water regulation. Am J Physiol Renal Physiol. 2019;317(1):F73–76. doi:10.1152/ajprenal.00563.2018.
  • Herman-Edelstein M, Weinstein T, Levi M. Bile acid receptors and the kidney. Curr Opin Nephrol Hypertens. 2018;27(1):56–62. doi:10.1097/MNH.0000000000000374.
  • Verhaar BJH, Prodan A, Nieuwdorp M, Muller M. GuT microbiota in hypertension and Atherosclerosis: a review. Nutrients. 2020;12(10):2982. doi:10.3390/nu12102982.
  • D’agati VD, Chagnac A, de Vries AP, Levi M, Porrini E, Herman-Edelstein M, Praga M. Obesity-related glomerulopathy: clinical and pathologic characteristics and pathogenesis. Nat Rev Nephrol. 2016;12(8):453–71. doi:10.1038/nrneph.2016.75.
  • Cheng L, Huang C, Chen Z. Tauroursodeoxycholic acid Ameliorates Lipopolysaccharide-induced depression like behavior in mice via the inhibition of Neuroinflammation and oxido-Nitrosative stress. Pharmacology. 2019;103(1–2):93–100. doi:10.1159/000494139.
  • Kumar DP, Rajagopal S, Mahavadi S, Mirshahi F, Grider JR, Murthy KS, Sanyal AJ. Activation of transmembrane bile acid receptor TGR5 stimulates insulin secretion in pancreatic β cells. Biochem Biophys Res Commun. 2012;427(3):600–05. doi:10.1016/j.bbrc.2012.09.104.
  • Weber GJ, Foster J, Pushpakumar SB, Sen U. Altered microRNA regulation of short chain fatty acid receptors in the hypertensive kidney is normalized with hydrogen sulfide supplementation. Pharmacol res. 2018;134:157–65. doi:10.1016/j.phrs.2018.06.012.
  • Tomasova L, Dobrowolski L, Jurkowska H, Wrobel M, Huc T, Ondrias K, Ostaszewski R, Ufnal M. Intracolonic hydrogen sulfide lowers blood pressure in rats. Nitric Oxide. 2016;60:50–58. doi:10.1016/j.niox.2016.09.007.
  • Huc T, Jurkowska H, Wrobel M, Jaworska K, Onyszkiewicz M, Ufnal M. Colonic hydrogen sulfide produces portal hypertension and systemic hypotension in rats. Exp Biol Med (Maywood). 2018;243(1):96–106. doi:10.1177/1535370217741869.
  • Weber GJ, Pushpakumar S, Tyagi SC, Sen U. Homocysteine and hydrogen sulfide in epigenetic, metabolic and microbiota related renovascular hypertension. Pharmacol res. 2016;113:300–12. doi:10.1016/j.phrs.2016.09.002.
  • Tomasova L, Konopelski P, Ufnal M. Gut bacteria and Hydrogen Sulfide: the new old players in circulatory system Homeostasis. Molecules. 2016;21(11):1558. doi:10.3390/molecules21111558.
  • Battson ML, Lee DM, Weir TL, Gentile CL. The gut microbiota as a novel regulator of cardiovascular function and disease. J Nutr Biochem. 2018;56:1–15. doi:10.1016/j.jnutbio.2017.12.010.
  • Sun Y, Zhong S, Yu J, Zhu J, Ji D, Hu G, Wu C, Li Y, Singh UP. The aqueous extract of Phellinus igniarius (SH) ameliorates dextran sodium sulfate-induced colitis in C57BL/6 mice. Plos One. 2018;13(10):e0205007. doi:10.1371/journal.pone.0205007.
  • Grylls A, Seidler K, Neil J. Link between microbiota and hypertension: focus on LPS/TLR4 pathway in endothelial dysfunction and vascular inflammation, and therapeutic implication of probiotics. Biomed Pharmacother. 2021;137:111334. 10.1016/j.biopha.2021.111334
  • Toral M, Robles-Vera I, de la Visitacion N, Romero M, Yang T, Sanchez M, Gomez-Guzman M, Jimenez R, Raizada MK, Duarte J. CriticaL role of the interaction gut microbiota - Sympathetic nervous system in the regulation of blood pressure. Front Physiol. 2019;10:231. doi:10.3389/fphys.2019.00231.
  • Kim TT, Parajuli N, Sung MM, Bairwa SC, Levasseur J, Soltys CM, Wishart DS, Madsen K, Schertzer JD, Dyck JRB. Fecal transplant from resveratrol-fed donors improves glycaemia and cardiovascular features of the metabolic syndrome in mice. Am J Physiol Endocrinol Metab. 2018;315(4):E511–19. doi:10.1152/ajpendo.00471.2017.
  • Khalesi S, Sun J, Buys N, Jayasinghe R. Effect oF probiotics on blood pressure a systematic review and meta-analysis of randomized, controlled trials. Hypertension. 2014;64(4):897±. doi:10.1161/Hypertensionaha.114.03469.
  • Aoyagi Y, Park S, Matsubara S, Honda Y, Amamoto R, Kushiro A, Miyazaki K, Shephard RJ. Habitual intake of fermented milk products containing Lactobacillus casei strain Shirota and a reduced risk of hypertension in older people. Benef Microbes. 2017;8(1):23–29. doi:10.3920/Bm2016.0135.
  • Upadrasta A, Madempudi RS. Probiotics and blood pressure: current insights. Integr Blood Press Control. 2016;9:33–42. doi:10.2147/IBPC.S73246.
  • Robles-Vera I, Toral M, Romero M, Jimenez R, Sanchez M, Perez-Vizcaino F, Duarte J. Antihypertensive Effects of Probiotics. Curr Hypertens Rep. 2017;19(4):26. doi:10.1007/s11906-017-0723-4.
  • Sipola M, Finckenberg P, Santisteban J, Korpela R, Vapaatalo H, Nurminen ML. Long-term intake of milk peptides attenuates development of hypertension in spontaneously hypertensive rats. J Physiol Pharmacol. 2001;52(4 Pt 2):745–54.
  • Mizushima S, Ohshige K, Watanabe J, Kimura M, Kadowaki T, Nakamura Y, Tochikubo O, Ueshima H. Randomized controlled trial of sour milk on blood pressure in borderline hypertensive men. Am J Hypertens. 2004;17(8):701–06. doi:10.1016/j.amjhyper.2004.03.674.
  • Inoue K, Shirai T, Ochiai H, Kasao M, Hayakawa K, Kimura M, Sansawa H. Blood-pressure-lowering effect of a novel fermented milk containing γ-aminobutyric acid (GABA) in mild hypertensives. Eur J Clin Nutr. 2003;57(3):490–95. doi:10.1038/sj.ejcn.1601555.
  • Galla S, Chakraborty S, Cheng X, Yeo JY, Mell B, Chiu N, Wenceslau CF, Vijay-Kumar M, Joe B, Exposure to Amoxicillin in early life is associated with changes in gut microbiota and reduction in blood pressure: findings from a study on Rat Dams and Offspring. J Am Heart Assoc. 2020;9(2):e014373. doi:10.1161/JAHA.119.014373
  • Yoo HH, Kim IS, Yoo DH, Kim DH. Effects of orally administered antibiotics on the bioavailability of amlodipine: gut microbiota-mediated drug interaction. J Hypertens. 2016;34(1):156–62. doi:10.1097/HJH.0000000000000773.
  • Qi Y, Aranda JM, Rodriguez V, Raizada MK, Pepine CJ. Impact of antibiotics on arterial blood pressure in a patient with resistant hypertension - a case report. Int J Cardiol. 2015;201:157–58. doi:10.1016/j.ijcard.2015.07.078.
  • Galla S, Chakraborty S, Cheng X, Yeo J, Mell B, Zhang H, Mathew AV, Vijay-Kumar M, Joe B. Disparate effects of antibiotics on hypertension. Physiol Genomics. 2018;50(10):837–45. doi:10.1152/physiolgenomics.00073.2018.
  • Wang XH, Yang YN, Liang Y, Lang R, Zeng Q, Yan L, Yu RH, Wu CM. Structural modulation of gut microbiota during alleviation of experimental passive Heymann nephritis in rats by a traditional Chinese herbal formula. Biomed Pharmacother. 2022;145:112475. doi:10.1016/j.biopha.2021.112475.
  • Zhang F, Zhang X, Yu J, Tan Y, Guo P, Wu C. The gut microbiota confers the lipid-lowering effect of bitter melon (Momordica charantia L.) in high-fat diet (HFD)-Induced hyperlipidemic mice. Biomed Pharmacother. 2020;131:110667. doi:10.1016/j.biopha.2020.110667.
  • Wu C, Tian Y, Yu J, Zhang R, Zhang X, Guo P. The pandanus tectorius fruit extract (PTF) modulates the gut microbiota and exerts anti-hyperlipidaemic effects. Phytomedicine. 2019;58:152863. doi:10.1016/j.phymed.2019.152863.
  • Clarke SF, Murphy EF, O’sullivan O, Lucey AJ, Humphreys M, Hogan A, Hayes P, O’reilly M, Jeffery IB, Wood-Martin R, et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut. 2014;63(12):1913–20. doi:10.1136/gutjnl-2013-306541.
  • Barton W, Penney NC, Cronin O, Garcia-Perez I, Molloy MG, Holmes E, Shanahan F, Cotter PD, O’sullivan O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut. 2018;67:625–33. doi:10.1136/gutjnl-2016-313627.
  • Robles-Vera I, Toral M, de la Visitacion N, Sanchez M, Gomez-Guzman M, Munoz R, Algieri F, Vezza T, Jimenez R, Galvez J, et al. Changes to the gut microbiota induced by losartan contributes to its antihypertensive effects. Brit J Pharmacol. 2020;177(9):2006–23. doi:10.1111/bph.14965.
  • Xu XH, Yu XY, Jin H, Yan CL, Zhang QJ. The influence of Benazepril and Amlodipine on blood pressure and intestinal microbiom in spontaneously hypertensive rats. Chin J Microecol. 2020;32:379–83. doi:10.13381/j.cnki.cjm.202004002.
  • Yang YN, Deng YT, Zang CC, Zhang F, Huang ZB, Dong L, Lu WY, Zhang XP, Wu CM. The gut microbial Co-Abundance Gene Groups (CAGs) differentially respond to the flavor (Yao-Wei) of Chinese Materia Medica. Am J Chin Med. 2022;50(08):2223–44. doi:10.1142/S0192415X22500963.
  • Ma XC, Xiong XJ, Mo Y, Yue GH, Cai T, Huang J. Study on changes of intestinal microflora in Spontaneously Hypertensive Rats based on 16S rDNA sequencing and intervention of traditional Chinese Medicine. Chin Arch Tradit Chin Med. 2020; 38: 71-74+265-266. DOI:10.13193/j.issn.1673-7717.2020.08.017.
  • Yang Y, Cao S, Xu W, Zang C, Zhang F, Xie Y, Wu C. Dual modulation of gut bacteria and fungi manifests the gut-based anti-hyperlipidemic effect of Coptidis Rhizoma. Biomed Pharmacother. 2022;153:113542. doi:10.1016/j.biopha.2022.113542.
  • Wu J, Nakashima S, Nakamura S, Matsuda H. Effects of Sanoshashinto on left ventricular hypertrophy and gut microbiota in spontaneously hypertensive rats. J Nat Med. 2020;74(2):482–86. doi:10.1007/s11418-020-01387-9.
  • Han C, Jiang YH, Li W, Liu Y, Qi ZQ, Bai WZ. Study on the mechanism of Astragalus and Salvia miltiorrhiza on intestinal flora of spontaneously hypertensive rats based on 16S rDNA sequencing technology. China J Tradit Chin Med Pharm. 2019;34:2233–37. doi:10.1155/2019/5418796.
  • Qi YZ, Jiang YH, Jiang LY, Shao LL, Liu CY, Wang YG, Tao YN, Yang CH. Effects of Eucommia ulmoides-Tribulus terrestris on intestinal microbiota in aged spontaneously rats. Chin J Hypertens. 2019;27:454–62. doi:10.16439/j.cnki.1673-7245.2019.05.017.
  • Xiong MQ, Tang XT, Cui JG, Li LL, Tang XM, Wang PW, Chen Y, Zhang T. Effects and mechanisms of Qinggan Yishen Qufeng formula in the treatment against gut barrier Dysfunction in Hypertension. Modernization Tradit Chin Med Materia Medica World Sci Technol. 2019;21:1939–47. doi:10.11842/wst.20190129002.
  • Yu X, Zhang X, Jin H, Wu Z, Yan C, Liu Z, Xu X, Liu S, Zhu F. Zhengganxifeng decoction affects gut microbiota and reduces blood pressure via Renin–Angiotensin system. Biol Pharm Bull. 2019;42(9):1482–90. doi:10.1248/bpb.b19-00057.
  • Shi Y, Hu J, Geng J, Hu T, Wang B, Yan W, Jiang Y, Li J, Liu S. Berberine treatment reduces atherosclerosis by mediating gut microbiota in apoE-/- mice. Biomed Pharmacother. 2018;107:1556–63. doi:10.1016/j.biopha.2018.08.148.
  • Gautam A, Paudel YN, Abidin S, Bhandari U. Guggulsterone, a farnesoid X receptor antagonist lowers plasma trimethylamine-N-oxide levels: an evidence from in vitro and in vivo studies. Hum Exp Toxicol. 2019;38(3):356–70. doi:10.1177/0960327118817862.
  • Chen ML, Yi L, Zhang Y, Zhou X, Ran L, Yang J, Zhu JD, Zhang QY, Mi MT. Resveratrol Attenuates Trimethylamine-N-Oxide (TMAO)-Induced Atherosclerosis by regulating TMAO Synthesis and Bile Acid Metabolism via Remodeling of the Gut Microbiota. mBio. 2016;7(2):e02210–15. doi:10.1128/mBio.02210-15.
  • Zhou X, Liu J, Li JF. Effects and mechanism of quercetin on blood pressure,intestinal flora and ventricular remodeling in spontaneously hypertensive rats. Nat Prod Res Dev. 2020;32:1449–55. doi:10.16333/j.1001-6880.2020.9.001.
  • Han C, Jiang YH, Li W, Liu Y, Qi ZQ. Study on the Antihypertensive mechanism of Astragalus membranaceus and Salvia miltiorrhiza based on Intestinal Flora-Host Metabolism. Evid Based Complement Alternat Med. 2019;2019:5418796. doi:10.1155/2019/5418796.