Gut microbiota-dependent phenylacetylglutamine in cardiovascular disease: current knowledge and new insights

Yaonan Song , Haoran Wei , Zhitong Zhou , Huiqing Wang , Weijian Hang , Junfang Wu , Dao Wen Wang

Front. Med. ›› 2024, Vol. 18 ›› Issue (1) : 31 -45.

PDF (1265KB)
Front. Med. ›› 2024, Vol. 18 ›› Issue (1) : 31 -45. DOI: 10.1007/s11684-024-1055-9
REVIEW

Gut microbiota-dependent phenylacetylglutamine in cardiovascular disease: current knowledge and new insights

Author information +
History +
PDF (1265KB)

Abstract

Phenylacetylglutamine (PAGln) is an amino acid derivate that comes from the amino acid phenylalanine. There are increasing studies showing that the level of PAGln is associated with the risk of different cardiovascular diseases. In this review, we discussed the metabolic pathway of PAGln production and the quantitative measurement methods of PAGln. We summarized the epidemiological evidence to show the role of PAGln in diagnostic and prognostic value in several cardiovascular diseases, such as heart failure, coronary heart disease/atherosclerosis, and cardiac arrhythmia. The underlying mechanism of PAGln is now considered to be related to the thrombotic potential of platelets via adrenergic receptors. Besides, other possible mechanisms such as inflammatory response and oxidative stress could also be induced by PAGln. Moreover, since PAGln is produced across different organs including the intestine, liver, and kidney, the cross-talk among multiple organs focused on the function of this uremic toxic metabolite. Finally, the prognostic value of PAGln compared to the classical biomarker was discussed and we also highlighted important gaps in knowledge and areas requiring future investigation of PAGln in cardiovascular diseases.

Keywords

PAGln / cardiovascular disease / gut microbiota / uremic metabolite / biomarker

Cite this article

Download citation ▾
Yaonan Song, Haoran Wei, Zhitong Zhou, Huiqing Wang, Weijian Hang, Junfang Wu, Dao Wen Wang. Gut microbiota-dependent phenylacetylglutamine in cardiovascular disease: current knowledge and new insights. Front. Med., 2024, 18(1): 31-45 DOI:10.1007/s11684-024-1055-9

登录浏览全文

4963

注册一个新账户 忘记密码

1 Introduction

The gut microbiota is described as the “microbial organ” of the human body, which participates in the host’s energy metabolism, regulates innate and acquired immunity, and is regarded as the biological barrier of the human intestine [1]. Recently, two “back-to-back” randomized controlled trials found that the occurrence of cardiovascular disease is closely related to changes in the host intestinal microbiota [2], and intestinal microbiota dysbiosis often appears earlier than the clinical manifestations [3], confirming that intestinal microbial imbalance may be the initiation of cardiovascular diseases. Such intestinal imbalance could promote cholesterol accumulation [4], oxidative stress, and inflammation [5], while probiotics modulation showed protective effects in cardiovascular disease [6], implying the gut microbiota is a significant contributor to cardiovascular homeostasis.

The gut microbiome and its related metabolites have been reported to be involved in cardiovascular disease [7] since the epoch-making studies about trimethylamine-N-oxide (TMAO) [4,8]. The levels of TMAO were found closely associated with cardiovascular disease risk because TMAO enhances cholesterol accumulation in macrophages, increases cell surface expression of proatherogenic scavenger receptors [4], prolongs hypertensive effects of angiotensin II, as well as enhances platelet activation [9], which may contribute to platelet hyperresponsiveness and thrombosis. Besides, other gut microbial-host co-metabolites, such as bile acids, short-chain fatty acids, and tryptophan-related metabolites also served as essential mediators between gut microbiota and the cardiovascular system. For example, the elevated bile acids [10] and indole-sulfate [11] showed a close link with the occurrence of cardiovascular disease risk. On the contrary, the short-chain fatty acid propionate could ameliorate vascular calcification by intestinal microbiota Akkermansia muciniphila remodeling [12] as well as the indole-3-carboxaldehyde alleviated atherosclerosis by reducing ROS levels and inflammatory factors in endothelial cell [13]. Furthermore, the short-chain fatty acid acetate and propionate were reported to lower the heart rate and cardiac contractility along with blood pressure in animal models [14] or multi-center clinical trials [15], implying the role of microbial-related metabolite in the physiological regulation of the cardiovascular system.

Phenylacetylglutamine (PAGln) is a newly gut microbiome-related metabolite reported by Nemet et al. in a cohort with cardiovascular disease from an untargeted metabolomics profiling platform. The increased levels of PAGln were associated with major adverse cardiovascular events (MACEs) such as myocardial infarction, stroke, or death in the population [16]. Recently, a series of studies found an association between PAGln and cardiovascular diseases, such as heart failure, coronary heart disease, and atrial fibrillation. Here we reviewed the metabolic pathway of PAGln, the role of gut microbiota-dependent PAGln in several common clinical cardiovascular diseases and its possible mechanism. On the other hand, since the production of PAGln across multi-organs such as the intestine, liver, heart, and kidney, we also discussed the crosstalk among different organs of this uremic metabolite and explored possible preventive ways for cardiovascular disease by PAGln modulation. Finally, we highlighted important gaps currently in knowledge and areas requiring future investigation of PAGln in cardiovascular diseases.

2 Production and measurement of PAGln

2.1 The metabolic pathway of PAGln production

The PAGln comes from the essential amino acid, phenylalanine [17]. Since the human body cannot synthesize phenylalanine itself, phenylalanine is mainly dependent on food intake and is absorbed in the small intestine [18]. Previous studies have found that amino acid catabolism associated with proteolysis is significantly increased due to higher intake and consumption of cardiac energy substances in heart failure conditions [19]. In addition to its involvement in energy metabolism, the unabsorbed phenylalanine is then metabolized in the large intestine by the gut microbiota to produce phenylpyruvic acid [20] (Fig.1). There are two distinct gut microbial pathways for conversion from phenylpyruvic acid into phenylacetic acid in the presence of Christensenellaceae, Lachnospiraceae, and Ruminococcaceae [21]. One is catalyzed by phenylpyruvate ferredoxin oxidoreductase to form phenylacetyl-CoA. The other is catalyzed by phenylpyruvate decarboxylase to form phenylacetaldehyde [22]. Further microbial colonization in germ-free mice found that the microbial porA gene is associated with PAGln production which favored the conversion of phenylpyruvate to phenylacetic acid [16]. Then the phenylacetic acid enters the liver through the portal system, conjugating with glutamine to form PAGln in humans and with glycine to form phenylacetylglycine (PAGly) in rodents [22].

PAGln is a normal constituent of human urine that is excreted from the kidney primarily as tubular secretion [23,24]. The PAGln clearance is greatly reduced when glomerular filtration is impaired (as measured by eGFR) [24,25] and usually, is accumulated in the plasma of uremic patients [26,27]. Such accumulation is considered as an early marker of renal dysfunction previously [28] and is applied to predict clinical endpoints in patients with chronic kidney disease [29]. Furthermore, PAGln was also known as the main product of the treatment of acute/severe hepatic encephalopathy [30,31]. The urinary PAGln can be used as a non-invasive biomarker to facilitate daily monitoring of phenylalanine metabolism in phenylketonuria patients [32] and to set up as an index for monitoring “non-invasive chemical biopsy of human liver” and is assumed to reflect liver citric acid cycle intermediates [33,34].

2.2 Measurement of PAGln

The PAGln is an amino acid derivative that can be measured by several analytical techniques including nuclear magnetic resonance (NMR), liquid chromatograph-mass spectrometer (LC-MS), and gas chromatograph-mass spectrometer (GC-MS) (Tab.1).

In fact, PAGln is not a newly-discovered metabolite. Shockcor et al. reported that urinary PAGln is a typical uremic toxin under the existence of gut microbiota since the 1990s by the NMR platform [35]. Because the level of urinary PAGln is relatively high (34–47 μM/mM creatinine) [23], it is easy to measure based on the NMR-dominated untargeted metabolic platform previously. Fukui et al. identified urinary PAGln as a novel marker of interstitial cystitis [36], which indicated the disturbance of the gut microbiome in these patients. Compared to the mass spectrometer, the sensitivity of NMR platform is relatively low even though it possesses the advantage of simple sample preparation and high throughput [37,38]. Hence, it is hard to quantify the level of PAGln in plasma on the NMR platform because of its relatively lower concentration.

Several research groups tried to quantify the level of PAGln in blood plasma/serum samples with the aid of a state-of-the-art mass spectrometry platform [39] since the level of plasma PAGln is less than ten percent of that in urine. Usually, the physiologic median value of PAGln level in plasma is 0.8 μM in healthy control subjects and 2.0 μM in heart failure patients [16,40]. With the aid of the deuterated metabolite analog, the PAGln could be quantified accurately and its measurement stability was verified in the LC-MS/MS platform [40,41]. Besides, the levels of PAGln could also be measured by another analytical technique, gas chromatography mass spectrometry [42]. However, because PAGln is not a commonly volatile organic compound, this method is seldom used by other groups.

In addition to the analytical method, the ELISA measurement of PAGln has also been reported in recent studies. The levels of PAGln reported from ELISA studies [43,44] are relatively lower when compared with studies measured by the LC-MS/MS platform. It might be related to the wide coverage range and fewer exogenous interferences of LC-MS/MS, with the selective multiple-reaction monitoring capable of choosing the specific ions [45]. However, despite these technique differences, the scientific conclusion was not affected significantly when comparing the level of PAGln between the cardiovascular disease group and healthy controls. The LC-MS/MS platform takes advantage of high precision and high coverage range for PAGln measurement. That is also why the LC-MS/MS platform is the most widely used analytical tool for PAGln measurement clinically.

Except for the plasma and urine, the level of PAGln in the saliva has also been explored recently by a French group. The saliva PAGln level is approximately ten percent of its plasma concentration by LC-MS/MS platform [46]. In a small healthy volunteer cohort, Fabresse et al. found the PAGln level in saliva was not correlated with the PAGln level in serum. Such inconsistency might be due to the degradation by oral microbiota [47] or lack of organic anion transmembrane transport from the blood to saliva [48].

3 Role of PAGln in cardiovascular disease based on epidemiological evidence

In Nemet’s studies, they found that the increased levels of PAGln showed prognostic value in MACEs of those patients undergoing routine cardiac evaluation. That is, the incidences of the MACEs are not specified to which kind of cardiovascular disease [16,49]. The epidemiological evidence from other different cardiovascular disease cohorts showed the diagnosis or prognostic value of PAGln in heart failure, coronary heart disease, atrial fibrillation, and atherosclerosis (Tab.2).

3.1 Heart failure

Heart failure is a heterogeneous disease caused by many different etiologies [50]. Recently, the understanding of heart failure progression has been further enhanced by the aspect of “gut hypothesis” [51,52]. That is, the reduced cardiac output causes ischemic and/or edematous change in the gut, leading to translocation of the gut microbiota and increased production of circulating gut-microbiota-related metabolites and endotoxins, which in turn leads to further damage to the cardiovascular system [15,16]. As a consequence, the dynamic levels of circulating microbial-related metabolites possess the potential to reflect the role of gut microbiota in the development of heart failure [53].

Relatively few studies on PAGln and heart failure are currently available. In a prospective study of 1744 African Americans, a variety of metabolites including PAGln were screened for risk of heart failure using an untargeted metabolomics approach [54]. In a small population-based study, plasma levels of PAGln were higher in patients with heart failure-related death or re-hospitalization within one year than those in the non-event group in acute decompensated heart failure [55]. Several clinical studies based on multi-center cohorts of heart failure with reduced ejection fraction showed that elevated PAGln level is an independent risk factor for heart failure and is associated with an increased risk of cardiovascular mortality [56,57]. Considering that many of these studies were based on non-targeted methods and relatively small sample sizes, the plasma PAGln in patients with heart failure using targeted strategy LC/MS-MS in a larger cohort to date was measured in our recent studies [40]. On the other hand, because of the different causes of main subtypes of heart failure, that is, heart failure with reduced EF (HFrEF) and heart failure with preserved EF (HFpEF), it is still unknown whether the role of PAGln in the heart failure subtypes could be different or not from previous studies. Our results confirmed that PAGln is an independent risk predictor for cardiovascular events in different subtypes of heart failure (including HFrEF and HFpEF). The level of PAGln is highly associated with deleterious cardiac function which was evaluated by the New York Heart Association class, especially for the increasing risk of cardiovascular death or hospital readmission, regardless of HFpEF and HFrEF [40]. This is the first study that manifested the role of PAGln in heart failure patients with preserved ejection fraction.

3.2 Coronary heart disease/atherosclerosis

The level of PAGln is reported to be associated with coronary heart disease/atherosclerosis patients in the Asian [58] or Western [59] population. In two independent prospective cohorts, Ottosson et al. found that PAGln was associated with an increased risk of coronary heart disease and was independent of other cardiovascular factors [60]. Recently the increased PAGln is also reported in lower arterial stiffness women patients. The carotid-femoral pulse wave velocity, a measure of arterial stiffness, was associated with the gut-microbiome derived PAGln and they found that the microbiome factors could explain 8.3% of the variance in arterial stiffness [61]. Yu et al. showed that plasma PAGln levels are not only higher in patients with ischemic stroke than in healthy subjects [62] but also associated with increased white matter hyperintensity burden in patients with acute ischemic stroke [63]. Furthermore, serum PAGln level is also elevated in patients with peripheral artery disease compared with non-peripheral artery controls [64]. When considering the coronary computed tomographic angiography together, Liu et al. found that the level of PAGln is independently correlated with the coronary atherosclerotic burden among patients with suspected coronary artery disease [58]. For those coronary artery patients undergoing percutaneous transluminal coronary intervention, Fu et al. found that the increased levels of PAGln could distinguish the coronary in-stent restenosis group from the in-stent patency in a small cohort of coronary artery disease. But it is not helpful for differentiating the in-stent hyperplasia group with a marginal statistical P value (P = 0.094), which might be due to the small sample size [65]. These results suggested that PAGln is a valuable index for predicting coronary in-stent restenosis and gut microbes may be a promising intervention target to prevent its progression. Such dysbiosis of microbial diversity and composition was confirmed by the fecal 16S rRNA sequencing in coronary artery disease patients with stent stenosis. The significant enrichment of Roseburia, Blautia, and Ruminococcus was enhanced in coronary artery disease with in-stent stenosis when compared with in-stent intimal hyperplasia patients [43].

Except for the common coronary artery disease patients, the gut-derived PAGln is also helpful for differentiating the explainable and unexplainable atherosclerosis patients. In a nested case-control study (316/3056 patients) of atherosclerosis, Bogiatze et al. found increased PAGln in the atherosclerosis patients with explained reasons when compared to the controls. Interestingly, for those unexplainable patients, that is, 5% with extremely low levels of known risk factors but with a very high plaque burden, the concentration of PAGln is also higher than the patients with explainable reasons. It is worth noting that there were no significant differences in PAGln precursors (phenylalanine and glutamine) among the comparable three groups (control, explainable, and unexplainable atherosclerosis patients). It implies the disturbance of the gut microbiome that contributes to the residual risk of atherosclerosis [66].

3.3 Cardiac arrhythmia

In recent decades, intricate impacts of the gut microbiome on cardiac arrhythmia have been identified as prospective approaches for its prevention, development, treatment, and prognosis. The direct evidence comes from the metagenomic and metabolomic profiling of matched atrial fibrillation patients. Except for the increased relative abundance of porA, the dramatic elevation in microbial diversity and overgrowth of Ruminococcus, Streptococcus, and Enterococcus, as well as reduction of Faecalibacterium, Alistipes, Oscillibacter, and Bilophila were detected in both fecal and serum samples in patients with atrial fibrillation [67]. A series of gut microbial-related metabolites, such as TMAO, indoxyl sulfate, and primary bile acids have also been proven to mediate molecular pathways linked to atrial fibrillation [68]. The association between newly gut-microbial related PAGln and atrial fibrillation was explored in 92 patients. Plasma PAGln levels were higher in atrial fibrillation patients and its increased levels were correlated with the left atrial diameter. The increased PAGln exerted a predictive potential of atrial fibrillation, and the combination of PAGln and atrial fibrillation score helps improve the predictive model [44].

Except for atrial fibrillation, ventricular arrhythmias are more dangerous caused by an electrical malfunction, which can drastically reduce blood flow to the rest of the body. In a thoracic aortic coarctation-induced ventricular arrhythmias model, Fu et al. found that PAGln deteriorated the susceptibility of ventricular arrhythmias [69], which suggests the role of PAGln in the modulation of cardiac arrhythmia.

4 Potential mechanisms of PAGln in CVD

4.1 Adrenergic receptor mediated thrombosis

Along with the findings that the increased PAGln was associated with MACEs from all patients without specific cardiovascular etiology, Nemet et al. proposed a possible mechanism that PAGln could promote the activation of platelets and thrombosis [16]. According to multiple genetic and pharmacology studies, PAGln activates platelet responsiveness and thrombosis potential through G-protein coupled receptors (including α2A, α2B, and β2-adrenergic receptors) in both cellular and animal models (Fig.2). In the FeCl3-induced carotid artery injury model, intraperitoneal injection of PAGln/PAGly (the PAGln analog in rodent mice) promotes platelet thrombus formation and reduces the time to cessation, while the use of β-blocker carvedilol reverses this pro-thrombotic effect [16]. Since most previous studies focused on the correlation between the level of PAGln and cardiovascular diseases in the view of epidemiology, the potential mechanisms are relatively few. This work by Nemet et al. not only confirms that PAGln is a new intestinal flora-dependent metabolite associated with cardiovascular disease but also leads the study of the association of intestinal flora in the field of cardiovascular disease to causal-orientated research [70].

It is worth noting that in recent studies, the interaction effects between the level of PAGln and the use of β-blocker were not significant in a relatively large heart failure population [40]. For comparing the effects of β-blocker in the different quartiles of PAGln level to those without the usage of β-blocker, the Kaplan–Meier plots show that the usage of β-blocker takes benefits for heart failure patients, regardless of each stratified PAGln quartile. That means the presence or absence of β-blocker did not affect the relationship between PAGln and heart failure prognosis in the view of epidemiology [40]. These results also indicate that there might be more complex underlying mechanisms except for the adrenergic receptor-mediated thrombosis in the disease development of heart failure.

4.2 Inflammatory responses

Previously, the classical microbiome derived metabolite TMAO has been found to activate the NLR family pyrin domain containing 3 inflammasome-related inflammatory pathways [71,72] and exacerbate cardiac fibrosis in the carotid artery ligation mouse model [72,73]. The other gut microbial derived indoxyl sulfate increased the expression of pro-inflammatory and pro-fibrotic signaling molecules and oxidative stress in atrial fibrillation patients [74], which revealed the role of microbial related metabolites in promoting inflammatory responses in cardiovascular diseases. Here the administration of PAGln was also reported to deteriorate myocardial inflammatory cell infiltration and increase the inflammatory response by activating Toll-like receptor 4 and its downstream protein kinase and mammalian target of the rapamycin signal pathway [69], implying the activated inflammatory responses by PAGln. Actually, the inflammatory response index was found promoted in the thoracic aortic coarctation induced heart failure mouse model, as well as in the heart failure patients [75], but the gut microbial related PAGln could deteriorate such inflammatory responses. Such elevated inflammatory indices (such as IL-1β and IL-6) induced by PAGln were also confirmed in isoprenaline stimulated neonatal rat cardiac myocytes [40]. These above results imply that the increased inflammatory response and activated immune systems induced by PAGln contribute to the disease progression of cardiovascular disease.

4.3 Protective effects of PAGln

Even elevated PAGln was found to increase the predictive value of cardiovascular event risk in many cardiovascular clinical cohorts. However, there are also some preliminary basic studies suggesting potential protective effects of PAGln/PAGly under certain conditions. For example, Xu et al. found that the analog of PAGln, PAGly, activates the Gαi/PI3K/AKT anti-apoptotic signaling pathway via B2AR and reduces hypoxia/reoxygenation injury in neonatal mouse cardiomyocytes [76]. Administration of a moderate dosage of PAGly to mice with hypoxia/reoxygenation injury reduced the size of the myocardial infarct zone, but high dosage of PAGln failed to have this effect, which may be related to the heterogeneity within the high-dosage group [76]. Another in vitro study demonstrated the anti-inflammatory properties of PAGln in mouse spleen cells and peritoneal cavity cells [77] by increasing ROS production, reducing cell activity, and increasing apoptosis in HL-1 cells [78]. Therefore, more rigorous clinical studies and deeper mechanistic studies are necessary to explore the relationship between PAGln and cardiovascular diseases in the future.

4.4 Inter-organ communication in PAGln uremic toxicity

As previously mentioned, the PAGln is produced from protein metabolism and influenced by colonic microbiota and enterohepatic circulation [49]. This gut-derived low molecular weight uremic toxin [79] is removed by glomerular filtration in the kidney. Generally, the kidney-intestinal cross-talk involves intestinal epithelial damage, dysbiosis, and generation of uremic toxins. Recently, Zhong et al. revealed that kidney injury expands the intestinal lymphatics, which is a route for transporting uremic toxins and alters the composition of mesenteric lymph in kidney disease [80].

On the other hand, these small molecules could be transported into circulation across the barrier in the intestine, the heart, and the kidney and play roles in inter-organ and inter-organism communication [81]. From a perspective of the remote sensing and signaling theory of these gut derived uremic toxins, Glorieux et al. raised a point that these uremic toxins including PAGln, are critical for the maintenance of homeostasis along the gut–heart–kidney axis and for responding to homeostatic perturbations [81]. The remote sensing and signaling theory has been used to understand the roles of transporters, enzymes, and kinases in inter-organ and inter-organism communication via small endogenous molecules [82,83]. For example, these uremic toxins including PAGln have been demonstrated as fibrogenic and prohypertrophic effects [84] as well as aortic calcification, endothelial cell dysfunction, and tubular damage in the kidneys, indicating their roles on the heart, vessels, and kidneys. Furthermore, this inter-organ crosstalk was also confirmed by gastrointestinal and hepatic involvement in heart failure patients [85]. In our previous studies, the increased level of PAGln showed a close correlation with creatinine in heart failure patients [40]. The close link between gut–heart–kidney crosstalk reminds us not to put this gut-derived metabolite PAGln as a biomarker only specific for cardiovascular or kidney diseases. The disturbance of PAGln could be modulated from each view of the gut, heart, and kidney. In other words, even if the potential mechanism was poorly revealed, those individuals with relatively normal renal function or normal intestinal activity might be at a higher risk of cardiovascular events due to the elevated PAGln and hence should be given sufficient attention. On the contrary, as Drs. Spence and Urquhart suggested, the high cardiovascular disease risk in chronic kidney disease patients also requires consideration of therapies due to this kind of gut-derived uremic metabolite [86].

5 Gaps in knowledge and future directions

5.1 Comparison between PAGln and classical biomarkers in cardiovascular diseases

Since PAGln has proven to be a potential biomarker that possesses the value in diagnosis and prognosis of cardiovascular disease, its comparison with the traditional biomarker could not be ignored. The highly sensitive cardiac troponin (hs-cTnI) and N-terminal proB-type natriuretic peptide (NT-proBNP) are classical biomarkers in managing ischemic cardiomyopathy [87,88] and heart failure [89]. The level of NT-proBNP takes the priority that is recommended in the diagnosis and prognosis of heart failure by the American Heart Association [89]. Due to the recruited sample numbers of the heart failure cohort being small and the lack of follow-up records, previous studies related to the interaction between PAGln and NT-proBNP are relatively few. On the other hand, most of the previous studies focused on typical heart failure whose ejection fraction is less than 40% (HFrEF). In a small sample size cohort, Tang suggested that four metabolites including PAGln could increase the predictive value of BNP in heart failure patients [55]. Recently, in our group including 3152 heart failure patients, we stratified the heart failure patients based on the levels of PAGln and NT-proBNP, respectively. The group with the highest quartile and PAGln and NT-proBNP demonstrated the highest risks for getting major adverse cardiac events, compared to a reference group with the lowest quartile of PAGln and NT-proBNP, confirming the joint effects of PAGln and NT-proBNP in a larger population [40]. Besides, when the reference was set to group with its first quartile of NT-proBNP, the PAGln showed predictive value for a group with lower (or normal) NT-proBNP, especially in heart failure patients with preserved ejection fraction, implying the complementary effects of PAGln to the classical index NT-proBNP. Our studies got editorial comments for highlighting their potential application in the prognostic value of cardiovascular death or hospital readmission in HFpEF patients [90].

The consequent question is why the PAGln possesses more predictive value in heart failure with normal NT-proBNP. The main causes of HFpEF are cardiac metabolic factors, such as higher body mass index (BMI), hypertension, diabetes, and obesity. The BMI index was inversely associated with the level of PAGln not only in total HF patients but also in different subtypes of HF, i.e., HFpEF and HFrEF in our previous studies. Such results agreed with findings from a recent study by Romano et al. [91] and urinary PAGln in the human adiposity population [92]. Considering that obesity is the main cause of HFpEF, it was believed that low BNP levels were more commonly seen in HFpEF because of two possible reasons. The first is that androgens produced by adipose tissue could inhibit the expression of the natriuretic peptide precursor B gene, resulting in reduced release of BNP [93]. On the other hand, it may be related to the abundance expression of natriuretic peptide receptors on adipocytes, which increases the clearance of BNP and leads to a decrease in BNP levels [94]. Therefore, obesity should be an important mediator of heart failure, especially for HFpEF patients.

Furthermore, except for BMI, the increased level of PAGln is also related to the level of glucose [95]. The relationship between PAGln and T2DM and its complications has been corroborated by a series of subsequent modest clinical studies [96]. PAGln was positively correlated with the severity of T2DM after adjusting for sex and age [41]. Among diabetic patients, those who progressed to distal symmetric polyneuropathy [97], diabetic nephropathy [98], and diabetic retinopathy [99] had even further elevated levels of PAGln in the circulation. In contrast, Hua et al. reported the increased PAGln associated with chronic kidney diseases and heart failure, but not diabetes in their cohort [96]. These studies suggested that metabolic syndrome could not be ignored when evaluating the role of PAGln in cardiovascular diseases.

5.2 Modulation of PAGln

Usually, the classical intervention ways that modulate the gut ecosystem include diet [100,101], microbial modification (such as facel microbial transplantation and probiotics modulation), and specific inhibitors [102,103]. For example, a high-protein diet did not affect serum TMAO levels whereas PAGln was significantly elevated in an elderly woman cohort, which suggests that the effect of dietary intake of different components on TMAO/PAGln in human circulation may differ [104]. A recent study shows that PAGln may be involved in the regulation of blood pressure by the Dietary Approaches to Stop Hypertension Diet (DASH) [105]. That is because the dietary fibers in these diets which are regarded as common prebiotics, were not absorbed by the upper gastrointestinal tract and utilized by colonic microbiota. Kaye et al. found that dietary deficiency of prebiotic fiber led to increased blood pressure and pathological myocardial remodeling in mouse models [106], which could be reversed by high fiber diet treatment [107] or green tea containing the precursor of PAGln [108].

Tang et al. provided alternative ideas to modulate the gut microbiota directly. For example, the antibiotic-treated mice had significantly increased mortality after infarction, and probiotic supplementation or fecal transplantation was able to repair damaged myocardium via SCFAs [5]. The use of antibiotics by modulating gut microbiota for the treatment of cardiovascular diseases has become a major research interest. The use of antibiotics in rats with myocardial infarction was able to modulate microbial metabolites and reduce the size of the infarction part [109]. In animal models of HF, antibiotic administration reduced plasma TMAO levels, attenuated myocardial hypertrophy, and fibrosis [110], and exerted a protective effect [111]. However, the usage of antibiotics has to be carefully considered, because the gut microbiota is involved in normal physiologic processes, and its potential for new dysbiosis and resistance is an unavoidable problem [112]. Beyond this, a 10-year CLARICOR RCT study also showed that clarithromycin administration in patients with stable heart disease increased cardiovascular mortality [113].

Thirdly, the specific inhibitors, such as the choline analog (3,3-dimethylbutanol [114] or fluoromethycholine [103]) for inhibiting choline trimethylamine lyase activity and β-blocker carvedilol for inhibiting the adrenergic receptor in PAGln production, were used in previous studies [16]. However, further clinical application still needs to be carried out based on the safety and reliability of human studies. Besides, based on the uremic toxicity of PAGln, Fatani et al. found that hemodialysis could be a useful tool for lowering the levels of PAGln in those patients with high uremic metabolites. But in this case, the extra fiber supplementation does not modulate the microbial-generated uremic molecules in a randomized controlled trial [115].

In a word, even though the close association of PAGln in cardiovascular diseases, as well as the dietary intervention, showed promising application in the regulation of microbial metabolites, there is still a lack of specific way or clear dietary guideline for lowering the level of this uremic toxin metabolite [116]. Consequently, the modulation based on the level of PAGln remains a long way off.

6 Conclusions

The new microbial metabolite PAGln has provided ideas for the study of the relationship between gut microbiota and cardiovascular diseases. With the development of metabolomics and metagenomics, the microbial metabolites not only serve as biomarkers to help clinicians identify high-risk cardiovascular patients for timely prevention and treatment but also provide promising targets for interventions based on the existing treatment system, such as precise dietary interventions, the use of antibiotics and probiotics.

Considering the complexity of the gut microbiota and its associated metabolites, there are still a lot of gaps that need to be filled in the current stage of research. The current findings lack more evidence from multi-center clinical trials, especially for interventional studies on the regulation of gut microbiota and metabolites. Secondly, the specific mechanisms by which microbial metabolites affect cardiovascular disease remain to be elucidated. There is also a lack of clear dietary or microbial guidelines associated with PAGln for the treatment of cardiovascular diseases. Furthermore, the cross-talk of this uremic toxicity among multi-organs in cardiovascular or kidney disease needs further investigation. The collaboration of public health scientists, biologists, and clinicians is the only way to overcome these challenges, and we expect that research on PAGln and its related microbial metabolite will make a greater contribution to the prevention and treatment of cardiovascular diseases.

References

[1]

Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science 2005; 307(5717): 1915–1920

[2]

Fromentin S, Forslund SK, Chechi K, Aron-Wisnewsky J, Chakaroun R, Nielsen T, Tremaroli V, Ji B, Prifti E, Myridakis A, Chilloux J, Andrikopoulos P, Fan Y, Olanipekun MT, Alves R, Adiouch S, Bar N, Talmor-Barkan Y, Belda E, Caesar R, Coelho LP, Falony G, Fellahi S, Galan P, Galleron N, Helft G, Hoyles L, Isnard R, Le Chatelier E, Julienne H, Olsson L, Pedersen HK, Pons N, Quinquis B, Rouault C, Roume H, Salem JE, Schmidt TSB, Vieira-Silva S, Li P, Zimmermann-Kogadeeva M, Lewinter C, Søndertoft NB, Hansen TH, Gauguier D, Gøtze JP, Køber L, Kornowski R, Vestergaard H, Hansen T, Zucker JD, Hercberg S, Letunic I, Bäckhed F, Oppert JM, Nielsen J, Raes J, Bork P, Stumvoll M, Segal E, Clément K, Dumas ME, Ehrlich SD, Pedersen O. Microbiome and metabolome features of the cardiometabolic disease spectrum. Nat Med 2022; 28(2): 303–314

[3]

Talmor-Barkan Y, Bar N, Shaul AA, Shahaf N, Godneva A, Bussi Y, Lotan-Pompan M, Weinberger A, Shechter A, Chezar-Azerrad C, Arow Z, Hammer Y, Chechi K, Forslund SK, Fromentin S, Dumas ME, Ehrlich SD, Pedersen O, Kornowski R, Segal E. Metabolomic and microbiome profiling reveals personalized risk factors for coronary artery disease. Nat Med 2022; 28(2): 295–302

[4]

Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung YM, Wu Y, Schauer P, Smith JD, Allayee H, Tang WH, DiDonato JA, Lusis AJ, Hazen SL. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011; 472(7341): 57–63

[5]

Tang TWH, Chen HC, Chen CY, Yen CYT, Lin CJ, Prajnamitra RP, Chen LL, Ruan SC, Lin JH, Lin PJ, Lu HH, Kuo CW, Chang CM, Hall AD, Vivas EI, Shui JW, Chen P, Hacker TA, Rey FE, Kamp TJ, Hsieh PCH. Loss of gut microbiota alters immune system composition and cripples postinfarction cardiac repair. Circulation 2019; 139(5): 647–659

[6]

Ettinger G, MacDonald K, Reid G, Burton JP. The influence of the human microbiome and probiotics on cardiovascular health. Gut Microbes 2014; 5(6): 719–728

[7]

Shalon D, Culver RN, Grembi JA, Folz J, Treit PV, Shi H, Rosenberger FA, Dethlefsen L, Meng X, Yaffe E, Aranda-Díaz A, Geyer PE, Mueller-Reif JB, Spencer S, Patterson AD, Triadafilopoulos G, Holmes SP, Mann M, Fiehn O, Relman DA, Huang KC. Profiling the human intestinal environment under physiological conditions. Nature 2023; 617(7961): 581–591

[8]

Tang WH, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med 2013; 368(17): 1575–1584

[9]

Zhu W, Gregory JC, Org E, Buffa JA, Gupta N, Wang Z, Li L, Fu X, Wu Y, Mehrabian M, Sartor RB, McIntyre TM, Silverstein RL, Tang WHW, DiDonato JA, Brown JM, Lusis AJ, Hazen SL. Gut microbial metabolite TMAO enhances platelet hyperreactivity and thrombosis risk. Cell 2016; 165(1): 111–124

[10]

Charach G, Rabinovich A, Argov O, Weintraub M, Rabinovich P. The role of bile acid excretion in atherosclerotic coronary artery disease. Int J Vasc Med 2012; 2012: 949672

[11]

Cao XS, Chen J, Zou JZ, Zhong YH, Teng J, Ji J, Chen ZW, Liu ZH, Shen B, Nie YX, Lv WL, Xiang FF, Tan X, Ding XQ. Association of indoxyl sulfate with heart failure among patients on hemodialysis. Clin J Am Soc Nephrol 2015; 10(1): 111–119

[12]

Yan J, Pan Y, Shao W, Wang C, Wang R, He Y, Zhang M, Wang Y, Li T, Wang Z, Liu W, Wang Z, Sun X, Dong S. Beneficial effect of the short-chain fatty acid propionate on vascular calcification through intestinal microbiota remodelling. Microbiome 2022; 10(1): 195

[13]

Lu Y, Yang W, Qi Z, Gao R, Tong J, Gao T, Zhang Y, Sun A, Zhang S, Ge J. Gut microbe-derived metabolite indole-3-carboxaldehyde alleviates atherosclerosis. Signal Transduct Target Ther 2023; 8(1): 378

[14]

Poll BG, Xu J, Jun S, Sanchez J, Zaidman NA, He X, Lester L, Berkowitz DE, Paolocci N, Gao WD, Pluznick JL. Acetate, a short-chain fatty acid, acutely lowers heart rate and cardiac contractility along with blood pressure. J Pharmacol Exp Ther 2021; 377(1): 39–50

[15]

Verhaar BJH, Collard D, Prodan A, Levels JHM, Zwinderman AH, Bäckhed F, Vogt L, Peters MJL, Muller M, Nieuwdorp M, van den Born BH. Associations between gut microbiota, faecal short-chain fatty acids, and blood pressure across ethnic groups: the HELIUS study. Eur Heart J 2020; 41(44): 4259–4267

[16]

Nemet I, Saha PP, Gupta N, Zhu W, Romano KA, Skye SM, Cajka T, Mohan ML, Li L, Wu Y, Funabashi M, Ramer-Tait AE, Naga Prasad SV, Fiehn O, Rey FE, Tang WHW, Fischbach MA, DiDonato JA, Hazen SL. A cardiovascular disease-linked gut microbial metabolite acts via adrenergic receptors. Cell 2020; 180(5): 862–877.e22

[17]

Seakins JW. The determination of urinary phenylacetylglutamine as phenylacetic acid. Studies on its origin in normal subjects and children with cystic fibrosis. Clin Chim Acta 1971; 35(1): 121–131

[18]

Agergaard J, Justesen TEH, Jespersen SE, Tagmose Thomsen T, Holm L, van Hall G. Even or skewed dietary protein distribution is reflected in the whole-body protein net-balance in healthy older adults: a randomized controlled trial. Clin Nutr 2023; 42(6): 899–908

[19]

Murashige D, Jang C, Neinast M, Edwards JJ, Cowan A, Hyman MC, Rabinowitz JD, Frankel DS, Arany Z. Comprehensive quantification of fuel use by the failing and nonfailing human heart. Science 2020; 370(6514): 364–368

[20]

Liu Y, Hou Y, Wang G, Zheng X, Hao H. Gut microbial metabolites of aromatic amino acids as signals in host-microbe interplay. Trends Endocrinol Metab 2020; 31(11): 818–834

[21]

Barrios C, Beaumont M, Pallister T, Villar J, Goodrich JK, Clark A, Pascual J, Ley RE, Spector TD, Bell JT, Menni C. Gut-microbiota-metabolite axis in early renal function decline. PLoS One 2015; 10(8): e0134311

[22]

Zhu Y, Dwidar M, Nemet I, Buffa JA, Sangwan N, Li XS, Anderson JT, Romano KA, Fu X, Funabashi M, Wang Z, Keranahalli P, Battle S, Tittle AN, Hajjar AM, Gogonea V, Fischbach MA, DiDonato JA, Hazen SL. Two distinct gut microbial pathways contribute to meta-organismal production of phenylacetylglutamine with links to cardiovascular disease. Cell Host Microbe 2023; 31(1): 18–32.e9

[23]

Bouatra S, Aziat F, Mandal R, Guo AC, Wilson MR, Knox C, Bjorndahl TC, Krishnamurthy R, Saleem F, Liu P, Dame ZT, Poelzer J, Huynh J, Yallou FS, Psychogios N, Dong E, Bogumil R, Roehring C, Wishart DS. The human urine metabolome. PLoS One 2013; 8(9): e73076

[24]

Mair RD, Lee S, Plummer NS, Sirich TL, Meyer TW. Impaired tubular secretion of organic solutes in advanced chronic kidney disease. J Am Soc Nephrol 2021; 32(11): 2877–2884

[25]

Poesen R, Claes K, Evenepoel P, de Loor H, Augustijns P, Kuypers D, Meijers B. Microbiota-derived phenylacetylglutamine associates with overall mortality and cardiovascular disease in patients with CKD. J Am Soc Nephrol 2016; 27(11): 3479–3487

[26]

Mair RD, Sirich TL, Meyer TW. Uremic toxin clearance and cardiovascular toxicities. Toxins (Basel) 2018; 10(6): 226

[27]

Brusilow SW. Phenylacetylglutamine may replace urea as a vehicle for waste nitrogen excretion. Pediatr Res 1991; 29(2): 147–150

[28]

Sirich TL, Funk BA, Plummer NS, Hostetter TH, Meyer TW. Prominent accumulation in hemodialysis patients of solutes normally cleared by tubular secretion. J Am Soc Nephrol 2014; 25(3): 615–622

[29]

Posada-Ayala M, Zubiri I, Martin-Lorenzo M, Sanz-Maroto A, Molero D, Gonzalez-Calero L, Fernandez-Fernandez B, de la Cuesta F, Laborde CM, Barderas MG, Ortiz A, Vivanco F, Alvarez-Llamas G. Identification of a urine metabolomic signature in patients with advanced-stage chronic kidney disease. Kidney Int 2014; 85(1): 103–111

[30]

Safadi R, Rahimi RS, Thabut D, Bajaj JS, Ram Bhamidimarri K, Pyrsopoulos N, Potthoff A, Bukofzer S, Wang L, Jamil K, Devarakonda KR. Pharmacokinetics/pharmacodynamics of L-ornithine phenylacetate in overt hepatic encephalopathy and the effect of plasma ammonia concentration reduction on clinical outcomes. Clin Transl Sci 2022; 15(6): 1449–1459

[31]

Wang X, Tseng J, Mak C, Poola N, Vilchez RA. Exposures of phenylacetic acid and phenylacetylglutamine across different subpopulations and correlation with adverse events. Clin Pharmacokinet 2021; 60(12): 1557–1567

[32]

Andrade F, Cano A, Unceta Suarez M, Arza A, Vinuesa A, Ceberio L, López-Oslé N, de Frutos G, López-Oceja R, Aznal E, González-Lamuño D, de Las Heras J. Urine phenylacetylglutamine determination in patients with hyperphenylalaninemia. J Clin Med 2021; 10(16): 3674

[33]

Magnusson I, Schumann WC, Bartsch GE, Chandramouli V, Kumaran K, Wahren J, Landau BR. Noninvasive tracing of Krebs cycle metabolism in liver. J Biol Chem 1991; 266(11): 6975–6984

[34]

Esenmo E, Chandramouli V, Schumann WC, Kumaran K, Wahren J, Landau BR. Use of 14CO2 in estimating rates of hepatic gluconeogenesis. Am J Physiol 1992; 263(1): E36–E41

[35]

Shockcor JP, Unger SE, Wilson ID, Foxall PJ, Nicholson JK, Lindon JC. Combined HPLC, NMR spectroscopy, and ion-trap mass spectrometry with application to the detection and characterization of xenobiotic and endogenous metabolites in human urine. Anal Chem 1996; 68(24): 4431–4435

[36]

Fukui Y, Kato M, Inoue Y, Matsubara A, Itoh K. A metabonomic approach identifies human urinary phenylacetylglutamine as a novel marker of interstitial cystitis. J Chromatogr B Analyt Technol Biomed Life Sci 2009; 877(30): 3806–3812

[37]

Blaise BJ, Correia GDS, Haggart GA, Surowiec I, Sands C, Lewis MR, Pearce JTM, Trygg J, Nicholson JK, Holmes E, Ebbels TMD. Statistical analysis in metabolic phenotyping. Nat Protoc 2021; 16(9): 4299–4326

[38]

Marchev AS, Vasileva LV, Amirova KM, Savova MS, Balcheva-Sivenova ZP, Georgiev MI. Metabolomics and health: from nutritional crops and plant-based pharmaceuticals to profiling of human biofluids. Cell Mol Life Sci 2021; 78(19–20): 6487–6503

[39]

Piszcz J, Lemancewicz D, Dudzik D, Ciborowski M. Differences and similarities between LC-MS derived serum fingerprints of patients with B-cell malignancies. Electrophoresis 2013; 34(19): 2857–2864

[40]

Wei H, Wu J, Wang H, Huang J, Li C, Zhang Y, Song Y, Zhou Z, Sun Y, Xiao L, Peng L, Chen C, Zhao C, Wang DW. Increased circulating phenylacetylglutamine concentration elevates the predictive value of cardiovascular event risk in heart failure patients. J Intern Med 2023; 294(4): 515–530

[41]

Tang Y, Zou Y, Cui J, Ma X, Zhang L, Yu S, Qiu L. Analysis of two intestinal bacterial metabolites (trimethylamine N-oxide and phenylacetylglutamine) in human serum samples of patients with T2DM and AMI using a liquid chromatography tandem mass spectrometry method. Clin Chim Acta 2022; 536: 162–168

[42]

Yang D, Beylot M, Agarwal KC, Soloviev MV, Brunengraber H. Assay of the human liver citric acid cycle probe phenylacetylglutamine and of phenylacetate in plasma by gas chromatography-mass spectrometry. Anal Biochem 1993; 212(1): 277–282

[43]

Fang C, Zuo K, Fu Y, Li J, Wang H, Xu L, Yang X. Dysbiosis of gut microbiota and metabolite phenylacetylglutamine in coronary artery disease patients with stent stenosis. Front Cardiovasc Med 2022; 9: 832092

[44]

Fang C, Zuo K, Jiao K, Zhu X, Fu Y, Zhong J, Xu L, Yang X. PAGln, an atrial fibrillation-linked gut microbial metabolite, acts as a promoter of atrial myocyte injury. Biomolecules 2022; 12(8): 1120

[45]

Kahl KW, Seither JZ, Reidy LJ. LC-MS-MS vs ELISA: validation of a comprehensive urine toxicology screen by LC-MS-MS and a comparison of 100 forensic specimens. J Anal Toxicol 2019; 43(9): 734–745

[46]

Fabresse N, Larabi IA, Abe E, Lamy E, Rigothier C, Massy ZA, Alvarez JC. Correlation between saliva levels and serum levels of free uremic toxins in healthy volunteers. Toxins (Basel) 2023; 15(2): 150

[47]

Kuc D, Rahnama M, Tomaszewski T, Rzeski W, Wejksza K, Urbanik-Sypniewska T, Parada-Turska J, Wielosz M, Turski WA. Kynurenic acid in human saliva—does it influence oral microflora? Pharmacol Rep 2006; 58(3): 393–398

[48]

Wu W, Bush KT, Nigam SK. Key role for the organic anion transporters, OAT1 and OAT3, in the in vivo handling of uremic toxins and solutes. Sci Rep 2017; 7(1): 4939

[49]

Nemet I, Li XS, Haghikia A, Li L, Wilcox J, Romano KA, Buffa JA, Witkowski M, Demuth I, König M, Steinhagen-Thiessen E, Bäckhed F, Fischbach MA, Tang WHW, Landmesser U, Hazen SL. Atlas of gut microbe-derived products from aromatic amino acids and risk of cardiovascular morbidity and mortality. Eur Heart J 2023; 44(32): 3085–3096

[50]

Damasceno A, Lunet N. Comorbidities and heart failure: heterogeneity and challenges to fill in the gaps. Lancet Glob Health 2023; 11(12): e1830–e1831

[51]

Niebauer J, Volk HD, Kemp M, Dominguez M, Schumann RR, Rauchhaus M, Poole-Wilson PA, Coats AJ, Anker SD. Endotoxin and immune activation in chronic heart failure: a prospective cohort study. Lancet 1999; 353(9167): 1838–1842

[52]

Tang WH, Wang Z, Fan Y, Levison B, Hazen JE, Donahue LM, Wu Y, Hazen SL. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-N-oxide in patients with heart failure: refining the gut hypothesis. J Am Coll Cardiol 2014; 64(18): 1908–1914

[53]

Braunwald E. Heart failure: a 70 year Odyssey. Eur Heart J 2022; 43(18): 1697–1699

[54]

Zheng Y, Yu B, Alexander D, Manolio TA, Aguilar D, Coresh J, Heiss G, Boerwinkle E, Nettleton JA. Associations between metabolomic compounds and incident heart failure among African Americans: the ARIC Study. Am J Epidemiol 2013; 178(4): 534–542

[55]

Tang HY, Wang CH, Ho HY, Lin JF, Lo CJ, Huang CY, Cheng ML. Characteristic of metabolic status in heart failure and its impact in outcome perspective. Metabolites 2020; 10(11): 437

[56]

Zong X, Fan Q, Yang Q, Pan R, Zhuang L, Tao R. Phenylacetylglutamine as a risk factor and prognostic indicator of heart failure. ESC Heart Fail 2022; 9(4): 2645–2653

[57]

TangWHWNemet ILiXSWuYHaghikiaA WitkowskiMKoeth RADemuthIKönigMSteinhagen-Thiessen EBäckhedFFischbachMADebA LandmesserUHazen SL. Prognostic value of gut microbe-generated metabolite phenylacetylglutamine in patients with heart failure. Eur J Heart Fail 2023; [Epub ahead of print] doi: 10.1002/ejhf.3111

[58]

Liu Y, Liu S, Zhao Z, Song X, Qu H, Liu H. Phenylacetylglutamine is associated with the degree of coronary atherosclerotic severity assessed by coronary computed tomographic angiography in patients with suspected coronary artery disease. Atherosclerosis 2021; 333: 75–82

[59]

Yap IK, Brown IJ, Chan Q, Wijeyesekera A, Garcia-Perez I, Bictash M, Loo RL, Chadeau-Hyam M, Ebbels T, De Iorio M, Maibaum E, Zhao L, Kesteloot H, Daviglus ML, Stamler J, Nicholson JK, Elliott P, Holmes E. Metabolome-wide association study identifies multiple biomarkers that discriminate north and south Chinese populations at differing risks of cardiovascular disease: INTERMAP study. J Proteome Res 2010; 9(12): 6647–6654

[60]

Ottosson F, Brunkwall L, Smith E, Orho-Melander M, Nilsson PM, Fernandez C, Melander O. The gut microbiota-related metabolite phenylacetylglutamine associates with increased risk of incident coronary artery disease. J Hypertens 2020; 38(12): 2427–2434

[61]

Menni C, Lin C, Cecelja M, Mangino M, Matey-Hernandez ML, Keehn L, Mohney RP, Steves CJ, Spector TD, Kuo CF, Chowienczyk P, Valdes AM. Gut microbial diversity is associated with lower arterial stiffness in women. Eur Heart J 2018; 39(25): 2390–2397

[62]

Yu F, Li X, Feng X, Wei M, Luo Y, Zhao T, Xiao B, Xia J. Phenylacetylglutamine, a novel biomarker in acute ischemic stroke. Front Cardiovasc Med 2021; 8: 798765

[63]

Yu F, Feng X, Li X, Luo Y, Wei M, Zhao T, Xia J. Gut-derived metabolite phenylacetylglutamine and white matter hyperintensities in patients with acute ischemic stroke. Front Aging Neurosci 2021; 13: 675158

[64]

Azab SM, Zamzam A, Syed MH, Abdin R, Qadura M, Britz-McKibbin P. Serum metabolic signatures of chronic limb-threatening ischemia in patients with peripheral artery disease. J Clin Med 2020; 9(6): 1877

[65]

Fu Y, Yang Y, Fang C, Liu X, Dong Y, Xu L, Chen M, Zuo K, Wang L. Prognostic value of plasma phenylalanine and gut microbiota-derived metabolite phenylacetylglutamine in coronary in-stent restenosis. Front Cardiovasc Med 2022; 9: 944155

[66]

Bogiatzi C, Gloor G, Allen-Vercoe E, Reid G, Wong RG, Urquhart BL, Dinculescu V, Ruetz KN, Velenosi TJ, Pignanelli M, Spence JD. Metabolic products of the intestinal microbiome and extremes of atherosclerosis. Atherosclerosis 2018; 273: 91–97

[67]

Zuo K, Li J, Li K, Hu C, Gao Y, Chen M, Hu R, Liu Y, Chi H, Wang H, Qin Y, Liu X, Li S, Cai J, Zhong J, Yang X. Disordered gut microbiota and alterations in metabolic patterns are associated with atrial fibrillation. Gigascience 2019; 8(6): giz058

[68]

Gawałko M, Agbaedeng TA, Saljic A, Müller DN, Wilck N, Schnabel R, Penders J, Rienstra M, van Gelder I, Jespersen T, Schotten U, Crijns HJGM, Kalman JM, Sanders P, Nattel S, Dobrev D, Linz D. Gut microbiota, dysbiosis and atrial fibrillation. Arrhythmogenic mechanisms and potential clinical implications. Cardiovasc Res 2022; 118(11): 2415–2427

[69]

Fu H, Kong B, Zhu J, Huang H, Shuai W. Phenylacetylglutamine increases the susceptibility of ventricular arrhythmias in heart failure mice by exacerbated activation of the TLR4/AKT/mTOR signaling pathway. Int Immunopharmacol 2023; 116: 109795

[70]

Witkowski M, Weeks TL, Hazen SL. Gut microbiota and cardiovascular disease. Circ Res 2020; 127(4): 553–570

[71]

Zhang X, Li Y, Yang P, Liu X, Lu L, Chen Y, Zhong X, Li Z, Liu H, Ou C, Yan J, Chen M. Trimethylamine-N-oxide promotes vascular calcification through activation of NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3) inflammasome and NF-κB (nuclear factor κB) signals. Arterioscler Thromb Vasc Biol 2020; 40(3): 751–765

[72]

Zhao J, Zhang Q, Cheng W, Dai Q, Wei Z, Guo M, Chen F, Qiao S, Hu J, Wang J, Chen H, Bao X, Mu D, Sun X, Xu B, Xie J. Heart-gut microbiota communication determines the severity of cardiac injury after myocardial ischaemia/reperfusion. Cardiovasc Res 2023; 119(6): 1390–1402

[73]

Li X, Geng J, Zhao J, Ni Q, Zhao C, Zheng Y, Chen X, Wang L. Trimethylamine N-oxide exacerbates cardiac fibrosis via activating the NLRP3 inflammasome. Front Physiol 2019; 10: 866

[74]

Aoki K, Teshima Y, Kondo H, Saito S, Fukui A, Fukunaga N, Nawata T, Shimada T, Takahashi N, Shibata H. Role of indoxyl sulfate as a predisposing factor for atrial fibrillation in renal dysfunction. J Am Heart Assoc 2015; 4(10): e002023

[75]

Adamo L, Rocha-Resende C, Prabhu SD, Mann DL. Reappraising the role of inflammation in heart failure. Nat Rev Cardiol 2020; 17(5): 269–285

[76]

Xu X, Lu WJ, Shi JY, Su YL, Liu YC, Wang L, Xiao CX, Chen C, Lu Q. The gut microbial metabolite phenylacetylglycine protects against cardiac injury caused by ischemia/reperfusion through activating β2AR. Arch Biochem Biophys 2021; 697: 108720

[77]

Hazekawa M, Ono K, Nishinakagawa T, Kawakubo-Yasukochi T, Nakashima M. In vitro anti-inflammatory effects of the phenylbutyric acid metabolite phenylacetyl glutamine. Biol Pharm Bull 2018; 41(6): 961–966

[78]

Fang C, Zuo K, Jiao K, Zhu X, Fu Y, Zhong J, Xu L, Yang X. PAGln, an atrial fibrillation-linked gut microbial metabolite, acts as a promoter of atrial myocyte injury. Biomolecules 2022; 12(8): 1120

[79]

Glassock RJ, Massry SG. Chapter 6 — Uremic toxins: an integrated overview of classification and pathobiology. In: Kopple JD, Massry SG, Kalantar-Zadeh K, Fouque D. Nutritional Management of Renal Disease (Fourth Edition). Academic Press, 2022: 77–89

[80]

Zhong J, Kirabo A, Yang HC, Fogo AB, Shelton EL, Kon V. Intestinal lymphatic dysfunction in kidney disease. Circ Res 2023; 132(9): 1226–1245

[81]

Glorieux G, Nigam SK, Vanholder R, Verbeke F. Role of the microbiome in gut-heart-kidney cross talk. Circ Res 2023; 132(8): 1064–1083

[82]

Ahn SY, Nigam SK. Toward a systems level understanding of organic anion and other multispecific drug transporters: a remote sensing and signaling hypothesis. Mol Pharmacol 2009; 76(3): 481–490

[83]

Rosenthal SB, Bush KT, Nigam SK. A network of SLC and ABC transporter and DME genes involved in remote sensing and signaling in the gut-liver-kidney axis. Sci Rep 2019; 9(1): 11879

[84]

Lekawanvijit S, Kompa AR, Wang BH, Kelly DJ, Krum H. Cardiorenal syndrome: the emerging role of protein-bound uremic toxins. Circ Res 2012; 111(11): 1470–1483

[85]

Sundaram V, Fang JC. Gastrointestinal and liver issues in heart failure. Circulation 2016; 133(17): 1696–1703

[86]

Spence JD, Urquhart BL. Cerebrovascular disease, cardiovascular disease, and chronic kidney disease: interplays and influences. Curr Neurol Neurosci Rep 2022; 22(11): 757–766

[87]

Tsutamoto T, Kawahara C, Nishiyama K, Yamaji M, Fujii M, Yamamoto T, Horie M. Prognostic role of highly sensitive cardiac troponin I in patients with systolic heart failure. Am Heart J 2010; 159(1): 63–67

[88]

Sandoval Y, Apple FS, Mahler SA, Body R, Collinson PO, Jaffe AS; International Federation of Clinical Chemistry and Laboratory Medicine Committee on the Clinical Application of Cardiac Biomarkers. High-sensitivity cardiac troponin and the 2021 AHA/ACC/ASE/CHEST/SAEM/SCCT/SCMR guidelines for the evaluation and diagnosis of acute chest pain. Circulation 2022; 146(7): 569–581 doi:10.1161/CIRCULATIONAHA.122.059678

[89]

McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, Burri H, Butler J, Čelutkienė J, Chioncel O, Cleland JGF, Coats AJS, Crespo-Leiro MG, Farmakis D, Gilard M, Heymans S, Hoes AW, Jaarsma T, Jankowska EA, Lainscak M, Lam CSP, Lyon AR, McMurray JJV, Mebazaa A, Mindham R, Muneretto C, Francesco Piepoli M, Price S, Rosano GMC, Ruschitzka F, Kathrine Skibelund A; ESC Scientific Document Group. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J 2021; 42(36): 3599–3726

[90]

Shiels PG. ‘Debugging’ heart failure. J Intern Med 2023; 294(4): 374–376

[91]

Romano KA, Nemet I, Prasad Saha P, Haghikia A, Li XS, Mohan ML, Lovano B, Castel L, Witkowski M, Buffa JA, Sun Y, Li L, Menge CM, Demuth I, König M, Steinhagen-Thiessen E, DiDonato JA, Deb A, Bäckhed F, Tang WHW, Naga Prasad SV, Landmesser U, Van Wagoner DR, Hazen SL. Gut microbiota-generated phenylacetylglutamine and heart failure. Circ Heart Fail 2023; 16(1): e009972

[92]

Elliott P, Posma JM, Chan Q, Garcia-Perez I, Wijeyesekera A, Bictash M, Ebbels TM, Ueshima H, Zhao L, van Horn L, Daviglus M, Stamler J, Holmes E, Nicholson JK. Urinary metabolic signatures of human adiposity. Sci Transl Med 2015; 7(285): 285ra62

[93]

Chang AY, Abdullah SM, Jain T, Stanek HG, Das SR, McGuire DK, Auchus RJ, de Lemos JA. Associations among androgens, estrogens, and natriuretic peptides in young women: observations from the Dallas Heart Study. J Am Coll Cardiol 2007; 49(1): 109–116

[94]

Mueller C, McDonald K, de Boer RA, Maisel A, Cleland JGF, Kozhuharov N, Coats AJS, Metra M, Mebazaa A, Ruschitzka F, Lainscak M, Filippatos G, Seferovic PM, Meijers WC, Bayes-Genis A, Mueller T, Richards M, Januzzi JL Jr; Heart Failure Association of the European Society of Cardiology. Heart Failure Association of the European Society of Cardiology practical guidance on the use of natriuretic peptide concentrations. Eur J Heart Fail 2019; 21(6): 715–731

[95]

Landau BR, Chandramouli V, Schumann WC, Ekberg K, Kumaran K, Kalhan SC, Wahren J. Estimates of Krebs cycle activity and contributions of gluconeogenesis to hepatic glucose production in fasting healthy subjects and IDDM patients. Diabetologia 1995; 38(7): 831–838

[96]

Hua S, Lv B, Qiu Z, Li Z, Wang Z, Chen Y, Han Y, Tucker KL, Wu H, Jin W. Microbial metabolites in chronic heart failure and its common comorbidities. EMBO Mol Med 2023; 15(6): e16928

[97]

Xu J, Cai M, Wang Z, Chen Q, Han X, Tian J, Jin S, Yan Z, Li Y, Lu B, Lu H. Phenylacetylglutamine as a novel biomarker of type 2 diabetes with distal symmetric polyneuropathy by metabolomics. J Endocrinol Invest 2023; 46(5): 869–882

[98]

Zuo J, Lan Y, Hu H, Hou X, Li J, Wang T, Zhang H, Zhang N, Guo C, Peng F, Zhao S, Wei Y, Jia C, Zheng C, Mao G. Metabolomics-based multidimensional network biomarkers for diabetic retinopathy identification in patients with type 2 diabetes mellitus. BMJ Open Diabetes Res Care 2021; 9(1): e001443

[99]

Tan YM, Gao Y, Teo G, Koh HWL, Tai ES, Khoo CM, Choi KP, Zhou L, Choi H. Plasma metabolome and lipidome associations with type 2 diabetes and diabetic nephropathy. Metabolites 2021; 11(4): 228

[100]

Pan A, Sun Q, Bernstein AM, Schulze MB, Manson JE, Stampfer MJ, Willett WC, Hu FB. Red meat consumption and mortality: results from 2 prospective cohort studies. Arch Intern Med 2012; 172(7): 555–563

[101]

Mohan D, Mente A, Dehghan M, Rangarajan S, O’Donnell M, Hu W, Dagenais G, Wielgosz A, Lear S, Wei L, Diaz R, Avezum A, Lopez-Jaramillo P, Lanas F, Swaminathan S, Kaur M, Vijayakumar K, Mohan V, Gupta R, Szuba A, Iqbal R, Yusuf R, Mohammadifard N, Khatib R, Yusoff K, Gulec S, Rosengren A, Yusufali A, Wentzel-Viljoen E, Chifamba J, Dans A, Alhabib KF, Yeates K, Teo K, Gerstein HC, Yusuf S; PURE, ONTARGET investigators. Associations of fish consumption with risk of cardiovascular disease and mortality among individuals with or without vascular disease from 58 countries. JAMA Intern Med 2021; 181(5): 631–649

[102]

Hitch TCA, Hall LJ, Walsh SK, Leventhal GE, Slack E, de Wouters T, Walter J, Clavel T. Microbiome-based interventions to modulate gut ecology and the immune system. Mucosal Immunol 2022; 15(6): 1095–1113

[103]

Witkowski M, Witkowski M, Friebel J, Buffa JA, Li XS, Wang Z, Sangwan N, Li L, DiDonato JA, Tizian C, Haghikia A, Kirchhofer D, Mach F, Räber L, Matter CM, Tang WHW, Landmesser U, Lüscher TF, Rauch U, Hazen SL. Vascular endothelial tissue factor contributes to trimethylamine N-oxide-enhanced arterial thrombosis. Cardiovasc Res 2022; 118(10): 2367–2384

[104]

Dahl WJ, Hung WL, Ford AL, Suh JH, Auger J, Nagulesapillai V, Wang Y. In older women, a high-protein diet including animal-sourced foods did not impact serum levels and urinary excretion of trimethylamine-N-oxide. Nutr Res 2020; 78: 72–81

[105]

Chan Q, Wren GM, Lau CE, Ebbels TMD, Gibson R, Loo RL, Aljuraiban GS, Posma JM, Dyer AR, Steffen LM, Rodriguez BL, Appel LJ, Daviglus ML, Elliott P, Stamler J, Holmes E, Van Horn L. Blood pressure interactions with the DASH dietary pattern, sodium, and potassium: the International Study of Macro-/Micronutrients and Blood Pressure (INTERMAP). Am J Clin Nutr 2022; 116(1): 216–229

[106]

Kaye DM, Shihata WA, Jama HA, Tsyganov K, Ziemann M, Kiriazis H, Horlock D, Vijay A, Giam B, Vinh A, Johnson C, Fiedler A, Donner D, Snelson M, Coughlan MT, Phillips S, Du XJ, El-Osta A, Drummond G, Lambert GW, Spector TD, Valdes AM, Mackay CR, Marques FZ. Deficiency of prebiotic fiber and insufficient signaling through gut metabolite-sensing receptors leads to cardiovascular disease. Circulation 2020; 141(17): 1393–1403

[107]

Marques FZ, Nelson E, Chu PY, Horlock D, Fiedler A, Ziemann M, Tan JK, Kuruppu S, Rajapakse NW, El-Osta A, Mackay CR, Kaye DM. 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): 964–977

[108]

Zhou Y, Zhang N, Arikawa AY, Chen C. Inhibitory effects of green tea polyphenols on microbial metabolism of aromatic amino acids in humans revealed by metabolomic analysis. Metabolites 2019; 9(5): 96

[109]

Lam V, Su J, Hsu A, Gross GJ, Salzman NH, Baker JE. Intestinal microbial metabolites are linked to severity of myocardial infarction in rats. PLoS One 2016; 11(8): e0160840

[110]

Li Z, Wu Z, Yan J, Liu H, Liu Q, Deng Y, Ou C, Chen M. Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Lab Invest 2019; 99(3): 346–357

[111]

Riba A, Deres L, Eros K, Szabo A, Magyar K, Sumegi B, Toth K, Halmosi R, Szabados E. Doxycycline protects against ROS-induced mitochondrial fragmentation and ISO-induced heart failure. PLoS One 2017; 12(4): e0175195

[112]

Palleja A, Mikkelsen KH, Forslund SK, Kashani A, Allin KH, Nielsen T, Hansen TH, Liang S, Feng Q, Zhang C, Pyl PT, Coelho LP, Yang H, Wang J, Typas A, Nielsen MF, Nielsen HB, Bork P, Wang J, Vilsbøll T, Hansen T, Knop FK, Arumugam M, Pedersen O. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat Microbiol 2018; 3(11): 1255–1265

[113]

Winkel P, Hilden J, Hansen JF, Kastrup J, Kolmos HJ, Kjøller E, Jensen GB, Skoog M, Lindschou J, Gluud C; CLARICOR trial group. Clarithromycin for stable coronary heart disease increases all-cause and cardiovascular mortality and cerebrovascular morbidity over 10years in the CLARICOR randomised, blinded clinical trial. Int J Cardiol 2015; 182: 459–465

[114]

Wang G, Kong B, Shuai W, Fu H, Jiang X, Huang H. 3,3-Dimethyl-1-butanol attenuates cardiac remodeling in pressure-overload-induced heart failure mice. J Nutr Biochem 2020; 78: 108341

[115]

Fatani AMN, Suh JH, Auger J, Alabasi KM, Wang Y, Segal MS, Dahl WJ. Pea hull fiber supplementation does not modulate uremic metabolites in adults receiving hemodialysis: a randomized, double-blind, controlled trial. Front Nutr 2023; 10: 1179295

[116]

Tang WW, Hazen SL. Dietary metabolism, gut microbiota and acute heart failure. Heart 2016; 102(11): 813–814

[117]

Zhang Z, Cai B, Sun Y, Deng H, Wang H, Qiao Z. Alteration of the gut microbiota and metabolite phenylacetylglutamine in patients with severe chronic heart failure. Front Cardiovasc Med 2023; 9: 1076806

[118]

Zong X, Fan Q, Yang Q, Pan R, Zhuang L, Tao R. Phenylacetylglutamine as a risk factor and prognostic indicator of heart failure. ESC Heart Fail 2022; 9(4): 2645–2653

RIGHTS & PERMISSIONS

The Author(s) 2024. This article is published with open access at link.springer.com and journal.hep.com.cn

AI Summary AI Mindmap
PDF (1265KB)

4730

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/