3,105
views
1
recommends
+1 Recommend
1 collections
    0
    shares

      CVIA now indexed by SCOPUS from February 2024. CVIA received its first Journal Impact Factor (0.5) in the 2023 Journal Citation Reports Release. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Gut Microbiota and Vascular Diseases: An Update

      Published
      review-article
      Bookmark

            Abstract

            Vascular diseases, including atherosclerosis, aneurysms, and vascular calcification, are a leading cause of morbidity and mortality worldwide. In past decades, the gut microbiota has been found to be an indispensable population exerting effects on hosts under physiological and pathological conditions. Gut microbiota-derived metabolites, such as trimethylamine-N-oxide and short-chain fatty acids, mediate these effects by regulating vascular cells systematically. Translation of research knowledge to clinical scenarios has led to the development of new therapies including dietary interventions and metabolite inhibitors. This review describes recent advancements in understanding of the interplay between the gut microbiota and vascular dysfunction, and potential treatments for vascular diseases.

            Main article text

            Introduction

            Vascular systems, consisting primarily of the tunica intima, tunica media, and adventitia, circulate blood through the entire body, and provide oxygen and nutrients for diverse tissues. Blood vessels are not simply elastic tubes but are a dynamic microenvironment responding to stimuli. Vascular homeostasis is maintained by cell types including endothelial cells, vascular smooth muscle cells (VSMCs), fibroblasts, macrophages, and adipocytes. Disruption of vascular homeostasis by various risk factors, such as diabetes, hyperlipidemia, smoking, high blood pressure, and obesity, lead to vascular remodeling, in a structural and functional adaptive process. Consequently, various vascular diseases, such as atherosclerosis, vascular calcification (VC), and aneurysms, can arise and threaten human health.

            In the human gastrointestinal tract, more than 100 trillion microorganisms, termed the gut microbiota, symbiotically evolved together with the host [1, 2]. Substantial evidence has demonstrated that the gut microbiota acts as an “invisible organ” affecting human physiology, metabolism, and immunity homeostasis [13]. A well-balanced gut microbiota preserves host health, and gut microbiota dysbiosis facilitates inflammatory and metabolic diseases, such as vascular diseases [1, 4]. A deeper understanding of the mechanisms of gut microbiota-host interaction has enabled novel therapeutic options for delaying or even reversing vascular diseases. In this narrative review, we summarize recent progress in the advancing field of the gut microbiota and vascular remodeling processes. We searched manuscripts by using the keywords (“gut microbiota” or “microorganism” or “TMAO”) and (“atherosclerosis” or “vascular calcification” or “aneurysms”) in PubMed, and focused on studies published in the past 3 years.

            Atherosclerosis

            Atherosclerosis, one of the most common vascular diseases, is characterized by endothelial dysfunction, lipid accumulation, infiltration of inflammatory cells, and the formation of plaques, thus ultimately leading to vascular stenosis and ischemic events including myocardial infarction, cerebral infarction, mesenteric artery ischemia, and peripheral artery disease (PAD) [5]. Low and oscillatory shear stress activates endothelial inflammation and contributes to the pathogenesis of atherosclerosis and restenosis [6, 7]. Although gut microbiota dysbiosis has been observed in several studies, the conclusions have been ambiguous, largely because of differences in study populations, criteria, and methods [8, 9]. Recently, Choroszy’s analysis of 21 studies has highlighted that Enterobacteriaceae, Lactobacillus, and Streptococcus are elevated, while Bacteroidetes and Lachnospiraceae are diminished, in patients with coronary artery disease (CAD) [10]. These changes might correlate with the progression of atherosclerosis. Gut microbiota alterations in patients with PAD remain unclear, including whether these patients show similar trends in microbiota changes to patients with CAD. The precise roles of specific taxa in atherosclerosis also remain uncertain. For instance, Lactobacillus has traditionally been considered to have anti-atherosclerotic roles by lowering lipids but is paradoxically elevated in patients with CAD [11, 12]. Whether quantitative changes in Lactobacillus protects against CAD development remains to be determined.

            A plethora of evidence, including shifts in gut microbiota-modified metabolites in patients with CAD, has suggested that metabolites directly link microbiota dysbiosis to vascular atherosclerosis.

            Trimethylamine-N-oxide (TMAO), a metabolite produced from dietary precursors in the liver by trimethylamine (TMA) oxidization, has been explored in previous studies [13, 14]. Extensive clinical and animal studies have concluded that TMAO promotes atherosclerosis through multiple mechanisms, such as cholesterol accumulation, inflammatory cell recruitment, and endothelial dysfunction [3, 1315]. Elevated TMAO levels predict greater incidence of adverse events in patients with CAD and PAD [16]. Promisingly, TMAO precursors and TMAO are targets for vascular protection. In contrast to TMAO, short-chain fatty acids (SCFAs), generated from fermented fibers, exert beneficial effects on vascular inflammation and lipid disruption [17, 18]. Recently, Haghikia’s research has emphasized that propionic acid (PA), an important SCFA, significantly decreases atherosclerotic lesions in HFD-induced Apoe/ mice [19]. This protective effect is mediated by inhibition of cholesterol transporters and lowering of cholesterol levels (Figure 1) [19]. Interestingly, oral administration of 500 mg of PA twice daily for 8 weeks markedly decreases cholesterol levels in patients with hypercholesterolemia, thus suggesting translational potential of this treatment in the future [19]. A larger long-term study is warranted to comprehensively evaluate the effects of PA on vascular walls and other tissues.

            Figure 1

            Effects and Mechanisms of SCFAs in Vascular Diseases.

            Illustration made with Figdraw.

            Considerable investigations have indicated the involvement of microbiota-derived tryptophan metabolites in vascular inflammation and atherosclerosis [20, 21]. Xue’s updated work has established that, in addition to indolamine-2,3-dioxygenase-1 (IDO1), indole-3-propionic acid (IPA), another tryptophan metabolite, is correlated with atherosclerosis, on the basis of microbiome-metabolome sequencing in patients [22]. IPA is diminished in atherosclerotic patients and ApoE/ mice, whereas administration of IPA hinders atherosclerosis development in ApoE/ mice [22]. The beneficial effects are due to increased cholesterol efflux in macrophages through modulation of the miR-142-5p/ABCA1 signaling pathway [22]. Phenylacetylglutamine plays a crucial role in atherosclerosis and in-stent stenosis, but the exact mechanisms remain unsubstantiated [23, 24]. Further studies on phenylacetylglutamine are anticipated.

            Natural products are a group of bioactive components with robust ability to protect against vascular inflammation [25]. A body of updated studies have affirmed that these protective effects result from modulation of the gut microbiota, because the oral bioavailability of these natural products is low. Although previous investigations have suggested that berberine minimizes plaque development, Ma’s work has further verified that this observation is strongly associated with decreased TMAO generation through a vitamin-like effect downregulating the choline-TMA-TMAO pathway [26]. Importantly, oral administration of berberine for 4 months down-regulates plaque scores and TMAO levels in patients with atherosclerosis compared with those receiving a statin plus antiplatelet drugs [26]. However, this evaluation was conducted through ultrasonography, which is less accurate than CT scans. Bicyclol, another herbal extract, has shown strong anti-atherosclerotic effects in high fat diet-induced murine models [27]. A metagenome-wide association study has revealed that bicyclol maintains the gut microbiota, and further preserves gut integrity and immunity [27]. Ginsenoside Rc, like other family members, alleviates atherosclerosis development by restoring the gut microbiota, particularly Muribaculaceae, Lactobacillus, Ileibacterium, Bifidobacterium, Faecalibaculum, Oscillibacter, Blautia, and Eubacterium_coprostanoligenes_group [28]. Consequently, the gut microbiota is an important mediator of the effects of natural products in vivo, and natural products may therefore find clinical applications. The results for berberine have been particularly encouraging, because berberine is an over-the-counter drug with demonstrated safety in time course studies. The long-term effects of berberine on atherosclerosis and the gut microbiota will be a topic of interest for further studies.

            Vascular Calcification

            Unlike atherosclerosis, VC is characterized by the deposition of calcium phosphate in the vascular intima and media [29, 30]. Progression of VC is a strictly regulated biological process associated with aging, diabetes, and chronic kidney disease (CKD) [30]. An elevated incidence of VC is observed in patients with CKD, largely because of excessive uremic toxins, and dysmetabolism of calcium and phosphate. Gut microbiota dysbiosis and abnormal metabolites also contribute to VC [3133]. In a pilot study including 44 patients with CKD with peritoneal dialysis, with or without VC, Merino-Ribas’s research has identified alterations in Coprobacter, Coprococcus 3, Lactobacillus, and Eubacterium eligens group in the gut, and Cutibacterium, Pajaroellobacter, Devosia, Hyphomicrobium, and Pelomonas in the blood [34]. Moreover, Eubacterium eligens in the gut and Devosia genus in the blood correlate with mortality in patients with CKD and therefore may predict prognosis in these patients [34]. Additionally, Bao and colleagues have analyzed the gut microbiota of patients with high versus low calcification scores, undergoing hemodialysis [35]. In accordance with findings from a previous study, Firmicutes, Actinobacteriota, Proteobacteria, and Bacteroidota were found to be major bacteria in patients with CKD [35]. Unexpectedly, the researchers identified another two phyla: Escherichia-Shigella and Ruminococcus, which were positively and negatively associated with vascular calcification, respectively [35]. Discrepancies in findings across studies may be due to differences in dialysis styles, patient race, diet, and methods. To date, conclusions have been largely based on the population with CKD. Investigating the gut microbiota in patients with age-associated calcification in clinical settings will be a necessary next step to advance research [36].

            An altered gut microbiota in patients with CKD with VC detrimentally affects vascular homeostasis and further contributes to VC progression [31, 32]. Various uremic toxins, such as indoxyl sulfate and p-cresyl sulfate, originate from the gut microbiota [37]. These factors facilitate VC development through inflammatory activation and VSMC osteogenic changes [38, 39]. Although dietary interventions greatly decrease levels of indoxyl sulfate and p-cresyl sulfate, direct evidence of the effects of dietary intervention on VC remains insufficient [40]. TMAO is elevated in patients with CKD with VC, and has been verified as an independent risk factor for VC in patients undergoing hemodialysis [41, 42]. Furthermore, excessive supplementation with TMAO in rats with CKD promotes aortic calcification, whereas this effect is reversed by antibiotic treatment [42]. In vitro and ex vivo studies have demonstrated that TMAO enhances calcium/phosphate induced osteogenic differentiation of VSMCs, in a process mediated by NLRP3 and NF-κB stimulation [42]. Intriguingly, the influence of SCFAs on VC varies (Figure 1). In a cohort of 92 patients, the level of propionate has been negatively associated with vascular calcification [43]. To validate the role of propionate in VC, one study has supplemented vitamin D3 and nicotine induced rats with propionate, orally and rectally [43]. In this model, propionate significantly decreases VC progression and is accompanied by Akkermansia enrichment [43]. Furthermore, addition of Akkermansia hinders the development of VC; consequently, Akkermansia may mediate the protective effects of propionate [43]. Although butyrate has been speculated to be a protective factor against VC, Zhong’s work has indicated that butyrate accelerates VC in vitamin D3 induced mice [44]. In VSMCs with high phosphate, butyrate promotes calcification and activates osteogenic genes [44]. Mechanistic analysis has revealed that this phenomenon is correlated with HDAC inhibition and NF-κB signaling [44]. Nevertheless, observations have confirmed that propionate does not change mRNA expressions of osteogenic genes in high phosphate induced VSMCs, in contrast to Yan’s data [43, 44]. The use of different inducers of VC, namely, vitamin D3 and nicotine, or high phosphate, may account for the inconsistent results across studies. Another VC animal model induced by a high fat diet was not studied previously [45]. Given the diverse types of gut microbiota-derived metabolites, the precise roles of metabolites must be explored in the future [37] to provide further evidence regarding therapeutic targets of VC, given that no effective pharmacological strategies for VC have been developed in clinical settings.

            Abdominal Aortic Aneurysm and Intracranial Aneurysm

            Abdominal aortic aneurysm (AAA) is defined as segmental enlargement of the abdominal aorta above 50% of its normal diameter [46]. Rupture of AAA always causes death, although no effective pharmacological therapy is available to delay AAA development [47]. Emerging evidence suggests that the gut microbiota is robustly involved in AAA progression [48]. In an analysis of 30 AAA patients, diminished Bacteroidetes has been observed [49]. Importantly, bacteria such as Streptococcus have been identified in blood samples and aneurysmal tissues, thus suggesting that bacterial translocation may contribute to human AAA development [49]. Although Streptococcus is associated with mycotic AAA, the precise mechanisms underlying the role of Streptococcus in chronic AAA progression remain unclear [50, 51]. Recently, Tian’s work has further profiled gut dysbiosis in patients with AAA [52]. Proteobacteria and Actinobacteria are markedly elevated, whereas Firmicutes and Bacteroidetes are diminished, in patients with AAA [52]. Metabolic detection has further indicated that patients with AAA have low levels of beneficial metabolites and significantly elevated LPS levels [52]. Detailed exploration of the gut microbiota has also been conducted in an Ang II-induced AAA mouse model. Akkermansia is considered an important participant, and Akkermansia enrichment is negatively associated with AAA diameter [52]. Consistently, network analysis has suggested that Bacteroidetes tightly correlates with AAA progression [53]. However, no direct evidence has shown a causal relationship between Bacteroidetes and AAA, and this potential relationship must be validated in future work.

            Recently, several studies have evaluated the effects of the gut microbiota on AAA progression. Reduction of the gut microbiota by oral antibiotic treatment markedly delays AAA expansion and rupture, thus suggesting a relationship between AAA and the gut microbiota [53]. These beneficial effects are accompanied by a decrease in mobilization of monocytes from spleen, and gut microbiota-derived factors, such as Nod1 ligands, have been suggested to maintain splenic monocytes [54]. In AAA, as in atherosclerosis, TMAO has been suggested to be a crucial mediator facilitating disease progression [55]. Disrupting the production of TMAO by antibiotics, inhibitors, or genetic ablation alleviates AAA progression, even that of established AAA [55]. A mechanistic study has suggested that increased TMAO promotes endoplasmic reticulum stress and apoptosis in VSMCs [55]. The results of delaying establishment of AAA in mice might extend to patients diagnosed with AAA. Studies have estimated the overall effects of the gut microbiota. Interestingly, Tian’s comprehensive work has indicated that supplementation with R. Intestinalis decreases CaCl2-induced AAA expansion, thus suggesting promising application potential for R. Intestinalis in chronic AAA management (Figure 1) [52]. Butyrate, a metabolite of R. Intestinalis accounts for the decrease in neutrophil infiltration and neutrophil extracellular trap formation in aneurysmal tissues, in a NOX-dependent manner [52]. Application of R. Intestinalis or butyrate in the clinical management of AAA should be further explored, including its safety and convenience. Dosages will also be an important part of developing R. Intestinalis therapies.

            Intracranial aneurysm (IA) is the localized dilation of blood vessels in the brain, and IA rupture is a life-threatening condition for patients [56]. Recently, the relationship between IA and the gut microbiota has gained increasing attention. Shikata’s report first suggested that oral antibiotic administration markedly decreases IA incidence in a murine model, even if the antibiotic administration is stopped before IA induction [57]. Pathological observations have revealed that macrophage infiltration is significantly decreased, and inflammatory cytokines including IL-1β, IL-6, and TNF-α synchronously decline [57]. Furthermore, a metagenome-wide association study of fecal samples from patients with IA has identified that Bacteroides, Parabacteroides, Ruminococcus, and Blautiawere are relatively enriched, and transplantation of fecal samples from patients with IA facilitates aneurysm progression in murine models [58]. Analyses including metabolic profiling have suggested a low abundance of Hungatella hatheway and diminished circulating taurine levels in both patients with IA and mouse models [58]. Promisingly, oral Hungatella hatheway treatment alleviates IA progression and rupture, and is accompanied by elevated taurine levels [58]. These protective effects are mimicked by taurine addition, thus indicating that the Hungatella hatheway-taurine pathway is crucial in IA development [58]. Hungatella hatheway or taurine supplementation might become a feasible IA intervention. Another species of gut microbiota, Campylobacter, is highly elevated in patients with ruptured IA, compared with stable IA [59]. A recent Mendelian randomization study conducted by Ma has demonstrated that Streptococcus, Adlercreutzia, Clostridia, Rhodospirillaceae, Sutterella, Victivallis, and Peptostreptococcaceae tend to promote IA, whereas Oscillospira and Paraprevotella have opposite roles [60]. Nevertheless, the causal relationship and exact mechanisms remain unclear, and animal studies and mechanistic explorations will be required in the future.

            The pathogenesis of moyamoya disease (MMD), another relatively rare intracranial artery disease, has not been thoroughly elucidated [61]. Recent studies have correlated MMD with gut microbiota alterations, although the results have been inconsistent. A 16S-sequencing study has revealed elevated Ruminococcus gnavus in 27 patients with MMD—a finding associated with elevated risk [62]. However, Takayanagi’s work has shown more compelling differences in the oral microbiota than the gut microbiota, in 16 patients with MMD compared with 15 controls [63]. A comprehensive case-control investigation including 60 patients with MMD and 60 controls [64] has observed a marked elevation in Fusobacteriota and a marked decline in Actinobacteria in patients with MMD, at the phylum level. Further analysis has indicated that Lachnoclostridium and Fusobacterium are abundant, and Bifidobacterium and Enterobacter are diminished in patients with MMD [64]. By combining these four genera, the authors have developed a promising predictive model to stratify patients with MMD [64]. In contrast to previous findings, Ruminococcus gnavus and Peptostreptococcaceae were not elevated in patients with MMD, and Peptostreptococcaceae was even higher in controls. These discrepancies might stem from differences in sample sizes, geographic origins, and analytic methods across studies. Multicenter cohorts of patients with MMD will be indispensable to overcome these limitations. Whether these alterations in the gut microbiota affect the progression of MMD remains undetermined.

            Vasculitis

            Vasculitis is a group of autoimmune diseases causing persistent vascular inflammation and injury [65]. Although the exact etiology of vasculitis remains unclear, environmental factors such as the gut microbiota are important triggers of pathogenesis [65, 66]. Kawasaki disease (KD), an acute febrile disease characterized by destruction of small and mid-sized arteries, is the leading cause of acquired heart disease in children [65, 67]. The interplay between the gut microbiota and KD has gradually been revealed [65, 68]. Microbial diversity has consistently been found to be diminished in patients with KD in various studies, although the alterations in the gut microbiota have varied, partially because of differences in study populations and methods [67, 69, 70]. Notably, Enterococcus and Helicobacter were positively associated with IL-6 levels in patients with KD, and Fusobacteria, Shigella, and Streptococcus were positively with KD activity [67, 71]. Recently, Wang’s data have revealed that gut microbiota producing SCFAs are significantly diminished in KD model mice [68]. Intriguingly, administration of Clostridium butyricum enriches SCFA-producing bacteria and abolishes the progression of KD, and is accompanied by decreases in IL-1β and IL-6, and maintenance of intestinal barrier integrity [68]. Mechanistic explorations have further demonstrated that butyrate, but not other SCFAs, contribute to the protective effects through elevated phosphatase MKP-1 and downregulated MAPK pathways [68]. Rebalanced Th17s/Tregs are another potential mechanism in the regulation of KD by SCFAs [72]. These studies have provided evidence of application of the probiotic Clostridium butyricum in children with KD to minimize KD complications such as coronary artery aneurysms. Clinical trials will be the next step in comprehensively evaluating the effects and safety of this intervention.

            Potential Therapy Targeting the Gut Microbiota and Metabolites

            Accumulating evidence indicates crosstalk between the gut microbiota and vascular dysfunction, thus extending the scope of therapy for atherosclerosis, aneurysm, and VC (Table 1) [73, 74]. Dietary modulation is a viable and acceptable means of decreasing chronic vascular inflammation [3, 15]. Specifically, the abovementioned natural products show powerful anti-inflammatory effects with minimal adverse effects [25]. According to high throughput sequencing and animal studies, direct intake of probiotics effectively inhibits atherosclerotic progression [75, 76]. However, the dosages and safety of these treatments should be further evaluated in clinical settings. Clinical evidence of probiotic treatment for VC and aneurysms remains limited. Fecal microbiota transplantation (FMT) has also gained extensive attention and is already used in the treatment of Clostridium difficile infection [77]. FMT is has been adequately demonstrated to be involved in vascular atherosclerosis, but has not been used in patients under long-term administration [78, 79]. The FMT process carries a high risk of transplanting other pathogenic microorganisms. Given that metabolites such as TMAO profoundly damage vascular walls, the development of selective TMAO inhibitors is another direction for protecting cardiac health [80, 81]. Elucidation of the interaction between the gut microbiota and vascular disease has helped develop new therapies for the management of atherosclerosis, aneurysm, and VC.

            Table 1

            Recent Findings in Links between the Gut Microbiota and Vascular Disease, from a Translational Perspective.

            Vascular dysfunctionFunctional gut microbiotaFunctional metabolitesIntervention and causal relationshipMechanismsRefs
            Atherosclerosis-Propionate200 mg/kg in Apoe −/− mice;
            500 mg twice daily in patients;
            atherosclerosis decreased
            Tregs IL-10 increased;
            Npc1l1 decreased
            [19]
            Atherosclerosis Clostridium and Peptostreptococcus decreasedIndole-3-propionic acid decreased50 mg/kg in ApoE −/− mice;
            atherosclerosis decreased
            Macrophage reverse cholesterol transport increased;
            SPI1/miR-142-5p/ABCA1 pathway decreased
            [22]
            Atherosclerosis-TMAO increasedBerberine 100 mg/kg/d, 200 mg/kg/d in HFD-induced hamsters; oral berberine 0.5 g twice daily in patients;
            atherosclerosis decreased
            Choline-TMA-TMAO production decreased[26]
            Vascular calcification Akkermansia decreasedPropionate decreased1 g/kg in VDN-treated rats;
            calcification decreased
            Intestinal barrier function increased;
            TNF-α IL-1β, and IL-6 decreased
            [43]
            Vascular calcification-Butyrate1000 mg/kg/day in vitamin D3-induced mice;
            calcification increased
            NF-κB, Wnt, and Akt signaling increased;
            HDAC expression decreased
            [44]
            Abdominal aortic aneurysm R. Intestinalis decreasedButyrate decreased R. Intestinalis (1 × 109 CFUs/ mouse) or butyrate (400 mg/kg/day) in CaCl2-induced AAA mice;
            AAA decreased
            Neutrophil infiltration and NOX2-dependent neutrophil extracellular trap formation decreased; LPS, TNF-α, IL-1β, IL-6, and MCP-1 decreased;
            synthetic phenotype changes in VSMCs decreased
            [52]
            Abdominal aortic aneurysm-TMAO increasedAng II and elastase induced AAA models treated with antibiotics, TMAO inhibitor, or FMO3 genetic ablation;
            AAA decreased
            Endoplasmic reticulum stress decreased;
            VSMC apoptosis decreased
            [55]
            Intracranial aneurysms Hungatella hathewayi
            decreased
            Taurine decreased Hungatella hathewayi 1 × 109 CFU/mouse in IA induction surgery;
            IA decreased
            MMP-2 and MMP-9 decreased;
            VSMC apoptosis decreased
            [58]
            Kawasaki diseaseSCFA-producing bacteria decreasedButyrate, acetate, and propionate decreased Clostridium butyricum (5 × 106 CFU/g) in CAWS-induced KD mice;
            KD decreased
            IL-1β and IL-6 decreased;
            intestinal barrier integrity maintained;
            phosphatase MKP-1 increased
            [68, 72]

            Conclusion and Outlook

            Emerging investigations have strongly correlated gut microbiota dysbiosis and vascular dysfunction, including atherosclerosis, aneurysms, and VC. Alterations in the gut microbiota and metabolites, and their functions, have been partially clarified through the use of sequencing technologies and bioinformatics analysis. The causal relationships between these alterations and vascular pathogenesis remain unclear. A paucity of human studies have explored signaling pathways. Given the complexity of the gut microbiota and diverse metabolites, more comprehensive research is warranted to deepen understanding of the crosstalk between the gut microbiota and host under physiological and pathological conditions. Mechanistic animal studies are also necessary. Recently, several encouraging results have identified targeted species and metabolites. Translational studies including clinical trials should be conducted to better manage vascular diseases. Dietary therapy is considered a viable and acceptable strategy to modify the gut microbiota and minimize adverse vascular events. Long-term effects and safety should be a focus of future work.

            Emerging artificial intelligence technology has been applied to establish diagnostic tools for early screening, personalized diagnosis, and risk stratification for cardiovascular diseases [82]. Analysis of the gut microbiota provides extensive information on the development of vascular disease, and the gut microbiota is anticipated to be integrated into current artificial intelligence tools to enrich understanding of vascular diseases [83], with far-reaching clinical implications.

            Conflict of Interest

            The authors declare they have no conflicts of interest.

            Citation Information

            References

            1. , , , , , , et al. Recent advances in modulation of cardiovascular diseases by the gut microbiota. J Hum Hypertens 2022;36(11):952–9.

            2. , . Gut microbiota in human metabolic health and disease. Nat Rev Microbiol 2021;19(1):55–71.

            3. , . Diet-gut microbiota interactions on cardiovascular disease. Comput Struct Biotec 2022;20:1528–40.

            4. . Gut microbiota dysbiosis: triggers, consequences, diagnostic and therapeutic options. Microorganisms 2022;10(3):578.

            5. . The changing landscape of atherosclerosis. Nature 2021;592(7855):524–33.

            6. , , , , , , et al. Estimating current and long-term risks of coronary artery in silico by fractional flow reserve, wall shear stress and low-density lipoprotein filtration rate. Biomed Phys Eng Express 2018;4:025006.

            7. , , , , , , et al. Effects of stent shape on focal hemodynamics in intracranial atherosclerotic stenosis: a simulation study with computational fluid. Front Neurol 2022;13:1067566.

            8. , , , , , , et al. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome 2019;7(1):68.

            9. , , , , , , et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun 2017;8(1):845.

            10. , , , , , , et al. Human gut microbiota in coronary artery disease: a systematic review and meta-analysis. Metabolites 2022;12(12):1165.

            11. , , , . Probiotics bring new hope for atherosclerosis prevention and treatment. Oxid Med Cell Longev 2022;2022:1–13.

            12. , , . Lactobacilli and Bifidobacterium as anti-atherosclerotic agents. Iran J Basic Med Sci 2022;25(8):934–46.

            13. , , , , , . Gut microbiota-derived TMAO: a causal factor promoting atherosclerotic cardiovascular disease? Int J Mol Sci 2023;24(3):1940.

            14. , , , , , . Trimethylamine N-Oxide generated by the gut microbiota: potential atherosclerosis treatment strategies. Curr Pharm Des 2022;28(35):2914–9.

            15. , , , , . Role of gut microbiota in the immunopathology of atherosclerosis: focus on immune cells. Scand J Immunol 2022;96(1):e13174.

            16. , , , , . Gut microbiota metabolites and risk of major adverse cardiovascular disease events and death: a systematic review and met-analysis of prospective studies. J Am Heart Assoc 2017;6(7):e004947.

            17. , , , , , , et al. Microbiota-derived short-chain fatty acids: implications for cardiovascular and metabolic disease. Front Cardiovasc Med 2022;9:900381.

            18. , , , , , , et al. The correlation of short-chain fatty acids with peripheral arterial disease in diabetes mellitus patients. Life 2022;12(10):1464.

            19. , , , , , , et al. Propionate attenuates atherosclerosis by immune-dependent regulation of intestinal cholesterol metabolism. Eur Heart J 2022;43(6):518–33.

            20. , , , , , , et al. Microbiota-derived tryptophan metabolites in vascular inflammation and cardiovascular disease. Amino Acids 2022;54(10):1339–56.

            21. , , , , , , et al. Plasma gut microbe-derived metabolites associated with peripheral artery disease and major adverse cardiac events. Microorganisms 2022;10(10):2065.

            22. , , , , , , et al. Gut microbially produced indole-3-propionic acid inhibits atherosclerosis by promoting reverse cholesterol transport and its deficiency is causally related to atherosclerotic cardiovascular disease. Circ Res 2022;131(5):404–20.

            23. , , , , , , et al. Dysbiosis of gut microbiota and metabolite phenylacetylglutamine in coronary artery disease patients with stent stenosis. Front Cardiovasc Med 2022;9:832092.

            24. , , , , , . 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.

            25. , , . Natural products against inflammation and atherosclerosis: targeting on gut microbiota. Front Microbiol 2022;13:997056.

            26. , , , , , , et al. Berberine treats atherosclerosis via a vitamine-like effect down-regulating Choline-TMA-TMAO production pathway in gut microbiota. Signal Transduct Target Ther 2022;7(1):207.

            27. , , , , , , et al. Bicyclol alleviates atherosclerosis by manipulating gut microbiota. Small 2022;18(9):2105021.

            28. , , , , , . Ginsenoside Rc ameliorated atherosclerosis via regulating gut microbiota and fecal metabolites. Front Pharmacol 2022;13:990476.

            29. . Vascular calcification: mechanisms of vascular smooth muscle cell calcification. Trends Cardiovas Med 2015;25(4):267–74.

            30. , , , , , , et al. Medial arterial calcification. J Am Coll Cardiol 2021;78(11):1145–65.

            31. , , , , . Interplay between gut microbiota, bone health and vascular calcification in chronic kidney disease. Eur J Clin Invest 2021;51(9):e13588.

            32. , , , , , . Role of gut microbiota-derived metabolites on vascular calcification in CKD. J Cell Mol Med 2021;25(3):1332–41.

            33. , , , . Contribution of gut microbiota-derived uremic toxins to the cardiovascular system mineralization. Toxins 2021;13(4):274.

            34. , , , , , , et al. Vascular calcification and the gut and blood microbiome in chronic kidney disease patients on peritoneal dialysis: a pilot study. Biomolecules 2022;12(7):867.

            35. , , , , , . Relationship between gut microbiota and vascular calcification in hemodialysis patients. Ren Fail 2023;45(1):2148538.

            36. , , . Age-associated arterial calcification: the current pursuit of aggravating and mitigating factors. Curr Opin Lipidol 2020;31(5):265–72.

            37. , , , , , . Gut microbiota as a source of uremic toxins. Int J Mol Sci 2022;23(1):483.

            38. , , , , , , et al. Indoxyl sulfate and p-cresyl sulfate promote vascular calcification and associate with glucose intolerance. J Am Soc Nephrol 2019;30(5):751–66.

            39. , , , , , , et al. Indoxyl sulfate-induced vascular calcification is mediated through altered notch signaling pathway in vascular smooth muscle cells. Int J Med Sci 2020;17(17):2703–17.

            40. , , , . Decreasing microbiota-derived uremic toxins to improve CKD outcomes. Clin Kidney J 2022;15(12):2214–9.

            41. , , , , . Higher serum trimethylamine-N-oxide levels are associated with increased abdominal aortic calcification in hemodialysis patients. Ren Fail 2022;44(1):2019–27.

            42. , , , , , , et al. 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):751–65.

            43. , , , , , , et al. Beneficial effect of the short-chain fatty acid propionate on vascular calcification through intestinal microbiota remodelling. Microbiome 2022;10(1):195.

            44. , , , , , , et al. The short-chain fatty acid butyrate accelerates vascular calcification via regulation of histone deacetylases and NF-κB signaling. Vasc Pharmacol 2022;146:107096.

            45. , , , , . Research models for studying vascular calcification. Int J Mol Sci 2020;21(6):2204.

            46. , , , , , . Abdominal aortic aneurysm: roles of inflammatory cells. Front Immunol 2021;11:609161.

            47. , , , . Pharmacological therapy of abdominal aortic aneurysm: an update. Curr Vasc Pharmacol 2018;16(2):114–24.

            48. , , , , , , et al. Gut microbiome sheds light on the development and treatment of abdominal aortic aneurysm. Front Cardiovasc Med 2022;9:1063683.

            49. , , , , , , et al. Gut dysbiosis and bacterial translocation in the aneurysmal wall and blood in patients with abdominal aortic aneurysm. Plos One 2022;17(12):e278995.

            50. , , , , , . Infected abdominal aortic aneurysm caused by Streptococcus pneumoniae diagnosed using polymerase chain reaction. Surgical Case Reports 2015;1(1):83.

            51. , , , . Group B streptococcus mycotic aneurysm of the abdominal aorta: report of a case and review of the Literature. Yale J Biol Med 2012;85(1):97–104.

            52. , , , , , , et al. Gut microbiome dysbiosis contributes to abdominal aortic aneurysm by promoting neutrophil extracellular trap formation. Cell Host Microbe 2022;30(10):1450–63.

            53. , , , , , . The gut microbiota in experimental abdominal aortic aneurysm. Front Cardiovasc Med 2023;10:1051648.

            54. , , , , , , et al. Gut microbiota influence the development of abdominal aortic aneurysm by suppressing macrophage accumulation in mice. Hypertension 2022;79(12):2821–9.

            55. , , , , , , et al. Gut microbiota-derived trimethylamine N-oxide contributes to abdominal aortic aneurysm through inflammatory and apoptotic mechanisms. Circulation 2023;147(14):1079–96.

            56. , . Unruptured intracranial aneurysms: development, rupture and preventive management. Nat Rev Neurol 2016;12(12):699–713.

            57. , , , , , , et al. Potential influences of gut microbiota on the formation of intracranial aneurysm. Hypertension 2019;73(2):491–6.

            58. , , , , , , et al. Alterations of gut microbiota contribute to the progression of unruptured intracranial aneurysms. Nat Commun 2020;11(1):3218.

            59. , , , , , , et al. Dysbiosis of gut microbiome is associated with rupture of cerebral aneurysms. Stroke 2022;53(3):895–903.

            60. , , , , , , et al. Association of gut microbiome with risk of intracranial aneurysm: a mendelian randomization study. BMC Neurol 2023;23(1):269.

            61. , , , , , . Efficacy assessment of superficial temporal artery-middle cerebral artery bypass surgery in treating moyamoya disease from a hemodynamic perspective: a pilot study using computational modeling and perfusion imaging. Acta Neurochir (Wien) 2023;165(3):613–23.

            62. , , , , , , et al. Increased abundance of Ruminococcus gnavus in gut microbiota is associated with moyamoya disease and non-moyamoya intracranial large artery disease. Sci Rep 2022;12(1):20244.

            63. , , , , , , et al. Higher abundance of Campylobacter in the oral microbiome of Japanese patients with moyamoya disease. Sci Rep 2023;13(1):18545.

            64. , , , , , , et al. Gut microbiota in adults with moyamoya disease: characteristics and biomarker identification. Front Cell Infect Microbiol 2023;13:1252681.

            65. , , . Findings on the relationship between intestinal microbiome and vasculitis. Front Cell Infect Microbiol 2022;12:908352.

            66. , . An update on the microbiome in vasculitis. Curr Opin Rheumatol 2021;33(1):15–23.

            67. , , , , , , et al. Altered gut microbiota correlated with systemic inflammation in children with Kawasaki disease. Sci Rep 2020;10(1):14525.

            68. , , , , , , et al. The reduced SCFA-producing gut microbes are involved in the inflammatory activation in Kawasaki disease. Front Immunol 2023;14:1124118.

            69. , , , , . Effects of changes on gut microbiota in children with acute Kawasaki disease. Peer J 2020;8:e9698.

            70. , , . Alteration of the oral and gut microbiota in patients with Kawasaki disease. Peer J 2023;11:e15662.

            71. , , , , , , et al. Correlation of gut microbial compositions to the development of Kawasaki disease vasculitis in children. Future Microbiol 2020;15:591–600.

            72. , , , , . Our evolving understanding of Kawasaki disease pathogenesis: role of the gut microbiota. Front Immunol 2020;11:1616.

            73. , , , , , , et al. The gut microbiota (microbiome) in cardiovascular disease and its therapeutic regulation. Front Cell Infect Microbiol 2022;12:903570.

            74. , , , , , , et al. Transplantation of fecal microbiota rich in short chain fatty acids and butyric acid treat cerebral ischemic stroke by regulating gut microbiota. Pharmacol Res 2019;148:104403.

            75. , , , , , , et al. Lactobacillus acidophilus ATCC 4356 attenuates the atherosclerotic progression through modulation of oxidative stress and inflammatory process. Int Immunopharmacol 2013;17(1):108–15.

            76. , , , , . Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe-/- mice. Circulation 2016;133(24):2434–46.

            77. , , , , , , et al. Fecal microbiota transplantation: review and update. J Formos Med Assoc 2019;118:S23–31.

            78. , , , , , , et al. Fecal microbiota transplantation ameliorates atherosclerosis in mice with C1q/TNF-related protein 9 genetic deficiency. Exp Mol Med 2022;54(2):103–14.

            79. , , , , , , et al. Transmission of atherosclerosis susceptibility with gut microbial transplantation. J Biol Chem 2015;290(9):5647–60.

            80. , , , , , , et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med 2018;24(9):1407–17.

            81. , , , , , , et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 2015;163(7):1585–95.

            82. , , , , , , et al. Healthcare big data in Hong Kong: development and implementation of artificial intelligence-enhanced predictive models for risk stratification. Curr Probl Cardiol 2023;21:102168.

            83. , , , , , et al. A possible link between gut microbiome composition and cardiovascular comorbidities in psoriatic patients. J Pers Med 2022;12(7):1118.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            24 January 2024
            : 9
            : 1
            : e990
            Affiliations
            [1] 1Department of Gastroenterology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
            [2] 2Translational Medicine Center, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, China
            [3] 3Clinical Center for HIV/AIDS, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
            [4] 4Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
            Author notes
            Correspondence: Yidong Wang, Department of Cardiology, State Key Laboratory of Transvascular Implantation Devices, Provincial Key Laboratory of Cardiovascular Research, The Second Affliated Hospital Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou 310009, P.R. China, E-mail: wangyidong@ 123456zju.edu.cn

            aJiaqi Wu and Yuxuan Li contributed equally to this work.

            Article
            cvia.2023.0090
            10.15212/CVIA.2023.0090
            973a0a1d-3d2c-4ef4-9366-c85a5266ec90
            Copyright © 2024 Cardiovascular Innovations and Applications

            This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 Unported License (CC BY-NC 4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. See https://creativecommons.org/licenses/by-nc/4.0/.

            History
            : 24 August 2023
            : 05 November 2023
            : 21 November 2023
            Page count
            Figures: 1, Tables: 1, References: 83, Pages: 11
            Funding
            Funded by: National Natural Science Foundation of China
            Award ID: 82100500
            This work was supported by the National Natural Science Foundation of China (grant no.82100500).
            Categories
            Review Article

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            aneurysms,Gut microbiota,therapy,vascular calcification,atherosclerosis

            Comments

            Comment on this article