Next Article in Journal
The Association Between the Mediterranean Diet and Fatty Acids in Red Blood Cells of Spanish Adolescents
Previous Article in Journal
Effects of Two Compound Probiotic Formulations on Gastrointestinal Symptoms and Gut Microbiota: A 4-Week Randomized, Double-Blind Intervention Trial
Previous Article in Special Issue
Distinct Effects of Wheat and Black Bean Consumption on Postprandial Vascular Responses in People with Arterial Stiffness: A Pilot Randomized Cross-Over Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Gut Microbiota to Combat Vascular Aging and Cardiovascular Disease: Mechanisms and Therapeutic Potential

Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2025, 17(17), 2887; https://doi.org/10.3390/nu17172887
Submission received: 9 June 2025 / Revised: 22 August 2025 / Accepted: 4 September 2025 / Published: 6 September 2025
(This article belongs to the Special Issue Nutritional Strategies for Arterial Health)

Abstract

Vascular aging, characterized by arterial thickening, reduced elasticity, and endothelial dysfunction, significantly compromises vascular health and accelerates the progression of cardiovascular diseases (CVDs). Emerging research highlights the gut–vascular axis as a critical mediator of vascular health, with the gut microbiota (GM) playing a pivotal role in modulating vascular aging and CVDs. This review presents a thorough and up-to-date discussion of the fundamental mechanisms driving vascular aging and explores how GM and its metabolites influence these processes. Furthermore, we place particular emphasis on therapeutic strategies, including probiotics and food-derived natural products, that foster beneficial bacterial growth and support cardiovascular health, while also exploring the underlying mechanisms. By targeting GM composition and function, these approaches offer promising strategies to mitigate vascular aging and lower CVD risk. Future studies aimed at deepening our understanding of the interplay between GM, its metabolites, and vascular health could pave the way for novel preventive and therapeutic interventions against vascular aging and CVDs.

1. Introduction

Cardiovascular diseases (CVDs) encompass a wide range of heart and blood vessel disorders, such as coronary artery disease, stroke, rheumatic heart disease, and other vascular conditions. They remain the leading cause of death and disability worldwide, accounting for an estimated 17.9 million deaths each year—approximately one-third of all global deaths [1]. According to the reports from Centers for Diseases Control, 919,032 people died from CVDs in 2023. The economic burden is also substantial, with heart disease costing an estimated $417.9 billion between 2020 and 2021, including expenses for medical care, prescription drugs, and productivity losses. Vascular aging, characterized by arterial thickening, plaque formation, and reduced elasticity, contributes significantly to the rising prevalence of CVDs and accelerates the progression of conditions such as coronary heart disease, hypertension, and stroke [2,3]. Recent research has highlighted gut microbiota (GM) as a critical factor in vascular health, with dysbiosis contributing to the onset and progression of various CVDs [4,5]. Imbalances in gut bacteria can promote vascular aging, while beneficial microbes play a protective role in cardiovascular health.
Probiotics from beneficial bacterial strains like Lactobacillus, Bifidobacterium, and Faecalibacterium show promise in mitigating vascular aging and CVDs [6,7,8]. These probiotics enhance immune function, reduce inflammation, and improve endothelial health. Additionally, natural products, such as dietary fibers and phytochemicals, support cardiovascular health by modulating the GM composition and maintaining intestinal balance [9]. Targeting the GM with these compounds offers a potential strategy to combat vascular aging and reduce CVD risk in aging populations. This review examines the role of GM in vascular aging and CVDs, emphasizing the therapeutic potential of probiotics and natural products.

2. Mechanisms of Vascular Aging and CVDs

Vascular aging is a complex biological process characterized by structural and functional changes in the blood vessels that occur with chronological and biological aging, which leads to increased stiffness, endothelial dysfunction, and a higher risk of cardiovascular diseases [10]. While these changes are age-related, they can be accelerated by pathological conditions such as chronic inflammation, oxidative stress, and metabolic disorders. Understanding these underlying mechanisms of vascular aging is crucial before exploring the potential role of GM in vascular aging. In the following sections, we will discuss the key mechanisms contributing to vascular aging.

2.1. Inflammation and Oxidative Stress

Vascular inflammation, or “inflammaging”, is a hallmark of vascular aging, characterized by elevated pro-inflammatory cytokines such as Tumor Necrosis Factor alpha (TNF-α) and interleukins (IL-1β, IL-6, IL-8, IL-13, IL-18) [11]. This leads to endothelial dysfunction and impaired vasodilation in aging arteries [12] (Figure 1i). A major driver of vascular inflammation is oxidative stress, where reactive oxygen species (ROS) react with nitric oxide (NO), reducing its availability and impairing vasodilation. ROS also trigger lipid peroxidation, DNA damage, and protein oxidation [13,14]. A key consequence of oxidative stress is the age-related decline in NO bioavailability, driven by multiple factors, including increased arginase activity, which depletes L-arginine (the substrate for NO synthesis), and the loss of tetrahydrobiopterin (BH4), an essential cofactor for nitric oxide synthase (NOS) [15,16]. Oxidative stress activates Nuclear Factor kappa B (NFκB), upregulating pro-inflammatory genes and matrix metalloproteinases (MMPs), thereby compromising extracellular matrix integrity and vascular homeostasis [17]. The NLR family pyrin domain containing 3 (NLRP3) inflammasome further amplifies inflammaging by promoting IL-1β and IL-18 production [18].

2.2. Mitochondrial Dysfunction

Vascular aging is associated with impaired mitochondrial function, resulting in reduced electron transport efficiency and ATP synthesis, which contribute to aging-related vascular diseases [20,21]. A major contributor to this dysfunction is the excessive production of mitochondrial reactive oxygen species (mtROS) by pro-oxidative enzymes such as NADPH oxidase (NOX) [22,23]. Elevated mtROS depletes antioxidants, damages mitochondrial DNA (mtDNA), and disrupts mitochondrial homeostasis (Figure 1ii). The depletion of antioxidants like glutathione (GSH) and Mn-SOD occurs due to peroxynitrite-mediated inhibition, weakening oxidative defense mechanisms [24]. Simultaneously, the Nrf-2 signaling pathway, crucial for antioxidant responses, is compromised, further exacerbating oxidative stress [9]. Notably, the deletion of p66Shc has been shown to reduce oxidative stress and mitigate endothelial dysfunction, highlighting its role in vascular aging [25].
Aging cells also exhibit high mtDNA mutation rates, which drive increased ROS production, apoptosis, and senescence [26,27,28]. Additionally, mitochondrial biogenesis is impaired, as indicated by reduced levels of PGC-1α and TFAM, key regulators of mitochondrial function [29,30]. Dysfunctional mitochondria accumulate in aged vascular cells due to insufficient mitophagy, imbalanced fission-fusion dynamics, and decreased oxygen consumption [31,32,33]. The decline of SIRT1, a key regulator of mitochondrial quality control, further exacerbates these age-related mitochondrial impairments [34].

2.3. Loss of Proteostasis

Proteostasis ensures proper protein synthesis, folding, and degradation through molecular chaperones and systems like the ubiquitin-proteasome system (UPS) and autophagy [35]. With aging, this network malfunctions, leading to misfolded protein accumulation and cytotoxic aggregates (Figure 1iii) [36]. Molecular chaperones, such as heat shock proteins (HSPs), prevent protein aggregation, particularly under stress [37,38,39]. Hsp60 increases in atherosclerosis, while mitochondrial Hsp90 accumulation is linked to pulmonary hypertension and vascular remodeling [40,41]. However, Hsp70 inducibility declines in aging aortic tissues, potentially impacting vascular health [42,43,44]. The UPS and autophagy pathways degrade misfolded proteins, but their function declines with age [45]. While proteasome activity is elevated in atherosclerotic plaques as a compensatory response, it eventually decreases, contributing to vascular cell senescence, oxidative stress, and inflammation [46,47]. Dysregulated autophagy, including mitophagy, further accelerates vascular aging, as deletion of key autophagy-related genes increases susceptibility to endothelial dysfunction [48,49,50,51]. Maintaining proteostasis is therefore essential for vascular health, and its disruption plays a significant role in vascular aging.

2.4. Cellular Senescence, Apoptosis, and Necroptosis

Vascular cells undergo senescence due to chronic stressors like DNA damage, oxidative stress, and telomere dysfunction, contributing to disease progression through the pro-inflammatory senescence-associated secretory phenotype (SASP) (Figure 1iv) [52,53]. Senescence markers p16, p21, and p53 are elevated in atherosclerotic plaques of older adults [54,55]. Senescent endothelial cells (ECs) increase oxidative stress and reduce NO bioavailability, leading to endothelial dysfunction, impaired vascular tone, and mitochondrial dysfunction [56,57]. Similarly, vascular smooth muscle cell (VSMC) senescence promotes atherosclerosis, hypertension, and diabetes by increasing arterial stiffness, inflammation, and vascular calcification [58,59]. Aging also disrupts programmed cell death, including apoptosis and necroptosis [60]. Aged ECs show impaired NO synthesis and heightened apoptosis, while in vascular injury, increased VSMC proliferation resists apoptotic signals, leading to excessive neointima formation [61]. Necroptosis, marked by cell rupture and DAMP release, activates inflammation and is linked to atherosclerotic plaques and AAA, playing a key role in inflammaging [62,63,64,65,66,67,68].

3. Key Mediators of Gut–Vascular Communication in Vascular Aging and Cardiovascular Health

The GM influences the cardiovascular system, often through direct communication via microbial signals crossing the intestinal epithelium. These signals include structural components like lipopolysaccharides (LPS) and peptidoglycans, which interact with host cells via pattern recognition receptors, as well as metabolites such as trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and bile acids (BAs). In the following, we will explore how these microbial components and metabolites influence vascular aging by modulating key factors such as inflammation, oxidative stress, and mitochondrial function (Figure 2).

3.1. Trimethylamine N-Oxide (TMAO)

TMAO is a metabolite produced by GM from dietary nutrients such as choline, phosphatidylcholine, and L-carnitine. Certain gut bacteria, including Desulfovibrio and Clostridium species, harbor trimethylamine (TMA) lyases that convert these dietary precursors into TMA [69]. TMA is absorbed into the bloodstream and oxidized to TMAO by hepatic flavin monooxygenase enzymes, primarily FMO3 [70].
Elevated TMAO levels are strongly linked to CVDs, contributing to inflammation, oxidative stress, endothelial dysfunction, atherosclerosis, VSMCs proliferation, and platelet activation [71,72,73,74,75]. Specifically, TMAO promotes vascular aging by disrupting endothelial nitric oxide synthase (eNOS), stimulating leukocyte recruitment, and increasing adhesion molecule expression, leading to impaired vasodilation and vascular remodeling. TMAO causes mutations in mitochondrial DNA, disrupting oxidative phosphorylation (OXPHOS), raising ROS levels, and lowering ATP/ADP ratios. This results in mitochondrial dysfunction, increased cell death [76]. Additionally, TMAO influences cholesterol and bile acid metabolism and enhances foam cell formation via scavenger receptors like cluster of differentiation 36 (CD36) and steroid receptor RNA activator 1 (SRA1) on macrophages [77].
Although TMAO is closely associated with adverse cardiovascular events, its role as a causative factor versus a biomarker remains debated. TMAO has been shown to amplify angiotensin II-induced hypertension but does not affect normal blood pressure [78], leaving its causative role in question. Despite this, targeting TMAO pathways is under exploration as a potential therapeutic strategy to mitigate vascular aging and cardiovascular risk.

3.2. Short-Chain Fatty Acids (SCFAs)

SCFAs are produced by gut bacteria like Faecalibacterium prausnitzii, Bifidobacterium, and Akkermansia muciniphila during the fermentation of fiber. The main SCFAs, acetate, propionate, and butyrate, have emerged as key modulators in preventing and managing vascular aging and its associated CVDs. These metabolites exert their effects through various biological processes, including anti-inflammatory and antioxidant activities, regulation of endothelial function, blood pressure control, and mitochondrial health.
SCFAs help mitigate the chronic inflammation and oxidative stress that characterize vascular aging. By activating free fatty acid receptors (FFA2 and FFA3), SCFAs modulate immune cell function, promoting anti-inflammatory responses [79]. These receptors, found on immune cells like dendritic cells, macrophages, and T cells, help decrease pro-inflammatory cytokine production and foster regulatory immune cell phenotypes, reducing the inflammatory environment in the vasculature. Butyrate, in particular, is known to inhibit histone deacetylases (HDACs), which modulate gene expression related to inflammation and oxidative stress, further dampening inflammatory pathways and promoting vascular health [80,81,82].
In addition to their anti-inflammatory properties, SCFAs also support mitochondrial function and endothelial health. Butyrate enhances endothelial NO production [80], which is crucial for vascular dilation and overall endothelial function. Propionate has been linked to a reduction in blood pressure, especially in hypertensive models [83]. Furthermore, SCFAs like acetate provide energy to vascular cells [84,85], potentially aiding in the maintenance of cellular function and reducing age-related vascular decline.

3.3. Lipopolysaccharide (LPS)

LPS, produced by Gram-negative bacteria such as Escherichia coli, Bacteroides fragilis, and Enterobacter cloacae, links GM to vascular aging by driving chronic inflammation, oxidative stress, and cellular dysfunction. Elevated LPS levels activate TLR4 on ECs, immune cells, and VSMCs, leading to ROS production and pro-inflammatory cytokines [86,87]. These responses impair mitochondrial function, disrupt proteostasis, and induce cellular senescence.
LPS-triggered inflammation promotes endothelial dysfunction by reducing NO bioavailability, increasing leukocyte adhesion, and inducing vascular stiffness and fibrosis [88,89]. Oxidative stress from LPS further exacerbates mitochondrial dysfunction, promoting apoptosis and necroptosis in vascular cells, accelerating tissue damage. Recruitment of monocytes and macrophages into arterial walls under LPS influence contributes to foam cell formation and atherosclerosis [90,91]. LPS also activates platelets, increasing thrombotic risks [92,93], a hallmark of vascular aging and CVDs. Targeting GM producing LPS, such as Escherichia coli and Bacteroides fragilis, offers potential therapeutic strategies against vascular-related diseases.

3.4. Bile Acids (BAs)

BAs are amphipathic cholesterol metabolites produced in the liver and modified by GM, including Clostridium, Bacteroides, and Lactobacillus. These bacteria transform primary BAs into secondary BAs such as deoxycholic acid (DCA) and lithocholic acid (LCA), which significantly influence vascular aging.
Several mechanisms link BAs to vascular dysfunction, primarily through inflammation, oxidative stress, and cellular damage. In ECs, BAs such as chenodeoxycholic acid promote ROS production, activating NF-κB and p38 MAPK signaling pathways, leading to increased adhesion molecule expression and inflammation [94]. Hydrophobic BAs exacerbate oxidative stress in VSMCs, causing tissue damage and remodeling [95].
GM-modulated BAs also affect vascular calcification and cardiac function. Hydrophobic BAs like DCA contribute to vascular calcification by inducing VSMCs trans-differentiation through calcium-phosphate metabolism. Elevated BA levels disrupt vascular tone via nuclear and G-protein-coupled receptors, including the farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5), with detrimental effects on endothelial function and arterial stiffness [96]. Moreover, chronic exposure to high concentrations of hydrophobic BAs, influenced by dysbiosis, increases arrhythmia risk by modulating ion channels in cardiac cells and altering electrical excitability [97]. These effects are further amplified by the inflammatory and oxidative stress environment driven by BA dysregulation, contributing to the progression of atherosclerosis and heart failure.
In conclusion, the interplay between GM and vascular aging forms a dynamic system where microbial metabolites and components significantly impact cardiovascular health. Gut-derived metabolites such as TMAO, SCFAs, LPS, and BAs modulate key factors like inflammation and oxidative stress, playing dual roles in either accelerating or mitigating vascular aging processes.

4. Utilization of Beneficial Gut Bacteria as Probiotics in Mitigating Vascular Aging and Enhancing Vascular Health

GM consists of beneficial bacteria that naturally reside in the human gut and play a role in mitigating vascular aging. Some of these bacterial strains are now being investigated as probiotics with potential to support vascular health. These probiotics offer direct benefits, such as immune modulation, anti-inflammatory effects, maintenance of endothelial function, regulation of blood pressure, and improvements in metabolic health. A summary of these probiotic strains and their effects is presented in Table 1 [98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130].
Specific probiotics, such as Lactobacillus, Bifidobacterium, and Faecalibacterium strains, modulate immune responses by balancing pro-inflammatory and anti-inflammatory activities, helping to alleviate vascular aging [104,131,132,133,134,135,136,137]. For example, Lactobacillus plantarum 299v supplementation reduces IL-8, IL-12, and leptin levels, independent of traditional risk factors and TMAO changes [104]. Bifidobacterium bifidum PRL2010 increases IL-12 in the kidneys and reduces IL-10 in the liver and kidneys in ischemia/reperfusion injury models [138], while Faecalibacterium prausnitzii reduces CD45-positive leukocytes in subcutaneous adipose tissue [139]. Certain Lactobacillus and Bifidobacterium species also enhance endothelial function and regulate blood pressure. Lactobacillus helveticus, for example, lowers blood pressure in hypertensive individuals by producing bioactive peptides that inhibit ACE [121,140,141]. Probiotics also show potential for improving metabolic health, which can reduce vascular aging and cardiovascular diseases. Lactobacillus casei improves lipid profiles by lowering total cholesterol, LDL cholesterol, and triglycerides [142]. Lactobacillus plantarum enhances insulin sensitivity, glucose metabolism, and lipid profiles [143,144], while Lactobacillus acidophilus improves insulin sensitivity and reduces total and LDL cholesterol [145,146,147,148]. Bifidobacterium breve and Bifidobacterium lactis benefit glucose metabolism and insulin sensitivity [149,150,151].

5. Nutrient-Derived Natural Products for Modulating GM and Mitigating Vascular Aging

Nutrient-derived natural products have been integral to managing chronic diseases, including CVDs, due to their diverse pharmacological properties [108,152,153]. Functional foods rich in bioactive compounds provide cardioprotective benefits by mitigating vascular aging through mechanisms such as anti-inflammatory and antioxidant effects, improving endothelial function, regulating blood pressure, and optimizing lipid metabolism.
Recent research underscores the critical interplay between GM and these natural products. After ingestion, compounds like dietary fiber, flavonoids, polyphenols, and alkaloids undergo metabolic transformations by GM, including fermentation, deglycosylation, oxidation, and conjugation. These processes yield secondary metabolites with modified bioactivity, which can influence their therapeutic efficacy. Furthermore, natural products contribute to maintaining microbial diversity, restoring GM balance, and enhancing nutrient absorption, indirectly promoting vascular health (Figure 3).
Given the emerging role of GM in vascular aging, ongoing studies aim to identify natural products that can modulate GM to delay age-related vascular changes and elucidate their underlying mechanisms. In the following, this review explores the potential of food-derived phytochemicals (Table 2: Chemical structure and food source of these natural products) to simultaneously impact GM and vascular health, emphasizing GM-targeted natural compounds as promising anti-vascular aging agents.

5.1. Flavonoids

Flavonoids, naturally occurring phenolic compounds found in fruits, vegetables, and plant-based foods, have been shown to offer numerous health benefits, particularly in cardiovascular health [154]. These compounds, present in foods like berries, citrus fruits, apples, onions, broccoli, green tea, cocoa, and red wine, include well-known flavonoids such as quercetin, epigallocatechin-3-gallate (EGCG), and kaempferol. Increasing evidence suggests that flavonoids can modulate GM, highlighting their potential as natural interventions against vascular aging through the GM. By targeting pathways such as SCFAs production, oxidative stress, and inflammation, flavonoids bridge the gut–vascular axis.
Quercetin, a well-studied flavanol, quercetin, is abundant in apples, berries, onions, citrus fruits, and leafy greens. It has prebiotic-like effects, promoting beneficial gut bacteria such as Bifidobacterium and Lactobacillus while inhibiting pathogens like Escherichia coli [155,156]. Quercetin enhances the production of SCFAs, reduces TMAO levels, and regulates BA metabolism [157,158,159]. These actions mitigate inflammation and oxidative stress, factors integral to vascular aging. Quercetin’s vascular benefits include alleviating endothelial dysfunction, reducing VSMCs senescence, and combating atherosclerosis by modulating microbial populations, such as increasing Firmicutes and decreasing Verrucomicrobia [160,161,162].
Epigallocatechin-3-Gallate (EGCG), predominantly found in green tea, cocoa, and grapes, alters GM associated with obesity and diabetes. It increases the Firmicutes-to-Bacteroidetes ratio, promotes beneficial bacteria like Lactobacillus and Akkermansia, and reduces harmful Desulfovibrionaceae [163,164]. By addressing chronic inflammation, oxidative stress, and endothelial dysfunction, common pathways in diabetes and vascular aging, EGCG aids vascular health. It enhances SCFA levels, inhibits vascular permeability via matrix metallopeptidase 9 (MMP-9) suppression, and improves endothelial function through the nuclear factor erythroid 2-related factor 2 (Nrf2)/cysteine-aspartic acid protease 3 (Caspase-3) pathway [165,166,167].
Kaempferol, present in apple skins, kale, spinach, celery, and broccoli, kaempferol restores gut flora balance in conditions like obesity, diabetes, and colitis. It reverses high-fat diet-induced changes in GM, such as increasing Bacteroidetes and reducing Firmicutes, and modulates metabolites linked to energy production and bile acid metabolism [168,169]. In vascular systems, kaempferol protects against oxidative and inflammatory damage via the Nrf2/heme oxygenase 1 (HO-1) pathway and prevents endothelial apoptosis through autophagy regulation via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway [170,171].

5.2. Polyphenols

Polyphenols, including resveratrol, chlorogenic acid, ellagic acid (EA), ferulic acid, and secoisolariciresinol diglucoside (SDG), have garnered increasing attention for their potential role in managing vascular aging and CVDs, particularly through modulation of GM [172]. As vascular aging is characterized by increased arterial stiffness, which impairs the ability of arteries to expand and contract with pressure changes, polyphenols present a promising strategy to target GM-related metabolism and mitigate these age-related changes.
Resveratrol, a stilbene found in foods such as peanuts, grapes, and berries, has been shown to interact with GM to protect against aging and age-related CVDs [173,174,175]. Resveratrol supplementation increases populations of beneficial bacteria like Akkermansia, Lactobacillus, and Bifidobacterium, while reducing the Firmicutes-to-Bacteroidetes ratio and inhibiting the growth of Enterococcus faecalis [173,174,175,176,177,178]. Resveratrol also enhances SCFAs production, modulates BA metabolism, and reduces TMAO levels [179], all of which helps to reduce vascular inflammation and oxidative stress, crucial factors in vascular aging. Additionally, resveratrol directly benefits vascular health by enhancing NO production, upregulating endothelial nitric oxide synthase (eNOS), reducing endothelin-1 synthesis, inhibiting VSMCs proliferation, and protecting against arterial stiffness and vascular remodeling [180,181].
Chlorogenic acid, a phenolic acid found in coffee, apples, and blueberries, selectively modulates GM to improve metabolic functions [182,183,184,185]. In animal models, chlorogenic acid increases beneficial bacteria such as Akkermansia and enhances SCFA production [184,185]. Chlorogenic acid also activates the Nrf2 pathway in endothelial cells, improving endothelial function and reducing oxidative damage from oxLDL [186,187]. Furthermore, it exhibits antihypertensive effects and delays vascular senescence through the Nrf2/HO-1 pathway [188,189].
Ellagic acid (EA), found in berries, pomegranates, and nuts, influences GM composition by increasing Lactobacillus and reducing Escherichia coli populations [190]. EA is converted into bioavailable urolithins by GM [191], and these derivatives have been studied for their anti-aging and anti-CVD potential. EA and urolithins regulate VSMCs proliferation, endothelial cell dysfunction, lipid metabolism, and calcium handling, providing a multifaceted approach to preventing CVDs [192,193,194].
Ferulic acid, present in whole grains, fruits, vegetables, and coffee, acts as a prebiotic, promoting the growth of beneficial gut bacteria like Olsenella and Faecalibaculum while producing SCFAs [195]. Ferulic acid is also a potent antioxidant and anti-inflammatory agent that protects against endothelial dysfunction and may help lower blood pressure [196]. By modulating GM and lipid metabolism through the AMP-activated protein kinase α (AMPKα)/sterol regulatory element binding protein 1 (SREBP1)/acetyl-CoA carboxylase 1 (ACC1) pathway, ferulic acid has been shown to improve atherosclerotic injury in animal models [197].
Secoisolariciresinol diglucoside (SDG), found in flaxseeds and whole grains, has significant anti-inflammatory effects, partly attributed to its modulation of GM [198]. SDG influences the diversity and composition of GM, promoting bacteria that produce SCFAs while decreasing inflammation-related bacteria. It has been shown to slow the progression of atherosclerosis and suppress endothelial inflammation through inhibition of the Akt/IκB/NF-κB pathway [199].

5.3. Dietary Fiber

Dietary fiber, which is resistant to digestion in the upper gastrointestinal tract and undergoes fermentation in the colon, where it supports the expansion of beneficial microbes. This fermentation process promotes the growth of beneficial bacteria, such as Bifidobacterium and Lactobacillus species, which are known to contribute to gut health by supporting intestinal integrity and reducing inflammation, preventing CVDs and mitigating the effects of vascular aging.
Inulin is a type of dietary fiber found in plants like chicory root, Jerusalem artichoke, garlic, onions, asparagus, and bananas. In the colon, inulin is fermented by beneficial bacteria, producing SCFAs like butyrate [200]. These SCFAs help nourish colon cells, enhance intestinal barrier integrity, and exert anti-inflammatory effects. Additionally, inulin can restore microbiota balance and reduce microbiota–mucosa separation under high-fat diet conditions [201,202]. Studies also suggest that inulin improves vascular health, with effects such as improved endothelial function and reduced atherosclerotic lesions in animal models [203,204]. Inulin supplementation reverses endothelial dysfunction in mesenteric and carotid arteries by activating the NOS pathway, influenced by GM composition and BA production [205].
Pectin, found in the cell walls of fruits such as citrus, apples, and pears, is a polysaccharide rich in galacturonic acid. Although indigestible by human enzymes, pectin can be fermented by gut bacteria to produce SCFAs. Pectin supplementation influences GM diversity, stimulating the growth of beneficial bacteria like Bacteroides and Eubacterium [206,207]. Although research on pectin’s vascular effects is limited, recent studies suggest that high-esterified pectin supplementation can improve blood pressure, reduce heart lipid content, and regulate cardiac gene expression in rat models [208].
β-Glucan, a soluble fiber found in mushrooms, yeast, oats, and barley, has various health benefits, including antioxidant effects, blood sugar regulation, cholesterol reduction, and immune support [209]. β-Glucan consumption has been linked to increased levels of beneficial bacteria, including Akkermansia, Lactobacillus, and Lachnospiraceae [209]. Research indicates that β-glucan positively modulates vascular function in both animal models and human studies, improving outcomes such as blood pressure and GM composition [210,211,212,213,214]. In atherosclerotic mice and hypercholesterolemic human subjects, β-glucan supplementation decreased body weight, blood pressure, and modified GM, promoting the production of anti-inflammatory metabolites.

5.4. Alkaloids

Alkaloids are a group of nitrogen-containing compounds found in plants, fungi, bacteria, and marine organisms, with caffeine and capsaicin being notable examples. Research has explored their pharmacological effects on CVDs and, more recently, the role of GM in mediating these effects.
Caffeine, a widely consumed substance in coffee, tea, chocolate, and soft drinks, caffeine’s impact on GM is still under study, but it has been shown to influence gut motility and acid production [215,216]. Some studies suggest that caffeine increases the abundance of beneficial bacteria, such as Faecalibacterium and Roseburia, while decreasing harmful bacteria like Erysipelatoclostridium [217]. In metabolic syndrome mouse models, caffeine improved the profile of SCFAs and BA metabolism [218,219]. Caffeine has well-established vascular benefits, including enhancing endothelial function through NO production, and may protect against cognitive decline in older women with vascular disordersts anti-inflammatory, antioxidant, and cardioprotective effect [220,221,222].
Capsaicin, primarily found in chili peppers, is a widely used spice known for its analgesic properties. Studies show that it can alter GM composition, increasing the abundance of bacteria such as Faecalibacterium, Akkermansia, and Roseburi [223]. In addition to modulating GM, capsaicin has been linked to vascular health, with potential benefits in preventing vascular aging in diabetes by reducing EC senescence [224]. Capsaicin also protects against arterial calcification and improves endothelial function [225,226], suggesting its role in cardiovascular protection and anti-aging effects.

5.5. Phytosterols

Phytosterols are plant-derived steroids with a similar structure to cholesterol, primarily found in vegetable oils, nuts, seeds, and whole grains. They play a vital role in maintaining the stability of cellular membranes. Consumption of phytosterols helps lower LDL cholesterol by competing with cholesterol for absorption in the digestive system, making them popular ingredients in functional foods like margarine, spreads, and yogurt. Studies suggest that GM may prefer phytosterols over cholesterol as a substrate [227], which could influence GM composition and contribute to cardiovascular health.
β-Sitosterol, the most abundant phytosterol, is found in plant oils, nuts, seeds, and grains. It has been shown to support cardiovascular health by reducing atherosclerotic plaques in mice [228,229,230], improving gut health by increasing beneficial bacteria like Bifidobacterium and Lactobacillus, and decreasing harmful bacteria such as Desulfovibrionaceae [231,232]. β-Sitosterol also has direct vascular effects, inhibiting vascular smooth muscle cell proliferation through the AMPK and mTOR pathways [233].
Stigmasterol, found in legumes, nuts, whole grains, and vegetables, has emerged for its hepatoprotective effects, particularly by remodeling GM [234,235,236]. It alters GM diversity, increasing beneficial bacteria and improving the balance of regulatory T cells to CD8+ T cells [235]. Stigmasterol also improves liver function by modulating bile acid profiles and reversing gut dysbiosis induced by a high-fat diet [236]. Vascular protection from stigmasterol includes inhibiting the proliferation of smooth muscle cells and ECs through various signaling pathways, including the MAPK pathway [237].
Overall, the impact of food-based natural products on gut microbiota, their derived metabolites, and mechanisms associated with vascular aging is summarized in Table 3 [160,162,174,205,206,208,224,233,238,239,240,241,242,243,244,245,246,247,248]. Table 4 [177,197,208,214,249,250,251,252,253,254,255,256,257,258,259,260,261,262,263,264,265] provides an overview of their effects on cardiovascular diseases, including atherosclerosis, hypertension, and abdominal aortic aneurysm (AAA), through gut microbiota modulation. These findings highlight the promising potential of incorporating gut microbiota-targeted dietary strategies into therapeutic approaches for managing vascular aging and preventing cardiovascular diseases.

6. Conclusions and Perspectives

Vascular aging is a complex process shaped by inflammation, oxidative stress, mitochondrial dysfunction, and cellular senescence. Emerging research emphasizes the central role of GM in these processes, highlighting a dynamic gut–vascular axis that significantly influences cardiovascular health. Dysbiosis exacerbates vascular aging by inducing oxidative stress, inflammation, and endothelial dysfunction, mediated by metabolites such as TMAO, SCFAs, LPS, and BAs. These microbial factors accelerate arterial stiffening, atherosclerosis, and mitochondrial dysfunction, hallmark features of vascular aging. While substantial progress has been made, further studies are essential to deepen our understanding of the molecular pathways connecting GM to vascular aging. Recent findings underscore the need to explore additional mechanisms, including mitochondrial dysfunction, loss of proteostasis, cellular senescence, apoptosis, and necroptosis, as potential links. Furthermore, discovering novel microbiota-derived metabolites and their impacts on vascular health could open new therapeutic avenues to mitigate aging-related vascular decline.
Nutrient-derived natural products offer promising strategies for regulating GM and combating vascular aging, addressing the challenge of promoting cardiovascular health. Compounds such as flavonoids, polyphenols, dietary fibers, alkaloids, and phytosterols not only exert direct cardiovascular benefits through anti-inflammatory, antioxidant, and metabolic effects but also indirectly enhance vascular health by modulating GM composition and activity. The intricate interplay between these natural products and GM results in the production of bioactive metabolites, restoration of microbial diversity, and improved gut barrier function, thereby influencing vascular integrity and function. Future research should prioritize identifying novel GM-derived metabolites, modulated by nutrient-derived natural products, linked to vascular health and developing personalized interventions tailored to individual GM profiles, enabling precise dietary strategies to combat vascular aging effectively.
Despite the promising potential of GM-targeted interventions, several limitations and challenges must be addressed before these strategies can be effectively translated into clinical practice. One major hurdle is the significant interindividual variability in GM composition, which can influence the efficacy of probiotics, prebiotics, and nutrient-derived natural products. Factors such as genetics, age, diet, medication use, and disease status all contribute to differences in microbial communities, resulting in variable responses to the same therapeutic intervention [266]. For instance, a probiotic strain that promotes vascular benefits in one individual may be ineffective or even counterproductive in another due to differences in microbial ecology or metabolite profiles [267]. Additionally, the long-term effects and safety of chronic modulation of the microbiota remain unclear, particularly in elderly or immunocompromised populations. The complexity of host-microbiota interactions and the need for precise, personalized approaches underscore the importance of developing robust biomarkers and diagnostic tools to stratify patients and tailor interventions accordingly. Addressing these challenges will be critical for harnessing the full therapeutic potential of GM-based strategies in the prevention and treatment of vascular aging and related CVDs.
While this review provides a comprehensive synthesis of current evidence on the interplay between GM, vascular aging, and cardiovascular health, several limitations should be acknowledged. First, the scope of the literature reviewed is constrained by the availability of published studies, and certain aspects of gut–vascular interactions remain underexplored, particularly in human cohorts and aged animal models. Second, substantial heterogeneity exists in the methodologies used among the cited studies, including variations in experimental models, microbiota profiling techniques, and outcome measures, which may limit direct comparability and the ability to draw definitive conclusions. Third, potential publication bias, favoring studies with positive or significant results, may skew the overall understanding of the field. Additionally, variability in study populations, such as differences in age, sex, genetic background, dietary patterns, and environmental exposures, may influence the generalizability of findings. These factors should be considered when interpreting the current evidence and in designing future research to address these gaps.

Author Contributions

Conceptualization, J.L., Y.W., Y.D. and J.Z.; methodology, J.L., Y.W. and S.S.; writing—original draft preparation, J.L., Y.W. and S.S.; writing—review and editing, A.T.G., Y.D. and J.Z.; supervision, Y.D. and J.Z.; project administration, Y.D. and J.Z.; funding acquisition, Y.D. and J.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Heart, Lung, and Blood Institute Grants R01HL168465 (JZ) and R01HL153333 (YD).

Acknowledgments

The author(s) acknowledge the use of ChatGPT (GPT-5, OpenAI) for assistance with language editing and improving textual clarity during the preparation of this manuscript. Following the use of this tool, the author(s) carefully reviewed and edited the content to ensure accuracy, appropriateness.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

BAs, bile acids; CMA, chaperone-mediated autophagy; CVDs, cardiovascular diseases; EA, ellagic acid; ECs, endothelial cells; EGCG, epigallocatechin-3-gallate; FMO, flavin mono-oxygenase; GM, gut microbiota; HFD, high fat diet; HSP, heat shock protein; IL, interleukin; LPS, lipopolysaccharides; MMP, matrix metalloproteinase; NO, nitric oxide; NOS, nitric oxide synthase; ROS, reactive oxygen species; SCFAs, short-chain fatty acids; SDG, secoisolariciresinol diglucoside; TMAO, trimethylamine-N-oxide; UPS, ubiquitin-proteasome system; VSMC, vascular smooth muscle cells.

References

  1. Di Cesare, M.; Perel, P.; Taylor, S.; Kabudula, C.; Bixby, H.; Gaziano, T.A.; McGhie, D.V.; Mwangi, J.; Pervan, B.; Narula, J.; et al. The heart of the world. Glob. Heart 2024, 19, 11. [Google Scholar] [CrossRef]
  2. Laurent, S. Defining vascular aging and cardiovascular risk. J. Hypertens. 2012, 30, S3–S8. [Google Scholar] [CrossRef]
  3. Kim, H.-L. Arterial stiffness and hypertension. Clin. Hypertens. 2023, 29, 31. [Google Scholar] [CrossRef]
  4. Datta, S.; Pasham, S.; Inavolu, S.; Boini, K.M.; Koka, S. Role of gut microbial metabolites in cardiovascular diseases—Current insights and the road ahead. Int. J. Mol. Sci. 2024, 25, 10208. [Google Scholar] [CrossRef] [PubMed]
  5. Escobar, C.; Aldeguer, X.; Vivas, D.; Manzano Fernández, S.; Gonzalez Caballero, E.; Garcia Martín, A.; Barrios, V.; Freixa-Pamias, R. The gut microbiota and its role in the development of cardiovascular disease. Expert Rev. Cardiovasc. Ther. 2025, 23, 23–34. [Google Scholar] [CrossRef]
  6. Tang, J.; Wei, Y.; Pi, C.; Zheng, W.; Zuo, Y.; Shi, P.; Chen, J.; Xiong, L.; Chen, T.; Liu, H.; et al. The therapeutic value of bifidobacteria in cardiovascular disease. npj Biofilms Microbiomes 2023, 9, 82. [Google Scholar] [CrossRef]
  7. Agnoletti, D.; Piani, F.; Cicero, A.F.; Borghi, C. The Gut Microbiota and Vascular Aging: A State-of-the-Art and Systematic Review of the Literature. J. Clin. Med. 2022, 11, 3557. [Google Scholar] [CrossRef]
  8. Qu, J.; Meng, F.; Wang, Z.; Xu, W. Unlocking Cardioprotective Potential of Gut Microbiome: Exploring Therapeutic Strategies. J. Microbiol. Biotechnol. 2024, 34, 2413–2424. [Google Scholar] [CrossRef]
  9. Mirmohammadali, S.N.; Rosenkranz, S.K. Dietary phytochemicals, gut microbiota composition, and health outcomes in human and animal models. Biosci. Microbiota Food Health 2023, 42, 152–171. [Google Scholar] [CrossRef]
  10. Qiu, Y.; Liu, Y.; Tao, J. Progress of clinical evaluation for vascular aging in humans. J. Transl. Intern. Med. 2021, 9, 17–23. [Google Scholar] [CrossRef]
  11. Minciullo, P.L.; Catalano, A.; Mandraffino, G.; Casciaro, M.; Crucitti, A.; Maltese, G.; Morabito, N.; Lasco, A.; Gangemi, S.; Basile, G. Inflammaging and anti-inflammaging: The role of cytokines in extreme longevity. Arch. Immunol. Ther. Exp. 2016, 64, 111–126. [Google Scholar] [CrossRef]
  12. Pacinella, G.; Ciaccio, A.M.; Tuttolomondo, A. Endothelial Dysfunction and Chronic Inflammation: The Cornerstones of Vascular Alterations in Age-Related Diseases. Int. J. Mol. Sci. 2022, 23, 15722. [Google Scholar] [CrossRef]
  13. Koutsaliaris, I.K.; Moschonas, I.C.; Pechlivani, L.M.; Tsouka, A.N.; Tselepis, A.D. Inflammation, Oxidative Stress, Vascular Aging and Atherosclerotic Ischemic Stroke. Curr. Med. Chem. 2022, 29, 5496–5509. [Google Scholar] [CrossRef]
  14. Yang, Y.; Dong, M. Exploring the role of oxidative stress in carotid atherosclerosis: Insights from transcriptomic data and single-cell sequencing combined with machine learning. Biol. Direct 2025, 20, 15. [Google Scholar] [CrossRef] [PubMed]
  15. Shaito, A.; Aramouni, K.; Assaf, R.; Parenti, A.; Orekhov, A.; El Yazbi, A.; Pintus, G.; Eid, A.H. Oxidative Stress-Induced Endothelial Dysfunction in Cardiovascular Diseases. Front. Biosci. (Landmark Ed) 2022, 27, 105. [Google Scholar] [CrossRef] [PubMed]
  16. Springo, Z.; Tarantini, S.; Toth, P.; Tucsek, Z.; Koller, A.; Sonntag, W.E.; Csiszar, A.; Ungvari, Z. Aging exacerbates pressure-induced mitochondrial oxidative stress in mouse cerebral arteries. J. Gerontol. Ser. A Biomed. Sci. Med. Sci. 2015, 70, 1355–1359. [Google Scholar] [CrossRef] [PubMed]
  17. Cabral-Pacheco, G.A.; Garza-Veloz, I.; Castruita-De la Rosa, C.; Ramirez-Acuña, J.M.; Perez-Romero, B.A.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef]
  18. Liang, R.; Qi, X.; Cai, Q.; Niu, L.; Huang, X.; Zhang, D.; Ling, J.; Wu, Y.; Chen, Y.; Yang, P.; et al. The role of NLRP3 inflammasome in aging and age-related diseases. Immun. Ageing 2024, 21, 14. [Google Scholar] [CrossRef]
  19. Sadaf, M.Z.N.; Shingatgeri, V.; Uddin, M.; Izhari, M.A.; Akhtar, M.S.; Kathait, A.; Kar, S.; Jain, I.; Datt, P. The Pivotal Role of Senescence in Cell Death and Aging: Where Do We Stand? Curr. Mol. Biol. Rep. 2020, 6, 91–101. [Google Scholar] [CrossRef]
  20. Owens, C.D.; Mukli, P.; Csipo, T.; Lipecz, A.; Silva-Palacios, F.; Dasari, T.W.; Tarantini, S.; Gardner, A.W.; Montgomery, P.S.; Waldstein, S.R.; et al. Microvascular dysfunction and neurovascular uncoupling are exacerbated in peripheral artery disease, increasing the risk of cognitive decline in older adults. Am. J. Physiol. Heart Circ. Physiol. 2022, 322, H924–H935. [Google Scholar] [CrossRef]
  21. Tracy, E.P.; Hughes, W.; Beare, J.E.; Rowe, G.; Beyer, A.; LeBlanc, A.J. Aging-Induced Impairment of Vascular Function: Mitochondrial Redox Contributions and Physiological/Clinical Implications. Antioxid. Redox Signal. 2021, 35, 974–1015. [Google Scholar] [CrossRef] [PubMed]
  22. Lee, H.; Jose, P.A. Coordinated Contribution of NADPH Oxidase- and Mitochondria-Derived Reactive Oxygen Species in Metabolic Syndrome and Its Implication in Renal Dysfunction. Front. Pharmacol. 2021, 12, 670076. [Google Scholar] [CrossRef] [PubMed]
  23. Frey, R.S.; Ushio–Fukai, M.; Malik, A.B. NADPH oxidase-dependent signaling in endothelial cells: Role in physiology and pathophysiology. Antioxid. Redox Signal. 2009, 11, 791–810. [Google Scholar] [CrossRef] [PubMed]
  24. Ungvari, Z.; Tarantini, S.; Donato, A.J.; Galvan, V.; Csiszar, A. Mechanisms of vascular aging. Circ. Res. 2018, 123, 849–867. [Google Scholar] [CrossRef]
  25. Ungvari, Z.; Bailey-Downs, L.; Sosnowska, D.; Gautam, T.; Koncz, P.; Losonczy, G.; Ballabh, P.; de Cabo, R.; Sonntag, W.E.; Csiszar, A. Vascular oxidative stress in aging: A homeostatic failure due to dysregulation of NRF2-mediated antioxidant response. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H363–H372. [Google Scholar] [CrossRef]
  26. Mistriotis, P.; Andreadis, S.T. Vascular aging: Molecular mechanisms and potential treatments for vascular rejuvenation. Ageing Res. Rev. 2017, 37, 94–116. [Google Scholar] [CrossRef]
  27. Maldonado, E.; Morales-Pison, S.; Urbina, F.; Solari, A. Aging Hallmarks and the Role of Oxidative Stress. Antioxidants 2023, 12, 651. [Google Scholar] [CrossRef]
  28. Miwa, S.; Kashyap, S.; Chini, E.; von Zglinicki, T. Mitochondrial dysfunction in cell senescence aging. J. Clin. Investig. 2022, 132, e158447. [Google Scholar] [CrossRef]
  29. Abu Shelbayeh, O.; Arroum, T.; Morris, S.; Busch, K.B. PGC-1α Is a Master Regulator of Mitochondrial Lifecycle and ROS Stress Response. Antioxidants 2023, 12, 1075. [Google Scholar] [CrossRef]
  30. Chen, L.; Qin, Y.; Liu, B.; Gao, M.; Li, A.; Li, X.; Gong, G. PGC-1α-Mediated Mitochondrial Quality Control: Molecular Mechanisms and Implications for Heart Failure. Front. Cell Dev. Biol. 2022, 10, 871357. [Google Scholar] [CrossRef]
  31. Zhang, H.; Muhetarijiang, M.; Chen, R.J.; Hu, X.; Han, J.; Zheng, L.; Chen, T. Mitochondrial Dysfunction: A Roadmap for Understanding and Tackling Cardiovascular Aging. Aging Dis. 2024, 16, 2575. [Google Scholar] [CrossRef]
  32. Ishida, M. Mitochondrial fusion and fission in vascular disease. Hypertens. Res. 2024, 47, 2935–2938. [Google Scholar] [CrossRef]
  33. Vásquez-Trincado, C.; García-Carvajal, I.; Pennanen, C.; Parra, V.; Hill, J.A.; Rothermel, B.A.; Lavandero, S. Mitochondrial dynamics, mitophagy and cardiovascular disease. J. Physiol. 2016, 594, 509–525. [Google Scholar] [CrossRef]
  34. Zhang, T.; Wang, L.; Duan, X.; Niu, Y.; Li, M.; Yun, L.; Sun, H.; Ma, Y.; Guo, Y. Sirtuins mediate mitochondrial quality control mechanisms: A novel therapeutic target for osteoporosis. Front. Endocrinol. 2023, 14, 1281213. [Google Scholar] [CrossRef] [PubMed]
  35. Li, Y.; Li, S.; Wu, H. Ubiquitination-Proteasome System (UPS) and Autophagy Two Main Protein Degradation Machineries in Response to Cell Stress. Cells 2022, 11, 851. [Google Scholar] [CrossRef] [PubMed]
  36. Hipp, M.S.; Park, S.-H.; Hartl, F.U. Proteostasis impairment in protein-misfolding and -aggregation diseases. Trends Cell Biol. 2014, 24, 506–514. [Google Scholar] [CrossRef] [PubMed]
  37. Hibshman, J.D.; Carra, S.; Goldstein, B. Tardigrade small heat shock proteins can limit desiccation-induced protein aggregation. Commun. Biol. 2023, 6, 121. [Google Scholar] [CrossRef]
  38. Gonçalves, C.C.; Sharon, I.; Schmeing, T.M.; Ramos, C.H.I.; Young, J.C. The chaperone HSPB1 prepares protein aggregates for resolubilization by HSP70. Sci. Rep. 2021, 11, 17139. [Google Scholar] [CrossRef]
  39. Albinhassan, T.H.; Alharbi, B.M.; AlSuhaibani, E.S.; Mohammad, S.; Malik, S.S. Small Heat Shock Proteins: Protein Aggregation Amelioration and Neuro- and Age-Protective Roles. Int. J. Mol. Sci. 2025, 26, 1525. [Google Scholar] [CrossRef]
  40. Hu, L.; Zhao, R.; Liu, Q.; Li, Q. New Insights Into Heat Shock Protein 90 in the Pathogenesis of Pulmonary Arterial Hypertension. Front. Physiol. 2020, 11, 1081. [Google Scholar] [CrossRef]
  41. Grundtman, C.; Kreutmayer, S.B.; Almanzar, G.; Wick, M.C.; Wick, G. Heat shock protein 60 and immune inflammatory responses in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 960–968. [Google Scholar] [CrossRef]
  42. Ahmed, B.; Rahman, A.A.; Lee, S.; Malhotra, R. The Implications of Aging on Vascular Health. Int. J. Mol. Sci. 2024, 25, 11188. [Google Scholar] [CrossRef]
  43. de Oliveira, A.A.; Mendoza, V.O.; Priviero, F.; Webb, R.C.; Nunes, K.P. Age-Related Decline in Vascular Responses to Phenylephrine is Associated with Reduced Levels of HSP70. Biomolecules 2022, 12, 1125. [Google Scholar] [CrossRef]
  44. de Oliveira, A.A.; Priviero, F.; Tostes, R.C.; Webb, R.C.; Nunes, K.P. Dissecting the interaction between HSP70 and vascular contraction: Role of Ca2+ handling mechanisms. Sci. Rep. 2021, 11, 1420. [Google Scholar] [CrossRef] [PubMed]
  45. Herrmann, J.; Lerman, L.O.; Lerman, A. On to the road to degradation: Atherosclerosis and the proteasome. Cardiovasc. Res. 2010, 85, 291–302. [Google Scholar] [CrossRef] [PubMed]
  46. Nakayama, H.; Nishida, K.; Otsu, K. Macromolecular degradation systems and cardiovascular aging. Circ. Res. 2016, 118, 1577–1592. [Google Scholar] [CrossRef] [PubMed]
  47. Wang, F.; Lerman, A.; Herrmann, J. Dysfunction of the ubiquitin-proteasome system in atherosclerotic cardiovascular disease. Am. J. Cardiovasc. Dis. 2015, 5, 83–100. [Google Scholar] [PubMed]
  48. Lv, X.-F.; Zhang, Y.-J.; Liu, X.; Zheng, H.-Q.; Liu, C.-Z.; Zeng, X.-L.; Li, X.-Y.; Lin, X.-C.; Lin, C.-X.; Ma, M.-M.; et al. TMEM16A ameliorates vascular remodeling by suppressing autophagy via inhibiting Bcl-2-p62 complex formation. Theranostics 2020, 10, 3980–3993. [Google Scholar] [CrossRef]
  49. Jin, J.; Liu, Y.; Huang, L.; Tan, H. Advances in epigenetic regulation of vascular aging. Rev. Cardiovasc. Med. 2019, 20, 19–25. [Google Scholar] [CrossRef]
  50. Grootaert, M.O.; da Costa Martins, P.A.; Bitsch, N.; Pintelon, I.; De Meyer, G.R.; Martinet, W.; Schrijvers, D.M. Defective autophagy in vascular smooth muscle cells accelerates senescence and promotes neointima formation and atherogenesis. Autophagy 2015, 11, 2014–2032. [Google Scholar] [CrossRef]
  51. Bharath, L.P.; Mueller, R.; Li, Y.; Ruan, T.; Kunz, D.; Goodrich, R.; Mills, T.; Deeter, L.; Sargsyan, A.; Babu, P.V.A.; et al. Impairment of autophagy in endothelial cells prevents shear-stress-induced increases in nitric oxide bioavailability. Can. J. Physiol. Pharmacol. 2014, 92, 605–612. [Google Scholar] [CrossRef] [PubMed]
  52. Di Micco, R.; Krizhanovsky, V.; Baker, D.; di Fagagna, F.D. Cellular senescence in ageing: From mechanisms to therapeutic opportunities. Nat. Rev. Mol. Cell Biol. 2021, 22, 75–95. [Google Scholar] [CrossRef] [PubMed]
  53. Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef] [PubMed]
  54. Luan, Y.; Zhu, X.; Jiao, Y.; Liu, H.; Huang, Z.; Pei, J.; Xu, Y.; Yang, Y.; Ren, K. Cardiac cell senescence: Molecular mechanisms, key proteins and therapeutic targets. Cell Death Discov. 2024, 10, 78. [Google Scholar] [CrossRef]
  55. Katsuumi, G.; Shimizu, I.; Yoshida, Y.; Minamino, T. Vascular Senescence in Cardiovascular and Metabolic Diseases. Front. Cardiovasc. Med. 2018, 5, 18. [Google Scholar] [CrossRef]
  56. Jia, G.; Aroor, A.R.; Jia, C.; Sowers, J.R. Endothelial cell senescence in aging-related vascular dysfunction. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2019, 1865, 1802–1809. [Google Scholar] [CrossRef]
  57. Bhayadia, R.; Schmidt, B.M.; Melk, A.; Hömme, M. Senescence-induced oxidative stress causes endothelial dysfunction. J. Gerontol. Ser. A Biomed. Sci. Med. Sci. 2016, 71, 161–169. [Google Scholar] [CrossRef]
  58. Chi, C.; Li, D.J.; Jiang, Y.J.; Tong, J.; Fu, H.; Wu, Y.H.; Shen, F.M. Vascular smooth muscle cell senescence and age-related diseases: State of the art. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2019, 1865, 1810–1821. [Google Scholar] [CrossRef]
  59. Ma, S.; Xie, X.; Yuan, R.; Xin, Q.; Miao, Y.; Leng, S.X.; Chen, K.; Cong, W. Vascular Aging and Atherosclerosis: A Perspective on Aging. Aging Dis. 2024, 16, 33–48. [Google Scholar] [CrossRef]
  60. Tower, J. Programmed cell death in aging. Ageing Res. Rev. 2015, 23, 90–100. [Google Scholar] [CrossRef]
  61. Pourbagher-Shahri, A.M.; Farkhondeh, T.; Talebi, M.; Kopustinskiene, D.M.; Samarghandian, S.; Bernatoniene, J. An Overview of NO Signaling Pathways in Aging. Molecules 2021, 26, 4533. [Google Scholar] [CrossRef]
  62. Yang, Y.; Li, X.; Zhang, T.; Xu, D. RIP kinases and necroptosis in aging and aging-related diseases. Life Med. 2022, 1, 2–20. [Google Scholar] [CrossRef] [PubMed]
  63. Thadathil, N.; Nicklas, E.H.; Mohammed, S.; Lewis Jr, T.L.; Richardson, A.; Deepa, S.S. Necroptosis increases with age in the brain and contributes to age-related neuroinflammation. Geroscience 2021, 43, 2345–2361. [Google Scholar] [CrossRef]
  64. Mohammed, S.; Thadathil, N.; Selvarani, R.; Nicklas, E.H.; Wang, D.; Miller, B.F.; Richardson, A.; Deepa, S.S. Necroptosis contributes to chronic inflammation and fibrosis in aging liver. Aging Cell 2021, 20, e13512. [Google Scholar] [CrossRef]
  65. Royce, G.H.; Brown-Borg, H.M.; Deepa, S.S. The potential role of necroptosis in inflammaging and aging. Geroscience 2019, 41, 795–811. [Google Scholar] [CrossRef]
  66. Deepa, S.S.; Unnikrishnan, A.; Matyi, S.; Hadad, N.; Richardson, A. Necroptosis increases with age and is reduced by dietary restriction. Aging Cell 2018, 17, e12770. [Google Scholar] [CrossRef]
  67. Wang, Q.; Liu, Z.; Ren, J.; Morgan, S.; Assa, C.; Liu, B. Receptor-interacting protein kinase 3 contributes to abdominal aortic aneurysms via smooth muscle cell necrosis and inflammation. Circ. Res. 2015, 116, 600–611. [Google Scholar] [CrossRef]
  68. Meng, L.; Jin, W.; Wang, X. RIP3-mediated necrotic cell death accelerates systematic inflammation and mortality. Proc. Natl. Acad. Sci. USA 2015, 112, 11007–11012. [Google Scholar] [CrossRef]
  69. Rath, S.; Heidrich, B.; Pieper, D.H.; Vital, M. Uncovering the trimethylamine-producing bacteria of the human gut microbiota. Microbiome 2017, 5, 54. [Google Scholar] [CrossRef]
  70. Seth, M.; Mondal, P.; Ghosh, D.; Mukhopadhyay, S.K. The foul play of two dietary metabolites trimethylamine (TMA) and trimethylamine N-oxide (TMAO) on human health and the role of microbes in mitigating their effects. Nutrire 2023, 48, 52. [Google Scholar] [CrossRef]
  71. Benson, T.W.; Conrad, K.A.; Li, X.S.; Wang, Z.; Helsley, R.N.; Schugar, R.C.; Coughlin, T.M.; Wadding-Lee, C.; Fleifil, S.; Russell, H.M.; et al. Gut microbiota–derived trimethylamine N-oxide contributes to abdominal aortic aneurysm through inflammatory and apoptotic mechanisms. Circulation 2023, 147, 1079–1096. [Google Scholar] [CrossRef]
  72. Liu, Y.; Dai, M. Trimethylamine N-oxide generated by the gut microbiota is associated with vascular inflammation: New insights into atherosclerosis. Mediat. Inflamm. 2020, 2020, 4634172. [Google Scholar] [CrossRef]
  73. Brunt, V.E.; Gioscia-Ryan, R.A.; Casso, A.G.; VanDongen, N.S.; Ziemba, B.P.; Sapinsley, Z.J.; Richey, J.J.; Zigler, M.C.; Neilson, A.P.; Davy, K.P.; et al. Trimethylamine-N-oxide promotes age-related vascular oxidative stress and endothelial dysfunction in mice and healthy humans. Hypertension 2020, 76, 101–112. [Google Scholar] [CrossRef] [PubMed]
  74. Li, T.; Chen, Y.; Gua, C.; Li, X. Elevated circulating trimethylamine N-oxide levels contribute to endothelial dysfunction in aged rats through vascular inflammation and oxidative stress. Front. Physiol. 2017, 8, 350. [Google Scholar] [CrossRef] [PubMed]
  75. Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-oxide: The good, the bad and the unknown. Toxins 2016, 8, 326. [Google Scholar] [CrossRef] [PubMed]
  76. Dabravolski, S.A.; Khotina, V.A.; Sukhorukov, V.N.; Kalmykov, V.A.; Mikhaleva, L.M.; Orekhov, A.N. The role of mitochondrial DNA mutations in cardiovascular diseases. Int. J. Mol. Sci. 2022, 23, 952. [Google Scholar] [CrossRef]
  77. Yang, Y.; Karampoor, S.; Mirzaei, R.; Borozdkin, L.; Zhu, P. The interplay between microbial metabolites and macrophages in cardiovascular diseases: A comprehensive review. Int. Immunopharmacol. 2023, 121, 110546. [Google Scholar] [CrossRef]
  78. Mutengo, K.H.; Masenga, S.K.; Mweemba, A.; Mutale, W.; Kirabo, A. Gut microbiota dependant trimethylamine N-oxide and hypertension. Front. Physiol. 2023, 14, 1075641. [Google Scholar] [CrossRef]
  79. Li, M.; Van Esch, B.C.; Wagenaar, G.T.; Garssen, J.; Folkerts, G.; Henricks, P.A. Pro-and anti-inflammatory effects of short chain fatty acids on immune and endothelial cells. Eur. J. Pharmacol. 2018, 831, 52–59. [Google Scholar]
  80. Dias, M.T.S.; Aguilar, E.C.; Campos, G.P.; do Couto, N.F.; Capettini, L.D.S.A.; Braga, W.F.; de Oliveira Andrade, L.; Alvarez-Leite, J. Butyrate inhibits LPC-induced endothelial dysfunction by regulating nNOS-produced NO and ROS production. Nitric Oxide 2023, 138, 42–50. [Google Scholar] [CrossRef]
  81. Modoux, M.; Rolhion, N.; Lefevre, J.H.; Oeuvray, C.; Nádvorník, P.; Illes, P.; Emond, P.; Parc, Y.; Mani, S.; Dvorak, Z.; et al. Butyrate acts through HDAC inhibition to enhance aryl hydrocarbon receptor activation by gut microbiota-derived ligands. Gut Microbes 2022, 14, 2105637. [Google Scholar] [CrossRef] [PubMed]
  82. Kibbie, J.J.; Dillon, S.M.; Thompson, T.A.; Purba, C.M.; McCarter, M.D.; Wilson, C.C. Butyrate directly decreases human gut lamina propria CD4 T cell function through histone deacetylase (HDAC) inhibition and GPR43 signaling. Immunobiology 2021, 226, 152126. [Google Scholar] [CrossRef] [PubMed]
  83. Najmanová, I.; Pourová, J.; Vopršalová, M.; Pilařová, V.; Semecký, V.; Nováková, L.; Mladěnka, P. Flavonoid metabolite 3-(3-hydroxyphenyl) propionic acid formed by human microflora decreases arterial blood pressure in rats. Mol. Nutr. Food Res. 2016, 60, 981–991. [Google Scholar] [CrossRef] [PubMed]
  84. Cobb, K.M.; Chavez, D.A.; Kenyon, J.D.; Hutelin, Z.; Webster, M.J. Acetic Acid Supplementation: Effect on Resting and Exercise Energy Expenditure and Substrate Utilization. Int. J. Exerc. Sci. 2021, 14, 222–229. [Google Scholar] [CrossRef]
  85. Tan, S.M.; Lee, S.M.; Dykes, G.A. Acetic acid induces pH-independent cellular energy depletion in Salmonella enterica. Foodborne Pathog. Dis. 2015, 12, 183–189. [Google Scholar] [CrossRef]
  86. Qi, H.-M.; Cao, Q.; Liu, Q. TLR4 regulates vascular smooth muscle cell proliferation in hypertension via modulation of the NLRP3 inflammasome. Am. J. Transl. Res. 2021, 13, 314–325. [Google Scholar]
  87. Balistreri, C.R.; Ruvolo, G.; Lio, D.; Madonna, R. Toll-like receptor-4 signaling pathway in aorta aging and diseases:“its double nature”. J. Mol. Cell. Cardiol. 2017, 110, 38–53. [Google Scholar] [CrossRef]
  88. Strela, F.B.; Brun, B.F.; Berger, R.C.M.; Melo, S.; de Oliveira, E.M.; Barauna, V.G.; Vassallo, P.F. Lipopolysaccharide exposure modulates the contractile and migratory phenotypes of vascular smooth muscle cells. Life Sci. 2020, 241, 117098. [Google Scholar] [CrossRef]
  89. Echeverría, C.; Montorfano, I.; Sarmiento, D.; Becerra, A.; Nuñez-Villena, F.; Figueroa, X.F.; Cabello-Verrugio, C.; Elorza, A.A.; Riedel, C.; Simon, F. Lipopolysaccharide induces a fibrotic-like phenotype in endothelial cells. J. Cell. Mol. Med. 2013, 17, 800–814. [Google Scholar] [CrossRef]
  90. Tellides, G.; Pober, J.S. Inflammatory and immune responses in the arterial media. Circ. Res. 2015, 116, 312–322. [Google Scholar] [CrossRef]
  91. Surmi, B.K.; Hasty, A.H. The role of chemokines in recruitment of immune cells to the artery wall and adipose tissue. Vasc. Pharmacol. 2010, 52, 27–36. [Google Scholar] [CrossRef]
  92. Nocella, C.; Carnevale, R.; Bartimoccia, S.; Novo, M.; Cangemi, R.; Pastori, D.; Calvieri, C.; Pignatelli, P.; Violi, F. Lipopolysaccharide as trigger of platelet aggregation via eicosanoid over-production. Thromb. Haemost. 2017, 117, 1558–1570. [Google Scholar] [CrossRef]
  93. Zhang, G.; Han, J.; Welch, E.J.; Ye, R.D.; Voyno-Yasenetskaya, T.A.; Malik, A.B.; Du, X.; Li, Z. Lipopolysaccharide stimulates platelet secretion and potentiates platelet aggregation via TLR4/MyD88 and the cGMP-dependent protein kinase pathway. J. Immunol. 2009, 182, 7997–8004. [Google Scholar] [CrossRef]
  94. Qin, P.; Tang, X.; Elloso, M.M.; Harnish, D.C. Bile acids induce adhesion molecule expression in endothelial cells through activation of reactive oxygen species, NF-κB, and p38. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H741–H747. [Google Scholar] [CrossRef]
  95. Wang, C.; Ma, Q.; Yu, X. Bile Acid Network and Vascular Calcification-Associated Diseases: Unraveling the Intricate Connections and Therapeutic Potential. Clin. Interv. Aging 2023, 18, 1749–1767. [Google Scholar] [CrossRef]
  96. Khurana, S.; Raufman, J.P.; Pallone, T.L. Bile acids regulate cardiovascular function. Clin. Transl. Sci. 2011, 4, 210–218. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, R.; Ma, W.-Q.; Fu, M.-J.; Li, J.; Hu, C.H.; Chen, Y.; Zhou, M.M.; Gao, Z.J.; He, Y.L. Overview of bile acid signaling in the cardiovascular system. World J. Clin. Cases 2021, 9, 308–320. [Google Scholar] [CrossRef] [PubMed]
  98. Zanotti, I.; Turroni, F.; Piemontese, A.; Mancabelli, L.; Milani, C.; Viappiani, A.; Prevedini, G.; Sanchez, B.; Margolles, A.; Elviri, L.; et al. Evidence for cholesterol-lowering activity by Bifidobacterium bifidum PRL2010 through gut microbiota modulation. Appl. Microbiol. Biotechnol. 2015, 99, 6813–6829. [Google Scholar] [CrossRef]
  99. Wang, Q.; Guo, M.; Liu, Y.; Xu, M.; Shi, L.; Li, X.; Zhao, J.; Zhang, H.; Wang, G.; Chen, W. Bifidobacterium breve and Bifidobacterium longum attenuate choline-induced plasma trimethylamine N-oxide production by modulating gut microbiota in mice. Nutrients 2022, 14, 1222. [Google Scholar] [CrossRef] [PubMed]
  100. Robles-Vera, I.; de la Visitación, N.; Toral, M.; Sánchez, M.; Romero, M.; Gómez-Guzmán, M.; Yang, T.; Izquierdo-García, J.L.; Guerra-Hernández, E.; Ruiz-Cabello, J.; et al. Probiotic Bifidobacterium breve prevents DOCA-salt hypertension. Fed. Am. Soc. Exp. Biol. J. 2020, 34, 13626–13640. [Google Scholar] [CrossRef]
  101. Bernini, L.J.; Simão, A.N.C.; de Souza, C.H.; Alfieri, D.F.; Segura, L.G.; Costa, G.N.; Dichi, I. Effect of Bifidobacterium lactis HN019 on inflammatory markers and oxidative stress in subjects with and without the metabolic syndrome. Br. J. Nutr. 2018, 120, 645–652. [Google Scholar] [CrossRef]
  102. Al-Sheraji, S.H.; Ismail, A.; Manap, M.Y.; Mustafa, S.; Yusof, R.M.; Hassan, F.A. Hypocholesterolaemic effect of yoghurt containing Bifidobacterium pseudocatenulatum G4 or Bifidobacterium longum BB536. Food Chem. 2012, 135, 356–361. [Google Scholar] [CrossRef]
  103. Jiang, J.; Wu, C.; Zhang, C.; Zhang, Q.; Yu, L.; Zhao, J.; Zhang, H.; Narbad, A.; Chen, W.; Zhai, Q. Strain-specific effects of Bifidobacterium longum on hypercholesterolemic rats and potential mechanisms. Int. J. Mol. Sci. 2021, 22, 1305. [Google Scholar] [CrossRef] [PubMed]
  104. Malik, M.; Suboc, T.M.; Tyagi, S.; Salzman, N.; Wang, J.; Ying, R.; Tanner, M.J.; Kakarla, M.; Baker, J.E.; Widlansky, M.E. Lactobacillus plantarum 299v supplementation improves vascular endothelial function and reduces inflammatory biomarkers in men with stable coronary artery disease. Circ. Res. 2018, 123, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  105. Qiu, L.; Tao, X.; Xiong, H.; Yu, J.; Wei, H. Lactobacillus plantarum ZDY04 exhibits a strain-specific property of lowering TMAO via the modulation of gut microbiota in mice. Food Funct. 2018, 9, 4299–4309. [Google Scholar] [CrossRef]
  106. Zheng, Z.-Y.; Cao, F.-W.; Wang, W.-J.; Yu, J.; Chen, C.; Chen, B.; Liu, J.X.; Firrman, J.; Renye, J.; Ren, D.X. Probiotic characteristics of Lactobacillus plantarum E680 and its effect on Hypercholesterolemic mice. BMC Microbiol. 2020, 20, 239. [Google Scholar] [CrossRef]
  107. Huang, Y.; Wang, X.; Wang, J.; Wu, F.; Sui, Y.; Yang, L.; Wang, Z. Lactobacillus plantarum strains as potential probiotic cultures with cholesterol-lowering activity. J. Dairy Sci. 2013, 96, 2746–2753. [Google Scholar] [CrossRef]
  108. Zhao, X.; Oduro, P.K.; Tong, W.; Wang, Y.; Gao, X.; Wang, Q. Therapeutic potential of natural products against atherosclerosis: Targeting on gut microbiota. Pharmacol. Res. 2021, 163, 105362. [Google Scholar] [CrossRef]
  109. Oh, N.S.; Koh, J.H.; Park, M.R.; Kim, Y.; Kim, S.H. Hypolipidemic and antiinflammatory effects of fermented Maillard reaction products by Lactobacillus fermentum H9 in an animal model. J. Dairy. Sci. 2016, 99, 9415–9423. [Google Scholar] [CrossRef]
  110. Gómez-Guzmán, M.; Toral, M.; Romero, M.; Jiménez, R.; Galindo, P.; Sanchez, M.; Zarzuelo, M.J.; Olivares, M.; Galvez, J.; Duarte, J. Antihypertensive effects of probiotics Lactobacillus strains in spontaneously hypertensive rats. Mol. Nutr. Food Res. 2015, 59, 2326–2336. [Google Scholar] [CrossRef]
  111. Robles-Vera, I.; Toral, M.; de la Visitación, N.; Sánchez, M.; Romero, M.; Olivares, M.; Jiménez, R.; Duarte, J. The probiotic Lactobacillus fermentum prevents dysbiosis and vascular oxidative stress in rats with hypertension induced by chronic nitric oxide blockade. Mol. Nutr. Food Res. 2018, 62, 1800298. [Google Scholar] [CrossRef]
  112. Tomaro-Duchesneau, C.; Saha, S.; Malhotra, M.; Jones, M.L.; Labbé, A.; Rodes, L.; Kahouli, I.; Prakash, S. Effect of orally administered L. fermentum NCIMB 5221 on markers of metabolic syndrome: An in vivo analysis using ZDF rats. Appl. Microbiol. Biotechnol. 2014, 98, 115–126. [Google Scholar] [CrossRef]
  113. Watanabe, S.; Katsube, T.; Hattori, H.; Sato, H.; Ishijima, T.; Nakai, Y.; Abe, K.; Sonomoto, K. Effect of Lactobacillus brevis 119-2 isolated from Tsuda kabu red turnips on cholesterol levels in cholesterol-administered rats. J. Biosci. Bioeng. 2013, 116, 45–51. [Google Scholar] [CrossRef]
  114. London, L.E.; Kumar, A.H.; Wall, R.; Casey, P.G.; O’Sullivan, O.; Shanahan, F.; Hill, C.; Cotter, P.D.; Fitzgerald, G.F.; Ross, R.P.; et al. Exopolysaccharide-producing probiotic Lactobacilli reduce serum cholesterol and modify enteric microbiota in ApoE-deficient mice. J. Nutr. 2014, 144, 1956–1962. [Google Scholar] [CrossRef]
  115. Kalani, M.; Hodjati, H.; Sajedi Khanian, M.; Doroudchi, M. Lactobacillus acidophilus increases the anti-apoptotic micro RNA-21 and decreases the pro-inflammatory micro RNA-155 in the LPS-treated human endothelial cells. Probiotics Antimicrob. Proteins 2016, 8, 61–72. [Google Scholar] [CrossRef]
  116. Seo, K.-H.; Lee, H.G.; Eor, J.Y.; Jeon, H.J.; Yokoyama, W.; Kim, H. Effects of kefir lactic acid bacteria-derived postbiotic components on high fat diet-induced gut microbiota and obesity. Food Res. Int. 2022, 157, 111445. [Google Scholar] [CrossRef] [PubMed]
  117. Gan, G.; Lin, S.; Luo, Y.; Zeng, Y.; Lu, B.; Zhang, R.; Chen, S.; Lei, H.; Cai, Z.; Huang, X. Unveiling the oral-gut connection: Chronic apical periodontitis accelerates atherosclerosis via gut microbiota dysbiosis and altered metabolites in apoE−/− Mice on a high-fat diet. Int. J. Oral Sci. 2024, 16, 39. [Google Scholar] [CrossRef] [PubMed]
  118. Chiang, S.-S.; Pan, T.-M. Beneficial effects of Lactobacillus paracasei subsp. paracasei NTU 101 and its fermented products. Appl. Microbiol. Biotechnol. 2012, 93, 903–916. [Google Scholar] [PubMed]
  119. Tanida, M.; Yamano, T.; Maeda, K.; Okumura, N.; Fukushima, Y.; Nagai, K. Effects of intraduodenal injection of Lactobacillus johnsonii La1 on renal sympathetic nerve activity and blood pressure in urethane-anesthetized rats. Neurosci. Lett. 2005, 389, 109–114. [Google Scholar] [CrossRef]
  120. Pihlanto, A.; Virtanen, T.; Korhonen, H. Angiotensin I converting enzyme (ACE) inhibitory activity and antihypertensive effect of fermented milk. Int. Dairy J. 2010, 20, 3–10. [Google Scholar] [CrossRef]
  121. Jauhiainen, T.; Vapaatalo, H.; Poussa, T.; Kyrönpalo, S.; Rasmussen, M.; Korpela, R. Lactobacillus helveticus fermented milk lowers blood pressure in hypertensive subjects in 24-h ambulatory blood pressure measurement. Am. J. Hypertens. 2005, 18, 1600–1605. [Google Scholar] [CrossRef]
  122. Wang, Q.; He, Y.; Li, X.; Zhang, T.; Liang, M.; Wang, G.; Zhao, J.; Zhang, H.; Chen, W. Lactobacillus reuteri CCFM8631 alleviates hypercholesterolaemia caused by the paigen atherogenic diet by regulating the gut microbiota. Nutrients 2022, 14, 1272. [Google Scholar] [CrossRef] [PubMed]
  123. Hsieh, M.-C.; Tsai, W.-H.; Jheng, Y.-P.; Su, S.L.; Wang, S.Y.; Lin, C.C.; Chen, Y.H.; Chang, W.W. The beneficial effects of Lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus: A randomized, double-blinded, placebo-controlled trial. Sci. Rep. 2018, 8, 16791. [Google Scholar] [CrossRef] [PubMed]
  124. Qiu, L.; Yang, D.; Tao, X.; Yu, J.; Xiong, H.; Wei, H. Enterobacter aerogenes ZDY01 attenuates choline-induced trimethylamine N-oxide levels by remodeling gut microbiota in mice. J. Microbiol. Biotechnol. 2017, 27, 1491–1499. [Google Scholar] [CrossRef] [PubMed]
  125. Mikeš, Z.; Ferenčík, M.; Jahnová, E.; Ebringer, L.; Čižnár, I. Hypocholesterolemic and immunostimulatory effects of orally applied Enterococcus faecium M-74 in man. Folia Microbiol. 1995, 40, 639–646. [Google Scholar] [CrossRef]
  126. Armstrong, L.; do Carmo, M.A.V.; Wu, Y.; Esmerino, L.A.; Azevedo, L.; Zhang, L.; Granato, D. Optimizing the extraction of bioactive compounds from pu-erh tea (Camellia sinensis var assamica) and evaluation of antioxidant, cytotoxic, antimicrobial, antihemolytic, and inhibition of α-amylase and α-glucosidase activities. Food Res. Int. 2020, 137, 109430. [Google Scholar]
  127. Mizoguchi, T.; Kasahara, K.; Yamashita, T.; Sasaki, N.; Yodoi, K.; Matsumoto, T.; Emoto, T.; Hayashi, T.; Kitano, N.; Yoshida, N.; et al. Oral administration of the lactic acid bacterium Pediococcus acidilactici attenuates atherosclerosis in mice by inducing tolerogenic dendritic cells. Heart Vessel. 2017, 32, 768–776. [Google Scholar] [CrossRef]
  128. Yang, M.; Wang, J.-H.; Shin, J.-H.; Lee, D.; Lee, S.N.; Seo, J.G.; Shin, J.H.; Nam, Y.D.; Kim, H.; Sun, X. Pharmaceutical efficacy of novel human-origin Faecalibacterium prausnitzii strains on high-fat-diet-induced obesity and associated metabolic disorders in mice. Front. Endocrinol. 2023, 14, 1220044. [Google Scholar] [CrossRef]
  129. Yoshida, N.; Emoto, T.; Yamashita, T.; Watanabe, H.; Hayashi, T.; Tabata, T.; Hoshi, N.; Hatano, N.; Ozawa, G.; Sasaki, N.; et al. Bacteroides vulgatus and Bacteroides dorei reduce gut microbial lipopolysaccharide production and inhibit atherosclerosis. Circulation 2018, 138, 2486–2498. [Google Scholar] [CrossRef]
  130. Katiraei, S.; de Vries, M.R.; Costain, A.H.; Thiem, K.; Hoving, L.R.; van Diepen, J.A.; Smits, H.H.; Bouter, K.E.; Rensen, P.C.; Quax, P.H.; et al. Akkermansia muciniphila exerts lipid-lowering and immunomodulatory effects without affecting neointima formation in hyperlipidemic APOE* 3-Leiden. CETP mice. Mol. Nutr. Food Res. 2020, 64, 1900732. [Google Scholar] [CrossRef]
  131. Chen, Y.-H.; Tsai, W.-H.; Wu, H.-Y.; Chen, C.Y.; Yeh, W.L.; Chen, Y.H.; Hsu, H.Y.; Chen, W.W.; Chen, Y.W.; Chang, W.W.; et al. Probiotic Lactobacillus spp. act against Helicobacter pylori-induced inflammation. J. Clin. Med. 2019, 8, 90. [Google Scholar] [CrossRef] [PubMed]
  132. Quévrain, E.; Maubert, M.; Michon, C.; Chain, F.; Marquant, R.; Tailhades, J.; Miquel, S.; Carlier, L.; Bermúdez-Humarán, L.G.; Pigneur, B.; et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016, 65, 415–425. [Google Scholar] [CrossRef] [PubMed]
  133. Cao, Y.; Shen, J.; Ran, Z.H. Association between Faecalibacterium prausnitzii reduction and inflammatory bowel disease: A meta-analysis and systematic review of the literature. Gastroenterol. Res. Pract. 2014, 2014, 872725. [Google Scholar] [CrossRef] [PubMed]
  134. Khokhlova, E.V.; Smeianov, V.V.; Efimov, B.A.; Kafarskaia, L.I.; Pavlova, S.I.; Shkoporov, A.N. Anti-inflammatory properties of intestinal Bifidobacterium strains isolated from healthy infants. Microbiol. Immunol. 2012, 56, 27–39. [Google Scholar] [CrossRef]
  135. Mohamadzadeh, M.; Pfeiler, E.A.; Brown, J.B.; Zadeh, M.; Gramarossa, M.; Managlia, E.; Bere, P.; Sarraj, B.; Khan, M.W.; Pakanati, K.C.; et al. Regulation of induced colonic inflammation by Lactobacillus acidophilus deficient in lipoteichoic acid. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4623–4630. [Google Scholar] [CrossRef]
  136. Zhang, L.L.; Chen, X.; Zheng, P.Y.; Luo, Y.; Lu, G.F.; Liu, Z.Q.; Huang, H.; Yang, P.C. Oral Bifidobacterium modulates intestinal immune inflammation in mice with food allergy. J. Gastroenterol. Hepatol. 2010, 25, 928–934. [Google Scholar] [CrossRef]
  137. Liu, Y.; Fatheree, N.Y.; Mangalat, N.; Rhoads, J.M. Human-derived probiotic Lactobacillus reuteri strains differentially reduce intestinal inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G1087–G1096. [Google Scholar] [CrossRef]
  138. Duranti, S.; Vivo, V.; Zini, I.; Milani, C.; Mangifesta, M.; Anzalone, R.; Mancabelli, L.; Viappiani, A.; Cantoni, A.M.; Barocelli, E.; et al. Bifidobacterium bifidum PRL2010 alleviates intestinal ischemia/reperfusion injury. PLoS ONE 2018, 13, e0202670. [Google Scholar] [CrossRef]
  139. Munukka, E.; Rintala, A.; Toivonen, R.; Nylund, M.; Yang, B.; Takanen, A.; Hänninen, A.; Vuopio, J.; Huovinen, P.; Jalkanen, S.; et al. Faecalibacterium prausnitzii treatment improves hepatic health and reduces adipose tissue inflammation in high-fat fed mice. ISME J. 2017, 11, 1667–1679. [Google Scholar] [CrossRef]
  140. Aihara, K.; Kajimoto, O.; Hirata, H.; Takahashi, R.; Nakamura, Y. Effect of powdered fermented milk with Lactobacillus helveticus on subjects with high-normal blood pressure or mild hypertension. J. Am. Coll. Nutr. 2005, 24, 257–265. [Google Scholar] [CrossRef]
  141. Tuomilehto, J.; Lindström, J.; Hyyrynen, J.; Korpela, R.; Karhunen, M.L.; Mikkola, L.; Jauhiainen, T.; Seppo, L.; Nissinen, A. Effect of ingesting sour milk fermented using Lactobacillus helveticus bacteria producing tripeptides on blood pressure in subjects with mild hypertension. J. Hum. Hypertens. 2004, 18, 795–802. [Google Scholar] [CrossRef]
  142. Li, X.; Liu, Y.; Guo, X.; Ma, Y.; Zhang, H.; Liang, H. Effect of Lactobacillus casei on lipid metabolism and intestinal microflora in patients with alcoholic liver injury. Eur. J. Clin. Nutr. 2021, 75, 1227–1236. [Google Scholar] [CrossRef]
  143. Lee, E.; Jung, S.-R.; Lee, S.-Y.; Lee, N.K.; Paik, H.D.; Lim, S.I. Lactobacillus plantarum strain Ln4 attenuates diet-induced obesity, insulin resistance, and changes in hepatic mRNA levels associated with glucose and lipid metabolism. Nutrients 2018, 10, 643. [Google Scholar] [CrossRef] [PubMed]
  144. Li, X.; Wang, N.; Yin, B.; Fang, D.; Jiang, T.; Fang, S.; Zhao, J.; Zhang, H.; Wang, G.; Chen, W. Effects of Lactobacillus plantarum CCFM0236 on hyperglycaemia and insulin resistance in high-fat and streptozotocin-induced type 2 diabetic mice. J. Appl. Microbiol. 2016, 121, 1727–1736. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, F.; Zhu, W.-J.; Edirisuriya, P.; Ai, Q.; Nie, K.; Ji, X.M.; Li, Y.; Zhou, K. Beneficial effects of a combination of Clostridium cochlearium and Lactobacillus acidophilus on body weight gain, insulin sensitivity, and gut microbiota in high-fat diet–induced obese mice. Nutrition 2022, 93, 111439. [Google Scholar] [CrossRef] [PubMed]
  146. Tang, C.; Cao, G.; Zhao, W.; Bie, X.; Lu, F.; Lu, Z.; Lu, Y. Lactobacillus acidophilus NX2-6 improved high-fat diet-induced glucose metabolism disorder independent of promotion of insulin secretion in mice. J. Agric. Food Chem. 2021, 69, 15598–15610. [Google Scholar] [CrossRef]
  147. Yan, F.; Li, N.; Shi, J.; Li, H.; Yue, Y.; Jiao, W.; Wang, N.; Song, Y.; Huo, G.; Li, B. Lactobacillus acidophilus alleviates type 2 diabetes by regulating hepatic glucose, lipid metabolism and gut microbiota in mice. Food Funct. 2019, 10, 5804–5815. [Google Scholar] [CrossRef]
  148. Andreasen, A.S.; Larsen, N.; Pedersen-Skovsgaard, T.; Berg, R.M.; Møller, K.; Svendsen, K.D.; Jakobsen, M.; Pedersen, B.K. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br. J. Nutr. 2010, 104, 1831–1838. [Google Scholar] [CrossRef]
  149. Minami, J.-i.; Kondo, S.; Yanagisawa, N.; Odamaki, T.; Xiao, J.Z.; Abe, F.; Nakajima, S.; Hamamoto, Y.; Saitoh, S.; Shimoda, T. Oral administration of Bifidobacterium breve B-3 modifies metabolic functions in adults with obese tendencies in a randomised controlled trial. J. Nutr. Sci. 2015, 4, e17. [Google Scholar] [CrossRef]
  150. Kim, S.H.; Huh, C.S.; Choi, I.D.; Jeong, J.W.; Ku, H.K.; Ra, J.H.; Kim, T.Y.; Kim, G.B.; Sim, J.H.; Ahn, Y.T. The anti-diabetic activity of Bifidobacterium lactis HY8101 in vitro and in vivo. J. Appl. Microbiol. 2014, 117, 834–845. [Google Scholar] [CrossRef]
  151. Kondo, S.; Xiao, J.Z.; Satoh, T.; Odamaki, T.; Takahashi, S.; Sugahara, H.; Yaeshima, T.; Iwatsuki, K.; Kamei, A.; Abe, K. Antiobesity effects of Bifidobacterium breve strain B-3 supplementation in a mouse model with high-fat diet-induced obesity. Biosci. Biotechnol. Biochem. 2010, 74, 1656–1661. [Google Scholar] [CrossRef]
  152. Sharifi-Rad, J.; Rodrigues, C.F.; Sharopov, F.; Docea, A.O.; Karaca, C.A.; Sharifi-Rad, M.; Karıncaoglu, K.D.; Gülseren, G.; Şenol, E.; Demircan, E.; et al. Diet, lifestyle and cardiovascular diseases: Linking pathophysiology to cardioprotective effects of natural bioactive compounds. Int. J. Environ. Res. Public Health 2020, 17, 2326. [Google Scholar] [CrossRef]
  153. Shaito, A.; Thuan, D.T.B.; Phu, H.T.; Nguyen, T.H.D.; Hasan, H.; Halabi, S.; Abdelhady, S.; Nasrallah, G.K.; Eid, A.H.; Pintus, G. Herbal medicine for cardiovascular diseases: Efficacy, mechanisms, and safety. Front. Pharmacol. 2020, 11, 422. [Google Scholar] [CrossRef]
  154. Ciumărnean, L.; Milaciu, M.V.; Runcan, O.; Vesa, Ș.C.; Răchișan, A.L.; Negrean, V.; Perné, M.G.; Donca, V.I.; Alexescu, T.G.; Para, I.; et al. The effects of flavonoids in cardiovascular diseases. Molecules 2020, 25, 4320. [Google Scholar] [CrossRef]
  155. Benito, I.; Encío, I.J.; Milagro, F.I.; Alfaro, M.; Martínez-Peñuela, A.; Barajas, M.; Marzo, F. Microencapsulated Bifidobacterium bifidum and Lactobacillus gasseri in combination with Quercetin inhibit colorectal cancer development in ApcMin/+ mice. Int. J. Mol. Sci. 2021, 22, 4906. [Google Scholar] [CrossRef] [PubMed]
  156. Xue, Y.; Du, M.; Zhu, M.-J. Quercetin prevents Escherichia coli O157: H7 adhesion to epithelial cells via suppressing focal adhesions. Front. Microbiol. 2019, 9, 3278. [Google Scholar] [CrossRef] [PubMed]
  157. Zhang, L.; Wu, Q.; Wang, N.; Zhang, L.; Yang, X.; Zhao, Y. Quercetin inhibits hepatotoxic effects by reducing trimethylamine-N-oxide formation in C57BL/6J mice fed with a high l-carnitine diet. Food Funct. 2023, 14, 206–214. [Google Scholar] [CrossRef] [PubMed]
  158. Liu, S.; Loo, Y.T.; Li, Z.; Ng, K. Alginate-inulin-chitosan based microspheres alter metabolic fate of encapsulated quercetin, promote short chain fatty acid production, and modulate pig gut microbiota. Food Chem. 2023, 418, 135802. [Google Scholar] [CrossRef]
  159. Zhang, M.; Xie, Z.; Gao, W.; Pu, L.; Wei, J.; Guo, C. Quercetin regulates hepatic cholesterol metabolism by promoting cholesterol-to-bile acid conversion and cholesterol efflux in rats. Nutr. Res. 2016, 36, 271–279. [Google Scholar] [CrossRef]
  160. Fan, T.; Du, Y.; Zhang, M.; Zhu, A.R.; Zhang, J. Senolytics cocktail dasatinib and quercetin alleviate human umbilical vein endothelial cell senescence via the TRAF6-MAPK-NF-κB axis in a YTHDF2-dependent manner. Gerontology 2022, 68, 920–934. [Google Scholar] [CrossRef]
  161. Kim, S.G.; Sung, J.Y.; Kim, J.-R.; Choi, H.H. Quercetin-induced apoptosis ameliorates vascular smooth muscle cell senescence through AMP-activated protein kinase signaling pathway Korean. J. Physiol. Pharmacol. 2020, 24, 69–79. [Google Scholar]
  162. Nie, J.; Zhang, L.; Zhao, G.; Du, X. Quercetin reduces atherosclerotic lesions by altering the gut microbiota and reducing atherogenic lipid metabolites. J. Appl. Microbiol. 2019, 127, 1824–1834. [Google Scholar] [CrossRef] [PubMed]
  163. Park, J.-M.; Shin, Y.; Kim, S.H.; Jin, M.; Choi, J.J. Dietary epigallocatechin-3-gallate alters the gut microbiota of obese diabetic db/db mice: Lactobacillus is a putative target. J. Med. Food 2020, 23, 1033–1042. [Google Scholar] [CrossRef] [PubMed]
  164. Ushiroda, C.; Naito, Y.; Takagi, T.; Uchiyama, K.; Mizushima, K.; Higashimura, Y.; Yasukawa, Z.; Okjubo, T.; Inoue, R.; Honda, A.; et al. Green tea polyphenol (epigallocatechin-3-gallate) improves gut dysbiosis and serum bile acids dysregulation in high-fat diet-fed mice. J. Clin. Biochem. Nutr. 2019, 65, 34–46. [Google Scholar] [CrossRef] [PubMed]
  165. Khor, Y.Y.; Lee, S.-K.; Devi, M.D.; Ling, W.C. Epigallocatechin-3-gallate exerts antihypertensive effects and improves endothelial function in spontaneously hypertensive rats. Asian Pac. J. Trop. Biomed. 2023, 13, 287–295. [Google Scholar] [CrossRef]
  166. Zhou, X.; Liang, L.; Zhao, Y.; Zhang, H. Epigallocatechin-3-gallate ameliorates angiotensin II-induced oxidative stress and apoptosis in human umbilical vein endothelial cells through the activation of Nrf2/caspase-3 signaling. J. Vasc. Res. 2017, 54, 299–308. [Google Scholar] [CrossRef]
  167. Lee, H.S.; Jun, J.-H.; Jung, E.-H.; Koo, B.A.; Kim, Y.S. Epigalloccatechin-3-gallate inhibits ocular neovascularization and vascular permeability in human retinal pigment epithelial and human retinal microvascular endothelial cells via suppression of MMP-9 and VEGF activation. Molecules 2014, 19, 12150–12172. [Google Scholar] [CrossRef]
  168. Bian, Y.; Lei, J.; Zhong, J.; Wang, B.; Wan, Y.; Li, J.; Liao, C.; He, Y.; Liu, Z.; Ito, K.; et al. Kaempferol reduces obesity, prevents intestinal inflammation, and modulates gut microbiota in high-fat diet mice. J. Nutr. Biochem. 2022, 99, 108840. [Google Scholar] [CrossRef]
  169. Aa, L.-X.; Fei, F.; Qi, Q.; Sun, R.B.; Gu, S.H.; Di, Z.Z.; Aa, J.Y.; Wang, G.J.; Liu, C.X. Rebalancing of the gut flora and microbial metabolism is responsible for the anti-arthritis effect of kaempferol. Acta Pharmacol. Sin. 2020, 41, 73–81. [Google Scholar] [CrossRef]
  170. Yao, H.; Sun, J.; Wei, J.; Zhang, X.; Chen, B.; Lin, Y. Kaempferol protects blood vessels from damage induced by oxidative stress and inflammation in association with the Nrf2/HO-1 signaling pathway. Front. Pharmacol. 2020, 11, 1118. [Google Scholar] [CrossRef]
  171. Che, J.; Liang, B.; Zhang, Y.; Wang, Y.; Tang, J.; Shi, G. Kaempferol alleviates ox-LDL-induced apoptosis by up-regulation of autophagy via inhibiting PI3K/Akt/mTOR pathway in human endothelial cells. Cardiovasc. Pathol. 2017, 31, 57–62. [Google Scholar] [CrossRef]
  172. De Bruyne, T.; Steenput, B.; Roth, L.; De Meyer, G.R.Y.; dos Santos, C.N.; Valentová, K.; Dambrova, M.; Hermans, N. Dietary polyphenols targeting arterial stiffness: Interplay of contributing mechanisms and gut microbiome-related metabolism. Nutrients 2019, 11, 578. [Google Scholar] [CrossRef]
  173. Zhou, D.-D.; Luo, M.; Huang, S.-Y.; Saimaiti, A.; Shang, A.; Gan, R.Y.; Li, H.B. Effects and mechanisms of resveratrol on aging and age-related diseases. Oxidative Med. Cell. Longev. 2021, 2021, 9932218. [Google Scholar] [CrossRef]
  174. Man, A.W.; Li, H.; Xia, N. Resveratrol and the interaction between gut microbiota and arterial remodelling. Nutrients 2020, 12, 119. [Google Scholar] [CrossRef]
  175. Chaplin, A.; Carpéné, C.; Mercader, J. Resveratrol, metabolic syndrome, and gut microbiota. Nutrients 2018, 10, 1651. [Google Scholar] [CrossRef] [PubMed]
  176. Zhao, L.; Zhang, Q.; Ma, W.; Tian, F.; Shen, H.; Zhou, M. A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota. Food Funct. 2017, 8, 4644–4656. [Google Scholar] [CrossRef] [PubMed]
  177. Chen, M.-l.; Yi, L.; Zhang, Y.; Zhou, X.; Ran, L.; Yang, J.; Zhu, J.D.; Zhang, Q.Y.; Mi, M.T. Resveratrol attenuates trimethylamine-N-oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. MBio 2016, 7, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  178. Qiao, Y.; Sun, J.; Xia, S.; Tang, X.; Shi, Y.; Le, G. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct. 2014, 5, 1241–1249. [Google Scholar] [CrossRef]
  179. Sung, M.M.; Kim, T.T.; Denou, E.; Soltys, C.L.M.; Hamza, S.M.; Byrne, N.J.; Masson, G.; Park, H.; Wishart, D.S.; Madsen, K.L.; et al. Improved glucose homeostasis in obese mice treated with resveratrol is associated with alterations in the gut microbiome. Diabetes 2017, 66, 418–425. [Google Scholar] [CrossRef]
  180. Gianchecchi, E.; Fierabracci, A. Insights on the effects of resveratrol and some of its derivatives in cancer and autoimmunity: A molecule with a dual activity. Antioxidants 2020, 9, 91. [Google Scholar] [CrossRef]
  181. Li, H.; Xia, N.; Hasselwander, S.; Daiber, A. Resveratrol and vascular function. Int. J. Mol. Sci. 2019, 20, 2155. [Google Scholar] [CrossRef]
  182. Shi, A.; Li, T.; Zheng, Y.; Song, Y.; Wang, H.; Wang, N.; Dong, L.; Shi, H. Chlorogenic acid improves NAFLD by regulating gut microbiota and GLP-1. Front. Pharmacol. 2021, 12, 693048. [Google Scholar] [CrossRef]
  183. Wang, Z.; Lam, K.L.; Hu, J.; Ge, S.; Zhou, A.; Zheng, B.; Zeng, S.; Lin, S. Chlorogenic acid alleviates obesity and modulates gut microbiota in high-fat-fed mice. Food Sci. Nutr. 2019, 7, 579–588. [Google Scholar] [CrossRef] [PubMed]
  184. Ye, X.; Liu, Y.; Hu, J.; Gao, Y.; Ma, Y.; Wen, D. Chlorogenic acid-induced gut microbiota improves metabolic endotoxemia. Front. Endocrinol. 2021, 12, 762691. [Google Scholar] [CrossRef] [PubMed]
  185. Xie, M.G.; Fei, Y.Q.; Wang, Y.; Wang, W.Y.; Wang, Z. Chlorogenic acid alleviates colon mucosal damage induced by a high-fat diet via gut microflora adjustment to increase short-chain fatty acid accumulation in rats. Oxidative Med. Cell. Longev. 2021, 2021, 3456542. [Google Scholar] [CrossRef]
  186. Wuttimongkolchai, N.; Kanlaya, R.; Nanthawuttiphan, S.; Subkod, C.; Thongboonkerd, V. Chlorogenic acid enhances endothelial barrier function and promotes endothelial tube formation: A proteomics approach and functional validation. Biomed. Pharmacother. 2022, 153, 113471. [Google Scholar] [CrossRef]
  187. Tsai, K.L.; Hung, C.H.; Chan, S.H.; Hsieh, P.L.; Ou, H.C.; Cheng, Y.H.; Chu, P.M. Chlorogenic acid protects against oxLDL-induced oxidative damage and mitochondrial dysfunction by modulating SIRT1 in endothelial cells. Mol. Nutr. Food Res. 2018, 62, 1700928. [Google Scholar] [CrossRef]
  188. Hada, Y.; Uchida, H.A.; Otaka, N.; Onishi, Y.; Okamoto, S.; Nishiwaki, M.; Takemoto, R.; Takeuchi, H.; Wada, J. The protective effect of chlorogenic acid on vascular senescence via the Nrf2/HO-1 pathway. Int. J. Mol. Sci. 2020, 21, 4527. [Google Scholar] [CrossRef]
  189. Zhao, Y.; Wang, J.; Ballevre, O.; Luo, H.; Zhang, W. Antihypertensive effects and mechanisms of chlorogenic acids. Hypertens. Res. 2012, 35, 370–374. [Google Scholar] [CrossRef]
  190. Xu, Q.; Shen, M.; Han, Y.; Diao, H. Effects of ellagic acid supplementation on jejunal morphology, digestive enzyme activities, antioxidant capacity, and microbiota in mice. Front. Microbiol. 2021, 12, 793576. [Google Scholar] [CrossRef]
  191. Zhang, M.; Cui, S.; Mao, B.; Zhang, Q.; Zhao, J.; Zhang, H.; Tang, X.; Chen, W. Ellagic acid and intestinal microflora metabolite urolithin A: A review on its sources, metabolic distribution, health benefits, and biotransformation. Crit. Rev. Food Sci. Nutr. 2023, 63, 6900–6922. [Google Scholar] [CrossRef]
  192. Xian, W.; Deng, Y.; Yang, Y.; Tan, Z.; Chen, C.; Li, W.; Yang, R. Ameliorative effect of ellagic acid on aging in rats with the potential mechanism relying on the gut microbiota and urolithin a-producing ability. J. Agric. Food Chem. 2023, 71, 7396–7407. [Google Scholar] [CrossRef]
  193. da Silva, F.B.; Romero, W.G.; Rouver, W.D.N.; Silva, K.; de Almeida, S.A.; Mengal, V.; Peluso, A.A.; Endlich, P.W.; Bissoli, N.S.; Claudio, E.R.G.; et al. Ellagic Acid prevents vascular dysfunction in small mesenteric arteries of ovariectomized hypertensive rats. J. Nutr. Biochem. 2022, 105, 108995. [Google Scholar] [CrossRef]
  194. D’Amico, D.; Andreux, P.A.; Valdés, P.; Singh, A.; Rinsch, C.; Auwerx, J. Impact of the natural compound urolithin A on health, disease, and aging. Trends Mol. Med. 2021, 27, 687–699. [Google Scholar] [CrossRef]
  195. Tian, B.; Geng, Y.; Wang, P.; Cai, M.; Neng, J.; Hu, J.; Xia, D.; Cao, W.; Yang, K.; Sun, P. Ferulic acid improves intestinal barrier function through altering gut microbiota composition in high-fat diet-induced mice. Eur. J. Nutr. 2022, 61, 3767–3783. [Google Scholar] [CrossRef] [PubMed]
  196. Liu, Y.; Shi, L.; Qiu, W.; Shi, Y. Ferulic acid exhibits anti-inflammatory effects by inducing autophagy and blocking NLRP3 inflammasome activation. Mol. Cell. Toxicol. 2022, 18, 509–519. [Google Scholar] [CrossRef] [PubMed]
  197. Gu, Y.; Zhang, Y.; Li, M.; Huang, Z.; Jiang, J.; Chen, Y.; Chen, J.; Jia, Y.; Zhang, L.; Zhou, F. Ferulic acid ameliorates atherosclerotic injury by modulating gut microbiota and lipid metabolism. Front. Pharmacol. 2021, 12, 621339. [Google Scholar] [CrossRef] [PubMed]
  198. Zhang, L.; Lan, Y.; Wang, Y.; Yang, Y.; Han, W.; Li, J.; Wang, Y.; Liu, X. Secoisolariciresinol diglucoside ameliorates high fat diet-induced colon inflammation and regulates gut microbiota in mice. Food Funct. 2022, 13, 3009–3022. [Google Scholar] [CrossRef]
  199. Zhang, S.; Cheng, M.; Wang, Z.; Liu, Y.; Ren, Y.; Rong, S.; Wang, X. Secoisolariciresinol diglucoside exerts anti-inflammatory and antiapoptotic effects through inhibiting the Akt/IκB/NF-κB pathway on human umbilical vein endothelial cells. Mediat. Inflamm. 2020, 2020, 3621261. [Google Scholar] [CrossRef]
  200. Moens, F.; De Vuyst, L. Inulin-type fructan degradation capacity of Clostridium cluster IV and XIVa butyrate-producing colon bacteria and their associated metabolic outcomes. Benef. Microbes 2017, 8, 473–490. [Google Scholar] [CrossRef]
  201. Zou, J.; Reddivari, L.; Shi, Z.; Li, S.; Wang, Y.; Bretin, A.; Ngo, V.; Flythe, M.; Pellizzon, M.; Chassaing, B.; et al. Inulin fermentable fiber ameliorates type I diabetes via IL22 and short-chain fatty acids in experimental models. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 983–1000. [Google Scholar] [CrossRef] [PubMed]
  202. Zou, J.; Chassaing, B.; Singh, V.; Pellizzon, M.; Ricci, M.; Fythe, M.; Kumar, M.; Gewirtz, A. Fiber-mediated nourishment of gut microbiota protects against diet-induced obesity by restoring IL-22-mediated colonic health. Cell Host Microbe 2018, 23, 41–53.e4. [Google Scholar] [CrossRef] [PubMed]
  203. Azuma, N.; Saito, Y.; Nishijima, T.; Aoki, R.; Nishihira, J. Effect of daily ingestion of Bifidobacterium and dietary fiber on vascular endothelial function: A randomized, double-blind, placebo-controlled, parallel-group comparison study. Biosci. Biotechnol. Biochem. 2024, 88, 86–96. [Google Scholar] [CrossRef] [PubMed]
  204. Ruskovska, T.; Konic-Ristic, A.; Mazur, A.; Milenkovic, D. Molecular mechanisms underlying hypertensive effect of fructose and the preventive properties of inulin–Global transcriptomic analysis in rat aorta. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 441–456. [Google Scholar] [CrossRef]
  205. Catry, E.; Bindels, L.B.; Tailleux, A.; Lestavel, S.; Netrinck, A.; Goossens, J.; Lobysheva, I.; Plovier, H.; Essaghir, A.; Demoulin, J.; et al. Targeting the gut microbiota with inulin-type fructans: Preclinical demonstration of a novel approach in the management of endothelial dysfunction. Gut 2018, 67, 271–283. [Google Scholar] [CrossRef]
  206. Elshahed, M.S.; Miron, A.; Aprotosoaie, A.C.; Farag, M.A. Pectin in diet: Interactions with the human microbiome, role in gut homeostasis, and nutrient-drug interactions. Carbohydr. Polym. 2021, 255, 117388. [Google Scholar] [CrossRef]
  207. Chung, W.S.F.; Walker, A.W.; Louis, P.; Parkhill, J.; Vermeiren, J.; Bosscher, D.; Duncan, S.; Flint, H. Modulation of the human gut microbiota by dietary fibres occurs at the species level. BMC Biol. 2016, 14, 3. [Google Scholar] [CrossRef]
  208. García-Carrizo, F.; Galmés, S.; Picó, C.; Palou, A.; Rodriguez, A.M. Supplementation with the prebiotic high-esterified pectin improves blood pressure and cardiovascular risk biomarker profile counteracting metabolic malprogramming. J. Agric. Food Chem. 2022, 70, 13200–13211. [Google Scholar]
  209. Sung, M.; Yoon, Y.; Lee, J. The immunomodulatory effect of β-glucan depends on the composition of the gut microbiota. Foods 2023, 12, 3148. [Google Scholar] [CrossRef]
  210. Gao, H.; Song, R.; Li, Y.; Zhang, W.; Wan, Z.; Wang, Y.; Zhang, H.; Han, S. Effects of oat fiber intervention on cognitive behavior in LDLR–/– mice modeling atherosclerosis by targeting the microbiome–Gut–brain axis. J. Agric. Food Chem. 2020, 68, 14480–14491. [Google Scholar] [CrossRef]
  211. Thandapilly, S.J.; Ndou, S.P.; Wang, Y.; Nyachoti, C.M.; Ames, N.P. Barley β-glucan increases fecal bile acid excretion and short chain fatty acid levels in mildly hypercholesterolemic individuals. Food Funct. 2018, 9, 3092–3096. [Google Scholar] [CrossRef]
  212. Bezerra, L.S.; Magnani, M.; Castro-Gomez, R.J.H.; Cavalcante, H.C.; da Silva, T.A.F.; Vieira, R.L.P.; de Medeiros, I.A.; Veras, R.C. Modulation of vascular function and anti-aggregation effect induced by (1→ 3)(1→ 6)-β-d-glucan of Saccharomyces cerevisiae and its carboxymethylated derivative in rats. Pharmacol. Rep. 2017, 69, 448–455. [Google Scholar] [CrossRef]
  213. Wang, Y.; Harding, S.V.; Eck, P.; Thandapilly, S.J.; Gamel, T.H.; Abdel-Aal, E.M.; Crow, G.H.; Tosh, S.M.; Jones, P.J.; Ames, N.P. High-molecular-weight β-glucan decreases serum cholesterol differentially based on the CYP7A1 rs3808607 polymorphism in mildly hypercholesterolemic adults. J. Nutr. 2015, 146, 720–727. [Google Scholar] [CrossRef] [PubMed]
  214. Smith, K.N.; Queenan, K.M.; Thomas, W.; Fulcher, R.G.; Slavin, J.L. Physiological effects of concentrated barley β-glucan in mildly hypercholesterolemic adults. J. Am. Coll. Nutr. 2008, 27, 434–440. [Google Scholar] [CrossRef] [PubMed]
  215. Gounaris, A.K.; Grivea, I.N.; Baltogianni, M.; Gounari, E.; Antonogeorgos, G.; Kokori, F. Caffeine and gastric emptying time in very preterm neonates. J. Clin. Med. 2020, 9, 1676. [Google Scholar] [CrossRef] [PubMed]
  216. Liszt, K.I.; Ley, J.P.; Lieder, B.; Behrens, M.; Stöger, V.; Reiner, A.; Hochkogler, C.M.; Köck, E.; Marchiori, A.; Hans, J.; et al. Caffeine induces gastric acid secretion via bitter taste signaling in gastric parietal cells. Proc. Natl. Acad. Sci. USA 2017, 114, E6260–E6269. [Google Scholar] [CrossRef]
  217. Gurwara, S.; Dai, A.; Ajami, N.; El-Serag, H.B.; Graham, D.Y.; Jiao, L. 196 caffeine consumption and the colonic mucosa-associated gut microbiota. Off. J. Am. Coll. Gastroenterol.|ACG 2019, 114, S119–S120. [Google Scholar] [CrossRef]
  218. Chen, L.; Wang, X.-J.; Chen, J.-X.; Yang, J.C.; Lin, L.; Cai, X.B.; Chen, Y.S. Caffeine ameliorates the metabolic syndrome in diet-induced obese mice through regulating the gut microbiota and serum metabolism. Diabetol. Metab. Syndr. 2023, 15, 37. [Google Scholar] [CrossRef]
  219. Nishitsuji, K.; Watanabe, S.; Xiao, J.; Nagatomo, R.; Ogawa, H.; Tsunematsu, T.; Umemoto, H.; Morimoto, Y.; Akatsu, H.; Inoue, K.; et al. Effect of coffee or coffee components on gut microbiome and short-chain fatty acids in a mouse model of metabolic syndrome. Sci. Rep. 2018, 8, 16173. [Google Scholar] [CrossRef]
  220. Vercambre, M.-N.; Berr, C.; Ritchie, K.; Kang, J.H. Caffeine and cognitive decline in elderly women at high vascular risk. J. Alzheimer’s Dis. 2013, 35, 413–421. [Google Scholar] [CrossRef]
  221. Shechter, M.; Shalmon, G.; Scheinowitz, M.; Koren-Morag, N.; Feinberg, M.S.; Harats, D.; Sela, B.A.; Sharabi, Y.; Chouraqui, P. Impact of acute caffeine ingestion on endothelial function in subjects with without coronary artery disease. Am. J. Cardiol. 2011, 107, 1255–1261. [Google Scholar] [CrossRef] [PubMed]
  222. Umemura, T.; Ueda, K.; Nishioka, K.; Hidaka, T.; Takemoto, H.; Nakamura, S.; Jitsuiki, D.; Soga, J.; Goto, C.; Chayama, K.; et al. Effects of acute administration of caffeine on vascular function. Am. J. Cardiol. 2006, 98, 1538–1541. [Google Scholar] [CrossRef] [PubMed]
  223. Wang, F.; Huang, X.; Chen, Y.; Zhang, D.; Chen, D.; Chen, L.; Lin, J. Study on the effect of capsaicin on the intestinal flora through high-throughput sequencing. ACS Omega 2020, 5, 1246–1253. [Google Scholar] [CrossRef] [PubMed]
  224. Zhu, S.L.; Wang, M.L.; He, Y.T.; Guo, S.W.; Li, T.T.; Peng, W.J.; Luo, D. Capsaicin ameliorates intermittent high glucose-mediated endothelial senescence via the TRPV1/SIRT1 pathway. Phytomedicine 2022, 100, 154081. [Google Scholar] [CrossRef]
  225. Luo, D.; Li, W.; Xie, C.; Yin, L.; Su, X.; Chen, J.; Huang, H. Capsaicin attenuates arterial calcification through promoting SIRT6-mediated deacetylation and degradation of Hif1α (Hypoxic-Inducible Factor-1 alpha). Hypertension 2022, 79, 906–917. [Google Scholar] [CrossRef]
  226. Chularojmontri, L.; Suwatronnakorn, M.; Wattanapitayakul, S.K. Influence of capsicum extract and capsaicin on endothelial health. J. Med. Assoc. Thail. 2011, 93, 92. [Google Scholar]
  227. Cuevas-Tena, M.; Bermúdez, J.D.; de los Ángeles Silvestre, R.; Alegría, A.; Lagarda, M.J. Impact of colonic fermentation on sterols after the intake of a plant sterol-enriched beverage: A randomized, double-blind crossover trial. Clin. Nutr. 2019, 38, 1549–1560. [Google Scholar] [CrossRef]
  228. Jayaraman, S.; Devarajan, N.; Rajagopal, P.; Babu, S.; Ganesan, S.K.; Veeraraghavan, V.P.; Palanisamy, C.P.; Cui, B.; Periyasamy, V.; Chandrasekar, K. β-Sitosterol circumvents obesity induced inflammation and insulin resistance by down-regulating IKKβ/NF-κB and JNK signaling pathway in adipocytes of type 2 diabetic rats. Molecules 2021, 26, 2101. [Google Scholar] [CrossRef]
  229. McCarron, A.; Donnelley, M.; Parsons, D. Airway disease phenotypes in animal models of cystic fibrosis. Respir. Res. 2018, 19, 54. [Google Scholar] [CrossRef]
  230. Gogoi, D.; Pal, A.; Chattopadhyay, P.; Paul, S.; Deka, R.C.; Mukherjee, A.K. First report of plant-derived β-sitosterol with antithrombotic, in vivo anticoagulant, and thrombus-preventing activities in a mouse model. J. Nat. Prod. 2018, 81, 2521–2530. [Google Scholar] [CrossRef]
  231. Xiao, X.; Wang, J.; Zhu, Y.; Deng, B.; Liu, Y.; Wang, S.; Hou, T.; Song, T. Phytosterols Protect against Osteoporosis by Regulating Gut Microbiota. J. Agric. Food Chem. 2023, 71, 14539–14549. [Google Scholar] [CrossRef]
  232. Ma, L.; Ma, Y.; Liu, Y. β-Sitosterol protects against food allergic response in BALB/c mice by regulating the intestinal barrier function and reconstructing the gut microbiota structure. Food Funct. 2023, 14, 4456–4469. [Google Scholar] [CrossRef]
  233. He, S.; He, S.; Chen, Y.; Jin, X.; Mei, W.; Lu, Q. Beta-sitosterol modulates the migration of vascular smooth muscle cells via the PPARG/AMPK/mTOR pathway. Pharmacology 2022, 107, 495–509. [Google Scholar] [CrossRef] [PubMed]
  234. Xin, Y.; Li, X.; Zhu, X.; Lin, X.; Luo, M.; Xiao, Y.; Ruan, Y.; Guo, H. Stigmasterol Protects Against Steatohepatitis Induced by High-Fat and High-Cholesterol Diet in Mice by Enhancing the Alternative Bile Acid Synthesis Pathway. J. Nutr. 2023, 153, 1903–1914. [Google Scholar] [CrossRef] [PubMed]
  235. Huo, R.; Yang, W.-J.; Liu, Y.; Liu, T.; Li, T.; Wang, C.Y.; Pan, B.S.; Wang, B.L.; Guo, W. Stigmasterol: Remodeling Gut Microbiota and Suppressing Tumor Growth through Treg and CD8+ T Cells in Hepatocellular Carcinoma. Phytomedicine 2023, 129, 155225. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, Y.; Gu, Y.; Jiang, J.; Cui, X.; Cheng, S.; Liu, L.; Huang, Z.; Liao, R.; Zhao, P.; Yu, J.; et al. Stigmasterol attenuates hepatic steatosis in rats by strengthening the intestinal barrier and improving bile acid metabolism. npj Sci. Food 2022, 6, 38. [Google Scholar] [CrossRef]
  237. Sun, J.; Zhao, Y. Stigmasterol Inhibits High Glucose Induced Proliferation and Angiogenesis of Retinal Endothelial Cells by Regulating Mitogen-Activated Protein Kinase Signaling Pathway. Curr. Top. Nutraceutical Res. 2022, 20, 617. [Google Scholar] [CrossRef]
  238. Corral-Jara, K.F.; Nuthikattu, S.; Rutledge, J.; Villablanca, A.; Fong, R.; Heiss, C.; Ottaviani, J.I.; Milenkovic, D. Structurally related (−)-epicatechin metabolites and gut microbiota derived metabolites exert genomic modifications via VEGF signaling pathways in brain microvascular endothelial cells under lipotoxic conditions: Integrated multi-omic study. J. Proteom. 2022, 263, 104603. [Google Scholar] [CrossRef]
  239. Chu, T.; Yu, R.; Gu, Y.; Wang, Y.; Chang, H.; Li, Y.; Li, J.; Bian, Y. Kaempferol protects gut-vascular barrier from high glucose-induced disorder via NF-κB pathway. J. Nutr. Biochem. 2024, 123, 109496. [Google Scholar] [CrossRef]
  240. Godos, J.; Romano, G.L.; Gozzo, L.; Laudani, S.; Paladino, N.; Dominguez Azpíroz, I.; Martínez López, N.M.; Giampieri, F.; Quiles, J.L.; Battino, M.; et al. Resveratrol and vascular health: Evidence from clinical studies and mechanisms of actions related to its metabolites produced by gut microbiota. Front. Pharmacol. 2024, 15, 1368949. [Google Scholar] [CrossRef]
  241. Zhu, Q.; Zhu, Y.; Liu, Y.; Tao, Y.; Lin, Y.; Lai, S.; Liang, Z.; Chen, Y.; Chen, Y.; Wang, L. Moderation of gut microbiota and bile acid metabolism by chlorogenic acid improves high-fructose-induced salt-sensitive hypertension in mice. Food Funct. 2022, 13, 6987–6999. [Google Scholar] [CrossRef]
  242. Nishimoto, Y.; Fujisawa, K.; Ukawa, Y.; Kudoh, M.; Funahashi, K.; Kishimoto, Y.; Fukuda, S. Effect of urolithin A on the improvement of vascular endothelial function depends on the gut microbiota. Front. Nutr. 2023, 9, 1077534. [Google Scholar] [CrossRef] [PubMed]
  243. Li, J.; Liao, R.; Zhang, S.; Weng, H.; Liu, Y.; Tao, T.; Yu, F.; Li, G.; Wu, J. Promising remedies for cardiovascular disease: Natural polyphenol ellagic acid and its metabolite urolithins. Phytomedicine 2023, 116, 154867. [Google Scholar] [CrossRef] [PubMed]
  244. Yu, L.; Xu, Q.; Wang, P.; Luo, J.; Zheng, Z.; Zhou, J.; Zhang, L.; Sun, L.; Zuo, D. Secoisolariciresinol diglucoside-derived metabolite, enterolactone, attenuates atopic dermatitis by suppressing Th2 immune response. Int. Immunopharmacol. 2022, 111, 109039. [Google Scholar] [CrossRef]
  245. Boon, E.A.; Croft, K.D.; Shinde, S.; Hodgson, J.M.; Ward, N.C. The acute effect of coffee on endothelial function and glucose metabolism following a glucose load in healthy human volunteers. Food Funct. 2017, 8, 3366–3373. [Google Scholar] [CrossRef]
  246. Rosca, A.E.; Iesanu, M.I.; Zahiu, C.D.M.; Voiculescu, S.E.; Paslaru, A.C.; Zagrean, A.M. Capsaicin and gut microbiota in health and disease. Molecules 2020, 25, 5681. [Google Scholar] [CrossRef]
  247. Yu, Y.; Cao, Y.; Huang, W.; Liu, Y.; Lu, Y.; Zhao, J. β-Sitosterol ameliorates endometrium receptivity in PCOS-like mice: The mediation of gut microbiota. Front. Nutr. 2021, 8, 667130. [Google Scholar] [CrossRef]
  248. Zhang, X.; Wang, J.; Zhu, L.; Wang, X.; Meng, F.; Xia, L.; Zhang, H. Advances in Stigmasterol on its anti-tumor effect and mechanism of action. Front. Oncol. 2022, 12, 1101289. [Google Scholar] [CrossRef]
  249. Duan, R.; Guan, X.; Huang, K.; Zhang, Y.; Li, S.; Xia, J.A.; Shen, M. Flavonoids from whole-grain oat alleviated high-fat diet-induced hyperlipidemia via regulating bile acid metabolism gut microbiota in mice. J. Agric. Food Chem. 2021, 69, 7629–7640. [Google Scholar] [CrossRef]
  250. Xu, M.; Lv, C.; Wang, H.; Lu, Q.; Ye, M.; Zhu, X.; Liu, R. Peanut skin extract ameliorates high-fat diet-induced atherosclerosis by regulating lipid metabolism, inflammation reaction and gut microbiota in ApoE−/− mice. Food Res. Int. 2022, 154, 111014. [Google Scholar] [CrossRef]
  251. Wu, M.; Wu, X.; Zhu, J.; Li, F.; Wei, X.; Wang, Y. Selenium-enriched and ordinary green tea extracts prevent high blood pressure and alter gut microbiota composition of hypertensive rats caused by high-salt diet. Food Sci. Hum. Wellness 2022, 11, 738–751. [Google Scholar] [CrossRef]
  252. Cheng, X.; Han, X.; Zhou, L.; Sun, Y.; Zhou, Q.; Lin, X.; Gao, Z.; Wang, J.; Zhao, W. Cabernet sauvignon dry red wine ameliorates atherosclerosis in mice by regulating inflammation and endothelial function, activating AMPK phosphorylation, and modulating gut microbiota. Food Res. Int. 2023, 169, 112942. [Google Scholar] [CrossRef] [PubMed]
  253. Guan, Y.; Chen, K.; Quan, D.; Kang, L.; Yang, D.; Wu, H.; Yan, M.; Wu, S.; Lv, L.; Zhang, G. The combination of Scutellaria baicalensis Georgi and Sophora japonica L. ameliorate renal function by regulating gut microbiota in spontaneously hypertensive rats. Front. Pharmacol. 2021, 11, 575294. [Google Scholar] [CrossRef] [PubMed]
  254. Milton-Laskibar, I.; Marcos-Zambrano, L.J.; Gómez-Zorita, S.; Fernández-Quintela, A.; Carrillo de Santa Pau, E.; Martínez, J.A.; Portillo, M.P. Gut microbiota induced by pterostilbene and resveratrol in high-fat-high-fructose fed rats: Putative role in steatohepatitis onset. Nutrients 2021, 13, 1738. [Google Scholar] [CrossRef] [PubMed]
  255. Chowdhury, F.I.; Yasmin, T.; Akter, R.; Islam, M.N.; Hossain, M.M.; Khan, F.; Aldhahrani, A.; Soliman, M.M.; Subhan, N.; Haque, M.A.; et al. Resveratrol treatment modulates several antioxidant and anti-inflammatory genes expression and ameliorated oxidative stress mediated fibrosis in the kidneys of high-fat diet-fed rats. Saudi Pharm. J. 2022, 30, 1454–1463. [Google Scholar] [CrossRef]
  256. Suzuki, A.; Nomura, T.; Jokura, H.; Kitamura, N.; Saiki, A.; Fujii, A. Chlorogenic acid-enriched green coffee bean extract affects arterial stiffness assessed by the cardio-ankle vascular index in healthy men: A pilot study. Int. J. Food Sci. Nutr. 2019, 70, 901–908. [Google Scholar] [CrossRef]
  257. Bhandarkar, N.S.; Brown, L.; Panchal, S.K. Chlorogenic acid attenuates high-carbohydrate, high-fat diet–induced cardiovascular, liver, and metabolic changes in rats. Nutr. Res. 2019, 62, 78–88. [Google Scholar] [CrossRef]
  258. Hidalgo-Lozada, G.M.; Villarruel-López, A.; Martínez-Abundis, E.; Vázquez-Paulino, O.; González-Ortiz, M.; Pérez-Rubio, K.G. Ellagic acid effect on the components of metabolic syndrome, insulin sensitivity and insulin secretion: A randomized, double-blind, placebo-controlled clinical trial. J. Clin. Med. 2022, 11, 5741. [Google Scholar] [CrossRef]
  259. Hoving, L.R.; De Vries, M.R.; de Jong, R.C.; Katiraei, S.; Pronk, A.; Quax, P.H.; Van Harmelen, V.; Willems van Dijk, K. The prebiotic inulin aggravates accelerated atherosclerosis in hypercholesterolemic APOE* 3-Leiden mice. Nutrients 2018, 10, 172. [Google Scholar] [CrossRef]
  260. Chen, Y.; Xu, C.; Huang, R.; Song, J.; Li, D.; Xia, M. Butyrate from pectin fermentation inhibits intestinal cholesterol absorption and attenuates atherosclerosis in apolipoprotein E-deficient mice. J. Nutr. Biochem. 2018, 56, 175–182. [Google Scholar] [CrossRef]
  261. Han, S.; Zhang, W.; Zhang, R.; Jiao, J.; Fu, C.; Tong, X.; Zhang, W.; Qin, L. Cereal fiber improves blood cholesterol profiles and modulates intestinal cholesterol metabolism in C57BL/6 mice fed a high-fat, high-cholesterol diet. Food Nutr. Res. 2019, 63, 10–29219. [Google Scholar] [CrossRef]
  262. Panchal, S.K.; Poudyal, H.; Waanders, J.; Brown, L. Coffee extract attenuates changes in cardiovascular and hepatic structure and function without decreasing obesity in high-carbohydrate, high-fat diet-fed male rats. J. Nutr. 2012, 142, 690–697. [Google Scholar] [CrossRef]
  263. Dai, Z.; Li, S.; Meng, Y.; Zhao, Q.; Zhang, Y.; Suonan, Z.; Sun, Y.; Shen, Q.; Liao, X.; Xue, Y. Capsaicin Ameliorates High-Fat Diet-Induced Atherosclerosis in ApoE−/− Mice via Remodeling Gut Microbiota. Nutrients 2022, 14, 4334. [Google Scholar] [CrossRef]
  264. Wu, W.; Liu, W.; Wang, H.; Wang, W.; Chu, W.; Jin, J. β-sitosterol inhibits trimethylamine production by regulating the gut microbiota and attenuates atherosclerosis in ApoE–/– mice. Front. Cardiovasc. Med. 2022, 9, 986905. [Google Scholar] [CrossRef]
  265. Yan, H.; Hu, Y.; Akk, A.; Wickline, S.A.; Pan, H.; Pham, C.T. Peptide-siRNA nanoparticles targeting NF-κB p50 mitigate experimental abdominal aortic aneurysm progression and rupture. Biomater. Adv. 2022, 139, 213009. [Google Scholar] [CrossRef]
  266. Walker, R.L.; Vlamakis, H.; Lee, J.W.J.; Besse, L.A.; Xanthakis, V.; Vasan, R.S.; Shaw, S.Y.; Xavier, R.J. Population study of the gut microbiome: Associations with diet, lifestyle, and cardiometabolic disease. Genome Med. 2021, 13, 188. [Google Scholar] [CrossRef]
  267. Bubnov, R.; Spivak, M. Pathophysiology-based individualized use of probiotics and prebiotics for metabolic syndrome: Implementing predictive, preventive, and personalized medical approach. In Microbiome in 3P Medicine Strategies: The First Exploitation Guide; Springer International Publishing: Cham, Switzerland, 2023; pp. 133–196. [Google Scholar]
Figure 1. This model predicts that the mechanisms of vascular aging mainly include (i) inflammation and oxidative stress, (ii) mitochondrial dysfunction, (iii) loss of proteostasis, and (iv) cellular senescence and apoptosis, all potentially modulated by gut microbiota. Through these shifts in biological function with aging, structural and functional alterations occur in vascular cells (ECs and VSMCs), which result in vascular aging and age-related vascular diseases. (i) During aging, the increased level of O2 generated by the electron transport chain in senescent EC are dismutated to H2O2 in mitochondria by SOD. The excessive H2O2 contributes to the activation of NFκB, resulting in an accumulation of pro-inflammatory cytokines. On the other hand, up-regulation of TNFα in senescent EC, at least in part, promotes the production of O2 by the NAD (P)H oxidase and down-regulates eNOS, which is responsible for the impaired bioavailability of NO and endothelial dysfunction. (ii) Vascular aging is associated with progressive mitochondrial dysfunction that occurs due to excessive ROS generation, accumulation of damaged mtDNA, impaired oxygen consumption rate, and suppressed mitochondrial activities (mitochondrial dynamic, mitochondrial biogenesis, and mitophagy). The disordered mitochondria accumulated in vascular cells, leading to cellular damage and metabolic disorders. (iii) Aging stress on the vascular system triggers increased demand for protein folding, whereas the proteotasis system, including ubiquitin-proteasome system (UPS), chaperone-mediated autophagy (CMA), and macro-autophagy, are significantly declined with aging, which causes the accumulation of aggregated proteins and thus aggravates aging stress. (iv) A verities of external aging stress factors, including replicative stress, telomere attrition, oncogene activation, radiations, and chemo-therapeutic drugs, can trigger the exit of senescence-associated cell cycle, which is regulated with the initiation of p16INK4a/Rb and p53/p21 pathways. Idea adapted from [19].
Figure 1. This model predicts that the mechanisms of vascular aging mainly include (i) inflammation and oxidative stress, (ii) mitochondrial dysfunction, (iii) loss of proteostasis, and (iv) cellular senescence and apoptosis, all potentially modulated by gut microbiota. Through these shifts in biological function with aging, structural and functional alterations occur in vascular cells (ECs and VSMCs), which result in vascular aging and age-related vascular diseases. (i) During aging, the increased level of O2 generated by the electron transport chain in senescent EC are dismutated to H2O2 in mitochondria by SOD. The excessive H2O2 contributes to the activation of NFκB, resulting in an accumulation of pro-inflammatory cytokines. On the other hand, up-regulation of TNFα in senescent EC, at least in part, promotes the production of O2 by the NAD (P)H oxidase and down-regulates eNOS, which is responsible for the impaired bioavailability of NO and endothelial dysfunction. (ii) Vascular aging is associated with progressive mitochondrial dysfunction that occurs due to excessive ROS generation, accumulation of damaged mtDNA, impaired oxygen consumption rate, and suppressed mitochondrial activities (mitochondrial dynamic, mitochondrial biogenesis, and mitophagy). The disordered mitochondria accumulated in vascular cells, leading to cellular damage and metabolic disorders. (iii) Aging stress on the vascular system triggers increased demand for protein folding, whereas the proteotasis system, including ubiquitin-proteasome system (UPS), chaperone-mediated autophagy (CMA), and macro-autophagy, are significantly declined with aging, which causes the accumulation of aggregated proteins and thus aggravates aging stress. (iv) A verities of external aging stress factors, including replicative stress, telomere attrition, oncogene activation, radiations, and chemo-therapeutic drugs, can trigger the exit of senescence-associated cell cycle, which is regulated with the initiation of p16INK4a/Rb and p53/p21 pathways. Idea adapted from [19].
Nutrients 17 02887 g001
Figure 2. This model predicts the effects of Immunoregulatory microbe and GM metabolites, including TMAO, SCFAs, LPS, and BAs, on inflammation and oxidative stress as well as how they affect vascular health.
Figure 2. This model predicts the effects of Immunoregulatory microbe and GM metabolites, including TMAO, SCFAs, LPS, and BAs, on inflammation and oxidative stress as well as how they affect vascular health.
Nutrients 17 02887 g002
Figure 3. This model predicts how natural products regulate vascular aging-related diseases. Some food-based bioactive natural products could achieve direct vascular benefits in their prototype forms, whereas some are metabolized to active substances, which are then absorbed by intestinal epithelial cells and transported into circulating. The bioactive components cause GM alterations in either bacteria composition or metabolites or both, which contribute to gut-aorta axis.
Figure 3. This model predicts how natural products regulate vascular aging-related diseases. Some food-based bioactive natural products could achieve direct vascular benefits in their prototype forms, whereas some are metabolized to active substances, which are then absorbed by intestinal epithelial cells and transported into circulating. The bioactive components cause GM alterations in either bacteria composition or metabolites or both, which contribute to gut-aorta axis.
Nutrients 17 02887 g003
Table 1. Gut-origin microbes act as functional probiotics attenuating vascular aging-related diseases.
Table 1. Gut-origin microbes act as functional probiotics attenuating vascular aging-related diseases.
Category (Phylum)Bacteria StrainsAthero-sclerosisHyper-tensionAAAGM MetabolitesStudy ModelRef.
ActinobacteriaBif. bifidum PRL 2010°°°Bacteria-fed ApoE−/− mice[98]
Bif. breve Bb4°°↓ TMAOCholine chloride-fed C57BL/6J mice[99]
Bif. breve CECT7263°°°Spontaneously hypertensive rats[100]
Bif. lactis HN019°°°Hypertensive women[101]
Bif. pseudocatenulatum G4°°↑ BAsCholesterol-enriched-diet-fed SD rats[102]
Bif. longum BB536°°↑ BAsCholesterol-enriched-diet-fed SD rats[102]
Bif. longum CCFM 1077°°↑ BAsHigh-cholesterol-diet-fed SD rats[103]
Bif. longum BL1 and BL7°°↓ TMAOCholine chloride-fed C57BL/6J mice[103]
FirmicutesLb. plantarum 299v°°Humans with coronary artery disease[104]
Lb. plantarum ZDY04°°↓ TMAO1.3% choline-fed ApoE−/− mice[105]
Lb. plantarum E680°°°High-fat emulsion-fed ICR mice[106]
Lb. plantarum ATCC 8014°°°Propylthiouracil & cholesterol-fed mice[107]
Lb. rhamnosus GG°°°HFD-fed ApoE−/− mice[108]
Lb. rhamnosus GR-1°°°HFD-fed ApoE−/− mice[108]
Lb. fermentum H9°°°HFD-fed SD rats[109]
Lb. fermentum CEC 5716°°°Spontaneously hypertensive rats[110]
Lb. fermentum CEC 5716°°L-NAME-treated Wister rats[111]
Lb. fermentum NCIMB 5221°°°Zucker diabetic fatty rats[112]
Lb. brevis 119-2°°°HFD-fed SD rats[113]
Lb. mucosae DPC 6426°°°HFD-fed ApoE−/− mice[114]
Lb. acidophilus ATCC 4356°°↓ LPSLPS-treated HUVECs[115]
Lb. amylovorus CP1563°°HFD-fed C57BL/6J mice[116]
Lb. casei Shirota°°HFD-fed ApoE−/− mice[117]
Lb. paracasei NTU 101°°Hyperlipidemic hamsters[118]
Lb. johnsonii La1°°°Bacteria-fed Wistar rats[119]
Lb. jensenii ATCC 25258°°°Spontaneously hypertensive rats[120]
Lb. helveticus°°Hypertensive subjects[121]
Lb. reuteri CCFM8631°°°Paigen atherogenic diet-fed mice[122]
Lb. reuteri ADR-3°°°High-fructose-fed rats[123]
E. aerogenes ZDY01°°↓ TMAO1.3% choline-fed BALB/c mice[124]
E. faecium M-74°°°Healthy human subjects[125]
E. faecalis ATCC 19433°°°HFD-fed C57BL/6J mice[126]
P. acidilactici RO37°°°Female ApoE−/− mice[127]
F. prausnitzii°°↓ LPSHFD-fed ApoE−/− mice[128]
BacteroidetesB. vulgatus and B. dorei°°↓ LPSHuman patients and ApoE−/− mice[129]
VerrucomicrobiaA. muciniphila°°↓ LPSHFD-fed ApoE−/− mice[130]
Notes: • indicates Observed Effect; ° indicates Not Applicable; ↑ indicates Increase/Promote; ↓ indicates Decrease/Inhibit. Bif. indicates Bifidobacterium; Lb. indicates Lactobacillu; E. indicates Enterobacter; P. indicates Pediococcus; F. indicates Faecalibacterium; B. indicates Bacteroides; A. indicates Akkermansia; St. indicates Streptococcus. AAA indicates Abdominal Aortic Aneurysm; GM indicates Gut Microbiota; TMAO indicates Trimethylamine-N-oxide; LPS indicates Lipopolysaccharide; BAs indicates bile acids; SD indicates Sprague Dawley; HFD indicates High Fat Diet; Ref. indicates References.
Table 2. Chemical structure and food source of the mentioned natural products.
Table 2. Chemical structure and food source of the mentioned natural products.
CategoryCompoundsChemical StructureFood Source
FlavonoidsQuercetinNutrients 17 02887 i001Apples, berries, onions, capers, citrus fruits, and leafy greens
EGCGNutrients 17 02887 i002Grapes, wine, cocoa, apricots, beans, and green tea
KaempferolNutrients 17 02887 i003Apple skins, kale, spinach, celery, lettuce, and broccoli
PolyphenolsResveratrolNutrients 17 02887 i004Peanuts, grapes, blueberries, raspberries, and mulberries
Chlorogenic acidNutrients 17 02887 i005Coffee beans, apples, pears, blueberries, artichokes, tomatoes, and potatoes
Ellagic acidNutrients 17 02887 i006Berries, grapes, pomegranate, spinach, lettuce, walnuts, and pecans
SDGNutrients 17 02887 i007Flaxseeds, wheat bran, oats, barley, rye, and legumes
Ferulic AcidNutrients 17 02887 i008Wheat, oats, rice, corn, oranges, apple, cherries, tomato, onion, almonds, peanuts, flaxseeds, and coffee beans
PolysaccharidesInulinNutrients 17 02887 i009Chicory root, Jerusalem artichoke, garlic, onions, asparagus, and bananas
Pectin
(Galacturonic acid)
Nutrients 17 02887 i010Citrus fruits, apples, berries, guava, quince, and pears
β-glucan (Cellulose)Nutrients 17 02887 i011Oats, barley, whole grains, mushrooms, and yeast
AlkaloidsCaffeineNutrients 17 02887 i012Coffee, tea, chocolate, and soft drinks
CapsaicinNutrients 17 02887 i013Chili peppers
Phytosterolsβ-sitosterolNutrients 17 02887 i014Plant-based oils, nuts, seeds, and whole grains
StigmasterolNutrients 17 02887 i015Soybeans, pistachios, oats, avocados, spinach
PolyamineSpermidineNutrients 17 02887 i016Wheat germ, soybeans, mushroom, corn, peas, and cheese
Notes: EGCG indicates Epigallocatechin-3-gallate; SDG indicates Secoisolariciresinol diglucoside.
Table 3. The impact of food-based natural products on gut microbiota modulation and vascular aging-associated factors.
Table 3. The impact of food-based natural products on gut microbiota modulation and vascular aging-associated factors.
CategoryCompoundsGM Composition
(↑↓)
GM Metabolites (↑↓)I/OVFA/SMechanism of ActionsRefs.
FlavonoidsQuercetinBifidobacterium
Lactobacillus
Escherichia coli
↑ SCFAs
↑ BAs
↓ TMAO
Induce apoptosis in VSMCs.
Regulate TRAF6-MAPK pathway in EC.
[160,162]
EGCGFirmicutes
Lactobacillus
Desulfovibrionaceae
↑ SCFAs
Suppress MMP-9 and VEGF activation.
Activate Nrf2/Caspase-3 signaling.
[238]
KaempferolFirmicutes
Bacteroidetes
↓ BAs°
Regulate NF-κB signaling pathway.
Inhibit PI3K/Akt/mTOR pathway.
[239]
PolyphenolsResveratrolAkkermansia
Lactobacillus
Bifidobacterium
Enterococcus faecalis
↑ SCFAs
↓ TMAO
Upregulate eNOS activity.
Reduce endothelin-1 synthesis.
Inhibit VSMC proliferation.
[174,240]
Chlorogenic acidBlautia
Sutterella
Dubosiella
Romboutsia
↑ SCFAs
↑ BAs
Improve Nrf2 activation.
Regulate Rap1 and PI3K/AKT pathways.
Regulate Nrf2/HO-1 pathway.
[241]
Ellagic acidLactobacillus
Escherichia coli
Bacteroidetes
Akkermansia
°
Inhibit VSMC proliferation.
Improve endothelial function.
Modulate Ca2+ intake and release.
[242,243]
SDGProteobacteria
Roseburia
Blautia
↑ SCFAs
Inhibit the Akt/IκB/NF-κB pathway.
[244]
PolysaccharidesInulinBifidobacterium
Lactobacillus
Firmicutes
↑ SCFAs
↓ BAs
Activate NO synthase/NO pathway.
[241]
PectinFirmicutes
Eubacterium eligens
↑ SCFAs°°
Reduce blood pressure.
[242,244]
AlkaloidsCaffeineFaecalibacterium
Roseburia
Erysipelatoclostridium
↑ SCFAs
↓ BAs
°
Increase NO production.
[245]
CapsaicinFaecalibacterium
Akkermansia
°
Regulate TRPV1/SIRT1 pathway.
Upregulate SIRT6 pathway.
[224,246]
Phytosterolsβ-sitosterolBifidobacterium
Desulfovibrionaceae
↓ TMAO°
Activate AMPK pathway.
Suppress mTOR pathway.
[233,247]
StigmasterolLactobacillus
Erysipelatoclostridium
°°
Inactivate MAPK signaling.
Induce arrest of cell cycle.
[248]
Notes: • indicates Observed Effect; ° indicates Not applicable; ↑ indicates Increase/Promote; ↓ indicates Decrease/Inhibit. GM indicates Gut Microbiota; I/O indicates Inflammation and Oxidative Stress; VF indicates Vascular Function; A/S indicates Aging/Senescence; TMAO indicates Trimethylamine-N-oxide; LPS indicates Lipopolysaccharide; BAs indicates bile acids; SCFAs indicates short chain fatty acids. Ref. indicates References.
Table 4. The impact of food-based natural products or extracts on cardiovascular disease by modulating gut microbiota.
Table 4. The impact of food-based natural products or extracts on cardiovascular disease by modulating gut microbiota.
CategoryCompoundsAthero-sclerosisHyper-tensionAAAGM Modulation (↑↓)Study Model; DosageRef.
FlavonoidsQuercetin°°Verrocomicrobia
Actinobacteria, Cyanobacteria and Firmicutes
HFD-fed LDLR−/− mice; 10 µg/day Quercetin[249]
EGCG°°Roseburia, Rothia, Parabacteroides and Akkermansia
Bilophila and Alistipes
HFD-fed ApoE−/− mice; 150 mg/kg/day Extract of Peanut Skin containing EGCG[250]
EGCG°°↑ Paraprevotella and Bacteroides
↑ Allobaculum and Bifidobacterium
High salt diet-fed Wistar rats; 500 mg/kg/day Extract of Selenium-enriched and Ordinary Green Tea containing EGCG[251]
Kaempferol°°Bacteroidetes, Verrucomicrobiota, and AkkermansiaceaeHFD-fed ApoE−/− mice; 6.2 mL/kg Dry Red Wine containing Kaempferol[252]
Kaempferol°°Lactobacillus and SCFAs
↓ Ratio of Firmicutes/Bacteroidetes
Clostridiaceae
Spontaneously hypertensive rats; 0.9 g/kg Extracts of Scutellaria baicalensis Georgi and Sophora japonica L. containing Kaempferol[253]
PolyphenolsResveratrol°°Lactobacillus and Bifidobacterium
↓ TMAO and ileal BA
TMAO-induced atherosclerosis in ApoE−/− mice; 0.4% Resveratrol[177]
Resveratrol°°Bacteroidetes
Verrucomicrobia
Firmicutes to Proteobacteria ratio
High-fructose diet-fed SD rats; 50 mg/L Resveratrol[254]
Resveratrol°°Butyricicoccus and Acetic acid
Actinobacteria
0.5% adenine diet-fed SD rats; 10 mg/kg/day Resveratrol[255]
Chlorogenic acid°°Mogibacteriaceae, Coprococcus, Dorea, Ruminococcus, Firmicutes, and DesulfovibrioHFD-fed ApoE−/− mice; Extract of Green Coffee Bean, equivalent to 220 mg/kg of Chlorogenic acid[256]
Chlorogenic acid°°Lachnospiraceae, and OscillospiraHFD-fed Wistar rats; 2 g Chlorogenic acid per kg of food[257]
Ellagic acid°°Lactococcus
Bifidobacterium
Adult metabolic syndrome patients under secondary pharmacological prevention and without previous CVD events; 900 mg/day Pomegranate Extract containing Ellagic acid[258]
Ferulic acid°°Fimicutes, Erysipelotrichaceae, and IleibacteriumHFD-fed ApoE−/− mice; 40 mg/kg/day Ferulic Acid[197]
PolysaccharidesInulin°°↑ Ratio of propionate to acetateHigh cholesterol diet-fed ApoE−/− mice; 10% inulin of diet weight[259]
Pectin°°↑ SCFAsHigh-fat/cholesterol diet-fed ApoE−/− mice; 20% Pectin of diet weight[260]
Pectin°°Bacteroides/Prevotella
Lactobacillus, and Bifidobacterium
Clostridium coccoides
High sucrose diet-fed Wistar rats; 10% High-Esterified Pectin supplementation in diet[208]
β-glucan°°Eisenbergiella and Romboutsia ↑ SCFAs
↓ LPS
High-fat/high-cholesterol diet-fed LDLR−/− mice; 0.8% Oat fiber containing 22% β-glucan[261]
β-glucan°°Bacteroides
Firmicutes
Mildly hypercholesterolemic human subjects; 3 g/day High Molecular Weight Barley β-glucan[214]
AlkaloidsCaffeine°°↓ Ratio of Firmicutes to BacteroidetesHigh-carbohydrate/high-fat diet-fed Wistar rats; 5% Spent Coffee Grounds containing Caffeine[262]
Capsaicin°°Turicibacter, Odoribacter, and Ileibacterium
↓ Deoxycholic acid, cholic acid, hypoxanthine, and stercobilin
HFD-fed ApoE−/− mice; 0.01% Capsaicin of diet weight[263]
Phytosterolsβ-sitosterol°°↓ TMAO
Actinobacteriota, Bacteroidota, Desulfobacterota, and Firmicute
Proteobacteria, Verrucomicrobiota
High-choline diet-fed ApoE−/− mice; 400 mg/kg/d β-sitosterol[264]
PolyamineSpermidine°°Parabacteroides
Prevotella, Desulfovibrionaceae, Campylobacterales, and Helicobacter
Porcine Pancreatic Elastase-induced AAA model in C57BL/6 mice; 3 mM Spermidine via drinking water[265]
Notes: • indicates Observed Effect; ° indicates Not applicable; ↑ indicates Increase/Promote; ↓ indicates Decrease/Inhibit. EGCG indicates Epigallocatechin-3-gallate; AAA indicates Abdominal Aortic Aneurysm; GM indicates Gut Microbiota; TMAO indicates Trimethylamine-N-oxide; LPS indicates Lipopolysaccharide; BAs indicates bile acids; SD indicates Sprague Dawley; HFD indicates High Fat Diet; Ref. indicates References.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Li, J.; Wang, Y.; Shrestha, S.; Gewirtz, A.T.; Ding, Y.; Zou, J. Targeting Gut Microbiota to Combat Vascular Aging and Cardiovascular Disease: Mechanisms and Therapeutic Potential. Nutrients 2025, 17, 2887. https://doi.org/10.3390/nu17172887

AMA Style

Li J, Wang Y, Shrestha S, Gewirtz AT, Ding Y, Zou J. Targeting Gut Microbiota to Combat Vascular Aging and Cardiovascular Disease: Mechanisms and Therapeutic Potential. Nutrients. 2025; 17(17):2887. https://doi.org/10.3390/nu17172887

Chicago/Turabian Style

Li, Jian, Yadong Wang, Sanjiv Shrestha, Andrew T. Gewirtz, Ye Ding, and Jun Zou. 2025. "Targeting Gut Microbiota to Combat Vascular Aging and Cardiovascular Disease: Mechanisms and Therapeutic Potential" Nutrients 17, no. 17: 2887. https://doi.org/10.3390/nu17172887

APA Style

Li, J., Wang, Y., Shrestha, S., Gewirtz, A. T., Ding, Y., & Zou, J. (2025). Targeting Gut Microbiota to Combat Vascular Aging and Cardiovascular Disease: Mechanisms and Therapeutic Potential. Nutrients, 17(17), 2887. https://doi.org/10.3390/nu17172887

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop