Next Article in Journal
Phytopythium vexans Associated with Apple and Pear Decline in the Saïss Plain of Morocco
Next Article in Special Issue
Ligilactobacillus salivarius PS2 Supplementation during Pregnancy and Lactation Prevents Mastitis: A Randomised Controlled Trial
Previous Article in Journal
In Vivo Biofilm Formation of Pathogenic Leptospira spp. in the Vitreous Humor of Horses with Recurrent Uveitis
Previous Article in Special Issue
Cudrania tricuspidata Combined with Lacticaseibacillus rhamnosus Modulate Gut Microbiota and Alleviate Obesity-Associated Metabolic Parameters in Obese Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiome, Functional Food, Atherosclerosis, and Vascular Calcifications—Is There a Missing Link?

by
Dragos Cretoiu
1,2,†,
Ruxandra Florentina Ionescu
3,†,
Robert Mihai Enache
4,†,
Sanda Maria Cretoiu
1,* and
Silviu Cristian Voinea
5
1
Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
2
Alessandrescu-Rusescu National Institute for Mother and Child Health, Fetal Medicine Excellence Research Center, 020395 Bucharest, Romania
3
Central Military Emergency Hospital “Dr. Carol Davila”, Department of Cardiology I, 134 Plevnei Blvd, 010825 Bucharest, Romania
4
Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 258 Fundeni Road, 022328 Bucharest, Romania
5
Department of Surgical Oncology, Prof. Dr. Alexandru Trestioreanu Oncology Institute, Carol Davila University of Medicine and Pharmacy, 252 Fundeni Road, 022328 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2021, 9(9), 1913; https://doi.org/10.3390/microorganisms9091913
Submission received: 20 July 2021 / Revised: 21 August 2021 / Accepted: 7 September 2021 / Published: 9 September 2021
(This article belongs to the Special Issue Dialogue of Probiotics with the Host)

Abstract

:
The gut microbiome is represented by the genome of all microorganisms (symbiotic, potential pathogens, or pathogens) residing in the intestine. These ecological communities are involved in almost all metabolic diseases and cardiovascular diseases are not excluded. Atherosclerosis, with a continuously increasing incidence in recent years, is the leading cause of coronary heart disease and stroke by plaque rupture and intraplaque hemorrhage. Vascular calcification, a process very much alike with osteogenesis, is considered to be a marker of advanced atherosclerosis. New evidence, suggesting the role of dietary intake influence on the diversity of the gut microbiome in the development of vascular calcifications, is highly debated. Gut microbiota can metabolize choline, phosphatidylcholine, and L-carnitine and produce vasculotoxic metabolites, such as trimethylamine-N-oxide (TMAO), a proatherogenic metabolite. This review article aims to discuss the latest research about how probiotics and the correction of diet is impacting the gut microbiota and its metabolites in the atherosclerotic process and vascular calcification. Further studies could create the premises for interventions in the microbiome as future primary tools in the prevention of atherosclerotic plaque and vascular calcifications.

1. Introduction

The human body is populated by a multitude of microorganisms, ranging from bacteria and viruses to fungi. The most abundant and complex community of microbes is found in the gastrointestinal system, known as the gut microbiome [1]. There are five phyla that dominate the gut microbiome: Firmicutes, Bacteroidetes, Actinobacteria, Proteobacteria, and Verrucomicrobia [2,3]. Physiological functions of the gut microbiome are fermentation [4], vitamin synthesis [5], energy production through short-chain fatty acids (SCFAs) [6], and adjustment of the intestinal mucosal barrier [2,7]. SCFAs are directly responsible for gut–brain signaling, while bile acids (BA) and trimethylamine-N-oxide (TMAO) are associated with the atherosclerotic process [2,8,9].
Atherosclerosis, a disease affecting the arteries, is the leading cause of death by heart disease and stroke (~50% of all deaths in Western societies), while cardiovascular diseases (CVDs) represent 32% of all global deaths in 2019 [10]. Atherosclerosis can be regarded not only as an unavoidable degenerative result of aging, but also as a chronic inflammatory disorder, with possible acute complications, such as plaque rupture and thrombosis [11]. Both genetic and environmental factors can influence this condition. Atherosclerosis (and intimal calcifications) and arteriosclerosis (and medial calcifications) are intertwined, precipitating vascular impairment [12]. The evolution of atherosclerosis and its consequences are illustrated in Figure 1. The initial lesions of atherosclerosis are represented by subendothelial accumulations of macrophages containing cholesterol (foam cells). Fatty band deposits are the precursors of fibrous lesions, which contain necrotic debris rich in lipids and smooth muscle cells (SMCs). The fibrous cap of the plaque is represented by SMCs and extracellular matrix, which encapsulates a necrotic core. Plaques can evolve through calcification, ulceration, and hemorrhagic processes. Critical outcomes of plaque evolution can consist of acute occlusion of the artery through thrombus or clot formation, leading to stroke or myocardial infarction, especially if located in the coronary arteries [13]. Thrombosis of the plaque can be linked to erosion or breach of the lesion [11]. Animal studies revealed that deficiency of apolipoprotein E (apoE) or low-density lipoprotein (LDL) receptors can lead to advanced atherosclerotic lesions [14].
“Monckeberg sclerosis” is defined as the calcification of the medial layer of arteries, in the absence of atheroma, being more frequently located in the lower limb arteries, but also in the aorta. The calcification process affecting elastic or muscular arteries is ectopic and is usually observed in patients with chronic inflammatory conditions such as diabetes mellitus, chronic kidney disease (CKD), and hypertension. There can also be a genetic component involved [12].
This review analyzes the main findings related to gut microbiota involvement in the development of CVD, with an emphasis on the direct relation with the calcification of the atherosclerotic plaques. The roles of diverse macro- and micronutrients are also discussed, as well as the most recent findings related to the use of different probiotics as future therapies for CVD.

2. General Mechanisms of Vascular Calcification

Vascular calcifications occur frequently in the large and medium arteries, affected by atherosclerosis, either in the intima (focal plaques) or in the media. Calcifications can also occur in arteries without atherosclerosis, e.g., calcification of internal elastic lamina in patients infected with HIV [15]. These types of arterial calcifications not only vary morphologically and epidemiologically, but they also have different clinical significance [12]. Features that can favor vascular calcifications can be aging, male sex, hypertension, diabetes mellitus, CKD, dyslipidemia, and smoking. In addition, inflammation, oxidative stress, shear stress, advanced glycation end-products, elevated calcium-phosphate product, high angiotensin II concentrations, uremic toxins, and vitamin K deficiency (or antagonists) have significant effects on the vascular system [16].
Calcium deposits at intimal levels, diffuse or punctiforme, in the presence of atherosclerosis, are frequently found in coronary arteries and aorta. They are associated with the accumulation of lipids and macrophages [12,17]. Lipid accumulation and atherosclerotic plaque development can lead to the calcification of the intima, on the grounds of the inflammatory response. Both intimal and medial layer calcifications lead to increased vascular stiffness, diminishing the capacity to adapt to variations in blood pressure and therefore promoting injury and advancement of atherosclerotic plaques [12]. Considered for a long time to be a passive process, vascular calcification was demonstrated to be an actively regulated phenomenon, which resembles bone development. Several bone-regulating proteins, such as osteopontin, matrix Gla protein, osteocalcin, osteonectin, collagen I and II, alkaline phosphatase, bone sialoprotein, and bone morphogenic proteins are expressed at the level of human vascular smooth muscle cells (VSMCs) [17]. Several substances that can either favor or inhibit vascular calcifications in CKD are depicted in Table 1. Vascular calcifications can be considered predictive indicators for CVDs and are associated with a poor prognosis, including higher risk of mortality by adverse cardiovascular events (stroke, acute myocardial infarction, peripheral vascular diseases).

3. Particularities of Gut Microbiota in Atherosclerosis

Three different enterotypes were identified depending on the dominant genera: enterotype 1 dominated by Bacteroides, enterotype 2 dominated by Prevotella, and enterotype 3 dominated by Ruminococcus [26]. Bacteroides dominance was associated with the habits of eating animal protein, amino acids, and saturated fats [27]. Prevotella was found to be in lower levels in these patients, but in high concentrations in patients with diets rich in carbohydrates and simple sugars [27]. Individuals with diets low in fat and animal protein, but high in fiber, starch, and plant polysaccharides represent another category presenting abundant Prevotella [28]. Animal studies pointed out that diets high in unsaturated fats increase Actinobacteria (Bifidobacterium), lactic acid bacteria (Lactobacillus and Streptococcus), and Verrucomicrobia (Akkermansia muciniphila) [29,30], but also amplify the translocation of lipopolysaccharides [31]. Fruits and vegetables, naturally abundant in polyphenols [32], alongside probiotics, can raise levels of Bifidobacterium and bacteria that produce lactic acid and diminish the populations of enteropathogenic bacteria [30].
The adult microbiome is attracting more and more interest because there seems to be a link between the degree of dysbiosis and various pathologies, including cardiovascular diseases [33].
Symptomatic atherosclerotic patients seem to display numerous populations of Collinsella compared to healthy people [2,34]. Some studies concluded that the gut microbiome of patients with atherosclerosis had an abundance of Streptococcus spp. and 4 nterobacteriaceae (including Escherichia coli, Enterobacter aerogenes, and Klebsiella spp.). As a result, metabolism and molecule transport, including TMAO, are affected. Furthermore, the gut microbiome of patients with CVDs is more proinflammatory [2,35]. Studies showed the bacterial phylotypes found in the atherosclerotic plaque were like the ones in the oral cavity, especially in patients with periodontal disease. Moreover, dysbiosis is correlated with plasma cholesterol levels [36].
The main phyla and genera present in the oral cavity, gut, and atherosclerotic plaque can be observed in Figure 2. Patients suffering from coronary artery disease have dysbiosis, displaying increased levels of Lactobacillales order (Firmicutes phylum—Lactobacillus, Streptococcus, and Enterococcus genera) alongside a decrease in Bacteroidetes phylum (Bacteroides and Prevotella genera) [37]. Several studies found in this type of patient low concentrations of Faecalibacterium (possessing an anti-inflammatory role [38,39]), Subdoligranulum, Roseburia (involved in SCFA production [40,41]), Eubacterium rectale, and numerous populations of Escherichia/Shigella and Enterococcus [42]. Elevated bacterial diversity was described in patients with coronary artery disease. Staphylococcus species, Proteus vulgaris, Klebsiella pneumoniae, and Streptococcus spp. were identified in atheromatous plaques. Nevertheless, a definitive causal role between the gut microbiota composition and atherosclerosis was not clearly established. It can be affirmed, though, that the presence of certain bacteria may be involved with atherosclerosis evolution [43].

4. The Role of Inflammation and Dysbiosis in the Atherosclerotic Process

The human gut microbiota varies throughout life and with dietary changing patterns. Dysbiosis can lead to inflammatory or immune disorders by activating proinflammatory responses throughout the body. Gut dysbiosis is linked to dyslipidemia. Atherosclerosis is represented by a process of chronic inflammation, alongside lipid accumulation in vascular walls. The gut microbiome dysbiosis can accentuate low-grade inflammation at intestinal level, favoring the passage of bacterial and metabolism products into the bloodstream, perpetuating chronic inflammation [44].
There is evidence pointing out that cells of the immune system, from carotid or coronary atherosclerotic plaques in patients with acute coronary syndromes, can generate antibodies against gut microbial antigens, such as Klebsiella and Proteus [45]. Furthermore, inflammation is indicated to play an important role in the development of vascular calcifications [2,46]. Moreover, the composition of atherosclerotic plaques is like the gut or oral cavity microbial load, as previously discussed here [36,47].
Diet patterns can influence the composition of gut microbiota, which in turn can influence the general health of the individual by digestion and absorption of nutrients [48]. This way, the dietary pattern could represent one of the most uncomplicated and straightforward methods to change the human microbiome [27]. Representative of the Western diet is the high protein and fat intake, elevating the cardiovascular risk and diminishing Bifidobacterium populations [49]. In comparison, the Mediterranean diet is abundant in vegetables, grains, fruits, antioxidants, and fibers [50]. This diet can lower oxidative stress and inflammation, and accentuate antioxidant effects and nitric oxide bioavailability, thus ameliorating vascular and cardiac function. It can also be associated with a lower incidence and severity of heart failure [51].

5. The Microbiome, Dietary Nutrients, and Vasculotoxic Metabolites

The link between the intestinal microbiota and atherosclerosis has underlined the importance of dietary changes as a new method of vascular prevention [52]. In the past, cholesterol and saturated fats were at the forefront of nutritional management, but now it is established that lowering the intake of L-carnitine from red meat and phosphatidylcholine from egg yolks can also play a key role in the prevention of CVDs, including atherosclerosis (Figure 3) [52]. If so far, drugs such as statins were used as a first intention in lowering fasting LDL levels, studies have pointed out that after a high-cholesterol meal, arterial inflammation, oxidative stress [53,54], and endothelial dysfunction [55] increase for several hours [56].
The microbiota is strongly connected to our health by modulating, among others, the immune function, intestinal function, and bioactivation of nutrients and vitamins. Recently, studies have shown that intestinal microbiota produce vasculotoxic metabolites from dietary components [57]. The most studied pathway that links vasculotoxic metabolites of the microbiota to CVDs, including atherosclerosis, is the conversion of carnitine (found mainly in red meat) and phosphatidylcholine (from egg yolk) into trimethylamine (TMA) by intestinal microorganisms (Figure 3) [58,59].
Phosphatidylcholine, under the action of intestinal lipases, produces a variety of metabolic products, including the choline-containing nutrients glycerophosphocholine, phosphocholine, and choline. Further, these are metabolized by the gut microbiota, mainly from cecum and large bowel to TMA, which is oxidized by hepatic flavin monooxygenases (FMOs) to trimethylamine N-oxide (TMAO). TMAO, a proatherogenic compound, can stimulate macrophage scavenger receptors, increasing the amount of cholesterol in macrophages, the accretion of foam cells in artery walls, and, therefore, atherosclerosis [59]. Additionally, choline can be oxidized in the liver and kidneys to betaine, which represents another way for bacteria to form TMA [60].
L-carnitine consumption has increased in recent years in developed countries and represents a major risk factor for CVDs. It contains a TMA structure, like that of choline, that can activate the same pathogenic pathway [59].
As an assumption, other dietary components that have a TMA structure can also produce TMAO from gut microbiota and lead to atherosclerosis [60]. Consequently, it is important for future studies to raise awareness about this hypothesis and verify its veracity.
Vasculotoxic metabolites are filtered by the kidney and excreted into urine, so this must be considered in the case of patients with renal failure, as they must limit red meat and egg yolk ingestion. Otherwise, levels of TMAO increase, leading to a severe decline in glomerular filtration and a higher cardiovascular risk [61]. The intestinal microbiota also generates other metabolic products that can increase cardiovascular risk in these patients, such as indoxyl sulfate, indole-3-acetic acid, and p-cresyl sulfate [60,62].
Uremic toxins, represented by indoxyl sulfate [63] and indole 3-acetic acid [60], metabolized from tryptophan ingestion and serum p-cresyl sulfate [64], respectively, are accumulated in case of renal failure. They increase the concentration of reactive oxygen species (ROS) in endothelial cells, contributing to endothelial dysfunction, resulting in aortic calcification, vascular stiffness, and higher cardiovascular mortality [65].
An interesting aspect is that vegans lack the intestinal bacteria that produce trimethylamine (TMA), so the consumption of L-carnitine among vegans does not lead to an increased amount of TMAO [59].
Studies have underlined the hypothesis that the intestinal microbiota is adjustable [66] and one can speculate that stool transplantation, already used in the treatment of Clostridium difficile infections, could be useful in the prevention of atherosclerosis. However, evidence is conflicting regarding the subject. For instance, the study of Smits et al. pointed out that levels of TMAO and vascular inflammation markers are not influenced by fecal microbiota transplantation in patients with metabolic syndrome [67]. The concept of fecal transplantation and its other possible beneficial effects are illustrated in Figure 4.

6. The Contribution of Dietary Factors to Vascular Calcification

Cardiovascular calcifications are a predictor of cardiovascular events and mortality. Since there is no accurate treatment for cardiovascular calcifications, studies regarding the relationship between diet and the vascular calcification process were also considered over time. Some of the results are summarized in Table 2.
Establishing connections between the effects of certain foods and the development of atherosclerosis is a difficult process, due to the impossibility of forming batches to be subjected to the same living conditions and appropriate controls. The atherosclerotic process is influenced by numerous factors, including circulating lipid levels and the co-existence of chronic inflammation. Among other interventions in the management of atherosclerosis one can also include the functional foods, a term coined in Japan in the early 1980s. Functional foods are generally defined as edibles with physiological advantages, and which are responsible for short- or long-term benefits, other than their nutritional values [94]. Moreover, other terms were introduced, such as nutraceuticals or bioceuticals, known as a food or part of a food, that can sometimes be seen as dietary supplements providing health benefits (prevention and even treatment of a specific disease) [95,96]. In CVDs, the need for some nutrients may increase. Several functional foods were demonstrated to have beneficial effects in CVDs by lowering the total lipid concentration (and LDL cholesterol), through their antioxidative properties and by their effect on homocysteine levels (for extensive reviews see [97,98]).
Tomatoes, very rich in lycopene with antioxidant properties, are considered very useful in the maintenance of endothelial function and normal levels of blood glucose and lipids [99]. Cranberries, rich in polyphenols, isoprenoids, and other components, were shown to contribute to an increased resistance of LDL cholesterol to oxidation, inhibit platelet aggregation, and reduce blood pressure [100,101]. Relevant findings indicate the benefits of a diet rich in oily fish, long-chain omega 3 fats, and vegetables, while the avoidance of trans fats and simple sugars might also be protective [68].
Soluble fibers such as pectins from apples, pears, and potatoes are important because they can bind to cholesterol in the gastrointestinal tract and increase lipid excretion in feces, while β-glucans, found in oats and barley, are associated with a lower incidence of dyslipidemia, hypertension, and obesity [102,103]. Moreover, fibers from flaxseed and psyllium were proved to lower LDL cholesterol [104].
Polyphenols are nutrients that can be found in tea, coffee, wine, fruits, vegetables, cocoa, and mushrooms [105]. They can inhibit oxidases, decrease the production of superoxide and oxidized low-density lipoprotein, suppress VSMCs proliferation and migration, and diminish platelet aggregation, this way lowering the production of reactive oxygen species [105].
Polyphenols can be listed as flavonoids (the most abundant category), phenolic acids, stilbenes, and lignans [105,106]. They can restrain the evolution of arterial hypertension, diabetes mellitus, hyperlipidemia, and obesity [105].
Data from the literature suggest that polyphenols possess anti-inflammatory and antioxidant properties, being able to attenuate the atherosclerotic process in certain conditions [107]. The study of Loke and colleagues indicated that quercetin and theaflavin may reduce the formation of atherosclerotic lesions in Apo E -/- mice [107]. Other studies reveal the property of polyphenol-rich beverages to attenuate the atherosclerotic process in Apo E -/- mice, such as red wine, dealcoholized red wine [108,109], and tea [107,110].
Other micronutrients can display protective roles against cardiovascular calcifications: magnesium (≥380 mg/day) [68,75,76,77,78], phylloquinone and menaquinone (500 μg/day) [68,79,80,82,83], and 25(OH) vitamin D ≥ 75 nmol/L [68]. On the contrary, inorganic phosphorus from food preservatives may trigger the process of calcification [84,85,86]. In CKD patients, VSMCs transdifferentiation can be amplified by hyperphosphatemia, leading to the formation of an osteoid matrix containing calcium at the level of vascular media [111]. A plasmatic homocysteine level of >12 µmol/L could predict cardiovascular calcifications, with accelerated atherosclerotic plaque progression [70]. Even though oxidative stress can initiate vascular calcifications, antioxidant vitamins did not prove their efficacy, α-tocopherol even being able to elevate the risk for calcification [87]. However, other antioxidant substances, such as quercetin [90], resveratrol (red wine) [91], and epigallocatechin gallate (green tea) [68], were protective against the calcification process [68].
A cross-sectional study conducted by Machado et al. indicated a positive association between the dietary intake of calcium, phosphorus, and potassium and coronary artery calcification in patients suffering from CKD [112].
The relationship between calcium intake and cardiovascular disease is highly debated [113,114,115]. Evidence from clinical trials suggested that there could be an association between calcium dietary supplementation and a high risk for cardiovascular events, including myocardial infarction [116,117,118,119]. Calcium could influence the pathogenesis of cardiovascular events by acting on several pathways: lipid metabolism, inflammation, insulin sensitivity and secretion, thrombosis, and vascular calcification [119,120]. Moreover, a daily dose of more than 1400 mg of calcium supplement was associated with increased death rates from all causes, including from CVDs [119,121]. According to the study conducted by Anderson et al. over a 10-year follow-up period, calcium dietary supplementation was independently associated with incident coronary artery calcification, regardless of adjustment for total calcium intake. Nevertheless, a protective link between total calcium intake and incident coronary atherosclerosis, especially among non-supplement users, was shown [119]. Even so, calcium supplementation does not necessarily promote vascular calcifications in patients not suffering from renal disease or hyperparathyroidism [68,71,72,73].
Sun and colleagues conducted studies on ApoE-deficient mice and showed that low dietary potassium favored atherosclerotic vascular calcification and elevated aortic stiffness versus normal potassium-fed mice [88]. On the contrary, high dietary potassium lowered vascular calcification and aortic stiffness. How can that be explained? Diminished potassium concentrations, near the lower limit of the normal range, elevated intracellular calcium, thus activating cAMP response element-binding protein (CREB). CREB can accentuate autophagy and favor VSMCs calcification. VSMCs calcifications were reduced through the inhibition of calcium signals and downscaling of CREB or ATG7, an autophagy regulator (autophagy-related 7 protein). At the level of calcified arteries and aorta of low-potassium-diet-fed mice, high autophagy and CREB signaling were noted [88]. Moreover, a population-based study revealed that elevated dietary potassium levels diminished the proliferation of VSMCs, lowered monocyte adherence to vascular walls, and decelerated the evolution of atherosclerosis [88,89].
Concerning magnesium, in patients with advanced CKD was noted an association between low serum magnesium concentrations, frequent vascular calcifications, and elevated cardiovascular mortality. The results indicate that magnesium could protect VSMCs against calcifications through molecular mechanisms. Oral magnesium supplementation could lead to decreased vascular calcifications in patients suffering from CKD [81].
Regarding vitamin administration, supplements with vitamins A, B, C, D, and E do not seem to possess effects against calcification. Vitamin K1, K2, and magnesium supplementation, alongside aged garlic extract, could possibly slow the progression of calcifications [16].
Vitamin K is an essential cofactor for the activation of some extracellular matrix proteins, including matrix Gla-protein (MGP), which can inhibit vascular calcification. The vitamin K “concept” is represented by structurally related elements, which comprise phylloquinone (vitamin K1), one of the most utilized synthetic nutritional supplements, and menaquinones (vitamin K2) [16,83]. Newer vitamin K supplements were introduced, mostly containing menaquinone 4 (MK-4) and menaquinone 7 (MK-7: the most hydrophobic variant, with good bioavailability and long half-life) [16,83].
The role of vitamin K was also underlined by the study of McCabe and colleagues, which involved rats with adenine-induced chronic renal failure. High dietary vitamin K in rats with CKD decreased the evolution of warfarin-induced calcifications [122]. What is more, vitamin K1 can be transformed into menaquinone-4 [123]; as a result, dietary supplementation can be achieved with vitamin K2 or analogues, instead of high doses of vitamin K1 [16].
Vitamin K antagonists (VKA), among the most recommended oral anticoagulants, influence the regeneration of vitamin K1 and K2, needed for the activation of coagulation factors and matrix Gla protein, which in turn is responsible for the inhibition of arterial calcification. The study conducted by Hasific et al., evaluating the relationship between VKA treatment and the extent of coronary artery calcifications, showed that a longer period of treatment with VKA, but not novel oral anticoagulants, was associated with the risk of a higher category of coronary artery calcification in patients without previous CVDs [124].
Results from the CARDIA study point out that low-carbohydrate diets adopted from young ages are associated with a higher risk of developing coronary artery calcifications, especially when replacements for carbohydrates are animal proteins or fats [69].
Evidence concerning hyperhomocysteinemia, folic acid, and vitamin B12 as indicators for cardiovascular disease and cardiovascular mortality in CKD patients is conflicting. They were suggested to be considered risk factors for the progression of CKD, representing a potential therapeutic target. The pathophysiological chain of events in this condition can result in malnutrition, anorexia, gastroparesis, and diminished intestinal transit, which further reduce serum folic acid and vitamin B12 concentrations [125]. Reduced blood levels of vitamin B12 were associated with elevated body mass index, diabetes mellitus or insulin resistance, dyslipidemia, and CVDs [125,126]. Hyperhomocysteinemia has been noted as a risk factor for CVDs, but the data are still very debated [125,127,128,129]. High homocysteine blood levels, as well as folic acid and vitamin B12 metabolism abnormalities, can be found in patients suffering from CKD [125,130,131]. Results of a meta-analysis summing up to 5123 patients state that hyperhomocysteinemia is a risk factor for cardiovascular disease and mortality for the CKD patients who do not receive folic acid supplementation [125,132]. Hyperhomocysteinemia was also considered a cardiovascular risk factor in several studies [133,134,135,136,137,138,139]. On the other hand, opposite conclusions were drawn by other studies; for example, Suliman et al. [140] and Wrone et al. [141].
Soohoo et al. [142] indicate that low folic acid concentrations are associated with increased all-cause mortality in hemodialysis patients, therefore folic acid supplementation can diminish cardiovascular events, according to Righetti et al. [125,143]. What is to be done regarding diet supplementation? Data from the literature point into different directions concerning this topic. Several studies concluded that folic acid and vitamin B12 supplementation had no impact on all-cause mortality or cardiovascular events in CKD patients [144,145,146], while 5 mg folic acid supplementation in hemodialysis patients reduced cardiovascular events [125,143].

7. Pharmacological Approaches Impacting Microbiome, Atherosclerosis, and Vascular Calcifications

Calcium channel blockers and renin–angiotensin–aldosterone inhibitors proved to be able to decrease vascular calcifications in animal models [16,147,148,149,150,151]. Human studies, however, did not provide an accurate conclusion regarding this phenomenon [152,153,154,155]. Statins, although targeting cardiovascular morbidity and mortality, did not modify the evolution of coronary calcifications [156].
Up to now, there is conflicting evidence regarding possible interventions in the gut microbiome to diminish the intensity of atherosclerosis and vascular calcifications.
Patients with CKD represent a population of high interest. In CKD, as the glomerular filtration rate decreases, the colon takes up a bigger role in the excretion of urea and uric acid. Long exposure to high concentrations of urea can lead to excessive development of bacteria containing urease, generating high ammonia levels and increasing the intestinal pH [157]. Elevated concentrations of uric acid and high oxalate secretion in the digestive tract results in an elevated number of uricase-producing bacteria [158,159].
Correction of gut dysbiosis can improve the gut–vascular–bone axis in CKD patients, diminishing vascular calcification development and bone demineralization. Possible solutions can be diet changes, with high fiber intake, probiotics and prebiotics administration, and vitamin K supplementation, as a risk factor for vascular calcification and bone demineralization can be vitamin K deficiency, a common phenomenon in CKD [159,160]. Evidence suggests lower uremic toxin production because of probiotics administration, alongside lower levels of inflammation biomarkers and oxidative stress [161]. Prebiotics (prebiotic-resistant starch supplementation; oat β-glucan) have been linked to the minimization of uremic toxins and inflammatory markers in hemodialysis patients [162,163].
Several clinical studies presented the positive effect of probiotics [164] on blood lipid profile (Lactobacillus acidophilus, Bifidobacterium lactis, Lactobacillus plantarum, Lactobacillus helveticus, Enterococcus faecium) [165] and underlined their antioxidant effect (Lactobacillus fermentum, Lactobacillus plantarum, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus reuteri, Lactobacillus delbrueckii ssp. bulgaricus, Lactobacillus sporogenes, Lactococcus lactis, Lactobacillus bulgaricus, Lactobacillus gasser, Lactobacillus rhamnosus, Lactobacillus langum, Lactobacillus paracasei, Bifidobacterium lactis, Bifidobacterium bifidum, Bifidobacterium longum, Bifidobacterium BB-12, Bifidobacterium infantis, Bifidobacterium breve) [166], but not the amelioration of endothelial function. A promising new treatment for patients at risk could be, according to Matsumoto M., bacterial metabolites control. Some articles also suggested that oral supplementation with polyamines can be efficient in preventing CVDs [167].
Polyamines, spermidine and spermine for example, are bioactive compounds that can enhance autophagy and diminish inflammation, but also possess anti platelet aggregation properties [167,168]. Spermine can inhibit the inflammatory response through restraining proinflammatory cytokine production [167,169]. It can also alter the transmigration of inflammatory cells to the setting of inflammation by suppressing LFA-1, which binds to endothelial cells through intercellular adhesion molecule 1 [167,170].
Spermidine, according to LaRocca et al., elevates the bioavailability of nitric oxide, diminishing oxidative stress, altering structural factors, and promoting autophagy, thus having a powerful anti-aging influence on arteries [167,171]. Autophagy induced by spermidine can prevent atherosclerosis. Another study pointed out that spermidine supplementation, through raising autophagy in aged and salt-sensitive hypertensive animal models, prolonged the lifespan of mice, provided protective effects on the cardiovascular system, lowered blood pressure, and delayed the progression to heart failure [167,172]. Michiels et al. affirmed that spermidine supplements diminished atherosclerotic plaques formation by provoking autophagy in apolipoprotein E-deficient mice [167,173].
As vascular endothelial dysfunction is a premature symptom of atherosclerosis, intervention in early stages can restore the normal physiology. Matsumoto et al. evaluated the outcomes of putrescine, a precursor of spermidine and spermine, produced by the gut microbiome after administration of Bifidobacterium animalis spp. Lactis LKM512 (Bifal) and arginine (Arg), on endothelial dysfunction [167,174]. The results indicate that the Bifal and Arg group had a lower atherosclerotic risk compared to the placebo group, by upregulating blood spermidine concentrations, which in turn promote autophagy, with improved vascular endothelial function [167].
Studies on vitamin K show promising results. Kawashima and colleagues evaluated the effects of vitamin K2, menatetrenone, on atherosclerosis and coagulation in hypercholesterolemic rabbits. Vitamin K, indispensable for the gamma-carboxylation of glutamic acid (Gla) within proteins in the body, such as vitamin K-dependent clotting factors and bone Gla-protein [175,176], in doses from 1 to 10 mg/kg/day diminished the evolution of atherosclerotic plaques, intima thickening, and pulmonary atherosclerosis, and prevented the coagulation tendency through lowering total cholesterol levels, lipid peroxidation, and factor X plasmatic activity [177].
In hypercholesterolemic rabbits, the plasmatic activity of vitamin K-dependent clotting factors is increased compared to normolipidemic animals [178,179], along with elevated procoagulant activity [177,180]. Pharmacological doses of vitamin K2 reduced the ester-cholesterol deposition at aortic sites in hypercholesterolemic rabbits [177].
Vitamin K2 supplementation for 270 days in patients with stages 3–5 of CKD significantly modified the levels of regulators of calcification, such as desphosphorylated–uncarboxylated matrix Gla protein, osteocalcin, and osteoprotegerin, diminishing the progression of atherosclerosis, but did not significantly influence the progression of calcification [181].
Osteocalcin, matrix Gla protein, and Gla-rich protein are inhibitors of calcifications of the soft tissue and require vitamin K-dependent carboxylation for activation. Circulating desphosphorylated–uncarboxylated matrix Gla protein was shown to be predictive for cardiovascular risk and mortality and the prevalence of arterial calcifications was associated with circulating total uncarboxylated matrix Gla. Over a 3-year period of supplementation of vitamin K, vascular elasticity was observed, compared to a 12% loss in the placebo group [182].
A Gla protein found in atherosclerotic plaques possesses rather controversial roles; it is linked to atherosclerotic calcification on one hand, according to Levy et al. [177,183,184], and on the other hand it can limit calcium deposits in vitro, according to Gijsbers et al. [177,185].
It is known that the gut metabolite from trimethylamine (TMA), trimethylamine-N-oxide (TMAO), is an independent risk factor for atherosclerosis. A hypothesis regarding the potential capacity of berberine to modify gut microbiota and treat diabetes, obesity, and atherosclerosis was raised, based on the inhibition of TMA/TMAO production by gut microbiota mediated by berberine. According to the work of Li Xingxing et al., berberine proved to attenuate TMA/TMAO production, so it diminished atherosclerotic lesions in animal subjects fed with cereal-based diets, supplemented with choline [186].
There is an indisputable role of diet in balancing the composition and activity of the gut microbiota. The gut microbiota and its complex composition display numerous roles, including immunomodulation. Studies on Akkermansia muciniphila, a mucin-degrading bacterium, were conducted due to its numerous beneficial properties on human health. Berberine intake was indicated to elevate intestinal Akkermansia levels and reduce atherosclerosis in ApoE -/- mice on high-fat diets. Mice fed with berberine had diminished high-fat-diet-induced atherosclerotic lesions and elevated concentrations of Akkermansia spp. Berberine proved to mitigate the expression of proinflammatory cytokines, reduce endotoxemia and inflammation promoted by high fat diets, and improve the gut barrier function by promoting expression of tight junction proteins. Thus, berberine induced gut changes, accentuating Akkermansia spp. abundance, highlighting the antiatherosclerotic effects of berberine. Similar effects were shown for resveratrol, which, by changing the gut microbiota composition, decreases TMAO-mediated atherosclerosis in ApoE -/- mice [187]. Resveratrol can restrict TMAO-mediated atherosclerosis through lowering TMAO levels and elevating bile acid secretion [187,188].
One class of microbial metabolites, known as SCFAs, represented mainly by acetate, propionate, and butyrate, which are most abundant in the human gut, were shown to function like an extra energy source, leading to de novo lipogenesis [189]. The action of SCFAs is possible after their binding to G-protein-coupled receptors, present on endothelial cells and on SMCs (in tunica media), consisting mainly of regulation of blood pressure and of the vascular tone [190]. The general effects of SCFAs might be attributed, in relation to the atherosclerotic process, to their anti-inflammatory properties by reducing migration and proliferation of immune cells, decreasing the levels of several cytokines (IL-1, IL-6 and TNF-α), and triggering apoptosis [191].
Another class of microbial metabolites is represented by secondary bile acids derived from primary bile acids [192]. Different in vivo and in vitro studies were carried out to draw attention to their involvement in atherosclerosis (for detailed review see [193]). Secondary biliary acids modulate bile salt hydrolase activity, which hydrolyzes glycine and taurine conjugates to liberate free biliary acids. If high quantities are present, it leads to increased cholesterol levels and foam cell formation, and it directly influences the size of atherosclerotic plaque [194]. The phenomenon is possible due to the presence of Gram-positive bacteria, such as Bifidobacterium, Lactobacillus, Clostridium, and Enterococcus, as well as Gram-negative bacteria, such as Bacteroides [195]. Plant-based protein intake, compared to animal-based protein intake, has been associated with decreases in Bacteroides and increases in Bifidobacterium and Lactobacillus concentrations, resulting in health advantages [196]. LDL cholesterol concentrations are decreased in populations consuming whole-grain foods, where Bifidobacterium and Lactobacillus are well represented and contribute to biliary acid deconjugation [197].
Although discussing all functional foods as such is considered beyond the scope of this review, we considered it more useful to synthesize recent studies, either on humans or in laboratory conditions, that establish connections between dietary components and their effects on vascular calcification, due to changes produced in the microbiome and in the microbial metabolites. The results are summarized in Table 3.

8. Conclusions

The very important advances in gut microbiome research recently showed its involvement in different metabolic diseases that are usually followed by the formation of atherosclerotic plaques. Bacterial metabolites, SCFA, TMAO, and biliary acids are directly affected by dietary patterns responsible for different enterotypes. Microbiome involvement in atherosclerotic plaque formation and stability should not be overlooked, since future vascular events are correlated with plaque vulnerability, undoubtedly related to its calcification. Certainly, the role of the microbiome in the prevention or management of atherosclerosis and vascular calcifications is not to be neglected. Numerous studies attest to the favorable or unfavorable effect of diet on the phenomenon of atherosclerosis. However, it is becoming clear that the microbiome is very sensitive to the composition of diets and that the products of bacterial metabolism play an overwhelming role in general health and prevention of CVDs. From now on, numerous detailed studies on the link between the microbiota and functional foods are needed. There is still much to learn regarding the connections between gut microbiome, vascular health, and cardiovascular risk, but novel therapies, including here the manipulation of diet, the use of pre/probiotics, or administration of different supplements, should be taken into consideration. Therefore, one can consider that the connections between microbiome, diet, functional foods, and the administration of pre/probiotics will represent the future means for the prevention and treatment of CVDs in an integrated context.

Author Contributions

Conceptualization, S.C.V., R.F.I., and S.M.C.; methodology, R.F.I.; software, D.C., R.M.E., S.M.C., and S.C.V.; validation, S.M.C., and D.C.; formal analysis, R.M.E., S.C.V., S.M.C., and D.C.; investigation R.F.I., R.M.E., and D.C.; resources, R.M.E., S.C.V., S.M.C., and D.C.; data curation, R.F.I., R.M.E., and S.M.C.; writing—original draft preparation, R.F.I., R.M.E., D.C., and S.M.C.; writing—review and editing, S.M.C., and S.C.V.; visualization, R.F.I., R.M.E., and S.M.C.; supervision, S.C.V., and S.M.C.; project administration, S.M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  2. Ahmad, A.F.; Dwivedi, G.; O’Gara, F.; Caparros-Martin, J.; Ward, N.C. The gut microbiome and cardiovascular disease: Current knowledge and clinical potential. Am. J. Physiol. Heart Circ. Physiol. 2019, 317, H923–H938. [Google Scholar] [CrossRef] [PubMed]
  3. Zabell, A.; Tang, W.H. Targeting the microbiome in heart failure. Curr. Treat. Opt. Cardiovasc. Med. 2017, 19, 27. [Google Scholar] [CrossRef]
  4. Liu, C.; Zhang, J.; Li, M.; Zhao, L.; Ji, C.; Ma, Q. Alterations and structural resilience of the gut microbiota under dietary fat perturbations. J. Nutr. Biochem. 2018, 61, 91–100. [Google Scholar] [CrossRef]
  5. Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [Green Version]
  6. Blacher, E.; Levy, M.; Tatirovsky, E.; Elinav, E. Microbiome-modulated metabolites at the interface of host immunity. J. Immunol. 2017, 198, 572–580. [Google Scholar] [CrossRef] [Green Version]
  7. Kitai, T.; Tang, W.H.W. Gut microbiota in cardiovascular disease and heart failure. Clin. Sci. 2018, 132, 85–91. [Google Scholar] [CrossRef] [PubMed]
  8. Smolinska, A.; Tedjo, D.I.; Blanchet, L.; Bodelier, A.; Pierik, M.J.; Masclee, A.A.M.; Dallinga, J.; Savelkoul, P.H.M.; Jonkers, D.M.A.E.; Penders, J.; et al. Volatile metabolites in breath strongly correlate with gut microbiome in CD patients. Anal. Chim. Acta 2018, 1025, 1–11. [Google Scholar] [CrossRef]
  9. Ding, L.; Chang, M.; Guo, Y.; Zhang, L.; Xue, C.; Yanagita, T.; Zhang, T.; Wang, Y. Trimethylamine-N-oxide (TMAO)-induced atherosclerosis is associated with bile acid metabolism. Lipids Health Dis. 2018, 17, 286. [Google Scholar] [CrossRef] [Green Version]
  10. World Health Organization. Cardiovascular Diseases (CVDs). Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 17 July 2021).
  11. Lusis, A.J. Atherosclerosis. Nature 2000, 407, 233–241. [Google Scholar] [CrossRef]
  12. Mackey, R.H.; Venkitachalam, L.; Sutton-Tyrrell, K. Calcifications, arterial stiffness and atherosclerosis. Adv. Cardiol. 2007, 44, 234–244. [Google Scholar] [CrossRef]
  13. Otsuka, F.; Yasuda, S.; Noguchi, T.; Ishibashi-Ueda, H. Pathology of coronary atherosclerosis and thrombosis. Cardiovasc. Diagn. Ther. 2016, 6, 396–408. [Google Scholar] [CrossRef] [Green Version]
  14. Tamminen, M.; Mottino, G.; Qiao, J.H.; Breslow, J.L.; Frank, J.S. Ultrastructure of early lipid accumulation in apoE-deficient mice. Arterioscler. Thromb. Vasc. Biol. 1999, 19, 847–853. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Micheletti, R.G.; Fishbein, G.A.; Currier, J.S.; Singer, E.J.; Fishbein, M.C. Calcification of the internal elastic lamina of coronary arteries. Mod. Pathol. 2008, 21, 1019–1028. [Google Scholar] [CrossRef]
  16. Vossen, L.M.; Kroon, A.A.; Schurgers, L.J.; de Leeuw, P.W. Pharmacological and Nutritional Modulation of Vascular Calcification. Nutrients 2019, 12, 100. [Google Scholar] [CrossRef] [Green Version]
  17. Proudfoot, D.; Shanahan, C.M. Biology of calcification in vascular cells: Intima versus media. Herz 2001, 26, 245–251. [Google Scholar] [CrossRef]
  18. Bain, M.A.; Faull, R.; Fornasini, G.; Milne, R.W.; Evans, A.M. Accumulation of trimethylamine and trimethylamine-N-oxide in end-stage renal disease patients undergoing haemodialysis. Nephrol. Dial. Transplant. 2006, 21, 1300–1304. [Google Scholar] [CrossRef] [Green Version]
  19. Cockayne, S.; Adamson, J.; Lanham-new, S.; Shearer, M.J.; Gilbody, S.; Torgerson, D.J. Vitamin K and the prevention of fractures. Arch. Intern. Med. 2006, 166, 1256–1261. [Google Scholar] [CrossRef] [PubMed]
  20. Moe, S.M.; Chen, N.X. Inflammation and vascular calcification. Blood Purif. 2005, 23, 64–71. [Google Scholar] [CrossRef] [PubMed]
  21. Vervloet, M.; Cozzolino, M. Vascular calcification in chronic kidney disease: Different bricks in the wall? Kidney Int. 2017, 91, 808–817. [Google Scholar] [CrossRef] [PubMed]
  22. Evenepoel, P.; Opdebeeck, B.; David, K.; D’Haese, P.C. Bone-vascular axis in chronic kidney disease. Adv. Chronic Kidney Dis. 2019, 26, 472–483. [Google Scholar] [CrossRef] [PubMed]
  23. Yamada, S.; Giachelli, C.M. Vascular Calcification in CKD-MBD: Roles for phosphate, FGF23, and Klotho. Bone 2017, 100, 87–93. [Google Scholar] [CrossRef] [PubMed]
  24. Liabeuf, S.; Barreto, D.V.; Barreto, F.C.; Meert, N.; Glorieux, G.; Schepers, E.; Temmar, M.; Choukroun, G.; Vanholder, R.; Massy, Z.A.; et al. Free p-cresylsulphate is a predictor of mortality in patients at different stages of chronic kidney disease. Nephrol. Dial. Transplant. 2010, 25, 1183–1191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lano, G.; Burtey, S.; Sallée, M. Indoxyl sulfate, a uremic endotheliotoxin. Toxins 2020, 12, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180, Erratum in: Nature 2011, 474, 666. Erratum in: Nature 2014, 506, 516. [Google Scholar] [CrossRef]
  27. Wu, G.D.; Chen, J.; Hoffmann, C.; Bittinger, K.; Chen, Y.Y.; Keilbaugh, S.A.; Bewtra, M.; Knights, D.; Walters, W.A.; Knight, R.; et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011, 334, 105–108. [Google Scholar] [CrossRef] [Green Version]
  28. De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Coelho, O.G.L.; Candido, F.G.; Alfenas, R.C.G. Dietary fat and gut microbiota: Mechanisms involved in obesity control. Crit. Rev. Food Sci. Nutr. 2018, 59, 3045–3053. [Google Scholar] [CrossRef]
  30. Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef] [Green Version]
  31. Jamar, G.; Ribeiro, D.A.; Pisani, L.P. High-fat or high-sugar diets as trigger inflammation in the microbiota-gut-brain axis. Crit. Rev. Food Sci. Nutr. 2021, 61, 836–854. [Google Scholar] [CrossRef]
  32. Pérez-Jiménez, J.; Neveu, V.; Vos, F.; Scalbert, A. Identification of the 100 richest dietary sources of polyphenols: An application of the Phenol-Explorer database. Eur. J. Clin. Nutr. 2010, 64 (Suppl. S3), S112–S120. [Google Scholar] [CrossRef]
  33. Ionescu, R.F.; Boroghina, S.C.; Cretoiu, S.M. Is there a link between the gut microbiome and arterial hypertension? J. Hypertens. Res. 2021, 7, 12–17. [Google Scholar]
  34. Karlsson, F.H.; Fåk, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Bäckhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012, 3, 1245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Jie, Z.; Xia, H.; Zhong, S.L.; Feng, Q.; Li, S.; Liang, S.; Zhong, H.; Liu, Z.; Gao, Y.; Zhao, H.; et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat. Commun. 2017, 8, 845. [Google Scholar] [CrossRef] [Green Version]
  36. Koren, O.; Spor, A.; Felin, J.; Fåk, F.; Stombaugh, J.; Tremaroli, V.; Behre, C.J.; Knight, R.; Fagerberg, B.; Ley, R.E.; et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4592–4598. [Google Scholar] [CrossRef] [Green Version]
  37. Emoto, T.; Yamashita, T.; Sasaki, N.; Hirota, Y.; Hayashi, T.; So, A.; Kasahara, K.; Yodoi, K.; Matsumoto, T.; Mizoguchi, T.; et al. Analysis of gut microbiota in coronary artery disease patients: A possible link between gut microbiota and coronary artery disease. J. Atheroscler. Thromb. 2016, 23, 908–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Lopez-Siles, M.; Martinez-Medina, M.; Surís-Valls, R.; Aldeguer, X.; Sabat-Mir, M.; Duncan, S.H.; Flint, H.J.; Garcia-Gil, L.J. Changes in the abundance of Faecalibacterium prausnitzii phylogroups I and II in the intestinal mucosa of inflammatory bowel disease and patients with colorectal cancer. Inflamm. Bowel. Dis. 2016, 22, 28–41. [Google Scholar] [CrossRef] [Green Version]
  39. Machiels, K.; Joossens, M.; Sabino, J.; De Preter, V.; Arijs, I.; Eeckhaut, V.; Ballet, V.; Claes, K.; Van Immerseel, F.; Verbeke, K.; et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014, 63, 1275–1283. [Google Scholar] [CrossRef] [PubMed]
  40. Trøseid, M.; Andersen, G.Ø.; Broch, K.; Hov, J.R. The gut microbiome in coronary artery disease and heart failure: Current knowledge and future directions. EBioMedicine 2020, 52, 102649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhu, Q.; Gao, R.; Zhang, Y.; Pan, D.; Zhu, Y.; Zhang, X.; Yang, R.; Jiang, R.; Xu, Y.; Qin, H. Dysbiosis signatures of gut microbiota in coronary artery disease. Physiol. Genom. 2018, 50, 893–903. [Google Scholar] [CrossRef] [PubMed]
  43. Ott, S.J.; El Mokhtari, N.E.; Musfeldt, M.; Hellmig, S.; Freitag, S.; Rehman, A.; Kühbacher, T.; Nikolaus, S.; Namsolleck, P.; Blaut, M.; et al. Detection of diverse bacterial signatures in atherosclerotic lesions of patients with coronary heart disease. Circulation 2006, 113, 929–937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Komaroff, A.L. The microbiome and risk for atherosclerosis. JAMA 2018, 319, 2381–2382. [Google Scholar] [CrossRef] [PubMed]
  45. Canducci, F.; Saita, D.; Foglieni, C.; Piscopiello, M.R.; Chiesa, R.; Colombo, A.; Cianflone, D.; Maseri, A.; Clementi, M.; Burioni, R. Cross-reacting antibacterial auto-antibodies are produced within coronary atherosclerotic plaques of acute coronary syndrome patients. PLoS ONE 2012, 7, e42283. [Google Scholar] [CrossRef]
  46. Pedicino, D.; Severino, A.; Ucci, S.; Bugli, F.; Flego, D.; Giglio, A.F.; Trotta, F.; Ruggio, A.; Lucci, C.; Iaconelli, A.; et al. Epicardial adipose tissue microbial colonization and inflammasome activation in acute coronary syndrome. Int. J. Cardiol. 2017, 236, 95–99. [Google Scholar] [CrossRef]
  47. Chhibber-Goel, J.; Singhal, V.; Bhowmik, D.; Vivek, R.; Parakh, N.; Bhargava, B.; Sharma, A. Linkages between oral commensal bacteria and atherosclerotic plaques in coronary artery disease patients. NPJ Biofilmss Microb. 2016, 2, 7. [Google Scholar] [CrossRef]
  48. Schiattarella, G.G.; Sannino, A.; Esposito, G.; Perrino, C. Diagnostics and therapeutic implications of gut microbiota alterations in cardiometabolic diseases. Trends Cardiovasc. Med. 2019, 29, 141–147. [Google Scholar] [CrossRef]
  49. Battson, M.L.; Lee, D.M.; Jarrell, D.K.; Hou, S.; Ecton, K.E.; Weir, T.L.; Gentile, C.L. Suppression of gut dysbiosis reverses Western diet-induced vascular dysfunction. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E468–E477. [Google Scholar] [CrossRef] [Green Version]
  50. Eckel, R.H.; Jakicic, J.M.; Ard, J.D.; de Jesus, J.M.; Houston Miller, N.; Hubbard, V.S.; Lee, I.M.; Lichtenstein, A.H.; Loria, C.M.; Millen, B.E.; et al. American College of Cardiology/American Heart Association Task Force on Practice Guidelines. 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2014, 63, 2960–2984, Erratum in 2014, 63, 3027–3028. [Google Scholar] [CrossRef] [Green Version]
  51. Kerley, C.P. Dietary patterns and components to prevent and treat heart failure: A comprehensive review of human studies. Nutr. Res. Rev. 2019, 32, 1–27. [Google Scholar] [CrossRef]
  52. Spence, J.D. Effects of the Intestinal Microbiome on Constituents of Red Meat and Egg Yolks: A New Window Opens on Nutrition and Cardiovascular Disease. Can. J. Cardiol. 2014, 30, 150–151. [Google Scholar] [CrossRef] [PubMed]
  53. Ghanim, H.; Abuaysheh, S.; Sia, C.L.; Korzeniewski, K.; Chaudhuri, A.; Fernandez-Real, J.M.; Dandona, P. Increase in plasma endotoxin concentrations and the expression of Toll-like receptors and suppressor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: Implications for insulin resistance. Diabetes Care 2009, 32, 2281–2287. [Google Scholar] [CrossRef] [Green Version]
  54. Dandona, P.; Ghanim, H.; Chaudhuri, A.; Dhindsa, S.; Kim, S.S. Macronutrient intake induces oxidative and inflammatory stress: Potential relevance to atherosclerosis and insulin resistance. Exp. Mol. Med. 2010, 42, 245–253. [Google Scholar] [CrossRef] [PubMed]
  55. Vogel, R.A.; Corretti, M.C.; Plotnick, G.D. Effect of a single high-fat meal on endothelial function in healthy subjects. Am. J. Cardiol. 1997, 79, 350–354. [Google Scholar] [CrossRef]
  56. Spence, J.D. Fasting lipids: The carrot in the snowman. Can. J. Cardiol. 2003, 19, 890–892. [Google Scholar]
  57. Spence, J.D. Recent advances in pathogenesis, assessment, and treatment of atherosclerosis. F1000Research 2016, 5, F1000 Faculty Rev-1880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; Dugar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.M.; et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011, 472, 57–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Koeth, R.A.; Wang, Z.; Levison, B.S.; Buffa, J.A.; Org, E.; Sheehy, B.T.; Britt, E.B.; Fu, X.; Wu, Y.; Li, L.; et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 2013, 19, 576–585. [Google Scholar] [CrossRef] [Green Version]
  60. Spence, J.D.; Urquhart, B.L.; Bang, H. Effect of renal impairment on atherosclerosis: Only partially mediated by homocysteine. Nephrol. Dial. Transplant. 2016, 31, 937–944. [Google Scholar] [CrossRef] [Green Version]
  61. Tang, W.H.; Wang, Z.; Kennedy, D.J.; Wu, Y.; Buffa, J.A.; Agatisa-Boyle, B.; Li, X.S.; Levison, B.S.; Hazen, S.L. Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease. Circ. Res. 2015, 116, 448–455. [Google Scholar] [CrossRef] [Green Version]
  62. Poesen, R.; Claes, K.; Evenepoel, P.; de Loor, H.; Augustijns, P.; Kuypers, D.; Meijers, B. Microbiota-Derived Phenylacetylglutamine Associates with Overall Mortality and Cardiovascular Disease in Patients with CKD. J. Am. Soc. Nephrol. 2016, 27, 3479–3487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Poesen, R.; Viaene, L.; Verbeke, K.; Claes, K.; Bammens, B.; Sprangers, B.; Naesens, M.; Vanrenterghem, Y.; Kuypers, D.; Evenepoel, P.; et al. Renal clearance and intestinal generation of p-cresyl sulfate and indoxyl sulfate in CKD. Clin. J. Am. Soc. Nephrol. 2013, 8, 1508–1514. [Google Scholar] [CrossRef] [Green Version]
  64. Wu, I.W.; Hsu, K.H.; Hsu, H.J.; Lee, C.C.; Sun, C.Y.; Tsai, C.J.; Wu, M.S. Serum free p-cresyl sulfate levels predict cardiovascular and all-cause mortality in elderly hemodialysis patients—A prospective cohort study. Nephrol. Dial. Transplant. 2012, 27, 1169–1175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Dou, L.; Sallée, M.; Cerini, C.; Poitevin, S.; Gondouin, B.; Jourde-Chiche, N.; Fallague, K.; Brunet, P.; Calaf, R.; Dussol, B.; et al. The cardiovascular effect of the uremic solute indole-3 acetic Acid. J. Am. Soc. Nephrol. 2015, 26, 876–887. [Google Scholar] [CrossRef]
  66. Petrof, E.O.; Gloor, G.B.; Vanner, S.J.; Weese, S.J.; Carter, D.; Daigneault, M.C.; Brown, E.M.; Schroeter, K.; Allen-Vercoe, E. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: ‘RePOOPulating’ the gut. Microbiome 2013, 1, 3. [Google Scholar] [CrossRef] [Green Version]
  67. Smits, L.P.; Kootte, R.S.; Levin, E.; Prodan, A.; Fuentes, S.; Zoetendal, E.G.; Wang, Z.; Levison, B.S.; Cleophas, M.C.P.; Kemper, E.M.; et al. Effect of Vegan Fecal Microbiota Transplantation on Carnitine- and Choline-Derived Trimethylamine-N-Oxide Production and Vascular Inflammation in Patients With Metabolic Syndrome. J. Am. Heart Assoc. 2018, 7, e008342. [Google Scholar] [CrossRef]
  68. Nicoll, R.; Howard, J.M.; Henein, M.Y. A review of the effect of diet on cardiovascular calcification. Int. J. Mol. Sci. 2015, 16, 8861–8883. [Google Scholar] [CrossRef] [Green Version]
  69. Gao, J.W.; Hao, Q.Y.; Zhang, H.F.; Li, X.Z.; Yuan, Z.M.; Guo, Y.; Wang, J.F.; Zhang, S.L.; Liu, P.M. Low-Carbohydrate Diet Score and Coronary Artery Calcium Progression: Results from the CARDIA Study. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 491–500. [Google Scholar] [CrossRef]
  70. Rasouli, M.L.; Nasir, K.; Blumenthal, R.S.; Park, R.; Aziz, D.C.; Budoff, M.J. Plasma homocysteine predicts progression of atherosclerosis. Atherosclerosis 2005, 181, 159–165. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, T.K.; Bolland, M.J.; van Pelt, N.C.; Horne, A.M.; Mason, B.H.; Ames, R.W.; Grey, A.B.; Ruygrok, P.N.; Gamble, G.D.; Reid, I.R. Relationships between vascular calcification, calcium metabolism, bone density, and fractures. J. Bone Miner. Res. 2010, 25, 2777–2785. [Google Scholar] [CrossRef] [PubMed]
  72. Agata, U.; Park, J.H.; Hattori, S.; Iimura, Y.; Ezawa, I.; Akimoto, T.; Omi, N. The effect of different amounts of calcium intake on bone metabolism and arterial calcification in ovariectomised rats. J. Nutr. Sci. Vitaminol. 2013, 59, 29–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Hsu, H.H.; Culley, N.C. Effects of dietary calcium on atherosclerosis, aortic calcification, and icterus in rabbits fed a supplemental cholesterol diet. Lipids Health Dis. 2006, 5, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Moe, S.M.; Chen, N.X.; Newman, C.L.; Gattone, V.H., 2nd; Organ, J.M.; Chen, X.; Allen, M.R. A comparison of calcium to zoledronic acid for improvement of cortical bone in an animal model of CKD. J. Bone Miner. Res. 2014, 29, 902–910. [Google Scholar] [CrossRef] [Green Version]
  75. Hruby, A.; O’Donnell, C.J.; Jacques, P.F.; Meigs, J.B.; Hoffmann, U.; McKeown, N.M. Magnesium intake is inversely associated with coronary artery calcification: The framingham heart study. JACC Cardiovasc. Imaging 2014, 7, 59–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Ishimura, E.; Okuno, S.; Kitatani, K.; Tsuchida, T.; Yamakawa, T.; Shioi, A.; Inaba, M.; Nishizawa, Y. Significant association between the presence of peripheral vascular calcification and lower serum magnesium in hemodialysis patients. Clin. Nephrol. 2007, 68, 222–227. [Google Scholar] [CrossRef] [PubMed]
  77. Meema, H.E.; Oreopoulos, D.G.; Rapoport, A. Serum magnesium level and arterial calcification in end-stage renal disease. Kidney Int. 1987, 32, 388–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Tzanakis, I.; Virvidakis, K.; Tsomi, A.; Mantakas, E.; Girousis, N.; Karefyllakis, N.; Papadaki, A.; Kallivretakis, N.; Mountokalakis, T. Intra- and extracellular magnesium levels and atheromatosis in haemodialysis patients. Magnes. Res. 2004, 17, 102–108. [Google Scholar]
  79. Beulens, J.W.; Bots, M.L.; Atsma, F.; Bartelink, M.L.; Prokop, M.; Geleijnse, J.M.; Witteman, J.C.; Grobbee, D.E.; van der Schouw, Y.T. High dietary menaquinone intake is associated with reduced coronary calcification. Atherosclerosis 2009, 203, 489–493. [Google Scholar] [CrossRef]
  80. Geleijnse, J.M.; Vermeer, C.; Grobbee, D.E.; Schurgers, L.J.; Knapen, M.H.; van der Meer, I.M.; Hofman, A.; Witteman, J.C. Dietary intake of menaquinone is associated with a reduced risk of coronary heart disease: The Rotterdam Study. J. Nutr. 2004, 134, 3100–3105. [Google Scholar] [CrossRef]
  81. Hénaut, L.; Massy, Z.A. Magnesium as a calcification inhibitor. Adv. Chronic Kidney Dis. 2018, 25, 281–290. [Google Scholar] [CrossRef]
  82. Shea, M.K.; O’Donnell, C.J.; Hoffmann, U.; Dallal, G.E.; Dawson-Hughes, B.; Ordovas, J.M.; Price, P.A.; Williamson, M.K.; Booth, S.L. Vitamin K supplementation and progression of coronary artery calcium in older men and women. Am. J. Clin. Nutr. 2009, 89, 1799–1807. [Google Scholar] [CrossRef] [PubMed]
  83. Schurgers, L.J.; Teunissen, K.J.F.; Hamulyák, K.; Knapen, M.H.J.; Vik, H.; Vermeer, C. Vitamin K-containing dietary supplements: Comparison of synthetic vitamin K1 and natto-derived menaquinone-7. Blood 2007, 109, 3279–3283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Linefsky, J.P.; O’Brien, K.D.; Katz, R.; de Boer, I.H.; Barasch, E.; Jenny, N.S.; Siscovick, D.S.; Kestenbaum, B. Association of serum phosphate levels with aortic valve sclerosis and annular calcification: The cardiovascular health study. J. Am. Coll. Cardiol. 2011, 58, 291–297. [Google Scholar] [CrossRef] [Green Version]
  85. Linefsky, J.P.; O’Brien, K.D.; Sachs, M.; Katz, R.; Eng, J.; Michos, E.D.; Budoff, M.J.; de Boer, I.; Kestenbaum, B. Serum phosphate is associated with aortic valve calcification in the Multi-ethnic Study of Atherosclerosis (MESA). Atherosclerosis 2014, 233, 331–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. McCarty, M.F.; Lerner, A.; DiNicolantonio, J.J.; Iloki-Assanga, S.B. Bioavailable dietary phosphate, a mediator of cardiovascular disease, may be decreased with plant-based diets, phosphate binders, niacin, and avoidance of phosphate additives. Nutrition 2014, 30, 739–747. [Google Scholar] [CrossRef]
  87. Hatzigeorgiou, C.; Taylor, A.J.; Feuerstein, I.M.; Bautista, L.; O’Malley, P.G. Antioxidant vitamin intake and subclinical coronary atherosclerosis. Prev. Cardiol. 2006, 9, 75–81. [Google Scholar] [CrossRef]
  88. Sun, Y.; Byon, C.H.; Yang, Y.; Bradley, W.E.; Dell’Italia, L.J.; Sanders, P.W.; Agarwal, A.; Wu, H.; Chen, Y. Dietary potassium regulates vascular calcification and arterial stiffness. JCI Insight 2017, 2, e94920. [Google Scholar] [CrossRef] [Green Version]
  89. Young, D.B.; Lin, H.; McCabe, R.D. Potassium’s cardiovascular protective mechanisms. Am. J. Physiol. 1995, 268, R825–R837. [Google Scholar] [CrossRef]
  90. Beazley, K.E.; Lima, F.; Borras, T.; Nurminskaya, M. Attenuation of chondrogenic transformation in vascular smooth muscle by dietary quercetin in the MGP-deficient mouse model. PLoS ONE 2013, 8, e76210. [Google Scholar] [CrossRef] [Green Version]
  91. Tomayko, E.J.; Cachia, A.J.; Chung, H.R.; Wilund, K.R. Resveratrol supplementation reduces aortic atherosclerosis and calcification and attenuates loss of aerobic capacity in a mouse model of uremia. J. Med. Food 2014, 17, 278–283. [Google Scholar] [CrossRef] [Green Version]
  92. Rehman, H.; Krishnasamy, Y.; Haque, K.; Thurman, R.G.; Lemasters, J.J.; Schnellmann, R.G.; Zhong, Z. Green tea polyphenols stimulate mitochondrial biogenesis and improve renal function after chronic cyclosporin a treatment in rats. PLoS ONE 2013, 38, e65029. [Google Scholar] [CrossRef]
  93. Held, C.; Sumner, G.; Sheridan, P.; McQueen, M.; Smith, S.; Dagenais, G.; Yusuf, S.; Lonn, E. Correlations between plasma homocysteine and folate concentrations and carotid atherosclerosis in high-risk individuals: Baseline data from the Homocysteine and Atherosclerosis Reduction Trial (HART). Vasc. Med. 2008, 13, 245–253. [Google Scholar] [CrossRef] [Green Version]
  94. Ross, S. Functional foods: The food and drug administration perspective. Am. J. Clin. Nutr. 2000, 71, 1735S–1738S. [Google Scholar] [CrossRef] [Green Version]
  95. Thomas, P.R.; Earl, R. (Eds.) Enhancing the food supply. In Opportunities in the Nutrition and Food Sciences; Institute of Medicine, National Academy Press: Washington, DC, USA, 1994; pp. 98–142. [Google Scholar] [CrossRef]
  96. Block, G.; Patterson, B.B.; Subar, A. Fruit, vegetables, and cancer prevention: A review of the epidemiological evidence. Nutr. Cancer 1992, 18, 1–29. [Google Scholar] [CrossRef]
  97. Asgary, S.; Rastqar, A.; Keshvari, M. Functional Food and Cardiovascular Disease Prevention and Treatment: A Review. J. Am. Coll. Nutr. 2018, 37, 5–429. [Google Scholar] [CrossRef]
  98. Karagodin, V.P.; Sukhorukov, V.N.; Orekhov, A.N.; Yet, S.F.; Sobenin, I.A. Prevention of atherosclerosis: The role of special diets and functional foods. Front. BioSci. 2020, 12, 57–69. [Google Scholar] [CrossRef] [PubMed]
  99. Yanai, H.; Adachi, H.; Kawaguchi, A.; Hakoshima, M.; Waragai, Y.; Harigae, T.; Masui, Y.; Kakuta, K.; Hamasaki, H.; Katsuyama, H.; et al. The anti-atherosclerotic effects of tomatoes. Funct. Foods Health Dis. 2017, 7, 411–428. [Google Scholar] [CrossRef] [Green Version]
  100. McKay, D.L.; Blumberg, J.B. Cranberries (Vaccinium macrocarpon) and cardiovascular disease risk factors. Nutr. Rev. 2007, 65, 490–502. [Google Scholar] [CrossRef]
  101. Blumberg, J.B.; Basu, A.; Krueger, C.G.; Lila, M.A.; Neto, C.C.; Novotny, J.A.; Reed, J.D.; Rodriguez-Mateos, A.; Toner, C.D. Impact of cranberries on gut microbiota and cardiometabolic health: Proceedings of the cranberry health research conference 2015. Adv. Nutr. 2016, 7, 759S–770S. [Google Scholar] [CrossRef] [PubMed]
  102. Soliman, G.A. Dietary Fiber, Atherosclerosis, and Cardiovascular Disease. Nutrients 2019, 11, 1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. El Khoury, D.; Cuda, C.; Luhovyy, B.L.; Anderson, G.H. Beta glucan: Health benefits in obesity and metabolic syndrome. J. Nutr. Metab. 2012, 2012, 851362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kristensen, M.; Jensen, M.G.; Aarestrup, J.; Petersen, K.E.; Søndergaard, L.; Mikkelsen, M.S.; Astrup, A. Flaxseed dietary fibers lower cholesterol and increase fecal fat excretion, but magnitude of effect depend on food type. Nutr. Metab. 2012, 9, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Cheng, Y.C.; Sheen, J.M.; Hu, W.L.; Hung, Y.C. Polyphenols and Oxidative Stress in Atherosclerosis-Related Ischemic Heart Disease and Stroke. Oxid Med. Cell. Longev. 2017, 8526438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Neveu, V.; Perez-Jimenez, J.; Vos, F.; Crespy, V.; du Chaffaut, L.; Mennen, L.; Knox, C.; Eisner, R.; Cruz, J.; Wishart, D.; et al. Phenol-explorer: An online comprehensive database on polyphenol contents in foods. Database 2010, 2010, bap024. [Google Scholar] [CrossRef] [PubMed]
  107. Loke, W.M.; Proudfoot, J.M.; Hodgson, J.M.; McKinley, A.J.; Hime, N.; Magat, M.; Stocker, R.; Croft, K.D. Specific dietary polyphenols attenuate atherosclerosis in apolipoprotein E-knockout mice by alleviating inflammation and endothelial dysfunction. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 749–757. [Google Scholar] [CrossRef] [Green Version]
  108. Waddington, E.; Puddey, I.B.; Croft, K.D. Red wine polyphenolic compounds inhibit atherosclerosis in apolipoprotein E-deficient mice independently of effects on lipid peroxidation. Am. J. Clin. Nutr. 2004, 79, 54–61. [Google Scholar] [CrossRef]
  109. Stocker, R.; O’Halloran, R.A. Dealcoholized red wine decreases atherosclerosis in apolipoprotein E gene-deficient mice independently of inhibition of lipid peroxidation in the artery wall. Am. J. Clin. Nutr. 2004, 79, 123–130. [Google Scholar] [CrossRef]
  110. Miura, Y.; Chiba, T.; Tomita, I.; Koizumi, H.; Miura, S.; Umegaki, K.; Hara, Y.; Ikeda, M.; Tomita, T. Tea catechins prevent the development of atherosclerosis in apoprotein E-deficient mice. J. Nutr. 2001, 131, 27–32. [Google Scholar] [CrossRef] [Green Version]
  111. Chen, S.I.; Chiang, C.L.; Chao, C.T.; Chiang, C.K.; Huang, J.W. Gustatory function and the uremic toxin, phosphate, are modulators of the risk of vascular calcification among patients with chronic kidney disease: A pilot study. Toxins 2020, 12, 420. [Google Scholar] [CrossRef]
  112. Machado, A.D.; Gómez, L.M.; Marchioni, D.M.L.; Dos Anjos, F.S.N.; Molina, M.D.C.B.; Lotufo, P.A.; Benseñor, I.J.M.; Titan, S.M.d.O. Association between Dietary Intake and Coronary Artery Calcification in Non-Dialysis Chronic Kidney Disease: The PROGREDIR Study. Nutrients 2018, 10, 372. [Google Scholar] [CrossRef] [Green Version]
  113. Samelson, E.J.; Booth, S.L.; Fox, C.S.; Tucker, K.L.; Wang, T.J.; Hoffmann, U.; Cupples, L.A.; O’Donnell, C.J.; Kiel, D.P. Calcium intake is not associated with increased coronary artery calcification: The Framingham Study. Am. J. Clin. Nutr. 2012, 96, 1274–1280. [Google Scholar] [CrossRef] [Green Version]
  114. Lutsey, P.L.; Michos, E.D. Vitamin D, calcium, and atherosclerotic risk: Evidence from serum levels and supplementation studies. Curr. Atheroscler. Rep. 2013, 15, 293. [Google Scholar] [CrossRef] [Green Version]
  115. Li, K.; Kaaks, R.; Linseisen, J.; Rohrmann, S. Associations of dietary calcium intake and calcium supplementation with myocardial infarction and stroke risk and overall cardiovascular mortality in the Heidelberg cohort of the European Prospective Investigation into Cancer and Nutrition study (EPIC-Heidelberg). Heart 2012, 98, 920–925. [Google Scholar] [CrossRef] [Green Version]
  116. Bolland, M.J.; Grey, A.; Avenell, A.; Gamble, G.D.; Reid, I.R. Calcium supplements with or without vitamin D and risk of cardiovascular events: Reanalysis of the Women’s Health Initiative limited access dataset and meta-analysis. BMJ 2011, 342, d2040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Bolland, M.J.; Avenell, A.; Baron, J.A.; Grey, A.; MacLennan, G.S.; Gamble, G.D.; Reid, I.R. Effect of calcium supplements on risk of myocardial infarction and cardiovascular events: Meta-analysis. BMJ 2010, 341, c3691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Bolland, M.J.; Barber, P.A.; Doughty, R.N.; Mason, B.; Horne, A.; Ames, R.; Gamble, G.D.; Grey, A.; Reid, I.R. Vascular events in healthy older women receiving calcium supplementation: Randomised controlled trial. BMJ 2008, 336, 262–266. [Google Scholar] [CrossRef] [Green Version]
  119. Anderson, J.J.; Kruszka, B.; Delaney, J.A.; He, K.; Burke, G.L.; Alonso, A.; Bild, D.E.; Budoff, M.; Michos, E.D. Calcium Intake from Diet and Supplements and the Risk of Coronary Artery Calcification and its Progression Among Older Adults: 10-Year Follow-up of the Multi-Ethnic Study of Atherosclerosis (MESA). J. Am. Heart Assoc. 2016, 5, e003815. [Google Scholar] [CrossRef] [PubMed]
  120. Rautiainen, S.; Wang, L.; Manson, J.E.; Sesso, H.D. The role of calcium in the prevention of cardiovascular disease–a review of observational studies and randomized clinical trials. Curr. Atheroscler. Rep. 2013, 15, 362. [Google Scholar] [CrossRef] [PubMed]
  121. Michaelsson, K.; Melhus, H.; Warensjo Lemming, E.; Wolk, A.; Byberg, L. Long term calcium intake and rates of all cause and cardiovascular mortality: Community based prospective longitudinal cohort study. BMJ 2013, 346, f228. [Google Scholar] [CrossRef] [Green Version]
  122. McCabe, K.M.; Booth, S.L.; Fu, X.; Shobeiri, N.; Pang, J.J.; Adams, M.A.; Holden, R.M. Dietary vitamin K and therapeutic warfarin alter the susceptibility to vascular calcification in experimental chronic kidney disease. Kidney Int. 2013, 83, 835–844. [Google Scholar] [CrossRef] [Green Version]
  123. Thijssen, H.H.; Drittij-Reijnders, M.J.; Fischer, M.A. Phylloquinone and menaquinone-4 distribution in rats: Synthesis rather than uptake determines menaquinone-4 organ concentrations. J. Nutr. 1996, 126, 537–543. [Google Scholar] [CrossRef] [Green Version]
  124. Hasific, S.; Øvrehus, K.A.; Gerke, O.; Hallas, J.; Busk, M.; Lambrechtsen, J.; Urbonaviciene, G.; Sand, N.P.R.; Nielsen, J.S.; Diederichsen, L.; et al. Extent of arterial calcification by conventional vitamin K antagonist treatment. PLoS ONE 2020, 15, e0241450. [Google Scholar] [CrossRef] [PubMed]
  125. Capelli, I.; Cianciolo, G.; Gasperoni, L.; Zappulo, F.; Tondolo, F.; Cappuccilli, M.; La Manna, G. Folic Acid and Vitamin B12 Administration in CKD, Why Not? Nutrients 2019, 11, 383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Bamonti-Catena, F.; Buccianti, G.; Porcella, A.; Valenti, G.; Como, G.; Finazzi, S.; Maiolo, A.T. Folate measurements in patients on regular hemodialysis treatment. Am. J. Kidney Dis. 1999, 33, 492–497. [Google Scholar] [CrossRef]
  127. Chrysant, S.G.; Chrysant, G.S. The current status of homocysteine as a risk factor for cardiovascular disease: A mini review. Expert Rev. Cardiovasc. Ther. 2018, 16, 559–565. [Google Scholar] [CrossRef]
  128. Wald, D.S.; Law, M.; Morris, J.K. Homocysteine and cardiovascular disease: Evidence on causality from a metaanalysis. BMJ 2002, 325, 1202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Homocysteine Studies Collaboration. Homocysteine and risk of ischemic heart disease and stroke: A meta-analysis. JAMA 2002, 288, 2015–2022. [Google Scholar] [CrossRef] [PubMed]
  130. Robinson, K. Renal disease, homocysteine, and cardiovascular complications. Circulation 2004, 109, 294–295. [Google Scholar] [CrossRef] [PubMed]
  131. Langan, R.C.; Goodbred, A.J. Vitamin B12 Deficiency: Recognition and Management. Am. Fam. Physician 2017, 96, 384–389. [Google Scholar]
  132. Heinz, J.; Kropf, S.; Luley, C.; Dierkes, J. Homocysteine as a risk factor for cardiovascular disease in patients treated by dialysis: A meta-analysis. Am. J. Kidney Dis. 2009, 54, 478–489. [Google Scholar] [CrossRef]
  133. Haraki, T.; Takegoshi, T.; Kitoh, C.; Kajinami, K.; Wakasugi, T.; Hirai, J.; Shimada, T.; Kawashiri, M.; Inazu, A.; Koizumi, J.; et al. Hyperhomocysteinemia, diabetes mellitus, and carotid atherosclerosis independently increase atherosclerotic vascular disease outcome in Japanese patients with end-stage renal disease. Clin. Nephrol. 2001, 56, 132–139. [Google Scholar]
  134. Kunz, K.; Petitjean, P.; Lisri, M.; Chantrel, F.; Koehl, C.; Wiesel, M.L.; Cazenave, J.P.; Moulin, B.; Hannedouche, T.P. Cardiovascular morbidity and endothelial dysfunction in chronic haemodialysis patients: Is homocyst(e)ine the missing link? Nephrol. Dial. Transpl. 1999, 14, 1934–1942. [Google Scholar] [CrossRef] [Green Version]
  135. Manns, B.J.; Burgess, E.D.; Hyndman, M.E.; Parsons, H.G.; Schaefer, J.P.; Scott-Douglas, N.W. Hyperhomocyst(e)inemia and the prevalence of atherosclerotic vascular disease in patients with end-stage renal disease. Am. J. Kidney Dis. 1999, 34, 669–677. [Google Scholar] [CrossRef]
  136. Robinson, K.; Gupta, A.; Dennis, V.; Arheart, K.; Chaudhary, D.; Green, R.; Vigo, P.; Mayer, E.L.; Selhub, J.; Kutner, M.; et al. Hyperhomocysteinemia confers an independent increased risk of atherosclerosis in end-stage renal disease and is closely linked to plasma folate and pyridoxine concentrations. Circulation 1996, 94, 2743–2748. [Google Scholar] [CrossRef] [PubMed]
  137. Bachmann, J.; Tepel, M.; Raidt, H.; Riezler, R.; Graefe, U.; Langer, K.; Zidek, W. Hyperhomocysteinemia and the risk for vascular disease in hemodialysis patients. J. Am. Soc. Nephrol. 1995, 6, 121–125. [Google Scholar] [CrossRef] [PubMed]
  138. Cianciolo, G.; De Pascalis, A.; Di Lullo, L.; Ronco, C.; Zannini, C.; La Manna, G. Folic Acid and Homocysteine in Chronic Kidney Disease and Cardiovascular Disease Progression: Which Comes First? Cardioren. Med. 2017, 7, 255–266. [Google Scholar] [CrossRef]
  139. Vychytil, A.; Födinger, M.; Wölfl, G.; Enzenberger, B.; Auinger, M.; Prischl, F.; Buxbaum, M.; Wiesholzer, M.; Mannhalter, C.; Hörl, W.H.; et al. Major determinants of hyperhomocysteinemia in peritoneal dialysis patients. Kidney Int. 1998, 53, 1775–1782. [Google Scholar] [CrossRef] [Green Version]
  140. Suliman, M.E.; Qureshi, A.R.; Bárány, P.; Stenvinkel, P.; Filho, J.C.; Anderstam, B.; Heimbürger, O.; Lindholm, B.; Bergström, J. Hyperhomocysteinemia, nutritional status, and cardiovascular disease in hemodialysis patients. Kidney Int. 2000, 57, 1727–1735. [Google Scholar] [CrossRef] [Green Version]
  141. Wrone, E.M.; Zehnder, J.L.; Hornberger, J.M.; McCann, L.M.; Coplon, N.S.; Fortmann, S.P. An MTHFR variant, homocysteine, and cardiovascular comorbidity in renal disease. Kidney Int. 2001, 60, 1106–1113. [Google Scholar] [CrossRef] [Green Version]
  142. Soohoo, M.; Ahmadi, S.F.; Qader, H.; Streja, E.; Obi, Y.; Moradi, H.; Rhee, C.M.; Kim, T.H.; Kovesdy, C.P.; Kalantar-Zadeh, K. Association of serum vitamin B12 and folate with mortality in incident hemodialysis patients. Nephrol. Dial. Transpl. 2017, 32, 1024–1032. [Google Scholar] [CrossRef] [Green Version]
  143. Righetti, M.; Serbelloni, P.; Milani, S.; Ferrario, G. Homocysteine-lowering vitamin B treatment decreases cardiovascular events in hemodialysis patients. Blood Purif. 2006, 24, 379–386. [Google Scholar] [CrossRef] [PubMed]
  144. Heinz, J.; Kropf, S.; Domröse, U.; Westphal, S.; Borucki, K.; Luley, C.; Neumann, K.H.; Dierkes, J. B vitamins and the risk of total mortality and cardiovascular disease in end-stage renal disease: Results of a randomized controlled trial. Circulation 2010, 121, 1432–1438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Mann, J.F.; Sheridan, P.; McQueen, M.J.; Held, C.; Arnold, J.M.; Fodor, G.; Yusuf, S.; Lonn, E.M. HOPE-2 investigators. Homocysteine lowering with folic acid and B vitamins in people with chronic kidney disease—Results of the renal Hope-2 study. Nephrol. Dial. Transpl. 2008, 23, 645–653. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Jamison, R.L.; Hartigan, P.; Kaufman, J.S.; Goldfarb, D.S.; Warren, S.R.; Guarino, P.D.; Gaziano, J.M. Veterans Affairs Site Investigators. Effect of Homocysteine Lowering on Mortality and Vascular Disease in Advanced Chronic Kidney Disease and End-stage Renal Disease. JAMA 2007, 298, 1163–1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Fleckenstein-Grün, G.; Thimm, F.; Frey, M.; Czirfusz, A. Role of calcium in arteriosclerosis—Experimental evaluation of antiarteriosclerotic potencies of Ca antagonists. Study group for calcium antagonism. Basic Res. Cardiol. 1994, 89 (Suppl. S1), 145–159. [Google Scholar] [CrossRef]
  148. Armstrong, Z.B.; Boughner, D.R.; Drangova, M.; Rogers, K.A. Angiotensin II type 1 receptor blocker inhibits arterial calcification in a pre-clinical model. Cardiovasc. Res. 2011, 90, 165–170. [Google Scholar] [CrossRef] [Green Version]
  149. Henry, P.D.; Bentley, K.I. Suppression of atherogenesis in cholesterol-fed rabbit treated with nifedipine. J. Clin. Investig. 1981, 68, 1366–1369. [Google Scholar] [CrossRef] [Green Version]
  150. Pitt, B.; Byington, R.P.; Furberg, C.D.; Hunninghake, D.B.; Mancini, G.B.; Miller, M.E.; Riley, W. Effect of amlodipine on the progression of atherosclerosis and the occurrence of clinical events. PREVENT Investigators. Circulation 2000, 102, 1503–1510. [Google Scholar] [CrossRef] [Green Version]
  151. Sugano, M.; Nakashima, Y.; Tasaki, H.; Takasugi, M.; Kuroiwa, A.; Koide, O. Effects of diltiazem on suppression and regression of experimental atherosclerosis. Br. J. Exp. Pathol. 1988, 69, 515–523. [Google Scholar] [PubMed]
  152. Lichtlen, P.R.; Hugenholtz, P.G.; Rafflenbeul, W.; Hecker, H.; Jost, S.; Nikutta, P.; Deckers, J.W. Retardation of coronary artery disease in humans by the calcium-channel blocker nifedipine: Results of the INTACT study (International Nifedipine Trial on Antiatherosclerotic Therapy). Cardiovasc. Drugs Ther. 1990, 4 (Suppl. 5), 1047–1068. [Google Scholar] [CrossRef] [PubMed]
  153. Motro, M.; Kirwan, B.A.; de Brouwer, S.; Poole-Wilson, P.A.; Shemesh, J.; ACTION CC Side-Arm Study. Tracking coronary calcification and atherosclerotic lesions in patients with stable angina pectoris undergoing nifedipine therapy. Cardiology 2007, 107, 165–171. [Google Scholar] [CrossRef]
  154. Motro, M.; Shemesh, J. Calcium channel blocker nifedipine slows down progression of coronary calcification in hypertensive patients compared with diuretics. Hypertension 2001, 37, 1410–1413. [Google Scholar] [CrossRef] [Green Version]
  155. Simon, A.; Gariépy, J.; Moyse, D.; Levenson, J. Differential effects of nifedipine and co-amilozide on the progression of early carotid wall changes. Circulation 2001, 103, 2949–2954. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Henein, M.Y.; Owen, A. Statins moderate coronary stenoses but not coronary calcification: Results from meta-analyses. Int. J. Cardiol. 2011, 153, 31–35. [Google Scholar] [CrossRef] [PubMed]
  157. Tsai, C.W.; Lin, S.Y.; Kuo, C.C.; Huang, C.C. Serum Uric Acid and Progression of Kidney Disease: A Longitudinal Analysis and Mini-Review. PLoS ONE 2017, 12, e0170393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Wong, J.; Piceno, Y.M.; DeSantis, T.Z.; Pahl, M.; Andersen, G.L.; Vaziri, N.D. Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am. J. Nephrol. 2014, 39, 230–237. [Google Scholar] [CrossRef] [Green Version]
  159. Rodrigues, F.G.; Ormanji, M.S.; Heilberg, I.P.; Bakker, S.J.L.; de Borst, M.H. Interplay between gut microbiota, bone health and vascular calcification in chronic kidney disease. Eur. J. Clin. Investig. 2021, e13588, advance online publication. [Google Scholar] [CrossRef]
  160. Cozzolino, M.; Fusaro, M.; Ciceri, P.; Gasperoni, L.; Cianciolo, G. The role of vitamin K in vascular calcification. Adv. Chronic Kidney Dis. 2019, 26, 437–444. [Google Scholar] [CrossRef]
  161. Armani, R.G.; Ramezani, A.; Yasir, A.; Sharama, S.; Canziani, M.E.F.; Raj, D.S. Gut microbiome in chronic kidney disease. Curr. Hypertens. Rep. 2017, 19, 29. [Google Scholar] [CrossRef] [PubMed]
  162. Esgalhado, M.; Kemp, J.A.; Azevedo, R.; Paiva, B.R.; Stockler-Pinto, M.B.; Dolenga, C.J.; Borges, N.A.; Nakao, L.S.; Mafra, D. Could resistant starch supplementation improve inflammatory and oxidative stress biomarkers and uremic toxins levels in hemodialysis patients? A pilot randomized controlled trial. Food Funct. 2018, 9, 6508–6516. [Google Scholar] [CrossRef] [PubMed]
  163. Hill, E.; Sapa, H.; Negrea, L.; Bame, K.; Hostetter, T.; Barkoukis, H.; Dusso, A.; Dobre, M. Effect of Oat b -glucan supplementation on chronic kidney disease: A feasibility study. J. Ren. Nutr. 2019, 30, 208–215. [Google Scholar] [CrossRef]
  164. Iorga, R.; Bratu, O.G.; Bacalbasa, N.; Gaman, M.A.; Diaconu, C.C. Intestinal microbiota—A possible contributor to cardiovascular diseases? J. Mind Med. Sci. 2020, 7, 168–172. [Google Scholar] [CrossRef]
  165. Cho, Y.A.; Kim, J. Effect of Probiotics on Blood Lipid Concentrations: A Meta-Analysis of Randomized Controlled Trials. Medicine 2015, 94, e1714. [Google Scholar] [CrossRef]
  166. Pourrajab, B.; Fatahi, S.; Sohouli, M.H.; Găman, M.A.; Shidfar, F. The effects of probiotic/synbiotic supplementation compared to placebo on biomarkers of oxidative stress in adults: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2020, 4, 1–18. [Google Scholar] [CrossRef]
  167. Matsumoto, M. Prevention of Atherosclerosis by the Induction of Microbial Polyamine Production in the Intestinal Lumen. Biol. Pharm. Bull. 2020, 43, 221–229. [Google Scholar] [CrossRef]
  168. De la Peña, N.C.; Sosa-Melgarejo, J.A.; Ramos, R.R.; Méndez, J.D. Inhibition of platelet aggregation by putrescine, spermidine, and spermine in hypercholesterolemic rabbits. Arch. Med. Res. 2000, 31, 546–550. [Google Scholar] [CrossRef]
  169. Zhang, M.; Caragine, T.; Wang, H.; Cohen, P.S.; Botchkina, G.; Soda, K.; Bianchi, M.; Ulrich, P.; Cerami, A.; Sherry, B.; et al. Spermine inhibits proinflammatory cytokine synthesis in human mononuclear cells: A counterregulatory mechanism that restrains the immune response. J. Exp. Med. 1997, 185, 1759–1768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Soda, K.; Kano, Y.; Nakamura, T.; Kasono, K.; Kawakami, M.; Konishi, F. Spermine, a natural polyamine, suppresses LFA-1 expression on human lymphocyte. J. Immunol. 2005, 175, 237–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. LaRocca, T.J.; Gioscia-Ryan, R.A.; Hearon, C.M., Jr.; Seals, D.R. The autophagy enhancer spermidine reverses arterial aging. Mech. Ageing Dev. 2013, 134, 314–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Eisenberg, T.; Abdellatif, M.; Schroeder, S.; Primessnig, U.; Stekovic, S.; Pendl, T.; Harger, A.; Schipke, J.; Zimmermann, A.; Schmidt, A.; et al. Cardioprotection and lifespan extension by the natural polyamine spermidine. Nat. Med. 2016, 22, 1428–1438. [Google Scholar] [CrossRef]
  173. Michiels, C.F.; Kurdi, A.; Timmermans, J.P.; De Meyer, G.R.Y.; Martinet, W. Spermidine reduces lipid accumulation and necrotic core formation in atherosclerotic plaques via induction of autophagy. Atherosclerosis 2016, 251, 319–327. [Google Scholar] [CrossRef] [PubMed]
  174. Matsumoto, M.; Kitada, Y.; Naito, Y. Endothelial Function is improved by Inducing Microbial Polyamine Production in the Gut: A Randomized Placebo-Controlled Trial. Nutrients 2019, 11, 1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Doherty, T.M.; Detrano, R.C. Coronary arterial calcification as an active process: A new perspective on an old problem. Calcif. Tissue Int. 1994, 54, 224–230. [Google Scholar] [CrossRef]
  176. Shearer, M.J. Vitamin K metabolism and nutriture. Blood Rev. 1992, 6, 92–104. [Google Scholar] [CrossRef]
  177. Kawashima, H.; Nakajima, Y.; Matubara, Y.; Nakanowatari, J.; Fukuta, T.; Mizuno, S.; Takahashi, S.; Tajima, T.; Nakamura, T. Effects of vitamin K2 (menatetrenone) on atherosclerosis and blood coagulation in hypercholesterolemic rabbits. Jpn. J. Pharmacol. 1997, 75, 135–143. [Google Scholar] [CrossRef]
  178. Mitropoulos, K.A.; Esnouf, M.P.; Meade, T.W. Increased Factor VII coagulant activity in the rabbit following diet-induced hypercholesterolaemia: Evidence for increased conversion of VII to aVIIa and higher flux within the coagulation pathway. Atherosclerosis 1987, 63, 43–52. [Google Scholar] [CrossRef]
  179. Mitropoulos, K.A.; Esnouf, M.P. Turnover of factor X and of prothrombin in rabbits fed on a standard or cholesterol supplemented diet. Biochem. J. 1987, 244, 263–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Lupu, F.; Moldovan, N.; Ryan, J.; Stern, D.; Simionescu, N. Intrinsic procoagulant surface induced by hypercholesterolaemia on rabbit aortic endothelium. Blood Coagul. Fibrinolysis 1993, 4, 743–752. [Google Scholar] [CrossRef]
  181. Kurnatowska, I.; Grzelak, P.; Masajtis-Zagajewska, A.; Kaczmarska, M.; Stefańczyk, L.; Vermeer, C.; Maresz, K.; Nowicki, M. Effect of vitamin K2 on progression of atherosclerosis and vascular calcification in nondialyzed patients with chronic kidney disease stages 3–5. Pol. Arch. Med. Wewn. 2015, 125, 631–640. [Google Scholar] [CrossRef]
  182. Theuwissen, E.; Smit, E.; Vermeer, C. The role of vitamin K in soft-tissue calcification. Adv. Nutr. 2012, 3, 166–173. [Google Scholar] [CrossRef] [Green Version]
  183. Levy, R.J.; Lian, J.B.; Gallop, P. Atherocalcin, a gamma-carboxyglutamic acid containing protein from atherosclerotic plaque. Biochem. Biophys. Res. Commun. 1979, 91, 41–49. [Google Scholar] [CrossRef]
  184. Levy, R.J.; Zenker, J.A.; Lian, J.B. Vitamin K-dependent calcium binding proteins in aortic valve calcification. J. Clin. Investig. 1980, 65, 563–566. [Google Scholar] [CrossRef] [PubMed]
  185. Gijsbers, B.L.M.G.; van Haarlem, L.J.M.; Soute, B.A.M.; Ebberink, R.H.M.; Vermeer, C. Characterization of a Gla-containing protein from calcified human atherosclerotic plaques. Arteriosclerosis 1990, 10, 991–995. [Google Scholar] [CrossRef] [Green Version]
  186. Li, X.; Su, C.; Jiang, Z.; Yang, Y.; Zhang, Y.; Yang, M.; Zhang, X.; Du, Y.; Zhang, J.; Wang, L.; et al. Berberine attenuates choline-induced atherosclerosis by inhibiting trimethylamine and trimethylamine-N-oxide production via manipulating the gut microbiome. NPJ Biofilms Microbiomes 2021, 7, 36. [Google Scholar] [CrossRef] [PubMed]
  187. Jayachandran, M.; Chung, S.S.M.; Xu, B. A critical review of the relationship between dietary components, the gut microbe Akkermansia muciniphila, and human health. Crit. Rev. Food Sci. Nutr. 2020, 60, 2265–2276. [Google Scholar] [CrossRef]
  188. Chen, M.L.; Yi, L.; Zhang, Y.; Zhou, X.; Ran, L.; Yang, J.; Zhu, J.D.; Zhang, Q.Y.; Mi, M.T. Resveratrol attenuates trimethyl-amine-N-oxide (TMAO)-induced atherosclerosis by regulating TMAO synthesis and bile acid metabolism via remodeling of the gut microbiota. MBio 2016, 7, e02210–e02215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Li, X.; Shimizu, Y.; Kimura, I. Gut microbial metabolite short-chain fatty acids and obesity. BioSci. Microbiota Food Health. 2017, 36, 135–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Natarajan, N.; Hori, D.; Flavahan, S.; Steppan, J.; Flavahan, N.A.; Berkowitz, D.E.; Pluznick, J.L. Microbial short chain fatty acid metabolites lower blood pressure via endothelial G protein-coupled receptor 41. Physiol. Genom. 2016, 48, 826–834. [Google Scholar] [CrossRef]
  191. Ohira, H.; Tsutsui, W.; Fujioka, Y. Are Short Chain Fatty Acids in Gut Microbiota Defensive Players for Inflammation and Atherosclerosis? J. Atheroscler. Thromb. 2017, 24, 660–672. [Google Scholar] [CrossRef] [Green Version]
  192. Wahlström, A.; Sayin, S.I.; Marschall, H.U.; Bäckhed, F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Rodríguez-Morató, J.; Matthan, N.R. Nutrition and Gastrointestinal Microbiota, Microbial-Derived Secondary Bile Acids, and Cardiovascular Disease. Curr. Atheroscler. 2020, 22, 47. [Google Scholar] [CrossRef] [PubMed]
  194. Ma, J.; Li, H. The role of gut microbiota in atherosclerosis and hypertension. Front. Pharmacol. 2018, 9, 1082. [Google Scholar] [CrossRef]
  195. Busnelli, M.; Manzini, S.; Chiesa, G. The gut microbiota affects host pathophysiology as an endocrine organ: A focus on cardiovascular disease. Nutrients 2019, 12, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Tomova, A.; Bukovsky, I.; Rembert, E.; Yonas, W.; Alwarith, J.; Barnard, N.D.; Kahleova, H. The Effects of Vegetarian and Vegan Diets on Gut Microbiota. Front. Nutr. 2019, 6, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Connolly, M.L.; Tzounis, X.; Tuohy, K.M.; Lovegrove, J.A. Hypocholesterolemic and prebiotic effects of a whole-grain oatbased granola breakfast cereal in a cardio-metabolic “at risk” population. Front. Microbiol. 2016, 7, 1675. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Evolution of atherosclerosis and its consequences. The process of atherosclerosis begins with an endothelial injury, leading to accumulation of macrophage foam cells and infiltration of smooth muscle cells with development of fatty streaks. From this point, atherosclerosis, through an inflammatory process, will lead to the development of a vulnerable plaque, consisting of necrotic core, micro- and macrocalcification, and intraplaque hemorrhage. When the thin fibrous cap of the vulnerable plaque develops a fissure, platelets will begin to aggregate, leading to thrombosis and acute coronary syndrome, but also other pathologies. Created with BioRender.com (Last accessed on 21 July 2021).
Figure 1. Evolution of atherosclerosis and its consequences. The process of atherosclerosis begins with an endothelial injury, leading to accumulation of macrophage foam cells and infiltration of smooth muscle cells with development of fatty streaks. From this point, atherosclerosis, through an inflammatory process, will lead to the development of a vulnerable plaque, consisting of necrotic core, micro- and macrocalcification, and intraplaque hemorrhage. When the thin fibrous cap of the vulnerable plaque develops a fissure, platelets will begin to aggregate, leading to thrombosis and acute coronary syndrome, but also other pathologies. Created with BioRender.com (Last accessed on 21 July 2021).
Microorganisms 09 01913 g001
Figure 2. Main bacterial phyla found in atherosclerotic plaque. Bacterial analysis from atherosclerotic plaque samples proved the existence of microorganisms common with the gut microbiota or oral cavity microbiota. The oral cavity and the gut are dominated by Firmicutes, while atherosclerotic plaques are rich in Proteobacteria. The Bacteroidetes phylum is very well represented in the gut but is present to a lesser extent in atherosclerotic plaques. The ratio of Firmicutes/Bacteroidetes is demonstrated to be higher in patients suffering from coronary artery disease. Created with BioRender.com (Last accessed on 21 July 2021).
Figure 2. Main bacterial phyla found in atherosclerotic plaque. Bacterial analysis from atherosclerotic plaque samples proved the existence of microorganisms common with the gut microbiota or oral cavity microbiota. The oral cavity and the gut are dominated by Firmicutes, while atherosclerotic plaques are rich in Proteobacteria. The Bacteroidetes phylum is very well represented in the gut but is present to a lesser extent in atherosclerotic plaques. The ratio of Firmicutes/Bacteroidetes is demonstrated to be higher in patients suffering from coronary artery disease. Created with BioRender.com (Last accessed on 21 July 2021).
Microorganisms 09 01913 g002
Figure 3. The TMA/FMO/TMAO pathway is highly dependent on the diet constituents and plays a role in the pathogenesis of CVD. Trimethylamine (TMA), resulting from bacterial metabolism, will interact with the enzyme flavin monooxygenase 3 (FMO), which is a powerful modifier of cholesterol metabolism and responsible for trimethylamine N-oxide (TMAO) production. Created with BioRender.com (Last accessed on 21 July 2021).
Figure 3. The TMA/FMO/TMAO pathway is highly dependent on the diet constituents and plays a role in the pathogenesis of CVD. Trimethylamine (TMA), resulting from bacterial metabolism, will interact with the enzyme flavin monooxygenase 3 (FMO), which is a powerful modifier of cholesterol metabolism and responsible for trimethylamine N-oxide (TMAO) production. Created with BioRender.com (Last accessed on 21 July 2021).
Microorganisms 09 01913 g003
Figure 4. Beneficial effects of fecal microbiota transplantation. The concept of fecal transplantation is represented by the transfer of beneficial bacteria from the stools of a healthy donor to a patient with a disease caused by dysbiosis of their gut microbiota, in order to increase microbial diversity. Created with BioRender.com (Last accessed on 21 July 2021).
Figure 4. Beneficial effects of fecal microbiota transplantation. The concept of fecal transplantation is represented by the transfer of beneficial bacteria from the stools of a healthy donor to a patient with a disease caused by dysbiosis of their gut microbiota, in order to increase microbial diversity. Created with BioRender.com (Last accessed on 21 July 2021).
Microorganisms 09 01913 g004
Table 1. Influence on vascular calcifications in patients with chronic kidney disease and gut dysbiosis.
Table 1. Influence on vascular calcifications in patients with chronic kidney disease and gut dysbiosis.
Promoters of Vascular
Calcifications
Inhibitors of Vascular
Calcifications
TMAO [18]Vitamin K [19]
Inflammatory cytokines [20]Osteoprotegerin [21]
Oxidative stress [22]Pyrophosphate [23]
Free p-cresylsulphate [24]Fetuin-A [21]
Serum calcium [22,23]Matrix Gla protein [21]
Serum phosphate [22,23]Osteopontin [23]
Serum PTH [22]Bone morphogenetic protein 7 [21]
Serum FGF-23 [22]
Bone morphogenetic protein 2 [21]
Osteocalcin [21]
Osteonectin [21]
Indoxyl sulfate [25]
Table 2. Dietary recommendations and their contribution to vascular calcification.
Table 2. Dietary recommendations and their contribution to vascular calcification.
May Protect against Cardiovascular
Calcifications
Potential Triggers of Cardiovascular
Calcifications
Avoidance of trans fats and simple sugars [68]Diets low in carbohydrates from young ages [69]
Diet rich in vegetables, oily fish, and long-chain omega 3 fats [68]Plasmatic homocysteine > 12 µmol/L [70]
Calcium (800 μg/day) [68,71,72,73,74]Oxidative stress [68]
Magnesium (≥380 mg/day) [75,76,77,78]
Menaquinone (vitamin K2) [79,80]
Low serum magnesium [81]
Phylloquinone (vitamin K1) [16,82,83]Phosphorus [84,85,86]
Serum 25(OH)D ≥ 75 nmol/L [68]α-tocopherol [87]
High dietary potassium [88,89]Low dietary potassium [88]
Quercetin [90]
Resveratrol [91]
Epigallocatechin gallate [92]
Plasmatic folate > 39.4 nmol/L [68,93]
Table 3. Summary of recent studies linking gut microbiome, dietary interventions, and atherosclerosis.
Table 3. Summary of recent studies linking gut microbiome, dietary interventions, and atherosclerosis.
StudyStudy DetailsTreatment/InterventionResults
Study DesignSubject NumberDurationTreatmentGut MicrobiomeMicrobial MetabolitesMarkers of Atherosclerosis
Esgalhado, M., 2018 [162]Randomized, double-blind, placebo-controlled trial43 CKD patients4 weeksResistant
starch (Hi-Maize® 260, Ingredion, Westchester, IL, USA) or placebo (manioc flour, Yoki) supplementation
Bifidobacteria with balancing the Bacteroidetes and Firmicutes ratio↓indoxyl sulfate; p-Cresyl sulfate not affected↓PC; ↓TBARS; ↓IL-6; ↓hs-CRP
Matsumoto, M., 2019 [174]Randomized, double-blinded, placebo-controlled, parallel-group comparative study44 healthy subjects12 weeksNormal yogurt containing Bifal and Arg or placebo (normal yogurt)Citrobacter; ↑Escherichia/Shigella ratio; ↑Enterococcus;Bacteroidetes/Firmicutes ratio↑putrescine production↑RHI; ↓BP; ↓Serum platelet; ↓triglyceride concentrations; ↑HDL-cholesterol
Li, X., 2021 [186]Laboratory study5 C57BL/6J strain mice and 5 ApoE KO model mice6 and 16 weeks respectivelyStandard chow diet (0.1% choline) or choline diet (chow diet with 1% additional choline) +/− BBRthe choline I group: ↑Clostridium, Eubacterium, Lachnoclostridium, Roseburia, Odoribacter; the C + BBR group: ↑Bacteroides, Prevotella, Parabacteroides, Alloprevotella↓TMA↓serum TMAO level; ↓progression of atherosclerotic plaque; ↓macrophage-specific biomarkers of macrophage-derived foam cells
Chen, M., 2016 [188]Laboratory study10 female C57BL/6J strain mice and 10 ApoE model mice30 daysStandard chow diet (NIH31 modified mouse/rat diet) or resveratrolLactobacillus; ↑Bifidobacterium; ↑Bacteroides; ↑Akkermansia↓TMA ↑BA deconjugation↓serum TMAO level; ↑hepatic BA neosynthesis; ↓TMAO-induced atherosclerosis
CKD—chronic kidney disease, PC—protein carbonyl, TBARS—thiobarbituric-acid-reactive substances, IL-6—interleukin-6, hs-CRP—high-sensitivity C-reactive protein, RHI—reactive hyperemia index, BP—blood pressure, BBR—berberine, TMA—trimethylamine, TMAO—trimethylamine N-oxide, BA—bile acid, ↑—elevation, ↓—diminution.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cretoiu, D.; Ionescu, R.F.; Enache, R.M.; Cretoiu, S.M.; Voinea, S.C. Gut Microbiome, Functional Food, Atherosclerosis, and Vascular Calcifications—Is There a Missing Link? Microorganisms 2021, 9, 1913. https://doi.org/10.3390/microorganisms9091913

AMA Style

Cretoiu D, Ionescu RF, Enache RM, Cretoiu SM, Voinea SC. Gut Microbiome, Functional Food, Atherosclerosis, and Vascular Calcifications—Is There a Missing Link? Microorganisms. 2021; 9(9):1913. https://doi.org/10.3390/microorganisms9091913

Chicago/Turabian Style

Cretoiu, Dragos, Ruxandra Florentina Ionescu, Robert Mihai Enache, Sanda Maria Cretoiu, and Silviu Cristian Voinea. 2021. "Gut Microbiome, Functional Food, Atherosclerosis, and Vascular Calcifications—Is There a Missing Link?" Microorganisms 9, no. 9: 1913. https://doi.org/10.3390/microorganisms9091913

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