Next Article in Journal
Validity of the Mediterranean Diet and Culinary Index (MediCul) for Online Assessment of Adherence to the ‘Traditional’ Diet and Aspects of Cuisine in Older Adults
Next Article in Special Issue
Effects of Grape Pomace Polyphenolic Extract (Taurisolo®) in Reducing TMAO Serum Levels in Humans: Preliminary Results from a Randomized, Placebo-Controlled, Cross-Over Study
Previous Article in Journal
A Metabolomics Approach to the Identification of Urinary Biomarkers of Pea Intake
Previous Article in Special Issue
Alcohol Pattern Consumption Differently Affects the Efficiency of Macrophage Reverse Cholesterol Transport in Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dietary Composition and Cardiovascular Risk: A Mediator or a Bystander?

Second Department of Internal Medicine and Research Institute, University General Hospital Attikon, 124 62 Haidari, Greece
*
Author to whom correspondence should be addressed.
Nutrients 2018, 10(12), 1912; https://doi.org/10.3390/nu10121912
Submission received: 29 October 2018 / Revised: 28 November 2018 / Accepted: 29 November 2018 / Published: 4 December 2018
(This article belongs to the Special Issue Nutrients and Atherosclerosis)

Abstract

:
The role of nutrition in the pathogenesis of cardiovascular disease has long been debated. The established notion of the deleterious effects of fat is recently under question, with numerous studies demonstrating the benefits of low-carbohydrate, high-fat diets in terms of obesity, diabetes, dyslipidemia, and metabolic derangement. Monounsaturated and polyunsaturated fatty acids, especially n-3 PUFAs (polyunsaturated fatty acids), are the types of fat that favor metabolic markers and are key components of the Mediterranean Diet, which is considered an ideal dietary pattern with great cardioprotective effects. Except for macronutrients, however, micronutrients like polyphenols, carotenoids, and vitamins act on molecular pathways that affect oxidative stress, endothelial function, and lipid and glucose homeostasis. In relation to these metabolic markers, the human gut microbiome is constantly revealed, with its composition being altered by even small dietary changes and different microbial populations being associated with adverse cardiovascular outcomes, thus becoming the target for potential new treatment interventions. This review aims to present the most recent data concerning different dietary patterns at both the macro- and micronutrient level and their association with atherosclerosis, obesity, and other risk factors for cardiovascular disease.

1. Introduction

Cardiovascular disease (CVD) is the most common cause of death in Westernized societies, being responsible for one of every three deaths in the United States and one of every four deaths in Europe. The prevalence of obesity, type 2 diabetes mellitus (T2DM), and metabolic syndrome (MetS) may be the most common risk factors for it and has increased dramatically in recent years. According to the World Health Organization (WHO), the global prevalence of diabetes in adults has increased from 4.7% in 1980 to 8.5% in 2014, whereas worldwide obesity has almost doubled since the 1980s [1].
The role of nutrition in terms of reducing calorie intake and therefore body weight so as to prevent disease and lengthen life span has been well known since the 1930s, with the scientific society (along with popular belief) insisting specifically on the reduction of fat consumption. The Seven Countries Study was the first to demonstrate a clear link between the consumption of fat and the risk for cardiovascular events and set specific dietary rules for decades [2,3]. On the other hand, results from recent large randomized controlled trials (RCTs), like the PURE study, have raised serious questions as to what the ideal nutritional regimen is and to how each type of macronutrient, namely carbohydrates, proteins, and fats, affect body weight, glycaemic control, oxidative stress, inflammation markers, and other predisposing factors for CVD.
A notion that seems to prevail, however, is the beneficial role of the Mediterranean Diet (Med Diet) in the prevention of CVD. This benefit derives from the high consumption of fruits, vegetables, nuts, fish, poultry, and olive oil that this diet entails, together with a small quantity of red meat, emphasizing the importance of low-fat (LF) animal protein sources, complex plant-based carbohydrates, and high fiber content, as well as mono- and polyunsaturated lipids [4]. The majority of these foods contain an increased content of micronutrients that have anti-inflammatory capacities, decrease the oxidization of lipoproteins, and have a favorable effect on oxidative stress in general. Many studies have demonstrated the correlation between the Mediterranean Diet and every aspect of cardiovascular disease and its predisposing factors. The exact mechanism of how these micronutrients work is constantly unraveled.
In addition, it is now known that gut microflora plays a significant role in human health. Changes in the composition of gut microbiota associated with disease, namely, dysbiosis, have been related to cardiometabolic derangement such as atherosclerosis and insulin resistance [5]. Hence, the effects of dietary patterns on the composition of intestinal microflora and the pathways through which different strains influence the main risk factors for CVD are in the spotlight and have emerged as possible treatment choices for the general population.
The objective of this review is to present and discuss the latest data regarding dietary patterns and their effect on cardiovascular disease through the compilation of different studies that have been published in recent years. The physiologic pathways through which macro- and micronutrients affect metabolic health are presented, along with the effect of dietary composition on gut microbiome and how this is connected to different cardiometabolic outcomes.

2. From Physiology to Disease: A Briefing on the Main CVD Risk Factors

A variety of factors have been associated with cardiovascular disease, such as metabolic syndrome, obesity, smoking, diabetes, hyperlipidemia, family history, hypertension, exercise, and more. The main risk factors and a summary of the pathophysiological mechanisms that lead to cardiovascular disease are described below.

2.1. Obesity

Obesity has risen sharply in the past decades. Worldwide, the proportion of adults with a body mass index (BMI) of 25 kg/m2 or greater increased between 1980 and 2013 from 28.8% to 36.9% in men and from 29.8% to 38% in women [6]. However, BMI estimates overall adiposity; it is also the distribution of body fat that counts. Fat accumulation in visceral adipose tissue leads to an influx of macrophages, the production of proinflammatory cytokines, dysregulated levels of adipokines, insulin resistance, and atherogenesis, as adiponectin can no longer suppress foam cell formation. In addition, accumulation of lipids within muscle cells and hepatocytes leads to further insulin resistance and abnormal glucose uptake [7]. It should be noted, however, that the optimal BMI with respect to mortality is yet to be defined, as even lean individuals can be insulin resistant, and obese persons can have no manifestations of metabolic syndrome. According to a meta-analysis [8], mortality was lowest at BMI 22.5–25.0 kg/m2. When adjusted for other traits, such as waist circumference or MetS, BMI is a much weaker predictor for CVD risk [9].

2.2. Insulin Resistance and Diabetes

As we mentioned above, insulin resistance can severely alter adipokine levels. It decreases the production of nitric oxide through inhibition of the phosphoinositol-3 kinase (PI3-K) pathway, thus resulting in endothelial dysfunction. In addition, it favors the secretion of endothelin-1, which leads to vasoconstriction and vascular smooth muscle cell (VSMC) proliferation. Insulin resistance also leads to reduced apoptosis of macrophage cells in atherosclerotic lesions, which eventually leads to plaque rupture [10]. Contrary to obesity, insulin resistance (and the consequent hyperinsulinemia) is an independent risk factor for cardiovascular disease, with a one-unit increase in the Homeostasis Model Assessment (HOMA-IR, which quantifies insulin resistance) being associated with a 5.4% increase in CVD risk [11].

2.3. Hyperlipidemia

Hyperlipidemia is characterized by the change in the lipid concentrations in plasma with the accumulation of one or more classes of lipoproteins. The major categories of lipoproteins consist of low-density lipoprotein (LDL), very-low-density lipoprotein (VLDL), high-density lipoprotein (HDL), and chylomicrons. They contain multiple proteins, called apolipoproteins, with the main one of LDL and VLDL being apoB-100, which is considered extremely atherogenic. VLDL also contains apoC and apoE. LDL has long been established as a major risk factor for CVD [12], and the mechanisms that lead to atherosclerosis are well described [13]. As LDL particles filter into the arterial intima, they are attacked by macrophages which are then transformed to foam cells. Accumulation of foam cells gives birth to fatty streaks. As foam cells die and smooth muscle cells produce fibrous connective tissue, the fatty streak turns into a fibrous plaque (fibroatheroma), which eventually becomes unstable and prone to rupture after years. Lp(a) is a complex lipoprotein that consists of an LDL and an apoB-100 particle linked to the plasminogen-like apolipoprotein (a) and is considered an even more atherogenic particle than LDL-C (low-density lipoprotein cholesterol). Lp(a) 3.5-fold higher than normal increases the risk for CVD events, particularly when LDL-C is more than 130 mg/dL [14], through mechanisms such as platelet aggregation, alteration of fibrin clot structure, and endothelial dysfunction. HDL-C (high-density lipoprotein cholesterol) contains the apolipoproteins apoA1 and apoA2 and is associated with favorable effects on CVD [15]. Hypertriglyceridemia occurs when the concentration of the triglyceride-rich VLDL particles is increased, along with a reduction in HDL-C. It is the result of the inhibition of lipoprotein lipase (LPL), which breaks down triglycerides so that free fatty acids (FAs) can be used by adipose tissue and muscles [16]. On fatty acid levels, the consumption of saturated fatty acids (SFA) increases inflammation by promoting the production of the proinflammatory type M2 macrophages and inhibiting the production of the anti-inflammatory type M1 macrophages. On the other hand, monounsaturated (MUFA) and polyunsaturated fatty acids (PUFA) ameliorate glucose tolerance and increase the levels of adiponectine, which is associated with higher HDL-C and lower triglycerides [17].

2.4. Oxidative Stress

Oxidative stress is a state of imbalance between oxidants and antioxidants in favor of the oxidants. Its association with vascular CVD risk has been established in many studies [18]. Oxidants, also called reactive oxygen species (ROS), include free radicals such as superoxide and peroxynitrite and nonradicals such as hydrogen peroxide. Their sources include enzymes like myeloperoxidases, uncoupled nitric oxide synthase (NOS), peroxidases and NADPH oxidase. Antioxidants include enzymes such as superoxide dismutase and catalase, nonenzyme molecules such as glutathione, and micronutrients [19]. Oxidative stress and inflammation are closely interrelated, and many studies have shown the association of inflammatory markers like high-sensitive CRP (hsCRP) and interleukin-6 (IL-6) with adverse metabolic outcomes [20]. The detrimental effects of oxidative stress in various pathways that are related to diabetes, obesity, endothelial dysfunction, atherosclerosis, hypertension, and generally every predisposing factor for cardiovascular disease are numerous, and their description is beyond the scope of this review. An excellent example of them is the oxidized LDL (ox-LDL), which is the result of the interaction of LDL particles with free radicals and is considered highly atherogenic. In endothelial cells, the main ox-LDL receptor is the LOX-1. TNF-α, adhesion molecules VCAM-1 and ICAM-1, ROS generated from NADPH oxidase, and other proinflammatory molecules increase the production and expression of LOX-1; on the contrary, the suppression of the inflammatory NF-kB pathway downregulates its expression, thus limiting the formation of foam cells and preventing atherosclerosis [21].

3. Low-Carbohydrate Diets vs. Low-Fat Diets: A Long-Standing Debate about the Role of Macronutrients on CVD

The Seven Countries Study [2], beginning in 1956 and originally published in 1978, was the first epidemiological study that examined the relationship between diet and cardiovascular disease in different populations and parts of the world. Its results consolidated the belief that higher fat intake (and especially high saturated fatty acids) was directly associated with higher prevalence of CVD, despite the existence of several randomized trial and studies throughout these decades that did not support this conclusion [22,23,24,25,26]. For example, the Nurses’ Health Study, originally established in 1976, showed an inverse association between total fat and total mortality, along with a higher risk of ischemic stroke due to a higher glycemic load [22]. Similar results were obtained from the Health Professionals Follow-up study (1985–2008) [23,24]. However, it was not until recently, with the publication of the Prospective Urban Rural Epidemiology (PURE) study in 2017, that the common belief about the adverse role of fat was again questioned.
In alignment with established notions, the 2016 European Society of Cardiology (ESC) guidelines has recommended a total fat intake of below 30%, of which 10% should only consist of saturated fats. Moreover, substitution of saturated fat with polyunsaturated fatty acids is strongly emphasized [27]. On top of that, in June 2017, the American Heart Association’s (AHA) presidential advisory on the lipid content of nutrition concluded that replacing saturated fat with polyunsaturated vegetable oil can reduce CVD by 30% [28,29].
The PURE study has questioned the guidance relating to fat consumption when it assessed the association between consumption of carbohydrate, total fat, and each type of fat with cardiovascular disease and total mortality. The study was a very large epidemiological one, with over 135,000 patients enrolled from 18 different countries of different income statuses across 5 continents. Patients were followed up for a median of 7.4 years. Outcomes included total mortality and major adverse cardiovascular events (MACEs) (CVD death, nonfatal MI, stroke, and heart failure). Higher carbohydrate intake was associated with an increased risk of total mortality but not with the risk of cardiovascular disease or cardiovascular disease mortality. On the contrary, fat intake was associated with a decreased risk of total mortality, regardless of the type of fat examined, and saturated fat intake was associated with a lower risk of stroke and total mortality [30]. A cross-sectional analysis of the PURE study also demonstrated that total fat intake, regardless of type, was associated with higher total cholesterol (TC) and LDL cholesterol, higher HDL cholesterol and apolipoprotein A1 (ApoA1), lower total:HDL cholesterol ratio, lower triglycerides:HDL ratio, lower triglycerides, and lower ApoB:ApoA1 ratio. The replacement of saturated fatty acids with carbohydrates or monounsaturated or polyunsaturated fats improved blood pressure, while surprisingly, the replacement of saturated fats with unsaturated fats had mixed results on the lipid profile of the patients [31].
In conclusion, the PURE study showed that total fat intake had at least neutral or even beneficial effects on cardiovascular health, while a high carbohydrate intake (>60% of diet) did not have a positive impact on atherosclerosis and CVD. It also proved that mortality is increased when refined carbohydrates replace saturated fat in an attempt to minimize fat consumption. Finally, the study emphasizes that the largest benefit in mortality is observed when refined carbohydrates are reduced and replaced by polyunsaturated fat [32]. Nonetheless, the study has drawn serious criticism in some points. The fact that most study participants came from low-income countries makes poverty and undernutrition a serious confounding factor regarding mortality. In addition, the reliability of dietary intake data has been questioned, as in China, for instance, other surveys have shown an average intake of around 30% of daily calories from fat, compared to 17.7% in PURE. Finally, the macronutrient analysis did not qualify the type of dietary carbohydrate as either simple (with low glycemic index) or complex (high in fiber and low glycemic index), which significantly differ in terms of incidence of cardiovascular events [33].
Low-carb, high-fat (LCHF) diets have proved to be at least as effective as low-fat, high-carb (LFHC) diets in weight loss [34,35,36]. A randomized controlled trial by Bazzano et al., in 2014 indicated that the LCHF diet group lost more weight (−5.3 kg compared to −1.8 kg) and had a 1.3% decrease in % body fat compared with a 0.3% gain in the LFHC group [37], a finding that is supported in other trials with both diabetic and nondiabetic patients [38,39,40,41]. Similarly, Shai et al., showed that after 24 months of follow-up, the LCHF diet group, despite being the only group that ate ad libitum (meaning, without energy restriction), experienced the greatest weight loss (−4.7 kg) compared with an LFHC group (−2.9 kg) and a Mediterranean Diet group (−4.4 kg) [42]. On the other hand, a meta-analysis by Bueno et al., showed no significant difference in weight loss between LCHF and LFHC diets [43]. Hjorth et al., observed that prediabetic and diabetic individuals lost more weight when they were on an LCHF diet, whereas individuals with normoglycemia had better results with a low-fat diet [44].
Many studies have also shown a beneficial impact of LCHF diets on lipid markers. LCHF diets have proved more effective than LFHC ones at lowering triglycerides and ApoB—a risk predictor for coronary artery disease—and increasing HDL cholesterol [45,46,47]. On the other hand, a common argument against LCHF diets is the increase in LDL cholesterol that follows higher fat intake [48]. Tay et al., showed an increase of total cholesterol (+0.7 vs. +0.1 mmol/L) and LDL-C (+0.6 vs. +0.1 mmol/l) in the LCHF group [49], while Hu et al., also documented higher decrease of TC and LDL-C in the LFHC groups [50]. However, what should be taken into account is that the increase in HDL-C by itself is considered highly cardioprotective [51], and that the differences in LDL-C concentrations should be evaluated along with the changes in the LDL particles (the small ones are more atherogenic) [52]. Morgan et al., documented that although the subject’s initial LDL-C phenotype affected the changes in the size of LDL particles, the LCHF diets were associated with greater and less dense LDL particles in general [53]. However, it is true that the increase in LDL remains a serious concern regarding the low-carbohydrate diets which needs to be assessed in more studies in the future.
Finally, a recent study published in the Lancet Public Health has added fuel to the fire, enrolling 15,428 adults from the Atherosclerosis Risk in Communities (ARIC) study, who completed a dietary questionnaire. The researchers investigated the association between the percentage of energy from carbohydrate intake and all-cause mortality. The results suggested that both high- and low-carbohydrate diets were associated with increased mortality, with an optimal carb intake of 50–55%. Higher mortality was observed when carbohydrates were substituted by animal-derived protein and fat, while lower mortality was achieved when protein and fat were derived from plant sources and whole grains [54]. The paper made headlines around the world stating that a low-carb diet will shorten life. However, a lot of criticism has risen, mainly due to the observational nature of the study, the possible confounders like the whole unhealthy lifestyle (i.e., alcohol consumption, environmental toxins, smoking) that could interfere with the results apart from the diet, and the weak associations found that actually are just epidemiological observations that contradict strong data so far [55].

4. Choosing the Right Type of Fat: A Comparison of Fatty Acids

The consumption of different types of fatty acids causes different metabolic changes. As T2DM is a leading predisposing factor for CVD, the effects of fatty acids on insulin metabolism are of profound significance. Many human studies have shown an association between diets rich in saturated fatty acids (SFAs) and poor in polyunsaturated fatty acids (PUFAs) with insulin resistance [56,57,58,59]. Mullner et al., showed that the supplementation of PUFA-rich plants in the daily dietary regimen of diabetic and nondiabetic individuals resulted in a significant reduction of HbA1c [60], and similar were the results in a study by Lee et al., where subjects with T2DM or metabolic syndrome were provided with capsules of fish oil compared to other types of oil. In diabetics, the supplementation of 3 g of alfa-linolenic acid (ALA) per day resulted in improved insulin sensitivity, as did the supplementation of conjugated linoleic acid (CLA), a group of isomers of linoleic acid rich in PUFAs, in obese children. Among the SFAs, palmitic acid is thought to have the most detrimental effect on pancreatic β-cell function [61]. Koska et al., showed that a diet rich in SFAs resulted in higher glucose and insulin concentration after 24 h compared to a diet where saturated fats comprised only 5% of total calories ingested [62]. As for animal studies, Malinska et al., found that the supplementation of hypertriglyceridemia-induced dyslipidemic rats with CLA resulted in enhanced insulin sensitivity [63]. An in vitro study in mice showed that the administration of palmitate induced insulin resistance in cardiovascular cells, contrary to the favorable effect of oleate [64]. MUFAs also exhibit a beneficial role regarding glucose metabolism, being associated with an important decrease in HbA1c levels, while stimulating GLP-1 secretion, at least in animal models [65].
Regarding dyslipidemia, SFAs have a strong atherogenic effect due to the reduction in the expression of the LDL receptor gene, thus increasing the concentration of LDL in circulation [66]. On the contrary, PUFAs are considered to ameliorate lipid markers, with n-3 PUFA consumption resulting in reduction of plasma triacylglycerols and ApoB-100, which in turn reduces the concentration of VLDL and LDL [67]. Docosahexanoic acid (DHA)—the main n-3 PUFA together with eicosapentanoic acid (EPA)—improves cellular membrane fluids, an action that results in the removal of lipids and inflammatory agents from the arterial wall [68], while both these n-3 PUFAs have been found to decrease the expression of sterol regulatory element-binding protein-1c (SREBP-1c), hence reducing the production of VLDL and triacylglycerols (TAGs) [69]. EPA and DHA have also been shown to increase HDL when supplemented in various daily doses in humans, which, however, exceed the normal daily intake [70,71]. In an RCT where patients with impaired glucose metabolism and coronary artery disease were supplemented with 1800 mg/day EPA for six months, postprandial hypertriglyceridemia, hyperglycemia, and insulin secretion were ameliorated compared to placebo [72]. The REDUCE-IT trial showed that the supplementation of 2 g per day of icosapent ethyl, a highly purified eicosapentaenoic acid ethyl ester, led to a reduction in ischemic events, including cardiovascular death, compared to placebo [73]. Unlike these, n-6 PUFAs have not been found to have such beneficial effects on lipid markers universally, according to several studies with rodents [74,75]. Regarding MUFAs, the data are more limited, but in vivo studies in Wistar rats like those of Macri et al., and Alsina et al., found that olive oil and fish oil, rich in MUFAs, were associated with a decrease in TC and LDL and better fat distribution in tissues [76,77]. Contrary to the aforementioned findings, however, the VITAL trial, an RCT which examined the results of the supplementation of both vitamin D3 (at a dose of 2000 IU per day) and n-3 fatty acids (at a dose of 1 g per day) among adults in the United States, showed no statistically significant difference in the incidence of cardiovascular events compared to the placebo group. [78]
Fatty acids also intervene with inflammation markers which provoke oxidative stress. A study in adult individuals which compared four diets with different FA composition found that n-3 PUFAs decreased postprandial endotoxin levels, unlike the n-6 ones [79]. Matsumoto et al., showed that, in ApoE−/− mice, supplementation of EPA resulted in amelioration of atherosclerotic lesions with less macrophage infiltration and more collagen concentration [80]. Similarly, Simopoulos et al., showed that n-6 PUFAs increase cellular triglycerides and the permeability of the membrane, thus increasing adipose tissue fat, which has strong inflammatory capacities [81]. In another study in obese adults, a diet rich in palmitic acid, compared to a diet rich in oleic acid, decreased the concentrations of the inflammatory cytokines IL-1B, IL-10, IL-18, and TNF-α [82]. In agreement with these findings in human studies, Moya-Perez et al., showed that high-fat diets increase the infiltration of lymphocytes B in rats, which in turn increases the release of proinflammatory macrophages, IL-8, and IFN-γ cytokines [83]. A higher size of adipocyte cells has also been observed with high-fat diets in rats, leading to an increase of NF-γ, TNF-α, and other inflammatory markers [84].

5. Mediterranean Diet: The Golden Standard against Cardiovascular Disease?

Mediterranean Diet is the dietary pattern of countries around the Mediterranean Sea. Despite the relatively high fat intake (35–45%), it has long been considered to be one of the most cardioprotective diets because of its abundance in unsaturated fats and other micronutrients with a beneficial impact on CVD risk factors. It comprises a high intake of olive oil, fruits, vegetables, legumes, nuts, and cereals; a moderate intake of fish and poultry; low consumption of red meat and sweets; and a moderate consumption of wine, mainly with meals [4]. The Lyon Diet Heart Study was the first secondary prevention trial that found that adherence to this dietary pattern was associated with a reduced rate of recurrence after a first myocardial infarction [85], and since then, a number of observational studies or randomized controlled trials have favored these cardioprotective effects [86,87,88,89,90].
One landmark study is the PREDIMED (Prevención con Dieta Mediterránea) study, which took place in Spain. Seven thousand four hundred and forty-seven participants at high cardiovascular risk, but with no cardiovascular disease at enrollment, were divided in three groups according to the dietary pattern that they had to follow: a Mediterranean Diet supplemented with extra-virgin olive oil, a Mediterranean Diet supplemented with nuts, and a control low-fat intake group. The median follow-up was about 4.8 years. The study found that the groups adherent to the Mediterranean Diet had a relative risk reduction of about 30% for major cardiovascular events, namely, myocardial infarction, stroke, and death from cardiovascular causes [91]. In addition, a cross-sectional analysis of a subgroup of the participants in the PREDIMED by Salas-Salvadó et al., demonstrated that after a follow-up of four years, diabetes incidence was 10.1%, 11%, and 17.9% in the extra-virgin oil group, the nuts group, and the control group, respectively, while when the two Med Diet groups were compared together with the control group, diabetes incidence was reduced by 52% in total [92]. Decreases in blood pressure and the total cholesterol:HDL-C ratio and waist circumference were also pointed out.
However, in June, 2017, concerns were raised about the implausibility of the distribution of baseline data in the original primary publication of PREDIMED. After an investigation, the New England Journal of Medicine retracted and republished the primary PREDIMED paper with a modified analysis to account for protocol deviations and departures from individual randomization in a subsample of patients during the trial [93].
Again in favor of the Mediterranean Diet, Tektonidis et al., found that, after a 10-year follow-up, higher adherence to a Mediterranean Diet was associated with a statistically significant reduction of 26%, 21%, and 22% risk for myocardial infarction, heart failure, and ischemic stroke, respectively, while no association was found regarding the risk of hemorrhagic stroke [94]. These results were consistent with results from another cohort study of 30,000 participants in the United States [95]. In a report from the CORDIOPREV study, the endothelial function of 805 participants was measured using ultrasonography of the brachial artery to calculate flow mediated vasodilatation before and after 1.5 years of adherence to a Mediterranean dietary pattern or low-fat diet. Results found that Med Diet improved the flow-mediated dilatation (FMD) (and, thus, endothelial function) in patients with diabetes and prediabetes [96]. The CARDIA study showed that the consumption of fruits and vegetables at a rate of seven to nine servings per day in early adulthood had an inverse association with the prevalence of coronary artery calcium after 20 years of follow-up [97]. In the Multiethnic Study of Atherosclerosis (MESA), a higher Med Diet score was associated with better left ventricular (LV) structure and function, which was attributed to a higher LV volume, higher ejection fraction, and higher stroke volume, despite some increase in LV mass [98]. In addition, the higher Med Diet score was associated with less visceral and peripheral fat (but not subcutaneous fat) and less hepatic steatosis [99]. Finally, the recently published EPIC study, which used nine dietary components of the Med Diet (score 0–18), found an inverse association of Med Diet with risk of developing T2DM [100].
The positive effects of Mediterranean Diet cannot be attributed specifically to every single constituent of it. It seems that it is the combination of the different nutrients that is responsible for its metabolic benefits. Olive oil, having oleic acid, a MUFA, as its major component, is highly effective in decreasing the oxidization of LDL [101]. Wine’s favorable effects on HDL-C, waist circumference, blood pressure, and hyperglycemia have been shown in several studies [102,103], in accordance with the PREDIMED study, where moderate wine intake was associated with a 44% reduction in the prevalence of MetS compared with abstainers. Studies on fish, and especially fatty fish which are rich in n-3 PUFAs, have shown a positive effect on body weight, blood pressure, glucose homeostasis, as well as lipid markers, an effect that possibly has to do with changes in HDL particles [104,105,106]. A meta-analysis of six prospective studies showed that nut consumption protects from coronary heart disease and reduces the risk for development of T2DM [107], while another meta-analysis of 49 RCTs reported improvements in at least one criterion of MetS with higher nut consumption [108]. Finally, dairy products, and especially low-fat ones, have been associated with reduced risk of MetS [109], a finding that was repeated in a recent meta-analysis of 2016, where high consumption led to a 15% reduction in risk [110]. Other meta-analyses have reported an inverse association of dairy consumption with T2DM risk [111,112,113]. However, all these data refer mainly to low-fat products and yoghurt, with cheese (especially yellow cheese) having rather controversial effects on metabolic health. Interestingly, recent data imply that dairy consumption and in particular odd-chain saturated fatty acids, like pentadecanoic acid (C15:0) and heptadecanoic acid (17:0), may have a beneficial effect on beta-cell function and in diabetes prevention [114].
In recent years, some alternative dietary regimens to the Mediterranean Diet have emerged. The Dietary Approaches to Stop Hypertension (DASH) Diet has been created by the US-based National Heart, Lung, and Blood Institute of Health, and enjoys high approval both in the United States and worldwide. It consists of fruits, vegetables, whole grains, poultry, fish, nuts, and minimal consumption of red meat, sweets, and sugar-sweetened beverages. The main differences with the Med Diet have to do with the higher content in fat in the latter, through the consumption of olive oil and the higher consumption of dairy products in the former, along with a limitation in sodium intake. The DASH Diet has proved to be effective mainly in decreasing blood pressure [115,116], and a subanalysis in the MESA study has also shown favorable effects on left ventricular function in terms of a higher end-diastolic volume and a (marginally significant) increase in ejection fraction [117]. However, data regarding the association of the DASH Diet and MetS are rather scarce. Another dietary pattern that has recently been proposed is the Nordic Diet, due to poor adherence of people in Nordic countries to the Mediterranean Diet. It includes high intake of fish, apples, cabbages, root vegetables, pears, and rapeseed oil. In an RCT of 200 individuals with MetS, adherence to the Nordic diet resulted in amelioration of lipid markers [118], while in a Danish study by Hansen et al., high adherence to the Nordic Diet was associated with a 14% lower risk for ischemic stroke compared to low adherence, according to a Healthy Nordic Food Index score [119]. Rapeseed oil is probably the most highlighted component of this diet, as it is an excellent source of fat due to its high concentration in mono- and polyunsaturated fatty acids and to its lowest omega-6:omega-3 ratio of the commonly used vegetable oils.
In accordance with the aforementioned results, a recent systematic review of 14 studies and a total of 188,470 participants compared the Mediterranean, DASH, vegetarian, and Paleolithic diets for primary prevention of heart failure and demonstrated that both the Mediterranean and DASH diets exert a protective effect on the incidence of heart failure and of cardiac function parameters’ deterioration rate [120]. Another recent systematic review included 12 studies and 4201 participants and examined the effect of several dietary patterns on the secondary prevention of heart failure. The results favored the DASH Diet as the beneficiary for the secondary prevention of heart failure, with the Mediterranean Diet exhibiting a positive but less robust correlation with some factors of secondary prevention [121].

6. Micronutrients: Functional Patterns and Benefits in Metabolism and Cardiovascular Disease

Micronutrients are essential elements that are required in very small quantities in the human organism, estimated at milligrams per day, and have a vast array of biochemical functions that regulate metabolism and homeostasis in general. As diabetes, atherosclerosis, and metabolic syndrome are associated with chronic systemic inflammation and oxidative stress, micronutrients have drawn popular interest due to their antioxidant capacities. By intervening in various molecular pathways, they are potent scavengers of ROS or they can limit the generation of free radicals and inflammatory agents [122]. The main micronutrients and their effect on metabolic health are presented below.

6.1. Polyphenols

Polyphenols are the most abundant antioxidants in diet and, in general, can be found mainly in fruits, vegetables, green tea, red wine, nuts, spices, and extra-virgin olive oil. Flavonoids are the most common polyphenols, which in turn are divided in six subclasses: flavanols, flavones, flavanones, anthocyanins, flavonols, and isoflavones. Polyphenols have been associated with an inverse relationship with T2DM, as they inhibit α-glucosidase and α-amylase and increase GIP and GLP-1 peptides, thus resulting in a better glucose homeostasis, as it was shown both in a small study in which humans who consumed caffeinated and decaffeinated coffee in a usual dose were compared [123], and in diabetic mice where high doses of resveratrol (a polyphenol analyzed below) were supplemented for weeks [124,125]. Phenols like tannic and chlorogenic acids can interact with SGLT-1 and SGLT-2 co-transporters and thus inhibit glucose absorption [126]. Beneficial effects have also been mentioned regarding stroke. Diets rich in anthocyanins in rats have been associated with better neuroprotection after stroke and reduced cerebral ischemia and oxidative stress in both human and animal models [127,128]. Epigallocatechin-3-gallate (EGCG), a polyphenol in green tea, also proved to have neuroprotective impact in cases of cerebral ischemia in rats in vivo [129], a finding that applied also to quercetin, which protected the integrity of the blood-brain barrier in rats where it was administered intraperitoneally by reducing levels of matrix metallopeptidase 9 (MMP-9) [130].
Resveratrol is a flavonoid that is mainly found in red wine. In some in vivo studies in rats, resveratrol has been found to reduce plasma triglycerides and LDL-C and to increase HDL-C [131]. It decreases the oxidization of LDL particles and increases the expression of LDL receptors in hepatocytes in vitro [132]. By activating SIRT-1, eNOS, and Nrf2 pathways, it decreases the concentration of TNF-α, a major inflammatory cytokine [133]. It also decreases the expression of adhesion molecules ICAM-1 and VCAM-1 via inhibition of the NF-κB pathway activation [134] and reduces the formation of foam cells via inhibition of NADPH oxidase-1 in mouse macrophages [135]. The cumulative result of the aforementioned actions is a reduction of vascular inflammation and enhanced endothelial function. In studies with diabetic patients [136], healthy obese men [137], and healthy adult smokers [138], resveratrol showed beneficial effects on the lipid profile of the subjects, yet this result was achieved through the supplementation of high doses of resveratrol (250–1000, 150, and 500 mg/day, respectively), which cannot be achieved by normal daily food intake. As for hypertension, resveratrol increases the bioavailability of nitric oxide (NO) in human umbilical vein endothelial cells (HUVECs) in vitro [139] as well as its production in animal models [140], while in clinical studies, antihypertensive results have been shown only in high concentrations [141]. In some studies in rodents, pretreatment with resveratrol after a myocardial infarction decreased infarct size and arrhythmias [142,143]. In general, the low availability of resveratrol due to its rapid metabolism seems to limit its clinical potential.
Luteolin is a flavonoid present in many medicinal plants and in vegetables such as celery and parsley. Many studies have demonstrated its antioxidant and anti-inflammatory capacities [144,145]. It inhibits oxidization of LDL and expression of VCAM-1, while it decreases plasma lipids and benefits vascular dilatation through enhanced expression of eNOS gene [146,147]. These results were demonstrated both in in vivo and in vitro studies with humans or rodents. The main concern regarding many of these findings was that they took place in pharmacological (>10 μΜ) rather than physiological (<2 μΜ) concentrations of luteolin. However, a recent study in mice and in human endothelial cells by Zhenquan et al., showed that, even in low doses, luteolin inhibited TNF-α-induced binding of monocytes to endothelial cells, activation of NF-κB pathway signaling, vascular inflammation, and changes in the intima layer of the aorta, thus confirming its cardioprotective role regardless of its levels [148].
Quercetin is a flavonol that can be found in many vegetables, like red onions, capers, and kale. As mentioned above, it has a beneficial role in stroke and also improves lipidemic profile. In vitro studies have shown that quercetin increases the expression of PPAR-γ receptors and ATP-binding cassette transporter (ABCA1), which leads to reduced formation of foam cells [149]. It also reduces oxidative stress. In ApoE-knockout mice, quercetin reduced hydrogen peroxide and leukotrien B4 in vessels and increased endothelial nitric oxide synthase (eNOS) [150]. Kim et al., found that quercetin, through activating LKB1-AMPK signaling pathway, inhibited myosin light chain kinase (MLCK) and phosphorylated myosin light chain (p-MLC), hence inhibiting vascular smooth muscle cell (VSMC) contraction in rats [151]. In vitro studies in mice have indicated that quercetin promotes glucose transport within adipose and muscle cells through GLUT4 transporters’ translocation and the AMP-activated protein kinase pathway, favoring normoglycemia [152]. Though it is true that most studies regarding quercetin have been conducted in animal models, there are also human studies that support its beneficial role. Obese subjects aged 25–65 years with metabolic syndrome were randomized to receive 150 mg quercetin/day for six-week treatment times. Compared to placebo, quercetin reduced systolic blood pressure by 2.6 mmHg in the entire group, by 2.9 mmHg in the subgroup of hypertensive patients, and by 3.7 mmHg in the subgroup of younger adults (25–50-years old), as well as reducing the total concentration of HDL-C and oxidized LDL [153]. In another study by Lee et al. [154], quercetin-rich supplements based on onion peel extract given to adult subjects for 10 weeks significantly reduced LDL-C, total cholesterol, systolic and diastolic blood pressure, and increased HDL-C. An RCT in humans showed that 730 mg of quercetin/day for four weeks decreased systolic and diastolic blood pressure in stage 1 hypertensive patients [155]. Another trial in diabetic women indicated that 500 mg of quercetin/day for 10 weeks reduced systolic blood pressure [156]. A large meta-analysis by Serban et al., on RCTs about the effect of quercetin on blood pressure reported that quercetin achieved a statistically significant decrease of blood pressure only in doses of >500 mg/day [157]. As it is clear, quercetin’s beneficial effects take place only in pharmacological doses that highly exceed normal daily food intake, taking into account that in the United States, for example, the daily intake of quercetin is about 10 mg.
Curcumin is a flavonoid that is mainly found in turmeric, curry spice, and ginger. Its role in improving oxidative stress has been marked in many studies in animal models, all of which attribute this capacity to the inhibition of the TLR4 signaling pathway [158,159]. Studies in ApoE−/− mice indicate that supplementation with curcumin leads to less macrophage infiltration in atherosclerosis plaque, reduced aortic NF-κB activation, reduced levels of IL-1B and TNF-α, and reduced expression of the adhesion molecules ICAM-1 and VCAM-1 [160].

6.2. Carotenoids

Carotenoids are lipophylic antioxidants that are found in plants and some photosynthetic bacteria and fungi. They are classified into carotenes and xanthophylls. Carotenes include beta-carotene and lycopene, and xanthophylls include lutein, fucoxanthin, zeaxanthin, canthaxanthin, astaxanthin, beta-criptoxanthin, and capsorubin [161].
Astaxanthin is found in plankton, fish (mainly salmon—200 g of salmon contains about 0.5–1.25 mg of astaxanthin), and other seafood. It is a strong free radical scavenger and decreases LDL-C and triglycerides, increases HDL-C, and ameliorates inflammation markers both in human and animal models [162,163]. An RCT by Yoshida et al., in nonobese humans aged 20–65 years reported that the supplementation of astaxanthin (0, 6, 12, and 18 mg/day, respectively, in each group) for 12 weeks improved HDL-C and TG, as well as adiponectin levels [164]. Similarly, Iwamoto et al., reported improvements in LDL-C levels after supplementation with astaxanthin (from 1.8 to 21.6 mg/day) for two weeks in healthy volunteers [165]. In another study by Hussein et al., astaxanthin lowered blood pressure in hypertensive rats, an effect that was attributed to modulation of nitric oxide levels [166].
Lycopene is found mainly in tomatoes, watermelons, and red grapefruits. Many studies have associated low plasma levels of lycopene with early carotid atherosclerotic lesions [167]. The Rotterdam Study, a prospective cohort study on adults aged 55 years or older, revealed an inverse association between lycopene and calcified plaques in the abdominal aorta, a result that was more evident in current and former smokers [168], whereas another report by Rissanen et al., in middle-aged adults supports that lycopene has a beneficial impact on carotid intima media thickness and cardiovascular events in general, at least in men [169]. It increases HDL-C, lowers triacylglycerols and ox-LDL [170], and contributes to vasodilatation through modulation of NO. Its relationship with T2DM risk remains controversial.
Lutein is found mainly in dark green vegetables such as spinach, parsley, and broccoli. It is a ROS scavenger and inhibits the NF-κB pathway [171]. It has been proved to decrease TNF-α, IL-6, PGE2, and oxidative stress in general, at least in rodents for which high pharmacological doses of lutein were administered [172]. A diet with high lutein intake is beneficial for atherosclerosis and arterial stiffness, as large studies like the ARIC [173] and the CUDAS [174] have demonstrated.
Beta-carotene is found primarily in carrots, tomatoes, and spinach. It decreases oxidization of LDL [175] and inhibits the NF-κB-induced expression of adhesion molecules in vitro [176]. It also reduces non-HDL cholesterol levels and the expression of IL-1a and VCAM-1 in mice in vitro [177]. In mature adipocytes, beta-carotene is metabolized to retinoic acid, which decreases the expression of PPAR-α and CCAAT/enhancer-binding protein, thus reducing the lipid content of adipocytes [178]. Finally, there seems to exist an inverse association between T2DM and consumption of beta-carotene with diet [179].

6.3. Trace Elements

Although a number of epidemiological studies have marked a selenium deficiency in people with diabetes, RCTs have not managed to prove that supplementation with selenium decreases the risk for T2DM and cardiovascular mortality at all [180]. In a recent systematic review, Sarmento et al., indicated that in diabetic patients, low levels of selenium were associated with an increased risk for CVD, but clearly pointed out the need for more studies in humans to solidify these results [181]. On the contrary, data that show that zinc deficiency is associated with vascular diseases are more stable. Low levels of zinc cause increased oxidative stress in endothelial cells in vitro [182], and they have been associated with increased cardiovascular events in diabetic patients. In a recent study in diabetic mice by Miao et al., supplementation of zinc (5 mg ZnSO4/kg per day for three months) reduced aortic tunica media thickness and collagen accumulation, apoptotic cell death and expression of inflammatory markers VCAM-1 and PAI-1, and increased the expression of Nrf2 and metallothioneins in the aorta, both of which have strong antioxidant properties [183]. It should be noted, however, that this dose does not represent daily human intake, which is about 0.07–0.23 mg/kg/day.

6.4. Vitamins

The data regarding the cardioprotective effects of B-complex vitamins is controversial. However, according to some studies in humans, high intakes of B6, B12, and folic acid may be beneficial for both ischemic and hemorrhagic stroke prevention, possibly due to the reduction of plasma homocysteine levels, and a diet rich in B vitamins can reduce poststroke functional decline [184,185]. Chambers et al., found that supplementation of folic acid and B12 at a dose of 5 and 1 mg per day, respectively, for eight weeks significantly improved endothelial dilatation [186] (the daily intake for adults in the United States is about 500 mcg/day for folic acid and 3.4 mcg/day for B12). Coenzyme Q10 or ubiquinone, a lipid-soluble benzoquinone that acts as a vitamin, has also been associated with favorable cardiovascular effects when supplemented to the normal diet [187,188]. Q10 also serves as an antioxidant, reducing lipid peroxidation and increasing the levels of antioxidant enzymes catalase (CAT) and superoxide dismutase (SOD) in vitro, after a daily treatment with 150 mg for 12 weeks, when its normal daily intake is no more than 3–5 mg [189]. Vitamins C (mainly found in citrus fruits) and E (mainly found in seeds and vegetable oil) are associated with a reduction in coronary artery disease (Law et al.) [190] and have antioxidant and anti-inflammatory properties, but their supplementation beyond the usual dietary intake does not have any proven benefits for humans [191,192]. Vitamin E is also considered to have favorable effects on glucose homeostasis, as it inhibits the formation of advanced glycosylation end products, reduces HbA1c, and prevents oxidative stress in pancreatic β-cells [193,194]. As for vitamin D, the recently published VITAL study (described above) showed that supplementation with vitamin D did not result in a lower incidence of cardiovascular events [195].

7. Gut Microbiota, Diet, and Cardiovascular Disease: A Recently Discovered Field

In recent years, a lot of interest has been drawn to the role of gut microbiota in human health. The gut microbiome encompasses 1014 microorganisms. The bacterial population is composed mainly of five phyla: Firmicutes (60–65%), Bacteroidetes (20–25%), Proteobacteria (5–10%), Actinobacteria (about 3%), and Cerrucomicrobia [196]. These microorganisms participate in the food digestion process through two catabolic pathways. In the saccharolytic pathway, gut microbiota break down sugars and produce most of the short-chain fatty acids (SCFAs), whose byproducts, mainly acetate, butyrate and propionate, are a major source of energy for intestinal epithelium. More specifically, SCFAs signal to the host through four different pathways at least. First, they are an energy substrate for colonocytes. Second, butyrate and acetate act as histone deacetylase inhibitors. Third, propionate can induce intestinal gluconeogenesis, with a favorable effect on glucose tolerance, and fourth, they activate G-protein-coupled receptors like GPR41 and GPR43, triggering the release of GLP-1, which plays a crucial role in glucose homeostasis. Fatty acid oxidation is activated by SCFAs, while de novo synthesis and lipolysis are inhibited, leading to a decreased concentration of free fatty acids in plasma and weight loss [197,198,199]. In the proteolytic pathway, protein fermentation takes place, along with SCFA formation and the production of many other metabolites, some of which are toxic for the host [200]. Primary bile acids, cholic acid (CA), and chenodeoxycholic acid (CDCA) are metabolized into the secondary bile acids deoxycholic acid (DCA) and lithocholic acid (LCA), which signal to the host through G-protein-coupled receptor 1 (also called TGR5) and the bile acid receptor FXR, both of which affect metabolism [201]. On the other hand, microbes produce endotoxins (also called lipopolysaccharides) which promote systemic inflammation and subsequently lead to insulin resistance. Another important pathway is that of the TMAO production. As microbes metabolize phosphatidylcholine and L-carnitine, they produce trimethylamine (TMA), which is then oxidized in the liver to trimethylamine N-oxide (TMAO) that has been associated with atherosclerosis both in humans [202] and mice. It is also well known that that the microbiota contribute both to local intestinal and systemic immunity through their effects on toll-like receptor (TLR) expression, macrophages, T cells, antibodies, and other mechanisms [203,204]. Changes in the composition of the gut microbiota have been associated with dietary changes, even short and acute ones, and have been linked to metabolic syndrome and cardiovascular disease in many aspects that are analyzed below.

7.1. Diet and Gut Microbiota

(a) Proteins. Many studies have shown associations between protein intake and gut microbial diversity [205,206]. In small human studies, whey and pea protein increased the population of the beneficial Bifidobacterium and Lactobacillus species, together with levels of SCFAs [207,208,209]. On the contrary, animal protein increases anaerobes such as Bacteroides and Alistipes [210]. Obese men were given a high protein/low carbohydrate diet for four weeks, which eventually reduced the concentrations of Roseburia and Eubacterium rectale populations in fecal samples with a concomitant reduction in the concentration of butyrate in feces [211], a finding that was confirmed in another study by De Filippo et al. This study compared the fecal microbiota of European children on a high-protein diet to those of children in Burkina Faso, where a high-fiber diet is followed, and demonstrated a lower concentration of fecal SCFAs in the first group [212]. The high intake of red meat as a source of protein has also been related to increased levels of trimethylamine-n-oxide (TMAO), which is an oxidation product of the microbial metabolite TMA and has been shown to have detrimental effects on atherogenesis [213].
(b) Fats. High fat intake increases the population of Bacteroides, Clostridiales, and Enterobacteriales and decreases the counts of the beneficial Lactobacillus intestinalis species, as studies in rats have indicated [214,215]. On the contrary, a low-fat diet increases fecal levels of Bifidobacterium, with positive effects on blood glucose homeostasis and total cholesterol levels. The type of fat consumed is also important. Mice supplemented with fish oil for 11 weeks had increased counts of Lactobacillus and Actinobacteria, while mice supplemented on lard oil had increased counts of Bacteroides and Bilophila, which led to increased TLR activation, impaired insulin sensitivity, and white adipose tissue inflammation [216]. Diets with n-6 PUFAs increase the number of Firmicutes, Actinobacteria, and Proteobacteria strains and decrease that of Bifidobacteria, which are associated with chronic inflammation and metabolic syndrome, mediated by an increase of lipopolysaccharides (LPS) [217].
(c) Carbohydrates. Digestible carbohydrates, which include starch and sugars (glucose, fructose, sucrose, and lactose) increase the number of Bifidobacteria strains and decrease that of Bacteroides in studies in vitro [218]. A study in which infants with cow’s milk allergy received lactose for two months showed that lactose increased SCFAs in fecal samples compared to the placebo group [219]. Nondigestible carbohydrates such as fiber act as prebiotics—nondigestible dietary components that selectively stimulate the growth and/or activity of certain microorganisms, thus affecting the host’s health beneficially [220]. In two studies where adult subjects received a whole-grain breakfast contrary to the placebo group, prebiotics increased Bifidobacteria and Lactobacilli strains and had favorable effects on glucose homeostasis and inflammation markers, such as IL-6 and IL-10 [221,222].
(d) Dairy products. Milk and yoghurt are rich in lactic acid bacteria and are considered probiotics (live microorganisms that, in appropriate amounts, benefit the host’s health) [223]. They increase Bifidobacteria and Lactobacilli population, as has been indicated in various human studies [224,225,226]. In an RCT where overweight adults were supplemented with probiotics, an enhancement in lipid markers and hsCRP was demonstrated [227]. Hanie et al., found that the administration of probiotic yoghurt to diabetic patients decreased fasting blood glucose and HbA1c compared with the control group after a follow-up period of six weeks [228].

7.2. Gut Microbiota and CVD

(a) Atherosclerosis and coronary artery disease: The bacteria that were found in atherosclerotic plaques were also found in the gut and oral cavity of the same individuals in a number of studies, implying that these bacteria could be a source of plaque colonization [229,230]. After sequencing of the gut metagenome in patients with unstable plaques, differences in microbiota composition were observed, associated with lower fecal levels of the genus Roseburiam and higher levels of the genus Collinsella, together with increased oxidative stress [231]. In rodents, increased TMAO levels were associated with an enhanced aortic atherosclerotic plaque [232], and similar findings have emerged in humans. In a large cohort study with over 4000 patients undergoing elective coronary angiography, it was reported that high TMAO levels were associated with higher incidence of CVD events over a three-year follow-up period [202]. Such results have been confirmed in other trials with over 1800 patients undergoing elective coronary angiography, where TMAO levels were associated with higher atherosclerotic plaque size, vulnerable atherosclerotic plaque, and higher all-cause mortality [233,234].
(b) Dyslipidemia: As mentioned above, gut microbiota produce secondary bile acids, which can alter both hepatic and systemic lipid metabolism through FXR and GPR131 [235,236]. Increased TMAO levels have been associated with deleterious effects on the lipid profile, affecting mechanisms such as reverse cholesterol transport and sterol metabolism, as studies in obese mice which underwent vertical sleeve gastrectomy indicated [237].
(c) Heart failure: The gut hypothesis in heart failure claims that the impairment in hemodynamics leads to intestinal mucosal ischemia and edema and increases bacterial translocation and production of endotoxins that exacerbate the already-existing oxidative stress and systemic inflammation associated with heart failure [238]. A confirmation of this hypothesis was provided by Pacini et al., who found increased concentrations of microorganisms in feces of patients with heart failure, which were associated with increased membrane permeability [239]. In a study by Tang et al., the relationship between fasting plasma TMAO and all-cause mortality over a five-year period in stable patients with heart failure compared to healthy controls was examined. Patients had higher levels of TMAO than controls, and these levels were associated with higher mortality risk (hazard ratio: 2.2 after adjustment for other risk factors). The mechanisms that link heart failure to TMAO levels, however, remain to be clarified. [240,241].
(d) Obesity, T2DM, and metabolic syndrome. Several studies have related obesity to an increased ratio of Firmicutes to Bacteroidetes [242,243]. In obese humans, there is decreased abundance in Bacteroidetes compared to lean individuals, and weight loss in these individuals increased their population. Overweight children also show an increased relative abundance of Enterobacteriaceae compared to children with normal BMI [244], while in human and animal models, there has been noticed an increased population of Bifidobacteria in obese subjects. Nevertheless, data about which specific species promote or prevent obesity are still conflicting. An increased population of Firmicutes, especially those of the Ruminococcaceae family, had a favorable impact on insulin sensitivity, plasma acetate and triglycerides, and was associated with positive changes in markers of brown adipocytes in subcutaneous (but not in visceral) fat in a study with diabetic, morbidly obese women and men who underwent elective gastric-bypass surgery [245]. After one year’s adherence to a Mediterranean Diet or an LFHC diet, the abundance of the Roseburia genus and F. prausnitzii, respectively, led to less insulin resistance in obese subjects [246]. A study by Scwiertz et al., which examined the fecal microbiota in normal, overweight, and obese subjects, showed that SCFAs, especially propionate, were increased in the feces of overweight and obese subjects [247]. A 24-week study involving 60 overweight adults where propionate was specifically delivered to the colon through a novel inulin-propionate ester stimulated the release of anorexigenic peptide YY (PYY) and GLP-1 from colonic cells, leading to eventual weight loss [248]. Similar to this result, other studies have shown an association between T2DM and decreased population of butyrate-producing bacteria and an increased abundance of Lactobacillus spp. [249].
(e) Hypertension. Few studies have directly linked the composition of gut microbiota to hypertension, mainly in animal models. Yang et al., showed an increased Firmicutes/Bacteroidetes ratio in hypertensive rats [250]. SCFAs could also play a role through the GPR pathways that impact renin secretion [251]. A recent meta-analysis of nine trials showed that the consumption of probiotics significantly changed systolic BP by −3.56 mmHg and diastolic BP by −2.38 mmHg, changes that were more profound in hypertensive subjects and when the probiotic intervention included many different species and lasted at least eight weeks [252]. However, many further studies are needed to elucidate the association between hypertension and microbiota.

7.3. A Novel Therapeutic Target?

The relationships between gut microbiota, diet, and cardiovascular disease imply that any intervention in the composition of the microbiome could be considered a therapeutic prospect. Currently, diet intervention is the main proposed regimen [253]. The exact dietary changes, however, that need to take place are difficult to be determined. As we mentioned above, even short changes in diet can dramatically affect the composition of the microbiota; the switch between plant- and meat-based diets or the addition of more than 30 g per day of specific fibers in daily dietary regimen, even for 10 days, results in shifts in microbiota composition and function in just over 1–2 days [254]. Contrary to these rapid dynamics, a 10-day feeding study in 10 people did not modulate the major characteristics of their microbiota, implying that long-term dietary habits also play a fundamental role in the synthesis of gut microbiome [255]. Even more intriguingly, a specific change in diet can have variable effects on different people due to the distinct combination of each individual’s gut microbial species, as molecular analyses of recent decades, including The Human Microbiome Project, have made evident [256]. The use of probiotics and prebiotics is on the increase [257,258]. However, changes in microbial populations are often relatively small, and generally persist only for as long as the period of the intervention. The use of antibiotics to change intestinal microflora is not widely accepted, as it is considered to do more harm than good, given the adverse effects that drugs have and the obvious inability to be administered for prolonged periods. Fecal microbiota transplantation (FMT) is a promising new therapeutic procedure for cardiometabolic disorders. In a study where overweight patients were transferred microbiota from healthy controls, hepatic and peripheral insulin sensitivity were improved by more than 100% compared to subjects that were transferred microbiota from their own feces [259]. However, the danger of transferring also harmful endotoxins and other agents with this procedure imposes some limitations to this technique, and more studies are needed to assure its safety [260,261].

8. Conclusions

As cardiovascular diseases are on a constant rise, the role of diet as a mediator gathers more and more interest. Dietary manipulation can reduce cardiovascular disease by 50%. Despite some controversial data and the definite need for more studies to come, dietary patterns that combine a high fat intake with low carbohydrates are no longer out of the question. Different kinds of fatty acids act differently in terms of atherosclerosis, inflammation, and oxidative stress. There is no doubt that ingestion of industrialized transunsaturated fats accelerates atherosclerosis and augments mortality. Saturated fats are also considered a potential macronutrient enemy for the heart and the endothelium while reducing them can mitigate CVD risk. Polyunsaturated fats and especially n-3 PUFAs seem to possess the most favorable potential. The role of the Mediterranean Diet in preventing CVD has been highlighted in recent years by many studies, and it is attributed to the combination of foods and micronutrients that this diet entails. These facts, combined with the new data supporting that different strains in the human gut microbiome lead to different metabolic responses and have various effects on cardiometabolic disorders, make dietary interventions a promising and exciting prospect for the treatment of cardiovascular disease in the future. What is important to remember, however, is that we do not consume macro- or micronutrients solely, but we rather integrate them within a holistic lifestyle pattern, where moderation is probably the key to the best outcome. For these reasons, larger RCTs are needed to actually prove the best individualized dietary approach for each metabolic milieu.

Author Contributions

E.K. and V.L.: assistance with structuring of the review, writing, and literature review; G.D. and A.R.: assistance with writing.

Funding

This research received no external funding.

Conflicts of Interest

The authors report no conflict of interest.

Abbreviations

CVDcardiovascular disease
T2DMtype 2 diabetes mellitus
WHOWorld Health Organization
ESCEuropean Society of Cardiology
AHAAmerican Heart Association
MACEmajor adverse cardiovascular event
MImyocardial infarction
LDLlow-density lipoprotein
HDLhigh-density lipoprotein
ApoA1apolipoprotein A1
ApoBapolipoprotein B
ApoEapolipoprotein E
LCHFlow-carb, high-fat
LFHClow-fat, high-carb
HbA1chemoglobin A1c
SFAsaturated fatty acid
CLAConjugated Linoleic Acid
MUFAmonounsaturated fatty acid
PUFApolyunsaturated fatty acid
GLP-1glucagon-like peptide-1
ApoB-100apolipoprotein B-100
VLDLvery low-density lipoprotein
DHADocosahexanoic acid
EPAeicosapentanoic acid
SREBP-1csterol regulatory element-binding protein-1c
TAGtriacylglycerol
TCtotal cholesterol
TGtriglycerides
FAfatty acid
ILinterleukin
TNF-αtumor necrosis factor-α
IFN-γinterferon-γ
NF-κΒnuclear factor-κΒ
Med-DietMediterranean Diet
FMDflow-mediated dilatation
LVleft ventricle
MetSmetabolic syndrome
RCTrandomized controlled trial
DASHDietary Approaches to Stop Hypertension
EGCGEpigallocatechin-3-gallate
GIPGastric inhibitory polypeptide
SGLTsodium-glucose transport protein
MMP-9metallopeptidase 9
SIRT-1sirtuin-1
eNOSendothelial nitric oxide synthase
Nrf2nuclear factor-like 2
ICAM-1intercellular adhesion molecule-1
VCAMvascular cell adhesion molecule-1
NADPHnicotinamide adenine dinucleotide phosphate
NOnitric oxide
PPAR-γperoxisome proliferator-activated receptor-γ
ABCA1ATP-binding cassette transporter
LKB1liver kinase B1
AMPKAMP-activated protein kinase
MLCKmyosin light chain kinase
p-MLCphosphorylated myosin light chain.
VSMCvascular smooth muscle cells
GLUT-4glucose transporter type 4
AMPadenosine monophosphate
TLR4toll-like receptor 4
ox-LDLoxidized LDL

References

  1. World Health Organization. Obesity and Overweight. 2014. Available online: http://www.who.int/mediacentre/factsheets/fs311/en/.
  2. Keys, A.; Aravanis, C.; Buchem, F.S.P.; Blackburn, H. The diet and all-causes death rate in the Seven Countries Study. Lancet 1981, 2, 58–61. [Google Scholar]
  3. Fung, T.T.; Rexrode, K.M.; Mantzoros, C.S.; Manson, J.E.; Willett, W.C.; Hu, F.B. Mediterranean diet and incidence of and mortality from coronary heart disease and stroke in women. Circulation 2009, 119, 1093–1100. [Google Scholar] [CrossRef] [PubMed]
  4. Willett, W.C.; Sacks, F.; Trichopoulou, A.; Drescher, G.; Ferro-Luzzi, A.; Helsing, E.; Trichopoulos, D. Mediterranean diet pyramid: A cultural model for healthy eating. Am. J. Clin. Nutr. 1995, 61 (Suppl. 6), 1402s–1406s. [Google Scholar] [CrossRef] [PubMed]
  5. Festi, D.; Schiumerini, R.; Eusebi, L.H.; Marasco, G.; Taddia, M.; Colecchia, A. Gut microbiota and metabolic syndrome. World J. Gastroenterol. 2014, 20, 16079–16094. [Google Scholar] [CrossRef] [PubMed]
  6. Ng, M.; Fleming, T.; Robinson, M.; Thomson, B.; Graetz, N.; Margono, C.; Mullany, E.C.; Biryukov, S.; Abbafati, C.; Abera, S.F.; et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014, 384, 766–781. [Google Scholar] [CrossRef]
  7. Van Gaal, L.F.; Mentens, I.L.; De Block, C.E. Mechanisms linking obesity with cardiovascular disease. Nature 2006, 444, 875–880. [Google Scholar] [CrossRef]
  8. Prospective Studies Collaboration. Body-mass index and cause-specific mortality in 900000 adults: Collaborative analyses of 57 prospective studies. Lancet 2009, 373, 1083–1096. [Google Scholar] [CrossRef]
  9. Yusuf, S.; Hawken, S.; Ounpuu, S.; Bautista, L.; Franzosi, M.G.; Commerford, P.; Lang, C.C.; Rumboldt, Z.; Onen, C.L.; Lisheng, L.; et al. Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: A case-control study. Lancet 2005, 366, 1640–1649. [Google Scholar] [CrossRef]
  10. Bansilal, S.; Farkouh, M.E.; Fuster, V. Role of insulin resistance and hyperglycemia in the development of atherosclerosis. Am. J. Cardiol. 2007, 99, 6B–14B. [Google Scholar] [CrossRef]
  11. Hanley, A.J.; Williams, K.; Stern, M.P.; Haffner, S.M. Homeostasis model assessment of insulin resistance in relation to the incidence of cardiovascular disease: The San Antonio Heart Study. Diabetes Care 2002, 25, 1177–1184. [Google Scholar] [CrossRef]
  12. National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation 2002, 106, 3143–3421. [Google Scholar] [CrossRef]
  13. Tabas, I.; García-Cardeña, G.; Owens, G.K. Recent insights into the cellular biology of atherosclerosis. J. Cell Biol. 2015, 209, 13–22. [Google Scholar] [CrossRef] [PubMed]
  14. Bucci, M.; Tana, C.; Giamberardino, M.A.; Cipollone, F. Lp(a) and cardiovascular risk: Investigating the hidden side of the moon. Nutr. Metab. Cardiovasc. Dis. 2016, 26, 980–986. [Google Scholar] [CrossRef] [PubMed]
  15. Rosenson, R.S.; Brewer Jr, H.B.; Ansell, B.; Barter, P.; Chapman, M.J.; Heinecke, J.W.; Kontush, A.; Tall, A.R.; Webb, N.R. Translation of high-density lipoprotein function into clinical practice: Current prospects and future challenges. Circulation 2013, 128, 1256–1267. [Google Scholar] [CrossRef] [PubMed]
  16. Tanaka, A.; Ai, M.; Kobayashi, Y.; Tamura, M.; Shimokado, K.; Numano, F. Metabolism of triglyceride-rich lipoproteins and their role in atherosclerosis. Ann. N. Y. Acad. Sci. 2001, 947, 207–212. [Google Scholar] [CrossRef] [PubMed]
  17. Chan, K.L.; Pillon, N.J.; Sivaloganathan, D.M.; Costford, S.R.; Liu, Z.; Théret, M.; Chazaud, B.; Klip, A. Palmitoleate reverses high fat-induced proinflammatory macrophage polarization via amp-activated protein kinase (AMPK). J. Biol. Chem. 2015, 290, 16979–16988. [Google Scholar] [CrossRef] [PubMed]
  18. Griendling, K.K.; FitzGerald, G.A. Oxidative stress and cardiovascular injury part 1: Basic mechanisms and in vivo monitoring of ROS. Circulation 2003, 108, 1912–1916. [Google Scholar] [CrossRef] [PubMed]
  19. Sies, H. Oxidative stress: Oxidants and antioxidants. Exp. Physiol. 1997, 82, 291–295. [Google Scholar] [CrossRef]
  20. Weiner, S.D.; Ahmed, H.N.; Jin, Z.; Cushman, M.; Herrington, D.M.; Nelson, J.C.; Di Tullio, M.R.; Homma, S. Systemic inflammation and brachial artery endothelial function in the Multi-Ethnic Study of Atherosclerosis (MESA). Heart 2014, 100, 862–866. [Google Scholar] [CrossRef]
  21. Ryoo, S.; Bhiunia, A.; Chang, F.; Shoukas, A.; Berkowitz, D.E.; Romer, L.H. Ox-LDL-dependent activation of arginase II is dependent on the LOX-1 receptor and downstream RhoA signalling. Atherosclerosis 2011, 214, 279–287. [Google Scholar] [CrossRef]
  22. Liu, S.; Willett, W.C.; Stampfer, M.J.; Hu, F.B.; Franz, M.; Sampson, L.; Hennekens, C.H.; Manson, J.E. A prospective study of dietary glycemic load, carbohydrate intake, and risk of coronary heart disease in US women. Am. J. Clin. Nutr. 2000, 71, 1455–1461. [Google Scholar] [CrossRef] [PubMed]
  23. Ascherio, A.; Rimm, E.B.; Giovannucci, E.L.; Spiegelman, D.; Stampfer, M.; Willett, W.C. Dietary fat and risk of coronary heart disease in men: Cohort follow up study in the United States. BMJ 1996, 313, 84–90. [Google Scholar] [CrossRef] [PubMed]
  24. Schwingshackl, L.; Hoffmann, G. Dietary fatty acids in the secondary prevention of coronary heart disease: A systematic review, meta-analysis and meta-regression. BMJ Open 2014, 4, e004487. [Google Scholar] [CrossRef] [PubMed]
  25. Hamley, S. The effect of replacing saturated fat with mostly n-6 polyunsaturated fat on coronary heart disease: A meta-analysis of randomised controlled trials. Nutr. J. 2017, 16, 30. [Google Scholar] [CrossRef] [PubMed]
  26. Ramsden, C.E.; Zamora, D.; Majchrzak-Hong, S.; Faurot, K.R.; Broste, S.K.; Frantz, R.P.; Davis, J.M.; Ringel, A.; Suchindran, C.M.; Hibbeln, J.R. Re-evaluation of the traditional diet-heart hypothesis: Analysis of recovered data from Minnesota Coronary Experiment (1968–1973). BMJ 2016, 353, i1246. [Google Scholar] [CrossRef] [PubMed]
  27. Piepoli, M.F.; Hoes, A.W.; Agewall, S.; Albus, C.; Brotons, C.; Catapano, A.L.; Cooney, M.T.; Corra, U.; Cosyns, B.; Deaton, C.; et al. European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts)Developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR). Eur. Heart J. 2016, 37, 2315–2381. [Google Scholar] [PubMed]
  28. Sacks, F.M.; Lichtenstein, A.H.; Wu, J.H.; Appel, L.J.; Creager, M.A.; Kris-Etherton, P.M.; Miller, M.; Rimm, E.B.; Rudel, L.L.; Robinson, J.G.; et al. Dietary Fats and Cardiovascular Disease: A Presidential Advisory From the American Heart Association. Circulation 2017, 136, e1–e23. [Google Scholar] [CrossRef]
  29. Malhotra, A.; Sahdev, N.; Sharma, S. Diet and Nutrition after the PURE study. Eur. Heart J. 2018, 39, 1503–1513. [Google Scholar] [CrossRef]
  30. Dehghan, M.; Mente, A.; Zhang, X.; Swaminathan, S.; Li, W.; Mohan, V.; Iqbal, R.; Kumar, R.; Wentzel-Viljoen, E.; Rosengren, A.; et al. Associations of fats and carbohydrate intake with cardiovascular disease and mortality in 18 countries from five continents (PURE): A prospective cohort study. Lancet 2017, 390, 2050–2062. [Google Scholar] [CrossRef]
  31. Mente, A.; Dehghan, M.; Rangarajan, S.; McQueen, M.; Dagenais, G.; Wielgosz, A.; Lear, S.; Li, W.; Chen, H.; Yi, S.; et al. Association of dietary nutrients with blood lipids and blood pressure in 18 countries: A cross-sectional analysis from the PURE study. Lancet Diabetes Endocrinol. 2017, 10, 774–787. [Google Scholar] [CrossRef]
  32. Li, Y.; Hruby, A.; Bernstein, A.M.; Ley, S.H.; Wang, D.D.; Chiuve, S.E.; Sampson, L.; Rexrode, K.M.; Rimm, E.B.; Willett, W.C.; et al. Saturated Fat as Compared with Unsaturated Fats and Sources of Carbohydrates in Relation to Risk of Coronary Heart Disease: A Prospective Cohort Study. J. Am. Coll. Cardiol. 2015, 66, 1538–1548. [Google Scholar] [CrossRef] [PubMed]
  33. Gianos, E.; Williams, K.A.; Freeman, A.M.; Kris-Etherton, P.; Aggarwal, M. How Pure is PURE? Dietary Lessons Learned and Not Learned from the PURE Trials. Am. J. Med. 2018, 131, 457–458. [Google Scholar] [CrossRef] [PubMed]
  34. Santos, F.L.; Esteves, S.S.; Da Costa, P.A.; da Costa Pereira, A.; Yancy, W.S., Jr.; Nunes, J.P.L. Systematic review and meta-analysis of clinical trials of the effects of low carbohydrate diets on cardiovascular risk factors. Obes. Rev. 2012, 13, 1048–1066. [Google Scholar] [CrossRef] [PubMed]
  35. Tobias, D.K.; Chen, M.; Manson, J.E.; Ludwig, D.S.; Willett, W.; Hu, F.B. Effect of low-fat diet interventions versus other diet interventions on long-term weight change in adults: A systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2015, 3, 968–979. [Google Scholar] [CrossRef]
  36. Mansoor, N.; Vinknes, K.J.; Veierød, M.B.; Retterstøl, K. Effects of low-carbohydrate diets v. low-fat diets on body weight and cardiovascular risk factors: A meta-analysis of randomised controlled trials. Br. J. Nutr. 2016, 115, 466–479. [Google Scholar] [CrossRef] [PubMed]
  37. Bazzano, L.A.; Hu, T.; Reynolds, K.; Yao, L.; Bunol, C.; Liu, Y.; Chen, C.S.; Klag, M.J.; Whelton, P.K.; He, J. Effects of low-carbohydrate and low-fat diets: A randomized trial. Ann. Intern. Med. 2014, 161, 309–318. [Google Scholar] [CrossRef] [PubMed]
  38. Daly, M.E.; Paisey, R.; Paisey, R.; Millward, B.A.; Eccles, C.; Williams, K.; Hammersley, S.; MacLeod, K.M.; Gale, T.J. Short-term effects of severe dietary carbohydrate-restriction advice in Type 2 diabetes—A randomized controlled trial. Diabet. Med. 2006, 23, 15–20. [Google Scholar] [CrossRef] [PubMed]
  39. Dyson, P.A.; Beatty, S.; Matthews, D.R. A low-carbohydrate diet is more effective in reducing body weight than healthy eating in both diabetic and non-diabetic subjects. Diabet. Med. 2007, 24, 1430–1435. [Google Scholar] [CrossRef] [PubMed]
  40. Saslow, L.R.; Kim, S.; Daubenmier, J.J.; Moskowitz, J.T.; Phinney, S.D.; Goldman, V.; Murphy, E.J.; Cox, R.M.; Moran, P.; Hecht, F.M. A randomized pilot trial of a moderate carbohydrate diet compared to a very low carbohydrate diet in overweight or obese individuals with type 2 diabetes mellitus or prediabetes. PLoS ONE 2014, 9, e91027. [Google Scholar] [CrossRef] [PubMed]
  41. Foster, G.D.; Wyatt, H.R.; Hill, J.O.; McGuckin, B.G.; Brill, C.; Mohammed, B.S.; Szapary, P.O.; Rader, D.J.; Edman, J.S.; Klein, S. A randomized trial of a low-carbohydrate diet for obesity. N. Engl. J. Med. 2003, 348, 2082–2090. [Google Scholar] [CrossRef] [PubMed]
  42. Shai, I.; Schwarzfuchs, D.; Henkin, Y.; Shahar, D.R.; Witkow, S.; Greenberg, I.; Golan, R.; Fraser, D.; Bolotin, A.; Vardi, H.; et al. Weight loss with a low-carbohydrate, Mediterranean, or low-fat diet. N. Engl. J. Med. 2008, 359, 229–241. [Google Scholar] [CrossRef] [PubMed]
  43. Bueno, N.B.; De Melo, I.S.; De Oliveira, S.L.; da Rocha Ataide, T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: A meta-analysis of randomised controlled trials. Br. J. Nutr. 2013, 110, 1178–1187. [Google Scholar] [CrossRef] [PubMed]
  44. Hjorth, M.F.; Ritz, C.; Blaak, E.E.; Saris, W.H.; Langin, D.; Poulsen, S.K.; Larsen, T.M.; Sørensen, T.I.; Zohar, Y.; Astrup, A. Pretreatment fasting plasma glucose and insulin modify dietary weight loss success: Results from 3 randomized clinical trials. Am. J. Clin. Nutr. 2017, 106, 499–505. [Google Scholar] [CrossRef] [PubMed]
  45. Yancy, W.S., Jr.; Olsen, M.K.; Guyton, J.R.; Bakst, R.P.; Westman, E.C. A low-carbohydrate, ketogenic diet versus a low-fat diet to treat obesity and hyperlipidemia: A randomized, controlled trial. Ann. Intern. Med. 2004, 140, 769–777. [Google Scholar] [CrossRef] [PubMed]
  46. Brinkworth, G.D.; Noakes, M.; Buckley, J.D.; Keogh, J.B.; Peter, M.C. Long-term effects of a very-low-carbohydrate weight loss diet compared with an isocaloric low-fat diet after 12 mo. Am. J. Clin. Nutr. 2009, 90, 23–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Keogh, J.B.; Brinkworth, G.D.; Noakes, M.; Belobrajdic, D.P.; Jonathan, D.B.; Clifton, P.M. Effects of weight loss from a very low-carbohydrate diet on endothelial function and markers of cardiovascular disease risk in subjects with abdominal obesity. Am. J. Clin. Nutr. 2008, 87, 567–576. [Google Scholar] [CrossRef] [PubMed]
  48. Hernandez, T.L.; Sutherland, J.P.; Wolfe, P.; Allian-Sauer, M.; Capell, W.H.; Talley, N.D.; Wyatt, H.R.; Foster, G.D.; Hill, J.O.; Eckel, R.H. Lack of suppression of circulating free fatty acids and hypercholesterolemia during weight loss on a high-fat, low-carbohydrate diet. Am. J. Clin. Nutr. 2010, 91, 578–585. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Tay, J.; Brinkworth, G.D.; Noakes, M.; Keogh, J.; Clifton, P.M. Metabolic effects of weight loss on a very-low-carbohydrate diet compared with an isocaloric high-carbohydrate diet in abdominally obese subjects. J. Am. Coll. Cardiol. 2008, 51, 59–67. [Google Scholar] [CrossRef] [PubMed]
  50. Hu, T.; Mills, K.T.; Yao, L.; Demanelis, K.; Eloustaz, M.; Yancy, W.S., Jr.; Kelly, T.N.; He, J.; Bazzano, L.A. Effects of low-carbohydrate diets versus low-fat diets on metabolic risk factors: A metaanalysis of randomized controlled clinical trials. Am. J. Epidemiol. 2012, 176 (Suppl. 7), S44–S54. [Google Scholar] [CrossRef]
  51. Menown, I.B.; Murtagh, G.; Maher, V.; Cooney, M.T.; Graham, I.M.; Tomkin, G. Dyslipidemia therapy update: The importance of full lipid profile assessment. Adv. Ther. 2009, 26, 711–718. [Google Scholar] [CrossRef]
  52. Superko, R.H. Lipoprotein subclasses and atherosclerosis. Front. Biosci. 2001, 6, D355–D365. [Google Scholar] [CrossRef] [PubMed]
  53. Morgan, L.M.; Griffin, B.A.; Millward, D.J.; DeLooy, A.; Fox, K.R.; Baic, S.; Bonham, M.P.; Wallace, J.M.W.; MacDonald, I.; Taylor, M.A.; et al. Comparison of the effects of four commercially available weight-loss programmes on lipid-based cardiovascular risk factors. Public Health Nutr. 2009, 12, 799–807. [Google Scholar] [CrossRef] [PubMed]
  54. Seidelmann, S.B.; Claggett, B.; Cheng, S.; Henglin, M.; Shah, A.; Steffen, L.M.; Folsom, A.R.; Rimm, E.B.; Willett, W.C.; Solomon, S.D. Dietary carbohydrate intake and mortality: A prospective cohort study and meta-analysis. Lancet Public Health 2018, 3, e419–e428. [Google Scholar] [CrossRef]
  55. Ioannidis, J.P. The Challenge of Reforming Nutritional Epidemiologic Research. JAMA 2018, 320, 969–970. [Google Scholar] [CrossRef] [PubMed]
  56. Crochemore, I.C.C.; Souza, A.F.P.; de Souza, A.C.F.; Rosado, E.L. ω-3 polyunsaturated fatty acid supplementation does not influence body composition, insulin resistance, and lipemia in women with type 2 diabetes and obesity. Nutr. Clin. Pract. 2012, 27, 553–560. [Google Scholar] [CrossRef] [PubMed]
  57. Lee, T.C.; Ivester, P.; Hester, A.G.; Sergeant, S.; Case, L.D.; Morgan, T.; Kouba, E.O.; Chilton, F.H. The impact of polyunsaturated fatty acid-based dietary supplements on disease biomarkers in a metabolic syndrome/diabetes population. Lipids Health Dis. 2014, 13, 196. [Google Scholar] [CrossRef] [Green Version]
  58. Gomes, P.M.; Hollanda-Miranda, W.R.; Beraldo, R.A.; Castro, A.V.B.; Geloneze, B.; Foss, M.C.; Foss-Freitas, M.C. Supplementation of α-linolenic acid improves serum adiponectin levels and insulin sensitivity in patients with type 2 diabetes. Nutrition 2015, 31, 853–857. [Google Scholar] [CrossRef]
  59. Garibay-Nieto, N.; Queipo-Garcia, G.; Alvarez, F.; Bustos, M.; Villanueva, E.; Ramirez, F.; Leon, M.; Laresgoiti-Servitje, E.; Duggirala, R.; Macias, T.; et al. Effects of conjugated linoleic acid and metformin on insulin sensitivity in obese children: Randomized clinical trial. J. Clin. Endocrinol. Metab. 2017, 102, 132–140. [Google Scholar]
  60. Müllner, E.; Plasser, E.; Brath, H.; Waldschütz, W.; Forster, E.; Kundi, M.; Wagner, K.-H. Impact of polyunsaturated vegetable oils on adiponectin levels, glycaemia and blood lipids in individuals with type 2 diabetes: A randomised, double-blind intervention study. J. Hum. Nutr. Diet. 2014, 27, 468–478. [Google Scholar] [CrossRef]
  61. Bermudez, B.; Ortega-Gomez, A.; Varela, L.M.; Villar, J.; Abia, R.; Muriana, F.J.G.; Lopez, S.; Gillingham, L.G.; Harris-Janz, S.; Jones, P.J.; et al. Clustering effects on postprandial insulin secretion and sensitivity in response to meals with different fatty acid compositions. Food Funct. 2014, 5, 1374–1380. [Google Scholar] [CrossRef]
  62. Koska, J.; Ozias, M.K.; Deer, J.; Kurtz, J.; Salbe, A.D.; Harman, S.M.; Reaven, P.D. A human model of dietary saturated fatty acid induced insulin resistance. Metab. Clin. Exp. 2016, 65, 1621–1628. [Google Scholar] [CrossRef] [PubMed]
  63. Malinska, H.; Huttl, M.; Oliyarnyk, O.; Bratova, M.; Kazdova, L. Conjugated linoleic acid reduces visceral and ectopic lipid accumulation and insulin resistance in chronic severe hypertriacylglycerolemia. Nutrition 2015, 31, 1045–1051. [Google Scholar] [CrossRef] [PubMed]
  64. Finucane, O.M.; Lyons, C.L.; Murphy, A.M.; Reynolds, C.M.; Klinger, R.; Healy, N.P.; Cooke, A.; Coll, R.; McAllan, L.; Nilaweera, L.; et al. Monounsaturated fatty acid-enriched high-fat diets impede adipose nlrp3 inflammasome-mediated IL-1β secretion and insulin resistance despite obesity. Diabetes 2015, 64, 2116–2128. [Google Scholar] [CrossRef] [PubMed]
  65. Rocca, A.S.; Lagreca, J.; Kalitsky, J.; Brubaker, P.L. Monounsaturated fatty acid diets improve glycemic tolerance through increased secretion of glucagon-like peptide-1. Endocrinology 2001, 142, 1148–1155. [Google Scholar] [CrossRef] [PubMed]
  66. Fernandez, M.L.; West, K.L. Mechanisms by which dietary fatty acids modulate plasma lipids. J. Nutr. 2005, 135, 2075–2078. [Google Scholar] [CrossRef]
  67. Benes, L.B.; Bassi, N.S.; Davidson, M.H. Omega-3 carboxylic acids monotherapy and combination with statins in the management of dyslipidemia. Vasc. Health Risk Manag. 2016, 12, 481–490. [Google Scholar] [CrossRef]
  68. Singh, S.; Arora, R.R.; Singh, M.; Khosla, S. Eicosapentaenoic acid versus docosahexaenoic acid as options for vascular risk prevention. Am. J. Ther. 2016, 23, e905–e910. [Google Scholar] [CrossRef]
  69. Harris, W.S.; Bulchandani, D. Why do omega-3 fatty acids lower serum triglycerides? Curr. Opin. Lipidol. 2006, 17, 387–393. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, F.; Wang, Y.; Zhu, Y.; Liu, X.; Xia, H.; Yang, X.; Sun, G. Treatment for 6 months with fish oil-derived n-3 polyunsaturated fatty acids has neutral effects on glycemic control but improves dyslipidemia in type 2 diabetic patients with abdominal obesity: A randomized, double-blind, placebo-controlled trial. Eur. J. Nutr. 2016, 56, 2415–2422. [Google Scholar] [CrossRef]
  71. Dittrich, M.; Jahreis, G.; Bothor, K.; Drechsel, C.; Kiehntopf, M.; Blüher, M.; Dawczynski, C. Benefits of foods supplemented with vegetable oils rich in α-linolenic, stearidonic or docosahexaenoic acid in hypertriglyceridemic subjects: A double-blind, randomized, controlled trail. Eur. J. Nutr. 2015, 54, 881–893. [Google Scholar] [CrossRef]
  72. Sawada, T.; Tsubata, H.; Hashimoto, N.; Takabe, M.; Miyata, T.; Aoki, K.; Yamashita, S.; Oishi, S.; Osue, T.; Yokoi, K.; et al. Effects of 6-month eicosapentaenoic acid treatment on postprandial hyperglycemia, hyperlipidemia, insulin secretion ability, and concomitant endothelial dysfunction among newly-diagnosed impaired glucose metabolism patients with coronary artery disease. An open label, single blinded, prospective randomized controlled trial. Cardiovasc. Diabetol. 2016, 15, 121. [Google Scholar] [PubMed]
  73. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2018. [Google Scholar] [CrossRef] [PubMed]
  74. Li, F.; Duan, Y.; Li, Y.; Tang, Y.; Geng, M.; Oladele, O.A.; Kim, S.W.; Yin, Y. Effects of dietary n-6:N-3 pufa ratio on fatty acid composition, free amino acid profile and gene expression of transporters in finishing pigs. Br. J. Nutr. 2015, 113, 739–748. [Google Scholar] [CrossRef]
  75. Figueiredo, P.S.; Candido, C.J.; Jaques, J.A.S.; Nunes, Â.A.; Caires, A.R.L.; Michels, F.S.; Almeida, J.A.; Filiú, W.F.O.; Hiane, P.A.; Nascimento, V.A.; et al. Oxidative stability of sesame and flaxseed oils and their effects on morphometric and biochemical parameters in an animal model. J. Sci. Food Agric. 2017, 97, 3359–3364. [Google Scholar] [CrossRef] [PubMed]
  76. Macri, E.V.; Lifshitz, F.; Alsina, E.; Juiz, N.; Zago, V.; Lezón, C.; Rodriguez, P.N.; Schreier, L.; Boyer, P.M.; Friedman, S.M. Monounsaturated fatty acids-rich diets in hypercholesterolemic-growing rats. Int. J. Food Sci. Nutr. 2015, 66, 400–408. [Google Scholar] [CrossRef] [PubMed]
  77. Alsina, E.; Macri, E.V.; Lifshitz, F.; Bozzini, C.; Rodriguez, P.N.; Boyer, P.M.; Friedman, S.M. Efficacy of phytosterols and fish-oil supplemented high-oleic-sunflower oil rich diets in hypercholesterolemic growing rats. Int. J. Food Sci. Nutr. 2016, 67, 441–453. [Google Scholar] [CrossRef] [PubMed]
  78. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Albert, C.M.; Gordon, D.; Copeland, T.; et al. Marine n-3 Fatty Acids and Prevention of Cardiovascular Disease and Cancer. N. Engl. J. Med. 2018. [Google Scholar] [CrossRef]
  79. Lyte, J.M.; Gabler, N.K.; Hollis, J.H. Postprandial serum endotoxin in healthy humans is modulated by dietary fat in a randomized, controlled, cross-over study. Lipids Health Dis. 2016, 15, 186. [Google Scholar] [CrossRef]
  80. Matsumoto, M.; Sata, M.; Fukuda, D.; Tanaka, K.; Soma, M.; Hirata, Y.; Nagai, R. Orally administered eicosapentaenoic acid reduces and stabilizes atherosclerotic lesions in ApoE-deficient mice. Atherosclerosis 2008, 197, 524–533. [Google Scholar] [CrossRef]
  81. Simopoulos, A.P. An increase in the omega-6/omega-3 fatty acid ratio increases the risk for obesity. Nutrients 2016, 8, 128. [Google Scholar] [CrossRef]
  82. Kien, C.L.; Bunn, J.Y.; Fukagawa, N.K.; Anathy, V.; Matthews, D.E.; Crain, K.I.; Ebenstein, D.B.; Tarleton, E.K.; Pratley, R.E.; Poynter, M.E. Lipidomic evidence that lowering the typical dietary palmitate to oleate ratio in humans decreases the leukocyte production of proinflammatory cytokines and muscle expression of redox-sensitive genes. J. Nutr. Biochem. 2015, 26, 1599–1606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Moya-Pérez, A.; Neef, A.; Sanz, Y. Bifidobacterium pseudocatenulatum cect 7765 reduces obesity-associated inflammation by restoring the lymphocyte-macrophage balance and gut microbiota structure in high-fat diet-fed mice. PLoS ONE 2015, 10, e0126976. [Google Scholar] [CrossRef] [PubMed]
  84. Masi, L.N.; Martins, A.R.; Crisma, A.R.; Amaral, C.L.; Davanso, M.R.; Serdan, T.D.A.; de Sá, R.D.C.; Cruz, M.M.; Alonso-Vale, M.I.C.; Torres, R.P.; et al. Combination of a high-fat diet with sweetened condensed milk exacerbates inflammation and insulin resistance induced by each separately in mice. Sci. Rep. 2017, 7, 3937. [Google Scholar] [CrossRef] [PubMed]
  85. de Lorgeril, M.; Salen, P.; Martin, J.L.; Monjaud, I.; Delaye, J.; Mamelle, N. Mediterranean diet, traditional risk factors, and the rate of cardiovascular complications after myocardial infarction: Final report of the Lyon Diet Heart Study. Circulation 1999, 99, 779–785. [Google Scholar] [CrossRef] [PubMed]
  86. Tognon, G.; Lissner, L.; Sæbye, D.; Walker, K.Z.; Heitmann, B.L. The Mediterranean diet in relation to mortality and CVD: A Danish cohort study. Br. J. Nutr. 2014, 111, 151–159. [Google Scholar] [CrossRef] [PubMed]
  87. Knoops, K.T.; de Groot, L.C.; Kromhout, D.; Perrin, A.E.; Moreiras-Varela, O.; Menotti, A.; Van Staveren, W.A. Mediterranean diet, lifestyle factors, and 10-year mortality in elderly European men and women: The HALE project. JAMA 2004, 292, 1433–1439. [Google Scholar] [CrossRef] [PubMed]
  88. Lopez-Garcia, E.; Rodriguez-Artalejo, F.; Li, T.Y.; Fung, T.T.; Li, S.; Willett, W.C.; Rimm, E.B.; Hu, F.B. The Mediterranean-style dietary pattern and mortality among men and women with cardiovascular disease. Am. J. Clin. Nutr. 2014, 99, 172–180. [Google Scholar] [CrossRef]
  89. Mitrou, P.N.; Kipnis, V.; Thiébaut, A.C.; Reedy, J.; Subar, A.F.; Wirfält, E.; Flood, A.; Mouw, T.; Hollenbeck, A.R.; Leitzmann, M.F.; et al. Mediterranean dietary pattern and prediction of all-cause mortality in a US population: Results from the NIHAARP diet and health study. Arch. Intern. Med. 2007, 167, 2461–2468. [Google Scholar] [CrossRef]
  90. Sofi, F.; Abbate, R.; Gensini, G.F.; Casini, A. Accruing evidence on benefits of adherence to the Mediterranean diet on health: An updated systematic review and meta-analysis. Am. J. Clin. Nutr. 2010, 92, 1189–1196. [Google Scholar] [CrossRef]
  91. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. PREDIMED Study Investigators. Primary prevention of cardiovascular disease with a Mediterranean diet. N. Engl. J. Med. 2013, 368, 1279–1290. [Google Scholar] [CrossRef]
  92. Estruch, R.; Martínez-González, M.A.; Corella, D.; Salas-Salvadó, J.; Fitó, M.; Chiva-Blanch, G.; Fiol, M.; Gómez-Gracia, E.; Arós, F.; Lapetra, J.; et al. PREDIMED Study Investigators. Effect of a high-fat Mediterranean diet on bodyweight and waist circumference: A prespecified secondary outcomes analysis of the PREDIMED randomised controlled trial. Lancet Diabetes Endocrinol. 2016, 4, 666–676. [Google Scholar] [CrossRef]
  93. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Retraction and Republication: Primary Prevention of Cardiovascular Disease with a Mediterranean Diet. N. Engl. J. Med. 2018, 378, 2441–2442. [Google Scholar] [CrossRef] [PubMed]
  94. Tektonidis, T.G.; Åkesson, A.; Gigante, B.; Wolk, A.; Larsson, S.C. A Mediterranean diet and risk of myocardial infarction, heart failure and stroke: A population-based cohort study. Atherosclerosis 2015, 243, 93–98. [Google Scholar] [CrossRef] [PubMed]
  95. Tsivgoulis, G.; Psaltopoulou, T.; Wadley, V.G.; Alexandrov, A.V.; Howard, G.; Unverzagt, F.W.; Moy, C.; Howard, V.J.; Kissela, B.; Judd, S.E. Adherence to a Mediterranean diet and prediction of incident stroke. Stroke 2015, 46, 780–785. [Google Scholar] [CrossRef]
  96. Torres-Peña, J.D.; Garcia-Rios, A.; Delgado-Casado, N.; Gomez-Luna, P.; Alcala-Diaz, J.F.; Yubero-Serrano, E.M.; Gomez-Delgado, F.; Leon-Acuña, A.; Lopez-Moreno, J.; Camargo, A.; et al. Mediterranean diet improves endothelial function in patients with diabetes and prediabetes: A report from the CORDIOPREV study. Atherosclerosis 2018, 269, 50–56. [Google Scholar] [CrossRef] [PubMed]
  97. Miedema, M.D.; Petrone, A.; Shikany, J.M.; Greenland, P.; Lewis, C.E.; Pletcher, M.J.; Gaziano, J.M.; Djousse, L. The Association of Fruit and Vegetable Consumption During Early Adulthood with the Prevalence of Coronary Artery Calcium After 20 Years of Follow-Up: The CARDIA Study. Circulation 2015, 132, 1990–1998. [Google Scholar] [CrossRef] [PubMed]
  98. Levitan, E.B.; Ahmed, A.; Arnett, D.K.; Polak, J.F.; Hundley, W.G.; Bluemke, D.A.; Heckbert, S.R.; Jacobs, D.R., Jr.; Nettleton, J.A. Mediterranean diet score and left ventricular structure and function: The Multi-Ethnic Study of Atherosclerosis. Am. J. Clin. Nutr. 2016, 104, 595–602. [Google Scholar] [CrossRef] [PubMed]
  99. Shah, R.V.; Murthy, V.L.; Allison, M.A.; Ding, J.; Budoff, M.; Frazier-Wood, A.C.; Lima, J.A.; Steffen, L.; Siscovick, D.; Tucker, K.L.; et al. Diet and Adipose Tissue Distributions: The Multi-Ethnic Study of Atherosclerosis. Nutr. Metab. Cardiovasc. Dis. 2016, 26, 185–193. [Google Scholar] [CrossRef] [PubMed]
  100. InterAct Consortium. Mediterranean diet and type 2 diabetes risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) study: The InterAct project. Diabetes Care 2011, 34, 1913–1918. [Google Scholar] [CrossRef] [PubMed]
  101. Covas, M.I.; de la Torre, R.; Fito, M. Virgin olive oil: A key food for cardiovascular risk protection. Br. J. Nutr. 2015, 113 (Suppl. 2), S19–S28. [Google Scholar] [CrossRef]
  102. Chiva-Blanch, G.; Urpi-Sarda, M.; Ros, E.; Valderas-Martinez, P.; Casas, R.; Arranz, S.; Guillén, M.; Lamuela-Raventós, R.M.; Llorach, R.; Andres-Lacueva, C.; et al. Effects of red wine polyphenols and alcohol on glucose metabolism and the lipid profile: A randomized clinical trial. Clin. Nutr. 2013, 32, 200–206. [Google Scholar] [CrossRef]
  103. Gepner, Y.; Golan, R.; Harman-Boehm, I.; HenkiN, Y.; Schwarzfuchs, D.; Shelef, I.; Durst, R.; Kovsan, J.; Bolotin, A.; Leitersdorf, E.; et al. Effects of initiating moderate alcohol intake on cardiometabolic risk in adults with type 2 diabetes: A 2-year randomized, controlled trial. Ann. Intern. Med. 2015, 163, 569–579. [Google Scholar] [CrossRef]
  104. Abete, I.; Goyenechea, E.; Zulet, M.A.; Martínez, J.A. Obesity and metabolic syndrome: Potential benefit from specific nutritional components. Nutr. Metab. Cardiovasc. Dis. 2011, 21 (Suppl. 2), B1–B15. [Google Scholar] [CrossRef] [PubMed]
  105. Puglisi, M.J.; Hasty, A.H.; Saraswathi, V. The role of adipose tissue in mediating the beneficial effects of dietary fish oil. J. Nutr. Biochem. 2011, 22, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Lankinen, M.; Schwab, U.; Erkkila, A.; Seppänen-Laakso, T.; Hannila, M.-L.; Mussalo, H.; Lehto, S.; Uusitupa, M.; Gylling, H.; Orešič, M. Fatty fish intake decreases lipids related to inflammation and insulin signaling—A lipidomics approach. PLoS ONE 2009, 4, e5258. [Google Scholar] [CrossRef] [PubMed]
  107. Afshin, A.; Micha, R.; Khatibzadeh, S.; Mozaffarian, D. Consumption of nuts and legumes and risk of incident ischemic heart disease, stroke, and diabetes: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2014, 100, 278–288. [Google Scholar] [CrossRef] [PubMed]
  108. Blanco Mejia, S.; Kendall, C.W.; Viguiliouk, E.; Augustin, L.S.; Ha, V.; Cozma, A.I.; Mirrahimi, A.; Maroleanu, A.; Chiavaroli, L.; Leiter, L.A.; et al. Effect of tree nuts on metabolic syndrome criteria: A systematic review and meta-analysis of randomised controlled trials. BMJ Open 2014, 4, e004660. [Google Scholar] [CrossRef]
  109. PREDIMED Investigators. Consumption of yogurt, low-fat milk, and other low-fat dairy products is associated with lower risk of metabolic syndrome incidence in an elderly Mediterranean population. J. Nutr. 2015, 145, 2308–2316. [Google Scholar] [CrossRef]
  110. Kim, Y.; Je, Y. Dairy consumption and risk of metabolic syndrome: A meta-analysis. Diabet. Med. 2016, 33, 428–440. [Google Scholar] [CrossRef]
  111. Tong, X.; Dong, J.Y.; Wu, Z.W.; Li, W.; Qin, L.Q. Dairy consumption and risk of type 2 diabetes mellitus: A meta-analysis of cohort studies. Eur. J. Clin. Nutr. 2011, 65, 1027–1031. [Google Scholar] [CrossRef]
  112. Gao, D.; Ning, N.; Wang, C.; Wang, Y.; Li, Q.; Meng, Z.; Liu, Y.; Li, Q. Dairy products consumption and risk of type 2 diabetes: Systematic review and dose-response meta-analysis. PLoS ONE 2013, 8, e73965. [Google Scholar] [CrossRef] [PubMed]
  113. Gijsbers, L.; Ding, E.L.; Malik, V.S.; de Goede, J.; Geleijnse, J.M.; Soedamah-Muthu, S.S. Consumption of dairy foods and diabetes incidence: A dose-response meta-analysis of observational studies. Am. J. Clin. Nutr. 2016, 103, 1111–1124. [Google Scholar] [CrossRef] [PubMed]
  114. Risérus, U.; Marklund, M. Milk fat biomarkers and cardiometabolic disease. Curr. Opin. Lipidol. 2017, 28, 46–51. [Google Scholar] [CrossRef] [PubMed]
  115. Appel, L.J.; Moore, T.J.; Obarzanek, E.; Vollmer, W.M.; Svetkey, L.P.; Sacks, F.M.; Bray, G.A.; Vogt, T.M.; Cutler, J.A.; Windhauser, M.M. A clinical trial of the effects of dietary patterns on blood pressure. N. Engl. J. Med. 1997, 336, 1117–1124. [Google Scholar] [CrossRef] [PubMed]
  116. Sacks, F.M.; Svetkey, L.P.; Vollmer, W.M.; Appel, L.J.; Bray, G.A.; Harsha, D.; Obarzanek, E.; Conlin, P.R.; Miller, E.R., 3rd; Simons-Morton, D.G. Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) Diet. N. Engl. J. Med. 2001, 344, 3–10. [Google Scholar] [CrossRef] [PubMed]
  117. Nguyen, H.T.; Bertoni, A.G.; Nettleton, J.A.; Bluemke, D.A.; Levitan, E.B.; Burke, G.L. DASH Eating Pattern Is Associated with Favorable Left Ventricular Function in the Multi-Ethnic Study of Atherosclerosis. J. Am. Coll. Nutr. 2012, 31, 401–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Uusitupa, M.; Hermansen, K.; Savolainen, M.J.; Schwab, U.; Kolehmainen, M.; Brader, L.; Mortensen, L.S.; Cloetens, L.; Johansson-Persson, A.; Önning, G.; et al. Effects of an isocaloric healthy Nordic diet on insulin sensitivity, lipid profile and inflammation markers in metabolic syndrome—A randomized study (SYSDIET). J. Intern. Med. 2013, 274, 52–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Hansen, C.P.; Overvad, K.; Kyrø, C.; Olsen, A.; Tjønneland, A.; Johnsen, S.P.; Jakobsen, M.U.; Dahm, C.C. Adherence to a Healthy Nordic Diet and Risk of Stroke. A Danish Cohort Study. Stroke 2017, 48. [Google Scholar] [CrossRef] [PubMed]
  120. Sanches Machado d’Almeida, K.; Ronchi Spillere, S.; Zuchinali, P.; Corrêa Souza, G. Mediterranean Diet and Other Dietary Patterns in Primary Prevention of Heart Failure and Changes in Cardiac Function Markers: A Systematic Review. Nutrients 2018, 10, 58. [Google Scholar] [CrossRef] [PubMed]
  121. dos Reis Padilha, G.; Sanches Machado d’Almeida, K.; Ronchi Spillere, S.; Corrêa Souza, G. Dietary Patterns in Secondary Prevention of Heart Failure: A Systematic Review. Nutrients 2018, 10, 828. [Google Scholar] [CrossRef] [PubMed]
  122. Vassort, G.; Turan, B. Protective role of antioxidants in diabetes-induced cardiac dysfunction. Cardiovasc. Toxicol. 2010, 10, 73–86. [Google Scholar] [CrossRef]
  123. Johnston, K.L.; Clifford, M.N.; Morgan, L.M. Coffee acutely modifies gastrointestinal hormone secretion and glucose tolerance in humans: Glycemic effects of chlorogenic acid and caffeine. Am. J. Clin. Nutr. 2003, 2, 728–733. [Google Scholar] [CrossRef] [PubMed]
  124. Tadera, K.; Minami, Y.; Takamatsu, K.; Matsuoka, T. Inhibition of alpha-glucosidase and alpha-amylase by flavonoids. J. Nutr. Sci. Vitaminol. 2006, 12, 149–153. [Google Scholar] [CrossRef]
  125. Dao, T.M.; Waget, A.; Klopp, P.; Serino, M.; Vachoux, C.; Pechere, L.; Drucker, D.; Champion, S.; Barthélemy, S.; Barra, Y.; et al. Resveratrol increases glucose induced GLP-1 secretion in mice: A mechanism which contributes to the glycemic control. PLoS ONE 2011, 12, e20700. [Google Scholar] [CrossRef] [Green Version]
  126. Kobayashi, Y.; Suzuki, M.; Satsu, H.; Arai, S.; Hara, Y.; Suzuki, K.; Miyamoto, Y.; Shimizu, M. Green tea polyphenols inhibit the sodiumdependent glucose transporter of intestinal epithelial cells by a competitive mechanism. J. Agric. Food Chem. 2000, 12, 5618–5623. [Google Scholar] [CrossRef]
  127. Basu, A.; Rhone, M.; Lyons, T.J. Berries: Emerging impact on cardiovascular health. Nutr. Rev. 2010, 68, 168–177. [Google Scholar] [CrossRef] [PubMed]
  128. Del Rio, D.; Borges, G.; Crozier, A. Berry flavonoids and phenolics: Bioavailability and evidence of protective effects. Br. J. Nutr. 2010, 104 (Suppl. 3), S67–S90. [Google Scholar] [CrossRef]
  129. Park, J.W.; Hong, J.S.; Lee, K.S.; Kim, H.Y.; Lee, J.J.; Lee, S.R. Green tea polyphenol (−)-epigallocatechin gallate reduces matrix metalloproteinase-9 activity following transient focal cerebral ischemia. J. Nutr. Biochem. 2010, 21, 1038–1044. [Google Scholar] [CrossRef]
  130. Lee, J.K.; Kwak, H.J.; Piao, M.S.; Jang, J.W.; Kim, S.H.; Kim, H.S. Quercetin reduces the elevated matrix metalloproteinases-9 level and improves functional outcome after cerebral focal ischemia in rats. Acta Neurochir. 2011, 153, 1321–1329. [Google Scholar] [CrossRef]
  131. Göçmen, A.Y.; Burgucu, D.; Gümü¸slü, S. Effect of resveratrol on platelet activation in hypercholesterolemic rats: CD40-CD40L system as a potential target. Appl. Physiol. Nutr. Metab. 2011, 36, 323–330. [Google Scholar] [CrossRef]
  132. Yashiro, T.; Nanmoku, M.; Shimizu, M.; Inoue, J.; Sato, R. Resveratrol increases the expression and activity of the low density lipoprotein receptor in hepatocytes by the proteolytic activation of the sterol regulatory element-binding proteins. Atherosclerosis 2012, 220, 369–374. [Google Scholar] [CrossRef] [PubMed]
  133. Haskó, G.; Pacher, P. Endothelial Nrf2 activation: A new target for resveratrol? Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H10–H12. [Google Scholar] [CrossRef] [PubMed]
  134. Deng, Y.H.; Alex, D.; Huang, H.Q.; Wang, N.; Yu, N.; Wang, Y.T.; Leung, G.P.; Lee, S.M. Inhibition of TNF-α-mediated endothelial cell-monocyte cell adhesion and adhesion molecules expression by the resveratrol derivative, trans-3,5,41-trimethoxystilbene. Phytother. Res. 2011, 25, 451–457. [Google Scholar] [PubMed]
  135. Park, D.W.; Baek, K.; Kim, J.R.; Lee, J.J.; Ryu, S.H.; Chin, B.R.; Baek, S.H. Resveratrol inhibits foam cell formation via NADPH oxidase 1- mediated reactive oxygen species and monocyte chemotactic protein-1. Exp. Mol. Med. 2009, 41, 171–179. [Google Scholar] [CrossRef] [PubMed]
  136. Bo, S.; Ciccone, G.; Castiglione, A.; Gambino, R.; de Michieli, F.; Villois, P.; Durazzo, M.; Cavallo-Perin, P.; Cassader, M. Anti-inflammatory and antioxidant effects of resveratrol in healthy smokers a randomized, double-blind, placebo-controlled, cross-over trial. Curr. Med. Chem. 2013, 20, 1323–1331. [Google Scholar] [CrossRef] [PubMed]
  137. Van der Made, S.M.; Plat, J.; Mensink, R.P. Resveratrol does not influence metabolic risk markers related to cardiovascular health in overweight and slightly obese subjects: A randomized, placebo controlled crossover trial. PLoS ONE 2015, 10, e0118393. [Google Scholar] [CrossRef]
  138. Faghihzadeh, F.; Adibi, P.; Hekmatdoost, A. The effects of resveratrol supplementation on cardiovascular risk factors in patients with non-alcoholic fatty liver disease: Arandomised, double blind, placebo-controlled study. Br. J. Nutr. 2015, 114, 796–803. [Google Scholar] [CrossRef]
  139. Gordish, K.L.; Beierwaltes, W.H. Resveratrol induces acute endothelium-dependent renal vasodilation mediated through nitric oxide and reactive oxygen species scavenging. Am. J. Physiol. Ren. Physiol. 2014, 306, F542–F550. [Google Scholar] [CrossRef]
  140. Rush, J.W.; Quadrilatero, J.; Levy, A.S.; Ford, R.J. Chronic resveratrol enhances endothelium dependent relaxation but does not alter eNOS levels in aorta of spontaneously hypertensive rats. Exp. Biol. Med. 2007, 232, 814–822. [Google Scholar]
  141. Liu, Y.; Ma, W.; Zhang, P.; He, S.; Huang, D. Effect of resveratrol on blood pressure: A meta-analysis of randomized controlled trials. Clin. Nutr. 2015, 34, 27–34. [Google Scholar] [CrossRef]
  142. Hung, L.M.; Su, M.J.; Chen, J.K. Resveratrol protects myocardial ischemia-reperfusion injury through both NO-dependent and NO-independent mechanisms. Free Radic. Biol. Med. 2004, 36, 774–781. [Google Scholar] [CrossRef] [PubMed]
  143. Shen, M.; Jia, G.L.; Wang, Y.M.; Ma, H. Cardioprotective effect of resvaratrol pretreatment on myocardial ischemia-reperfusion induced injury in rats. Vasc. Pharmacol. 2006, 45, 122–126. [Google Scholar] [CrossRef] [PubMed]
  144. Nazari, Q.A.; Kume, T.; Takada-Takatori, Y.; Izumi, Y.; Akaike, A. Protective effect of luteolin on an oxidative-stress model induced by microinjection of sodium nitroprusside in mice. J. Pharmacol. Sci. 2013, 122, 109–117. [Google Scholar] [CrossRef] [PubMed]
  145. Si, H.; Wyeth, R.P.; Liu, D. The flavonoid luteolin induces nitric oxide production and arterial relaxation. Eur. J. Nutr. 2014, 53, 269–275. [Google Scholar] [CrossRef] [PubMed]
  146. Englisch, W.; Beckers, C.; Unkauf, M.; Ruepp, M.; Zinserling, V. Efficacy of Artichoke dry extract in patients with hyperlipoproteinemia. Arzneimittelforschung 2000, 50, 260–265. [Google Scholar] [CrossRef]
  147. Li, H.; Xia, N.; Brausch, I.; Yao, Y.; Forstermann, U. Flavonoids from artichoke (Cynara scolymus L.) up-regulate endothelial-type nitric-oxide synthase gene expression in human endothelial cells. J. Pharmacol. Exp. Ther. 2004, 310, 926–932. [Google Scholar] [CrossRef] [PubMed]
  148. Jia, Z.; Nallasamy, P.; Liu, D.; Shah, H.; Li, J.Z.; Chitrakar, R.; Si, H.; McCormick, J.; Zhu, H.; Zhen, W.; et al. Luteolin protects against vascular inflammation in mice and TNF-alpha-induced monocyte adhesion to endothelial cells via suppressing IKBα/NF-κB signaling pathway. J. Nutr. Biochem. 2015, 26, 293–302. [Google Scholar] [CrossRef]
  149. Sun, L.; Li, E.; Wang, F.; Wang, T.; Qin, Z.; Niu, S.; Qiu, C. Quercetin increases macrophage cholesterol efflux to inhibit foam cell formation through activating PPAR-ABCA1 pathway. Int. J. Clin. Exp. Pathol. 2015, 8, 10854–10860. [Google Scholar]
  150. 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]
  151. Kim, S.G.; Kim, J.R.; Choi, H.C. Quercetin-Induced AMP-Activated Protein Kinase Activation Attenuates Vasoconstriction Through LKB1-AMPK Signaling Pathway. J. Med. Food. 2018, 21, 146–153. [Google Scholar] [CrossRef]
  152. Park, C.E.; Kim, M.J.; Lee, J.H.; Min, B.I.; Bae, H.; Choe, W.; Kim, S.S.; Ha, J. Resveratrol stimulates glucose transport in C2C12 myotubes by activating AMP-activated protein kinase. Mol. Med. 2007, 12, 222–229. [Google Scholar] [CrossRef] [PubMed]
  153. Egert, S.; Bosy-Westphal, A.; Seiberl, J.; Kürbitz, C.; Settler, U.; Plachta-Danielzik, S.; Wagner, A.E.; Frank, J.; Schrezenmeir, J.; Rimbach, G.; et al. Quercetin reduces systolic blood pressure and plasma oxidised lowdensity lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: A double-blinded, placebo-controlled cross-over study. Br. J. Nutr. 2009, 102, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  154. Lee, K.H.; Park, E.; Lee, H.J.; Kim, M.O.; Cha, Y.J.; Kim, J.M.; Lee, H.; Shin, M.J. Effects of daily quercetin-rich supplementation on cardiometabolic risks in male smokers. Nutr. Res. Pract. 2011, 5, 28–33. [Google Scholar] [CrossRef] [PubMed]
  155. Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin reduces blood pressure in hypertensive subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef] [PubMed]
  156. Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does quercetin improve cardiovascular risk factors and inflammatory biomarkers in women with type 2 diabetes: A double-blind randomized controlled clinical trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar] [PubMed]
  157. Serban, M.C.; Sahebkar, A.; Zanchetti, A.; Mikhailidis, D.P.; Howard, G.; Antal, D.; Andrica, F.; Ahmed, A.; Aronow, W.S.; Muntner, P.; et al. Effects of quercetin on blood pressure: A systematic review and meta-analysis of randomized controlled trials. J. Am. Heart Assoc. 2016, 5, e002713. [Google Scholar] [CrossRef]
  158. Meng, Z.; Yan, C.; Deng, Q.; Gao, D.F.; Niu, X.L. Curcumin inhibits LPS-induced inflammation in rat vascular smooth muscle cells in vitro via ROS-relative TLR4-MAPK/NF-kappaB pathways. Acta Pharmacol. Sin. 2013, 34, 901–911. [Google Scholar] [CrossRef]
  159. Kong, F.; Ye, B.; Cao, J.; Cai, X.; Lin, L.; Huang, S.; Huang, W.; Huang, Z. Curcumin represses NLRP3 inflammasome activation via TLR4/MyD88/NF-kappaB and P2X7R signaling in PMA-induced macrophages. Front. Pharmacol. 2016, 399, 369. [Google Scholar]
  160. Zhang, S.; Zou, J.; Li, P.; Zheng, X.; Feng, D. Curcumin attenuates atherosclerosis in apolipoprotein-E knockout mice by inhibiting Toll-like receptor 4 expression. J. Agric. Food Chem. 2018, 66, 449–456. [Google Scholar] [CrossRef]
  161. McNulty, H.; Jacob, R.F.; Mason, R.P. Biologic activity of carotenoids related to distinct membrane physicochemical interactions. Am. J. Cardiol. 2008, 101, S20–S29. [Google Scholar] [CrossRef]
  162. Fassett, R.G.; Coombes, J.S. Astaxanthin in cardiovascular health and disease. Molecules 2012, 17, 2030–2048. [Google Scholar] [CrossRef] [PubMed]
  163. Choi, S.K.; Park, Y.S.; Choi, D.K.; Chang, H.I. Effects of astaxanthin on the production of NO and the expression of COX-2 and iNOS in LPS-stimulated BV2 microglial cells. J. Microbiol. Biotechnol. 2008, 18, 1990–1996. [Google Scholar] [PubMed]
  164. Yoshida, H.; Yanai, H.; Ito, K.; Tomono, Y.; Koikeda, T.; Tsukahara, H.; Tada, N. Administration of natural astaxanthin increases serum HDL-cholesterol and adiponectin in subjects with mild hyperlipidemia. Atherosclerosis 2010, 209, 520–523. [Google Scholar] [CrossRef]
  165. Iwamoto, T.; Hosoda, K.; Hirano, R.; Kurata, H.; Matsumoto, A.; Miki, W.; Kamiyama, M.; Itakura, H.; Yamamoto, S.; Kondo, K. Inhibition of low-density lipoprotein oxidation by astaxanthin. J. Atheroscler. Thromb. 2000, 7, 216–222. [Google Scholar] [CrossRef] [PubMed]
  166. Hussein, G.; Nakamura, M.; Zhao, Q.; Iguchi, T. Anti-hypertensive and neuro-protective effects of astaxanthin in experimental animals. Biol. Pharm. Bull. 2005, 28, 47–52. [Google Scholar] [CrossRef] [PubMed]
  167. Riccioni, G.; Bucciarelli, T.; D’Orazio, N.; Palumbo, N.; DiIlio, E.; Corradi, F.; Pennelli, A.; Bazzano, L.A. Plasma Antioxidants and asymptomatic carotid atherosclerotic disease. Ann. Nutr. Metab. 2008, 53, 86–90. [Google Scholar] [CrossRef] [PubMed]
  168. Grobusch, K.K.; Launer, L.J.; Geleijnse, J.M.; Boeing, H.; Hofman, A.; Witteman, J.C. Serum carotenoids and ATS: The Rotterdam study. Atherosclerosis 2000, 148, 49–56. [Google Scholar] [CrossRef]
  169. Rissanen, T.H.; Voutilainen, S.; Nyyssonen, K.; Salonen, J.T. Lycopene, atherosclerosis, and coronary heart disease. Exp. Biol. Med. 2002, 227, 900–907. [Google Scholar] [CrossRef]
  170. Kaliora, A.C.; Dedoussis, G.V.; Schmidt, H. Dietary antioxidants in preventing atherogenesis. Atherosclerosis 2006, 187, 1–17. [Google Scholar] [CrossRef] [PubMed]
  171. Krishaswamy, R.; Devaraj, S.N.; Padma, V.V. Lutein protects HT- 29 cells against Deoxynivalenol-induced oxidative stress and apoptosis: Prevention of NF-κB nuclear localization and down regulation of NF-κB and COX-2 expression. Free Radic. Biol. Med. 2010, 49, 50–60. [Google Scholar] [CrossRef]
  172. Jin, X.H.; Ohgami, K.; Shiratori, K.; Suzuki, Y.; Hirano, T.; Koyama, Y.; Yoshida, K.; Ilieva, I.; Iseki, K.; Ohno, S. Inhibitory effects of lutein on endo toxin-induced uveitis in Lewis rats. Investig. Ophthalmol. Vis. Sci. 2006, 47, 2562–2568. [Google Scholar] [CrossRef] [PubMed]
  173. Kritchevsky, S.B.; Tell, G.S.; Shimakawa, T. Provitamin A carotenoid intake and carotid artery plaques: The atherosclerosis risk in communities study. Am. J. Clin. Nutr. 1998, 68, 726–733. [Google Scholar] [CrossRef] [PubMed]
  174. McQuillan, B.M.; Hung, J.; Beilby, J.P. Antioxidant vitamins and the risk of carotid atherosclerosis. The perth carotid ultrasound disease assessment study (CUDAS). J. Am. Coll. Cardiol. 2001, 38, 1788–1794. [Google Scholar] [CrossRef]
  175. Jialal, I.; Norkus, E.P.; Cristol, L.; Grundy, S.M. Beta-Carotene inhibits the oxidative modification of low-density lipoprotein. Biochem. Biophys. Acta 1991, 1086, 134–138. [Google Scholar] [CrossRef]
  176. DiTomo, P.; Canali, R.; Ciavardelli, D.; DiSilvestre, S.; DeMarco, A.; Giardinelli, A.; Pipino, C.; Di Pietro, N.; Virgili, F.; Pandolfi, A. B-carotene and lycopene affect endothelial response to TNF-a reducing nitro-oxidative stress and interaction with monocytes. Mol. Nutr. Food Res. 2012, 56, 217–227. [Google Scholar] [CrossRef] [PubMed]
  177. Harari, A.; Harats, D.; Marko, D.; Cohen, H.; Barshack, I.; Kamari, Y.; Gonen, A.; Gerber, Y.; Ben-Amotz, A.; Shaish, A. A 9-cis b-Carotene-enriched diet inhibits atherogenesis and fatty liver formation in LDL-R knockout mice. J. Nutr. Dis. 2008, 138, 1923–1930. [Google Scholar] [CrossRef] [PubMed]
  178. Lobo, G.P.; Amengual, J.; Li, H.N.M.; Golczak, M.; Bonet, M.L.; Palczewski, K.; VonLintig, J. Beta-Carotene decreases PPR-alpha activity and reduces lipid storage capacity of adipocytes in a beta-carotene oxygenase 1-dependent manner. J. Biol. Chem. 2010, 285, 27891–27899. [Google Scholar] [CrossRef]
  179. Ramakrishna, V.; Jailkhani, R. Oxidative stress in non insulin dependent diabetes mellitus (NIDDM) patients. Acta Diabetol. 2008, 45, 41–46. [Google Scholar] [CrossRef]
  180. Stranges, S.; Marshall, J.R.; Natarajan, R.; Donahue, R.P.; Trevisan, M.; Combs, G.F.; Cappuccio, F.P.; Ceriello, A.; Reid, M.E. Effects of long-term selenium supplementation on the incidence of type 2 diabetes: A randomized trial. Ann. Intern. Med. 2007, 147, 217–223. [Google Scholar] [CrossRef]
  181. Sarmento, R.A.; Silva, F.M.; Sbruzzi, G.; Schaan, B.D.; Almeida, J.C. Antioxidant micronutrients and cardiovascular risk in patients with diabetes: A systematic review. Arq. Bras. Cardiol. 2013, 101, 240–248. [Google Scholar] [CrossRef]
  182. McClain, C.; Morris, P.; Hennig, B. Zinc and endothelial function. Nutrition 1995, 11 (Suppl. 1), 117–120. [Google Scholar] [PubMed]
  183. Miao, X.; Wang, Y.; Sun, J.; Sun, W.; Tan, Y.; Cai, L.; Zheng, Y.; Su, G.; Liu, Q.; Wang, Y. Zinc protects against diabetes-induced pathogenic changes in the aorta: Roles of metallothionein and nuclear factor (erythroid-derived 2)-like 2. Cardiovasc. Diabetol. 2013, 12, 54. [Google Scholar] [CrossRef]
  184. Sarkar, P.K.; Lambert, L.A. Aetiology and treatment of hyperhomocysteinaemia causing ischaemic stroke. Int. J. Clin. Pract. 2001, 55, 262–268. [Google Scholar] [PubMed]
  185. Sánchez-Moreno, C.; Jiménez-Escrig, A.; Martín, A. Stroke: Roles of B vitamins, homocysteine and antioxidants. Nutr. Res. Rev. 2009, 22, 49–67. [Google Scholar] [CrossRef] [PubMed]
  186. Chambers, J.C.; Ueland, P.M.; Obeid, O.A.; Wrigley, J.; Refsum, H.; Kooner, J.S. Improved vascular endothelial function after oral B vitamins: An effect mediated through reduced concentrations of free plasma homocysteine. Circulation 2000, 102, 2479–2483. [Google Scholar] [CrossRef] [PubMed]
  187. Littarru, G.P.; Tiano, L. Clinical aspects of coenzyme Q10: An update. Nutrition 2010, 26, 250–254. [Google Scholar] [CrossRef]
  188. Pepe, S.; Marasco, S.F.; Hass, S.J.; Sheeran, F.L.; Krum, H.; Rosenfeldt, F.L. Coenzyme Q10 in cardiovascular disease. Mitochondrion 2007, 7, S154–S167. [Google Scholar] [CrossRef]
  189. Lee, B.J.; Huang, Y.C.; Chen, S.J.; Lin, P.T. Coenzyme Q10 supplementation reduces oxidative stress and increases antioxidant enzyme activity in patients with coronary artery disease. Nutrition 2012, 28, 250–255. [Google Scholar] [CrossRef]
  190. Law, M.R.; Morris, J.K. By how much does fruit and vegetable consumption reduce the risk of ischemic heart disease. Eur. J. Clin. Nutr. 1998, 52, 549–556. [Google Scholar] [CrossRef]
  191. Montonen, J.; Knekt, P.; Jrvinen, R.; Reunanen, A. Dietary antioxidant intake and risk of type 2 diabetes. Diabetes Care 2004, 27, 362–366. [Google Scholar] [CrossRef]
  192. Manzella, D.; Barbieri, M.; Ragno, E.; Paolisso, G. Chronic administration of pharmacologic doses of vitamin E improves the cardiac autonomic nervous system in patients with type 2 diabetes. Am. J. Clin. Nutr. 2001, 73, 1052–1057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Minamiyama, Y.; Takemura, S.; Bito, Y.; Shinkawa, H.; Tsukioka, T.; Nakahira, A.; Suehiro, S.; Okada, S. Supplementation of alpha-tocopherol improves cardiovascular risk factors via the insulin signalling pathway and reduction of mitochondrial-reactive oxygen species in type II diabetic rats. Free Radic. Res. 2008, 42, 261–271. [Google Scholar] [CrossRef] [PubMed]
  194. Pazdro, R.; Burgess, J.R. The role of vitamin E and oxidative stress in diabetes complications. Mech. Ageing Dev. 2010, 131, 276–286. [Google Scholar] [CrossRef] [PubMed]
  195. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Gordon, D.; Copeland, T.; D’Agostino, D.; et al. Vitamin D Supplements and Prevention of Cancer and Cardiovascular Disease. N. Engl. J. Med. 2018. [Google Scholar] [CrossRef] [PubMed]
  196. Walker, A.W.; Ince, J.; Duncan, S.H.; Webster, L.M.; Holtrop, G.; Ze, X.; Brown, D.; Stares, M.D.; Scott, P.; et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2011, 5, 220–230. [Google Scholar] [CrossRef] [PubMed]
  197. Donohoe, D.R.; Wali, A.; Brylawski, B.P.; Bultman, S.J. Microbial regulation of glucose metabolism and cell-cycle progression in mammalian colonocytes. PLoS ONE 2012, 7, e46589. [Google Scholar] [CrossRef] [PubMed]
  198. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Di, Y.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Lin, H.V.; Frassetto, A.; Kowalik, E.J., Jr.; Nawrocki, A.R.; Lu, M.M.; Kosinski, J.R.; Hubert, J.A.; Szeto, D.; Yao, X.; Forrest, G.; et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3 independent mechanisms. PLoS ONE 2012, 7, e35240. [Google Scholar]
  200. Sekirov, I.; Russell, S.L.; Antunes, L.C.; Finlay, B.B. Gut microbiota in health and disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef]
  201. Thomas, C.; Pellicciari, R.; Pruzanski, M.; Auwerx, J.; Schoonjans, K. Targeting bile-acid signalling for metabolic diseases. Nat. Rev. Drug Discov. 2008, 7, 678–693. [Google Scholar] [CrossRef]
  202. Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [PubMed]
  203. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [PubMed]
  204. Lundin, A.; Bok, C.M.; Aronsson, L.; Björkholm, B.; Gustafsson, J.A.; Pott, S.; Arulampalam, V.; Hibberd, M.; Rafter, J.; Pettersson, S. Gut flora, toll-like receptors and nuclear receptors: A tripartite communication that tunes innate immunity in large intestine. Cell. Microbiol. 2008, 10, 1093–1103. [Google Scholar] [CrossRef] [PubMed]
  205. Romond, M.B.; Ais, A.; Guillemot, F.; Bounouader, R.; Cortot, A.; Romond, C. Cell-free whey from milk fermented with Bifidobacterium breve C50 used to modify the colonic microflora of healthy subjects. J. Dairy Sci. 1998, 81, 1229–1235. [Google Scholar] [CrossRef]
  206. Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; Kerins, D.M.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Świątecka, D.; Arjan, N.; Karyn, R.P.; Henryk, K. The study on the impact of glycated pea proteins on human intestinal bacteria. Int. J. Food Microbiol. 2011, 145, 267–272. [Google Scholar]
  208. Meddah, A.T.; Yazourh, A.; Desmet, I.; Risbourg, B.; Verstraete, W.; Romond, M.B. The regulatory effects of whey retentate from bifidobacteria fermented milk on the microbiota of the simulator of the human intestinal microbial ecosystem (SHIME). J. Appl. Microbiol. 2001, 91, 1110–1117. [Google Scholar] [CrossRef]
  209. Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277. [Google Scholar] [CrossRef]
  210. Reddy, B.S.; Weisburger, J.H.; Wynder, E.L. Effects of high risk and low risk diets for colon carcinogenesis on fecal microflora and steroids in man. J. Nutr. 1975, 105, 878–884. [Google Scholar] [CrossRef]
  211. Russell, W.R.; Gratz, S.W.; Duncan, S.H.; Holtrop, G.; Ince, J.; Scobbie, L.; Duncan, G.; Johnstone, A.; Lobley, G.; Wallace, R. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am. J. Clin. Nutr. 2011, 93, 1062–1072. [Google Scholar] [CrossRef] [Green Version]
  212. 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]
  213. De Filippis, F.; Pellegrini, N.; Vannini, L.; Jefery, I.B.; La Storia, A.; Laghi, L.; Serrazanetti, D.I.; Di Cagno, R.; Ferrocino, I.; Lazzi, C.; et al. High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome. Gut 2016, 65, 1812–1821. [Google Scholar] [CrossRef] [PubMed]
  214. Fava, F.; Gitau, R.; Grifn, B.A.; Gibson, G.R.; Tuohy, K.M.; Lovegrove, J.A. The type and quantity of dietary fat and carbohydrate alter faecal microbiome and short-chain fatty acid excretion in a metabolic syndrome “at-risk” population. Int. J. Obes. 2013, 37, 216–223. [Google Scholar] [CrossRef] [PubMed]
  215. Lecomte, V.; Kaakoush, N.O.; Maloney, C.A.; Raipuria, M.; Huinao, K.D.; Mitchell, H.M.; Morris, M.J. Changes in gut microbiota in rats fed a high fat diet correlate with obesity-associated metabolic parameters. PLoS ONE 2015, 10, e0126931. [Google Scholar] [CrossRef] [PubMed]
  216. Caesar, R.; Tremaroli, V.; Kovatcheva-Datchary, P.; Cani, P.D.; Bäckhed, F. Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signaling. Cell Metab. 2015, 22, 658–668. [Google Scholar] [CrossRef] [PubMed]
  217. De la Serre, C.; Ellis, C.L.; Lee, J.; Hartman, A.L.; Rutledge, J.C.; Raybould, H.E. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am. J. Physiol. 2010, 299, G440–G448. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Eid, N.; Enani, S.; Walton, G.; Corona, G.; Costabile, A.; Gibson, G.; Rowland, I.; Spencer, J.P.E. The impact of date palm fruits and their component polyphenols, on gut microbial ecology, bacterial metabolites and colon cancer cell proliferation. J. Nutr. Sci. 2014, 3, e46. [Google Scholar] [CrossRef]
  219. Francavilla, R.; Calasso, M.; Calace, L.; Siragusa, S.; Ndagijimana, M.; Vernocchi, P.; Brunetti, L.; Mancino, G.; Tedeschi, G.; Guerzoni, E.; et al. Effect of lactose on gut microbiota and metabolome of infants with cow’s milk allergy. Pediatr. Allergy Immunol. 2012, 23, 420–427. [Google Scholar] [CrossRef]
  220. Gibson, G.R.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar]
  221. Costabile, A.; Klinder, A.; Fava, F.; Napolitano, A.; Fogliano, V.; Leonard, C.; Gibson, G.R.; Tuohy, K.M. Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: A double-blind, placebo-controlled, crossover study. Br. J. Nutr. 2008, 99, 110–120. [Google Scholar] [CrossRef]
  222. West, N.P.; Christophersen, C.T.; Pyne, D.B.; Cripps, A.W.; Conlon, M.A.; Topping, D.L.; Kang, S.; McSweeney, C.S.; Fricker, P.A.; Aguirre, D.; et al. Butyrylated starch increases colonic butyrate concentration but has limited effects on immunity in healthy physically active individuals. Exerc. Immunol. Rev. 2013, 19, 102–119. [Google Scholar] [PubMed]
  223. Champagne, C.P.; Ross, R.P.; Saarela, M.; Hansen, K.F.; Charalampopoulos, D. Recommendations for the viability assessment of probiotics as concentrated cultures and in food matrices. Int. J. Food Microbiol. 2011, 149, 185–193. [Google Scholar] [CrossRef] [PubMed]
  224. TormoCarnicer, R.; Infante Piña, D.; Roselló Mayans, E.; Bartolomé Comas, R. Intake of fermented milk containing Lactobacillus casei DN-114 001 and its effect on gut flora. An. Pediatr. 2006, 65, 448–453. [Google Scholar] [CrossRef]
  225. Wang, S.; Zhu, H.; Lu, C.; Kang, Z.; Luo, Y.; Feng, L.; Lu, X. Fermented milk supplemented with probiotics and prebiotics can effectively alter the intestinal microbiota and immunity of host animals. J. Dairy Sci. 2012, 95, 4813–4822. [Google Scholar] [CrossRef] [PubMed]
  226. He, T.; Priebe, M.G.; Zhong, Y.; Huang, C.; Harmsen, H.J.M.; Raangs, G.C.; Antoine, J.-M.; Welling, G.W.; Vonk, R.J. Effects of yogurt and bifidobacteria supplementation on the colonic microbiota in lactose-intolerant subjects. J. Appl. Microbiol. 2008, 104, 595–604. [Google Scholar] [CrossRef] [PubMed]
  227. Rajkumar, H.; Mahmood, N.; Kumar, M.; Varikuti, S.R.; Challa, H.R.; Myakala, S.P. Effect of probiotic (VSL#3) and omega-3 on lipid profile, insulin sensitivity, inflammatory markers, and gut colonization in overweight adults: A randomized, controlled trial. Med. Inflamm. 2014, 2014, 348959. [Google Scholar]
  228. Ejtahed, Hanie S.; Mohtadi-Nia, J.; Homayouni-Rad, A.; Niafar, M.; Asghari-Jafarabadi, M.; Mofid, V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 2012, 28, 539–543. [Google Scholar]
  229. Koren, O.; Spor, A.; Felin, J.; Fak, 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. 1), 4592–4598. [Google Scholar] [CrossRef] [PubMed]
  230. Ott, S.J.; El Mokhtari, N.E.; Musfeldt, M.; Hellmig, S.; Freitag, S.; Rehman, A.; Kuhbacher, 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]
  231. Karlsson, F.H.; Fak, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Backhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012, 3, 1245. [Google Scholar] [CrossRef] [Green Version]
  232. 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]
  233. Fu, Q.; Zhao, M.; Wang, D.; Hu, H.; Guo, C.; Chen, W.; Li, Q.; Zheng, L.; Chen, B. Coronary plaque characterization assessed by optical coherence tomography and plasma trimethylamine-n-oxide levels in patients with coronary artery disease. Am. J. Cardiol. 2016, 118, 1311–1315. [Google Scholar] [CrossRef] [PubMed]
  234. Li, X.S.; Obeid, S.; Klingenberg, R.; Gencer, B.; Mach, F.; Raber, L.; Windecker, S.; Rodondi, N.; Nanchen, D.; Muller, O.; et al. Gut microbiota-dependent trimethylamine n-oxide in acute coronary syndromes: A prognostic marker for incident cardiovascular events beyond traditional risk factors. Eur. Heart J. 2017, 11, 814–824. [Google Scholar] [CrossRef] [PubMed]
  235. Ryan, K.K.; Tremaroli, V.; Clemmensen, C.; Kovatcheva-Datchary, P.; Myronovych, A.; Karns, R.; Wilson-Perez, H.E.; Sandoval, D.A.; Kohli, R.; Backhed, F.; et al. Fxr is a molecular target for the effects of vertical sleeve gastrectomy. Nature 2014, 509, 183–188. [Google Scholar] [CrossRef] [PubMed]
  236. Thomas, C.; Gioiello, A.; Noriega, L.; Strehle, A.; Oury, J.; Rizzo, G.; Macchiarulo, A.; Yamamoto, H.; Mataki, C.; Pruzanski, M.; et al. Tgr5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009, 10, 167–177. [Google Scholar] [CrossRef] [PubMed]
  237. 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] [PubMed] [Green Version]
  238. Sandek, A.; Bauditz, J.; Swidsinski, A.; Buhner, S.; Weber-Eibel, J.; von Haehling, S.; Schroedl, W.; Karhausen, T.; Doehner, W.; Rauchhaus, M.; et al. Altered intestinal function in patients with chronic heart failure. J. Am. Coll. Cardiol. 2007, 50, 1561–1569. [Google Scholar] [CrossRef]
  239. Pasini, E.; Aquilani, R.; Testa, C.; Baiardi, P.; Angioletti, S.; Boschi, F.; Verri, M.; Dioguardi, F. Pathogenic gut flora in patients with chronic heart failure. JACC Heart Fail. 2016, 4, 220–227. [Google Scholar] [CrossRef]
  240. Tang, W.H.; Wang, Z.; Fan, Y.; Levison, B.; Hazen, J.E.; Donahue, L.M.; Wu, Y.; Hazen, S.L. Prognostic value of elevated levels of intestinal microbe-generated metabolite trimethylamine-n-oxide in patients with heart failure: Refining the gut hypothesis. J. Am. Coll. Cardiol. 2014, 64, 1908–1914. [Google Scholar] [CrossRef]
  241. Wang, Z.; Tang, W.H.; Buffa, J.A.; Fu, X.; Britt, E.B.; Koeth, R.A.; Levison, B.S.; Fan, Y.; Wu, Y.; Hazen, S.L. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-n-oxide. Eur. Heart J. 2014, 35, 904–910. [Google Scholar] [CrossRef]
  242. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
  243. Ley, R.E.; Backhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Karlsson, C.L.; Onnerfalt, J.; Xu, J.; Molin, G.; Ahrné, S.; Thorngren-Jerneck, K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity 2012, 20, 2257–2261. [Google Scholar] [CrossRef] [PubMed]
  245. Moreno-Navarrete, J.M.; Serino, M.; Blasco-Baque, V.; Azalbert, V.; Barton, R.H.; Cardellini, M.; Latorre, J.; Ortega, F.; Sabater-Masdeu, M.; Burcelin, R.; et al. Gut Microbiota Interacts with Markers of Adipose Tissue Browning, Insulin Action and Plasma Acetate in Morbid Obesity. Mol. Nutr. Food Res. 2018, 62. [Google Scholar] [CrossRef] [PubMed]
  246. Haro, C.; Montes-Borrego, M.; Rangel-Zuniga, O.A.; Alcalá-Díaz, J.F.; Gómez-Delgado, F.; Pérez-Martínez, P.; Delgado-Lista, J.; Quintana-Navarro, G.M.; Tinahones, F.J.; et al. Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population. J. Clin. Endocrinol. Metab. 2016, 101, 233–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Schwiertz, A.; Taras, D.; Schäfer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef] [PubMed]
  248. Chambers, E.S.; Viardot, A.; Psichas, A.; Morrison, D.; Murphy, K.G.; Zac-Varghese, S.E.K.; MacDougall, K.; Preston, T.; Tedford, C.; Finlayson, G.S.; et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut 2015, 64, 1744–1754. [Google Scholar] [CrossRef] [PubMed]
  249. Larsen, N.; Vogensen, F.K.; van den Berg, F.W.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sorensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef]
  250. Yang, T.; Santisteban, M.M.; Rodriguez, V.; Li, E.; Ahmari, N.; Carvajal Jessica, M.; Zadeh, M.; Gong, M.; Qi, Y.; Zubcevic, J.; et al. Gut dysbiosis is linked to hypertension. Hypertension 2015, 65, 1331–1340. [Google Scholar] [CrossRef]
  251. Pluznick, J.L.; Protzko, R.J.; Gevorgyan, H.; Peterlin, Z.; Sipos, A.; Han, J.; Brunet, I.; Wan, L.X.; Rey, F.; Wang, T.; et al. Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation. Proc. Natl. Acad. Sci. USA 2013, 110, 4410–4415. [Google Scholar] [CrossRef] [Green Version]
  252. Khalesi, S.; Sun, J.; Buys, N.; Jayasinghe, R. Effect of probiotics on blood pressure: A systematic review and meta-analysis of randomized, controlled trials. Hypertension 2014, 64, 897–903. [Google Scholar] [CrossRef] [PubMed]
  253. Duncan, S.H.; Lobley, G.E.; Holtrop, G.; Ince, J.; Johnstone, A.M.; Louis, P.; Flint, H.J. Human colonic microbiota associated with diet, obesity and weight loss. Int. J. Obes. 2008, 32, 1720–1724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.; Varma, Y.F.; Michael, A.; et al. Sloan Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. 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] [PubMed]
  256. Cotillard, A.; Kennedy, S.P.; Kong, L.C.; Prifti, E.; Pons, N.; Le Chatelier, E.; Almeida, M.; Quinquis, B.; Levenez, F.; Galleron, N.; et al. Dietary intervention impact on gut microbial gene richness. Nature 2013, 500, 585–588. [Google Scholar] [CrossRef] [PubMed]
  257. Simon, M.C.; Strassburger, K.; Nowotny, B.; Kolb, H.; Nowotny, P.; Burkart, V.; Zivehe, F.; Hwang, J.H.; Stehle, P.; Pacini, G.; et al. Intake of lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: A proof of concept. Diabetes Care 2015, 38, 1827–1834. [Google Scholar] [CrossRef] [PubMed]
  258. Karlsson, C.; Ahrne, S.; Molin, G.; Berggren, A.; Palmquist, I.; Fredrikson, G.N.; Jeppsson, B. Probiotic therapy to men with incipient arteriosclerosis initiates increased bacterial diversity in colon: A randomized controlled trial. Atherosclerosis 2010, 208, 228–233. [Google Scholar] [CrossRef] [Green Version]
  259. Vrieze, A.; Van Nood, E.; Holleman, F.; Salojarvi, J.; Kootte, R.S.; Bartelsman, J.F.; Dallinga-Thie, G.M.; Ackermans, M.T.; Serlie, M.J.; Oozeer, R.; et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012, 143, 913–916. [Google Scholar] [CrossRef]
  260. De Leon, L.M.; Watson, J.B.; Kelly, C.R. Transient flare of ulcerative colitis after fecal microbiota transplantation for recurrent clostridium difficile infection. Clin. Gastroenterol. Hepatol. 2013, 11, 1036–1038. [Google Scholar] [CrossRef]
  261. Brandt, L.J. Fmt: First step in a long journey. Am. J. Gastroenterol. 2013, 108, 1367–1368. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Korakas, E.; Dimitriadis, G.; Raptis, A.; Lambadiari, V. Dietary Composition and Cardiovascular Risk: A Mediator or a Bystander? Nutrients 2018, 10, 1912. https://doi.org/10.3390/nu10121912

AMA Style

Korakas E, Dimitriadis G, Raptis A, Lambadiari V. Dietary Composition and Cardiovascular Risk: A Mediator or a Bystander? Nutrients. 2018; 10(12):1912. https://doi.org/10.3390/nu10121912

Chicago/Turabian Style

Korakas, Emmanouil, George Dimitriadis, Athanasios Raptis, and Vaia Lambadiari. 2018. "Dietary Composition and Cardiovascular Risk: A Mediator or a Bystander?" Nutrients 10, no. 12: 1912. https://doi.org/10.3390/nu10121912

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