Next Article in Journal
Deer Skin Collagen Peptides Bound to Calcium: In Vitro Gastrointestinal Simulation of Digestion, Cellular Uptake and Analysis of Antioxidant Activity
Next Article in Special Issue
Differences in Vitamin A Levels and Their Association with the Atherogenic Index of Plasma and Subclinical Hypothyroidism in Adults: A Cross-Sectional Analysis in China
Previous Article in Journal
Lack of Association between Insufficient Intake of Multiple Vitamins and Frailty in Older Adults Who Consume Sufficient Energy and Protein: A Nationwide Cross-Sectional Study
Previous Article in Special Issue
Adherence to a Healthy Diet and Risk of Multiple Carotid Atherosclerosis Subtypes: Insights from the China MJ Health Check-Up Cohort
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Antioxidants in the Therapy of Cardiovascular Diseases—A Literature Review

1
Department of Nephrocardiology, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
2
Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, Ul. Zeromskiego 113, 90-549 Lodz, Poland
*
Author to whom correspondence should be addressed.
Submission received: 25 June 2024 / Revised: 24 July 2024 / Accepted: 30 July 2024 / Published: 6 August 2024
(This article belongs to the Special Issue Diet, Nutrition and Cardiovascular Health)

Abstract

:
Antioxidants are endogenous and exogenous substances with the ability to inhibit oxidation processes by interacting with reactive oxygen species (ROS). ROS, in turn, are small, highly reactive substances capable of oxidizing a wide range of molecules in the human body, including nucleic acids, proteins, lipids, carbohydrates, and even small inorganic compounds. The overproduction of ROS leads to oxidative stress, which constitutes a significant factor contributing to the development of disease, not only markedly diminishing the quality of life but also representing the most common cause of death in developed countries, namely, cardiovascular disease (CVD). The aim of this review is to demonstrate the effect of selected antioxidants, such as coenzyme Q10 (CoQ10), flavonoids, carotenoids, and resveratrol, as well as to introduce new antioxidant therapies utilizing miRNA and nanoparticles, in reducing the incidence and progression of CVD. In addition, new antioxidant therapies in the context of the aforementioned diseases will be considered. This review emphasizes the pleiotropic effects and benefits stemming from the presence of the mentioned substances in the organism, leading to an overall reduction in cardiovascular risk, including coronary heart disease, dyslipidaemia, hypertension, atherosclerosis, and myocardial hypertrophy.

1. Introduction

Reactive oxygen species (ROS) are small, reactive agents, generated both under pathological and physiological processes. ROS function as a secondary signaler and take part in modulating certain biological functions, including regulation of cell death. This is particularly significant because disruption of cell death regulation contributes to the development of the leading cause of death globally, namely, cardiovascular disease (CVD), and this process is disturbed as a result of the occurrence of oxidative stress, secondary to overproduction of ROS [1,2,3]. ROS vigorously engage with numerous molecules, encompassing various small inorganic compounds, carbohydrates, lipids, proteins, and nucleic acids. This may result in irreversible degradation of the target molecule’s function [4]. Apart from regulating cell death, ROS also participate in modulating the inflammatory response, vascular tone regulation, oxidation of LDL-cholesterol (LDL-c), and cell growth. Their concentration in the arterial wall increases in conditions such as diabetes, dyslipidemia, arterial hypertension, and cigarette smoking, contributing to the development of atherosclerosis [5]. Thereby, they may contribute to the onset of metabolic syndrome [6]. The role of ROS in the development and progression of atherosclerosis also encompasses DNA oxidation in vessel wall cells, endothelial dysfunction, and exertion of a negative impact on fibrous cap stability. This provokes atherosclerotic plaque ruptures and may adversely affect the frequency of cardiovascular complications, such as myocardial infarction [7]. ROS can also cause a decrease in the bioavailability of nitric oxide, thereby reducing endothelium-dependent vasodilatation, contributing to the development of arterial hypertension [8]. They also exhibit a negative impact on ryanodine receptor type 2 (RyR2), responsible for regulating calcium ion homeostasis in the atria of the heart, inducing its dysfunction, which may further contribute to the development of atrial fibrillation (AF) [9]. Other disturbances can also be observed in cardiac tissue, where ROS initiate signaling cascades implicated in inflammation, impaired contractility, interstitial fibrosis, or myocardial hypertrophy, influencing cellular architecture and function, and playing a role in cardiac injury. All of the aforementioned mechanisms contribute to the development of CVD [10]. Sources of ROS include mitochondrial dysfunction, nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, xanthine oxidase, nitric oxide oxidase, endoplasmic reticulum (ER), or others, such as xenobiotics, radiation, polluted air, chemicals, or certain medications [11]. Selected sources of ROS are presented in Figure 1 [11].
Currently, CVD is a major cause of both low quality of life and mortality in developed countries, with oxidative stress playing an undisputed role in this regard [12]. However, the organism is not defenseless against ROS. It possesses a range of antioxidants, which are substances that slow down or completely inhibit the oxidation process, even when their concentration is relatively low. Antioxidants can be categorized into endogenous and exogenous types, as well as those that indirectly eliminate ROS by modulating various signaling pathways and those that directly scavenge ROS. Endogenous ones are also subdivided as enzymatic and non-enzymatic [13]. Within the category of endogenous enzymatic antioxidants, the main representatives include superoxide dismutase (SOD), which is present in every cell of the body, followed by glutathione peroxidase (GPx) and catalase (CAT). Non-enzymatic antioxidants include, among others, vitamin E (alpha-tocopherol), vitamin A (beta-carotene), vitamin C (ascorbic acid), uric acid, glutathione (GSH), flavonoids, albumin, and ceruloplasmin [14]. In addition to the body’s natural antioxidative capacities, there is increasing interest in the potential utilization of antioxidants in the therapy of diseases rooted in oxidative stress, including CVD [15]. The aim of this review is to demonstrate the effect of selected antioxidants, such as coenzyme Q10 (CoQ10), flavonoids, carotenoids, or resveratrol, in reducing the incidence and progression of CVD. In addition, new antioxidant therapies in the context of the aforementioned diseases will be considered. This review did not use specific selection criteria or research methodology to identify sources and references.

2. Coenzyme Q10

2.1. Chemical Characteristics

CoQ10 is a naturally occurring lipid-soluble quinone in the human body, possessing numerous properties, with its most significant being its antioxidative and anti-inflammatory characteristics. The designation “10” signifies the number of isoprenyl units, accounting for its high solubility in fats, low polarity, and rapid penetration through the inner mitochondrial membrane [16]. It is noteworthy that CoQ10 exists in three forms—oxidized (ubiquinone), reduced (ubiquinol), and partially reduced (ubisemiquinone) [17]. These forms are presented in Figure 2. CoQ10 is endogenously synthesized from acetyl-CoA in the inner mitochondrial membrane, where it fulfils its primary biological role of electron transfer from complexes I and II to complex III [18]. This electron transfer contributes to the establishment of a proton gradient, ultimately leading to the efficient production of ATP. In addition, CoQ10 participates in the metabolism of other antioxidants, such as vitamin E and vitamin C [19]. It attains its highest concentration in tissues with high energy demands, such as muscles, kidneys, liver, and the heart, while its lowest concentration is found in the lungs and serum [16]. With age, the concentration of CoQ10 decreases in the heart, serum, and pancreas, accompanied by a shift in its redox status to oxidized from reduced. This process is associated with a decline in antioxidant capacity, thereby influencing the protection of lipoproteins and tissues in the blood [20]. Due to its antioxidative properties and significant role in ATP synthesis, CoQ10 plays a crucial role in meeting the energy demands of the cardiac muscle and other tissues. Among patients with CVD, studies have shown that CoQ10 concentrations are significantly lower compared to healthy individuals [21]. Over the past few years, numerous studies have been conducted to explore the impact of CoQ10 supplementation on the course of CVD.

2.2. Q10 Role’s in Heart Failure

ROS can induce cellular damage by interacting with protein centers, DNA, and cell membranes, as well as stimulate the proliferation of myocardial cells [22,23]. CoQ10 inhibits the initiation of lipid peroxidation processes, thereby exerting a protective influence on the cardiac muscle and other tissues [24]. In heart failure (HF), there is a decline in adenosine triphosphate (ATP) synthesis within the cardiac muscle, accompanied by an elevated generation of ROS and an alteration in calcium exchange, primarily attributed to ineffective electron transport chain function [16]. Referring to the meta-analysis of data investigating the impact of CoQ10 on HF, one can infer that supplementation of this compound demonstrates favorable effects on cardiac function, correlating with a reduction in mortality and hospitalization rates [25]. For instance, in 2014, a study was conducted, revealing that patients in the experimental group, receiving a daily dose of 300 mg of CoQ10, exhibited a reduced risk of major adverse cardiovascular events and experienced an improvement in disease symptoms compared to the control group [26]. Another study by Fladerer et al. from 2023 [27] found that while supplementation of the oxidized form of CoQ10 (ubiquinone) reduced the rate of cardiovascular death in patients with HF, the same effect was not observed for the reduced form (ubiquinol). Moreover, a recently published overview by Alarcón-Vieco et al. [25] analyzed preexisting data on CoQ10 supplementation in HF and found that it possessed a beneficial effect on heart function and mortality rates. CoQ10 was found to improve the ejection fraction (EF) by between 1.77% and 3.81%, which was an effect smaller than that exerted by traditional drugs, such as metoprolol (which increases EF by 7.4%) [25,28]. However, the data are inconsistent between different studies, the optimal supplementation dosage is unknown, and more trials evaluating the effect of CoQ10 in combination with drugs commonly used in HF are needed [25].

2.3. Q10 Role’s in Coronary Artery Disease and Dyslipidemia

Coronary artery disease (CAD) is a prevalent cardiovascular condition characterized by the formation of atherosclerotic plaques, leading to the narrowing of coronary artery lumens. This condition results in anginal symptoms, reduced tolerance to physical exertion, increased mortality, and a decline in the quality of life for affected individuals. Contributing factors to atherosclerotic plaque formation include oxidative stress and a state of chronic inflammation [29]. A systematic review conducted in 2019 revealed that CoQ10 supplementation contributes to a reduction in the levels of inflammatory markers among CAD patients. Additionally, higher concentrations of enzymes exhibiting antioxidative properties were detected, along with a decrease in malondialdehyde (MDA) levels, a marker of lipid peroxidation. These findings suggest that CoQ10 supplementation may be beneficial for CAD patients. However, to confirm these hypotheses, further research in this area is warranted [30]. One of the most significant risk factors for the development of CAD is dyslipidemia. It is widely acknowledged that elevated levels of LDL-C exert an atherogenic effect, whereas high concentrations of HDL-C prevent the formation of atherosclerotic plaques and serve as a favorable prognostic factor among patients with CVD. Studies have demonstrated that CoQ10 supplementation among CAD patients significantly reduces total cholesterol levels while concurrently increasing HDL-C concentrations [31]. On the other hand, in a randomized, double-blind, placebo-controlled study, it was demonstrated that CoQ10 supplementation did not significantly influence the lipid profile [32]. A recently published meta-analysis by Liu et al. [33] found that CoQ10 supplementation decreased total cholesterol, LDL-C, and triglycerides levels. Noteworthy, the meta-analysis also showed that the dosage of 400 to 500 mg/day exerted the greatest beneficial effect on total cholesterol [33]. Yet, investigations into the impact of CoQ10 on lipid concentrations have yielded inconclusive results, necessitating further research to conclusively elucidate the effects of supplementing this antioxidant on individual lipid fractions [34]. In 2022, a comprehensive study was conducted to investigate the effects of specific antioxidants and other micronutrients on CVD. In this study, it was demonstrated that CoQ10 reduces the concentration of specific lipoproteins by an average of 0.81 mmol/L for total cholesterol TC, 0.57 mmol/L for LDL-c, and 0.25 mmol/L for triglycerides TG. These results are significant compared to other studied micronutrients. Based on this study, it can be inferred that CoQ10 exhibits a more effective hypolipidemic effect than vitamin C, vitamin D, lycopene, resveratrol, quercetin, and isoflavones [35].

2.4. Q10 Role’s in Hypertension

Hypertension stands as a pivotal risk factor for the development of the majority of CVDs. Substantiated evidence suggests that CoQ10 diminishes arterial blood pressure and mitigates vasoconstriction, primarily through its anti-inflammatory and antioxidative properties [36]. In a systematic review conducted for primary prevention, the authors observed that CoQ10 supplementation led to a significant decrease in both systolic and diastolic blood pressure among patients who did not undergo lifestyle intervention [37]. In a randomized, double-blinded, controlled clinical trial conducted on a cohort of patients suffering from hyperlipidemia and myocardial infarction, supplementation with 200 mg of CoQ10 daily for 12 weeks resulted in a reduction in both systolic and diastolic blood pressure values [38]. Moreover, a recent meta-analysis by Zhao et al. [39] found that supplementation of CoQ10 was able to reduce systolic blood pressure by approximately −4.77 mmHg compared to the control group; however, it had no significant effect on diastolic blood pressure. The analysis also suggested that the CoQ10 dose of 100–200 mg/day had the best efficacy in decreasing systolic blood pressure [39]. However, the quality of the analyzed evidence was rated as moderate for systolic blood pressure and low for diastolic blood pressure according to the Grading of Recommendations, Assessment, Development, and Evaluation approach (GRADE) [39]. Moreover, CoQ10 reduced systolic blood pressure by an average of 3.55 mmHg and diastolic blood pressure by 0.92 mmHg. This indicates that CoQ10 was found to be more effective in lowering blood pressure than vitamin C, lycopene, isoflavones, and flavonoids, but less effective than resveratrol or L-arginine [35].

3. Polyphenols

3.1. Chemical Characteristics

Polyphenols, chemically described as aromatic compounds featuring at least two phenolic groups, are bioactive compounds, predominantly found in foods such as fruits, grains, vegetables, chocolate, and beverages, such as coffee, tea, and wine, and they play a significant role in the prevention and treatment of CVD, offering protection against numerous chronic illnesses [40]. Polyphenols are typically classified based solely on the chemical structure of their aglycones, specifically focusing on the number of phenol rings and the binding chemical structures between them. All plant phenolic chemicals are derived from the same intermediate, phenylalanine, or a close predecessor, shikimic acid. They often occur in conjugated forms, with one or more sugar residues attached to hydroxyl groups; however, direct connections of the sugar (polysaccharide or monosaccharide) to an aromatic carbon are also possible. Associations with other molecules, such as carboxylic and organic acids, amines, and lipids, as well as coupling with other phenols, are prevalent. Polyphenols can be categorized into distinct categories based on the amount of phenol rings they contain and the structural components that tie these rings together. Consequently, they are categorized into four groups: phenolic acids, stilbenes, lignans, and flavonoids (Figure 3) [41]. In our review, we aimed to offer an analysis of the most recent scientific insights into the effects of polyphenols on various cardiovascular diseases, with a specific focus on flavonoids and stilbenes.
The advantageous effects of polyphenols on cardiovascular health are attributed to their direct antioxidant action, which involves scavenging ROS and reactive nitrogen species [42]. Nonetheless, increasing evidence indicates alternative mechanisms that could also explain the observed decrease in CVD risk linked to the intake of polyphenols. These mechanisms include direct anti-inflammatory effects, modulation of intracellular signaling pathways and gene expression, maintenance of nitric oxide homeostasis, and antiplatelet aggregation capacity. However, polyphenols differ in their site of absorption in humans. Some of the polyphenols are well absorbed in the gastrointestinal tract, while others in the intestine or other parts of the digestive tract. Flavonoids, except flavanols, exist in glycosylated forms in foods. Absorption in the stomach is unclear, but some flavonoids, such as quercetin, can be absorbed at the gastric level. Glucosides may be transported into enterocytes by SGLT1 and hydrolyzed by β-glucosidase. Proanthocyanidins, due to their polymeric nature and high molecular weight, may limit absorption through the gut barrier [43].

3.2. Flavonoids

Flavonoids are secondary metabolites that mostly consist of a benzopyrone ring with phenolic or polyphenolic groups at various locations [44]. They are mostly present in fruits, herbs, stems, grains, nuts, vegetables, flowers, and seeds. The presence of bioactive phytochemical elements in these various plant sections provides them with therapeutic potential and biological activity. More than 10,000 flavonoid compounds have been isolated and identified. Most flavonoids are extensively recognized as medicinal agents. These are naturally generated via the phenylpropanoid pathway, with bioactivity determined by the absorption mechanism and bioavailability. Flavonoids have been employed in natural colors, cosmetics and skin care products, and anti-wrinkle creams [45]. However, these polyphenols are most commonly used in medicine.
Flavonoids have been widely employed as anticancer, antibacterial, antiviral, antiangiogenic, antimalarial, antioxidant, neuroprotective, antitumor, and anti-proliferative drugs (Figure 4) [46]. Apple peel preparations high in flavonoids inhibit acetylcholinesterase (ACE) in vitro and are an efficient antihypertensive drug [47]. It also protects against cardiometabolic illnesses and improves cognitive function with age. These compounds act as potent antioxidants as they reduce low-density lipoprotein (LDL) cholesterol oxidation, modulate cell signaling pathways, and reduce platelet aggregation [44]. Epidemiological studies show that flavonoids have health benefits, which are often linked to their antioxidant activity. In vitro investigations, however, revealed that many flavonoids hinder organification in thyroid cells and follicles. Studies in vivo and in vitro using synthetic and natural flavonoids revealed T4 displacement from transthyretin, resulting in changes in thyroid hormone availability in tissues. Radioactively tagged flavonoids appeared to be quickly removed from the body, primarily by fecal excretion. In pregnant rats, synthetic flavonoids pass the placenta and accumulate in the fetal compartment, including the brain. As a result, consuming excessive amounts of flavonoids is not recommended [48].
They are categorized into several categories based on their chemical structure, level of unsaturation, and carbon ring oxidation. Flavonoids are classified into many subgroups: anthoxanthins (flavanone and flavanol), flavanones, flavanonols, flavans, chalcones, anthocyanidins, and isoflavonoids. Each of these flavonoids is commonly found in nature. An increased consumption of flavonoid-rich foods provides a range of health advantages, and because these natural substances have beneficial benefits on human health, there has been an increased attempt to separate them from diverse plants. Citrus fruits are an excellent source of flavonoids. Oranges, lemons, and grapes contain two flavonoids called narigenin and hesperetin [49]. Mulberry contains anthocyanins and quercetin glycosides, both flavonoids.

3.2.1. Anthoxanthins (Flavanone and Flavanol)

Kaempferol is a flavonoid with anti-inflammatory properties [45], which prevent atherogenesis in THP-1 macrophages via regulating monocyte-to-macrophage differentiation, pro-inflammatory gene expression, monocyte mobility, IFN-γ-mediated inflammation, and cholesterol export. Kaempferol might be used to lower the risk of atherosclerotic diseases by altering the expression of disease-related genes, perhaps leading to the creation of a new additional treatment for CVD [45]. Kaempferol, a phytochemical rich in plant-derived foods, is commonly consumed as a phytochemical in a well-balanced diet due to its anti-inflammatory and anti-atherosclerotic effects. Its mechanism of action involves inhibiting the production of pro-inflammatory genes, MCP-1 and ICAM-1, involved in atherosclerosis progression. Kaempferol, along with other flavonoids, possesses diverse biological effects that can be either beneficial or harmful depending on the situation. Research suggests that kaempferol may have mutagenic and genotoxic properties, primarily because it can induce oxidative stress in laboratory conditions. This occurs when kaempferol interacts with free radicals, acting either as an antioxidant by neutralizing them or as a pro-oxidant, leading to the production of reactive oxygen species. Moreover, kaempferol’s pro-oxidant activity is associated with its ability to reduce metal ions, which can further enhance oxidative damage by generating hydroxyl radicals [50].
Oxidative stress, which results from an imbalance between cellular oxidants and antioxidants, can lead to damage to proteins, lipids, and DNA, triggering inflammatory reactions. Kaempferol demonstrates strong antioxidant properties by scavenging various reactive oxygen and nitrogen species, inhibiting lipid peroxidation, and safeguarding against oxidative damage in various cell types. Furthermore, it regulates cellular responses to inflammation by affecting the activity of redox-sensitive transcription factors, such as Nrf-2, thus enhancing cellular defense mechanisms against oxidative stress-induced harm [51].
Kaempferol inhibits ERK1/2 by downregulating cytokine receptors, reducing cardiac failure and hypertrophy. Additionally, CYP1B1 plays a crucial role in the metabolism of various xenobiotics, arachidonic acid, estrogen, and cholesterol, which are important in the etiology of CVD, including heart hypertrophy and hypertension [52].
Rutin is a flavonoid molecule that has anti-inflammatory and vascular protective properties. The available data show that rutin improves cardiovascular structure and function in rats, including hypertrophy, inflammation, and fibrosis. It prevents atherosclerosis by lowering total cholesterol, triglycerides, non-esterified fatty acids, and insulin levels in rat blood. Rutin improves metabolic disorders and slows the premature aging of vascular smooth-muscle cells, lowering the burden of atherosclerosis and stabilizing plaques, and may have a role in the treatment of diabetic atherosclerosis [45]. The study looks at the function of rutin in vascular calcification, which is a possible risk factor for CVD [53]. Air pollution affects around 80% of the Chinese population, and prolonged exposure to air pollution is highly related with atherosclerosis and CVD. Winter coal production in Taiyuan, a hilly region in northern China, contributes to the high organic content, concentration, and major heavy metal components in winter PM2.5. Vascular calcification is an important pathophysiological substrate for CVD development.
The findings revealed that PM2.5 exposure accelerated vascular calcification, which was linked to the activation of oxidative stress and OPG/RANKL in vivo and in vitro. Rutin supplementation reduced PM2.5-induced vascular calcification via the OPG/RANKL signal pathway and ROS production. Following PM2.5 exposure, RANKL expression rose while OPG expression decreased, showing that PM2.5 promoted vascular calcification through the OPG/RANKL signaling pathway both in vivo and in vitro. PM2.5 exposure activated the nuclear translocation of NF-κB, which plays a crucial role in pro-inflammatory responses. This activation is critical for RANKL-mediated osteogenic differentiation of smooth-muscle cells, which contributes to vascular calcification [54]. PM2.5 exposure also causes OS, which results in oxidative damage. PM2.5 exposure boosts the expression of NOX4 and p22phox, two components of the superoxide-producing NAPDH (reduced form of nicotinamide-adenine dinucleotide phosphate) oxidase system, which is also linked to vascular calcification [45].

3.2.2. Flavanones

Viscosine is a flavonoid extracted from Dodonea viscosa that exhibits anti-inflammatory, antipyretic, and antioxidant effects. Baicalin is a flavonoid found in medicinal plants, such as Scutellaria baicalensis Georgi and Oroxylum indicum. This flavonoid has antioxidant and anti-inflammatory properties and is used to treat conditions such as asthma, liver and kidney disease, inflammatory bowel disease, carcinogenesis, and CVD. Chrysin, a flavonoid, also exhibits anti-inflammatory and antioxidant properties [55].

3.2.3. Chalcones

Chalcones are the natural precursors to flavonoids and iso-flavonoids. They are present in many plants and vegetables and have a wide range of biological functions. Chalcone is an aromatic ketone and enone that can trigger the nuclear factor erythroid 2-related factor (2NRF2) pathway [56]. Several novel dihydroxy chalcones were synthesized and investigated for their ability to lower ROS and oxidative stress, acting as an anti-ischemic stroke agent by activating the KEAP1/NRF2/ARE pathway [56]. Rat models were utilized to study cerebral ischemia-reperfusion injury (CIRI) after a stroke. Not only did these compounds efficiently protect neuron-like PC12 cells from H2O2-induced oxidative damage, but they also showed neuroprotective capabilities against ischemia/reperfusion-related brain injury in animals. These inflammatory genes are activated after a stroke. Stem cells are capable of producing anti-inflammatory substances. Following an ischemic stroke, endothelial progenitor cells (EPCs) exert a direct impact on the inflammation-associated stroke vasculome [57]. Fisetin, a flavonoid, inhibits LPS-induced TNFα production and decreases nuclear factor jB activation, making it a neuroprotective and anti-inflammatory medication for post-ischemia damage in vitro. Focal cortical dysplasia (FCD) develops when neurons in the brain do not migrate correctly during their development in utero. Rutin, a naturally occurring flavonoid, has been used in animal studies to treat FCD. Motor neurons recovered considerably when administered 50 mg/kg. This might be employed as a medicinal drug in the future [58].

3.3. Stilbenes

Resveratrol, also known chemically as 3,4′,5-trihydroxystilbene, is a naturally occurring stilbene, a bioactive molecule with pleiotropic biofunctions, naturally produced by many plants, primarily grapes and peanuts. This polyphenol has been presented in numerous studies as a substance with a wide range of beneficial effects on human health, including its impact on CVD [59,60]. The pleiotropic effect of resveratrol arises from its ability to interact with diverse targets, including kinases, receptors, and signaling molecules [61].

3.3.1. Resveratrol’s Impact on Oxidative Stress, Inflammation, and NO Synthesis

Early research demonstrates that resveratrol effectively reduces oxidative stress by inhibiting human LDL oxidation and lipid peroxidation. In mice, resveratrol supplementation decreased lethality from lipopolysaccharide (LPS) exposure and enhanced antioxidative enzyme activities in the myocardium and aorta. Resveratrol exhibits notable anti-inflammatory effects by inhibiting COX-1, thus reducing pro-inflammatory eicosanoid synthesis. Furthermore, resveratrol enhances vasculoprotective nitric oxide (NO) synthesis through eNOS activation, which is mediated by estrogen receptors, MAPK signaling, and SIRT1 activation. Animal studies confirmed these findings, with resveratrol improving endothelial functionality in hypercholesterolemic rabbits and enhancing eNOS and NO levels in the plasma [59]. Research findings indicate that resveratrol reduces ROS production and lowers levels of oxidative stress and blood pressure by enhancing the capability of redox proteins to regulate redox balance within cells [62]. Moreover, by activating SIRT1, it can augment the activity of endothelial nitric oxide synthase (eNOS), thereby suppressing the expression of the angiotensin II receptor. This impedes vessel constriction induced by angiotensin II, leading to vasodilation and a reduction in blood pressure. Studies unequivocally suggest that resveratrol supplementation could, therefore, influence the prevention and treatment of hypertension [63,64].

3.3.2. Resveratrol and Lipid Oxidation

Joao Tomé-Carneiro et al. evaluated the impact of a grape supplement containing 8 mg of resveratrol on oxidized LDL, apolipoprotein B (ApoB), and serum lipids in patients undergoing statin therapy as part of primary prevention of CVD. Patients with CVD were administered GE-RES, GE, or a placebo in combination with statins, commonly prescribed drugs for CVD [65]. The GE-RES formulation was created by blending 8 mg of resveratrol with grape extract (GE). Within the GE-RES group, a notable reduction in LDLc, ApoB, and the ratio of oxidized LDL/ApoB was observed. Conversely, in the GE group, only a decrease in LDLc was evident [65]. Additionally, GE-RES significantly reduced hsCRP levels, which correlated well with the decrease in TNF-α and plasminogen activator inhibitor type 1 (PAI-1) levels [66]. Considering PAI-1′s involvement in the pathogenesis of obesity, diabetes, and CVD, it could be deemed an attractive target for addressing these conditions [67].
In a comprehensive cross-sectional study, called PREDIMED, which focused on individuals at a heightened risk of CVD, findings revealed that a high level of resveratrol metabolite in urine correlated with a decreased risk of CVD. Moreover, the combination of moderate wine consumption with resveratrol supplementation was linked to enhancements in the blood lipid profile, fasting blood glucose levels, and heart rate. Nevertheless, it is worth noting that resveratrol on its own may only lead to a reduction in fasting blood glucose levels [68].

3.3.3. Resveratrol on Atherosclerosis

Resveratrol, as many different polyphenols, revealed many cardiovascular benefits. (Table 1). Resveratrol has demonstrated significant anti-atherosclerotic effects by inhibiting the dysregulated and excessive proliferation of vascular smooth-muscle cells (VSMCs), a key contributor to atherosclerosis and restenosis post-vascular surgery. Studies indicate that resveratrol suppresses VSMC proliferation induced by advanced glycation end-products (AGEs), oxidized low-density lipoprotein (ox-LDL), and endothelin-1 (ET-1). Mechanistically, these effects are associated with the suppression of MAPK ERK1/2 and a reduction in ROS and H2O2 production. Additionally, resveratrol attenuates VSMC proliferation induced by hypoxia and homocysteine, while also reducing collagen synthesis and promoting the expression of protective factors. Resveratrol’s vasculoprotective benefits extend to inhibiting platelet aggregation, further mitigating the atherosclerosis risk. In animal models, resveratrol supplementation has been shown to reduce platelet aggregation and enhance endothelial function, highlighting its potential as a therapeutic agent in managing atherosclerosis [59].

3.3.4. Resveratrol and Protective Effect for Heart Injury

Resveratrol’s positive effects on cardiomyocytes in an ischemia-reperfusion paradigm include superoxide suppression, potassium channel activation, and increased endothelium-dependent vasodilation. In rats fed a high-cholesterol diet and exposed to an experimentally induced myocardial infarction, oral resveratrol supplementation altered specific cardiologic parameters, such as ejection fraction and fractional shortening, and promoted neovascularization in the damaged myocardium. Furthermore, feeding rats resveratrol for three weeks protected them against ischemia/reperfusion damage and restored an aberrant microRNA pattern. Resveratrol improved survival, hemodynamics, and energetics in rats in a model of hypertension that leads to heart failure [53].
Numerous studies also demonstrated significant cardioprotective properties of resveratrol and its potential therapeutic effects in patients with CHD and post-myocardial infarction [60,61]. Chekalina et al. demonstrated an improvement in both systolic and diastolic function of the left ventricle as well as left ventricular ejection fraction in patients with CHD following treatment with resveratrol [52]. Following myocardial infarction, patients exhibited improvements in endothelial function and left ventricular diastolic function, as well as reduced LDL levels, even with very low doses of resveratrol (10 mg/day; 3 months) [63].
In another study on patients diagnosed with stable angina pectoris, a combination therapy of resveratrol (20 mg/day) and calcium fructoborate (112 mg/day) was administered. The study evaluated the levels of hsCRP and brain natriuretic peptide (BNP), recognizing prognostic markers for inflammation and left ventricular function in cardiovascular patients. The combined treatment significantly decreased levels of hsCRP and BNP, credited to the synergistic effect of both agents [62].
It is worth mentioning a relatively new double-blind clinical trial that assessed the potential benefits of RES in patients with symptomatic systolic HF. This study enrolled sixty patients with New York Heart Association (NYHA) class II–III HF who were randomly assigned to one of two groups: (1) receiving RES supplementation at a dose of 100 mg/day for 3 months or (2) receiving a placebo. All patients were receiving maximally tolerated medical therapy for HF at the time of study inclusion, in accordance with the guidelines. At the end of the study, improvements in systolic and diastolic cardiac function and global cardiovascular burden were observed in the RES group, along with reductions in serum levels of biomarkers (NT-proBNP and galectin-3) and inflammatory cytokines (IL-1 and IL-6); furthermore, the treatment group exhibited a higher exercise capacity and reported better quality of life [69].

3.3.5. Resveratrol and Limitations

Although resveratrol shows therapeutic potential, its low bioavailability after oral administration is a significant limitation [70,71,72]. Studying the metabolism of resveratrol reveals that it is rapidly absorbed after ingestion. Despite this quick absorption, its levels remain very low due to rapid metabolism [70]. Gambini et al. conducted a study in which, after the oral consumption of 25 mg of resveratrol, they observed a peak serum concentration of less than 10 ng/mL after 0.5 h [71]. Resveratrol is also rapidly eliminated from the body. Approximately 77–80% of ingested resveratrol is absorbed in the intestines, with 49–60% of that being excreted in the urine. Consequently, 75% of the ingested resveratrol is removed from the body. The remaining portion is metabolized, with the maximum concentration of free resveratrol being only 1.7–1.9% [70].
Various strategies have been described to improve the pharmacokinetic properties and beneficial effects of resveratrol. These methodological approaches include nanoencapsulation in lipid nanocarriers or liposomes, nano-emulsions, micelles, incorporation into polymeric particles, solid dispersions, and nanocrystals [73]. One of the methods used in an in vitro study aiming to improve resveratrol’s bioavailability was drug yeast encapsulation. Despite the absence of in vivo studies, the authors concluded that the rapid metabolism and elimination contributing to the poor bioavailability of resveratrol could be partially mitigated through yeast cell encapsulation technology [74].
The focus of recent research is on innovative resveratrol delivery systems based on lipid nanoparticles. These sophisticated controlled-release systems are specifically engineered to transport and shield this bioactive compound from degradation. By doing so, they significantly enhance its physical stability and boost its oral bioavailability. Lipid nanoparticles, which are submicron colloidal carriers, are made from biodegradable and biocompatible lipids. These materials are widely acknowledged for their safety and appropriateness in encapsulating lipophilic and poorly water-soluble substances, such as resveratrol. This encapsulation process facilitates the efficient absorption of resveratrol when taken orally [75]. Neves et al. developed resveratrol nano-delivery systems based on solid-lipid nanoparticles and nanostructured lipid carriers. The research revealed that resveratrol was released only minimally over several hours from both nanocarrier systems, indicating that these lipid nanoparticles are highly stable. Simulations of the gastrointestinal tract in vitro showed that resveratrol largely remained associated with the lipid nanoparticles even after exposure to digestive fluids. Consequently, these delivery nano-systems are deemed effective for oral administration, as they protect the encapsulated resveratrol and facilitate its controlled release upon absorption, thereby improving the bioavailability of the compound [75].
Resveratrol exhibits a different effective dosage range in vitro (micromolar range in cell culture media) compared to its in vivo bioavailability (nanomolar range in the blood), complicating the determination of the biologically effective concentration for human supplementation. Concerns have been raised about achieving the effective in vitro concentrations in vivo. Therefore, the actual biologically effective concentration range of resveratrol in vivo remains undetermined. Although human tissue and organ levels are still being studied, evidence from rodent studies shows that resveratrol can accumulate in specific tissues or organs at concentrations comparable to those used in many in vitro experiments [60].
In conclusion, current literature shows that resveratrol possesses substantial therapeutic potential in addressing CVD. Despite recent advances in our understanding of how resveratrol affects CVD, and in particular CHD, further research is still required to appreciate what clinical significance this has at a population-based level.
Table 1. The source and role of different polyphenols and their cardiovascular benefits.
Table 1. The source and role of different polyphenols and their cardiovascular benefits.
PolyphenolSourceCardiovascular Benefits
ResveratrolGrapes, red wine, berriesImproves endothelial function, reduces blood pressure, anti-inflammatory effects, decreases LDL oxidation, and inhibits platelet aggregation [76].
EpicatechinDark chocolate, green teaEnhances endothelial function, improves blood flow, lowers blood pressure, reduces oxidative stress, and improves cholesterol profiles [77].
QuercetinApples, onions, berriesAnti-inflammatory effects, reduces blood pressure, improves endothelial function, and decreases LDL oxidation [78].
CatechinsGreen tea, cocoa, applesAntioxidant effects, improves endothelial function, reduces blood pressure, lowers cholesterol levels, and enhances nitric oxide availability [79].
AnthocyaninsBerries, red grapes, red cabbageReduces oxidative stress, anti-inflammatory effects, improves endothelial function, lowers blood pressure, and enhances nitric oxide production [48].
FlavonolsTea, onions, kaleReduces blood pressure and has anti-inflammatory effects [80].

4. Carotenoids

4.1. Chemical Characteristics

Carotenoids are organic pigments, belonging to the tetraterpenes family, that can be found in many foods, such as fruits, vegetables, and fish [81,82]. The typical human diet contains around 40 out of more than 600 carotenoids, and only 20 carotenoids have been found to be present in human blood and tissues [81,83].
Most carotenoids are composed of a central carbon chain of alternating single and double bonds and different cyclic or acyclic end groups [84]. The group can be classified into provitamin A (namely β-carotene, α-carotene, and β-cryptoxanthin) and non-provitamin A compounds [84]. Based on which functional group they contain, carotenoids can also be divided into xanthophylls (such as lutein or zeaxanthin), which contain oxygen and carotenes (such as α-carotene, β-carotene, and lycopene) with only a hydrocarbon chain and no functional group [85].
One of the significant features of the carotenoids is their strong coloration, which is a consequence of light absorption in the presence of a conjugated chain [86,87]. For example, β-carotene, α-carotene, and β-cryptoxanthin are orange, lutein is yellow, and lycopene is red [86].

4.2. Antioxidant Capabilities

Carotenoids possess beneficial antioxidant properties because of conjugated double bonds, and they are able to accept electrons from reactive species and neutralize free radicals [88]. Carotenoids act as ROS scavengers—they are capable of trapping peroxyl radicals and quenching the singlet oxygen [89]. The scavenging occurs in three steps: electron transfer (oxidation and reduction), hydrogen abstraction, and addition [90]. Moreover, carotenoids are involved in the process of deactivation of electronically excited sensitizer molecules, which partake in the production of singlet oxygen and radicals [91]. The carotenoids’ efficacy in quenching the singlet oxygen is dependent on the number of conjugated double bonds present in the molecule; therefore, the most effective quencher is the open-ring carotenoid lycopene [84].

4.3. Carotenoids in the Human Body

Carotenoids are absorbed from food into gastrointestinal mucosal cells, formed into the chylomicrons, and released into the lymphatic system, and then they bind the lipoprotein at the liver and are released into the bloodstream [92,93,94]. Carotenoids are primarily accumulated in adipose tissue and the liver, but also in the cervix, lungs, skin, and eyes [95,96,97]. Particularly, carotenoids are stored in greater amounts in tissues that contain more low-density lipoprotein receptors (LDL-R), probably as a result of a non-specific uptake by lipoprotein carriers [98]. The positive health effect of carotenoids is primarily dependent on their antioxidant activities [98,99]. For instance, they act as photo-protectants and are involved in protecting the skin and eyes [98]. Their antioxidant abilities have also been suggested to be one of the mechanisms of their cardiovascular beneficial qualities [98].

4.4. Carotenoids and Cardiovascular Diseases

Oxidatively modified LDL plays a major role in the initiation and promotion of atherosclerosis and coronary heart disease (CHD) [100]. Free radicals, which cause LDL oxidation, partake in the production of foam cells and, thus, promote atherogenesis [101]. Therefore, it has been speculated that antioxidants, which hamper LDL oxidation, may exhibit a protective effect against CHD [82]. It has been observed that patients with CAD presented lower plasma levels of oxygenated carotenoids compared to the general population [102]. Moreover, it has been found that the reduced levels of lutein, zeaxanthin, and β-cryptoxanthin correlated with smoking, high body mass index, and low high-density lipoprotein cholesterol (HDL-C) [102]. Studies have shown that low plasma levels of lycopene were associated with subclinical atherosclerosis (understood as an increase in intima-media thickness of the common carotid artery) [103].
Furthermore, evidence has shown that higher serum levels of carotenoids were correlated with a decreased risk of elevated serum NT-proBNP levels, which might indicate that carotenoids can partake in preventing cardiac overload [104]. It has also been suggested that high plasma levels of β-cryptoxanthin and lutein might be linked to a lower risk of acute myocardial infarction [105]. Moreover, a study by Akbaraly et al. [106] confirmed that high plasma carotenoid levels correlate to a reduced risk of dysglycemia. The CARDIA study [107] found that the risk of developing hypertension was lower in individuals with higher concentrations of carotenoids.
A study by Xu et al. [108] showed that serum carotenoids were associated with a reduced risk of atherosclerosis and inversely associated with inflammatory cytokines. Therefore, it is speculated that carotenoids can reduce the risk of CVD by improving the lipid profile, preventing lipid peroxidation, contrasting vascular wall inflammation, stabilizing membrane properties, and thus acting in opposition to the pathophysiologic steps of atherosclerosis [109].
Since β-carotene and lycopene are carotenoids primarily transported in LDL, it has been suggested that they may be the most effective in protecting LDL from oxidation; however, many other carotenoids have been studied as well [110].

4.5. β-Carotene

β-Carotene is a quencher of singlet oxygen and a radical-trapping antioxidant [111]. It is able to inhibit LDL oxidation and, as a result, reduce its degradation by macrophages; thus, it is speculated that it may prevent atherosclerosis [111]. However, a study by Shaish et al. [112] showed that, while all-trans β-carotene inhibited atherogenesis in hypercholesterolemic rabbits, the effect may be separated from the LDL resistance to oxidation and may depend on stereospecific interactions with retinoic acid receptors in the artery wall. A study by Di Tomo et al. [113] found that β-carotene and lycopene were able to significantly decrease tumor necrosis factor-α-induced inflammation in human endothelial cells, meaning that they exerted a positive effect against CVD by opposing inflammatory oxidative stress.
An observational epidemiologic study by D’Odorico et al. [114] provided evidence that β-carotene had a protective effect on early atherogenesis and CVD, whereby the antioxidant properties countered the development of atherosclerotic lesions. Studies on the population of middle-aged Finnish men found that low plasma levels of β-carotene increased the risk of CVD mortality as well as sudden cardiac death [115,116]. Moreover, a study by Street et al. [117] found that decreased serum levels of β-carotene were associated with an increased risk of myocardial infarction in the population of smokers.
However, some clinical trials have not found β-carotene supplementation to have any beneficial effect on CVD or death incidence [118,119]. It is speculated that the conflicting results may be a result of differences in the trials’ performances or the form of dietary β-carotene; nevertheless, the beneficial effect of β-carotene remains uncertain [109].

4.6. Lycopene

Lycopene has been found to exert an anti-atherogenic effect, which is linked to its anti-inflammatory capabilities, improved lipid homeostasis, and inhibition of IL-1 secretion [97,120]. Lycopene is also able to positively influence nitric oxide levels, which contribute to vasodilatation, therefore, slowing the progression of atherosclerosis [121].
A study by Alvi et al. [122] showed that lycopene was able to target the expression of the liver genes PCSK-9 and HMGR, which resulted in an increase in LDL receptor activity and, therefore, a lowering of hypercholesterolemia. Lycopene was also found to improve the LDL/HDL ratio, reduce the accumulation of cholesterol in the aorta, lower the synthesis of dysfunctional HDL, and inhibit vascular smooth-muscle cell proliferation and foam cell formation, and thus, to have a beneficial effect on the initial stages of atherosclerosis [123,124,125].
Several studies have shown that lower blood levels of lycopene were correlated with an increased risk of atherosclerotic lesions and an increased risk of acute coronary events or stroke [126,127,128]. Consequently, high lycopene levels were found to be associated with a lower risk of CVD in women [129]. Moreover, a study by Kim et al. [130] found that an increased dietary intake of lycopene correlated with reduced hypertension in overweight and obese individuals, while another study by Han et al. [131] suggested that a higher serum concentration of lycopene was associated with a reduced risk of mortality in the population with metabolic syndrome. Another study by Polidori et al. [132] found that in the population with HF, the left ventricular ejection fraction was significantly and positively correlated with plasma lycopene levels, and increased consumption of antioxidant micronutrients, such as lycopene, might help in achieving cardiovascular health.
However, some studies have shown that lycopene exerted no beneficial effect on CVD risk [133,134]. Yet, an epidemiologic follow-up study by Ito et al. [135] found that high serum levels of lycopene were indeed associated with low hazard ratios for cardiovascular mortality. A systematic review and meta-analysis of 43 studies by Tierney et al. [136] found that the data on lycopene’s efficacy in improving cardiovascular risk were conflicting.
Role of different carotenoids in preventing CVD is presented in Table 2.

5. Novel Experimental Antioxidant Therapies

Imbalanced ROS production during intense oxidative stress leads to exacerbation of pathophysiological processes in humans [137]. In many CVDs, such as myocardial infarction, hypertension, atherosclerosis, myocardial hypertrophy, HF, and restenosis after angioplasty or venous bypass, excessive ROS production plays an important role in their development [138]. There is, therefore, great hope that antioxidants and substances with antioxidant activity might minimize the negative effects of ROS and, at the same time, help to improve the prognosis of patients with CVD.
Novel antioxidants include compounds that are activators of endogenous antioxidant defense systems, compounds that are inhibitors of oxidative stress generation, and compounds that enable functional repair of ROS-induced damage.
Activators of endogenous antioxidant defense systems include the activator NRF2, which is a basic transcription factor that recognizes an enhancer of the antioxidant response element. Its reduced expression and activity have been shown to predispose to the development of hypertension or atherosclerosis [139]. The drug targeting NRF2 is dimethyl fumarate (DMF) [140]. Experiments have shown that it reduces infarct size after ischemia/reperfusion injury [140] and has a protective role against cardiomyocytes after this injury [141]. DMF also reduced the development of atherosclerosis in an apolipoprotein-E-deficient study [142]. DMF was shown to prevent endothelial dysfunction [143].
Inhibitors of oxidative stress generation include drugs targeting Xo, NOX, and MPO, among others. Allopurinol is an Xo inhibitor that has shown beneficial effects in clinical trials in hypertension, HF, and ischemia/reperfusion injury by reducing oxidative stress in endothelial cells [144]. In a meta-analysis evaluating its effects on blood pressure, it showed a moderate reduction in systolic and diastolic blood pressure in patients [145], which may be related to its ability to improve endothelial function [146]. It was also shown to reduce in-hospital mortality and cardiac complications in patients undergoing primary percutaneous coronary intervention or coronary artery bypass grafting [147,148]. Allopurinol has been shown to improve myocardial oxygen consumption and myocardial blood flow [149]. It may improve exercise capacity in patients with chronic HF [146]. The use of allopurinol in patients with ischemic cardiomyopathy has been associated with a significant improvement in left ventricular ejection fraction and a reduction in left ventricular end-systolic volume [150].
GKT137831 is a NOX inhibitor in clinical trials. When used in ApoE knock-out mice, the drug had a potent anti-atherosclerotic effect [151]. It also improved cardiac function after ischemia/reperfusion injury [152]. In contrast, MPO inhibitors led to changes in the composition of atherosclerotic lesions and cardiac remodeling in mouse studies [153].
Compounds that enable functional repair of ROS-induced damage include compounds that affect nitric oxide–cyclic guanosine monophosphate (NO-cGMP) signaling, including HNO donors, such as CXL-1427, L-citrulline, or L-arginine [154]. In clinical trials, CXL-1427 showed a favorable safety profile and hemodynamic effects in HFrEF patients [155]. For L-arginine and L-citrulline, meta-analyses of randomized clinical trials showed that oral administration of these compounds was associated with reductions in systolic and diastolic blood pressure [156].

5.1. miRNA

miRNAs are involved in the oxidative stress response and play a key role in its regulation, and therefore represent important compounds for therapeutic intervention against various pathological conditions. Future clinical applications of miRNAs in the treatment of CVD include the use of induction (restoring miRNAs that have lost function) as well as inhibition of miRNA expression [157]. In addition, modifications or carriers must be used to increase the stability and bioavailability of the molecules [158,159].
During hypoxia, miRNA-210 levels are significantly increased, leading to improved cardiac function by promoting angiogenesis and inhibiting cardiomyocyte apoptosis [160,161,162]. Direct injection of miRNA-210 into the myocardium in an animal model of myocardial infarction resulted in improved myocardial function [163]. Circulating miRNA-210 levels were found to be significantly associated with mortality in patients with acute coronary syndrome [164].
The most abundant of these, which also plays a key role in muscle cell differentiation and proliferation, is miRNA-1. It is a regulator of cardiomyocyte growth and a pro-apoptotic factor in anemic myocardium, as observed in diseases such as hypertrophy, myocardial infarction, and arrhythmias [165,166,167]. Studies have shown that its overexpression is associated with increased ROS and decreased SOD production [168]. Furthermore, H2O2 increased miRNA-1 in cardiomyocytes in a rat model [168]. Increased miRNA-1 was associated with a significant reduction in infarct size [169] and its serum levels correlated with circulating troponin T, suggesting that it may be used as a biomarker for myocardial infarction [170,171]. Administration of miRNA-1 to mice after myocardial infarction improved myocardial function [172].
It has been observed that some miRNAs may play an important role in regulating atherosclerotic plaque formation. miRNA-133 was elevated in the presence of symptomatic atherosclerotic plaques and in patients with CVD [173,174]. Its inhibition mainly targets NOS and may prevent vascular endothelial dysfunction [175]. Regulation of endothelial NOS expression may also be influenced by miRNA-92a [176], which further reduces plaque inflammation and increases plaque stability by promoting endothelial cell proliferation and angiogenesis [177]. In the case of miRNA-206, which modulates VEGF expression, there is inhibition of viability and invasion and increased apoptosis of endothelial progenitor cells in patients with CAD [178]. In contrast, inhibition of miRNA-377 had a protective effect. It reduced myocardial fibrosis and improved myocardial function [179]. The functions of the miRNAs mentioned above are shown in Table 3.

5.2. Nanoparticles

Nanoparticles could have a groundbreaking impact on the treatment of CVD due to their size and properties, which allow them to be easily modified [180]. Among other things, research is testing H2O2-responsive nanoparticles that would target the site of ischemia/reperfusion injury in the myocardium. Such particles have shown potent anti-inflammatory and anti-apoptotic effects in various animal models, leading to a reduction in further organ damage [181]. Nanoparticles with antioxidant properties can be used in anticoagulant therapy, alongside existing thrombolytic agents, due to fibrin aggregation and increased H2O2 levels in thrombi [182]. This involves imaging the thrombus and then inhibiting its formation by scavenging H2O2. Other studies have also used nanoparticles to reduce oxidative stress by modifying its production or removal system. In one study, nanoparticles linked to the small-interfering RNA (siRNA) NOX2 were injected directly into the heart muscle of mice after a heart attack, resulting in improved heart function [183]. Studies are also focusing on the protective role of SOD, using nanoparticles capable of carrying it. When injected into the ischemic area of the myocardium in a rat model of ischemia/reperfusion, it resulted in reduced myocyte apoptosis and improved myocardial function [184]. Nanoparticles designed to carry N-acetylcysteine showed effective attenuation of myocardial fibrosis in rat models of ischemia/reperfusion [185]. In addition, selenium-based nanoparticles showed improved biological effects in ischemic cardiomyocytes due to their ROS-quenching properties [186]. In studies, nanoparticles attenuated ROS-induced inflammation and cell apoptosis in macrophages by scavenging intracellularly generated ROS, effectively preventing foam cell formation and reducing internalization of oxidized LDL [187]. Potential roles of nanoparticles in the prevention and treatment of CVDs are presented in Table 4.
The use of antioxidant therapies is mainly based on enhancing their effects through supplementation. This is related to the continuing failure of therapies designed to modulate oxidative stress. This may be due to non-selective modulation of ROS, which would interfere with physiological ROS-dependent signaling pathways, or to insufficient efficacy of modulation [134]. Moreover, the efficacy of therapy is hampered by the lack of available methods to quantify ROS and ROS damage in vivo in tissues and blood vessels, and a study design that considers patient differences in ROS-generating systems or cellular antioxidants [108,188]. It is also important to better understand the effects of oxidants and antioxidants in clinically relevant models of human disease [189]. Several alternative antioxidant-based experimental approaches are being developed, but further research and understanding of oxidative signaling is needed before they can be used in the treatment of CVD. Although there have been many animal studies with promising results, unfortunately, the results of randomized clinical trials do not support the positive effects of antioxidants on the cardiovascular system. Evidence from randomized clinical trials on the effectiveness of antioxidants in CVD, especially assessing their long-term effects, is still lacking.
Emerging therapies, however, represent a promising source for potential future clinical use. By regulating multiple genes involved in the response of biological systems to oxidative stress, miRNAs may represent biomarkers of CVD, including myocardial infarction, HF, or endothelial dysfunction. Modifying their expression may be a useful therapeutic option. In addition, the use of nanomaterials with unique ROS-regulating properties represents a promising option for the development of new therapeutic strategies for CVD.

6. Conclusions

ROS are small, highly reactive substances capable of oxidizing a wide range of molecules in the human body, including nucleic acids, proteins, lipids, carbohydrates, and even small inorganic compounds. The overproduction of ROS leads to oxidative stress, which constitutes a significant factor contributing to the development of diseases, especially cardiovascular.
CoQ10 inhibits the initiation of lipid peroxidation processes, thereby exerting a protective influence on the cardiac muscle and other tissues. Supplementation demonstrates favorable effects on cardiac function, correlating with a reduction in mortality and hospitalization rates in patients with HF. Moreover, it contributes to a reduction in inflammatory markers and total cholesterol levels concurrently increasing HDL-c concentrations. Evidence suggests that CoQ10 diminishes arterial blood pressure and mitigates vasoconstriction.
Effect of polyphenols on cardiovascular health are attributed to their direct antioxidant action, which involves scavenging ROS and reactive nitrogen species. Kaempferol might be used to lower the risk of atherosclerotic diseases by altering the expression of disease-related genes, such as MCP-1 or ICAM-1. It also inhibits ERK1/2 by downregulating cytokine receptors and reducing cardiac failure and hypertrophy. However, studies suggest that kaempferol may have mutagenic and genotoxic properties. Studies show that rutin improves cardiovascular structure and function, including hypertrophy, inflammation, and fibrosis. It prevents atherosclerosis by lowering total cholesterol, triglycerides, and insulin levels in rat blood. Resveratrol has been shown to improve in both systolic and diastolic function of the left ventricle as well as the left ventricular ejection fraction in patients with CHD.
Higher serum levels of carotenoids were correlated with a decreased risk of elevated serum NT-proBNP levels, which might indicate that carotenoids can partake in preventing cardiac overload. Moreover, they were associated with a reduced risk of atherosclerosis and inversely associated with inflammatory cytokines.
There is great hope that antioxidants and substances with antioxidant activity might minimize the negative effects of ROS and, at the same time, help to improve the prognosis of patients with CVD. Therefore, novel therapies based on antioxidant activity are researched, such as drugs targeting NRF2, which has a protective role against cardiomyocytes, XO inhibitors with a potential role in decreasing blood pressure, and NOX inhibitors with potent anti-atherosclerotic effects. In addition, more and more studies are emerging examining the potential impact of miRNAs on cardiovascular disease, but these remain in the animal phase. Similar is the case of nanoparticles, which have great potential but require further research.
Although there have been many animal studies with promising results, unfortunately, the results of randomized clinical trials do not support the positive effects of antioxidants on the cardiovascular system. Evidence from randomized clinical trials on the effectiveness of antioxidants in CVD, especially assessing their long-term effects, is still lacking. The effectiveness of antioxidant defenses is constrained by the role oxidative stress plays in disease pathology. Typically, oxidative stress is a secondary rather than a primary factor in disease, so mitigating it might not significantly alter disease progression. This limitation is frequently overlooked in clinical trials of antioxidants. Additionally, there is concern that compounds meant to boost antioxidant defenses may not achieve effective concentrations in vivo. Adopting a beneficial diet rich in antioxidant-containing foods over the long term is supported by evidence for reducing CVD risk factors. However, the same benefits are not conclusively seen with antioxidant supplements, whether these are short- or long-term clinical trials. The optimal timing for follow-up on the use of antioxidants to reduce cardiovascular disease risk factors remains uncertain and warrants further investigation due to the variability in outcomes observed across studies. For specific recommendations and duration, consulting with a healthcare provider is advisable.
This review has limitations. There was no specific selection criteria or research methodology used to identify sources and references for this review. In addition, due to the extent of the topic, the focus was on several major antioxidants and their potential role in the treatment of cardiovascular disease. The review has also strengths. We have presented overviews for the most known antioxidants, their chemical characteristics, and their impact on CVD diseases.

Author Contributions

Conceptualization, E.M., B.F., and J.R.; methodology, E.M., J.H., W.C., P.F., K.L., W.L., and G.M.; validation, E.M., B.F., and J.R.; formal analysis, E.M., J.H., W.C., P.F., K.L., W.L., and G.M.; investigation E.M., J.H., W.C., P.F., K.L., W.L., and G.M.; resources, E.M., B.F., and J.R.; data curation, E.M.; writing—original draft preparation, E.M., J.H., W.C., P.F., K.L., W.L., and G.M.; writing—review and editing, E.M.; visualization, E.M., J.H., W.C., P.F., K.L., W.L., and G.M.; supervision, E.M., B.F., and J.R.; project administration, E.M.; funding acquisition, B.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

The data used in this article were sourced from materials mentioned in the References Section.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Xu, T.; Ding, W.; Ji, X.; Ao, X.; Liu, Y.; Yu, W.; Wang, J. Oxidative Stress in Cell Death and Cardiovascular Diseases. Oxid. Med. Cell Longev. 2019, 2019, 9030563. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  2. Yan, Q.; Liu, S.; Sun, Y.; Chen, C.; Yang, S.; Lin, M.; Long, J.; Yao, J.; Lin, Y.; Yi, F.; et al. Targeting oxidative stress as a preventive and therapeutic approach for cardiovascular disease. J. Transl. Med. 2023, 21, 519. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Kaminsky, L.A.; German, C.; Imboden, M.; Ozemek, C.; Peterman, J.E.; Brubaker, P.H. The importance of healthy lifestyle behaviors in the prevention of cardiovascular disease. Prog. Cardiovasc. Dis. 2022, 70, 8–15. [Google Scholar] [CrossRef] [PubMed]
  4. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef] [PubMed]
  5. Kattoor, A.J.; Pothineni, N.V.K.; Palagiri, D.; Mehta, J.L. Oxidative Stress in Atherosclerosis. Curr. Atheroscler. Rep. 2017, 19, 42. [Google Scholar] [CrossRef] [PubMed]
  6. Masenga, S.K.; Kabwe, L.S.; Chakulya, M.; Kirabo, A. Mechanisms of Oxidative Stress in Metabolic Syndrome. Int. J. Mol. Sci. 2023, 24, 7898. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  7. Batty, M.; Bennett, M.R.; Yu, E. The Role of Oxidative Stress in Atherosclerosis. Cells 2022, 11, 3843. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  8. Amponsah-Offeh, M.; Diaba-Nuhoho, P.; Speier, S.; Morawietz, H. Oxidative Stress, Antioxidants and Hypertension. Antioxidants 2023, 12, 281. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  9. Senoner, T.; Dichtl, W. Oxidative Stress in Cardiovascular Diseases: Still a Therapeutic Target? Nutrients 2019, 11, 2090. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Halliwell, B. Understanding mechanisms of antioxidant action in health and disease. Nat. Rev. Mol. Cell Biol. 2024, 25, 13–33. [Google Scholar] [CrossRef] [PubMed]
  11. Pagan, L.U.; Gomes, M.J.; Gatto, M.; Mota, G.A.F.; Okoshi, K.; Okoshi, M.P. The Role of Oxidative Stress in the Aging Heart. Antioxidants 2022, 11, 336. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Steven, S.; Frenis, K.; Oelze, M.; Kalinovic, S.; Kuntic, M.; Bayo Jimenez, M.T.; Vujacic-Mirski, K.; Helmstädter, J.; Kröller-Schön, S.; Münzel, T.; et al. Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. Oxid. Med. Cell Longev. 2019, 2019, 7092151. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  13. Velena, A.; Zarkovic, N.; Gall Troselj, K.; Bisenieks, E.; Krauze, A.; Poikans, J.; Duburs, G. 1,4-Dihydropyridine Derivatives: Dihydronicotinamide Analogues-Model Compounds Targeting Oxidative Stress. Oxid. Med. Cell Longev. 2016, 2016, 1892412. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Sharma, G.N.; Gupta, G.; Sharma, P. A Comprehensive Review of Free Radicals, Antioxidants, and Their Relationship with Human Ailments. Crit. Rev. Eukaryot. Gene Expr. 2018, 28, 139–154. [Google Scholar] [CrossRef] [PubMed]
  15. Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709, Erratum in Nat. Rev. Drug Discov. 2021, 20, 652. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  16. Zozina, V.I.; Covantev, S.; Goroshko, O.A.; Krasnykh, L.M.; Kukes, V.G. Coenzyme Q10 in Cardiovascular and Metabolic Diseases: Current State of the Problem. Curr. Cardiol. Rev. 2018, 14, 164–174. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  17. Rabanal-Ruiz, Y.; Llanos-González, E.; Alcain, F.J. The Use of Coenzyme Q10 in Cardiovascular Diseases. Antioxidants 2021, 10, 755. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  18. Crane, F.L.; Hatefi, Y.; Lester, R.L.; Widmer, C. Isolation of a quinone from beef heart mitochondria. Biochim. Biophys. Acta 1957, 25, 220–221. [Google Scholar] [CrossRef] [PubMed]
  19. Sohal, R.S. Coenzyme Q and vitamin E interactions. Methods Enzymol. 2004, 378, 146–151. [Google Scholar] [CrossRef] [PubMed]
  20. Miles, M.V.; Horn, P.S.; Tang, P.H.; Morrison, J.A.; Miles, L.; DeGrauw, T.; Pesce, A.J. Age-related changes in plasma coenzyme Q10 concentrations and redox state in apparently healthy children and adults. Clin. Chim. Acta 2004, 347, 139–144. [Google Scholar] [CrossRef] [PubMed]
  21. Gutierrez-Mariscal, F.M.; de la Cruz-Ares, S.; Torres-Peña, J.D.; Alcalá-Diaz, J.F.; Yubero-Serrano, E.M.; López-Miranda, J. Coenzyme Q10 and Cardiovascular Diseases. Antioxidants 2021, 10, 906. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Bergamini, C.; Cicoira, M.; Rossi, A.; Vassanelli, C. Oxidative stress and hyperuricaemia: Pathophysiology, clinical relevance, and therapeutic implications in chronic heart failure. Eur. J. Heart Fail. 2009, 11, 444–452. [Google Scholar] [CrossRef]
  23. Lim, J.Y.; Park, S.J.; Hwang, H.Y.; Park, E.J.; Nam, J.H.; Kim, J.; Park, S.I. TGF-beta1 induces cardiac hypertrophic responses via PKC-dependent ATF-2 activation. J. Mol. Cell Cardiol. 2005, 39, 627–636. [Google Scholar] [CrossRef] [PubMed]
  24. Turunen, M.; Olsson, J.; Dallner, G. Metabolism and function of coenzyme Q. Biochim. Biophys. Acta 2004, 1660, 171–199. [Google Scholar] [CrossRef] [PubMed]
  25. Alarcón-Vieco, E.; Martínez-García, I.; Sequí-Domínguez, I.; Rodríguez-Gutiérrez, E.; Moreno-Herráiz, N.; Pascual-Morena, C. Effect of coenzyme Q10 on cardiac function and survival in heart failure: An overview of systematic reviews and meta-analyses. Food Funct. 2023, 14, 6302–6311. [Google Scholar] [CrossRef] [PubMed]
  26. Mortensen, S.A.; Rosenfeldt, F.; Kumar, A.; Dolliner, P.; Filipiak, K.J.; Pella, D.; Alehagen, U.; Steurer, G.; Littarru, G.P.; Q-SYMBIO Study Investigators. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: Results from Q-SYMBIO: A randomized double-blind trial. JACC Heart Fail. 2014, 2, 641–649. [Google Scholar] [CrossRef]
  27. Fladerer, J.P.; Grollitsch, S. Comparison of Coenzyme Q10 (Ubiquinone) and Reduced Coenzyme Q10 (Ubiquinol) as Supplement to Prevent Cardiovascular Disease and Reduce Cardiovascular Mortality. Curr. Cardiol. Rep. 2023, 25, 1759–1767. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. van Campen, L.C.; Visser, F.C.; Visser, C.A. Ejection fraction improvement by beta-blocker treatment in patients with heart failure: An analysis of studies published in the literature. J. Cardiovasc. Pharmacol. 1998, 32 (Suppl. 1), S31–S35. [Google Scholar] [CrossRef] [PubMed]
  29. Libby, P.; Theroux, P. Pathophysiology of coronary artery disease. Circulation 2005, 111, 3481–3488. [Google Scholar] [CrossRef] [PubMed]
  30. Jorat, M.V.; Tabrizi, R.; Kolahdooz, F.; Akbari, M.; Salami, M.; Heydari, S.T.; Asemi, Z. The effects of coenzyme Q10 supplementation on biomarkers of inflammation and oxidative stress in among coronary artery disease: A systematic review and meta-analysis of randomized controlled trials. Inflammopharmacology 2019, 27, 233–248. [Google Scholar] [CrossRef] [PubMed]
  31. Jorat, M.V.; Tabrizi, R.; Mirhosseini, N.; Lankarani, K.B.; Akbari, M.; Heydari, S.T.; Mottaghi, R.; Asemi, Z. The effects of coenzyme Q10 supplementation on lipid profiles among patients with coronary artery disease: A systematic review and meta-analysis of randomized controlled trials. Lipids Health Dis. 2018, 17, 230. [Google Scholar] [CrossRef]
  32. Lee, Y.J.; Cho, W.J.; Kim, J.K.; Lee, D.C. Effects of coenzyme Q10 on arterial stiffness, metabolic parameters, and fatigue in obese subjects: A double-blind randomized controlled study. J. Med. Food 2011, 14, 386–390. [Google Scholar] [CrossRef] [PubMed]
  33. Sharifi, N.; Tabrizi, R.; Moosazadeh, M.; Mirhosseini, N.; Lankarani, K.B.; Akbari, M.; Chamani, M.; Kolahdooz, F.; Asemi, Z. The Effects of Coenzyme Q10 Supplementation on Lipid Profiles Among Patients with Metabolic Diseases: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Curr. Pharm. Des. 2018, 24, 2729–2742. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, Z.; Tian, Z.; Zhao, D.; Liang, Y.; Dai, S.; Liu, M.; Hou, S.; Dong, X.; Zhaxinima Yang, Y. Effects of Coenzyme Q10 Supplementation on Lipid Profiles in Adults: A Meta-analysis of Randomized Controlled Trials. J. Clin. Endocrinol. Metab. 2022, 108, 232–249. [Google Scholar] [CrossRef] [PubMed]
  35. An, P.; Wan, S.; Luo, Y.; Luo, J.; Zhang, X.; Zhou, S.; Xu, T.; He, J.; Mechanick, J.I.; Wu, W.C.; et al. Micronutrient Supplementation to Reduce Cardiovascular Risk. J. Am. Coll. Cardiol. 2022, 80, 2269–2285. [Google Scholar] [CrossRef] [PubMed]
  36. Digiesi, V.; Cantini, F.; Oradei, A.; Bisi, G.; Guarino, G.C.; Brocchi, A.; Bellandi, F.; Mancini, M.; Littarru, G.P. Coenzyme Q10 in essential hypertension. Mol. Aspects Med. 1994, 15, s257–s263. [Google Scholar] [CrossRef] [PubMed]
  37. Flowers, N.; Hartley, L.; Todkill, D.; Stranges, S.; Rees, K. Co-enzyme Q10 supplementation for the primary prevention of cardiovascular disease. Cochrane Database Syst. Rev. 2014, 2014, CD010405. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Mohseni, M.; Vafa, M.R.; Hajimiresmail, S.J.; Zarrati, M.; Rahimi-Forushani, A.; Bitarafan, V.; Shidfar, F. Effects of coenzyme q10 supplementation on serum lipoproteins, plasma fibrinogen, and blood pressure in patients with hyperlipidemia and myocardial infarction. Iran. Red. Crescent Med. J. 2014, 16, e16433. [Google Scholar] [CrossRef] [PubMed]
  39. Zhao, D.; Liang, Y.; Dai, S.; Hou, S.; Liu, Z.; Liu, M.; Dong, X.; Zhan, Y.; Tian, Z.; Yang, Y. Dose-Response Effect of Coenzyme Q10 Supplementation on Blood Pressure among Patients with Cardiometabolic Disorders: A Grading of Recommendations Assessment, Development, and Evaluation (GRADE)-Assessed Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2022, 13, 2180–2194. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  40. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef]
  41. Rodrigo, R.; Gil, D.; Miranda-Merchak, A.; Kalantzidis, G. Antihypertensive role of polyphenols. Adv. Clin. Chem. 2012, 58, 225–254. [Google Scholar] [CrossRef] [PubMed]
  42. Tangney, C.C.; Rasmussen, H.E. Polyphenols, inflammation, and cardiovascular disease. Curr. Atheroscler. Rep. 2013, 15, 324. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  43. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Important Flavonoids and Their Role as a Therapeutic Agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  45. Paramita, V.; Kusumayanti, H.; Amalia, R.; Leviana, W.; Nisa, Q.A. Application of Flavonoid and Anthocyanin Contents from Rambutan (Nephelium lappaceum) Peel as Natural Dyes on Cotton Fabric. Adv. Sci. Lett. 2018, 24, 9853–9855. [Google Scholar] [CrossRef]
  46. Zhao, L.; Yuan, X.; Wang, J.; Feng, Y.; Ji, F.; Li, Z.; Bian, J. A review on flavones targeting serine/threonine protein kinases for potential anticancer drugs. Bioorganic Med. Chem. 2019, 27, 677–685. [Google Scholar] [CrossRef]
  47. Lagunas-Herrera, H.; Tortoriello, J.; Herrera-Ruiz, M.; Martínez-Henández, G.B.; Zamilpa, A.; Santamaría, L.A.; Lorenzana, M.G.; Lombardo-Earl, G.; Jiménez-Ferrer, E. Acute and Chronic Antihypertensive Effect of Fractions, Tiliroside and Scopoletin from Malva parviflora. Biol. Pharm. Bull 2019, 42, 18–25. [Google Scholar] [CrossRef]
  48. Zang, Z.; Tang, S.; Li, Z.; Chou, S.; Shu, C.; Chen, Y.; Chen, W.; Yang, S.; Yang, Y.; Tian, J.; et al. An updated review on the stability of anthocyanins regarding the interaction with food proteins and polysaccharides. Compr. Rev. Food Sci. Food Saf. 2022, 21, 4378–4401. [Google Scholar] [CrossRef] [PubMed]
  49. Hofer, S.; Geisler, S.; Lisandrelli, R.; Ngoc, H.N.; Ganzera, M.; Schennach, H.; Fuchs, D.; Fuchs, J.E.; Gostner, J.M.; Kurz, K. Pharmacological Targets of Kaempferol Within Inflammatory Pathways—A Hint Towards the Central Role of Tryptophan Metabolism. Antioxidants 2020, 9, 180. [Google Scholar] [CrossRef]
  50. Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry to medicine. Pharmacol. Res. 2015, 99, 1–10. [Google Scholar] [CrossRef] [PubMed]
  51. Rajendran, P.; Rengarajan, T.; Nandakumar, N.; Palaniswami, R.; Nishigaki, Y.; Nishigaki, I. Kaempferol, a potential cytostatic and cure for inflammatory disorders. Eur. J. Med. Chem. 2014, 86, 103–112. [Google Scholar] [CrossRef] [PubMed]
  52. Chang, T.K.; Chen, J.; Yeung, E.Y. Effect of Ginkgo biloba extract on procarcinogen-bioactivating human CYP1 enzymes: Identification of isorhamnetin, kaempferol, and quercetin as potent inhibitors of CYP1B1. Toxicol. Appl. Pharmacol. 2006, 213, 18–26. [Google Scholar] [CrossRef] [PubMed]
  53. Su, R.; Jin, X.; Zhao, W.; Wu, X.; Zhai, F.; Li, Z. Rutin ameliorates the promotion effect of fine particulate matter on vascular calcification in calcifying vascular cells and ApoE−/− mice. Ecotoxicol. Environ. Saf. 2022, 234, 113410. [Google Scholar] [CrossRef] [PubMed]
  54. Nishikawa, M.; Kada, Y.; Kimata, M.; Sakaki, T.; Ikushiro, S. Comparison of metabolism and biological properties among positional isomers of quercetin glucuronide in LPS- and RANKL-challenged RAW264.7 cells. Biosci. Biotechnol. Biochem. 2022, 86, 1670–1679. [Google Scholar] [CrossRef] [PubMed]
  55. Day, A.J.; Mellon, F.; Barron, D.; Sarrazin, G.; Morgan, M.R.; Williamson, G. Human metabolism of dietary flavonoids: Identification of plasma metabolites of quercetin. Free Radical Res. 2001, 35, 941–952. [Google Scholar] [CrossRef]
  56. Bussmann, A.J.C.; Zaninelli, T.H.; Saraiva-Santos, T.; Fattori, V.; Guazelli, C.F.S.; Bertozzi, M.M.; Andrade, K.C.; Ferraz, C.R.; Camilios-Neto, D.; Casella, A.M.B.; et al. The Flavonoid Hesperidin Methyl Chalcone Targets Cytokines and Oxidative Stress to Reduce Diclofenac-Induced Acute Renal Injury: Contribution of the Nrf2 Redox-Sensitive Pathway. Antioxidants 2022, 11, 1261. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Ni, H.; Li, J.; Zheng, J.; Zhou, B. Cardamonin attenuates cerebral ischemia/reperfusion injury by activating the HIF-1α/VEGFA pathway. Phytother. Res. 2022, 36, 1736–1747. [Google Scholar] [CrossRef] [PubMed]
  58. Zhang, T.; Zhang, C.; Luo, Y.; Liu, S.; Li, S.; Li, L.; Ma, Y.; Liu, J. Protective effect of rutin on spinal motor neuron in rats exposed to acrylamide and the underlying mechanism. Neurotoxicology 2023, 95, 127–135. [Google Scholar] [CrossRef] [PubMed]
  59. Xia, N.; Daiber, A.; Förstermann, U.; Li, H. Antioxidant effects of resveratrol in the cardiovascular system. Br. J. Pharmacol. 2017, 174, 1633–1646. [Google Scholar] [CrossRef]
  60. Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef]
  61. Kulkarni, S.S.; Cantó, C. The molecular targets of resveratrol. Biochim. Biophys. Acta 2015, 1852, 1114–1123. [Google Scholar] [CrossRef] [PubMed]
  62. Chekalina, N.I. Resveratrol has a positive effect on parameters of central hemodynamics and myocardial ischemia in patients with stable coronary heart disease. Wiad. Lek. 2017, 70 Pt 2, 286–291. [Google Scholar] [PubMed]
  63. Chen, H.-E.; Lin, Y.-J.; Lin, I.-C.; Yu, H.-R.; Sheen, J.-M.; Tsai, C.-C.; Huang, L.-T.; Tain, Y.-L. Resveratrol prevents combined prenatal NG-nitro-L-arginine-methyl ester (L-NAME) treatment plus postnatal high-fat diet induced programmed hypertension in adult rat offspring: Interplay between nutrient-sensing signals, oxidative stress and gut microbiota. J. Nutr. Biochem. 2019, 70, 28–37. [Google Scholar] [CrossRef] [PubMed]
  64. Tain, Y.L.; Lee, W.C.; Wu, K.L.H.; Leu, S.; Chan, J.Y.H. Resveratrol Prevents the Development of Hypertension Programmed by Maternal Plus Post-Weaning High-Fructose Consumption through Modulation of Oxidative Stress, Nutrient-Sensing Signals, and Gut Microbiota. Mol. Nutr. Food Res. 2018, 62, e1800066. [Google Scholar] [CrossRef] [PubMed]
  65. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; García-Almagro, F.J.; Avilés-Plaza, F.; Parra, S.; Yáñez-Gascón, M.J.; Ruiz-Ros, J.A.; García-Conesa, M.T.; Tomás-Barberán, F.A.; et al. Consumption of a grape extract supplement containing resveratrol decreases oxidized LDL and ApoB in patients undergoing primary prevention of cardiovascular disease: A triple-blind, 6-month follow-up, placebo-controlled, randomized trial. Mol. Nutr. Food Res. 2012, 56, 810–821. [Google Scholar] [CrossRef] [PubMed]
  66. Tomé-Carneiro, J.; Gonzálvez, M.; Larrosa, M.; Yáñez-Gascón, M.J.; García-Almagro, F.J.; Ruiz-Ros, J.A.; Conesa, M.T.G.; Tomás-Barberán, F.A.; Espín, J.C. One-year consumption of a grape nutraceutical containing resveratrol improves the inflammatory and fibrinolytic status of patients in primary prevention of cardiovascular disease. Am. J. Cardiol. 2012, 110, 356–363. [Google Scholar] [CrossRef] [PubMed]
  67. De Taeye, B.; Smith, L.H.; Vaughan, D.E. Plasminogen activator inhibitor-1: A common denominator in obesity, diabetes and cardiovascular disease. Curr. Opin. Pharmacol. 2005, 5, 149–154. [Google Scholar] [CrossRef] [PubMed]
  68. Zamora-Ros, R.; Urpi-Sarda, M.; Lamuela-Raventós, R.M.; Martínez-González, M.; Salas-Salvadó, J.; Arós, F.; Fitó, M.; Lapetra, J.; Estruch, R.; Andres-Lacueva, C. High urinary levels of resveratrol metabolites are associated with a reduction in the prevalence of cardiovascular risk factors in high-risk patients. Pharmacol. Res. 2012, 65, 615–620. [Google Scholar] [CrossRef]
  69. Gal, R.; Deres, L.; Horvath, O.; Eros, K.; Sandor, B.; Urban, P.; Soos, S.; Marton, Z.; Sumegi, B.; Toth, K.; et al. Resveratrol Improves Heart Function by Moderating Inflammatory Processes in Patients with Systolic Heart Failure. Antioxidants 2020, 9, 1108. [Google Scholar] [CrossRef]
  70. Pannu, N.; Bhatnagar, A. Resveratrol: From enhanced biosynthesis and bioavailability to multitargeting chronic diseases. Biomed. Pharmacother. 2019, 109, 2237–2251. [Google Scholar] [CrossRef]
  71. Gambini, J.; Inglés, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. Properties of Resveratrol: In Vitro and In Vivo Studies about Metabolism, Bioavailability, and Biological Effects in Animal Models and Humans. Oxid. Med. Cell Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef] [PubMed]
  72. Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed]
  73. Chimento, A.; De Amicis, F.; Sirianni, R.; Sinicropi, M.S.; Puoci, F.; Casaburi, I.; Saturnino, C.; Pezzi, V. Progress to Improve Oral Bioavailability and Beneficial Effects of Resveratrol. Int. J. Mol. Sci. 2019, 20, 1381. [Google Scholar] [CrossRef]
  74. Shi, G.; Rao, L.; Yu, H.; Xiang, H.; Yang, H.; Ji, R. Stabilization and encapsulation of pho tosensitive resveratrol within yeast cell. Int. J. Pharm. 2008, 349, 83–93. [Google Scholar] [CrossRef] [PubMed]
  75. Neves, A.R.; Lúcio, M.; Martins, S.; Lima, J.L.; Reis, S. Novel resveratrol nanodelivery systems based on lipid nanoparticles to enhance its oral bioavailability. Int. J. Nanomed. 2013, 8, 177–187. [Google Scholar] [CrossRef]
  76. Molfino, A.; Gioia, G.; Rossi Fanelli, F.; Muscaritoli, M. The role for dietary omega-3 fatty acids supplementation in older adults. Nutrients 2014, 6, 4058–4073. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  77. Allen, R.R.; Carson, L.; Kwik-Uribe, C.; Evans, E.M.; Erdman, J.W., Jr. Daily consumption of a dark chocolate containing flavanols and added sterol esters affects cardiovascular risk factors in a normotensive population with elevated cholesterol. J. Nutr. 2008, 138, 725–731. [Google Scholar] [CrossRef] [PubMed]
  78. Hosseini, A.; Razavi, B.M.; Banach, M.; Hosseinzadeh, H. Quercetin and metabolic syndrome: A review. Phytother. Res. 2021, 35, 5352–5364. [Google Scholar] [CrossRef] [PubMed]
  79. Abudureheman, B.; Yu, X.; Fang, D.; Zhang, H. Enzymatic Oxidation of Tea Catechins and Its Mechanism. Molecules 2022, 27, 942. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  80. Aherne, S.A.; O’Brien, N.M. Dietary flavonols: Chemistry, food content, and metabolism. Nutrition 2002, 18, 75–81. [Google Scholar] [CrossRef] [PubMed]
  81. Magyar, K.; Halmosi, R.; Palfi, A.; Feher, G.; Czopf, L.; Fulop, A.; Battyany, I.; Sumegi, B.; Toth, K.; Szabados, E. Cardioprotection by resveratrol: A human clinical trial in patients with stable coronary artery disease. Clin. Hemorheol. Microcirc. 2012, 50, 179–187. [Google Scholar] [CrossRef]
  82. Milani, A.; Basirnejad, M.; Shahbazi, S.; Bolhassani, A. Carotenoids: Biochemistry, pharmacology and treatment. Br. J. Pharmacol. 2017, 174, 1290–1324. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Tapiero, H.; Townsend, D.M.; Tew, K.D. The role of carotenoids in the prevention of human pathologies. Biomed. Pharmacother. 2004, 58, 100–110. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Parker, R.S. Carotenoids in human blood and tissues. J. Nutr. 1989, 119, 101–104. [Google Scholar] [CrossRef] [PubMed]
  85. Stahl, W.; Sies, H. Bioactivity and protective effects of natural carotenoids. Biochim. Biophys. Acta 2005, 1740, 101–107. [Google Scholar] [CrossRef] [PubMed]
  86. Saini, R.K.; Nile, S.H.; Park, S.W. Carotenoids from fruits and vegetables: Chemistry, analysis, occurrence, bioavailability and biological activities. Food Res. Int. 2015, 76 Pt 3, 735–750. [Google Scholar] [CrossRef] [PubMed]
  87. Tan, B.L.; Norhaizan, M.E. Carotenoids: How Effective Are They to Prevent Age-Related Diseases? Molecules 2019, 24, 1801. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  88. Fiedor, J.; Burda, K. Potential role of carotenoids as antioxidants in human health and disease. Nutrients 2014, 6, 466–488. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Rutz, J.K.; Borges, C.D.; Zambiazi, R.C.; da Rosa, C.G.; da Silva, M.M. Elaboration of microparticles of carotenoids from natural and synthetic sources for applications in food. Food Chem. 2016, 202, 324–333. [Google Scholar] [CrossRef] [PubMed]
  90. Nishino, A.; Yasui, H.; Maoka, T. Reaction of Paprika Carotenoids, Capsanthin and Capsorubin, with Reactive Oxygen Species. J. Agric. Food Chem. 2016, 64, 4786–4792. [Google Scholar] [CrossRef] [PubMed]
  91. El-Agamey, A.; Lowe, G.M.; McGarvey, D.J.; Mortensen, A.; Phillip, D.M.; Truscott, T.G.; Young, A.J. Carotenoid radical chemistry and antioxidant/pro-oxidant properties. Arch. Biochem. Biophys. 2004, 430, 37–48. [Google Scholar] [CrossRef] [PubMed]
  92. Young, A.J.; Lowe, G.M. Antioxidant and prooxidant properties of carotenoids. Arch. Biochem. Biophys. 2001, 385, 20–27. [Google Scholar] [CrossRef] [PubMed]
  93. Parker, R.S. Absorption, metabolism, and transport of carotenoids. FASEB J. 1996, 10, 542–551. [Google Scholar] [CrossRef] [PubMed]
  94. Erdman, J.W., Jr.; Bierer, T.L.; Gugger, E.T. Absorption and transport of carotenoids. Ann. N. Y Acad. Sci. 1993, 691, 76–85. [Google Scholar] [CrossRef] [PubMed]
  95. Rao, A.V.; Rao, L.G. Carotenoids and human health. Pharmacol. Res. 2007, 55, 207–216. [Google Scholar] [CrossRef] [PubMed]
  96. Stahl, W.; Schwarz, W.; Sundquist, A.R.; Sies, H. cis-trans isomers of lycopene and beta-carotene in human serum and tissues. Arch. Biochem. Biophys. 1992, 294, 173–177. [Google Scholar] [CrossRef] [PubMed]
  97. Darvin, M.E.; Sterry, W.; Lademann, J.; Vergou, T. The role of carotenoids in human skin. Molecules 2011, 16, 10491–10506. [Google Scholar] [CrossRef]
  98. Gammone, M.A.; Pluchinotta, F.R.; Bergante, S.; Tettamanti, G.; D’Orazio, N. Prevention of cardiovascular diseases with Carotenoids. Front Biosci. 2017, 9, 165–171. [Google Scholar] [CrossRef] [PubMed]
  99. Gammone, M.A.; Riccioni, G.; D’Orazio, N. Carotenoids: Potential allies of cardiovascular health? Food Nutr. Res. 2015, 59, 26762. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  100. Seifried, H.E.; Anderson, D.E.; Fisher, E.I.; Milner, J.A. A review of the interaction among dietary antioxidants and reactive oxygen species. J. Nutr. Biochem. 2007, 18, 567–579. [Google Scholar] [CrossRef] [PubMed]
  101. Witztum, J.L. The oxidation hypothesis of atherosclerosis. Lancet 1994, 344, 793–795. [Google Scholar] [CrossRef] [PubMed]
  102. Salonen, J.T.; Ylä-Herttuala, S.; Yamamoto, R.; Butler, S.; Korpela, H.; Salonen, R.; Nyyssönen, K.; Palinski, W.; Witztum, J.L. Autoantibody against oxidised LDL and progression of carotid atherosclerosis. Lancet 1992, 339, 883–887. [Google Scholar] [CrossRef] [PubMed]
  103. Lidebjer, C.; Leanderson, P.; Ernerudh, J.; Jonasson, L. Low plasma levels of oxygenated carotenoids in patients with coronary artery disease. Nutr. Metab. Cardiovasc. Dis. 2007, 17, 448–456. [Google Scholar] [CrossRef] [PubMed]
  104. Rissanen, T.; Voutilainen, S.; Nyyssönen, K.; Salonen, R.; Salonen, J.T. Low plasma lycopene concentration is associated with increased intima-media thickness of the carotid artery wall. Arterioscler. Thromb. Vasc. Biol. 2000, 20, 2677–2681. [Google Scholar] [CrossRef] [PubMed]
  105. Suzuki, K.; Ishii, J.; Kitagawa, F.; Kuno, A.; Kusuhara, Y.; Ochiai, J.; Ichino, N.; Osakabe, K.; Sugimoto, K.; Yamada, H.; et al. Association of serum carotenoid levels with N-terminal pro-brain-type natriuretic peptide: A cross-sectional study in Japan. J. Epidemiol. 2013, 23, 163–168. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Koh, W.P.; Yuan, J.M.; Wang, R.; Lee, Y.P.; Lee, B.L.; Yu, M.C.; Ong, C.N. Plasma carotenoids and risk of acute myocardial infarction in the Singapore Chinese Health Study. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 685–690. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Akbaraly, T.N.; Fontbonne, A.; Favier, A.; Berr, C. Plasma carotenoids and onset of dysglycemia in an elderly population: Results of the Epidemiology of Vascular Ageing Study. Diabetes Care 2008, 31, 1355–1359. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  108. Hozawa, A.; Jacobs, D.R. Jr.; Steffes, M.W.; Gross, M.D.; Steffen, L.M.; Lee, D.H. Circulating carotenoid concentrations and incident hypertension: The Coronary Artery Risk Development in Young Adults (CARDIA) study. J. Hypertens. 2009, 27, 237–242. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  109. Xu, X.R.; Zou, Z.Y.; Huang, Y.M.; Xiao, X.; Ma, L.; Lin, X.M. Serum carotenoids in relation to risk factors for development of atherosclerosis. Clin. Biochem. 2012, 45, 1357–1361. [Google Scholar] [CrossRef] [PubMed]
  110. Ciccone, M.M.; Cortese, F.; Gesualdo, M.; Carbonara, S.; Zito, A.; Ricci, G.; De Pascalis, F.; Scicchitano, P.; Riccioni, G. Dietary intake of carotenoids and their antioxidant and anti-inflammatory effects in cardiovascular care. Mediators Inflamm. 2013, 2013, 782137. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  111. Goulinet, S.; Chapman, M.J. Plasma LDL and HDL subspecies are heterogenous in particle content of tocopherols and oxygenated and hydrocarbon carotenoids. Relevance to oxidative resistance and atherogenesis. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 786–796. [Google Scholar] [CrossRef] [PubMed]
  112. Jialal, I.; Norkus, E.P.; Cristol, L.; Grundy, S.M. beta-Carotene inhibits the oxidative modification of low-density lipoprotein. Biochim. Biophys. Acta 1991, 1086, 134–138. [Google Scholar] [CrossRef] [PubMed]
  113. Shaish, A.; Daugherty, A.; O’Sullivan, F.; Schonfeld, G.; Heinecke, J.W. Beta-carotene inhibits atherosclerosis in hypercholesterolemic rabbits. J. Clin. Investig. 1995, 96, 2075–2082. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  114. Di Tomo, P.; Canali, R.; Ciavardelli, D.; Di Silvestre, S.; De Marco, A.; Giardinelli, A.; Pipino, C.; Di Pietro, N.; Virgili, F.; Pandolfi, A. β-Carotene and lycopene affect endothelial response to TNF-α reducing nitro-oxidative stress and interaction with monocytes. Mol. Nutr. Food Res. 2012, 56, 217–227. [Google Scholar] [CrossRef] [PubMed]
  115. D’Odorico, A.; Martines, D.; Kiechl, S.; Egger, G.; Oberhollenzer, F.; Bonvicini, P.; Sturniolo, G.C.; Naccarato, R.; Willeit, J. High plasma levels of alpha- and beta-carotene are associated with a lower risk of atherosclerosis: Results from the Bruneck study. Atherosclerosis 2000, 153, 231–239. [Google Scholar] [CrossRef] [PubMed]
  116. Karppi, J.; Laukkanen, J.A.; Mäkikallio, T.H.; Ronkainen, K.; Kurl, S. Low β-carotene concentrations increase the risk of cardiovascular disease mortality among Finnish men with risk factors. Nutr. Metab. Cardiovasc. Dis. 2012, 22, 921–928. [Google Scholar] [CrossRef] [PubMed]
  117. Karppi, J.; Laukkanen, J.A.; Mäkikallio, T.H.; Ronkainen, K.; Kurl, S. Serum β-carotene and the risk of sudden cardiac death in men: A population-based follow-up study. Atherosclerosis 2013, 226, 172–177. [Google Scholar] [CrossRef] [PubMed]
  118. Street, D.A.; Comstock, G.W.; Salkeld, R.M.; Schüep, W.; Klag, M.J. Serum antioxidants and myocardial infarction. Are low levels of carotenoids and alpha-tocopherol risk factors for myocardial infarction? Circulation 1994, 90, 1154–1161. [Google Scholar] [CrossRef] [PubMed]
  119. Hennekens, C.H.; Buring, J.E.; Manson, J.E.; Stampfer, M.; Rosner, B.; Cook, N.R.; Belanger, C.; LaMotte, F.; Gaziano, J.M.; Ridker, P.M.; et al. Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease. N. Engl. J. Med. 1996, 334, 1145–1149. [Google Scholar] [CrossRef] [PubMed]
  120. Omenn, G.S.; Goodman, G.E.; Thornquist, M.D.; Balmes, J.; Cullen, M.R.; Glass, A.; Keogh, J.P.; Meyskens, F.L.; Valanis, B.; Williams, J.H.; et al. Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N. Engl. J. Med. 1996, 334, 1150–1155. [Google Scholar] [CrossRef] [PubMed]
  121. Kaliora, A.C.; Dedoussis, G.V.; Schmidt, H. Dietary antioxidants in preventing atherogenesis. Atherosclerosis 2006, 187, 1–17. [Google Scholar] [CrossRef] [PubMed]
  122. Hu, M.Y.; Li, Y.L.; Jiang, C.H.; Liu, Z.Q.; Qu, S.L.; Huang, Y.M. Comparison of lycopene and fluvastatin effects on atherosclerosis induced by a high-fat diet in rabbits. Nutrition 2008, 24, 1030–1038. [Google Scholar] [CrossRef] [PubMed]
  123. Sultan Alvi, S.; Ansari, I.A.; Khan, I.; Iqbal, J.; Khan, M.S. Potential role of lycopene in targeting proprotein convertase subtilisin/kexin type-9 to combat hypercholesterolemia. Free Radic. Biol. Med. 2017, 108, 394–403. [Google Scholar] [CrossRef] [PubMed]
  124. Thies, F.; Mills, L.M.; Moir, S.; Masson, L.F. Cardiovascular benefits of lycopene: Fantasy or reality? Proc. Nutr. Soc. 2016, 76, 122–129. [Google Scholar] [CrossRef] [PubMed]
  125. McEneny, J.; Wade, L.; Young, I.S.; Masson, L.; Duthie, G.; McGinty, A.; McMaster, C.; Thies, F. Lycopene intervention reduces inflammation and improves HDL functionality in moderately overweight middle-aged individuals. J. Nutr. Biochem. 2013, 24, 163–168. [Google Scholar] [CrossRef] [PubMed]
  126. Slivnick, J.; Lampert, B.C. Hypertension and Heart Failure. Heart Fail. Clin. 2019, 15, 531–541. [Google Scholar] [CrossRef] [PubMed]
  127. Rissanen, T.H.; Voutilainen, S.; Nyyssönen, K.; Salonen, R.; Kaplan, G.A.; Salonen, J.T. Serum lycopene concentrations and carotid atherosclerosis: The Kuopio Ischaemic Heart Disease Risk Factor Study. Am. J. Clin. Nutr. 2003, 77, 133–138. [Google Scholar] [CrossRef] [PubMed]
  128. Klipstein-Grobusch, K.; Launer, L.J.; Geleijnse, J.M.; Boeing, H.; Hofman, A.; Witteman, J.C. Serum carotenoids and atherosclerosis. The Rotterdam Study. Atherosclerosis 2000, 148, 49–56. [Google Scholar] [CrossRef] [PubMed]
  129. Rissanen, T.H.; Voutilainen, S.; Nyyssönen, K.; Lakka, T.A.; Sivenius, J.; Salonen, R.; Kaplan, G.A.; Salonen, J.T. Low serum lycopene concentration is associated with an excess incidence of acute coronary events and stroke: The Kuopio Ischaemic Heart Disease Risk Factor Study. Br. J. Nutr. 2001, 85, 749–754. [Google Scholar] [CrossRef] [PubMed]
  130. Sesso, H.D.; Buring, J.E.; Norkus, E.P.; Gaziano, J.M. Plasma lycopene, other carotenoids, and retinol and the risk of cardiovascular disease in women. Am. J. Clin. Nutr. 2004, 79, 47–53. [Google Scholar] [CrossRef] [PubMed]
  131. Kim, J.Y.; Paik, J.K.; Kim, O.Y.; Park, H.W.; Lee, J.H.; Jang, Y.; Lee, J.H. Effects of lycopene supplementation on oxidative stress and markers of endothelial function in healthy men. Atherosclerosis 2011, 215, 189–195. [Google Scholar] [CrossRef] [PubMed]
  132. Han, G.M.; Meza, J.L.; Soliman, G.A.; Islam, K.M.; Watanabe-Galloway, S. Higher levels of serum lycopene are associated with reduced mortality in individuals with metabolic syndrome. Nutr. Res. 2016, 36, 402–407. [Google Scholar] [CrossRef] [PubMed]
  133. Polidori, M.C.; Savino, K.; Alunni, G.; Freddio, M.; Senin, U.; Sies, H.; Stahl, W.; Mecocci, P. Plasma lipophilic antioxidants and malondialdehyde in congestive heart failure patients: Relationship to disease severity. Free Radic. Biol. Med. 2002, 32, 148–152. [Google Scholar] [CrossRef] [PubMed]
  134. Sesso, H.D.; Liu, S.; Gaziano, J.M.; Buring, J.E. Dietary lycopene, tomato-based food products and cardiovascular disease in women. J. Nutr. 2003, 133, 2336–2341. [Google Scholar] [CrossRef] [PubMed]
  135. Sesso, H.D.; Buring, J.E.; Norkus, E.P.; Gaziano, J.M. Plasma lycopene, other carotenoids, and retinol and the risk of cardiovascular disease in men. Am. J. Clin. Nutr. 2005, 81, 990–997. [Google Scholar] [CrossRef] [PubMed]
  136. Ito, Y.; Kurata, M.; Suzuki, K.; Hamajima, N.; Hishida, H.; Aoki, K. Cardiovascular disease mortality and serum carotenoid levels: A Japanese population-based follow-up study. J. Epidemiol. 2006, 16, 154–160. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  137. Tierney, A.C.; Rumble, C.E.; Billings, L.M.; George, E.S. Effect of Dietary and Supplemental Lycopene on Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis. Adv. Nutr. Int. Rev. J. 2020, 11, 1453–1488. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Thannickal, V.J.; Fanburg, B.L. Reactive oxygen species in cell signaling. Am. J. Physiol. Lung Cell Mol. Physiol. 2000, 279, L1005–L1028. [Google Scholar] [CrossRef] [PubMed]
  139. Sugamura, K.; Keaney, J.F., Jr. Reactive oxygen species in cardiovascular disease. Free Radic. Biol. Med. 2011, 51, 978–992. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  140. Cuadrado, A.; Manda, G.; Hassan, A.; Alcaraz, M.J.; Barbas, C.; Daiber, A.; Ghezzi, P.; León, R.; López, M.G.; Oliva, B.; et al. Transcription Factor NRF2 as a Therapeutic Target for Chronic Diseases: A Systems Medicine Approach. Pharmacol. Rev. 2018, 70, 348–383. [Google Scholar] [CrossRef] [PubMed]
  141. Meili-Butz, S.; Niermann, T.; Fasler-Kan, E.; Barbosa, V.; Butz, N.; John, D.; Brink, M.; Buser, P.T.; Zaugg, C.E. Dimethyl fumarate, a small molecule drug for psoriasis, inhibits Nuclear Factor-kappaB and reduces myocardial infarct size in rats. Eur. J. Pharmacol. 2008, 586, 251–258. [Google Scholar] [CrossRef] [PubMed]
  142. Kuang, Y.; Zhang, Y.; Xiao, Z.; Xu, L.; Wang, P.; Ma, Q. Protective effect of dimethyl fumarate on oxidative damage and signaling in cardiomyocytes. Mol. Med. Rep. 2020, 22, 2783–2790. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  143. Luo, M.; Sun, Q.; Zhao, H.; Tao, J.; Yan, D. The Effects of Dimethyl Fumarate on Atherosclerosis in the Apolipoprotein E-Deficient Mouse Model with Streptozotocin-Induced Hyperglycemia Mediated by the Nuclear Factor Erythroid 2-Related Factor 2/Antioxidant Response Element (Nrf2/ARE) Signaling Pathway. Med. Sci. Monit. 2019, 25, 7966–7975. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  144. Sharma, A.; Rizky, L.; Stefanovic, N.; Tate, M.; Ritchie, R.H.; Ward, K.W.; de Haan, J.B. The nuclear factor (erythroid-derived 2)-like 2 (Nrf2) activator dh404 protects against diabetes-induced endothelial dysfunction. Cardiovasc. Diabetol. 2017, 16, 33. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  145. Okafor, O.N.; Farrington, K.; Gorog, D.A. Allopurinol as a therapeutic option in cardiovascular disease. Pharmacol. Ther. 2017, 172, 139–150. [Google Scholar] [CrossRef] [PubMed]
  146. Agarwal, V.; Hans, N.; Messerli, F.H. Effect of allopurinol on blood pressure: A systematic review and meta-analysis. J. Clin. Hypertens. 2013, 15, 435–442. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  147. Landmesser, U.; Drexler, H. Allopurinol and endothelial function in heart failure: Future or fantasy? Circulation 2002, 106, 173–175. [Google Scholar] [CrossRef] [PubMed]
  148. Rashid, M.A.; William-Olsson, G. Influence of allopurinol on cardiac complications in open heart operations. Ann. Thorac. Surg. 1991, 52, 127–130. [Google Scholar] [CrossRef] [PubMed]
  149. Guan, W.; Osanai, T.; Kamada, T.; Hanada, H.; Ishizaka, H.; Onodera, H.; Iwasa, A.; Fujita, N.; Kudo, S.; Ohkubo, T.; et al. Effect of allopurinol pretreatment on free radical generation after primary coronary angioplasty for acute myocardial infarction. J. Cardiovasc. Pharmacol. 2003, 41, 699–705. [Google Scholar] [CrossRef] [PubMed]
  150. Zarrabi, A.; Eftekhari, H.; Casscells, S.W.; Madjid, M. The open-artery hypothesis revisited. Tex. Heart Inst. J. 2006, 33, 345–352. [Google Scholar] [PubMed] [PubMed Central]
  151. Baldus, S.; Müllerleile, K.; Chumley, P.; Steven, D.; Rudolph, V.; Lund, G.K.; Staude, H.J.; Stork, A.; Köster, R.; Kähler, J.; et al. Inhibition of xanthine oxidase improves myocardial contractility in patients with ischemic cardiomyopathy. Free Radic. Biol. Med. 2006, 41, 1282–1288. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  152. Altenhöfer, S.; Radermacher, K.A.; Kleikers, P.W.; Wingler, K.; Schmidt, H.H. Evolution of NADPH Oxidase Inhibitors: Selectivity and Mechanisms for Target Engagement. Antioxid. Redox Signal 2015, 23, 406–427. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  153. Yu, L.; Yang, G.; Zhang, X.; Wang, P.; Weng, X.; Yang, Y.; Li, Z.; Fang, M.; Xu, Y.; Sun, A.; et al. Megakaryocytic Leukemia 1 Bridges Epigenetic Activation of NADPH Oxidase in Macrophages to Cardiac Ischemia-Reperfusion Injury. Circulation 2018, 138, 2820–2836. [Google Scholar] [CrossRef] [PubMed]
  154. Roth Flach, R.J.; Su, C.; Bollinger, E.; Cortes, C.; Robertson, A.W.; Opsahl, A.C.; Coskran, T.M.; Maresca, K.P.; Keliher, E.J.; Yates, P.D.; et al. Myeloperoxidase inhibition in mice alters atherosclerotic lesion composition. PLoS ONE 2019, 14, e0214150. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  155. Dao, V.T.; Casas, A.I.; Maghzal, G.J.; Seredenina, T.; Kaludercic, N.; Robledinos-Anton, N.; Di Lisa, F.; Stocker, R.; Ghezzi, P.; Jaquet, V.; et al. Pharmacology and Clinical Drug Candidates in Redox Medicine. Antioxid. Redox Signal 2015, 23, 1113–1129. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  156. Tita, C.; Gilbert, E.M.; Van Bakel, A.B.; Grzybowski, J.; Haas, G.J.; Jarrah, M.; Dunlap, S.H.; Gottlieb, S.S.; Klapholz, M.; Patel, P.C.; et al. A Phase 2a dose-escalation study of the safety, tolerability, pharmacokinetics and haemodynamic effects of BMS-986231 in hospitalized patients with heart failure with reduced ejection fraction. Eur. J. Heart Fail. 2017, 19, 1321–1332. [Google Scholar] [CrossRef]
  157. Khalaf, D.; Krüger, M.; Wehland, M.; Infanger, M.; Grimm, D. The Effects of Oral l-Arginine and l-Citrulline Supplementation on Blood Pressure. Nutrients 2019, 11, 1679. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  158. Broderick, J.A.; Zamore, P.D. MicroRNA therapeutics. Gene Ther. 2011, 18, 1104–1110. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  159. Iorio, M.V.; Croce, C.M. MicroRNA dysregulation in cancer: Diagnostics, monitoring and therapeutics. A comprehensive review. EMBO Mol. Med. 2012, 4, 143–159, Erratum in EMBO Mol. Med. 2017, 9, 852. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  160. Baumann, V.; Winkler, J. miRNA-based therapies: Strategies and delivery platforms for oligonucleotide and non-oligonucleotide agents. Future Med. Chem. 2014, 6, 1967–1984. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  161. Guan, Y.; Song, X.; Sun, W.; Wang, Y.; Liu, B. Effect of Hypoxia-Induced MicroRNA-210 Expression on Cardiovascular Disease and the Underlying Mechanism. Oxid. Med. Cell Longev. 2019, 2019, 4727283. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  162. Wang, L.; Jia, Q.; Xinnong, C.; Xie, Y.; Yang, Y.; Zhang, A.; Liu, R.; Zhuo, Y.; Zhang, J. Role of cardiac progenitor cell-derived exosome-mediated microRNA-210 in cardiovascular disease. J. Cell Mol. Med. 2019, 23, 7124–7131. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  163. Hu, S.; Huang, M.; Li, Z.; Jia, F.; Ghosh, Z.; Lijkwan, M.A.; Fasanaro, P.; Sun, N.; Wang, X.; Martelli, F.; et al. MicroRNA-210 as a novel therapy for treatment of ischemic heart disease. Circulation 2010, 122 (Suppl. 11), S124–S131. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  164. Karakas, M.; Schulte, C.; Appelbaum, S.; Ojeda, F.; Lackner, K.J.; Münzel, T.; Schnabel, R.B.; Blankenberg, S.; Zeller, T. Circulating microRNAs strongly predict cardiovascular death in patients with coronary artery disease-results from the large AtheroGene study. Eur. Heart J. 2017, 38, 516–523. [Google Scholar] [CrossRef] [PubMed]
  165. Schulte, C.; Zeller, T. microRNA-based diagnostics and therapy in cardiovascular disease-Summing up the facts. Cardiovasc. Diagn. Ther. 2015, 5, 17–36. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  166. Cai, B.; Pan, Z.; Lu, Y. The roles of microRNAs in heart diseases: A novel important regulator. Curr. Med. Chem. 2010, 17, 407–411. [Google Scholar] [CrossRef] [PubMed]
  167. Silvestri, P.; Di Russo, C.; Rigattieri, S.; Fedele, S.; Todaro, D.; Ferraiuolo, G.; Altamura, G.; Loschiavo, P. MicroRNAs and ischemic heart disease: Towards a better comprehension of pathogenesis, new diagnostic tools and new therapeutic targets. Recent. Pat. Cardiovasc. Drug Discov. 2009, 4, 109–118. [Google Scholar] [CrossRef] [PubMed]
  168. Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Cardiac-specific miRNA in cardiogenesis, heart function, and cardiac pathology (with focus on myocardial infarction). J. Mol. Cell Cardiol. 2016, 94, 107–121. [Google Scholar] [CrossRef] [PubMed]
  169. Cheng, Y.; Tan, N.; Yang, J.; Liu, X.; Cao, X.; He, P.; Dong, X.; Qin, S.; Zhang, C. A translational study of circulating cell-free microRNA-1 in acute myocardial infarction. Clin. Sci. 2010, 119, 87–95. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  170. Li, Y.Q.; Zhang, M.F.; Wen, H.Y.; Hu, C.L.; Liu, R.; Wei, H.Y.; Ai, C.M.; Wang, G.; Liao, X.X.; Li, X. Comparing the diagnostic values of circulating microRNAs and cardiac troponin T in patients with acute myocardial infarction. Clinics 2013, 68, 75–80. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  171. D’Alessandra, Y.; Devanna, P.; Limana, F.; Straino, S.; Di Carlo, A.; Brambilla, P.G.; Rubino, M.; Carena, M.C.; Spazzafumo, L.; De Simone, M.; et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur. Heart J. 2010, 31, 2765–2773. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  172. Duan, L.; Xiong, X.; Liu, Y.; Wang, J. miRNA-1: Functional roles and dysregulation in heart disease. Mol. BioSyst. 2014, 10, 2775–2782. [Google Scholar] [CrossRef]
  173. Polyakova, E.A.; Zaraiskii, M.I.; Mikhaylov, E.N.; Baranova, E.I.; Galagudza, M.M.; Shlyakhto, E.V. Association of myocardial and serum miRNA expression patterns with the presence and extent of coronary artery disease: A cross-sectional study. Int. J. Cardiol. 2021, 322, 9–15. [Google Scholar] [CrossRef] [PubMed]
  174. Zampetaki, A.; Dudek, K.; Mayr, M. Oxidative stress in atherosclerosis: The role of microRNAs in arterial remodeling. Free Radic. Biol. Med. 2013, 64, 69–77. [Google Scholar] [CrossRef] [PubMed]
  175. Li, P.; Yin, Y.-L.; Guo, T.; Sun, X.-Y.; Ma, H.; Zhu, M.-L.; Zhao, F.-R.; Xu, P.; Chen, Y.; Wan, G.-R.; et al. Inhibition of Aberrant MicroRNA-133a Expression in Endothelial Cells by Statin Prevents Endothelial Dysfunction by Targeting GTP Cyclohydrolase 1 in Vivo. Circulation 2016, 134, 1752–1765. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  176. Lee, D.Y.; Chiu, J.J. Atherosclerosis and flow: Roles of epigenetic modulation in vascular endothelium. J. Biomed. Sci. 2019, 26, 56. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  177. Loyer, X.; Potteaux, S.; Vion, A.C.; Guérin, C.L.; Boulkroun, S.; Rautou, P.E.; Ramkhelawon, B.; Esposito, B.; Dalloz, M.; Paul, J.L.; et al. Inhibition of microRNA-92a prevents endothelial dysfunction and atherosclerosis in mice. Circ. Res. 2014, 114, 434–443. [Google Scholar] [CrossRef] [PubMed]
  178. Wang, Y.; Huang, Q.; Liu, J.; Wang, Y.; Zheng, G.; Lin, L.; Yu, H.; Tang, W.; Huang, Z. Vascular endothelial growth factor A polymorphisms are associated with increased risk of coronary heart disease: A meta-analysis. Oncotarget 2017, 8, 30539–30551. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  179. Wen, Z.; Huang, W.; Feng, Y.; Cai, W.; Wang, Y.; Wang, X.; Liang, J.; Wani, M.; Chen, J.; Zhu, P.; et al. MicroRNA-377 Regulates Mesenchymal Stem Cell-Induced Angiogenesis in Ischemic Hearts by Targeting VEGF. PLoS ONE 2014, 9, e104666. [Google Scholar] [CrossRef]
  180. Kim, K.S.; Song, C.G.; Kang, P.M. Targeting Oxidative Stress Using Nanoparticles as a Theranostic Strategy for Cardiovascular Diseases. Antioxid. Redox Signal 2019, 30, 733–746. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  181. Lee, D.; Bae, S.; Hong, D.; Lim, H.; Yoon, J.H.; Hwang, O.; Park, S.; Ke, Q.; Khang, G.; Kang, P.M. H2O2-responsive molecularly engineered polymer nanoparticles as ischemia/reperfusion-targeted nanotherapeutic agents. Sci. Rep. 2013, 3, 2233. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  182. Kang, C.; Gwon, S.; Song, C.; Kang, P.M.; Park, S.C.; Jeon, J.; Hwang, D.W.; Lee, D. Fibrin-Targeted and H2O2-Responsive Nanoparticles as a Theranostics for Thrombosed Vessels. ACS Nano 2017, 11, 6194–6203. [Google Scholar] [CrossRef] [PubMed]
  183. Somasuntharam, I.; Boopathy, A.V.; Khan, R.S.; Martinez, M.D.; Brown, M.E.; Murthy, N.; Davis, M.E. Delivery of Nox2-NADPH oxidase siRNA with polyketal nanoparticles for improving cardiac function following myocardial infarction. Biomaterials 2013, 34, 7790–7798. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  184. Seshadri, G.; Sy, J.C.; Brown, M.; Dikalov, S.; Yang, S.C.; Murthy, N.; Davis, M.E. The delivery of superoxide dismutase encapsulated in polyketal microparticles to rat myocardium and protection from myocardial ischemia-reperfusion injury. Biomaterials 2010, 31, 1372–1379. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  185. Gray, W.D.; Che, P.; Brown, M.; Ning, X.; Murthy, N.; Davis, M.E. N-acetylglucosamine conjugated to nanoparticles enhances myocyte uptake and improves delivery of a small molecule p38 inhibitor for post-infarct healing. J. Cardiovasc. Transl. Res. 2011, 4, 631–643. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  186. Soumya, R.S.; Vineetha, V.P.; Raj, P.S.; Raghu, K.G. Beneficial properties of selenium incorporated guar gum nanoparticles against ischemia/reperfusion in cardiomyoblasts (H9c2). Met. Integr. Biometal Sci. 2014, 6, 2134–2147. [Google Scholar] [CrossRef] [PubMed]
  187. Wang, Y.; Li, L.; Zhao, W.; Dou, Y.; An, H.; Tao, H.; Xu, X.; Jia, Y.; Lu, S.; Zhang, J.; et al. Targeted Therapy of Atherosclerosis by a Broad-Spectrum Reactive Oxygen Species Scavenging Nanoparticle with Intrinsic Anti-inflammatory Activity. ACS Nano 2018, 12, 8943–8960. [Google Scholar] [CrossRef] [PubMed]
  188. Abrescia, P.; Golino, P. Free radicals and antioxidants in cardiovascular diseases. Expert. Rev. Cardiovasc. Ther. 2005, 3, 159–171. [Google Scholar] [CrossRef] [PubMed]
  189. Islam, M.M.T.; Shekhar, H.U. Antioxidant therapy in cardiovascular diseases: Still a matter of debate. Adv. Cytol. Pathol. 2017, 2, 87–88. [Google Scholar] [CrossRef]
Figure 1. Selected sources of reactive oxygen species [11]. Abbreviations: ROS—reactive oxygen species; NADPH—nicotinamide adenine dinucleotide phosphate; NO—nitric oxide; ER—endoplasmic reticulum.
Figure 1. Selected sources of reactive oxygen species [11]. Abbreviations: ROS—reactive oxygen species; NADPH—nicotinamide adenine dinucleotide phosphate; NO—nitric oxide; ER—endoplasmic reticulum.
Nutrients 16 02587 g001
Figure 2. Existing forms of CoQ10.
Figure 2. Existing forms of CoQ10.
Nutrients 16 02587 g002
Figure 3. Chemical structures of the different classes of polyphenols. Extracted and modified from [36].
Figure 3. Chemical structures of the different classes of polyphenols. Extracted and modified from [36].
Nutrients 16 02587 g003
Figure 4. Flavonoids’ properties.
Figure 4. Flavonoids’ properties.
Nutrients 16 02587 g004
Table 2. The role of different carotenoids in preventing CVD [97,111,113,120,121,122,123,125].
Table 2. The role of different carotenoids in preventing CVD [97,111,113,120,121,122,123,125].
CarotenoidsFunctionRole in Preventing CVD
β-CaroteneInhibits LDL oxidationPrevents atherosclerosis
Decreases TNF-α-induced inflammation in endothelial cellsReduces risk of CVD by opposing inflammatory oxidative stress
LycopeneDecreases TNF-α-induced inflammation in endothelial cellsReduces risk of CVD by opposing inflammatory oxidative stress
Inhibits IL-1 secretionExerts an anti-atherogenic effect
Increases NO levelsDilates blood vessels, slowing the progression of atherosclerosis
Regulates PCSK9 and HMGR genes, and increases LDL-R activityLowers hypercholesterolemia
Improves the LDL/HDL ratioReduces the risk of atherosclerosis and postpones its progression
Reduces accumulation of cholesterol in the aorta
Inhibits vascular smooth-muscle cell proliferation and foam cell formation
Table 3. Role of miRNAs and the potential target of novel treatments for cardiovascular diseases. Abbreviations: ROS, reactive oxygen species; NOS, nitric oxide synthase; VEGF, vascular endothelial growth factor; CVD, cardiovascular disease.
Table 3. Role of miRNAs and the potential target of novel treatments for cardiovascular diseases. Abbreviations: ROS, reactive oxygen species; NOS, nitric oxide synthase; VEGF, vascular endothelial growth factor; CVD, cardiovascular disease.
Type of miRNARole in Onset of CVD
miRNA-210During hypoxia, miRNA-210 promotes angiogenesis and inhibits cardiomyocyte apoptosis.
miRNA-1Involved in the differentiation and proliferation of muscle cells.
In anemic myocardium, it regulates cardiomyocyte growth and proapoptotic factors.
Overexpression increases ROS production.
miRNA-133Inhibition results in NOS production and may prevent endothelial dysfunction.
miRNA-92aRegulates NOS expression, reduces plaque inflammation, and increases its stability by promoting cell proliferation and angiogenesis.
miRNA-206Regulates VEGF expression, inhibits viability, and increases apoptosis of endothelial progenitor cells.
miRNA-377Inhibition of miRNA-377 reduces myocardial fibrosis and improves its function.
Table 4. Types of nanoparticles and their functions related to prevention and treatment of cardiovascular diseases based on animal models.
Table 4. Types of nanoparticles and their functions related to prevention and treatment of cardiovascular diseases based on animal models.
Type of NanoparticlesFunction of Nanoparticles
H2O2-responsive nanoparticlesAnti-inflammatory and anti-apoptotic effects and reductions in further organ damage.
Nanoparticles with antioxidant propertiesEnables imaging the thrombus and inhibits its formation by scavenging H2O2 and reduces oxidative stress.
Nanoparticles carrying SODReduces myocyte apoptosis and improves myocardial function.
Nanoparticles carrying N-acetylcysteineAttenuates myocardial fibrosis.
Selenium-based nanoparticlesReduces ROS production in ischemic cardiomyocytes.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Młynarska, E.; Hajdys, J.; Czarnik, W.; Fularski, P.; Leszto, K.; Majchrowicz, G.; Lisińska, W.; Rysz, J.; Franczyk, B. The Role of Antioxidants in the Therapy of Cardiovascular Diseases—A Literature Review. Nutrients 2024, 16, 2587. https://doi.org/10.3390/nu16162587

AMA Style

Młynarska E, Hajdys J, Czarnik W, Fularski P, Leszto K, Majchrowicz G, Lisińska W, Rysz J, Franczyk B. The Role of Antioxidants in the Therapy of Cardiovascular Diseases—A Literature Review. Nutrients. 2024; 16(16):2587. https://doi.org/10.3390/nu16162587

Chicago/Turabian Style

Młynarska, Ewelina, Joanna Hajdys, Witold Czarnik, Piotr Fularski, Klaudia Leszto, Gabriela Majchrowicz, Wiktoria Lisińska, Jacek Rysz, and Beata Franczyk. 2024. "The Role of Antioxidants in the Therapy of Cardiovascular Diseases—A Literature Review" Nutrients 16, no. 16: 2587. https://doi.org/10.3390/nu16162587

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