Next Article in Journal
Leukocyte Imbalances in Mucopolysaccharidoses Patients
Next Article in Special Issue
Macrophages of the Cardiorenal Axis and Myocardial Infarction
Previous Article in Journal
Phase Angle in Head and Neck Cancer: A Sex-Differential Analysis from Biological and Clinical Behavior to Health-Related Quality of Life
Previous Article in Special Issue
Evidence and Uncertainties on Lipoprotein(a) as a Marker of Cardiovascular Health Risk in Children and Adolescents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiovascular Remodeling Post-Ischemia: Herbs, Diet, and Drug Interventions

by
Ayodeji A. Olabiyi
* and
Lisandra E. de Castro Brás
Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27858, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(6), 1697; https://doi.org/10.3390/biomedicines11061697
Submission received: 18 May 2023 / Revised: 8 June 2023 / Accepted: 9 June 2023 / Published: 13 June 2023

Abstract

:
Cardiovascular disease (CVD) is a serious health burden with increasing prevalence, and CVD continues to be the principal global source of illness and mortality. For several disorders, including CVD, the use of dietary and medicinal herbs instead of pharmaceutical drugs continues to be an alternate therapy strategy. Despite the prevalent use of synthetic pharmaceutical medications, there is currently an unprecedented push for the use of diet and herbal preparations in contemporary medical systems. This urge is fueled by a number of factors, the two most important being the common perception that they are safe and more cost-effective than modern pharmaceutical medicines. However, there is a lack of research focused on novel treatment targets that combine all these strategies—pharmaceuticals, diet, and herbs. In this review, we looked at the reported effects of pharmaceutical drugs and diet, as well as medicinal herbs, and propose a combination of these approaches to target independent pathways that could synergistically be efficacious in treating cardiovascular disease.

1. Introduction

According to the World Health Organization, each year 17.9 million people worldwide die from cardiovascular disease (CVD), accounting for 31% of all mortality [1]. The American Heart Association reports that roughly half of all Americans have a type of CVD [2]. Of these, heart attacks (i.e., myocardial infarction, MI) and strokes account for 80% of CVD fatalities [2]. An MI occurs when an artery to the heart is clogged, and the heart does not receive enough blood or oxygen for a significant amount of time leading to ischemia/infarction of the tissue downstream of the obstruction. Upon infarction, if reperfusion does not occur in a timely manner, cardiac cells will become ischemic and metabolically compromised.
Cardiac cells (including cardiomyocytes), capillaries, and extracellular matrix (ECM), which is made up of various collagen fiber types and ensures structural integrity, are the three crucial components of heart muscle that enable effective contraction and pumping of the blood to the whole body. When cells die due to ischemia, a cascade of events is triggered to clear and replace the damaged cells and tissues. These events are dynamic and are broadly divided into three stages: inflammation/necrosis to remove dead cells/tissue; proliferation/fibrosis to replace cells, ECM, and set up tissue vascularization; and long-term remodeling/maturation to regenerate the tissue and/or mature the scar and provide tissue integrity and function [3]. Different risk cues, either exogenous or endogenous, may start the inflammatory response. Microorganism invasion is considered exogenous, whereas tissue injury is considered endogenous. Pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) are the names given to the external and endogenous signaling molecules that are released, respectively, by pathogens or necrotic cells [4]. Toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), C-type lectins, and receptors for advanced glycation end-products (RAGE) are just a few of the pattern recognition receptors (PRRs) that may detect PAMPs and DAMPs. A described genus of specific pro-resolving mediators (SPMs) including lipoxins, resolvins, protectins, and maresins is activated during the resolution phase of inflammation [5]. These mediators have strong, direct anti-inflammatory and pro-resolution properties that limit the recruitment of inflammatory leukocytes while encouraging macrophage clearance of apoptotic neutrophils. By controlling ventricular size, shape, and function, left ventricular (LV) remodeling defines the heart’s (mal)adaptation to injury and/or mechanical, neurohormonal, and hereditary alterations. While the increased microcirculatory blood supply provided during the proliferation phase will support any live cardiomyocytes, continual LV remodeling (due to uncontrolled inflammation) leads to cardiomyocyte loss, expansion of the ischemic area, persistent collagenase activity, and degradation of high-tensile collagens; this in turn leads to LV dilation, with wall thinning and fibrosis [6]. This “adverse” or “pathological” remodeling following MI is maladaptive, carries a disproportionate risk of heart failure (HF), and dramatically lowers survival [7]. Over the last twenty years, the prevalence of HF has increased [8]. Despite immediate revascularization and subsequent treatment options that significantly lower the mortality of acute MI, ischemic heart disease remains the main cause of HF [8,9]. Comorbidities foretell the severity and progression of HF [10,11]; in fact, more than 50% of HF patients have more than seven comorbidities, with age and sex being the two main predictors of HF [12]. Men have a larger heart mass and a higher incidence of subclinical coronary artery disease than women [13,14]. Women show less severe incidence than men in terms of atherosclerosis, left ventricular hypertrophy, and cardiomyocyte apoptosis [15,16]. Similarly, mortality post-MI correlates with advancing age, regardless of infarct size [17], which may be related to a higher prevalence of ventricular hypertrophy but also to a decreased immunological response, scarring, and autophagy [18].
Medications that have been effective in treating HF include neurohormonal antagonists (beta blockers, angiotensin receptor blockers, and aldosterone antagonists), which can prevent or reverse remodeling and have been shown to lower mortality and morbidity [19]. The use of medications has been practiced over the years for the prevention of HF. However, some of these medications produce adverse effects and due to high cost are not easily available to every patient. Over the past three decades, there has been a significant rise in the use of herbal supplements to prevent, avoid, and/or treat different conditions, including CVD; this rise results from a perceived safety (natural, non-toxic sources), efficiency with few or no adverse effects (unlike synthetic/chemical medications), and low cost [20]. The US Dietary Supplement Health and Education Act (DSHEA) of 1994 categorizes herbal products as dietary supplements. In addition to herbal supplements, epidemiological research showed that consuming fruits, vegetables, olive oil, wine, legumes, and whole grains, as found in the Mediterranean diet, has a cardioprotective effect [21]. Foods high in polyphenols, such as berries and olive oil, have been demonstrated to have cardioprotective benefits that enhance endothelial function and plasma lipid profiles while preventing aberrant platelet aggregation and decreasing inflammation [22]. Thus, diet and dietary supplements have important roles in maintaining homeostasis and reducing CVD and warrant further investigation. Preload, afterload, and molecular mechanisms of remodeling have been targeted using synthetic drug therapy to date; can this also be achieved through diet and natural supplements? This review focuses on the use of prescription drugs, herbal products, and diet modification on cardiovascular remodeling.

2. Cardiovascular Health and Drugs Treatment

Over the past two centuries, the pathways of discovery, innovation, and therapeutic improvement in cardiovascular science and medicine have been really astounding. Nabel and Braunwald [23] discussed the remarkable decrease in cardiovascular disease deaths due to scientific advancements from 1950, when it was over 450 deaths per 100,000 of the population, to approximately 100 deaths per 100,000 by 2010. Even though many clinical advancements have contributed to this impressive decline, they emphasized the significance of β-blocking therapy and 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG Co-A) reductase inhibition, as well as the introduction of novel antihypertensive agents in the context of the national high blood pressure education program. Nevertheless, notwithstanding these medical advances, concurrent disorders such as diabetes mellitus, obesity, and congestive HF contribute to the consistently high prevalence of CVD. Additionally, because of the remarkable decline in MI mortality rates, affected individuals live longer but nearly 40% develop HF, which has a worse prognosis than acute MI. Despite prior advancements in pharmacological therapy, such as oral diuretics and medication treatment for hypertension, progress is still being made in the management of cardiovascular disorders such as HF and cardiac arrhythmias.
HF patients with systolic LV dysfunction treated with angiotensin-converting enzyme (ACE) inhibitors present reduced mortality and morbidity [24]. ACE inhibitors act as vasodilators and their effectiveness in reducing remodeling is supported by both experimental and clinical studies, particularly in the post-infarction setting [20,21,22,25,26,27]. However, with ACE inhibitors, reverse remodeling has infrequently been observed in sizable patient cohorts. One of the aspects of this therapy that may limit its ability to provide long-term benefit is protection without reversion of remodeling. In clinical studies with ACE inhibitors, the survival curves of patients assigned to either the active treatment or the placebo groups typically show an early divergence of the curves followed by a parallel trend through time [28,29]. This shows that these medications more often delay the onset of the disease than stop it [30].
Angiotensin receptor blockers (ARBs) are also used in HF treatment, instead of or in combination with ACE inhibitors. ARBs selectively reduce the activity of type 2 receptors (AT2R), which mediate vasodilation and may prevent myocardial fibrosis, without affecting type 1 receptors (AT1R), which primarily mediate cardiac hypertrophy, aldosterone production, fibrosis, and vasoconstriction [31].
Matrix metalloproteinase (MMP) activation and maladaptive LV remodeling have a clear cause-and-effect relationship, as shown by experimental research [32,33,34]. Thus, MMPs have long been recognized as an attractive target to decrease or prevent maladaptive cardiac remodeling. MMP inhibitors have been suggested as a treatment for several illnesses in order to lessen matrix deterioration, blunt inflammation, and enhance tissue regeneration. However, difficulties in producing MMP specific inhibitors and the many functions of MMPS (both beneficial and detrimental) have led to little progress in clinical trials and still need further investigation [35,36,37].

3. Cardiovascular Health and Diet

According to data from the Centers for Disease Control (CDC) and Prevention, some of the most common health problems in the United States are chronic diseases such as cancer, diabetes, heart disease, and stroke. Many of these chronic illnesses can be prevented, though, as they are linked to unhealthy eating and lifestyle choices. Dietary intervention allows for a better blending of various diets and nutrients. Healthy eating practices therefore encourage a greater magnitude of favorable impacts than the potential effects of a single nutrient supplementation because of the synergistic health effects among them. Research has shown that a high intake of fiber, antioxidants, vitamins, minerals, polyphenols, monounsaturated fatty acids (MUFA), and polyunsaturated fatty acids (PUFA), low intakes of salt, saturated fats, and trans fats, and a high intake of carbohydrates with low glycemic loads are all indicators of healthy eating habits [38].
Although some cardiovascular diseases are congenital, years of investigation into the relationship between diet and heart disease concentrated on specific vitamins and minerals, types of fats, and individual nutrients such as cholesterol (and foods high in dietary cholesterol, such eggs). This has been eye-opening, but it has also led to some dead ends and perpetuated misconceptions about what a heart-healthy diet is. People eat food, not nutrition, which explains this [38]. A growing heart need nutritious grains, vegetables, and fruits, as well as a mineral balance (sodium and potassium) and body weight maintenance, as well as sufficient sleep. People who follow this eating pattern have a 31% lower risk of heart disease, a 33% lower risk of diabetes, and a 20% lower risk of stroke [39]. Neonatal malnutrition, inflammation, and growth retardation are linked to a lifelong decrease in cardiomyocyte number, systolic and diastolic dysfunction, ischemia sensitivity, and hypertension [40,41,42,43]. A significant body of scientific research supports nutrition as one of the most effective ways to avoid CVD associated mortality [44] and may even be able to reverse heart disease [45]. Even more, the management of other risk factors, such as excess weight, hypertension, diabetes, or dyslipidemia, appears to depend heavily on nutrition and exercise [45]. In this regard, foods or dietary habits that can improve CVD prevention have been identified and categorized.

3.1. Mediterranean Diet

The Mediterranean diet (MeDiet), known as a mainly plant-based diet, characterized by a high consumption of fruits, vegetables, and whole grains as well as fish, nuts, and legumes, unsaturated fatty acids, and low to moderate intake of alcohol (consumed with a meal) has been reported to have a great impact on the arterial wall, which could account for the association between MeDiet and low CVD prevalence [46]. Intriguingly, MeDiet appears to modulate the expression of pro-atherogenic genes like cyclooxygenase-2 (COX-2), monocyte chemoattractant protein-1 (MCP-1), and low-density lipoprotein receptor-related protein (LRP1), while decreasing plasmatic levels of molecules negatively linked to plaque stability and rupture, such as MMP-9 and interleukins (IL-10, IL-13, or IL-18) [47,48]. Furthermore, in order to reduce the risk of CVD, the American Heart Association Nutrition Committee and the European Society of Cardiology both strongly recommend daily consumption of several portions of both fruits and vegetables [49,50], hallmarks of the MeDiet. These recommendations are mostly supported by epidemiological studies and meta-analyses [49,50,51,52,53]. According to a meta-analysis [52] of 83 studies, an increase in the consumption of fruits and vegetables was significantly inversely related to C-reactive protein (CRP) and tumor necrosis factor (TNF) levels and directly related to an increase in the proliferation of T-cell populations (71 clinical trials and 12 observational studies). The main component of the MeDiet, extra virgin olive oil (EVOO), has bioactive components shown to improve endothelial dysfunction, oxidative stress, and overall inflammatory state [54]. A meta-analysis by Schwingshackl et al. [55] found that, compared to controls, daily ingestion of 1 mg and 50 mg of olive oil significantly reduced CRP in 30 randomized control studies with a total of 3106 participants. Additionally, those who consumed the most olive oil had a substantially higher flow-mediated dilatation score (0.6%, p < 0.002). Similar to EVOO, nuts contain monosaturated fats; numerous sizable studies have shown that nuts significantly lower CVD morbidity and death [56,57,58,59]. Hence, a MeDiet rich in nuts had been linked to better weight reduction, lower low-density lipoprotein-cholesterol (LDL-c) levels [60,61,62], lower risk of developing hypertension [61,63], and lower levels of inflammatory and oxidative mediators [64,65].

3.2. Paleo Diet

Another eating plan based on foods popular during the Old Stone Age is the Paleolithic diet (Paleo), which primarily consists of lean meat, fish, eggs, fruits, vegetables, roots, and nuts while avoiding grains, dairy products, processed foods, added sugars, and salts [66]. This eating plan has less salt but more protein, several micronutrients such as vitamins C and E, carotenes, and fiber [67]. It also has fewer calories from refined carbohydrates and fat [67,68,69]. According to a meta-analysis of 8 studies, a Paleo diet significantly improved circulating concentrations of total cholesterol, triglycerides, LDL-c, and CRP while also significantly lowering body weight, waist circumference, body mass index, body fat percentage, and systolic and diastolic blood pressure. It was also reported to significantly increase HDL cholesterol [70]. Another meta-analysis of four randomized control trials (RCTs) suggested short-term improvements in metabolic syndrome components following adoption of a Paleo diet, as reported by Manheimer et al. [71].

3.3. Keto Diet

Diets low in carbohydrates, but usually high in fats and/or proteins, have also become increasingly popular, referred to as ketogenic diets (keto diet). A keto diet restricts carbohydrate intake to less than 25 to 50 g per day, in an attempt to induce tissues to use fat or ketones as fuel during caloric restriction. As a result, the liver begins to produce ketone bodies, which are then utilized as an alternative energy source, particularly by the central nervous system [62]. From several reports, the keto diet may be linked to some improvements in various cardiovascular risk factors, such as obesity, type 2 diabetes, and HDL cholesterol levels [72,73,74,75,76,77]. On the other hand, a recent report describes keto diet causing cardiac fibrosis and inhibiting mitochondrial biogenesis [78]. According to their study, keto diet or repeated deep fasting impaired mitochondrial biogenesis, decreased cell respiration, and enhanced cardiac fibrosis and death in cardiomyocytes. As revealed by the research, increased levels of the histone deacetylase 2 (HDAC2) and inhibitor ketone body -hydroxybutyrate (-OHB), which promotes histone acetylation of the Sirt7 promoter and activates Sirt7 transcription, caused an increase in cardiac fibrosis and cardiomyocyte apoptosis [75]. Sirt7 transcription also prevents the transcription of mitochondrial ribosome-encoding genes. According to Xu and colleagues, consuming a keto diet over an extended period or accumulating -OHB may raise the chance of developing cardiovascular disease. In another population-based study, a low-carbohydrate diet, similar to keto, resulted in deleterious effects on the coronary artery [79]. In contrast, a research study in 2020 showed a keto diet was beneficial in diabetic patients by promoting cardiomyocyte survival and reducing interstitial fibrosis [79]. Although it is not clear what the reason(s) are for the discrepancies observed within these studies, it could result from differences between the animal models.

3.4. DASH

In 1990, dietary approaches to stop hypertension (DASH) were initiated; these approaches have received attention and several grants from the National Institute of Health (NIH) to see if specific dietary interventions were useful in treating hypertension. Some of the individuals enrolled showed a 6 to 11 mm Hg reduction in systolic blood pressure from the food intervention alone [80]. Both hypertensive and normotensive individuals experienced this effect, and as a result, DASH has occasionally been recommended as the first-line pharmacologic therapy in conjunction with lifestyle modifications. DASH encourages the eating of fresh produce, lean meat, low-fat dairy products, and foods rich in micronutrients. Additionally, it encourages limiting daily sodium intake to 1500 mg. DASH places a strong emphasis on eating fresh, minimally processed foods. The DASH diet is very similar to other dietary styles that are supported for cardiovascular health. The DASH diet is a synthesis of the old and new [81,82,83].

4. Cardiovascular Health and Herbal Medicine

Traditional herbal remedies have been used for decades and are typically regarded as safer than synthetic pharmaceuticals [84,85,86]. Approaches influenced by traditional medicine are still crucial, particularly for the management of chronic illnesses and to speed up the development of natural product drugs [87,88]. When administered with contemporary synthetic treatments, combinations of herbal medications or phytochemical active ingredients have been demonstrated to be useful in treating several disorders [89]. While the market for dietary botanical supplements is expanding, it is strongly recommended that more thorough clinical and scientific research be conducted on herbal and traditional medicines to increase their acceptance and visibility. Moreover, it is necessary to increase control on the quality of the materials used for formulation of the herbal/dietary supplements to ensure efficacy and safety.
In plants, certain polyphenols are present with adequate bioavailability singly or in combination with others (acting additively or synergistically). According to laboratory data gathered over the years, medicinal herbs can affect several CVD risks factors and, therefore, may have therapeutic utility in treating CVDs (Table 1). To effectively use herbs in CVD therapy, there have been numerous initiatives to shift studies on medicinal herbs from the bench to the bedside [90,91]. Natural products’ biological activity and structural variety outperform any existing synthetic drug screening library, which is another factor leading to a return of interest in them [90].
The herbs ginseng, ginkgo biloba, ganoderma lucidum, and gynostemma pentaphyllum have been reported to prevent CVDs [92,93]. These herbs are becoming more and more well-known due to their presence in commercial commodities in numerous markets around the world and their established therapeutic potential in a variety of settings, including cardiovascular conditions [92]. These organic compounds profoundly alter important cellular, molecular, and metabolic processes that regulate the etiology and pathogenesis of CVDs [94]. Recent research shows that these herbal remedies have strong therapeutic effects and can improve pathological diseases linked to CVDs [92,93]. Clear clinical therapeutic advantages have not, however, been established. Rafacho et al. [95] demonstrated rosemary supplementation (Rosmarinus oficinallis L.) to cause reduced myocardial damage and blood pressure in hypertensive rats fed a high-fructose diet and protected the heart against cardiac dysfunction and fibrosis after MI in rats. From their findings, it was concluded that the mechanism could involve improved energy metabolism and reduced oxidative stress. In general, herbs could prevent or treat more than one disease since they have several therapeutic properties. As an illustration, the herb Crocus sativus L. was discovered to contain therapeutic properties that may be used to treat five different types of heart illness, including hypertension, heart attacks, blood fat reduction, antioxidants, and cardiac tonics [96,97,98]. This herb’s role in preventing cardiovascular disorders was reportedly attributed to its anti-inflammatory and antioxidant properties [84,99].
In another study, due to strong antioxidant and free radical-scavenging activity, Citrus medica L. has also been reported to have cardioprotective potential [100]. In addition, a Crataegus species was observed to be a safe, nontoxic therapy option for ischemic heart disease and cardiovascular disease [101]. For example, the fruit or, alternatively, the leaves and flowers of Crataegus monogyna, which are high in polyphenols, are used medicinally. Direct scavenging of reactive oxygen species (ROS), increased catalase and superoxide dismutase activities, antioxidant activity, and downregulation of the caspase 3 gene are some of the known mechanisms of action of the Crataegus species [101]. Hawthorns are frequently utilized in a holistic approach to treat circulatory problems and congestive heart failure [102]. Moreover, stage 1 hypertension individuals who consume Elettaria cardamom have significantly lower blood pressure, increased fibrinolysis, and improved antioxidant status [103]. A recent study investigated the ability of cardamom oil to reestablish lipid homeostasis in the presence of hypercholesterolemia [104]. This study reduced atherogenicity index with dietary intervention with cardamom powder and oil, demonstrating the cardioprotective benefit of cardamom [104]. Researchers have also discovered two major methods through which the bark of Terminalia arjuna demonstrates cardioprotective advantages against induced cardiotoxicity which includes increased coronary artery flow and protection of the myocardium from ischemic damage [105].
Table 1. List of herbal remedies traditionally used for the treatment of different forms of CVDs.
Table 1. List of herbal remedies traditionally used for the treatment of different forms of CVDs.
HerbsForms of CVDsReference
GinsengOxidative stress, hypertension, cardiac disease, hyperlipidemia and ion regulation[93,106]
Ginkgo bilobaCardiac activity, vasorelaxant and vasoconstriction activity, hypertension[93]
Ganoderma lucidumAtherosclerosis, hyperlipidemia[107]
Gnostemma pentaphyllumHyperlipidemia[92]
Rosemary oficinalis L.Cardiac dysfunction and fibrosis[95]
Cocus sativus L.Systolic hypertension, oxidative stress, inflammation[86,92]
Citrus medica L.Ischemia heart disease[100]
Crataegus monogynaCongestive heart failure[101]
Elettaria cardamomHypercholesterolemia[104]
Terminalia arjunaCardiotoxicity[105]
Punica granatum L.Blood pressure, inflammation[108,109]
Apple (Malus pumila)Blood lipid levels[110,111]
Watermelon (Citrullus lanatus)Heart attacks, ischemic strokes, atherosclerosis[112,113,114]
BerriesMyocardial infarction, oxidative stress, inflammation, platelet aggregation[115,116,117]
Grapes (Vitis vinifera L.)Cardiac fibrosis, hyperlipidemia[118,119]
Garlic (Allium satinum L.)Hypertension, hypercholesterolemia[120]
Cinnamon (Cinnamomum verum)Oxidative stress, inflammation, artherosclerosis[121]

5. Polyphenols and Cardiovascular Health

Plants and plant products have been documented to have over 8000 known polyphenols [122]. Human health and wellness have been shown to directly be affected by a polyphenol-rich diet. According to Quideau et al. [123], polyphenols are substances with several phenolic units and no nitrogen-based functionalities that are generated from the polyketide and/or shikimate/phenyl propanoid pathways. After being metabolized, most polyphenols are glycosylated and either can bind to other phenols or conjugate with glucuronic acid, galacturonic acid, or glutathione [124]. Polyphenols are categorized into four main types: phenolic acids, stilbenes, lignans, and flavonoids (Figure 1).

5.1. Phenolic Acids

Phenolic acids include caffeic acid, which can be found in almost all fruits; chlorogenic acid, found in strawberries, pineapple, and other foods; and p-coumaric acid, present in cereal grains. The two families of phenolic acids—derivatives of benzoic acid and derivatives of cinnamic acid—are widely present in foods. Except for some red fruits, black radishes, and onions, which can have concentrations of many tens of milligrams per kilogram fresh weight of hydroxybenzoic acid, the hydroxybenzoic acid content of food plants are often modest [125].

5.2. Stilbenes

Stilbenes are phenolic substances with two phenyl groups joined by a bridge made of two carbon atoms in the methylene ring. Most stilbenes in plants function as phytoalexins, which are substances that are often only produced as a result of infection or injury. Resveratrol is one of the most thoroughly investigated stilbenes. Red wines, red grape juice, and peanuts all contain stilbenes [126,127].

5.3. Lignans

A wide class of low molecular weight polyphenols called lignans is present in many types of plants, especially seeds, whole grains, and vegetables. The word “wood” in Latin is the source of the name. Phytoestrogens’ predecessors are lignans [128]. They might act as antifeedants to protect seeds and plants from herbivores [129]. Secoisolariciresinol from flaxseed [130] and sesamin from sesame seed [131] are two major examples of lignans.

5.4. Flavonoids

Fruits, vegetables, legumes, red wine, and green tea all contain flavonoids. They are further divided into chalcones, anthocyanins, proanthocyanidins, flavanols, flavonols, flavones, isoflavones, and flavanones [97]. Green and black tea include flavanols, such as epigallocatechin gallate [132]. Onions, broccoli, and blueberries all contain flavonols, such as kaempferol and quercetin [133]. In strongly colored fruit, anthocyanins can be discovered (cyanin glucoside is one of the examples) as reported by Ly et al. [124]. Parsley and celery both contain flavones, such as apigenin, chrysin, and luteolin [124]. Soya and its processed derivatives contain isoflavones, such as daidzein and genistein [134]. Grapefruit contains flavanones, such as naringenin [124]. Chalcones are primarily plentiful in hops used in beer making. They are also plentiful in numerous plants and spices, as well as in several vegetables and fruits, such as shallots, tomatoes, potatoes, and bean sprouts (as well as licorice and cardamom). The primary prenylated chalcone is xanthohumol, which is mostly present in hops and hence in beer [135]. Condensed flavan-3-ols known as proanthocyanidins are present in numerous plants, including apples, grape seeds, grape skin, and cocoa beans (the main source) [136]. Another group of polyphenols is the curcuminoids which includes turmeric or the compound curcumin.
Generally, the most prevalent polyphenols among these several types are phenolic acids and flavonoids [137]. Various protective properties of natural polyphenols against cardiovascular illnesses have been reported [59,88,138,139]. It has been noted that polyphenols belonging to various subclasses can reduce heart fibrosis and dysfunction after a cardiac injury. Quercetin with its glycoside derivative, rutin, or solitary quercetin reduced cardiac dysfunction and myocardial damage brought on by isoproterenol, prevented cardiac fibrosis, and inhibited the synthesis of connective tissue growth factor (CTGF), transforming growth factor-β (TGF-β1), and ECM components [140]. Panchal et al. [141] reported that by blocking the NF-κB signaling pathway and promoting nuclear factor erythroid 2-related factor 2 (Nrf-2) and its downstream components, heart remodeling was prevented in an obese rat model fed a Western diet supplemented with quercetin.
The most common and powerful catechin in green tea is epigallocatechin-3-gallate (EGCG) [142]. In rats, EGCG has been reported to reduce heart hypertrophy and the proliferation of cardiac fibroblasts (CFs) [143]. Other findings revealed EGCG inhibited oxidative stress, which slowed heart hypertrophy both in vivo and in vitro [144]. In the meantime, it reduced the activation of rat CFs induced by AngII through the involvement of β -arrestin1 [145]. By blocking the NF-κB signaling pathway during hypertrophic stimulation, Cai et al. [146] discovered that EGCG might inhibit the expression of fibronectin and collagen formation in rat CFs brought on by AngII. It also significantly improved excessive CTGF expression and cardiac fibrosis [146]. An appropriate dose is necessary for EGCG to play its protective effect in reducing heart fibrosis, though. In contrast, a high dose of EGCG causes the creation of cardiac collagen and worsens cardiac fibrosis in mice [147].
NADPH oxidases (NOXs), a class of enzymes linked to the production of ROS, were found to contribute to the advancement of myocardial fibrosis and HF [148]. The myocardium expresses all forms of NOX, but NOX2 and NOX4 are predominant. In disease, the human heart’s end-stage failure exhibits enhanced NOX expression, and cardiac hypertrophy is evidently a significant source of elevated cardiac ROS. According to Wang et al. [149], luteolin inhibits CFs proliferation by reducing oxidative stress in vitro. The mechanism underlying this effect is dependent on the regulation of NOX2 and NOX4 in cardiac hypertrophy, which reduces the production of c-Jun N-terminal kinase (JNK) and TGF-1 β and lowers cardiac fibrosis [150]. Additionally, the anthocyanins found in grape skins, such as malvidin-3-glucoside, delphinidin-3-glucoside (Dp3G), cyanidin-3-glucoside (Cy3G), petunidin-3-glucoside (Pg3G), and peonidin-3-glucoside, guard against problems from ischemia/reperfusion and diabetic mellitus [151]. Dp3G and Cy3G, but not Pg3G, were able to restore complex I of the mitochondrial respiratory chain to its original condition and increase ischemia-depleted ATP levels via promoting oxidative phosphorylation [152]. Because of its high potential to decrease cytosolic cytochrome c, Cy3G, but not Pg3G, protects the rat heart against ischemia/reperfusion-induced apoptosis and necrosis [153]. Administration of Cy3G improves cardiac dysfunction and cardiac inflammation in STZ-induced diabetic cardiomyopathy by activating MMP-9 and lowering the level of tissue inhibitor of MMP-9 (TIMP-1) found in diabetic rat hearts [154].
Curcumin, a natural inhibitor of p300-specific histone acetyltransferase, has been demonstrated to prevent HF [155]. Curcumin alone or in combination with enalapril reduces severe perivascular fibrosis in rats after MI and enhances left ventricular systolic function via reducing nuclear expression of p300 [155,156]. According to a different study, curcumin prevents unfavorable cardiac repair and lessens the fibrotic response to ischemia and reperfusion by slowing the breakdown of the extracellular matrix and preventing the synthesis of collagens through the TGF-β/Smad pathway [157]. Curcumin dramatically reduced collagen buildup in vivo, as well as CF proliferation, migration, and MMP production, according to another line of study [158]. According to Zeng et al. [159], curcumin’s ability to up-regulate Nrf-2 expression and suppress NF-κB activation was directly related to its protective role. Resveratrol administration has been shown to prevent and/or slow down the progression of cardiac remodeling in an animal model of HF [160].
Due to their biological actions as an antioxidant, an anti-inflammatory, and an anticancer agent, polyphenols are commonly present in many foods and have received significant attention. Polyphenols are chemicals with a benzene ring structure with two or more phenolic hydroxyl groups and, depending on their structural properties, they can be divided into flavonoids and phenolic acids. Several positive impacts of polyphenols on health have been documented [161,162,163,164,165,166]. They are endowed with special functional capabilities to exercise their good effects on human health and are added to meals owing to their remarkable biological activities, such as antioxidant and antibacterial effects, as well as their natural availability and biocompatibility. In the prevention of numerous chronic diseases such as diabetes, hypertension, and cancer, the potential significance of functional foods containing polyphenolic chemicals is of importance.
Flavonoids are divided into six main subclasses based on the variety in the type of heterocycle involved: flavonols, flavanones, flavanols, flavones, anthocyanins, and isoflavones. Individual variances within each group are caused by differences in the quantity, arrangement, alkylation, and/or glycosylation levels of the hydroxyl groups. The C ring of flavonols (such as quercetin and kaempferol) has a 3-hydroxy pyran-4-one group. The C ring of flavanones (such as naringenin and taxifolin) contains an unsaturated carbon-carbon bond. The 4-one structure in the C ring and the 3-hydroxyl group are absent from flavanols (such as the catechins). There is no hydroxyl group in the 3-position on the C ring of flavones (such as luteolin). Isoflavones, such as genistein, have the B ring connected to the C ring in the 3-position rather than the 2-position as is the case with other flavonoids. Anthocyanins, such as cyanidin, are characterized by the presence of an oxonium ion on the C ring and are strongly colored as a result.

6. Food-Drug and Herb-Drug Interactions

Drug release, absorption, distribution, metabolism, and/or elimination can be significantly impacted by the concurrent consumption of food or herbs and medications, which can also have a negative/positive impact on the efficacy and safety of pharmacotherapy. Food and herb consumption alters the human gastrointestinal (GI) tract’s physiological environment in a number of ways that can influence a drug’s release, absorption, distribution, metabolism, and/or elimination. These interactions between food, herbs, and drugs are general, thus they will apply to any oral formulation [167]. Their applicability, however, depends on the drug’s formulation and physical characteristics. It is possible to lessen negative drug reactions and iatrogenic disorders by increasing understanding of interindividual variation in drug breakdown capability and results regarding the influence of drugs, nutrition, and herbal items. Medical care needs to be increasingly individually adjusted to each patient, and when diet and herbal treatments are administered in conjunction with medications, this can boost therapeutic efficacy and reduce drug toxicity. Designing a patient’s ideal personal regimen can be aided by understanding how dietary components might increase or decrease pharmacological efficacy. Understanding all the variables can aid in creating individualized medicine, nutrition, and herbal remedy plans for patients, preventing negative side effects, and fostering healing and health [168]. The synergistic impact of some herbs with some drugs to lessen xenobiotic inputs may be intriguing in that situation. Herbs/foods that boost a prescription drug’s effectiveness may allow for lower drug doses when taken concurrently—a potentially advantageous interaction.

7. Conclusions

Numerous epidemiological studies have found a link between consuming natural polyphenols and a lower risk of developing chronic diseases. The risk of developing major cardiovascular illnesses, MI, and cardiovascular mortality is inversely correlated with higher fruit and vegetable consumption [169]. Synthetic medications, herbal medicine and diets have been utilized in the management of CVDs for decades. It is important to note that though many reports establish that the molecular mechanisms of cardiac remodeling have been improved using these therapies individually and not as combinations medication of HF, little research has investigated new therapeutic targets that incorporate all three tactics. While their possible interactions should be considered, a combination of treatments that target independent pathways could proof very efficacious in the treatment of CVD (Figure 2).

Author Contributions

Conceptualization, A.A.O. and L.E.d.C.B.; writing—original draft preparation, A.A.O.; writing—review and editing, A.A.O. and L.E.d.C.B.; supervision and project leader, L.E.d.C.B. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the NIH/NHLBI (grant HL152297).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. Cardiovascular Diseases (CVDs) (World Health Organization). 2017. Available online: https://www.who.int/news-room/factsheets/detail/cardiovascular-diseases-(cvds) (accessed on 18 October 2020).
  2. Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; De Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart disease and stroke statistics—2017 update: A report from the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef]
  3. Frangogiannis, N.G. The inflammatory response in myocardial injury, repair, and remodelling. Nat. Rev. Cardiol. 2014, 11, 255–265. [Google Scholar] [CrossRef] [Green Version]
  4. Lugrin, J.; Rosenblatt-Velin, N.; Parapanov, R.; Liaudet, L. The role of oxidative stress during inflammatory processes. Biol. Chem. 2014, 395, 203–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Serhan CN Novel lipid mediators and resolution mechanisms in acute inflammation: To resolve or not? Am. J. Pathol. 2010, 177, 1576–1591. [CrossRef] [PubMed]
  6. Janicki, J.S.; Brower, G.L.; Henegar, J.R.; Wang, L. Ventricular remodeling in heart failure: The role of myocardial collagen. Adv. Exp. Med. Biol. 1995, 382, 239–245. [Google Scholar] [CrossRef] [PubMed]
  7. Bolognese, L.; Neskovic, A.N.; Parodi, G.; Cerisano, G.; Buonamici, P.; Santoro, G.M.; Antoniucci, D. Left ventricular remodeling after primary coronary angioplasty: Patterns of left ventricular dilation and long-term prognostic implications. Circulation 2002, 106, 2351–2357. [Google Scholar] [CrossRef] [Green Version]
  8. Virani, S.S.; Alonso, A.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Delling, F.N.; et al. Heart disease and stroke statistics—2020 update: A report from the American Heart Association. Circulation 2020, 141, e139–e596. [Google Scholar] [CrossRef] [PubMed]
  9. Conrad, N.; Judge, A.; Tran, J.; Mohseni, H.; Hedgecott, D.; Crespillo, A.P.; Allison, M.; Hemingway, H.; Cleland, J.G.; McMurray, J.J.; et al. Temporal trends and patterns in heart failure incidence: A population-based study of 4 million individuals. Lancet 2018, 391, 572–580. [Google Scholar] [CrossRef] [Green Version]
  10. Störk, S.; Hense, H.W.; Zentgraf, C.; Uebelacker, I.; Jahns, R.; Ertl, G.; Angermann, C.E. Pharmacotherapy according to treatment guidelines is associated with lower mortality in a community-based sample of patients with chronic heart failure A prospective cohort study. Eur. J. Heart Fail. 2008, 10, 1236–1245. [Google Scholar] [CrossRef]
  11. Desta, L.; Jernberg, T.; Spaak, J.; Hofman-Bang, C.; Persson, H. Risk and predictors of readmission for heart failure following a myocardial infarction between 2004 and 2013: A Swedish nationwide observational study. Int. J. Cardiol. 2017, 248, 221–226. [Google Scholar] [CrossRef]
  12. Romiti, G.F.; Recchia, F.; Zito, A.; Visioli, G.; Basili, S.; Raparelli, V. Sex and Gender-Related Issues in Heart Failure. Cardiol. Clin. 2022, 40, 259–268. [Google Scholar] [CrossRef] [PubMed]
  13. Salton, C.J.; Chuang, M.L.; O’Donnell, C.J.; Kupka, M.J.; Larson, M.G.; Kissinger, K.V.; Edelman, R.R.; Levy, D.; Manning, W.J. Gender differences and normal left ventricular anatomy in an adult population free of hypertension: A cardiovascular magnetic resonance study of the Framingham Heart Study Offspring cohort. J. Am. Coll. Cardiol. 2002, 39, 1055–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Laufer, E.M.; Mingels, A.M.; Winkens, M.H.; Joosen, I.A.; Schellings, M.W.; Leiner, T.; Wildberger, J.E.; Narula, J.; Van Dieijen-Visser, M.P. and Hofstra, L. The extent of coronary atherosclerosis is associated with increasing circulating levels of high sensitive cardiac troponin T. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 1269–1275. [Google Scholar] [CrossRef] [PubMed]
  15. Piro, M.; Della Bona, R.; Abbate, A.; Biasucci, L.M.; Crea, F. Sex-related differences in myocardial remodeling. J. Am. Coll. Cardiol. 2010, 55, 1057–1065. [Google Scholar] [CrossRef]
  16. Westerman, S.; Wenger, N.K. Women and heart disease, the underrecognized burden: Sex differences, biases, and unmet clinical and research challenges. Clin. Sci. 2016, 130, 551–563. [Google Scholar] [CrossRef]
  17. Maggioni, A.A.; Maseri, A.; Fresco, C.; Franzosi, M.G.; Mauri, F.; Santoro, E.; Tognoni, G. Age-related increase in mortality among patients with first myocardial infarctions treated with thrombolysis. N. Engl. J. Med. 1993, 329, 1442–1448. [Google Scholar] [CrossRef]
  18. Bujak, M.; Kweon, H.J.; Chatila, K.; Li, N.; Taffet, G.; Frangogiannis, N.G. Aging-related defects are associated with adverse cardiac remodeling in a mouse model of reperfused myocardial infarction. J. Am. Coll. Cardiol. 2008, 51, 1384–1392. [Google Scholar] [CrossRef] [Green Version]
  19. Koitabashi, N.; Kass, D.A. Reverse remodeling in heart failure--mechanisms and therapeutic opportunities. Nat. Rev. Cardiol. 2011, 9, 147–157. [Google Scholar] [CrossRef]
  20. Nisar, B.; Sultan, A.; Rubab, S.L. Comparison of medicinally important natural products versus synthetic drugs-a short commentary. Nat. Prod. Chem. Res. 2018, 6, 308. [Google Scholar] [CrossRef]
  21. Martinez-Gonzalez, M.A.; Fernández-Jarne, E.; Serrano-Martínez, M.; Wright, M.; Gomez-Gracia, E. Development of a short dietary intake questionnaire for the quantitative estimation of adherence to a cardioprotective Mediterranean diet. Eur. J. Clin. Nutr. 2004, 58, 1550–1552. [Google Scholar] [CrossRef] [Green Version]
  22. Goszcz, K.; Deakin, S.J.; Duthie, G.G.; Stewart, D.; Megson, I.L. Bioavailable concentrations of delphinidin and its metabolite, gallic acid, induce antioxidant protection associated with increased intracellular glutathione in cultured endothelial cells. Oxidative Med. Cell. Longev. 2017, 2017, 9260701. [Google Scholar] [CrossRef] [Green Version]
  23. Nabel, E.G.; Braunwald, E.A. Tale of coronary artery disease and myocardial infarction. N. Engl. J. Med. 2012, 366, 54–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Gustafsson, I.; Torp-Pedersen, C.; Køber, L.; Gustafsson, F.; Hildebrandt, P.; Trace Study Group. Effect of the angiotensin-converting enzyme inhibitor trandolapril on mortality and morbidity in diabetic patients with left ventricular dysfunction after acute myocardial infarction. J. Am. Coll. Cardiol. 1999, 34, 83–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Greenberg, B.; Quinones, M.A.; Koilpillai, C.; Limacher, M.; Shindler, D.; Benedict, C.; Shelton, B. Effects of long-term enalapril therapy on cardiac structure and function in patients with left ventricular dysfunction: Results of the SOLVD echocardiography substudy. Circulation 1995, 91, 2573–2581. [Google Scholar] [CrossRef] [PubMed]
  26. Quiñones, M.A.; Greenberg, B.H.; Kopelen, H.A.; Koilpillai, C.; Limacher, M.C.; Shindler, D.M.; Shelton, B.J.; Weiner, D.H.; SOLVD Investigators∗∗. Echocardiographic predictors of clinical outcome in patients with left ventricular dysfunction enrolled in the SOLVD registry and trials: Significance of left ventricular hypertrophy. J. Am. Coll. Cardiol. 2000, 35, 1237–1244. [Google Scholar] [CrossRef] [Green Version]
  27. Anand, I.S.; Florea, V.G. Structural Remodeling in the Development of Chronic Systolic Heart Failure: Implication for Treatment. In Congestive Heart Failure and Cardiac Transplantation; Springer: Cham, Switzerland, 2017; pp. 247–265. [Google Scholar]
  28. Pfeffer, M.A.; Braunwald, E.; Moyé, L.A.; Basta, L.; Brown, E.J., Jr.; Cuddy, T.E.; Davis, B.R.; Geltman, E.M.; Goldman, S.; Flaker, G.C.; et al. Effect of captopril on mortality and morbidity in patients with left ventricular dysfunction after myocardial infarction: Results of the Survival and Ventricular Enlargement Trial. N. Engl. J. Med. 1992, 327, 669–677. [Google Scholar] [CrossRef] [Green Version]
  29. Lopez-Sendon, J.; Swedberg, K.; McMurray, J.; Tamargo, J.; Maggioni, A.P.; Dargie, H.; Tendera, M.; Waagstein, F.; Kjekshus, J.; Lechat, P.; et al. Expert consensus document on angiotensin converting enzyme inhibitors in cardiovascular disease: The Task Force on ACE-inhibitors of the European Society of Cardiology. Eur. Heart J. 2004, 25, 1454–1470. [Google Scholar]
  30. Fedak, P.W.; Verma, S.; Weisel, R.D.; Li, R.K. Cardiac remodeling and failure: From molecules to man (Part I). Cardiovasc. Pathol. 2005, 14, 1–11. [Google Scholar] [CrossRef]
  31. Azizi, M.; Ménard, J.; Bissery, A.; Guyenne, T.T.; Bura-Rivière, A.; Vaidyanathan, S.; Camisasca, R.P. Pharmacologic demonstration of the synergistic effects of a combination of the renin inhibitor aliskiren and the AT1 receptor antagonist valsartan on the angiotensin II–renin feedback interruption. J. Am. Soc. Nephrol. 2004, 15, 3126–3133. [Google Scholar] [CrossRef] [Green Version]
  32. King, M.K.; Coker, M.L.; Goldberg, A.; McElmurray, J.H.; Gunasinghe, H.R., 3rd; Mukherjee, R.; Zile, M.R.; O’Neill, T.P.; Spinale, F.G. Selective matrix metalloproteinase inhibition with developing heart failure: Effects on left ventricular function and structure. Circ. Res. 2003, 92, 177–185. [Google Scholar] [CrossRef] [Green Version]
  33. Ikonomidis, J.S.; Hendrick, J.W.; Parkhurst, A.M.; Herron, A.R.; Escobar, P.G.; Dowdy, K.B.; Stroud, R.E.; Hapke, E.; Zile, M.R.; Spinale, F.G. Accelerated LV remodeling after myocardial infarction in TIMP-1-deficient mice: Effects of exogenous MMP inhibition. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H149–H158. [Google Scholar] [CrossRef]
  34. Matsumura, S.; Iwanaga, S.; Mochizuki, S.; Okamoto, H.; Ogawa, S.; Okada, Y. Targeted deletion or pharmacological inhibition of MMP-2 prevents cardiac rupture after myocardial infarction in mice. J. Clin. Investig. 2005, 115, 599–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Hu, J.; Van den Steen, P.E.; Sang, Q.X.A.; Opdenakker, G. Matrix metalloproteinase inhibitors as therapy for inflammatory and vascular diseases. Nat. Rev. Drug Discov. 2007, 6, 480–498. [Google Scholar] [CrossRef] [PubMed]
  36. Vandenbroucke, R.E.; Libert, C. Is there new hope for therapeutic matrix metalloproteinase inhibition? Nat. Rev. Drug Discov. 2014, 13, 904–927. [Google Scholar] [CrossRef]
  37. Fields, G.B. The rebirth of matrix metalloproteinase inhibitors: Moving beyond the dogma. Cells 2019, 8, 984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Mozaffarian, D. Dietary and policy priorities for cardiovascular disease, diabetes, and obesity: A comprehensive review. Circulation 2016, 133, 187–225. [Google Scholar] [CrossRef]
  39. Chiuve, S.E.; Fung, T.T.; Rimm, E.B.; Hu, F.B.; McCullough, M.L.; Wang, M.; Stampfer, M.J.; Willett, W.C. Alternative dietary indices both strongly predict risk of chronic disease. J. Nutr. 2012, 142, 1009–1018. [Google Scholar] [CrossRef] [Green Version]
  40. van Abeelen, A.F.; Elias, S.G.; Bossuyt, P.M.; Grobbee, D.E.; van der Schouw, Y.T.; Roseboom, T.J.; Uiterwaal, C.S. Cardiovascular consequences of famine in the young. Eur. Heart J. 2012, 33, 538–545. [Google Scholar] [CrossRef] [Green Version]
  41. Ferguson, D.P.; Monroe, T.O.; Heredia, C.P.; Fleischmann, R.; Rodney, G.G.; Taffet, G.E.; Fiorotto, M.L. Postnatal undernutrition alters adult female mouse cardiac structure and function leading to limited exercise capacity. J. Physiol. 2019, 597, 1855–1872. [Google Scholar] [CrossRef]
  42. Bensley, J.G.; Stacy, V.K.; De Matteo, R.; Harding, R.; Black, M.J. Cardiac remodelling as a result of pre-term birth: Implications for future cardiovascular disease. Eur. Heart J. 2010, 31, 2058–2066. [Google Scholar] [CrossRef]
  43. Anatskaya, O.V.; Sidorenko, N.V.; Beyer, T.V.; Vinogradov, A.E. Neonatal cardiomyocyte ploidy reveals critical windows of heart development. Int. J. Cardiol. 2010, 141, 81–91. [Google Scholar] [CrossRef]
  44. Mozaffarian, D.; Ludwig, D.S. Dietary guidelines in the 21st century—A time for food. JAMA 2010, 304, 681–682. [Google Scholar] [CrossRef] [PubMed]
  45. LaCroix, A.Z.; Rillamas-Sun, E.; Buchner, D.; Evenson, K.R.; Di, C.; Lee, I.M.; Marshall, S.; LaMonte, M.J.; Hunt, J.; Tinker, L.F.; et al. The objective physical activity and cardiovascular disease health in older women (OPACH) study. BMC Public Health 2017, 17, 192. [Google Scholar] [CrossRef] [Green Version]
  46. Esposito, K.; Ciotola, M.; Giugliano, D. Mediterranean diet, endothelial function and vascular inflammatory markers. Public Health Nutr. 2006, 9, 1073–1076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Casas, R.; Sacanella, E.; Urpi-Sarda, M.; Chiva-Blanch, G.; Ros, E.; Martinez-Gonzalez, M.A.; Estruch, R. The effects of the mediterranean diet on biomarkers of vascular wall inflammation and plaque vulnerability in subjects with high risk for cardiovascular disease. A randomized trial. PLoS ONE 2014, 9, e100084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Casas, R.; Urpi-Sardà, M.; Sacanella, E.; Arranz, S.; Corella, D.; Castañer, O.; Lamuela-Raventós, R.M.; Salas-Salvadó, J.; Lapetra, J.; Portillo, M.P.; et al. Anti-inflammatory effects of the Mediterranean diet in the early and late stages of atheroma plaque development. Mediat. Inflamm. 2017, 2017, 3674390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Dauchet, L.; Amouyel, P.; Dallongeville, J. Fruits, vegetables and coronary heart disease. Nat. Rev. Cardiol. 2009, 6, 599–608. [Google Scholar] [CrossRef] [PubMed]
  50. Dauchet, L.; Amouyel, P.; Hercberg, S.; Dallongeville, J. Fruit and vegetable consumption and risk of coronary heart disease: A meta-analysis of cohort studies. J. Nutr. 2006, 136, 2588–2593. [Google Scholar] [CrossRef] [Green Version]
  51. Corley, J.; Kyle, J.A.; Starr, J.M.; McNeill, G.; Deary, I.J. Dietary factors and biomarkers of systemic inflammation in older people: The Lothian Birth Cohort 1936. Br. J. Nutr. 2015, 114, 1088–1098. [Google Scholar] [CrossRef] [Green Version]
  52. Hosseini, B.; Berthon, B.S.; Saedisomeolia, A.; Starkey, M.R.; Collison, A.; Wark, P.A.; Wood, L.G. Effects of fruit and vegetable consumption on inflammatory biomarkers and immune cell populations: A systematic literature review and meta-analysis. Am. J. Clin. Nutr. 2018, 108, 136–155. [Google Scholar] [CrossRef] [Green Version]
  53. Arouca, A.; Michels, N.; Moreno, L.A.; González-Gil, E.M.; Marcos, A.; Gómez, S.; Díaz, L.E.; Widhalm, K.; Molnár, D.; Manios, Y.; et al. Associations between a Mediterranean diet pattern and inflammatory biomarkers in European adolescents. Eur. J. Nutr. 2018, 57, 1747–1760. [Google Scholar] [CrossRef]
  54. Wongwarawipat, T.; Papageorgiou, N.; Bertsias, D.; Siasos, G.; Tousoulis, D. Olive oil-related anti-inflammatory effects on atherosclerosis: Potential clinical implications. Endocr. Metab. Immune Disord.-Drug Targets 2018, 18, 51–62. [Google Scholar] [CrossRef] [PubMed]
  55. Schwingshackl, L.; Christoph, M.; Hoffmann, G. Effects of olive oil on markers of inflammation and endothelial function—A systematic review and meta-analysis. Nutrients 2015, 7, 7651–7675. [Google Scholar] [CrossRef] [Green Version]
  56. Mente, A.; de Koning, L.; Shannon, H.S.; Anand, S.S. A systematic review of the evidence supporting a causal link between dietary factors and coronary heart disease. Arch. Intern. Med. 2009, 169, 659–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Aune, D.; Keum, N.; Giovannucci, E.; Fadnes, L.T.; Boffetta, P.; Greenwood, D.C.; Tonstad, S.; Vatten, L.J.; Riboli, E.; Norat, T. Nut consumption and risk of cardiovascular disease, total cancer, all-cause and cause-specific mortality: A systematic review and dose-response meta-analysis of prospective studies. BMC Med. 2016, 14, 207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Guasch-Ferré, M.; Bulló, M.; Martínez-González, M.Á.; Ros, E.; Corella, D.; Estruch, R.; Fitó, M.; Arós, F.; Wärnberg, J.; Fiol, M.; et al. Frequency of nut consumption and mortality risk in the PREDIMED nutrition intervention trial. BMC Med. 2013, 11, 164. [Google Scholar] [CrossRef] [Green Version]
  59. Olabiyi, A.A.; Carvalho, F.B.; Bottari, N.B.; Lopes, T.F.; da Costa, P.; Stefanelo, N.; Morsch, V.M.; Akindahunsi, A.A.; Oboh, G.; Schetinger, M.R. Dietary supplementation of tiger nut alters biochemical parameters relevant to erectile function in L-NAME treated rats. Food Res. Int. 2018, 109, 358–367. [Google Scholar] [CrossRef]
  60. Estruch, R.; Martínez-González, M.A.; Corella, D.; Salas-Salvadó, J.; Ruiz-Gutiérrez, V.; Covas, M.I.; Fiol, M.; Gómez-Gracia, E.; López-Sabater, M.C.; Vinyoles, E.; et al. Effects of a Mediterranean-style diet on cardiovascular risk factors: A randomized trial. Ann. Intern. Med. 2006, 145, 1–11. [Google Scholar] [CrossRef]
  61. Del Gobbo, L.C.; Falk, M.C.; Feldman, R.; Lewis, K.; Mozaffarian, D. Effects of tree nuts on blood lipids, apolipoproteins, and blood pressure: Systematic review, meta-analysis, and dose-response of 61 controlled intervention trials. Am. J. Clin. Nutr. 2015, 102, 1347–1356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Martini, D.; Godos, J.; Marventano, S.; Tieri, M.; Ghelfi, F.; Titta, L.; Lafranconi, A.; Trigueiro, H.; Gambera, A.; Alonzo, E.; et al. Nut and legume consumption and human health: An umbrella review of observational studies. Int. J. Food Sci. Nutr. 2021, 72, 871–878. [Google Scholar] [CrossRef] [PubMed]
  63. Zhou, D.; Yu, H.; He, F.; Reilly, K.H.; Zhang, J.; Li, S.; Zhang, T.; Wang, B.; Ding, Y.; Xi, B. Nut consumption in relation to cardiovascular disease risk and type 2 diabetes: A systematic review and meta-analysis of prospective studies. Am. J. Clin. Nutr. 2014, 100, 270–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Aramwit, P.; Kanokpanont, S.; De-Eknamkul, W.; Srichana, T. Monitoring of inflammatory mediators induced by silk sericin. J. Biosci. Bioeng. 2009, 107, 556–561. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, L.; Teng, H.; Jia, Z.; Battino, M.; Miron, A.; Yu, Z.; Cao, H. and Xiao, J. Intracellular signaling pathways of inflammation modulated by dietary flavonoids: The most recent evidence. Crit. Rev. Food Sci. Nutr. 2018, 58, 2908–2924. [Google Scholar] [CrossRef] [PubMed]
  66. Jönsson, T.; Granfeldt, Y.; Ahrén, B.; Branell, U.C.; Pålsson, G.; Hansson, A.; Söderström, M.; Lindeberg, S. Beneficial effects of a Paleolithic diet on cardiovascular risk factors in type 2 diabetes: A randomized cross-over pilot study. Cardiovasc. Diabetol. 2009, 8, 35. [Google Scholar] [CrossRef] [Green Version]
  67. Jew, S.; AbuMweis, S.S.; Jones, P.J. Evolution of the human diet: Linking our ancestral diet to modern functional foods as a means of chronic disease prevention. J. Med. Food 2009, 12, 925–934. [Google Scholar] [CrossRef] [Green Version]
  68. Cordain, L. The nutritional characteristics of a contemporary diet based upon Paleolithic food groups. J. Am. Nutraceutical. Assoc. 2002, 5, 15–24. [Google Scholar]
  69. Österdahl, M.; Kocturk, T.; Koochek, A.; Wändell, P. Effects of a short-term intervention with a paleolithic diet in healthy volunteers. Eur. J. Clin. Nutr. 2008, 62, 682–685. [Google Scholar] [CrossRef]
  70. Ghaedi, E.; Mohammadi, M.; Mohammadi, H.; Ramezani-Jolfaie, N.; Malekzadeh, J.; Hosseinzadeh, M.; Salehi-Abargouei, A. Effects of a Paleolithic Diet on Cardiovascular Disease Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2019, 10, 634–646, Erratum in Adv. Nutr. 2020, 11, 1054. [Google Scholar] [CrossRef]
  71. Manheimer, E.W.; van Zuuren, E.J.; Fedorowicz, Z.; Pijl, H. Paleolithic nutrition for metabolic syndrome: Systematic review and meta-analysis. Am. J. Clin. Nutr. 2015, 102, 922–932. [Google Scholar] [CrossRef] [Green Version]
  72. Owen, O.E.; Morgan, A.P.; Kemp, H.G.; Sullivan, J.M.; Herrera, M.G.; Cahill, G.F. Brain metabolism during fasting. J. Clin. Investig. 1967, 46, 1589–1595. [Google Scholar] [CrossRef]
  73. Santos, F.L.; Esteves, S.S.; da Costa Pereira, A.; Yancy, W.S.; Nunes, J.P., Jr. Systematic review and meta-analysis of clinical trials of the effects of low carbohydrate diets on cardiovascular risk factors: Low carbohydrate diets and cardiovascular risk factors. Obes. Rev. 2012, 13, 1048–1066. [Google Scholar] [CrossRef] [PubMed]
  74. Bueno, N.B.; de Melo, I.S.; de Oliveira, S.L.; da Rocha Ataide, T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: A meta-analysis of randomised controlled trials. Br. J. Nutr. 2013, 110, 1178–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Naude, C.E.; Schoonees, A.; Senekal, M.; Young, T.; Garner, P.; Volmink, J. Low carbohydrate versus isoenergetic balanced diets for reducing weight and cardiovascular risk: A systematic review and meta-analysis. PLoS ONE 2014, 9, e100652. [Google Scholar] [CrossRef] [Green Version]
  76. Kosinski, C.; Jornayvaz, F.R. Effects of Ketogenic Diets on Cardiovascular Risk Factors: Evidence from Animal and Human Studies. Nutrients 2017, 9, 517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. O’Neill, B.; Raggi, P. The ketogenic diet: Pros and cons. Atherosclerosis 2020, 292, 119–126. [Google Scholar] [CrossRef] [Green Version]
  78. Xu, S.; Tao, H.; Cao, W.; Cao, L.; Lin, Y.; Zhao, S.M.; Xu, W.; Cao, J.; Zhao, J.Y. Ketogenic diets inhibit mitochondrial biogenesis and induce cardiac fibrosis. Signal Transduct. Target Ther. 2021, 6, 54. [Google Scholar] [CrossRef]
  79. Gao, J.W.; Hao, Q.Y.; Zhang, H.F.; Li, X.Z.; Yuan, Z.M.; Guo, Y.; Wang, J.F.; Zhang, S.L.; Liu, P.M. Low-carbohydrate diet score and coronary artery calcium progression: Results from the CARDIA Study. Arterioscler. Thromb. Vasc. Biol. 2020, 41, 491–500. Available online: https://www.ahajournals.org/doi/10.1161/ATVBAHA.120.314838 (accessed on 29 October 2020). [CrossRef]
  80. Garcia-Rios, A.; Ordovas, J.M.; Lopez-Miranda, J.; Perez-Martinez, P. New diet trials and cardiovascular risk. Curr. Opin. Cardiol. 2018, 33, 423–428. [Google Scholar] [CrossRef]
  81. Juraschek, S.P.; Miller III, E.R.; Chang, A.R.; Anderson, C.A.; Hall, J.E.; Appel, L.J. Effects of sodium reduction on energy, metabolism, weight, thirst, and urine volume: Results from the DASH (dietary approaches to stop hypertension)-sodium trial. Hypertension 2020, 75, 723–729. [Google Scholar] [CrossRef] [PubMed]
  82. Kerley, C.P. Dietary patterns and components to prevent and treat heart failure: A comprehensive review of human studies. Nutr. Res. Rev. 2019, 32, 1–27. [Google Scholar] [CrossRef]
  83. Kerley, C.P. A review of plant-based diets to prevent and treat heart failure. Card. Fail. Rev. 2018, 4, 54. [Google Scholar] [CrossRef] [PubMed]
  84. Oboh, G.; Olabiyi, A.A.; Akinyemi, A.J. Inhibitory effect of aqueous extract of different parts of unripe pawpaw (Carica papaya) fruit on Fe2+-induced oxidative stress in rat pancreas in vitro. Pharm. Biol. 2013, 51, 1165–1174. [Google Scholar] [CrossRef] [Green Version]
  85. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661. [Google Scholar] [CrossRef] [Green Version]
  86. Ray, S.; Saini, M.K. Cure and prevention of cardiovascular diseases: Herbs for heart. Clin. Phytoscience 2021, 7, 5741198. [Google Scholar] [CrossRef]
  87. Patwardhan, B.; Mashelkar, R.A. Traditional medicine-inspired approaches to drug discovery: Can Ayurveda show the way forward? Drug Discov. Today 2009, 14, 804–811. [Google Scholar] [CrossRef] [PubMed]
  88. Olabiyi, A.A.; Afolabi, B.A.; Reichert, K.P.; Palma, T.V.; Morsch, V.M.; Oboh, G.; Schetinger, M.R.C. Assessment of sexual behavior and neuromodulation of Cyperus esculentus L. and Tetracarpidium conophorum Müll. Arg dietary supplementation regulating the purinergic system in the cerebral cortex of L-NAME-challenged rats. J. Food Biochem. 2021, 45, e13862. [Google Scholar] [CrossRef] [PubMed]
  89. Badole, S.L.; Bodhankar, S.L.; Patel, N.M.; Bhardwaj, S. Acute and chronic diuretic effect of ethanolic extract of leaves of Cocculus hirsutus (L.) Diles in normal rats. J. Pharm. Pharmacol. 2009, 61, 387–393. [Google Scholar] [CrossRef]
  90. Davison, E.K.; Brimble, M.A. Natural product derived privileged scaffolds in drug discovery. Curr. Opin. Chem. Biol. 2019, 52, 1–8. [Google Scholar] [CrossRef]
  91. Olabiyi, A.A.; Ajayi, K. Diet, herbs and erectile function: A good friendship! Andrologia 2022, 54, e14424. [Google Scholar] [CrossRef]
  92. Shaito, A.; Thuan, D.T.B.; Phu, H.T.; Nguyen, T.H.D.; Hasan, H.; Halabi, S.; Abdelhady, S.; Nasrallah, G.K.; Eid, A.H.; Pintus, G. Herbal medicine for cardiovascular diseases: Efficacy, mechanisms, and safety. Front. Pharmacol. 2020, 11, 422. [Google Scholar] [CrossRef] [Green Version]
  93. Silva, H.; Martins, F.G. Cardiovascular Activity of Ginkgo biloba—An Insight from Healthy Subjects. Biology 2022, 12, 15. [Google Scholar] [CrossRef] [PubMed]
  94. Upadhyay, S.; Dixit, M. Role of polyphenols and other phytochemicals on molecular signaling. Oxidative Med. Cell. Longev. 2015, 2015, 504253. [Google Scholar] [CrossRef] [Green Version]
  95. Murino Rafacho, B.P.; Portugal dos Santos, P.; Goncalves, A.D.F.; Fernandes, A.A.H.; Okoshi, K.; Chiuso-Minicucci, F.; Azevedo, P.S.; Mamede Zornoff, L.A.; Minicucci, M.F.; Wang, X.D.; et al. Rosemary supplementation (Rosmarinus oficinallis L.) attenuates cardiac remodeling after myocardial infarction in rats. PLoS ONE 2017, 12, e0177521. [Google Scholar] [CrossRef] [Green Version]
  96. Butnariu, M.; Quispe, C.; Herrera-Bravo, J.; Sharifi-Rad, J.; Singh, L.; Aborehab, N.M.; Bouyahya, A.; Venditti, A.; Sen, S.; Acharya, K.; et al. The pharmacological activities of Crocus sativus L.: A review based on the mechanisms and therapeutic opportunities of its phytoconstituents. Oxidative Med. Cell. Longev. 2022, 2022, 8214821. [Google Scholar] [CrossRef] [PubMed]
  97. Khazdair, M.R.; Boskabady, M.H.; Hosseini, M.; Rezaee, R.; Tsatsakis, A.M. The effects of Crocus sativus (saffron) and its constituents on nervous system: A review. Avicenna J. Phytomedicine 2015, 5, 376. [Google Scholar]
  98. Mzabri, I.; Addi, M.; Berrichi, A. Traditional and modern uses of saffron (Crocus sativus). Cosmetics 2019, 6, 63. [Google Scholar] [CrossRef] [Green Version]
  99. Sargolzaei, J.; Shabestari, M.M. The effects of Crocus Sativus, L. and its main constituents against cardiovascular diseases. Der Pharm. Lett. 2016, 8, 38–41. [Google Scholar]
  100. Al-Yahya, M.A.; Mothana, R.A.; Al-Said, M.S.; El-Tahir, K.E.; Al-Sohaibani, M.; Rafatullah, S. Citrus medica “Otroj”: Attenuates oxidative stress and cardiac dysrhythmia in isoproterenol-induced cardiomyopathy in rats. Nutrients 2013, 5, 4269–4283. [Google Scholar] [CrossRef] [Green Version]
  101. Tassell, M.C.; Kingston, R.; Gilroy, D.; Lehane, M.; Furey, A. Hawthorn (Crataegus spp.) in the treatment of cardiovascular disease. Pharmacogn. Rev. 2010, 4, 32. [Google Scholar]
  102. Altinterim, B. Cardiovascular effects of Hawthorn (Crataegus monogyna). KSÜ Doğa Bilim. Derg. 2012, 15, 16–18. [Google Scholar]
  103. Verma, S.K.; Jain, V.; Katewa, S.S. Blood pressure lowering, fibrinolysis enhancing and antioxidant activities of cardamom (Elettaria cardamomum). Indian J. Biochem. Biophys. 2009, 46, 503–506. [Google Scholar]
  104. Nagashree, S.; Archana, K.K.; Srinivas, P.; Srinivasan, K.; Sowbhagya, H.B. Anti-hypercholesterolemic influence of the spice cardamom (Elettaria cardamomum) in experimental rats. J. Sci. Food Agric. 2017, 97, 3204–3210. [Google Scholar] [CrossRef]
  105. Arbeláez, L.F.G.; Pardo, A.C.; Fantinelli, J.C.; Schinella, G.R.; Mosca, S.M.; Ríos, J.L. Cardioprotection and natural polyphenols: An update of clinical and experimental studies. Food Funct. 2018, 9, 6129–6145. [Google Scholar] [CrossRef]
  106. Lee, C.H.; Kim, J.H. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. J. Ginseng Res. 2014, 38, 161–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Chan, S.W.; Tomlinson, B.; Chan, P.; Lam, C.W.K. The beneficial effects of Ganoderma lucidum on cardiovascular and metabolic disease risk. Pharm. Biol. 2021, 59, 1161–1171. [Google Scholar] [CrossRef]
  108. Aviram, M.; Dornfeld, L. Pomegranate juice consumption inhibits serum angiotensin converting enzyme activity and reduces systolic blood pressure. Atherosclerosis 2001, 158, 195–198. [Google Scholar] [CrossRef]
  109. Asgary, S.; Haghjooyjavanmard, S.; Setorki, M.; Rafieian, M.; Haghighi, S.; Eidi, A.; Rohani, A.H. The postprandial effect of apple juice intake on some of the biochemical risk factors of atherosclerosis in male rabbit. J. Med. Plants Res. 2009, 3, 785–790. [Google Scholar]
  110. Asgary, S.; Sahebkar, A.; Afshani, M.R.; Keshvari, M.; Haghjooyjavanmard, S.; Rafieian-Kopaei, M. Clinical evaluation of blood pressure lowering, endothelial function improving, hypolipidemic and anti-inflammatory effects of pomegranate juice in hypertensive subjects. Phytother. Res. 2014, 28, 193–199. [Google Scholar] [CrossRef]
  111. Setorki, M.; Nazari, B.; Asgary, S.; Eidi, A.; Rohani, A.H. Acute effects of apple cider vinegar intake on some biochemical risk factors of atherosclerosis in rabbits fed with a high cholesterol diet. Qom Univ. Med. Sci. J. 2010, 3, 10. [Google Scholar]
  112. Setorki, M.; Asgary, S.; Eidi, A.; Rohani, A.H.; Esmaeil, N. Effects of apple juice on risk factors of lipid profile, inflammation and coagulation, endothelial markers and atherosclerotic lesions in high cholesterolemic rabbits. Lipids Health Dis. 2009, 8, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Omoni, A.O.; Aluko, R.E. The anti-carcinogenic and anti-atherogenic effects of lycopene: A review. Trends Food Sci. Technol. 2005, 16, 344–350. [Google Scholar] [CrossRef]
  114. Naz, A.; Butt, M.S.; Sultan, M.T.; Qayyum, M.M.N.; Niaz, R.S. Watermelon lycopene and allied health claims. EXCLI J. 2014, 13, 650. [Google Scholar] [PubMed]
  115. Chiva-Blanch, G.; Visioli, F. Polyphenols and health: Moving beyond antioxidants. J. Berry Res. 2012, 2, 63–71. [Google Scholar] [CrossRef] [Green Version]
  116. Niki, E. Antioxidant capacity: Which capacity and how to assess it? J. Berry Res. 2011, 1, 169–176. [Google Scholar] [CrossRef] [Green Version]
  117. Cassidy, A.; Mukamal, K.J.; Liu, L.; Franz, M.; Eliassen, A.H.; Rimm, E.B. High anthocyanin intake is associated with a reduced risk of myocardial infarction in young and middle-aged women. Circulation 2013, 127, 188–196. [Google Scholar] [CrossRef] [Green Version]
  118. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.; Fong, H.H.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; et al. Cancer chemopreventive activity of resveratrol, a natural product derived from grapes. Science 1997, 275, 218–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Seymour, E.M.; Singer, A.A.; Bennink, M.R.; Parikh, R.V.; Kirakosyan, A.; Kaufman, P.B.; Bolling, S.F. Chronic intake of a phytochemical-enriched diet reduces cardiac fibrosis and diastolic dysfunction caused by prolonged salt-sensitive hypertension. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2008, 63, 1034–1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Varshney, R.; Budoff, M.J. Garlic and heart disease. J. Nutr. 2016, 146, 416S–421S. [Google Scholar] [CrossRef] [Green Version]
  121. Ko, F.N.; Yu, S.M.; Kang, Y.F.; Teng, C.M. Characterization of the thromboxane (TP-) receptor subtype involved in proliferation in cultured vascular smooth muscle cells of rat. Br. J. Pharmacol. 1995, 116, 1801–1808. [Google Scholar] [CrossRef] [Green Version]
  122. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxidative Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [Green Version]
  123. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant polyphenols: Chemical properties, biological activities, and synthesis. Angew. Chem. Int. Ed. 2011, 50, 586–621. [Google Scholar] [CrossRef] [PubMed]
  124. Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef] [Green Version]
  125. Kumar, N.; Goel, N. Phenolic acids: Natural versatile molecules with promising therapeutic applications. Biotechnol. Rep. 2019, 24, e00370. [Google Scholar] [CrossRef]
  126. Vitrac, X.; Monti, J.P.; Vercauteren, J.; Deffieux, G.; Mérillon, J.M. Direct liquid chromatographic analysis of resveratrol derivatives and flavanonols in wines with absorbance and fluorescence detection. Anal. Chim. Acta 2002, 458, 103–110. [Google Scholar] [CrossRef]
  127. Prasad, R.; Kawaguchi, S.; Ng, D.T. Biosynthetic mode can determine the mechanism of protein quality control. Biochem. Biophys. Res. Commun. 2012, 425, 689–695. [Google Scholar] [CrossRef] [PubMed]
  128. Korkina, L.; Kostyuk, V.; De Luca, C.; Pastore, S. Plant phenylpropanoids as emerging anti-inflammatory agents. Mini Rev. Med. Chem. 2011, 11, 823–835. [Google Scholar] [CrossRef] [PubMed]
  129. Saleem, M.; Kim, H.J.; Ali, M.S.; Lee, Y.S. An update on bioactive plant lignans. Nat. Prod. Rep. 2005, 22, 696–716. [Google Scholar] [CrossRef]
  130. Ly, C.; Yockell-Lelievre, J.; Ferraro, Z.M.; Arnason, J.T.; Ferrier, J.; Gruslin, A. The effects of dietary polyphenols on reproductive health and early development. Hum. Reprod. Update 2015, 21, 228–248. [Google Scholar] [CrossRef] [Green Version]
  131. Smeds, A.I.; Eklund, P.C.; Willför, S.M. Content, composition, and stereochemical characterisation of lignans in berries and seeds. Food Chem. 2012, 134, 1991–1998. [Google Scholar] [CrossRef]
  132. Khan, N.; Mukhtar, H. Tea polyphenols for health promotion. Life Sci. 2007, 81, 519–533. [Google Scholar] [CrossRef] [Green Version]
  133. Hollman, P.C.H.; Arts, I.C.W. Flavonols, flavones and flavanols–nature, occurrence and dietary burden. J. Sci. Food Agric. 2000, 80, 1081–1093. [Google Scholar] [CrossRef]
  134. Cassidy, A.; Hanley, B.; Lamuela-Raventos, R.M. Isoflavones, lignans and stilbenes–origins, metabolism and potential importance to human health. J. Sci. Food Agric. 2000, 80, 1044–1062. [Google Scholar] [CrossRef]
  135. Brglez Mojzer, E.; Knez Hrnčič, M.; Škerget, M.; Knez, Ž.; Bren, U. Polyphenols: Extraction Methods, Antioxidative Action, Bioavailability and Anticarcinogenic Effects. Molecules 2016, 21, 901. [Google Scholar] [CrossRef] [PubMed]
  136. Mateos-Martín, M.L.; Fuguet, E.; Quero, C.; Pérez-Jiménez, J.; Torres, J.L. New identification of proanthocyanidins in cinnamon (Cinnamomum zeylanicum L.) using MALDI-TOF/TOF mass spectrometry. Anal. Bioanal. Chem. 2012, 402, 1327–1336. [Google Scholar] [CrossRef]
  137. Han, X.; Shen, T.; Lou, H. Dietary polyphenols and their biological significance. Int. J. Mol. Sci. 2007, 8, 950–988. [Google Scholar] [CrossRef] [Green Version]
  138. Raj, P.; Louis, X.L.; Thandapilly, S.J.; Movahed, A.; Zieroth, S.; Netticadan, T. Potential of resveratrol in the treatment of heart failure. Life Sci. 2014, 95, 63–71. [Google Scholar] [CrossRef]
  139. Niu, L.; He, X.H.; Wang, Q.W.; Fu, M.Y.; Xu, F.; Xue, Y.; Wang, Z.Z.; An, X.J. Polyphenols in regulation of redox signaling and inflammation during cardiovascular diseases. Cell Biochem. Biophys. 2015, 72, 485–494. [Google Scholar] [CrossRef]
  140. Li, M.; Jiang, Y.; Jing, W.; Sun, B.; Miao, C.; Ren, L. Quercetin provides greater cardioprotective effect than its glycoside derivative rutin on isoproterenol-induced cardiac fibrosis in the rat. Can. J. Physiol. Pharmacol. 2013, 91, 951–959. [Google Scholar] [CrossRef]
  141. Panchal, S.K.; Poudyal, H.; Brown, L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J. Nutr. 2012, 142, 1026–1032. [Google Scholar] [CrossRef] [Green Version]
  142. Mak, J.C. Potential role of green tea catechins in various disease therapies: Progress and promise. Clin. Exp. Pharmacol. Physiol. 2012, 39, 265–273. [Google Scholar] [CrossRef]
  143. Sheng, R.; Gu, Z.L.; Xie, M.L.; Zhou, W.X.; Guo, C.Y. EGCG inhibits cardiomyocyte apoptosis in pressure overload-induced cardiac hypertrophy and protects cardiomyocytes from oxidative stress in rats 1. Acta Pharmacol. Sin. 2007, 28, 191–201. [Google Scholar] [CrossRef] [PubMed]
  144. Sheng, R.; Gu, Z.L.; Xie, M.L.; Zhou, W.X.; Guo, C.Y. EGCG inhibits proliferation of cardiac fibroblasts in rats with cardiac hypertrophy. Planta Med. 2009, 75, 113–120. [Google Scholar] [CrossRef] [PubMed]
  145. Han, Y.S.; Lan, L.; Chu, J.; Kang, W.Q.; Ge, Z.M. Epigallocatechin gallate attenuated the activation of rat cardiac fibroblasts induced by angiotensin II via regulating β-arrestin1. Cell. Physiol. Biochem. 2013, 31, 338–346. [Google Scholar] [CrossRef]
  146. Cai, Y.; Yu, S.S.; Chen, T.T.; Gao, S.; Geng, B.; Yu, Y.; Ye, J.T.; Liu, P.Q. EGCG inhibits CTGF expression via blocking NF-κB activation in cardiac fibroblast. Phytomedicine 2013, 20, 106–113. [Google Scholar] [CrossRef]
  147. Cai, Y.; He, S.Q.; Hong, H.Q.; Cai, Y.P.; Zhao, L.; Zhang, M. High doses of (−)-epigallocatechin-3-gallate from green tea induces cardiac fibrosis in mice. Biotechnol. Lett. 2015, 37, 2371–2377. [Google Scholar] [CrossRef] [PubMed]
  148. Heymans, S.; González, A.; Pizard, A.; Papageorgiou, A.P.; López-Andrés, N.; Jaisser, F.; Thum, T.; Zannad, F.; Díez, J. Searching for new mechanisms of myocardial fibrosis with diagnostic and/or therapeutic potential. Eur. J. Heart Fail. 2015, 17, 764–771. [Google Scholar] [CrossRef] [Green Version]
  149. Wang, T.; Pan, D.; Zhang, Y.; Li, D.; Zhang, Y.; Xu, T.; Luo, Y.; Ma, Y. Luteolin antagonizes angiotensin II-dependent proliferation and collagen synthesis of cultured rat cardiac fibroblasts. Curr. Pharm. Biotechnol. 2015, 16, 430–439. [Google Scholar] [CrossRef]
  150. Nakayama, A.; Morita, H.; Nakao, T.; Yamaguchi, T.; Sumida, T.; Ikeda, Y.; Kumagai, H.; Motozawa, Y.; Takahashi, T.; Imaizumi, A.; et al. A food-derived flavonoid luteolin protects against angiotensin II-induced cardiac remodeling. PLoS ONE 2015, 10, e0137106. [Google Scholar] [CrossRef] [Green Version]
  151. Sun, C.D.; Zhang, B.; Zhang, J.K.; Xu, C.J.; Wu, Y.L.; Li, X.; Chen, K.S. Cyanidin-3-glucoside-rich extract from Chinese bayberry fruit protects pancreatic β cells and ameliorates hyperglycemia in streptozotocin-induced diabetic mice. J. Med. Food 2012, 15, 288–298. [Google Scholar] [CrossRef] [Green Version]
  152. Skemiene, K.; Liobikas, J.; Borutaite, V. Anthocyanins as substrates for mitochondrial complex I–protective effect against heart ischemic injury. FEBS J. 2015, 282, 963–971. [Google Scholar] [CrossRef]
  153. Škėmienė, K.; Jablonskienė, G.; Liobikas, J.; Borutaitė, V. Protecting the heart against ischemia/reperfusion-induced necrosis and apoptosis: The effect of anthocyanins. Medicina 2013, 49, 15. [Google Scholar] [CrossRef]
  154. Chen, Y.F.; Shibu, M.A.; Fan, M.J.; Chen, M.C.; Viswanadha, V.P.; Lin, Y.L.; Lai, C.H.; Lin, K.H.; Ho, T.J.; Kuo, W.W.; et al. Purple rice anthocyanin extract protects cardiac function in STZ-induced diabetes rat hearts by inhibiting cardiac hypertrophy and fibrosis. J. Nutr. Biochem. 2016, 31, 98–105. [Google Scholar] [CrossRef] [PubMed]
  155. Sunagawa, Y.; Morimoto, T.; Wada, H.; Takaya, T.; Katanasaka, Y.; Kawamura, T.; Yanagi, S.; Marui, A.; Sakata, R.; Shimatsu, A.; et al. A natural p300-specific histone acetyltransferase inhibitor, curcumin, in addition to angiotensin-converting enzyme inhibitor, exerts beneficial effects on left ventricular systolic function after myocardial infarction in rats. Circ. J. 2011, 75, 2151–2159. [Google Scholar] [CrossRef] [Green Version]
  156. Sunagawa, Y.; Sono, S.; Katanasaka, Y.; Funamoto, M.; Hirano, S.; Miyazaki, Y.; Hojo, Y.; Suzuki, H.; Morimoto, E.; Marui, A.; et al. Optimal dose-setting study of curcumin for improvement of left ventricular systolic function after myocardial infarction in rats. J. Pharmacol. Sci. 2014, 126, 329–336. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Wang, N.P.; Wang, Z.F.; Tootle, S.; Philip, T.; Zhao, Z.Q. Curcumin promotes cardiac repair and ameliorates cardiac dysfunction following myocardial infarction. Br. J. Pharmacol. 2012, 167, 1550–1562. [Google Scholar] [CrossRef] [Green Version]
  158. Xiao, J.; Sheng, X.; Zhang, X.; Guo, M.; Ji, X. Curcumin protects against myocardial infarction-induced cardiac fibrosis via SIRT1 activation in vivo and in vitro. Drug Des. Dev. Ther. 2016, 10, 1267. [Google Scholar] [CrossRef]
  159. Zeng, C.; Zhong, P.; Zhao, Y.; Kanchana, K.; Zhang, Y.; Khan, Z.A.; Chakrabarti, S.; Wu, L.; Wang, J.; Liang, G. Curcumin protects hearts from FFA-induced injury by activating Nrf2 and inactivating NF-κB both in vitro and in vivo. J. Mol. Cell. Cardiol. 2015, 79, 1–12. [Google Scholar] [CrossRef]
  160. Sung, M.M.; Dyck, J.R. Therapeutic potential of resveratrol in heart failure. Ann. N. Y. Acad. Sci. 2015, 1348, 32–45. [Google Scholar] [CrossRef]
  161. Song, H.; Wang, Q.; He, A.; Li, S.; Guan, X.; Hu, Y.; Feng, S. Antioxidant activity, storage stability and in vitro release of epigallocatechin-3-gallate (EGCG) encapsulated in hordein nanoparticles. Food Chem. 2022, 388, 132903. [Google Scholar] [CrossRef]
  162. Qiu, C.; McClements, D.J.; Jin, Z.; Qin, Y.; Hu, Y.; Xu, X.; Wang, J. Resveratrol-loaded core-shell nanostructured delivery systems: Cyclodextrin-based metal-organic nanocapsules prepared by ionic gelation. Food Chem. 2020, 317, 126328. [Google Scholar] [CrossRef]
  163. Ding, H.W.; Huang, A.L.; Zhang, Y.L.; Li, B.; Huang, C.; Ma, T.T.; Meng, X.M.; Li, J. Design, synthesis and biological evaluation of hesperetin derivatives as potent anti-inflammatory agent. Fitoterapia 2017, 121, 212–222. [Google Scholar] [CrossRef]
  164. Lee, S.H.; Lee, Y.J. Synergistic anticancer activity of resveratrol in combination with docetaxel in prostate carcinoma cells. Nutr. Res. Pract. 2021, 15, 12–25. [Google Scholar] [CrossRef]
  165. Quiñones, M.; Guerrero, L.; Suarez, M.; Pons, Z.; Aleixandre, A.; Arola, L.; Muguerza, B. Low-molecular procyanidin rich grape seed extract exerts antihypertensive effect in males spontaneously hypertensive rats. Food Res. Int. 2013, 51, 587–595. [Google Scholar] [CrossRef]
  166. Rains, T.M.; Agarwal, S.; Maki, K.C. Antiobesity effects of green tea catechins: A mechanistic review. J. Nutr. Biochem. 2011, 22, 1–7. [Google Scholar] [CrossRef] [PubMed]
  167. Koziolek, M.; Alcaro, S.; Augustijns, P.; Basit, A.W.; Grimm, M.; Hens, B.; Hoad, C.L.; Jedamzik, P.; Madla, C.M.; Maliepaard, M.; et al. The mechanisms of pharmacokinetic food-drug interactions—A perspective from the UNGAP group. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2019, 134, 31–59. [Google Scholar] [CrossRef]
  168. O’Shea, J.P.; Holm, R.; O’Driscoll, C.M.; Griffin, B.T. Food for thought: Formulating away the food effect–a PEARRL review. J. Pharm. Pharmacol. 2019, 71, 510–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Miller, V.; Mente, A.; Dehghan, M.; Rangarajan, S.; Zhang, X.; Swaminathan, S.; Dagenais, G.; Gupta, R.; Mohan, V.; Lear, S.; et al. Fruit, vegetable, and legume intake, and cardiovascular disease and deaths in 18 countries (PURE): A prospective cohort study. Lancet 2017, 390, 2037–2049. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Chemical compositions of the various polyphenol classes. The amount of phenol rings a polyphenol contains and the structural components that hold these rings together are used to categorize polyphenols. They can be generally divided into four classes: stilbenes, lignans, phenolic acids, and flavonoids. Hydroxyl benzoic and hydroxyl cinnamic acids are two more subgroups of phenolic acids. Phenolic acids, which are present in all plant material but are more prevalent in fruits with an acidic flavor, make up around one-third of the polyphenolic substances in our diet. Some common phenolic acids include ferulic acid, gallic acid, and caffeic acid. The most prevalent polyphenols in the human diet are flavonoids, which have a basic structure that consists of two aromatic rings connected by three carbon atoms to create an oxygenated heterocycle. Biogenetically, one ring typically develops from a resorcinol molecule, while the other ring develops from the shikimate route. Two phenyl moieties are joined by a two-carbon methylene bridge in stilbenes. The majority of stilbenes in plants function as phytoalexins, which are substances that are only produced in reaction to infection or damage. Resveratrol has been the stilbene that has been investigated the most. The 2,3-dibenzylbutane structure of lignans, which are diphenolic substances, is created when two cinnamic acid residues dimerize.
Figure 1. Chemical compositions of the various polyphenol classes. The amount of phenol rings a polyphenol contains and the structural components that hold these rings together are used to categorize polyphenols. They can be generally divided into four classes: stilbenes, lignans, phenolic acids, and flavonoids. Hydroxyl benzoic and hydroxyl cinnamic acids are two more subgroups of phenolic acids. Phenolic acids, which are present in all plant material but are more prevalent in fruits with an acidic flavor, make up around one-third of the polyphenolic substances in our diet. Some common phenolic acids include ferulic acid, gallic acid, and caffeic acid. The most prevalent polyphenols in the human diet are flavonoids, which have a basic structure that consists of two aromatic rings connected by three carbon atoms to create an oxygenated heterocycle. Biogenetically, one ring typically develops from a resorcinol molecule, while the other ring develops from the shikimate route. Two phenyl moieties are joined by a two-carbon methylene bridge in stilbenes. The majority of stilbenes in plants function as phytoalexins, which are substances that are only produced in reaction to infection or damage. Resveratrol has been the stilbene that has been investigated the most. The 2,3-dibenzylbutane structure of lignans, which are diphenolic substances, is created when two cinnamic acid residues dimerize.
Biomedicines 11 01697 g001
Figure 2. Diagrammatic representation of the influence of synthetic pharmaceutical medications, dietary and herbal products in CVD management, with emphasis in common pathways that are targeted by each intervention. DAMPs = damage-associated molecular patterns, SPMs = specific pro-resolving mediators, CFs = cardiac fibroblasts, NOX = NADPH oxidases, TNF-α = tumor necrosis factor, MMPs = metalloproteinases, DNA = deoxyribonucleic acid, ECM = extracellular matrix. Figure created with Biorender.com.
Figure 2. Diagrammatic representation of the influence of synthetic pharmaceutical medications, dietary and herbal products in CVD management, with emphasis in common pathways that are targeted by each intervention. DAMPs = damage-associated molecular patterns, SPMs = specific pro-resolving mediators, CFs = cardiac fibroblasts, NOX = NADPH oxidases, TNF-α = tumor necrosis factor, MMPs = metalloproteinases, DNA = deoxyribonucleic acid, ECM = extracellular matrix. Figure created with Biorender.com.
Biomedicines 11 01697 g002
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

Olabiyi, A.A.; de Castro Brás, L.E. Cardiovascular Remodeling Post-Ischemia: Herbs, Diet, and Drug Interventions. Biomedicines 2023, 11, 1697. https://doi.org/10.3390/biomedicines11061697

AMA Style

Olabiyi AA, de Castro Brás LE. Cardiovascular Remodeling Post-Ischemia: Herbs, Diet, and Drug Interventions. Biomedicines. 2023; 11(6):1697. https://doi.org/10.3390/biomedicines11061697

Chicago/Turabian Style

Olabiyi, Ayodeji A., and Lisandra E. de Castro Brás. 2023. "Cardiovascular Remodeling Post-Ischemia: Herbs, Diet, and Drug Interventions" Biomedicines 11, no. 6: 1697. https://doi.org/10.3390/biomedicines11061697

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