Next Article in Journal
Treatment Options for Troublesome Itch
Next Article in Special Issue
Looking beyond the Skin: Pathophysiology of Cardiovascular Comorbidity in Psoriasis and the Protective Role of Biologics
Previous Article in Journal
A Versatile Class of 1,4,4-Trisubstituted Piperidines Block Coronavirus Replication In Vitro
Previous Article in Special Issue
Pharmaceutical Prevention and Management of Cardiotoxicity in Hematological Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Pharmaceutical Applications of Quercetin in Cardiovascular Diseases

1
Laboratory of Biopharmaceutics-Pharmacokinetics, Department of Pharmacy, School of Health Sciences, National & Kapodistrian University of Athens, 15784 Athens, Greece
2
Medical School, University of Cyprus, Nicosia 2029, Cyprus
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2022, 15(8), 1019; https://doi.org/10.3390/ph15081019
Submission received: 22 July 2022 / Revised: 9 August 2022 / Accepted: 11 August 2022 / Published: 18 August 2022

Abstract

:
Quercetin, as a member of flavonoids, has emerged as a potential therapeutic agent in cardiovascular diseases (CVDs) in recent decades. In this comprehensive literature review, our goal was a critical appraisal of the pathophysiological mechanisms of quercetin in relation to the classical cardiovascular risk factors (e.g., hyperlipidemia), atherosclerosis, etc. We also assessed experimental and clinical data about its potential application in CVDs. Experimental studies including both in vitro methods and in vivo animal models mainly outline the following effects of quercetin: (1) antihypertensive, (2) hypolipidemic, (3) hypoglycemic, (4) anti-atherosclerotic, and (5) cardioprotective (suppressed cardiotoxicity). From the clinical point of view, there are human studies and meta-analyses implicating its beneficial effects on glycemic and lipid parameters. In contrast, other human studies failed to demonstrate consistent favorable effects of quercetin on other cardiometabolic risk factors such as MS, obesity, and hypertension, underlying the need for further investigation. Analyzing the reason of this inconsistency, we identified significant drawbacks in the clinical trials’ design, while the absence of pharmacokinetic/pharmacodynamic tests prior to the studies attenuated the power of clinical results. Therefore, additional well-designed preclinical and clinical studies are required to examine the therapeutic mechanisms and clinical efficacy of quercetin in CVDs.

1. Introduction

Flavonol quercetin has recently received extensive attention from the medical and research communities because of its significant potential therapeutical applications, including antioxidant, antitumor, antiatherogenic, antithrombotic, and cardioprotective properties [1,2,3]. In 1930, rutin (quercetin 3-O-rutinoside), one of quercetin’s most common forms, was isolated from oranges, and it was initially believed to be a vitamin; for a while, rutin was called vitamin P [4]. Quercetin, from Latin quercetum (oak forest), was first described in 1936 [5,6]. It is a low-cost substance found widely in many plant species [1]. Fruits, vegetables, tea, and red wine are the major sources of flavonols. However, some characteristics of the molecule (Figure 1), including low water solubility, poor bioavailability, instability in the biological environment, and enzymatic degradation with very small half-life, have largely prevented its clinical usage [2,3]. Trying to overcome those drawbacks, other studies examined its binding ability with iron [7] and copper [8] to improve its bioavailability.
Recently, preventive and therapeutic effects of flavonoids on the cardiovascular system, alone or in combination with other agents, have been outlined in a plethora of studies [3]. The first randomized controlled trials (RCTs) examining the cardioprotective effects of quercetin on healthy subjects were conducted in 1998 [9,10]. Subsequently, a growing body of evidence has supported the clinical importance of modified dosage forms of quercetin, using other carriers such as nanoparticles and microemulsions [2,11].
The aim of this comprehensive review was to summarize and critically discuss the main pharmacological mechanisms of quercetin in relation to atherosclerosis and the classical cardiovascular risk factors such as diabetes mellitus (DM), hyperlipidemia, etc. We also tried to assess the potential application (protective and/or therapeutic) in cardiovascular diseases (CVDs) based on experimental and clinical studies. The main cardiovascular protective properties of quercetin are depicted with the possible mechanisms of action in Figure 1.

2. Literature Search Strategy

For the needs of this review paper, we searched MEDLINE and EMBASE, Web of Science, Cochrane, and Google Scholar databases for English-language publications from 1990 to May 2022. We also checked the reference lists of the identified articles to find any additional relevant articles. Our search included the titles, abstracts, and medical subject headings (MeSH), and we used the following search terms: quercetin, atherosclerosis, diabetes mellitus, hyperglycemia, hypertension, hyperlipidemia, metabolic syndrome, cardiovascular diseases, coronary artery disease. Four investigators (N.B., C.P., M.V., and P.P.) performed the literature search independently. In our search, we included both in vitro and in vivo preclinical experimental studies, as well as clinical studies (both clinical investigations and clinical meta-analyses). To draw firm conclusions, we excluded the following studies: full text unavailable, publication language other than English, conference abstracts, and mixing of quercetin and other substances in interventional arms.
Using the abovementioned terms, we initially found 2120 hits. After the abstracts’ screening, we removed 192 duplicated studies, and also 1865 irrelevant studies; 110 full-text studies were screened for eligibility. After removing the studies with wrong design, irrelevant outcomes, unavailable full text, we ended up with a total of 30 experimental studies, 10 systematic reviews and meta-analyses of clinical data, and 20 additional clinical studies which were not included in those meta-analyses.

3. Results and Discussion

3.1. Chemical Structure of Quercetin and Its Main Derivatives

Flavonoids are a group of different phytonutrients with variable phenolic structures found in almost all plants [12]. These natural low-molecular-weight substances are secondary metabolites characterized by a polyphenolic chemical structure [4,12]. The main feature of all flavonoids is the basic flavan skeleton: a 15-carbon phenylpropanoid chain, C6–C3–C6, which consists of two aromatic rings (A and B) and a heterocyclic pyrane ring (C) [13] (Figure 1). The different subgroups of flavonoids result from the carbon attached between the two rings (B and C) of the molecule and the degree of unsaturation and oxidation of the C ring [3,12] (Figure 2). Flavonoids can be subdivided into anthocyanins, chalcones, flavanones, flavones, flavonols, and isoflavonoids [3,13]. Quercetin (3,3′,4′,5,7-pentahydroxyflavone) belongs to flavonols with a ketone group [12] (Figure 1). Flavonols are characterized by an unsaturated C ring at the C2–C3 position, which can be hydroxylated at C3 and oxidized at C4 [13]. There are variable methylation, hydroxylation, and glycosylation patterns leading to the formation of quercetin derivatives (Figure 2). Glycosylation may occur at any of the five hydroxyl groups of quercetin, but most commonly occurs at the 3-OH group [1,12,14,15]. Quercetin has strong antioxidant properties because of its chemical structure, including its highly reactive hydroxyl groups, catechol functionality (ortho-dihydroxyl) on the B ring, and a Δ2 double bond adjacent to the 4-oxo group in the C ring [16]. Studies have shown that quercetin can exist conjugated with carbohydrates, lipids, alcohol, and sulfate group or unconjugated as an aglycone [16]. Although quercetin metabolites have fewer antioxidant capabilities, there are some metabolic derivatives of quercetin which effectively remove reactive species of the body [16].

3.2. Quercetin’s Metabolic Pathways

Per os administration of quercetin is slightly absorbed in the stomach, followed by extensive intestinal absorption via the Na+-dependent cotransporter (SGLT1) and the first pass metabolism [17,18]. During metabolic phase II, quercetin is conjugated in the small intestine via glucuronidation, sulphuration, and methylation as it is confirmed by the presence of the corresponding metabolites in human small intestine microsomes [19]. Several enzymes are responsible for the catalysis of conjugation reactions such as sulfotransferases (SULTs), uridine-5′-diphosphate glucuronosyl transferases (UGTs), and catechol-O-methyl- transferases (COMTs). Namely, UGT-mediated glucuronidation is required for the production of the active metabolite quercetin-3-O-b-D-glucuronide, as well as of tamarixetin-7-glucuronide and isorhamnetin-3-glucuronide. Moreover, the sulphuration regulated by SULTs lead to the metabolite quercetin-3′-sulphate, while ring fission—to 3,4-dihydroxyphenylacetic acid [20]. Most of the metabolites in the bloodstream are glucurono–sulfo-conjugates of isorhamnetin and quercetin, while a minor part are quercetin glucuronides and methoxylated forms [21]. However, oral administration of quercetin in rats has shown that quercetin-3-O-b-D-glucuronide is the major bioactive metabolite in plasma [22]. Moreover, tamarixetin-7-glucuronide and isorhamnetin-3-glucuronide are accumulated in tissues, and they have been associated with anti-inflammatory and anti-atherosclerotic effects, respectively [23,24]. The half-life of quercetin metabolites shows a range of 11 to 28 h, which is considered high compared to other phytochemicals [18]. These metabolites are released into the blood and lymphatic circulation, as evidenced by the presence of quercetin glycosides in both the portal vein and lymph, while quercetin levels have been found below the detection limits or equal to 6.7% of the administered dose [17,25]. Subsequently, the metabolites reach the liver and enter hepatocytes via either passive diffusion or through organic anion transporters (OAT) and the organic anion transport protein (OATP) [26]. After hepatic uptake, they are further metabolized by phase II conjugating enzymes and pass into the circulation or are localized in biliary excretion [25]. Quercetin is excreted with feces and urine, while there are reports of lung elimination when the flavonoid is administered in high doses [18].
The limited absorption due to the low solubility of the compound in gastrointestinal fluids and the extensive metabolism into inactive metabolites that are excreted rapidly from the body contribute to quercetin’s low oral bioavailability. It is worth mentioning that the bioavailability has not yet been accurately determined due to the high variability of the reported pharmacokinetic parameters even after the consumption of the same dose and form of quercetin. This fact is likely attributed to the interindividual variability of β-glucosidase activity, as well as variations in the performance of metabolic enzymes [20].

3.3. Quercetin and Cardiovascular Prevention Based on Preclinical Studies

3.3.1. Hypertension

The mechanism of hypertension development has not been clearly understood. However, it is strongly associated with the vascular endothelial dysfunction [27]. Moreover, oxidative stress accompanied by reactive oxygen species (ROS) contribute to this dysfunction uncoupling endothelial nitric oxide synthase (eNOS) and reducing the nitric oxide (NO) bioavailability [28]. NO acts as an endogenous relaxation factor that regulates the vascular tone, as well as cardiac and vascular remodeling. The reduction of NO levels is related to hypoxia and cardiovascular diseases progression in patients with already existing vascular dysfunction, while its contribution to vasodilation of healthy subjects is ambiguous [29].
The protective effect of quercetin on endothelial dysfunction has been proposed to be related to its antihypertensive effect. This effect is assisted by its scavenging activity against ROS, leading to the reduction of endoplasmic stress [7]. An in vitro study has proven that 20 μM of quercetin can decrease intracellular ROS levels in endothelial cells of mesenteric arteries isolated from both hypertensive and normotensive animals [30]. Furthermore, ex vivo endothelial functional studies showed that quercetin can improve the vascular function via the activated protein kinase (AMPK) pathway inducing eNOS activation and, consequently, NO production [31] (Shen et al., 2012). In vitro studies by both Pereira et al. (2018) [32] and Lin et al. (2020) [30] support the involvement of quercetin-induced autophagy, which improves the quality of endothelial cells and NO production. Tumor studies also confirm the ability of quercetin to act as an inducer of autophagy [33,34,35]. Quercetin as a flavonoid compound of Ugni molinae Turcz. (murtilla) fruits has shown a vasodilatory and potentially hypotensive effect via calcium-dependent potassium channels [36]. Notably, quercetin may selectively stimulate vasorelaxant KCa1.1 channels and concomitantly inhibit Ca channels’ activity with potential blood pressure-lowering impact [37].
Furthermore, quercetin is correlated with the reduction of oxidative stress in aortas of 2K1C rats. However, its effect on systolic blood pressure (SBP) values is controversial in the case of 2K1C rats [32], while in SHRs, a dose-dependent decrease is observed. In particular, a high dose of quercetin (>7 mg/kg) is required to obtain a significant decrease (p < 0.05) in both systolic and diastolic blood pressure (DBP) [38,39] (Elbarbry et al., 2020; Duarte et al., 2001). Notably, SBP decrease was observed either in adult (12 weeks) or young hypertensive rats (5 weeks) [30,38]. In contrast, Carlstrom et al. (2007) [40] failed to show the suppressive effects of a quercetin-supplemented diet on arterial hypertension and its complications, such as cardiac hypertrophy, vascular dysfunction, vascular remodeling, in SHRs, but they attributed this discrepancy to the mode of administration.
In several in vitro and in vivo studies, quercetin is proposed to inhibit the angiotensin-converting enzyme (ACE) by binding a zinc molecule at the active site of the enzyme and via this mechanism slows down the conversion of angiotensin I to angiotensin II [41,42]. This effect is attributed to the chemical structure of flavonoids and mainly in the presence of the double bond and the ketone group at the C ring, as well as of the 3′,4′-catechol group in the B ring (Figure 1) [43].

3.3.2. Diabetes

Diabetes is a heterogeneous endocrine disorder characterized by persistent hyperglycemia due to inadequate insulin secretion or insulin resistance. Several in vitro and in vivo studies have attempted to prove the beneficial effect of flavonoids and quercetin specifically on the prevention and nutritional management of diabetes [44,45,46,47,48,49] (Table 1). Namely, studies on C2C12 and L6 skeletal muscle cell lines showed that quercetin and its most active glycosides (quercetin-3-O-glycosides) can increase the glucose uptake via a mechanism involving AMPK, stimulating the glucose transporter type 4 (GLUT4) translocation and its expression in skeletal muscles [47,50] (p < 0.05). Those findings are in line with those of Jiang et al. (2019) [51] where treatment of L6 myoblasts derived from rat skeletal muscles with quercetin and its glycoside isorhamnetin promoted glucose uptake. Furthermore, in vitro and in vivo experiments in the same cell line and in alloxan-induced type 2 diabetic mice, respectively, demonstrated that quercetin can reduce oxidative stress induced by ROS in various concentrations (1 μM, 10 μM, 100 μM). Moreover, quercetin may reduce malonaldehyde (MDA) levels, an index of lipid peroxidation (p ≤ 0.05) [45,49]. Notably, in the same study [33], 3-week-long treatment of mice with 20 mg/kg/day significantly lowered the fasting blood glucose levels (p ≤ 0.05), which was accompanied by the recovery of the activity of antioxidant enzymes (p ≤ 0.05).
In their systematic review, Shi et al. (2019) [52] summarized the antidiabetic effects of quercetin in diabetic animal models employed in several studies. Based on their analysis, quercetin significantly decreased blood glucose levels when administered intraperitoneally or orally at various doses ranging from 2.5 mg/kg/day to 200 mg/kg/day for a time period varying from 2 to 10 weeks.

3.3.3. Hyperlipidemia

Hyperlipidemia constitutes one of the major risk factors for the occurrence of CVDs. Quercetin is a fundamental flavonoid of the human diet capable of reversing the effects of hyperlipidemia. In rabbits with diet-induced hyperlipidemia, quercetin reduced the serum triglycerides and cholesterol levels in those receiving a high-fat diet for 12 weeks and, to a lesser extent, in those receiving a high-fat diet for 4 weeks with a carotid artery injury. Both animal models presented a reduction in atherosclerotic lesions (aortic atherosclerosis and injured carotid artery, respectively) due to the antioxidant properties and the anti-inflammatory activity of the flavonoid [53]. Furthermore, quercetin exerted a protective effect against cardiac diastolic dysfunction induced by a high-cholesterol diet in hyperglycemic rats. This effect is expressed via the impediment of cholesterol accumulation and ATP reduction preventing the alteration of PGC-1α, UCP2, and PPARγ receptors’ expression [54]. An in vitro study on human THP-1 derived macrophage cells showed that quercetin causes an outflow of cholesterol and decreases foam cells formation, which potentially halt the progression of atherosclerosis. This effect of quercetin is mediated via the upregulation of the ABCA1 cholesterol transporter and the PPARγ transcription factor [55]. The same findings were also reported by Cui et al. (2017) [56] after an 8-week-long treatment of apolipoprotein E-deficient atherosclerotic mice with quercetin. Recently, in vitro and in silico studies have proven that quercetin could prevent overexpression of the ICAM-1 and MCP-1 genes and impede cell migration in atherosclerotic plaques, reducing the risk of stroke [57]. In their studies, Liang et al. (2021) [58] underlined the importance of the development of novel delivery systems for quercetin solubility enhancement in both in vitro cell lines and an in vivo animal model.

3.3.4. Quercetin and Cardiac Protection: Cardiomyopathies, Cardiotoxicity, Myocardial Perfusion Injury, Myocardial Infarction

Cardiac Injury

The vasculoprotective effects of quercetin have involved several mechanisms and different signaling pathways, such as intracellular protein kinase cascades, the reduction of oxidative stress, the inhibition of cell migration towards atherosclerotic plaques, etc. Furthermore, quercetin has been characterized as a cardioprotective agent in different kinds of cardiac injury or drug-induced cardiotoxicity [59]. Quercetin has a capability to protect the myocardium from cardiac ischemia/reperfusion injury. In this context, it represses the inflammatory cascade and apoptosis, mainly via the PI3K/Akt pathway [60].
Quercetin protected cardiomyocytes from apoptosis, limited myocardial injury from mechanical trauma, and improved cardiac dysfunction via intracellular Ca2+ overproduction and the elimination of ROS [61]. However, the accumulated data are scarce and controversial [62]. Quercetin seems to inhibit Ca2+ influx via L-type Ca2+ channels, leading to myocardium protection against ischemia [63]. Thus, regulation of calcium channels can be a potential cardioprotective mechanism of quercetin which requires further investigation. In parallel, the decrease in ROS production after the treatment with quercetin has also been reported in the model of a 4-hydroxynonenal-induced toxicity H9c2 cardiac tissue-derived cell line. This reduction occurred along with the attenuation of apoptotic stress markers (pMAPKAPK-2, p-SAPK/JNK, p-Hsp27, p-c-JUN, and cl-CASP3), leading to the improvement of cell viability after 24 h quercetin treatment in concentrations ranging from 0.1 to 10 μM [64]. Moreover, H9c2 cells exposed to hypoxia/reoxygenation (H/R) were rescued by quercetin treatment (10–16 μM) [65], while 24 h pretreatment with PLGA-encapsulated quercetin nanoparticles was also preventive [66]. In human cardiomyocytes cultured in H/R medium quercetin can improve the mitophagy mechanism and reduce endoplasmic stress via stimulation of SIRT1/TMBIM6 pathways. Both pathways can increase autophagy and mitophagy events and reverse ROS overproduction induced by hypoxia. Thus, cellular homeostasis can be improved, as well as the vulnerability of cardiomyocytes against H/R events [67]. Similarly, isolated heart models for the ischemia/reperfusion (I/R) injury have shown that heart recovery is improved via the HMGB1 (high mobility group box 1) pathway after 30 min of ischemia followed by 30 min of reperfusion [68]. Moreover, quercetin treatment yielded reduced expression of both TNF-alpha (TNF-α) and interleukin 10 (IL-10) along with their serum levels in rats after acute myocardial ischemia and reperfusion injury [69]. The protective effect of quercetin against adverse remodeling of myocardial infarction has been confirmed in infarction-affected rats treated with 50 mg/kg/orally for 30 days. That effect was attributed to the attenuation of TGF-b1/Smad3 signaling which is associated with cardiac fibrosis, hypertrophy, and cardiac dysfunction [70]. In addition, the treatment of rats with quercetin (10 mg/kg) after a myocardial I/R injury stimulated the phosphoinositide 3-kinases (PI3Ks) pathway, which is responsible for the favorable regulation of cell proliferation, as well as of cellular survival and apoptosis [71].

Drug-Induced Cardiotoxicity

Drug-induced cardiotoxicity is one of the most commonly reported adverse effects related with several medicines of different categories. The underlying mechanisms mostly involve drug–protein interactions, which can lead mainly to myocardial infarction or ischemia, heart failure, valvular heart disease, and QT prolongation associated, in several cases, with sudden cardiac death [72]. ROS formation at levels higher than those that can be counterbalanced by cellular detoxification mechanisms causes progressive mitochondrial dysfunction leading to cardiomyocyte apoptosis. Quercetin can scavenge free radicals thanks to the catechol groups in the B ring, the 2,3-double bond in conjugation with the 4-oxo group in the C ring, and the OH groups located at positions 3 and 5 in the heterocyclic ring [73]. ROS deactivation by quercetin takes place in three stages, including formation of a superoxide ion, generation of hydroxyl (or cryptohydroxyl) radicals, and formation of lipid peroxy radicals [74]. Several drugs, such as anthracyclines or protein tyrosine kinase inhibitors, express their cardiotoxicity inducing ROS production and accumulation in cells. Doxorubicin is a chemotherapy drug classified as an anthracycline and employed in different types of cancer [72]. Quercetin given orally to rats before their treatment with daunorubicin showed significant cardiac protection. Moreover, preadministration of the flavonoid combined with angiotensin II receptor blocker losartan can increase the ability of the drug to impede lipid peroxidation and, thus, progression of atherosclerotic plaques. In doxycycline-treated rats, coadministration of these two agents reduced the signs of cardiomyopathy and the induced inflammation [75]. Nazmi et al. (2016) [76] proved the antioxidative and renoprotective activity of quercetin against doxorubicin-induced cardiotoxicity and nephrotoxicity in Wistar albino rats. After the 7-day-long per os treatment with 2 mg/kg/day quercetin, the blood glutathione levels restored, while the levels of lipid peroxidation products were reduced. Most importantly, the quercetin-treated rats had no signs of vascular myopathy and featured normal appearance of their myocardium [76]. However, the effect of quercetin on the less cardiotoxic epirubicin was found to be negative for cell viability. In particular, quercetin decreased the elimination of the drug from cardiomyocytes, impeding its transformation to the less toxic secondary alcohol metabolite epirubicinol [77].

Metabolic Syndrome (MS)

Metabolic syndrome (MS) is a medical term describing the accumulation of several risk factors increasing the incidence of cardiovascular pathology, such as coronary heart disease, diabetes mellitus, and stroke. The cluster of these risk factors includes hypertension, hyperlipidemia, obesity, and diabetes mellitus [78]. The correlation between the incidence of these factors and quercetin consumption has been extensively investigated in in vitro and preclinical studies performed using models of metabolic syndrome [32,44,79,80] (Table 1). Recently, Hosseini et al. (2021) [5] accumulated the animal studies implicating the beneficial effects of quercetin on metabolic syndrome factors, including T2DM, hypertension, dyslipidemia, and obesity, in their review about quercetin and metabolic syndrome. The given doses in those studies ranged from 5 mg/kg/day to 240 mg/kg/day administered either via intraperitoneal injection or orally (gavage method or via drinking water). The administration period lasted from 10 to 90 days. The studies included either rats or mice, but streptozotocin-induced diabetic rats were the most common animal model used. From the mechanistic perspective, quercetin-induced insulin secretion and insulin sensitization were the principal pathways for the protective effect of quercetin against T2DM [81,82]. Furthermore, quercetin exerted a hepatoprotective action in animal models [83], while the anti-obesity effect of quercetin was associated with its antioxidant and anti-inflammatory activity [84].
The decrease in triglycerides, total cholesterol, and low-density lipoprotein cholesterol has been reported after quercetin administration in several studies [85,86,87]. However, its effect on HDL levels is considered controversial. It has been found substantially increased in apolipoprotein E-deficient mice fed a high-fat diet, whereas a nonsignificant increase was noted in dyslipidemic mice treated with 500 mg/L of quercetin in drinking water [56,87,88]. In the study of Gao et al. (2021) [89], the protective effect of this flavonoid on a nonalcoholic fatty liver disease (NAFLD) rat model was also demonstrated. Taken together, quercetin administration seems to exert a beneficial impact on almost all components of metabolic syndrome simultaneously, presenting a multifactorial cardioprotective agent.

Atherosclerosis

Cardiovascular diseases and especially atherosclerosis constitute a complex process influenced by both genetic and environmental factors. There is evidence that quercetin suppresses the progression of atherosclerosis in animal studies, and several anti-atherosclerotic mechanisms have been proposed [53,90]. Quercetin supplementation, alone or combined with exercise, was found to prevent atherosclerotic plaque formation in C57Bl6 LDLr-deficient mice fed an atherogenic diet. This effect is expressed via the stimulation of cholesterol reverse transport genes promoting lipid reduction [91]. A few studies showed atheromatous plaques regression through the induction of autophagy and the delay of senescence, possibly via the activation of the ERK signaling pathway [92,93,94]. A number of in vivo and in vitro studies indicated suppression of the expression of many inflammatory factors, including IL-6, TNF-α, PCSK9, MCP-1, CD36, and others [88,91,95,96,97], as well as upregulation of ABCA1 and LXR-α when quercetin is delivered to mice [88,97]. In the same anti-inflammatory context, quercetin mediates maturation of dendritic cells, inhibition of VCAM-1 and ICAM-1 [98,99,100,101], and inhibition of the galectin–3NLRP3 signaling pathway [102]. Quercetin also seems to confer an atheroprotective effect via the activation of the caspase-3 and NFK-β factors [103], the upregulation of para-oxonase 1 gene expression [104,105], and the suppression of the ER stress-CHOP pathway with a concomitant increase in Bcl-2 gene expression via inhibiting ATF6 and IRE1 activation [106].
Based on mice studies, quercetin has the potential to stabilize atherosclerotic plaques through the inhibition of elastin degradation, macrophage infiltration, MMP-9 and VCfAM-1 expression [106]. Moreover, it can reduce the lipid content, the levels of sICAM-1, IL-6, and VCAM-1 and increase Sirt1 within atheromatous plaques in the aorta of the animal model [107]. Sirt1 is a key molecule for basal-level autophagy [108]. In vitro, quercetin blocks the ERK signaling pathway, which in turn inhibits MMP-1 and MMP-9, two important mediators of plaques’ vulnerability [109,110]. From the aforementioned studies, it can be suggested that quercetin has a promising effect on atherosclerotic lesions, but this remains to be confirmed in humans.
Lipid decrease and the suppressive effect of quercetin administration on fat accumulation and the increase in blood lipids has been proposed as an additional antiatherogenic mechanism [111,112,113]. Consequently, lower lipid deposition in macrophages is noted, along with the promotion of cholesterol efflux and blockage of foam cell formation [107]. Furthermore, downregulation of the PCSK9 protein and platelet glycoprotein 4 (CD36) as well as increased expression of the PPARγ and LXRα receptors and the ABCA1 transports in apoE-/- mouse aortas treated with 12.5 mg/kg/day of quercetin were found to be associated with the antiatherogenic effect of the flavonoid. High levels of PCSK9 force the expression of CD36, which is followed by a cascade of events that contribute to the progression of atherosclerotic plaques [114].

Obesity

Obesity is associated not only with adipose tissue accumulation, but also with its dysfunction and the occurrence of CVDs [115]. Obesity is an inflammatory process where overexpression of inflammatory cytokines such as TNF-a, IL-6, and MCP-1 is observed. This proinflammatory state leads to endothelial and microvascular dysfunction due to a significant increase in ROS [116]. Quercetin is a CYP2E1 inhibitor capable of neutralizing free radicals and downregulating the inflammatory process, normalizing the main biochemical parameters of blood serum such as the cholesterol, glucose, and creatinine levels [117]. Furthermore, it acts as a blocker of adipogenesis, impeding the mitogen-activated protein kinase (MAPK) signal pathway [118]. Increased levels of the MAPK were found to be related with an increase in body weight and a reduction in energy expenditure [119]. Treatment of 3T3-L1 fibroblast cells with quercetin showed that adipogenesis is downregulated due to the impediment of extracellular signal-regulated kinases’ (ERK) and c-Jun N-terminal kinases’ (JNK) phosphorylation and the activation of adipocyte apoptosis [118]. In their work on C57B1/6 mice, Jung et al. (2012) [111] proved the anti-obesity effect of quercetin on high-fat diet-induced obesity, and this effect was attributed to its involvement in lipogenesis and lipid metabolism.

3.4. Quercetin and Cardiovascular Prevention Based on Clinical Studies

Cardiovascular diseases (CVD) are the main cause of morbidity and mortality worldwide, with atherosclerosis being considered the underlying mechanism [122]. Traditional cardiovascular risk factors including hypertension, dyslipidemia, diabetes, and obesity often coexist and share common pathophysiologic mechanisms [123]. Certain antihypertensive, antithrombotic, anti-dyslipidemic, and anti-atherosclerotic properties of quercetin have been proposed for CVD protection [124]. Clinical studies and meta-analyses summarized in Table 2 and Table 3 evaluated the impact of quercetin on the cardiovascular risk factors described below.

3.4.1. Metabolic Syndrome (MS)

A recent systematic review [125] of seventeen RCTs evaluated the effect of daily supplementation with quercetin in patients with MS, including hypertension, T2DM, PCOS, and obesity, as well as in healthy individuals. The dosage of quercetin ranged from 30 to 1000 mg/day and the duration of intervention varied between 2 and 12 weeks along the studies. The cumulative data demonstrated significantly reduced both systolic and diastolic blood pressure. Although the previous systematic review failed to report any significant change in lipid or glucose profiles after the administration of quercetin, subgroup analysis showed significant amelioration of HDL and triglycerides concentrations in trials where the duration of quercetin administration exceeded 8 weeks. Another meta-analysis [126] of eighteen RCTs assessed the effect of flavonol supplementation for 2 to 12 weeks on MS parameters, including plasma lipids, glucose, and blood pressure. Significant decrease was observed in blood pressure, fasting glucose, LDL, and total cholesterol, as well as a significant increase in HDL cholesterol levels. However, those results should be interpreted with caution because of the studied population. Most of the selected studies (n = 13 studies) enrolled healthy subjects, while the rest recruited patients with MS (n = 4 studies). Notably, only one study included patients with hypertension. Among the selected RCTs, nine used pure quercetin supplementation, while the rest (n = 9 studies) investigated interventions of either pure flavonol supplements or enriched mixtures of flavonols. Most recently, an observational prospective study of 6417 participants [127] showed that a higher dietary intake of flavonoids (combination of quercetin with magnesium) was associated with a lower incidence of MS. Similarly, a recent RCT [128] confirmed the favorable effects of quercetin administration on such MS parameters as body weight, cholesterol metabolism, fasting plasma insulin, and blood pressure. In the context of MS, quercetin may additionally protect against the associated liver disease [129]. After all, the up-to-date data are conflicting, and there is no robust evidence that supplementary quercetin treatment effectively modifies MS parameters.

3.4.2. Obesity

A recent meta-analysis [125] of nine RCTs evaluated the effects of quercetin administration on body weight. Overall, supplementation with quercetin had no significant impact on body weight, BMI, waist circumference, or waist-to-hip ratio among the participants with obesity, hypertension, and PCOS. Pfeuffer et al. (2011) [130] introduced the influence of genetic polymorphism on quercetin’s efficacy since they showed significantly decreased BMI, body weight, and waist circumference in APOE3/3 but not in APOE4 patients. The association between quercetin and APOE isoforms was observed in another study [131] of overweight/obese subjects with MS. Moreover, the APOE3 group, but not the APOE4 group, responded to quercetin therapy with a decrease in systolic blood pressure. On the other hand, significant reductions in plasma-oxidized LDL and TNF-a were observed in both groups.

3.4.3. Hypertension

There are controversial results about the impact of quercetin supplementation on blood pressure. A systematic review [132] of seven RCTs demonstrated significant reductions in both systolic and diastolic blood pressure after quercetin supplementation in patients with hypertension [133]. It seems like the hypotensive effects of quercetin are more evident with a higher dosage (>500 mg/day) and in subjects with certain characteristics, including MS and smoking [125,132,133]. Another recent meta-analysis [134] of eight RCTs showed a significant decrease in systolic blood pressure only and not in diastolic blood pressure following supplementation with quercetin in patients with MS. In contrast, a recent large (n = 15,662 participants) prospective cohort study [135] failed to show any significant association between quercetin dietary intake as a measure of primary prevention and the incidence of hypertension [135]. In line, other eight clinical studies [9,131,136,137,138,139,140,141] conducted between 1998 and 2019 failed to find any significant effect of quercetin supplementation on blood pressure in healthy and hypertensive subjects, while other cardiovascular risk factors (lipid levels, inflammatory mediators, or thrombogenic risk factors) remained unaffected as well. Presumably, the validity of the latter studies is limited since they enrolled a small number of participants, had a short duration of intervention, and the majority of the participants were healthy individuals or patients with prehypertension or, at maximum, stage I hypertension.

3.4.4. Dyslipidemia

In a recent meta-analysis by Sahebkar et al. (2017) [142], quercetin supplementation significantly reduced plasma TG at doses above 50 mg/day. However, available evidence from the five RCTs included in that meta-analysis showed a nonsignificant relationship between quercetin supplementation and the rest of the lipid parameters (total cholesterol, LDL, HDL). Another systematic review of nine RCTs, by Guo et al. (2019) [143], failed to find any significant change in plasma lipids after quercetin treatment unless higher doses of quercetin (≥250 mg/d) were administered to overweight and obese individuals which could reduce LDL cholesterol. Hence, the pure effect of quercetin on lipids remains controversial.
Other investigators have supported the favorable effects of quercetin in dyslipidemic patients when used as an adjunctive therapy or in patients with MS. In particular, Mazza et al. (2021) [144] showed improved TG and LDL plasma levels in dyslipidemic and hypertensive patients with statin intolerance when receiving quercetin in combination with ezetimibe. A recent meta-analysis of sixteen RCTs (Tabrizi et al., 2020) [145] indicated that quercetin supplementation significantly decreased plasma levels of total cholesterol, LDL, and CRP in patients with MS. However, it did not affect the levels of TG, HDL, IL-6, and TNF-α. Furthermore, two more studies [146,147] reported that consumption of quercetin-rich onion juice or powder significantly reduced plasma lipids and prevented obesity. It is undoubtable that these beneficial effects differ significantly depending on the formulation and dosage of quercetin, as well as the duration of administration.

3.4.5. Hyperglycemia

In a recent systematic review (Ostadmohammadi et al., 2019) [148] of nine RCTs, quercetin supplementation did not affect fasting glucose or hemoglobin A1 levels or insulin resistance in patients with MS including hypertension, T2DM, PCOS, or obesity. The duration of intervention varied between 4 and 12 weeks and the dosage of quercetin ranged from 100 to 1000 mg/day. However, subgroup analysis revealed that longer duration (≥8 weeks) and higher dosages of quercetin (≥500 mg/day) were significantly correlated with reduced fasting plasma glucose. Similarly, higher doses of quercetin (≥500 mg/day) effectively decreased insulin levels only in studies recruiting younger subjects (aged <45 years old). A large prospective study by Song et al. (2005) of 38,018 healthy women failed to demonstrate a preventive effect of dietary flavonoids, including quercetin, on T2DM [149]. In contrast, a very small noncontrolled pilot study of 15 T2DM patients who for 3 months received Eugenia punicifolia, mainly containing quercetin, showed a significant decrease in hemoglobin A1 and basal insulin [150]. Further large-scale trials are needed to investigate the impact of quercetin on T2DM management and prevention.
The inconsistent data about the impact of quercetin on cardiometabolic risk factors highlights the need for further human studies.

3.4.6. Quercetin and CVDs

Recent evidence has emerged that changes in gut microbiota are related to the occurrence of chronic low-grade inflammation and, thus, to the development of CVDs, including coronary artery disease, heart failure, and hypertension. Quercetin’s dose-dependent effects on the gut microbiota have an impact on CVD progression [151,152]. Quercetin-like natural plant compounds usually affect multiple targets to prevent cardiovascular events. Thus, the activity of quercetin-like plant compounds as cofactors of COX-2 is just one mechanism by which they decrease the risk of cardiovascular diseases, and more research is needed to confirm this hypothesis [153]. Endothelial dysfunction is an important step seen early in the progression of atherosclerosis and hypertension [154].
A randomized controlled trial investigated the effect of quercetin (120 mg per day for 2 months) on 30 patients with coronary artery disease and showed a positive effect on chronic systemic inflammation indicated by reduction in IL-1β and the IkBa gene in blood mononuclear cells [155]. In a cohort of 5133 healthy adults, lower dietary intake of flavonoids, including quercetin, was correlated with higher risks of coronary artery disease [156]. Quercetin administration is generally well-tolerated, with some minor side effects such as mild headache, nausea, and tingling of the extremities observed in long-term supplementation at 1000 mg/day. Nephrotoxicity has been reported with high IV doses in cancer patients [154].
Table 2. Published meta-analyses of randomized clinical trials investigating the impact of quercetin on classical cardiovascular risk factors.
Table 2. Published meta-analyses of randomized clinical trials investigating the impact of quercetin on classical cardiovascular risk factors.
ReferenceStudies and CohortsStudy DesignOutcomes
Huang et al., 2019 [130]Nine RCTs, 525 pts; obese, HTN, PCOS, healthy individualsQuercetin daily; dose: 100–1000 mg; duration: 2–12 weeks↔ body weight, BMI, waist circumference, waist-to-hip ratio
Huang et al., 2020 [125]17 RCTs, 896 pts; MS, T2DM, PCOS, obesityQuercetin daily; dose: 30–1000 mg; duration: 2–12 weeks↓ SBP and DBP
↔ lipid and glucose profile Subgroup analysis (> 8 weeks after intervention): ↑ HDL, ↓ TG
Menezes et al., 2017 [126]18 RCTs, 530 pts; healthy individuals, MS, HTNFlavonol daily; dose: 16–1200 mg; duration: 2–12 weeks↓ BP, FPG, LDL, Tchol, triacylglycerol
↑ HDL
Subgroup analysis: favorable in dyslipidemic pts and pts of Asian origin
Ostadmohammadi et al., 2019 [150]Nine RCTs, 781 pts;
MS
Quercetin daily; dose: 150–1000 mg; duration 4–12 weeks↔ FPG, HbA1c, insulin resistance
Sahebkar et al., 2017 [144]Five RCTs, 442 pts; central obesity, hypertriglyceridemia, T2DM, HTNQuercetin daily; dose: 30–730 mg; duration: 2–10 weeks↓ TG (at doses > 400 mg/day)
↔ Tchol, LDL, HDL
Serban et al., 2016 [133]Seven RCTs, 587 pts; HTNQuercetin daily; dose: 100–1000 mg; duration: 4–10 weeks↓ SBP and DBP
Tabrizi et al., 2020 [147]16 RCTs, 1575 pts; MSQuercetin daily; dose: 3.12–3000 mg; duration: 2 h postprandially for 12 weeks↓ Tchol, LDL, CRP
↔ TG, HDL, IL-6, TNF-α
BMI, body mass index; CRP, C-reactive protein; DBP, diastolic blood pressure; FPG, fasting plasma glucose; HDL, high-density lipoprotein; HTN, hypertension; IL-6, interleukin 6; LDL, low-density lipoprotein; MS, metabolic syndrome; PCOS, polycystic ovarian syndrome; pts, participants; RCTs, randomized controlled trials; SBP, systolic blood pressure; T2DM, type 2 diabetes mellitus; Tchol, total cholesterol; TG, triglycerides; TNF-α, tumor necrosis factor-α; wks, weeks; ↓, decrease; ↑, increase; ↔, non-significant change.
Table 3. Published clinical studies investigating the impact of quercetin on the classical cardiovascular risk factors not included in meta-analyses.
Table 3. Published clinical studies investigating the impact of quercetin on the classical cardiovascular risk factors not included in meta-analyses.
ReferenceStudy Cohort/and ConditionStudy DesignOutcomes
Biesinger et al., 2016 [138]18 pts; MSCrossover RCT; quercetin dehydrate daily vs. placebo; dose: 25 mg; duration: 28 days;
washout period: 2 weeks
↔ HTN incidence
Brull et al., 2015 [141]68 pts; MS, overweight or obese, prehypertension and stage I HTNRCT, crossover study; quercetin from onion skin extract daily vs. placebo; dose: 162 mg; duration: 6 wks; washout period: 6 weeks↔ HTN incidence, endothelin-1, ADMA, ACE activity, CRP, endothelin-1, sE-selectin, sVCAM-1, sICAM-1, RHI, AI
Burak et al., 2019 [143]67 healthy nonobese volunteers aged 19–35 yearsRandomized double-blind placebo-controlled crossover study; 3.6 g/day ALA plus 190 mg/day quercetin vs. placebo; duration: 8 wks; washout period: 8 weeks↔ office systolic BP, mean 24-hour ambulatory BP, mean ambulatory BP, HDL, apolipoprotein A1, glucose, uric acid, oxidized LDL, CRP
Chekalina et al., 2018 [155]85 pts; CADRCT; intervention group (30 pts): quercetin, 120 mg daily; control group (55 pts): placebo; duration: 2 mo↓ IL-1b, TNF-α, expression of the IkBa gene in blood mononuclear cells
Conquer et al., 1998 [9]27 pts; healthyRCT; intervention group (13 pts): quercetin, 1000 mg daily↔ HTN incidence, Tchol, LDL, HDL, TG, platelet aggregation, platelet thromboxane B2 production, resting heart rate
Edwards et al., 2007 [139]41 pts; prehypertension and stage I HTNCrossover RCT; quercetin aglycone twice/day vs. placebo; dose: 365 mg; duration: 4 weeks; washout period: 2 weeks↔ HTN incidence, weight, BMI, indices of oxidative stress, TG, LDL, VLDL, HDL, Tchol, FPG
Prehypertensive subjects: ↓ SBP, DBP, MAP
Egert et al., 2009 [137]93 pts; overweight or obese pts, MSDouble-blind placebo-controlled crossover study; quercetin daily vs. placebo; dose: 150 mg; duration: 6 weeks; washout period: 5 weeks↓ HDL, oxidized LDL
↔ incidence of HTN, Tchol, TG, LDL:HDL cholesterol, TG:HDL cholesterol ratios, TNF-α, CRP
Egert et al., 2010 [132]93 overweight/obese pts; MS, APOE3/3, 3/4, 4/4, 2/3, 2/4Double-blind placebo-controlled crossover study; quercetin daily vs. placebo; dose: 150 mg; duration: 6 weeks; washout period: 5 weeks↓ oxidized LDL, TNF-α
↔ CRP, body weight, waist circumference, fat mass, fat-free mass
APOE3/3 group: ↓ SBP
APOE4 group: ↓ HDL
Jin et al., 2021 [127]6417 subjectsObservational prospective cohort study; dietary intake flavonoids (quercetin + magnesium)↓ incidence of MS
Knekt et al., 1996 [156]5133 healthy adults aged 30–69 yearsCohort study; dietary intake of flavonoids↓ incidence of CAD
Larson et al., 2012 [142]17 men; normotensive and stage I HTNDouble-blind placebo-controlled crossover study; quercetin aglycone (dose: 1095 mg) vs. placebo;
duration: acute single dose
Normotensive:
↔ BP, ACE activity
Stage I HTN: ↓ SBP, DBP, mean BP
↔ ACE activity, NO metabolites, ET-1, ET-1:NO ratio metabolites
Lu et al., 2015 [148]24 healthy subjects; mild hypercholesterolemiaPilot RCT; intervention group (12 pts): 100 ml quercetin-rich onion juice daily; control group (12 pts): placebo; duration: 8 weeks↓ waist circumference, Tchol, LDL
Mazza et al., 2021 [146]96 pts; dyslipidemia, HTN, statin intoleranceRCT; intervention group (48 pts): ezetimibe/quercetin, 10/100 mg daily; control group (48 pts): ezetimibe monotherapy; duration: 3 mo↓ TG, LDL
Nishimura et al., 2020 [149]70 healthy subjectsRCT; intervention group (35 pts): 9 g quercetin-rich onion powder daily; control group (35 pts): placebo; duration: 12 weeks↓ ALT
↔ visceral fat area, BP
Pfeuffer et al., 2013 [131]49 healthy men; APOE3/3, 3/4, 4/4Double-blind crossover study; quercetin daily vs. placebo; dose: 150 mg; washout period: 3 weeks↔ endothelial function
APOE3/3 group: ↓ BMI, body weight, waist circumference
Sales et al., 2014 [152]15 pts; T2DMPilot study; intervention: capsules containing 200 mg dried leaves of E. punicifolia; duration: 3 mo↓ HbA1c, basal insulin, thyroid-stimulating
hormone, CRP, SBP, DBP
Shatylo et al., 2021 [128]110 pts aged >60 years; MSRCT; intervention group (55 pts): 240 mg quercetin daily; control group (55 pts): placebo; duration: 3 mo↓ SBP and DBP, body weight, BMI, Tchol, LDL, insulin, 2-hour glucose level
↔ HDL, TG, oxidative stress (CAT, SOD, AGEs)
Song et al., 2005 [151]38,018 healthy women aged >45 yearsCross-sectional study; dietary intake of flavonoids↔ incidence of T2DM
Yao et al., 2021 [136]15,662 subjectsProspective cohort study; dietary intake of quercetin daily↔ HTN incidence
Zahedi et al., 2013 [140]62 women aged 35–55 years; T2DMRCT; intervention group (34 pts): quercetin, 500 mg daily; control group (28 pts): placebo; duration: 10 wks↓ SBP, HDL
↔ HTN incidence, DBP, Tchol, LDL, TG, ratio of TG/HDL and LDL/HDL, TNF-α, IL-6, hs-CRP
ACE, angiotensin-converting enzyme; ADMA, asymmetric dimethylarginine; AGEs, advanced glycoxidation end products; AI, augmentation index; ALT, alanine transaminase; APOE, apolipoprotein E; BMI, body mass index; CAD, coronary artery disease; CRP, C-reactive protein; DBP, diastolic blood pressure; FPG, fasting plasma glucose; HDL, high-density lipoprotein; HTN, hypertension; IL, interleukin; LDL, low-density lipoprotein; mo, months; MS, metabolic syndrome; PCOS, polycystic ovarian syndrome; RCTs, randomized controlled trials; RHI, reactive hyperemia index; SBP, systolic blood pressure; sICAM-1, soluble intercellular adhesion molecule 1; sVCAM-1, soluble vascular cell adhesion molecule 1; T2DM, type 2 diabetes mellitus; Tchol, total cholesterol; TNF-α, tumor necrosis factor-α; TG, triglycerides; wks, weeks; ↓, decrease; ↑, increase; ↔, non-significant change.

4. Study Limitations

The published data are quite promising; however, there are several drawbacks before considering the net results of the effects of quercetin on cardiovascular risk factors and diseases. The mechanistic explanations are derived from mostly in vitro rather than in vivo studies. Despite the growing number of studies, the underlying mechanisms of quercetin action remain unclear or somehow speculative and require further investigation using appropriate animal models. Most of the clinical studies summarized in Table 3 are of small sample size, while quercetin was administered as a plant extract or within the dietary intake of flavonoids as a food supplement. Furthermore, the dose administered varied from 3.12 mg to 3000 mg daily, while the clinical studies also differed with respect to study duration and patients’ characteristics. It should also be mentioned that literature lacks pharmacokinetic/pharmacodynamic and dose linearity studies of quercetin that are essential in the optimum design of clinical trials before any clinical application of quercetin and significantly compromises the validity and reproducibility of the clinical studies.

5. Conclusions

Flavonol quercetin is the main pharmacologically active ingredient of many fruits, vegetables, tea, and red wine with antioxidant, anti-inflammatory, hypotensive, glucose- and lipid-lowering activity. A part of those observations is also supported by results of the existing clinical studies that have shown an improvement in glycemic and lipid profile. However, the inconsistent data about the impact of quercetin on cardiometabolic risk factors such as MS, obesity, and hypertension point out the need for further human studies. Accordingly, additional properly designed clinical trials for specific cardiac conditions using standard dosages based on the previously performed pharmacokinetic/pharmacodynamic studies are required to test its value as a food supplement to conventional drug therapies.

Author Contributions

P.P., N.V. and E.K. contributed equally; P.P., N.V. and E.K.: literature search; P.P., N.V., E.K., and I.K.: writing—original draft preparation; I.K., N.P.K. and G.V.: writing—review and editing; I.K., N.P.K. and G.V.: conceptualization; N.P.K.: supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable.

Acknowledgments

We thank University of Cyprus for supporting the publication of the present paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, J.; Hao, K.; Yu, F.; Shen, L.; Wang, F.; Yang, J.; Su, C. Field application of nanoliposomes delivered quercetin by inhibiting specific hsp70 gene expression against plant virus disease. J. Nanobiotechnol. 2022, 20, 1–16. [Google Scholar] [CrossRef] [PubMed]
  2. Alizadeh, S.R.; Ebrahimzadeh, M.A. Quercetin derivatives: Drug design, development, and biological activities, a review. Eur. J. Med. Chem. 2022, 229, 114068. [Google Scholar] [CrossRef] [PubMed]
  3. Khan, J.; Deb, P.K.; Priya, S.; Medina, K.D.; Devi, R.; Walode, S.G.; Rudrapal, M. Dietary Flavonoids: Cardioprotective Potential with Antioxidant Effects and Their Pharmacokinetic, Toxicological and Therapeutic Concerns. Molecules 2021, 26, 4021. [Google Scholar] [CrossRef] [PubMed]
  4. Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 162750. [Google Scholar] [CrossRef]
  5. Hosseini, A.; Razavi, B.M.; Banach, M.; Hosseinzadeh, H. Quercetin and metabolic syndrome: A review. Phytother. Res. 2021, 35, 5352–5364. [Google Scholar] [CrossRef]
  6. Rusznyák, S.; Szent-Györgyi, A. Vitamin P: Flavonols as Vitamins. Nature 1936, 138, 27. [Google Scholar] [CrossRef]
  7. Xu, D.; Hu, M.J.; Wang, Y.Q.; Cui, Y.L. Antioxidant Activities of Quercetin and Its Complexes for Medicinal Application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef]
  8. Chen, K.T.J.; Anantha, M.; Leung, A.W.Y.; Kulkarni, J.A.; Militao, G.G.C.; Wehbe, M.; Sutherland, B.; Cullis, P.R.; Bally, M.B. Characterization of a liposomal copper (II)-quercetin formulation suitable for parenteral use. Drug Deliv. Transl. Res. 2020, 10, 202–215. [Google Scholar] [CrossRef]
  9. Conquer, J.A.; Maiani, G.; Azzini, E.; Raguzzini, A.; Holub, B.J. Supplementation with quercetin markedly increases plasma quercetin concentration without effect on selected risk factors for heart disease in healthy subjects. J. Nutr. 1998, 128, 593–597. [Google Scholar] [CrossRef]
  10. Janssen, K.; Mensink, R.P.; Cox, F.J.; Harryvan, J.L.; Hovenier, R.; Hollman, P.C.; Katan, M.B. Effects of the flavonoids quercetin and apigenin on hemostasis in healthy volunteers: Results from an in vitro and a dietary supplement study. Am. J. Clin. Nutr. 1998, 67, 255–262. [Google Scholar] [CrossRef]
  11. Nam, J.S.; Sharma, A.R.; Nguyen, L.T.; Chakraborty, C.; Sharma, G.; Lee, S.S. Application of Bioactive Quercetin in Oncotherapy: From Nutrition to Nanomedicine. Molecules 2016, 21, 108. [Google Scholar] [CrossRef] [PubMed]
  12. Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 29, e47. [Google Scholar] [CrossRef] [PubMed]
  13. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef] [PubMed]
  14. Deng, Q.; Li, X.X.; Fang, Y.; Chen, X.; Xue, J. Therapeutic Potential of Quercetin as an Antiatherosclerotic Agent in Atherosclerotic Cardiovascular Disease: A Review. Evid Based Complement Altern. Med. 2020, 5926381. [Google Scholar] [CrossRef]
  15. Hai, Y.; Zhang, Y.; Liang, Y.; Ma, X.; Qi, X.; Xiao, J.; Xue, W.; Luo, Y.; Yue, T. Advance on the absorption, metabolism, and efficacy exertion of quercetin and its important derivatives. Food Front. 2020, 1, 420–434. [Google Scholar] [CrossRef]
  16. Khan, F.; Niaz, K.; Maqbool, F.; Ismail Hassan, F.; Abdollahi, M.; Nagulapalli Venkata, K.C.; Nabavi, S.M.; Bishayee, A. Molecular Targets Underlying the Anticancer Effects of Quercetin: An Update. Nutrients 2016, 8, 529. [Google Scholar] [CrossRef]
  17. Chen, X.; Yin, O.Q.P.; Zuo, Z.; Chow, M.S.S. Pharmacokinetics and Modeling of Quercetin and Metabolites. Pharm. Res. 2005, 22, 892–901. [Google Scholar] [CrossRef]
  18. Ulusoy, H.G.; Sanlier, N. A minireview of quercetin: From its metabolism to possible mechanisms of its biological activities. Crit. Rev. Food Sci. Nutr. 2020, 60, 3290–3303. [Google Scholar] [CrossRef]
  19. Van der Woude, H.; Boersma, M.G.; Vervoort, J.; Rietjens, I.M. Identification of 14 quercetin phase II mono- and mixed conjugates and their formation by rat and human phase II in vitro model systems. Chem. Res. Toxicol. 2004, 17, 1520–1530. [Google Scholar] [CrossRef]
  20. Guo, Y.; Bruno, R.S. Endogenous and exogenous mediators of quercetin bioavailability. J. Nutr. Biochem. 2015, 26, 201–210. [Google Scholar] [CrossRef]
  21. Morand, C.; Crespy, V.; Manach, C.; Besson, C.; Demigné, C.; Rémésy, C. Plasma metabolites of quercetin and their antioxidant properties. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 1998, 275, 212–219. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, L.L.; Xiao, N.; Li, X.W.; Fan, Y.; Alolga, R.N.; Sun, X.Y.; Wang, S.L.; Li, P.; Qi, L.W. Pharmacokinetic comparison between quercetin and quercetin 3-O-β-glucuronide in rats by UHPLC-MS/MS. Sci. Rep. 2016, 6, 35460. [Google Scholar] [CrossRef] [PubMed]
  23. Park, H.J.; Lee, S.J.; Cho, J.; Gharbi, A.; Han, H.D.; Kang, T.H.; Lee, Y.; Park, W.-S.; Jung, I.-D.; Park, Y.-M. Tamarixetin Exhibits Anti-inflammatory Activity and Prevents Bacterial Sepsis by Increasing IL-10 Production. J. Nat. Prod. 2018, 81, 1435–1443. [Google Scholar] [CrossRef] [PubMed]
  24. Gong, G.; Guan, Y.-Y.; Zhang, Z.-L.; Rahman, K.; Wang, S.-J.; Zhou, S.; Luan, X.; Zhang, H. Isorhamnetin: A review of pharmacological effects. Biomed. Pharmacother. 2020, 128, 110301. [Google Scholar] [CrossRef] [PubMed]
  25. Arts, I.C.; Sesink, A.L.; Faassen-Peters, M.; Hollman, P.C. The type of sugar moiety is a major determinant of the small intestinal uptake and subsequent biliary excretion of dietary quercetin glycosides. Br. J. Nutr. 2004, 91, 841–847. [Google Scholar] [CrossRef]
  26. Wong, C.C.; Akiyama, Y.; Abe, T.; Lippiat, J.D.; Orfila, C.; Williamson, G. Carrier-mediated transport of quercetin conjugates: Involvement of organic anion transporters and organic anion transporting polypeptides. Biochem. Pharmacol. 2012, 84, 564–570. [Google Scholar] [CrossRef]
  27. Taddei, S.; Virdis, A.; Ghiadoni, L.; Sudano, I.; Salvetti, A. Endothelial Dysfunction in Hypertension. J. Cardiovasc. Pharmacol. 2001, 38, S11–S14. [Google Scholar] [CrossRef]
  28. Incalza, M.A.; D’Oria, R.; Natalicchio, A.; Perrini, S.; Laviola, L.; Giorgino, F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul. Pharmacol. 2018, 100, 1–19. [Google Scholar] [CrossRef]
  29. Ling, W.; Murugan, D.; Lau, Y.; Vanhoutte, P.M.; Mustafa, M.R. Sodium nitrite exerts an antihypertensive effect and improves endothelial function through activation of eNOS in the SHR. Sci. Rep. 2016, 6, 33048. [Google Scholar] [CrossRef]
  30. Lin, X.; Han, T.; Fan, Y.; Wu, S.; Wang, F.; Wang, C. Quercetin improves vascular endothelial function through promotion of autophagy in hypertensive rats. Life Sci. 2020, 258, 118106. [Google Scholar] [CrossRef]
  31. Shen, Y.; Croft, K.D.; Hodgson, J.M.; Kyle, R.; Lee, I.L.; Wang, Y.; Stocker, R.; Ward, N.C. Quercetin and its metabolites improve vessel function by inducing eNOS activity via phosphorylation of AMPK. Biochem. Pharmacol. 2012, 84, 1036–1044. [Google Scholar] [CrossRef] [PubMed]
  32. Pereira, S.C.; Parente, J.M.; Belo, V.A.; Mendes, A.S.; Gonzaga, N.A.; do Vale, G.T.; Ceron, C.S.; Tanus-Santos, J.E.; Tirapelli, C.R.; Castro, M.M. Quercetin decreases the activity of matrix metalloproteinase-2 and ameliorates vascular remodeling in renovascular hypertension. Atherosclerosis 2018, 270, 146–153. [Google Scholar] [CrossRef] [PubMed]
  33. Hasima, N.; Ozpolat, B. Regulation of autophagy by polyphenolic compounds as a potential therapeutic strategy for cancer. Cell Death Dis. 2014, 5, e1509. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, K.; Liu, R.; Li, J.; Mao, J.; Lei, Y.; Wu, J.; Zeng, J.; Zhang, T.; Wu, H.; Chen, L.; et al. Quercetin induces protective autophagy in gastric cancer cells: Involvement of Akt-mTOR- and hypoxia-induced factor 1α-mediated signaling. Autophagy 2011, 7, 966–978. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, Y.; Gong, W.; Yang, Z.Y.; Zhou, X.S.; Gong, C.; Zhang, T.R.; Wei, X.; Ma, D.; Ye, F.; Gao, Q.L. Quercetin induces protective autophagy and apoptosis through ER stress via the p-STAT3/Bcl-2 axis in ovarian cancer. Apoptosis 2017, 22, 544–557. [Google Scholar] [CrossRef] [PubMed]
  36. Jofré, I.; Pezoa, C.; Cuevas, M.; Scheuermann, E.; Freires, I.A.; Rosalen, P.L.; de Alencar, S.M.; Romero, F. Antioxidant and Vasodilator Activity of Ugni molinae Turcz. (Murtilla) and Its Modulatory Mechanism in Hypotensive Response. Oxid. Med. Cell. Longev. 2016, 6513416. [Google Scholar] [CrossRef]
  37. Carullo, G.; Ahmed, A.; Trezza, A.; Spiga, O.; Brizzi, A.; Saponara, S.; Fusi, F.; Aiello, F. Design, synthesis and pharmacological evaluation of ester-based quercetin derivatives as selective vascular KCa1.1 channel stimulators. Bioorgan. Chem. 2020, 105, 104404. [Google Scholar] [CrossRef]
  38. Elbarbry, F.; Abdelkawy, K.; Moshirian, N.; Abdel-Megied, A.M. The Antihypertensive Effect of Quercetin in Young Spontaneously Hypertensive Rats; Role of Arachidonic Acid Metabolism. Int. J. Mol. Sci. 2020, 21, 6554. [Google Scholar] [CrossRef]
  39. Duarte, J.; Perez-Palencia, R.; Vargas, F.; Ocete, M.A.; Perez-Vizcaino, F.; Zarzuelo, A.; Tamargo, J. Antihypertensive effects of the flavonoid quercetin in spontaneously hypertensive rats. Br. J. Pharmacol. 2001, 133, 117–124. [Google Scholar] [CrossRef]
  40. Carlstrom, J.; Symons, J.D.; Wu, T.C.; Bruno, R.S.; Litwin, S.E.; Jalili, T. A quercetin supplemented diet does not prevent cardiovascular complications in spontaneously hypertensive rats. J. Nutr. 2007, 137, 628–633. [Google Scholar] [CrossRef]
  41. Häckl, L.P.; Cuttle, G.; Dovichi, S.S.; Lima-Landman, M.T.; Nicolau, M. Inhibition of angiotesin-converting enzyme by quercetin alters the vascular response to brandykinin and angiotensin I. Pharmacology 2002, 65, 182–186. [Google Scholar] [CrossRef] [PubMed]
  42. Loizzo, M.R.; Said, A.; Tundis, R.; Rashed, K.; Statti, G.A.; Hufner, A.; Menichini, F. Inhibition of angiotensin converting enzyme (ACE) by flavonoids isolated from Ailanthus excelsa (Roxb) (Simaroubaceae). Phytother. Res. 2007, 21, 32–36. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, X.; Raghuvanshi, R.; Ceylan, F.D.; Bolling, B.W. Quercetin and Its Metabolites Inhibit Recombinant Human Angiotensin-Converting Enzyme 2 (ACE2) Activity. J. Agric. Food Chem. 2020, 68, 13982–13989. [Google Scholar] [CrossRef] [PubMed]
  44. Abdelkarem, H.M.; Fadda, L.H. Flaxseed and quercetin improve anti-inflammatory cytokine level and insulin sensitivity in animal model of metabolic syndrome, the fructose-fed rats. Arab. J. Chem. 2017, 10, 3015–3020. [Google Scholar] [CrossRef]
  45. Alam, M.M.; Meerza, D.; Naseem, I. Protective effect of quercetin on hyperglycemia, oxidative stress and DNA damage in alloxan induced type 2 diabetic mice. Life Sci. 2014, 109, 8–14. [Google Scholar] [CrossRef] [PubMed]
  46. Gomes, I.B.S.; Porto, M.L.; Santos, M.C.L.F.S.; Campagnaro, B.P.; Gava, A.L.; Meyrelles, S.S.; Pereira, T.M.C.; Vasquez, E.C. Theprotective effects of oral low-dose quercetin on diabetic nephropathy in hypercholesterolemic mice. Front. Physiol. 2015, 6, 247. [Google Scholar] [CrossRef] [PubMed]
  47. Eid, H.M.; Nachar, A.; Thong, F.; Sweeney, G.; Haddad, P.S. The molecular basis of the antidiabetic action of quercetin in cultured skeletal muscle cells and hepatocytes. Pharmacogn. Mag. 2015, 11, 74–81. [Google Scholar]
  48. Dhanya, R.; Arya, A.D.; Nisha, P.; Jayamurthy, P. Quercetin, a Lead Compound against Type 2 Diabetes Ameliorates Glucose Uptake via AMPK Pathway in Skeletal Muscle Cell Line. Front. Pharmacol. 2017, 8, 336. [Google Scholar] [CrossRef]
  49. Dhanya, R.; Arun, K.B.; Syama, H.P.; Nisha, P.; Sundaresan, A.; Santhosh Kumar, T.R.; Jayamurthy, P. Rutin and quercetin enhance glucose uptake in L6 myotubes under oxidative stress induced by tertiary butyl hydrogen peroxide. Food Chem. 2014, 158, 546–554. [Google Scholar] [CrossRef]
  50. Eid, H.M.; Martineau, L.C.; Saleem, A.; Muhammad, A.; Vallerand, D.; Benhaddou-Andaloussi, A.; Nistor, L.; Afshar, A.; Arnason, J.T.; Haddad, P.S. Stimulation of AMP-activated protein kinase and enhancement of basal glucose uptake in muscle cells by quercetin and quercetin glycosides, active principles of the antidiabetic medicinal plant Vaccinium vitis-idaea. Mol. Nutr. Food Res. 2010, 54, 991–1003. [Google Scholar] [CrossRef]
  51. Jiang, H.; Yamashita, Y.; Nakamura, A.; Croft, K.; Ashida, H. Quercetin and its metabolite isorhamnetin promote glucose uptake through different signalling pathways in myotubes. Sci. Rep. 2019, 9, 2690. [Google Scholar] [CrossRef] [PubMed]
  52. Shi, G.-J.; Li, Y.; Cao, Q.-H.; Wu, H.-X.; Tang, X.-Y.; Gao, X.-H.; Yub, J.Q.; Chen, Z.; Yang, Y. In vitro and in vivo evidence that quercetin protects against diabetes and its complications: A systematic review of the literature. Biomed. Pharmacother. 2019, 109, 1085–1099. [Google Scholar] [CrossRef] [PubMed]
  53. Juźwiak, S.; Wójcicki, J.; Mokrzycki, K.; Marchlewicz, M.; Białecka, M.; Wenda-Rózewicka, L.; Gawrońska-Szklarz, B.; Droździk, M. Effect of quercetin on experimental hyperlipidemia and atherosclerosis in rabbits. Pharmacol. Rep. 2005, 57, 604–609. [Google Scholar] [PubMed]
  54. Castillo, R.L.; Herrera, E.A.; Gonzalez-Candia, A.; Reyes-Farias, M.; de la Jara, N.; Peña, J.P.; Carrasco-Pozo, C. Quercetin Prevents Diastolic Dysfunction Induced by a High-Cholesterol Diet: Role of Oxidative Stress and Bioenergetics in Hyperglycemic Rats. Oxid. Med. Cell. Longev. 2018, 1–14, 7239123. [Google Scholar] [CrossRef]
  55. Sun, L.; Li, E.; Wang, F.; Wang, T.; Qin, Z.; Niu, S.; Qiu, C. Quercetin increases macrophage cholesterol efflux to inhibit foam cell formation through activating PPARγ-ABCA1 pathway. Int. J. Clin. Exp. Pathol. 2015, 8, 10854–10860. [Google Scholar]
  56. Cui, Y.; Hou, P.; Li, F.; Liu, Q.; Qin, S.; Zhou, G.; Xu, X.; Si, Y.; Guo, S. Quercetin improves macrophage reverse cholesterol transport in apolipoprotein E-deficient mice fed a high-fat diet. Lipids Health Dis. 2017, 16, 1–7. [Google Scholar] [CrossRef]
  57. Huwait, E.A.; Saddeek, S.Y.; Al-Massabi, R.F.; Almowallad, S.J.; Pushparaj, P.N.; Kalamegam, G. Antiatherogenic Effects of Quercetin in the THP-1 Macrophage Model In Vitro, With Insights into Its Signaling Mechanisms Using In Silico Analysis. Front. Pharmacol. 2021, 12, 698138. [Google Scholar] [CrossRef]
  58. Liang, N.; Li, Y.M.; He, Z.; Hao, W.; Zhao, Y.; Liu, J.; Zhu, H.; Kwek, E.; Ma, K.Y.; He, W.S.; et al. Rutin and Quercetin Decrease Cholesterol in HepG2 Cells but Not Plasma Cholesterol in Hamsters by Oral Administration. Molecules 2021, 26, 3766. [Google Scholar] [CrossRef]
  59. Ferenczyova, K.; Kalocayova, B.; Bartekova, M. Potential Implications of Quercetin and its Derivatives in Cardioprotection. Int. J. Mol. Sci. 2020, 21, 1585. [Google Scholar] [CrossRef]
  60. Patel, R.V.; Mistry, B.M.; Shinde, S.K.; Syed, R.; Singh, V.; Shin, H.S. Therapeutic potential of quercetin as a cardiovascular agent. Eur. J. Med. Chem. 2018, 155, 889–904. [Google Scholar] [CrossRef]
  61. Jing, Z.; Wang, Z.; Li, X.; Li, X.; Cao, T.; Bi, Y.; Zhou, J.; Chen, X.; Yu, D.; Zhu, L.; et al. Protective Effect of Quercetin on Posttraumatic Cardiac Injury. Sci. Rep. 2016, 6, 30812. [Google Scholar] [CrossRef] [PubMed]
  62. Santos, M.S.; Oliveira, E.D.; Santos-Miranda, A.; Cruz, J.S.; Gondim, A.N.S.; Menezes-Filho, J.E.R.; Souza, D.S.; Pinho-da-Silva, L.; Jesus, I.C.G.; Roman-Campos, D.; et al. Dissection of the Effects of Quercetin on Mouse Myocardium. Basic Clin. Pharmacol. Toxicol. 2017, 120, 550–559. [Google Scholar] [CrossRef] [PubMed]
  63. Guo, X.; Chen, M.; Zeng, H.; Liu, P.; Zhu, X.; Zhou, F.; Liu, J.; Zhang, J.; Dong, Z.; Tang, Y.; et al. Quercetin Attenuates Ethanol-Induced Iron Uptake and Myocardial Injury by Regulating the Angiotensin II-L-Type Calcium Channel. Mol. Nutr. Food Res. 2018, 62, 1700772. [Google Scholar]
  64. Bali, E.B.; Ergin, V.; Rackova, L.; Bayraktar, O.; Küçükboyaci, N.; Karasu, Ç. Olive leaf extracts protect cardiomyocytes against 4-hydroxynonenal-induced toxicity in vitro: Comparison with oleuropein, hydroxytyrosol, and quercetin. Planta Med. 2014, 80, 984–992. [Google Scholar] [CrossRef]
  65. Li, C.; Wang, T.; Zhang, C.; Xuan, J.; Su, C.; Wang, Y. Quercetin attenuates cardiomyocyte apoptosis via inhibition of JNK and p38 mitogen-activated protein kinase signaling pathways. Gene 2016, 577, 275–280. [Google Scholar] [CrossRef] [PubMed]
  66. Lozano, O.; Lázaro-Alfaro, A.; Silva-Platas, C.; Oropeza-Almazán, Y.; Torres-Quintanilla, A.; Bernal-Ramírez, J.; Alves-Figueiredo, H.; García-Rivas, G. Nanoencapsulated Quercetin Improves Cardioprotection during Hypoxia-Reoxygenation Injury through Preservation of Mitochondrial Function. Oxid. Med. Cell. Longev. 2019, 7683051. [Google Scholar] [CrossRef] [PubMed]
  67. Chang, X.; Zhang, T.; Meng, Q.; Yan, P.; Wang, X.; Luo, D.; Zhou, X.; Ji, R. Quercetin Improves Cardiomyocyte Vulnerability to Hypoxia by Regulating SIRT1/TMBIM6-Related Mitophagy and Endoplasmic Reticulum Stress. Oxid. Med. Cell. Longev. 2021, 5529913. [Google Scholar] [CrossRef] [PubMed]
  68. Dong, L.Y.; Chen, F.; Xu, M.; Yao, L.P.; Zhang, Y.J.; Zhuang, Y. Quercetin attenuates myocardial ischemia-reperfusion injury via downregulation of the HMGB1-TLR4-NF-κB signaling pathway. Am. J. Transl. Res. 2018, 10, 1273–1283. [Google Scholar]
  69. Jin, H.B.; Yang, Y.B.; Song, Y.L.; Zhang, Y.C.; Li, Y.R. Protective roles of quercetin in acute myocardial ischemia and reperfusion injury in rats. Mol. Biol. Rep. 2012, 39, 11005–11009. [Google Scholar] [CrossRef]
  70. Albadrani, G.M.; BinMowyna, M.N.; Bin-Jumah, M.N.; El-Akabawy, G.; Aldera, H.; AL-Farga, A.M. Quercetin prevents myocardial infarction adverse remodeling in rats by attenuating TGF-β1/Smad3 signaling: Different mechanisms of action. Saudi J. Biol. Sci. 2021, 28, 2772–2782. [Google Scholar] [CrossRef]
  71. Wang, Y.; Zhang, Z.Z.; Wu, Y.; Ke, J.J.; He, X.H.; Wang, Y.L. Quercetin postconditioning attenuates myocardial ischemia/reperfusion injury in rats through the PI3K/Akt pathway. Braz. J. Med. Biol. 2013, 46, 861–867. [Google Scholar] [CrossRef] [PubMed]
  72. Mamoshina, P.; Rodriguez, B.; Bueno-Orovio, A. Toward a broader view of mechanisms of drug cardiotoxicity. Cell Rep. Med. 2021, 2, 100216. [Google Scholar] [CrossRef] [PubMed]
  73. Manta, K.; Papakyriakopoulou, P.; Chountoulesi, M.; Diamantis, D.A.; Spaneas, D.; Vakali, V.; Naziris, N.; Chatziathanasiadou, M.V.; Andreadelis, I.; Moschovou, K.; et al. Preparation and Biophysical Characterization of Quercetin Inclusion Complexes with β-Cyclodextrin Derivatives to be Formulated as Possible Nose-to-Brain Quercetin Delivery Systems. Mol. Pharm. 2020, 17, 4241–4255. [Google Scholar] [CrossRef] [PubMed]
  74. Hanasaki, Y.; Ogawa, S.; Fukui, S. The correlation between active oxygens scavenging and antioxidative effects of flavonoids. Free Radic. Biol. Med. 1994, 16, 845–850. [Google Scholar] [CrossRef]
  75. Matouk, A.I.; Taye, A.; Heeba, G.H.; El-Moselhy, M.A. Quercetin augments the protective effect of losartan against chronic doxorubicin cardiotoxicity in rats. Environ. Toxicol. Pharmacol. 2013, 36, 443–450. [Google Scholar] [CrossRef]
  76. Nazmi, A.S.; Ahmad, S.J.; Pillai, K.K.; Akhtar, M.; Ahmad, A.; Najmi, A.K. Protective effects of Bombyx mori, quercetin and benazepril against doxorubicin induced cardiotoxicity and nephrotoxicity. J. Saudi Chem. Soc. 2016, 20, 573–578. [Google Scholar] [CrossRef]
  77. Salvatorelli, E.; Guarnieri, S.; Menna, P.; Liberi, G.; Calafiore, A.M.; Mariggiò, M.A.; Mordente, A.; Gianni, L.; Minotti, G. Defective one- or two-electron reduction of the anticancer anthracycline epirubicin in human heart. Relative importance of vesicular sequestration and impaired efficiency of electron addition. J. Biol. Chem. 2006, 281, 10990–11001. [Google Scholar] [CrossRef]
  78. Prasun, P. Mitochondrial dysfunction in metabolic syndrome. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165838. [Google Scholar] [CrossRef]
  79. Iwara, I.; Mboso, E.O.; Eteng, O.; Elot, K.N.; Igile, G.O.; Ebong, P.E. Peristrophe Bicalyculata Extract and Quercetin Ameliorate High Fat Diet- Streptozotocin- Induced Type II Diabetes in Wistar Rats. SSRN 2022, 2, 100060. [Google Scholar] [CrossRef]
  80. Zhou, J.-F.; Wang, W.-J.; Yin, Z.-P.; Zheng, G.-D.; Chen, J.-G.; Li, J.-E.; Chen, L.L.; Zhang, Q.-F. Quercetin is a promising pancreatic lipase inhibitor in reducing fat absorption in vivo. Food Biosci. 2021, 43, 101248. [Google Scholar] [CrossRef]
  81. Boydens, C.; Pauwels, B.; Vanden Daele, L.; Van de Voorde, J. Protective effect of resveratrol and quercetin on in vitro-induced diabetic mouse corpus cavernosum. Cardiovasc. Diabetol. 2016, 15, 46. [Google Scholar] [CrossRef] [PubMed]
  82. Xie, J.; Song, W.; Liang, X.; Zhang, Q.; Shi, Y.; Liu, W.; Shi, X. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed. Pharmacother. 2020, 127, 110147. [Google Scholar] [CrossRef] [PubMed]
  83. Mariee, A.D.; Abd-Allah, G.M.; El-Beshbishy, H.A. Protective effect of dietary flavonoid quercetin against lipemic-oxidative hepatic injury in hypercholesterolemic rats. Pharm. Biol. 2012, 50, 1019–1025. [Google Scholar] [CrossRef] [PubMed]
  84. Ting, Y.; Chang, W.T.; Shiau, D.K.; Chou, P.H.; Wu, M.F.; Hsu, C.L. Antiobesity Efficacy of Quercetin-Rich Supplement on Diet-Induced Obese Rats: Effects on Body Composition, Serum Lipid Profile, and Gene Expression. J. Agric. Food Chem. 2018, 66, 70–80. [Google Scholar] [CrossRef]
  85. Kuipers, E.N.; Dam, A.; Held, N.M.; Mol, I.M.; Houtkooper, R.H.; Rensen, P.; Boon, M.R. Quercetin Lowers Plasma Triglycerides Accompanied by White Adipose Tissue Browning in Diet-Induced Obese Mice. Int. J. Mol. Sci. 2018, 19, 1786. [Google Scholar] [CrossRef]
  86. Wang, M.; Xiao, F.L.; Mao, Y.J.; Ying, L.L.; Zhou, B.; Li, Y. Quercetin decreases the triglyceride content through the PPAR signalling pathway in primary hepatocytes of broiler chickens. Biotechnol. Biotechnol. Equip. 2019, 33, 1000–1010. [Google Scholar] [CrossRef]
  87. Muselin, F.; Cristina, R.T.; Dumitrescu, E.; Doma, A.O.; Radulov, I.; Berbecea, A.A.; Horablaga, A.; Morariu, F.E.; Manea, D.N.; Horablaga, N.M. Quercetin Beneficial Role in the Homeostatic Variation of Certain Trace Elements in Dyslipidemic Mice. Evid.-Based Complement. Altern. Med. 2022, 3299505. [Google Scholar] [CrossRef]
  88. Jia, Q.; Cao, H.; Shen, D.; Li, S.; Yan, L.; Chen, C.; Xing, S.; Dou, F. Quercetin protects against atherosclerosis by regulating the expression of PCSK9, CD36, PPARγ, LXRα and ABCA1. Int. J. Mol. Med. 2019, 44, 893–902. [Google Scholar] [CrossRef]
  89. Gao, X.R.; Chen, Z.; Fang, K.; Xu, J.X.; Ge, J.F. Protective effect of quercetin against the metabolic dysfunction of glucose and lipids and its associated learning and memory impairments in NAFLD rats. Lipids Health Dis. 2021, 20, 164. [Google Scholar] [CrossRef]
  90. Xiao, L.; Liu, L.; Guo, X.; Zhang, S.; Wang, J.; Zhou, F.; Liu, L.; Tang, Y.; Yao, P. Quercetin attenuates high fat diet-induced atherosclerosis in apolipoprotein E knockout mice: A critical role of NADPH oxidase. Food Chem. Toxicol. 2017, 105, 22–33. [Google Scholar] [CrossRef]
  91. Garelnabi, M.; Mahini, H.; Wilson, T. Quercetin intake with exercise modulates lipoprotein metabolism and reduces atherosclerosis plaque formation. J. Int. Soc. Sports Nutr. 2014, 11, 22. [Google Scholar] [CrossRef] [PubMed]
  92. Hayek, T.; Fuhrman, B.; Vaya, J.; Rosenblat, M.; Belinky, P.; Coleman, R.; Elis, A.; Aviram, M. Reduced progression of atherosclerosis in apolipoprotein E-deficient mice following consumption of red wine, or its polyphenols quercetin or catechin, is associated with reduced susceptibility of LDL to oxidation and aggregation. Arterioscler. Thromb. Vasc. Biol. 1997, 17, 2744–2752. [Google Scholar] [CrossRef] [PubMed]
  93. Zhi, K.; Li, M.; Bai, J.; Wu, Y.; Zhou, S.; Zhang, X.; Qu, L. Quercitrin treatment protects endothelial progenitor cells from oxidative damage via inducing autophagy through extracellular signal-regulated kinase. Angiogenesis 2016, 19, 311–324. [Google Scholar] [CrossRef] [PubMed]
  94. Cao, H.; Jia, Q.; Shen, D.; Yan, L.; Chen, C.; Xing, S. Quercetin has a protective effect on atherosclerosis via enhancement of autophagy in ApoE-/- mice. Exp. Ther. Med. 2019, 18, 2451–2458. [Google Scholar] [CrossRef]
  95. Xue, F.; Nie, X.; Shi, J.; Liu, Q.; Wang, Z.; Li, X.; Zhou, J.; Su, J.; Xue, M.; Chen, W.D.; et al. Quercetin Inhibits LPS-Induced Inflammation and ox-LDL-Induced Lipid Deposition. Front. Pharmacol. 2017, 8, 40. [Google Scholar] [CrossRef]
  96. Nie, J.; Zhang, L.; Zhao, G.; Du, X. Quercetin reduces atherosclerotic lesions by altering the gut microbiota and reducing atherogenic lipid metabolites. J. Appl. Microbiol. 2019, 127, 1824–1834. [Google Scholar] [CrossRef]
  97. Li, S.; Cao, H.; Shen, D.; Chen, C.; Xing, S.; Dou, F.; Jia, Q. Effect of Quercetin on Atherosclerosis Based on Expressions of ABCA1, LXR-α and PCSK9 in ApoE-/- Mice. Chin. J. Integr. Med. 2020, 26, 114–121. [Google Scholar] [CrossRef]
  98. Bhaskar, S.; Sudhakaran, P.; Helen, A. Quercetin attenuates atherosclerotic inflammation and adhesion molecule expression by modulating TLR-NF-κB signaling pathway. Cell. Immunol. 2016, 310, 131–140. [Google Scholar] [CrossRef]
  99. Lin, W.; Wang, W.; Wang, D.; Ling, W. Quercetin protects against atherosclerosis by inhibiting dendritic cell activation. Mol. Nutr. Food Res. 2017, 61, 1700031. [Google Scholar] [CrossRef]
  100. Bian, Y.; Liu, P.; Zhong, J.; Hu, Y.; Zhuang, S.; Fan, K.; Liu, Z. Quercetin Attenuates Adhesion Molecule Expression in Intestinal Microvascular Endothelial Cells by Modulating Multiple Pathways. Dig. Dis. Sci. 2018, 63, 3297–3304. [Google Scholar] [CrossRef]
  101. Atrahimovich, D.; Samson, A.; Khattib, A.; Vaya, J.; Khatib, S. Punicalagin Decreases Serum Glucose Levels and Increases PON1 Activity and HDL Anti-Inflammatory Values in Balb/c Mice Fed a High-Fat Diet. Oxid. Med. Cell. Longev. 2018, 2673076. [Google Scholar] [CrossRef] [PubMed]
  102. Li, H.; Xiao, L.; He, H.; Zeng, H.; Liu, J.; Jiang, C.; Mei, G.; Yu, J.; Chen, H.; Yao, P.; et al. Quercetin Attenuates Atherosclerotic Inflammation by Inhibiting Galectin-3-NLRP3 Signaling Pathway. Mol. Nutr. Food Res. 2021, 65, e2000746. [Google Scholar] [CrossRef] [PubMed]
  103. Lu, X.; Zhao, C.; Yao, X.; Zhang, H. Quercetin attenuates high fructose feeding-induced atherosclerosis by suppressing inflammation and apoptosis via ROS-regulated PI3K/AKT signaling pathway. Biomed. Pharmacother. 2017, 85, 658–671. [Google Scholar] [CrossRef] [PubMed]
  104. Gong, M.; Garige, M.; Varatharajalu, R.; Marmillot, P.; Gottipatti, C.; Leckey, L.; Lakshman, R.M. Quercetin up-regulates paraoxonase 1 gene expression with concomitant protection against LDL oxidation. Biochem. Biophys. Res. Commun. 2009, 379, 1001–1004. [Google Scholar] [CrossRef]
  105. Yao, S.; Sang, H.; Song, G.; Yang, N.; Liu, Q.; Zhang, Y.; Jiao, P.; Zong, C.; Qin, S. Quercetin protects macrophages from oxidized low-density lipoprotein-induced apoptosis by inhibiting the endoplasmic reticulum stress-C/EBP homologous protein pathway. Exp. Biol. Med. 2012, 237, 822–831. [Google Scholar] [CrossRef]
  106. Kondo, M.; Izawa-Ishizawa, Y.; Goda, M.; Hosooka, M.; Kagimoto, Y.; Saito, N.; Matsuoka, R.; Zamami, Y.; Chuma, M.; Yagi, K.; et al. Preventive Effects of Quercetin against the Onset of Atherosclerosis-Related Acute Aortic Syndromes in Mice. Int. J. Mol. Sci. 2020, 21, 7226. [Google Scholar] [CrossRef]
  107. Jiang, Y.; Jiang, L.; Wang, Y.; Ma, D.; Li, X. Quercetin Attenuates Atherosclerosis via Modulating Oxidized LDL-Induced Endothelial Cellular Senescence. Front. Pharmacol. 2020, 11, 512. [Google Scholar]
  108. Song, S.; Jang, S.; Kang, H.; Wei, B.; Jeoun, U.; Yoon, G.; Hwang, E.S. Modulation of Mitochondrial Membrane Potential and ROS Generation by Nicotinamide in a Manner Independent of SIRT1 and Mitophagy. Mol. Cells 2017, 40, 503–514. [Google Scholar]
  109. Song, L.; Xu, M.; Lopes-Virella, M.; Huang, Y. Quercetin inhibits matrix metalloproteinase-1 expression in human vascular endothelial cells through extracellular signal-regulated kinase. Arch. Biochem. Biophys. 2001, 391, 72–78. [Google Scholar] [CrossRef]
  110. Saragusti, A.; Ortega, M.; Cabrera, J.; Estrin, D.; Marti, M.; Chiabrando, G. Inhibitory effect of quercetin on matrix metalloproteinase 9 activity molecular mechanism and structure-activity relationship of the flavonoid-enzyme interaction. Eur. J. Pharmacol. 2010, 644, 138–145. [Google Scholar] [CrossRef]
  111. Jung, C.H.; Cho, I.; Ahn, J.; Jeon, T.-I.; Ha, T.-Y. Quercetin Reduces High-Fat Diet-Induced Fat Accumulation in the Liver by Regulating Lipid Metabolism Genes. Phyt. Res. 2012, 27, 139–143. [Google Scholar] [CrossRef] [PubMed]
  112. Fukaya, M.; Sato, Y.; Kondo, S.; Adachi, S.; Yoshizawa, F.; Sato, Y. Quercetin enhances fatty acid β-oxidation by inducing lipophagy in AML12 hepatocytes. Heliyon 2021, 7, e07324. [Google Scholar] [CrossRef] [PubMed]
  113. Rasheed, R.A.; Elshikh, M.S.; Mohamed, M.O.; Darweesh, M.F.; Hussein, D.S.; Almutairi, S.M.; Embaby, A.S. Quercetin mitigates the adverse effects of high fat diet on pancreatic and renal tissues in adult male albino rats. J. King Saud Univ. Sci. 2022, 34, 101946. [Google Scholar] [CrossRef]
  114. Puteri, M.U.; Azmi, N.U.; Kato, M.; Saputri, F.C. PCSK9 Promotes Cardiovascular Diseases: Recent Evidence about Its Association with Platelet Activation-Induced Myocardial Infarction. Life 2022, 12, 190. [Google Scholar] [CrossRef] [PubMed]
  115. Kadoglou, N.P.; Kapelouzou, A.; Tsanikidis, H.; Vitta, I.; Liapis, C.D.; Sailer, N. Effects of rosiglitazone/metformin fixed-dose combination therapy and metformin monotherapy on serum vaspin, adiponectin and IL-6 levels in drug-naïve patients with type 2 diabetes. Exp. Clin. Endocrinol. Diabetes 2011, 119, 63–68. [Google Scholar] [CrossRef]
  116. Ellulu, M.S.; Patimah, I.; Khaza’ai, H.; Rahmat, A.; Abed, Y. Obesity and inflammation: The linking mechanism and the complications. Arch. Med. Sci. 2017, 13, 851–863. [Google Scholar] [CrossRef]
  117. Rushchak, V.; Voronina, A.; Shayakhmetova, A.; Chashchyn, M. Quercetin effectiveness in the prevention of pathological processes at the metabolic syndrome. Toxicol. Lett. 2013, 221, 171. [Google Scholar] [CrossRef]
  118. Ahn, J.; Lee, H.; Kim, S.; Park, J.; Ha, T. The anti-obesity effect of quercetin is mediated by the AMPK and MAPK signaling pathways. Biochem. Biophys. Res. Commun. 2008, 373, 545–549. [Google Scholar] [CrossRef] [PubMed]
  119. Rodrigues, B.A.; Muñoz, V.R.; Kuga, G.K.; Gaspar, R.C.; Nakandakari, S.; Crisol, B.M.; Botezelli, J.D.; Pauli, L.; da Silva, A.; de Moura, L.P.; et al. Obesity Increases Mitogen-Activated Protein Kinase Phosphatase-3 Levels in the Hypothalamus of Mice. Front. Cell. Neurosci. 2017, 11, 313. [Google Scholar] [CrossRef]
  120. Hemmati, M.; Mostafavi, S.E.; Zarban, A.; Hoshyar, R. Protective Effects of Quercetin on Hyperglycemia and Stress Proteins Expression in Rats with Streptozocin-Induced Diabetes. Brieflands 2018, 15, e64964. [Google Scholar] [CrossRef]
  121. Le, N.H.; Kim, C.S.; Park, T.; Park, J.H.; Sung, M.K.; Lee, D.G.; Hong, S.M.; Choe, S.Y.; Goto, T.; Kawada, T.; et al. Quercetin protects against obesity-induced skeletal muscle inflammation and atrophy. Mediat. Inflamm. 2014, 834294. [Google Scholar] [CrossRef]
  122. Feng, J.; Ge, C.; Li, W.; Li, R. 3-(3-Hydroxyphenyl)propionic acid, a microbial metabolite of quercetin, inhibits monocyte binding to endothelial cells via modulating E-selectin expression. Fitoterapia 2022, 156, 105071. [Google Scholar] [CrossRef] [PubMed]
  123. Kadoglou, N.P.E.; Velidakis, N.; Khattab, E.; Kassimis, G.; Patsourakos, N. The interplay between statins and adipokines. Is this another explanation of statins’ ‘pleiotropic’ effects? Cytokine 2021, 148, 155698. [Google Scholar] [CrossRef] [PubMed]
  124. Mirsafaei, L.; Reiner, Ž.; Shafabakhsh, R.; Asemi, Z. Molecular and Biological Functions of Quercetin as a Natural Solution for Cardiovascular Disease Prevention and Treatment. Plant Foods Hum. Nutr. 2020, 75, 307–315. [Google Scholar] [CrossRef]
  125. Huang, H.; Liao, D.; Dong, Y.; Pu, R. Effect of quercetin supplementation on plasma lipid profiles, blood pressure, and glucose levels: A systematic review and meta-analysis. Nutr. Rev. 2020, 78, 615–626. [Google Scholar] [CrossRef] [PubMed]
  126. Menezes, R.; Rodriguez-Mateos, A.; Kaltsatou, A.; González-Sarrías, A.; Greyling, A.; Giannaki, C.; Andres-Lacueva, C.; Milenkovic, D.; Gibney, E.R.; Dumont, J.; et al. Impact of Flavonols on Cardiometabolic Biomarkers: A Meta-Analysis of Randomized Controlled Human Trials to Explore the Role of Inter-Individual Variability. Nutrients 2017, 9, 117. [Google Scholar] [CrossRef] [PubMed]
  127. Jin, S.; Liu, J.; Jia, Y.; Han, T.; Zhao, X.; Sun, C.; Na, L. The association of dietary flavonoids, magnesium and their interactions with the metabolic syndrome in Chinese adults: A prospective cohort study. Br. J. Nutr. 2021, 126, 892–902. [Google Scholar] [CrossRef]
  128. Shatylo, V.; Antoniuk-Shcheglova, I.; Naskalova, S.; Bondarenko, O.; Havalko, A.; Krasnienkov, D.; Zabuga, O.; Kukharskyy, V.; Guryanov, V.; Vaiserman, A. Cardio-metabolic benefits of quercetin in elderly patients with metabolic syndrome. PharmaNutrition 2021, 15, 100250. [Google Scholar] [CrossRef]
  129. Cano-Martínez, A.; Bautista-Pérez, R.; Castrejón-Téllez, V.; Carreón-Torres, E.; Pérez-Torres, I.; Díaz-Díaz, E.; Flores-Estrada, J.; Guarner-Lans, V.; Rubio-Ruíz, M.E. Resveratrol and Quercetin as Regulators of Inflammatory and Purinergic Receptors to Attenuate Liver Damage Associated to Metabolic Syndrome. Int. J. Mol. Sci. 2021, 22, 8939. [Google Scholar] [CrossRef]
  130. Pfeuffer, M.; Auinger, A.; Bley, U.; Kraus-Stojanowic, I.; Laue, C.; Winkler, P.; Rüfer, C.E.; Frank, J.; Bösch-Saadatmandi, C.; Rimbach, G.; et al. Effect of quercetin on traits of the metabolic syndrome, endothelial function and inflammation in men with different APOE isoforms. Nutr. Metab. Cardiovasc. Dis. 2013, 23, 403–409. [Google Scholar] [CrossRef]
  131. Egert, S.; Bosy-Westphal, A.; Seiberl, J.; Kürbitz, C.; Settler, U.; Plachta-Danielzik, S.; Wagner, A.E.; Frank, J.; Schrezenmeir, J.; Rimbach, G.; et al. Quercetin reduces systolic blood pressure and plasma oxidised low-density lipoprotein concentrations in overweight subjects with a high-cardiovascular disease risk phenotype: A double-blinded, placebo-controlled cross-over study. Br. J. Nutr. Camb. 2009, 102, 1065–1074. [Google Scholar] [CrossRef] [PubMed]
  132. Serban, M.C.; Sahebkar, A.; Zanchetti, A.; Mikhailidis, D.P.; Howard, G.; Antal, D.; Andrica, F.; Ahmed, A.; Aronow, W.S.; Muntner, P.; et al. Lipid and Blood Pressure Meta-analysis Collaboration (LBPMC) Group. Effects of Quercetin on Blood Pressure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Am. Heart Assoc. 2016, 5, e002713. [Google Scholar] [CrossRef] [PubMed]
  133. Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: From antioxidant to nutraceutical. Eur. J. Pharmacol. 2008, 585, 325–337. [Google Scholar] [CrossRef] [PubMed]
  134. Tamtaji, O.R.; Milajerdi, A.; Dadgostar, E.; Kolahdooz, F.; Chamani, M.; Amirani, E.; Mirzaei, H.; Asemi, Z. The Effects of Quercetin Supplementation on Blood Pressures and Endothelial Function Among Patients with Metabolic Syndrome and Related Disorders: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Curr. Pharm. Des. 2019, 25, 1372–1384. [Google Scholar] [CrossRef] [PubMed]
  135. Yao, Z.; Dai, K.; Meng, G.; Zhang, Q.; Liu, L.; Wu, H.; Gu, Y.; Sun, S.; Wang, X.; Jia, Q.; et al. Low dietary quercetin intake by food frequency questionnaire analysis is not associated with hypertension occurrence. Clin. Nutr. 2021, 40, 3748–3753. [Google Scholar] [CrossRef] [PubMed]
  136. Biesinger, S.; Michaels, H.A.; Quadros, A.S.; Qian, Y.; Rabovsky, A.B.; Badger, R.S.; Jalili, T. A combination of isolated phytochemicals and botanical extracts lowers diastolic blood pressure in a randomized controlled trial of hypertensive subjects. Eur. J. Clin. Nutr. 2016, 70, 10–16. [Google Scholar] [CrossRef]
  137. Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin reduces blood pressure in hypertensive subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef]
  138. Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does Quercetin Improve Cardiovascular Risk factors and Inflammatory Biomarkers in Women with Type 2 Diabetes: A Double-blind Randomized Controlled Clinical Trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar]
  139. Brüll, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Müller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Naaf, S.; et al. Effects of a quercetin-rich onion skin extract on 24 h ambulatory blood pressure and endothelial function in overweight-to-obese patients with (pre-)hypertension: A randomised double-blinded placebo-controlled cross-over trial. Br. J. Nutr. 2015, 114, 1263–1277. [Google Scholar] [CrossRef]
  140. Larson, A.; Witman, M.A.; Guo, Y.; Ives, S.; Richardson, R.S.; Bruno, R.S.; Jalili, T.; Symons, J.D. Acute, quercetin-induced reductions in blood pressure in hypertensive individuals are not secondary to lower plasma angiotensin-converting enzyme activity or endothelin-1: Nitric oxide. Nutr. Res. 2012, 32, 557–564. [Google Scholar] [CrossRef]
  141. Burak, C.; Wolffram, S.; Zur, B.; Langguth, P.; Fimmers, R.; Alteheld, B.; Stehle, P.; Egert, S. Effect of alpha-linolenic acid in combination with the flavonol quercetin on markers of cardiovascular disease risk in healthy, non-obese adults: A randomized, double-blinded placebo-controlled crossover trial. Nutrition 2019, 58, 47–56. [Google Scholar] [CrossRef] [PubMed]
  142. Sahebkar, A. Effects of quercetin supplementation on lipid profile: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2017, 57, 666–676. [Google Scholar] [CrossRef] [PubMed]
  143. Guo, W.; Gong, X.; Li, M. Quercetin Actions on Lipid Profiles in Overweight and Obese Individuals: A Systematic Review and Meta-Analysis. Curr. Pharm. Des. 2019, 25, 3087–3095. [Google Scholar] [CrossRef] [PubMed]
  144. Mazza, A.; Nicoletti, M.; Lenti, S.; Torin, G.; Rigatelli, G.; Fratter, A. Effectiveness and safety of nutraceutical compounds added to ezetimibe treatment in hypertensive and hyper-cholesterolemic subjects with statinintolerance. J. Hypertens. 2021, 39 (Suppl. S1), e371. [Google Scholar] [CrossRef]
  145. Tabrizi, R.; Tamtaji, O.R.; Mirhosseini, N.; Lankarani, K.B.; Akbari, M.; Heydari, S.T.; Dadgostar, E.; Asemi, Z. The effects of quercetin supplementation on lipid profiles and inflammatory markers among patients with metabolic syndrome and related disorders: A systematic review and meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2020, 60, 1855–1868. [Google Scholar] [CrossRef] [PubMed]
  146. Lu, T.M.; Chiu, H.F.; Shen, Y.C.; Chung, C.C.; Venkatakrishnan, K.; Wang, C.K. Hypocholesterolemic Efficacy of Quercetin Rich Onion Juice in Healthy Mild Hypercholesterolemic Adults: A Pilot Study. Plant Foods Hum. Nutr. 2015, 70, 395–400. [Google Scholar] [CrossRef] [PubMed]
  147. Nishimura, M.; Muro, T.; Kobori, M.; Nishihira, J. Effect of Daily Ingestion of Quercetin-Rich Onion Powder for 12 Weeks on Visceral Fat: A Randomised, Double-Blind, Placebo-Controlled, Parallel-Group Study. Nutrients 2019, 12, 91. [Google Scholar] [CrossRef] [PubMed]
  148. Ostadmohammadi, V.; Milajerdi, A.; Ayati, E.; Kolahdooz, F.; Asemi, Z. Effects of quercetin supplementation on glycemic control among patients with metabolic syndrome and related disorders: A systematic review and meta-analysis of randomized controlled trials. Phytother. Res. 2019, 33, 1330–1340. [Google Scholar] [CrossRef]
  149. Song, Y.; Manson, J.E.; Buring, J.E.; Sesso, H.D.; Liu, S. Associations of dietary flavonoids with risk of type 2 diabetes, and markers of insulin resistance and systemic inflammation in women: A prospective study and cross-sectional analysis. J. Am. Coll. Nutr. 2005, 24, 376–384. [Google Scholar] [CrossRef]
  150. Sales, D.S.; Carmona, F.; de Azevedo, B.C.; Taleb-Contini, S.H.; Bartolomeu, A.C.; Honorato, F.B.; Martinez, E.Z.; Pereira, A.M. Eugenia punicifolia (Kunth) DC. as an adjuvant treatment for type-2 diabetes mellitus: A non-controlled, pilot study. Phytother. Res. 2014, 28, 1816–1821. [Google Scholar] [CrossRef]
  151. Tabasco, R.; Sánchez-Patán, F.; Monagas, M.; Bartolomé, B.; Victoria Moreno-Arribas, M.; Peláez, C.; Requena, T. Effect of grape polyphenols on lactic acid bacteria and bifidobacteria growth: Resistance and metabolism. Food Microbiol. 2011, 28, 1345–1352. [Google Scholar] [CrossRef] [PubMed]
  152. Li, Q.; Gao, B.; Siqin, B.; He, Q.; Zhang, R.; Meng, X.; Zhang, N.; Zhang, N.; Li, M. Gut Microbiota: A Novel Regulator of Cardiovascular Disease and Key Factor in the Therapeutic Effects of Flavonoids. Front. Pharmacol. 2021, 12, 651926. [Google Scholar] [CrossRef] [PubMed]
  153. Chen, W.; Zhong, Y.; Feng, N.; Guo, Z.; Wang, S.; Xing, D. New horizons in the roles and associations of COX-2 and novel natural inhibitors in cardiovascular diseases. Mol. Med. 2021, 27, 123. [Google Scholar] [CrossRef] [PubMed]
  154. Dagher, O.; Mury, P.; Thorin-Trescases, N.; Noly, P.; Thorin, E.; Carrier, M. Therapeutic Potential of Quercetin to Alleviate Endothelial Dysfunction in Age-Related Cardiovascular Diseases. Front. Cardiovasc. Med. 2021, 8, 658400. [Google Scholar] [CrossRef]
  155. Chekalina, N.; Burmak, Y.; Petrov, Y.; Borisova, Z.; Manusha, Y.; Kazakov, Y.; Kaidashev, I. Quercetin reduces the transcriptional activity of NF-kB in stable coronary artery disease. Indian Heart J. 2018, 70, 593–597. [Google Scholar] [CrossRef]
  156. Knekt, P.; Jarvinen, R.; Reunanen, A.; Maatela, J. Flavonoid intake and coronary mortality in Finland: A cohort study. BMJ 1996, 312, 478–481. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the most important cardiovascular protective properties of quercetin and the related possible mechanism of action. Arrows indicate increase (↑) or decrease (↓) of the respective biomarker expression and the resulting formation (→). Key: ABCA1: ATP-binding cassette transporter A1; ACE: angiotensin-converting enzyme; Bcl-2: B cell lymphoma-2; CHOP: C/EBP homologous protein; ECs: endothelial cells; eNOS: endothelial NO synthase; ER: endoplasmic reticulum; ERK: extracellular signal-regulated kinase; ICAM-1: intercellular adhesion molecule 1; IL-6: interleukin 6; JNK: Jun N-terminal kinase; LXR-α: liver X receptor; MMP: matrix metalloproteinase; PCSK9: proprotein convertase subtilisin/kexin type 9; PG: prostaglandin; PGC-1α: peroxisome proliferator-activated receptor γ coactivator-1α; PPARγ: peroxisome proliferator-activated receptor γ; ROS: reactive oxygen species; SIRT1: sirtuin 1; UCP2: uncoupling protein 2.
Figure 1. Schematic representation of the most important cardiovascular protective properties of quercetin and the related possible mechanism of action. Arrows indicate increase (↑) or decrease (↓) of the respective biomarker expression and the resulting formation (→). Key: ABCA1: ATP-binding cassette transporter A1; ACE: angiotensin-converting enzyme; Bcl-2: B cell lymphoma-2; CHOP: C/EBP homologous protein; ECs: endothelial cells; eNOS: endothelial NO synthase; ER: endoplasmic reticulum; ERK: extracellular signal-regulated kinase; ICAM-1: intercellular adhesion molecule 1; IL-6: interleukin 6; JNK: Jun N-terminal kinase; LXR-α: liver X receptor; MMP: matrix metalloproteinase; PCSK9: proprotein convertase subtilisin/kexin type 9; PG: prostaglandin; PGC-1α: peroxisome proliferator-activated receptor γ coactivator-1α; PPARγ: peroxisome proliferator-activated receptor γ; ROS: reactive oxygen species; SIRT1: sirtuin 1; UCP2: uncoupling protein 2.
Pharmaceuticals 15 01019 g001
Figure 2. Chemical structure of quercetin and its main derivatives (metabolites).
Figure 2. Chemical structure of quercetin and its main derivatives (metabolites).
Pharmaceuticals 15 01019 g002
Table 1. Published preclinical studies investigating the impact of quercetin on classical cardiovascular risk factors.
Table 1. Published preclinical studies investigating the impact of quercetin on classical cardiovascular risk factors.
ReferenceAnimal ModelStudy DesignOutcomes
Alam et al., 2014 [45]Swiss albino mice20 mg/kg/day quercetin orally for 3 weeks↓ FBG
↑ GLUT-4
Albadrani et al., 2020 [70]Wistar albino rats50 mg/kg/day quercetin orally for 4 weeks↓ TGF-b1/Smad3 signaling
Abdelkarem and Fadda, 2017 [44]Sprague–Dawley rats50 mg/kg/day quercetin orally for 4 weeks↓ serum glucose, TC, LDL-C, TG, leptin, adiponectin
Carlstrom et al., 2007 [40]Spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY)1.5 g quercetin/kg/day quercetin orally (gavage) for 5 or 11 weeks (SHR)↔ BP, cardiac hypertrophy, vascular dysfunction, vascular remodeling, and indices of oxidative stress in SHR
Castillo et al., 2018 [54]Wistar albino ratsHC diet supplemented with 0.5% w/w quercetin for 4 weeks↓ TG, TC, glucose, oxidative stress suppression, LDL-C and VLDL-C increase, HDL-C decrease
Cui et al., 2017 [56]apoE−/− mice12.5 mg/kg/day quercetin via gavage↑ RCT
Duarte et al., 2009 [39]Spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto rats (WKY)10 mg/kg/day quercetin orally (gavage) for 5 weeks↓ SBP, DBP, and MAP in SHR
Elbarbry et al., 2020 [38]Spontaneously hypertensive rats (SHR)10, 30, and 60 mg/L quercetin in drinking water for 7 weeks↓ MAP at a high dose of quercetin
Gao et al., 2021 [89]Nonalcoholic fatty liver disease (NAFLD) rat model80, 40, or 20 mg/kg/day quercetin via gavage for 4 weeks↓ TC, TG, blood glucose levels
Garelnabi et al., 2022 [91]C57BL6 LDLr−/− mice100 μg/day quercetin orally for 4 weeks↓ AP, MCP-1
Gomes et al., 2015 [46]apoE−/− mice10 mg/kg/day quercetin orally for 4 weeks↓ plasma glucose, TG, TC, tendency to reduce proteinuria and glomerular injury
Häckl et al., 2002 [41]Wistar albino ratsOral or IV preadministration
of 88.7 μmol/kg and 14.7 μmol/kg quercetin, respectively, for 45 and 5 min before a bradykinin IV injection
↑ hypotensive effect of bradykinin
↓ MAP
Hemmati et al., 2018 [120]Wistar albino rats15 mg/kg/day quercetin (i.p. injection) for 3 weeks↓ MDA, mRNA levels of HSP27, HSP70, HSF-1, and glucose-6-phosphatase
↑ glucokinase expression
Iwara et al., 2022 [79]Albino rats10 mg/kg/day quercetin orally for 3 weeks↓ FBG
↑ BW, AST, ALP, ALT, albumin
Jia et al., 2019 [88]apoE−/− mice12.5 mg/kg/day quercetin orally for 12 weeks↓ TC, LDL-C, oxLDL, TNF-α, IL-6, plaques
↑ IL-10, PPAR-γ, LXRα ABCA1
Jin et al., 2012 [96]Sprague–Dawley rats1 mg/kg quercetin IV↓ TNF-α, IL-10
Jung et al., 2012 [111]C57BL/6J miceHC diet supplemented with 0.025% w/w quercetin for 9 weeks↓ BW, size of the epididymal adipose tissue and liver tissue, TBARS, fat, altered expression of the lipid metabolism-related genes
Juźwiak et al., 2005 [53]Mongrel rabbits0.05 mg/kg/day quercetin orally for 4 and 12 weeks↓ TG, TC, plaque formation, thickening of the tunica intima of the aorta
Kuipers et al., 2018 [85]C57Bl/6J miceHC diet supplemented with 0.1% w/w quercetin for 12 weeks↓ TG, white adipose tissue browning
Le et al., 2014 [121]C57BL/6J miceHC diet supplemented with 0.05% and 0.1% w/w quercetin for 9 weeks↓ TNF-α, MCP-1 skeletal muscle atrophy
Liang et al., 2021 [58]Hypercholesterolemia hamsters2.5 g/kg/day quercetin orally for 8 weeks↔ TC
Lin et al., 2020 [30]Spontaneously hypertensive rats (SHR)10 mg/kg/day quercetin orally (gavage) for 6 weeks↓ SBP, DBP
↑ autophagy
Mariee et al., 2012 [83]Sprague–Dawley rats15 mg/kg/day quercetin orally for 2 weeks↓ TG, TC, LDL-C, ALT, AST, γ-GT, liver TBARS
↑ HDL-C, GSH
Matouk et al., 2013 [75]Wistar albino rats10 mg/kg/day quercetin orally for 4 weeks↓ TNF-α, LDH, CK-MB, MDA, NO
↓ CAT, ↓SOD
Muselin et al., 2022 [87]BALB/c mice500 mg/L quercetin in drinking water (duration of the study not reported)↓ TC, LDL-C, TG
Nazmi et al., 2016 [76]Wistar albino rats2 mg/kg/day quercetin orally for 1 week↑ AST, LDH, BUN, creatinine, GSH
Pereira et al., 2018 [32]2K1C hypertensive Wistar albino rats10 mg/kg/day quercetin via gavage for 3 weeks↓ SBP, BW, ROS, MLP
Rasheed et al., 2022 [113]Albino rats50 mg/kg/day of quercetin orally for 12 weeksImprovement of the histopathological degenerative and inflammatory changes
↓ mean area % of collagen fibers
Ting et al., 2018 [84]Wistar albino rats13 mg/kg/day quercetin orally for 8 weeks↓ BW, ALT, TG, TC, size of perirenal adipocytes
↑ adiponectin expression, AST
Wang et al., 2013 [71]Sprague–Dawley rats10 mg/kg quercetin (i.p. injection) 5 min before reperfusion↓ infarct size, serum levels of creatine kinase and lactate dehydrogenase, caspase-3 immunoreactivity, and Bax expression
↑ Akt phosphorylation and Bcl-2 expression
Zhou et al., 2021 [80]Sprague–Dawley ratsOral preadministration of 5 and 10 mg/kg quercetin↓ TG, fat absorption
↑ fat excretion
ALP, alkaline phosphatase; ALT, alanine aminotransferase; AP, atherosclerotic plaques; AST, aspartate aminotransferase; BP, blood pressure; BUN, blood urea nitrogen; BW, body weight; CAT: myocardial catalase; CK-MB, creatine kinase; DBP, diastolic blood pressure; FBG, fasting blood glucose; GLUT-4, glucose transporter type 4; GSH, glutathione; HC, high cholesterol; HDL, high-density lipoprotein; HR, heart rate; HSF1, heat shock factor; 1HSP27, heat shock protein 27; HSP70, heat shock protein 70; IL-10, interleukin 10; i.p., intraperitoneal; IV, intravenously; LDH, lactate dehydrogenase; LDL-C, low-density lipoprotein cholesterol; LXRα, liver X receptor alpha; MAP, mean arterial pressure; MCP-1, monocyte chemoattractant protein-1; MDA, malondialdehyde; MLP: myocardial lipid peroxidation; oxLDL, oxidative modification of a low-density lipoprotein; PPAR-γ, peroxisome proliferator-activated receptor-γ; RCT, reverse cholesterol transport; ROS, reactive oxygen species; SBP, systolic blood pressure; SOD, superoxide dismutase; TBARS, thiobarbituric acid reactive substances; TC, total cholesterol; TG, triglycerides; TGF-b1, transforming growth factor beta 1; TNF-α, tumor necrosis factor; VLDL-C, very low-density lipoprotein cholesterol; ↓, decrease; ↑, increase; ↔, non-significant change.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Papakyriakopoulou, P.; Velidakis, N.; Khattab, E.; Valsami, G.; Korakianitis, I.; Kadoglou, N.P. Potential Pharmaceutical Applications of Quercetin in Cardiovascular Diseases. Pharmaceuticals 2022, 15, 1019. https://doi.org/10.3390/ph15081019

AMA Style

Papakyriakopoulou P, Velidakis N, Khattab E, Valsami G, Korakianitis I, Kadoglou NP. Potential Pharmaceutical Applications of Quercetin in Cardiovascular Diseases. Pharmaceuticals. 2022; 15(8):1019. https://doi.org/10.3390/ph15081019

Chicago/Turabian Style

Papakyriakopoulou, Paraskevi, Nikolaos Velidakis, Elina Khattab, Georgia Valsami, Ioannis Korakianitis, and Nikolaos PE Kadoglou. 2022. "Potential Pharmaceutical Applications of Quercetin in Cardiovascular Diseases" Pharmaceuticals 15, no. 8: 1019. https://doi.org/10.3390/ph15081019

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