Next Article in Journal
The Caenorhabditis elegans sdhb-1(R244H) Model Shows Characteristics of Human PPGL Tumor Cells
Previous Article in Journal
HLA Class II Alleles and Suicidal Behavior: Evidence from a Case–Control Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Navigating the Effects of Anti-Atherosclerotic Supplements and Acknowledging Associated Bleeding Risks

by
Maria-Zinaida Dobre
1,
Bogdana Virgolici
1,*,
Ioana-Cristina Doicin
2,
Horia Vîrgolici
3 and
Iulia-Ioana Stanescu-Spinu
4
1
Department of Biochemistry, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
2
Cardiology Resident at the Central Military Emergency University Hospital “Dr. Carol Davila”, 88 Mircea Vulcanescu Street, 010825 Bucharest, Romania
3
Department of Marketing and Medical Technology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
4
Department of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(20), 10183; https://doi.org/10.3390/ijms262010183
Submission received: 8 September 2025 / Revised: 16 October 2025 / Accepted: 18 October 2025 / Published: 20 October 2025
(This article belongs to the Special Issue Platelets at the Intersection of Atherosclerosis Processes)

Abstract

Several nutraceuticals demonstrate potential cardiovascular benefits through lipid-lowering, antithrombotic, and vascular protective mechanisms. Omega-3 fatty acids, berberine, garlic, and nattokinase exert favorable metabolic and vascular effects, yet their clinical efficacy depends on formulation, dosage, and patient characteristics and may be limited by bleeding risk or drug interactions. Antioxidant agents such as vitamin C, vitamin E, resveratrol, astaxanthin, and coenzyme Q provide additional vascular protection but can interfere with hemostasis, metabolism, or redox-sensitive pathways. Similarly, ginkgo biloba, ginger, ginseng, and curcumin exhibit anti-inflammatory vascular activity but also increase the risk of bleeding when combined with antithrombotic therapy. Given the variability in evidence and product quality, their use should be individualized, with further large-scale clinical trials needed to establish safety and efficacy.

1. Introduction

The development of atherosclerosis, an essential promoter of cardiovascular diseases and stroke via thrombus formation [1], is supported by the interplay between platelets and endothelial cells [2]. Taking into account that atherosclerosis is considered to be the main cause of mortality on a global scale, its pathogenesis has been a hot research topic for decades [3]. In order to reduce the risk of thrombosis, patients with atherosclerotic cardiovascular disease are often prescribed antiplatelet therapy. Nevertheless, platelet aggregation inhibition can also result in an increased bleeding risk, which is why the clinical approach must consider the benefit of single or dual antiplatelet treatments. The bleeding risk in atherosclerosis can arise from plaque rupture or the presence of intraplaque hemorrhage.
Vascular chronic inflammation, which leads to the formation of atheroma plaque and its rupture, begins in the endothelium of the blood vessels, where cells disposed on a single layer, called endothelial cells (EC), detect signals produced by biologically active substances and transmit them to the vascular wall [4].
The main events that characterize the development of atherosclerosis are as follows: endothelial cells are activated as a result of damage to the glycocalyx, which loses its function as a barrier and increases its permeability for low-density lipoprotein cholesterol (LDC-C); leukocytes, especially monocytes, which are transformed into macrophages, internalize oxidized LDL-C (ox-LDL-C) and become foam cells; and atheroma plaque and the atheromatous nucleus are formed [5]. In atherosclerosis, leukocyte activation induced by platelets leads to vascular inflammation, elevated endothelial permeability and plasma protein accumulation in the interstitium, events promoted by upregulation of intercellular adhesion molecules (ICAM), vascular cell adhesion molecule 1 (VCAM-1) and E-selectin [6]. The resulting EC damage leads to an imbalance between vasodilation and vasoconstriction processes, reactive oxygen species (ROS) formation and cytokine release (IL-1, IL-6, TNF-alpha), decreased NO availability and finally endothelial dysfunction and thrombosis [7,8]. Upon plaque rupture, platelet aggregation is stimulated, and the thrombus can result in ischemia and infarction [9].
P-selectin together with von-Willbrand factor are responsible for the interaction of platelets with activated endothelial cells, leading to platelet activation. Moreover, P-selectin aggravates atherosclerosis by distributing the proinflammatory mediators produced by platelets to the vascular wall [10]. Binding P-selectin present on platelets to an analogue receptor called P-selectin glycoprotein ligand-1 (PSGL-1) results in the formation of leukocyte–platelet aggregates. As a result, leukocyte β2 integrins macrophage antigen-1 (Mac-1) and lymphocyte function-associated antigen-1 (LFA-1) are activated and platelets produce chemokines, including platelet factor 4 (PF4), a vital mediator of atherosclerosis, to enhance the adhesion of monocytes, which will further turn into macrophages and will finally become foam cells by lipid uptake [11]. PF4 directly favors the internalization of ox-LDL-C by macrophages and prevents the destruction of the receptor for LDL-C [12].
Therefore, in atherosclerosis, a significant role is played by platelets, also known as thrombocytes, which interact with ECs, progenitor cells, and leukocytes, playing a significant pro-inflammatory role [13]. In fact, in atherosclerosis, they are considered the connection between plaque formation, thrombosis and inflammation, as well as intervening after plaque rupture [14]; a meta-analysis of 50 randomized clinical trials showed that antiplatelet and anticoagulant therapies increase the bleeding risk [15]. Therefore, platelets are not only responsible for promoting the development of atherosclerotic disease but could also play key roles as therapeutic targets in order to reduce the risk of complications, including decreasing the bleeding risk [16].
Furthermore, in atherosclerosis, neovascularization (vasa vasorum) in the arterial wall is favored by the release of vascular endothelial growth factor (VEGF), cytokines and chemokines from platelets. Plaque immature new blood vessels present abnormal structural and functional properties, with discontinuation in the base membrane and decreased normal junctions between the ECs, and promote plaque instability and intraplaque hemorrhage, usually in the proximity of the necrotic core [17].
However, hemorrhage can also occur, favored by inflammation, which causes endothelial damage of mature blood vessels, mediated by free hemoglobin, which stimulates ROS formation [18]. Previous studies revealed a link between lesion instability and intraplaque hemorrhage [19], which can lead to rupture and cause internal bleeding.
Figure 1 summarizes the atherothrombotic cascade, highlighting how supplements exert anti-atherosclerotic and antiplatelet effects that may overlap and influence bleeding risk.

2. Nutraceuticals and Cardiovascular Health: Anti-Atherosclerotic Potential and Safety Concerns

The use of dietary supplements for cardiovascular disease (CVD) prevention has risen in recent years, often promoted as natural alternatives or adjuncts to lipid-lowering therapies. Nutraceuticals—bioactive compounds from foods or plants—are of particular interest. Agents such as omega-3 fatty acids, plant sterols, soluble fibers, niacin, red yeast rice, and berberine have demonstrated lipid-lowering effects, including reductions in total cholesterol, LDL-C, and triglycerides [20].
However, clinical benefits remain uncertain. Evidence from randomized trials and meta-analyses shows wide variability, influenced by supplement composition, dosage, study design, and patient characteristics [21]. In the United States, supplements are regulated under the FDA’s Dietary Supplement Health and Education Act (DSHEA), which imposes limited requirements for safety, efficacy, and labeling, raising public health concerns [22,23]. Despite this, use remains widespread, often driven by low consumer awareness.
In selected patients, such as those with statin intolerance or seeking integrative approaches, certain supplements may provide incremental benefit, though routine use for CVD prevention is not supported by high-quality evidence [24,25]. Importantly, nutraceuticals should not replace proven therapies.
Some supplements promoted for anti-atherosclerotic effects also possess antiplatelet activity and may increase bleeding risk, particularly in patients receiving anticoagulant or antiplatelet drugs. Examples include omega-3 fatty acids, garlic, ginkgo biloba, ginseng, curcumin, vitamin E, ginger, green tea extract, and high-dose fish oil. Caution is advised in patients on anticoagulation or undergoing surgery.
This review highlights mechanisms by which dietary supplements may exert anti-atherosclerotic effects and identifies those associated with elevated bleeding risk, especially in patients already receiving antithrombotic therapy.

2.1. Anti-Atherosclerotic Supplements Acting as Lipid Lowering Agents

2.1.1. Omega-3 Fatty Acids

Among the most extensively studied nutraceuticals for lipid modulation are omega-3 fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). These long-chain polyunsaturated fatty acids, obtained primarily from marine sources such as fish and krill oil, have consistently demonstrated the ability to reduce serum triglyceride levels by 20–50%, depending on dosage and baseline lipid status [26]. The proposed mechanisms of action include inhibition of hepatic very-low-density lipoprotein (VLDL) synthesis, enhancement of fatty acid β-oxidation, and suppression of hepatic lipogenesis [27,28].
Beyond their lipid-modifying properties, omega-3 fatty acids exert antiplatelet effects primarily through inhibition of thromboxane A2 (TxA2) synthesis and attenuation of TxA2-dependent platelet activation. This occurs through competition between arachidonic acid and EPA for enzymatic pathways involved in thromboxane biosynthesis, as well as through the generation of EPA-derived metabolites, such as resolvins, which interact with TxA2 receptors and modulate their activity [29,30,31,32].
EPA and DHA also appear to influence platelet function by reducing membrane fluidity, which decreases the externalization of procoagulant phospholipids, particularly phosphatidylserine. This alteration leads to impaired calcium influx, thereby attenuating activation of the coagulation cascade and inhibiting platelet aggregation [33,34].
In addition, omega-3 fatty acids are presumed to modulate platelet activation through effects on the glycoprotein VI (GPVI) collagen receptor. Proposed mechanisms include direct interference with GPVI signaling pathways or attenuation of collagen-dependent platelet reactivity via protein kinase A activation [35,36,37].
Atherosclerosis is closely linked to the adiponectin/leptin ratio, an important marker of metabolic and inflammatory balance. In a systematic review, nine studies reported significantly lower leptin and/or higher adiponectin levels following EPA + DHA intake. The doses, supplementation duration, and population characteristics varied across studies. The EPA + DHA dose ranged from 0.52 to 4.2 g/day, with supplementation lasting 4 to 24 weeks [38].
Table 1 highlights key clinical studies from the most recent trials addressing the potential benefits of omega-3 fatty acids in atherosclerosis, including triglyceride reduction, anti-inflammatory effects, improved endothelial function, and plaque stabilization.

2.1.2. Berberine

Berberine, an isoquinoline alkaloid from Coptis chinensis and Berberis species, exhibits broad pharmacological activity with therapeutic potential in cancer, metabolic, cardiovascular, digestive, and neurological diseases [44]. Berberine improves glucose and lipid metabolism by activating AMPK in adipocytes and myocytes, which enhances GLUT4 translocation, boosts lipid oxidation, and inhibits lipid synthesis [45]. Berberine has a lipid-lowering effect by stabilizing LDLR mRNA, thereby increasing LDL receptor levels and enhancing LDL cholesterol clearance from the blood [46]. Additionally, berberine reduces cholesterol levels by inhibiting its intestinal absorption and limiting its release into the bloodstream [47].
In vitro, berberine and its metabolite berberrubine (M2) inhibited ADP-induced platelet activation by blocking Glycoprotein IIb/IIIa activation, reducing P-selectin expression, and limiting fibrinogen binding. Both compounds selectively targeted PI3Kβ, likely through interaction with its active site [48].
Network pharmacology analysis revealed that berberine acts against atherosclerosis through pathways involving the cell cycle, ubiquitin-mediated proteolysis, MAPK, and PI3K-Akt signaling. Overall, these mechanisms indicate that berberine may reduce atherosclerosis by inhibiting inflammation and vascular cell proliferation [49].

2.1.3. Red Yeast Rice

Red yeast rice (RYR) is a fermented product obtained by growing the fungus Monascus purpureus on rice, producing bioactive compounds such as monacolin K and pigments [50]. Similar to traditional statins, monacolins inhibit HMG-CoA reductase resulting in reduction of endogenous cholesterol synthesis and reduced blood cholesterol levels [51]. RYR can lower LDL-C to a degree comparable to low-dose statins and is particularly useful for individuals with mild-to-moderate hypercholesterolemia who are not eligible for statins, as well as for those who are intolerant to statins or prefer nutraceuticals [52]. RYR may also help avoid some of the ‘nocebo’ effects observed with statins [53].
Liu et al. demonstrated that combined nattokinase and red yeast rice supplementation safely enhances cardiometabolic profiles in patients with coronary artery disease, showing stronger reductions in thromboxane B2 and increases in antithrombin III than placebo, suggesting lowered thrombosis risk [54]. Moreover, this year, a double-blind, parallel-controlled trial is being conducted to evaluate the effect of a Natto Red Yeast Rice (NRYR) supplement combined with statins on lipid levels in individuals with dyslipidemia [55].

2.1.4. Garlic

Garlic (Allium sativum) has a long-standing history of medicinal use, including for cardiovascular protection. Its active compounds—particularly allicin and S-allyl cysteine—have lipid-lowering, anti-inflammatory, and antioxidative effects [56].
Garlic’s lipid-lowering effects are multifactorial, involving inhibition of hepatic cholesterol synthesis—potentially through suppression of HMG-CoA reductase—enhancement of biliary excretion of cholesterol and bile acids, and antioxidant activity that reduces LDL oxidation and atherosclerotic plaque formation [57].
An early systematic review reported an average 12% reduction in total cholesterol, supporting garlic’s modest but consistent lipid-lowering effect after more than four weeks of supplementation [58]. A meta-analysis of 14 studies (1981–2016) further confirmed significant decreases in total cholesterol and LDL-C, though effects on HDL and triglycerides were not significant [57]. A recent 2024 meta-analysis by Du et al., including 21 randomized controlled trials, found that garlic significantly reduced total cholesterol, LDL-C, and triglycerides, with a slight increase in HDL-C [59]. The observed heterogeneity in study outcomes was likely influenced by differences in garlic formulation, dosage, treatment duration, and participant characteristics, as well as methodological limitations such as the absence of subgroup analyses [57,59]. Thus, greater lipid-lowering effects were observed in individuals with high baseline cholesterol, particularly with aged garlic extract, which also improved oxidative stress markers [60].
Garlic also modulates gut microbiota composition, which may indirectly influence lipid metabolism and bile acid homeostasis [61].
Garlic extract and other phytochemicals may regulate lipid homeostasis by modulating the mevalonate pathway, which is key to cholesterol metabolism and tumorigenesis, offering potential benefits for dyslipidemia, obesity-related complications, and cancer [62].
Allicin improves metabolic function in diabetic rats by lowering blood glucose and lipids, reducing liver fat, modulating gut microbiota and bile acids, increasing intestinal GLP-1, suppressing FGF15, and upregulating hepatic CYP7A1 [61].
Garlic inhibits platelet aggregation by suppressing cyclooxygenase activity and thromboxane A2 synthesis, reducing intraplatelet Ca2+, increasing cAMP and cGMP, and decreasing fibrinogen binding and platelet shape change [63].
In addition, its antioxidant properties and stimulation of nitric oxide synthase (NOS) enhance platelet-derived NO production. Garlic compounds may also directly interfere with GPIIb/IIIa receptors, reducing platelet–fibrinogen interactions [60]. Garlic exerts anti-inflammatory effects by modulating cytokine production and improving endothelial function [40].

2.1.5. Nattokinase

Natto is a traditional Japanese food produced by fermenting soybeans with Bacillus natto. This fermentation generates several bioactive compounds, among which nattokinase is the most studied. Nattokinase (NK) is a serine protease known for its enzymatic stability and resistance to freeze–thaw cycles. It contributes to lipid regulation and anti-atherosclerotic activity by stimulating hormone-sensitive lipase, suppressing HMG-CoA reductase, and promoting lipoprotein lipase function [64].
By combining anti-inflammatory, antioxidant, hemorheological, and thrombolytic actions with plaque stabilization and endothelial protection, nattokinase emerges as a promising cardioprotective agent. Through modulation of TLR-4/JAK-STAT and TRAF-6/NF-κB-MAPK signaling, nattokinase decreases IL-6 mediated inflammation and ROS-driven caspase activation, ultimately reducing apoptosis in vascular and myocardial tissue [65]. Nattokinase supplementation has no impact on the progression of subclinical atherosclerosis in healthy, low–cardiovascular-risk individuals [66].
Nattokinase not only degrades fibrin directly but also enhances fibrinolysis by increasing tissue plasminogen activator (tPA) release and inactivating PAI-1 [67].
It facilitates the conversion of prourokinase to urokinase, further promoting clot lysis [68]. In addition, NK suppresses thromboxane formation, thereby inhibiting platelet aggregation without inducing bleeding [69]. Human studies have shown reductions in fibrinogen, factor VII, and factor VIII, as well as shortened euglobulin lysis time and increased fibrin/fibrinogen degradation products, indicating potent anticoagulant and fibrinolytic effects [70].
In a clinical trial involving 113 patients with dyslipidemia, participants were randomly assigned to receive either nattokinase-monascus supplementation or placebo. Treatment with nattokinase-monascus supplementation significantly reduced total cholesterol and low-density lipoprotein cholesterol, while no significant effects on coagulation parameters were observed [71].

2.2. Nutraceutical and Botanical Bioactives That Inhibit LDL Oxidation

The oxidative modification of LDL into its atherogenic form is widely recognized as a critical event in the initiation and progression of atherosclerosis [8]. Oxidized LDL (oxLDL) disrupts endothelial homeostasis, intensifies pro-inflammatory signaling, and accelerates foam cell formation within the arterial wall [72]. A range of bioactive molecules—including plant-derived antioxidants and lipid-phase cofactors such as coenzyme Q10—have been investigated for their ability to counteract this process by directly scavenging reactive species or by strengthening endogenous redox defenses. When optimized in terms of formulation and bioavailability, these agents may offer complementary vascular protection, though potential pharmacokinetic interactions with conventional cardiovascular therapies must be considered [73].

2.2.1. Astaxanthin

Astaxanthin is a lipid-soluble xanthophyll carotenoid with a high affinity for cellular membranes, where it integrates into lipoproteins and phospholipid bilayers to protect polyunsaturated fatty acids from oxidative degradation. This spatial localization enables efficient quenching of singlet oxygen and lipid radicals, thereby terminating the propagation phase of lipid peroxidation [74]. Clinical investigations—particularly in individuals with impaired glucose metabolism and elevated oxidative stress—have reported enhanced LDL oxidative stability and reductions in lipid peroxidation markers such as malondialdehyde following supplementation with astaxanthin at 12 mg/day for 12 weeks [75,76,77]. Although no clinically significant drug–nutrient interactions have been established, the strong redox activity of astaxanthin suggests a theoretical potential to influence the pharmacokinetics of redox-sensitive agents, a hypothesis warranting further mechanistic investigation [78]. Small randomized studies indicate improvements in oxidative stress markers and LDL stability; however, multicenter outcome trials confirming cardiovascular benefits are still lacking [79,80,81].

2.2.2. Resveratrol

Resveratrol, a stilbenoid polyphenol found abundantly in red grapes and Polygonum cuspidatum, supports vascular health by reducing oxidative stress and enhancing endogenous antioxidant defenses [82]. It inhibits NADPH oxidase and stimulates antioxidant enzymes such as superoxide dismutase and catalase, thereby mitigating the oxidative modifications that render LDL particles atherogenic [83,84]. In addition, resveratrol modulates lipid homeostasis by activating the EGFR–ERK signaling pathway, leading to upregulation of hepatic LDL receptor expression and improved clearance of circulating LDL, which may slow the development of early atheromatous plaques [85]. Preclinical studies suggest potential synergy with statins through enhanced endothelial repair and accelerated vascular healing following injury [86]. Mechanistic investigations further demonstrate that resveratrol restores autophagic flux in endothelial cells exposed to oxidized LDL by upregulating SIRT1, promoting lysosomal degradation pathways, and reducing oxLDL accumulation, underscoring a reparative role that extends beyond its classical antioxidant activity [87].
Small randomized trials and meta-analyses in humans report reductions in inflammatory biomarkers (e.g., CRP, TNF-α) and improvements in endothelium-dependent vasodilation assessed by flow-mediated dilation (FMD), albeit with heterogeneous effects across studies; however, large multicenter outcome trials showing reductions in major adverse cardiovascular events are lacking [88,89,90,91].

2.2.3. Coenzyme Q10 (CoQ10)

Coenzyme Q10 (Ubiquinone/Ubiquinol) functions both as an essential cofactor in the mitochondrial electron transport chain and as a lipid-phase antioxidant that protects cellular and lipoprotein membranes. By integrating into LDL particles, CoQ10 helps preserve structural integrity under oxidative stress, thereby limiting the formation of atherogenic oxidized LDL (oxLDL). Clinically, supplementation at doses of 200–400 mg/day has been associated with reductions in circulating oxLDL and improvements in endothelium-dependent vasodilation, as assessed by flow-mediated dilation (FMD), particularly among statin-treated patients [92]. This is mechanistically relevant because statins inhibit the mevalonate pathway, reducing endogenous CoQ10 synthesis and potentially aggravating mitochondrial dysfunction [93]. Restoring CoQ10 levels is therefore a rational adjunctive strategy to mitigate statin-associated oxidative and bioenergetic deficits. Recent meta-analyses support these findings, reporting significant improvements in FMD with CoQ10 supplementation, consistent with enhanced endothelial function [94,95]. In a randomized controlled trial among statin users, CoQ10 improved mitochondrial function and antioxidant capacity, although effects on muscle symptoms were variable [96]. Overall, CoQ10 exhibits a favorable safety profile when used at customary doses.
Meta-analyses indicate modest improvements in endothelium-dependent vasodilation assessed by flow-mediated dilation (FMD) and oxidative biomarkers—particularly in statin-treated populations—yet robust atherosclerotic outcome data are limited. A multicenter RCT in chronic heart failure (Q-SYMBIO) reported reductions in major adverse cardiovascular events with adjunctive CoQ10, but its population and endpoints differ from atherosclerosis prevention [94,96,97,98,99]. In elderly with low selenium status, combined selenium + CoQ10 reduced cardiovascular mortality in the randomized KiSel-10 trial and its long-term follow-ups; however, effects cannot be attributed to CoQ10 alone [100,101,102]. A recent RCT in MASLD reported improvements in vascular and myocardial function with high-dose CoQ10, extending evidence to a metabolic-risk cohort, though without hard CV endpoints [103].

2.2.4. Vitamins C and E

Vitamins C and E act synergistically to limit oxidative vascular injury through complementary mechanisms. Vitamin E refers to a family of lipid-soluble tocopherols and tocotrienols that incorporate into lipoproteins and cellular membranes, where α-tocopherol terminates lipid peroxidation chain reactions and reduces LDL susceptibility to oxidative modification [104]. Tocotrienols may confer additional benefits: in a 2025 randomized trial, a tocotrienol-rich formulation (300 mg/day) significantly lowered LDL-C and C-reactive protein compared with α-tocopherol–dominant preparations [105]. The inter-vitamin recycling mechanism—where ascorbate regenerates oxidized α-tocopherol—underlies the observed synergy during co-supplementation, yielding up to ~40% greater reductions in lipid peroxidation compared with either vitamin alone [106]. Clinical studies employing approximately 1 g/day of vitamin C have reported reductions in inflammatory biomarkers (CRP, IL-6), decreases in lipid peroxidation markers (e.g., malondialdehyde), and improvements in flow-mediated dilation (FMD) among patients with diabetes or metabolic syndrome [107].
Across randomized trials and meta-analyses, vitamins C and E yield heterogeneous effects on inflammatory and oxidative stress biomarkers and modest improvements in endothelium-dependent vasodilation assessed by flow-mediated dilation (FMD); however, large multicenter trials have not consistently demonstrated reductions in major cardiovascular events with high-dose vitamin C or E [108,109,110,111]. High-dose vitamin E (≥400 IU/day) has also been associated with increased all-cause mortality in a dose–response meta-analysis [112,113].

2.3. Supplements with Anti-Atherosclerotic Effects That Reduce Vascular Inflammation

2.3.1. Ginkgo Biloba

Ginkgo biloba (Maidenhair tree) is an ancient, long-lived species valued for its distinctive fan-shaped leaves and medicinal properties. EGb 761, a standardized extract of Ginkgo biloba leaves, is widely used in phytotherapy and clinical research. Its principal constituents—ginkgolide B and flavonoids—upregulate endothelial nitric oxide synthase (eNOS) expression and promote Akt-dependent eNOS phosphorylation, thereby enhancing nitric oxide (NO) release and vasorelaxation [114]. At the molecular level, ginkgolide B mitigates endothelial oxidative stress by downregulating LOX-1 (lectin-like oxidized LDL receptor 1) and NOX4 (NADPH oxidase 4), leading to reduced reactive oxygen species (ROS) generation and decreased expression of vascular adhesion molecules (ICAM-1, VCAM-1, MCP-1) [115]. In vitro, Ginkgo biloba extract suppresses cytokine-induced endothelial adhesiveness by upregulating heme oxygenase-1 (HO-1) through activation of the p38 and Nrf2 signaling pathways. In vivo, it reduces leukocyte adhesion to injured arteries and increases HO-1 expression in monocytes and arterial tissue following wire-induced injury [116]. Furthermore, EGb 761 activates HO-1 in endothelial progenitor cells, supporting re-endothelialization and vascular repair [117].
Human evidence derives mainly from small, short-duration randomized trials with standardized EGb 761 (typically 120–240 mg/day over 4–24 weeks). These studies show modest, inconsistent improvements in endothelium-dependent vasodilation assessed by FMD and microvascular perfusion surrogates, with neutral or variable effects on lipids, inflammation, or blood pressure; trials are heterogeneous in extract, dose, and endpoints and are underpowered for clinical events. Crucially, no large multicenter RCT has demonstrated reductions in myocardial infarction, stroke, or cardiovascular death: in the 3069-participant GEM program (EGb 761, 120 mg twice daily), secondary analyses found no reduction in incident CVD or CVD mortality [118,119,120,121]. Recent clinical work continues to focus on surrogate and inflammatory/oxidative markers rather than hard cardiovascular outcomes (e.g., randomized/open-label EGb 761 in mild cognitive impairment reporting changes in circulating inflammation/oxidative stress biomarkers). These newer trials reinforce the surrogate-heavy and underpowered nature of the literature [122,123].

2.3.2. Ginger

Ginger (Zingiber officinale) is a widely used medicinal and culinary plant, valued for its bioactive phenolic constituents—6-gingerol and 6-shogaol—which exhibit potent anti-inflammatory and antioxidant properties. These compounds attenuate systemic inflammation by lowering circulating levels of C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α), biomarkers consistently associated with endothelial dysfunction [124]. At the cellular level, 6-gingerol suppresses reactive oxygen species (ROS) production and restores redox homeostasis in endothelial cells exposed to oxidative stress, in part through activation of the Nrf2/HO-1 signaling pathway [125].
Meta-analyses of randomized trials report that ginger supplementation (typically 1–3 g/day for 8–12 weeks) significantly reduces hs-CRP and TNF-α across diverse adult populations, with inconsistent effects on IL-6 [126]. In metabolic-risk cohorts (e.g., metabolic syndrome, type 2 diabetes), ginger has been associated with lower triglycerides, improved fasting glycemia/HbA1c, and enhanced systemic antioxidant status, while effects on blood pressure or HDL/LDL cholesterol are variable [127]. Collectively, these changes are compatible with improved endothelial milieu and vasodilator responsiveness. The clinical literature is dominated by small RCTs and meta-analyses of surrogate outcomes (inflammation, oxidative stress, glycemic indices, lipids). While signals are directionally favorable for hs-CRP/TNF-α and selected metabolic parameters, large multicenter outcome trials showing reductions in myocardial infarction, stroke, or cardiovascular death are not yet available [126,128,129]. Most RCTs used powdered/extract ginger at 1–3 g/day for 8–12 weeks [129].

2.3.3. Ginseng

Asian ginseng (Panax ginseng) is a medicinal plant traditionally used in East Asia, whose principal saponins—ginsenosides (e.g., Rg3, Rb1, Rd, Rp1, Rp3)—exert adaptogenic and immunomodulatory effects [130,131]. Ginsenosides activate PI3K/Akt–AMPK–eNOS signaling, enhance nitric oxide (NO) production, and suppress endothelin-1 expression, thereby promoting vasorelaxation. Antioxidant defenses are strengthened via Nrf2/HO-1 induction, while NF-κB signaling and endothelial adhesion molecules (ICAM-1, VCAM-1) are downregulated in experimental models [132]. Ginseng also modulates platelet-related pathways (e.g., GPVI–PLCγ2, PI3K/Akt, MAPKs), which may contribute to reduced thrombo-inflammatory activation and improved plaque stability [133]. Small randomized trials have reported modest improvements in endothelium-dependent vasodilation, as assessed by flow-mediated dilation (FMD) and other vascular surrogates, with heterogeneous effects on blood pressure and lipid profiles. Examples include acute FMD enhancement after administration of 3 g Korean red ginseng in healthy adults and variable findings among individuals with prehypertension or metabolic risk [134,135,136]. Clinical literature largely comprises small, short-duration randomized controlled trials (RCTs) employing surrogate endpoints; results vary according to preparation and dose, and most studies are underpowered for major cardiovascular outcomes. Meta-analyses focusing on vascular surrogates suggest improvements in FMD; however, no large multicenter RCTs have demonstrated reductions in myocardial infarction, stroke, or cardiovascular mortality [133,137,138].

2.3.4. Curcumin

Curcumin/turmeric (Curcuma longa) is a medicinal and culinary plant whose rhizome contains curcumin, a polyphenol with potent anti-inflammatory and antioxidant properties. At the endothelial level, curcumin suppresses NF-κB activity, thereby reducing the expression of adhesion molecules (ICAM-1, VCAM-1) and limiting cytokine release (IL-6, TNF-α) [139]. Concurrently, it activates Nrf2-dependent antioxidant pathways, enhancing heme oxygenase-1 (HO-1) and other cytoprotective enzymes to restore redox balance under oxidative stress [140]. Clinical findings broadly support these mechanisms. An umbrella meta-analysis of randomized trials (2024) reported modest but significant vascular improvements with curcumin supplementation, including reductions in diastolic blood pressure (−0.94 mmHg), pulse wave velocity, and circulating VCAM-1, along with an approximate 1.6% increase in flow-mediated dilation (FMD) [141]. Benefits were more consistent in studies employing optimized formulations (e.g., nanoparticles, phytosomes, phospholipid complexes) designed to overcome curcumin’s poor oral bioavailability [142,143].
Across small-to-moderate randomized controlled trials (RCTs) and meta-analyses, curcumin demonstrates modest improvements in endothelial surrogates—most consistently in endothelium-dependent vasodilation assessed by FMD, arterial stiffness (PWV), and reductions in VCAM-1—accompanied by favorable changes in inflammatory and oxidative biomarkers. However, large multicenter outcome trials confirming reductions in myocardial infarction, stroke, or cardiovascular mortality remain unavailable [141,144,145,146].

3. Hemostatic Safety of Nutraceutical and Botanical Bioactives: Bleeding Risks and Drug–Drug Interactions

Nutraceutical and botanical use is common yet often under-disclosed, with perioperative societies advising discontinuation 1–2 weeks before elective procedures; formulation variability and limited standardization further complicate risk appraisal [147]. This section synthesizes hemostatic safety signals across three domains: (1) pharmacodynamic antiplatelet and fibrinolytic effects (e.g., Ginkgo biloba, ginger, curcumin, resveratrol, omega-3 fatty acids, garlic); (2) micronutrient-related effects on coagulation, particularly with high-dose vitamin E; and (3) pharmacokinetic interactions influencing warfarin or direct oral anticoagulant (DOAC) exposure [148].

3.1. Pharmacodynamic (Antiplatelet/Fibrinolytic) Effects

Ginkgo biloba standardized extracts (EGb 761) contain terpene trilactones—most notably ginkgolide B—which act as potent antagonists of the platelet-activating factor (PAF) receptor, a well-established antiplatelet mechanism demonstrated in biochemical and platelet models [149,150]. Authoritative safety sources therefore caution against concurrent use of ginkgo with aspirin, clopidogrel, nonsteroidal anti-inflammatory drugs (NSAIDs), or warfarin due to the potential for additive antiplatelet effects and altered coagulation parameters [151]. In large clinical datasets, concomitant use with vitamin K antagonists has been associated with an increased risk of major bleeding—for example, a chart review of approximately 807,399 patients found higher rates of major bleeding with ginkgo plus warfarin compared with warfarin alone [152], while a Veterans Administration electronic health record cohort reported a 38% higher hazard of bleeding with co-prescribed ginkgo (HR 1.38, 95% CI 1.20–1.58) [153]. Beyond vitamin K antagonists, a 2025 hospital-based retrospective analysis identified that ginkgo interactions most frequently involved aspirin or clopidogrel and were associated with abnormal coagulation tests and a small but significant increase in clinical bleeding events [154]. Complementing these observations, case reports have described intracranial and ocular hemorrhages temporally associated with ginkgo use, typically in patients receiving concurrent antithrombotic therapy [155].
Ginger’s principal phenolics (6-gingerol and 6-shogaol) inhibit platelet thromboxane synthesis and attenuate cyclooxygenase-1 (COX-1)-dependent aggregation, with human and ex vivo evidence showing reduced platelet reactivity [156,157]. Clinically, concurrent warfarin and ginger use has been linked to altered anticoagulation and bleeding in case reports, including supratherapeutic INR and prolonged bleeding, suggesting a plausible pharmacodynamic interaction [158]. In a prospective cohort of warfarin users, ginger intake was independently associated with a higher risk of self-reported bleeding [159]. However, expert reviews note that at modest doses, ginger (and ginkgo) does not consistently affect warfarin activity, highlighting dose, preparation, and patient context as key modifiers and supporting a cautious, individualized approach [160]. In clinical practice, drug-interaction compendia and perioperative guidelines advise avoiding concomitant use with antiplatelet or anticoagulant agents when feasible and discontinuing ginger 1–2 weeks before elective procedures, given the potential for additive bleeding risk [161].
Curcumin’s principal actions on platelets include suppression of thromboxane-dependent aggregation and inhibition of downstream signaling (Akt/MAPK/Src) with blockade of dense-granule secretion—mechanisms demonstrated in human/ex vivo models [162,163]. Clinically, the bleeding signal emerges largely in the setting of concomitant antithrombotic therapy, with pharmacovigilance alerts and case reports describing supratherapeutic INR and hemorrhagic events when turmeric/curcumin is used alongside warfarin or fluindione [164,165]. Randomized data quantifying bleeding outcomes are lacking—the evidence base is predominantly mechanistic, case-based, or observational—which underpins conservative perioperative and cardiovascular guidance [166]. In practice, co-administration with antiplatelet agents or warfarin warrants caution, and theoretical DOAC interactions remain a concern given curcumin’s inhibition of P-glycoprotein and reported modulation of CYP3A4, pathways central to apixaban/rivaroxaban disposition [167,168]. Consistent with multi-society recommendations on herbal products, discontinuation of curcumin-containing supplements 1–2 weeks before elective procedures is reasonable to minimize additive antithrombotic effects and simplify management [169].
Resveratrol demonstrates antiplatelet activity in human/ex vivo models, attenuating thromboxane A2–dependent platelet activation and aggregation and reducing platelet activation markers at concentrations achievable with moderate dietary exposure [170,171]. Clinically, bleeding events attributable to resveratrol are sparsely documented, but precaution is warranted: preclinical work shows potentiation of warfarin’s anticoagulant effect, and contemporary clinical reviews flag a plausible bleeding risk—particularly in patients receiving antithrombotic therapy [172,173]. Beyond pharmacodynamics, resveratrol modulates drug disposition pathways—in healthy volunteers, high-dose resveratrol inhibited CYP3A4 activity, and multiple studies indicate interactions with P-glycoprotein—supporting a theoretical interaction with DOACs and other CYP3A4/P-gp substrates even though clinical confirmation is limited [174,175,176]. In perioperative care, expert consensus favors temporary discontinuation 1–2 weeks before elective procedures when used alongside antiplatelet/anticoagulant therapy, given uncertain benefit and potential additive bleeding risk [147,169].
Omega-3 PUFA consumption decreases arachidonic acid- and thromboxane-derived metabolites from omega-6 pathways, promoting anti-inflammatory and antithrombotic effects. This shift reduces platelet activation and clot formation potential, which may slightly increase bleeding tendency. However, a systematic review and meta-analysis of randomized clinical trials found no association between omega-3 PUFA intake and an increased risk of overall, hemorrhagic, intracranial, or gastrointestinal bleeding. In contrast, high-dose purified EPA was linked to a modest, dose-dependent increase in bleeding risk, independent of antiplatelet therapy [177]. Similarly, another systematic review and meta-analysis reported that EPA monotherapy, compared with control, was associated with a higher risk of total bleeding and atrial fibrillation [178]. The results of clinical studies on adverse events associated with omega-3 fatty acid supplements are summarized in Table 1. Also, a systematic review of randomized controlled trials conducted between 1987 and 2023 found that individuals taking omega-3 polyunsaturated fatty acids (PUFAs) were more likely to experience minor adverse effects, including diarrhea, taste disturbances, and a tendency to bleed, while reporting less back pain. Importantly, no serious adverse events directly linked to omega-3 PUFAs were observed, emphasizing the need for comprehensive monitoring to detect subtle side effects [179].
Another systematic review relevant for surgical practice found no evidence to support discontinuing fish oil supplements prior to surgery or other invasive procedures [180]. Moreover, omega-3 PUFAs have demonstrated benefits in liver surgery and acute respiratory distress syndrome, suggesting potential value in perioperative care for trauma patients by reducing hospital and intensive care unit stays [181].
However, omega-3 use in early pregnancy was associated with a higher risk of postpartum hemorrhage, supporting recommendations to discontinue supplementation in late pregnancy [182]. Despite its benefits, omega-3 supplementation can increase bleeding risk by inhibiting platelet function, particularly in patients on anticoagulants, as illustrated by an elderly patient on warfarin and fish oil whose coagulopathy was uncorrectable after blunt head trauma [183].
Garlic extract offers a safe and modestly effective adjunct for lipid management, particularly in individuals with mild hyperlipidemia or those intolerant to statins. While it is not a substitute for statin therapy in high-risk patients, its lipid-lowering, antioxidant, and potential anti-atherogenic effects justify its inclusion in dietary and lifestyle-based approaches to cardiovascular risk reduction [184].
Garlic inhibits platelet aggregation by suppressing cyclooxygenase activity and thromboxane A2 synthesis, reducing intraplatelet Ca2+, increasing cAMP and cGMP, and decreasing fibrinogen binding and platelet shape change [63]. Garlic compounds may also directly interfere with GPIIb/IIIa receptors, reducing platelet–fibrinogen interactions [60].
Importantly, garlic may exert mild antiplatelet effects, which can increase bleeding risk and may be clinically relevant in patients receiving anticoagulant or antiplatelet therapy. But overall, the use of garlic appeared to be safe among individuals on warfarin [185,186].
Nattokinase was shown to be capable of blocking thromboxane formation resulting in an inhibition of platelet aggregation without producing the side effect of bleeding [69].
Thus, nattokinase shows promise for cardiovascular and thrombotic disorders, offering antithrombotic, antihypertensive, antiatherosclerotic, and neuroprotective benefits with a strong safety profile. But, short-term, low-dose nattokinase may not significantly reduce lipid levels, but it shows potential as a supportive therapy for managing hypertension [187].
In a clinical trial involving 113 patients with dyslipidemia, participants were randomly assigned to receive either nattokinase-monascus supplementation or placebo. Treatment with nattokinase-monascus supplementation significantly reduced total cholesterol and low-density lipoprotein cholesterol, while no significant effects on coagulation parameters were observed [71].
Recent studies have demonstrated that NK exerts therapeutic effects against atherosclerotic pathology by modulating the “Lipid and Atherosclerosis” pathway. High-dose NK treatment (900 FU/kg body weight) for six weeks significantly reduced atherosclerotic lesions in ApoE−/− mice. Liver proteomic analysis revealed downregulation of PXDN 9 (peroxidasin) and PNLIP (pancreatic lipase)—proteins associated with lipid oxidation and absorption—leading to enhanced lipid metabolism, reduced lipid peroxidation, and overall attenuation of atherosclerosis. In the same study, the researchers also showed that NK exhibited a dose-dependent thrombolytic effect on both fresh and aged blood clots, achieving a 30–40% thrombolysis rate at a concentration of 5000 μg/mL [188].
Currently, clinical data on the potential risks of long-term nattokinase use—such as bleeding tendency and liver burden—are limited. Although human trials show encouraging results, further research on its pharmacokinetics and drug interactions is needed before it can be considered a substitute for conventional cardiovascular medications. medications [189].
Recent studies suggest that nattokinase, as a functional food ingredient, may benefit aging-related diseases without significant side effects [190].

3.2. Micronutrient-Related Coagulation Effects

Vitamin E at pharmacologic doses (≥300–400 IU/day) can antagonize vitamin K-dependent coagulation through CYP4F2-mediated pathways and may also inhibit platelet aggregation [191,192]. In anticoagulated patients, higher serum α-tocopherol concentrations have been associated with increased bleeding risk during warfarin or acenocoumarol therapy, and recent case reports describe reversible coagulopathy precipitated by excessive vitamin E intake [193,194]. Consequently, perioperative and anesthesia guidelines recommend avoiding high-dose vitamin E in patients receiving anticoagulants and discontinuing supplementation 1–2 weeks prior to elective procedures [169,195].
Unlike vitamin E, vitamin C has no consistent clinical bleeding signal; a controlled study of high-dose oral vitamin C (500 mg twice daily, 14 days) found no overall significant effect on CYP3A4 activity (a modest induction was seen only in men), and contemporary clinical meta-analyses have not linked vitamin C to excess hemorrhage [196,197].

3.3. Pharmacokinetic Interactions with Antithrombotics

Asian ginseng, with its diverse ginsenoside profile, poses a potential pharmacokinetic interaction with antithrombotic therapy, primarily through attenuation of warfarin’s anticoagulant effect. In a randomized, double-blind, placebo-controlled trial in healthy volunteers, American ginseng significantly reduced warfarin’s anticoagulant response—reflected by lower INR and diminished pharmacodynamic effect—consistent with increased drug clearance [198]. Clinical probe studies demonstrate that Asian ginseng induces CYP3A activity, reducing oral midazolam exposure with minimal influence on P-glycoprotein (P-gp), a mechanism expected to preferentially decrease R-warfarin exposure and thereby blunt overall anticoagulation [199]. Contemporary reviews corroborate the directionality of this interaction, while noting heterogeneity across species and extract types, and recommend close INR monitoring or avoidance when ginseng is initiated or discontinued in patients receiving vitamin K antagonists [200,201]. For DOACs, direct clinical data are lacking, but potential CYP3A4/P-gp modulation implies a theoretical risk of reduced exposure (notably apixaban/rivaroxaban), warranting caution in high-risk settings and around elective procedures, where many perioperative sources advise discontinuation ~1–2 weeks pre-operatory [200].
Coenzyme Q10 (ubiquinone) is structurally related to vitamin K, and multiple clinical reports/interaction databases describe reduced warfarin anticoagulation with INR returning to baseline after CoQ10 discontinuation, consistent with a vitamin-K–like antagonism [202]. Mechanistically and clinically, the principal risk is therefore subtherapeutic anticoagulation (↓INR) rather than bleeding, warranting caution when CoQ10 is added to or withdrawn from stable warfarin therapy [186]. Notably, the only randomized, double-blind, placebo-controlled crossover trial in 24 stable VKA-treated outpatients (CoQ10 100 mg/day for 4 weeks) found no significant change in INR or warfarin dose versus placebo—highlighting heterogeneity and dose/preparation effects across the literature [203]. Contemporary references therefore recommend closer INR monitoring or avoidance when initiating or stopping CoQ10 in patients on VKAs [202]. For DOACs, there is no robust clinical data showing a meaningful interaction with CoQ10; given the lack of a clear CYP3A4/P-gp mechanism, any effect is theoretical, so practice should default to clinical vigilance rather than a blanket contraindication [160].
At pharmacologic doses, resveratrol modulates human drug-metabolizing enzymes: in a randomized crossover study of healthy volunteers given 1 g/day for 4 weeks, resveratrol inhibited CYP3A4, CYP2D6 and CYP2C9 activity (phenotypic probe cocktail), a profile that could increase exposure to CYP3A4-metabolized antithrombotics and co-medications [174]. Preclinical work further suggests potentiation of warfarin anticoagulation at higher resveratrol concentrations, underscoring a plausible PK/PD interaction even though definitive clinical bleeding trials are lacking [172]. In parallel, curcumin demonstrably modulates intestinal transport and metabolism: contemporary mechanistic studies show P-glycoprotein inhibition and interaction with CYP3A4, while a human PK trial found oral curcumin reduced bioavailability of a P-gp/CYP3A4 probe substrate via apparent CYP3A4 activation, highlighting formulation- and context-dependent effects [173,204]. Regulatory and clinical safety sources flag turmeric/curcumin as potential contributors to excess anticoagulation/bleeding when combined with warfarin, with case reports of INR elevation and hemorrhage and professional compendia recommending caution [171]. For DOACs, direct clinical data are sparse, but expert reviews note theoretical risk of altered exposure where CYP3A4/P-gp pathways are relevant (e.g., apixaban/rivaroxaban), justifying conservative perioperative practice and careful co-medication review rather than blanket prohibition [205].
Berberine is a multifunctional compound with potential benefits in a variety of conditions, including cardiovascular disease, type 2 diabetes mellitus, gastrointestinal disorders, polycystic ovary syndrome, nonalcoholic fatty liver disease, hyperlipidemia, metabolic syndrome, obesity, and schizophrenia. According to the latest evidence—including the first systematic review on berberine published this year—this compound appears to support cardiovascular health by modulating gut microbiota and regulating energy metabolism through the AMPK–PGC1α pathway [206]. Also, recent studies suggest that berberine may help treat atherosclerosis by inhibiting both the proliferation and apoptosis of vascular smooth muscle cells under mechanical stretch, as well as exerting regulatory effects on platelets [207,208].
Paul et al. reported that berberine inhibits platelet aggregation and superoxide production by modulating aldose reductase, NADPH oxidase, and glutathione reductase. Berberine also decreases calcium release, ERK activation, secretion of α- and dense granules, and platelet adhesion. By inhibiting the p38–p53 pathway, it prevents Bax activation, mitochondrial dysfunction, and platelet apoptosis under high-glucose conditions, suggesting potential vascular benefits in diabetes mellitus [207].
Additionally, berberine exhibits anticoagulant and thrombolytic effects by stimulating antithrombin III, lowering plasminogen activator inhibitor-1 (PAI-1), enhancing urokinase and streptokinase activity, and inhibiting platelet aggregation induced by ADP, thrombin, collagen, or arachidonic acid. Experimental studies show that berberine reduces hypercoagulation induced by a high-fat diet and suppresses thrombus formation, underscoring its therapeutic potential against thrombosis in metabolic diseases [209]. In vivo, oral administration of berberine effectively suppressed platelet activation and carrageenan-induced thrombosis in mice, without prolonging bleeding time [48].
In a recent systematic review and meta-analysis of randomized clinical trials, berberine monotherapy lowers blood lipids and glucose, improves insulin resistance, and shows stronger effects after more than three months of treatment, proving beneficial for older adults with metabolic disorders. Berberine is considered clinically safe and well-tolerated in humans, with few reported adverse effects and no observed impact on participants’ dietary habits [210].
In an umbrella review on berberine and health outcomes, potential side effects identified include constipation, diarrhea, abdominal bloating, and a bitter taste [211].
Several clinical trials (NCT01697735) on berberine 500 mg, administered twice daily as monotherapy or in combination with statins, are ongoing or recently completed. However, the number of participants remains small, and the results have not yet been made publicly [212].
Berberine hydrochloride (produced by the National Institute for the Control of Pharmaceutical and Biological Products) competes to bind warfarin-binding plasma proteins. So, Berberine can displace warfarin from plasma proteins, increasing its free concentration and bleeding risk [213].
Red Yeast Rice (RYR) has been shown in studies to potentially improve outcomes after major surgeries, being associated with fewer postoperative complications, lower mortality rates, and reduced hospital stay and costs [214]. However, a single case reported by Mazzanti et al. indicated a possible interaction between RYR supplementation and an anticoagulant, although treatment was not discontinued in that instance to confirm causality [215].
RYR extracts with hepatotoxic properties may form during maturation. Additionally, if poorly manufactured, red yeast rice can contain citrinin, a mycotoxin with hepatotoxic and nephrotoxic potential [216].
Because monacolin K acts like a statin, it carries risks of drug–drug interactions, especially with CYP3A4 inhibitors, leading to possible serious side effects [217].
Although RYR extract is generally considered safe and has not been linked to life-threatening or frequent adverse events or an increased risk of musculoskeletal disorders its safety remains debated [52,218]. The EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) recently reassessed the safety of monacolins from RYR under EU scrutiny (Regulation (EC) No 1925/2006, Part C, Annex III). Based on updated analytical, toxicological, and clinical evidence, the Panel reaffirmed earlier concerns (EFSA ANS Panel, 2018) that exposure to monacolin K—even at doses as low as 3 mg/day—may lead to serious hepatic and musculoskeletal adverse effects, including rhabdomyolysis. Therefore, the safety of RYR supplements remains inconclusive and continues to be the subject of scientific debate [219].

3.4. Agents with Limited or No Consistent Clinical Bleeding Signal

Astaxanthin has not been associated with a consistent clinical bleeding signal in human trials, and regulatory safety evaluations deem typical intakes well tolerated; nonetheless, caution is advisable in patients receiving anticoagulant or antiplatelet therapy [220].
Table 2 summarizes the multi-mechanistic actions of selected anti-atherosclerotic nutraceuticals—highlighting hypolipidemic, vasodilatory, and anti-inflammatory pathways alongside fibrinolytic, antiplatelet, and anticoagulant effects.

3.5. Extracellular Vesicles at the Thrombosis–Bleeding Interface

In atherosclerosis, extracellular vesicles (EVs) released by endothelial cells, platelets, smooth-muscle cells, and macrophages act as key mediators of intercellular communication. By exposing phosphatidylserine and tissue factor, and carrying adhesion molecules, lipids, and regulatory RNAs, they amplify vascular inflammation, leukocyte recruitment, and thrombin generation, contributing to plaque progression and rupture. Conversely, distinct EV profiles may also reflect plaque stabilization and are being explored as biomarkers and therapeutic carriers. Anti-atherosclerotic nutraceuticals such as omega-3 fatty acids, curcumin, resveratrol, ginkgo, garlic, berberine, and nattokinase may influence EV biogenesis or cargo, potentially modulating platelet reactivity and bleeding risk when combined with antithrombotic drugs. Including EV-related endpoints in nutraceutical safety studies could help clarify these mechanisms and refine risk assessment [221,222].

4. Future Research Directions

Future research on nutraceuticals should prioritize methodological rigor, standardization, and reproducibility. Well-designed randomized controlled trials with sufficient sample sizes, clearly defined populations, and standardized formulations are essential to confirm efficacy and safety. Attention should be given to dose–response relationships, bioavailability, treatment duration, and interactions with conventional therapies. Long-term safety and possible rebound effects after discontinuation warrant further investigation.
Ensuring product quality and consistency is crucial, as variability among commercial preparations often limits comparability and reliability of findings. Mechanistic studies are also needed to clarify the molecular pathways underlying the metabolic, anti-inflammatory, and vascular effects of nutraceuticals.
Collaborative efforts among clinicians, nutrition scientists, and regulatory agencies will be key to establishing evidence-based guidelines. Overall, future studies should provide robust evidence to support the safe and effective integration of nutraceuticals into clinical practice as complementary tools for improving cardiometabolic and overall health outcomes.

5. Conclusions

Omega-3 fatty acids and garlic demonstrate modest lipid-lowering effects, with purified EPA showing more consistent cardiovascular benefits than mixed EPA and DHA formulations. Nattokinase and red yeast rice (RYR) may also support cardiovascular health, though their efficacy depends heavily on formulation, dosage, and manufacturing quality.
Vitamin C, vitamin E, resveratrol, astaxanthin, and coenzyme Q10 may offer vascular benefits but can interfere with coagulation-related therapies through effects on hemostasis, drug metabolism, or redox-sensitive pathways. Their use alongside antithrombotic agents or CYP3A4-metabolized drugs should therefore be individualized and undertaken with caution.
Ginkgo biloba, ginger, ginseng, and curcumin show promising anti-atherosclerotic and vascular anti-inflammatory properties, but all carry a clinically significant risk of increased bleeding, particularly when combined with antiplatelet or anticoagulant therapy. While mechanistic data supports their vascular benefits, inconsistent evidence, variability in formulations, and the lack of standardized clinical trials highlight the need for caution and further research before routine clinical use.
Strict adherence to Good Manufacturing Practices is essential to ensure the safety and reproducibility of supplements.

Author Contributions

Conceptualization, B.V. and H.V.; writing—original draft preparation, B.V., M.-Z.D., I.-C.D., H.V. and I.-I.S.-S.; writing—review and editing, B.V., M.-Z.D., I.-C.D., H.V. and I.-I.S.-S.; visualization, H.V.; supervision, B.V., M.-Z.D. and I.-I.S.-S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
cAMP Cyclic Adenosine Monophosphate
COX1 Cyclooxygenase1
CRP C-Reactive Protein
CVD Cardiovascular Disease
CYP3A Cytochrome P450 3A
DHA Docosahexaenoic Acid
DSHEA Dietary Supplement Health and Education Act
EGFR Epidermal Growth Factor Receptor
EPA Eicosapentaenoic Acid
ERK Extracellular Signal-Regulated Kinase
FDA Food and Drug Administration
FMD Flow-Mediated Dilation
GPVI Glycoprotein VI
HDLC High-Density Lipoprotein Cholesterol
HO1 Heme Oxygenase1
hsCRP High-Sensitivity C-Reactive Protein
ICAM1 Intercellular Adhesion Molecule1
IL6 Interleukin6
INR International Normalized Ratio
IP3Inositol1,4,5trisphosphate
JAKSTAT Janus Kinase Signal Transducer and Activator of Transcription
LDL Low-Density Lipoprotein
LDLC Low-Density Lipoprotein Cholesterol
LDLR Low-Density Lipoprotein Receptor
LOX1 Lectinlike Oxidized LDL Receptor1
MAPK Mitogen-Activated Protein Kinase
MCP1 Monocyte Chemoattractant Protein1
MI Myocardial Infarction
NFκB Nuclear Factor kappa light chain enhancer of Activated B cells
NK Nattokinase
NO Nitric Oxide
NOX4 NADPH Oxidase 4
Nrf2 Nuclear Factor Erythroid 2related Factor 2
NRYR Natto Red Yeast Rice
oxLDL Oxidized Low-Density Lipoprotein
PAF Platelet-Activating Factor
PAI1 Plasminogen Activator Inhibitor1
PCSK9 Proprotein Convertase Subtilisin/Kexin Type 9
Pgp P-glycoprotein
PI3K Phosphoinositide 3Kinase
PI3Kβ Phosphoinositide 3Kinase Beta
PLC Phospholipase C
PNLIP Pancreatic lipase
PXDN Peroxidasin
PXR Pregnane X Receptor
ROS Reactive Oxygen Species
RYR Red Yeast Rice
SIRT1 Sirtuin 1
tPA Tissue Plasminogen Activator
TNFα Tumor Necrosis Factor alpha
TLR4 Toll-like Receptor 4
TRAF6 TNF Receptor Associated Factor 6
TXA2Thromboxane A2
VCAM1 Vascular Cell Adhesion Molecule1
VASP Vasodilator-Stimulated Phosphoprotein
VLDL Very-Low-Density Lipoprotein

References

  1. Björkegren, J.L.; Lusis, A.J. Atherosclerosis: Recent developments. Cell 2022, 185, 1630–1645. [Google Scholar] [CrossRef] [PubMed]
  2. Siegel-Axel, D.I.; Gawaz, M. Platelets and endothelial cells. Semin. Thromb. Hemost. 2007, 33, 128–135. [Google Scholar] [CrossRef] [PubMed]
  3. Fan, J.; Watanabe, T. Atherosclerosis: Known and unknown. Pathol. Int. 2022, 72, 151–160. [Google Scholar] [CrossRef] [PubMed]
  4. Jebari-Benslaiman, S.; Galicia-García, U.; Larrea-Sebal, A.; Olaetxea, J.R.; Alloza, I.; Vandenbroeck, K.; Benito-Vicente, A.; Martín, C. Pathophysiology of atherosclerosis. Int. J. Mol. Sci. 2022, 23, 3346. [Google Scholar] [CrossRef]
  5. Gusev, E.; Sarapultsev, A. Atherosclerosis and inflammation: Insights from the theory of general pathological processes. Int. J. Mol. Sci. 2023, 24, 7910. [Google Scholar] [CrossRef]
  6. Mussbacher, M.; Schossleitner, K.; Kral-Pointner, J.B.; Salzmann, M.; Schrammel, A.; Schmid, J.A. More than just a monolayer: The multifaceted role of endothelial cells in the pathophysiology of atherosclerosis. Curr. Atheroscler. Rep. 2022, 24, 483–492. [Google Scholar] [CrossRef]
  7. Wu, X.-M.; Zhang, N.; Li, J.-S.; Yang, Z.-H.; Huang, X.-L.; Yang, X.-F. Purinergic receptors mediate endothelial dysfunction and participate in atherosclerosis. Purinergic Signal. 2023, 19, 265–272. [Google Scholar] [CrossRef]
  8. Jiang, H.; Zhou, Y.; Nabavi, S.M.; Sahebkar, A.; Little, P.J.; Xu, S.; Weng, J.; Ge, J. Mechanisms of oxidized LDL-mediated enothelial dysfunction and its consequences for the development of atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 925923. [Google Scholar] [CrossRef]
  9. Batty, M.; Bennett, M.R.; Yu, E. The role of oxidative stress in atherosclerosis. Cells 2022, 11, 3843. [Google Scholar] [CrossRef]
  10. Momi, S.; Falcinelli, E.; Petito, E.; Ciarrocca Taranta, G.; Ossoli, A.; Gresele, P. Matrix metalloproteinase-2 on activated platelets triggers endothelial PAR-1 initiating atherosclerosis. Eur. Heart J. 2022, 43, 504–514. [Google Scholar] [CrossRef]
  11. Gardin, C.; Ferroni, L.; Leo, S.; Tremoli, E.; Zavan, B. Platelet-derived exosomes in atherosclerosis. Int. J. Mol. Sci. 2022, 23, 12546. [Google Scholar] [CrossRef] [PubMed]
  12. Theofilis, P.; Sagris, M.; Antonopoulos, A.S.; Oikonomou, E.; Tsioufis, C.; Tousoulis, D. Inflammatory mediators of platelet activation: Focus on atherosclerosis and COVID-19. Int. J. Mol. Sci. 2021, 22, 11170. [Google Scholar] [CrossRef] [PubMed]
  13. Bravo, G.M.; Annarapu, G.; Carmona, E.; Nawarskas, J.; Clark, R.; Novelli, E.; Alvidrez, R.I.M. Platelets in thrombosis and atherosclerosis: A double-edged sword. Am. J. Pathol. 2024, 194, 1608–1621. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, L.; Tang, C. Targeting platelet in atherosclerosis plaque formation: Current knowledge and future perspectives. Int. J. Mol. Sci. 2020, 21, 9760. [Google Scholar] [CrossRef]
  15. Serebruany, V.L.; Malinin, A.I.; Eisert, R.M.; Sane, D.C. Risk of bleeding complications with antiplatelet agents: Meta-analysis of 338,191 patients enrolled in 50 randomized controlled trials. Am. J. Hematol. 2004, 75, 40–47. [Google Scholar] [CrossRef]
  16. Nording, H.; Baron, L.; Langer, H.F. Platelets as therapeutic targets to prevent atherosclerosis. Atherosclerosis 2020, 307, 97–108. [Google Scholar] [CrossRef]
  17. Parma, L.; Baganha, F.; Quax, P.H.; de Vries, M.R. Plaque angiogenesis and intraplaque hemorrhage in atherosclerosis. Eur. J. Pharmacol. 2017, 816, 107–115. [Google Scholar] [CrossRef]
  18. Levy, A.; Moreno, P. Intraplaque hemorrhage. Curr. Mol. Med. 2006, 6, 479–488. [Google Scholar] [CrossRef]
  19. Kolodgie, F.D.; Gold, H.K.; Burke, A.P.; Fowler, D.R.; Kruth, H.S.; Weber, D.K.; Farb, A.; Guerrero, L.; Hayase, M.; Kutys, R. Intraplaque hemorrhage and progression of coronary atheroma. N. Engl. J. Med. 2003, 349, 2316–2325. [Google Scholar] [CrossRef]
  20. AlAli, M.; Alqubaisy, M.; Aljaafari, M.N.; AlAli, A.O.; Baqais, L.; Molouki, A.; Abushelaibi, A.; Lai, K.-S.; Lim, S.-H.E. Nutraceuticals: Transformation of Conventional Foods into Health Promoters/Disease Preventers and Safety Considerations. Molecules 2021, 26, 2540. [Google Scholar] [CrossRef]
  21. Mirzai, S.; Laffin, L.J. Supplements for Lipid Lowering: What Does the Evidence Show? Curr. Cardiol. Rep. 2023, 25, 795–805. [Google Scholar] [CrossRef]
  22. Vogel, J.H.K.; Bolling, S.F.; Costello, R.B.; Guarneri, E.M.; Krucoff, M.W.; Longhurst, J.C.; Olshansky, B.; Pelletier, K.R.; Tracy, C.M.; Vogel, R.A.; et al. Integrating complementary medicine into cardiovascular medicine: A report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. J. Am. Coll. Cardiol. 2005, 46, 123–136. [Google Scholar] [CrossRef]
  23. Starr, R.R. Too little, too late: Ineffective regulation of dietary supplements in the United States. Am. J. Public Health 2015, 105, 478–485. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  24. Collins, R.; Reith, C.; Emberson, J.; Armitage, J.; Baigent, C.; Blackwell, L.; Blumenthal, R.; Danesh, J.; Davey Smith, G.; DeMets, D.; et al. Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet 2016, 388, 2532–2561, Correction in Lancet 2017, 389, 602. [Google Scholar] [CrossRef] [PubMed]
  25. Grant, J.K.; Dangl, M.; Ndumele, C.E.; Michos, E.D.; Martin, S.S. A historical, evidence-based, and narrative review on commonly used dietary supplements in lipid-lowering. J. Lipid Res. 2024, 65, 100493. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  26. Skulas-Ray, A.C.; Wilson, P.W.F.; Harris, W.S.; Brinton, E.A.; Kris-Etherton, P.M.; Richter, C.K. Omega-3 fatty acids for the management of hypertriglyceridemia: A science advisory from the American Heart Association. Circulation 2019, 140, e673–e691. [Google Scholar] [CrossRef]
  27. Backes, J.; Anzalone, D.; Hilleman, D.; Catini, J. The clinical relevance of omega-3 fatty acids in the management of hypertriglyceridemia. Lipids Health Dis. 2016, 15, 118. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  28. Green, C.J.; Pramfalk, C.; Charlton, C.A.; Gunn, P.J.; Cornfield, T.; Pavlides, M.; Karpe, F.; Hodson, L. Hepatic de novo lipogenesis is suppressed and fat oxidation is increased by omega-3 fatty acids at the expense of glucose metabolism. BMJ Open Diabetes Res. Care 2020, 8, e000871. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Adili, R.; Hawley, M.; Holinstat, M. Regulation of platelet function and thrombosis by omega-3 and omega-6 polyunsaturated fatty acids. Prostaglandins Other Lipid Mediat. 2018, 139, 10–18. [Google Scholar] [CrossRef]
  30. DeFilippis, A.; Rai, S.N.; Cambon, A.; Miles, R.; Jaffe, A.S.; Moser, A.B.; O Jones, R.; Bolli, R.; Schulman, S.P. Fatty acids and TxA2 generation, in the absence of platelet-COX-1 activity. Nutr. Metab. Cardiovasc. Dis. 2014, 24, 428–433. [Google Scholar] [CrossRef]
  31. Bäck, M. Omega-3 fatty acids in atherosclerosis and coronary artery disease. Futur. Sci. OA 2017, 3, FSO236. [Google Scholar] [CrossRef]
  32. Fredman, G.; Van Dyke, T.E.; Serhan, C.N. Resolvin E1 Regulates Adenosine Diphosphate Activation of Human Platelets. Arter. Thromb. Vasc. Biol. 2010, 30, 2005–2013. [Google Scholar] [CrossRef] [PubMed]
  33. Kacik, M.; Olivan-Viguera, A.; Köhler, R. Modulation of KCa3.1 Channels by Eicosanoids, Omega-3 Fatty Acids, and Molecular Determinants. PLoS ONE 2014, 9, e112081. [Google Scholar] [CrossRef] [PubMed]
  34. Larson, M.K.; Tormoen, G.W.; Weaver, L.J.; Luepke, K.J.; Patel, I.A.; Hjelmen, C.E.; Ensz, N.M.; McComas, L.S.; Mccarty, O.J.T. Exogenous modification of platelet membranes with the omega-3 fatty acids EPA and DHA reduces platelet procoagulant activity and thrombus formation. Am. J. Physiol. Physiol. 2013, 304, C273–C279. [Google Scholar] [CrossRef] [PubMed]
  35. Yamaguchi, A.; Stanger, L.; Freedman, C.J.; Standley, M.; Hoang, T.; Adili, R.; Tsai, W.; Van Hoorebeke, C.; Holman, T.R.; Holinstat, M. DHA 12-LOX-derived oxylipins regulate platelet activation and thrombus formation through a PKA-dependent signaling pathway. J. Thromb. Haemost. 2020, 19, 839–851. [Google Scholar] [CrossRef]
  36. Larson, M.K.; Shearer, G.C.; Ashmore, J.H.; Anderson-Daniels, J.M.; Graslie, E.L.; Tholen, J.T.; Vogelaar, J.L.; Korth, A.J.; Nareddy, V.; Sprehe, M.; et al. Omega-3 fatty acids modulate collagen signaling in human platelets. Prostaglandins Leukot. Essent. Fat. Acids. 2011, 84, 93–98. [Google Scholar] [CrossRef]
  37. Golanski, J.; Szymanska, P.; Rozalski, M. Effects of Omega-3 Polyunsaturated Fatty Acids and Their Metabolites on Haemostasis-Current Perspectives in Cardiovascular Disease. Int. J. Mol. Sci. 2021, 22, 2394. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  38. Rausch, J.; Gillespie, S.; Orchard, T.; Tan, A.; McDaniel, J.C. Systematic review of marine-derived omega-3 fatty acid supplementation effects on leptin, adi-ponectin, and the leptin-to-adiponectin ratio. Nutr. Res. 2021, 85, 135–152. [Google Scholar] [CrossRef]
  39. Bowman, L.; Mafham, M.; Wallendszus, K.; Stevens, W.; Buck, G.; Barton, J.; Murphy, K.; Aung, T.; Haynes, R.; Cox, J.; et al. Effects of Aspirin for Primary Prevention in Persons with Diabetes Mellitus. N. Engl. J. Med. 2018, 18, 379, 1529–1539. [Google Scholar] [CrossRef] [PubMed]
  40. Bhatt, D.L.; Steg, P.G.; Miller, M.; Brinton, E.A.; Jacobson, T.A.; Ketchum, S.B.; Doyle, R.T., Jr.; Juliano, R.A.; Jiao, L.; Granowitz, C.; et al. REDUCE-IT Investigators. Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N. Engl. J. Med. 2019, 380, 11–22. [Google Scholar] [CrossRef] [PubMed]
  41. Zivkovic, S.; Maric, G.; Cvetinovic, N.; Lepojevic-Stefanovic, D.; Bozic Cvijan, B. Anti-Inflammatory Effects of Lipid-Lowering Drugs and Supplements-A Narrative Review. Nutrients 2023, 15, 1517. [Google Scholar] [CrossRef]
  42. Nicholls, S.J.; Lincoff, A.M.; Garcia, M.; Bash, D.; Ballantyne, C.M.; Barter, P.J.; Davidson, M.H.; Kastelein, J.J.P.; Koenig, W.; McGuire, D.K.; et al. Effect of high-dose omega-3 fatty acids vs corn oil on major adverse cardiovascular events in patients at high cardiovascular risk: The STRENGTH randomized clinical trial. JAMA 2020, 324, 2268–2280. [Google Scholar] [CrossRef]
  43. Manson, J.E.; Cook, N.R.; Lee, I.M.; Christen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Albert, C.M.; Gordon, D.; Copeland, T.; et al. Marine n−3 fatty acids and prevention of cardiovascular disease and cancer. N. Engl. J. Med. 2019, 380, 23–32. [Google Scholar] [CrossRef] [PubMed]
  44. Song, D.; Hao, J.; Fan, D. Biological properties and clinical applications of berberine. Front. Med. 2020, 14, 564–582. [Google Scholar] [CrossRef] [PubMed]
  45. Lee, Y.S.; Kim, W.S.; Kim, K.H.; Yoon, M.J.; Cho, H.J.; Shen, Y.; Ye, J.M.; Lee, C.H.; Oh, W.K.; Kim, C.T.; et al. Berberine, a natural plant product, activates AMP-activated protein kinase with beneficial metabolic effects in diabetic and insulin-resistant states. Diabetes 2006, 55, 2256–2264. [Google Scholar] [CrossRef] [PubMed]
  46. Kong, W.; Wei, J.; Abidi, P.; Lin, M.; Inaba, S.; Li, C.; Wang, Y.; Wang, Z.; Si, S.; Pan, H.; et al. Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat. Med. 2004, 10, 1344–1351. [Google Scholar] [CrossRef] [PubMed]
  47. Ai, X.; Yu, P.; Peng, L.; Luo, L.; Liu, J.; Li, S.; Lai, X.; Luan, F.; Meng, X. Berberine: A Review of its Pharmacokinetics Properties and Therapeutic Potentials in Diverse Vascular Diseases. Front. Pharmacol. 2021, 12, 762654. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  48. Wang, C.; Cheng, Y.; Zhang, Y.; Jin, H.; Zuo, Z.; Wang, A.; Huang, J.; Jiang, J.; Kong, W. Berberine and Its Main Metabolite Berberrubine Inhibit Platelet Activation Through Suppressing the Class I PI3Kβ/Rasa3/Rap1 Pathway. Front. Pharmacol. 2021, 12, 734603. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Xie, X.; Ma, X.; Zeng, S.; Tang, W.; Xiao, L.; Zhu, C.; Yu, R. Mechanisms of Berberine for the Treatment of Atherosclerosis Based on Network Pharmacology. Evid.-Based Complement. Altern. Med. 2020, 2020, 3568756. [Google Scholar] [CrossRef]
  50. Dempsey, P. Red Yeast Rice: Lipid Lowering Properties and Safety Considerations. Online J. Complement. Altern. Med. 2021, 6, 101752366. [Google Scholar] [CrossRef]
  51. Peng, D.; Fong, A.; Pelt, A. The effects of red yeast rice supplementation on cholesterol levels in adults. Am. J. Nurs. 2017, 117, 46–54. [Google Scholar] [CrossRef]
  52. Trogkanis, E.; Karalexi, M.A.; Sergentanis, T.N.; Kornarou, E.; Vassilakou, T. Safety and Efficacy of the Consumption of the Nutraceutical “Red Yeast Rice Extract” for the Reduction of Hypercholesterolemia in Humans: A Systematic Review and Meta-Analysis. Nutrients 2024, 16, 1453. [Google Scholar] [CrossRef] [PubMed]
  53. Cicero, A.F.G.; Fogacci, F.; Stoian, A.P.; Toth, P.P. Red Yeast Rice for the Improvement of Lipid Profiles in Mild-to-Moderate Hypercholesterolemia: A Narrative Review. Nutrients 2023, 15, 2288. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Liu, M.; Xu, Z.; Wang, Z.; Wang, D.; Yang, M.; Li, H.; Zhang, W.; He, R.; Cheng, H.; Guo, P.; et al. Lipid-lowering, antihypertensive, and antithrombotic effects of nattokinase combined with red yeast rice in patients with stable coronary artery disease: A randomized, double-blinded, placebo-controlled trial. Front. Nutr. 2024, 11, 1380727. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, S.; Liu, F.; Li, J.; Liang, F.; Li, J.; Cao, J.; Liu, D.; Huang, K.; Li, H.; Lu, X.; et al. Evaluation of NaTto Red Yeast Rice on Regulating Blood Lipid (ENTRY) Study: A Multicenter, Double-Placebo, Double-Blinded, Randomized Controlled Trial in Chinese Adults. Chronic Dis. Transl. Med. 2025, 11, 122–129. [Google Scholar] [CrossRef]
  56. Tesfaye, A. Revealing the Therapeutic Uses of Garlic (Allium sativum) and Its Potential for Drug Discovery. Sci. World J. 2021, 2021, 8817288. [Google Scholar] [CrossRef]
  57. Sun, Y.E.; Wang, W.; Qin, J. Anti-Hyperlipidemia of Garlic by Reducing the Level of Total Cholesterol and Low-Density Lipoprotein: A Meta-Analysis. Medicine 2018, 97, e0255. [Google Scholar] [CrossRef]
  58. Silagy, C.; Neil, A. Garlic as a Lipid Lowering Agent-A Meta-Analysis. J. R. Coll. Physicians Lond. 1994, 28, 39–45. [Google Scholar] [CrossRef] [PubMed]
  59. Du, Y.; Zhou, H.; Zha, W. Garlic Consumption Can Reduce the Risk of Dyslipidemia: A Meta-Analysis of Randomized Controlled Trials. J. Health Popul. Nutr. 2024, 43, 113. [Google Scholar] [CrossRef]
  60. Rahman, K.; Lowe, G.M.; Smith, S. Aged Garlic Extract Inhibits Human Platelet Aggregation by Altering Intracellular Signaling and Platelet Shape Change. J. Nutr. 2016, 146, 410S–415S. [Google Scholar] [CrossRef]
  61. Wang, Z.; Ding, L.; Liu, J.; Savarin, P.; Wang, X.; Zhao, K.; Ding, W.; Hou, Y. Allicin Ameliorates Glucose and Lipid Metabolism via Modulation of Gut Microbiota and Bile Acid Profile in Diabetic Rats. J. Funct. Foods 2023, 111, 105899. [Google Scholar] [CrossRef]
  62. Laka, K.; Makgoo, L.; Mbita, Z. Cholesterol-Lowering Phytochemicals: Targeting the Mevalonate Pathway for Anticancer Interventions. Front. Genet. 2022, 13, 841639. [Google Scholar] [CrossRef]
  63. Rahman, K. Effects of Garlic on Platelet Biochemistry and Physiology. Mol. Nutr. Food Res. 2007, 51, 1335–1344. [Google Scholar] [CrossRef]
  64. Wei, C.; Cai, R.; Song, Y.; Liu, X.; Xu, H.L. Research Progress of Nattokinase in Reducing Blood Lipid. Nutrients 2025, 17, 1784. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Muric, M.; Nikolic, M.; Todorovic, A.; Jakovljevic, V.; Vucicevic, K. Comparative Cardioprotective Effectiveness: NOACs vs. Nattokinase-Bridging Basic Research to Clinical Findings. Biomolecules 2024, 14, 956. [Google Scholar] [CrossRef] [PubMed]
  66. Hodis, H.N.; Mack, W.J.; Meiselman, H.J.; Kalra, V.; Liebman, H.; Hwang-Levine, J.; Dustin, L.; Kono, N.; Mert, M.; Wenby, R.B.; et al. Nattokinase atherothrombotic prevention study: A randomized controlled trial. Clin. Hemorheol. Microcirc. 2021, 78, 339–353. [Google Scholar] [CrossRef] [PubMed]
  67. Urano, T.; Ihara, H.; Umemura, K.; Suzuki, Y.; Oike, M.; Akita, S.; Tsukamoto, Y.; Suzuki, I.; Takada, A. The profibrinolytic enzyme subtilisin NAT purified from Bacillus subtilis cleaves and inactivates plasminogen activator inhibitor type 1. J. Biol. Chem. 2001, 276, 24690–24696. [Google Scholar] [CrossRef]
  68. Milner, M.; Makise, K. Natto and its active ingredient nattokinase: A potent and safe thrombolytic agent. Altern. Complement. Therap. 2002, 8, 157–164. [Google Scholar] [CrossRef]
  69. Jang, J.Y.; Kim, T.S.; Cai, J.; Kim, J.; Kim, Y.; Shin, K.; Kim, K.S.; Park, S.K.; Lee, S.P.; Choi, E.K.; et al. Nattokinase improves blood flow by inhibiting platelet aggregation and thrombus formation. Lab. Anim. Res. 2013, 29, 221–225. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Hsia, C.H.; Shen, M.C.; Lin, J.S.; Wen, Y.K.; Hwang, K.L.; Cham, T.M.; Yang, N.C. Nattokinase decreases plasma levels of fibrinogen, factor VII, and factor VIII in human subjects. Nutr. Res. 2009, 29, 190–196. [Google Scholar] [CrossRef]
  71. Liu, X.; Zeng, X.; Mahe, J.; Guo, K.; He, P.; Yang, Q.; Zhang, Z.; Li, Z.; Wang, D.; Zhang, Z.; et al. The Effect of Nattokinase-Monascus Supplements on Dyslipidemia: A Four-Month Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2023, 15, 4239. [Google Scholar] [CrossRef] [PubMed]
  72. Luquain-Costaz, C.; Delton, I. Oxysterols in vascular cells and role in atherosclerosis. In Implication of Oxysterols and Phy-tosterols in Aging and Human Diseases; Springer International Publishing: Cham, Switzerland, 2023; pp. 213–229. [Google Scholar]
  73. Samimi, F.; Namiranian, N.; Sharifi-Rigi, A.; Siri, M.; Abazari, O.; Dastghaib, S. Coenzyme Q10: A Key Antioxidant in the Management of Diabetes-Induced Cardiovascular Complications—An Overview of Mechanisms and Clinical Evidence. Int. J. Endocrinol. 2024, 2024, 2247748. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, S.; Wang, J.; Feng, J.; Xuan, R. Research progress of Astaxanthin nano-based drug delivery system: Applications, prospects and challenges? Front. Pharmacol. 2023, 14, 1102888. [Google Scholar] [CrossRef] [PubMed]
  75. Gao, C.; Gong, N.; Chen, F.; Hu, S.; Zhou, Q.; Gao, X. The Effects of Astaxanthin on Metabolic Syndrome: A Comprehensive Review. Mar. Drugs. 2024, 23, 9. [Google Scholar] [CrossRef]
  76. Ciaraldi, T.P.; Boeder, S.C.; Mudaliar, S.R.; Giovannetti, E.R.; Henry, R.R.; Pettus, J.H. Astaxanthin, a natural antioxidant, lowers cholesterol and markers of cardiovascular risk in individuals with prediabetes and dyslipidaemia. Diabetes Obes. Metab. 2023, 25, 1985–1994. [Google Scholar] [CrossRef]
  77. Davinelli, S.; Saso, L.; D’Angeli, F.; Calabrese, V.; Intrieri, M.; Scapagnini, G. Astaxanthin as a modulator of Nrf2, NF-κB, and their crosstalk: Molecular mechanisms and possible clinical applications. Molecules 2022, 27, 502. [Google Scholar] [CrossRef]
  78. Pereira, C.P.M.; Souza, A.C.R.; Vasconcelos, A.R.; Prado, P.S.; Name, J.J. Antioxidant and anti-inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review). Int. J. Mol. Med. 2021, 47, 37–48. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  79. Heidari, M.; Chaboksafar, M.; Alizadeh, M.; Sohrabi, B.; Kheirouri, S. Effects of Astaxanthin supplementation on selected metabolic parameters, anthropometric indices, Sirtuin1 and TNF-α levels in patients with coronary artery disease: A randomized, double-blind, placebo-controlled clinical trial. Front. Nutr. 2023, 10, 1104169. [Google Scholar] [CrossRef]
  80. Wu, D.; Xu, H.; Chen, J.; Zhang, L. Effects of astaxanthin supplementation on oxidative stress. Int. J. Vitam. Nutr. Res. 2020, 90, 179–194. [Google Scholar] [CrossRef]
  81. Fassett, R.G.; Coombes, J.S. Astaxanthin in cardiovascular health and disease. Molecules 2012, 17, 2030–2048. [Google Scholar] [CrossRef]
  82. Xia, N.; Daiber, A.; Förstermann, U.; Li, H. Antioxidant effects of resveratrol in the cardiovascular system. Br. J. Pharmacol. 2017, 174, 1633–1646. [Google Scholar] [CrossRef] [PubMed]
  83. Obeme-Nmom, J.I.; Abioye, R.O.; Reyes Flores, S.S.; Udenigwe, C.C. Regulation of redox enzymes by nutraceuticals: A review of the roles of antioxidant polyphenols and peptides. Food Funct. 2024, 15, 10956–10980. [Google Scholar] [CrossRef] [PubMed]
  84. Li, H.; Förstermann, U. Resveratrol: A Multifunctional Compound Improving Endothelial Function: Editorial to:“Resveratrol Supplementation Gender Independently Improves Endothelial Reactivity and Suppresses Superoxide Production in Healthy Rats” by S. Soylemez et al. Cardiovasc. Drugs Ther. 2009, 23, 425–429. [Google Scholar] [CrossRef] [PubMed]
  85. Hu, D.; Wang, L.; Qi, L.; Yang, X.; Jin, Y.; Yin, H.; Huang, Y.; Sheng, J.; Wang, X. Resveratrol improved atherosclerosis by increasing LDLR levels via the EGFR-ERK1/2 signaling pathway. Lipids Health Dis. 2025, 24, 167. [Google Scholar] [CrossRef]
  86. Penumathsa, S.V.; Thirunavukkarasu, M.; Koneru, S.; Juhasz, B.; Zhan, L.; Pant, R.; Menon, V.P.; Otani, H.; Maulik, N. Statin and resveratrol in combination induces cardioprotection against myocardial infarction in hypercholesterolemic rat. J. Mol. Cell. Cardiol. 2007, 42, 508–516. [Google Scholar] [CrossRef]
  87. Zhang, Y.; Cao, X.; Zhu, W.; Liu, Z.; Liu, H.; Zhou, Y.; Cao, Y.; Liu, C.; Xie, Y. Resveratrol enhances autophagic flux and promotes Ox-LDL degradation in HUVECs via upregulation of SIRT1. Oxid. Med. Cell. Longev. 2016, 2016, 7589813. [Google Scholar] [CrossRef]
  88. Teimouri, M.; Homayouni-Tabrizi, M.; Rajabian, A.; Amiri, H.; Hosseini, H. Anti-inflammatory effects of resveratrol in patients with cardiovascular disease: A systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2022, 70, 102863. [Google Scholar] [CrossRef]
  89. Mohammadipoor, N.; Shafiee, F.; Rostami, A.; Kahrizi, M.S.; Soleimanpour, H.; Ghodsi, M.; Ansari, M.J.; Bokov, D.O.; Jannat, B.; Mosharkesh, E.; et al. Resveratrol supplementation efficiently improves endothelial health: A systematic review and meta-analysis of randomized controlled trials. Phytother. Res. 2022, 36, 3529–3539. [Google Scholar] [CrossRef]
  90. Akbari, M.; Tamtaji, O.R.; Lankarani, K.B.; Tabrizi, R.; Dadgostar, E.; Kolahdooz, F.; Jamilian, M.; Mirzaei, H.; Asemi, Z. The effects of resveratrol supplementation on endothelial function and blood pressures among patients with metabolic syndrome and related disorders: A systematic review and meta-analysis of randomized controlled trials. High Blood Press. Cardiovasc. Prev. 2019, 26, 305–319. [Google Scholar] [CrossRef]
  91. Godos, J.; Romano, G.L.; Gozzo, L.; Laudani, S.; Paladino, N.; Dominguez Azpíroz, I.; Martínez López, N.M.; Giampieri, F.; Quiles, J.L.; Battino, M.; et al. Resveratrol and vascular health: Evidence from clinical studies and mechanisms of actions related to its metabolites produced by gut microbiota. Front. Pharmacol. 2024, 15, 1368949. [Google Scholar] [CrossRef]
  92. Cirilli, I.; Damiani, E.; Dludla, P.V.; Hargreaves, I.; Marcheggiani, F.; Millichap, L.E.; Orlando, P.; Silvestri, S.; Tiano, L. Role of Coenzyme Q10 in Health and Disease: An Update on the Last 10 Years (2010–2020). Antioxidants 2021, 10, 1325. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  93. Mollazadeh, H.; Tavana, E.; Fanni, G.; Bo, S.; Banach, M.; Pirro, M.; Von Haehling, S.; Jamialahmadi, T.; Sahebkar, A. Effects of statins on mitochondrial pathways. J. Cachexia Sarcopenia Muscle 2021, 12, 237–251. [Google Scholar] [CrossRef]
  94. Daei, S.; Ildarabadi, A.; Goodarzi, S.; Mohamadi-Sartang, M. Effect of coenzyme Q10 supplementation on vascular endothelial function: A systematic review and meta-analysis of randomized controlled trials. High Blood Press. Cardiovasc. Prev. 2024, 31, 113–126. [Google Scholar] [CrossRef] [PubMed]
  95. Borges, J.Y. The Role of Coenzyme Q10 in Cardiovascular Disease Treatment: An Updated 2024 Systematic Review and Meta-Analysis of Prospective Cohort Studies (1990–2024). medRxiv 2024. [Google Scholar] [CrossRef]
  96. Dohlmann, T.L.; Kuhlman, A.B.; Morville, T.; Dahl, M.; Asping, M.; Orlando, P.; Silvestri, S.; Tiano, L.; Helge, J.W.; Dela, F.; et al. Coenzyme Q10 supplementation in statin treated patients: A double-blinded randomized placebo-controlled trial. Antioxidants 2022, 11, 1698. [Google Scholar] [CrossRef] [PubMed]
  97. Gao, L.; Mao, Q.; Cao, J.; Wang, Y.; Zhou, X.; Fan, L. Effects of coenzyme Q10 on vascular endothelial function in humans: A meta-analysis of randomized controlled trials. Atherosclerosis 2012, 221, 311–316. [Google Scholar] [CrossRef]
  98. Hamilton, S.J.; Chew, G.T.; Watts, G.F. Coenzyme Q10 improves endothelial dysfunction in statin-treated type 2 diabetic patients. Diabetes Care 2009, 32, 810–812. [Google Scholar] [CrossRef]
  99. Mortensen, S.A.; Rosenfeldt, F.; Kumar, A.; Dolliner, P.; Filipiak, K.J.; Pella, D.; Alehagen, U.; Steurer, G.; Littarru, G.P. The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure. Eur. J. Heart Fail. 2013, 12, S21. [Google Scholar]
  100. Alehagen, U.; Johansson, P.; Björnstedt, M.; Rosén, A.; Dahlström, U. Cardiovascular mortality and N-terminal-proBNP reduced after combined selenium and coenzyme Q10 supplementation: A 5-year prospective randomized double-blind placebo-controlled trial among elderly Swedish citizens. Int. J. Cardiol. 2013, 167, 1860–1866. [Google Scholar] [CrossRef]
  101. Alehagen, U.; Aaseth, J.; Johansson, P. Reduced cardiovascular mortality 10 years after supplementation with selenium and coenzyme Q10 for four years: Follow-up results of a prospective randomized double-blind placebo-controlled trial in elderly citizens. PLoS ONE 2015, 10, e0141641. [Google Scholar] [CrossRef]
  102. Alehagen, U.; Aaseth, J.; Alexander, J.; Johansson, P. Still reduced cardiovascular mortality 12 years after supplementation with selenium and coenzyme Q10 for four years: A validation of previous 10-year follow-up results of a prospective randomized double-blind placebo-controlled trial in elderly. PLoS ONE 2018, 13, e0193120. [Google Scholar] [CrossRef] [PubMed]
  103. Vrentzos, E.; Ikonomidis, I.; Pavlidis, G.; Katogiannis, K.; Korakas, E.; Kountouri, A.; Pliouta, L.; Michalopoulou, E.; Pelekanou, E.; Boumpas, D.; et al. Six-month supplementation with high dose coenzyme Q10 improves liver steatosis, endothelial, vascular and myocardial function in patients with metabolic-dysfunction associated steatotic liver disease: A randomized double-blind, placebo-controlled trial. Cardiovasc. Diabetol. 2024, 23, 245. [Google Scholar] [CrossRef] [PubMed]
  104. Gulcin, İ. Antioxidants: A comprehensive review. Arch. Toxicol. 2025, 99, 1893–1997. [Google Scholar] [CrossRef] [PubMed]
  105. Trugilho, L.; Alvarenga, L.; Cardozo, L.; Paiva, B.; Brito, J.; Barboza, I.; Almeida, J.; Dos Anjos, J.; Khosla, P.; Ribeiro-Alves, M.; et al. Effects of Tocotrienol on Cardiovascular Risk Markers in Patients with Chronic Kidney Disease: A Randomized Controlled Trial. J. Nutr. Metab. 2025, 2025, 8482883. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Samsam Shariat, S.Z.; Mostafavi, S.A.; Khakpour, F. Antioxidant effects of vitamins C and e on the low-density lipoprotein oxidation mediated by myeloperoxidase. Iran. Biomed. J. 2013, 17, 22–28. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  107. Dludla, P.V.; Nkambule, B.B.; Nyambuya, T.M.; Ziqubu, K.; Mabhida, S.E.; Mxinwa, V.; Mokgalaboni, K.; Ndevahoma, F.; Hanser, S.; Mazibuko-Mbeje, S.E.; et al. Vitamin C intake potentially lowers total cholesterol to improve endothelial function in diabetic patients at increased risk of cardiovascular disease: A systematic review of randomized controlled trials. Front. Nutr. 2022, 9, 1011002. [Google Scholar] [CrossRef]
  108. Ashor, A.W.; Siervo, M.; Lara, J.; Oggioni, C.; Afshar, S.; Mathers, J.C. Effect of vitamin C and vitamin E supplementation on endothelial function: A systematic review and meta-analysis of randomised controlled trials. Br. J. Nutr. 2015, 113, 1182–1194. [Google Scholar] [CrossRef] [PubMed]
  109. Ye, Y.; Li, J.; Yuan, Z. Effect of antioxidant vitamin supplementation on cardiovascular outcomes: A meta-analysis of randomized controlled trials. PLoS ONE 2013, 8, e56803. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  110. Lonn, E.; Bosch, J.; Yusuf, S.; Sheridan, P.; Pogue, J.; Arnold, J.M.; Ross, C.; Arnold, A.; Sleight, P.; Probstfield, J.; et al. Effects of long-term vitamin E supplementation on cardiovascular events and CancerA randomized controlled trial. J. Am. Med. Assoc. 2005, 293, 1338–1347. [Google Scholar]
  111. Lee, I.M.; Cook, N.R.; Gaziano, J.M.; Gordon, D.; Ridker, P.M.; Manson, J.E.; Hennekens, C.H.; Buring, J.E. Vitamin E in the primary prevention of cardiovascular disease and cancer: The Women’s Health Study: A randomized controlled trial. JAMA 2005, 294, 56–65. [Google Scholar] [CrossRef]
  112. Miller III, E.R.; Pastor-Barriuso, R.; Dalal, D.; Riemersma, R.A.; Appel, L.J.; Guallar, E. Meta-analysis: High-dosage vitamin E supplementation may increase all-cause mortality. Ann. Intern. Med. 2005, 142, 37–46. [Google Scholar] [CrossRef]
  113. Simon, J.A. Combined vitamin E and vitamin C supplement use and risk of cardiovascular disease mortality. Arch. Intern. Med. 2002, 162, 2630. [Google Scholar] [CrossRef]
  114. Koltermann, A. Influence of Ginkgo Biloba Extract EGb 761 on Signaling Pathways in Endothelial Cells. Ph.D. Dissertation, Ludwig-Maximilians-University, Munich, Germany, 2008. [Google Scholar] [CrossRef]
  115. Ma, L.; Liu, X.; Zhao, Y.; Chen, B.; Li, X.; Qi, R. Ginkgolide B reduces LOX-1 expression by inhibiting Akt phosphorylation and increasing Sirt1 expression in oxidized LDL-stimulated human umbilical vein endothelial cells. PLoS ONE 2013, 8, e74769. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Chen, J.S.; Huang, P.H.; Wang, C.H.; Lin, F.Y.; Tsai, H.Y.; Wu, T.C.; Lin, S.J.; Chen, J.W. Nrf-2 mediated heme oxygenase-1 expression, an antioxidant-independent mechanism, contributes to anti-atherogenesis and vascular protective effects of Ginkgo biloba extract. Atherosclerosis 2011, 214, 301–309. [Google Scholar] [CrossRef]
  117. Wu, T.-C.; Chen, J.-S.; Wang, C.-H.; Huang, P.-H.; Lin, F.-Y.; Lin, L.-Y.; Lin, S.-J.; Chen, J.-W. Activation of heme oxygenase-1 by Ginkgo biloba extract differentially modulates endothelial and smooth muscle-like progenitor cells for vascular repair. Sci. Rep. 2019, 9, 17316. [Google Scholar] [CrossRef] [PubMed]
  118. Gardner, C.D.; Taylor-Piliae, R.E.; Kiazand, A.; Nicholus, J.; Rigby, A.J.; Farquhar, J.W. Effect of Ginkgo biloba (EGb 761) on treadmill walking time among adults with peripheral artery disease: A randomized clinical trial. J. Cardiopulm. Rehabil. Prev. 2008, 28, 258–265. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  119. Wu, Y.; Li, S.; Cui, W.; Zu, X.; Wang, F.; Du, J. Ginkgo biloba extract improves coronary blood flow in patients with coronary artery disease: Role of endothelium-dependent vasodilation. Planta Med. 2007, 73, 624–628. [Google Scholar] [CrossRef] [PubMed]
  120. Kuller, L.H.; Ives, D.G.; Fitzpatrick, A.L.; Carlson, M.C.; Mercado, C.; Lopez, O.L.; Burke, G.L.; Furberg, C.D.; DeKosky, S.T. Does Ginkgo biloba reduce the risk of cardiovascular events? Circ. Cardiovasc. Qual. Outcomes 2010, 3, 41–47. [Google Scholar] [CrossRef]
  121. Snitz, B.E.; O’Meara, E.S.; Carlson, M.C.; Arnold, A.M.; Ives, D.G.; Rapp, S.R.; Saxton, J.; Lopez, O.L.; Dunn, L.O.; Sink, K.M.; et al. Ginkgo biloba for preventing cognitive decline in older adults: A randomized trial. JAMA 2009, 302, 2663–2670. [Google Scholar] [CrossRef]
  122. Morató, X.; Marquié, M.; Tartari, J.P.; Lafuente, A.; Abdelnour, C.; Alegret, M.; Jofresa, S.; Buendía, M.; Pancho, A.; Aguilera, N.; et al. A randomized, open-label clinical trial in mild cognitive impairment with EGb 761 examining blood markers of inflammation and oxidative stress. Sci. Rep. 2023, 13, 5406. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  123. Dong, H.; Liu, M.; Rong, L.; Yang, S.; Wang, J. Efficacy and Hemorheology of Ginkgo biloba Extract (EGb 761) in the Treatment of Sudden Sensorineural Hearing Loss: A Retrospective Study. Noise Health 2024, 26, 383–389. [Google Scholar] [CrossRef]
  124. Ballester, P.; Cerdá, B.; Arcusa, R.; Marhuenda, J.; Yamedjeu, K.; Zafrilla, P. Effect of Ginger on Inflammatory Diseases. Molecules 2022, 27, 7223. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  125. Wang, S.; Song, X.; Gao, H.; Zhang, Y.; Zhou, X.; Wang, F. 6-Gingerol Inhibits Ferroptosis in Endothelial Cells in Atherosclerosis by Activating the NRF2/HO-1 Pathway. Appl. Biochem. Biotechnol. 2025, 197, 3890–3906. [Google Scholar] [CrossRef] [PubMed]
  126. Morvaridzadeh, M.; Fazelian, S.; Agah, S.; Khazdouz, M.; Rahimlou, M.; Agh, F.; Potter, E.; Heshmati, S.; Heshmati, J. Effect of ginger (Zingiber officinale) on inflammatory markers: A systematic review and meta-analysis of randomized controlled trials. Cytokine 2020, 135, 155224. [Google Scholar] [CrossRef] [PubMed]
  127. Preciado-Ortiz, M.E.; Gembe-Olivarez, G.; Martínez-López, E.; Rivera-Valdés, J.J. Immunometabolic Effects of Ginger (Zingiber officinale Roscoe) Supplementation in Obesity: A Comprehensive Review. Molecules 2025, 30, 2933. [Google Scholar] [CrossRef]
  128. Askari, G.; Aghajani, M.; Salehi, M.; Najafgholizadeh, A.; Keshavarzpour, Z.; Fadel, A.; Venkatakrishnan, K.; Salehi-Sahlabadi, A.; Hadi, A.; Pourmasoumi, M. The effects of ginger supplementation on biomarkers of inflammation and oxidative stress in adults: A systematic review and meta-analysis of randomized controlled trials. J. Herb. Med. 2020, 22, 100364. [Google Scholar] [CrossRef]
  129. Garza, M.C.; Pérez-Calahorra, S.; Rodrigo-Carbó, C.; Sánchez-Calavera, M.A.; Jarauta, E.; Mateo-Gallego, R.; Gracia-Rubio, I.; Lamiquiz-Moneo, I. Effect of aromatic herbs and spices present in the Mediterranean diet on the glycemic profile in type 2 diabetes subjects: A systematic review and meta-analysis. Nutrients 2024, 16, 756. [Google Scholar] [CrossRef]
  130. Lee, C.H.; Kim, J.H. A review on the medicinal potentials of ginseng and ginsenosides on cardiovascular diseases. J. Ginseng Res. 2014, 38, 161–166. [Google Scholar] [CrossRef]
  131. Song, X.; Gao, W.; Shi, Y.; Li, J.; Zheng, Z. Panax Ginseng and its derivatives: Promoting angiogenesis in ischemic dis-eases–A mechanistic overview. J. Funct. Foods 2023, 109, 105762. [Google Scholar] [CrossRef]
  132. Rodthongdee, K.; Watanapa, W.B.; Ruamyod, K.; Semprasert, N.; Nambundit, P.; Kooptiwut, S.; Boontaveekul, L. Ginsenoside Re increases human coronary artery endothelial SKCa current and nitric oxide release via glucocorticoid recep-tor-PI3K-Akt/PKB pathway. J. Ginseng Res. 2025, 49, 523–531. [Google Scholar] [CrossRef]
  133. Zhang, H.; Hu, C.; Xue, J.; Jin, D.; Tian, L.; Zhao, D.; Li, X.; Qi, W. Ginseng in vascular dysfunction: A review of therapeutic potentials and molecular mechanisms. Phytother. Res. 2022, 36, 857–872. [Google Scholar] [CrossRef]
  134. Jovanovski, E.; Peeva, V.; Sievenpiper, J.L.; Jenkins, A.L.; Desouza, L.; Rahelic, D.; Sung, M.K.; Vuksan, V. Modulation of en-dothelial function by Korean red ginseng (Panax ginseng CA Meyer) and its components in healthy individuals: A randomized controlled trial. Cardiovasc. Ther. 2014, 32, 163–169. [Google Scholar] [CrossRef]
  135. Chung, I.M.; Lim, J.W.; Chung, H.Y.; Seo, J.Y.; Shin, G.J.; Park, S.H.; Kim, H. 7. Korean Panax Red Ginseng Improves Endothelial Dysfunction and Arterial Stiffness in Patients with Coronary Artery Disease Probably by Decreasing Rho-Associated Kinase Activity of Peripheral Blood Mononuclear Cells. Artery Res. 2009, 3, 93. [Google Scholar] [CrossRef]
  136. Cha, T.W.; Kim, M.; Kim, M.; Chae, J.S.; Lee, J.H. Blood pressure-lowering effect of Korean red ginseng associated with decreased circulating Lp-PLA2 activity and lysophosphatidylcholines and increased dihydrobiopterin level in prehy-pertensive subjects. Hypertens. Res. 2016, 39, 449–456. [Google Scholar] [CrossRef] [PubMed]
  137. Esmaeili, A.; Khalili, N.; Najafi, N.; Hajizadeh-Sharafabad, F. Ginseng supplementation and vascular function: A sys-tematic review and meta-analysis of clinical trials. BMC Complement. Med. Ther. 2025, 25, 259. [Google Scholar] [CrossRef] [PubMed]
  138. Jafari, A.; Kordkatuli, K.; Mardani, H.; Mehdipoor, F.; Jami, P.B.; Abbastabar, M.; Vakili, M.; Besharat, S.; Alaghi, A. Ginseng supplementation and cardiovascular disease risk factors: A protocol for GRADE-assessed systematic review and dose-response meta-analysis. BMJ Open 2024, 14, e080926. [Google Scholar] [CrossRef]
  139. Huang, J.; Yi, Q.; Chen, Y.; Li, Y.; Xu, G.; Zhang, J.; Niu, T.; You, Y.; Zou, W.; Qian, S.; et al. Curcumin suppresses oxidative stress via regulation of ROS/NF-κB signaling pathway to protect retinal vascular endothelial cell in diabetic reti-nopathy. Mol. Cell. Toxicol. 2021, 17, 367–376. [Google Scholar] [CrossRef]
  140. Shahcheraghi, S.H.; Salemi, F.; Peirovi, N.; Ayatollahi, J.; Alam, W.; Khan, H.; Saso, L. Nrf2 regulation by curcumin: Molecular aspects for therapeutic prospects. Molecules 2021, 27, 167. [Google Scholar] [CrossRef]
  141. Tang, W.W.; Huang, F.F.; Haedi, A.R.; Shi, Q.Y. The effect of curcumin supplementation on endothelial function and blood pressure in patients with metabolic disorders: A meta-analysis of meta-analyses. Prostaglandins Other Lipid Mediat. 2024, 175, 106900. [Google Scholar] [CrossRef]
  142. Hegde, M.; Girisa, S.; BharathwajChetty, B.; Vishwa, R.; Kunnumakkara, A.B. Curcumin formulations for better bioavailability: What we learned from clinical trials thus far? ACS Omega 2023, 8, 10713–10746. [Google Scholar] [CrossRef]
  143. Yakubu, J.; Pandey, A.V. Innovative delivery systems for curcumin: Exploring nanosized and conventional formulations. Pharmaceutics 2024, 16, 637. [Google Scholar] [CrossRef]
  144. Dehzad, M.J.; Ghalandari, H.; Askarpour, M. Curcumin/turmeric supplementation could improve blood pressure and endothelial function: A grade-assessed systematic review and dose–response meta-analysis of randomized controlled trials. Clin. Nutr. ESPEN 2024, 59, 194–207. [Google Scholar] [CrossRef]
  145. Karimi, A.; Jazani, A.M.; Darzi, M.; Azgomi, R.N.; Vajdi, M. Effects of curcumin on blood pressure: A systematic review and dose-response meta-analysis. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 2089–2101. [Google Scholar] [CrossRef]
  146. Gimblet, C.J.; Kruse, N.T.; Geasland, K.; Michelson, J.; Sun, M.; Ten Eyck, P.; Linkenmeyer, C.; Mandukhail, S.R.; Rossman, M.J.; Sambharia, M.; et al. Curcumin supplementation and vascular and cognitive function in chronic kidney disease: A randomized controlled trial. Antioxidants 2024, 13, 983. [Google Scholar] [CrossRef]
  147. Lazo, O.L.; White, P.F.; Lee, C.; Eng, H.C.; Matin, J.M.; Lin, C.; Del Cid, F.; Yumul, R. Use of herbal medication in the perioperative period: Potential adverse drug interactions. J. Clin. Anesth. 2024, 95, 111473. [Google Scholar] [CrossRef]
  148. Chow, S.L.; Bozkurt, B.; Baker, W.L.; Bleske, B.E.; Breathett, K.; Fonarow, G.C.; Greenberg, B.; Khazanie, P.; Leclerc, J.; Morris, A.A.; et al. Complementary and alternative medicines in the management of heart failure: A scientific statement from the American Heart Association. Circulation 2023, 147, e4–e30. [Google Scholar] [CrossRef] [PubMed]
  149. Kim, H.; Kang, S.; Go, G.W. Exploring the multifaceted role of ginkgolides and bilobalide from Ginkgo biloba in mitigating metabolic disorders. Food Sci. Biotechnol. 2024, 33, 2903–2917. [Google Scholar] [CrossRef] [PubMed]
  150. Vogensen, S.B.; Strømgaard, K.; Shindou, H.; Jaracz, S.; Suehiro, M.; Ishii, S.; Shimizu, T.; Nakanishi, K. Preparation of 7-substituted ginkgolide derivatives: Potent platelet activating factor (PAF) receptor antagonists. J. Med. Chem. 2003, 46, 601–608. [Google Scholar] [CrossRef] [PubMed]
  151. National Center for Complementary and Integrative Health (NCCIH). Ginkgo: Usefulness and Safety. Updated February 2025. Available online: https://www.nccih.nih.gov/health/ginkgo (accessed on 8 October 2025).
  152. Hatfield, J.; Saad, S.; Housewright, C. Dietary supplements and bleeding. Bayl. Univ. Med. Cent. Proc. 2022, 35, 802–807. [Google Scholar] [CrossRef]
  153. Stoddard, G.J.; Archer, M.; Shane-McWhorter, L.; Bray, B.E.; Redd, D.F.; Proulx, J.; Zeng-Treitler, Q. Ginkgo and warfarin interaction in a large veterans administration population. AMIA Annu. Symp. Proc. 2015, 2015, 1174. [Google Scholar]
  154. Mai, N.T.; Hieu, N.V.; Ngan, T.T.; Van Anh, T.; Van Linh, P.; Thu Phuong, N.T. Impact of Ginkgo biloba drug interactions on bleeding risk and coagulation profiles: A comprehensive analysis. PLoS ONE 2025, 20, e0321804. [Google Scholar] [CrossRef]
  155. Bent, S.; Goldberg, H.; Padula, A.; Avins, A.L. Spontaneous bleeding associated with Ginkgo biloba: A case report and systematic review of the literature. J. Gen. Intern. Med. 2005, 20, 657–661. [Google Scholar] [CrossRef]
  156. Marx, W.; McKavanagh, D.; McCarthy, A.L.; Bird, R.; Ried, K.; Chan, A.; Isenring, L. The effect of ginger (Zingiber officinale) on platelet aggregation: A systematic literature review. PLoS ONE 2015, 10, e0141119, Correction in PLoS ONE 2015, 10, e0143675. [Google Scholar]
  157. Modi, M.; Modi, K. Ginger Root. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK565886/ (accessed on 29 August 2025).
  158. Rubin, D.; Patel, V.; Dietrich, E. Effects of oral ginger supplementation on the INR. Case Rep. Med. 2019, 2019, 8784029. [Google Scholar] [CrossRef]
  159. Chua, Y.T.; Ang, X.L.; Zhong, X.M.; Khoo, K.S. Interaction between warfarin and Chinese herbal medicines. Singap. Med. J. 2015, 56, 11. [Google Scholar] [CrossRef]
  160. Mar, P.L.; Gopinathannair, R.; Gengler, B.E.; Chung, M.K.; Perez, A.; Dukes, J.; Ezekowitz, M.D.; Lakkireddy, D.; Lip, G.Y.; Miletello, M.; et al. Drug interactions affecting oral anticoagulant use. Circ. Arrhythmia Electrophysiol. 2022, 15, e007956. [Google Scholar] [CrossRef]
  161. Drugs.com. Drug Interaction Report: Clopidogrel and Ginger (Professional Version). Available online: https://www.drugs.com/interactions-check.php?drug_list=705-0,1173-0&professional=1 (accessed on 8 October 2025).
  162. Hussain, Y.; Abdullah Khan, F.; Alsharif, K.F.; Alzahrani, K.J.; Saso, L.; Khan, H. Regulatory effects of curcumin on platelets: An update and future directions. Biomedicines 2022, 10, 3180. [Google Scholar] [CrossRef] [PubMed]
  163. Shah, B.H.; Nawaz, Z.; Pertani, S.A.; Roomi, A.; Mahmood, H.; Saeed, S.A.; Gilani, A.H. Inhibitory effect of curcumin, a food spice from turmeric, on platelet-activating factor-and arachidonic acid-mediated platelet aggregation through inhibition of thromboxane formation and Ca2+ signaling. Biochem. Pharmacol. 1999, 58, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  164. Talasaz, A.H.; McGonagle, B.; HajiQasemi, M.; Ghelichkhan, Z.A.; Sadeghipour, P.; Rashdi, S.; Cuker, A.; Lech, T.; Goldhaber, S.Z.; Jennings, D.L.; et al. Pharmacokinetic and pharmacodynamic interactions between food or herbal products and oral anticoagulants: Evidence review, practical recommendations, and knowledge gaps. In Seminars in Thrombosis and Hemostasis; Thieme Medical Publishers, Inc.: New York, NY, USA, 2024. [Google Scholar]
  165. Medsafe. Turmeric. Early Warning System. 2018. Available online: https://medsafe.govt.nz/safety/ews/2018/Turmeric.asp (accessed on 8 October 2025).
  166. Flory, S.; Männle, R.; Frank, J. The inhibitory activity of curcumin on P-glycoprotein and its uptake by and efflux from LS180 cells is not affected by its galenic formulation. Antioxidants 2021, 10, 1826. [Google Scholar] [CrossRef] [PubMed]
  167. Hsieh, Y.W.; Huang, C.Y.; Yang, S.Y.; Peng, Y.H.; Yu, C.P.; Chao, P.D.; Hou, Y.C. Oral intake of curcumin markedly activated CYP 3A4: In vivo and ex-vivo studies. Sci. Rep. 2014, 4, 6587. [Google Scholar] [CrossRef]
  168. Nowinski, K.; Chaireti, R. Discrepancies in Recommendations on Pharmacokinetic Drug Interactions for Anticancer Medications and Direct Oral Anticoagulants (DOAC): A Comparative Analysis of Different Clinical Decision Support Systems and Sources. Pharmaceuticals 2025, 18, 1044. [Google Scholar] [CrossRef]
  169. Cummings, K.C., III; Keshock, M.; Ganesh, R.; Sigmund, A.; Kashiwagi, D.; Devarajan, J.; Grant, P.J.; Urman, R.D.; Mauck, K.F. Preoperative management of surgical patients using dietary supplements: Society for Perioperative Assessment and Quality Improvement (SPAQI) consensus statement. Mayo Clin. Proc. 2021, 96, 1342–1355. [Google Scholar] [CrossRef]
  170. Yang, Y.M.; Chen, J.Z.; Wang, X.X.; Wang, S.J.; Hu, H.; Wang, H.Q. Resveratrol attenuates thromboxane A2 receptor ago-nist-induced platelet activation by reducing phospholipase C activity. Eur. J. Pharmacol. 2008, 583, 148–155. [Google Scholar] [CrossRef]
  171. Gresele, P.; Pignatelli, P.; Guglielmini, G.; Carnevale, R.; Mezzasoma, A.M.; Ghiselli, A.; Momi, S.; Violi, F. Resveratrol, at Concentrations Attainable with Moderate Wine Consumption, Stimulates Human Platelet Nitric Oxide Production3. J. Nutr. 2008, 138, 1602–1608. [Google Scholar] [CrossRef] [PubMed]
  172. Chiba, T.; Kimura, Y.; Suzuki, S.; Tatefuji, T.; Umegaki, K. Trans-resveratrol enhances the anticoagulant activity of warfarin in a mouse model. J. Atheroscler. Thromb. 2016, 23, 1099–1110. [Google Scholar] [CrossRef] [PubMed]
  173. Shaito, A.; Posadino, A.M.; Younes, N.; Hasan, H.; Halabi, S.; Alhababi, D.; Al-Mohannadi, A.; Abdel-Rahman, W.M.; Eid, A.H.; Nasrallah, G.K.; et al. Potential adverse effects of resveratrol: A literature review. Int. J. Mol. Sci. 2020, 21, 2084. [Google Scholar] [CrossRef] [PubMed]
  174. Chow, H.S.; Garland, L.L.; Hsu, C.H.; Vining, D.R.; Chew, W.M.; Miller, J.A.; Perloff, M.; Crowell, J.A.; Alberts, D.S. Resveratrol modulates drug-and carcinogen-metabolizing enzymes in a healthy volunteer study. Cancer Prev. Res. 2010, 3, 1168–1175. [Google Scholar] [CrossRef]
  175. Guthrie, A.R.; Chow, H.H.; Martinez, J.A. Effects of resveratrol on drug-and carcinogen-metabolizing enzymes, implica-tions for cancer prevention. Pharmacol. Res. Perspect. 2017, 5, e00294. [Google Scholar] [CrossRef]
  176. Aires, V.; Colin, D.J.; Doreau, A.; Di Pietro, A.; Heydel, J.M.; Artur, Y.; Latruffe, N.; Delmas, D. P-glycoprotein 1 affects chemoactivities of resveratrol against human colorectal cancer cells. Nutrients 2019, 11, 2098. [Google Scholar] [CrossRef]
  177. Javaid, M.; Kadhim, K.; Bawamia, B.; Cartlidge, T.; Farag, M.; Alkhalil, M. Bleeding Risk in Patients Receiving Omega-3 Polyunsaturated Fatty Acids: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. J. Am. Heart Assoc. 2024, 13, e032390. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  178. Khan, S.U.; Lone, A.N.; Khan, M.S.; Virani, S.S.; Blumenthal, R.S.; Nasir, K.; Miller, M.; Michos, E.D.; Ballantyne, C.M.; Boden, W.E.; et al. Effect of omega-3 fatty acids on cardiovascular outcomes: A systematic review and meta-analysis. eClinicalMedicine 2021, 38, 100997. [Google Scholar] [CrossRef]
  179. Chang, J.P.-C.; Tseng, P.-T.; Ze, B.-S. Safety of Supplementation of Omega-3 Polyunsaturated Fatty Acids: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv. Nutr. 2023, 14, 1326–1336. [Google Scholar] [CrossRef]
  180. Begtrup, K.M.; Krag, A.E.; Hvas, A.M. No impact of fish oil supplements on bleeding risk: A systematic review. Dan. Med. J. 2017, 64, A5366. [Google Scholar] [PubMed]
  181. Ouagueni, A.; Al-Zoubi, R.M.; Zarour, A.; Al-Ansari, A.; Bawadi, H. Effects of Omega-3 Polyunsaturated Fatty Acids, Docosahexaenoic Acid and Eicosapentaenoic Acid, on Post-Surgical Complications in Surgical Trauma Patients: Mechanisms, Nutrition, and Challenges. Mar. Drugs 2024, 22, 207. [Google Scholar] [CrossRef] [PubMed]
  182. Lichtenstein, J.; Sterpu, I.; Lindqvist, P.G. Does Omega-3 supplementation increase profuse postpartum hemorrhage? A hospital-based register study. Acta Obstet. Gynecol. Scand. 2024, 103, e14987. [Google Scholar] [CrossRef] [PubMed]
  183. Gross, B.W.; Gillio, M.; Rinehart, C.D.; Lynch, C.A.; Rogers, F.B. Omega-3 Fatty Acid Supplementation and Warfarin: A Lethal Combination in Traumatic Brain Injury. J. Trauma Nurs. 2017, 24, 15–18. [Google Scholar] [CrossRef]
  184. Ried, K.; Toben, C.; Fakler, P. Effect of Garlic on Serum Lipids: An Updated Meta-Analysis. Nutr. Rev. 2013, 71, 282–299. [Google Scholar] [CrossRef]
  185. Macan, H.; Uykimpang, R.; Alconcel, M.; Takasu, J.; Razon, R.; Amagase, H.; Niihara, Y. Aged garlic extract may be safe for patients on warfarin therapy. J. Nutr. 2006, 136, 793S–795S. [Google Scholar] [CrossRef]
  186. Tan, C.S.S.; Lee, S.W.H. Warfarin and Food, Herbal or Dietary Supplement Interactions: A Systematic Review. Br. J. Clin. Pharmacol. 2020, 86, 1165–1182. [Google Scholar] [CrossRef]
  187. Li, X.; Long, J.; Gao, Q.; Pan, M.; Wang, J.; Yang, F.; Zhang, Y. Nattokinase Supplementation and Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Rev. Cardiovasc. Med. 2023, 24, 234. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  188. Zhou, Y.; Fu, Z.; Dou, P.; Yu, C.; Dong, X.; Hong, H.; Luo, Y.; Tan, Y. Decoding nattokinase efficacy: From digestion and absorption to lipid pathway modulation in high-fat diet-induced atherosclerosis. Food Biosci. 2025, 66, 106203. [Google Scholar] [CrossRef]
  189. Chen, H.; McGowan, E.M.; Ren, N.; Lal, S.; Nassif, N.; Shad-Kaneez, F.; Qu, X.; Lin, Y. Nattokinase: A Promising Alternative in Prevention and Treatment of Cardiovascular Diseases. Biomark. Insights 2018, 13, 1177271918785130. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  190. Wu, H.; Zhang, Q.; Suo, H.; Xu, F.; Huang, W.; Wang, D.O. Nattokinase as a Functional Food Ingredient: Therapeutic Ap-plications and Mechanisms in Age-Related Diseases. Food Sci. Hum. Wellness 2024, 13, 2401–2409. [Google Scholar] [CrossRef]
  191. Zubiaur, P.; Rodríguez-Antona, C.; Boone, E.C.; Daly, A.K.; Tsermpini, E.E.; Khasawneh, L.Q.; Sangkuhl, K.; Duconge, J.; Botton, M.R.; Savieo, J.; et al. PharmVar GeneFocus: CYP4F2. Clin. Pharmacol. Ther. 2024, 116, 963–975. [Google Scholar] [CrossRef] [PubMed]
  192. Medsafe. Can Vitamin E Cause Bleeding? 2022. Available online: https://www.medsafe.govt.nz/profs/PUArticles/June2022/Can-Vitamin-E-cause-bleeding.html (accessed on 8 October 2025).
  193. Pastori, D.; Carnevale, R.; Cangemi, R.; Saliola, M.; Nocella, C.; Bartimoccia, S.; Vicario, T.; Farcomeni, A.; Violi, F.; Pignatelli, P. Vitamin E serum levels and bleeding risk in patients receiving oral anticoagulant therapy: A retrospective cohort study. J. Am. Heart Assoc. 2013, 2, e000364. [Google Scholar] [CrossRef] [PubMed]
  194. Abrol, R.; Kaushik, R.; Goel, D.; Sama, S.; Kaushik, R.M.; Kala, M. Vitamin E-induced coagulopathy in a young patient: A case report. J. Med. Case Rep. 2023, 17, 107. [Google Scholar] [CrossRef] [PubMed]
  195. Özkan, M.; Güneş, H. Do the natural and herbal remedies used for fighting against COVID-19 pose a risk for surgical patients? J. Herb. Med. 2024, 46, 100902. [Google Scholar] [CrossRef]
  196. van Heeswijk, R.P.; Cooper, C.L.; Foster, B.C.; Chauhan, B.M.; Shirazi, F.; Seguin, I.; Phillips, E.J.; Mills, E. Effect of high-dose vitamin C on hepatic cytochrome P450 3A4 activity. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2005, 25, 1725–1728. [Google Scholar] [CrossRef]
  197. Sharma, Y.; Sumanadasa, S.; Shahi, R.; Woodman, R.; Mangoni, A.A.; Bihari, S.; Thompson, C. Efficacy and safety of vitamin C supplementation in the treatment of community-acquired pneumonia: A systematic review and meta-analysis with trial sequential analysis. Sci. Rep. 2024, 14, 11846. [Google Scholar] [CrossRef]
  198. Yuan, C.S.; Wei, G.; Dey, L.; Karrison, T.; Nahlik, L.; Maleckar, S.; Kasza, K.; Ang-Lee, M.; Moss, J. Brief communication: American ginseng reduces warfarin’s effect in healthy patients: A randomized, controlled trial. Ann. Intern. Med. 2004, 141, 23–27. [Google Scholar] [CrossRef]
  199. Malati, C.Y.; Robertson, S.M.; Hunt, J.D.; Chairez, C.; Alfaro, R.M.; Kovacs, J.A.; Penzak, S.R. Influence of Panax ginseng on cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp) activity in healthy participants. J. Clin. Pharmacol. 2012, 52, 932–939. [Google Scholar] [CrossRef] [PubMed]
  200. Park, J.D. Metabolism and drug interactions of Korean ginseng based on the pharmacokinetic properties of ginseno-sides: Current status and future perspectives. J. Ginseng Res. 2024, 48, 253–265. [Google Scholar] [CrossRef] [PubMed]
  201. Dong, H.; Ma, J.; Li, T.; Xiao, Y.; Zheng, N.; Liu, J.; Gao, Y.; Shao, J.; Jia, L. Global deregulation of ginseng products may be a safety hazard to warfarin takers: Solid evidence of ginseng-warfarin interaction. Sci. Rep. 2017, 7, 5813. [Google Scholar] [CrossRef] [PubMed]
  202. Sood, B.; Patel, P.; Keenaghan, M. Coenzyme Q10. In StatPearls [Internet]; StatPearls Publishing: Treasure Island, FL, USA, 2025. Available online: https://www.ncbi.nlm.nih.gov/books/NBK531491/ (accessed on 5 September 2025).
  203. Engelsen, J.; Nielsen, J.D.; Hansen, K.F. Effect of Coenzyme Q10 and Ginkgo biloba on warfarin dosage in patients on long-term warfarin treatment. A randomized, double-blind, placebo-controlled cross-over trial. Ugeskr. Laeger 2003, 165, 1868–1871. [Google Scholar] [PubMed]
  204. Volak, L.P.; Ghirmai, S.; Cashman, J.R.; Court, M.H. Curcuminoids inhibit multiple human cytochromes P450, UDP-glucuronosyltransferase, and sulfotransferase enzymes, whereas piperine is a relatively selective CYP3A4 inhibitor. Drug Metab. Dispos. 2008, 36, 1594–1605. [Google Scholar] [CrossRef]
  205. Sanchez-Fuentes, A.; Rivera-Caravaca, J.M.; Lopez-Galvez, R.; Marin, F.; Roldan, V. Non-vitamin K antagonist oral anticoagulants and drug-food interactions: Implications for clinical practice and potential role of probiotics and prebiotics. Front. Cardiovasc. Med. 2022, 8, 787235. [Google Scholar] [CrossRef]
  206. Shi, L.; Wang, W.; Jing, C.; Hu, J.; Liao, X. Berberine and health outcomes: An overview of systematic reviews. BMC Complement. Med. Ther. 2025, 25, 147. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  207. Paul, M.; Hemshekhar, M.; Kemparaju, K.; Girish, K.S. Berberine mitigates high glucose-potentiated platelet aggregation and apoptosis by modulating aldose reductase and NADPH oxidase activity. Free Radic. Biol. Med. 2019, 130, 196–205. [Google Scholar] [CrossRef]
  208. Rui, R.; Yang, H.; Liu, Y.; Zhou, Y.; Xu, X.; Li, C.; Liu, S. Effects of Berberine on Atherosclerosis. Front. Pharmacol. 2021, 12, 764175. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  209. Wang, C.; Wu, Y.-B.; Wang, A.-P.; Jiang, J.-D.; Kong, W.-J. Evaluation of Anticoagulant and Antithrombotic Activities of Berberine: A Focus on the Ameliorative Effect on Blood Hypercoagulation. Int. J. Pharmacol. 2018, 14, 1087–1098. [Google Scholar] [CrossRef]
  210. Ye, Y.; Liu, X.; Wu, N.; Han, Y.; Wang, J.; Yu, Y.; Chen, Q. Efficacy and Safety of Berberine Alone for Several Metabolic Disorders: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Front. Pharmacol. 2021, 12, 653887. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  211. Li, Z.; Wang, Y.; Xu, Q.; Ma, J.; Li, X.; Yan, J.; Tian, Y.; Wen, Y.; Chen, T. Berberine and health outcomes: An umbrella review. Phytother Res. 2023, 37, 2051–2066. [Google Scholar] [CrossRef] [PubMed]
  212. The Therapeutic Effects of Statins and Berberine on the Hyperlipemia. Available online: https://clinicaltrials.gov/study/NCT01697735 (accessed on 20 September 2025).
  213. Zhuang, W.; Liu, S.; Zhao, X.; Sun, N.; He, T.; Wang, Y.; Jia, B.; Lin, X.; Chu, Y.; Xi, S. Interaction Between Chinese Medicine and Warfarin: Clinical and Research Update. Front. Pharmacol. 2021, 12, 751107. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  214. Chen, T.L.; Yeh, C.C.; Lin, C.S.; Shih, C.C.; Liao, C.C. Effects of red yeast rice prescription (LipoCol Forte) on adverse outcomes of surgery. QJM 2019, 112, 253–259. [Google Scholar] [CrossRef] [PubMed]
  215. Mazzanti, G.; Moro, P.A.; Raschi, E.; Da Cas, R.; Menniti-Ippolito, F. Adverse reactions to dietary supplements containing red yeast rice: Assessment of cases from the Italian surveillance system. Br. J. Clin. Pharmacol. 2017, 83, 894–908. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  216. Twarużek, M.; Ałtyn, I.; Kosicki, R. Dietary Supplements Based on Red Yeast Rice—A Source of Citrinin? Toxins 2021, 13, 497. [Google Scholar] [CrossRef]
  217. Rowan, C.G.; Brunelli, S.M.; Munson, J.; Flory, J.; Reese, P.P.; Hennessy, S.; Lewis, J.; Mines, D.; Barrett, J.S.; Bilker, W.; et al. Clinical importance of the drug interaction between statins and CYP3A4 inhibitors: A retrospective cohort study in the Health Improvement Network. Pharmacoepidemiol. Drug Saf. 2012, 21, 494–506. [Google Scholar] [CrossRef]
  218. Fogacci, F.; Banach, M.; Mikhailidis, D.P.; Bruckert, E.; Toth, P.P.; Watts, G.F.; Reiner, Ž.; Mancini, J.; Rizzo, M.; Mitchenko, O.; et al. Safety of red yeast rice supplementation: A systematic review and meta-analysis of randomized controlled trials. Pharmacol. Res. 2019, 143, 1–16. [Google Scholar] [CrossRef]
  219. EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA); Turck, D.; Bohn, T.; Cámara, M.; Castenmiller, J.; De Henauw, S.; Hirsch-Ernst, K.I.; Jos, Á.; Mangelsdorf, I.; McNulty, B.; et al. Scientific Opinion on additional scientific data related to the safety of monacolins from red yeast rice submitted pursuant to Article 8 of Regulation (EC) No 1925/2006. EFSA J. 2025, 23, 9276. [Google Scholar] [CrossRef]
  220. Renaud, D.; Höller, A.; Michel, M. Potential Drug–Nutrient Interactions of 45 Vitamins, Minerals, Trace Elements, and Associated Dietary Compounds with Acetylsalicylic Acid and Warfarin—A Review of the Literature. Nutrients 2024, 16, 950. [Google Scholar] [CrossRef]
  221. Fang, F.; Yang, H.; Wang, X.; Zhao, T.; Zhao, P.; Liu, X. Extracellular Vesicles in Atherosclerosis: From Pathogenesis to Theranostic Applications. Small 2025, 21, e2504761. [Google Scholar] [CrossRef] [PubMed]
  222. Zhang, Y.; Zhang, W.; Wu, Z.; Chen, Y. Diversity of extracellular vesicle sources in atherosclerosis: Role and therapeutic application. Angiogenesis 2025, 28, 34. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The overlap between anti-atherosclerotic mechanisms and antiplatelet effects.
Figure 1. The overlap between anti-atherosclerotic mechanisms and antiplatelet effects.
Ijms 26 10183 g001
Table 1. Clinical Trials Evaluating the Cardioprotective Role of Omega-3 Fatty Acid Supplements.
Table 1. Clinical Trials Evaluating the Cardioprotective Role of Omega-3 Fatty Acid Supplements.
FeatureASCEND Trial [39]REDUCE-IT Trial [40,41]STRENGTH Trial [42]VITAL Trial
[43]
Patient
Characteristics
Diabetic, no prior CV eventsDiabetic with additional CV risk factors or patients with established CVDHigh-risk CVD patients, statin-treatedGenerally healthy, men ≥ 50 yrs, women ≥ 55 yrs
Number of
Patients
15,480817913,07825,871
Dosage1 g/day Omega-3 (EPA 460 mg + DHA 380 mg) and placebo
-a subgroup with 100 mg/day Aspirin
4 g/day Icosapent Ethyl (EPA only) and
placebo-mineral oil
4 g/day Omega-3 (EPA + DHA) and
placebo—corn oil
1 g/day Omega-3 (Omacor, fish oil) with or without Vitamin D3: 2000 IU/day and
placebo
Follow-up
Duration
~7.4 years4.9 years~3.5 years5.3 years
Main FindingsAspirin reduced CV events
Omega-3 showed no CV benefit
Strong evidence supporting this dose of EPA for reducing cardio-vascular events in high-risk patientsOmega-3 showed no CV benefitOmega-3 showed no reduction in major cardiovascular events, but a modest reduction in myocardial infarction in those with low baseline fish intake
Mechanism of action for
cardiovascular benefits
Aspirin: antithrombotic
Omega-3: the relatively low dose may have been inadequate to produce meaningful cardiovascular benefit
Reduced triglycerides
anti-inflammatory
improved endothelial function
plaque stabilization antioxidant antithrombotic
DHA may counteract EPA’s beneficial effects, so
formulation rather than dose explains the lack of cardiovascular benefits
Omega-3: the relatively low dose may have been inadequate to produce meaningful cardiovascular benefit
Safety/
Adverse
Effects
Increased major bleeding after aspirin intake
No major issues with omega-3
Increased atrial fibrillation risk, a trend toward higher serious bleeding riskIncreased atrial fibrillation risk, gastrointestinal side effects No major safety concerns
Table 2. Anti-atherosclerotic mechanisms of selected nutraceuticals; hemostatic actions (antiplatelet, fibrinolytic) highlighted in red.
Table 2. Anti-atherosclerotic mechanisms of selected nutraceuticals; hemostatic actions (antiplatelet, fibrinolytic) highlighted in red.
CompoundKey Mechanisms/ActionsReferences
Omega-3 fatty acids (EPA, DHA)↓ Triglycerides, modest ↑ HDL-C [27]
Hepatic de novo lipogenesis and increased postprandial fatty acid oxidation [39]
↑ Resolvin = anti-inflammatory effects [32]
↓ TxAz synthesis & GPVI modulation, ↓ Ca2+ influx & membrane fluidity[29,32,36]
Berberine↓ Triglycerides and LDL-C, modest ↑ HDL-C [46]
↓ ROS anti-inflammatory effects [47]
Improves insulin sensitivity[45]
Blocks GPIIb/IIIa, modulates PI3Kβ/Ca2+[48]
Red yeast riceMonacolin K acts like a statin = ↓LDL-C [50]
↓ TXB2 and ↑ antithrombin III [51]
Garlic ↓ TC, ↓ TG, ↓ LDL-C slightly ↑ HDL-C [57]
↓ ROS, ↓ NF-kB, ↑ NO, ↑ H2S ↑ ANP, ↓ SRAA, ↑ VSMC proliferation ⟶ vasodilation and lower blood pressure[62]
Decrease the absorption of cholesterol, -HMG-CoA Reductase [53]
Increased secondary bile acids, increase GLP-1 [61]
Inhibits platelet activation & GP IIb/IIIa binding[63]
Inhibits fibrinogen binding and platelet shape change[60]
Nattokinase (NK)↓ TG, ↓ LDL-C, ↓ ox LDL, ↑ HDL-C[188]
Down-regulated PXDN and PNLIP [189]
-HMGcoA reductase, +LPLase [64]
↓ ROS, ↓ IL-6 [65]
↓ Blood pressure [71]
↑ tPA release, ↓ TXA2, ↓ Fibrinogen & clotting factors (VII, VIII)[67,69,70]
Ginkgo biloba↑ NO bioavailability[114]
↓ ICAM-1, VCAM-1[115]
↓ ROS, ↓cytokines[116]
↓ PAF[150,151]
Ginger↓ CRP, ↓ Il-6, ↓ TNFα[126]
↓ ROS[124]
↓ TXA2 GPVI modulation[157,158]
Ginseng↑ NO [131]
↓ ICAM-1, VCAM-1[132]
↓ GPIIb/IIIa, ↓ TXB2, GPVI modulation [133]
Legend: ↑ = increase and ↓ = decrease. The abbreviations are defined at the end of the article.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Dobre, M.-Z.; Virgolici, B.; Doicin, I.-C.; Vîrgolici, H.; Stanescu-Spinu, I.-I. Navigating the Effects of Anti-Atherosclerotic Supplements and Acknowledging Associated Bleeding Risks. Int. J. Mol. Sci. 2025, 26, 10183. https://doi.org/10.3390/ijms262010183

AMA Style

Dobre M-Z, Virgolici B, Doicin I-C, Vîrgolici H, Stanescu-Spinu I-I. Navigating the Effects of Anti-Atherosclerotic Supplements and Acknowledging Associated Bleeding Risks. International Journal of Molecular Sciences. 2025; 26(20):10183. https://doi.org/10.3390/ijms262010183

Chicago/Turabian Style

Dobre, Maria-Zinaida, Bogdana Virgolici, Ioana-Cristina Doicin, Horia Vîrgolici, and Iulia-Ioana Stanescu-Spinu. 2025. "Navigating the Effects of Anti-Atherosclerotic Supplements and Acknowledging Associated Bleeding Risks" International Journal of Molecular Sciences 26, no. 20: 10183. https://doi.org/10.3390/ijms262010183

APA Style

Dobre, M.-Z., Virgolici, B., Doicin, I.-C., Vîrgolici, H., & Stanescu-Spinu, I.-I. (2025). Navigating the Effects of Anti-Atherosclerotic Supplements and Acknowledging Associated Bleeding Risks. International Journal of Molecular Sciences, 26(20), 10183. https://doi.org/10.3390/ijms262010183

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