Next Article in Journal
The lncRNAs at X Chromosome Inactivation Center: Not Just a Matter of Sex Dosage Compensation
Previous Article in Journal
Surface Modification of Bacterial Cellulose for Biomedical Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Impacts of Commonly Used Edible Plants on the Modulation of Platelet Function

by
Dina A. I. Albadawi
1,
Divyashree Ravishankar
1,
Thomas M. Vallance
1,
Ketan Patel
2,
Helen M. I. Osborn
1,* and
Sakthivel Vaiyapuri
1,*
1
School of Pharmacy, University of Reading, Reading RG6 6UB, UK
2
School of Biological Sciences, University of Reading, Reading RG6 6UB, UK
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(2), 605; https://doi.org/10.3390/ijms23020605
Submission received: 11 November 2021 / Revised: 28 December 2021 / Accepted: 3 January 2022 / Published: 6 January 2022
(This article belongs to the Section Molecular Plant Sciences)

Abstract

:
Cardiovascular diseases (CVDs) are a primary cause of deaths worldwide. Thrombotic diseases, specifically stroke and coronary heart diseases, account for around 85% of CVDs-induced deaths. Platelets (small circulating blood cells) are responsible for the prevention of excessive bleeding upon vascular injury, through blood clotting (haemostasis). However, unnecessary activation of platelets under pathological conditions, such as upon the rupture of atherosclerotic plaques, results in thrombus formation (thrombosis), which can cause life threatening conditions such as stroke or heart attack. Therefore, antiplatelet medications are usually prescribed for people who are at a high risk of thrombotic diseases. The currently used antiplatelet drugs are associated with major side effects such as excessive bleeding, and some patients are resistant to these drugs. Therefore, numerous studies have been conducted to develop new antiplatelet agents and notably, to establish the relationship between edible plants, specifically fruits, vegetables and spices, and cardiovascular health. Indeed, healthy and balanced diets have proven to be effective for the prevention of CVDs in diverse settings. A high intake of fruits and vegetables in regular diet is associated with lower risks for stroke and coronary heart diseases because of their plethora of phytochemical constituents. In this review, we discuss the impacts of commonly used selected edible plants (specifically vegetables, fruits and spices) and/or their isolated compounds on the modulation of platelet function, haemostasis and thrombosis.

1. Introduction

According to the World Health Organisation, cardiovascular diseases (CVDs) are a major cause of deaths worldwide [1]. In 2017, more than 85% of total CVDs-associated deaths were caused by thrombotic diseases (primarily strokes and coronary heart diseases), and they are largely triggered by the rupture of atherosclerotic plaques [1,2]. Numerous studies have demonstrated that the effective management of risk factors for CVDs is critical for their prevention and management, as well as to improve the quality of life for patients in the long-term [3]. An unhealthy diet is a key risk factor for CVDs, as the regular consumption of food which has high levels of saturated fats, sugar, sodium, animal proteins and low in fibre (fruit and vegetables) can cause hypertension, hyperglycaemia, hyperlipidaemia and obesity, which ultimately result in CVDs [4,5,6].
CVDs are a group of pathological conditions that affect the heart and blood vessels due to both non-modifiable and modifiable risk factors. Non-modifiable risk factors include age, gender, ethnicity, and family history for CVDs. However, modifiable risk factors involve high blood pressure, increased cholesterol level, high blood glucose, unhealthy diet, obesity, reduced physical activities, smoking and stress [7]. The most common type of CVDs, coronary artery disease, is mainly caused by the formation/rupture of atherosclerotic plaques within coronary arteries leading to reduced blood supply to heart muscles [7]. Atherosclerotic plaques are formed due to the accumulation of low-density lipoproteins (LDL) in the arterial intima, which results in endothelial dysfunction, initiation of inflammatory responses and the formation of oxidised LDL [8]. As a result, immune cells such as monocytes are attracted to the damaged site by chemokines, and inflammatory markers are released from the affected region. The migrated monocytes differentiate into macrophages that engulf oxidised LDL and become foam cells [8,9]. The accumulation of an excessive amount of lipids over time causes necrosis, fibrous tissue formation and calcification which increases the plaque size and subsequently leads to a reduction in blood flow inside the affected arteries [7]. Notably, the plaque can rupture and trigger the formation of blood clots (thrombi) within the blood vessels. Thrombosis can restrict the blood flow to vital organs such as the heart and brain, and can cause myocardial infarction or ischemic stroke, respectively [10].
At the site of vascular damage, the subendothelial matrix and its contents, mainly collagen, are exposed to circulation. Collagen adheres to circulating platelets by binding to von Willebrand factor (vWF) (immobilised on fibrillar collagen type I and III), platelet glycoprotein (GP) Ib-V-IX receptor complex and GPVI receptor and facilitates their activation at the injury site [11]. As a result, a monolayer of platelets is formed to cover the damaged region. Notably, GPVI plays an essential role in collagen-mediated platelet activation, and also promotes a stable adhesion of collagen via integrin α2β1. GPVI-mediated signalling activates phosphoinositol-3-kinase (PI3K) and phospholipase Cγ2 leading to intracellular calcium mobilisation, inside-out signalling to integrin αIIbβ3, granule secretion and finally, platelet aggregate or thrombus formation [11,12]. In addition to collagen-mediated activation, more platelets are activated by autocrine and paracrine signalling through adenosine diphosphate (ADP) (released from platelet dense granules) and thromboxane A2 (TXA2, synthesised and released from activated platelets). Fibrinogen (present in plasma) is the major ligand for integrin αIIbβ3 on the surface of activated platelets and it promotes platelet aggregation by acting as a bridge between platelets [13]. In addition, thrombin is produced as a result of coagulation cascades and it activates more platelets via protease-activated receptors (PARs) and converts fibrinogen into fibrin to form a polymerised fibrin network, resulting in a stable clot/thrombus formation [14,15].
Platelets are small, anucleated circulating blood cells derived from megakaryocytes in the bone marrow. They have a life span of around 8–10 days and their normal count in blood is between 150–400 × 109/L. The primary function of platelets is to avoid excessive blood loss upon vascular injury by forming a blood clot under physiological conditions (this process is commonly known as ‘haemostasis’) [11]. However, as detailed above, platelets become unnecessarily activated under various pathological conditions, leading to thrombosis [11,16]. Therefore, antiplatelet medications are predominantly prescribed for the primary and secondary prevention of thrombotic diseases [17,18]. The most commonly prescribed antiplatelet drugs are aspirin and clopidogrel either as monotherapy (single drug) or dual therapy (two drugs) to achieve their optimal/maximal effects [19,20]. However, even dual antiplatelet treatments may not prevent the recurrence of thrombotic incidents in some patients and many patients develop resistance to aspirin and/or clopidogrel due to a number of reasons, including inadequate dosing, drug–drug interactions and poor patient compliance [21,22]. Moreover, excessive bleeding is a major side effect of these medications and may lead to the need for blood transfusion in some cases. The bioavailability of clopidogrel may be affected by co-administered drugs that are metabolised in the liver by CYP2C19 and CYP3A4/5 enzymes and this may affect the decision on prescribing it for some patients [22].
Notably, an unhealthy diet that consists of high sugar, sodium and saturated fats and lower amounts of fruit, vegetables, legumes, fibres, nuts and fish is a critical modifiable risk factor for several chronic diseases, specifically CVDs [23,24,25]. In most cases, a healthy and balanced diet is a part of the treatment/prevention plan for CVDs, including for thrombotic diseases [26]. Several studies have demonstrated the effects of plants-based diets in reducing risk factors for CVDs and the associated mortality rate [27]. In general, a daily intake of five servings of fruits/vegetables (plant-based foods) ranging from 400–800 g/day is recommended to reduce the risk for CVD [28]. The beneficial effects of fruits, vegetables and other plant-based foods are mainly related to their antioxidant, anti-inflammatory, hypotensive and hypoglycaemic effects as a result of a plethora of phytochemicals, including flavonoids, phenolic acids, alkaloids and glycosides as well as vitamins, minerals and fibres [29,30,31,32].
The outcomes of many observational studies demonstrated that a vegetarian diet has favourable effects on cardiovascular health to control obesity, hyperlipidaemia, hypertension and type II diabetes compared to non-vegetarian diets [33]. Vegetarians displayed lower body mass index (BMI), serum cholesterol level and diastolic blood pressure than non-vegetarians [33,34,35]. In addition, it is reported that vegetarian diets offer significantly lower risks for heart attacks and strokes [36,37,38]. Moreover, inflammatory biomarkers, including interleukin-6 (IL-6), C-reactive protein (CRP) and tumour necrosis factor-alpha (TNF-α) were significantly lower among vegetarians [39,40].
Overall, although antiplatelet drugs are the first line of defence for the prevention and treatment of CVDs (specifically thrombotic diseases), they are associated with serious side effects. Therefore, there is an urgent need to develop novel antiplatelet agents that are more effective, safer, and affordable for short and long-term management of thrombotic diseases/CVDs. Accordingly, numerous studies have investigated the impacts of various edible plants and/or their isolated active compounds on the modulation of platelet function. In this review, the effects of various vegetables, fruits, spices, and edible fungi and their isolated compounds in the modulation of platelet activation and thrombosis are discussed. Whilst some reviews were published on a specific plant compound or a group of compounds, specific plants and their effects on platelets, this review will focus on the effects of both plant extracts and their isolated compounds on the modulation of platelet activation under diverse settings in an integrated manner.

2. Vegetables

2.1. Onions

Allium cepa (onion) is one of the main Allium spices that were studied for their beneficial effects on human health, specifically on the cardiovascular system. Their antioxidant, antihypertensive, antiplatelet, anti-inflammatory and antihyperlipidemic effects were analysed in various settings [41,42]. Onions are a widely cultivated and consumed vegetable all over the world, although they originate from Central Asia. Traditionally, onions were used to treat cold, flu, dysentery, wound healing, and alleviate pain [42]. Several studies have explored the different classes of phytochemicals present in A. cepa including volatile oil, and sulphur-containing compounds such as methyl 5-methylfuryl sulphide and dimethyl disulphide which are responsible for their characteristic flavour. In addition, phenolic compounds such as phenolic acids (e.g., p-hydroxybenzoic acid and gallic acid) and flavonoids (e.g., anthocyanins, quercetin and kaempferol) were isolated and characterised from onions [43,44,45]. These active constituents are associated with the biological effects of A. cepa, including antioxidant, antibiotic, anti-cancer, anti-diabetic, anti-inflammatory and anti-allergic activities. In addition, they significantly reduce CVD risks through hypolipidemic, hypotensive, hypoglycaemic and antiplatelet effects [41,42].
To evaluate the antiplatelet effects of A. cepa bulb, different concentrations of its aqueous extracts (250 and 500 mg/mL) were tested in human isolated platelets using an aggregation assay upon stimulation with a TXA2 receptor agonist, U46619 (2 µM). Both concentrations of the extract (250 and 500 mg/mL) significantly inhibited platelet aggregation by around 85–100% [46]. In addition, an ethanolic extract of A. cepa bulb showed significant antiplatelet effects when 5 µg/mL collagen was used as an agonist in rat isolated platelets through reducing intracellular Ca2+ levels, cyclooxygenase 1 enzyme (COX-1) and TXA2 synthase activities. It also increased cAMP levels in a concentration dependent manner [47]. The anti-aggregatory effects of A. cepa are attributable to the abundant flavonoid, quercetin (Figure 1a) and its glycosides, quercetin-3,4′-O-diglucoside (Figure 1b) and quercetin-4′-O-monoglucoside (Figure 1c). These compounds were isolated from the methanolic extract of A. cepa and at a concentration of 2 mg/mL, they completely inhibited 6 µg/mL collagen-induced platelet (rat) aggregation [48].
Furthermore, to test the impact of cooking methods and cooking time on A. cepa bulb-mediated antiplatelet effects, conventional (200 °C) and microwave (500 W, which is almost equivalent to 200 °C) ovens were selected to cook the samples for 10, 20 or 30 min. For conventional oven cooking, A. cepa samples were divided into; whole (intact) bulb, chopped into quarters, and crushed samples. For microwave cooking, only the whole bulbs and crushed samples were tested. Then, all samples were tested in human whole blood aggregation upon stimulation with 1 µg/mL collagen. First, a raw crushed sample of A. cepa was tested and it significantly inhibited platelet aggregation by around 85%. Samples that were cooked using the conventional oven showed different effects on platelet aggregation compared to the raw samples as shown in Table 1. These data suggested that the antiplatelet effects of A. cepa bulb can be lost due to aggressive processing using high temperature. In addition, the long cooking time can change the anti-aggregatory effects of A. cepa. On the other hand, samples that were cooked using the microwave method did not exert any inhibitory effects on platelets, irrespective of the cooking time [49].
Furthermore, a human pilot study (n = 6) tested the acute effects of low (8.1 mg/L) and high (114.8 mg/L) quercetin (Figure 1a) contents in onion soups on human platelet activity via oral consumption. The anti-aggregatory effects of onion soups were evaluated in human isolated platelets upon activation with collagen (0.5, 1, 2 and 3 µg/mL) after 1 and 3 h of consumption. The soup with high quercetin content significantly inhibited platelet aggregation induced by different concentrations of collagen. In addition, the effect of both soups on tyrosine phosphorylation of spleen tyrosine kinase (Syk) and phospholipase C gamma 2 (PLCγ2) were evaluated, as they are crucial molecules in the signalling pathways of GPVI (a major collagen receptor). The high quercetin soup significantly inhibited the phosphorylation of Syk and PLCγ2 in 25 µg/mL collagen-induced platelets, when samples collected after 1 and 3 h of ingestion. In addition, the low quercetin soup insignificantly inhibited platelet aggregation induced by collagen while it stimulated tyrosine phosphorylation of Syk and PLCγ2 compared to the control at 1 and 3 h after ingestion (Figure 2) [50].

2.2. Garlic

Allium sativum (garlic) is a common used vegetable and plays a critical role in the traditional medicine of many ancient cultures, such as the Egyptian, Indian, Chinese, Sumerian, and Greek. It was widely used to treat persistent cough, arthritis, constipation, snakebites and as a general antibiotic [51]. Various studies demonstrated that A. sativum exerts antioxidant, anti-inflammatory, anti-tumor, antibiotic, hypoglycaemic and renal protective effects [51,52]. In addition, it was reported that A. sativum has positive effects on CVDs through its antioxidant, hypotensive and hypocholesterolaemic effects. These biological effects are linked to sulphur-containing phytochemicals (including alliin and allicin) and enzymes (including alliinase and peroxidase) as well as flavonoids (e.g., quercetin) in A. sativum [52]. The antiplatelet effects of aqueous and methanolic extracts of A. sativum bulbs were examined in human PRP aggregation using different agonists; 20 µM ADP, 190 µg/mL collagen and 20 µM epinephrine. The aqueous extracts (10 mg/mL) significantly reduced ADP-induced aggregation by around 86% but did not affect aggregation induced by other agonists. However, the methanolic extracts (10 mg/mL) significantly inhibited ADP, epinephrin and collagen-induced aggregation by approximately 89%, 66% and 32%, respectively. The antiaggregatory effects of the methanolic extracts were suggested to be as a result of high contents of alliin (Figure 3a) and allicin (Figure 3b) in this plant [53]. However, the tested concentrations of agonists were higher than the commonly used concentrations (ADP: 0.5–10 µM; collagen: 1–5 µg/mL; epinephrin: 0.5–10 µM) for platelet aggregation [53]. Therefore, the inhibitory effects were not apparent.
Allicin is an organosulfur compound that accounts for around 70% of total thiosulfinates [contain the functional group, R-S(O)-S-R] in A. sativum. It is produced upon the physical disruption (e.g., by crushing or cutting) of tissues of A. sativum as the alliinase enzyme converts alliin to allicin upon damage [53]. Allicin and alliin are the main compounds responsible for the biological activities of A. sativum [52,54,55]. In human PRP and isolated platelets, the effect of 40 µM allicin was tested using 5 µg/mL collagen or 10 µM ADP and 55 µM epinephrine (combined)-induced platelet aggregation. In PRP, allicin did not affect aggregation, whilst in isolated platelets allicin markedly inhibited aggregation by around 98% in collagen and ADP-epinephrine activated platelets which indicates that the anti-aggregatory effects of allicin may be affected by plasma proteins in PRP. In addition, 40 µM allicin demonstrated significant inhibitory effects on fibrinogen binding and P-selectin exposure by around 80% and 90%, respectively, in ADP-epinephrine activated platelets [55]. However, in this study the combined usage of ADP-epinephrine as agonists was not justified. Moreover, the epinephrine concentration that was used appears to be higher than the commonly used concentrations (0.5–10 µM).
In addition, N-feruloyltyramine (Figure 4a) is an amide alkaloid isolated from methanolic extracts of A. sativum and it is known to exhibit antioxidant, anti-fungal, anti-bacterial and cytotoxic effects [56]. This compound was tested in mouse whole blood along with its synthetic analogues, N-caffeoylnorephedrine (Figure 4b) and N-caffeoyltyramine (Figure 4c) to evaluate their effects on platelet function, specifically on COX-I enzyme and P-selectin exposure. N-feruloyltyramine and its analogues at the concentration of 0.05 µM significantly reduced the activity of COX-1. However, N-feruloyltyramine exhibited the highest inhibitory effect of around 43% compared to a COX-1 inhibitor, ibuprofen. In addition, N-feruloyltyramine, N-caffeoyltyramine and N-caffeoylnorephedrine (0.05 µM) significantly inhibited P-selectin exposure by 31%, 30% and 39%, respectively, in mouse whole blood upon stimulation with 2.5 µg/mL collagen [57].
Moreover, the extracts of A. sativum were evaluated after 20 months following their storage (aged extracts) for their antiplatelet effects. The aged extract was prepared by placing the chopped A. sativum bulb in ethanol or water/ethanol mixture at room temperature for 20 months. During this process, allicin, which is an unstable compound, is converted to a stable compound, namely S-allylcysteine. In addition, this extract has a higher content of total phenolic compounds compared to fresh A. sativum extracts (around 129 ± 1.8 mg/g compared to 56 ± 1.2 mg/g). Aged extracts showed better antioxidant, hypotensive, hypoglycaemic and hypolipidemic effects than fresh A. sativum extracts [58]. The aged extract prepared in 15–20% water: ethanol was tested in 8 µM ADP-induced human PRP aggregation at different concentrations; 0.29%, 0.58%, 1.56%, 3.12% and 6.25% (v/v). All these concentrations significantly inhibited platelet aggregation and fibrinogen binding (Figure 2) in a concentration dependant manner. Additionally, the extract markedly affected the change of platelet shape by blocking filopodia formation upon stimulation with ADP [59]. In another study, an aged extract [1.56%, 3.12%, 6.25%,12.5% and 25% (v/v)] significantly reduced human PRP aggregation induced by 8 µM ADP in a concentration dependant manner with almost 75% inhibition achieved at 25% (v/v) of the extract. In addition, 25% (v/v) extract significantly suppressed intracellular Ca2+ levels in 5 µM A23187 (a calcium ionophore) activated human isolated platelets [60]. Moreover, at concentrations of 3.12% and 12.5% (v/v), it markedly inhibited ADP-induced human PRP aggregation by around 40% by reducing the binding of platelets to fibrinogen and increasing the levels of intracellular cAMP (Figure 2) [61].
Furthermore, the antiplatelet effects of the aged extract were tested in rat PRP following oral administration. Three doses (1, 2 or 5 g/kg/day) of the extract were administered for 7 or 14 days in different cohorts of rats and then PRP was tested using 10 µg/mL collagen. All doses that were administered for 14 days significantly inhibited aggregation in a dose dependent manner. In isolated platelets obtained from rats treated with a 5 g/kg/day dose for 14 days, a significant inhibition of the phosphorylation of the mitogen-activated protein (MAP) kinases; p38, c-JUN NH2-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) (following activation with collagen), which are all important for platelets signalling was observed [62].

2.3. Wild Garlic

Allium ursinum has a less pungent taste than A. sativum (common garlic) and all of its parts are edible, although the leaves are typically consumed (raw or cooked) rather than the cloves, due to their high content of biologically active compounds, specifically organosulfur molecules [63]. The leaves were used in Asian, Middle Eastern and European folk medicine for their anti-bacterial, digestion stimulating and hypotensive effects [64]. Recent studies have demonstrated its antibiotic, cytotoxic, hypotensive, hypolipidemic and antiplatelet effects [63].
A study tested the antiplatelet effects of aqueous, chloroform and methanolic extracts of leaves of A. ursinum as well as the ethyl acetate fractions of the ethanolic extract (prepared by liquid-liquid extraction) in human PRP upon activation with different agonists. The ethanolic extract (10 mg/mL) showed a significant reduction in 20 µM ADP- induced aggregation by around 66%. However, the inhibitory effects against A23178 (4 µg/mL) and epinephrine (20 µM) were not significant. In addition, chloroform and ethyl acetate fractions of the methanolic extract, at a concentration of 5 mg/mL, inhibited ADP-induced platelet aggregation by almost 98% [53]. Similarly, the ethanolic extract of A. ursinum leaves, essential oil, ethyl acetate and chloroform factions showed significant inhibitory effects, specifically against 20 µM ADP-induced human PRP aggregation. In addition, isolated compounds from chloroform fractions such as 1,2-di-O-α-linolenoyl-3-O-β-d-galactopyranosyl-sn-glycerol (Figure 5a) and β-sitosterol-3-O-β-d-glucoside (Figure 5b) showed inhibitory effects on ADP-induced human PRP aggregation [64].

2.4. Cruciferous Vegetables

Cruciferous vegetables (family Brassicaceae) such as cabbage, Chinese cabbage, cauliflower, broccoli and kale are cultivated and consumed globally [65]. Various studies have reported on the protective effects of cruciferous vegetables against different types of cancers including lung, breast, gastric, bladder and prostate cancers [65,66,67,68]. Cruciferous vegetables are a good source of vitamins (including vitamin C, E and folic acid), minerals (including calcium, iron and zinc), flavonoids (mainly anthocyanins), phenolic acids (including hydroxycinnamic acid) and tannins [65]. However, most of the biological effects of cruciferous vegetables result from their organosulfur compounds (glucosinolates) that are hydrolysed into isothiocyanates and indole-3-carbinol by myrosinase present in plant cells during the process of chopping, cooking or freezing, or in human gut [65,69,70]. In addition, cruciferous vegetables significantly reduce CVDs-associated mortalities and exhibit antioxidant, hypotensive, hypolipidemic, hypoglycaemic and antiplatelet effects [70,71].
The ethyl acetate and n-butanol extracts of Brassica oleracea L. var. capitata (cabbage) leaves, Brassica oleracea var. Italica (broccoli) florets, Brassica oleracea var. botrytis L. (cauliflower) floral head, Brassica rapa subsp. rapa (turnip) root and Wasabia japonica rhizome (wasabi) were evaluated for their antiplatelet effects in human PRP using 10 µM ADP and 0.5 mM arachidonic acid (AA) as agonists (Table 2). In AA-induced PRP aggregation, the ethyl acetate extracts of Wasabia japonica, Brassica oleracea L. var. capitata and Brassica rapa subsp. rapa showed significant inhibitory effects by around 90%, 88% and 80%, respectively. The ethyl acetate extract of Wasabia japonica inhibited platelets by 60%, while the n-butanol extract inhibited platelets by 58% upon stimulation with ADP in platelets [72].
In addition, methanolic extracts of Brassica oleraceae L. var. acephala (kale) leaves exhibited significant reduction in P-selectin exposure in AA (250 µg/mL)-induced whole blood samples that were collected from patients who were diagnosed with pathological conditions such as obesity, hypertension, hyperglycaemia, and hyperlipidaemia. However, its effect on agonist-induced fibrinogen binding was insignificant [73].
The antiplatelet effects of the anthocyanin-rich extract were evaluated in human platelets. Anthocyanins are a group of plant pigments that belong to the flavonoid class of phytochemicals. Their basic structures consist of a flavylium cation (Figure 6) and based on the substitutions at C3, C5–C7and C3′–C5′ positions, their structures are different in diverse anthocyanins [74]. Various studies reported the health benefits of anthocyanins including antioxidant, anti-inflammatory, anti-cancer, anti-bacterial, anti-thrombotic and neuroprotective effects [74,75]. Brassica oleracea var. capitata F. rubra (red cabbage) leaves, which are abundant in anthocyanins (around 322 mg of anthocyanins/100 g of fresh weight), were prepared as a methanolic extract. The anthocyanin-rich extract at 5, 10 and 15 µM (calculated based on absorption coefficient) showed a significant inhibition on human isolated platelet aggregation induced by 0.5 U/mL thrombin to around 66%, 53% and 38%, respectively [76]. This extract also exhibited an inhibitory effect on lipid peroxidation in platelets which leads to inappropriate platelet activation [76,77]. At a concentration of 15 µM, it significantly reduced the production of superoxide anion in human isolated platelets activated by thrombin (6 U/mL) by around 80%, while at the concentration of 10 µM, it significantly inhibited the metabolism of AA and subsequently the formation of TXA2 [75,76]. At 10 µM, it significantly suppressed lipid peroxidation in platelets activated by lipopolysaccharides (LPS) from Escherichia coli and Pseudomonas aeruginosa (0.15 and 1.5 µg/mL) by around 50% (with E. coli LPS) and 60% (P. aeruginosa LPS) [77].
Sulforaphane (Figure 7) is a sulphur compound that is abundant in Brassica oleracea var. Italica (broccoli) florets (62.64–982.36 µg/g of dry weight) and stem (18.11 to 274.00 µg/g of dry weight) [78] and in Brassica oleracea var. capitata F. rubra (red cabbage) leaves (48–101.99 µg/g of dry weight) and Brassica oleracea L. var. capitata (green cabbage) leaves (7.58–540 µg/g of dry weight) [79,80]. It has been reported that sulforaphane exhibits anti-cancer, antibiotic, antioxidant, anti-inflammatory, hypotensive and hypoglycaemic effects [69].
The effect of sulforaphane on platelet aggregation has been evaluated in previous studies [81,82]. In human isolated platelets, 25 µM and 50 µM sulforaphane markedly inhibited 1 µg/mL collagen-induced platelet aggregation by around 60% and 90%, respectively. However, the inhibitory effects of sulforaphane (25–200 µM) in platelets induced by 60 µM AA, 1 µM U46619 or 0.05 U/mL thrombin were not significantly different from controls. Moreover, 25 µM and 50 µM sulforaphane markedly inhibited the phosphorylation of PLCγ2 in platelets (Figure 2) [81]. Upon platelet activation, PLCγ2 hydrolyses membrane phospholipid, phosphatidylinositol 4,5 bisphosphate (PIP2) to produce the second messengers 1,4,5-trisphosphate (IP3), which increases the cytosolic calcium levels and diacylglycerol (DAG) to stimulate protein kinase C (PKC) which in turn activates platelet degranulation, TXA2 synthesis and release, and activation of integrin αIIbβ3 leading to platelet aggregation [83]. Furthermore, sulforaphane at 0.125 and 0.25 mg/kg showed a significant reduction in mortality rate due to ADP (700 mg/kg) induced acute pulmonary thrombosis in mice by 58.3% and 41.7%, respectively [81]. Similarly, the effect of 60 µM and 100 µM of sulforaphane on platelet aggregation was tested in human isolated platelets after 30 min of incubation. Upon activation with thrombin (0.1 U/mL), 100 µM sulforaphane showed 50% inhibition. However, in collagen (2.5 µg/mL)-activated platelets, both 60 µM and 100 µM of sulforaphane inhibited aggregation by nearly 100%. However, neither concentrations affected ADP (1.56 µM)-activated platelets [84]. In addition, aggregation using mouse platelets activated by 3 U/mL thrombin was significantly suppressed by sulforaphane at 10, 20 and 50 µM. In vivo tail bleeding times in mice were also prolonged to 58.2 ± 1.4 and 76.4 ± 1.2 s when 7.1 µg/mouse and 17.7 µg/mouse concentrations were used, respectively [82].
Another anti-cancer compound isolated from cruciferous vegetables is indole-3-carbinol (Figure 8). This compound is produced by the hydrolysis of glucobrassicin (which is a glucosinolate) via myrosinase, during plant tissue processing, i.e., chopping, crushing, or chewing or by the human gut microflora. Glucobrassicin is a major compound in Brassica oleracea var. Italica (broccoli), Brassica oleraceae var. capitata F. alba (white cabbage) and Brassica oleracea var. botrytis L. (cauliflower) as it accounts for almost 50% of the total glucosinolate in those vegetables [85]. Indole-3-carbinol has been reported to possess anticancer effects, specifically against hormone-dependent cancers such as breast, uterine, ovarian and prostate [86,87].
Additionally, the antiplatelet effects of indole-3-carbinol were evaluated using various platelet functional assays. Different concentrations of indole-3-carbinol (3, 6, 12 and 25 µM) showed a significant reduction in human isolated platelet aggregation activated by 5 µg/mL collagen by around 70%. Thromboxane B2 (TXB2, a stable metabolite of TXA2) production was also significantly inhibited by 25 µM indole-3-carbinol by around 70%. Moreover, in a thromboembolism model in mice, the oral administration of 4.4, 8.8, 17.7 or 36.8 mg/kg of indole-3-carbinol protected mice against mortality by 67%, 70%, 70% and 89%, respectively upon stimulation with a mixture of 114 µg collagen and 13.20 µg epinephrine [88].
In another study, indole-3-carbinol (3, 6, 12 and 25 µM) displayed significant inhibitory effects on 10 µM ADP-induced human PRP aggregation by 64%, 76%, 84% and 90%, respectively. Additionally, in ADP (10 µM) activated rat PRP, oral doses of 12.5, 25, and 50 mg/kg/day of indole-3-carbinol for 14 days significantly suppressed PRP activation by 33%, 55%, 66% and 77%, respectively. The same doses significantly reduced rat brain infarction volume in a middle-cerebral artery occlusion rat model [89].

2.5. Green Leafy Vegetables

Green leafy vegetables are rich in flavonoids, vitamins such as vitamin A and C, glucosinolates, carotenoids, essential polyunsaturated fatty acids and nitrate [90,91]. Nitrates (NO3) that consist of nitrogen and oxygen atoms are crucial for plant growth as they are used to synthesise amino acids and subsequently proteins. They are also important for chlorophyll formation. Plants absorb nitrates from soil through their roots and then store them in leaves [92]. Therefore, green leafy vegetables have a high content of nitrates. Nitrates display different pharmacological effects, including antioxidant, gastroprotective, antibacterial, hypotensive, and antiplatelet effects and have been found to improve endothelial function and blood flow to ischaemic tissues [92,93]. Spinacia oleracea (spinach) leaves have many beneficial effects on human health including antioxidant, anti-cancer, anti-inflammatory, hypolipidemic and hypoglycaemic effects [94]. Cho et al. [95,96], studied the effect of Spinacia oleracea (spinach) leaf extract which is a saponin-rich extract on platelet activity. Saponins are natural compounds that have shown positive impacts on cardiovascular health by acting as antioxidant, hypotensive, hypoglycaemic and hypocholesterolaemic agents [97]. Different concentrations (100, 300 and 500 µg/mL) of the S. oleracea saponin-rich extract showed significant inhibitory effects on aggregation in rat isolated platelets upon activation by 10 µg/mL collagen in a concentration dependent manner (53%, 50% and 40%, respectively). In addition, 500 µg/mL saponins-rich extract significantly increased cAMP and cGMP levels in platelets by around 60% compared to the controls [96].
Another leafy vegetable that demonstrated antiplatelet effects is Eruca sativa (Rocket) leaves [98]. 1 mg/mL concentration of 30% methanolic extract of E. sativa leaves was tested in human PRP, activated by 8 µM ADP, 1 mM AA and 1.5 µg/mL collagen. However, only the ADP-stimulated platelet aggregation was inhibited by around 50% (IC50 of 0.71 mg/mL). In addition, it inhibited P-selectin exposure from around 58% to 42% and the release of TXB2 in platelets. A single dose of 200 mg/kg of the extract was tested in a thrombosis model in mice and it postponed the artery occlusion time to 60 min compared to 30 min in the control group and it significantly reduced the maximum occlusion from 100% to almost 56% [98].

3. Fruits

3.1. Tomatoes

Solanum lycopersicum (tomatoes) is widely consumed all over the world in different forms, i.e., raw, cooked or processed. They are known for their antioxidant, anti-inflammatory, anticarcinogenic, hypoglycaemic and lipid-lowering effects due to their active components including phytosterols, flavonoids, nucleosides and carotenoids [99,100,101]. Specifically, lycopene, which accounts for more than 80% of total carotenoids in tomatoes, and β-carotene, which accounts for around 10%, are two important molecules that display biological effects in tomatoes [101].
Due to the beneficial effects of S. lycopersicum on CVD risk factors, various studies have investigated the effects of S. lycopersicum extracts or their isolated compounds on platelet function. Fuentes et al. [102] demonstrated the effect of methanolic extracts of S. lycopersicum ripe fruit and processed products (paste and pomace) and their liquid-liquid fractions (petroleum ether, ethyl acetate, and aqueous) on platelet activation. In human PRP stimulated by ADP (8 µM), 1 mg/mL of methanolic extract of ripe fruits and aqueous fractions significantly inhibited platelet aggregation by around 50%. Additionally, adenosine (Figure 9a), a purine nucleotide was isolated from the aqueous fraction of tomatoes and it significantly reduced PRP aggregation by around 45% at a concentration of 4.6 µM [103,104]. The aqueous extract (1 mg/mL) of S. lycopersicum pomace (industrial tomato by-product consists mainly of peels and crushed seeds) and paste (cooked and concentrated whole fruits) significantly supressed ADP-induced PRP activation by around 35–40% [102]. These data indicate that processing of S. lycopersicum fruits, which involves heating up to 100 °C, may not affect their antiplatelet effects.
In another similar study, the methanolic extract of S. lycopersicum and its petroleum ether, ethyl acetate and aqueous fractions as well as guanosine (Figure 9b) (another purine nucleotide), which was isolated from the aqueous fraction, were tested on human isolated platelets. The aqueous fraction (1 mg/mL) was found to have the highest (around 54%) inhibition on platelet aggregation, stimulated by 8 µM ADP, followed by the petroleum ether (43 ± 6%) and ethyl acetate (39 ± 8%) fractions. Moreover, 4 mM guanosine suppressed ADP-induced aggregation by 95% and ATP release by 92%. However, in collagen (1.5 µg/mL) activated PRP, aggregation was significantly inhibited by approximately 97%, with ATP release inhibited by around 72%. Guanosine also inhibited platelet spreading and sCD40L release in thrombin-activated platelets (Figure 2). Collagen-induced platelet adhesion under controlled flow was reduced by nearly 70% with 2 mM guanosine [105]. Data from the previous two studies indicate that the aqueous extract of S. lycopersicum is more effective as an antiplatelet agent in ADP- and collagen-activated platelets than the total extract and its fractions. This is mostly due to the purine nucleotides (adenosine and guanosine), with guanosine showing higher antiplatelet activities than adenosine.
Moreover, 40 µL aqueous extract of S. lycopersicum fruit significantly reduced ADP (10 µM) and collagen (2 µg/mL)-induced human PRP aggregation by approximately 70% and 41%, respectively. However, the extract did not affect platelet aggregation stimulated by 0.5 mM AA. In this study, the effects of different incubation times (5, 10, 15 and 30 min) of the extract with PRP in collagen-induced platelet aggregation were probed and the results demonstrated that the inhibitory effect of the extract (40 µL) was positively correlated with the incubation time. Platelet activation was inhibited by around 23%, 25%, 30% and 49%, respectively, for those incubation times. Furthermore, the authors suggested that the extract inhibits platelet aggregation through inhibiting phospholipase C (Figure 2), and not by increasing cAMP levels [106]. Although it was suggested that the inhibitory effects are positively correlated with the concentrations of extract tested, the precise concentrations of the extract were not stated and instead the volumes were used to express the amount of extract utilised in this study.
The effect of oral intake of S. lycopersicum fruit pomace on platelet function was evaluated in a human pilot, randomised, single-blind, and placebo-controlled intervention study. A total of 99 participants were divided equally into three groups. Two doses (1 g and 2.5 g) of the extract were tested against placebo and all were administered in the form of flavoured beverages for five consecutive days. The blood samples were collected on day 1 and 5 and the PRP was tested upon activation with 4 µM ADP. The inhibition (around 35%) of platelet aggregation was found to be significant, after day 5 in the group that received 1 g dose. However, the inhibitory effect of the 2.5 g dose was not found to be significant [107].
A previous study evaluated the differences in antiplatelet effects between 1 mg/mL aqueous extract of fresh S. lycopersicum fruit and the following commercially available processed products; sauce, ketchup, juice, and pomace in human PRP aggregation using four different agonists; 8 µM ADP, 1.5 µg/mL collagen, 1 µM AA and 30 µM thrombin receptor activator peptide 6 (TRAP-6) (Table 3). All extracts showed significant reductions in aggregation upon stimulation with ADP and collagen. In ADP activated PRP, ketchup and sauce showed the highest inhibitory effects of around 50%. In collagen activated PRP, ketchup and pomace exerted the highest inhibitory effect by around 40%. In addition, the abundant phenolic compounds in the extracts, chlorogenic, p-coumaric, ferulic and caffeic acids were identified and their antiplatelet effects were evaluated at a concentration of 500 µM using the same four agonists (Table 3). Generally, these compounds exhibited significant anti-aggregatory effects on ADP and collagen-stimulated platelets, but p-coumaric and chlorogenic acids showed the highest inhibitory effects [108]. Although the phenolic compounds showed antiplatelet effects, the toxicity of the tested (high) concentration, 500 µM was not evaluated in platelets.
In another study, the effect of different concentrations of an active compound from S. lycopersicum, lycopene (Figure 10) on platelet function was examined [109]. Lycopene is a natural, red pigment that belongs to carotenoid phytochemicals (specifically hydrocarbon carotenoids). The content of lycopene in fresh S. lycopersicum fruit is around 0.88–7.74 mg/100 g and it differs according to the stage of fruit ripeness [110,111]. It is reported that lycopene content is higher in processed products because heating facilitates the release of lycopene from plant tissues and increases its bioavailability [111]. For example, ketchup contains 9.9–13.44 mg/100 g of lycopene. The beneficial effects of lycopene were previously documented on cardiovascular health, as it acts as an antioxidant, anti-inflammatory, hypotensive, hypolipidemic and antiplatelet agent [111].
The antiplatelet effects of different concentrations (4, 6, 8, 10 and 12 µM) of lycopene were examined in human PRP stimulated by 2.5 µM ADP and 1 µg/mL collagen (Table 4). All tested concentrations of lycopene displayed significant inhibitory effects in ADP- and collagen-induced platelets. However, when compared to aspirin (140 µM), inhibitory effects of lycopene were insignificant, as aspirin reduced aggregation by 47.79% ± 15.99% and 70.37% ± 7.49% upon stimulation with ADP and collagen, respectively. In addition, the synergistic inhibitory effects of lycopene and aspirin combinations were investigated but they were not significant [109].
Furthermore, Zhang et al. [112] examined the antiplatelet effects of a pharmaceutical preparation of S. lycopersicum, Fruitflow® powder in rat isolated platelets [113]. Fruitflow® is a pharmaceutical preparation of an aqueous extract of S. lycopersicum fruits. It is available in a syrup and powder form and it mainly consists of active ingredients, adenosine, chlorogenic acid, rutin and lycopene all of which are reported to have antiplatelet effects [108]. At concentrations of 4 and 6 g/L Fruitflow® inhibited by around 55% and 74%, respectively in 2.5 µM ADP-induced platelets. However, in collagen stimulated platelets, aggregation was reduced by 40% and 71%, respectively. Additionally, the binding of fibrinogen to integrin αIIbβ3 was significantly inhibited by 32% at 6 g/L of Fruitflow® in 10 µM ADP activated isolated platelets. Moreover, the antiplatelet effects of Fruitflow® were tested in 2.5 µM ADP-induced isolated platelets after 4 weeks of oral administration of 25, 75 and 150 mg/kg doses of Fruitflow®. The 150 mg/kg dose markedly inhibited platelet aggregation to 24% [114].

3.2. Berries

Several studies on many varieties of berries have shown that the regular intake of berries may decrease CVD risk factors due to their antioxidant, anti-inflammatory, hypoglycaemic, and hypotensive effects. These effects have been attributed to their high content of vitamins specifically vitamin C and E, phenolic compounds mainly flavonols, phytoestrogen, minerals and essential fatty acids [115,116]. Thus, several studies have evaluated the effects of different species of berries on platelet function.
Fragaria ananassa (strawberry) intake has a protective effect on cardiovascular health, mainly because it has the highest antioxidant effects compared to other berries as well as other fruits and vegetables due to their high total polyphenolics and vitamin C contents [117,118]. Thus, the antiplatelet effects of F. ananassa were evaluated by testing the aqueous extract of fruits (0.1, 0.5, and 1 mg/mL) in human isolated platelets upon stimulation with ADP (8 µM), collagen (15 µg/mL), AA (1 mM) and TRAP-6 (30 µM). The extract inhibited platelet activation induced by AA and ADP by around 65% and 55%, respectively at a concentration of 1 mg/mL. The same concentration of extract markedly reduced the release of inflammatory markers such as sCD40L, P-selectin, chemokine ligand 5 (CCL5) and interleukin-1β (IL-1β) from platelets (Figure 2) by around 43%, 37%, 41% and 37%, respectively following stimulation with 2 U/mL thrombin. In an in vivo thrombosis model, a dose of 200 mg/kg of the extract significantly delayed thrombus formation (60 min compared to 20 min in the control group) with a maximum occlusion of 35% compared to control which resulted in 100% occlusion [119].
The antiplatelet effects of Aronia melanocarpa (black chokeberry fruit) were examined during hyperhomocysteinemia, which induces platelet activation. Hyperhomocysteinemia is caused by increased plasma levels of homocysteine (Hcy) (or its metabolite, homocysteine thiolactone), which is produced in the body as a result of the metabolism of methionine [120]. A high level of homocysteine is a risk factor for CVDs as it induces oxidative stress, inflammation, endothelial dysfunction and platelet aggregation. The phenolic rich extract (2.5, 5 and 10 µg/mL) of A. melanocarpa was combined with 100 µM homocysteine or 1 µM homocysteine thiolactone (HTL) and tested in thrombin- (0.1 U/mL) stimulated human isolated platelets (Table 5), A. melanocarpa extracts showed anti-aggregatory effects in a concentration dependant manner. However, the inhibitory effects of the extract were found to be insignificant on platelet adhesion to collagen and fibrinogen. In addition, when the extract was combined with Hcy or HTL, it was able to significantly reduce the stimulatory effects of both Hcy and HTL in thrombin-induced platelet aggregation and adhesion in a concentration dependant manner [121]. These effects are attributable to the antioxidant effects of A. melanocarpa’s phenolic rich extracts (that mainly contain flavonoids) as reported by other studies [121,122].
In a recent human study, the effects of 5, 10 and 50 µg/mL of the phenolic fraction of 80% methanolic extract of Hippophae rhamnoides (sea buckthorn berry fruit) were tested on platelet adhesion to collagen and fibrinogen upon stimulation with 0.2 U/mL thrombin or 10 µM ADP activated human isolated platelets. In resting platelets, platelet adhesion was significantly reduced in a concentration-dependent manner. However, in thrombin and ADP stimulated platelets, adhesion to fibrinogen was significantly inhibited by around 65% and 55%, respectively at 50 µg/mL. In ADP activated PRP, the extract showed insignificant inhibition on aggregation [123] which indicates that the extract may have no effect on platelet ADP receptors. In a similar study, 10 µg/mL of isorhamnetin (Figure 11), an abundant flavonol in H. rhamnoides, and its glycoside derivative [isorhamnetin 3-O-beta-glucoside-7-O-alfa-(3′′′-isovaleryl)-rhamnoside] were tested in human PRP using 10 µM ADP, collagen 2 µg/mL and 0.1 U/mL thrombin as agonists. Isorhamnetin accounts for around 44.5–78.3% of total flavonol content in H. rhamnoides [124] and is reported to exert antioxidant, anti-inflammatory, anti-microbial, anti-cancer and hepatoprotective effects [125]. The antiaggregatory effects of both compounds were significant in thrombin activated platelets with around 25% effect while it was only minor in ADP and collagen activated platelets [126].

4. Spices

4.1. Turmeric

Curcuma longa rhizome, which is known as turmeric, is widely used as a spice and food colouring agent, and it was routinely used in Asian traditional medicine. It was used to treat malaria, rheumatoid arthritis, hyperglycaemia and wound healing [127]. The curcuminoids such as curcumin (Figure 12a), demethoxycurcumin (Figure 12b) and bisdemethoxycurcumin (Figure 12c) are its major active constituents [128]. However, curcumin is more abundant, comprising, on average, 70–80% (w/w) of the total extract, whereas demethoxycurcumin accounts for 11.47–23.81% (w/w) and bisdemethoxycurcumin accounts for 5.97–13.88% w/w [129].
A recent study showed that 250 μg/mL of ethanolic extract of C. longa and 25 µM of its another active compound, cyclocurcumin, (Figure 13) significantly reduced shear-stress induced platelet aggregation in a dose-dependent manner in human PRP by around 75% and 70%, respectively, with an IC50 value of 6.33 ± 3.29 µM for cyclocurcumin [130].
Shear-stress describes the force applied by blood flow within blood vessels which increases in blocked vessels due to thrombosis [131]. In addition, the effects of 25 µM of nine active constituents isolated from C. longa ethanolic extract: artumerone, bisabolatraen, bisacurone, bisdemethoxycurcumin, curcumin, 4-dehydroxybisacurone, demethoxycurcumin, β-hydroxycinnamic acid and β-sitosterol were examined (Table 6). Although these compounds reduced aggregation to different extents, they were insignificant compared to cyclocurcumin. Moreover, cyclocurcumin showed similar inhibitory effects on aggregation in human isolated platelets. Cyclocurcumin (1,5 and 10 µM) also markedly reduced intracellular Ca+2 levels, serotonin release, dense and α- granules secretion, fibrinogen and von Willebrand factor (vWF) binding to platelets in shear-stress-induced human isolated platelets (Figure 2) [130].
To examine the antiplatelet effects of curcuminoids, Maheswaraiah et al. [132] tested different concentrations of curcumin (10, 30 and 60 µg/mL) and the curcuminoids rich-ethanolic fraction (10, 20 and 30 µg/mL) on platelets. The curcuminoids rich fraction consists of 33% curcumin, 18% demethoxycurcumin and 48% bisdemethoxycurcumin and was tested in rat PRP aggregation stimulated by ADP (40 µM), collagen (15 µg/mL) and AA (0.75 mM) (Table 7). Generally, all tested concentrations of curcuminoids and curcumin showed reductions in aggregation to different levels when three agonists were used, but curcuminoids showed higher inhibitory effects. In addition, 20 µg/mL of curcuminoids enhanced the release of nitric oxide in rat platelets upon activation with ADP, AA and collagen.
Another compound isolated from C. Longa that possesses antiplatelet effects is the sesquiterpene ketone, aromatic-turmerone or ar-turmerone (Figure 14), which is the major component of the essential oil (61.79%) and has antioxidant, anti-inflammatory, and anticancer effects [133]. In animal models, it demonstrated positive impacts on neurogenerative diseases, including Parkinson’s and epilepsy by exhibiting anticonvulsant and protective effects on neurons [134,135,136]. Antiplatelet effects of ar-turmerone were tested in rabbit isolated platelets induced by 2 µg/mL collagen at concentrations of 1, 5 and 10 µg/mL. It inhibited aggregation significantly by 60% and 80%, respectively, with an IC50 value of 1.2 µM [133].
In a myocardial ischemia-reperfusion injury study in rats, the effect of 500 mg/kg oral dose of C. longa oil on platelet function was evaluated. The oil exhibited significant improvement on neurological impairment and reduced the percentage of apoptotic cells as well as reactive oxygen species resulting from ischemic injury in rats [137,138]. The oil exhibited an inhibitory effect in rat PRP aggregation after 1 and 24 h of administration upon stimulation with 10 µg/mL collagen, 10 µM ADP and 0.64 U/mL thrombin at around 28%, 31% and 34%, respectively. However, C. longa oil did not affect A23187, AA and 12-phorbol 13-myristate acetate (PMA) induced platelet aggregation. In addition, oral administration of C. longa oil at 500 mg/kg and 1000 mg/kg markedly reduced tyrosine phosphorylation of signalling proteins with molecular weights between 55–60, 70–75, 80–85 and 90–120 kDa in rat isolated platelets upon activation with collagen, ADP, and thrombin. Furthermore, in a mouse pulmonary thromboembolism model, both doses of oil showed anti-thrombotic effects of 43 ± 7% and 63 ± 5%, respectively, after 1 h of administration compared to the 38 ± 3% obtained by 30 mg/kg aspirin. Moreover, rats treated with C. longa oil displayed a prolonged tail bleeding time by 18% and 25% for the 500 mg/kg and 1000 mg/kg doses, respectively compared to the controls (3–5 min) [139].
Another study reported that curcumin (1, 10, 20, 50 and 100 µM) greatly reduced platelet aggregation in aspirin pre-treated (for synergistic effects) human isolated platelets stimulated by a snake venom toxin, convulxin. Additionally, the inhibitory effects of 50 µM curcumin upon activation with 50, 100, 200, 500 ng/mL convulxin were approximately 98%, 75%, 40% and 30%, respectively. Curcumin (50 µM) also significantly suppressed platelet aggregation induced by 20 µg/mL collagen and 5 µg/mL cross-linked collagen related peptide (CRP-XL). However, it did not affect aggregation induced by a protease activated receptor 4 (PAR4) agonist, AYPGKF. Curcumin considerably suppressed the degranulation of dense granules in a concentration-dependent manner and phosphorylation of linker for activated T-cells (LAT) and PLCγ2 upon activation by 100 ng/mL convulxin (Figure 2) [140].

4.2. Ginger

In ancient Chinese, Indian and Roman cultures Zingiber officinale rhizome (ginger) was widely used as a spice and for treating nausea, indigestion, diarrhoea, cough, and blood stasis [141]. Several research studies demonstrated the pharmacological effects of Z. officinale including, antioxidant, anti-inflammatory, anti-cancer and pain-relieving effects [141,142]. The most abundant active constituents of Z. officinale are gingerols, followed by shogaols and paradols. Gingerols are phenolic compounds that differ in the number of their carbon chain (after the carbonyl group) (Figure 15a). They are thermolabile compounds, as they are transformed into shogaols (Figure 15b) at high temperature (40 °C or higher) during drying or prolonged cooking [143]. After ingestion, shogaols are metabolised to paradols inside the body [144]. Thus, the most abundant gingerol in Z. officinale is 6-gingerol (Figure 15c) which is reported to exhibit analgesic, anti-pyretic and cytotoxic effects as well as inhibiting nitric oxide production in macrophages [142,143].
To evaluate the antiplatelet effects of Z. officinale powder, a placebo-controlled study examined the effect of a daily dose of 4 g of Z. officinale extract capsules on patients (n = 30) with acute myocardial infarction (more than 6 months). Patients were asked to stop their daily dose of aspirin for 2 weeks before the beginning of the study. Blood samples were collected after 45 and 90 days of administration and PRP aggregation was performed using ADP and epinephrin as agonists. Although the effect of Z. officinale on platelet aggregation was insignificant, a single 10 g dose led to a reduction in PRP aggregation of nearly 25% against ADP and epinephrin-induced activation after 4 h from administration. In addition, the n-hexane extract (5, 10, 25 and 125 µg/mL) decreased PRP aggregation induced by 0.5 µM AA in a concentration dependent manner [145].
Another study compared the antiplatelet effects of 10 µM of isolated compounds of Z. officinale and their synthetic analogues in 0.5 mM AA activated human whole blood aggregation (Table 8). All tested compounds showed significant antiplatelet effects. The aggregation was totally inhibited by 12-gingerol, 8-paradol, 8-shogaol, 5-hydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl)-hept-6-ene-3-one and 3-hydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl) heptane [146].

5. Edible Fungi

In the past, mushrooms were categorised within the plant kingdom and considered as vegetables. However, mushrooms are now categorised as fungi [147]. Mushrooms (also known as macrofungi) are different from microfungi (such as moulds, smuts and plant rusts) due to their visible fruiting bodies. Mushrooms lack chlorophyll, as they do not undergo photosynthesis and they do not consist of many organs such as fully developed roots, stems, leaves or flowers [148]. In recent years, there has been an increasing interest in exploring the pharmacological effects of different species of edible mushrooms due to their high mineral, vitamins, essential amino acids and fibre content along with low fat content [149,150]. Some commonly consumed mushrooms such as Ramaria flava (changle), Pleurotaceae ostreatus (oyster mushroom), Agaricus bisporus (button mushroom), Lentinus edodes (oak mushroom) and Flammulina velutipes (winter fungus) have demonstrated many biological effects including anti-cancer, antioxidant, antibiotic, immune enhancing, hypoglycaemic, hypocholesterolaemic, hepatoprotective and cardioprotective effects [151,152,153].
Some species of edible mushrooms were demonstrated to affect platelet activation. The methanolic extract of fruit bodies of Pleurotus florida (500 µg/mL) was reported to significantly reduce human isolated platelets aggregation upon stimulation by 1 mM ADP by around 88% and 95% after 5- and 15 min incubation, respectively. It was suggested that this effect was due to flavonoids and polysaccharide in the extract [154]. In addition, methanolic, ethyl acetate and aqueous (5 mg/mL) extracts of Pleurotus eous (pink oyster mushroom) showed significant anti-aggregatory effects upon stimulation with ADP (1 mM) in human isolated platelets by 45%, 35% and 36%, respectively [155]. Poniedziałek et al. [156] studied the effect of the hot aqueous extract of eight edible mushrooms: Agaricus bisporus, Auricularia auricularia-judae, Coprinus comatus, Ganoderma lucidum, Hericium erinaceus, Lentinula edodes, Pleurotus eryngii and Pleurotus ostreatus in human whole blood platelet aggregation induced by 6.5 µM ADP. However, only P. eryngii, A. bisporus, A. auricularia-judae and C. comatus, showed significant inhibition effects, of 65.1%, 58.0%, 54.3%, and 51.6%, respectively. These effects were more significant than aspirin’s (140 µM) inhibitory effects in this study. Additionally, these four extracts, as well as the extract of G. lucidum, inhibited aggregation induced by 0.5 mM AA (30–34%). The inhibitory effects were linked to total polysaccharides and ergosterol (Figure 16) content and the antioxidant effects of the extracts [156].
Polysaccharides of mushrooms such as galactans, chitin and mannans are consumed as prebiotics to stimulate the growth of human gut bacteria and improve the gut health. They were also reported to have anti-tumor and immune enhancing effects [157]. Ergosterol is a sterol that found in mushroom cell membrane, and it is used as a vitamin D precursor in vitamin D supplement preparations and known to have anti-inflammatory and anti-tumor activities [158].
Moreover, isolated compounds from the ethanolic extract of Hericium erinaceus (lion’s mane) mushroom; hericenone B (Figure 17) significantly inhibited 3 µg/mL collagen activated human isolated platelet aggregation with an IC50 value of 3 µM [159].
In an animal study, davallialactone (Figure 18), isolated from Inonotus xeranticus mushroom, significantly reduced rat isolated platelet aggregation stimulated by thrombin (0.1 U/mL), collagen (2.5 μg/mL) and ADP (10 µM) in a dose-dependent manner. In addition, it suppressed the concentration of intracellular Ca2+ following activation with collagen. Moreover, it reduced the phosphorylation of p38 mitogen-activated kinase (MAPK) and extracellular signal-regulated kinase (ERK2) in collagen (2.5 μg/mL) and thrombin (0.1 U/mL) stimulated platelets [160].
Additionally, ethanolic extract of Hypsizygus marmoreus (white beech mushroom) markedly inhibited rat isolated platelet aggregation, ATP release and intracellular Ca2+ levels after activation with 1 μg/mL collagen, although it did not affect platelet activation by ADP (5 µM) or thrombin (0.05 U/mL) [161]. The ethanolic extract of Cordyceps militaris mushroom reduced rat PRP aggregation induced by 10 µM ADP and collagen 5 μg/mL in a dose-dependent manner after oral administration of 30 mg/kg, 100 mg/kg or 300 mg/kg of the extract for 3 weeks [162].

6. Clinical Trials for Plant Extracts and Their Isolated Compounds

Although natural products do not require Food and Drug Administration (FDA) approval to be released in the market, some pharmaceutical companies apply for FDA approval as a proof of drug’s efficacy and safety [163,164,165]. The FDA drug-review process consists of preclinical studies (on animals) followed by three phases of human studies. These phases determine the efficacy and safety of potential drug molecules using different cohorts of human volunteers. Some examples of natural plant extracts/compounds (that are already sold in the market as supplements) with antiplatelet effects that are currently in the process of FDA approval (no data released) are shown in Table 9 [166,167,168].

7. Conclusions

CVDs are a primary cause of deaths worldwide and they are mainly caused by impaired platelet function as well as other risk factors such as unhealthy diet, hypercholesterolemia, hyperglycaemia, hypertension, and smoking. Therefore, antiplatelet agents are predominantly used in the treatment regimen for CVD patients. Although there are various classes of antiplatelet agents that act through different mechanisms, they are associated with serious side effects and the development of resistance. Some patients with aspirin and/or clopidogrel resistance are at higher risk of recurrent strokes, large infarct size and early neurological deterioration. Some high-risk patients with recurrent ischemic stroke or transient ischemic attacks and taking three antiplatelet agents (aspirin, clopidogrel and dipyridamole) for three months experienced bleeding incidents ranging from mild to fatal although the rate of stroke recurrence was not significantly reduced. Therefore, many studies were conducted to evaluate the effects of numerous edible plants on platelet activation, with the aim of discovering novel antiplatelets agents with better bioavailability, activity and safety profiles whilst promoting the regular intake of healthy diet.
Indeed, plants are important sources for drug discovery to treat different diseases including CVDs. Several prescribed CVD medications are derived from plants, and they mainly comprise alkaloid, cardiac glycosides and polyphenolic phytochemicals. Most of the alkaloid-derived CVD drugs (such are deserpidine and reserpine) are used to treat hypertension. In addition, digoxin and digitoxin (common heart failure and antiarrhythmic drugs) are cardiac glycosides, whereas the phenolic compounds aspirin and hesperidin are used as antiplatelet drugs. However, developing new medications for CVDs is challenging because of the complicated treatment plans due to coexistence of comorbidities and other chronic conditions such as hyperlipidaemia and hypertension. These conditions increase the possibility of developing serious side effects and drug–drug interactions that might affect the bioavailability. Moreover, the complex plant extracts that contain numerous active compounds are often difficult to separate in reasonable quantities (large scale extraction and isolation are needed) and semisynthetic or synthetic strategies may be required.
Plant extracts or their active constituents can be consumed as supplements to act synergistically with prescribed medications to improve outcomes. In Chinese clinical practice, certain plant-based supplement formulations are used as adjunct treatments with CVD medications for better outcomes. For example, Rhodiola sacra and Rhodiola kirilowii capsules and injection formulations are prescribed regularly for ischemic heart disease patients (doses are adjusted according to patient’s condition) [169]. The roots and rhizomes of both species are known to act as antioxidant, cytotoxic, antidepressant and cardioprotective agents. Data from a meta-analysis study (n = 1672) demonstrate that using one of the Rhodiola formulations significantly improved ischemic heart disease symptoms (chest pressure, chest pain and shortness of breath) and electrocardiography compared to registered medications alone [169,170,171]. However, the use of plants supplements in CVDs is not always supported by clinical practice. The main concerns are that plants supplements do not need approval by the FDA or European Medicines Agency (EMA) to be released in the market. There is a lack of controlled clinical studies regarding the efficacy and safety, and insufficient knowledge of possible interactions between supplements active constituent (s) and drugs. The underreporting of adverse effects or poisoning due to consumption of plant products to the local and international regulatory organisations is another major concern. Indeed, plant supplements may be used to improve patients’ outcomes, but must be used under the guidance/supervision of health care providers. Finally, in studies that investigate the effect of plant extracts and/or phytochemicals on CVDs, human clinical trials should be considered as part of the FDA drug-approval process to determine the efficacy, safety and their interactions with other molecules.
Overall, plants are known to be a rich source for bioactive compounds that have numerous beneficial effects on cardiovascular health. They can be consumed safely as part of our diet in the form of fresh raw fruit and vegetables as recommended by health organisations. The WHO recommends the daily consumption of at least 400 g of fruit and vegetables (excluding starchy vegetables), which is equivalent to five portions daily. Based on this recommendation, many countries began a 5-a-day campaign in 1990. However, its importance has been better acknowledged since 2000, as governments have started to promote its importance in schools and big supermarkets, which are used to advertise this via their ready to eat fruit and vegetables [172].
The effects of consumption of five servings of fruit and vegetables on CVDs have been evaluated by many longitudinal studies as reviewed in this article. They reported that consuming five servings showed a significant reduction in the mortality rate of CVDs by almost 10% [173,174]. In addition, incidence of stroke was decreased regardless of sex and consumption duration compared to the group that consumed three or less servings [175,176,177]. Moreover, in hypotensive patients, the consumption of five servings for 12 weeks showed a significant improvement in blood flow through vasodilatation [178]. Furthermore, patients who were followed up for venous thromboembolism, who consumed 3–5 portions of fruit and vegetables, were at significantly lower risk of developing venous thromboembolism (around 27–53%) [179]. In addition, patients who survived stroke were at a lower risk (34%) of recurrence when this dietary plan was followed. Hence, consuming a minimum of 400g/day of fresh fruit and vegetables daily must be a part of the daily diet for children and adults, and especially for CVD patients. We believe that this article provides a plethora of information on the antiplatelet effects of selected and commonly used edible plants consumed regularly. This article aims to create awareness among members of the public and CVD patients on the necessity to adhere to a healthy diet. Moreover, it will encourage scientific communities to further investigate the impacts of these plants for the prevention and treatment of CVDs. Although we covered a wide range of commonly available edible vegetables, fruits, spices and fungi in this article, we were unable to discuss several other edible plants (e.g., green tea) that also exhibit antiplatelet effects due to their occasional and/or limited medicinal uses.

Author Contributions

D.A.I.A., H.M.I.O. and S.V., writing original draft preparation; D.A.I.A., D.R., T.M.V., K.P., S.V. and H.M.I.O., review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to thank the Ministry of Education, Saudi Arabia for their funding support.

Institutional Review Board Statement

Not applicable as this is a review article.

Informed Consent Statement

Not applicable as this is a review article.

Data Availability Statement

All data and appropriate references are included within this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. The World Health Organization. Cardiovascular Diseases (CVDs). 2017. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 28 October 2020).
  2. Benjamin, E.J.; Virani, S.S.; Callaway, C.W.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Chiuve, S.E.; Cushman, M.; Delling, F.N.; Deo, R.; et al. Heart Disease and Stroke Statistics-2018 Update: A Report From the American Heart Association. Circulation 2018, 137, e67–e492. [Google Scholar] [CrossRef]
  3. Karunathilake, S.P.; Ganegoda, G.U. Secondary Prevention of Cardiovascular Diseases and Application of Technology for Early Diagnosis. BioMed Res. Int. 2018, 2018, 5767864. [Google Scholar] [CrossRef]
  4. Zhao, C.-N.; Meng, X.; Li, Y.; Li, S.; Liu, Q.; Tang, G.-Y.; Li, H.-B. Fruits for Prevention and Treatment of Cardiovascular Diseases. Nutrients 2017, 9, 598. [Google Scholar] [CrossRef] [Green Version]
  5. Salas-Salvadó, J.; Becerra-Tomás, N.; García-Gavilán, J.F.; Bulló, M.; Barrubés, L. Mediterranean Diet and Cardiovascular Disease Prevention: What Do We Know? Prog. Cardiovasc. Dis. 2018, 61, 62–67. [Google Scholar] [CrossRef]
  6. Zheng, J.; Zhou, Y.; Li, S.; Zhang, P.; Zhou, T.; Xu, D.-P.; Li, H.-B. Effects and Mechanisms of Fruit and Vegetable Juices on Cardiovascular Diseases. Int. J. Mol. Sci. 2017, 18, 555. [Google Scholar] [CrossRef] [Green Version]
  7. Malakar, A.K.; Choudhury, D.; Halder, B.; Paul, P.; Uddin, A.; Chakraborty, S. A review on coronary artery disease, its risk factors, and therapeutics. J. Cell. Physiol. 2019, 234, 16812–16823. [Google Scholar] [CrossRef]
  8. Moore, K.J.; Sheedy, F.J.; Fisher, E.A. Macrophages in atherosclerosis: A dynamic balance. Nat. Rev. Immunol. 2013, 13, 709–721. [Google Scholar] [CrossRef]
  9. Yu, X.-H.; Fu, Y.-C.; Zhang, D.-W.; Yin, K.; Tang, C.-K. Foam cells in atherosclerosis. Clin. Chim. Acta 2013, 424, 245–252. [Google Scholar] [CrossRef] [Green Version]
  10. Previtali, E.; Bucciarelli, P.; Passamonti, S.M.; Martinelli, I. Risk factors for venous and arterial thrombosis. Blood Transfus. 2011, 9, 120–138. [Google Scholar] [CrossRef]
  11. Gibbins, J.M. Platelet adhesion signalling and the regulation of thrombus formation. J. Cell Sci. 2004, 117, 3415–3425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sokol, J.; Skerenova, M.; Biringer, K.; Simurda, T.; Kubisz, P.; Stasko, J. Glycoprotein VI Gene Variants Affect Pregnancy Loss in Patients With Platelet Hyperaggregability. Clin. Appl. Thromb./Hemost. 2018, 24, 202S–208S. [Google Scholar] [CrossRef]
  13. Simurda, T.; Zolkova, J.; Snahnicanova, Z.; Loderer, D.; Skornova, I.; Sokol, J.; Hudecek, J.; Stasko, J.; Lasabova, Z.; Kubisz, P. Identification of Two Novel Fibrinogen Bβ Chain Mutations in Two Slovak Families with Quantitative Fibrinogen Disorders. Int. J. Mol. Sci. 2018, 19, 100. [Google Scholar] [CrossRef] [Green Version]
  14. Xu, X.R.; Carrim, N.; Neves, M.A.D.; McKeown, T.; Stratton, T.W.; Coelho, R.M.P.; Lei, X.; Chen, P.; Xu, J.; Dai, X.; et al. Platelets and platelet adhesion molecules: Novel mechanisms of thrombosis and anti-thrombotic therapies. Thromb. J. 2016, 14, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. De Candia, E. Mechanisms of platelet activation by thrombin: A short history. Thromb Res. 2012, 129, 250–256. [Google Scholar] [CrossRef]
  16. Nording, H.M.; Seizer, P.; Langer, H.F. Platelets in Inflammation and Atherogenesis. Front. Immunol. 2015, 6. [Google Scholar] [CrossRef] [Green Version]
  17. Nording, H.; Baron, L.; Langer, H.F. Platelets as therapeutic targets to prevent atherosclerosis. Atherosclerosis 2020, 307, 97–108. [Google Scholar] [CrossRef]
  18. Koupenova, M.; Kehrel, B.E.; Corkrey, H.A.; Freedman, J.E. Thrombosis and platelets: An update. Eur. Heart J. 2016, 38, 785–791. [Google Scholar] [CrossRef]
  19. Bhatt, D.L.; Fox, K.A.A.; Hacke, W.; Berger, P.B.; Black, H.R.; Boden, W.E.; Cacoub, P.; Cohen, E.A.; Creager, M.A.; Easton, J.D.; et al. Clopidogrel and Aspirin versus Aspirin Alone for the Prevention of Atherothrombotic Events. N. Engl. J. Med. 2006, 354, 1706–1717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. De Luca, L.; Danchin, N.; Valgimigli, M.; Goldstein, P. Effectiveness of Pretreatment With Dual Oral Antiplatelet Therapy. Am. J. Cardiol. 2015, 116, 660–668. [Google Scholar] [CrossRef]
  21. Spanos, K.; Kouvelos, G.; Matsagkas, M.; Giannoukas, A.D. Antiplatelet Resistance in Ischaemic Stroke Patients. Eur. J. Vasc. Endovasc. Surg. 2017, 54, 3–4. [Google Scholar] [CrossRef] [Green Version]
  22. Sweeny, J.M.; Gorog, D.A.; Fuster, V. Antiplatelet drug ‘resistance’. Part 1: Mechanisms and clinical measurements. Nat. Rev. Cardiol. 2009, 6, 273–282. [Google Scholar] [CrossRef]
  23. Rodríguez-Monforte, M.; Flores-Mateo, G.; Sánchez, E. Dietary patterns and CVD: A systematic review and meta-analysis of observational studies. Br. J. Nutr. 2015, 114, 1341–1359. [Google Scholar] [CrossRef] [Green Version]
  24. Shridhar, K.; Dhillon, P.K.; Bowen, L.; Kinra, S.; Bharathi, A.V.; Prabhakaran, D.; Reddy, K.S.; Ebrahim, S.; for the Indian Migration Study, g. The Association between a Vegetarian Diet and Cardiovascular Disease (CVD) Risk Factors in India: The Indian Migration Study. PLoS ONE 2014, 9, e110586. [Google Scholar] [CrossRef]
  25. Aune, D.; Giovannucci, E.; Boffetta, P.; Fadnes, L.T.; Keum, N.; Norat, T.; Greenwood, D.C.; Riboli, E.; Vatten, L.J.; Tonstad, S. Fruit and vegetable intake and the risk of cardiovascular disease, total cancer and all-cause mortality—A systematic review and dose-response meta-analysis of prospective studies. Int. J. Epidemiol. 2017, 46, 1029–1056. [Google Scholar] [CrossRef]
  26. Dinu, M.; Pagliai, G.; Sofi, F. A Heart-Healthy Diet: Recent Insights and Practical Recommendations. Curr. Cardiol. Rep. 2017, 19, 95. [Google Scholar] [CrossRef]
  27. Baden, M.Y.; Liu, G.; Satija, A.; Li, Y.; Sun, Q.; Fung, T.T.; Rimm, E.B.; Willett, W.C.; Hu, F.B.; Bhupathiraju, S.N. Changes in Plant-Based Diet Quality and Total and Cause-Specific Mortality. Circulation 2019, 140, 979–991. [Google Scholar] [CrossRef]
  28. Yu, E.; Malik, V.S.; Hu, F.B. Cardiovascular Disease Prevention by Diet Modification: JACC Health Promotion Series. J. Am. Coll. Cardiol. 2018, 72, 914–926. [Google Scholar] [CrossRef]
  29. Peluso, I.; Raguzzini, A.; Catasta, G.; Cammisotto, V.; Perrone, A.; Tomino, C.; Toti, E.; Serafini, M. Effects of High Consumption of Vegetables on Clinical, Immunological, and Antioxidant Markers in Subjects at Risk of Cardiovascular Diseases. Oxidative Med. Cell. Longev. 2018, 2018, 5417165. [Google Scholar] [CrossRef]
  30. Zhao, C.; Li, S.; Zhang, J.; Huang, Y.; Zhang, L.; Zhao, F.; Du, X.; Hou, J.; Zhang, T.; Shi, C.; et al. Current state and future perspective of cardiovascular medicines derived from natural products. Pharmacol. Ther. 2020, 216, 107698. [Google Scholar] [CrossRef]
  31. Khurana, S.; Venkataraman, K.; Hollingsworth, A.; Piche, M.; Tai, T.C. Polyphenols: Benefits to the Cardiovascular System in Health and in Aging. Nutrients 2013, 5, 3779–3827. [Google Scholar] [CrossRef]
  32. Chen, C.; Yang, F.-Q.; Zhang, Q.; Wang, F.-Q.; Hu, Y.-J.; Xia, Z.-N. Natural Products for Antithrombosis. Evid.-Based Complementary Altern. Med. 2015, 2015, 876426. [Google Scholar] [CrossRef] [Green Version]
  33. Djekic, D.; Shi, L.; Brolin, H.; Carlsson, F.; Särnqvist, C.; Savolainen, O.; Cao, Y.; Bäckhed, F.; Tremaroli, V.; Landberg, R.; et al. Effects of a Vegetarian Diet on Cardiometabolic Risk Factors, Gut Microbiota, and Plasma Metabolome in Subjects With Ischemic Heart Disease: Randomized, Crossover Study. J. Am. Heart Assoc. 2020, 9, e016518. [Google Scholar] [CrossRef]
  34. Crowe, F.L.; Appleby, P.N.; Travis, R.C.; Key, T.J. Risk of hospitalization or death from ischemic heart disease among British vegetarians and nonvegetarians: Results from the EPIC-Oxford cohort study. Am. J. Clin. Nutr. 2013, 97, 597–603. [Google Scholar] [CrossRef] [Green Version]
  35. Wright, N.; Wilson, L.; Smith, M.; Duncan, B.; McHugh, P. The BROAD study: A randomised controlled trial using a whole food plant-based diet in the community for obesity, ischaemic heart disease or diabetes. Nutr. Diabetes 2017, 7, e256. [Google Scholar] [CrossRef]
  36. Chiu, T.H.T.; Chang, H.-R.; Wang, L.-Y.; Chang, C.-C.; Lin, M.-N.; Lin, C.-L. Vegetarian diet and incidence of total, ischemic, and hemorrhagic stroke in 2 cohorts in Taiwan. Neurology 2020, 94, e1112–e1121. [Google Scholar] [CrossRef] [Green Version]
  37. Kahleova, H.; Levin, S.; Barnard, N. Cardio-Metabolic Benefits of Plant-Based Diets. Nutrients 2017, 9, 848. [Google Scholar] [CrossRef]
  38. Wang, D.; Li, Y.; Ho, Y.-L.; Nguyen, X.-M.; Song, R.J.; Hu, F.B.; Willett, W.; Wilson, P.W.F.; Cho, K.; Gaziano, J.M.; et al. Plant-Based Diet and the Risk of Cardiovascular Disease and Mortality: The Million Veteran Program. Curr. Dev. Nutr. 2020, 4, 1502. [Google Scholar] [CrossRef]
  39. Bujtor, M.; Turner, A.I.; Torres, S.J.; Esteban-Gonzalo, L.; Pariante, C.M.; Borsini, A. Associations of Dietary Intake on Biological Markers of Inflammation in Children and Adolescents: A Systematic Review. Nutrients 2021, 13, 356. [Google Scholar] [CrossRef]
  40. Holt, E.M.; Steffen, L.M.; Moran, A.; Basu, S.; Steinberger, J.; Ross, J.A.; Hong, C.-P.; Sinaiko, A.R. Fruit and Vegetable Consumption and Its Relation to Markers of Inflammation and Oxidative Stress in Adolescents. J. Am. Diet. Assoc. 2009, 109, 414–421. [Google Scholar] [CrossRef] [Green Version]
  41. Galavi, A.; Hosseinzadeh, H.; Razavi, B.M. The effects of Allium cepa L. (onion) and its active constituents on metabolic syndrome: A review. Iran. J. Basic Med. Sci. 2021, 24, 3–16. [Google Scholar] [CrossRef]
  42. Teshika, J.D.; Zakariyyah, A.M.; Zaynab, T.; Zengin, G.; Rengasamy, K.R.R.; Pandian, S.K.; Fawzi, M.M. Traditional and modern uses of onion bulb (Allium cepa L.): A systematic review. Crit. Rev. Food Sci. Nutr. 2019, 59, S39–S70. [Google Scholar] [CrossRef]
  43. Kuete, V. Chapter 14—Allium cepa. In Medicinal Spices and Vegetables from Africa; Kuete, V., Ed.; Academic Press: Cambridge, MA, USA, 2017; pp. 353–361. [Google Scholar] [CrossRef]
  44. Lanzotti, V.; Romano, A.; Lanzuise, S.; Bonanomi, G.; Scala, F. Antifungal saponins from bulbs of white onion, Allium cepa L. Phytochemistry 2012, 74, 133–139. [Google Scholar] [CrossRef]
  45. Marrelli, M.; Amodeo, V.; Statti, G.; Conforti, F. Biological Properties and Bioactive Components of Allium cepa L.: Focus on Potential Benefits in the Treatment of Obesity and Related Comorbidities. Molecules 2019, 24, 119. [Google Scholar] [CrossRef] [Green Version]
  46. Moon, C.H.; Jung, Y.S.; Kim, M.H.; Lee, S.H.; Baik, E.J.; Park, S.W. Mechanism for antiplatelet effect of onion: AA release inhibition, thromboxane A2synthase inhibition and TXA2/PGH2receptor blockade. Prostaglandins Leukot. Essent. Fat. Acids (PLEFA) 2000, 62, 277–283. [Google Scholar] [CrossRef]
  47. Ro, J.-Y.; Ryu, J.-H.; Park, H.-J.; Cho, H.-J. Onion (Allium cepa L.) peel extract has anti-platelet effects in rat platelets. Springerplus 2015, 4, 17. [Google Scholar] [CrossRef] [Green Version]
  48. Ko, E.Y.; Nile, S.H.; Jung, Y.-S.; Keum, Y.S. Antioxidant and antiplatelet potential of different methanol fractions and flavonols extracted from onion (Allium cepa L.). 3 Biotech 2018, 8, 155. [Google Scholar] [CrossRef]
  49. Cavagnaro, P.F.; Galmarini, C.R. Effect of Processing and Cooking Conditions on Onion (Allium cepa L.) Induced Antiplatelet Activity and Thiosulfinate Content. J. Agric. Food Chem. 2012, 60, 8731–8737. [Google Scholar] [CrossRef]
  50. Hubbard, G.P.; Wolffram, S.; de Vos, R.; Bovy, A.; Gibbins, J.M.; Lovegrove, J.A. Ingestion of onion soup high in quercetin inhibits platelet aggregation and essential components of the collagen-stimulated platelet activation pathway in man: A pilot study. Br. J. Nutr. 2006, 96, 482–488. [Google Scholar]
  51. Bayan, L.; Koulivand, P.H.; Gorji, A. Garlic: A review of potential therapeutic effects. Avicenna J. Phytomed. 2014, 4, 1–14. [Google Scholar] [PubMed]
  52. El-Saber Batiha, G.; Magdy Beshbishy, A.; Wasef, L.G.; Elewa, Y.H.A.; Al-Sagan, A.A.; Abd El-Hack, M.E.; Taha, A.E.; Abd-Elhakim, Y.M.; Prasad Devkota, H. Chemical Constituents and Pharmacological Activities of Garlic (Allium sativum L.): A Review. Nutrients 2020, 12, 872. [Google Scholar] [CrossRef] [Green Version]
  53. Hiyasat, B.; Sabha, D.; Grötzinger, K.; Kempfert, J.; Rauwald, J.W.; Mohr, F.W.; Dhein, S. Antiplatelet Activity of Allium ursinum and Allium sativum. Pharmacology 2009, 83, 197–204. [Google Scholar] [CrossRef]
  54. Briggs, W.H.; Xiao, H.; Parkin, K.L.; Shen, C.; Goldman, I.L. Differential Inhibition of Human Platelet Aggregation by Selected Allium Thiosulfinates. J. Agric. Food Chem. 2000, 48, 5731–5735. [Google Scholar] [CrossRef]
  55. Manaster, Y.; Shenkman, B.; Rosenberg, N.; Savion, N. Allicin and disulfiram enhance platelet integrin αIIbβ3-fibrinogen binding. Thromb. Res. 2009, 124, 477–482. [Google Scholar] [CrossRef] [PubMed]
  56. Gao, X.; Wang, C.; Chen, Z.; Chen, Y.; Santhanam, R.K.; Xue, Z.; Ma, Q.; Guo, Q.; Liu, W.; Zhang, M.; et al. Effects of N-trans-feruloyltyramine isolated from laba garlic on antioxidant, cytotoxic activities and H2O2-induced oxidative damage in HepG2 and L02 cells. Food Chem. Toxicol. 2019, 130, 130–141. [Google Scholar] [CrossRef]
  57. Park, J.B. Isolation and Characterization of N-Feruloyltyramine as the P-Selectin Expression Suppressor from Garlic (Allium sativum). J. Agric. Food Chem. 2009, 57, 8868–8872. [Google Scholar] [CrossRef]
  58. Elosta, A.; Slevin, M.; Rahman, K.; Ahmed, N. Aged garlic has more potent antiglycation and antioxidant properties compared to fresh garlic extract in vitro. Sci. Rep. 2017, 7, 39613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. 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] [PubMed] [Green Version]
  60. Allison, G.L.; Lowe, G.M.; Rahman, K. Aged garlic extract and its constituents inhibit platelet aggregation through multiple mechanisms. J. Nutr. 2006, 136, 782s–788s. [Google Scholar] [CrossRef]
  61. Allison, G.L.; Lowe, G.M.; Rahman, K. Aged garlic extract inhibits platelet activation by increasing intracellular cAMP and reducing the interaction of GPIIb/IIIa receptor with fibrinogen. Life Sci. 2012, 91, 1275–1280. [Google Scholar] [CrossRef]
  62. Morihara, N.; Hino, A. Aged garlic extract suppresses platelet aggregation by changing the functional property of platelets. J. Nat. Med. 2017, 71, 249–256. [Google Scholar] [CrossRef]
  63. Sobolewska, D.; Podolak, I.; Makowska-Wąs, J. Allium ursinum: Botanical, phytochemical and pharmacological overview. Phytochem. Rev. 2015, 14, 81–97. [Google Scholar] [CrossRef] [Green Version]
  64. Sabha, D.; Hiyasat, B.; Grötzinger, K.; Hennig, L.; Schlegel, F.; Mohr, F.W.; Rauwald, H.W.; Dhein, S. Allium ursinum L.: Bioassay-Guided Isolation and Identification of a Galactolipid and a Phytosterol Exerting Antiaggregatory Effects. Pharmacology 2012, 89, 260–269. [Google Scholar] [CrossRef]
  65. Manchali, S.; Chidambara Murthy, K.N.; Patil, B.S. Crucial facts about health benefits of popular cruciferous vegetables. J. Funct. Foods 2012, 4, 94–106. [Google Scholar] [CrossRef]
  66. Lam, T.K.; Gallicchio, L.; Lindsley, K.; Shiels, M.; Hammond, E.; Tao, X.; Chen, L.; Robinson, K.A.; Caulfield, L.E.; Herman, J.G.; et al. Cruciferous Vegetable Consumption and Lung Cancer Risk: A Systematic Review. Cancer Epidemiol. Biomark. Prev. 2009, 18, 184–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Tang, L.; Zirpoli, G.R.; Guru, K.; Moysich, K.B.; Zhang, Y.; Ambrosone, C.B.; McCann, S.E. Consumption of Raw Cruciferous Vegetables is Inversely Associated with Bladder Cancer Risk. Cancer Epidemiol. Biomark. Prev. 2008, 17, 938–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Wu, Q.J.; Yang, Y.; Wang, J.; Han, L.H.; Xiang, Y.B. Cruciferous vegetable consumption and gastric cancer risk: A meta-analysis of epidemiological studies. Cancer Sci. 2013, 104, 1067–1073. [Google Scholar] [CrossRef]
  69. Vazquez-Prieto, M.A.; Miatello, R.M. Organosulfur compounds and cardiovascular disease. Mol. Asp. Med. 2010, 31, 540–545. [Google Scholar] [CrossRef] [PubMed]
  70. Esteve, M. Mechanisms Underlying Biological Effects of Cruciferous Glucosinolate-Derived Isothiocyanates/Indoles: A Focus on Metabolic Syndrome. Front. Nutr. 2020, 7, 111. [Google Scholar] [CrossRef]
  71. Blekkenhorst, L.C.; Bondonno, C.P.; Lewis, J.R.; Devine, A.; Zhu, K.; Lim, W.H.; Woodman, R.J.; Beilin, L.J.; Prince, R.L.; Hodgson, J.M. Cruciferous and Allium Vegetable Intakes are Inversely Associated With 15-Year Atherosclerotic Vascular Disease Deaths in Older Adult Women. J. Am. Heart Assoc. 2017, 6, e006558. [Google Scholar] [CrossRef] [Green Version]
  72. Morimitsu, Y.; Hayashi, K.; Nakagawa, Y.; Fujii, H.; Horio, F.; Uchida, K.; Osawa, T. Antiplatelet and anticancer isothiocyanates in Japanese domestic horseradish, Wasabi. Mech. Ageing Dev. 2000, 116, 125–134. [Google Scholar] [CrossRef]
  73. Konić-Ristić, A.; Srdić-Rajić, T.; Kardum, N.; Aleksić-Veličković, V.; Kroon, P.A.; Hollands, W.J.; Needs, P.W.; Boyko, N.; Hayran, O.; Jorjadze, M.; et al. Effects of bioactive-rich extracts of pomegranate, persimmon, nettle, dill, kale and Sideritis and isolated bioactives on arachidonic acid induced markers of platelet activation and aggregation. J. Sci. Food Agric. 2013, 93, 3581–3587. [Google Scholar] [CrossRef]
  74. Khoo, H.E.; Azlan, A.; Tang, S.T.; Lim, S.M. Anthocyanidins and anthocyanins: Colored pigments as food, pharmaceutical ingredients, and the potential health benefits. Food Nutr. Res. 2017, 61, 1361779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Li, H.; Deng, Z.; Zhu, H.; Hu, C.; Liu, R.; Young, J.C.; Tsao, R. Highly pigmented vegetables: Anthocyanin compositions and their role in antioxidant activities. Food Res. Int. 2012, 46, 250–259. [Google Scholar] [CrossRef]
  76. Saluk, J.; Bijak, M.; Kołodziejczyk-Czepas, J.; Posmyk, M.; Janas, K.; Wachowicz, B. Anthocyanins from red cabbage extract—evidence of protective effects on blood platelets. Open Life Sci. 2012, 7, 655–663. [Google Scholar] [CrossRef] [Green Version]
  77. Saluk, J.; Bijak, M.; Posmyk, M.M.; Zbikowska, H.M. Red cabbage anthocyanins as inhibitors of lipopolysaccharide-induced oxidative stress in blood platelets. Int. J. Biol. Macromol. 2015, 80, 702–709. [Google Scholar] [CrossRef]
  78. Li, Z.; Liu, Y.; Fang, Z.; Yang, L.; Zhuang, M.; Zhang, Y.-y.; Sun, P. Development and verification of sulforaphane extraction method in cabbage (Brassica oleracea L. var. capitata) and broccoli (Brassica oleracea L. var. italica Planch.). J. Med. Plants Res. 2012, 6, 4796–4803. [Google Scholar] [CrossRef]
  79. Farag, M.A.; Motaal, A.A.A. Sulforaphane composition, cytotoxic and antioxidant activity of crucifer vegetables. J. Adv. Res. 2010, 1, 65–70. [Google Scholar] [CrossRef] [Green Version]
  80. Glade, M.J.; Meguid, M.M. A Glance at… Broccoli, glucoraphanin, and sulforaphane. Nutrition 2015, 31, 1175–1178. [Google Scholar] [CrossRef] [PubMed]
  81. Jayakumar, T.; Chen, W.-F.; Lu, W.-J.; Chou, D.-S.; Hsiao, G.; Hsu, C.-Y.; Sheu, J.-R.; Hsieh, C.-Y. A novel antithrombotic effect of sulforaphane via activation of platelet adenylate cyclase: Ex vivo and in vivo studies. J. Nutr. Biochem. 2013, 24, 1086–1095. [Google Scholar] [CrossRef]
  82. Ku, S.-K.; Bae, J.-S. Antithrombotic activities of sulforaphane via inhibiting platelet aggregation and FIIa/FXa. Arch. Pharmacal. Res. 2014, 37, 1454–1463. [Google Scholar] [CrossRef] [PubMed]
  83. Harper, M.T.; Poole, A.W. Diverse functions of protein kinase C isoforms in platelet activation and thrombus formation. J. Thromb. Haemost. 2010, 8, 454–462. [Google Scholar] [CrossRef]
  84. Gillespie, S.; Holloway, P.M.; Becker, F.; Rauzi, F.; Vital, S.A.; Taylor, K.A.; Stokes, K.Y.; Emerson, M.; Gavins, F.N.E. The isothiocyanate sulforaphane modulates platelet function and protects against cerebral thrombotic dysfunction. Br. J. Pharm. 2018, 175, 3333–3346. [Google Scholar] [CrossRef] [PubMed]
  85. Possenti, M.; Baima, S.; Raffo, A.; Durazzo, A.; Giusti, A.M.; Natella, F. Glucosinolates in Food. In Glucosinolates; Mérillon, J.-M., Ramawat, K.G., Eds.; Springer International Publishing: Cham, Switzerland, 2016; pp. 1–46. [Google Scholar]
  86. Martín-Ruiz, A.; Peña, L.; González-Gil, A.; Díez-Córdova, L.T.; Cáceres, S.; Illera, J.C. Effects of indole-3-carbinol on steroid hormone profile and tumor progression in a mice model of canine inflammatory mammarycancer. BMC Cancer 2018, 18, 626. [Google Scholar] [CrossRef]
  87. Fujioka, N.; Fritz, V.; Upadhyaya, P.; Kassie, F.; Hecht, S.S. Research on cruciferous vegetables, indole-3-carbinol, and cancer prevention: A tribute to Lee W. Wattenberg. Mol. Nutr. Food Res. 2016, 60, 1228–1238. [Google Scholar] [CrossRef]
  88. Park, M.K.; Rhee, Y.H.; Lee, H.J.; Lee, E.O.; Kim, K.H.; Park, M.J.; Jeon, B.H.; Shim, B.S.; Jung, C.H.; Choi, S.H.; et al. Antiplatelet and antithrombotic activity of indole-3-carbinol in vitro and in vivo. Phytother. Res. 2008, 22, 58–64. [Google Scholar] [CrossRef] [PubMed]
  89. Paliwal, P.; Chauhan, G.; Gautam, D.; Dash, D.; Patne, S.C.U.; Krishnamurthy, S. Indole-3-carbinol improves neurobehavioral symptoms in a cerebral ischemic stroke model. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2018, 391, 613–625. [Google Scholar] [CrossRef]
  90. Bryan, N.S. Functional Nitric Oxide Nutrition to Combat Cardiovascular Disease. Curr. Atheroscler. Rep. 2018, 20, 21. [Google Scholar] [CrossRef] [PubMed]
  91. Morris, M.C.; Wang, Y.; Barnes, L.L.; Bennett, D.A.; Dawson-Hughes, B.; Booth, S.L. Nutrients and bioactives in green leafy vegetables and cognitive decline: Prospective study. Neurology 2018, 90, e214–e222. [Google Scholar] [CrossRef] [PubMed]
  92. Weitzberg, E.; Lundberg, J.O. Novel Aspects of Dietary Nitrate and Human Health. Annu. Rev. Nutr. 2013, 33, 129–159. [Google Scholar] [CrossRef]
  93. Lidder, S.; Webb, A.J. Vascular effects of dietary nitrate (as found in green leafy vegetables and beetroot) via the nitrate-nitrite-nitric oxide pathway. Br. J. Clin. Pharm. 2013, 75, 677–696. [Google Scholar] [CrossRef] [Green Version]
  94. Roberts, J.L.; Moreau, R. Functional properties of spinach (Spinacia oleracea L.) phytochemicals and bioactives. Food Funct. 2016, 7, 3337–3353. [Google Scholar] [CrossRef]
  95. Cho, H.J.; Choi, S.A.; Kim, C.G.; Hong, J.H.; Park, H.J.; Park, H.J. Dietary spinach saponin-enriched lipophilic fraction inhibits platelet aggregation and blood coagulation. J. Med. Food 2011, 14, 784–791. [Google Scholar] [CrossRef]
  96. Cho, H.-J.; Choi, S.-A.; Kim, C.-G.; Jung, T.-S.; Hong, J.H.; Rhee, M.; Park, H.; Park, H.-J. Spinach Saponin-Enriched Fraction Inhibits Platelet Aggregation in cAMP- and cGMP-Dependent Manner by Decreasing TXA2 Production and Blood Coagulation. Biomol. Ther. 2011, 19, 218–223. [Google Scholar] [CrossRef] [Green Version]
  97. Shi, J.; Arunasalam, K.; Yeung, D.; Kakuda, Y.; Mittal, G.; Jiang, Y. Saponins from Edible Legumes: Chemistry, Processing, and Health Benefits. J. Med. Food 2004, 7, 67–78. [Google Scholar] [CrossRef] [PubMed]
  98. Fuentes, E.; Alarcón, M.; Fuentes, M.; Carrasco, G.; Palomo, I. A Novel Role of Eruca sativa Mill. (Rocket) Extract: Antiplatelet (NF-κB Inhibition) and Antithrombotic Activities. Nutrients 2014, 6, 5839–5852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Perveen, R.; Suleria, H.A.R.; Anjum, F.M.; Butt, M.S.; Pasha, I.; Ahmad, S. Tomato (Solanum lycopersicum) Carotenoids and Lycopenes Chemistry; Metabolism, Absorption, Nutrition, and Allied Health Claims—A Comprehensive Review. Crit. Rev. Food Sci. Nutr. 2015, 55, 919–929. [Google Scholar] [CrossRef] [PubMed]
  100. Chaudhary, P.; Sharma, A.; Singh, B.; Nagpal, A.K. Bioactivities of phytochemicals present in tomato. J. Food Sci. Technol. 2018, 55, 2833–2849. [Google Scholar] [CrossRef]
  101. Frusciante, L.; Carli, P.; Ercolano, M.R.; Pernice, R.; Di Matteo, A.; Fogliano, V.; Pellegrini, N. Antioxidant nutritional quality of tomato. Mol. Nutr. Food Res. 2007, 51, 609–617. [Google Scholar] [CrossRef] [PubMed]
  102. Fuentes, E.; Castro, R.; Astudillo, L.; Carrasco, G.; Alarcón, M.; Gutiérrez, M.; Palomo, I. Bioassay-Guided Isolation and HPLC Determination of Bioactive Compound That Relate to the Antiplatelet Activity (Adhesion, Secretion, and Aggregation) from Solanum lycopersicum. Evid.-Based Complementary Altern. Med. 2012, 2012, 147031. [Google Scholar] [CrossRef]
  103. Burch, L.R.; Stuchbury, T. Metabolism of purine nucleotides in the tomato plant. Phytochemistry 1986, 25, 2445–2449. [Google Scholar] [CrossRef]
  104. Stasolla, C.; Katahira, R.; Thorpe, T.A.; Ashihara, H. Purine and pyrimidine nucleotide metabolism in higher plants. J. Plant Physiol. 2003, 160, 1271–1295. [Google Scholar] [CrossRef]
  105. Fuentes, E.; Alarcón, M.; Astudillo, L.; Valenzuela, C.; Gutiérrez, M.; Palomo, I. Protective Mechanisms of Guanosine from Solanum lycopersicum on Agonist-Induced Platelet Activation: Role of sCD40L. Molecules 2013, 18, 8120–8135. [Google Scholar] [CrossRef]
  106. Lazarus, S.A.; Garg, M.L. Tomato extract inhibits human platelet aggregation in vitro without increasing basal cAMP levels. Int. J. Food Sci. Nutr. 2004, 55, 249–256. [Google Scholar] [CrossRef] [PubMed]
  107. Palomo, I.; Concha-Meyer, A.; Lutz, M.; Said, M.; Sáez, B.; Vásquez, A.; Fuentes, E. Chemical Characterization and Antiplatelet Potential of Bioactive Extract from Tomato Pomace (Byproduct of Tomato Paste). Nutrients 2019, 11, 456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Fuentes, E.; Forero-Doria, O.; Carrasco, G.; Maricán, A.; Santos, L.S.; Alarcón, M.; Palomo, I. Effect of Tomato Industrial Processing on Phenolic Profile and Antiplatelet Activity. Molecules 2013, 18, 11526–11536. [Google Scholar] [CrossRef] [Green Version]
  109. Sawardekar, S.; Patel, T.; Uchil, D. Comparative evaluation of antiplatelet effect of lycopene with aspirin and the effect of their combination on platelet aggregation: An in vitro study. Indian J. Pharmacol. 2016, 48, 26–31. [Google Scholar] [CrossRef]
  110. Mayeaux, M.; King, J.; Prinyawiwatkul, W. Effects of Cooking Conditions on the Lycopene Content in Tomatoes. J. Food Sci. 2006, 71, C461–C464. [Google Scholar] [CrossRef]
  111. Story, E.N.; Kopec, R.E.; Schwartz, S.J.; Harris, G.K. An Update on the Health Effects of Tomato Lycopene. Annu. Rev. Food Sci. Technol. 2010, 1, 189–210. [Google Scholar] [CrossRef] [Green Version]
  112. Zhang, Q.; Zhang, X.-G.; Liu, L.; Zhang, Q.-L.; Ding, S.-L.; Chen, Y.; Wang, J.-Y.; Wang, L.; Liang, R.-X.; Liao, F.-L.; et al. Effects of water-soluble tomato concentrate on platelet aggregation. World J. Tradit. Chin. Med. 2019, 5, 260–268. [Google Scholar] [CrossRef]
  113. O’Kennedy, N.; Raederstorff, D.; Duttaroy, A.K. Fruitflow®: The first European Food Safety Authority-approved natural cardio-protective functional ingredient. Eur. J. Nutr. 2017, 56, 461–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Willcox, J.K.; Catignani, G.L.; Lazarus, S. Tomatoes and cardiovascular health. Crit. Rev. Food Sci. Nutr. 2003, 43, 1–18. [Google Scholar] [CrossRef] [PubMed]
  115. Olas, B. The multifunctionality of berries toward blood platelets and the role of berry phenolics in cardiovascular disorders. Platelets 2017, 28, 540–549. [Google Scholar] [CrossRef] [PubMed]
  116. Sayegh, M.; Miglio, C.; Ray, S. Potential cardiovascular implications of Sea Buckthorn berry consumption in humans. Int. J. Food Sci. Nutr. 2014, 65, 521–528. [Google Scholar] [CrossRef]
  117. Basu, A.; Nguyen, A.; Betts, N.M.; Lyons, T.J. Strawberry As a Functional Food: An Evidence-Based Review. Crit. Rev. Food Sci. Nutr. 2014, 54, 790–806. [Google Scholar] [CrossRef] [PubMed]
  118. Proteggente, A.R.; Pannala, A.S.; Paganga, G.; Buren, L.v.; Wagner, E.; Wiseman, S.; Put, F.v.d.; Dacombe, C.; Rice-Evans, C.A. The Antioxidant Activity of Regularly Consumed Fruit and Vegetables Reflects their Phenolic and Vitamin C Composition. Free Radic. Res. 2002, 36, 217–233. [Google Scholar] [CrossRef]
  119. Alarcón, M.; Fuentes, E.; Olate, N.; Navarrete, S.; Carrasco, G.; Palomo, I. Strawberry extract presents antiplatelet activity by inhibition of inflammatory mediator of atherosclerosis (sP-selectin, sCD40L, RANTES, and IL-1β) and thrombus formation. Platelets 2015, 26, 224–229. [Google Scholar] [CrossRef]
  120. Maron, B.A.; Loscalzo, J. The Treatment of Hyperhomocysteinemia. Annu. Rev. Med. 2009, 60, 39–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Malinowska, J.; Babicz, K.; Olas, B.; Stochmal, A.; Oleszek, W. Aronia melanocarpa extract suppresses the biotoxicity of homocysteine and its metabolite on the hemostatic activity of fibrinogen and plasma. Nutrition 2012, 28, 793–798. [Google Scholar] [CrossRef]
  122. Oszmiański, J.; Wojdylo, A. Aronia melanocarpa phenolics and their antioxidant activity. Eur. Food Res. Technol. 2005, 221, 809–813. [Google Scholar] [CrossRef]
  123. Olas, B.; Kontek, B.; Szczesna, M.; Grabarczyk, L.; Stochmal, A.; Zuchowski, J. Inhibition of blood platelet adhesion by phenolics’ rich fraction of Hippophae rhamnoides L. fruits. J. Physiol. Pharm. 2017, 68, 223–229. [Google Scholar]
  124. Ma, X.; Laaksonen, O.; Zheng, J.; Yang, W.; Trépanier, M.; Kallio, H.; Yang, B. Flavonol glycosides in berries of two major subspecies of sea buckthorn (Hippophaë rhamnoides L.) and influence of growth sites. Food Chem. 2016, 200, 189–198. [Google Scholar] [CrossRef]
  125. Settu, K.; Manju, V. Pharmacological Applications of Isorhamnetin. Int. J. Trend Sci. Res. Dev. 2017, 1, 672–678. [Google Scholar]
  126. Skalski, B.; Lis, B.; Pecio, Ł.; Kontek, B.; Olas, B.; Żuchowski, J.; Stochmal, A. Isorhamnetin and its new derivatives isolated from sea buckthorn berries prevent H2O2/Fe—Induced oxidative stress and changes in hemostasis. Food Chem. Toxicol. 2019, 125, 614–620. [Google Scholar] [CrossRef] [PubMed]
  127. Oyemitan, I.A.; Elusiyan, C.A.; Onifade, A.O.; Akanmu, M.A.; Oyedeji, A.O.; McDonald, A.G. Neuropharmacological profile and chemical analysis of fresh rhizome essential oil of Curcuma longa (turmeric) cultivated in Southwest Nigeria. Toxicol. Rep. 2017, 4, 391–398. [Google Scholar] [CrossRef] [PubMed]
  128. Monton, C.; Charoenchai, L.; Suksaeree, J.; Sueree, L. Quantitation of curcuminoid contents, dissolution profile, and volatile oil content of turmeric capsules produced at some secondary government hospitals. J. Food Drug Anal. 2016, 24, 493–499. [Google Scholar] [CrossRef] [Green Version]
  129. Pothitirat, W.; Gritsanapan, W. Quantitative Analysis of Curcumin, Demethoxycurcumin and Bisdemethoxycurcumin in the Crude Curcuminoid Extract from Curcuma longa in Thailand by TLC-Densitometry. J. Pharm. Sci. 2005, 32, 23–30. [Google Scholar]
  130. Ngo, T.; Kim, K.; Bian, Y.; An, G.-J.; Bae, O.-N.; Lim, K.-M.; Chung, J.-H. Cyclocurcumin from Curcuma longa selectively inhibits shear stress-induced platelet aggregation. J. Funct. Foods 2019, 61, 103462. [Google Scholar] [CrossRef]
  131. Rana, A.; Westein, E.; Niego, B.E.; Hagemeyer, C.E. Shear-Dependent Platelet Aggregation: Mechanisms and Therapeutic Opportunities. Front. Cardiovasc. Med. 2019, 6, 141. [Google Scholar] [CrossRef]
  132. Maheswaraiah, A.; Rao, L.J.; Naidu, K.A. Anti-platelet activity of water dispersible curcuminoids in rat platelets. Phytother. Res. 2015, 29, 450–458. [Google Scholar] [CrossRef]
  133. Lee, H.S. Antiplatelet property of Curcuma longa L. rhizome-derived ar-turmerone. Bioresour. Technol. 2006, 97, 1372–1376. [Google Scholar] [CrossRef]
  134. Orellana-Paucar, A.M.; Afrikanova, T.; Thomas, J.; Aibuldinov, Y.K.; Dehaen, W.; de Witte, P.A.M.; Esguerra, C.V. Insights from Zebrafish and Mouse Models on the Activity and Safety of Ar-Turmerone as a Potential Drug Candidate for the Treatment of Epilepsy. PLoS ONE 2013, 8, e81634. [Google Scholar] [CrossRef]
  135. Hori, Y.; Tsutsumi, R.; Nasu, K.; Boateng, A.; Ashikari, Y.; Sugiura, M.; Nakajima, M.; Kurauchi, Y.; Hisatsune, A.; Katsuki, H.; et al. Aromatic-Turmerone Analogs Protect Dopaminergic Neurons in Midbrain Slice Cultures through Their Neuroprotective Activities. Cells 2021, 10, 1090. [Google Scholar] [CrossRef] [PubMed]
  136. Hucklenbroich, J.; Klein, R.; Neumaier, B.; Graf, R.; Fink, G.R.; Schroeter, M.; Rueger, M.A. Aromatic-turmerone induces neural stem cell proliferation in vitro and in vivo. Stem Cell Res. Ther. 2014, 5, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Dohare, P.; Varma, S.; Ray, M. Curcuma oil modulates the nitric oxide system response to cerebral ischemia/reperfusion injury. Nitric Oxide 2008, 19, 1–11. [Google Scholar] [CrossRef] [PubMed]
  138. Dohare, P.; Garg, P.; Sharma, U.; Jagannathan, N.R.; Ray, M. Neuroprotective efficacy and therapeutic window of curcuma oil: In rat embolic stroke model. BMC Complementary Altern. Med. 2008, 8, 55. [Google Scholar] [CrossRef] [Green Version]
  139. Prakash, P.; Misra, A.; Surin, W.R.; Jain, M.; Bhatta, R.S.; Pal, R.; Raj, K.; Barthwal, M.K.; Dikshit, M. Anti-platelet effects of Curcuma oil in experimental models of myocardial ischemia-reperfusion and thrombosis. Thromb. Res. 2011, 127, 111–118. [Google Scholar] [CrossRef]
  140. Mayanglambam, A.; Dangelmaier, C.A.; Thomas, D.; Damodar Reddy, C.; Daniel, J.L.; Kunapuli, S.P. Curcumin inhibits GPVI-mediated platelet activation by interfering with the kinase activity of Syk and the subsequent activation of PLCγ2. Platelets 2010, 21, 211–220. [Google Scholar] [CrossRef] [PubMed]
  141. Dissanayake, K.G.C.; Liyanage, R.; Waliwita, W.A.L.C. A review on the pharmacological activity of ginger (Zingiber officinale). Int. J. Health Sci. Res. 2020, 10, 142–148. [Google Scholar]
  142. Mohd Yusof, Y.A. Gingerol and Its Role in Chronic Diseases. In Drug Discovery from Mother Nature; Gupta, S.C., Prasad, S., Aggarwal, B.B., Eds.; Springer International Publishing: Cham, Switzerland, 2016; pp. 177–207. [Google Scholar]
  143. Semwal, R.B.; Semwal, D.K.; Combrinck, S.; Viljoen, A.M. Gingerols and shogaols: Important nutraceutical principles from ginger. Phytochemistry 2015, 117, 554–568. [Google Scholar] [CrossRef]
  144. Wei, C.-K.; Tsai, Y.-H.; Korinek, M.; Hung, P.-H.; El-Shazly, M.; Cheng, Y.-B.; Wu, Y.-C.; Hsieh, T.-J.; Chang, F.-R. 6-Paradol and 6-Shogaol, the Pungent Compounds of Ginger, Promote Glucose Utilization in Adipocytes and Myotubes, and 6-Paradol Reduces Blood Glucose in High-Fat Diet-Fed Mice. Int. J. Mol. Sci. 2017, 18, 168. [Google Scholar] [CrossRef] [Green Version]
  145. Bordia, A.; Verma, S.K.; Srivastava, K.C. Effect of ginger (Zingiber officinale Rosc.) and fenugreek (Trigonella foenumgraecum L.) on blood lipids, blood sugar and platelet aggregation in patients with coronary artery disease. Prostaglandins Leukot. Essent. Fat. Acids 1997, 56, 379–384. [Google Scholar] [CrossRef]
  146. Nurtjahja-Tjendraputra, E.; Ammit, A.J.; Roufogalis, B.D.; Tran, V.H.; Duke, C.C. Effective anti-platelet and COX-1 enzyme inhibitors from pungent constituents of ginger. Thromb. Res. 2003, 111, 259–265. [Google Scholar] [CrossRef] [PubMed]
  147. Pennington, J.A.T.; Fisher, R.A. Classification of fruits and vegetables. J. Food Compos. Anal. 2009, 22, S23–S31. [Google Scholar] [CrossRef]
  148. Hongyun, L.; Lou, H.; Hu, J.; Liu, Z.; Chen, Q. Macrofungi: A review of cultivation strategies, bioactivity, and application of mushrooms. Compr. Rev. Food Sci. Food Saf. 2020, 19, 2333–2356. [Google Scholar] [CrossRef]
  149. Manninen, H.; Rotola-Pukkila, M.; Aisala, H.; Hopia, A.; Laaksonen, T. Free amino acids and 5′-nucleotides in Finnish forest mushrooms. Food Chem. 2018, 247, 23–28. [Google Scholar] [CrossRef] [PubMed]
  150. Reis, F.S.; Barros, L.; Martins, A.; Ferreira, I.C.F.R. Chemical composition and nutritional value of the most widely appreciated cultivated mushrooms: An inter-species comparative study. Food Chem. Toxicol. 2012, 50, 191–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Liu, K.; Wang, J.; Zhao, L.; Wang, Q. Anticancer, antioxidant and antibiotic activities of mushroom Ramaria flava. Food Chem. Toxicol. 2013, 58, 375–380. [Google Scholar] [CrossRef]
  152. Roncero-Ramos, I.; Delgado-Andrade, C. The beneficial role of edible mushrooms in human health. Curr. Opin. Food Sci. 2017, 14, 122–128. [Google Scholar] [CrossRef]
  153. Dai, X.; Stanilka, J.M.; Rowe, C.A.; Esteves, E.A.; Nieves, C.; Spaiser, S.J.; Christman, M.C.; Langkamp-Henken, B.; Percival, S.S. Consuming Lentinula edodes (Shiitake) Mushrooms Daily Improves Human Immunity: A Randomized Dietary Intervention in Healthy Young Adults. J. Am. Coll. Nutr. 2015, 34, 478–487. [Google Scholar] [CrossRef]
  154. Jose, N.; Ajith, T.A.; Janardhanan, K.K. Methanol extract of the oyster mushroom, Pleurotus florida, inhibits inflammation and platelet aggregation. Phytother. Res. 2004, 18, 43–46. [Google Scholar] [CrossRef]
  155. Suseem, S.R.; Saral, M. Inhibition of platelet aggregation and in vitro free radical scavenging activity of dried fruiting bodies of Pleurotus eous. Chin. J. Integr. Med. 2015, 21, 530–536. [Google Scholar] [CrossRef]
  156. Poniedziałek, B.; Siwulski, M.; Wiater, A.; Komaniecka, I.; Komosa, A.; Gąsecka, M.; Magdziak, Z.; Mleczek, M.; Niedzielski, P.; Proch, J.; et al. The Effect of Mushroom Extracts on Human Platelet and Blood Coagulation: In vitro Screening of Eight Edible Species. Nutrients 2019, 11, 3040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Singdevsachan, S.K.; Auroshree, P.; Mishra, J.; Baliyarsingh, B.; Tayung, K.; Thatoi, H. Mushroom polysaccharides as potential prebiotics with their antitumor and immunomodulating properties: A review. Bioact. Carbohydr. Diet. Fibre 2016, 7, 1–14. [Google Scholar] [CrossRef]
  158. Chen, S.; Yong, T.; Zhang, Y.; Su, J.; Jiao, C.; Xie, Y. Anti-tumor and Anti-angiogenic Ergosterols from Ganoderma lucidum. Front. Chem. 2017, 5, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Mori, K.; Kikuchi, H.; Obara, Y.; Iwashita, M.; Azumi, Y.; Kinugasa, S.; Inatomi, S.; Oshima, Y.; Nakahata, N. Inhibitory effect of hericenone B from Hericium erinaceus on collagen-induced platelet aggregation. Phytomedicine 2010, 17, 1082–1085. [Google Scholar] [CrossRef] [PubMed]
  160. Kim, S.D.; Lee, I.-K.; Lee, W.M.; Cho, J.Y.; Park, H.J.; Oh, J.-W.; Park, S.C.; Kim, S.K.; Kwak, Y.S.; Yun, B.-S.; et al. The mechanism of anti-platelet activity of davallialactone: Involvement of intracellular calcium ions, extracellular signal-regulated kinase 2 and p38 mitogen-activated protein kinase. Eur. J. Pharmacol. 2008, 584, 361–367. [Google Scholar] [CrossRef] [PubMed]
  161. Park, J.Y.; Oh, W.J.; Kwak, D.M.; Kim, M.G.; Seo, G.S.; Hong, S.B.; Rhee, M.H. The anti-platelet activity of Hypsizygus marmoreus extract is involved in the suppression of intracellular calcium mobilization and integrin αIIbβ3 activation. J. Med. Plants Res. 2011, 5, 2369–2377. [Google Scholar]
  162. Choi, E.; Oh, J.; Sung, G.-H. Antithrombotic and Antiplatelet Effects of Cordyceps militaris. Mycobiology 2020, 48, 228–232. [Google Scholar] [CrossRef]
  163. Liperoti, R.; Vetrano, D.L.; Bernabei, R.; Onder, G. Herbal Medications in Cardiovascular Medicine. J. Am. Coll. Cardiol. 2017, 69, 1188–1199. [Google Scholar] [CrossRef]
  164. Tachjian, A.; Maria, V.; Jahangir, A. Use of Herbal Products and Potential Interactions in Patients With Cardiovascular Diseases. J. Am. Coll. Cardiol. 2010, 55, 515–525. [Google Scholar] [CrossRef] [Green Version]
  165. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs from 1981 to 2014. J. Nat. Prod. 2016, 79, 629–661. [Google Scholar] [CrossRef] [Green Version]
  166. Shaito, A.; Thuan, D.T.B.; Phu, H.T.; Nguyen, T.H.D.; Hasan, H.; Halabi, S.; Abdelhady, S.; Nasrallah, G.K.; Eid, A.H.; Pintus, G. Herbal Medicine for Cardiovascular Diseases: Efficacy, Mechanisms, and Safety. Front. Pharmacol. 2020, 11. [Google Scholar] [CrossRef] [Green Version]
  167. Lin, T.-H.; Hsieh, C.-L. Pharmacological effects of Salvia miltiorrhiza (Danshen) on cerebral infarction. Chin. Med. 2010, 5, 22. [Google Scholar] [CrossRef] [Green Version]
  168. Caliceti, C.; Franco, P.; Spinozzi, S.; Roda, A.; FG Cicero, A. Berberine: New Insights from Pharmacological Aspects to Clinical Evidences in the Management of Metabolic Disorders. Curr. Med. Chem. 2016, 23, 1460–1476. [Google Scholar] [CrossRef]
  169. Yu, L.; Qin, Y.; Wang, Q.; Zhang, L.; Liu, Y.; Wang, T.; Huang, L.; Wu, L.; Xiong, H. The efficacy and safety of Chinese herbal medicine, Rhodiola formulation in treating ischemic heart disease: A systematic review and meta-analysis of randomized controlled trials. Complementary Ther. Med. 2014, 22, 814–825. [Google Scholar] [CrossRef] [PubMed]
  170. Grech-Baran, M.; Sykłowska-Baranek, K.; Pietrosiuk, A. Approaches of Rhodiola kirilowii and Rhodiola rosea field cultivation in Poland and their potential health benefits. Ann. Agric. Environ. Med. 2015, 22, 281–285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Liu, C.; Huang, Y. Chinese Herbal Medicine on Cardiovascular Diseases and the Mechanisms of Action. Front. Pharmacol. 2016, 7, 469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Ashfield-Watt, P.A. Fruits and vegetables, 5+ a day: Are we getting the message across? Asia Pac. J. Clin. Nutr. 2006, 15, 245–252. [Google Scholar] [PubMed]
  173. Wang, D.D.; Li, Y.; Bhupathiraju, S.N.; Rosner, B.A.; Sun, Q.; Giovannucci, E.L.; Rimm, E.B.; Manson, J.E.; Willett, W.C.; Stampfer, M.J.; et al. Fruit and Vegetable Intake and Mortality. Circulation 2021, 143, 1642–1654. [Google Scholar] [CrossRef]
  174. Hartley, L.; Igbinedion, E.; Holmes, J.; Flowers, N.; Thorogood, M.; Clarke, A.; Stranges, S.; Hooper, L.; Rees, K. Increased consumption of fruit and vegetables for the primary prevention of cardiovascular diseases. Cochrane Database Syst. Rev. 2013. [Google Scholar] [CrossRef] [Green Version]
  175. Joshipura, K.J.; Ascherio, A.; Manson, J.E.; Stampfer, M.J.; Rimm, E.B.; Speizer, F.E.; Hennekens, C.H.; Spiegelman, D.; Willett, W.C. Fruit and Vegetable Intake in Relation to Risk of Ischemic Stroke. JAMA 1999, 282, 1233–1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Ness, A.R.; Powles, J.W. The Role of Diet, Fruit and Vegetables and Antioxidants in the Aetiology of Stroke. J. Cardiovasc. Risk 1999, 6, 229–234. [Google Scholar] [CrossRef] [PubMed]
  177. Dauchet, L.; Amouyel, P.; Dallongeville, J. Fruit and vegetable consumption and risk of stroke. A Meta-Anal. Cohort Stud. 2005, 65, 1193–1197. [Google Scholar] [CrossRef]
  178. McCall, D.O.; McGartland, C.P.; McKinley, M.C.; Patterson, C.C.; Sharpe, P.; McCance, D.R.; Young, I.S.; Woodside, J.V. Dietary Intake of Fruits and Vegetables Improves Microvascular Function in Hypertensive Subjects in a Dose-Dependent Manner. Circulation 2009, 119, 2153–2160. [Google Scholar] [CrossRef] [Green Version]
  179. Steffen, L.M.; Folsom, A.R.; Cushman, M.; Jacobs, D.R.; Rosamond, W.D. Greater Fish, Fruit, and Vegetable Intakes Are Related to Lower Incidence of Venous Thromboembolism. Circulation 2007, 115, 188–195. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Chemical structures of quercetin (a), quercetin-3,4′-O-diglucoside (b) and quercetin-4′-O-monoglucoside (c) isolated from Allium cepa.
Figure 1. Chemical structures of quercetin (a), quercetin-3,4′-O-diglucoside (b) and quercetin-4′-O-monoglucoside (c) isolated from Allium cepa.
Ijms 23 00605 g001
Figure 2. Signalling pathways that are affected by selective plant extracts and their compounds in platelets. cAMP; cyclic adenosine monophosphate, DAG; diacylglycerol, IP3; inositol trisphosphate, PI3K; phosphoinositide 3-kinase, PKB; Protein kinase B, PKC; Protein kinase C, PLCγ2; phospholipase Cγ2, and Syk; spleen tyrosine kinase. This image was created in BioRender.com (accessed on 23 December 2021). using the information provided in this article.
Figure 2. Signalling pathways that are affected by selective plant extracts and their compounds in platelets. cAMP; cyclic adenosine monophosphate, DAG; diacylglycerol, IP3; inositol trisphosphate, PI3K; phosphoinositide 3-kinase, PKB; Protein kinase B, PKC; Protein kinase C, PLCγ2; phospholipase Cγ2, and Syk; spleen tyrosine kinase. This image was created in BioRender.com (accessed on 23 December 2021). using the information provided in this article.
Ijms 23 00605 g002
Figure 3. Structure of alliin (a) and (b) allicin isolated from the methanolic extract of A. sativum.
Figure 3. Structure of alliin (a) and (b) allicin isolated from the methanolic extract of A. sativum.
Ijms 23 00605 g003
Figure 4. Chemical structures of (a) N-feruloyltyramine, (b) N-caffeoylnorephedrine and (c) N-caffeoyltyramine isolated from methanolic extracts of A. sativum.
Figure 4. Chemical structures of (a) N-feruloyltyramine, (b) N-caffeoylnorephedrine and (c) N-caffeoyltyramine isolated from methanolic extracts of A. sativum.
Ijms 23 00605 g004
Figure 5. Chemical structures of (a) 1,2-di-O-α-linolenoyl-3-O-β-d-galactopyranosyl-sn-glycerol and (b) β-sitosterol-3-O-β-d-glucoside isolated from chloroform fractions of methonolic extract of A. ursinum.
Figure 5. Chemical structures of (a) 1,2-di-O-α-linolenoyl-3-O-β-d-galactopyranosyl-sn-glycerol and (b) β-sitosterol-3-O-β-d-glucoside isolated from chloroform fractions of methonolic extract of A. ursinum.
Ijms 23 00605 g005
Figure 6. Basic structure of anthocyanins (flavylium cation).
Figure 6. Basic structure of anthocyanins (flavylium cation).
Ijms 23 00605 g006
Figure 7. Structure of sulforaphane, an abundant sulphur compound in cruciferous vegetables.
Figure 7. Structure of sulforaphane, an abundant sulphur compound in cruciferous vegetables.
Ijms 23 00605 g007
Figure 8. Structure of indole-3-carbinol isolated from different Brassica species.
Figure 8. Structure of indole-3-carbinol isolated from different Brassica species.
Ijms 23 00605 g008
Figure 9. Structure of purine nucleotides isolated from S. lycopersicum; (a) adenosine and (b) guanosine.
Figure 9. Structure of purine nucleotides isolated from S. lycopersicum; (a) adenosine and (b) guanosine.
Ijms 23 00605 g009
Figure 10. Structure of lycopene, the major constituent of S. lycopersicum.
Figure 10. Structure of lycopene, the major constituent of S. lycopersicum.
Ijms 23 00605 g010
Figure 11. Structure of isorhamnetin isolated from H. rhamnoides.
Figure 11. Structure of isorhamnetin isolated from H. rhamnoides.
Ijms 23 00605 g011
Figure 12. Structure of curcuminoids: (a) curcumin, (b) demethoxycurcumin; (c) bisdemethoxycurcumin isolated from C. longa.
Figure 12. Structure of curcuminoids: (a) curcumin, (b) demethoxycurcumin; (c) bisdemethoxycurcumin isolated from C. longa.
Ijms 23 00605 g012
Figure 13. Structure of cyclocurcumin, an active constituent of C. Longa ethanolic extract.
Figure 13. Structure of cyclocurcumin, an active constituent of C. Longa ethanolic extract.
Ijms 23 00605 g013
Figure 14. Structure of ar-turmerone, a major active component of C. Loga essential oil.
Figure 14. Structure of ar-turmerone, a major active component of C. Loga essential oil.
Ijms 23 00605 g014
Figure 15. Chemical structures of different gingerols (a), shogaols (b) and paradols (c).
Figure 15. Chemical structures of different gingerols (a), shogaols (b) and paradols (c).
Ijms 23 00605 g015
Figure 16. Structure of ergosterol.
Figure 16. Structure of ergosterol.
Ijms 23 00605 g016
Figure 17. Structure of hericenone B isolated from ethanolic extract of Hericium erinaceus.
Figure 17. Structure of hericenone B isolated from ethanolic extract of Hericium erinaceus.
Ijms 23 00605 g017
Figure 18. Structure of davallialactone isolated from Inonotus xeranticus mushroom.
Figure 18. Structure of davallialactone isolated from Inonotus xeranticus mushroom.
Ijms 23 00605 g018
Table 1. The effects of conventional oven and microwave cooking, different preparation methods and cooking time on antiplatelet activities of A. cepa [49].
Table 1. The effects of conventional oven and microwave cooking, different preparation methods and cooking time on antiplatelet activities of A. cepa [49].
Cooking Time (min).Type of ProcessingEffects on Platelet Aggregation
Conventional Oven Cooking
10CrushedNo inhibition or activation effects
20 & 30Pro-aggregatory by ~25%
10Chopped into quarters85% inhibition of aggregation
20 & 30Pro-aggregatory by ~40%
10 & 20Whole (intact) bulb85% inhibition of aggregation
30Pro-aggregatory by ~30%
Microwave cooking
2CrushedInsignificant inhibition
4No inhibition or activation effects
8Pro-aggregatory by ~25%
2Whole (intact) bulbPro-aggregatory by ~20%
4Pro-aggregatory by ~30%
8Pro-aggregatory by ~40%
Table 2. Effects of extracts of selected cruciferous vegetables on human-platelet activation induced by ADP and AA [72].
Table 2. Effects of extracts of selected cruciferous vegetables on human-platelet activation induced by ADP and AA [72].
(%) Inhibition of Platelet Aggregation Induced by
Vegetable NameExtractADPAA
Brassica oleracea L. var. capitata (cabbage)Ethyl acetate28%88%
Brassica oleracea var. Italica (broccoli)40%17%
Brassica oleracea var. botrytis L. (cauliflower)8%10%
Brassica rapa subsp. rapa (turnip)30%80%
Wasabia japonica (wasabi)62%90%
Brassica oleracea L. var. capitata (cabbage)n-Butanol22%60%
Brassica oleracea var. Italica (broccoli)33%0%
Brassica oleracea var. botrytis L. (cauliflower)10%0%
Brassica rapa subsp. rapa (turnip)4%5%
Wasabia japonica (wasabi)58%62%
Table 3. Antiplatelet effects of aqueous extracts of S. lycopersicum and its isolated phenolic compounds [108].
Table 3. Antiplatelet effects of aqueous extracts of S. lycopersicum and its isolated phenolic compounds [108].
KERRYPNX (%) Inhibition on Aggregation
ADPCollagenAATRAP-6
Aqueous Extract
Fresh fruits40208 (ns)3 (ns)
Sauce48307 (ns)5 (ns)
Ketchup50406 (ns)10 (ns)
Juice302810 (ns)6 (ns)
Pomace38382022
Isolated Compounds
Caffeic acid35422025
Chlorogenic acid69502219
Ferulic acid4736ns *ns *
p-Coumaric acid716941ns *
ns = not significant and * ns = not significant and not mentioned in the paper.
Table 4. Inhibitory effects of lycopene and its combination with aspirin on ADP- and collagen-stimulated platelets [109].
Table 4. Inhibitory effects of lycopene and its combination with aspirin on ADP- and collagen-stimulated platelets [109].
(%) Inhibition on Aggregation
ADPCollagen
Lycopene Concentration (µM)
441.2250.01
643.1949.86
843.6251.89
1045.1851.48
1244.4749.20
Lycopene (L, µM) and Aspirin (A, µM) Combinations
L4 + A7053.1973.17
L8 + A7043.4766.97
L4 + A14053.1271.27
L8 + A14047.7168.49
Table 5. Inhibitory effects of different extracts of A. melanocarpa combined with Hcy (homocysteine) and HTL (homocysteine thiolactone) on platelet aggregation and adhesion [121].
Table 5. Inhibitory effects of different extracts of A. melanocarpa combined with Hcy (homocysteine) and HTL (homocysteine thiolactone) on platelet aggregation and adhesion [121].
(%) Inhibition
Platelet AggregationCollagen AdhesionFibrinogen Adhesion
Extract (µg/mL)
2.529.411.700.10
556.472.301.10
1065.884.204.10
Extract (µg/mL) + Hcy (100 µM)
2.5 + Hcy18.409.0012.00
5 + Hcy40.5018.9029.50
10 + Hcy48.9032.3043.80
Extract (µg/mL) + HTL (1 µM)
2.5 + HTL26.3016.3017.70
5 + HTL39.4035.0033.80
10 + HTL51.3047.4045.50
Table 6. Inhibitory effects of various compounds isolated from C. longa on platelets [130]. SIPA-shear stress induced platelet aggregation.
Table 6. Inhibitory effects of various compounds isolated from C. longa on platelets [130]. SIPA-shear stress induced platelet aggregation.
Isolated Compounds (µg/mL)(%) Inhibition of SIPA
Artumerone20
Bisabolatraen30
Bisacurone20
Bisdemethoxycurcumin18
Curcumin15
4-Dehydroxybisacurone16
Demethoxycurcumin5
b-Hydroxycinnamic acid19
b-Sitosterol22
Table 7. Inhibitory effects of curcuminoids and curcumin on rat PRP aggregation [132].
Table 7. Inhibitory effects of curcuminoids and curcumin on rat PRP aggregation [132].
(%) Inhibition
ADPCollagenAA
Curcuminoids (µg/mL)
10152018
20384045
30708078
Curcumin (µg/mL)
10151520
30203850
60707080
Table 8. Inhibitory effects of phenolic compounds of Z. officinale on whole blood aggregation [146].
Table 8. Inhibitory effects of phenolic compounds of Z. officinale on whole blood aggregation [146].
% Inhibition
Isolated Compounds
6-Gingerol86
8-Gingerol96
9-Gingerol86
12-Gingerol100
6-Paradol90
8-Paradol100
6-Shogaol91
8-Shogaol100
Synthetic analogues
3-Hydroxy-1-(4-hydroxy-3-methoxyphenyl) dec-4-ene92
2-Hydroxy-1-(4-hydroxy-3-methoxyphenyl) dodecan-3-one96
5-Hydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl)-hept-6-ene-3-one100
3-Hydroxy-1,7-bis(4-hydroxy-3-methoxyphenyl) heptane100
Table 9. Plant extracts and/or their compounds with antiplatelet effects that are currently in clinical trials [166,167,168].
Table 9. Plant extracts and/or their compounds with antiplatelet effects that are currently in clinical trials [166,167,168].
Antiplatelet ExtractsIsolated CompoundsStudy Phase
Ginseng root extract:
Panax ginseng (Korean red ginseng)
Panax notoginseng (Chinese ginseng)
Panax quinquefolium (American ginseng)
Panax japonicas (Japanese ginseng)
GinsenosidesPhase 3
Ginkgo biloba (maidenhair tree) leavesFlavonoids (ginkgo-flavone glycosides) and terpenoids (ginkgolides and bilobalide)Phase 3
Gynostemma pentaphyllum (miracle grass) leavesGypenoside saponinsPhase 2
Salvia miltiorrhiza (red sage) rootsSalvianolic acid BPhase 2
Not definedBerberinePhase 3
Colchicum autumnale (autumn crocus)ColchicinePhase 3
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Albadawi, D.A.I.; Ravishankar, D.; Vallance, T.M.; Patel, K.; Osborn, H.M.I.; Vaiyapuri, S. Impacts of Commonly Used Edible Plants on the Modulation of Platelet Function. Int. J. Mol. Sci. 2022, 23, 605. https://doi.org/10.3390/ijms23020605

AMA Style

Albadawi DAI, Ravishankar D, Vallance TM, Patel K, Osborn HMI, Vaiyapuri S. Impacts of Commonly Used Edible Plants on the Modulation of Platelet Function. International Journal of Molecular Sciences. 2022; 23(2):605. https://doi.org/10.3390/ijms23020605

Chicago/Turabian Style

Albadawi, Dina A. I., Divyashree Ravishankar, Thomas M. Vallance, Ketan Patel, Helen M. I. Osborn, and Sakthivel Vaiyapuri. 2022. "Impacts of Commonly Used Edible Plants on the Modulation of Platelet Function" International Journal of Molecular Sciences 23, no. 2: 605. https://doi.org/10.3390/ijms23020605

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