Plant extracts inhibit ADP-induced platelet activation in humans: their potential therapeutic role as ADP antagonists.
Journal: 2015/January - Purinergic Signalling
ISSN: 1573-9546
Abstract:
Adenosine diphosphate (ADP) plays a pivotal role in platelet activation. Platelet hyperactivity is associated with vascular disease and also has a key role in haemostasis and thrombosis. ADP activates platelets through three purinoceptor subtypes, the G(q)-coupled P2Y(1) receptor, G(i)-coupled P2Y(12) receptor and P2X(1) ligand-gated cation channel. Platelet ADP purinergic receptors are therefore suitable targets for antiplatelet drugs. Thienopyridines such as clopidogrel and ticlopidine, as well as other ADP receptor antagonists like prasugrel, ticagrelor, cangrelor and elinogrel have demonstrated clinical benefits via the inhibition of the selective purinergic ADP receptor, P2Y(12). However, they still have limitations in their mode of action and efficacy, like increased risk of bleeding. Thus, the ongoing pursuit to develop newer and more effective antiplatelet agents continues. There is a growing interest in the purinergic antiplatelet properties exhibited by plant extracts. This article considers the following: pomolic acid isolated from Licania pittieri, brazilin isolated from the heartwood of Caesalpinia sappan L, phylligenin isolated from the twigs of Muraltia vulpina, bark oil of Gonystylus velutinus, seed and bark extracts from Aesculus hippocastanum L. and red wine phenolics and catechins isolated from green tea. Moreover, the method used to investigate platelet purinergic receptors should be considered, since using a more sensitive, high-resolution platelet sizer can sometimes detect platelet variations when the light transmission method was not able to do so. The exact mechanisms by which these plant extracts work need further investigation. They all however inhibit ADP-induced activation in human platelets. This could explain, at least in part, the protective effect of plant extracts as antiplatelet agents.
Relations:
Content
Citations
(2)
References
(62)
Drugs
(2)
Chemicals
(4)
Organisms
(1)
Processes
(1)
Affiliates
(1)
Similar articles
Articles by the same authors
Discussion board
Purinergic Signal 10(2): 233-239

Plant extracts inhibit ADP-induced platelet activation in humans: their potential therapeutic role as ADP antagonists

Abstract

Adenosine diphosphate (ADP) plays a pivotal role in platelet activation. Platelet hyperactivity is associated with vascular disease and also has a key role in haemostasis and thrombosis. ADP activates platelets through three purinoceptor subtypes, the Gq-coupled P2Y1 receptor, Gi-coupled P2Y12 receptor and P2X1 ligand-gated cation channel. Platelet ADP purinergic receptors are therefore suitable targets for antiplatelet drugs. Thienopyridines such as clopidogrel and ticlopidine, as well as other ADP receptor antagonists like prasugrel, ticagrelor, cangrelor and elinogrel have demonstrated clinical benefits via the inhibition of the selective purinergic ADP receptor, P2Y12. However, they still have limitations in their mode of action and efficacy, like increased risk of bleeding. Thus, the ongoing pursuit to develop newer and more effective antiplatelet agents continues. There is a growing interest in the purinergic antiplatelet properties exhibited by plant extracts. This article considers the following: pomolic acid isolated from Licania pittieri, brazilin isolated from the heartwood of Caesalpinia sappan L, phylligenin isolated from the twigs of Muraltia vulpina, bark oil of Gonystylus velutinus, seed and bark extracts from Aesculus hippocastanum L. and red wine phenolics and catechins isolated from green tea. Moreover, the method used to investigate platelet purinergic receptors should be considered, since using a more sensitive, high-resolution platelet sizer can sometimes detect platelet variations when the light transmission method was not able to do so. The exact mechanisms by which these plant extracts work need further investigation. They all however inhibit ADP-induced activation in human platelets. This could explain, at least in part, the protective effect of plant extracts as antiplatelet agents.

Keywords: Adenosine diphosphate (ADP), Platelet, Plant extract, Antiplatelet drugs, Vascular disease
Academic Department of Surgery, Division of Surgical and Interventional Science, Royal Free Campus, University College London Medical School, University College London (UCL), Pond Street, London, NW3 2QG UK
Department of Clinical Biochemistry, Royal Free Campus, University College London Medical School, University College London (UCL), Pond Street, London, NW3 2QG UK
Indera Anita Jagroop, Phone: +44-20-7940500, Fax: +44-20-78302235, ku.ca.lcu@poorgaj.aredni.
Corresponding author.
Received 2013 Apr 23; Accepted 2013 Oct 10.

Adenosine-5′-diphosphate (ADP) plays a crucial role in platelet activation [1, 2]. In addition, platelet hyperactivity is known to be associated with vascular disease and has a key role in haemostasis and thrombosis [3, 4]. ADP is considered a weak platelet agonist, since it induces platelet shape change (PSC, an early phase of platelet activation) and reversible aggregation. Nevertheless, when ADP is secreted from its storage pools in platelet dense granules, it amplifies platelet responses induced by other platelet agonists [5, 6]. It is well established that ADP activates platelets through three purinoceptor subtypes, namely P2Y1, P2Y12 and P2X1 [1, 7, 8]. The P2Y1 purinergic receptor is coupled to Gq, which is involved in PSC and reversible aggregation, due to a transient increase in cytoplasmic Ca. The P2Y12 purinergic receptor is coupled to Gi which mediates the inhibition of adenylyl cyclase and amplifies the platelet aggregation response [6]. The P2X1 ligand-gated cation channel is activated by adenosine triphosphate (ATP) and is also involved in PSC [7, 9]. The simultaneous activation of both Gq and Gi pathways by ADP is necessary to elicit normal platelet aggregation [6]. Moreover, the limited distribution of the P2Y12 receptor, primarily expressed on platelets, makes it an especially attractive pharmacologic target for antithrombotic drugs [1012].

Over the last several decades, the thienopyridine family of P2Y12 receptor antagonists has provided the scientific data confirming the clinical benefit of selective purinergic ADP receptor, P2Y12, inhibition [1, 13]. In fact, the P2Y12 receptor is the target for thienopyridine compounds like ticlopidine and clopidogrel which have been used in the treatment of patients at risk for major adverse cardiovascular event (MACE) [1416]. Ticlopidine was first synthesized in 1974 [17], and clopidogrel was its successor [18]. Although thienopyridines have demonstrable antithrombotic efficacy, they still have limitations in their mode of action and efficacy. For example, both ticlopidine and clopidogrel are pro-drugs that need to be metabolised to its active metabolite. Also, antiplatelet agents like aspirin may cause gastric erosions and gastric ulcers, and patients may present with anaemia and gastric haemorrhage [19, 20]. These limitations have led to the search for other therapeutic P2Y12 antagonists.

Prasugrel, is a new FDA-approved ADP receptor antagonist. The metabolism of prasugrel to its active metabolite is much more efficient and consistent than that of clopidogrel; hence, it has a faster onset of action [21]. Prasugrel was also shown to be superior to clopidogrel in inhibiting P2Y12-dependent platelet function [21]. This accounts for the lower incidence of MACE, but the disadvantage is that it also increased bleeding among acute coronary syndrome (ACS) patients who required percutaneous coronary intervention (PCI) [21]. Furthermore, the results of the TRITON TIMI-38 trial showed that prasugrel was a more potent antiplatelet agent than clopidogrel when used only in high-risk patients or for a short period, while clopidogrel treatment is preferred in the remaining patient groups [22]. It is now known that clopidogrel treatment is ineffective in about 30 % of patients, thus other antithrombotic treatments have also been FDA-approved. One such drug is ticagrelor (previously known as AZD6140), a direct P2Y12 antagonist, which was shown to induce an appreciable inhibition of platelet function when given to patients who were poor responders to clopidogrel [12, 13, 23]. Ticagrelor has a rapid onset of actions and achieves a maximal inhibition of platelet function in about 2 h with much less inter-individual variability when compared to clopidogrel [23, 24]. Moreover, the DISPERSE study showed that overall, ticagrelor was superior to clopidogrel with regard to antiplatelet efficacy [25]. However, although ticagrelor shows some advantages over clopidogrel, the DISPERSE study also demonstrates that the incidence of bleeding (from 29 to 52 %) events was dose-related in ticagrelor-treated patients (50 mg b.i.d., 100 mg b.i.d., 200 mg b.i.d., or 400 mg q.d.), as compared to 32 % in clopidogrel-treated patients (75 mg once daily) [25].

Cangrelor (previously known as {"type":"entrez-protein","attrs":{"text":"ARC69931","term_id":"1169592793","term_text":"ARC69931"}}ARC69931[MX] is another direct and reversible P2Y12 antagonist, which can be given intravenously. It is immediately active since it does not have to be converted to an active metabolite [12, 13, 19]. Pharmacological studies showed that cangrelor inhibited ADP-induced aggregation in washed human platelets (IC50 = 9.4 with ADP 30 μM) [13, 26]. Moreover, Jagroop et al. [8] demonstrated that {"type":"entrez-protein","attrs":{"text":"ARC69931","term_id":"1169592793","term_text":"ARC69931"}}ARC69931[MX] inhibited ADP-induced PSC in a time-dependent manner in human platelets. This was the first group to actually quantify PSC inhibition by {"type":"entrez-protein","attrs":{"text":"ARC69931","term_id":"1169592793","term_text":"ARC69931"}}ARC69931[MX] or any other P2Y12 blocker. It is possible that {"type":"entrez-protein","attrs":{"text":"ARC69931","term_id":"1169592793","term_text":"ARC69931"}}ARC69931[MX] affects PSC due to its immediate effect and its high affinity for the P2Y12 receptors. Moreover, the findings of Jagroop et al. [8] suggest that the clinical effectiveness of P2Y12 antagonists may be linked to the fact that they inhibit both the early and late phases of platelet activation.

In addition to prasugrel, ticagrelor and cangrelor, the fourth P2Y12 inhibitor which has been investigated is elinogrel, which has direct and reversible actions. Elinogrel is the first P2Y12 antagonist to be available in both intravenous and oral formulations to enable a smooth transition from short- to long-term treatment [27, 28]. These four newly developed P2Y12 inhibitors offer a more potent, more predictable and faster onset of action than clopidogrel [1214]. However, their risk of major and even fatal bleeding needs to be considered, especially in elderly patients with comorbidities [29]. It should also be considered that in clinical trials, to assess the effectiveness of P2Y12 inhibitors, patients perceived to be at a higher risk of bleeding are usually excluded [29]. Thus, the ongoing pursuit to develop newer and more effective antiplatelet agents continues. As a result, there is a growing interest in the properties of plant extracts, and their antiplatelet effect on purinergic receptors. There is also a focus on the development of safe and effective antithrombotic drugs from plant-derived extracts. Furthermore, the importance of ADP in haemostasis and thrombosis greatly underscores the significance of understanding the function of platelet purinergic receptors with regard to drug development from plants.

The leaves of Licania pittieri plants (belonging to the Chrysobalanaceae family) are considered medicinal in South America and have historically been used to treat diseases such as diabetes and hypertension [30]. In an interesting study, Alvarado-Castillo et al. [31] evaluated pomolic acid (PA), a triterpenoid isolated from L. pittieri, as a competitive antagonist of ADP-induced aggregation of human platelets. The authors used light transmission aggregometry to assess the ability of PA (150 nM) to inhibit PSC induced by a selective P2Y1 receptor agonist, MRS 2365 (25 μM). They found that PA was not an antagonist of the P2Y1 receptor [31]. It would however be interesting to use another technique like a more sensitive, high-resolution platelet sizer to assess if there is indeed any P2Y1 purinergic effect associated with the plant isolate PA on PSC in human platelets [8]. There was also evidence in this study showing that PA (25–100 nM) acts as a competitive antagonist of ADP-induced (10–10 M) platelet aggregation in platelet-rich plasma (PRP). Thus, the authors concluded that PA may be a potent competitive antagonist of the P2Y12 receptor, a pharmacological characteristic shared by the new generation of P2Y12 receptor antagonists that are currently being investigated in clinical trials for their antiplatelet activity [31]. These results also provide evidence to suggest that purinergic receptor blockers isolated from the plants L. pittieri may be beneficial for their use as antiplatelet agents in humans [31]. However, further investigations are warranted to provide more data in support of this evidence.

With regard to using a more sensitive platelet sizer with a high-resolution channelyzer, I have shown that a P2Y1 receptor antagonist MRS 2179 (1.06–10.25 μmol/l) blocked ADP-induced (0.2–0.4 μM) PSC in human platelets by up to 100 % (median IC50 3.16 μmol/l) [8]. To the author's knowledge, this was the first study to demonstrate an IC50 for a P2Y1 receptor blocker using the human PSC phenomenon. These results are in line with the findings by Alvarado-Castillo et al. [31] who showed that MRS 2365-induced (25 μM, a P2Y1 agonist) PSC was completely blocked by MRS 2500 (100 nM, a P2Y1 antagonist) using human platelets in PRP. It was also demonstrated that the P2Y12 receptor antagonist AR-C69931MX (10 μM) significantly inhibited (p = 0.01 to 0.001) ADP-induced (0.2–0.4 μM) PSC [8]. The inhibition was of the order of 2.9 %, which provides support that this ‘relatively small’ change was detectable because of the sensitivity of the high-resolution (0.07 fl) channelyzer used. This allows lower concentrations of agonists and antagonists, and also has greater sensitivity to detect variations in PSC [3236]. Overall, the study by Jagroop et al. [8] suggested that both the P2Y1 and P2Y12 ADP receptors play a role in controlling PSC in humans [8]. It is possible that Alvarado-Castillo et al. [31] using the less sensitive optical technique to assess PSC could not detect such a relatively small P2Y1-dependent PSC inhibition.

In a noteworthy study, it is reported that PA proved to be a potent inhibitor of epinephrine-induced human platelet aggregation (IC50 20 nM) [30]. These were the first results to show the anti-aggregating effects of PA, suggesting its potential role in cardiovascular therapy. It is also possible that non-ADP mechanisms are involved. On the other hand, it can be suggested that PA could be acting on any ADP released by epinephrine. The mechanisms involved are perhaps complex because PA also acts as a vasorelaxant on norepinephrine-induced contraction in rat aortic rings. This effect was dependent on intact endothelium [37]. Furthermore, Barradas et al. [33] could not demonstrate an increase in median platelet volume in human platelets after adding epinephrine in vitro. Nevertheless, epinephrine enhanced serotonin-induced PSC [38]. Therefore, it may be possible to elucidate the effect of PA on epinephrine-induced platelet activation, assuming that PA does not influence serotonin-induced PSC. Estrada et al. [30] also reported that PA does not affect aggregation induced by several agonists (e.g. platelet-activating factor, collagen, U46619 (a thromboxane A2 analogue) and arachidonic acid). Furthermore, Barradas et al. [33] showed that all these agonists can induce PSC. It is very likely that PSC induced by the latter agonists is enhanced by epinephrine. This would help define the specific mode of action of PA on PSC. Overall, the findings of Jagroop et al. [8] suggest that both the P2Y1 and P2Y12 ADP receptors play a role in controlling PSC in humans. It would be interesting to assess the effect of PA on PSC using a high-resolution channelyzer. Further investigations are needed to understand the molecular mechanisms by which PA affects PSC as well as platelet aggregation.

Brazilin is isolated from the heartwood of Caesalpinia sappan L. (sappan wood, Leguminosae) [39] and for many years has been used as a traditional oriental or folk medicine. Brazilin has been involved in the treatment for sprains, convulsions and diabetic complications, and was used to improve blood circulation. In addition, brazilin was supposed to have antithrombotic activities, and it is interesting to note that sappan wood has been used as a remedy for thrombosis in the oriental traditional medicine [40]. Chang et al. [39] are the first to show the effect of brazilin in human platelet activation. They demonstrated that brazilin (1 to 50 μM) potentiated collagen-induced (0.1 μg/ml) platelet aggregation in washed human platelets, using the turbidimetric method for assessing aggregation [39]. Moreover, higher concentrations of brazilin (20 to 50 μM) directly triggered platelet aggregation in a concentration-dependant manner. They also demonstrated that ATP (50 μM, an ADP antagonist) inhibited platelet aggregation stimulated by ADP (20 μM) more effectively than that stimulated by brazilin (50 μM). These results indicated that brazilin-induced platelet aggregation was not mediated even partially by ADP receptors, but that brazilin acts as a collagen receptor agonist [39]. In contrast, another study reported that brazilin (10–10 M) inhibited thrombin- (0.5 U/ml), collagen- (1 mg/ml), and ADP-induced (2 μM) platelet aggregation in washed rat platelets [40]. The authors suggested that this discrepancy might be due to species-specific characteristics of platelets. If this is the case, then further investigations are needed. Chang et al. [39] stated that the plant-based natural product brazilin at relatively low concentrations (1–10 μM) has the potential to act as therapeutic antithrombotic agent for targeting collagen (0.1 μM) receptors. They also suggested that brazilin may also be a useful tool for the study of detailed mechanisms in collagen receptor-mediated platelet activation. Thus, the literature shows that brazilin which is of natural origin has the capacity to act on various platelet receptors. Undoubtedly, it is also of interest to determine if brazilin does indeed affect the P2 purinergic receptors in human platelets, as it can prospectively lead to the development of new ‘natural’ ADP receptor antagonists.

Mitrephora plants (of the Annonaceae family), located in Borneo, Philippines, north China, the west of India, and the south-east of Australia, have been investigated for their chemical components [41, 42]. In an exciting study, Moharam et al. [41] examined the biological activity of the methanol extract of the twigs of Muraltia vulpina. Phylligenin is one of the five compounds isolated from the twigs of M. vulpina and was shown to inhibit arachidonic acid- (AA, 0.5 mM) and ADP-induced (10 μM) aggregation in human platelets (in vitro) in a dose-dependent manner. Phylligenin demonstrated >60 % inhibition of ADP-induced aggregation, with an IC50 of 121.8 μM [41]. These results are of importance since the actions of endogenous agonists like AA and ADP can induce platelet aggregation in several cardiovascular disease states. Moreover, phylligenin has the potential to act as an antiplatelet agent possibly through purinergic receptors to inhibit platelet activation. The exact mechanism of the action of phylligenin is still unclear, although it has similar effects to aspirin, i.e. it inhibits thromboxane A2 formation [41, 42]. Furthermore, aspirin, a potent cyclooxygenase inhibitor, was used as a positive control for phylligenin and demonstrated a lower IC50 (24.8 μM) [41]. With the common knowledge that aspirin may cause gastric erosions and gastric ulcers, and that patients may suffer with anaemia and gastric haemorrhage, the search for more potent yet safe antiplatelet agents continues. In addition, since phylligenin has demonstrated ADP-induced platelet inhibition, further investigations should be carried out into the effects these oils (isolated from the twigs of M. vulpina) may have on ADP purinergic receptors.

Additional evidence of antiplatelet activity via the ADP purinergic receptors with ‘plant-derived’ extracts can be seen with essential oils of five Goniothalamus species found in tropical Southeast Asia, throughout Indochina and Malaysia [42]. Different parts of these plants are used in traditional medicine to treat conditions such as asthma, rheumatism, fever, malaria, cholera and stomach aches. The in vitro inhibitory effect of various oils from Goniothalamus species was observed for the first time, by Moharam et al. [42]. The bark oil, identified as Gonystylus velutinus (100 μg/ml, distilled from Goniothalamus) showed strong inhibition (51 %) of human platelet aggregation in whole blood, induced by ADP (10 μM, with an IC50 of 87.7 μg/ml). There was also inhibition (50 %) of AA-induced platelet aggregation (with an IC50 of 93.6 μg/ml). Aspirin, which was used as the positive control in the study, was found to be a more potent cyclooxygenase inhibitor, since its IC50 was much lower (4.5 μg/ml or 24.8 μM) than those of the oils investigated [42]. Furthermore, it is suggested that the antiplatelet activity of the Goniothalamus oils may not be due solely to any individual component but could be due to the synergistic effect of a combination of its compounds. This theory is supported by another experimental model (in mice) that shows for the first time that Lavandula hybrida oil (also known as lavender oil) has antiplatelet/antithrombotic properties which could also be due to a synergistic effect of its components [43]. Further studies are required to investigate the overall antiplatelet, synergistic effects caused by the combination of components making up the essential oils of the Goniothalamus species. It should however be noted that the collection of plant material from the Goniothalamus species is fresh from the forests and that its preparation and isolation into essential oils are time consuming and complicated, involving various stages of gas chromatography (GC) and gas chromatography–mass spectrometry (MS) [42]. Thus, it is worth considering that the quality and effectiveness of various batches of essential oils may not be uniform between different research groups and may thus influence results. There is however the ‘Wiley GC–MS Library’ database of some existing plant components which can be accessed by researchers to identify and compare relative retention indices of some isolated oils [42]. The information provided in the literature shows that the Goniothalamus species of trees and shrubs may prove to be a beneficial source for the therapeutic development of ADP receptor antagonists or other antiplatelet agents.

The extract from seeds and bark of the horse chestnut tree, Aesculus hippocastanum L. Hippocastanaceae, has been used as herbal medicine in Europe against chronic venous insufficiency, i.e. varicose veins accompanied with pain, oedema pruritus and a sense of heaviness [44]. Clinical studies have shown that oral horse chestnut extract was as effective as compression stockings in chronic venous insufficiency [45, 46]. The antithrombotic effect of the extract from the horse chestnut tree was first observed more than 40 years ago, but no thorough investigations have since been carried out [47]. Nonetheless, in 2010, Felixsson et al. investigated the pharmacological effects of horse chestnut extract [44]. It was shown that the mechanism of action was via the ADP receptors. That is to say that the extract from the horse chestnut tree (5 μl) inhibited ADP-induced (10 or 10 M) platelet aggregation in human platelet-rich plasma [44]. Moreover, when ketanserin (a 5HT2A receptor antagonist) was added to the horse chestnut extract-treated platelets, aggregation was further reduced. The authors suggest that platelet inhibition by horse chestnut extract appears to be the sum of both inhibitory (due to flavonoids) and stimulatory (due to 5HT2A receptor-mediated) effects [44]. It was clear from this study that horse chestnut extract had inhibitory action on ADP-induced human platelet aggregation. But, this antithrombotic effect also needs to be studied together with possible drug interactions like other antiplatelet drugs, commonly used in vascular disease. Thus, the clinical use of horse chestnut extract as an antiplatelet drug or ADP receptor antagonist remains to be further investigated.

Moderate and prolonged consumption of red wine is associated with a decrease in cardiovascular morbidity and mortality [48]. This may be linked to a lowering of the risk for stroke and sudden death, which have collectively come to be known as ‘The French Paradox’, highlighting the low incidence of coronary heart disease in a country known for its high-fat intake, little exercise and widespread cigarette smoking [49]. This phenomenon could be due to the presence of polyphenolic compounds in red wine. In an interesting study by de Lange et al. [49], the ingredients found in red wine were shown to have an inhibitory effect on ADP-induced (5 μM) platelet aggregation. This finding is consistent with other studies that suggest that wine and its polyphenolics, in particular, could significantly inhibit platelet aggregation and that this could explain, at least in part, the protective effect of red wine against atherosclerosis and coronary heart disease [50, 51]. Red wine contains polyphenols that belong to a complex group of compounds that determine its characteristics and qualities. Flavonoids and non-flavonoids are the two main groups of phenols. In grapes, the polyphenols' function is to scavenge free radicals and thus prevent the fruit from decay [52]. The inhibition of platelet function may provide some explanation for the cardio protective effects associated with red wine. Supporting studies have also shown that the consumption of polyphenolic-rich beverages and the addition of polyphenols to platelets in vitro are associated with the inhibition of platelet aggregation [53, 54]. Moreover, substantial progress in understanding the inhibition of ADP-induced platelet activation, as demonstrated by polyphenols, can play a key role in the development of novel antiplatelet drugs that target P2 receptors [55]. There has been great interest in exploiting cardiovascular purine P2 receptors as therapeutic targets [5658].

There is strong support that cigarette smoking is associated with cardiovascular events like myocardial infarction, peripheral vascular disease, stroke and even sudden death [59]. Hence, following on from The French Paradox, attention is now drawn to the ‘The Asian Paradox’, where, despite the high consumption of tobacco, a low incidence of atherosclerosis and lung cancer per capita is observed, particularly in Asia and Japan [60]. It has been suggested that this may be a result of the high consumption of green tea in Asia. Furthermore, most benefits occurred in regions where approximately 1.2 l of green tea is consumed every day [61]. Green tea is derived from the leaves of the plants Camellia sinensis and contains nearly 4,000 bioactive chemical compounds, one-third of which are polyphenols. The major catechin in green tea is (−)-epigallocatechin gallate (EGCG) and accounts for 10 % of the total weight. It has been shown that green tea catechins (GTC, 0.01–1 mg/ml) inhibited ADP- (20 μM) as well as collagen- (50 μg/ml), thrombin- (0.5 U/ml) and calcium ionophore A23187-induced (1 μM), aggregation of human platelets in vitro [62]. These results suggest that GTC and EGCG have antithrombotic activities and the modes of action may be due to their antiplatelet activities, which may account for the beneficial effect of green tea in cardiovascular disease [60]. Clearly, the role of platelet ADP purinergic receptor inhibition needs to be further examined. Therefore, a rigorous assessment of the effects of GTC on purinergic receptors in well-controlled human trials will be required for a better understanding of the effects of green tea in cardiovascular health [63].

In summary, this article gives a brief overview of some current thienopyridines and other P2Y12 receptor antagonists, highlighting their main disadvantages. Their delayed onset of actions and the increased risk for bleeding among this class of drugs are still of major concern [64]. Hence, these limitations have led to the search for new and improved ADP receptor antagonists. This review outlines a range of plant extracts and their inhibitory effect on ADP-induced platelet activation. The data from these papers indicate that plant extracts may potentially lead to the therapeutic development of ‘new’ ADP receptor antagonists. But, in some cases, the exact mechanisms of action are still unclear. These new plant extracts as drug candidates will have to prove beneficial in comparison to the currently FDA-approved ADP antagonists. Basically, they will have to display faster onset and offset of action, better efficacy and less prolongation of bleeding time. Thus, further investigations and toxicity studies in well controlled human trials are needed.

Conflict of interest

The author reports no conflict of interest.

Conflict of interest

References

  • 1. Gachet CThe platelet P2 receptors as molecular targets for old and new antiplatelet drugs. Pharmacol Ther. 2005;10:180–192. doi: 10.1016/j.pharmthera.2005.03.009.] [[PubMed][Google Scholar]
  • 2. Burnstock GIntroductory overview of purinergic signalling. Front Biosci (Elite Ed) 2011;3:896–900.[PubMed][Google Scholar]
  • 3. Jagroop IA, Kakafika AI, Mikhailidis DPPlatelets and vascular risk: an option for treatment. Curr Pharm Des. 2007;13:1669–1683. doi: 10.2174/138161207780831383.] [[PubMed][Google Scholar]
  • 4. Pang L, Weiss MJ, Poncz MMegakaryocyte biology and related disorders. J Clin Invest. 2005;115:3332–3338. doi: 10.1172/JCI26720.] [[Google Scholar]
  • 5. Hechler B, Cattaneo M, Gachet CThe P2 receptors in platelet function. Semin Thromb Hemost. 2005;31:150–161. doi: 10.1055/s-2005-869520.] [[PubMed][Google Scholar]
  • 6. Cattaneo MThe P2 receptors and congenital platelet function defects. Semin Thromb Hemost. 2005;31:168–173. doi: 10.1055/s-2005-869522.] [[PubMed][Google Scholar]
  • 7. Kunapuli SPP2 receptors and platelet activation. Sci World J. 2002;13:424–433. doi: 10.1100/tsw.2002.106.] [[Google Scholar]
  • 8. Jagroop IA, Burnstock G, Mikhailidis DPBoth the ADP receptors P2Y1 and P2Y12, play a role in controlling shape change in human platelets. Platelets. 2003;14:15–20. doi: 10.1080/0953710021000062914.] [[PubMed][Google Scholar]
  • 9. Mahaut-Smith MP, Jones S, Evans RJThe P2X1 receptor and platelet function. Purinergic Signal. 2011;7:341–356. doi: 10.1007/s11302-011-9224-0.] [[Google Scholar]
  • 10. Jennings LKMechanisms of platelet activation: need for new strategies to protect against platelet-mediated atherothrombosis. Thromb Haemost. 2009;102:248–257.[PubMed][Google Scholar]
  • 11. Xiang YZ, Kang LY, Gao XM, Shang HC, Zhang JH, Zhang BLStrategies for antiplatelet targets and agents. Thromb Res. 2008;123:35–49. doi: 10.1016/j.thromres.2008.05.003.] [[PubMed][Google Scholar]
  • 12. Barn K, Steinhubl SRA brief review of the past and future of platelet P2Y12 antagonist. Coron Artery Dis. 2012;23:368–374. doi: 10.1097/MCA.0b013e3283564930.] [[PubMed][Google Scholar]
  • 13. Cattaneo M, Lecchi AInhibition of the platelet P2Y12 receptor for adenosine diphosphate potentiates the antiplatelet effect of prostacyclin. J Thromb Haemost. 2007;5:577–582. doi: 10.1111/j.1538-7836.2007.02356.x.] [[PubMed][Google Scholar]
  • 14. Savi P, Pereillo JM, Uzabiaga MF, Combalbert J, Picard C, Maffrand JP, Pascal M, Herbert JMIdentification and biological activity of the active metabolite of clopidogrel. Thromb Haemost. 2000;84:891–896.[PubMed][Google Scholar]
  • 15. Bernlochner I, Sibbing DThienopyridines and other ADP-receptor antagonists. Handb Exp Pharmacol. 2012;210:165–198. doi: 10.1007/978-3-642-29423-5_7.] [[PubMed][Google Scholar]
  • 16. Kim L, Charitakis K, Swaminathan RV, Feldman DNNovel antiplatelet therapies. Curr Atheroscler Rep. 2012;14:78–84. doi: 10.1007/s11883-011-0216-x.] [[PubMed][Google Scholar]
  • 17. Panak E, Maffrand JP, Picard-Fraire C, Vallée E, Blanchard J, Roncucci RTiclopidine: a promise for the prevention and treatment of thrombosis and its complications. Haemostasis. 1983;13:1–54.[PubMed][Google Scholar]
  • 18. Packham MA, Mustard JFPlatelet aggregation and adenosine diphosphate/adenosine triphosphate receptors: a historical perspective. Semin Thromb Hemost. 2005;31:129–138. doi: 10.1055/s-2005-869518.] [[PubMed][Google Scholar]
  • 19. Cattaneo M, Podda GMState of the art of new P2Y12 antagonists. Intern Emerg Med. 2010;5:385–391. doi: 10.1007/s11739-010-0363-z.] [[PubMed][Google Scholar]
  • 20. Gresele P, Agnelli GNovel approaches to the treatment of thrombosis. Trends Pharmacol Sci. 2002;23:25–32. doi: 10.1016/S0165-6147(00)01885-X.] [[PubMed][Google Scholar]
  • 21. Wiviott SD, Trenk D, Frelinger AL, O'Donoghue M, Neumann FJ, Michelson AD, Angiolillo DJ, Hod H, Montalescot G, Miller DL, Jakubowski JA, Cairns R, Murphy SA, McCabe CH, Antman EM, Braunwald E, PRINCIPLE-TIMI 44 Investigators Prasugrel compared with high loading- and maintenance-dose clopidogrel in patients with planned percutaneous coronary intervention: the Prasugrel in Comparison to Clopidogrel for Inhibition of Platelet Activation and Aggregation-Thrombolysis in Myocardial Infarction 44 trial. Circulation. 2007;116:2923–2932. doi: 10.1161/CIRCULATIONAHA.107.740324.] [[PubMed]
  • 22. Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, Neumann FJ, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, Weerakkody G, Gibson CM, Antman EM, TRITON-TIMI 38 Investigators Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482.] [[PubMed]
  • 23. Gurbel PA, Bliden KP, Butler K, Antonino MJ, Wei C, Teng R, Rasmussen L, Storey RF, Nielsen T, Eikelboom JW, Sabe-Affaki G, Husted S, Kereiakes DJ, Henderson D, Patel DV, Tantry USResponse to ticagrelor in clopidogrel nonresponders and responders and effect of switching therapies: the RESPOND study. Circulation. 2010;121:1188–1199. doi: 10.1161/CIRCULATIONAHA.109.919456.] [[PubMed][Google Scholar]
  • 24. Storey RF, Angiolillo DJ, Patil SB, Desai B, Ecob R, Husted S, Emanuelsson H, Cannon CP, Becker RC, Wallentin LInhibitory effects of ticagrelor compared with clopidogrel on platelet function in patients with acute coronary syndromes: the PLATO (PLATelet inhibition and patient Outcomes) PLATELET substudy. J Am Coll Cardiol. 2010;56:1456–1462. doi: 10.1016/j.jacc.2010.03.100.] [[PubMed][Google Scholar]
  • 25. Husted S, Emanuelsson H, Heptinstall S, Sandset PM, Wickens M, Peters GPharmacodynamics, pharmacokinetics, and safety of the oral reversible P2Y12 antagonist AZD6140 with aspirin in patients with atherosclerosis: a double-blind comparison to clopidogrel with aspirin. Eur Heart J. 2006;27:1038–1047. doi: 10.1093/eurheartj/ehi754.] [[PubMed][Google Scholar]
  • 26. Norgard NBCangrelor: a novel P2Y12 receptor antagonist. Expert Opin Investig Drugs. 2009;18:1219–1230. doi: 10.1517/13543780903136708.] [[PubMed][Google Scholar]
  • 27. Oestreich JHElinogrel, a reversible P2Y12 receptor antagonist for the treatment of acute coronary syndrome and prevention of secondary thrombotic events. Curr Opin Investig Drugs. 2010;11:340–348.[PubMed][Google Scholar]
  • 28. Patel PA, Lane B, Augoustides JGProgress in platelet blockers: the target is the P2Y12 receptor. J Cardiothorac Vasc Anesth. 2013;27:620–624. doi: 10.1053/j.jvca.2013.01.008.] [[PubMed][Google Scholar]
  • 29. Collet JP, O'Connor SClinical effects and outcomes with new P2Y12 inhibitors in ACS. Fundam Clin Pharmacol. 2012;26:16–18. doi: 10.1111/j.1472-8206.2011.00984.x.] [[PubMed][Google Scholar]
  • 30. Estrada O, Alvarado-Castillo C, Fernandez A, Lopez M, Romero-Vecchione E, Vasquez J, et al Pomolic acid isolated from the leaves of Licania pittieri inhibits ADP- and epinephrine-induced platelet aggregation and has hypotensive effect on rats. Current Bioactive Compounds. 2009;5:219–225. doi: 10.2174/157340709789054786.[PubMed][Google Scholar]
  • 31. Alvarado-Castillo C, Estrada O, Carvajal EPomolic acid (PA), triterpenoid isolated from Licania pittieri, as a competitive antagonist of ADP-induced aggregation of human platelets. Phytomedicine. 2012;19:484–487. doi: 10.1016/j.phymed.2011.12.011.] [[PubMed][Google Scholar]
  • 32. Barradas MA, Jagroop IA, O'Donoghue S, Jeremy JY, Mikhailidis DPEffect of milrinone on human platelet shape change, aggregation and thromboxane A2 synthesis: an in vitro study. Thromb Res. 1993;71:227–236. doi: 10.1016/0049-3848(93)90097-8.] [[PubMed][Google Scholar]
  • 33. Barradas MA, O'Donoghue S, Mikhailidis DPMeasurement of platelet volume using a channelyzer: assessment of the effect of agonists and antagonists. In Vivo. 1992;6:629–634.[PubMed][Google Scholar]
  • 34. Jagroop IA, Clathworthy I, Lewin J, Mikhailidis DPShape change in human platelet: measurements with a channelyzer and visualisation by electron microscopy. Platelets. 2000;11:28–32. doi: 10.1080/09537100075760.] [[PubMed][Google Scholar]
  • 35. Jagroop IA, Mikhailidis DPDoxazosin, an alpha1-adrenoceptor antagonist, inhibits serotonin-induced shape change in human platelets. J Human Hypertens. 2001;15:203–207. doi: 10.1038/sj.jhh.1001146.] [[PubMed][Google Scholar]
  • 36. Jagroop IA, Mikhailidis DPThe effect of tirofiban on fibrinogen/agonist-induced platelet shape change and aggregation. Clin Appl Thromb Hemost. 2008;14:295–302. doi: 10.1177/1076029608316014.] [[PubMed][Google Scholar]
  • 37. Estrada O, González-Guzmán JM, Salazar-Bookaman M, Fernández AZ, Cardozo A, Alvarado-Castillo CPomolic acid of Licania pittieri elicits endothelium-dependent relaxation in rat aortic rings. Phytomedicine. 2011;18:464–469. doi: 10.1016/j.phymed.2010.10.008.] [[PubMed][Google Scholar]
  • 38. Barradas MA (1993) Serotonin and platelet studies in vascular diseaseDoctor of Philosophy Thesis, p 74, University of London
  • 39. Chang Y, Huang SK, Lu WJ, Chung CL, Chen WL, Lu SH, Lin KH, Sheu JRBrazilin isolated from Caesalpinia sappan L. acts as a novel collagen receptor agonist in human platelets. J Biomed Sci. 2013;20:4. doi: 10.1186/1423-0127-20-4.] [[Google Scholar]
  • 40. Hwang GS, Kim JY, Chang TS, Jeon SD, So DS, Moon CKEffects of brazilin on the phospholipase A2 activity and changes of intracellular free calcium concentration in rat platelets. Arch Pharm Res. 1998;21:774–778. doi: 10.1007/BF02976775.] [[PubMed][Google Scholar]
  • 41. Moharam BA, Jantan I, Jalil J, Shaari KInhibitory effects of phylligenin and quebrachitol isolated from Mitrephora vulpina on platelet activating factor receptor binding and platelet aggregation. Molecules. 2010;15:7840–7848. doi: 10.3390/molecules15117840.] [[Google Scholar]
  • 42. Moharam BA, Jantan I, Fb A, Jalil JAntiplatelet aggregation and platelet activating factor (PAF) receptor antagonistic activities of the essential oils of five Goniothalamus species. Molecules. 2010;15:5124–5138. doi: 10.3390/molecules15085124.] [[Google Scholar]
  • 43. Ballabeni V, Tognolini M, Chiavarini M, Impicciatore M, Bruni R, Bianchi A, Barocelli ENovel antiplatelet and antithrombotic activities of essential oil from Lavandula hybrida Reverchon "grosso". Phytomedicine. 2004;11:596–601. doi: 10.1016/j.phymed.2004.01.002.] [[PubMed][Google Scholar]
  • 44. Felixsson E, Persson IA, Eriksson AC, Persson KHorse chestnut extract contracts bovine vessels and affects human platelet aggregation through 5-HT(2A) receptors: an in vitro study. Phytother Res. 2010;24:1297–1301. doi: 10.1002/ptr.3103.] [[PubMed][Google Scholar]
  • 45. Tiffany N, Boon H, Ulbricht C, Basch E, Bent S, Barrette EP, Smith M, Sollars D, Dennehy CE, Szapary PHorse chestnut: a multidisciplinary clinical review. J Herb Pharmacother. 2002;2:71–85.[PubMed][Google Scholar]
  • 46. Siebert U, Brach M, Sroczynski G, Berla K. Efficacy, routine effectiveness, and safety of horse chestnut seed extract in the treatment of chronic venous insufficiency. A meta-analysis of randomized controlled trials and large observational studies. Int Angiol. 2002;21:305–315.[PubMed]
  • 47. Kronberger L, Gölles JOn the prevention of thrombosis with Aesculus extract. Med Klin. 1969;64:1207–1209.[PubMed][Google Scholar]
  • 48. Marmot MAetiology of coronary heart disease. BMJ. 2001;323:1261–1262. doi: 10.1136/bmj.323.7324.1261.] [[Google Scholar]
  • 49. de Lange DW, Verhoef S, Gorter G, Kraaijenhagen RJ, van de Wiel A, Akkerman JWPolyphenolic grape extract inhibits platelet activation through PECAM-1: an explanation for the French paradox. Alcohol Clin Exp Res. 2007;31:1308–1314. doi: 10.1111/j.1530-0277.2007.00439.x.] [[PubMed][Google Scholar]
  • 50. Ruf JCWine and polyphenols related to platelet aggregation and atherothrombosis. Drugs Exp Clin Res. 1999;25:125–131.[PubMed][Google Scholar]
  • 51. Pace-Asciak CR, Hahn S, Diamandis EP, Soleas G, Goldberg DMThe red wine phenolics trans-resveratrol and quercetin block human platelet aggregation and eicosanoid synthesis: implications for protection against coronary heart disease. Clin Chim Acta. 1995;235:207–219. doi: 10.1016/0009-8981(95)06045-1.] [[PubMed][Google Scholar]
  • 52. Soleas GJ, Diamandis EP, Goldberg DMWine as a biological fluid: history, production, and role in disease prevention. J Clin Lab Anal. 1997;11:287–313. doi: 10.1002/(SICI)1098-2825(1997)11:5<287::AID-JCLA6>3.0.CO;2-4.] [[PubMed][Google Scholar]
  • 53. Freedman JE, Parker C, 3rd, Li L, Perlman JA, Frei B, Ivanov V, Deak LR, Iafrati MD, Folts JDSelect flavonoids and whole juice from purple grapes inhibit platelet function and enhance nitric oxide release. Circulation. 2001;103:2792–2798. doi: 10.1161/01.CIR.103.23.2792.] [[PubMed][Google Scholar]
  • 54. Pignatelli P, Lenti L, Pulcinelli FM, Catasca R, Saccani G, Germanò G, Marcoccia A, Silvestri MA, Ghiselli A, Violi FRed and white wine differently affect collagen-induced platelet aggregation. Pathophysiol Haemost Thromb. 2002;32:356–358. doi: 10.1159/000073599.] [[PubMed][Google Scholar]
  • 55. Clutton P, Folts JD, Freedman JEPharmacological control of platelet function. Pharmacol Res. 2001;44:255–264. doi: 10.1006/phrs.2001.0861.] [[PubMed][Google Scholar]
  • 56. Ralevic V, Burnstock GInvolvement of purinergic signaling in cardiovascular diseases. Drug News Perspect. 2003;16:133–140. doi: 10.1358/dnp.2003.16.3.876886.] [[PubMed][Google Scholar]
  • 57. Jagroop IA, Matsagas MI, Geroulakos G, Mikhailidis DPThe effect of clopidogrel, aspirin and both antiplatelet drugs on platelet function in patients with peripheral arterial disease. Platelets. 2004;15:117–125. doi: 10.1080/09537105310001645960.] [[PubMed][Google Scholar]
  • 58. Erlinge D, Burnstock GP2 receptors in cardiovascular physiology and disease. Purinergic Signalling. 2008;4:1–20. doi: 10.1007/s11302-007-9078-7.] [[Google Scholar]
  • 59. Yun AJ, Bazar KA, Lee PY, Gerber A, Daniel SMThe smoking gun: many conditions associated with tobacco exposure may be attributable to paradoxical compensatory autonomic responses to nicotine. Med Hypotheses. 2005;64:1073–1079. doi: 10.1016/j.mehy.2004.11.040.] [[PubMed][Google Scholar]
  • 60. Sumpio BE, Cordova AC, Berke-Schlessel DW, Qin F, Chen QHGreen tea, the "Asian paradox," and cardiovascular disease. J Am Coll Surg. 2006;202:813–825. doi: 10.1016/j.jamcollsurg.2006.01.018.] [[PubMed][Google Scholar]
  • 61. Imai K, Nakachi KCross sectional study of effects of drinking green tea on cardiovascular and liver diseases. BMJ. 1995;310:693–696. doi: 10.1136/bmj.310.6981.693.] [[Google Scholar]
  • 62. Kang WS, Lim IH, Yuk DY, Chung KH, Park JB, Yoo HS, Yun YPAntithrombotic activities of green tea catechins and (−)-epigallocatechin gallate. Thromb Res. 1999;96:229–337. doi: 10.1016/S0049-3848(99)00104-8.] [[PubMed][Google Scholar]
  • 63. Islam MACardiovascular effects of green tea catechins: progress and promise. Recent Pat Cardiovasc Drug Discov. 2012;7:88–99. doi: 10.2174/157489012801227292.] [[PubMed][Google Scholar]
  • 64. Cattaneo MPlatelet P2 receptors: old and new targets for antithrombotic drugs. Expert Rev Cardiovasc Ther. 2007;5:45–55. doi: 10.1586/14779072.5.1.45.] [[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.