Next Article in Journal
Efficacy of BREATHOX® Device Inhalation on Acute Symptoms Associated with COVID-19 (BREATH Study): A Randomized Pilot Clinical Trial
Next Article in Special Issue
Relationships, Current Issues, Safety and Efficacy of Oral Anticoagulation in Cancer Patients with Atrial Fibrillation
Previous Article in Journal
Comment on Lunz et al. Impact and Modification of the New PJI-TNM Classification for Periprosthetic Joint Infections. J. Clin. Med. 2023, 12, 1262
Previous Article in Special Issue
ASA Allergy and Desensitization Protocols in the Management of CAD: A Review of Literature
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention

1
Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy
2
Third Medicine Division, Department of Medicine, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
3
Division of Cardiology, Ospedale degli Infermi, ASL Biella, 13875 Biella, Italy
4
Department of Translational Medicine, Eastern Piedmont University, 28100 Novara, Italy
5
Department of Cardiology, Humanitas Gavazzeni Hospital, 24125 Bergamo, Italy
6
U.O. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
7
Department of Internal Medicine and Medical Sciences, School of Medicine, University of Genova, 16126 Genova, Italy
8
Division of Cardiology, Hopital Erasmus, Universitè Libre de Bruxelles, 1050 Bruxelles, Belgium
9
Division of Cardiology, Ospedale Ruggi D’Aragona, Università di Salerno, 84084 Salerno, Italy
10
Department of Cardiology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
11
Cardiovascular Research Institute Maastricht (CARIM), 6229 ER Maastricht, The Netherlands
12
Department of Cardiology, Zuyderland Medical Center, 6419 PC Heerlen, The Netherlands
13
Department of Cardiovascular Medicine, IRCCS-Humanitas Research Hospital, 20089 Rozzano, Italy
14
Division of Cardiology, AOU “Policlinico G. Martino”, Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
15
Division of Cardiology, IRCCS Hospital Galeazzi-Sant’Ambrogio, 20157 Milan, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(18), 6074; https://doi.org/10.3390/jcm12186074
Submission received: 22 June 2023 / Revised: 7 September 2023 / Accepted: 8 September 2023 / Published: 20 September 2023

Abstract

:
The key role played by platelets in the atherosclerosis physiopathology, especially in the acute setting, is ascertained: they are the main actors during thrombus formation and, thus, one of the major investigated elements related to atherothrombotic process involving coronary arteries. Platelets have been studied from different points of view, according with the technology advances and the improvement in the hemostasis knowledge achieved in the last years. Morphology and reactivity constitute the first aspects investigated related to platelets with a significant body of evidence published linking a number of their values and markers to coronary artery disease and cardiovascular events. Recently, the impact of genetics on platelet activation has been explored with promising findings as additional instrument for patient risk stratification; however, this deserves further confirmations. Moreover, the interplay between immune system and platelets has been partially elucidated in the last years, providing intriguing elements that will be basic components for future research to better understand platelet regulation and improve cardiovascular outcome of patients.

Graphical Abstract

1. Introduction

Platelets enter the circulation after the separation of bone marrow megakaryocytes, as anucleate cells (2 μm in diameter) that lack genomic DNA [1]. Therefore, due to the absence of a nucleus and in the presence of little capacity for protein synthesis, which is only available by messenger RNA (mRNA) transferred from the megakaryocyte, the lifespan of platelets is limited to 7–10 days [2,3]; however, platelets contain mitochondria that have also been transferred from megakaryocyte origin, and therefore, like nucleated cells, they can still benefit from flexible aerobic and anaerobic metabolism in the presence of necrotic and apoptotic pathways that regulate their function and fate [4]. Thrombopoietin is a glycoprotein synthesized mostly in the liver but also in the kidneys, which regulates megakaryocytic proliferation and maturation, as well as platelet production [5]. Once they enter the circulation, platelets have a life span of 7 to 10 days in quiescent discoid state in the presence of healthy vessels without endothelium-impaired homeostasis. Platelets’ primary function is to stop hemorrhage after tissue trauma and vascular damage.
Injury to the intimal layer of the vessel exposes the underlying subendothelial matrix. Platelets move to sites of vascular disruption and adhere to the exposed matrix protein [6]. Adherent platelets undergo activation and subsequently release substances that recruit additional platelets to the site of injury. Additionally, they promote thrombin generation and subsequent fibrin formation. The potent platelet agonist thrombin, together with other amplifying factors, promotes platelet recruitment and activation. Activated platelets then aggregate to form a plug that seals the injury in the vasculature. The understanding of this highly integrated processes is essential to optimize the treatment in patients suffering from atherothrombotic disease [7,8,9]; therefore, the identification of makers of platelet regulation, whether inhibiting or promoting, has deserved increasing attention in the last decades.

2. Platelet Adhesion, Activation, and Aggregation

The first step of platelets at sites of vascular injury consists in adhesion, which is mediated by the glycoprotein complexes that allow platelets adhering to exposed factors and collagen. In particular, the glycoprotein (GP) Ib/V/IX mediates the initial capturing of free-flowing platelets binding the immobilized von Willebrand factor (vWF). The induced signals promote platelet main integrin activation, αIIbβ3, which binds fibrinogen at their ligation, which in turn induces a cascade of potent outside signals that significantly increase the cytosolic calcium influx [3]. Mouse models knock-out for the GP VI gene have shown platelets unable to form aggregate and occlusive arterial thrombi [10], and the progressive shedding of GP VI can affect the functional platelets properties of adhesion to the site of injury [11].
The vWF is synthesized by endothelial cells and megakaryocytes assembles into multimers with a size ranging from 550 kDa to more than 10,000 kDa [12]; the prevention of large multimer accumulation is mediated by the metalloprotease ADAMTS13, whose deficiency results in the thrombotic thrombocytopenia purpura.
When released from the α-granules of platelets or the Weibel–Palade bodies of endothelial cells, the majority of the vWF compounds enters the circulation, except for the vWF released from the abluminal surface of the endothelial cells: it accumulates in the subendothelial matrix and binds collagen via its A3 domain. The vWF anchored to the vessel subendothelial matrix changes its conformation, exposing the A1 domain that binds platelets.
The adhesion to collagen and vWF endorses the pathways that lead to platelet activation. The induced signals promote the integrin activation—firstly α2β1, which binds vWF, and then αIIbβ3, which binds fibrinogen. The integrin ligation produces, in turn, a cascade of signals increasing the cytosolic calcium influx [3]. During this process, platelets modify their morphology and stimulate the release of their granule-content-rich soluble agonists, including adenosine diphosphate (ADP), adhesion molecules, and coagulation factors, almost all of which amplify the thrombotic response [13]. Among these autocrine and paracrine mediators, platelet activation favors thromboxane A2 (TXA2) generation through cyclooxygenase1. The next step involves the conversion of prothrombin into thrombin through activated factor X [14]. Thrombin is the most potent known agonist for platelet activation, binding protease-activated receptor types 1 and 4 (PAR-1 and PAR-4), but it also plays a critical role in early thrombus formation, converting fibrinogen to fibrin, which effectively anchors the growing thrombus.
The final step in the formation of the platelet plug is represented by the aggregation that links platelets to each other to form clumps. Linkages are mediated by GP IIb/IIIa that has undergone conformational change after platelet activation to increase the affinity for its ligand, fibrinogen (Figure 1) [15].

3. Interplay Immune System

The immune system has a central key role in the atherothrombotic process [16,17]. Platelets have been involved in modulation of innate and adaptive immune responses, even if most mechanisms are still an enigma (Figure 2). Platelets α-granules contain many molecules with several effects not limited to platelet aggregation amplification and coagulation: a couple of molecules with a key role in several signaling pathways are CD40 and CD154, also known as CD40 ligand (CD40L) [18]. In particular, CD154 can interact both with endothelial and immune cells. The stimulation of endothelial cells leads to increased expression of adhesion molecules, mainly intercellular (I-CAM-1) and vascular (V-CAM-1) adhesion molecules [19], and consequent increased leukocytes recruitment and atherosclerotic plaque instability. Platelet-derived CD154 interacts with different type of immune cells: it has been described stimulating the B cell differentiation [20] and macrophages activity [21]. In a murine model of atherosclerosis, CD154 has been shown to increase plaque fragility and reduce its stability by inhibition of regulatory T cells [22]. Platelets have been found expressing different toll-like receptor (TLR) subtypes, enforcing the concept of inflammation modulatory action by platelets [23]. Among cytokines released by platelets, transforming growth factor-β1 (TGF-β1) is one of the most important and studied. It has been characterized with both anti-atherogenic and pro-atherogenic effects, given its number of potential targets. Briefly TGF-β1 may prevent the formation and progression of endothelial lipidic deposit by increasing the nitric oxide synthetase activity and polarization of macrophage into anti-inflammatory phenotypes [24], with consequent enhanced plaque stability [25]. The TGF-β1 pro-atherogenic effect has been elucidated in studies reporting its promotion of interstitial collagen deposition, fibrosis, and blockade of endothelial regeneration, with consequent growth of lesion and reduction in vascular lumen [26].
Moreover, platelets have been described to have the opportunity to recruit neutrophils and worse thrombo-inflammation [27].
In fact, leukocytes’ interaction with the endothelium is mediated by the different types of surface-expressed selectins and their specific ligands. Among selectins, E-selectin expression mainly occurs in presence of endothelial dysfunction [28] while P-selectin is expressed on both endothelial cells and platelets upon activation [29]. A damaged endothelium is rapidly covered by activated platelets which in turn capture circulating neutrophils into thrombus: in murine models, knock-out for P-selectin showed marked lower or absent leukocyte recruitment [30]. The main crosstalk element is constituted by the P-selectin that binds P-selectin glycoprotein ligand-1 expressed by neutrophils [31], but also chemokines CXCL4 (also named platelet factor-4, PF-4) and CXCL7 (also named neutrophil activating peptide, NAP-2) [32], and high mobility group box 1 (HMGB1) are released by platelets and contribute to neutrophil chemotaxis [33]. Neutrophil β2 integrin binds PF-4, while NAP-2 attracts and stimulates neutrophils through the engagement of CXCR1 and CXCR2 [34].
A further step of neutrophil–platelet interactions is the promotion of neutrophil extracellular trap (NET) [35,36]. This structure consists of extracellular chromatin supported by the histones. Activated platelets have been demonstrated inducing a rapid release of NET, not only using the P-selectin-dependent interplay, but also the TLR4 pathways, like in septic condition [37]. Moreover, NET itself may induce the thrombus formation, serving as a scaffold for both platelet binding and activation [38]. Several pathways have been suggested, including H3 histones and C3b attached to NET [39]: in murine models, the infusion of histone preparate was related to accelerated thrombus formation and vWF release [40]. Formed NET can reduce the endothelial integrity, which in turn determines platelet recruitment and activation and coagulation through factor XII, as well as NET physical interaction with activated fibrinogen providing stability to the thrombus [41,42].
From the clinical side, Hally et al. have investigated the potential utility of a composite biomarker score of NET activation and release, or NETosis, for predicting major cardiovascular adverse event (MACE) post-myocardial infarction (MI): authors demonstrated the importance of these combining biomarkers as risk predictors of MACE at 1 year [43]. Furthermore, Riegger et al. evaluated about two hundred and fifty thrombus specimens in patients with stent thrombosis, demonstrating the recruitment leukocytes, particularly neutrophils and the presence of NETosis in 23% of samples [44]. In the thrombus of culprit artery from patients with acute MI, the presence of NET formation has also been demonstrated [45]. Promising therapeutics have been investigated, inclacumab and crizanlizumab, that block interaction of P-selectin and neutrophils. In the SELECT-ACS study, inclacumab was reported to significantly reduce myocardial damage assessed through the CK-MB after percutaneous coronary intervention (PCI) in patients with non-ST segment elevation acute coronary syndrome (ACS) [46,47]. However, these findings were not confirmed in patients suffering of bypass graft failure after coronary artery bypass surgery [48]. Therefore, further studies are needed to better assess the potential of monoclonal antibodies therapy against NET in patients with coronary artery disease (CAD) [49].

4. Morphologic and Structural

Among the first proposed parameters linked to platelet aggregation, there is the mean platelet volume (MPV). MPV is a marker of platelet size, and it is usually provided by the majority of laboratories as a part of the full blood count [14]. It constitutes one of the most used indirect markers of platelet function. In fact, it has been shown that larger platelets are more active, from a metabolic point of view, thus leading to greater prothrombotic risk [50,51]. Early released platelets display higher MPV, while aging decreases platelet volume due to microvesiculation of fully activated platelets or apoptotic membrane fragmentation [52].
In their denser granules, larger platelets contain more β-thromboglobulin, serotonin, and TXA2 than smaller ones [53,54,55]. Moreover, it has been argued that MPV may reflect the platelet production rate [56], stimulation [57], and activation [58,59,60]. In particular, larger platelets are more adhesive and reactive [61,62], showing higher expression of glycoprotein Ib and GP IIb/IIIa receptors at the surface [63]. For these reasons, MPV has been variously associated to CAD, although with contrasting results [64]. In about 400 patients with ST segment elevation MI undergoing primary PCI, Huczek et al. found MPV to be a strong, independent predictor of impaired angiographic reperfusion and six-month mortality [65]. Similarly, Murat and colleagues showed MPV as an independent predictor of the severity of CAD among ACS patients [66].
However, the direct relationship between MPV and CAD has not been validated by other investigations [67,68]. Halbmayer et al., in fact, found no differences in MPV between healthy persons and patients with CAD as well as no significant variations of MPV values being reported between patients without prior MI and MI survivors [69]. In a large cohort of more than 1400 patients undergoing PCI, no relationship was found between MPV and the extent of CAD [70], neither with platelet aggregation [71].
Whether MPV might represent an independent predictor of cardiovascular (CV) events or on the contrary the consequence of other CV risk factors, such as diabetes, hypertension, or smoking is still debated, even if a recent meta-analysis suggests that MPV could be a useful prognostic marker in patients with CAD [72].
Another morphologic parameter of platelets that has been inquired upon for a potential relationship with CAD is platelet distribution width (PDW): it directly measures the variability in platelet size and has been used to differentiate primary disorders of platelets such as essential thrombocythemia from reactive thrombocytosis [73], therefore providing more information than MPV. Some studies have described a potential relationship between PDW and chronic total occlusion of CAD [74] and occluded saphenous vein graft in patients with coronary artery bypass graft surgery [75]. Opposite results were reported regarding the role of PDW in predicting CAD and its severity, suggesting the need of further investigations to clarify its contribution in patient management [76,77].
In addition, the platelet-large cell ratio (P-LCR), an index representing the percentage of platelets larger than 12 fL, has deserved attention as a marker of platelet activation [78]. Despite some promising results showing a relationship between P-LCR and inflammation in CAD patients [79,80], no significant contribution regarding the severity of CAD and platelet reactivity has been found for P-LCR [81,82].
Growing interest has been denoted about the fraction of reticulated platelets, which represent the proportion of younger platelets released from the bone marrow last, with a higher content in α-granules and RNA: these features have been hypothesized as leading to enhanced capability of proteins synthesis and then to a potentially increased overall reactivity [83,84]. McBane et al. reported that younger reticulated platelets appear to have a greater propensity for thrombus participation in presence of atherosclerotic stenosis and shear conditions compared to older ones. Among the suggested mechanisms, an increased receptor density of integrin-β3 in younger platelets may contribute to higher predisposition to thrombosis [85].
Immature platelet fraction (IPF) and the absolute immature platelet count (IPC) represent parameters for the identification of the reticulated platelet with a sample for a routine blood count [86]. A certain number of studies has reported higher IPF in patients with CAD, especially in the subgroups admitted for an ACS [87,88], with a potential role of IPF > 6.2 to predict mortality [87]. On the other hand, Berny-Lan et al. found no difference of IPF in patients with and without ACS [89] and Verdoia et al. showed no role of IPF to predict CAD and the severity of CAD in patients undergoing PCI, discussing IPF as a marker of platelet turnover rather than being directly involved in the pathogenesis of CAD. In patients on antiplatelet drugs, IPF has been linked with ineffective therapy, both mono- and dual-antiplatelet therapy [90,91], despite opposite evidences having also been provided [92,93].

5. Function and Reactivity

A direct assessment of platelet activation and aggregation could be performed though a platelet function test (PFT), of which the most used are displayed in Table 1. The identification of impairment in clot formation for hemorrhagic disorders was the rationale of the first test described by Duke, the evaluation of bleeding time [94]. To better assess congenital and acquired platelet disorders, the first platelet aggregation test was developed, adopting the light transmission aggregometry (LTA) with platelet-rich plasma (PRP) [95,96]. After them, several tests with different methods have been proposed to estimate platelet function. Apart from LTA, which requires elaborate management of blood samples before proceeding, impedance whole aggregometry allows to assess platelet aggregation using anticoagulated whole blood. It allows to measure platelet aggregation after several stimuli inquiring different activation signaling pathways with two main advantages: the small quantity of whole blood required and no manipulation before testing, preserving the physiological condition as much as possible [97]. Another aggregometry method uses a turbidimetric-based optical detection through a system cartridge containing fibrinogen-coated beads and platelet agonists [98]; specific assays for patients under antiplatelet therapy are commercially available [99]. A certain number of commercial assays are instead based on the evaluation of platelet adhesion under shear stress, or on viscoelastic methodologies [100,101,102,103].
Besides the different technologies used, the rationale of assessment of platelet function in patients with CAD is used for monitoring the response to antiplatelet drugs in order to identify subjects deserving modification in the composition and/or treatment duration [104]. The main reason of assessment is to recognize patients that are poor responders to antiplatelet therapy, both aspirin and P2Y12 inhibitors. Since these drugs represents the cornerstone in patients with CAD, undergoing PCI, their effectiveness is crucial to prevent adverse events [105]. However, the adoption of the expression “drug resistance” is not appropriate, considering that the baseline assessment of platelet function is unavailable in the vast majority of cases, especially in the acute setting. Therefore, it is recognized to refer to inappropriate platelet reactivity during antiplatelet therapy with the expression of “high residual platelet reactivity” (HRPR) or “high on-treatment platelet reactivity” (HTPR) [106]. Even if HRPR is a well-known independent predictor of several adverse events [107], strategies to tailor antiplatelet therapy according to platelet function provided contrasting results in demonstrating clinical benefits [104,108,109,110], suggesting other factors involved in the determinism of elevated platelet reactivity on top of antiplatelet treatment (Table 2).

6. Platelet Reactivity

The detection of patients displaying enhanced platelet reactivity despite antiplatelet therapy and deserving more clinical attention and stricter follow-up could improve the overall outcome of those subjects at higher risk for CV adverse events.
One of the first described elements affecting atherosclerosis and platelets is cigarette smoking. The impact on the CV system is recognized, while less detailed are the smoking mechanisms involved in the promotion of platelet reactivity. The increased smoking-induced lipid oxidation products bind platelet scavenger receptor CD36, which in turn results in increased platelet aggregation response [124]. Amplifying factors, including ADP and thrombin, have been shown to increase after cigarette smoking exposure, with consequent higher platelet aggregation response to these pathways [125,126], and nicotine displayed a direct role in increasing platelet aggregability [127]. However, some recent studies have found an opposite effect of smoking, especially on P2Y12 antagonists, whose platelet inhibition is enhanced after smoking exposure [128], despite concerns regarding basal lower platelet aggregation [129] and smoking effect on platelet morphology [130].
Higher platelet reactivity has been reported among diabetic patients, due to continuous environmental inflammation that characterizes diabetes mellitus. However, diabetes is not only per se a marker of higher risk [131], but the impaired glycemic control has also been linked to HRPR among clopidogrel-treated subjects [132] or under more potent P2Y12 inhibitors [133]. Conversely, Vivas et al. documented a significant reduction in aggregation in post-ACS patients receiving intensive glucose control treatment with insulin [134]. Indeed, hyperglycemia can impact platelet function both directly and by modulating the release of pro-oxidant and inflammatory substances or by the glycation platelet surface proteins, with consequent amplification of platelet adhesion [134].
Similar pathophysiological elements are shared by the excess of uric acid: it has been addressed as a main determinant of atherosclerosis and metabolic syndrome. In fact, hyperuricemia is a condition characterized by impaired nitric oxygen release and enhanced pro-inflammatory cytokines [135,136]. However, no direct impact has been demonstrated on platelet reactivity under antiplatelet therapy [137].
Increasing interest has grown regarding the potential CV impact of vitamin D [138]. Alongside the impact on endothelial dysfunction [139], a vitamin D receptor was detected on platelet surface, displaying antithrombotic effects [140]. Pivotal role in the platelet aggregation and thrombus formation has been described by Aihara et al., who demonstrated in murine models knock-out for the vitamin D receptor gene an enhanced thrombogenicity [141]. An indirect antiplatelet therapy has been previously reported by Lòpez-Farré and colleagues: the addition of vitamin D binding protein to whole blood in healthy subjects hampered the antiplatelet inhibitory effect of aspirin [142], and lower levels of vitamin D have been significantly associated to HRPR under ticagrelor treatment [143]. Genetics of vitamin-D-related genes constitutes an interesting ongoing field of investigation [144], despite more studies being needed [145].

7. Genetics and miRNAs

In relation to the antiplatelet therapy with P2Y12 inhibitors, several reports have addressed concerns on reduced effectiveness and platelet inhibition, especially during clopidogrel treatment. Because clopidogrel is a pro-drug, it first needs a metabolic modification by CYP2C19 before it becomes effective. However, about 5% of Caucasians and 15 to 20% of Asians display low or absent CYP2C19 activity, leading to a smaller or no clopidogrel effect on platelet function [146]. Several studies have reported higher number of CV adverse events in subjects who carry at least one non-functional copy of the CYP2C19 gene compared with patients with wild-type CYP2C19 gene [147,148], portraying these patients to be treated with higher clopidogrel dosage or with an alternative drug [149,150].
Guidelines to describe scenarios deserving modification in antiplatelet therapy have been proposed by the Clinical Pharmacogenetics Implementation Consortium (CPIC) and by Dutch Pharmacogenetics Working Group (DPWG) of the Royal Dutch Association for the Advancement of Pharmacy [151,152]. However, the recent TAILOR-PCI trial showed that in CYP2C19 loss of function carriers with ACS and stable CAD undergoing PCI, genotype-guided selection of an oral P2Y12 inhibitor, compared with conventional clopidogrel therapy without genotyping, resulted in non-statistically significant difference with respect to a composite end point of CV death, MI, stroke, stent thrombosis, and severe recurrent ischemia at 1 year [153], while a meta-analysis suggests the potential usefulness of genetic testing to guide de-escalation of antiplatelet therapy after a PCI for ACS [154].
In addition, more potent and expensive P2Y12 inhibitors were inquired for a potential impact of genetics on their effects: prasugrel, a pro-drug like clopidogrel, was found being influenced by some cytochrome P450 polymorphisms [155], even if the Food and Drug Administration stated that there were no relevant effects of genetic variation on pharmacokinetics of prasugrel’s active metabolite and, thus, on its antiplatelet effect [155]. On the other hand, ticagrelor, that is direct P2Y12 inhibitor not requiring activation, has no specific warning about potential genetic impact: Varenhorst et al. found some genetic loci influencing ticagrelor pharmacokinetics through a genome-wide association study, but that did not translate into any clinical detectable effect on ticagrelor efficacy and safety [156], even if the genetics of the adenosine signaling pathways may affect platelet reactivity under ticagrelor treatment (Figure 3) [157].
In the last years, increasing interest has been dedicated to RNA carried by platelets, specifically with regards to microRNAs (miRNAs). Considering that platelets are enriched with miRNAs and represent the second most abundant blood cell type, platelets are the major source of miRNAs in plasma and serum [158,159]. Moreover, the levels of both intraplatelet and circulating platelet-derived miRNAs are shown to correlate with platelet reactivity [160,161].
Of about 750 intraplatelet miRNAs identified, the most extensively investigated are miR-223, miR-21, and miR-126. With respect to the overall amount of platelets, miR-223 is the most represented, and it has been linked to P2Y12 receptor regulation, despite the exact mechanism needing to be clarified [162]. However, some suggestions come from murine model without miR-223 expression, which displayed increased thrombus size compared to normal miR-223 expression [163]; among diabetic patients, a lower level of miR-223 was detected with a concomitant increased platelet reactivity [164]. On the other hand, miR-21 together with other miRNAs were found upregulated in ACS patient with an enhanced response to clopidogrel [165]; however, this deserves further study to define its role in regulation of α-granules and platelet-derived pro-fibrotic factors, including TGF-β1 [166]. The role of miR-126 has been shown involving the regulation of P2Y12 receptor and, potentially, platelet-derived thrombin generation [167,168].
The role of circulating miRNAs in platelet activation is still under investigation, even if a progressive quantity of evidence has been published, underscoring their role in aggregation homeostasis [169,170]. Major shadows are related to the general mechanisms used by circulating miRNAs to act on their target, especially considering the abundance of RNases in the circulation that quickly degrade free RNA [171]: it has been proposed that circulating miRNAs might regulate the cell surface receptor through physical conformational interaction, but an adversative issue is that miRNAs are contained in vesicles or protein complexes to protect them by degradation [172]. Jansen et al. have described in 181 patients with stable CAD that miRNAs contained in microvesicles but not circulating miRNAs predict the occurrence of CV events [173]. In addition, Zampetaki et al. described the association of miRNAs expression patterns and the incidence of MI in a cohort of 820 patients [174], while other miRNAs levels were significantly associated with the risk of death in univariate and age- and sex-adjusted analyses in patients with ACS [175]. Further investigations will provide us a more detailed understanding of miRNAs pathways to design accurate studies for the definition of their functions in platelet aggregation and atherosclerosis progression.

8. Future Perspective and Conclusions

This overview aimed to show the different approaches and methods adopted to study platelet aggregation that appropriately warranted the attention of scientific research: not only because they are an essential component of the physiological hemostasis and pathological atherothrombotic process, but also since interventions on them have demonstrated dramatic improvements in patients with CAD, including both stable disease and acute presentation. Antiplatelet therapy represents a mandatory approach for subjects with clinically significant atherosclerosis, but the availability of effective parameters to assess platelet function is indispensable to select the best option for each patient in the era of a tailored therapeutic approach [176,177,178]. From the first instrument developed and of limited routinary usage, several morphological parameters and methods to assess platelet reactivity have shown significant contribution to daily clinical practice. Auspicious findings are from genetic investigations, in particular miRNAs, as well as from the progressively better understanding of the role of immune system cells in platelet thrombus formation. A single, definite biomarker of platelet aggregation would probably be chimeric, whilst the merging of different aspect assessments, including the most recent advances, will help—if not to close, then at least to move near the circle on a comprehensive valuation of the CV risk for patients with CAD.

Author Contributions

Conceptualization, methodology, G.D.L.; writing—original draft preparation, M.N. and M.V.; writing—review and editing, S.N., D.C., E.K. and G.G.; supervision, A.W.J.v.‘t.H., G.C. and G.D.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Ethical review and approval were waived for this study because this is a review.

Informed Consent Statement

Patient consent was waived because this is a review.

Data Availability Statement

Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACS = acute coronary syndrome; ASA = acetylsalicylic acid; CV = cardiovascular; GP IIb/IIIa = glycoprotein IIb/IIIa; LTA = light transmission platelet aggregation; MACCE = major adverse cardiovascular and cerebrovascular event; MACE = major adverse cardiovascular event; MI = myocardial infarction; PCI = percutaneous coronary intervention; PFT = platelet function test; TEG = thromboelastography; VASP = vasodilator stimulated phosphoprotein.

References

  1. Italiano, J.E.; Shivdasani, R.A. Megakaryocytes and beyond: The Birth of Platelets. J. Thromb. Haemost. 2003, 1, 1174–1182. [Google Scholar] [CrossRef]
  2. Healy, A.M.; Pickard, M.D.; Pradhan, A.D.; Wang, Y.; Chen, Z.; Croce, K.; Sakuma, M.; Shi, C.; Zago, A.C.; Garasic, J.; et al. Platelet Expression Profiling and Clinical Validation of Myeloid-Related Protein-14 as a Novel Determinant of Cardiovascular Events. Circulation 2006, 113, 2278–2284. [Google Scholar] [CrossRef]
  3. Hosseini, E.; Mohtashami, M.; Ghasemzadeh, M. Down-Regulation of Platelet Adhesion Receptors Is a Controlling Mechanism of Thrombosis, While Also Affecting Post-Transfusion Efficacy of Stored Platelets. Thromb. J. 2019, 17, 20. [Google Scholar] [CrossRef]
  4. Hosseini, E.; Nodeh, F.K.; Ghasemzadeh, M. Gamma Irradiation Induces a Pro-Apoptotic State in Longer Stored Platelets, without Progressing to an Overt Apoptosis by Day 7 of Storage. Apoptosis 2023, 28, 1141–1153. [Google Scholar] [CrossRef]
  5. Baigger, A.; Blasczyk, R.; Figueiredo, C. Towards the Manufacture of Megakaryocytes and Platelets for Clinical Application. Transfus. Med. Hemother. 2017, 44, 165–173. [Google Scholar] [CrossRef]
  6. Becker, R.C.; Sexton, T.; Smyth, S.S. Translational Implications of Platelets as Vascular First Responders. Circ. Res. 2018, 122, 506–522. [Google Scholar] [CrossRef]
  7. De Luca, G.; Suryapranata, H.; Stone, G.W.; Antoniucci, D.; Tcheng, J.E.; Neumann, F.-J.; Bonizzoni, E.; Topol, E.J.; Chiariello, M. Relationship between Patient’s Risk Profile and Benefits in Mortality from Adjunctive Abciximab to Mechanical Revascularization for ST-Segment Elevation Myocardial Infarction: A Meta-Regression Analysis of Randomized Trials. J. Am. Coll. Cardiol. 2006, 47, 685–686. [Google Scholar] [CrossRef]
  8. De Luca, G.; Navarese, E.P.; Cassetti, E.; Verdoia, M.; Suryapranata, H. Meta-Analysis of Randomized Trials of Glycoprotein IIb/IIIa Inhibitors in High-Risk Acute Coronary Syndromes Patients Undergoing Invasive Strategy. Am. J. Cardiol. 2011, 107, 198–203. [Google Scholar] [CrossRef]
  9. Verdoia, M.; Schaffer, A.; Barbieri, L.; Cassetti, E.; Piccolo, R.; Galasso, G.; Marino, P.; Sinigaglia, F.; De Luca, G. Benefits from New ADP Antagonists as Compared with Clopidogrel in Patients with Stable Angina or Acute Coronary Syndrome Undergoing Invasive Management: A Meta-Analysis of Randomized Trials. J. Cardiovasc. Pharmacol. 2014, 63, 339–350. [Google Scholar] [CrossRef]
  10. Nieswandt, B.; Pleines, I.; Bender, M. Platelet Adhesion and Activation Mechanisms in Arterial Thrombosis and Ischaemic Stroke. J. Thromb. Haemost. 2011, 9, 92–104. [Google Scholar] [CrossRef]
  11. Hosseini, E.; Beshkar, P.; Ghasemzadeh, M. Reverse Correlations of Collagen-Dependent Platelet Aggregation and Adhesion with GPVI Shedding during Storage. J. Thromb. Thrombolysis 2018, 46, 534–540. [Google Scholar] [CrossRef] [PubMed]
  12. Grover, S.P.; Mackman, N. Intrinsic Pathway of Coagulation and Thrombosis. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 331–338. [Google Scholar] [CrossRef]
  13. Gurbel, P.A.; Kuliopulos, A.; Tantry, U.S. G-Protein-Coupled Receptors Signaling Pathways in New Antiplatelet Drug Development. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 500–512. [Google Scholar] [CrossRef] [PubMed]
  14. Badimon, L.; Vilahur, G. Thrombosis Formation on Atherosclerotic Lesions and Plaque Rupture. J. Intern. Med. 2014, 276, 618–632. [Google Scholar] [CrossRef] [PubMed]
  15. Davì, G.; Patrono, C. Platelet Activation and Atherothrombosis. N. Engl. J. Med. 2007, 357, 2482–2494. [Google Scholar] [CrossRef] [PubMed]
  16. De Luca, G.; Verdoia, M.; Cassetti, E.; Schaffer, A.; Cavallino, C.; Bolzani, V.; Marino, P.; Novara Atherosclerosis Study Group (NAS). High Fibrinogen Level Is an Independent Predictor of Presence and Extent of Coronary Artery Disease among Italian Population. J. Thromb. Thrombolysis 2011, 31, 458–463. [Google Scholar] [CrossRef]
  17. Verdoia, M.; Schaffer, A.; Barbieri, L.; Aimaretti, G.; Marino, P.; Sinigaglia, F.; Suryapranata, H.; De Luca, G.; Novara Atherosclerosis Study Group (NAS). Impact of Diabetes on Neutrophil-to-Lymphocyte Ratio and Its Relationship to Coronary Artery Disease. Diabetes Metab. 2015, 41, 304–311. [Google Scholar] [CrossRef]
  18. Henn, V.; Slupsky, J.R.; Gräfe, M.; Anagnostopoulos, I.; Förster, R.; Müller-Berghaus, G.; Kroczek, R.A. CD40 Ligand on Activated Platelets Triggers an Inflammatory Reaction of Endothelial Cells. Nature 1998, 391, 591–594. [Google Scholar] [CrossRef]
  19. André, P.; Nannizzi-Alaimo, L.; Prasad, S.K.; Phillips, D.R. Platelet-Derived CD40L: The Switch-Hitting Player of Cardiovascular Disease. Circulation 2002, 106, 896–899. [Google Scholar] [CrossRef]
  20. von Hundelshausen, P.; Weber, C. Platelets as Immune Cells: Bridging Inflammation and Cardiovascular Disease. Circ. Res. 2007, 100, 27–40. [Google Scholar] [CrossRef]
  21. Semple, J.W.; Italiano, J.E.; Freedman, J. Platelets and the Immune Continuum. Nat. Rev. Immunol. 2011, 11, 264–274. [Google Scholar] [CrossRef]
  22. Lievens, D.; Zernecke, A.; Seijkens, T.; Soehnlein, O.; Beckers, L.; Munnix, I.C.A.; Wijnands, E.; Goossens, P.; van Kruchten, R.; Thevissen, L.; et al. Platelet CD40L Mediates Thrombotic and Inflammatory Processes in Atherosclerosis. Blood 2010, 116, 4317–4327. [Google Scholar] [CrossRef]
  23. Shiraki, R.; Inoue, N.; Kawasaki, S.; Takei, A.; Kadotani, M.; Ohnishi, Y.; Ejiri, J.; Kobayashi, S.; Hirata, K.-I.; Kawashima, S.; et al. Expression of Toll-Like Receptors on Human Platelets. Thromb. Res. 2004, 113, 379–385. [Google Scholar] [CrossRef] [PubMed]
  24. Singh, N.N.; Ramji, D.P. The Role of Transforming Growth Factor-Beta in Atherosclerosis. Cytokine Growth Factor Rev. 2006, 17, 487–499. [Google Scholar] [CrossRef] [PubMed]
  25. Lutgens, E.; Daemen, M.J. Transforming Growth Factor-Beta: A Local or Systemic Mediator of Plaque Stability? Circ. Res. 2001, 89, 853–855. [Google Scholar] [CrossRef] [PubMed]
  26. Grainger, D.J.; Wakefield, L.; Bethell, H.W.; Farndale, R.W.; Metcalfe, J.C. Release and Activation of Platelet Latent TGF-Beta in Blood Clots during Dissolution with Plasmin. Nat. Med. 1995, 1, 932–937. [Google Scholar] [CrossRef] [PubMed]
  27. Blair, P.; Flaumenhaft, R. Platelet α-Granules: Basic Biology and Clinical Correlates. Blood Rev. 2009, 23, 177–189. [Google Scholar] [CrossRef]
  28. Bevilacqua, M.P.; Stengelin, S.; Gimbrone, M.A.; Seed, B. Endothelial Leukocyte Adhesion Molecule 1: An Inducible Receptor for Neutrophils Related to Complement Regulatory Proteins and Lectins. Science 1989, 243, 1160–1165. [Google Scholar] [CrossRef]
  29. Geng, J.G.; Bevilacqua, M.P.; Moore, K.L.; McIntyre, T.M.; Prescott, S.M.; Kim, J.M.; Bliss, G.A.; Zimmerman, G.A.; McEver, R.P. Rapid Neutrophil Adhesion to Activated Endothelium Mediated by GMP-140. Nature 1990, 343, 757–760. [Google Scholar] [CrossRef]
  30. Gross, P.L.; Furie, B.C.; Merrill-Skoloff, G.; Chou, J.; Furie, B. Leukocyte-versus Microparticle-Mediated Tissue Factor Transfer during Arteriolar Thrombus Development. J. Leukoc. Biol. 2005, 78, 1318–1326. [Google Scholar] [CrossRef]
  31. Futosi, K.; Fodor, S.; Mócsai, A. Neutrophil Cell Surface Receptors and Their Intracellular Signal Transduction Pathways. Int. Immunopharmacol. 2013, 17, 638–650. [Google Scholar] [CrossRef] [PubMed]
  32. Brandt, E.; Van Damme, J.; Flad, H.D. Neutrophils Can Generate Their Activator Neutrophil-Activating Peptide 2 by Proteolytic Cleavage of Platelet-Derived Connective Tissue-Activating Peptide III. Cytokine 1991, 3, 311–321. [Google Scholar] [CrossRef] [PubMed]
  33. Bdeir, K.; Gollomp, K.; Stasiak, M.; Mei, J.; Papiewska-Pajak, I.; Zhao, G.; Worthen, G.S.; Cines, D.B.; Poncz, M.; Kowalska, M.A. Platelet-Specific Chemokines Contribute to the Pathogenesis of Acute Lung Injury. Am. J. Respir. Cell Mol. Biol. 2017, 56, 261–270. [Google Scholar] [CrossRef]
  34. Ghasemzadeh, M.; Ahmadi, J.; Hosseini, E. Platelet-Leukocyte Crosstalk in COVID-19: How Might the Reciprocal Links between Thrombotic Events and Inflammatory State Affect Treatment Strategies and Disease Prognosis? Thromb. Res. 2022, 213, 179–194. [Google Scholar] [CrossRef] [PubMed]
  35. Stark, K.; Philippi, V.; Stockhausen, S.; Busse, J.; Antonelli, A.; Miller, M.; Schubert, I.; Hoseinpour, P.; Chandraratne, S.; von Brühl, M.-L.; et al. Disulfide HMGB1 Derived from Platelets Coordinates Venous Thrombosis in Mice. Blood 2016, 128, 2435–2449. [Google Scholar] [CrossRef] [PubMed]
  36. Maugeri, N.; Campana, L.; Gavina, M.; Covino, C.; De Metrio, M.; Panciroli, C.; Maiuri, L.; Maseri, A.; D’Angelo, A.; Bianchi, M.E.; et al. Activated Platelets Present High Mobility Group Box 1 to Neutrophils, Inducing Autophagy and Promoting the Extrusion of Neutrophil Extracellular Traps. J. Thromb. Haemost. 2014, 12, 2074–2088. [Google Scholar] [CrossRef]
  37. Ghasemzadeh, M.; Hosseini, E. Platelet-Leukocyte Crosstalk: Linking Proinflammatory Responses to Procoagulant State. Thromb. Res. 2013, 131, 191–197. [Google Scholar] [CrossRef]
  38. Bonow, R.O.; Fonarow, G.C.; O’Gara, P.T.; Yancy, C.W. Association of Coronavirus Disease 2019 (COVID-19) with Myocardial Injury and Mortality. JAMA Cardiol. 2020, 5, 751. [Google Scholar] [CrossRef]
  39. Middleton, E.A.; He, X.-Y.; Denorme, F.; Campbell, R.A.; Ng, D.; Salvatore, S.P.; Mostyka, M.; Baxter-Stoltzfus, A.; Borczuk, A.C.; Loda, M.; et al. Neutrophil Extracellular Traps Contribute to Immunothrombosis in COVID-19 Acute Respiratory Distress Syndrome. Blood 2020, 136, 1169–1179. [Google Scholar] [CrossRef]
  40. Brill, A.; Fuchs, T.A.; Savchenko, A.S.; Thomas, G.M.; Martinod, K.; De Meyer, S.F.; Bhandari, A.A.; Wagner, D.D. Neutrophil Extracellular Traps Promote Deep Vein Thrombosis in Mice. J. Thromb. Haemost. 2012, 10, 136–144. [Google Scholar] [CrossRef]
  41. Massberg, S.; Grahl, L.; von Bruehl, M.-L.; Manukyan, D.; Pfeiler, S.; Goosmann, C.; Brinkmann, V.; Lorenz, M.; Bidzhekov, K.; Khandagale, A.B.; et al. Reciprocal Coupling of Coagulation and Innate Immunity via Neutrophil Serine Proteases. Nat. Med. 2010, 16, 887–896. [Google Scholar] [CrossRef] [PubMed]
  42. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D.; Wrobleski, S.K.; Wakefield, T.W.; Hartwig, J.H.; Wagner, D.D. Extracellular DNA Traps Promote Thrombosis. Proc. Natl. Acad. Sci. USA 2010, 107, 15880–15885. [Google Scholar] [CrossRef] [PubMed]
  43. Hally, K.E.; Parker, O.M.; Brunton-O’Sullivan, M.M.; Harding, S.A.; Larsen, P.D. Linking Neutrophil Extracellular Traps and Platelet Activation: A Composite Biomarker Score for Predicting Outcomes after Acute Myocardial Infarction. Thromb. Haemost. 2021, 121, 1637–1649. [Google Scholar] [CrossRef]
  44. Riegger, J.; Byrne, R.A.; Joner, M.; Chandraratne, S.; Gershlick, A.H.; ten Berg, J.M.; Adriaenssens, T.; Guagliumi, G.; Godschalk, T.C.; Neumann, F.-J.; et al. Histopathological Evaluation of Thrombus in Patients Presenting with Stent Thrombosis. A Multicenter European Study: A Report of the Prevention of Late Stent Thrombosis by an Interdisciplinary Global European Effort Consortium. Eur. Heart J. 2016, 37, 1538–1549. [Google Scholar] [CrossRef] [PubMed]
  45. Stakos, D.A.; Kambas, K.; Konstantinidis, T.; Mitroulis, I.; Apostolidou, E.; Arelaki, S.; Tsironidou, V.; Giatromanolaki, A.; Skendros, P.; Konstantinides, S.; et al. Expression of Functional Tissue Factor by Neutrophil Extracellular Traps in Culprit Artery of Acute Myocardial Infarction. Eur. Heart J. 2015, 36, 1405–1414. [Google Scholar] [CrossRef]
  46. Stähli, B.E.; Gebhard, C.; Duchatelle, V.; Cournoyer, D.; Petroni, T.; Tanguay, J.; Robb, S.; Mann, J.; Guertin, M.; Wright, R.S.; et al. Effects of the P-Selectin Antagonist Inclacumab on Myocardial Damage after Percutaneous Coronary Intervention According to Timing of Infusion: Insights from the SELECT-ACS Trial. J. Am. Heart Assoc. 2016, 5, e004255. [Google Scholar] [CrossRef]
  47. Tardif, J.-C.; Tanguay, J.-F.; Wright, S.R.; Duchatelle, V.; Petroni, T.; Grégoire, J.C.; Ibrahim, R.; Heinonen, T.M.; Robb, S.; Bertrand, O.F.; et al. Effects of the P-Selectin Antagonist Inclacumab on Myocardial Damage after Percutaneous Coronary Intervention for Non–ST-Segment Elevation Myocardial Infarction. J. Am. Coll. Cardiol. 2013, 61, 2048–2055. [Google Scholar] [CrossRef]
  48. Stähli, B.E.; Tardif, J.-C.; Carrier, M.; Gallo, R.; Emery, R.W.; Robb, S.; Cournoyer, D.; Blondeau, L.; Johnson, D.; Mann, J.; et al. Effects of P-Selectin Antagonist Inclacumab in Patients Undergoing Coronary Artery Bypass Graft Surgery. J. Am. Coll. Cardiol. 2016, 67, 344–346. [Google Scholar] [CrossRef]
  49. Kaiser, R.; Escaig, R.; Erber, J.; Nicolai, L. Neutrophil-Platelet Interactions as Novel Treatment Targets in Cardiovascular Disease. Front. Cardiovasc. Med. 2022, 8, 824112. [Google Scholar] [CrossRef]
  50. Chu, S.G.; Becker, R.C.; Berger, P.B.; Bhatt, D.L.; Eikelboom, J.W.; Konkle, B.; Mohler, E.R.; Reilly, M.P.; Berger, J.S. Mean Platelet Volume as a Predictor of Cardiovascular Risk: A Systematic Review and Meta-Analysis. J. Thromb. Haemost. 2010, 8, 148–156. [Google Scholar] [CrossRef]
  51. Jakubowski, J.A.; Thompson, C.B.; Vaillancourt, R.; Valeri, C.R.; Deykin, D. Arachidonic Acid Metabolism by Platelets of Differing Size. Br. J. Haematol. 1983, 53, 503–511. [Google Scholar] [CrossRef] [PubMed]
  52. Hosseini, E.; Ghasemzadeh, M.; Atashibarg, M.; Haghshenas, M. ROS Scavenger, N-Acetyl-L-Cysteine and NOX Specific Inhibitor, VAS2870 Reduce Platelets Apoptosis While Enhancing Their Viability during Storage. Transfusion 2019, 59, 1333–1343. [Google Scholar] [CrossRef] [PubMed]
  53. Kodiatte, T.A.; Manikyam, U.K.; Rao, S.B.; Jagadish, T.M.; Reddy, M.; Lingaiah, H.K.M.; Lakshmaiah, V. Mean Platelet Volume in Type 2 Diabetes Mellitus. J. Lab. Physicians 2012, 4, 5–9. [Google Scholar] [CrossRef]
  54. Hekimsoy, Z.; Payzin, B.; Ornek, T.; Kandoğan, G. Mean Platelet Volume in Type 2 Diabetic Patients. J. Diabetes Complicat. 2004, 18, 173–176. [Google Scholar] [CrossRef] [PubMed]
  55. Colwell, J.A.; Nesto, R.W. The Platelet in Diabetes: Focus on Prevention of Ischemic Events. Diabetes Care 2003, 26, 2181–2188. [Google Scholar] [CrossRef] [PubMed]
  56. Briggs, C. Quality Counts: New Parameters in Blood Cell Counting. Int. J. Lab. Hematol. 2009, 31, 277–297. [Google Scholar] [CrossRef]
  57. Thompson, C.B.; Eaton, K.A.; Princiotta, S.M.; Rushin, C.A.; Valeri, C.R. Size Dependent Platelet Subpopulations: Relationship of Platelet Volume to Ultrastructure, Enzymatic Activity, and Function. Br. J. Haematol. 1982, 50, 509–519. [Google Scholar] [CrossRef]
  58. Tsiara, S.; Elisaf, M.; Jagroop, I.A.; Mikhailidis, D.P. Platelets as Predictors of Vascular Risk: Is There a Practical Index of Platelet Activity? Clin. Appl. Thromb. Hemost. 2003, 9, 177–190. [Google Scholar] [CrossRef]
  59. Jagroop, I.A.; Clatworthy, I.; Lewin, J.; Mikhailidis, D.P. Shape Change in Human Platelets: Measurement with a Channelyzer and Visualisation by Electron Microscopy. Platelets 2000, 11, 28–32. [Google Scholar]
  60. Park, Y.; Schoene, N.; Harris, W. Mean Platelet Volume as an Indicator of Platelet Activation: Methodological Issues. Platelets 2002, 13, 301–306. [Google Scholar] [CrossRef]
  61. Schoene, N.W. Design Criteria: Tests Used to Assess Platelet Function. Am. J. Clin. Nutr. 1997, 65, 1665S–1668S. [Google Scholar] [CrossRef] [PubMed]
  62. Martin, J.F.; Trowbridge, E.A.; Salmon, G.; Plumb, J. The Biological Significance of Platelet Volume: Its Relationship to Bleeding Time, Platelet Thromboxane B2 Production and Megakaryocyte Nuclear DNA Concentration. Thromb. Res. 1983, 32, 443–460. [Google Scholar] [CrossRef]
  63. Giles, H.; Smith, R.E.; Martin, J.F. Platelet Glycoprotein IIb-IIIa and Size Are Increased in Acute Myocardial Infarction. Eur. J. Clin. Investig. 1994, 24, 69–72. [Google Scholar] [CrossRef] [PubMed]
  64. Pizzulli, L.; Yang, A.; Martin, J.F.; Lüderitz, B. Changes in Platelet Size and Count in Unstable Angina Compared to Stable Angina or Non-Cardiac Chest Pain. Eur. Heart J. 1998, 19, 80–84. [Google Scholar] [CrossRef] [PubMed]
  65. Huczek, Z.; Kochman, J.; Filipiak, K.J.; Horszczaruk, G.J.; Grabowski, M.; Piatkowski, R.; Wilczynska, J.; Zielinski, A.; Meier, B.; Opolski, G. Mean Platelet Volume on Admission Predicts Impaired Reperfusion and Long-Term Mortality in Acute Myocardial Infarction Treated with Primary Percutaneous Coronary Intervention. J. Am. Coll. Cardiol. 2005, 46, 284–290. [Google Scholar] [CrossRef]
  66. Murat, S.N.; Duran, M.; Kalay, N.; Gunebakmaz, O.; Akpek, M.; Doger, C.; Elcik, D.; Ocak, A.; Vatankulu, M.A.; Turfan, M.; et al. Relation between Mean Platelet Volume and Severity of Atherosclerosis in Patients with Acute Coronary Syndromes. Angiology 2013, 64, 131–136. [Google Scholar] [CrossRef] [PubMed]
  67. De Luca, G.; Verdoia, M.; Cassetti, E.; Schaffer, A.; Di Giovine, G.; Bertoni, A.; Di Vito, C.; Sampietro, S.; Aimaretti, G.; Bellomo, G.; et al. Mean Platelet Volume Is Not Associated with Platelet Reactivity and the Extent of Coronary Artery Disease in Diabetic Patients. Blood Coagul. Fibrinolysis 2013, 24, 619–624. [Google Scholar] [CrossRef] [PubMed]
  68. Verdoia, M.; Camaro, C.; Barbieri, L.; Schaffer, A.; Marino, P.; Bellomo, G.; Suryapranata, H.; De Luca, G. Mean Platelet Volume and the Risk of Periprocedural Myocardial Infarction in Patients Undergoing Coronary Angioplasty. Atherosclerosis 2013, 228, 136–141. [Google Scholar] [CrossRef]
  69. Halbmayer, W.M.; Haushofer, A.; Radek, J.; Schön, R.; Deutsch, M.; Fischer, M. Platelet Size, Fibrinogen and Lipoprotein(a) in Coronary Heart Disease. Coron. Artery Dis. 1995, 6, 397–402. [Google Scholar] [CrossRef]
  70. De Luca, G.; Santagostino, M.; Secco, G.G.; Cassetti, E.; Giuliani, L.; Franchi, E.; Coppo, L.; Iorio, S.; Venegoni, L.; Rondano, E.; et al. Mean Platelet Volume and the Extent of Coronary Artery Disease: Results from a Large Prospective Study. Atherosclerosis 2009, 206, 292–297. [Google Scholar] [CrossRef]
  71. Verdoia, M.; Pergolini, P.; Rolla, R.; Nardin, M.; Barbieri, L.; Schaffer, A.; Bellomo, G.; Marino, P.; Suryapranata, H.; De Luca, G. Mean Platelet Volume and High-Residual Platelet Reactivity in Patients Receiving Dual Antiplatelet Therapy with Clopidogrel or Ticagrelor. Expert Opin. Pharmacother. 2015, 16, 1739–1747. [Google Scholar] [CrossRef]
  72. Sansanayudh, N.; Numthavaj, P.; Muntham, D.; Yamwong, S.; McEvoy, M.; Attia, J.; Sritara, P.; Thakkinstian, A. Prognostic Effect of Mean Platelet Volume in Patients with Coronary Artery Disease. A Systematic Review and Meta-Analysis. Thromb. Haemost. 2015, 114, 1299–1309. [Google Scholar] [CrossRef]
  73. Vagdatli, E.; Gounari, E.; Lazaridou, E.; Katsibourlia, E.; Tsikopoulou, F.; Labrianou, I. Platelet Distribution Width: A Simple, Practical and Specific Marker of Activation of Coagulation. Hippokratia 2010, 14, 28–32. [Google Scholar]
  74. Vatankulu, M.A.; Sonmez, O.; Ertas, G.; Bacaksiz, A.; Turfan, M.; Erdogan, E.; Tasal, A.; Kul, S.; Uyarel, H.; Goktekin, O. A New Parameter Predicting Chronic Total Occlusion of Coronary Arteries. Angiology 2014, 65, 60–64. [Google Scholar] [CrossRef]
  75. Ege, M.R.; Guray, U.; Guray, Y.; Acıkgoz, S.; Demirkan, B. Platelet Distribution Width and Saphenous Vein Disease in Patients after CABG. Association with Graft Occlusion. Herz 2013, 38, 197–201. [Google Scholar] [CrossRef] [PubMed]
  76. Ihara, A.; Kawamoto, T.; Matsumoto, K.; Shouno, S.; Hirahara, C.; Morimoto, T.; Noma, Y. Relationship between Platelet Indexes and Coronary Angiographic Findings in Patients with Ischemic Heart Disease. Pathophysiol. Haemost. Thromb. 2006, 35, 376–379. [Google Scholar] [CrossRef] [PubMed]
  77. De Luca, G.; Venegoni, L.; Iorio, S.; Secco, G.G.; Cassetti, E.; Verdoia, M.; Schaffer, A.; Coppo, L.; Bellomo, G.; Marino, P.; et al. Platelet Distribution Width and the Extent of Coronary Artery Disease: Results from a Large Prospective Study. Platelets 2010, 21, 508–514. [Google Scholar] [CrossRef] [PubMed]
  78. Bessman, J.D.; Williams, L.J.; Gilmer, P.R. Mean Platelet Volume. The Inverse Relation of Platelet Size and Count in Normal Subjects, and an Artifact of Other Particles. Am. J. Clin. Pathol. 1981, 76, 289–293. [Google Scholar] [CrossRef] [PubMed]
  79. Lv, H.-C.; Wu, H.-Y.; Yin, J.-S.; Ge, J.-B. Thrombin Induced Platelet-Fibrin Clot Strength in Relation to Platelet Volume Indices and Inflammatory Markers in Patients with Coronary Artery Disease. Oncotarget 2017, 8, 64217–64223. [Google Scholar] [CrossRef]
  80. Larsen, S.B.; Grove, E.L.; Hvas, A.-M.; Kristensen, S.D. Platelet Turnover in Stable Coronary Artery Disease—Influence of Thrombopoietin and Low-Grade Inflammation. PLoS ONE 2014, 9, e85566. [Google Scholar] [CrossRef]
  81. De Luca, G.; Santagostino, M.; Secco, G.G.; Cassetti, E.; Giuliani, L.; Coppo, L.; Schaffer, A.; Fundaliotis, A.; Iorio, S.; Venegoni, L.; et al. Platelet-Large Cell Ratio and the Extent of Coronary Artery Disease: Results from a Large Prospective Study. J. Thromb. Thrombolysis 2010, 30, 426–433. [Google Scholar] [CrossRef] [PubMed]
  82. Verdoia, M.; Pergolini, P.; Rolla, R.; Nardin, M.; Barbieri, L.; Schaffer, A.; Bellomo, G.; Marino, P.; Suryapranata, H.; De Luca, G.; et al. Platelet Larger Cell Ratio and High-on Treatment Platelet Reactivity during Dual Antiplatelet Therapy. Cardiovasc. Drugs Ther. 2015, 29, 443–450. [Google Scholar] [CrossRef] [PubMed]
  83. Ault, K.A.; Rinder, H.M.; Mitchell, J.; Carmody, M.B.; Vary, C.P.H.; Hillman, R.S. The Significance of Platelets with Increased RNA Content (Reticulated Platelets) A Measure of the Rate of Thrombopoiesis. Am. J. Clin. Pathol. 1992, 98, 637–646. [Google Scholar] [CrossRef]
  84. Dusse, L.M.S.; Freitas, L.G. Clinical Applicability of Reticulated Platelets. Clin. Chim. Acta 2015, 439, 143–147. [Google Scholar] [CrossRef]
  85. McBane, R.D.; Gonzalez, C.; Hodge, D.O.; Wysokinski, W.E. Propensity for Young Reticulated Platelet Recruitment into Arterial Thrombi. J. Thromb. Thrombolysis 2014, 37, 148–154. [Google Scholar] [CrossRef] [PubMed]
  86. Hoffmann, J.J.M.L. Reticulated Platelets: Analytical Aspects and Clinical Utility. Clin. Chem. Lab. Med. 2014, 52, 1107–1117. [Google Scholar] [CrossRef]
  87. López-Jiménez, R.A.; Martín-Herrero, F.; González-Porras, J.R.; Sánchez-Barba, M.; Martín-Luengo, C.; Pabón-Osuna, P. Fracción de Plaquetas Inmaduras, Un Nuevo Marcador Pronóstico En El Síndrome Coronario Agudo. Rev. Esp. Cardiol. 2013, 66, 147–148. [Google Scholar] [CrossRef]
  88. Cesari, F.; Marcucci, R.; Gori, A.; Caporale, R.; Fanelli, A.; Casola, G.; Balzi, D.; Barchielli, A.; Valente, S.; Giglioli, C.; et al. Reticulated Platelets Predict Cardiovascular Death in Acute Coronary Syndrome Patients. Thromb. Haemost. 2013, 109, 846–853. [Google Scholar] [CrossRef]
  89. Berny-Lang, M.A.; Darling, C.E.; Frelinger, A.L.; Barnard, M.R.; Smith, C.S.; Michelson, A.D. Do Immature Platelet Levels in Chest Pain Patients Presenting to the Emergency Department Aid in the Diagnosis of Acute Coronary Syndrome? Int. J. Lab. Hematol. 2015, 37, 112–119. [Google Scholar] [CrossRef]
  90. Bernlochner, I.; Goedel, A.; Plischke, C.; Schüpke, S.; Haller, B.; Schulz, C.; Mayer, K.; Morath, T.; Braun, S.; Schunkert, H.; et al. Impact of Immature Platelets on Platelet Response to Ticagrelor and Prasugrel in Patients with Acute Coronary Syndrome. Eur. Heart J. 2015, 36, 3202–3210. [Google Scholar] [CrossRef]
  91. Perl, L.; Lerman-Shivek, H.; Rechavia, E.; Vaduganathan, M.; Leshem-Lev, D.; Zemer-Wassercug, N.; Dadush, O.; Codner, P.; Bental, T.; Battler, A.; et al. Response to Prasugrel and Levels of Circulating Reticulated Platelets in Patients with ST-Segment Elevation Myocardial Infarction. J. Am. Coll. Cardiol. 2014, 63, 513–517. [Google Scholar] [CrossRef] [PubMed]
  92. Verdoia, M.; Sartori, C.; Pergolini, P.; Nardin, M.; Rolla, R.; Barbieri, L.; Schaffer, A.; Marino, P.; Bellomo, G.; Suryapranata, H.; et al. Immature Platelet Fraction and High-on Treatment Platelet Reactivity with Ticagrelor in Patients with Acute Coronary Syndromes. J. Thromb. Thrombolysis 2016, 41, 663–670. [Google Scholar] [CrossRef] [PubMed]
  93. Verdoia, M.; Nardin, M.; Negro, F.; Rolla, R.; Carriero, A.; De Luca, G. Impact of Long-Term Therapy with Acetylsalicylic Acid on Immature Platelet Count. J. Cardiovasc. Med. 2019, 20, 306–312. [Google Scholar] [CrossRef] [PubMed]
  94. Duke, W.W. The Relation of Blood Platelets to Hemorrhagic Disease. J. Am. Med. Assoc. 1910, 55, 1185. [Google Scholar] [CrossRef]
  95. Born, G.V. Aggregation of Blood Platelets by Adenosine Diphosphate and Its Reversal. Nature 1962, 194, 927–929. [Google Scholar] [CrossRef]
  96. O’Brien, J. Platelet Aggregation: Part II Some Results from a New Method of Study. J. Clin. Pathol. 1962, 15, 452–455. [Google Scholar] [CrossRef]
  97. Tóth, O.; Calatzis, A.; Penz, S.; Losonczy, H.; Siess, W. Multiple Electrode Aggregometry: A New Device to Measure Platelet Aggregation in Whole Blood. Thromb. Haemost. 2006, 96, 781–788. [Google Scholar]
  98. Smith, J.W.; Steinhubl, S.R.; Lincoff, A.M.; Coleman, J.C.; Lee, T.T.; Hillman, R.S.; Coller, B.S. Rapid Platelet-Function Assay: An Automated and Quantitative Cartridge-Based Method. Circulation 1999, 99, 620–625. [Google Scholar] [CrossRef]
  99. Paniccia, R.; Antonucci, E.; Gori, A.M.; Marcucci, R.; Giglioli, C.; Antoniucci, D.; Gensini, G.F.; Abbate, R.; Prisco, D. Different Methodologies for Evaluating the Effect of Clopidogrel on Platelet Function in High-Risk Coronary Artery Disease Patients. J. Thromb. Haemost. 2007, 5, 1839–1847. [Google Scholar] [CrossRef]
  100. Kundu, S.K.; Heilmann, E.J.; Sio, R.; Garcia, C.; Davidson, R.M.; Ostgaard, R.A. Description of an in Vitro Platelet Function Analyzer—PFA-100. Semin. Thromb. Hemost. 1995, 21 (Suppl. S2), 106–112. [Google Scholar] [CrossRef]
  101. Varon, D.; Dardik, R.; Shenkman, B.; Kotev-Emeth, S.; Farzame, N.; Tamarin, I.; Savion, N. A New Method for Quantitative Analysis of Whole Blood Platelet Interaction with Extracellular Matrix under Flow Conditions. Thromb. Res. 1997, 85, 283–294. [Google Scholar] [CrossRef]
  102. Luddington, R.J. Thrombelastography/Thromboelastometry. Clin. Lab. Haematol. 2005, 27, 81–90. [Google Scholar] [CrossRef] [PubMed]
  103. Spurgeon, B.E.J.; Aburima, A.; Oberprieler, N.G.; Taskén, K.; Naseem, K.M. Multiplexed Phosphospecific Flow Cytometry Enables Large-Scale Signaling Profiling and Drug Screening in Blood Platelets. J. Thromb. Haemost. 2014, 12, 1733–1743. [Google Scholar] [CrossRef] [PubMed]
  104. Siller-Matula, J.M.; Francesconi, M.; Dechant, C.; Jilma, B.; Maurer, G.; Delle-Karth, G.; Gouya, G.; Ruzicka, K.; Podczeck-Schweighofer, A.; Christ, G. Personalized Antiplatelet Treatment after Percutaneous Coronary Intervention: The MADONNA Study. Int. J. Cardiol. 2013, 167, 2018–2023. [Google Scholar] [CrossRef] [PubMed]
  105. Collet, J.-P.; Thiele, H.; Barbato, E.; Barthélémy, O.; Bauersachs, J.; Bhatt, D.L.; Dendale, P.; Dorobantu, M.; Edvardsen, T.; Folliguet, T.; et al. 2020 ESC Guidelines for the Management of Acute Coronary Syndromes in Patients Presenting without Persistent ST-Segment Elevation. Eur. Heart J. 2021, 42, 1289–1367. [Google Scholar] [CrossRef] [PubMed]
  106. Cattaneo, M. High On-Treatment Platelet Reactivity—Definition and Measurement. Thromb. Haemost. 2013, 109, 792–798. [Google Scholar] [CrossRef]
  107. Stone, G.W.; Witzenbichler, B.; Weisz, G.; Rinaldi, M.J.; Neumann, F.-J.; Metzger, D.C.; Henry, T.D.; Cox, D.A.; Duffy, P.L.; Mazzaferri, E.; et al. Platelet Reactivity and Clinical Outcomes after Coronary Artery Implantation of Drug-Eluting Stents (ADAPT-DES): A Prospective Multicentre Registry Study. Lancet 2013, 382, 614–623. [Google Scholar] [CrossRef]
  108. Montalescot, G.; Rangé, G.; Silvain, J.; Bonnet, J.-L.; Boueri, Z.; Barthélémy, O.; Cayla, G.; Belle, L.; Van Belle, E.; Cuisset, T.; et al. High On-Treatment Platelet Reactivity as a Risk Factor for Secondary Prevention after Coronary Stent Revascularization: A Landmark Analysis of the ARCTIC Study. Circulation 2014, 129, 2136–2143. [Google Scholar] [CrossRef]
  109. Verdoia, M.; Pergolini, P.; Nardin, M.; Rolla, R.; Suryapranata, H.; Kedhi, E.; De Luca, G.; Novara Atherosclerosis Study Group (NAS). Ticagrelor and Prasugrel in Acute Coronary Syndrome: A Single-Arm Crossover Platelet Reactivity Study. J. Cardiovasc. Med. 2021, 22, 686–692. [Google Scholar] [CrossRef]
  110. Price, M.J.; Berger, P.B.; Angiolillo, D.J.; Teirstein, P.S.; Tanguay, J.-F.; Kandzari, D.E.; Cannon, C.P.; Topol, E.J. Evaluation of Individualized Clopidogrel Therapy after Drug-Eluting Stent Implantation in Patients with High Residual Platelet Reactivity: Design and Rationale of the GRAVITAS Trial. Am. Heart J. 2009, 157, 818–824.e1. [Google Scholar] [CrossRef]
  111. Aleil, B.; Jacquemin, L.; De Poli, F.; Zaehringer, M.; Collet, J.-P.; Montalescot, G.; Cazenave, J.-P.; Dickele, M.-C.; Monassier, J.-P.; Gachet, C. Clopidogrel 150 Mg/Day to Overcome Low Responsiveness in Patients Undergoing Elective Percutaneous Coronary Intervention: Results from the VASP-02 (Vasodilator-Stimulated Phosphoprotein-02) Randomized Study. JACC Cardiovasc. Interv. 2008, 1, 631–638. [Google Scholar] [CrossRef] [PubMed]
  112. Bonello, L.; Camoin-Jau, L.; Arques, S.; Boyer, C.; Panagides, D.; Wittenberg, O.; Simeoni, M.-C.; Barragan, P.; Dignat-George, F.; Paganelli, F. Adjusted Clopidogrel Loading Doses According to Vasodilator-Stimulated Phosphoprotein Phosphorylation Index Decrease Rate of Major Adverse Cardiovascular Events in Patients with Clopidogrel Resistance: A Multicenter Randomized Prospective Study. J. Am. Coll. Cardiol. 2008, 51, 1404–1411. [Google Scholar] [CrossRef] [PubMed]
  113. Cuisset, T.; Frere, C.; Quilici, J.; Morange, P.-E.; Mouret, J.-P.; Bali, L.; Moro, P.-J.; Lambert, M.; Alessi, M.-C.; Bonnet, J.L. Glycoprotein IIb/IIIa Inhibitors Improve Outcome after Coronary Stenting in Clopidogrel Nonresponders: A Prospective, Randomized Study. JACC Cardiovasc. Interv. 2008, 1, 649–653. [Google Scholar] [CrossRef] [PubMed]
  114. Valgimigli, M.; Campo, G.; de Cesare, N.; Meliga, E.; Vranckx, P.; Furgieri, A.; Angiolillo, D.J.; Sabatè, M.; Hamon, M.; Repetto, A.; et al. Intensifying Platelet Inhibition with Tirofiban in Poor Responders to Aspirin, Clopidogrel, or Both Agents Undergoing Elective Coronary Intervention: Results from the Double-Blind, Prospective, Randomized Tailoring Treatment with Tirofiban in Patients Sho. Circulation 2009, 119, 3215–3222. [Google Scholar] [CrossRef]
  115. Wang, X.; Zhang, D.; Zhuang, S.; Lai, Y. Modifying Clopidogrel Maintenance Doses According to Vasodilator-Stimulated Phosphoprotein Phosphorylation Index Improves Clinical Outcome in Patients with Clopidogrel Resistance. Clin. Cardiol. 2011, 34, 332–338. [Google Scholar] [CrossRef]
  116. Price, M.J.; Berger, P.B.; Teirstein, P.S.; Tanguay, J.-F.; Angiolillo, D.J.; Spriggs, D.; Puri, S.; Robbins, M.; Garratt, K.N.; Bertrand, O.F.; et al. Standard- vs. High-Dose Clopidogrel Based on Platelet Function Testing after Percutaneous Coronary Intervention: The GRAVITAS Randomized Trial. JAMA 2011, 305, 1097–1105. [Google Scholar] [CrossRef]
  117. Aradi, D.; Rideg, O.; Vorobcsuk, A.; Magyarlaki, T.; Magyari, B.; Kónyi, A.; Pintér, T.; Horváth, I.G.; Komócsi, A. Justification of 150 Mg Clopidogrel in Patients with High On-Clopidogrel Platelet Reactivity. Eur. J. Clin. Investig. 2012, 42, 384–392. [Google Scholar] [CrossRef]
  118. Hazarbasanov, D.; Velchev, V.; Finkov, B.; Postadjian, A.; Kostov, E.; Rifai, N.; Aradi, D. Tailoring Clopidogrel Dose According to Multiple Electrode Aggregometry Decreases the Rate of Ischemic Complications after Percutaneous Coronary Intervention. J. Thromb. Thrombolysis 2012, 34, 85–90. [Google Scholar] [CrossRef]
  119. Trenk, D.; Stone, G.W.; Gawaz, M.; Kastrati, A.; Angiolillo, D.J.; Müller, U.; Richardt, G.; Jakubowski, J.A.; Neumann, F.-J. A Randomized Trial of Prasugrel versus Clopidogrel in Patients with High Platelet Reactivity on Clopidogrel after Elective Percutaneous Coronary Intervention with Implantation of Drug-Eluting Stents: Results of the TRIGGER-PCI (Testing Platelet Reactivity in Patients Undergoing Elective Stent Placement on Clopidogrel to Guide Alternative Therapy with Prasugrel) Study. J. Am. Coll. Cardiol. 2012, 59, 2159–2164. [Google Scholar] [CrossRef]
  120. Collet, J.-P.; Cuisset, T.; Rangé, G.; Cayla, G.; Elhadad, S.; Pouillot, C.; Henry, P.; Motreff, P.; Carrié, D.; Boueri, Z.; et al. Bedside Monitoring to Adjust Antiplatelet Therapy for Coronary Stenting. N. Engl. J. Med. 2012, 367, 2100–2109. [Google Scholar] [CrossRef]
  121. Cayla, G.; Cuisset, T.; Silvain, J.; Leclercq, F.; Manzo-Silberman, S.; Saint-Etienne, C.; Delarche, N.; Bellemain-Appaix, A.; Range, G.; El Mahmoud, R.; et al. Platelet Function Monitoring to Adjust Antiplatelet Therapy in Elderly Patients Stented for an Acute Coronary Syndrome (ANTARCTIC): An Open-Label, Blinded-Endpoint, Randomised Controlled Superiority Trial. Lancet 2016, 388, 2015–2022. [Google Scholar] [CrossRef] [PubMed]
  122. Sibbing, D.; Aradi, D.; Jacobshagen, C.; Gross, L.; Trenk, D.; Geisler, T.; Orban, M.; Hadamitzky, M.; Merkely, B.; Kiss, R.G.; et al. Guided De-Escalation of Antiplatelet Treatment in Patients with Acute Coronary Syndrome Undergoing Percutaneous Coronary Intervention (TROPICAL-ACS): A Randomised, Open-Label, Multicentre Trial. Lancet 2017, 390, 1747–1757. [Google Scholar] [CrossRef] [PubMed]
  123. Tang, Y.-D.; Wang, W.; Yang, M.; Zhang, K.; Chen, J.; Qiao, S.; Yan, H.; Wu, Y.; Huang, X.; Xu, B.; et al. Randomized Comparisons of Double-Dose Clopidogrel or Adjunctive Cilostazol versus Standard Dual Antiplatelet in Patients with High Posttreatment Platelet Reactivity: Results of the CREATIVE Trial. Circulation 2018, 137, 2231–2245. [Google Scholar] [CrossRef] [PubMed]
  124. Podrez, E.A.; Byzova, T.V.; Febbraio, M.; Salomon, R.G.; Ma, Y.; Valiyaveettil, M.; Poliakov, E.; Sun, M.; Finton, P.J.; Curtis, B.R.; et al. Platelet CD36 Links Hyperlipidemia, Oxidant Stress and a Prothrombotic Phenotype. Nat. Med. 2007, 13, 1086–1095. [Google Scholar] [CrossRef]
  125. Togna, A.R.; Latina, V.; Orlando, R.; Togna, G.I. Cigarette Smoke Inhibits Adenine Nucleotide Hydrolysis by Human Platelets. Platelets 2008, 19, 537–542. [Google Scholar] [CrossRef]
  126. Hioki, H.; Aoki, N.; Kawano, K.; Homori, M.; Hasumura, Y.; Yasumura, T.; Maki, A.; Yoshino, H.; Yanagisawa, A.; Ishikawa, K. Acute Effects of Cigarette Smoking on Platelet-Dependent Thrombin Generation. Eur. Heart J. 2001, 22, 56–61. [Google Scholar] [CrossRef]
  127. Renaud, S.; Blache, D.; Dumont, E.; Thevenon, C.; Wissendanger, T. Platelet Function after Cigarette Smoking in Relation to Nicotine and Carbon Monoxide. Clin. Pharmacol. Ther. 1984, 36, 389–395. [Google Scholar] [CrossRef]
  128. Gremmel, T.; Steiner, S.; Seidinger, D.; Koppensteiner, R.; Panzer, S.; Kopp, C.W. Smoking Promotes Clopidogrel-Mediated Platelet Inhibition in Patients Receiving Dual Antiplatelet Therapy. Thromb. Res. 2009, 124, 588–591. [Google Scholar] [CrossRef]
  129. Crimi, G.; Somaschini, A.; Cattaneo, M.; Angiolillo, D.J.; Piscione, F.; Palmerini, T.; De Servi, S. Cigarette Smoking Reduces Platelet Reactivity Independently of Clopidogrel Treatment in Patients with Non-ST Elevation Acute Coronary Syndromes. Platelets 2018, 29, 309–311. [Google Scholar] [CrossRef]
  130. Nardin, M.; Verdoia, M.; Negro, F.; Rolla, R.; Tonon, F.; De Luca, G. Impact of Active Smoking on the Immature Platelet Fraction and Its Relationship with the Extent of Coronary Artery Disease. Eur. J. Clin. Investig. 2020, 50, e13181. [Google Scholar] [CrossRef]
  131. De Luca, G.; Dirksen, M.T.; Spaulding, C.; Kelbæk, H.; Schalij, M.; Thuesen, L.; van der Hoeven, B.; Vink, M.A.; Kaiser, C.; Musto, C.; et al. Impact of Diabetes on Long-Term Outcome after Primary Angioplasty. Diabetes Care 2013, 36, 1020–1025. [Google Scholar] [CrossRef] [PubMed]
  132. Wisman, P.P.; Roest, M.; Asselbergs, F.W.; de Groot, P.G.; Moll, F.L.; van der Graaf, Y.; de Borst, G.J. Platelet-Reactivity Tests Identify Patients at Risk of Secondary Cardiovascular Events: A Systematic Review and Meta-Analysis. J. Thromb. Haemost. 2014, 12, 736–747. [Google Scholar] [CrossRef] [PubMed]
  133. Nardin, M.; Verdoia, M.; Sartori, C.; Pergolini, P.; Rolla, R.; Barbieri, L.; Schaffer, A.; Bellomo, G.; Suryapranata, H.; De Luca, G. Diabetes Mellitus, Glucose Control Parameters and Platelet Reactivity in Ticagrelor Treated Patients. Thromb. Res. 2016, 143, 45–49. [Google Scholar] [CrossRef] [PubMed]
  134. Vivas, D.; Garcia-Rubira, J.C.; Bernardo, E.; Angiolillo, D.J.; Martin, P.; Calle-Pascual, A.; Nunez-Gil, I.; Macaya, C.; Fernandez-Ortiz, A. Effects of Intensive Glucose Control on Platelet Reactivity in Patients with Acute Coronary Syndromes. Results of the CHIPS Study (“Control de Hiperglucemia y Actividad Plaquetaria en Pacientes con Sindrome Coronario Agudo”). Heart 2011, 97, 803–809. [Google Scholar] [CrossRef]
  135. Filiopoulos, V.; Hadjiyannakos, D.; Vlassopoulos, D. New Insights into Uric Acid Effects on the Progression and Prognosis of Chronic Kidney Disease. Ren. Fail. 2012, 34, 510–520. [Google Scholar] [CrossRef]
  136. Elbasan, Z.; Şahin, D.Y.; Gür, M.; Kuloğlu, O.; Kıvrak, A.; İçen, Y.K.; Türkoğlu, C.; Yıldırım, A.; Özdoğru, İ.; Çaylı, M. Contrast-Induced Nephropathy in Patients with ST Elevation Myocardial Infarction Treated with Primary Percutaneous Coronary Intervention. Angiology 2014, 65, 37–42. [Google Scholar] [CrossRef]
  137. Barbieri, L.; Verdoia, M.; Pergolini, P.; Nardin, M.; Rolla, R.; Marino, P.; Bellomo, G.; Suryapranata, H.; De Luca, G. Uric Acid and High-Residual Platelet Reactivity in Patients Treated with Clopidogrel or Ticagrelor. Nutr. Metab. Cardiovasc. Dis. 2016, 26, 352–358. [Google Scholar] [CrossRef]
  138. Giovannucci, E.; Liu, Y.; Hollis, B.W.; Rimm, E.B. 25-Hydroxyvitamin D and Risk of Myocardial Infarction in Men: A Prospective Study. Arch. Intern. Med. 2008, 168, 1174–1180. [Google Scholar] [CrossRef]
  139. Tarcin, O.; Yavuz, D.G.; Ozben, B.; Telli, A.; Ogunc, A.V.; Yuksel, M.; Toprak, A.; Yazici, D.; Sancak, S.; Deyneli, O.; et al. Effect of Vitamin D Deficiency and Replacement on Endothelial Function in Asymptomatic Subjects. J. Clin. Endocrinol. Metab. 2009, 94, 4023–4030. [Google Scholar] [CrossRef]
  140. Silvagno, F.; De Vivo, E.; Attanasio, A.; Gallo, V.; Mazzucco, G.; Pescarmona, G. Mitochondrial Localization of Vitamin D Receptor in Human Platelets and Differentiated Megakaryocytes. PLoS ONE 2010, 5, e8670. [Google Scholar] [CrossRef]
  141. Aihara, K.; Azuma, H.; Akaike, M.; Ikeda, Y.; Yamashita, M.; Sudo, T.; Hayashi, H.; Yamada, Y.; Endoh, F.; Fujimura, M.; et al. Disruption of Nuclear Vitamin D Receptor Gene Causes Enhanced Thrombogenicity in Mice. J. Biol. Chem. 2004, 279, 35798–35802. [Google Scholar] [CrossRef] [PubMed]
  142. López-Farré, A.J.; Mateos-Cáceres, P.J.; Sacristán, D.; Azcona, L.; Bernardo, E.; de Prada, T.P.; Alonso-Orgaz, S.; Fernández-Arquero, M.; Fernández-Ortiz, A.; Macaya, C. Relationship between Vitamin D Binding Protein and Aspirin Resistance in Coronary Ischemic Patients: A Proteomic Study. J. Proteome Res. 2007, 6, 2481–2487. [Google Scholar] [CrossRef] [PubMed]
  143. Verdoia, M.; Pergolini, P.; Rolla, R.; Sartori, C.; Nardin, M.; Schaffer, A.; Barbieri, L.; Daffara, V.; Marino, P.; Bellomo, G.; et al. Vitamin D Levels and High-Residual Platelet Reactivity in Patients Receiving Dual Antiplatelet Therapy with Clopidogrel or Ticagrelor. Platelets 2016, 27, 576–582. [Google Scholar] [CrossRef] [PubMed]
  144. Verdoia, M.; Daffara, V.; Pergolini, P.; Rolla, R.; Marino, P.; Bellomo, G.; Carriero, A.; De Luca, G.; Novara Atherosclerosis Study Group (NAS). Vitamin D Binding Protein Rs7041 Polymorphism and High-Residual Platelet Reactivity in Patients Receiving Dual Antiplatelet Therapy with Clopidogrel or Ticagrelor. Vascul. Pharmacol. 2017, 93–95, 42–47. [Google Scholar] [CrossRef]
  145. Michos, E.D.; Misialek, J.R.; Selvin, E.; Folsom, A.R.; Pankow, J.S.; Post, W.S.; Lutsey, P.L. 25-Hydroxyvitamin D Levels, Vitamin D Binding Protein Gene Polymorphisms and Incident Coronary Heart Disease among Whites and Blacks: The ARIC Study. Atherosclerosis 2015, 241, 12–17. [Google Scholar] [CrossRef]
  146. Sibbing, D.; Koch, W.; Gebhard, D.; Schuster, T.; Braun, S.; Stegherr, J.; Morath, T.; Schömig, A.; von Beckerath, N.; Kastrati, A. Cytochrome 2C19*17 Allelic Variant, Platelet Aggregation, Bleeding Events, and Stent Thrombosis in Clopidogrel-Treated Patients with Coronary Stent Placement. Circulation 2010, 121, 512–518. [Google Scholar] [CrossRef]
  147. Mega, J.L.; Simon, T.; Collet, J.-P.; Anderson, J.L.; Antman, E.M.; Bliden, K.; Cannon, C.P.; Danchin, N.; Giusti, B.; Gurbel, P.; et al. Reduced-Function CYP2C19 Genotype and Risk of Adverse Clinical Outcomes among Patients Treated with Clopidogrel Predominantly for PCI: A Meta-Analysis. JAMA 2010, 304, 1821–1830. [Google Scholar] [CrossRef]
  148. Simon, T.; Verstuyft, C.; Mary-Krause, M.; Quteineh, L.; Drouet, E.; Méneveau, N.; Steg, P.G.; Ferrières, J.; Danchin, N.; Becquemont, L.; et al. Genetic Determinants of Response to Clopidogrel and Cardiovascular Events. N. Engl. J. Med. 2009, 360, 363–375. [Google Scholar] [CrossRef]
  149. Mega, J.L.; Hochholzer, W.; Frelinger, A.L.; Kluk, M.J.; Angiolillo, D.J.; Kereiakes, D.J.; Isserman, S.; Rogers, W.J.; Ruff, C.T.; Contant, C.; et al. Dosing Clopidogrel Based on CYP2C19 Genotype and the Effect on Platelet Reactivity in Patients with Stable Cardiovascular Disease. JAMA 2011, 306, 2221–2228. [Google Scholar] [CrossRef]
  150. Carreras, E.T.; Hochholzer, W.; Frelinger, A.L.; Nordio, F.; O’Donoghue, M.L.; Wiviott, S.D.; Angiolillo, D.J.; Michelson, A.D.; Sabatine, M.S.; Mega, J.L. Diabetes Mellitus, CYP2C19 Genotype, and Response to Escalating Doses of Clopidogrel. Insights from the ELEVATE-TIMI 56 Trial. Thromb. Haemost. 2016, 116, 69–77. [Google Scholar] [CrossRef]
  151. Scott, S.A.; Sangkuhl, K.; Stein, C.M.; Hulot, J.-S.; Mega, J.L.; Roden, D.M.; Klein, T.E.; Sabatine, M.S.; Johnson, J.A.; Shuldiner, A.R.; et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for CYP2C19 Genotype and Clopidogrel Therapy: 2013 Update. Clin. Pharmacol. Ther. 2013, 94, 317–323. [Google Scholar] [CrossRef] [PubMed]
  152. Royal Dutch Pharmacists Association (KNMP); Dutch Pharmacogenetics Working Group (DPWG). Pharmacogenetic Guidelines. Netherlands. Clopidogrel—CYP2C19. Available online: http://kennisbank.knmp.nl (accessed on 1 May 2023).
  153. Pereira, N.L.; Farkouh, M.E.; So, D.; Lennon, R.; Geller, N.; Mathew, V.; Bell, M.; Bae, J.-H.; Jeong, M.H.; Chavez, I.; et al. Effect of Genotype-Guided Oral P2Y12 Inhibitor Selection vs. Conventional Clopidogrel Therapy on Ischemic Outcomes after Percutaneous Coronary Intervention. JAMA 2020, 324, 761. [Google Scholar] [CrossRef] [PubMed]
  154. Tavenier, A.H.; Mehran, R.; Chiarito, M.; Cao, D.; Pivato, C.A.; Nicolas, J.; Beerkens, F.; Nardin, M.; Sartori, S.; Baber, U.; et al. Guided and Unguided De-Escalation from Potent P2Y12 Inhibitors among Patients with Acute Coronary Syndrome: A Meta-Analysis. Eur. Heart J. Cardiovasc. Pharmacother. 2022, 8, 492–502. [Google Scholar] [CrossRef] [PubMed]
  155. Mega, J.L.; Close, S.L.; Wiviott, S.D.; Shen, L.; Hockett, R.D.; Brandt, J.T.; Walker, J.R.; Antman, E.M.; Macias, W.L.; Braunwald, E.; et al. Cytochrome P450 Genetic Polymorphisms and the Response to Prasugrel: Relationship to Pharmacokinetic, Pharmacodynamic, and Clinical Outcomes. Circulation 2009, 119, 2553–2560. [Google Scholar] [CrossRef]
  156. Varenhorst, C.; Eriksson, N.; Johansson, Å.; Barratt, B.J.; Hagström, E.; Åkerblom, A.; Syvänen, A.-C.; Becker, R.C.; James, S.K.; Katus, H.A.; et al. Effect of Genetic Variations on Ticagrelor Plasma Levels and Clinical Outcomes. Eur. Heart J. 2015, 36, 1901–1912. [Google Scholar] [CrossRef]
  157. Nardin, M.; Verdoia, M.; Pergolini, P.; Rolla, R.; Barbieri, L.; Marino, P.; Bellomo, G.; Kedhi, E.; Suryapranata, H.; Carriero, A.; et al. Impact of Adenosine A2a Receptor Polymorphism Rs5751876 on Platelet Reactivity in Ticagrelor Treated Patients. Pharmacol. Res. 2018, 129, 27–33. [Google Scholar] [CrossRef]
  158. Laffont, B.; Corduan, A.; Plé, H.; Duchez, A.-C.; Cloutier, N.; Boilard, E.; Provost, P. Activated Platelets Can Deliver MRNA Regulatory Ago2•microRNA Complexes to Endothelial Cells via Microparticles. Blood 2013, 122, 253–261. [Google Scholar] [CrossRef]
  159. Kaudewitz, D.; Skroblin, P.; Bender, L.H.; Barwari, T.; Willeit, P.; Pechlaner, R.; Sunderland, N.P.; Willeit, K.; Morton, A.C.; Armstrong, P.C.; et al. Association of MicroRNAs and YRNAs with Platelet Function. Circ. Res. 2016, 118, 420–432. [Google Scholar] [CrossRef]
  160. Willeit, P.; Zampetaki, A.; Dudek, K.; Kaudewitz, D.; King, A.; Kirkby, N.S.; Crosby-Nwaobi, R.; Prokopi, M.; Drozdov, I.; Langley, S.R.; et al. Circulating MicroRNAs as Novel Biomarkers for Platelet Activation. Circ. Res. 2013, 112, 595–600. [Google Scholar] [CrossRef]
  161. Nagalla, S.; Shaw, C.; Kong, X.; Kondkar, A.A.; Edelstein, L.C.; Ma, L.; Chen, J.; McKnight, G.S.; López, J.A.; Yang, L.; et al. Platelet MicroRNA-MRNA Coexpression Profiles Correlate with Platelet Reactivity. Blood 2011, 117, 5189–5197. [Google Scholar] [CrossRef]
  162. Landry, P.; Plante, I.; Ouellet, D.L.; Perron, M.P.; Rousseau, G.; Provost, P. Existence of a MicroRNA Pathway in Anucleate Platelets. Nat. Struct. Mol. Biol. 2009, 16, 961–966. [Google Scholar] [CrossRef] [PubMed]
  163. Elgheznawy, A.; Shi, L.; Hu, J.; Wittig, I.; Laban, H.; Pircher, J.; Mann, A.; Provost, P.; Randriamboavonjy, V.; Fleming, I. Dicer Cleavage by Calpain Determines Platelet MicroRNA Levels and Function in Diabetes. Circ. Res. 2015, 117, 157–165. [Google Scholar] [CrossRef]
  164. Fejes, Z.; Póliska, S.; Czimmerer, Z.; Káplár, M.; Penyige, A.; Gál Szabó, G.; Debreceni, I.B.; Kunapuli, S.P.; Kappelmayer, J.; Nagy, B. Hyperglycaemia Suppresses MicroRNA Expression in Platelets to Increase P2RY12 and SELP Levels in Type 2 Diabetes Mellitus. Thromb. Haemost. 2017, 117, 529–542. [Google Scholar] [CrossRef] [PubMed]
  165. Peng, L.; Liu, J.; Qin, L.; Liu, J.; Xi, S.; Lu, C.; Yin, T. Interaction between Platelet-Derived MicroRNAs and CYP2C19*2 Genotype on Clopidogrel Antiplatelet Responsiveness in Patients with ACS. Thromb. Res. 2017, 157, 97–102. [Google Scholar] [CrossRef] [PubMed]
  166. Barwari, T.; Eminaga, S.; Mayr, U.; Lu, R.; Armstrong, P.C.; Chan, M.V.; Sahraei, M.; Fernández-Fuertes, M.; Moreau, T.; Barallobre-Barreiro, J.; et al. Inhibition of Profibrotic MicroRNA-21 Affects Platelets and Their Releasate. JCI Insight 2018, 3, e123335. [Google Scholar] [CrossRef]
  167. Wendel Garcia, P.D.; Fumeaux, T.; Guerci, P.; Heuberger, D.M.; Montomoli, J.; Roche-Campo, F.; Schuepbach, R.A.; Hilty, M.P.; Alfaro Farias, M.; Margarit, A.; et al. Prognostic Factors Associated with Mortality Risk and Disease Progression in 639 Critically Ill Patients with COVID-19 in Europe: Initial Report of the International RISC-19-ICU Prospective Observational Cohort. eClinicalMedicine 2020, 25, 100449. [Google Scholar] [CrossRef]
  168. Zapilko, V.; Fish, R.J.; Garcia, A.; Reny, J.-L.; Dunoyer-Geindre, S.; Lecompte, T.; Neerman-Arbez, M.; Fontana, P. MicroRNA-126 Is a Regulator of Platelet-Supported Thrombin Generation. Platelets 2020, 31, 746–755. [Google Scholar] [CrossRef]
  169. Becker, K.C.; Kwee, L.C.; Neely, M.L.; Grass, E.; Jakubowski, J.A.; Fox, K.A.A.; White, H.D.; Gregory, S.G.; Gurbel, P.A.; Carvalho, L.d.P.; et al. Circulating MicroRNA Profiling in Non-ST Elevated Coronary Artery Syndrome Highlights Genomic Associations with Serial Platelet Reactivity Measurements. Sci. Rep. 2020, 10, 6169. [Google Scholar] [CrossRef]
  170. Braza-Boïls, A.; Barwari, T.; Gutmann, C.; Thomas, M.R.; Judge, H.M.; Joshi, A.; Pechlaner, R.; Shankar-Hari, M.; Ajjan, R.A.; Sabroe, I.; et al. Circulating MicroRNA Levels Indicate Platelet and Leukocyte Activation in Endotoxemia Despite Platelet P2Y12 Inhibition. Int. J. Mol. Sci. 2020, 21, 2897. [Google Scholar] [CrossRef]
  171. Tsui, N.B.Y.; Ng, E.K.O.; Lo, Y.M.D. Stability of Endogenous and Added RNA in Blood Specimens, Serum, and Plasma. Clin. Chem. 2002, 48, 1647–1653. [Google Scholar] [CrossRef]
  172. Gutmann, C.; Mayr, M. Circulating MicroRNAs as Biomarkers and Mediators of Platelet Activation. Platelets 2022, 33, 512–519. [Google Scholar] [CrossRef]
  173. Jansen, F.; Yang, X.; Proebsting, S.; Hoelscher, M.; Przybilla, D.; Baumann, K.; Schmitz, T.; Dolf, A.; Endl, E.; Franklin, B.S.; et al. MicroRNA Expression in Circulating Microvesicles Predicts Cardiovascular Events in Patients with Coronary Artery Disease. J. Am. Heart Assoc. 2014, 3, e001249. [Google Scholar] [CrossRef]
  174. Zampetaki, A.; Willeit, P.; Tilling, L.; Drozdov, I.; Prokopi, M.; Renard, J.-M.; Mayr, A.; Weger, S.; Schett, G.; Shah, A.; et al. Prospective Study on Circulating MicroRNAs and Risk of Myocardial Infarction. J. Am. Coll. Cardiol. 2012, 60, 290–299. [Google Scholar] [CrossRef]
  175. Widera, C.; Gupta, S.K.; Lorenzen, J.M.; Bang, C.; Bauersachs, J.; Bethmann, K.; Kempf, T.; Wollert, K.C.; Thum, T. Diagnostic and Prognostic Impact of Six Circulating MicroRNAs in Acute Coronary Syndrome. J. Mol. Cell. Cardiol. 2011, 51, 872–875. [Google Scholar] [CrossRef]
  176. Capodanno, D.; Mehran, R.; Krucoff, M.W.; Baber, U.; Bhatt, D.L.; Capranzano, P.; Collet, J.-P.; Cuisset, T.; De Luca, G.; De Luca, L.; et al. Defining Strategies of Modulation of Antiplatelet Therapy in Patients with Coronary Artery Disease: A Consensus Document from the Academic Research Consortium. Circulation 2023, 147, 1933–1944. [Google Scholar] [CrossRef] [PubMed]
  177. De Servi, S.; Landi, A.; Savonitto, S.; De Luca, L.; De Luca, G.; Morici, N.; Montalto, C.; Crimi, G.; Cattaneo, M. Tailoring Oral Antiplatelet Therapy in Acute Coronary Syndromes: From Guidelines to Clinical Practice. J. Cardiovasc. Med. 2023, 24, 77–86. [Google Scholar] [CrossRef] [PubMed]
  178. Costa, F.; Montalto, C.; Branca, M.; Hong, S.-J.; Watanabe, H.; Franzone, A.; Vranckx, P.; Hahn, J.-Y.; Gwon, H.-C.; Feres, F.; et al. Dual Antiplatelet Therapy Duration after Percutaneous Coronary Intervention in High Bleeding Risk: A Meta-Analysis of Randomized Trials. Eur. Heart J. 2023, 44, 954–968. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Platelet activation pathways. The figure displays the intricate relationship between different stimuli and platelet activation. Created with BioRender.com. AC = adenylate cyclase; ADP = adenosine diphosphate; cAMP = cyclic adenosine monophosphate; DAG = diacylglycerol; IP3 = inositol triphosphate; MLKC = myosin light chain kinase; PAR-1 = protease activated receptor-1; PAR-4 = protease activated receptor-4; PIP2 = phosphatidylinositol bisphosphate; PKC = protein kinase C; PLC = phospholipase C; TP = thromboxane receptor; TXA2 = thromboxane A2; vWF = von Willebrand factor.
Figure 1. Platelet activation pathways. The figure displays the intricate relationship between different stimuli and platelet activation. Created with BioRender.com. AC = adenylate cyclase; ADP = adenosine diphosphate; cAMP = cyclic adenosine monophosphate; DAG = diacylglycerol; IP3 = inositol triphosphate; MLKC = myosin light chain kinase; PAR-1 = protease activated receptor-1; PAR-4 = protease activated receptor-4; PIP2 = phosphatidylinositol bisphosphate; PKC = protein kinase C; PLC = phospholipase C; TP = thromboxane receptor; TXA2 = thromboxane A2; vWF = von Willebrand factor.
Jcm 12 06074 g001
Figure 2. Activated platelet interplay between immune and cardiovascular systems. The figure shows complex interplays involving activated platelets between immune and cardiovascular system: on the left side, crosstalk between platelet and immune system is shown, reporting principal signaling pathways. On the right side, the main cardiovascular actions played by activated platelets are reported. The lines with arrows indicate promotion/activation, while the lines with final bar indicate blockage/inhibition. Created with BioRender.com. CAD = coronary artery disease; CXCL7 = chemokine (C-X-C motif) ligand 7; HMGB1 = high mobility group box 1; I-CAM = Intercellular adhesion molecule; NET = neutrophil extracellular trap; PF-4 = platelet factor-4; TGF-β1 = transforming growth factor-β1; V-CAM = Vascular adhesion molecule.
Figure 2. Activated platelet interplay between immune and cardiovascular systems. The figure shows complex interplays involving activated platelets between immune and cardiovascular system: on the left side, crosstalk between platelet and immune system is shown, reporting principal signaling pathways. On the right side, the main cardiovascular actions played by activated platelets are reported. The lines with arrows indicate promotion/activation, while the lines with final bar indicate blockage/inhibition. Created with BioRender.com. CAD = coronary artery disease; CXCL7 = chemokine (C-X-C motif) ligand 7; HMGB1 = high mobility group box 1; I-CAM = Intercellular adhesion molecule; NET = neutrophil extracellular trap; PF-4 = platelet factor-4; TGF-β1 = transforming growth factor-β1; V-CAM = Vascular adhesion molecule.
Jcm 12 06074 g002
Figure 3. Genetic and miRNAs. The figure depicts essential but crucial steps in pharmacokinetic of routinely used P2Y12 inhibitors, coupled with the potential target of the most studied miRNAs. The lines with arrows indicate promotion/activation, while the lines with final bar indicate blockage/inhibition. Created with BioRender.com. GPs = glycoproteins; ENT-1 = equilibrative nucleoside transporter-1; HRPR = high residual platelet reactivity; miRNAs = micro-RNAs; SNP = single nucleotide polymorphism; ? = hypothesized action.
Figure 3. Genetic and miRNAs. The figure depicts essential but crucial steps in pharmacokinetic of routinely used P2Y12 inhibitors, coupled with the potential target of the most studied miRNAs. The lines with arrows indicate promotion/activation, while the lines with final bar indicate blockage/inhibition. Created with BioRender.com. GPs = glycoproteins; ENT-1 = equilibrative nucleoside transporter-1; HRPR = high residual platelet reactivity; miRNAs = micro-RNAs; SNP = single nucleotide polymorphism; ? = hypothesized action.
Jcm 12 06074 g003
Table 1. Main platelet function test used in clinical practice.
Table 1. Main platelet function test used in clinical practice.
MethodsSampleApplicationPrincipleAdvantagesDisadvantages
Tests based on platelet aggregation
Light transmission platelet aggregation (LTA); also named optical platelet aggregationCitrated PRPScreening test for bleeding propensity
Diagnostic for platelet defects, both congenital and acquired
Monitoring antiplatelet treatment effect
Photo-optical measurement of light transmission increase in relation to specific agonist-induced platelet aggregationHistorical gold standard
Diagnostic method
Different platelet pathways investigated
Sensitive to different antiplatelet drugs therapy
Manual and long sample processing
Pre-analytic and analytic variables
High sample volume
Time consuming
Impedance platelet aggregationCitrated WBScreening test for bleeding propensity
Diagnostic for platelet defects, both congenital and acquired
Monitoring antiplatelet treatment effect
Measurement of electrical impedance between two electrodes after induction of platelet aggregation through a specific agonistNo sample processing
Diagnostic method
Flexible
Different platelet pathways investigated
Sensitive to anti-platelet therapy
Close to LTA
Sample preparation
Time consuming
Lumiaggregometry (i.e., VerifyNow)Citrated WBModified aggregometry for detection of storage/release disordersLTA or WB aggregometry combined with luminescenceNo WB processing
Quick and easy to do methods
Monitoring antiplatelet therapy
Nonflexible
Very expensive
Limited hematocrit and platelet count
PlateletworksCitrated WBMonitoring of the platelet response to antiplatelet agentsPlatelets’ counting and aggregates pre- and post-activation (use of ADP or arachidonic acid) in whole blood based in GP IIb/IIIa-dependent aggregationMinimal sample preparation
Easy, rapid screening test
Indirect assay
Performance within few minutes after sample collection
Required adjunctive platelet count
Scarce clinical data
Platelet function methods combined with viscoelastic test
TEG/r-TEG
platelet mapping system
Citrated WBAssessment of global hemostasis plus monitoring antiplatelet treatments effectAssessment of the rate and the strength of clot formation based on low shear-induced and agonist additionPoint of care for viscoelastic test
Global hemostasis test
Measure clot properties
Reduces blood transfusions
More studies are needed
ROTEM plateletCitrated WBAssessment of global hemostasis plus diagnostic of platelet defects plus monitoring antiplatelet treatments effectMeasurement of electrical impedance increase in relation to agonist-induced platelet aggregationAdaptation from TEG: results are identical
Predicts bleeding
Reduces blood transfusions
Improves clinical outcome
Global hemostasis test WB platelet aggregometry
Limited hematocrit and platelet count range (for platelet system)
Lack of clinical studies
Tests based on platelet adhesion under shear stress
PFA-100; Innovance PFA-200Citrated WBAssessment of bleeding risk and drug effects
Searching severe platelet dysfunctions, revealing of vWF disease
Time evaluation of high shear WB flow blocked by platelet plug into a hole in activated surface. Use of combination of collagen/epinephrine or collagen/ADPIn vitro standardized bleeding test
Quick and easy to do
Sensitive to severe platelet defects
Rigid closed system
Dependent on hematocrit, platelet count and vWF
Not sensitive to platelet granules defects
Contrasting evidence in thienopyridines treatment, especially for PFA-100
IMPACT—Cone and Plate(let) Analyzer (CPA)Citrated WBScreening of primary hemostasis and platelet defectsShear-induced platelet adhesion–aggregation upon specific surface covered by polystyrene. Ongoing studies on addition ADP and arachidonic acid for antiplatelet therapy monitoringGlobal platelet method
Small sample volume
Expensive
Experienced staff
Lack of clinical studies
Not widely available
Platelet analysis based on flow cytometry
Flow cytometryCitrated WB, PRP, washed plateletsCell counting, detection platelet activation by extent of expression of surface and/or cytoplasmic biomarkersEngineering laser-based detection of suspending fluorescent label platelets in a flowing solutionUseful into diagnose inherited platelet disordersExpensive
Experienced staff
Not widely available
Vasodilator Stimulated Phosphoprotein (VASP)Citrated WBIntracellular platelet pathwayImmunofluorescence on assay with a specific monoclonal antibodyUseful into monitoring antiplatelet drugExpensive
Evaluation of Thromboxane metabolites
Radio- or enzyme-linked immune assaysSerum, urine, citrated plasmaMeasurement of TXA2 metabolites (and β-TG, PF4, soluble P-selectin)
Ligand-binding assaysDirectly related to COX-1, the aspirin’s targetIndirect measure
No platelet specific
ADP = adenosine diphosphate; β-TG = β-thromboglobulin; COX-1 = cyclooxygenase-1; LTA = light transmission platelet aggregation; PF4 = platelet factor-4; PFA = platelet function assay; PRP = platelet-rich-plasma; ROTEM = rotational thromboelastometry; r-TEG = rapid thromboelastography; TEG = thromboelastography; TXA2 = thromboxane A2; WB = whole blood; vWF = von Willebrand factor.
Table 2. Main clinical trials using platelet function test for tailoring antiplatelet therapy.
Table 2. Main clinical trials using platelet function test for tailoring antiplatelet therapy.
StudyYearPFTInterventionSubjectsStudy designFindings
VASP-02 [111]2008VASPVASP-guided switch after 2 weeks in low responders150 patients undergoing elective PCIRandomized to 150 mg vs. 75 mg clopidogrelGreater platelet inhibition with 150 mg clopidogrel in poor responders
Bonello et al. [112]2008VASPVASP-based adjustment of clopidogrel loading dose162 patients undergoing PCI with basal VASP > 50%Randomized to VASP-guided vs. standard of careLower rate of 1-month MACE in VASP-guided group
Cuisset et al. [113] 2008LTAPFT-based identification of poor responder before randomization149 clopidogrel non-responders undergoing elective PCI Randomized to administration of additional GP IIb/IIIa antagonist vs. standard of careLower rate of 1-month CV event in intervention group
3T/2R [114] 2009VerifyNowPFT-based identification of poor responder before randomization263 ASA and/or clopidogrel non-responders undergoing PCIRandomized to tirofiban administration vs. standard of careLower MI and 1-months MACE in intervention group
Wang et al. [115]2011VASPPFT-based drug adjustment in tailored strategy306 patients undergoing PCI with basal VASP > 50%Randomized to VASP-guided vs. standard of careLower rate of 1-year MACE in VASP-guided group
GRAVITAS [116] 2011VerifyNowPFT-based identification of poor responder before randomization2796 clopidogrel non-responder patients undergoing PCIRandomized to higher dose of clopidogrel vs. standard of careNo differences in 6-month MACE rate
Aradi et al. [117]2012LTAPFT-based identification of poor responder before randomization200 clopidogrel non-responders undergoing elective PCIRandomized to higher dose of clopidogrel vs. standard of careLower MI and 1-months MACE in intervention group
Hazarbasanov et al. [118]2012Impedance aggregometryPFT-based drug adjustment in tailored strategy192 patients undergoing PCIRandomization to tailored strategy vs. standard of careLower rate of 6-months MACCE in intervention group
TRIGGER-PCI [119] 2012VerifyNowPFT-based identification of poor responder before randomization423 clopidogrel non-responders undergoing PCIRandomized to switch to prasugrel vs. clopidogrel maintenanceHigher platelet inhibition in prasugrel group.
ARTIC [120]2012VerifyNowPFT-based administration of additional bolus of clopidogrel, prasugrel or ASA along with GP IIb/IIIa antagonist2440 patients undergoing elective PCIRandomization to tailored strategy vs. standard of careNo differences in 1-year MACE
MADONNA study [104] 2013Impedance aggregometryPFT-based additional antiplatelet drug loading dose798 patients undergoing PCIRandomization to tailored strategy vs. standard of careLower stent thrombosis and ACS rate in tailored strategy
ANTARTIC [121] 2016VerifyNowPFT-based dose or drug adjustment in tailored strategy877 patients undergoing PCI for an ACSRandomization to tailored strategy vs. standard of careNo differences in 1-year MACE
TROPICAL-ACS [122]2017Impedance aggregometryPFT-based de-escalation strategy after 14 days2610 patients undergoing PCI for an ACSRandomization to tailored strategy vs. standard of careTailored de-escalation strategy non-inferior to standard of care
CREATIVE [123]2018TEGPFT-based identification of poor responder before randomization1087 clopidogrel poor responders undergoing PCIRandomized to higher dose of clopidogrel vs. standard dose of clopidogrel plus cilostazol vs. standard of careLower rate of 18-months MACCE with the adjunct of cilostazol
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nardin, M.; Verdoia, M.; Cao, D.; Nardin, S.; Kedhi, E.; Galasso, G.; van ‘t Hof, A.W.J.; Condorelli, G.; De Luca, G. Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention. J. Clin. Med. 2023, 12, 6074. https://doi.org/10.3390/jcm12186074

AMA Style

Nardin M, Verdoia M, Cao D, Nardin S, Kedhi E, Galasso G, van ‘t Hof AWJ, Condorelli G, De Luca G. Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention. Journal of Clinical Medicine. 2023; 12(18):6074. https://doi.org/10.3390/jcm12186074

Chicago/Turabian Style

Nardin, Matteo, Monica Verdoia, Davide Cao, Simone Nardin, Elvin Kedhi, Gennaro Galasso, Arnoud W. J. van ‘t Hof, Gianluigi Condorelli, and Giuseppe De Luca. 2023. "Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention" Journal of Clinical Medicine 12, no. 18: 6074. https://doi.org/10.3390/jcm12186074

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