Next Article in Journal
Th2 Cytokines (Interleukin-5 and -9) Polymorphism Affects the Response to Anti-TNF Treatment in Polish Patients with Ankylosing Spondylitis
Next Article in Special Issue
Frail Silk: Is the Hughes-Stovin Syndrome a Behçet Syndrome Subtype with Aneurysm-Involved Gene Variants?
Previous Article in Journal
Optical and Flame-Retardant Properties of a Series of Polyimides Containing Side Chained Bulky Phosphaphenanthrene Units
Previous Article in Special Issue
Anticoagulation Monitoring with Activated Partial ThromboPlastin Time and Anti-Xa Activity in Intensive Care Unit Patients: Interest of Thrombin Generation Assay
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases

by
Marco Heestermans
1,2,
Géraldine Poenou
3,
Anne-Claire Duchez
1,2,
Hind Hamzeh-Cognasse
1,
Laurent Bertoletti
1,3,4 and
Fabrice Cognasse
1,2,*
1
INSERM, U1059, SAINBIOSE, Jean Monnet University, F-42023 Saint-Etienne, France
2
French Blood Establishment (EFS) Auvergne Rhône Alpes-Scientific Department, F-42270 Saint-Etienne, France
3
Service de Médecine Vasculaire et Thérapeutique (Department of Vascular and Therapeutic Medicine), CHU de St-Etienne (St-Etienne University Hospital), F-42055 Saint-Etienne, France
4
INSERM, CIC-1408, CHU Saint-Etienne, F-42055 Saint-Etienne, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(21), 13176; https://doi.org/10.3390/ijms232113176
Submission received: 7 September 2022 / Revised: 25 October 2022 / Accepted: 27 October 2022 / Published: 29 October 2022
(This article belongs to the Special Issue Thromboinflammation: An Evolving Multifaceted Concept)

Abstract

:
Venous thromboembolism (VTE) is the third leading cardiovascular cause of death and is conventionally treated with anticoagulants that directly antagonize coagulation. However, recent data have demonstrated that also platelets play a crucial role in VTE pathophysiology. In the current review, we outline how platelets are involved during all stages of experimental venous thrombosis. Platelets mediate initiation of the disease by attaching to the vessel wall upon which they mediate leukocyte recruitment. This process is referred to as immunothrombosis, and within this novel concept inflammatory cells such as leukocytes and platelets directly drive the progression of VTE. In addition to their involvement in immunothrombosis, activated platelets can directly drive venous thrombosis by supporting coagulation and secreting procoagulant factors. Furthermore, fibrinolysis and vessel resolution are (partly) mediated by platelets. Finally, we summarize how conventional antiplatelet therapy can prevent experimental venous thrombosis and impacts (recurrent) VTE in humans.

1. Introduction

Platelets are small anucleated cells that are fragmented from megakaryocytes and released into the circulation. In healthy individuals, platelet levels range from 150,000 to 450,000 platelets per microliter of blood [1]. Many different functions are attributed to platelets and they are implicated in processes such as thrombosis and hemostasis, inflammation and regulation of tumor growth (reviewed in [2,3]). In the field of thrombosis and hemostasis, for many years platelets have been the main clinical target for managing arterial thrombotic diseases [4]. In contrast, platelets are believed to play a less prominent role during thrombosis within the venous system. Indeed, the vast platelet aggregates found in arterial thrombi are absent in venous thrombi [5]. Therefore, antiplatelet therapy is not commonly prescribed in the treatment of (recurrent) Venous Thrombo-Embolism (VTE), and anticoagulant drugs are more effective in combatting the disease. Anticoagulants inhibit secondary hemostasis or coagulation. The main driver of coagulation is thrombin, which cleaves the circulating glycoprotein fibrinogen to fibrin [6]. Fibrin can be crosslinked to generate large fibrin strands that can entangle blood cells leading to large thrombi that locally disrupt blood flow causing Deep Vein Thrombosis (DVT) [7]. When a fragment of the formed thrombus dislodges, it embolizes through the blood stream towards the lungs. This condition is known as Pulmonary Embolism (PE). VTE, the collective term for DVT and PE, affects 1–3 per 1000 individuals per year, and is the third leading cardiovascular cause of death [8].
The pathophysiology of venous thrombosis is different from that of arterial thrombosis, although an increasing overlap has been identified in recent years [9,10]. Venous thrombi are formed over a prolonged period (minutes versus hours/days). Initially, they mainly comprise erythrocytes and fibrin with the thrombi lacking the vast platelet aggregates found in larger quantities in arterial thrombi [11,12,13]. During the second stage of venous thrombosis, the fibrin gradually becomes dominant and replaces the blood cells. In the third stage of the disease, the thrombus is resolved in two phases. Collagen initially replaces the fibrinolyzing thrombus after which it is resolved in a process called collagenolysis.
In recent years, platelets have been viewed as mediators of immunothrombosis, a novel concept that directly links thrombotic diseases including VTE to inflammatory processes [14]. It is a well-documented fact that platelets play a pivotal role in inflammation [15,16]. Firstly, platelets express numerous receptors and store hundreds of secretory products that are instrumental in functional responses. All of these components provide potential new routes for drug targeting to combat inflammatory disorders [17,18,19]. Secondly, platelets are widely recognized as secretors of proinflammatory cytokines, chemokines and biological response modifiers, such as the CD40 ligand [20]. Thirdly, platelets secrete lipid mediators that can act as autocrine and/or paracrine mediators, although the related intracellular signaling pathways are less clear than in nucleated cells and have yet to be clarified [21,22].
Starting out as a relatively innocent bystander, platelets have been converted into a potentially crucial driver of inflammation and appear to be critical for immunothrombosis. This review aims to provide an overview of the current literature in which platelets have been linked to in vivo venous thrombosis. The role of platelets during different aspects of venous thrombosis will then be outlined from initial inflammation of the vessel wall and the recruitment of leukocytes to platelet activators known to be directly involved in thrombus formation. Furthermore, the role of platelets in coagulation, fibrinolysis, thrombus resolution and vessel restauration will be covered. Finally, the efficacy of antiplatelets in (pre)-clinical VTE will be summarized.

2. Platelets Interact with the Venous Vessel Wall

Venous thrombosis in large veins is initiated with the local inflammation of venous endothelial cells, which causes locally increased expression of surface selectins and VWF [23,24]. VWF is a multimeric glycoprotein produced by endothelial cells and stored in Weibel Palade bodies (WPBs) that reside in the cytoplasm [25]. Upon cellular stress, such as hypoxia or contact with inflammatory cytokines, WBPs fuse with the cellular membrane leading to the secretion of VWF. VWF functions in the circulation as a carrier protein for coagulation factor VIII (FVIII) by protecting the protease from rapid degradation, thus VWF and FVIII plasma levels are correlated [26]. FVIII is crucial for normal coagulation and a deficiency thereof results in hemophilia A, a severe bleeding disorder characterized by hemorrhages after vascular injury, deep bruising and joint pain and swelling [27]. Individuals with normal FVIII but a mutated form of VWF that lost its affinity for FVIII present a hemophilia A-like phenotype, emphasizing the critical role of VWF in stabilizing circulating FVIII. In addition to its role as a carrier protein, VWF mediates several other processes including primary hemostasis by directing platelet adhesion via its A1 domain that interacts with the platelet receptor glycoprotein GPIb [28] (Figure 1). Since the VWF A1 domain is exposed only upon high hemodynamic sheer stress, the consensus was that endothelial VWF-dependent platelet adhesion is not crucial for events in the venous vasculature. However, in mouse models for venous thrombosis based on the (partial) ligation of the inferior vena cava (IVC), VWF−/− mice were protected from the disease [29]. Interestingly, when mice were supplemented with FVIII, thromboprotection was maintained, thereby implying that platelet-VWF interaction is pivotal for thrombus formation. In addition, infusion of an antibody blocking platelet interaction with the VWF A1 domain prevented thrombosis in wild-type mice. In the event of an injury within the venous vasculature, circulating VWF can also directly interact with the exposed subendothelial collagen via its A3 domain [30,31]. This interaction exposes the A1 domain that once again allows recruitment of circulating platelets via GPIb.
Podoplanin is a well conserved, mucin-type transmembrane protein and is expressed in the vessel wall, although it is currently not entirely clear which precise cell type(s) express(es) the protein [32]. During hypoxia, a process comprising local stenosis and a risk factor for thrombus formation, the protein is upregulated. C-type lectin-like receptor 2 (CLEC-2) is a platelet receptor acting as a ligand for podoplanin. Indeed, platelet-specific CLEC-2−/− mice were protected from venous thrombosis in a stenosis model of the IVC [33]. Conversely, blocking podoplanin using a specific antibody significantly decreased thrombus formation. Interestingly, a recent study showed that deletion of the S1P exporter Mfsd2b impeded venous thrombosis, possibly through decreased expression of podoplanin in the vessel wall of the IVC [34]. Podoplanin is a molecule that is also expressed by certain types of cancer cells. Cancers in which high levels of podoplanin are expressed are associated with an increased risk of developing cancer-associated thrombosis, the most common cause of cancer-related death after cancer itself [35,36]. Pre-clinical studies have shown that blocking podoplanin decreased thrombosis onset in cancer models, thus demonstrating that targeting podoplanin in cancer patients may be a feasible strategy in reducing thrombosis [37,38,39].
Damage of the venous vessel wall is one of the best-documented risk factors for VTE, and a specific trigger such as surgery or trauma can lead to thrombosis in large veins. When subendothelial cells expressing the highly prothrombotic coagulation factor and tissue factor (TF) are exposed in the circulation, coagulation is catalyzed via the extrinsic coagulation pathway. The platelet receptor, GPVI, displays strong affinity for subendothelial collagen, and in certain clinical scenarios this interaction may contribute to platelet recruitment in the veins. Antibody-inhibited or GPVI−/− mice are less inclined to develop venous thrombosis [40,41]. However, it should be taken into account that platelet GPVI also binds to fibrin, which amplifies thrombin generation and maintains platelet recruitment [42]. Blocking platelet GPVI is linked to clot instability, possibly due to its interaction with fibrin [43,44].

3. Platelets Interact with and/or Recruit Other Immune Cells

The concept of immunothrombosis has been introduced in the last decade to describe the link between immunology and thrombotic processes in diseases such as viral and bacterial infections, and arterial thrombosis [45,46,47]. Immune cells are now also recognized as drivers of the pathology in VTE. It is often forgotten that platelets also function as immune cells and their traditional and novel roles in maintaining hemostasis and mediating inflammation, respectively, are not strictly separated during immunothrombosis [14]. In fact, in venous thrombosis, platelets most likely act more as mediators of leukocytes and less as hemostatic cells, in contrast to arterial thrombosis. Neutrophils and monocytes are two additional immune cell populations of interest and are known to play an important role in venous thrombus formation, with platelets often acting as mediators [48,49].
Neutrophils are granulocytes that are rapidly recruited towards sites of (sterile) inflammation. They also appear to be among the first leukocytes to arrive at the site where thrombus formation is initiated [50] (Figure 1). Locally, neutrophils are able to release so-called Neutrophil Extracellular Traps (NETs) that consist of DNA, histones and antimicrobial proteins. NETs are prothrombotic web-like structures in the extracellular space and dismantling NETs appears to be a tempting novel antithrombotic strategy for further investigation [51,52]. Several research groups have shown that the use of DNase-1, a compound that hydrolyses extracellular DNA, can prevent venous thrombosis in animal models [48,53,54]. Interestingly, a recently published study claims that DNase not only hydrolyses DNA but also Adenosine TriPhosphate (ATP) and Adenosine DiPhosphate (ADP) into adenosine [13]. ATP and ADP are potent agonists of both neutrophils and platelets while adenosine inhibits neutrophil function. Whether DNase prevents venous thrombosis via hydrolysis of DNA in NETs or the platelet agonists ATP/ADP, or a combination of both, has yet to be confirmed.
Platelets and neutrophils are closely interlinked as mediators of venous thrombosis, and several direct interactions between the two cell types have been described. In this instance, platelets mostly act as mediators to recruit or activate neutrophils. In addition to its role in platelet recruitment to the endothelium, platelet receptor GPIb interacts with the leukocyte complement receptor, Mac-1 (also known as αMβ2). Blocking this specific interaction using a GPIb specific antibody resulted in reduced neutrophil recruitment, decreased NET formation and lowered thrombus formation [48]. Conversely, blocking or a genetic deficiency of Mac-1 attenuated thrombosis in mouse models of arterial thrombosis and microvascular thrombosis [55]. It remains to be seen whether antagonizing Mac-1, also directly prevents venous thrombosis.
Solute Carrier Family 44 Member 2 (SLC44A2) is a choline transporter on which a single nucleotide polymorphism (SNP) has been correlated with an increased risk of VTE in humans [56,57]. Individuals carrying the SNP HNA3a have a 30% higher risk of developing VTE compared to those carrying the HNA3b variant. This finding was of considerable interest since it was the first gene not directly implicated in hemostasis to be identified in a genome wide association study (GWAS) on VTE. Mechanistically, platelet-expressed SLC44A2 promotes thrombin-mediated platelet activation by transporting choline to the mitochondria, thereby increasing mitochondrial oxygen consumption and ATP production [58]. SLC44A2 on neutrophils appears to have two binding partners that are relevant for thrombosis. Firstly, neutrophil SLC44A2 can directly interact with the A1 domain on endothelial VWF upon which NETosis takes place, thereby promoting thrombosis [59]. It was shown that the SNP found in the original GWAS studies determines the affinity of this interaction, possibly explaining the slightly increased risk of VTE in individuals with HNA3a compared to HNA3b [60]. Secondly, platelets can act as an intermediate between neutrophils and endothelial VWF. Platelets can bind to decrypted VWF via GPIb, thereby activating the αIIbβ3 receptor. Subsequently, activated αIIbβ3 is detected by neutrophil-expressed SLC44A2 under low sheer stress, again resulting in increased NETosis [61].
P-selectin (P-sel) is expressed by activated platelets and is probably the most commonly used marker to demonstrate platelet activation in vitro, making use of techniques such as flow cytometry or ELISA. P-sel expressed on the platelet membrane can interact with both neutrophils and monocytes via their P-selectin glycoprotein ligand-1 (PSGL-1) receptor, upon which firm adhesion is achieved through the binding of Mac-1 [62]. Consequently, monocytes are triggered to produce TF-positive microvesicles that promote coagulation and venous thrombosis [63,64]. Platelet P-sel also stimulates NET formation by interacting with neutrophils [65], and inhibiting P-sel function prevented experimental DVT [66]. Von Brühl et al., showed that P-sel deficient mice were protected from venous thrombosis, while the transfusion of wild type platelets did not correct the thrombotic phenotype [48]. These data imply that platelet P-sel per se is not crucial for leukocyte recruitment and that P-sel expressed by endothelial cells alone is sufficient to guide and activate neutrophils and monocytes at the site of immunothrombosis.
Additional direct or indirect interactions between platelets and neutrophils have been described, which possibly contribute to NETosis and thrombus formation. S1P exporter Mfsd2b deficient platelets demonstrated reduced interaction between platelets and neutrophils compared to wild type platelets [34]. In addition to supporting platelet activation and experimental arterial thrombosis, platelet proline-rich tyrosine kinase Pyk2 regulated platelet-induced NETosis [67]. Pyk2 deficient mice were protected from venous thrombosis induced by IVC ligation. Platelet membrane-expressed CD40L interacts with neutrophil CD40 and soluble CD40L promoted platelet-neutrophil interaction, leading to increased NET formation [68,69]. Mice deficient in platelet amyloid precursor protein (APP) demonstrated increased platelet-neutrophil aggregates, NETosis and developed larger venous thrombi in a venous thrombosis mouse model [70]. The absence of APP might boost the inflammatory status of thrombotic mice, as evidenced by elevated plasma C-reactive protein levels.
Platelets can physically interact with other circulating immune cells such as neutrophils and monocytes, and this process most probably contributes to the onset of (experimental) thrombosis in the venous vasculature. Another mode via which platelets can mediate various physiological processes is the secretion of active substances in the extracellular space [18]. Upon activation, platelets usually secrete various products stored in their lysosomes or α- or dense granules, thereby presenting different roles in inflammation, hemostasis and wound healing, for instance [71]. Platelet-secreted components also appear to be involved in venous thrombosis pathophysiology. SNAP23 deficient mice fail to release the contents of their platelet granules [72]. Interestingly, these mice are protected from both venous and arterial thrombosis, and although the exact mechanism for thromboprotection is currently unclear, these data emphasize the physiological relevance of granule secretion in thrombosis. P-sel can be expressed by platelets on the membrane and is used as an activation marker. However, P-sel can also be secreted from the platelet’s α-granules (soluble P-sel [73]). Soluble P-sel stemming from either platelets or endothelial cells can attract leukocytes to a site of (sterile) injury and may serve as a biomarker for VTE in humans [74,75,76]. Secreted platelet factor 4 (PF4 or CXCL4) may serve as a biomarker for VTE in humans [77,78,79], although its direct involvement in venous thrombosis pathophysiology is not exactly known. Myeloid-related protein-14 (MRP-14), a member of the alarmin or danger-associated molecular pattern molecules family, can be secreted by both platelets and neutrophils [80]. Platelet MRP-14 mediated NET formation and MRP-14 deficiency protected mice from venous thrombosis, which was corrected when mice were transfused with wild type platelets.

4. Platelet Activators Promoting Venous Thrombosis

Upon activation, platelets undergo a phenotypical change associated with a significant change in expression and secretion pattern [81]. In the context of coagulation and venous thrombosis, several components that modulate platelet activation have been identified. Toll-like receptors (TLRs) are expressed by various immune cells [82,83,84]. These receptors recognize Damage-Associated Molecular Patterns (DAMPs) or Pathogen-Associated Molecular Patterns (PAMPs), resulting in cells that become activated, generate oxygen and nitrogen radicals, and/or produce cytokines (Figure 1) [85]. Platelets also express TLRs at both their membrane and intracellular, and platelets are activated in response to DAMPs or PAMPs after which they mediate an immune response [82,86]. In mouse venous thrombosis, HMGB1 can function as a DAMP and is both secreted and recognized by platelets, presumably via TLR2, TLR4, and/or RAGE [87]. HMGB1 deficient mice or wild type mice treated with an HMGB1 inhibitor presented decreased thrombus formation, possibly because of reduced monocyte recruitment and signs of NETosis in the thrombus. In addition, supplementing mice with recombinant HMGB1 enhanced thrombus formation while platelet-specific HMGB1−/− mice were protected from thrombosis, coinciding with a decrease in the number of neutrophils present in the formed thrombi [53]. Kindlin-3, an integrin activator expressed in platelets, and paxillin appeared crucial for αIIbβ3 inside-out signaling [88,89,90]. In mice, disruption of the interaction between kindlin-3 and paxillin or a selected deficiency for platelet-Kindlin-3 demonstrated a significant decrease in experimental DVT.
Thrombin is the major coagulation mediator, and is involved in both inflammatory and thrombotic processes [91]. In humans, thrombin serves as a potent platelet activator mainly via protease-activated receptors (PAR) 1 [92], and the PAR1-specific thrombin derivative TRAP (thrombin receptor-activating peptide) is often used to activate platelets in vitro. In mice, PAR4 is the main thrombin receptor and platelet-specific PAR4−/− mice are indeed protected from venous thrombosis [93]. The complement cascade is an innate immune response that relies on the sequential activation of serine proteases, similar to the coagulation cascade. Platelets from complement factor C3 deficient mice displayed reduced platelet activation in response to a PAR4-specific stimulus [94]. In line with these results, Subramaniam et al., demonstrated that, upon activation, platelets from C3−/− mice showed reduced VWF binding, P-sel exposure, αIIbβ3 activation and phosphatidyl serine (PS) exposure [95]. Additionally, in an IVC ligation model, C3−/− mice were protected from thrombosis. Pro-inflammatory interleukins 9 and 17A promoted venous thrombosis in a mouse model based on IVC stasis, most likely due to direct stimulation of platelet activation [96,97]. Mice treated with estradiol demonstrated diminished platelet responsiveness and were protected from thromboembolic events and venous thrombosis [98,99]. Interestingly, estradiol treatment did not modulate the potency of the coagulation cascade, implying that the effects on thrombosis were platelet-dependent. Growth arrest-specific gene 6 (Gas6) is a vitamin-K dependent growth factor that is expressed and secreted by many cell types, including platelets [100]. Platelets from Gas6 deficient mice or mice with a platelet receptor deficiency (Tyro3, Axl and Mer) demonstrated a substantial decrease in the stabilization of platelet aggregates due to a disturbed inside-out signaling of αIIbβ3, resulting in protection from experimental venous thrombosis [101,102]. NADPH oxidase-derived reactive oxygen species (ROS) mediate normal platelet function and activation, and multiple studies have shown that altered ROS production modulates the risk for venous thrombosis, both in humans and in vivo models (reviewed in [103]).

5. Platelets and the Coagulation Cascade

Platelets are involved in the initiation of venous thrombosis by directing leukocytes to the site of (sterile) inflammation. However, they also directly participate in coagulation or secondary haemostasis [104,105]. Coagulation is the process that shapes blood from a liquid to a gel-like substance, following a process usually visualized as a complex cascade of bioactive serine proteases resulting in fibrin formation. A blood clot is characterized by these polymerized fibrin strands that entangle other circulating cells. Activated platelets, which play a supporting role in coagulation, express increased levels of phosphatidylserine (PS) on their surface that can be used as a marker of platelet activation (Figure 2). Pro- and anticoagulant factors with a domain rich in gamma-carboxylated amino acids (the so-called GLA domain of vitamin K-dependent coagulation factors) are recruited to PS-rich surfaces where they are efficiently activated [106,107,108]. Platelet membrane coagulation affects the tenase complex (factor Xa and IXa, and co-factor VIIIa) and the prothrombinase complex (factor Xa and cofactor Va). In addition, anticoagulants proteins C and S exert their effects on the PS-positive platelet surface, where they inactivate cofactors Va and VIIIa.
Platelet Pyk2 regulates PS expression, possibly explaining the thromboprotection observed in Pyk2 deficient mice [67]. In a mouse model relying on the transient inhibition of two important natural anticoagulants, antithrombin and protein C, complete platelet depletion corrected the onset of severe thrombotic coagulopathy [109]. Since traces of subclinical fibrin formation were found in the liver of the platelet-depleted mice, it was hypothesized that the absence of platelets did not prevent thrombosis entirely but rather limited the progression of massive coagulation for which a PS surface is required. These data suggest that restricting platelet PS exposure may be one way of limiting venous thrombosis progression.
In addition to the well-documented role of platelets in exposing PS to mediate coagulation, platelets also secrete several components that directly influence coagulation. Activated platelets are the main source of calcium required for the functioning of serine proteases such as thrombin and factor X [110]. In addition, recent studies have shown that platelets secrete several pro- and anticoagulants, such as fibrinogen [111], factor V [112,113], factor VIII [114,115], factor IX [116], factor XIII [117,118], VWF [119] and protein S [120]. All of these coagulation factors are not produced exclusively by platelets, hence the significance of the platelet-derived pool of each coagulation factor may vary.
Upon activation, platelets shed 0.1–1-μm fragments that express functional receptors and are PS-positive. These cellular fragments are commonly referred to as microvesicles or microparticles, and it has been shown that platelet-derived microvesicles are omnipresent within the circulation [121,122]. PS-positive microvesicles can catalyze coagulation, possibly in locations where platelets cannot penetrate [123,124], and it has been shown that they are enriched around the developing thrombus site [125]. Platelet-derived microvesicles promote venous thrombosis in mice [126] and may be useful as biomarkers for VTE in humans [127]. Another challenge is to investigate the role of microvesicles in transporting micro RNAs (miRNA) in immunothrombotic diseases. miRNAs are non-coding and modulate gene expression through mRNA degradation or translational repression of numerous targets [128]. Understanding the heterogeneity of (platelet) microvesicles will help them become biomarkers and may reveal specific roles for each microparticle subtype in health and disease [129].
Polyphosphates (polyp) are another prothrombotic component secreted by platelets from their dense granules that mediate coagulation. Interestingly, it has been shown that polyp are both procoagulant and proinflammatory and may thus mediate immunothrombosis [130]. Polyp’s function and activity depends on its polymer chain length [131]. Long chain polyp is insoluble and, following platelet activation, the polymer is retained on the surface in nanoparticles with divalent metal ions [132]. Here, it locally supports FXII activation leading to bradykinin formation and thrombin formation following the intrinsic pathway. A polyp-neutralizing agent prevents FXII-dependent mouse venous thrombosis, emphasizing its physiological relevance and clinical potential in preventing or curing VTE in humans [133]. Platelet-secreted soluble short chain polyp does not efficiently activate FXII. However, this form is able to modulate coagulation by supporting thrombin-dependent FXI activation [134] and FV activation [135], and also modifies fibrin polymerization [136].

6. Platelets during Thrombus Resolution and Vessel Restoration

Thrombus resolution starts with the process of breaking down crosslinked fibrin strands in a process called fibrinolysis. Defective fibrinolysis can lead to several chronic and acute pathologies, mostly linked to haematological disorders [137]. The main fibrinolysin is plasmin, a protease resulting from the cleavage of inactive circulating plasminogen. Comparable to coagulation, fibrinolysis is tightly controlled with several specific proteases, cofactors and protease inhibitors that all have a part to play in ensuring correct thrombus fibrinolysis. Platelets provide the PS-positive surface to allow efficient fibrinolysis (Figure 2). In addition, several reports show that platelets store and secrete both fibrinolytics, such as plasminogen [138,139] and antifibrinolytic agents including the plasminogen activator inhibitor-1 [140] and thrombin activatable fibrinolysis inhibitor [141]. Secretion of these factors make platelets key mediators of fibrinolysis.
Thrombus maturation, resolution and post-thrombotic vessel wall remodeling are the final stages in the lifespan of a thrombus, and platelets are also involved [142]. In a murine model for venous thrombosis, platelet depletion was seen to decrease thrombus fibrosis, smooth muscle cell invasion and intimal vessel wall thickening-a recognized histological feature associated with post-thrombotic syndrome. It was suggested that platelets stimulate smooth muscle cell invasion by secreting TGF-β, bFGF and PDGF, all of which are growth factors associated with this process. Interestingly, mice experimentally infected with Staphylococcus aureus also demonstrated a misguided thrombus resolution, possibly mediated by upregulation of similar growth factors [143]. Mice treated with statin, a compound associated with decreased platelet activity, showed improved thrombus resolution [144]. Whether the antiplatelet effect is responsible for altering thrombus resolution has yet to be investigated, since statins are known to have pleiotropic effects including dampening immune responses [145]. The platelet endothelial cell adhesion molecule-1 (PECAM-1) is a protein expressed by many different cells including platelets [146]. PECAM-1 deficient mice also displayed increased platelet activation and misguided thrombus resolution, although the precise role of platelet-expressed PECAM-1 was not addressed in the case of the latter [147].

7. Platelet Inhibition to Prevent Venous Thrombosis

Platelets appear to play a pivotal role in the initiation, progression and resolution of experimental venous thrombosis. Indeed, it has been demonstrated that platelet inhibitors/antiplatelet drugs prevent venous thrombosis in different animal models. Currently prescribed as antiplatelet drugs are Acetylsalicylic acid (ASA, better known as aspirin), the P2Y12 receptor antagonists clopidogrel, prasugrel, and ticagrelor, and the αIIbβ3 receptor antagonist, abciximab [148]. Aspirin is an irreversible inhibitor of platelet aggregation, which prevents platelet thromboxane A2 synthesis by direct binding to COX-1 and COX-2 [149]. Although the exact mechanism of action has not been fully elucidated in recent decades, the drug has been used for thousands of years as an effective pain killer or anti-inflammatory drug since it blocks the production of prostaglandins that can serve as molecular pain signals. P2Y12 inhibitors are part of a second class of antiplatelet drugs [150]. These compounds also inhibit platelet aggregation by preventing interaction between ADP and the P2Y12 platelet receptor that normally results in platelet activation. Finally, the αIIbβ3 receptor antagonist, abciximab, reduces fibrinogen-mediated platelet-platelet interactions thereby preventing the formation of aggregates [151,152].
In a laser injury model in rats, aspirin treatment decreased the number of thrombi and emboli formed [153]. More recently, a mouse model based on IVC ligation showed that aspirin decreased thrombus size, presumably because of reduced monocyte and neutrophil activation leading to lower TF activity and NET formation [154]. Multiple studies have assessed the effects of P2Y12 antagonists, mainly clopidogrel, on experimental venous thrombosis. Clopidogrel appeared to be effective in preventing venous thrombosis in dogs, rabbits and rats [155,156,157,158]. In mice treated with ticagrelor, venous thrombosis as a result of complete IVC ligation was significantly decreased [159]. Thrombus weight as a result of a ferric-chloride induced vascular injury of the vena cava was lowered in P2Y12−/− or wild type clopidogrel-treated mice compared to (untreated) wild type mice [160]. The accumulation of platelets in large vessel venous thrombosis in mice was decreased upon treatment with clopidogrel, while fibrin formation was not significantly altered [161]. In the mouse IVC stenosis model, aspirin and clopidogrel combined, but not alone, reduced thrombus formation [162]. Finally, the small molecule UNC2025, a compound that interferes with the Gas6 signalling pathway, decreased platelet activation alone or in combination with clopidogrel, and increased survival in a lethal PE mouse model [163].
In contrast to studies on experimental venous thrombosis, the efficacy of antiplatelet therapy in preventing human (secondary) VTE is still debated, and only aspirin has been tested in dedicated clinical studies. Current clinical practice guidelines from the American Academy of Orthopedic Surgeons, the American College of Chest Physicians, the American Society of Hematology and the National Institute of Health recommend aspirin as a potential thromboprophylactic agent, like Low Molecular Weight Heparin (LMWH) or Direct Oral AntiCoagulants (DOAC) without any preference expressed in terms of therapeutic options. In the EPCAT study, aspirin proved effective in preventing VTE after 5 days of treatment with the FXa inhibitor, rivaroxaban, in orthopedic surgery at the cost of a slightly increased risk of bleeding [164]. In contrast, the CRISTAL trial failed to demonstrate noninferiority of aspirin as compared to LMWH [165]. The results of another ongoing trial, the PEPPER trial (NCT02810704) compares aspirin to rivaroxaban and warfarin, and this trial may provide more inside on the use of aspirin to prevent primary VTE.
Two randomized clinical trials published in 2012 have highlighted encouraging results for aspirin in the secondary prevention or extended treatment of VTE. In the WARFASA and ASPIRE studies, the incidence of recurrent VTE was reduced by 40% and 35%, respectively [166,167]. In both studies, reduction of recurrent VTE coincided with a relatively low incidence of bleeding events. In 2017, Weitz et al., published the results of a randomized controlled phase 3 trial comparing rivaroxaban to aspirin for the extended treatment of VTE [168]. The incidence of adverse events was similar for both drugs but the risk of recurrence was lower with rivaroxaban. Consequently, aspirin was not validated for secondary prevention of VTE. In 2019, Mai et al., performed a meta-analysis of 18 independent randomized controlled trials, comparing the risk of recurrent thrombosis and major bleeding when treated with several antithrombotic drugs (LMWH, DOAC, VKA, or aspirin) [169]. For aspirin, a recurrence rate of 0.71 (0.55–0.91) was reported, which is about three times greater than the risk to patients treated with anticoagulants, there being no significant reduction in major bleeding. These data imply that the benefit of curing patients with recurrent thrombosis using aspirin is limited.
In a study on patients treated with both rivaroxaban/dabigatran and antiplatelet drugs, it was shown that the improved efficacy to prevent atherothrombosis did not stem from a direct antiplatelet effect of DOACs but from their inhibitory effect on platelet aggregation secondary to coagulation activation. This effect may differ per DOAC, depending on the targeted coagulation factor [170]. These results were confirmed in a dedicated pharmacokinetic study on blood samples from patients receiving rivaroxaban, a DOAC targeting FXa, in which the effect of several platelets agonists (ADP, arachidonic acid, epinephrine, collagen and thrombin) at different doses administrated were tested [171]. Thrombin antagonist dabigatran may inhibit platelet activation, in particular by thrombin [172]. Finally, FXa inhibitors apixaban and edoxaban might have the capacity to indirectly inhibit platelet aggregation [173,174].

8. Conclusions

For decades, platelets have been regarded as crucial players in arterial thrombosis, and this disease can be efficiently prevented or cured by antiplatelet therapy. More recently, pre-clinical data have provided an overwhelming amount of evidence to suggest that platelets also play a pivotal role in venous thrombosis as mediators of immunothrombosis. Platelets are involved in initial vascular inflammation, platelet secretion of biological response modifiers, recruitment and/or activation of leukocytes, thrombus progression and resolution as well as vessel wall remodeling. In this review, we aimed to provide an overview of the known roles of platelets in experimental venous thrombosis, and to outline the implications in the human VTE context. The versatility of platelets, particularly in terms of their inflammatory properties, is a constant source of amazement and many questions remain unanswered regarding their role in experimental venous thrombosis, and how to extrapolate these in vivo results to the human pathology. Finally, depending on the clinical scenario, it will be very interesting to establish whether antiplatelet therapy is a viable option in addition to or as a replacement for anticoagulation therapy in VTE patients.

Author Contributions

Conceptualization: M.H., L.B. and F.C. Literature searches: M.H. and G.P. Writing—Original Draft Preparation: M.H. and G.P. Visualization: A.-C.D. Revision of the original draft: A.-C.D., H.H.-C., L.B. and F.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the French Blood Establishment (EFS) and the Association “Les Amis de Rémi” Savigneux, France.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the medical staff and personnel of the Etablissement Français du Sang, France, for their medical and technical support during our investigations. We are also extremely grateful to the blood donors for taking part in this study.

Conflicts of Interest

The authors have no conflict of interest to declare.

References

  1. George, J.N. Platelets. Lancet 2000, 355, 1531–1539. [Google Scholar] [CrossRef]
  2. van der Meijden, P.E.J.; Heemskerk, J.W.M. Platelet biology and functions: New concepts and clinical perspectives. Nat. Rev. Cardiol. 2019, 16, 166–179. [Google Scholar] [CrossRef] [PubMed]
  3. Jenne, C.N.; Kubes, P. Platelets in inflammation and infection. Platelets 2015, 26, 286–292. [Google Scholar] [CrossRef] [PubMed]
  4. Stark, K.; Massberg, S. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat. Rev. Cardiol. 2021, 18, 666–682. [Google Scholar] [CrossRef]
  5. Koupenova, M.; Kehrel, B.E.; Corkrey, H.A.; Freedman, J.E. Thrombosis and platelets: An update. Eur. Heart J. 2016, 38, 785–791. [Google Scholar] [CrossRef]
  6. Versteeg, H.H.; Heemskerk, J.W.M.; Levi, M.; Reitsma, P.H. New Fundamentals in Hemostasis. Physiol. Rev. 2013, 93, 327–358. [Google Scholar] [CrossRef] [Green Version]
  7. Di Nisio, M.; van Es, N.; Büller, H.R. Deep vein thrombosis and pulmonary embolism. Lancet 2016, 388, 3060–3073. [Google Scholar] [CrossRef]
  8. Kahn, S.R.; de Wit, K. Pulmonary Embolism. N. Engl. J. Med. 2022, 387, 45–57. [Google Scholar] [CrossRef]
  9. Franchini, M.; Mannucci, P.M. Association between venous and arterial thrombosis: Clinical implications. Eur. J. Intern. Med. 2012, 23, 333–337. [Google Scholar] [CrossRef]
  10. Ageno, W. Arterial and Venous Thrombosis: Clinical Evidence for Mechanistic Overlap. Blood 2014, 124, SCI-3. [Google Scholar] [CrossRef]
  11. Delluc, A.; Lacut, K.; Rodger, M.A. Arterial and venous thrombosis: What’s the link? A narrative review. Thromb. Res. 2020, 191, 97–102. [Google Scholar] [CrossRef] [PubMed]
  12. Prandoni, P. Venous and arterial thrombosis: Two aspects of the same disease? Eur. J. Intern. Med. 2009, 20, 660–661. [Google Scholar] [CrossRef] [Green Version]
  13. Carminita, E.; Crescence, L.; Brouilly, N.; Altié, A.; Panicot-Dubois, L.; Dubois, C. DNAse-dependent, NET-independent pathway of thrombus formation in vivo. Proc. Natl. Acad. Sci. USA 2021, 118, e2100561118. [Google Scholar] [CrossRef] [PubMed]
  14. Martinod, K.; Deppermann, C. Immunothrombosis and thromboinflammation in host defense and disease. Platelets 2021, 32, 314–324. [Google Scholar] [CrossRef] [PubMed]
  15. Cognasse, F.; Duchez, A.C.; Audoux, E.; Ebermeyer, T.; Arthaud, C.A.; Prier, A.; Eyraud, M.A.; Mismetti, P.; Garraud, O.; Bertoletti, L.; et al. Platelets as Key Factors in Inflammation: Focus on CD40L/CD40. Front. Immunol. 2022, 13, 825892. [Google Scholar] [CrossRef] [PubMed]
  16. Cognasse, F.; Hamzeh-Cognasse, H.; Mismetti, P.; Tomas, T.; Eglin, D.; Marotte, H. The Non-Haemostatic Response of Platelets to Stress: An Actor of the Inflammatory Environment on Regenerative Medicine? Front. Immunol. 2021, 12, 741988. [Google Scholar] [CrossRef] [PubMed]
  17. Manne, B.K.; Xiang, S.C.; Rondina, M.T. Platelet secretion in inflammatory and infectious diseases. Platelets 2017, 28, 155–164. [Google Scholar] [CrossRef] [Green Version]
  18. Golebiewska, E.M.; Poole, A.W. Platelet secretion: From haemostasis to wound healing and beyond. Blood Rev. 2015, 29, 153–162. [Google Scholar] [CrossRef] [Green Version]
  19. Cognasse, F.; Garraud, O.; Pozzetto, B.; Laradi, S.; Hamzeh-Cognasse, H. How can non-nucleated platelets be so smart? J. Thromb. Haemost. 2016, 14, 794–796. [Google Scholar] [CrossRef] [Green Version]
  20. Cognasse, F.; Laradi, S.; Berthelot, P.; Bourlet, T.; Marotte, H.; Mismetti, P.; Garraud, O.; Hamzeh-Cognasse, H. Platelet Inflammatory Response to Stress. Front. Immunol. 2019, 10, 1478. [Google Scholar] [CrossRef]
  21. Shi, G.; Morrell, C.N. Platelets as initiators and mediators of inflammation at the vessel wall. Thromb. Res. 2011, 127, 387–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Koupenova, M.; Clancy, L.; Corkrey, H.A.; Freedman, J.E. Circulating Platelets as Mediators of Immunity, Inflammation, and Thrombosis. Circ. Res. 2018, 122, 337–351. [Google Scholar] [CrossRef] [PubMed]
  23. Yau, J.W.; Teoh, H.; Verma, S. Endothelial cell control of thrombosis. BMC Cardiovasc. Disord. 2015, 15, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Poredos, P.; Jezovnik, M.K. Endothelial Dysfunction and Venous Thrombosis. Angiology 2018, 69, 564–567. [Google Scholar] [CrossRef] [Green Version]
  25. Nightingale, T.; Cutler, D. The secretion of von W illebrand factor from endothelial cells; an increasingly complicated story. J. Thromb. Haemost. 2013, 11, 192–201. [Google Scholar] [CrossRef] [Green Version]
  26. Sadler, J.E. BIOCHEMISTRY AND GENETICS OF VON WILLEBRAND FACTOR. Annu. Rev. Biochem. 1998, 67, 395–424. [Google Scholar] [CrossRef]
  27. Franchini, M.; Mannucci, P. Past, present and future of hemophilia: A narrative review. Orphanet J. Rare Dis. 2012, 7, 24. [Google Scholar] [CrossRef] [Green Version]
  28. Denorme, F.; Vanhoorelbeke, K.; De Meyer, S.F. von Willebrand Factor and Platelet Glycoprotein Ib: A Thromboinflammatory Axis in Stroke. Front. Immunol. 2019, 10, 2884. [Google Scholar] [CrossRef]
  29. Brill, A.; Fuchs, T.A.; Chauhan, A.; Yang, J.J.; De Meyer, S.; Koellnberger, M.; Wakefield, T.W.; Lämmle, B.; Massberg, S.; Wagner, D.D. von Willebrand factor–mediated platelet adhesion is critical for deep vein thrombosis in mouse models. Blood 2011, 117, 1400–1407. [Google Scholar] [CrossRef] [Green Version]
  30. Lankhof, H.L.; van Hoeij, M.; E Schiphorst, M.; Bracke, M.; Wu, Y.-P.; Ijsseldijk, M.J.W.; Vink, T.; de Groot, P.G.; Sixma, J.J. A3 domain is essential for interaction of von Willebrand factor with collagen type III. Thromb. Haemost. 1996, 75, 950–958. [Google Scholar] [CrossRef]
  31. Flood, V.H.; Lederman, C.A.; Wren, J.S.; Christopherson, P.A.; Friedman, K.D.; Hoffmann, R.G.; Montgomery, R.R. Absent collagen binding in a VWF A3 domain mutant: Utility of the VWF:CB in diagnosis of VWD: Letters to the Editor. J. Thromb. Haemost. 2010, 8, 1431–1433. [Google Scholar] [CrossRef] [Green Version]
  32. Meng, D.; Luo, M.; Liu, B. The Role of CLEC-2 and Its Ligands in Thromboinflammation. Front. Immunol. 2021, 12, 688643. [Google Scholar] [CrossRef] [PubMed]
  33. Payne, H.; Ponomaryov, T.; Watson, S.P.; Brill, A. Mice with a deficiency in CLEC-2 are protected against deep vein thrombosis. Blood 2017, 129, 2013–2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Chandrakanthan, M.; Nguyen, T.Q.; Hasan, Z.; Muralidharan, S.; Vu, T.M.; Li, A.W.L.; Le, U.T.N.; Ha, H.T.T.; Baik, S.-H.; Tan, S.H.; et al. Deletion of Mfsd2b impairs thrombotic functions of platelets. Nat. Commun. 2021, 12, 2286. [Google Scholar] [CrossRef] [PubMed]
  35. Quintanilla, M.; Montero-Montero, L.; Renart, J.; Martín-Villar, E. Podoplanin in Inflammation and Cancer. Int. J. Mol. Sci. 2019, 20, 707. [Google Scholar] [CrossRef] [Green Version]
  36. Krishnan, H.; Rayes, J.; Miyashita, T.; Ishii, G.; Retzbach, E.P.; Sheehan, S.A.; Takemoto, A.; Chang, Y.; Yoneda, K.; Asai, J.; et al. Podoplanin: An emerging cancer biomarker and therapeutic target. Cancer Sci. 2018, 109, 1292–1299. [Google Scholar] [CrossRef] [Green Version]
  37. Tsukiji, N.; Osada, M.; Sasaki, T.; Shirai, T.; Satoh, K.; Inoue, O.; Umetani, N.; Mochizuki, C.; Saito, T.; Kojima, S.; et al. Cobalt hematoporphyrin inhibits CLEC-2–podoplanin interaction, tumor metastasis, and arterial/venous thrombosis in mice. Blood Adv. 2018, 2, 2214–2225. [Google Scholar] [CrossRef]
  38. Wang, X.; Liu, B.; Xu, M.; Jiang, Y.; Zhou, J.; Yang, J.; Gu, H.; Ruan, C.; Wu, J.; Zhao, Y. Blocking podoplanin inhibits platelet activation and decreases cancer-associated venous thrombosis. Thromb. Res. 2021, 200, 72–80. [Google Scholar] [CrossRef]
  39. Sasano, T.; Gonzalez-Delgado, R.; Muñoz, N.M.; Carlos-Alcade, W.; Cho, M.S.; Sheth, R.A.; Sood, A.K.; Afshar-Kharghan, V. Podoplanin promotes tumor growth, platelet aggregation, and venous thrombosis in murine models of ovarian cancer. J. Thromb. Haemost. 2022, 20, 104–114. [Google Scholar] [CrossRef]
  40. Nieswandt, B. Glycoprotein VI but not alpha2beta1 integrin is essential for platelet interaction with collagen. EMBO J. 2001, 20, 2120–2130. [Google Scholar] [CrossRef]
  41. Lockyer, S.; Okuyama, K.; Begum, S.; Le, S.; Sun, B.; Watanabe, T.; Matsumoto, Y.; Yoshitake, M.; Kambayashi, J.; Tandon, N.N. GPVI-deficient mice lack collagen responses and are protected against experimentally induced pulmonary thromboembolism. Thromb. Res. 2006, 118, 371–380. [Google Scholar] [CrossRef] [PubMed]
  42. Mammadova-Bach, E.; Ollivier, V.; Loyau, S.; Schaff, M.; Dumont, B.; Favier, R.; Freyburger, G.; Latger-Cannard, V.; Nieswandt, B.; Gachet, C.; et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood 2015, 126, 683–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ahmed, M.U.; Kaneva, V.; Loyau, S.; Nechipurenko, D.; Receveur, N.; Le Bris, M.; Janus-Bell, E.; Didelot, M.; Rauch, A.; Susen, S.; et al. Pharmacological Blockade of Glycoprotein VI Promotes Thrombus Disaggregation in the Absence of Thrombin. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 2127–2142. [Google Scholar] [CrossRef] [PubMed]
  44. Ahmed, M.U.; Receveur, N.; Janus-Bell, E.; Mouriaux, C.; Gachet, C.; Jandrot-Perrus, M.; Hechler, B.; Gardiner, E.E.; Mangin, P.H. Respective roles of Glycoprotein VI and FcγRIIA in the regulation of αIIbβ3-mediated platelet activation to fibrinogen, thrombus buildup, and stability. Res. Pract. Thromb. Haemost. 2021, 5, e12551. [Google Scholar] [CrossRef]
  45. Palacios-Acedo, A.L.; Mège, D.; Crescence, L.; Dignat-George, F.; Dubois, C.; Panicot-Dubois, L. Platelets, Thrombo-Inflammation, and Cancer: Collaborating with the Enemy. Front. Immunol. 2019, 10, 1805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Chen, R.; Zhang, X.; Gu, L.; Zhu, H.; Zhong, Y.; Ye, Y.; Xiong, X.; Jian, Z. New Insight Into Neutrophils: A Potential Therapeutic Target for Cerebral Ischemia. Front. Immunol. 2021, 12, 692061. [Google Scholar] [CrossRef]
  47. Liew, P.X.; Kubes, P. The Neutrophil’s Role During Health and Disease. Physiol. Rev. 2019, 99, 1223–1248. [Google Scholar] [CrossRef]
  48. von Brühl, M.-L.; Stark, K.; Steinhart, A.; Chandraratne, S.; Konrad, I.; Lorenz, M.; Khandoga, A.; Tirniceriu, A.; Coletti, R.; Köllnberger, M.; et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J. Exp. Med. 2012, 209, 819–835. [Google Scholar] [CrossRef]
  49. Swystun, L.L.; Liaw, P.C. The role of leukocytes in thrombosis. Blood 2016, 128, 753–762. [Google Scholar] [CrossRef] [Green Version]
  50. Kapoor, S.; Opneja, A.; Nayak, L. The role of neutrophils in thrombosis. Thromb. Res. 2018, 170, 87–96. [Google Scholar] [CrossRef]
  51. Fuchs, T.A.; Brill, A.; Duerschmied, D.; Schatzberg, D.; Monestier, M.; Myers, D.D., Jr.; 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] [Green Version]
  52. Martinod, K.; Wagner, D.D. Thrombosis: Tangled up in NETs. Blood 2014, 123, 2768–2776. [Google Scholar] [CrossRef]
  53. Dyer, M.R.; Chen, Q.; Haldeman, S.; Yazdani, H.; Hoffman, R.; Loughran, P.; Tsung, A.; Zuckerbraun, B.S.; Simmons, R.L.; Neal, M.D. Deep vein thrombosis in mice is regulated by platelet HMGB1 through release of neutrophil-extracellular traps and DNA. Sci. Rep. 2018, 8, 2068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kumar, R.; Sonkar, V.K.; Swamy, J.; Ahmed, A.; Sharathkumar, A.A.; Pierce, G.L.; Dayal, S. DNase 1 Protects from Increased Thrombin Generation and Venous Thrombosis during Aging: Cross-Sectional Study in Mice and Humans. J. Am. Heart Assoc. 2022, 11, e021188. [Google Scholar] [CrossRef]
  55. Wang, Y.; Gao, H.; Shi, C.; Erhardt, P.W.; Pavlovsky, A.; Soloviev, D.A.; Bledzka, K.; Ustinov, V.; Zhu, L.; Qin, J.; et al. Leukocyte integrin Mac-1 regulates thrombosis via interaction with platelet GPIbα. Nat. Commun. 2017, 8, 15559. [Google Scholar] [CrossRef] [Green Version]
  56. Germain, M.; Chasman, D.I.; de Haan, H.; Tang, W.; Lindström, S.; Weng, L.-C.; de Andrade, M.; de Visser, M.C.; Wiggins, K.L.; Suchon, P.; et al. Meta-analysis of 65,734 Individuals Identifies TSPAN15 and SLC44A2 as Two Susceptibility Loci for Venous Thromboembolism. Am. J. Hum. Genet. 2015, 96, 532–542. [Google Scholar] [CrossRef] [Green Version]
  57. Hinds, D.A.; Buil, A.; Ziemek, D.; Martinez-Perez, A.; Malik, R.; Folkersen, L.; Germain, M.; Mälarstig, A.; Brown, A.; Soria, J.M.; et al. Genome-wide association analysis of self-reported events in 6135 individuals and 252 827 controls identifies 8 loci associated with thrombosis. Hum. Mol. Genet. 2016, 25, 1867–1874. [Google Scholar] [CrossRef] [Green Version]
  58. Bennett, J.A.; Mastrangelo, M.A.; Ture, S.K.; Smith, C.O.; Loelius, S.G.; Berg, R.A.; Shi, X.; Burke, R.M.; Spinelli, S.L.; Cameron, S.J.; et al. The choline transporter Slc44a2 controls platelet activation and thrombosis by regulating mitochondrial function. Nat. Commun. 2020, 11, 3479. [Google Scholar] [CrossRef]
  59. Tilburg, J.; Coenen, D.M.; Zirka, G.; Dólleman, S.; Van Oeveren-Rietdijk, A.M.; Karel, M.F.A.; De Boer, H.C.; Cosemans, J.M.E.M.; Versteeg, H.H.; Morange, P.E.; et al. SLC44A2 deficient mice have a reduced response in stenosis but not in hypercoagulability driven venous thrombosis. J. Thromb. Haemost. 2020, 18, 1714–1727. [Google Scholar] [CrossRef]
  60. Zirka, G.; Robert, P.; Tilburg, J.; Tishkova, V.; Maracle, C.X.; Legendre, P.; van Vlijmen, B.; Alessi, M.C.; Lenting, P.J.; Morange, P.E.; et al. Impaired adhesion of neutrophils expressing Slc44a2/HNA-3b to VWF protects against NETosis under venous shear rates. Blood 2021, 137, 2256–2266. [Google Scholar] [CrossRef]
  61. Constantinescu-Bercu, A.; Grassi, L.; Frontini, M.; Salles-Crawley, I.I.; Woollard, K.; Crawley, J.T.B. Activated αIIbβ3 on platelets mediates flow-dependent NETosis via SLC44A2. eLife 2020, 9, e53353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Cerletti, C.; Evangelista, V.; de Gaetano, G. P-selectin-beta 2-integrin cross-talk: A molecular mechanism for polymorphonuclear leukocyte recruitment at the site of vascular damage. Thromb. Haemost. 1999, 82, 787–793. [Google Scholar]
  63. Kasthuri, R.S.; Glover, S.L.; Jonas, W.; McEachron, T.; Pawlinski, R.; Arepally, G.M.; Key, N.S.; Mackman, N. PF4/heparin-antibody complex induces monocyte tissue factor expression and release of tissue factor positive microparticles by activation of FcγRI. Blood 2012, 119, 5285–5293. [Google Scholar] [CrossRef] [PubMed]
  64. Ivanov, I.I.; Apta, B.H.R.; Bonna, A.M.; Harper, M.T. Platelet P-selectin triggers rapid surface exposure of tissue factor in monocytes. Sci. Rep. 2019, 9, 13397. [Google Scholar] [CrossRef] [Green Version]
  65. Etulain, J.; Martinod, K.; Wong, S.L.; Cifuni, S.M.; Schattner, M.; Wagner, D.D. P-selectin promotes neutrophil extracellular trap formation in mice. Blood 2015, 126, 242–246. [Google Scholar] [CrossRef] [Green Version]
  66. Wong, D.J.; Park, D.D.; Park, S.S.; Haller, C.A.; Chen, J.; Dai, E.; Liu, L.; Mandhapati, A.R.; Eradi, P.; Dhakal, B.; et al. A PSGL-1 glycomimetic reduces thrombus burden without affecting hemostasis. Blood 2021, 138, 1182–1193. [Google Scholar] [CrossRef]
  67. Momi, S.; Canino, J.; Vismara, M.; Galgano, L.; Falcinelli, E.; Guglielmini, G.; Taranta, G.C.; Guidetti, G.F.; Gresele, P.; Torti, M.; et al. Proline-rich tyrosine kinase Pyk2 regulates deep vein thrombosis. Haematologica 2022, 107, 1374–1383. [Google Scholar] [CrossRef]
  68. Fuchs, T.A.; Abed, U.; Goosmann, C.; Hurwitz, R.; Schulze, I.; Wahn, V.; Weinrauch, Y.; Brinkmann, V.; Zychlinsky, A. Novel cell death program leads to neutrophil extracellular traps. J. Cell Biol. 2007, 176, 231–241. [Google Scholar] [CrossRef]
  69. Jin, R.; Yu, S.; Song, Z.; Zhu, X.; Wang, C.; Yan, J.; Wu, F.; Nanda, A.; Granger, D.N.; Li, G. Soluble CD40 Ligand Stimulates CD40-Dependent Activation of the β2 Integrin Mac-1 and Protein Kinase C Zeda (PKCζ) in Neutrophils: Implications for Neutrophil-Platelet Interactions and Neutrophil Oxidative Burst. PLoS ONE 2013, 8, e64631. [Google Scholar] [CrossRef] [Green Version]
  70. Canobbio, I.; Visconte, C.; Momi, S.; Guidetti, G.F.; Zarà, M.; Canino, J.; Falcinelli, E.; Gresele, P.; Torti, M. Platelet amyloid precursor protein is a modulator of venous thromboembolism in mice. Blood 2017, 130, 527–536. [Google Scholar] [CrossRef] [Green Version]
  71. Opneja, A.; Kapoor, S.; Stavrou, E.X. Contribution of platelets, the coagulation and fibrinolytic systems to cutaneous wound healing. Thromb. Res. 2019, 179, 56–63. [Google Scholar] [CrossRef]
  72. Williams, C.M.; Li, Y.; Brown, E.; Poole, A.W. Platelet-specific deletion of SNAP23 ablates granule secretion, substantially inhibiting arterial and venous thrombosis in mice. Blood Adv. 2018, 2, 3627–3636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Furie, B.; Furie, B.C.; Flaumenhaft, R. A journey with platelet P-selectin: The molecular basis of granule secretion, signalling and cell adhesion. Thromb. Haemost. 2001, 86, 214–221. [Google Scholar] [CrossRef]
  74. Ay, C.; Jungbauer, L.V.; Sailer, T.; Tengler, T.; Koder, S.; Kaider, A.; Panzer, S.; Quehenberger, P.; Pabinger, I.; Mannhalter, C. High concentrations of soluble P-selectin are associated with risk of venous thromboembolism and the P-selectin Thr715 variant. Clin. Chem. 2007, 53, 1235–1243. [Google Scholar] [CrossRef] [Green Version]
  75. Ramacciotti, E.; Blackburn, S.; Hawley, A.E.; Vandy, F.; Ballard-Lipka, N.; Stabler, C.; Baker, N.; Guire, K.E.; Rectenwald, J.E.; Henke, P.K.; et al. Evaluation of soluble P-selectin as a marker for the diagnosis of deep venous thrombosis. Clin. Appl. Thromb. Off. J. Int. Acad. Clin. Appl. Thromb. 2011, 17, 425–431. [Google Scholar] [CrossRef] [Green Version]
  76. Antonopoulos, C.N.; Sfyroeras, G.S.; Kakisis, J.D.; Moulakakis, K.G.; Liapis, C.D. The role of soluble P selectin in the diagnosis of venous thromboembolism. Thromb. Res. 2014, 133, 17–24. [Google Scholar] [CrossRef]
  77. Furio, E.; García-Fuster, M.J.; Redon, J.; Marques, P.; Ortega, R.; Sanz, M.J.; Piqueras, L. CX3CR1/CX3CL1 Axis Mediates Platelet–Leukocyte Adhesion to Arterial Endothelium in Younger Patients with a History of Idiopathic Deep Vein Thrombosis. Thromb. Haemost. 2018, 118, 562–571. [Google Scholar] [CrossRef] [Green Version]
  78. Poruk, K.E.; Firpo, M.A.; Huerter, L.M.; Scaife, C.L.; Emerson, L.L.; Boucher, K.M.; Jones, K.A.; Mulvihill, S.J. Serum Platelet Factor 4 Is an Independent Predictor of Survival and Venous Thromboembolism in Patients with Pancreatic Adenocarcinoma. Cancer Epidemiol. Biomark. Prev. 2010, 19, 2605–2610. [Google Scholar] [CrossRef] [Green Version]
  79. Riedl, J.; Hell, L.; Kaider, A.; Koder, S.; Marosi, C.; Zielinski, C.; Pamzer, S.; Pabinger, I.; Ay, C. Association of platelet activation markers with cancer-associated venous thromboembolism. Platelets 2016, 27, 80–85. [Google Scholar] [CrossRef]
  80. Wang, Y.; Gao, H.; Kessinger, C.W.; Schmaier, A.; Jaffer, F.A.; Simon, D.I. Myeloid-related protein-14 regulates deep vein thrombosis. JCI Insight 2017, 2, e91356. [Google Scholar] [CrossRef] [Green Version]
  81. Yun, S.-H.; Sim, E.-H.; Goh, R.-Y.; Park, J.-I.; Han, J.-Y. Platelet Activation: The Mechanisms and Potential Biomarkers. BioMed Res. Int. 2016, 2016, 9060143. [Google Scholar] [CrossRef]
  82. Cognasse, F.; Hamzeh, H.; Chavarin, P.; Acquart, S.; Genin, C.; Garraud, O. Evidence of Toll-like receptor molecules on human platelets. Immunol. Cell Biol. 2005, 83, 196–198. [Google Scholar] [CrossRef]
  83. Beutler, B.A. TLRs and innate immunity. Blood 2009, 113, 1399–1407. [Google Scholar] [CrossRef] [Green Version]
  84. Blasius, A.L.; Beutler, B. Intracellular Toll-like Receptors. Immunity 2010, 32, 305–315. [Google Scholar] [CrossRef] [Green Version]
  85. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef]
  86. Ebermeyer, T.; Cognasse, F.; Berthelot, P.; Mismetti, P.; Garraud, O.; Hamzeh-Cognasse, H. Platelet Innate Immune Receptors and TLRs: A Double-Edged Sword. Int. J. Mol. Sci. 2021, 22, 7894. [Google Scholar] [CrossRef] [PubMed]
  87. 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]
  88. Moser, M.; Nieswandt, B.; Ussar, S.; Pozgajova, M.; Fässler, R. Kindlin-3 is essential for integrin activation and platelet aggregation. Nat. Med. 2008, 14, 325–330. [Google Scholar] [CrossRef] [PubMed]
  89. Yan, Y.; Yang, H.; Hu, X.; Zhang, Z.; Ge, S.; Xu, Z.; Gao, J.; Liu, J.; White, G.C.; Ma, Y.-Q. Kindlin-3 in platelets and myeloid cells differentially regulates deep vein thrombosis in mice. Aging 2019, 11, 6951–6959. [Google Scholar] [CrossRef] [PubMed]
  90. Nguyen, H.T.T.; Xu, Z.; Shi, X.; Liu, S.; Schulte, M.L.; White, G.C.; Ma, Y.-Q. Paxillin binding to the PH domain of kindlin-3 in platelets is required to support integrin αIIbβ3 outside-in signaling. J. Thromb. Haemost. 2021, 19, 3126–3138. [Google Scholar] [CrossRef]
  91. Larsen, J.B.; Hvas, A.-M. Thrombin: A Pivotal Player in Hemostasis and Beyond. Semin. Thromb. Hemost. 2021, 47, 759–774. [Google Scholar] [CrossRef] [PubMed]
  92. Andersen, H.; Greenberg, D.L.; Fujikawa, K.; Xu, W.; Chung, D.W.; Davie, E.W. Protease-activated receptor 1 is the primary mediator of thrombin-stimulated platelet procoagulant activity. Proc. Natl. Acad. Sci. USA 1999, 96, 11189–11193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lee, R.H.; Kawano, T.; Grover, S.P.; Bharathi, V.; Martinez, D.; Cowley, D.O.; Mackman, N.; Bergmeier, W.; Antoniak, S. Genetic deletion of platelet PAR4 results in reduced thrombosis and impaired hemostatic plug stability. J. Thromb. Haemost. 2022, 20, 422–433. [Google Scholar] [CrossRef] [PubMed]
  94. Gushiken, F.C.; Han, H.; Li, J.; Rumbaut, R.E.; Afshar-Kharghan, V. Abnormal platelet function in C3-deficient mice. J. Thromb. Haemost. 2009, 7, 865–870. [Google Scholar] [CrossRef] [Green Version]
  95. Subramaniam, S.; Jurk, K.; Hobohm, L.; Jäckel, S.; Saffarzadeh, M.; Schwierczek, K.; Wenzel, P.; Langer, F.; Reinhardt, C.; Ruf, W. Distinct contributions of complement factors to platelet activation and fibrin formation in venous thrombus development. Blood 2017, 129, 2291–2302. [Google Scholar] [CrossRef] [Green Version]
  96. Feng, Y.; Yu, M.; Zhu, F.; Zhang, S.; Ding, P.; Wang, M. IL-9 Promotes the Development of Deep Venous Thrombosis by Facilitating Platelet Function. Thromb. Haemost. 2018, 118, 1885–1894. [Google Scholar] [CrossRef] [Green Version]
  97. Ding, P.; Zhang, S.; Yu, M.; Feng, Y.; Long, Q.; Yang, H.; Li, J.; Wang, M. IL-17A promotes the formation of deep vein thrombosis in a mouse model. Int. Immunopharmacol. 2018, 57, 132–138. [Google Scholar] [CrossRef]
  98. Valéra, M.-C.; Gratacap, M.-P.; Gourdy, P.; Lenfant, F.; Cabou, C.; Toutain, C.E.; Marcellin, M.; Laurent, N.S.; Sié, P.; Sixou, M.; et al. Chronic estradiol treatment reduces platelet responses and protects mice from thromboembolism through the hematopoietic estrogen receptor α. Blood 2012, 120, 1703–1712. [Google Scholar] [CrossRef] [Green Version]
  99. Valera, M.-C.; Noirrit-Esclassan, E.; Dupuis, M.; Buscato, M.; Vinel, A.; Guillaume, M.; Briaux, A.; Garcia, C.; Benoit, T.; Lairez, O.; et al. Effect of chronic estradiol plus progesterone treatment on experimental arterial and venous thrombosis in mouse. PLoS ONE 2017, 12, e0177043. [Google Scholar] [CrossRef] [PubMed]
  100. Law, L.A.; Graham, D.K.; Di Paola, J.; Branchford, B.R. GAS6/TAM Pathway Signaling in Hemostasis and Thrombosis. Front. Med. 2018, 5, 137. [Google Scholar] [CrossRef] [Green Version]
  101. Angelillo-Scherrer, A.; De Frutos, P.G.; Aparicio, C.; Melis, E.; Savi, P.; Lupu, F.; Arnout, J.; Dewerchin, M.; Hoylaerts, M.F.; Herbert, J.-M.; et al. Deficiency or inhibition of Gas6 causes platelet dysfunction and protects mice against thrombosis. Nat. Med. 2001, 7, 215–221. [Google Scholar] [CrossRef]
  102. Angelillo-Scherrer, A.; Burnier, L.; Flores, N.; Savi, P.; DeMol, M.; Schaeffer, P.; Herbert, J.-M.; Lemke, G.; Goff, S.P.; Matsushima, G.K.; et al. Role of Gas6 receptors in platelet signaling during thrombus stabilization and implications for antithrombotic therapy. J. Clin. Investig. 2005, 115, 237–246. [Google Scholar] [CrossRef] [Green Version]
  103. Gutmann, C.; Siow, R.; Gwozdz, A.M.; Saha, P.; Smith, A. Reactive Oxygen Species in Venous Thrombosis. Int. J. Mol. Sci. 2020, 21, 1918. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Heemskerk, J.W.M.; Mattheij, N.J.A.; Cosemans, J.M.E.M. Platelet-based coagulation: Different populations, different functions: Platelet-based coagulation. J. Thromb. Haemost. 2013, 11, 2–16. [Google Scholar] [CrossRef] [PubMed]
  105. Sang, Y.; Roest, M.; de Laat, B.; de Groot, P.G.; Huskens, D. Interplay between platelets and coagulation. Blood Rev. 2021, 46, 100733. [Google Scholar] [CrossRef]
  106. Bevers, E.M.; Comfurius, P.; van Rijn, J.L.; Hemker, H.C.; Zwaal, R.F. Generation of prothrombin-converting activity and the exposure of phosphatidylserine at the outer surface of platelets. Eur. J. Biochem. 1982, 122, 429–436. [Google Scholar] [CrossRef] [PubMed]
  107. Lentz, B.R. Exposure of platelet membrane phosphatidylserine regulates blood coagulation. Prog. Lipid Res. 2003, 42, 423–438. [Google Scholar] [CrossRef]
  108. Reddy, E.C.; Rand, M.L. Procoagulant Phosphatidylserine-Exposing Platelets in vitro and in vivo. Front. Cardiovasc. Med. 2020, 7, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Heestermans, M.; Salloum-Asfar, S.; Streef, T.; Laghmani, E.H.; Salvatori, D.; Luken, B.M.; Zeerleder, S.S.; Spronk, H.M.H.; Korporaal, S.J.; Kirchhofer, D.; et al. Mouse venous thrombosis upon silencing of anticoagulants depends on tissue factor and platelets, not FXII or neutrophils. Blood 2019, 133, 2090–2099. [Google Scholar] [CrossRef]
  110. Sunnerhagen, M.; Drakenberg, T.; Forsén, S.; Stenflo, J. Effect of Ca2+ on the Structure of Vitamin K-Dependent Coagulation Factors. Pathophysiol. Haemost. Thromb. 1996, 26, 45–53. [Google Scholar] [CrossRef]
  111. Hur, W.S.; Paul, D.S.; Bouck, E.G.; Negrón, O.A.; Mwiza, J.M.N.; Poole, L.G.; Cline-Fedewa, H.M.; Clark, E.G.; Juang, L.J.; Leung, J.; et al. Hypofibrinogenemia with preserved hemostasis and protection from thrombosis in mice with an Fga truncation mutation. Blood 2022, 139, 1374–1388. [Google Scholar] [CrossRef] [PubMed]
  112. Chesney, C.M.; Pifer, D.; Colman, R.W. Subcellular localization and secretion of factor V from human platelets. Proc. Natl. Acad. Sci. USA 1981, 78, 5180–5184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Monković, D.D.; Tracy, P.B. Functional characterization of human platelet-released factor V and its activation by factor Xa and thrombin. J. Biol. Chem. 1990, 265, 17132–17140. [Google Scholar] [CrossRef]
  114. Bouma, B.N.; de Graaf, S.; Slot, J.W.; Zimmerman, T.S. Human blood platelet factor VIII-related antigen: Demonstration of release by α-chymotrypsin. Thromb. Res. 1979, 14, 687–696. [Google Scholar] [CrossRef]
  115. Yarovoi, H.V.; Kufrin, D.; Eslin, D.E.; Thornton, M.A.; Haberichter, S.L.; Shi, Q.; Zhu, H.; Camire, R.; Fakharzadeh, S.S.; Kowalska, M.A.; et al. Factor VIII ectopically expressed in platelets: Efficacy in hemophilia A treatment. Blood 2003, 102, 4006–4013. [Google Scholar] [CrossRef]
  116. Zhang, G.; Shi, Q.; Fahs, S.A.; Kuether, E.L.; Walsh, C.E.; Montgomery, R.R. Factor IX ectopically expressed in platelets can be stored in α-granules and corrects the phenotype of hemophilia B mice. Blood 2010, 116, 1235–1243. [Google Scholar] [CrossRef] [Green Version]
  117. Mitchell, J.L.; Lionikiene, A.S.; Fraser, S.R.; Whyte, C.S.; Booth, N.A.; Mutch, N.J. Functional factor XIII-A is exposed on the stimulated platelet surface. Blood 2014, 124, 3982–3990. [Google Scholar] [CrossRef] [Green Version]
  118. Somodi, L.; Debreceni, I.B.; Kis, G.; Cozzolino, M.; Kappelmayer, J.; Antal, M.; Panyi, G.; Bárdos, H.; Mutch, N.J.; Muszbek, L. Activation mechanism dependent surface exposure of cellular factor XIII on activated platelets and platelet microparticles. J. Thromb. Haemost. 2022, 20, 1223–1235. [Google Scholar] [CrossRef]
  119. Gralnick, H.R.; Williams, S.B.; MaKeown, L.P.; Magruder, L.; Hansmann, K.; Vail, M.; Parker, R.I. Platelet von Willebrand Factor. Mayo Clin. Proc. 1991, 66, 634–640. [Google Scholar] [CrossRef]
  120. Stavenuiter, F.; Davis, N.; Duan, E.; Gale, A.; Heeb, M. Platelet protein S directly inhibits procoagulant activity on platelets and microparticles. Thromb. Haemost. 2013, 109, 229–237. [Google Scholar] [CrossRef] [Green Version]
  121. Baj-Krzyworzeka, M.; Majka, M.; Pratico, D.; Ratajczak, J.; Vilaire, G.; Kijowski, J.; Reca, R.; Janowska-Wieczorek, A.; Ratajczak, M.Z. Platelet-derived microparticles stimulate proliferation, survival, adhesion, and chemotaxis of hematopoietic cells. Exp. Hematol. 2002, 30, 450–459. [Google Scholar] [CrossRef]
  122. Rank, A.; Nieuwland, R.; Delker, R.; Köhler, A.; Toth, B.; Pihusch, V.; Wilkowski, R.; Pihusch, R. Cellular origin of platelet-derived microparticles in vivo. Thromb. Res. 2010, 126, e255–e259. [Google Scholar] [CrossRef]
  123. Melki, I.; Tessandier, N.; Zufferey, A.; Boilard, E. Platelet microvesicles in health and disease. Platelets 2017, 28, 214–221. [Google Scholar] [CrossRef] [PubMed]
  124. Puhm, F.; Boilard, E.; Machlus, K.R. Platelet Extracellular Vesicles: Beyond the Blood. Arterioscler. Thromb. Vasc. Biol. 2020, 41, 87–96. [Google Scholar] [CrossRef]
  125. Dyer, M.R.; Alexander, W.; Hassoune, A.; Chen, Q.; Brzoska, T.; Alvikas, J.; Liu, Y.; Haldeman, S.; Plautz, W.; Loughran, P.; et al. Platelet-derived extracellular vesicles released after trauma promote hemostasis and contribute to DVT in mice. J. Thromb. Haemost. 2019, 17, 1733–1745. [Google Scholar] [CrossRef] [PubMed]
  126. Obermayer, G.; Afonyushkin, T.; Goederle, L.; Puhm, F.; Schrottmaier, W.C.; Taqi, S.; Schwameis, M.; Ay, C.; Pabinger, I.; Jilma, B.; et al. Natural IgM antibodies inhibit microvesicle-driven coagulation and thrombosis. Blood 2021, 137, 1406–1415. [Google Scholar] [CrossRef] [PubMed]
  127. Rectenwald, J.E.; Myers, D.D., Jr.; Hawley, A.E.; Longo, C.; Hemke, P.K.; Guire, K.E.; Schmaier, A.H.; Wakefield, T.W. D-dimer, P-selectin, and microparticles: Novel markers to predict deep venous thrombosis: A pilot study. Thromb. Haemost. 2005, 94, 1312–1317. [Google Scholar] [CrossRef]
  128. Sahu, A.; Jha, P.K.; Prabhakar, A.; Singh, H.D.; Gupta, N.; Chatterjee, T.; Tyagi, T.; Sharma, S.; Kumari, B.; Singh, S.; et al. MicroRNA-145 Impedes Thrombus Formation via Targeting Tissue Factor in Venous Thrombosis. EBioMedicine 2017, 26, 175–186. [Google Scholar] [CrossRef] [Green Version]
  129. Boilard, E.; Duchez, A.-C.; Brisson, A. The diversity of platelet microparticles. Curr. Opin. Hematol. 2015, 22, 437–444. [Google Scholar] [CrossRef]
  130. Müller, F.; Mutch, N.J.; Schenk, W.A.; Smith, S.A.; Esterl, L.; Spronk, H.M.; Schmidbauer, S.; Gahl, W.A.; Morrissey, J.H.; Renné, T. Platelet Polyphosphates Are Proinflammatory and Procoagulant Mediators In Vivo. Cell 2009, 139, 1143–1156. [Google Scholar] [CrossRef] [Green Version]
  131. Gajsiewicz, J.M.; Smith, S.A.; Morrissey, J.H. Polyphosphate and RNA Differentially Modulate the Contact Pathway of Blood Clotting. J. Biol. Chem. 2017, 292, 1808–1814. [Google Scholar] [CrossRef] [PubMed]
  132. Verhoef, J.J.F.; Barendrecht, A.D.; Nickel, K.F.; Dijkxhoorn, K.; Kenne, E.; Labberton, L.; Mccarty, O.J.T.; Schiffelers, R.; Heijnen, H.F.; Hendrickx, A.P.; et al. Polyphosphate nanoparticles on the platelet surface trigger contact system activation. Blood 2017, 129, 1707–1717. [Google Scholar] [CrossRef] [PubMed]
  133. Labberton, L.; Kenne, E.; Long, A.T.; Nickel, K.F.; Di Gennaro, A.; Rigg, R.A.; Hernandez, J.S.; Butler, L.; Maas, C.; Stavrou, E.; et al. Neutralizing blood-borne polyphosphate in vivo provides safe thromboprotection. Nat. Commun. 2016, 7, 12616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Choi, S.H.; Smith, S.A.; Morrissey, J.H. Polyphosphate is a cofactor for the activation of factor XI by thrombin. Blood 2011, 118, 6963–6970. [Google Scholar] [CrossRef] [Green Version]
  135. Choi, S.; Smith, S.; Morrissey, J. Polyphosphate accelerates factor V activation by factor XIa. Thromb. Haemost. 2015, 113, 599–604. [Google Scholar] [CrossRef] [Green Version]
  136. Whyte, C.S.; Chernysh, I.N.; Domingues, M.M.; Connell, S.; Weisel, J.W.; Ariens, R.A.S.; Mutch, N.J. Polyphosphate delays fibrin polymerisation and alters the mechanical properties of the fibrin network. Thromb. Haemost. 2016, 116, 897–903. [Google Scholar] [CrossRef] [Green Version]
  137. Chapin, J.C.; Hajjar, K.A. Fibrinolysis and the control of blood coagulation. Blood Rev. 2015, 29, 17–24. [Google Scholar] [CrossRef] [Green Version]
  138. Brogren, H.; Karlsson, L.; Andersson, M.; Wang, L.; Erlinge, D.; Jern, S. Platelets synthesize large amounts of active plasminogen activator inhibitor 1. Blood 2004, 104, 3943–3948. [Google Scholar] [CrossRef]
  139. Whyte, C.; Mitchell, J.; Mutch, N. Platelet-Mediated Modulation of Fibrinolysis. Semin. Thromb. Hemost. 2017, 43, 115–128. [Google Scholar] [CrossRef] [Green Version]
  140. Brogren, H.; Wallmark, K.; Deinum, J.; Karlsson, L.; Jern, S. Platelets Retain High Levels of Active Plasminogen Activator Inhibitor 1. PLoS ONE 2011, 6, e26762. [Google Scholar] [CrossRef]
  141. Mosnier, L.O.; Buijtenhuijs, P.; Marx, P.F.; Meijers, J.C.M.; Bouma, B.N. Identification of thrombin activatable fibrinolysis inhibitor (TAFI) in human platelets. Blood 2003, 101, 4844–4846. [Google Scholar] [CrossRef] [PubMed]
  142. DeRoo, E.; Martinod, K.; Cherpokova, D.; Fuchs, T.; Cifuni, S.; Chu, L.; Staudinger, C.; Wagner, D.D. The role of platelets in thrombus fibrosis and vessel wall remodeling after venous thrombosis. J. Thromb. Haemost. 2021, 19, 387–399. [Google Scholar] [CrossRef] [PubMed]
  143. Bonderman, D.; Jakowitsch, J.; Redwan, B.; Bergmeister, H.; Renner, M.-K.; Panzenböck, H.; Adlbrecht, C.; Georgopoulos, A.; Klepetko, W.; Kneussl, M.; et al. Role for Staphylococci in Misguided Thrombus Resolution of Chronic Thromboembolic Pulmonary Hypertension. Arterioscler. Thromb. Vasc. Biol. 2008, 28, 678–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kessinger, C.W.; Kim, J.W.; Henke, P.K.; Thompson, B.; McCarthy, J.R.; Hara, T.; Sillesen, M.; Margey, R.J.P.; Libby, P.; Weissleder, R.; et al. Statins Improve the Resolution of Established Murine Venous Thrombosis: Reductions in Thrombus Burden and Vein Wall Scarring. PLoS ONE 2015, 10, e0116621. [Google Scholar] [CrossRef] [Green Version]
  145. Violi, F.; Calvieri, C.; Ferro, D.; Pignatelli, P. Statins as Antithrombotic Drugs. Circulation 2013, 127, 251–257. [Google Scholar] [CrossRef] [Green Version]
  146. Xie, Y.; Muller, W.A. Molecular cloning and adhesive properties of murine platelet/endothelial cell adhesion molecule 1. Proc. Natl. Acad. Sci. USA 1993, 90, 5569–5573. [Google Scholar] [CrossRef] [Green Version]
  147. Kellermair, J.; Redwan, B.; Alias, S.; Jabkowski, J.; Panzenboeck, A.; Kellermair, L.; Winter, M.P.; Weltermann, A.; Lang, I.M. Platelet endothelial cell adhesion molecule 1 deficiency misguides venous thrombus resolution. Blood 2013, 122, 3376–3384. [Google Scholar] [CrossRef] [Green Version]
  148. McFadyen, J.D.; Schaff, M.; Peter, K. Current and future antiplatelet therapies: Emphasis on preserving haemostasis. Nat. Rev. Cardiol. 2018, 15, 181–191. [Google Scholar] [CrossRef]
  149. Vane, J.R.; Botting, R.M. The mechanism of action of aspirin. Thromb. Res. 2003, 110, 255–258. [Google Scholar] [CrossRef]
  150. Dorsam, R.T.; Kunapuli, S.P. Central role of the P2Y12 receptor in platelet activation. J. Clin. Investig. 2004, 113, 340–345. [Google Scholar] [CrossRef]
  151. Marciniak, S.; Furman, M.I.; Michelson, A.D.; Jakubowski, J.A.; Jordan, R.E.; Marchese, P.J.; Frelinger, A.L.; Mascelli, M.A. An additional mechanism of action of abciximab: Dispersal of newly formed platelet aggregates. Thromb. Haemost. 2002, 87, 1020–1025. [Google Scholar] [CrossRef] [PubMed]
  152. Ibbotson, T.; McGavin, J.K.; Goa, K.L. Abciximab: An Updated Review of its Therapeutic Use in Patients with Ischaemic Heart Disease Undergoing Percutaneous Coronary Revascularisation. Drugs 2003, 63, 1121–1163. [Google Scholar] [CrossRef] [PubMed]
  153. Imbault, P.; Doutremepuich, F.; Aguejouf, O.; Doutremepuich, C. Antithrombotic effects of aspirin and LMWH in a laser-induced model of arterials and venous thrombosis. Thromb. Res. 1996, 82, 469–478. [Google Scholar] [CrossRef]
  154. Tarantino, E.; Amadio, P.; Squellerio, I.; Porro, B.; Sandrini, L.; Turnu, L.; Cavalca, V.; Tremoli, E.; Barbieri, S.S. Role of thromboxane-dependent platelet activation in venous thrombosis: Aspirin effects in mouse model. Pharmacol. Res. 2016, 107, 415–425. [Google Scholar] [CrossRef] [PubMed]
  155. Herbert, J.; Bernat, A.; Maffrand, J. Importance of platelets in experimental venous thrombosis in the rat. Blood 1992, 80, 2281–2286. [Google Scholar] [CrossRef] [Green Version]
  156. Herbert, J.-M.; Bernat, A.; Maffrand, J.-P. Aprotinin reduces clopidogrel-induced prolongation of the bleeding time in the rat. Thromb. Res. 1993, 71, 433–441. [Google Scholar] [CrossRef]
  157. Wang, Y.-X.; Vincelette, J.; da Cunha, V.; Martin-McNulty, B.; Mallari, C.; Fitch, R.M.; Alexander, S.; Islam, I.; Buckman, B.O.; Yuan, S.; et al. A novel P2Y(12) adenosine diphosphate receptor antagonist that inhibits platelet aggregation and thrombus formation in rat and dog models. Thromb. Haemost. 2007, 97, 847–855. [Google Scholar]
  158. Hérault, J.P.; Dol, F.; Gaich, C.; Bernat, A.; Herbert, J.M. Effect of clopidogrel on thrombin generation in platelet-rich plasma in the rat. Thromb. Haemost. 1999, 81, 957–960. [Google Scholar] [CrossRef]
  159. Guenther, F.; Herr, N.; Mauler, M.; Witsch, T.; Roming, F.; Hein, L.; Boeynaems, J.-M.; Robaye, B.; Idzko, M.; Bode, C.; et al. Contrast ultrasound for the quantification of deep vein thrombosis in living mice: Effects of enoxaparin and P2Y 12 receptor inhibition. J. Thromb. Haemost. 2013, 11, 1154–1162. [Google Scholar] [CrossRef]
  160. Bird, J.E.; Wang, X.; Smith, P.L.; Barbera, F.; Huang, C.; Schumacher, W.A. A platelet target for venous thrombosis? P2Y1 deletion or antagonism protects mice from vena cava thrombosis. J. Thromb. Thrombolysis 2012, 34, 199–207. [Google Scholar] [CrossRef]
  161. Cooley, B.C.; Herrera, A.J. Cross-modulatory effects of clopidogrel and heparin on platelet and fibrin incorporation in thrombosis. Blood Coagul. Fibrinolysis 2013, 24, 593–598. [Google Scholar] [CrossRef] [PubMed]
  162. Mwiza, J.M.N.; Lee, R.H.; Paul, D.S.; Holle, L.A.; Cooley, B.C.; Nieswandt, B.; Schug, W.J.; Kawano, T.; Mackman, N.; Wolberg, A.S.; et al. Both G protein–coupled and immunoreceptor tyrosine-based activation motif receptors mediate venous thrombosis in mice. Blood 2022, 139, 3194–3203. [Google Scholar] [CrossRef] [PubMed]
  163. Branchford, B.R.; Stalker, T.J.; Law, L.; Acevedo, G.; Sather, S.; Brzezinski, C.; Wilson, K.M.; Minson, K.; Lee-Sherick, A.B.; Davizon-Castillo, P.; et al. The small-molecule MERTK inhibitor UNC2025 decreases platelet activation and prevents thrombosis. J. Thromb. Haemost. 2018, 16, 352–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Anderson, D.R.; Dunbar, M.; Murnaghan, J.; Kahn, S.R.; Gross, P.; Forsythe, M.; Pelet, S.; Fisher, W.; Belzile, E.; Dolan, S.; et al. Aspirin or Rivaroxaban for VTE Prophylaxis after Hip or Knee Arthroplasty. N. Engl. J. Med. 2018, 378, 699–707. [Google Scholar] [CrossRef]
  165. CRISTAL Study Group. Effect of Aspirin vs Enoxaparin on Symptomatic Venous Thromboembolism in Patients Undergoing Hip or Knee Arthroplasty: The CRISTAL Randomized Trial. JAMA 2022, 328, 719. [Google Scholar] [CrossRef]
  166. Becattini, C.; Agnelli, G.; Schenone, A.; Eichinger, S.; Bucherini, E.; Silingardi, M.; Bianchi, M.; Moia, M.; Ageno, W.; Vandelli, M.R.; et al. Aspirin for Preventing the Recurrence of Venous Thromboembolism. N. Engl. J. Med. 2012, 366, 1959–1967. [Google Scholar] [CrossRef] [Green Version]
  167. Brighton, T.A.; Eikelboom, J.W.; Mann, K.; Mister, R.; Gallus, A.; Ockelford, P.; Gibbs, H.; Hague, W.; Xavier, D.; Diaz, R.; et al. Low-Dose Aspirin for Preventing Recurrent Venous Thromboembolism. N. Engl. J. Med. 2012, 367, 1979–1987. [Google Scholar] [CrossRef] [Green Version]
  168. Weitz, J.I.; Lensing, A.W.; Prins, M.H.; Bauersachs, R.; Beyer-Westendorf, J.; Bounameaux, H.; Brighton, T.A.; Cohen, A.T.; Davidson, B.L.; Decousus, H.; et al. Rivaroxaban or Aspirin for Extended Treatment of Venous Thromboembolism. N. Engl. J. Med. 2017, 376, 1211–1222. [Google Scholar] [CrossRef]
  169. Mai, V.; Bertoletti, L.; Cucherat, M.; Jardel, S.; Grange, C.; Provencher, S.; Lega, J.-C. Extended anticoagulation for the secondary prevention of venous thromboembolic events: An updated network meta-analysis. PLoS ONE 2019, 14, e0214134. [Google Scholar] [CrossRef] [Green Version]
  170. Jourdi, G.; Bachelot-Loza, C.; Mazoyer, E.; Poirault-Chassac, S.; Duchemin, J.; Fontenay, M.; Gaussem, P. Effect of rivaroxaban and dabigatran on platelet functions: In vitro study. Thromb. Res. 2019, 183, 159–162. [Google Scholar] [CrossRef]
  171. Hernandez-Juarez, J.; Espejo-Godinez, H.G.; Mancilla-Padilla, R.; Hernandez-Lopez, J.R.; Moreno, J.A.A.; Majluf-Cruz, K.; Moreno-Hernández, M.; Isordia-Salas, I.; Majluf-Cruz, A. Effects of Rivaroxaban on Platelet Aggregation. J. Cardiovasc. Pharmacol. 2020, 75, 180–184. [Google Scholar] [CrossRef] [PubMed]
  172. Trabold, K.; Makhoul, S.; Gambaryan, S.; van Ryn, J.; Walter, U.; Jurk, K. The Direct Thrombin Inhibitors Dabigatran and Lepirudin Inhibit GPIbα-Mediated Platelet Aggregation. Thromb. Haemost. 2019, 119, 916–929. [Google Scholar] [PubMed]
  173. Kubisz, P.; Stanciakova, L.; Dobrotova, M.; Samos, M.; Mokan, M.; Stasko, J. Apixaban—Metabolism, Pharmacologic Properties and Drug Interactions. Curr. Drug Metab. 2017, 18, 609–621. [Google Scholar] [PubMed]
  174. Honda, Y.; Kamisato, C.; Morishima, Y. Edoxaban, a direct factor Xa inhibitor, suppresses tissue-factor induced human platelet aggregation and clot-bound factor Xa in vitro: Comparison with an antithrombin-dependent factor Xa inhibitor, fondaparinux. Thromb. Res. 2016, 141, 17–21. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The role of platelets in different aspects of venous thrombosis. Platelets are crucially involved in the pathophysiology of venous thrombosis. They can be recruited to the vessel wall and can interact with neutrophils and monocytes. In addition, activated platelets can mediate venous thrombosis including via stimulation of coagulation and fibrinolysis (see Figure 2).NETs: neutrophil extracellular traps, vWF: von Willebrand factor, GP1b: Glycoprotein 1b, CLEC2: C-type lectin-like receptor 2, GPVI: Glycoprotein VI, RAGE: TLR: Toll-like receptor, HMGB1: High mobility group box 1, CXCR4: C-X-C chemokine receptor type 4, PSGL-1: P-selectin glycoprotein ligand-1, SLC44A2: Solute Carrier Family 44 Member 2, αMβ2: integrin αMβ2 or Mac-1 (macrophage 1 antigen), MRP14: migration inhibitory factor-related protein 14, IL: Interleukin, PAR1-4: Protease-activated receptor 1–4, C3a: Complement factor 3a, GAS6: Growth arrest–specific 6. Image is created using Biorender.com.
Figure 1. The role of platelets in different aspects of venous thrombosis. Platelets are crucially involved in the pathophysiology of venous thrombosis. They can be recruited to the vessel wall and can interact with neutrophils and monocytes. In addition, activated platelets can mediate venous thrombosis including via stimulation of coagulation and fibrinolysis (see Figure 2).NETs: neutrophil extracellular traps, vWF: von Willebrand factor, GP1b: Glycoprotein 1b, CLEC2: C-type lectin-like receptor 2, GPVI: Glycoprotein VI, RAGE: TLR: Toll-like receptor, HMGB1: High mobility group box 1, CXCR4: C-X-C chemokine receptor type 4, PSGL-1: P-selectin glycoprotein ligand-1, SLC44A2: Solute Carrier Family 44 Member 2, αMβ2: integrin αMβ2 or Mac-1 (macrophage 1 antigen), MRP14: migration inhibitory factor-related protein 14, IL: Interleukin, PAR1-4: Protease-activated receptor 1–4, C3a: Complement factor 3a, GAS6: Growth arrest–specific 6. Image is created using Biorender.com.
Ijms 23 13176 g001
Figure 2. The role of platelets in coagulation and fibrinolysis. Platelets are involved in both coagulation and fibrinolysis. Upon activation, they can secrete microvesicles, coagulation and fibrinolysis factors from their α-granules, and Ca2+ and polyphosphates (polyp) from their dense granules. In addition, they expose phosphatidylserine on their membrane to support both coagulation and fibrinolysis. FV: coagulation factor V, FVIII: coagulation factor VIII, FIX: coagulation factor IX, FXIII: coagulation factor XIII, vWF: von Willebrand factor, FX: coagulation factor X, PAI1: plasminogen activator inhibitor 1, TAFI: thrombin activatable fibrinolysis inhibitor, uPA/tPA: urokinase/tissue plasminogen activator, δ-granules: dense granules. Image is created using Biorender.com.
Figure 2. The role of platelets in coagulation and fibrinolysis. Platelets are involved in both coagulation and fibrinolysis. Upon activation, they can secrete microvesicles, coagulation and fibrinolysis factors from their α-granules, and Ca2+ and polyphosphates (polyp) from their dense granules. In addition, they expose phosphatidylserine on their membrane to support both coagulation and fibrinolysis. FV: coagulation factor V, FVIII: coagulation factor VIII, FIX: coagulation factor IX, FXIII: coagulation factor XIII, vWF: von Willebrand factor, FX: coagulation factor X, PAI1: plasminogen activator inhibitor 1, TAFI: thrombin activatable fibrinolysis inhibitor, uPA/tPA: urokinase/tissue plasminogen activator, δ-granules: dense granules. Image is created using Biorender.com.
Ijms 23 13176 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Heestermans, M.; Poenou, G.; Duchez, A.-C.; Hamzeh-Cognasse, H.; Bertoletti, L.; Cognasse, F. Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases. Int. J. Mol. Sci. 2022, 23, 13176. https://doi.org/10.3390/ijms232113176

AMA Style

Heestermans M, Poenou G, Duchez A-C, Hamzeh-Cognasse H, Bertoletti L, Cognasse F. Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases. International Journal of Molecular Sciences. 2022; 23(21):13176. https://doi.org/10.3390/ijms232113176

Chicago/Turabian Style

Heestermans, Marco, Géraldine Poenou, Anne-Claire Duchez, Hind Hamzeh-Cognasse, Laurent Bertoletti, and Fabrice Cognasse. 2022. "Immunothrombosis and the Role of Platelets in Venous Thromboembolic Diseases" International Journal of Molecular Sciences 23, no. 21: 13176. https://doi.org/10.3390/ijms232113176

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