Next Article in Journal
Evidence of Validity and Normative Values of a New Auditory Backward Masking Test
Previous Article in Journal
Comparison between Carotid Distensibility-Based Vascular Age and Risk-Based Vascular Age in Middle-Aged Population Free of Cardiovascular Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders

by
Houssam Al-Koussa
1,†,‡,
Ibrahim AlZaim
1,2,†,§ and
Marwan E. El-Sabban
3,*
1
Department of Pharmacology and Toxicology, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
2
Department of Biochemistry and Molecular Genetics, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
3
Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, The American University of Beirut, Beirut P.O. Box 11-0236, Lebanon
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Current Affiliation: Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
§
Current Affiliation: Department of Biomedicine, Aarhus University, Høegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark.
J. Clin. Med. 2022, 11(16), 4932; https://doi.org/10.3390/jcm11164932
Submission received: 7 May 2022 / Accepted: 20 May 2022 / Published: 22 August 2022
(This article belongs to the Section Hematology)

Abstract

:
The notion of blood coagulation dates back to the ancient Greek civilization. However, the emergence of innovative scientific discoveries that started in the seventeenth century formulated the fundamentals of blood coagulation. Our understanding of key coagulation processes continues to evolve, as novel homeostatic and pathophysiological aspects of hemostasis are revealed. Hemostasis is a dynamic physiological process, which stops bleeding at the site of injury while maintaining normal blood flow within the body. Intrinsic and extrinsic coagulation pathways culminate in the homeostatic cessation of blood loss, through the sequential activation of the coagulation factors. Recently, the cell-based theory, which combines these two pathways, along with newly discovered mechanisms, emerged to holistically describe intricate in vivo coagulation mechanisms. The complexity of these mechanisms becomes evident in coagulation diseases such as hemophilia, Von Willebrand disease, thrombophilia, and vitamin K deficiency, in which excessive bleeding, thrombosis, or unnecessary clotting, drive the development and progression of diseases. Accumulating evidence implicates cell-derived and platelet-derived extracellular vesicles (EVs), which comprise microvesicles (MVs), exosomes, and apoptotic bodies, in the modulation of the coagulation cascade in hemostasis and thrombosis. As these EVs are associated with intercellular communication, molecular recycling, and metastatic niche creation, emerging evidence explores EVs as valuable diagnostic and therapeutic approaches in thrombotic and prothrombotic diseases.

1. Introduction

Hemostasis is a dynamic physiological process, which controls bleeding at the site of injury while maintaining normal blood flow within the body. This occurs through the induction of blood coagulation, a process by which the activation of clotting factors forms a blood clot that halts bleeding [1]. The concept of blood coagulation was recognized as early as ancient Greece. However, Marcello Malpighi, an Italian scientist, observed and documented the process of blood coagulation in the 1600s. The surge in interest in this field resulted in the discovery of several coagulation factors, and accordingly, numerous theories were presented. As such, the theory of hemostasis became well refined and understood during the twenty-first century. The complex regulation of coagulation factors in hemostasis relies on the interaction among a multitude of effectors such as platelets, blood vessels, and other coagulation factors. Coagulation is coordinated by pro-coagulation factors that are countered by the process of fibrinolysis, which comprises a set of anti-coagulation factors and inhibitors regulating the coagulation cascade and maintaining hemostasis while preventing thrombosis [2,3]. However, states of hypo-coagulation and hyper-coagulation emerge secondary to defects in the coagulation cascade leading to either abnormal bleeding or abnormal clotting [4].
An exciting novel opportunity in studying coagulopathies is the implication of extracellular vesicles (EVs), in the modulation of coagulation processes [5,6,7,8]. EVs comprise micro-vesicles (MVs), exosomes, and apoptotic bodies. Studies on the mechanisms of EV formation, cargo sorting, and release have witnessed a surge in the literature. Importantly, platelet-derived EVs are highly implicated in the regulation of coagulation physiology and the pathophysiology of hypercoagulable states. Here, we present a historical background of coagulation up to the discovery of the cell-based model. We also describe the process of hemostasis from injury occurrence to blood clot dispersion. Importantly, we focus on the emerging evidence implicating cell-derived and platelet-derived EVs in the modulation of hemostasis and coagulopathies.

2. Coagulation: A Historical Overview

Theories of blood clotting date back to ancient Greece when the words hema (blood) and thrombus (clump) were introduced [9,10]. In the seventeenth century, novel concepts regarding coagulation deviating from the “traditional Galen medicine” were tabooed, contested, and disputed. However, Dr. Marcello Malpighi championed these revolutionary theories and identified, using a single lense microscope, that blood consists of a combination of red blood cells and a meshwork of fibrous material, now referred to as fibers [3,11]. In the early 1720s, Jean-Louis Petit, a French surgeon, was the first to link coagulation with hemostasis after observing patients healing from limb amputation, after whom Dr. William Hewson, a young British surgeon, observed that blood coagulum arose from the liquid part of the blood, which is now known as plasma. Moreover, he discovered that the plasma contains different substances like albumin, globulin, and the precursor of fibrin [12,13]. In 1801, Fourcroy introduced the term “fibrin” and demonstrated that its precursor exists in the plasma. Four decades later in 1847, Virchow identified the precursor of fibrin and named it “fibrinogen” [14]. In 1872, Alexander Schmidt’s research showed that an enzymatic process leads to the conversion of fibrinogen into fibrin, and he called the enzyme “thrombin”. Schmidt realized that thrombin cannot exist in its active state in the blood, thus he postulated the presence of a precursor, prothrombin [15]. These discoveries laid the foundation of Paul Morawitz’s classic theory of coagulation in 1905, whereby the schema of coagulation consisted of four factors “prothrombin, thrombin, fibrinogen, and fibrin” in the presence of calcium [2,16].
This classic theory of coagulation persisted until Paul Owren introduced modern insights on the biochemical processes of coagulation in 1947 [13], when he identified new coagulation factors including von Willebrand factor (vWF) [17], factor five (FV) [18], FVII [19], FVIII [20], FIX [21], FXI [22], and FX [23]. In 1964, Macfarlane proposed the cascade model, which introduces the order in which the clotting factors are activated. This model was followed by the waterfall model by David and Ratnoff in which each clotting factor is a proenzyme (zymogen) awaiting predecessor-mediated proteolytic cleavage to initiate its activity thus preventing indiscriminate activation or degradation [24,25,26]. Coagulation can be initiated by two different pathways; the intrinsic pathway secondary to vascular endothelium damage and sub-endothelial collagen exposure and the TF-dependent extrinsic pathway. However, the initiation of either pathway results in the activation of FX of the cascade, leading to the formation of a fibrin clot [27].
In 1935, Henrik Dam discovered that vitamin K was an important player in coagulation [28]. Indeed, vitamin K plays a role in the activation of prothrombin, factors VII, IX, and X, and the production of proteins C, S, and Z [29]. Another key player in coagulation is calcium, which is required for the conversion of prothrombin into thrombin [13]. Recent work on coagulation led to the development of a new model “the cell-based model” in which the exposure of cells expressing TF on their surface initiates blood coagulation in vivo. This model pinpoints that the intrinsic pathway does not have a proper physiological role in hemostasis, adjusts the role of FXII, and highlights its contribution to pathological thrombosis [30,31].

3. Hemostasis

Hemostasis is achieved through an intricate balance between the levels of procoagulant and anti-coagulant factors [32,33]. Blood vessels are lined by endothelial cells that exhibit intrinsic antithrombotic properties. However, secondary to vascular injury, the sub-endothelial matrix comprising collagens, VWF, and adhesion proteins, makes the environment highly thrombogenic to blood [34]. Hemostasis comprises primary and secondary phases that culminate in the formation and stabilization of fibrin strands forming the platelet plug [35]. Key hemostatic mechanisms are described in Figure 1.

3.1. Primary Hemostasis, Secondary Hemostasis, and the Cell-Based Model of Coagulation

During primary hemostasis, platelets and vWF accumulate rapidly at the sub-endothelial matrix following endothelial damage. Platelet adhesion to collagen is facilitated by injured tissue-released vWF. This occurs following platelet activation and the interaction of platelet surface glycoproteins receptors GPIb-IX-V and GPVI with immobilized vWF, and sub-endothelial collagen, respectively [35,36]. Both receptors are critical for platelet adhesion to the sub-endothelial matrix at the site of injury [37,38,39,40,41]. Activated platelets exhibit increased activity of surface integrins, αIIbβ3, α2β1, and αvβ3, that ensure platelet-platelet interactions, as well as their binding to collagen, VWF, fibrinogen, and fibronectin [42,43]. Moreover, the platelet phospholipid bilayer membrane is rearranged following platelet activation and becomes negatively charged to support the coagulation cascade later during secondary hemostasis [44]. The platelet membrane acts as a surface for the assembly of activated coagulation cascade during secondary hemostasis.
Concurrently, and during secondary hemostasis, extravascular cells including smooth muscle, fibroblastic, and endothelial cells, release TF (also known as FIIIa). Platelet-released polyphosphates activate FVII into FVIIa, which then binds TF forming the FVIIa/TF complex. In the presence of calcium, the FVIIa/TF complex activates FX into FXa, which consequently activates prothrombin into thrombin in the presence of its cofactor FVa [45,46]. In the extrinsic pathway, FXII is activated into FXIIa by the negatively charged membrane phospholipid surface of the platelets. FXIIa then cleaves FXI producing FXIa, which activates, in the presence of calcium, FIX to FIXa. FIX can also be activated by FVIIa through the intrinsic pathway. FVIII is a cofactor produced by endothelial cells and hepatocytes and is found as a complex with circulating vWF. Following vascular injury, FVIIIa dissociates from the complex with vWF and interacts with FIX, forming a new complex in the presence of calcium ions and negatively charged membrane phospholipids that activate FX into FXa [47,48].
The extrinsic and intrinsic pathways culminate at FXa which activates the serine protease thrombin, which cleaves fibrinogen into fibrin [49]. Thrombin further activates platelets through the proteolytic processing of the G-coupled membrane receptors, protease-activated receptor 1 (PAR1) and PAR4 [50]. Thrombin also activates FXI, which later activates FIX through the intrinsic pathway; but also FVIII and FV, creating a positive feedback activation loop for coagulation [49].
More recently, a cell-based model of coagulation was proposed implicating elaborate interactions between platelets, vessel walls, and the coagulation system, and better describes the in vivo coagulation processes [34]. This model comprises three distinct phases; initiation, amplification, and propagation rather than independent pathways [31]. An example of such intricate processes includes FXa interaction with FVa, which produces limited amounts of thrombin that plays a key role in the activation of FXI, FVIII, FV, and platelet PARs during amplification [30,51,52,53]. Following its activation by thrombin, FVa is released from the fully-activated platelets and interacts with FXa (activated by the FVIIIa/FIXa complex) forming a complex that further activates thrombin. This process is referred to as the thrombin burst [54,55,56,57,58]. Thrombin can be directly inhibited by anti-thrombin III, which also inhibits FXa, FIXa, and FXIIa [59], and thrombin production could be halted by the tissue factor pathway inhibitor (TFPI), which inhibits the TF/FVIIa complex [60]. Finally, and in the presence of calcium, fibrin forms fibrin polymers [61], and thrombin activates FXIII to form FXIIIa, which crosslinks and strengthens fibrin polymers forming a fibrin mesh that strengthens the blood clot [62].

3.2. Fibrinolysis, Anti-Coagulation Factors, and Coagulation Inhibitors

Fibrinolytic mechanisms are activated following fibrin mesh formation. Plasminogen, the precursor of plasmin, an anti-coagulation proteolytic enzyme, is activated into plasmin by the urokinase-type plasminogen activator (uPA) and tissue-type plasminogen activator (tPA). PAs have a short half-life in circulation due to the presence of circulating inhibitor plasminogen activator inhibitors (PAIs). Plasmin cleaves fibrin into soluble fibrin degradation products (FDP). Plasminogen and PAs bind FDP exposed carboxy-terminal lysine amino acid, which leads to the activation of plasmin and the cleavage of fibrin to form FDP. Moreover, newly activated plasmin activates PAs by converting single-chain PAs to their two chain counterparts, exerting positive feedback on plasmin activation [63,64,65]. Thrombin activated fibrinolysis inhibitor (TAFI) is another zymogen that is activated by the thrombomodulin (TM)/thrombin complex to form TAFIa [66]. TAFIa downregulates fibrinolysis by removing the C-terminal lysine from the FDP, thus preventing tPA/plasminogen/fibrin complex formation and inhibiting further plasmin activation [66]. It is however worth noting that TM can act as a cofactor for APC activation and upregulate fibrinolysis by attenuation of thrombin production [67,68]. Thus, TAFIa and fibrinolysis regulation are linked to TM levels of the vasculature [69]. Moreover, the inhibition of coagulation is achieved through the presence of circulating protease inhibitors such as anti-thrombin, heparin cofactor II, TFPI, and CI inhibitors. Anti-thrombin targets thrombin, FXa, FIXa, FXIa, and FXIIa, while TFPI targets FXa and the TF/FVIIa complex. Heparin cofactor II and protein C target thrombin [34].
Protein C (PC), a vitamin K-dependent glycoprotein, exerts a pro-fibrinolytic effect by regulating thrombin formation and TAFI activation. Particularly, increasing thrombin formation leads to the binding and activation of PC-forming active protein C APC, secondary to increased thrombin interaction with TM [68]. PC activation is enhanced when bound to the endothelial cell protein C receptor (EPCR) [70]. Following APC formation, APC breaks from the EPCR and TM/thrombin complex, binds to its cofactor protein S (PS), and targets FVa and FVIIIa, thus preventing FXa formation and limiting the thrombin activity [68,71].

3.3. Pathophysiology of Coagulopathies

Two common hereditary hypercoagulable conditions include hemophilia and von Willebrand disease. Hemophilia A is the most frequent type of hemophilia and is correlated with FVIII deficiency, while Hemophilia B is correlated with FIX deficiency. Hemophilia C is the least frequent of the three hemophilia subtypes and is correlated with factor XI deficiency [72,73]. Von Willebrand disease is associated with the abnormal production of vWF [74]. Vitamin K is a crucial cofactor for the synthesis and activation of the coagulation factors FII, FIX, and FX, and the anticoagulation proteins (C and S). The lack of Vitamin K leads to the production of under-carboxylated factors that cannot bind to calcium [75].
Hypercoagulable conditions are associated with thrombophilia and are categorized into acquired and inherited conditions [4,76]. The majority of thrombosis is due to thrombophilia, especially in the case of deep vein thrombosis (DVT) [77]. DVT and pulmonary embolism (PE), both known as venous thromboembolism (VTE), are the most frequent conditions correlated with thrombophilia. Major inherited conditions causing thrombophilia include FV Leiden mutation, anti-thrombin III deficiency, PC-PS deficiency, and prothrombin-related thrombophilia [78]. FV Leiden thrombophilia is the most common type of thrombophilia and is associated with the emergence of an APC-resistant FV mutant [79]. Prothrombin-related thrombophilia is the second most common type of thrombophilia and arises from a prothrombin G20210A gene mutation, which enhances DVT occurrence secondary to increased thrombin levels [80]. Anti-thrombin deficiency is also associated with an increased incidence of thrombotic episodes [78,81]. Finally, PC and PS deficiencies enhance the development of DVT and PE [82,83]. There exist two major types of PC deficiencies, type I which is correlated with reduced circulating PC concentrations, and type II which is correlated with a reduction in PC functional activity [84].

4. Novel Avenues in Thrombosis Research

In addition to inherent disorders of coagulation, several pathological conditions such as cancer and sepsis are associated with a hypercoagulable state, to which extracellular vesicles significantly contribute. Extracellular vesicles can transfer prothrombotic molecules inter-cellularly and mediate the dissemination of coagulation resulting in arterial and venous thrombosis. As the role of cell-derived and platelet-derived EVs in physiological and pathophysiological states is being continuously revealed, an increased interest in the therapeutic manipulation of these entities is emerging [85]. The cellular release of vesicles is a common and evolutionary-conserved process [86]. The underlying mechanisms of vesicle formation, component sorting, and release have witnessed extensive research within the past decade. Here, we review key concepts of EV biology and thoroughly discuss the implication of cell-derived and platelet-derived EVs in hypercoagulable disease states, especially in cancer and sepsis.

4.1. Extracellular Vesicles as Novel Modulators of Coagulation

The term extracellular vesicle denotes numerous subpopulations of cell-shed, non-replicative lipid bilayer-enclosed vesicles [5,6,7]. EV subtypes are characterized by distinct cellular origins and different mechanisms of biogenesis, insights which were provided by means of transmission and immune-electron microscopy. EVs are widely distributed in human bodily fluids including urine, blood, saliva, and synovial and cerebrospinal fluids [86]. Advances in the standardization of EVs isolation and characterization are comprehensively reviewed elsewhere [5,86]. EVs, which include MVs, exosomes, membrane particles, and apoptotic bodies [8], have attracted significant interest as they are implicated in the regulation of coagulation physiology and the pathophysiology of hypercoagulable states. Although early observations highlighted procoagulant properties of EVs in healthy individuals by supporting low-grade thrombin formation [86], emerging evidence supports a fibrinolytic rather than a procoagulant activity of EVs in healthy humans [87].
Procoagulant EVs are released by a plethora of cells including endothelial cells, adipocytes, and macrophages among others implicating these entities in the pathogenesis of cardiovascular diseases and their associated procoagulant states as summarized in Table 1. [88,89,90,91]. The concentration of EVs in healthy individuals amounts to 103–1011 per mL depending on the methodologies used for EV quantification and their respective sensitivities [92,93]. The abundance of circulating EVs was shown to regress in an age-dependent manner [94]. It was, however, demonstrated that endothelial EVs from elderly subjects maintain a procoagulant activity contrary to that of young control subjects [95]. Moreover, increased EV plasma levels are observed in diseases associated with a procoagulant state such as thrombotic thrombocytopenic purpura [96], sickle cell disease [97], and heparin-induced thrombocytopenia [98]. Enhanced circulating EV levels are also observed in established cardiovascular diseases including acute coronary syndrome [99] and stroke [100,101], but also in metabolic disorders predisposing to cardiovascular dysfunction such as type 2 diabetes and metabolic syndrome [102,103,104]. Recent evidence suggests that cellular-derived and platelet-derived EVs modulate thrombogenicity in patients with atrial fibrillation [105]. AF is associated with elevated circulating levels of platelet-derived and mononuclear cell-derived EVs and reduced endothelial cell-derived EVs [105]. Elevated EVs including procoagulant TF-expressing, P-selectin-expressing, or PS-expressing EVs were demonstrated in AF patients and were suggested to contribute to AF-associated thrombogenicity [106,107,108]. Whether this increase in EVs concentration associated with these disorders drives their pathogenesis or is secondary to disease development requires further investigation. Importantly, the mechanisms by which coagulation factors modulate the release of EVs remain largely unknown. Nevertheless, recent evidence implicates FVIIa/EPCR/PAR1-mediated signaling in the induction of the release of procoagulant EVs from endothelial cells primarily through a ROCK-dependent pathway [88,89].
EVs represent a novel mechanism in intercellular communication in both physiological and pathophysiological states [109], carrying a cargo of bioactive molecules, such as lipids, proteins, messenger RNAs (mRNA), cellular metabolites, DNA, and non-coding RNA molecules including microRNAs (miRs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) into target cells (Figure 2) [110,111]. Particularly, intact EV-enclosed non-coding RNA molecules can be isolated from the circulation despite the presence of significant RNase activity [112,113]. Indeed, EVs enclosing miRNAs have been demonstrated to participate in the modulation of various diseases as miRNAs regulate key factors of hemostasis [114]. It is worth mentioning that platelets inherit a diverse array of coding and non-coding sequences and translational machinery from megakaryocytes and represent a major source of circulating miRs [115]. Nevertheless, the contribution of activated platelet-derived EVs to the pool of circulating non-coding RNAs requires further investigation and surely adds to the complexity of studying thrombosis-associated EV-encapsulated non-coding RNAs.
Table 1. Cardiovascular diseases are associated with increased levels of procoagulant circulating extracellular vesicles. Clinical evidence implicates procoagulant cellular-derived and platelet-derived extracellular vesicles in the promotion of cardiovascular disease-associated thrombogenicity. EVs, extracellular vesicles; MVs, microvesicles; PS, phosphatidylserine; TF, tissue factor.
Table 1. Cardiovascular diseases are associated with increased levels of procoagulant circulating extracellular vesicles. Clinical evidence implicates procoagulant cellular-derived and platelet-derived extracellular vesicles in the promotion of cardiovascular disease-associated thrombogenicity. EVs, extracellular vesicles; MVs, microvesicles; PS, phosphatidylserine; TF, tissue factor.
Disease Alteration of the Abundance of Circulating Procoagulant Extracellular VesiclesReferences
Thrombotic thrombocytopenic purpura (TTP)Increased levels of circulating platelet-derived EVs[96]
Idiopathic thrombocytopenic purpura (ITP)Increased levels of circulating platelet-derived EVs [116]
Heparin-induced thrombocytopenia (HIT)Increased levels of circulating TF- expressing platelet-derived EVs[98,117]
Sickle cell anemia Elevated circulating levels of erythrocyte, platelet, monocyte, and endothelial cell-derived EVs[93,118,119]
Disseminated intravascular coagulation (DIC)Increased levels of circulating endothelial cell-derived EVs (suggested as a biomarker of DIC caused by septic shock)[120,121,122]
Acute coronary syndromes (ACS)Elevated platelet and monocyte-derived MVs
Increased levels of circulating CD31+ CD42b MVs
[99,123,124]
Venous thromboembolism (VTE)Elevated levels of circulating endothelial cell and platelet-derived PSGL-1 and CD62P-expressing MVs [125,126]
Acute ischemic stroke (AIS)Elevated levels of circulating endothelial cell-derived MVs [127]
Paroxysmal nocturnal hemoglobinuria (PNH)Increased levels of circulating platelet, monocyte, and endothelial cell-derived EVs [128,129]
Coronary heart disease (CHD)Elevated levels of CD31+, CD42, and CD144+ endothelial cell-derived EVs[124]
Acute myocardial ischemia Elevated levels of circulating CD66b+, CD62E+, and CD142+ EVs[130]
ST-segment elevation myocardial infarction (STEMI)Elevated levels of circulating leukocyte-derived CD11+, endothelial cell-derived
CD105+, and TF-bearing MVs
Increased levels of erythrocyte-derived but not platelet-derived MVs
[131]
Acute stroke (AS)Elevated levels of circulating CD62E+ endothelial cell-derived EVs[101]
Acute pulmonary embolism (APE).Increased levels of circulating TF-expressing MVs[132,133]
Atrial Fibrillation (AF)Increased levels of circulating platelet-derived and mononuclear cell-derived EVs and reduced levels of circulating endothelial cell-derived EVs
Increased levels of circulating procoagulant EVs expressing TF, PS, and P-selectin
[105,106,107,108]

4.1.1. Microvesicles

First documented in the late 1960s and thought to be “cell dust”, microvesicles, also known as ectosomes or microparticles, are lipid bilayer-enclosed sacs ranging in diameter between 50 nm to 1.0 μm [8,134]. Although initially thought to be plasma membrane fragments released from platelets as part of the coagulation process [135,136], MVs are now believed to be important players in intercellular communication. Importantly, MVs were shown to transfer bioactive molecules such as hormones, cytokines, and coagulation factors, and are implicated in the modulation of coagulation processes [135]. Indeed, it was demonstrated that neutrophils and monocytes release TF-expressing MVs in circulating blood near the surface of platelets [137]. Two different mechanisms elicit MVs formation, apoptosis, and cell activation. During apoptosis, DNA fragments and the cell contracts resulting in an actin-myosin cytoskeleton-mediated membrane blebbing [138]. These blebs are distinct from those generated through cellular activation in size and protein composition. As the bleb formation continues, cells shrink into small apoptotic bodies as polymerized actin undergoes dissolution (Figure 2). The shedding of MVs can also occur in an apoptosis-independent manner following the exposure to certain physiological and pathophysiological stimuli among which are stimulation by proinflammatory cytokines, ROS, PMA, LPS, complement C5b-9, PAI-1, thrombin, and collagen [139,140]. It is worth mentioning that the mechanism by which cellular-derived and platelet-derived MVs release is induced determines their phenotype and function. Upon cellular activation, MVs are released in a calcium-dependent mechanism which triggers dynamic cytoskeletal rearrangement of the plasma membrane mediated by a consortium of enzymes including flippases, floppases, and scramblases [8]. Calcium also activates enzymes that modulate the cytoskeleton and promote the disassembly of sub-plasma membrane-localized actin such as gelsolin and calpain. All these changes in the structure and dynamics of the plasma membrane result in alterations of the membrane curvature and the formation of protrusions allowing the MVs to detach [141]. Dramatic extremes of temperature reduce the cellular capacity to internalize EVs and were shown to abolish the procoagulant activity of endothelial cell-derived MVs [142,143]. Moreover, calcium chelation, formalin fixation, and cytoskeleton disruption have been shown to reduce EVs cellular uptake [143].
The concentration of MVs in plasma has been estimated at 2–4 × 108 per mL [135]. Although the majority of these MVs are platelet-derived [144,145], MVs deriving from granulocytes, ECs, monocytes, and erythrocytes have been identified in states of health and disease. Indeed, human blood enrichment with circulating MVs from healthy subjects enhances platelet adhesion and fibrin deposition on the damaged porcine aorta and human atherosclerotic arteries [146]. Additionally, circulating thrombin-generating cell-derived MVs occur in healthy subjects and their procoagulant effects occur through TF-independent pathways and were inhibited, at least in part, through FXII, FXI, or FVIII-targeting antibodies [86]. Platelet-derived MVs express TF and contain procoagulant membrane components that potentiate coagulation due to their unusual lipid arrangements, including the externalization of phosphatidylserine (PS) [147,148,149]. Defects in systemic PS-expressing MVs clearance were found to instigate hypercoagulation in lactadherin-deficient mice [142,150,151,152]. TF-exposing MVs were shown to derive from platelets, neutrophils, erythrocytes, granulocytes, and monocytes, while their capacity to induce thrombin generation was reduced following the incubation of TF-neutralizing antibody [153]. In addition to TF and TFPI, other procoagulant molecules and receptors carried by MVs include P-selectin glycoprotein ligand-1 (PSGL-1), which mediates the interaction between MVs and CD62P expressed on activated platelets and endothelial cells [154]. Moreover, an increased number of circulating MVs was demonstrated in patients with vascular inflammation and heart failure, stroke, and myocardial infarction, at least in part secondary to platelet activation [124,150,151,155,156,157]. The abundance of circulating procoagulant MVs is also increased in ACS, chronic ischemic heart disease patients, CAD, heart failure, as well as atrial fibrillation patients, and following vascular stent implantation [130,151,157,158,159,160]. Particularly, increased procoagulant MVs in CAD and ACS are suggested to be endothelial cell-derived [99,124,161]. Indeed, the procoagulant potential of monocyte-derived MVs is the highest, followed by that of endothelial cells, platelets, and erythrocytes-derived MVs [162]. Indeed, several studies have shown that TF-expressing MVs derive from monocytes [163,164,165], while platelet-derived MVs were shown not to support FXa generation and to exhibit reduced procoagulant activity [166]. Furthermore, emerging evidence suggests that cellular phenotypes and polarization, such as the proinflammatory and anti-inflammatory polarization modalities of macrophages, modulate the procoagulant potential of their secreted MVs [167].

4.1.2. Platelet-Derived Microvesicles

Platelet-derived MVs can be distinguished by their expression of the surface markers CD41 and CD42b [168,169]. Platelet-derived MVs encompass a broad vesicle size range depending on their content of growth factors, chemokines, and plasma membrane receptors [170]. It is suggested that smaller MVs originate from α-granules, while larger ones derive from the plasma membrane. It was demonstrated that the number of MVs released from activated platelets does not match the number of MVs released by platelets in healthy subjects in vivo. This led scientists to theorize that MVs are derived from circulating quiescent platelets following shear stress exposure [171]. However, it is now hypothesized that CD41+, filamin-A-expressing MVs continuously derive from megakaryocytes and simultaneously exist with regular platelet-derived MVs in healthy humans [172]. Importantly, CD41+ MVs were shown not to be associated with an increased risk of CVDs, which reinforces the speculation that the non-active form of MVs possibly derives from megakaryocytes [173,174,175]. Nevertheless, it is suggested that following the activation of platelets, increased levels of platelet-derived MVs are released as depicted in Figure 3 [176]. Indeed, platelets activated by ADP, thrombin, and collagen were shown to release an enhanced number of MVs [177,178,179,180,181]. Anti-coagulant drugs including clopidogrel, which targets adenosine P2Y12 ADP receptor or ticagrelor, or the antiplatelet aspirin reduce the release of procoagulant platelet-derived EVs [182,183,184]. Direct thrombin inhibitors including dabigatran and melagatran reduce TF-expressing platelet-derived MVs release in response to thrombin or ADP stimulation [185]. Additionally, monoclonal antibodies targeting GPIbαinhibit procoagulant platelet-derived MVs release under high shear stress more efficiently than does the GPIIb/IIIa antagonist abciximab [186]. Moreover, activated platelet-derived MVs are 50–100 fold more procoagulant than the negatively-charged surface of platelets [187].
Although a balanced production of platelet-derived MVs is pivotal to the induction of angiogenesis and endothelial cell proliferation in vivo [175,188], aberrantly low or high production of platelet-derived MVs has been demonstrated in several pathological states. Individuals with Scott’s Syndrome, Glanzmann’s thrombasthenia, and Castaman defect have an impaired basal capacity to generate MVs resulting in an enhanced susceptibility for hemorrhage [189,190,191,192]. Alternatively, elevated levels of platelet-derived MVs were demonstrated in several thrombosis-associated diseases including arterial thrombosis [189], sickle cell disease [97], and CVDs such as angina and hypertension [189]. It has also been suggested that platelet-derived MVs transfer miRs to target cells as a form of intercellular communication [193,194]. For example, the transport of miRNA-133a, whereby levels are significantly elevated in patients with acute MI and angina pectoris occurs via MVs reaching target cells in the heart as well as other organs following these cardiovascular events [195,196]. Moreover, miR-containing platelet-derived MVs function beyond coagulation. For example, platelet-derived miR-1915-3p-containing MVs suppress the expression of Rho GTPase family member B, inducing megakaryopoiesis [197].

Exosomes

Exosomes are nano-sized particles that range in diameter between 50 and 150 nm and are secreted by almost all mammalian cells [85,198,199]. Exosomes’ protein content is characterized by a consortium of transmembrane and non-membrane bound proteins including Annexin V, GTPases, Rab, and flotillin, as well as protein components of the endosomal sorting complexes required for transport (ESCRT), such as AGLT-2-interacting protein X (Alix), heat shock proteins (HSPs), tumor susceptibility gene 101 (TSG101), tetraspanins (CD63, CD81, and CD9), and integrins (e.g., α4β1 and VCAM-1 on endothelial cells and αLβ2 on leukocytes) [8,200,201,202,203]. As such, patient treatment with abciximab, which targets the integrin αIIβ3, inhibits the uptake of PS-expressing EVs by human endothelial cells [204].
Exosomes derive from the fusion of the plasma membrane with the endosomal network. However, the mechanisms of exosome biogenesis, formation, and release are quite complicated, and only partly unraveled [8,205]. It has been proposed that exosomal biogenesis starts with the formation in specialized regions of the plasma membrane of endocytic EVs, which is followed by the formation of intraluminal vesicles (also called exosome precursors) within multi-vesicular bodies. Intraluminal vesicles then detach from the plasma membrane and move to the early endosome, where they fuse. In this compartment, internalized membrane receptors undergo conformational changes and dissociate from their ligands. The dissociated ligands are destined for degradation after their sorting into lysosomes. Alternatively, internalized receptors and retained ligands can be recycled to the plasma membrane or transferred to late endosomes. In late endosomes, proteins are either degraded through the lysosomal pathway or are incorporated in intraluminal vesicles, which upon budding from multi-vesicular bodies, are conveyed to the plasma membrane and are released (Figure 2) [8,206]. It is worth noting that the inhibition of exosome biosynthesis and release in senescent cells promoted DNA damage and accelerated cellular apoptosis-like cell death [207], suggesting that exosome-mediated release of detrimental DNA fragments as well as other cellular components during cellular senescence may play a protective role preventing the development of innate immune responses.
Accumulating evidence suggests the implication of various molecules in the membrane fusion and release of exosomes. Small Rab GTPases were shown to drive exosome secretion, while Rab27a and Rab27b are implicated in multi-vesicular body docking to the plasma membrane [208]. Multi-vesicular body formation and exosome secretion are also controlled by Ras-related GTPase homolog (RAL-1) [209]. Following secretion, circulating exosomes can be taken up by target cells by three different mechanisms; fusion, binding, and endocytosis [143]. In that sense, exosomes expressing membrane-bound ligands may bind to receptors of target cells to activate intracellular signaling [210]. For example, the expression of intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) on the surface of EVs allows their binding to recipient cells through receptor interactions with leukocyte function-associated antigen-1 (LFA-1) and very late antigen-4 (VLA-4), respectively [211,212]. Moreover, proteoglycans such as heparan sulfate proteoglycan receptors and glycoproteins such as the hyaluronan receptor CD44, or lectins present other protein groups implicated in the uptake of EVs by recipient cells [139]. Various mechanisms of endocytosis, being the major type of exosome uptake, exist and include micropinocytosis, phagocytosis, clathrin-mediated and clathrin-independent endocytosis, lipid raft-mediated endocytosis, and caveolae-mediated endocytosis [143,213]. As such, inhibiting clathrin-dependent endocytosis in endothelial cells reduces the uptake of PS-expressing EVs [204]. Moreover, it was demonstrated that inhibiting caveolae endocytosis, either pharmacologically or through knocking down caveolin-1, as well as cholesterol depletion, reduces exosomes and MVs uptake by target cells [163]. It is proposed that exosomes actively contribute toward a procoagulant phenotype through carrying key effectors of coagulation such as fibrinogen [214]. Nevertheless, exosomes were demonstrated to be less efficient in promoting coagulation than MVs [215].

4.1.3. Platelet-Derived Exosomes

Platelet-derived exosomes were shown to suppress athero-thrombotic processes by enhancing the proteolytic degradation of CD36 and by inhibiting platelet activation and subsequent thrombosis [216]. It was also shown that thrombin-activated platelets secrete miR-223 during inflammation, which downregulates the expression of ICAM-1 in endothelial cells [217]. Moreover, exosomes derived from thrombin-stimulated platelets contain miR-223, miR-339, and miR-21 and inhibit the expression of platelet-derived growth factor receptor-β in VSMCs [218]. In an miR-223-deficient mouse model of carotid thrombosis, prolonged times to occlusive thrombosis were observed demonstrating a protective effect of miR-223 deficiency [219]. The transfusion EVs from WT mice to miR-223-deficient mice reversed the reduction in thrombosis time. Another study suggested that platelet miR-223 levels were significantly lower in hypertensive patients and these levels were suggested as prognostic markers for cardiovascular disease in these patients [220]. Moreover, miR-233 platelet levels were elevated while their content in platelet-derived MVs decreased in a hypertensive, hyperlipidemic animal model of atherosclerosis [221]. This indicates an active mechanism of selective miR packing into platelet-derived MVs in different pathologies, which provides novel targets for the treatment of cardiovascular diseases. It was also demonstrated ex vivo that platelets support gymnosis, that is, the internalization of ectopic miRs in absence of conventional transfection reagents and their incorporation into RNA-induced silencing complexes (RISCs) by endocytic pathways [222]. The uptake of miR-223-3p by thrombin or fibrinogen-induced platelets inhibited exosome and MVs generation [222]. A recent study demonstrated that senescent platelets in vitro include miRNAs relevant to atherosclerosis and inflammation [223]. Moreover, senescent platelet-derived EVs contain upregulated levels of miRs involved in obesity, diabetes, and metabolic and vascular diseases (Figure 3) [223]. Although the multifaceted roles of EVs in atherosclerosis development and progression are appreciated [224,225], further investigation into the possible roles of platelet-derived exosomes in driving the pathogenesis of coagulopathies is warranted.

4.1.4. Apoptotic Bodies

Apoptotic bodies are large, permeable vesicles released following cellular apoptosis, with sizes ranging from 50 nm to 5.0 μm in diameter [150,226]. Importantly, apoptotic bodies are less regular in shape in comparison to MVs and are released in advanced stages of apoptosis as a result of cellular contractions and the increased hydrostatic pressure in dying cells [227,228]. Some MVs may also be released during this stage of membrane blebbing. These bodies can contain various cellular contents, cellular organelles, and DNA fragments and can thus participate in processes ranging from tissue regeneration to horizontal DNA transfer. For example, the DNA content of endothelial cell-derived apoptotic bodies was shown to promote the proliferation and differentiation of human endothelial progenitor cells [229]. Similarly, macrophage-derived apoptotic bodies were shown to contain miR-221/222 which promotes the proliferation of lung epithelial cells [230]. Fibroblast-derived apoptotic bodies were also demonstrated to contain amino acids that induce gene recombination in target cells [231]. Apoptotic bodies are cleared through phagocytosis by cells of the innate immune system and endothelial cells [232,233]. In comparison with MVs, apoptotic bodies have poor prothrombotic properties and were suggested to use phosphatidylserine for the recruitment of phagocytes to sites of cellular death rather than for platelet activation (Figure 2) [234]. Nevertheless, PS-expressing apoptotic EVs were shown to directly associate with FXII and support thrombin generation [149]. As such, the procoagulant roles of apoptotic bodies are yet to be elucidated and require further investigation.

4.2. Extracellular Vesicles as Drivers of Hypercoagulable States

4.2.1. Extracellular Vesicles in the Cancer-Associated Prothrombotic States

A significantly higher risk for the development of venous thrombosis in cancer patients is well documented [235]. Cancer-associated thrombosis is a multi-factorial process in which several mechanisms are implicated, the molecular underpinnings of which are still obscure [236]. Several studies highlighted a role for tumor-derived EVs expressing TF and PS in the thrombotic manifestations of many cancers including multiple myeloma, and breast and prostate cancers [237,238,239]. Indeed, procoagulant, PS-expressing tumor-derived EVs provide initiation sites for blood coagulation and tumor-derived EVs may activate platelets and promote their aggregation through both TF-dependent and TF-independent mechanisms [109,240,241,242]. TF-expressing EVs were first isolated from plasma samples of advanced colorectal cancer patients [243] and were then detected in the plasma of pancreatic, lung, ovarian, colorectal, prostate, and breast cancer patients [244,245,246,247]. Moreover, one study showed that tumor-derived TF-expressing EVs induce DVT and that these EVs cooperate with host TF in order to cause the prothrombotic state in pancreatic cancer [248].
Emerging evidence provides conflicting perspectives on the implication of tumor-derived and platelet-derived procoagulant EVs in the pathogenesis of cancer-associated thrombosis and their use as biomarkers predicting coagulopathy [249,250,251,252,253,254,255]. Whereas a strong correlation between TF-expressing EVs and VTE was demonstrated in patients with pancreatic cancer [256,257], other studies in different cancers failed to arrive at similar associations [256,258,259,260,261]. Particularly, a prospective cohort study demonstrated an association between MVs TF activity in cancer patients and total mortality and prognosis, but not thrombosis [260]. These conflicting outcomes probably arise from the differential use of various techniques for TF measurement, different EV purification methods, and variable antibody and assay sensitivity. As novel assays measuring the activity of TF in plasma EVs are developing, a better comprehension of the association between TF-expressing EVs and cancer-associated thrombosis will become possible. It is noteworthy that although cancer cell-derived exosomes and MVs display fibrinolytic activities, only cancer cell-derived MVs display prothrombotic activities [262]. Importantly, neither exosomes nor MVs displays fibrinolytic activity under physiological conditions.
As vascular and tumor cell-derived, TF-bearing EVs were shown to drive coagulopathy [263,264], it was shown that TF-expressing EVs promote prothrombotic states in murine models of Lewis lung carcinoma, melanoma, pancreatic cancer [200,264,265,266,267]. Mice bearing TF-positive tumors and elevated levels of circulating TF-expressing EVs were shown to exhibit enhanced thrombosis [268]. Mice injected with TF-overexpressing ovarian cancer cells exhibited enhanced venous thrombus formation following experimental stenosis [269], while mice bearing tumors under-expressing TF exhibited halted coagulation, platelet aggregation, and thrombus formation [270]. Coculturing prostate cancer cells with high TF expression with peripheral blood mononuclear cells and platelets enhanced EVs TF activity while no such observation was shown in prostate cancer cells with low TF expression [247]. It was also suggested that glioma-derived podoplanin and TF coexpressing EVs cooperatively enhance micro-thrombosis in glioma xenograft murine models [271]. It is worth noting that the procoagulant potential of TF-expressing MVs derived from human tumor cell lines is reduced in PAR2−/− mice following clopidogrel treatment. Moreover, recent evidence suggests that cancer cell-derived EVs expressing polyphosphates induce blood coagulation by activating FXII, with pancreatic and lung cancer cell lines exhibiting the most potent activity [272,273]. Degrading EV’s polyphosphates inhibits FXII activation [273] while blocking the polyphosphate/FXII pathway protects against thromboembolism without promoting hemorrhage in mice [274]. It was therefore suggested that the polyphosphate/FXII axis could be targeted to decrease cancer-associated thrombosis [275].
Moreover, cancer cells and their secreted EVs promote the formation of neutrophil extracellular traps (NETs), which in addition to modulating cancer growth and metastasis, contribute to cancer-associated coagulopathy [109,276,277,278,279]. NETs represent a molecular trap that is formed by DNA chromatin, histones, and serine proteases in addition to other components, and these components, rather than intact NETs are suggested to drive NETosis-associated coagulopathies [280,281]. Peptidylarginine deiminase 4 (PAD4), an enzyme that mediates chromatin decondensation, was shown to regulate NETosis and coagulopathy where PAD4−/− mice exhibit impaired NETs formation [282,283,284,285]. While P-selectin was shown to promote NETosis through binding to PSGL-1 [286], fibrin and APC were shown to inhibit NETosis [287,288]. Activated platelets promote NETs formation, which forms a viscous cycle that propagates thrombus formation [289,290,291].
NETs are present in both arterial and venous thrombi of both human and experimental mice and were shown to promote platelet activation and aggregation in histone-dependent and independent pathways [289,292,293,294]. Increased markers of NETs formation were also detected in cancer patients and were associated with an increased risk of thrombosis [295,296,297,298,299]. Interestingly, NETs can entrap tumor-derived pro-coagulant EVs which amplifies the establishment of the prothrombotic state [266,295]. Indeed, mice orthotopically injected with 4T1 breast cancer cells exhibit enhanced exosome-induced NETs formation, which is associated with enhanced thrombosis [295]. Targeting NETs exerted an antithrombotic effect which further stresses the importance of NETs generation on thrombus formation [283,289,295]. Epithelial-to-mesenchymal transition, another hallmark of cancer, may also promote the release of TF-bearing EVs from tumor cells [263]. Although tumor-derived EVs activate platelets, the platelets play a fundamental role in accumulating these vesicles at the site of thrombosis through PSGL-1 in an EVs membrane integrins-dependent manner [266,270].

4.2.2. Extracellular Vesicles in the Sepsis-Associated Prothrombotic States

Sepsis is a major cause of thrombosis and organ failure with a mortality rate approaching 50%, where early diagnosis and treatment are pivotal for patient survival. Elevated levels of circulating procoagulant and proinflammatory EVs were documented in sepsis and were suggested to contribute to coagulation disorders and organ dysfunction and are correlated with mortality [300,301,302,303,304,305,306]. Several factors expressed on the vesicle surface including TF and PS, and bioactive substances contained in vesicles such as nucleosomes and high-mobility group box 1 (HMGB1), contribute to this prothrombotic state [307,308,309]. Although platelet-derived EVs represent the major population of EVs in sepsis [310,311], endothelial cells, monocytes, neutrophils, and red blood cells were extensively studied and shown to participate in TF-positive EV expression. Therefore, further investigation is still required to delineate the exact cellular source of septic coagulopathy [312,313].
Although the procoagulant role of these EVs in sepsis is starting to unveil, the lack of standardized assays and procedures for the characterization of these vesicles in sepsis yielded inconclusive results [312]. The upregulation of TF expression in EVs was evident in EVs released from activated monocytes [314], endothelial cells, and platelets, and predicts the severity of sepsis [315,316,317]. Likewise, Zhang et al. reported that endothelial cells treated with EV-containing serum from septic patients demonstrated increased levels of exposed PS in comparison to controls [318]. In another study, Tripisciano et al. induced the generation of thrombin in vesicle-free human plasma to which platelet-derived EVs were added [319]. Importantly, this was abrogated by the PS-binding annexin V but not by anti-TF antibodies. In sepsis, damage-associated molecular patterns (DAMPs) are released from activated and damaged cells and are transferred by EVs [320]. Platelet-derived EVs were shown to preferentially associate with circulating monocytes, contributing to sepsis-associated prothrombotic state [321,322].
Exosomes play an important role in coagulation and inflammation processes as well as organ dysfunction in sepsis. It was suggested that elevated exosome plasma level predicts the severity of organ failure and mortality in septic patients [323]. Moreover, another study demonstrated a differential, dysregulated miR profile in serum exosomes, total serum, and platelets of sepsis patients [324]. This compartment-specific expression of sepsis-associated miRs, especially in exosomes, was suggested as a novel biomarker in sepsis [325]. Platelets also activate NET formation during sepsis. Treating neutrophils with septic shock patient-derived exosomes significantly increased NET components and correlated positively with disease severity and outcome [326]. This observation was also confirmed in an in vivo model of sepsis. Mechanistically, exosomal HMGB1, miR-15b-5p and miR-378a-3p induced NET formation through the Akt/mTOR pathway [327]. It was also suggested that IκB kinase controls the secretion of platelet-derived exosomes in sepsis [327].

5. Conclusions and Future Perspectives

Major advances in our comprehension of coagulation pathways and hemostasis have evolved since the early observations of Marcello Malpighi in the seventeenth century. As hemostasis is defined through the balance of procoagulant and anticoagulant mechanisms, a tilted balance towards either mechanism results in pronounced diseases such as hemorrhage and thrombosis, the magnitude of which depends on the scale of balance disruption. Extracellular vesicles represent an exciting novel avenue in thrombosis research as cellular and platelet-derived extracellular vesicles mediate, at least in part, the procoagulant phenotype observed in cardiovascular disorders, cancer, and sepsis. Indeed, clinical and experimental evidence demonstrates that the procoagulant activity of these EVs extends well beyond their exposure to TF or PS, but encompasses cellular components implicated in the multiplication of coagulation surfaces and intercellular cross-linking at sites of vascular injury. Their pervasive presence in all body fluids including blood and urine allows for their promising utilization as fluid biopsies for the diagnosis and prognosis of diseases in which EVs abundance and content are altered.
Nevertheless, it should be noted that clinical evidence more often than not derives from studies employing relatively small patient cohorts, thus, validation in larger cohorts of patients is warranted. Additionally, distinct methodologies utilized in the isolation and characterization of EVs subtypes such as differential ultracentrifugation, size exclusion chromatography, and density gradient chromatography, yield inconsistent EV preparations with differential purity and composition, halting the collective translational potential of these clinical findings. It is therefore of utmost importance that future endeavors into characterizing the hemostatic roles of EVs occur in accordance with published recommendations and guidelines [5]. Another limitation is presented by our incomprehensive knowledge regarding the effects of concurrent antiplatelet and anticoagulant treatments on the abundance of cellular-derived and platelet-derived EVs and the alteration in their contents. Indeed, further investigation into these research avenues is warranted.
Promising evidence implicates EVs as a drug delivery system and the modulation of their cellular uptake as an avenue in the design of future therapeutics [327]. As such, EVs can be genetically engineered to display antithrombotic, fibrinolytic, or regenerative properties achieving targeted in vivo effects with minimal off-target uptake [328,329]. Nevertheless, and before these applications become available for clinical trials, assessments of their limitations such as cytotoxicity, uptake kinetics, and half-life are warranted [330,331]. Given the overwhelming evidence implicating EVs in the modulation of hemostatic processes, as biomarkers in procoagulant disorders, and as potential drug delivery systems, it becomes evident that advancing our knowledge concerning the mechanisms underpinning EV formation and their pathological relevance will enhance their translation into clinical practice.

Author Contributions

H.A.-K. and I.A. equally participated in the literature review and screening and contributed to manuscript writing. M.E.E.-S. developed the idea, supervised the work, reviewed and modified the manuscript draft, and provided research funding support. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We are grateful to Hiba El Hajj for her critical reading of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Kahn, M.J. Hypercoagulability as a cause of stroke in adults.(Featured cme topic: Stroke). South. Med. J. 2003, 96, 350–353. [Google Scholar] [CrossRef]
  2. Riddel, J.P., Jr.; Aouizerat, B.E.; Miaskowski, C.; Lillicrap, D.P. Theories of blood coagulation. J. Pediatr. Oncol. Nurs. 2007, 24, 123–131. [Google Scholar] [CrossRef] [PubMed]
  3. Fughelli, P.; Stella, A.; Sterpetti, A.V. Marcello malpighi (1628–1694) the revolution in medicine. Circ. Res. 2019, 124, 1430–1432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Palta, S.; Saroa, R.; Palta, A. Overview of the coagulation system. Indian J. Anaesth. 2014, 58, 515–523. [Google Scholar] [CrossRef] [PubMed]
  5. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Nawaz, M.; Shah, N.; Zanetti, B.R.; Maugeri, M.; Silvestre, R.N.; Fatima, F.; Neder, L.; Valadi, H. Extracellular vesicles and matrix remodeling enzymes: The emerging roles in extracellular matrix remodeling, progression of diseases and tissue repair. Cells 2018, 7, 167. [Google Scholar] [CrossRef] [Green Version]
  7. Van der Pol, E.; Böing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [Green Version]
  8. Femminò, S.; Penna, C.; Margarita, S.; Comità, S.; Brizzi, M.F.; Pagliaro, P. Extracellular vesicles and cardiovascular system: Biomarkers and cardioprotective effectors. Vasc. Pharmacol. 2020, 135, 106790. [Google Scholar] [CrossRef]
  9. Meletis, J.; Konstantopoulos, K. The beliefs, myths, and reality surrounding the word hema (blood) from Homer to the present. Anemia 2010, 2010, 1–6. [Google Scholar] [CrossRef] [Green Version]
  10. Tsoucalas, G.; Karamanou, M.; Papaioannou, T.G.; Sgantzos, M. Theories about blood coagulation in the writings of ancient Greek medico-philosophers. Curr. Pharm. Des. 2017, 23, 1275–1278. [Google Scholar] [CrossRef]
  11. Babington, B.G. Some considerations with respect to the blood, founded on one or two very simple experiments on that fluid. Med. Chirurgical Trans. 1831, 16, 293. [Google Scholar] [CrossRef] [Green Version]
  12. Owen, C.A. A history of blood coagulation. Mayo Found. Med. 2002, 287, 1051–1052. [Google Scholar]
  13. Douglas, S. Coagulation history, Oxford 1951–53. Br. J. Haematol. 1999, 107, 22–32. [Google Scholar] [CrossRef] [PubMed]
  14. Costa-Filho, R.; Hochleitner, G.; Wendt, M.; Teruya, A.; Spahn, D.R. Over 50 years of fibrinogen concentrate. Clin. Appl. Thromb. Hemost. 2016, 22, 109–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Schmidt, A. Neue untersuchungen über die faserstoffgerinnung. Arch. Physiol. Menschen Tiere 1872, 6, 413–538. [Google Scholar] [CrossRef]
  16. Morawitz, P. The Chemistry of Blood Coagulation [Die Chemie der Blutgerinnung]; Hartmann, R.; Guenter, P.F., Translators; Charles C Thomas: Springfield, IL, USA, 1958. [Google Scholar]
  17. Willebrand, E.V. Über hereditäre Pseudohämophilie. Acta Med. Scand. 1931, 76, 521–550. [Google Scholar] [CrossRef]
  18. Owren, P.A. The coagulation of blood; investigations on a new clotting factor. Acta Med. Scand. 1947, 194, 1–7. [Google Scholar]
  19. Alexander, B.; Goldstein, R.; Landwehr, G.; Cook, C.; Addelson, E.; Wilson, C. Congenital SPCA deficiency: A hitherto unrecognized coagulation defect with hemorrhage rectified by serum and serum fractions. J. Clin. Investig. 1951, 30, 596–608. [Google Scholar] [CrossRef]
  20. Patek, A.J.; Stetson, R.P.; Hemophilia, I. The abnormal coagulation of the blood and its relation to the blood platelets. J. Clin. Investig. 1936, 15, 531–542. [Google Scholar] [CrossRef] [Green Version]
  21. Aggeler, P.M.; White, S.G.; Glendening, M.B.; Page, E.W.; Leake, T.B.; Bates, G. Plasma throinboplastin component (PTC) deficiency: A new disease resembling hemophilia. Exp. Biol. Med. 1952, 79, 692–694. [Google Scholar] [CrossRef]
  22. Rosenthal, R.L.; Dreskin, O.H.; Rosenthal, N. New hemophilia-like disease caused by deficiency of a third plasma thromboplastin factor. Exp. Biol. Med. 1953, 82, 171–174. [Google Scholar] [CrossRef] [PubMed]
  23. Neuenschwander, P. Coagulation cascade: Overview. Encyclopedia of Respiratory Medicine; Laurent, G.J., Dshapiro, S., Eds.; Elsevier Ltd.: Oxford, UK, 2006. [Google Scholar]
  24. Davie, E.W.; Ratnoff, O.D. Waterfall sequence for intrinsic blood clotting. Science 1964, 145, 1310–1312. [Google Scholar] [CrossRef] [PubMed]
  25. Macfarlane, R. An enzyme cascade in the blood clotting mechanism, and its function as a biochemical amplifier. Nature 1964, 202, 498–499. [Google Scholar] [CrossRef]
  26. Versteeg, H.H.; Heemskerk, J.W.; Levi, M.; Reitsma, P.H. New fundamentals in hemostasis. Physiol. Rev. 2013, 93, 327–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Luchtman-Jones, L.; Broze, G.J. The current status of coagulation. Ann. Med. 1995, 27, 47–52. [Google Scholar] [CrossRef]
  28. Dam, H. The antihæmorrhagic vitamin of the chick.: Occurrence and chemical nature. Nature 1935, 135, 652–653. [Google Scholar] [CrossRef]
  29. Berkner, K.L. The vitamin K–dependent carboxylase. Annu. Rev. Nutr. 2005, 25, 127–149. [Google Scholar] [CrossRef]
  30. Hoffman, M.; Monroe, D.M., III. A cell-based model of hemostasis. Thromb. Haemost. 2001, 85, 958–965. [Google Scholar]
  31. Loscalzo, J.; Schafer, A.I. Thrombosis and Hemorrhage; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2003. [Google Scholar]
  32. Lippi, G.; Favaloro, E.J. Laboratory hemostasis: Milestones in clinical chemistry and laboratory medicine. Clin. Chem. Lab. Med. (CCLM) 2012, 51, 91–97. [Google Scholar] [CrossRef]
  33. Bonar, R.A.; Lippi, G.; Favaloro, E.J. Overview of hemostasis and thrombosis and contribution of laboratory testing to diagnosis and management of hemostasis and thrombosis disorders. In Hemostasis and Thrombosis; Springer: Berlin/Heidelberg, Germany, 2017; pp. 3–27. [Google Scholar]
  34. Yau, J.W.; Teoh, H.; Verma, S. Endothelial cell control of thrombosis. BMC Cardiovasc. Disord. 2015, 15, 130. [Google Scholar] [CrossRef] [Green Version]
  35. Gale, A.J. Current understanding of hemostasis. Toxicol. Pathol. 2010, 39, 273–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Broos, K.; Feys, H.B.; de Meyer, S.F.; Vanhoorelbeke, K.; Deckmyn, H. Platelets at work in primary hemostasis. Blood Rev. 2011, 25, 155–167. [Google Scholar] [CrossRef] [PubMed]
  37. Ruggeri, Z.M. The role of von Willebrand factor in thrombus formation. Thromb. Res. 2007, 120, S5–S9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Kehrel, B.; Wierwille, S.; Clemetson, K.J.; Anders, O.; Steiner, M.; Knight, C.G.; Farndale, R.W.; Okuma, M.; Barnes, M.J. Glycoprotein VI is a major collagen receptor for platelet activation: It recognizes the platelet-activating quaternary structure of collagen, whereas CD36, glycoprotein IIb/IIIa, and von willebrand factor do not. Blood J. Am. Soc. Hematol. 1998, 91, 491–499. [Google Scholar] [CrossRef]
  39. Nieswandt, B.; Brakebusch, C.; Bergmeier, W.; Schulte, V.; Bouvard, D.; Mokhtari-Nejad, R.; Lindhout, T.; Heemskerk, J.W.; Zirngibl, H.; Fässler, R. Glycoprotein VI but not α2β1 integrin is essential for platelet interaction with collagen. EMBO J. 2001, 20, 2120–2130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Luo, B.-H.; Springer, T.A. Integrin structures and conformational signaling. Curr. Opin. Cell Biol. 2006, 18, 579–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Varga-Szabo, D.; Pleines, I.; Nieswandt, B. Cell adhesion mechanisms in platelets. Arter. Thromb. Vasc. Biol. 2008, 28, 403–412. [Google Scholar] [CrossRef]
  42. Brass, L.; Furie, B.; Lijnen, H.; Esmon, C. Hematology: Basic Principles and Practice; Churchill-Livingston, Inc.: New York, NY, USA, 2000. [Google Scholar]
  43. Kirchhofer, D.; Nemerson, Y. Initiation of blood coagulation: The tissue factor/factor VIIa complex. Curr. Opin. Biotechnol. 1996, 7, 386–391. [Google Scholar] [CrossRef]
  44. Dahlbäck, B. Blood coagulation. Lancet 2000, 355, 1627–1632. [Google Scholar] [CrossRef]
  45. Boron, W.; Boulpaep, E. Medical Physiology: Updated Edition; Schmitt, W.R., Dudlick, M., Eds.; Elsevier Saunders Inc.: Philadelphia, PA, USA, 2005. [Google Scholar]
  46. Smith, S.A.; Travers, R.J.; Morrissey, J.H. How it all starts: Initiation of the clotting cascade. Crit. Rev. Biochem. Mol. Biol. 2015, 50, 326–336. [Google Scholar] [CrossRef] [Green Version]
  47. Lane, D.A.; Philippou, H.; Huntington, J.A. Directing thrombin. Blood 2005, 106, 2605–2612. [Google Scholar] [CrossRef]
  48. Kahn, M.L.; Zheng, Y.-W.; Huang, W.; Bigornia, V.; Zeng, D.; Moff, S.; Farese, R.V.; Tam, C.; Coughlin, S.R. A dual thrombin receptor system for platelet activation. Nature 1998, 394, 690–694. [Google Scholar] [CrossRef]
  49. Monroe, D.; Hoffman, M.; Roberts, H. Transmission of a procoagulant signal from tissue factor-bearing cells to platelets. Blood Coagul. Fibrinolysis 1996, 7, 459–464. [Google Scholar] [CrossRef]
  50. Díaz-Ricart, M.; Estebanell, E.; Lozano, M.; Aznar-Salatti, J.; White, J.G.; Ordinas, A.; Escolar, G. Thrombin facilitates primary platelet adhesion onto vascular surfaces in the absence of plasma adhesive proteins: Studies under flow conditions. Haematologica 2000, 85, 280–288. [Google Scholar]
  51. Hung, D.; Vu, T.-K.; Wheaton, V.; Ishii, K.; Coughlin, S. Cloned platelet thrombin receptor is necessary for thrombin-induced platelet activation. J. Clin. Investig. 1992, 89, 1350–1353. [Google Scholar] [CrossRef]
  52. Hultin, M.B. Modulation of thrombin-mediated activation of factor VIII: C by calcium ions, phospholipid, and platelets. Blood 1985, 66, 53–58. [Google Scholar] [CrossRef] [Green Version]
  53. Monkovic, D.D.; Tracy, P.B. Activation of human factor V by factor Xa and thrombin. Biochemistry 1990, 29, 1118–1128. [Google Scholar] [CrossRef]
  54. Oliver, J.A.; Monroe, D.M.; Roberts, H.R.; Hoffman, M. Thrombin activates factor XI on activated platelets in the absence of factor XII. Arter. Thromb. Vasc. Biol. 1999, 19, 170–177. [Google Scholar] [CrossRef] [Green Version]
  55. Baglia, F.A.; Walsh, P.N. Prothrombin is a cofactor for the binding of factor XI to the platelet surface and for platelet-mediated factor XI activation by thrombin. Biochemistry 1998, 37, 2271–2281. [Google Scholar] [CrossRef] [PubMed]
  56. Baglia, F.A.; Walsh, P.N. Thrombin-mediated feedback activation of factor XI on the activated platelet surface is preferred over contact activation by factor XIIa or factor XIa. J. Biol. Chem. 2000, 275, 20514–20519. [Google Scholar] [CrossRef] [Green Version]
  57. Khatib, R.; Wilson, F. Pharmacology of Medications Used in the Treatment of Atherosclerotic Cardiovascular Disease. In Refrence Module in Biomedical Sciences; Elsevier Ltd.: Oxford, UK, 2018; pp. 68–88. [Google Scholar]
  58. Wood, J.P.; Ellery, P.E.; Maroney, S.A.; Mast, A.E. Biology of tissue factor pathway inhibitor. Blood J. Am. Soc. Hematol. 2014, 123, 2934–2943. [Google Scholar] [CrossRef] [Green Version]
  59. Weisel, J.W.; Litvinov, R.I. Mechanisms of fibrin polymerization and clinical implications. Blood J. Am. Soc. Hematol. 2013, 121, 1712–1719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Muszbek, L.; Katona, É.; Kerényi, A. Assessment of factor XIII. In Hemostasis and Thrombosis; Springer: Berlin/Heidelberg, Germany, 2017; pp. 277–293. [Google Scholar]
  61. Szymanski, L.M.; Pate, R.R.; Durstine, J.L. Effects of maximal exercise and venous occlusion on fibrinolytic activity in physically active and inactive men. J. Appl. Physiol. 1994, 77, 2305–2310. [Google Scholar] [CrossRef]
  62. Cesarman-Maus, G.; Hajjar, K.A. Molecular mechanisms of fibrinolysis. Br. J. Haematol. 2005, 129, 307–321. [Google Scholar] [CrossRef]
  63. Hoylaerts, M.; Rijken, D.; Lijnen, H.; Collen, D. Kinetics of the activation of plasminogen by human tissue plasminogen activator. Role of fibrin. J. Biol. Chem. 1982, 257, 2912–2919. [Google Scholar] [CrossRef]
  64. Bajzar, L.; Manuel, R.; Nesheim, M.E. Purification and characterization of TAFI, a thrombin-activable fibrinolysis inhibitor. J. Biol. Chem. 1995, 270, 14477–14484. [Google Scholar] [CrossRef] [Green Version]
  65. Bouma, B.N.; Mosnier, L.O. Thrombin activatable fibrinolysis inhibitor (TAFI)—How does thrombin regulate fibrinolysis? Ann. Med. 2006, 38, 378–388. [Google Scholar] [CrossRef] [PubMed]
  66. Esmon, C.T. The protein C pathway. Chest 2003, 124, 26S–32S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Mosnier, L.O.; Meijers, J.C.; Bouma, B.N. Regulation of fibrinolysis in plasma by TAFI and protein C is dependent on the concentration of thrombomodulin. Thromb. Haemost. 2001, 85, 5–11. [Google Scholar] [PubMed]
  68. Taylor, F.B., Jr.; Peer, G.T.; Lockhart, M.S.; Ferrell, G.; Esmon, C.T. Endothelial cell protein C receptor plays an important role in protein C activation in vivo. Blood J. Am. Soc. Hematol. 2001, 97, 1685–1688. [Google Scholar] [CrossRef] [Green Version]
  69. Bajzar, L.; Nesheim, M.; Tracy, P. The profibrinolytic effect of activated protein C in clots formed from plasma is TAFI-dependent. Blood 1996, 88, 2093–2100. [Google Scholar] [CrossRef] [PubMed]
  70. Edlund, M. Menorrhagia—A symptom not sufficiently surveyed. The path to diagnosis and treatment lined with ambiguity and misunderstandings. Lakartidningen 2001, 98, 5505–5506, 5509–5510. [Google Scholar] [PubMed]
  71. Martlew, V. Peri-operative management of patients with coagulation disorders. Br. J. Anaesth. 2000, 85, 446–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Kurth, A.; Ludwig, G.; Scharrer, I. Prevalence, pathophysiology, diagnosis, and treatment of Willebrand syndrome in orthopedic-trauma patients. Der Orthop. 1999, 28, 366–374. [Google Scholar] [CrossRef]
  73. Van, M.W.; De, R.B.; Van, S.D.V.; Van, S.B. Vitamin K, an update for the paediatrician. Eur. J. Pediatr. 2009, 168, 127–134. [Google Scholar]
  74. Pinjala, R.; Reddy, L.; Nihar, R.; Praveen, G.; Sandeep, M. Thrombophilia–How Far and How Much to Investigate? Indian J. Surg. 2012, 74, 157–162. [Google Scholar] [CrossRef] [Green Version]
  75. Kyrle, P.A.; Rosendaal, F.R.; Eichinger, S. Risk assessment for recurrent venous thrombosis. Lancet 2010, 376, 2032–2039. [Google Scholar] [CrossRef]
  76. Ambruso, D.R.; Leonard, B.D.; Bies, R.D.; Jacobson, L.; Hathaway, W.E.; Reeve, E.B. Antithrombin III deficiency: Decreased synthesis of a biochemically normal molecule. Blood 1982, 60, 78–83. [Google Scholar] [CrossRef] [Green Version]
  77. Segers, K.; Dahlbäck, B.; Nicolaes, G.A. Coagulation factor V and thrombophilia: Background and mechanisms. Thromb. Haemost. 2007, 98, 530–542. [Google Scholar] [CrossRef] [Green Version]
  78. Koster, T.; Vandenbroucke, J.; Rosendaal, F.; de Ronde, H.; Briët, E.; Bertina, R.M. Venous thrombosis due to poor anticoagulant response to activated protein C: Leiden thrombophilia study. Lancet 1993, 342, 1503–1506. [Google Scholar] [CrossRef]
  79. Tait, R.; Walker, I.D.; Perry, D.; Islam, S.; Daly, M.; McCall, F.; Conkie, J.; Carrell, R. Prevalence of antithrombin deficiency in the healthy population. Br. J. Haematol. 1994, 87, 106–112. [Google Scholar] [CrossRef] [PubMed]
  80. Foster, D.C.; Yoshitake, S.; Davie, E.W. The nucleotide sequence of the gene for human protein C. Proc. Natl. Acad. Sci. USA 1985, 82, 4673–4677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Broekmans, A.W.; Veltkamp, J.J.; Bertina, R.M. Congenital protein C deficiency and venous thromboembolism: A study of three Dutch families. N. Engl. J. Med. 1983, 309, 340–344. [Google Scholar] [CrossRef] [PubMed]
  82. Reitsma, P.; Bernardi, F.; Doig, R.; Gandrille, S.; Greengard, J.; Ireland, H.; Krawczak, M.; Lind, B.; Long, G.; Poort, S. Protein C deficiency: A database of mutations, 1995 update. Thromb. Haemost. 1995, 74, 876–889. [Google Scholar] [CrossRef]
  83. Puhm, F.; Boilard, E.; Machlus, K.R. Platelet extracellular vesicles: Beyond the blood. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 87–96. [Google Scholar] [CrossRef]
  84. Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular vesicles in physiological and pathological conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef] [Green Version]
  85. Doyle, L.M.; Wang, M.Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef] [Green Version]
  86. Berckmans, R.J.; Nieuwland, R.; Böing, A.N.; Romijn, F.P.; Hack, C.E.; Sturk, A. Cell-derived microparticles circulate in healthy humans and support low grade thrombin generation. Thromb. Haemost. 2001, 85, 639–649. [Google Scholar]
  87. Berckmans, R.J.; Lacroix, R.; Hau, C.M.; Sturk, A.; Nieuwland, R. Extracellular vesicles and coagulation in blood from healthy humans revisited. J. Extracell. Vesicles 2019, 8, 1688936. [Google Scholar] [CrossRef]
  88. Das, K.; Keshava, S.; Ansari, S.A.; Kondreddy, V.; Esmon, C.T.; Griffin, J.H.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa induces extracellular vesicles from the endothelium: A potential mechanism for its hemostatic effect. Blood 2021, 137, 3428–3442. [Google Scholar] [CrossRef]
  89. Das, K.; Keshava, S.; Pendurthi, U.R.; Rao, L.V.M. Factor VIIa suppresses inflammation and barrier disruption through the release of EEVs and transfer of microRNA 10a. Blood 2022, 139, 118–133. [Google Scholar] [CrossRef] [PubMed]
  90. Nakatani, E.; Naito, Y.; Ishibashi, K.; Ohkura, N.; Atsumi, G.I. Extracellular vesicles derived from 3T3-L1 adipocytes enhance procoagulant activity. Biol. Pharm. Bull. 2022, 45, 178–183. [Google Scholar] [CrossRef] [PubMed]
  91. Marchini, J.F.; Manica, A.; Crestani, P.; Dutzmann, J.; Folco, E.J.; Weber, H.; Libby, P.; Croce, K. Oxidized low-density lipoprotein induces macrophage production of prothrombotic microparticles. J. Am. Heart Assoc. 2020, 9, e015878. [Google Scholar] [CrossRef] [PubMed]
  92. Dragovic, R.A.; Gardiner, C.; Brooks, A.S.; Tannetta, D.S.; Ferguson, D.J.; Hole, P.; Carr, B.; Redman, C.W.; Harris, A.L.; Dobson, P.J. Sizing and phenotyping of cellular vesicles using nanoparticle tracking analysis. Nanomed. Nanotechnol. Biol. Med. 2011, 7, 780–788. [Google Scholar] [CrossRef] [Green Version]
  93. Shet, A.S.; Aras, O.; Gupta, K.; Hass, M.J.; Rausch, D.J.; Saba, N.; Koopmeiners, L.; Key, N.S.; Hebbel, R.P. Sickle blood contains tissue factor–positive microparticles derived from endothelial cells and monocytes. Blood 2003, 102, 2678–2683. [Google Scholar] [CrossRef] [Green Version]
  94. Eitan, E.; Green, J.; Bodogai, M.; Mode, N.A.; Bæk, R.; Jørgensen, M.M.; Freeman, D.W.; Witwer, K.W.; Zonderman, A.B.; Biragyn, A. Age-related changes in plasma extracellular vesicle characteristics and internalization by leukocytes. Sci. Rep. 2017, 7, 1342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Forest, A.; Pautas, E.; Ray, P.; Bonnet, D.; Verny, M.; Amabile, N.; Boulanger, C.; Riou, B.; Tedgui, A.; Mallat, Z. Circulating microparticles and procoagulant activity in elderly patients. J. Gerontol. Ser. A Biomed. Sci. Med. Sci. 2010, 65, 414–420. [Google Scholar] [CrossRef]
  96. Galli, M.; Grassi, A.; Barbui, T. Platelet-derived microvesicles in thrombotic thrombocytopenic purpura and hemolytic uremic syndrome. Thromb. Haemost. 1996, 75, 427–431. [Google Scholar] [CrossRef]
  97. Tomer, A.; Harker, L.A.; Kasey, S.; Eckman, J.R. Thrombogenesis in sickle cell disease. J. Lab. Clin. Med. 2001, 137, 398–407. [Google Scholar] [CrossRef]
  98. Hughes, M.; Hayward, C.P.; Warkentin, T.E.; Horsewood, P.; Chorneyko, K.A.; Kelton, J.G. Morphological analysis of microparticle generation in heparin-induced thrombocytopenia. Blood J. Am. Soc. Hematol. 2000, 96, 188–194. [Google Scholar]
  99. Mallat, Z.; Benamer, H.; Hugel, B.; Benessiano, J.; Steg, P.G.; Freyssinet, J.-M.; Tedgui, A. Elevated levels of shed membrane microparticles with procoagulant potential in the peripheral circulating blood of patients with acute coronary syndromes. Circulation 2000, 101, 841–843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Lee, Y.J.; Jy, W.; Horstman, L.L.; Janania, J.; Reyes, Y.; Kelley, R.E.; Ahn, Y.S. Elevated platelet microparticles in transient ischemic attacks, lacunar infarcts, and multiinfarct dementias. Thromb. Res. 1993, 72, 295–304. [Google Scholar] [CrossRef]
  101. Jung, K.H.; Chu, K.; Lee, S.T.; Park, H.K.; Bahn, J.J.; Kim, D.H.; Kim, J.H.; Kim, M.; Lee, S.K.; Roh, J.K. Circulating endothelial microparticles as a marker of cerebrovascular disease. Ann. Neurol. Off. J. Am. Neurol. Assoc. Child Neurol. Soc. 2009, 66, 191–199. [Google Scholar] [CrossRef] [PubMed]
  102. Deng, F.; Wang, S.; Zhang, L. Endothelial microparticles act as novel diagnostic and therapeutic biomarkers of diabetes and its complications: A literature review. BioMed Res. Int. 2016, 2016, 9802026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Martínez, M.C.; Andriantsitohaina, R. Extracellular vesicles in metabolic syndrome. Circ. Res. 2017, 120, 1674–1686. [Google Scholar] [CrossRef]
  104. Alaaeddine, R.A.; AlZaim, I.; Hammoud, S.H.; Arakji, A.; Eid, A.H.; Abd-Elrahman, K.S.; El-Yazbi, A.F. The pleiotropic effects of antithrombotic drugs in the metabolic–cardiovascular–neurodegenerative disease continuum: Impact beyond reduced clotting. Clin. Sci. 2021, 135, 1015–1051. [Google Scholar] [CrossRef]
  105. Berezin, A.E.; Berezin, A.A. Extracellular vesicles and thrombogenicity in atrial fibrillation. Int. J. Mol. Sci. 2022, 23, 1774. [Google Scholar] [CrossRef]
  106. Mørk, M.; Andreasen, J.J.; Rasmussen, L.H.; Lip, G.Y.H.; Pedersen, S.; Bæk, R.; Jørgensen, M.M.; Kristensen, S.R. Elevated blood plasma levels of tissue factor-bearing extracellular vesicles in patients with atrial fibrillation. Thromb. Res. 2019, 173, 141–150. [Google Scholar] [CrossRef]
  107. Wang, H.; Yan, H.M.; Tang, M.X.; Wang, Z.H.; Zhong, M.; Zhang, Y.; Deng, J.T.; Zhang, W. Increased serum levels of microvesicles in nonvalvular atrial fibrillation determinated by ELISA using a specific monoclonal antibody AD-1. Clin. Chim. Acta 2010, 411, 1700–1704. [Google Scholar] [CrossRef]
  108. Wang, L.; Bi, Y.; Yu, M.; Li, T.; Tong, D.; Yang, X.; Zhang, C.; Guo, L.; Wang, C.; Kou, Y.; et al. Phosphatidylserine-exposing blood cells and microparticles induce procoagulant activity in non-valvular atrial fibrillation. Int. J. Cardiol. 2018, 258, 138–143. [Google Scholar] [CrossRef]
  109. Almeida, V.H.; Rondon, A.M.; Gomes, T.; Monteiro, R.Q. Novel aspects of extracellular vesicles as mediators of cancer-associated thrombosis. Cells 2019, 8, 716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Li, C.; Ni, Y.Q.; Xu, H.; Xiang, Q.Y.; Zhao, Y.; Zhan, J.K.; He, J.Y.; Li, S.; Liu, Y.S. Roles and mechanisms of exosomal non-coding RNAs in human health and diseases. Signal Transduct. Target. Ther. 2021, 6, 383. [Google Scholar] [CrossRef] [PubMed]
  111. Li, Y.; Wen, Q.; Chen, H.; Wu, X.; Liu, B.; Li, H.; Su, L.; Tong, H. Exosomes derived from heat stroke cases carry miRNAs associated with inflammation and coagulation cascade. Front. Immunol. 2021, 12, 624753. [Google Scholar] [CrossRef] [PubMed]
  112. Jansen, F.; Yang, X.; Hoelscher, M.; Cattelan, A.; Schmitz, T.; Proebsting, S.; Wenzel, D.; Vosen, S.; Franklin, B.S.; Fleischmann, B.K.; et al. Endothelial microparticle-mediated transfer of microRNA-126 promotes vascular endothelial cell repair via spred1 and is abrogated in glucose-damaged endothelial microparticles. Circulation 2013, 128, 2026–2038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Fu, S.; Zhang, Y.; Li, Y.; Luo, L.; Zhao, Y.; Yao, Y. Extracellular vesicles in cardiovascular diseases. Cell Death Discov. 2020, 6, 68. [Google Scholar] [CrossRef]
  114. Arroyo, A.B.; de los Reyes-García, A.M.; Teruel-Montoya, R.; Vicente, V.; González-Conejero, R.; Martínez, C. microRNAs in the haemostatic system: More than witnesses of thromboembolic diseases? Thromb. Res. 2018, 166, 1–9. [Google Scholar] [CrossRef]
  115. Tay, J.; Tiao, J.; Hughes, Q.; Jorritsma, J.; Gilmore, G.; Baker, R. Circulating microRNA as thrombosis sentinels: Caveats and considerations. Semin. Thromb. Hemost. 2018, 44, 206–215. [Google Scholar] [CrossRef]
  116. Wenche, J.; Horstman, L.L.; Arce, M.; Ahn, Y.S. Clinical significance of platelet microparticles in autoimmune thrombocytopenias. J. Lab. Clin. Med. 1992, 119, 334–345. [Google Scholar]
  117. 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 J. Am. Soc. Hematol. 2012, 119, 5285–5293. [Google Scholar] [CrossRef]
  118. Lapping-Carr, G.; Gemel, J.; Mao, Y.; Beyer, E.C. Circulating extracellular vesicles and endothelial damage in sickle cell disease. Front. Physiol. 2020, 11, 1063. [Google Scholar] [CrossRef]
  119. Nader, E.; Garnier, Y.; Connes, P.; Romana, M. Extracellular vesicles in sickle cell disease: Plasma concentration, blood cell types origin distribution and biological properties. Front. Med. 2021, 8, 728693. [Google Scholar] [CrossRef] [PubMed]
  120. Biasucci, L.M.; Porto, I.; di Vito, L.; de Maria, G.L.; Leone, A.M.; Tinelli, G.; Tritarelli, A.; di Rocco, G.; Snider, F.; Capogrossi, M.C. Differences in microparticle release in patients with acute coronary syndrome and stable angina. Circ. J. 2012, 76, 2174–2182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Sun, C.; Zhao, W.-B.; Chen, Y.; Hu, H.-Y. Higher plasma concentrations of platelet microparticles in patients with acute coronary syndrome: A systematic review and meta-analysis. Can. J. Cardiol. 2016, 32, 1325.e1–1325.e10. [Google Scholar] [CrossRef] [PubMed]
  122. Delabranche, X.; Boisramé-Helms, J.; Asfar, P.; Berger, A.; Mootien, Y.; Lavigne, T.; Grunebaum, L.; Lanza, F.; Gachet, C.; Freyssinet, J.-M. Microparticles are new biomarkers of septic shock-induced disseminated intravascular coagulopathy. Intensiv. Care Med. 2013, 39, 1695–1703. [Google Scholar] [CrossRef] [Green Version]
  123. Matsumoto, N.; Nomura, S.; Kamihata, H.; Kimura, Y.; Iwasaka, T. Increased level of oxidized LDL-dependent monocytederived microparticles in acute coronary syndrome. Thromb. Haemost. 2004, 91, 146–154. [Google Scholar] [CrossRef]
  124. Wang, B.; Li, T.; Han, X.; Li, Y.; Cheng, W.; Wang, L.; Lu, Z.; Yang, J.; Zhao, M. The level of circulating microparticles in patients with coronary heart disease: A systematic review and meta-analysis. J. Cardiovasc. Transl. Res. 2019, 13, 702–712. [Google Scholar] [CrossRef]
  125. Chirinos, J.A.; Heresi, G.A.; Velasquez, H.; Jy, W.; Jimenez, J.J.; Ahn, E.; Horstman, L.L.; Soriano, A.O.; Zambrano, J.P.; Ahn, Y.S. Elevation of endothelial microparticles, platelets, and leukocyte activation in patients with venous thromboembolism. J. Am. Coll. Cardiol. 2005, 45, 1467–1471. [Google Scholar] [CrossRef]
  126. Jamaly, S.; Basavaraj, M.G.; Starikova, I.; Olsen, R.; Brækkan, S.K.; Hansen, J.B. Elevated plasma levels of P-selectin glycoprotein ligand-1-positive microvesicles in patients with unprovoked venous thromboembolism. J. Thromb. Haemost. 2018, 16, 1546–1554. [Google Scholar] [CrossRef]
  127. Simak, J.; Gelderman, M.; Yu, H.; Wright, V.; Baird, A. Circulating endothelial microparticles in acute ischemic stroke: A link to severity, lesion volume and outcome. J. Thromb. Haemost. 2006, 4, 1296–1302. [Google Scholar] [CrossRef]
  128. Wiedmer, T.; Hall, S.; Ortel, T.; Kane, W.; Rosse, W.; Sims, P. Complement-induced vesiculation and exposure of membrane prothrombinase sites in platelets of paroxysmal nocturnal hemoglobinuria. Blood 1993, 82, 1192–1196. [Google Scholar] [CrossRef] [Green Version]
  129. Kozuma, Y.; Sawahata, Y.; Takei, Y.; Chiba, S.; Ninomiya, H. Procoagulant properties of microparticles released from red blood cells in paroxysmal nocturnal haemoglobinuria. Br. J. Haematol. 2011, 152, 631–639. [Google Scholar] [CrossRef]
  130. Suades, R.; Padró, T.; Crespo, J.; Ramaiola, I.; Martin-Yuste, V.; Sabaté, M.; Sans-Roselló, J.; Sionis, A.; Badimon, L. Circulating microparticle signature in coronary and peripheral blood of ST elevation myocardial infarction patients in relation to pain-to-PCI elapsed time. Int. J. Cardiol. 2016, 202, 378–387. [Google Scholar] [CrossRef] [PubMed]
  131. Morel, O.; Pereira, B.; Averous, G.; Faure, A.; Jesel, L.; Germain, P.; Grunebaum, L.; Ohlmann, P.; Freyssinet, J.-M.; Bareiss, P. Increased levels of procoagulant tissue factor-bearing microparticles within the occluded coronary artery of patients with ST-segment elevation myocardial infarction: Role of endothelial damage and leukocyte activation. Atherosclerosis 2009, 204, 636–641. [Google Scholar] [CrossRef] [PubMed]
  132. Bal, L.; Ederhy, S.; di Angelantonio, E.; Toti, F.; Zobairi, F.; Dufaitre, G.; Meuleman, C.; Mallat, Z.; Boccara, F.; Tedgui, A. Factors influencing the level of circulating procoagulant microparticles in acute pulmonary embolism. Arch. Cardiovasc. Dis. 2010, 103, 394–403. [Google Scholar] [CrossRef] [Green Version]
  133. Bal, L.; Ederhy, S.; di Angelantonio, E.; Toti, F.; Zobairi, F.; Dufaitre, G.; Meuleman, C.; Mallat, Z.; Boccara, F.; Tedgui, A. Circulating procoagulant microparticles in acute pulmonary embolism: A case-control study. Int. J. Cardiol. 2010, 145, 321–322. [Google Scholar] [CrossRef]
  134. Wolf, P. The nature and significance of platelet products in human plasma. Br. J. Haematol. 1967, 13, 269–288. [Google Scholar] [CrossRef]
  135. Davidson, S.M.; Andreadou, I.; Barile, L.; Birnbaum, Y.; Cabrera-Fuentes, H.A.; Cohen, M.V.; Downey, J.M.; Girao, H.; Pagliaro, P.; Penna, C. Circulating blood cells and extracellular vesicles in acute cardioprotection. Cardiovasc. Res. 2019, 115, 1156–1166. [Google Scholar] [CrossRef]
  136. Sims, P.J.; Faioni, E.; Wiedmer, T.; Shattil, S. Complement proteins C5b-9 cause release of membrane vesicles from the platelet surface that are enriched in the membrane receptor for coagulation factor Va and express prothrombinase activity. J. Biol. Chem. 1988, 263, 18205–18212. [Google Scholar] [CrossRef]
  137. Giesen, P.L.; Rauch, U.; Bohrmann, B.; Kling, D.; Roqué, M.; Fallon, J.T.; Badimon, J.J.; Himber, J.; Riederer, M.A.; Nemerson, Y. Blood-borne tissue factor: Another view of thrombosis. Proc. Natl. Acad. Sci. USA 1999, 96, 2311–2315. [Google Scholar] [CrossRef] [Green Version]
  138. VanWijk, M.J.; VanBavel, E.; Sturk, A.; Nieuwland, R. Microparticles in cardiovascular diseases. Cardiovasc. Res. 2003, 59, 277–287. [Google Scholar] [CrossRef]
  139. Zifkos, K.; Dubois, C.; Schäfer, K. Extracellular vesicles and thrombosis: Update on the clinical and experimental evidence. Int. J. Mol. Sci. 2021, 22, 9317. [Google Scholar] [CrossRef]
  140. Ferreira, P.M.; Bozbas, E.; Tannetta, S.D.; Alroqaiba, N.; Zhou, R.; Crawley, J.T.B.; Gibbins, J.M.; Jones, C.I.; Ahnström, J.; Yaqoob, P. Mode of induction of platelet-derived extracellular vesicles is a critical determinant of their phenotype and function. Sci. Rep. 2020, 10, 18061. [Google Scholar] [CrossRef] [PubMed]
  141. Tucher, C.; Bode, K.; Schiller, P.; Claßen, L.; Birr, C.; Souto-Carneiro, M.M.; Blank, N.; Lorenz, H.-M.; Schiller, M. Extracellular vesicle subtypes released from activated or apoptotic T-lymphocytes carry a specific and stimulus-dependent protein cargo. Front. Immunol. 2018, 9, 534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Dasgupta, S.K.; Abdel-Monem, H.; Niravath, P.; Le, A.; Bellera, R.V.; Langlois, K.; Nagata, S.; Rumbaut, R.E.; Thiagarajan, P. Lactadherin and clearance of platelet-derived microvesicles. Blood J. Am. Soc. Hematol. 2009, 113, 1332–1339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Mulcahy, L.A.; Pink, R.C.; Carter, D.R.F. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tushuizen, M.E.; Diamant, M.; Sturk, A.; Nieuwland, R. Cell-derived microparticles in the pathogenesis of cardiovascular disease: Friend or foe? Arterioscler. Thromb. Vasc. Biol. 2011, 31, 4–9. [Google Scholar] [CrossRef] [Green Version]
  145. Flaumenhaft, R.; Mairuhu, A.T.; Italiano, J.E. Platelet-and megakaryocyte-derived microparticles. In Seminars in Thrombosis and Hemostasis; Thieme Medical Publishers: New York, NY, USA, 2010. [Google Scholar]
  146. Suades, R.; Padró, T.; Vilahur, G.; Badimon, L. Circulating and platelet-derived microparticles in human blood enhance thrombosis on atherosclerotic plaques. Thromb. Haemost. 2012, 108, 1208–1219. [Google Scholar] [CrossRef]
  147. Kay, J.G.; Grinstein, S. Phosphatidylserine-mediated cellular signaling. Lipid-Mediat. Protein Signal. 2013, 991, 177–193. [Google Scholar]
  148. Reddy, E.C.; Rand, M.L. Procoagulant phosphatidylserine-exposing platelets in vitro and in vivo. Front. Cardiovasc. Med. 2020, 7, 15. [Google Scholar] [CrossRef] [Green Version]
  149. Yang, A.; Chen, F.; He, C.; Zhou, J.; Lu, Y.; Dai, J.; Birge, R.B.; Wu, Y. The procoagulant activity of apoptotic cells is mediated by interaction with factor XII. Front. Immunol. 2017, 8, 1188. [Google Scholar] [CrossRef] [Green Version]
  150. Boulanger, C.M.; Loyer, X.; Rautou, P.-E.; Amabile, N. Extracellular vesicles in coronary artery disease. Nat. Rev. Cardiol. 2017, 14, 259–272. [Google Scholar] [CrossRef] [PubMed]
  151. Sluijter, J.P.G.; Davidson, S.M.; Boulanger, C.M.; Buzas, E.I.; de Kleijn, D.P.V.; Engel, F.B.; Giricz, Z.; Hausenloy, D.J.; Kishore, R.; Lecour, S. Extracellular vesicles in diagnostics and therapy of the ischaemic heart: Position paper from the working group on cellular biology of the heart of the European society of cardiology. Cardiovasc. Res. 2018, 114, 19–34. [Google Scholar] [CrossRef] [PubMed]
  152. Preston, R.A.; Jy, W.; Jimenez, J.J.; Mauro, L.M.; Horstman, L.L.; Valle, M.; Aime, G.; Ahn, Y.S. Effects of severe hypertension on endothelial and platelet microparticles. Hypertension 2003, 41, 211–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Biro, E.; Sturk-Maquelin, K.; Vogel, G.; Meuleman, D.; Smit, M.; Hack, C.; Sturk, A.; Nieuwland, R. Human cell-derived microparticles promote thrombus formation in vivo in a tissue factor-dependent manner. J. Thromb. Haemost. 2003, 1, 2561–2568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Falati, S.; Liu, Q.; Gross, P.; Merrill-Skoloff, G.; Chou, J.; Vandendries, E.; Celi, A.; Croce, K.; Furie, B.C.; Furie, B. Accumulation of tissue factor into developing thrombi in vivo is dependent upon microparticle P-selectin glycoprotein ligand 1 and platelet P-selectin. J. Exp. Med. 2003, 197, 1585–1598. [Google Scholar] [CrossRef]
  155. Kou, Y.; Zou, L.; Liu, R.; Zhao, X.; Wang, Y.; Zhang, C.; Dong, Z.; Kou, J.; Bi, Y.; Fu, L. Intravascular cells and circulating microparticles induce procoagulant activity via phosphatidylserine exposure in heart failure. J. Thromb. Thrombolysis 2019, 48, 187–194. [Google Scholar] [CrossRef]
  156. Chiva-Blanch, G.; Laake, K.; Myhre, P.; Bratseth, V.; Arnesen, H.; Solheim, S.; Badimon, L.; Seljeflot, I. Platelet-, monocyte-derived and tissue factor-carrying circulating microparticles are related to acute myocardial infarction severity. PLoS ONE 2017, 12, e0172558. [Google Scholar] [CrossRef]
  157. Wang, L.; Bi, Y.; Cao, M.; Ma, R.; Wu, X.; Zhang, Y.; Ding, W.; Liu, Y.; Yu, Q.; Zhang, Y. Microparticles and blood cells induce procoagulant activity via phosphatidylserine exposure in NSTEMI patients following stent implantation. Int. J. Cardiol. 2016, 223, 121–128. [Google Scholar] [CrossRef]
  158. Ueba, T.; Nomura, S.; Inami, N.; Nishikawa, T.; Kajiwara, M.; Iwata, R.; Yamashita, K. Plasma level of platelet-derived microparticles is associated with coronary heart disease risk score in healthy men. J. Atheroscler. Thromb. 2010, 17, 342–349. [Google Scholar] [CrossRef] [Green Version]
  159. Zhao, L.; Wu, X.; Si, Y.; Yao, Z.; Dong, Z.; Novakovic, V.A.; Guo, L.; Tong, D.; Chen, H.; Bi, Y. Increased blood cell phosphatidylserine exposure and circulating microparticles contribute to procoagulant activity after carotid artery stenting. J. Neurosurg. 2017, 127, 1041–1054. [Google Scholar] [CrossRef] [Green Version]
  160. Nylander, S.; Femia, E.; Scavone, M.; Berntsson, P.; Asztély, A.K.; Nelander, K.; Löfgren, L.; Nilsson, R.; Cattaneo, M. Ticagrelor inhibits human platelet aggregation via adenosine in addition to P2Y12 antagonism. J. Thromb. Haemost. 2013, 11, 1867–1876. [Google Scholar] [CrossRef]
  161. Bernal-Mizrachi, L.; Jy, W.; Jimenez, J.J.; Pastor, J.; Mauro, L.M.; Horstman, L.L.; de Marchena, E.; Ahn, Y.S. High levels of circulating endothelial microparticles in patients with acute coronary syndromes. Am. Heart J. 2003, 145, 962–970. [Google Scholar] [CrossRef]
  162. Lipets, E.; Antonova, O.; Shustova, O.; Losenkova, K.; Mazurov, A.; Ataullakhanov, F. Use of Thrombodynamics for revealing the participation of platelet, erythrocyte, endothelial, and monocyte microparticles in coagulation activation and propagation. PLoS ONE 2020, 15, e0227932. [Google Scholar] [CrossRef] [PubMed]
  163. Andrews, A.M.; Rizzo, V. Microparticle-induced activation of the vascular endothelium requires caveolin-1/caveolae. PLoS ONE 2016, 11, e0149272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Shustova, O.N.; Antonova, O.A.; Golubeva, N.V.; Khaspekova, S.G.; Yakushkin, V.V.; Aksuk, S.A.; Alchinova, I.B.; Karganov, M.Y.; Mazurov, A.V. Differential procoagulant activity of microparticles derived from monocytes, granulocytes, platelets and endothelial cells: Impact of active tissue factor. Blood Coagul. Fibrinolysis 2017, 28, 373–382. [Google Scholar] [CrossRef]
  165. Van Der Meijden, P.; van Schilfgaarde, M.; van Oerle, R.; Renne, T.; Cate, H.T.; Spronk, H. Platelet-and erythrocyte-derived microparticles trigger thrombin generation via factor XIIa. J. Thromb. Haemost. 2012, 10, 1355–1362. [Google Scholar] [CrossRef] [PubMed]
  166. Aleman, M.M.; Gardiner, C.; Harrison, P.; Wolberg, A.S. Differential contributions of monocyte-and platelet-derived microparticles towards thrombin generation and fibrin formation and stability. J. Thromb. Haemost. 2011, 9, 2251–2261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Hohensinner, P.J.; Mayer, J.; Kichbacher, J.; Kral-Pointner, J.; Thaler, B.; Kaun, C.; Hell, L.; Haider, P.; Mussbacher, M.; Schmid, J.A.; et al. Alternative activation of human macrophages enhances tissue factor expression and production of extracellular vesicles. Haematologica 2021, 106, 454–463. [Google Scholar] [CrossRef] [Green Version]
  168. George, J.N.; Thoi, L.L.; McManus, L.M.; Reimann, T.A. Isolation of human platelet membrane microparticles from plasma and serum. Blood 1982, 60, 834–840. [Google Scholar] [CrossRef] [Green Version]
  169. George, J.N.; Pickett, E.B.; Saucerman, S.; McEver, R.P.; Kunicki, T.J.; Kieffer, N.; Newman, P.J. Platelet surface glycoproteins. Studies on resting and activated platelets and platelet membrane microparticles in normal subjects, and observations in patients during adult respiratory distress syndrome and cardiac surgery. J. Clin. Investig. 1986, 78, 340–348. [Google Scholar] [CrossRef]
  170. Italiano, J.E., Jr.; Mairuhu, A.T.; Flaumenhaft, R. Clinical relevance of microparticles from platelets and megakaryocytes. Curr. Opin. Hematol. 2010, 17, 578–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Flaumenhaft, R. Formation and fate of platelet microparticles. Blood Cells Mol. Dis. 2006, 36, 182–187. [Google Scholar] [CrossRef] [PubMed]
  172. Flaumenhaft, R.; Dilks, J.R.; Richardson, J.; Alden, E.; Patel-Hett, S.R.; Battinelli, E.; Klement, G.L.; Sola-Visner, M.; Italiano, J.E., Jr. Megakaryocyte-derived microparticles: Direct visualization and distinction from platelet-derived microparticles. Blood J. Am. Soc. Hematol. 2009, 113, 1112–1121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. van der Zee, P.M.; Biró, E.; Ko, Y.; de Winter, R.J.; Hack, C.E.; Sturk, A.; Nieuwland, R. P-selectin- and CD63-exposing platelet microparticles reflect platelet activation in peripheral arterial disease and myocardial infarction. Clin. Chem. 2006, 52, 657–664. [Google Scholar] [CrossRef] [PubMed]
  174. Fackler, O.T.; Grosse, R. Cell motility through plasma membrane blebbing. J. Cell Biol. 2008, 181, 879–884. [Google Scholar] [CrossRef] [Green Version]
  175. Kim, H.K.; Song, K.S.; Chung, J.H.; Lee, K.R.; Lee, S.N. Platelet microparticles induce angiogenesis in vitro. Br. J. Haematol. 2004, 124, 376–384. [Google Scholar] [CrossRef]
  176. Stępień, E.; Stankiewicz, E.; Zalewski, J.; Godlewski, J.; Zmudka, K.; Wybrańska, I. Number of microparticles generated during acute myocardial infarction and stable angina correlates with platelet activation. Arch. Med. Res. 2012, 43, 31–35. [Google Scholar] [CrossRef]
  177. Gąsecka, A.; Rogula, S.; Eyileten, C.; Postuła, M.; Jaguszewski, M.J.; Kochman, J.; Mazurek, T.; Nieuwland, R.; Filipiak, K.J. Role of P2Y receptors in platelet extracellular vesicle release. Int. J. Mol. Sci. 2020, 21, 6065. [Google Scholar] [CrossRef]
  178. Ponomareva, A.; Nevzorova, T.; Mordakhanova, E.; Andrianova, I.; Rauova, L.; Litvinov, R.; Weisel, J. Intracellular origin and ultrastructure of platelet-derived microparticles. J. Thromb. Haemost. 2017, 15, 1655–1667. [Google Scholar] [CrossRef] [Green Version]
  179. Aatonen, M.T.; Öhman, T.; Nyman, T.A.; Laitinen, S.; Grönholm, M.; Siljander, P.R.-M. Isolation and characterization of platelet-derived extracellular vesicles. J. Extracell. Vesicles 2014, 3, 24692. [Google Scholar] [CrossRef]
  180. Siljander, P.; Carpen, O.; Lassila, R. Platelet-derived microparticles associate with fibrin during thrombosis. Blood 1996, 87, 4651–4663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Brisson, A.R.; Tan, S.; Linares, R.; Gounou, C.; Arraud, N. Extracellular vesicles from activated platelets: A semiquantitative cryo-electron microscopy and immuno-gold labeling study. Platelets 2017, 28, 263–271. [Google Scholar] [CrossRef] [PubMed]
  182. Goetzl, E.J.; Goetzl, L.; Karliner, J.S.; Tang, N.; Pulliam, L. Human plasma platelet-derived exosomes: Effects of aspirin. FASEB J. 2016, 30, 2058–2063. [Google Scholar] [CrossRef] [Green Version]
  183. Connor, D.E.; Ly, K.; Aslam, A.; Boland, J.; Low, J.; Jarvis, S.; Muller, D.W.; Joseph, J.E. Effects of antiplatelet therapy on platelet extracellular vesicle release and procoagulant activity in health and in cardiovascular disease. Platelets 2016, 27, 805–811. [Google Scholar] [CrossRef] [PubMed]
  184. Gasecka, A.; Nieuwland, R.; van der Pol, E.; Hajji, N.; Ćwiek, A.; Pluta, K.; Konwerski, M.; Filipiak, K.J. P2Y12 antagonist ticagrelor inhibits the release of procoagulant extracellular vesicles from activated platelets. Cardiol. J. 2020, 26, 782–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Christersson, C.; Johnell, M.; Siegbahn, A. The influence of direct thrombin inhibitors on the formation of platelet-leukocyte aggregates and tissue factor expression. Thromb. Res. 2010, 126, e327–e333. [Google Scholar] [CrossRef] [PubMed]
  186. Pontiggia, L.; Steiner, B.; Ulrichts, H.; Deckmyn, H.; Forestier, M.; Beer, J.H. Platelet microparticle formation and thrombin generation under high shear are effectively suppressed by a monoclonal antibody against GPIbα. Thromb. Haemost. 2006, 96, 774–780. [Google Scholar] [CrossRef] [PubMed]
  187. Shantsila, E.; Kamphuisen, P.W.; Lip, G.Y. Circulating microparticles in cardiovascular disease: Implications for atherogenesis and atherothrombosis. J. Thromb. Haemost. 2010, 8, 2358–2368. [Google Scholar] [CrossRef]
  188. Brill, A.; Dashevsky, O.; Rivo, J.; Gozal, Y.; Varon, D. Platelet-derived microparticles induce angiogenesis and stimulate post-ischemic revascularization. Cardiovasc. Res. 2005, 67, 30–38. [Google Scholar] [CrossRef]
  189. Burnier, L.; Fontana, P.; Kwak, B.R.; Angelillo-Scherrer, A. Cell-derived microparticles in haemostasis and vascular medicine. Thromb. Haemost. 2009, 101, 439–451. [Google Scholar] [CrossRef]
  190. Castaman, G.; Yu-Feng, L.; Battistin, E.; Rodeghiero, F. Characterization of a novel bleeding disorder with isolated prolonged bleeding time and deficiency of platelet microvesicle generation. Br. J. Haematol. 1997, 96, 458–463. [Google Scholar] [CrossRef] [PubMed]
  191. Gemmell, C.; Sefton, M.; Yeo, E. Platelet-derived microparticle formation involves glycoprotein IIb-IIIa. Inhibition by RGDS and a Glanzmann’s thrombasthenia defect. J. Biol. Chem. 1993, 268, 14586–14589. [Google Scholar] [CrossRef]
  192. Fujii, T.; Sakata, A.; Nishimura, S.; Eto, K.; Nagata, S. TMEM16F is required for phosphatidylserine exposure and microparticle release in activated mouse platelets. Proc. Natl. Acad. Sci. USA 2015, 112, 12800–12805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Diehl, P.; Fricke, A.; Sander, L.; Stamm, J.; Bassler, N.; Htun, N.; Ziemann, M.; Helbing, T.; El-Osta, A.; Jowett, J.B.; et al. Microparticles: Major transport vehicles for distinct microRNAs in circulation. Cardiovasc. Res. 2012, 93, 633–644. [Google Scholar] [CrossRef] [PubMed]
  194. Hunter, M.P.; Ismail, N.; Zhang, X.; Aguda, B.D.; Lee, E.J.; Yu, L.; Xiao, T.; Schafer, J.; Lee, M.L.; Schmittgen, T.D.; et al. Detection of microRNA expression in human peripheral blood microvesicles. PLoS ONE 2008, 3, e3694. [Google Scholar] [CrossRef] [Green Version]
  195. De Rosa, S.; Fichtlscherer, S.; Lehmann, R.; Assmus, B.; Dimmeler, S.; Zeiher, A.M. Transcoronary concentration gradients of circulating microRNAs. Circulation 2011, 124, 1936–1944. [Google Scholar] [CrossRef] [Green Version]
  196. Kuwabara, Y.; Ono, K.; Horie, T.; Nishi, H.; Nagao, K.; Kinoshita, M.; Watanabe, S.; Baba, O.; Kojima, Y.; Shizuta, S. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ. Cardiovasc. Genet. 2011, 4, 446–454. [Google Scholar] [CrossRef]
  197. Qu, M.; Zou, X.; Fang, F.; Wang, S.; Xu, L.; Zeng, Q.; Fan, Z.; Chen, L.; Yue, W.; Xie, X.; et al. Platelet-derived microparticles enhance megakaryocyte differentiation and platelet generation via miR-1915-3p. Nat. Commun. 2020, 11, 4964. [Google Scholar] [CrossRef]
  198. Harding, C.; Heuser, J.; Stahl, P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 1983, 97, 329–339. [Google Scholar] [CrossRef]
  199. Johnstone, R.; Mathew, A.; Mason, A.; Teng, K. Exosome formation during maturation of mammalian and avian reticulocytes: Evidence that exosome release is a major route for externalization of obsolete membrane proteins. J. Cell. Physiol. 1991, 147, 27–36. [Google Scholar] [CrossRef]
  200. Lima, L.G.; Oliveira, A.S.; Campos, L.C.; Bonamino, M.; Chammas, R.; Werneck, C.C.; Vicente, C.P.; Barcinski, M.A.; Petersen, L.C.; Monteiro, R.Q. Malignant transformation in melanocytes is associated with increased production of procoagulant microvesicles. Thromb. Haemost. 2011, 106, 712–723. [Google Scholar] [CrossRef] [PubMed]
  201. Rak, J. Microparticles in cancer. In Seminars in Thrombosis and Hemostasis; Thieme Medical Publishers: New York, NY, USA, 2010. [Google Scholar]
  202. Feigelson, S.W.; Grabovsky, V.; Shamri, R.; Levy, S.; Alon, R. The CD81 tetraspanin facilitates instantaneous leukocyte VLA-4 adhesion strengthening to vascular cell adhesion molecule 1 (VCAM-1) under shear flow. J. Biol. Chem. 2003, 278, 51203–51212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Reyes, R.; Monjas, A.; Yánez-Mó, M.; Cardeñes, B.; Morlino, G.; Gilsanz, A.; Machado-Pineda, Y.; Lafuente, E.; Monk, P.; Sánchez-Madrid, F. Different states of integrin LFA-1 aggregation are controlled through its association with tetraspanin CD9. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2015, 1853, 2464–2480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Dasgupta, S.K.; Le, A.; Chavakis, T.; Rumbaut, R.E.; Thiagarajan, P. Developmental endothelial locus-1 (Del-1) mediates clearance of platelet microparticles by the endothelium. Circulation 2012, 125, 1664–1672. [Google Scholar] [CrossRef] [Green Version]
  205. Pegtel, D.M.; Gould, S.J. Exosomes. Ann. Rev. Biochem. 2019, 88, 487–514. [Google Scholar] [CrossRef] [PubMed]
  206. Théry, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
  207. Takahashi, A.; Okada, R.; Nagao, K.; Kawamata, Y.; Hanyu, A.; Yoshimoto, S.; Takasugi, M.; Watanabe, S.; Kanemaki, M.T.; Obuse, C. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells. Nat. Commun. 2017, 8, 15287. [Google Scholar] [CrossRef] [Green Version]
  208. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [Green Version]
  209. Hyenne, V.; Apaydin, A.; Rodriguez, D.; Spiegelhalter, C.; Hoff-Yoessle, S.; Diem, M.; Tak, S.; Lefebvre, O.; Schwab, Y.; Goetz, J.G. RAL-1 controls multivesicular body biogenesis and exosome secretion. J. Cell Biol. 2015, 211, 27–37. [Google Scholar] [CrossRef] [Green Version]
  210. Patel, B.; Patel, J.; Cho, J.; Manne, S.; Bonala, S.; Henske, E.; Roegiers, F.; Markiewski, M.; Karbowniczek, M. Exosomes mediate the acquisition of the disease phenotypes by cells with normal genome in tuberous sclerosis complex. Oncogene 2016, 35, 3027–3036. [Google Scholar] [CrossRef]
  211. Akbar, N.; Digby, J.E.; Cahill, T.J.; Tavare, A.N.; Corbin, A.L.; Saluja, S.; Dawkins, S.; Edgar, L.; Rawlings, N.; Ziberna, K. Endothelium-derived extracellular vesicles promote splenic monocyte mobilization in myocardial infarction. JCI Insight 2017, 2, e93344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Lee, H.M.; Choi, E.-J.; Kim, J.H.; Kim, T.D.; Kim, Y.-K.; Kang, C.; Gho, Y.S. A membranous form of ICAM-1 on exosomes efficiently blocks leukocyte adhesion to activated endothelial cells. Biochem. Biophys. Res. Commun. 2010, 397, 251–256. [Google Scholar] [CrossRef] [PubMed]
  213. Van Niel, G.; d’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef] [PubMed]
  214. Toth, B.; Lok, C.A.; Böing, A.; Diamant, M.; van der Post, J.A.; Friese, K.; Nieuwland, R. Microparticles and exosomes: Impact on normal and complicated pregnancy. Am. J. Reprod. Immunol. 2007, 58, 389–402. [Google Scholar] [CrossRef] [PubMed]
  215. Azevedo, L.C.P.; Janiszewski, M.; Pontieri, V.; Pedro, M.d.; Bassi, E.; Tucci, P.J.F.; Laurindo, F.R.M. Platelet-derived exosomes from septic shock patients induce myocardial dysfunction. Crit. Care 2007, 11, R120. [Google Scholar] [CrossRef] [Green Version]
  216. Srikanthan, S.; Li, W.; Silverstein, R.L.; McIntyre, T.M. Exosome poly-ubiquitin inhibits platelet activation, downregulates CD 36 and inhibits pro-atherothombotic cellular functions. J. Thromb. Haemost. 2014, 12, 1906–1917. [Google Scholar] [CrossRef] [Green Version]
  217. Li, J.; Tan, M.; Xiang, Q.; Zhou, Z.; Yan, H. Thrombin-activated platelet-derived exosomes regulate endothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb. Res. 2017, 154, 96–105. [Google Scholar] [CrossRef]
  218. Tan, M.; Yan, H.-B.; Li, J.-N.; Li, W.-K.; Fu, Y.-Y.; Chen, W.; Zhou, Z. Thrombin stimulated platelet-derived exosomes inhibit platelet-derived growth factor receptor-beta expression in vascular smooth muscle cells. Cell. Physiol. Biochem. 2016, 38, 2348–2365. [Google Scholar] [CrossRef]
  219. Wang, H.; Wang, Q.; Kleiman, K.; Guo, C.; Eitzman, D.T. Hematopoietic deficiency of miR-223 attenuates thrombosis in response to photochemical injury in mice. Sci. Rep. 2017, 7, 1606. [Google Scholar] [CrossRef]
  220. Marketou, M.; Kontaraki, J.; Papadakis, J.; Kochiadakis, G.; Vrentzos, G.; Maragkoudakis, S.; Fragkiadakis, K.; Katsouli, E.; Plataki, M.; Patrianakos, A.; et al. Platelet microRNAs in hypertensive patients with and without cardiovascular disease. J. Hum. Hypertens. 2018, 33, 149–156. [Google Scholar] [CrossRef]
  221. Alexandru, N.; Constantin, A.; Nemecz, M.; Comarita, I.K.; Vilcu, A.; Procopciuc, A.; Tanko, G.; Georgescu, A. Hypertension associated with hyperlipidemia induced different microRNA expression profiles in plasma, platelets, and platelet-derived microvesicles; effects of endothelial progenitor cell therapy. Front. Med. 2019, 6, 280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  222. Lazar, S.; Wurtzel, J.G.T.; Chen, X.; Ma, P.; Goldfinger, L.E. High-efficiency unassisted transfection of platelets with naked double-stranded miRNAs modulates signal-activated translation and platelet function. Platelets 2021, 32, 794–806. [Google Scholar] [CrossRef] [PubMed]
  223. Pienimaeki-Roemer, A.; Konovalova, T.; Musri, M.M.; Sigruener, A.; Boettcher, A.; Meister, G.; Schmitz, G. Transcriptomic profiling of platelet senescence and platelet extracellular vesicles. Transfusion 2017, 57, 144–156. [Google Scholar] [CrossRef]
  224. Konkoth, A.; Saraswat, R.; Dubrou, C.; Sabatier, F.; Leroyer, A.S.; Lacroix, R.; Duchez, A.C.; Dignat-George, F. Multifaceted role of extracellular vesicles in atherosclerosis. Atherosclerosis 2021, 319, 121–131. [Google Scholar] [CrossRef] [PubMed]
  225. Williams, J.O.; Whelan, C.; Nash, J.; James, P.E. Extracellular vesicles in atherosclerosis research. Methods Mol. Biol. 2022, 2419, 349–359. [Google Scholar] [PubMed]
  226. Pfeffer, S.R. Unsolved mysteries in membrane traffic. Annu. Rev. Biochem. 2007, 76, 629–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Azevedo, L.C. Microparticles and exosomes: Are they part of important pathways in sepsis pathophysiology? In Severe Sepsis and Septic Shock-Understanding a Serious Killer; Intechopen: London, UK, 2012. [Google Scholar]
  228. van Dommelen, S.M.; Vader, P.; Lakhal, S.; Kooijmans, S.; van Solinge, W.W.; Wood, M.J.; Schiffelers, R.M. Microvesicles and exosomes: Opportunities for cell-derived membrane vesicles in drug delivery. J. Control. Release 2012, 161, 635–644. [Google Scholar] [CrossRef]
  229. Hristov, M.; Erl, W.; Linder, S.; Weber, P.C. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood 2004, 104, 2761–2766. [Google Scholar] [CrossRef]
  230. Zhu, Z.; Zhang, D.; Lee, H.; Menon, A.A.; Wu, J.; Hu, K.; Jin, Y. Macrophage-derived apoptotic bodies promote the proliferation of the recipient cells via shuttling microRNA-221/222. J. Leukoc. Biol. 2017, 101, 1349–1359. [Google Scholar] [CrossRef] [Green Version]
  231. Holmgren, L.; Szeles, A.; Rajnavölgyi, E.; Folkman, J.; Klein, G.; Ernberg, I.; Falk, K.I. Horizontal transfer of DNA by the uptake of apoptotic bodies. Blood J. Am. Soc. Hematol. 1999, 93, 3956–3963. [Google Scholar]
  232. Bergsmedh, A.; Szeles, A.; Henriksson, M.; Bratt, A.; Folkman, M.J.; Spetz, A.-L.; Holmgren, L. Horizontal transfer of oncogenes by uptake of apoptotic bodies. Proc. Natl. Acad. Sci. USA 2001, 98, 6407–6411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  233. Jansen, F.; Yang, X.; Hoyer, F.F.; Paul, K.; Heiermann, N.; Becher, M.U.; Hussein, N.A.; Kebschull, M.; Bedorf, J.; Franklin, B.S. Endothelial microparticle uptake in target cells is annexin I/phosphatidylserine receptor dependent and prevents apoptosis. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1925–1935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Janiszewski, M.; Carmo, A.O.D.; Pedro, M.A.; Silva, E.; Knobel, E.; Laurindo, F.R. Platelet-derived exosomes of septic individuals possess proapoptotic NAD(P)H oxidase activity: A novel vascular redox pathway. Crit. Care Med. 2004, 32, 818–825. [Google Scholar] [CrossRef] [PubMed]
  235. Khorana, A.A.; Francis, C.W.; Culakova, E.; Kuderer, N.M.; Lyman, G.H. Frequency, risk factors, and trends for venous thromboembolism among hospitalized cancer patients. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2007, 110, 2339–2346. [Google Scholar] [CrossRef] [PubMed]
  236. Abdol Razak, N.B.; Jones, G.; Bhandari, M.; Berndt, M.C.; Metharom, P. Cancer-associated thrombosis: An overview of mechanisms, risk factors, and treatment. Cancers 2018, 10, 380. [Google Scholar] [CrossRef] [Green Version]
  237. Al Saleh, H.A.; Haas-Neill, S.; Al-Hashimi, A.; Kapoor, A.; Shayegan, B.; Austin, R.C.; Al-Nedawi, K. Thrombotic characteristics of extracellular vesicles derived from prostate cancer cells. Prostate 2018, 78, 953–961. [Google Scholar] [CrossRef]
  238. Lima, L.G.; Leal, A.C.; Vargas, G.; Porto-Carreiro, I.; Monteiro, R.Q. Intercellular transfer of tissue factor via the uptake of tumor-derived microvesicles. Thromb. Res. 2013, 132, 450–456. [Google Scholar] [CrossRef]
  239. Zarfati, M.; Katz, T.; Avivi, I.; Brenner, B.; Aharon, A. PO-45—The role of microvesicles in multiple myeloma progression. Thromb. Res. 2016, 140 (Suppl. 1), S193. [Google Scholar] [CrossRef]
  240. Gomes, F.G.; Sandim, V.; Almeida, V.H.; Rondon, A.M.; Succar, B.B.; Hottz, E.D.; Leal, A.C.; Verçoza, B.R.F.; Rodrigues, J.C.F.; Bozza, P.T. Breast-cancer extracellular vesicles induce platelet activation and aggregation by tissue factor-independent and-dependent mechanisms. Thromb. Res. 2017, 159, 24–32. [Google Scholar] [CrossRef]
  241. Muhsin-Sharafaldine, M.-R.; Saunderson, S.C.; Dunn, A.C.; Faed, J.M.; Kleffmann, T.; McLellan, A.D. Procoagulant and immunogenic properties of melanoma exosomes, microvesicles and apoptotic vesicles. Oncotarget 2016, 7, 56279. [Google Scholar] [CrossRef]
  242. Zhang, C.; Yang, Z.; Zhou, P.; Yu, M.; Li, B.; Liu, Y.; Jin, J.; Liu, W.; Jing, H.; Du, J.; et al. Phosphatidylserine-exposing tumor-derived microparticles exacerbate coagulation and cancer cell transendothelial migration in triple-negative breast cancer. Theranostics 2021, 11, 6445–6460. [Google Scholar] [CrossRef] [PubMed]
  243. Hron, G.; Kollars, M.; Weber, H.; Sagaster, V.; Quehenberger, P.; Eichinger, S.; Kyrle, P.A.; Weltermann, A. Tissue factor-positive microparticles: Cellular origin and association with coagulation activation in patients with colorectal cancer. Thromb. Haemost. 2007, 97, 119–123. [Google Scholar] [CrossRef] [PubMed]
  244. Zwicker, J.I.; Liebman, H.A.; Neuberg, D.; Lacroix, R.; Bauer, K.A.; Furie, B.C.; Furie, B. Tumor-derived tissue factor–bearing microparticles are associated with venous thromboembolic events in malignancy. Clin. Cancer Res. 2009, 15, 6830–6840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Manly, D.A.; Wang, J.; Glover, S.L.; Kasthuri, R.; Liebman, H.A.; Key, N.S.; Mackman, N. Increased microparticle tissue factor activity in cancer patients with venous thromboembolism. Thromb. Res. 2010, 125, 511–512. [Google Scholar] [CrossRef] [Green Version]
  246. Campello, E.; Spiezia, L.; Radu, C.M.; Bulato, C.; Castelli, M.; Gavasso, S.; Simioni, P. Endothelial, platelet, and tissue factor-bearing microparticles in cancer patients with and without venous thromboembolism. Thromb. Res. 2011, 127, 473–477. [Google Scholar] [CrossRef]
  247. Hell, L.; Däullary, T.; Burghart, V.; Mauracher, L.M.; Grilz, E.; Moser, B.; Kramer, G.; Schmid, J.A.; Ay, C.; Pabinger, I.; et al. Extracellular vesicle-associated tissue factor activity in prostate cancer patients with disseminated intravascular coagulation. Cancers 2021, 13, 1487. [Google Scholar] [CrossRef]
  248. Stark, K.; Schubert, I.; Joshi, U.; Kilani, B.; Hoseinpour, P.; Thakur, M.; Grünauer, P.; Pfeiler, S.; Schmidergall, T.; Stockhausen, S. Distinct pathogenesis of pancreatic cancer microvesicle—Associated venous thrombosis identifies new antithrombotic targets in vivo. Arter. Thromb. Vasc. Biol. 2018, 38, 772–786. [Google Scholar] [CrossRef] [Green Version]
  249. Lacroix, R.; Vallier, L.; Bonifay, A.; Simoncini, S.; Mege, D.; Aubert, M.; Panicot-Dubois, L.; Dubois, C.; Dignat-George, F. Microvesicles and cancer associated thrombosis. In Seminars in Thrombosis and Hemostasis; Thieme Medical Publishers: New York, NY, USA, 2019. [Google Scholar]
  250. Hisada, Y.; Mackman, N. Cancer cell-derived tissue factor positive extracellular vesicles: Biomarkers of thrombosis and survival. Curr. Opin. Hematol. 2019, 26, 349. [Google Scholar] [CrossRef]
  251. van Es, N.; Hisada, Y.; di Nisio, M.; Cesarman, G.; Kleinjan, A.; Mahé, I.; Otten, H.-M.; Kamphuisen, P.W.; Berckmans, R.J.; Büller, H.R. Extracellular vesicles exposing tissue factor for the prediction of venous thromboembolism in patients with cancer: A prospective cohort study. Thromb. Res. 2018, 166, 54–59. [Google Scholar] [CrossRef]
  252. Lazar, S.; Goldfinger, L.E. Platelets and extracellular vesicles and their cross talk with cancer. Blood 2021, 137, 3192–3200. [Google Scholar] [CrossRef]
  253. Żmigrodzka, M.; Witkowska-Piłaszewicz, O.; Winnicka, A. Platelets extracellular vesicles as regulators of cancer progression—An updated perspective. Int. J. Mol. Sci. 2020, 21, 5195. [Google Scholar] [CrossRef] [PubMed]
  254. Gkolfinopoulos, S.; Jones, R.L.; Constantinidou, A. The emerging role of platelets in the formation of the micrometastatic niche: Current evidence and future perspectives. Front. Oncol. 2020, 10, 374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Hisada, Y.; Mackman, N. Tissue factor and extracellular vesicles: Activation of coagulation and impact on survival in cancer. Cancers 2021, 13, 3839. [Google Scholar] [CrossRef] [PubMed]
  256. Khorana, A.; Francis, C.; Menzies, K.; Wang, J.-G.; Hyrien, O.; Hathcock, J.; Mackman, N.; Taubman, M. Plasma tissue factor may be predictive of venous thromboembolism in pancreatic cancer. J. Thromb. Haemost. 2008, 6, 1983–1985. [Google Scholar] [CrossRef]
  257. Faille, D.; Bourrienne, M.-C.; de Raucourt, E.; de Chaisemartin, L.; Granger, V.; Lacroix, R.; Panicot-Dubois, L.; Hammel, P.; Lévy, P.; Ruszniewski, P. Biomarkers for the risk of thrombosis in pancreatic adenocarcinoma are related to cancer process. Oncotarget 2018, 9, 26453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  258. Tesselaar, M.; Romijn, F.; van der Linden, I.; Prins, F.; Bertina, R.; Osanto, S. Microparticle-associated tissue factor activity: A link between cancer and thrombosis? J. Thromb. Haemost. 2007, 5, 520–527. [Google Scholar] [CrossRef]
  259. Toth, B.; Liebhardt, S.; Steinig, K.; Ditsch, N.; Rank, A.; Bauerfeind, I.; Spannagl, M.; Friese, K.; Reininger, A.J. Platelet-derived microparticles and coagulation activation in breast cancer patients. Thromb. Haemost. 2008, 100, 663–669. [Google Scholar] [CrossRef] [Green Version]
  260. Hernández, C.; Orbe, J.; Roncal, C.; Alvarez-Hernandez, M.; de Lizarrondo, S.M.; Alves, M.T.; Mata, J.G.; Páramo, J.A. Tissue factor expressed by microparticles is associated with mortality but not with thrombosis in cancer patients. Thromb. Haemost. 2013, 110, 598–608. [Google Scholar] [CrossRef]
  261. Thaler, J.; Ay, C.; Mackman, N.; Bertina, R.; Kaider, A.; Marosi, C.; Key, N.; Barcel, D.; Scheithauer, W.; Kornek, G. Microparticle-associated tissue factor activity, venous thromboembolism and mortality in pancreatic, gastric, colorectal and brain cancer patients. J. Thromb. Haemost. 2012, 10, 1363–1370. [Google Scholar] [CrossRef]
  262. Durrieu, L.; Bharadwaj, A.; Waisman, D.M. Analysis of the thrombotic and fibrinolytic activities of tumor cell-derived extracellular vesicles. Blood Adv. 2018, 2, 1054–1065. [Google Scholar] [CrossRef] [Green Version]
  263. Garnier, D.; Magnus, N.; Lee, T.H.; Bentley, V.; Meehan, B.; Milsom, C.; Montermini, L.; Kislinger, T.; Rak, J. Cancer cells induced to express mesenchymal phenotype release exosome-like extracellular vesicles carrying tissue factor. J. Biol. Chem. 2012, 287, 43565–43572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Thomas, G.M.; Panicot-Dubois, L.; Lacroix, R.; Dignat-George, F.; Lombardo, D.; Dubois, C. Cancer cell—Derived microparticles bearing P-selectin glycoprotein ligand 1 accelerate thrombus formation in vivo. J. Exp. Med. 2009, 206, 1913–1927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Hisada, Y.; Ay, C.; Auriemma, A.; Cooley, B.; Mackman, N. Human pancreatic tumors grown in mice release tissue factor-positive microvesicles that increase venous clot size. J. Thromb. Haemost. 2017, 15, 2208–2217. [Google Scholar] [CrossRef] [Green Version]
  266. Thomas, G.; Brill, A.; Mezouar, S.; Crescence, L.; Gallant, M.; Dubois, C.; Wagner, D. Tissue factor expressed by circulating cancer cell-derived microparticles drastically increases the incidence of deep vein thrombosis in mice. J. Thromb. Haemost. 2015, 13, 1310–1319. [Google Scholar] [CrossRef]
  267. Davila, M.; Amirkhosravi, A.; Coll, E.; Desai, H.; Robles, L.; Colon, J.; Baker, C.; Francis, J. Tissue factor-bearing microparticles derived from tumor cells: Impact on coagulation activation. J. Thromb. Haemost. 2008, 6, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
  268. Wang, J.-G.; Geddings, J.E.; Aleman, M.M.; Cardenas, J.C.; Chantrathammachart, P.; Williams, J.C.; Kirchhofer, D.; Bogdanov, V.Y.; Bach, R.R.; Rak, J. Tumor-derived tissue factor activates coagulation and enhances thrombosis in a mouse xenograft model of human pancreatic cancer. Blood J. Am. Soc. Hematol. 2012, 119, 5543–5552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  269. Sasano, T.; Cho, M.S.; Rodriguez-Aguayo, C.; Bayraktar, E.; Taki, M.; Afshar-Kharghan, V.; Sood, A.K. Role of tissue-factor bearing extracellular vesicles released from ovarian cancer cells in platelet aggregation in vitro and venous thrombosis in mice. Thromb. Update 2021, 2, 100020. [Google Scholar] [CrossRef]
  270. Mezouar, S.; Darbousset, R.; Dignat-George, F.; Panicot-Dubois, L.; Dubois, C. Inhibition of platelet activation prevents the P-selectin and integrin-dependent accumulation of cancer cell microparticles and reduces tumor growth and metastasis in vivo. Int. J. Cancer 2014, 136, 462–475. [Google Scholar] [CrossRef] [Green Version]
  271. Tawil, N.; Bassawon, R.; Meehan, B.; Nehme, A.; Montermini, L.; Gayden, T.; de Jay, N.; Spinelli, C.; Chennakrishnaiah, S.; Choi, D.; et al. Glioblastoma cell populations with distinct oncogenic programs release podoplanin as procoagulant extracellular vesicles. Blood Adv. 2021, 5, 1682–1694. [Google Scholar] [CrossRef]
  272. Nickel, K.F.; Ronquist, G.; Langer, F.; Labberton, L.; Fuchs, T.A.; Bokemeyer, C.; Sauter, G.; Graefen, M.; Mackman, N.; Stavrou, E.X. The polyphosphate–factor XII pathway drives coagulation in prostate cancer-associated thrombosis. Blood J. Am. Soc. Hematol. 2015, 126, 1379–1389. [Google Scholar] [CrossRef] [Green Version]
  273. Shim, Y.J.; Chatterjee, V.; Swaidani, S.; Alluri, R.K.; Kundu, S.; Merkulova, A.; Angelini, D.; You, D.; Whitney, S.A.; Feener, E.P.; et al. Polyphosphate expression by cancer cell extracellular vesicles mediates binding of factor XII and contact activation. Blood Adv. 2021, 5, 4741–4751. [Google Scholar] [CrossRef] [PubMed]
  274. Larsson, M.; Rayzman, V.; Nolte, M.W.; Nickel, K.F.; Björkqvist, J.; Jämsä, A.; Hardy, M.P.; Fries, M.; Schmidbauer, S.; Hedenqvist, P. A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk. Sci. Transl. Med. 2014, 6, 222ra17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. Nickel, K.F.; Labberton, L.; Long, A.T.; Langer, F.; Fuchs, T.A.; Stavrou, E.X.; Butler, L.M.; Renné, T. The polyphosphate/factor XII pathway in cancer-associated thrombosis: Novel perspectives for safe anticoagulation in patients with malignancies. Thromb. Res. 2016, 141, S4–S7. [Google Scholar] [CrossRef]
  276. Demers, M.; Krause, D.S.; Schatzberg, D.; Martinod, K.; Voorhees, J.R.; Fuchs, T.A.; Scadden, D.T.; Wagner, D.D. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc. Natl. Acad. Sci. USA 2012, 109, 13076–13081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Demers, M.; Wagner, D.D. NETosis: A new factor in tumor progression and cancer-associated thrombosis. Semin. Thromb. Hemost. 2014, 40, 277–283. [Google Scholar] [CrossRef] [Green Version]
  278. Cedervall, J.; Zhang, Y.; Olsson, A.K. Tumor-induced NETosis as a risk factor for metastasis and organ failure. Cancer Res. 2016, 76, 4311–4315. [Google Scholar] [CrossRef] [Green Version]
  279. Noubouossie, D.F.; Reeves, B.N.; Strahl, B.D.; Key, N.S. Neutrophils: Back in the thrombosis spotlight. Blood 2019, 133, 2186–2197. [Google Scholar] [CrossRef]
  280. Chapman, E.A.; Lyon, M.; Simpson, D.; Mason, D.; Beynon, R.J.; Moots, R.J.; Wright, H.L. Caught in a trap? Proteomic analysis of neutrophil extracellular traps in rheumatoid arthritis and systemic lupus erythematosus. Front. Immunol. 2019, 10, 423. [Google Scholar] [CrossRef]
  281. Noubouossie, D.F.; Whelihan, M.F.; Yu, Y.B.; Sparkenbaugh, E.; Pawlinski, R.; Monroe, D.M.; Key, N.S. In vitro activation of coagulation by human neutrophil DNA and histone proteins but not neutrophil extracellular traps. Blood 2017, 129, 1021–1029. [Google Scholar] [CrossRef]
  282. Martinod, K.; Wagner, D.D. Thrombosis: Tangled up in NETs. Blood 2014, 123, 2768–2776. [Google Scholar] [CrossRef]
  283. Martinod, K.; Demers, M.; Fuchs, T.A.; Wong, S.L.; Brill, A.; Gallant, M.; Hu, J.; Wang, Y.; Wagner, D.D. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc. Natl. Acad. Sci. USA 2013, 110, 8674–8679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. McDonald, B.; Davis, R.P.; Kim, S.J.; Tse, M.; Esmon, C.T.; Kolaczkowska, E.; Jenne, C.N. Platelets and neutrophil extracellular traps collaborate to promote intravascular coagulation during sepsis in mice. Blood 2017, 129, 1357–1367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. Dou, H.; Kotini, A.; Liu, W.; Fidler, T.; Endo-Umeda, K.; Sun, X.; Olszewska, M.; Xiao, T.; Abramowicz, S.; Yalcinkaya, M.; et al. Oxidized phospholipids promote NETosis and arterial thrombosis in LNK(SH2B3) deficiency. Circulation 2021, 144, 1940–1954. [Google Scholar] [CrossRef] [PubMed]
  286. 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]
  287. Yu, X.; Diamond, S.L. Fibrin modulates shear-induced NETosis in sterile occlusive thrombi formed under haemodynamic flow. Thromb. Haemost. 2019, 119, 586–593. [Google Scholar] [CrossRef] [PubMed]
  288. Healy, L.D.; Puy, C.; Fernández, J.A.; Mitrugno, A.; Keshari, R.S.; Taku, N.A.; Chu, T.T.; Xu, X.; Gruber, A.; Lupu, F.; et al. Activated protein C inhibits neutrophil extracellular trap formation in vitro and activation in vivo. J. Biol. Chem. 2017, 292, 8616–8629. [Google Scholar] [CrossRef] [Green Version]
  289. Von Brühl, M.-L.; Stark, K.; Steinhart, A.; Chandraratne, S.; Konrad, I.; Lorenz, M.; Khandoga, A.; Tirniceriu, A.; Coletti, R.; Köllnberger, M. 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]
  290. Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair, G.D. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 2007, 13, 463–469. [Google Scholar] [CrossRef]
  291. Mizurini, D.M.; Aslan, J.S.; Gomes, T.; Ma, D.; Francischetti, I.M.; Monteiro, R.Q. Salivary thromboxane A2-binding proteins from triatomine vectors of Chagas disease inhibit platelet-mediated neutrophil extracellular traps (NETs) formation and arterial thrombosis. PLoS Negl. Trop. Dis. 2015, 9, e0003869. [Google Scholar] [CrossRef]
  292. Laridan, E.; Martinod, K.; de Meyer, S.F. Neutrophil extracellular traps in arterial and venous thrombosis. In Seminars in Thrombosis and Hemostasis; Thieme Publishing: New York, NY, USA, 2019. [Google Scholar]
  293. Fuchs, T.A.; Bhandari, A.A.; Wagner, D.D. Histones induce rapid and profound thrombocytopenia in mice. Blood 2011, 118, 3708–3714. [Google Scholar] [CrossRef] [Green Version]
  294. Elaskalani, O.; Razak, N.B.A.; Metharom, P. Neutrophil extracellular traps induce aggregation of washed human platelets independently of extracellular DNA and histones. Cell Commun. Signal. 2018, 16, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Leal, A.C.; Mizurini, D.M.; Gomes, T.; Rochael, N.C.; Saraiva, E.M.; Dias, M.S.; Werneck, C.C.; Sielski, M.S.; Vicente, C.P.; Monteiro, R.Q. Tumor-derived exosomes induce the formation of neutrophil extracellular traps: Implications for the establishment of cancer-associated thrombosis. Sci. Rep. 2017, 7, 6438. [Google Scholar] [CrossRef] [PubMed]
  296. Boone, B.A.; Murthy, P.; Miller-Ocuin, J.; Doerfler, W.R.; Ellis, J.T.; Liang, X.; Ross, M.A.; Wallace, C.T.; Sperry, J.L.; Lotze, M.T. Chloroquine reduces hypercoagulability in pancreatic cancer through inhibition of neutrophil extracellular traps. BMC Cancer 2018, 18, 678. [Google Scholar] [CrossRef] [Green Version]
  297. Mauracher, L.M.; Posch, F.; Martinod, K.; Grilz, E.; Däullary, T.; Hell, L.; Brostjan, C.; Zielinski, C.; Ay, C.; Wagner, D. Citrullinated histone H3, a biomarker of neutrophil extracellular trap formation, predicts the risk of venous thromboembolism in cancer patients. J. Thromb. Haemost. 2018, 16, 508–518. [Google Scholar] [CrossRef]
  298. Seo, J.D.; Gu, J.-Y.; Jung, H.S.; Kim, Y.J.; Kim, H.K. Contact system activation and neutrophil extracellular trap markers: Risk factors for portal vein thrombosis in patients with hepatocellular carcinoma. Clin. Appl. Thromb. Hemost. 2019, 25, 1076029618825310. [Google Scholar] [CrossRef] [Green Version]
  299. Hisada, Y.; Grover, S.P.; Maqsood, A.; Houston, R.; Ay, C.; Noubouossie, D.F.; Cooley, B.C.; Wallén, H.; Key, N.S.; Thålin, C. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2020, 105, 218–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  300. Nieuwland, R.; Berckmans, R.J.; McGregor, S.; Böing, A.N.; Romijn, F.P.T.M.; Westendorp, R.G.; Hack, C.E.; Sturk, A. Cellular origin and procoagulant properties of microparticles in meningococcal sepsis. Blood J. Am. Soc. Hematol. 2000, 95, 930–935. [Google Scholar] [CrossRef]
  301. Wang, J.G.; Manly, D.; Kirchhofer, D.; Pawlinski, R.; Mackman, N. Levels of microparticle tissue factor activity correlate with coagulation activation in endotoxemic mice. J. Thromb. Haemost. 2009, 7, 1092–1098. [Google Scholar] [CrossRef]
  302. Zafrani, L.; Gerotziafas, G.; Byrnes, C.; Hu, X.; Perez, J.; Lévi, C.; Placier, S.; Letavernier, E.; Leelahavanichkul, A.; Haymann, J.-P. Calpastatin controls polymicrobial sepsis by limiting procoagulant microparticle release. Am. J. Respir. Crit. Care Med. 2012, 185, 744–755. [Google Scholar] [CrossRef] [Green Version]
  303. Raeven, P.; Zipperle, J.; Drechsler, S. Extracellular vesicles as markers and mediators in sepsis. Theranostics 2018, 8, 3348. [Google Scholar] [CrossRef]
  304. Lehner, G.F.; Harler, U.; Haller, V.M.; Feistritzer, C.; Hasslacher, J.; Dunzendorfer, S.; Bellmann, R.; Joannidis, M. Characterization of microvesicles in septic shock using high-sensitivity flow cytometry. Shock Inj. Inflamm. Sepsis Lab. Clin. Approaches 2016, 46, 373–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Ohuchi, M.; Fujino, K.; Kishimoto, T.; Yamane, T.; Hamamoto, T.; Tabata, T.; Tsujita, Y.; Matsushita, M.; Takahashi, K.; Matsumura, K. Association of the plasma platelet-derived microparticles to platelet count ratio with hospital mortality and disseminated intravascular coagulopathy in critically lll patients. J. Atheroscler. Thromb. 2015, 22, 29439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Burgelman, M.; Vandendriessche, C.; Vandenbroucke, R.E. Extracellular vesicles: A double-edged sword in sepsis. Pharmaceuticals 2021, 14, 829. [Google Scholar] [CrossRef] [PubMed]
  307. Andersson, U.; Tracey, K.J. HMGB1 is a therapeutic target for sterile inflammation and infection. Ann. Rev. Immunol. 2011, 29, 139–162. [Google Scholar] [CrossRef] [Green Version]
  308. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  309. Zwicker, J.I. Tissue factor–bearing microparticles and cancer. In Seminars in Thrombosis and Hemostasis; Thieme Medical Publishers: New York, NY, USA, 2008. [Google Scholar]
  310. Freyssinet, J.-M.; Toti, F. Formation of procoagulant microparticles and properties. Thromb. Res. 2010, 125, S46–S48. [Google Scholar] [CrossRef]
  311. de Lizarrondo, S.M.; Roncal, C.; Calvayrac, O.; Rodríguez, C.; Varo, N.; Purroy, A.; Lorente, L.; Rodríguez, J.A.; Doeuvre, L.; Hervás-Stubbs, S. Synergistic effect of thrombin and CD40 ligand on endothelial matrix metalloproteinase-10 expression and microparticle generation in vitro and in vivo. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1477–1487. [Google Scholar] [CrossRef] [Green Version]
  312. Iba, T.; Ogura, H. Role of extracellular vesicles in the development of sepsis-induced coagulopathy. J. Intensiv. Care 2018, 6, 68. [Google Scholar] [CrossRef]
  313. Horstman, L.L.; Ahn, Y.S. Platelet microparticles: A wide-angle perspective. Crit. Rev. Oncol. Hematol. 1999, 30, 111–142. [Google Scholar] [CrossRef]
  314. Matsumoto, H.; Yamakawa, K.; Ogura, H.; Koh, T.; Matsumoto, N.; Shimazu, T. Clinical significance of tissue factor and CD13 double-positive microparticles in SIRS patients with trauma and severe sepsis. Shock Inj. Inflamm. Sepsis Lab. Clin. Approaches 2017, 47, 409–415. [Google Scholar] [CrossRef]
  315. Woei-A-Jin, F.S.H.; van der Starre, W.E.; Tesselaar, M.E.; Rodriguez, P.G.; van Nieuwkoop, C.; Bertina, R.M.; van Dissel, J.T.; Osanto, S. Procoagulant tissue factor activity on microparticles is associated with disease severity and bacteremia in febrile urinary tract infections. Thromb. Res. 2014, 133, 799–803. [Google Scholar] [CrossRef] [PubMed]
  316. Kleinjan, A.; Böing, A.N.; Sturk, A.; Nieuwland, R. Microparticles in vascular disorders: How tissue factor-exposing vesicles contribute to pathology and physiology. Thromb. Res. 2012, 130, S71–S73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  317. Hellum, M.; Øvstebø, R.; Brusletto, B.S.; Berg, J.P.; Brandtzaeg, P.; Henriksson, C.E. Microparticle-associated tissue factor activity correlates with plasma levels of bacterial lipopolysaccharides in meningococcal septic shock. Thromb. Res. 2014, 133, 507–514. [Google Scholar] [CrossRef] [PubMed]
  318. Zhang, Y.; Meng, H.; Ma, R.; He, Z.; Wu, X.; Cao, M.; Yao, Z.; Zhao, L.; Li, T.; Deng, R. Circulating microparticles, blood cells, and endothelium induce procoagulant activity in sepsis through phosphatidylserine exposure. Shock 2016, 45, 299–307. [Google Scholar] [CrossRef]
  319. Tripisciano, C.; Weiss, R.; Eichhorn, T.; Spittler, A.; Heuser, T.; Fischer, M.B.; Weber, V. Different potential of extracellular vesicles to support thrombin generation: Contributions of phosphatidylserine, tissue factor, and cellular origin. Sci. Rep. 2017, 7, 6522. [Google Scholar] [CrossRef]
  320. Radic, M.; Marion, T.; Monestier, M. Nucleosomes are exposed at the cell surface in apoptosis. J. Immunol. 2004, 172, 6692–6700. [Google Scholar] [CrossRef] [Green Version]
  321. Liaw, P.C.; Ito, T.; Iba, T.; Thachil, J.; Zeerleder, S. DAMP and DIC: The role of extracellular DNA and DNA-binding proteins in the pathogenesis of DIC. Blood Rev. 2016, 30, 257–261. [Google Scholar] [CrossRef]
  322. Weiss, R.; Gröger, M.; Rauscher, S.; Fendl, B.; Eichhorn, T.; Fischer, M.B.; Spittler, A.; Weber, V. Differential interaction of platelet-derived extracellular vesicles with leukocyte subsets in human whole blood. Sci. Rep. 2018, 8, 6598. [Google Scholar] [CrossRef]
  323. Im, Y.; Yoo, H.; Lee, J.Y.; Park, J.; Suh, G.Y.; Jeon, K. Association of plasma exosomes with severity of organ failure and mortality in patients with sepsis. J. Cell. Mol. Med. 2020, 24, 9439–9445. [Google Scholar] [CrossRef]
  324. Reithmair, M.; Buschmann, D.; Märte, M.; Kirchner, B.; Hagl, D.; Kaufmann, I.; Pfob, M.; Chouker, A.; Steinlein, O.K.; Pfaffl, M.W.; et al. Cellular and extracellular miRNAs are blood-compartment-specific diagnostic targets in sepsis. J. Cell. Mol. Med. 2017, 21, 2403–2411. [Google Scholar] [CrossRef]
  325. Hirschberger, S.; Hinske, L.C.; Kreth, S. MiRNAs: Dynamic regulators of immune cell functions in inflammation and cancer. Cancer Lett. 2018, 431, 11–21. [Google Scholar] [CrossRef]
  326. Jiao, Y.; Li, W.; Wang, W.; Tong, X.; Xia, R.; Fan, J.; Du, J.; Zhang, C.; Shi, X. Platelet-derived exosomes promote neutrophil extracellular trap formation during septic shock. Crit. Care 2020, 24, 380. [Google Scholar] [CrossRef] [PubMed]
  327. Usman, W.M.; Pham, T.C.; Kwok, Y.Y.; Vu, L.T.; Ma, V.; Peng, B.; Chan, Y.S.; Wei, L.; Chin, S.M.; Azad, A. Efficient RNA drug delivery using red blood cell extracellular vesicles. Nat. Commun. 2018, 9, 2359. [Google Scholar] [CrossRef] [PubMed]
  328. Lee, Y.; el Andaloussi, S.; Wood, M.J. Exosomes and microvesicles: Extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  329. Pawlowski, C.L.; Li, W.; Sun, M.; Ravichandran, K.; Hickman, D.; Kos, C.; Kaur, G.; Gupta, A.S. Platelet microparticle-inspired clot-responsive nanomedicine for targeted fibrinolysis. Biomaterials 2017, 128, 94–108. [Google Scholar] [CrossRef] [PubMed]
  330. Disharoon, D.; Marr, D.W.; Neeves, K.B. Engineered microparticles and nanoparticles for fibrinolysis. J. Thromb. Haemost. 2019, 17, 2004–2015. [Google Scholar] [CrossRef] [PubMed]
  331. Xu, R.; Greening, D.W.; Zhu, H.-J.; Takahashi, N.; Simpson, R.J. Extracellular vesicle isolation and characterization: Toward clinical application. J. Clin. Investig. 2016, 126, 1152–1162. [Google Scholar] [CrossRef] [Green Version]
Figure 1. The hemostatic pathways include the intrinsic and the extrinsic coagulation pathways. In the healthy state, endothelial cells maintain an antithrombotic property that ensures normal blood flow. Following vascular injury and the disruption of vascular endothelial integrity, FXII is activated by proteolytic cleavage resulting in the formation of FXIIa. FXIIa activates FXI into FXIa, which then activates FIX into FIXa in the presence of calcium. FIXa activates FX into FXa which binds thrombin-activated FVa forming the FXa/FVa complex that activates prothrombin into thrombin. Then, thrombin cleaves fibrinogen into fibrin forming the fibrin mesh and causing the cessation of blood loss. Thrombin also cleaves PAR1/4 and activates FV into FVa and FXI into FXIa, thus reinforcing the coagulation cascade. Thrombin also inhibits the binding of the endothelial-derived FVIII to exposed vWF and thus, accelerates the formation of the FVIII/vWF complex and the subsequent activation of FVIII into FVIIIa, which activates FIX into FIXa. Additionally, thrombin activity can be inhibited by ATIII. Parallel to the intrinsic pathway, the extrinsic pathway is activated in response to vascular trauma, which results in the secretion of TF. TF activates FVII into FVIIa and subsequently forms the TF-FVIIa complex, which activates FIX into FIXa. Activated platelets play a major role in the potentiation of both pathways by providing negatively charged surfaces following phospholipid bilayer rearrangement, which supports coagulation. Following primary and secondary hemostasis, fibrinolysis takes place. Fibrin degradation leads to the formation of FDP, which activates plasminogen into plasmin. TAFI, which is activated by low TM and the TM/thrombin complex into TAFIa, inhibits plasmin activity. TAFIa is inhibited by both high TM and protein C. Finally, protein C activates APC, which binds to protein S forming the APC/protein S complex, which inhibits the activity of FVa and FVIIIa. Abbreviations: APC, active protein C; ATIII, anti-thrombin III; EPCR, endothelial cell protein C receptor; F, factor; FDP, fibrin degradation products; GP, glycoprotein; PAR, protease-activated receptor; TAFI, thrombin-activatable fibrinolysis inhibitor; TF, tissue factor; TM, thrombomodulin; vWF, von Willebrand factor.
Figure 1. The hemostatic pathways include the intrinsic and the extrinsic coagulation pathways. In the healthy state, endothelial cells maintain an antithrombotic property that ensures normal blood flow. Following vascular injury and the disruption of vascular endothelial integrity, FXII is activated by proteolytic cleavage resulting in the formation of FXIIa. FXIIa activates FXI into FXIa, which then activates FIX into FIXa in the presence of calcium. FIXa activates FX into FXa which binds thrombin-activated FVa forming the FXa/FVa complex that activates prothrombin into thrombin. Then, thrombin cleaves fibrinogen into fibrin forming the fibrin mesh and causing the cessation of blood loss. Thrombin also cleaves PAR1/4 and activates FV into FVa and FXI into FXIa, thus reinforcing the coagulation cascade. Thrombin also inhibits the binding of the endothelial-derived FVIII to exposed vWF and thus, accelerates the formation of the FVIII/vWF complex and the subsequent activation of FVIII into FVIIIa, which activates FIX into FIXa. Additionally, thrombin activity can be inhibited by ATIII. Parallel to the intrinsic pathway, the extrinsic pathway is activated in response to vascular trauma, which results in the secretion of TF. TF activates FVII into FVIIa and subsequently forms the TF-FVIIa complex, which activates FIX into FIXa. Activated platelets play a major role in the potentiation of both pathways by providing negatively charged surfaces following phospholipid bilayer rearrangement, which supports coagulation. Following primary and secondary hemostasis, fibrinolysis takes place. Fibrin degradation leads to the formation of FDP, which activates plasminogen into plasmin. TAFI, which is activated by low TM and the TM/thrombin complex into TAFIa, inhibits plasmin activity. TAFIa is inhibited by both high TM and protein C. Finally, protein C activates APC, which binds to protein S forming the APC/protein S complex, which inhibits the activity of FVa and FVIIIa. Abbreviations: APC, active protein C; ATIII, anti-thrombin III; EPCR, endothelial cell protein C receptor; F, factor; FDP, fibrin degradation products; GP, glycoprotein; PAR, protease-activated receptor; TAFI, thrombin-activatable fibrinolysis inhibitor; TF, tissue factor; TM, thrombomodulin; vWF, von Willebrand factor.
Jcm 11 04932 g001
Figure 2. General mechanisms governing the formation and secretion of extracellular vesicles including microvesicles, apoptotic bodies, and exosomes. Microvesicles are 100 nm–1 μm in size and are secreted following cellular activation. Several stimuli promote microvesicle formation including shear stress, oxidative stress, several cytokines, as well as lipopolysaccharides, thrombin, and high plasma levels of homocysteine. Following cellular activation, flippases, floppases, and scramblases mediate the loss of the phospholipid bilayer integrity. Simultaneously, calcium-dependent, gelsolin and calponin-executed cytoskeletal rearrangement occur resulting in the alteration of the membrane curvature. This allows for the emergence of plasma membrane protrusions and the detachment of MVs. MVs contain several molecules such as miRNAs, lipids, proteins, and cellular metabolites that potentially modulate key functions in target cells. Apoptotic bodies, which have poor procoagulant activity, are released from apoptotic cells following membrane blebbing. Exosomes originate from an endocytotic extracellular vesicle that passes through the endosomal pathway, leading to the formation of multi-vesicular bodies. Exosomes are released from multi-vesicular bodies where they interact with target cells either by binding their surface receptors or through exosome-carried molecules following exosomal degradation. EV, extracellular vesicle; ICAM-1, intracellular adhesion molecule-1; LFA-1, leukocyte function-associated antigen-1; miRNA, micro RNA; MMP, matrix metalloproteinase; MV, microvesicle; RAL-1, Ras-related GTPase homolog; VCAM-1, vascular cell adhesion molecule-1; VLA-4, very late antigen-4.
Figure 2. General mechanisms governing the formation and secretion of extracellular vesicles including microvesicles, apoptotic bodies, and exosomes. Microvesicles are 100 nm–1 μm in size and are secreted following cellular activation. Several stimuli promote microvesicle formation including shear stress, oxidative stress, several cytokines, as well as lipopolysaccharides, thrombin, and high plasma levels of homocysteine. Following cellular activation, flippases, floppases, and scramblases mediate the loss of the phospholipid bilayer integrity. Simultaneously, calcium-dependent, gelsolin and calponin-executed cytoskeletal rearrangement occur resulting in the alteration of the membrane curvature. This allows for the emergence of plasma membrane protrusions and the detachment of MVs. MVs contain several molecules such as miRNAs, lipids, proteins, and cellular metabolites that potentially modulate key functions in target cells. Apoptotic bodies, which have poor procoagulant activity, are released from apoptotic cells following membrane blebbing. Exosomes originate from an endocytotic extracellular vesicle that passes through the endosomal pathway, leading to the formation of multi-vesicular bodies. Exosomes are released from multi-vesicular bodies where they interact with target cells either by binding their surface receptors or through exosome-carried molecules following exosomal degradation. EV, extracellular vesicle; ICAM-1, intracellular adhesion molecule-1; LFA-1, leukocyte function-associated antigen-1; miRNA, micro RNA; MMP, matrix metalloproteinase; MV, microvesicle; RAL-1, Ras-related GTPase homolog; VCAM-1, vascular cell adhesion molecule-1; VLA-4, very late antigen-4.
Jcm 11 04932 g002
Figure 3. General pathways through which extracellular vesicles participate in disease-associated procoagulant states. (A) Activated platelets secrete procoagulant microvesicles that increase in different cardiovascular disorders. Impaired production of platelet-derived microvesicles was shown to participate in different bleeding disorders such as Scott’s and Castman’s Syndromes. On the other hand, activated platelet-derived exosomes are thought to counteract the progression of atherosclerosis. (B) Cancer cell-derived extracellular vesicles are procoagulant through different pathways that initiate TF-dependent or independent platelet activation leading to disseminated coagulopathy. (C) Sepsis causes end-organ damage mainly through activating the coagulation system. In sepsis, proinflammatory and procoagulant EVS are released into the circulation, while procoagulant exosomes are released for activated platelets, where both participate in sepsis-associated mortality.
Figure 3. General pathways through which extracellular vesicles participate in disease-associated procoagulant states. (A) Activated platelets secrete procoagulant microvesicles that increase in different cardiovascular disorders. Impaired production of platelet-derived microvesicles was shown to participate in different bleeding disorders such as Scott’s and Castman’s Syndromes. On the other hand, activated platelet-derived exosomes are thought to counteract the progression of atherosclerosis. (B) Cancer cell-derived extracellular vesicles are procoagulant through different pathways that initiate TF-dependent or independent platelet activation leading to disseminated coagulopathy. (C) Sepsis causes end-organ damage mainly through activating the coagulation system. In sepsis, proinflammatory and procoagulant EVS are released into the circulation, while procoagulant exosomes are released for activated platelets, where both participate in sepsis-associated mortality.
Jcm 11 04932 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Al-Koussa, H.; AlZaim, I.; El-Sabban, M.E. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. J. Clin. Med. 2022, 11, 4932. https://doi.org/10.3390/jcm11164932

AMA Style

Al-Koussa H, AlZaim I, El-Sabban ME. Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders. Journal of Clinical Medicine. 2022; 11(16):4932. https://doi.org/10.3390/jcm11164932

Chicago/Turabian Style

Al-Koussa, Houssam, Ibrahim AlZaim, and Marwan E. El-Sabban. 2022. "Pathophysiology of Coagulation and Emerging Roles for Extracellular Vesicles in Coagulation Cascades and Disorders" Journal of Clinical Medicine 11, no. 16: 4932. https://doi.org/10.3390/jcm11164932

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