Skip to main content

MINI REVIEW article

Front. Cardiovasc. Med., 25 March 2022
Sec. Atherosclerosis and Vascular Medicine
Volume 9 - 2022 | https://doi.org/10.3389/fcvm.2022.863846

Implication of Platelets in Immuno-Thrombosis and Thrombo-Inflammation

Younes Zaid1,2* Yahye Merhi3
  • 1Laboratory of Materials, Nanotechnology and Environment, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
  • 2Immunology and Biodiversity Laboratory, Department of Biology, Faculty of Sciences, Hassan II University, Casablanca, Morocco
  • 3Laboratory of Thrombosis and Hemostasis, Montreal Heart Institute, Research Center, The Université de Montréal, Montreal, QC, Canada

In addition to their well-described hemostatic function, platelets are active participants in innate and adaptive immunity. Inflammation and immunity are closely related to changes in platelet reactions and enhanced platelet function in thrombo-inflammation, as well as in microbial and virus infections. A platelet’s immune function is incompletely understood, but an important balance exists between its protective and pathogenic responses and its thrombotic and inflammatory functions. As the mediator of vascular homeostasis, platelets interact with neutrophils, bacteria and virus by expressing specific receptors and releasing granules, transferring RNA, and secreting mitochondria, which controls hemostasis and thrombosis, infection, and innate and adaptive immunity. This review focuses on the involvement of platelets during immuno-thrombosis and thrombo-inflammation.

Introduction

Platelets are small (2–4 μm in diameter) anucleated cells derived from their megakaryocyte’s precursors with 7–10 days lifespan. Nearly one trillion platelets sentinel the blood vessels to monitor and preserve the integrity of the vasculature. There is no nucleus in platelets, but they are prepacked with proteins and various forms of RNA from their precursor cells. When damage to blood vessels occurs, it triggers the formation of a thrombus to stop bleeding (1). The discoid shape changes to a spherical one, resulting in long filopodia that facilitate adhesion. In order for platelets to function, they must involve an array of adhesive and activation receptors, secreted granule reservoirs, and dynamic cytoskeletal proteins (2). Extracellular vesicles (microparticles) can also play a role in the formation of thrombus mediated by platelets, as they provide anionic phospholipids that aid in the coagulation process (3). The role of platelets is not restricted to the hemostatic/thrombotic response, but platelets play a crucial role in inflammatory and immune responses (48). Indeed, not only do platelets express an array of molecules serving wound repair, but they also bear immune and inflammatory molecules such as interleukin IL1 (9), and an array of receptors including toll-like-receptors (TLRs), CD154, or CD40L (4), Fc receptor for IgG (FcγRIIA) (10), IgA (FcαRI) and IgE (FcεRI) (11, 12).

In response to harmful pathogens, platelets contribute to the immune system either directly, by producing cytokines and antimicrobial peptides, or indirectly, through interactions with neutrophils, monocytes, lymphocytes, and other cells (13, 14). Immuno-thrombosis may negatively affect hemostatic and immunological processes during a bacterial infection, resulting in adverse clinical outcomes (15). In this review, we will focus on the role of platelets in immuno-thrombosis and thrombo-inflammation.

Platelet-Associated Immunopathology: Immuno-Thrombosis and Thrombo-Inflammation

Mechanisms of Neutrophil Extracellular Trap-Induced Thrombosis

It is unsurprising that platelet-neutrophil interactions are greatly increased during inflammatory responses (1619). For the most part, soluble mediators initiate these interactions, which directly activate these cells. Platelets and neutrophils co-incubated with septic patient plasma induced platelet adhesion to neutrophils mediated by the TLR4 receptor (20, 21).

Neutrophils are the most abundant subset of leukocytes in arterial thrombi from patients with myocardial infarction (22). Activated neutrophils express adhesion molecules belonging to the selectin and integrin families promoting platelets and the endothelium binding (23). As well, activated platelets express adhesion molecules on their surface membrane, such as P-selectin that mediates binding of platelets to its main receptor on neutrophils, P-selectin-glycoprotein-ligand-1 or PSGL-1 (24). Indeed, neutrophils and activated platelets can recruit each other to inflamed or injured tissues, thereby causing thrombo-inflammation (25). In this regard, we found that platelets can modulate neutrophil adhesion to the injured arterial wall and that both elements influence the degree of post-injury vasoconstriction in in vivo porcine model involving arterial injury by angioplasty (26). More recently, we have revealed that platelet activation and binding to neutrophils enhance the secretion of platelet MMP-2 via an adhesive interaction between P-selectin and PSGL-1, which contributes to increase platelet-neutrophil aggregation (27). Inhibition of platelet-leukocyte binding, using a recombinant PSGL-1 reduced restenosis (28) and prevented in in-stent restenosis via reduction of thrombo-inflammatory reactions (29).

Neutrophil extracellular traps (NETs) are composed of DNA, histones, and antimicrobial peptides, and they are produced as part of an antimicrobial mechanism, which is affected by immune/immune-related cells during NETosis (30). Indeed, NETosis appears to be associated with many inflammatory disorders, including infections, cancers, endothelial dysfunction, atherosclerosis, thrombosis, and ischemia (31, 32).

Neutrophil extracellular traps contribute to thrombosis through direct and indirect mechanisms. Although the vast majority of studies use NET components rather than intact NETs, the role of intact versus NET component in activating coagulation is controversial (33). In addition to their ability to promote thrombin formation, NETs were also known for providing a scaffold for pro-coagulant molecules such as VWF, fibrinogen, FXII, and tissue factor, as well as pro-coagulant extracellular vesicles TF-bearing EVs for instance (3438).

Platelets can induce dysregulation of NET, resulting in tissue damage, hypercoagulability, and thrombosis (34). In addition, NETosis is well documented in its role in the pathogenesis of sepsis and ARDS, causing vascular tissue damage and spreading microthrombi that eventually cause multiorgan failure and death (39, 40).

The studies on NETosis provide growing evidence that NETosis is connected to inflammation, atherosclerosis, and atherothrombosis, as well as poor prognoses of ischemic/reperfusion injuries (41).

Mouse studies have examined the effects of NETosis and platelet aggregation on outcome after ischemia/reperfusion (42). Then, Cf-DNAs trigger DNA-platelet and DNA-platelet-granulocyte colonization. This form of NET and platelet aggravation leads to NETosis.

There are no doubts that NETs play a significant role in thrombosis and hemostasis (Figure 1). Both arterial and venous thrombosis are affected by NETs, and NETs are implicated in stenosis that regulate thrombosis in different ways (43, 44). Additionally, preventing the formation of NETs can reduce thrombogenicity, which could be useful in preventing thrombosis (44). A thorough understanding of how new therapeutic options targeting NETs affect thrombosis will require both preclinical and clinical trials.

FIGURE 1
www.frontiersin.org

Figure 1. Neutrophil extracellular trap (NET)-mediated platelet thrombosis. In the presence of pathogens, TNFα binds to its receptor (TNFR) to initiate diverse cellular responses, and platelets, neutrophils and monocytes collaborate to form NETs that are highly thrombotic and resistant to tissue plasminogen activator-mediated fibrinolysis. NETs also help to recruit platelets which support the immuno-thrombotic process by promoting fibrin generation along with endothelial cells. Activated platelets release large amounts of pro-inflammatory cytokines in platelet extracellular vesicles. Interactions between platelets and neutrophils or monocytes occur via direct contact between cell surface receptors, or by binding of secreted ligands. PSGL-1, P-selectin glycoprotein ligand-1; TNFα, tumor necrosis factor alpha; TNFR, tumor necrosis factor receptor; GPIb, glycoprotein Ib; SLC44A2, solute carrier family 44 member 2; CD40L, cluster of differentiation 40 lignad; CD40, cluster of differentiation 40; Mac-1, macrophage-1 antigen; GPIIbIIIa, glycoprotein IIb/IIIa.

Platelet Extracellular Vesicles

In view of the critical role for platelets in thrombosis, studying platelet function may provide novel biomarkers for arterial thrombosis (45). There are numerous platelet function tests available in clinical practice, but most commonly, aggregometry-based tests are carried out (46). The aggregometry method, however, provides information on platelet functionality in the presence of exogenous and soluble agonists, which does not represent in vivo platelet activation. Thus, so far results from platelet function tests in vitro have limited value as biomarkers of arterial thrombosis.

A biomolecule or metabolite associated with activated platelets may be able to provide information about arterial thrombosis (47). Platelet-derived extracellular vesicles (PEV) are among these biomarkers, covering platelet microparticles/microvesicles and exosomes (48).

A number of benefits attributed to platelets are likely mediated by platelet-derived extracellular vesicles (PEVs), which are small vesicles released from activated platelets. Indeed, the release of platelet α-granules have been considered as small vesicles, as revealed by electron microscopic analyses (49). Also known as microparticles, this release of vesicles from the platelet plasma membrane occurs as a result of the extrusion of the platelet cytomembrane structures (50). Later studies using electron microscopy confirmed the presence of the two types of PEVs: small vesicles with diameters of 80–200 nm, and larger vesicles with diameters of 400–600 nm, which retained procoagulant activity mediated by factor V-like activity and tissue factor (51).

Platelet-derived extracellular vesicle can serve as biomarkers for autoimmune diseases, cancer, cardiovascular diseases, and infectious diseases (5255). Rheumatoid arthritis patients have PEV detected in their synovial fluid (56), and increased levels of circulating PEV correlate with disease activity (57). Additionally, in murine models of atherosclerosis and autoimmune arthritis, PEV concentrations have been found to be increased in lymph (52, 58). As a result of systemic lupus erythematosus (SLE), PEV levels in blood can be increased. Higher levels of PEV have been associated with declining kidney function (59). Recently, an increase in PEV circulating in blood of COVID-19 patients has been observed (55, 60, 61).

Aside from transporting and producing inflammation-promoting mediators such as prostaglandins and leukotrienes, the PEV is also the transporter and producer of many lipid mediators (62).

Receptors in Platelet–Immune Cell Interactions

Platelets enhance leukocyte recruitment by expressing an arsenal of complement system receptors including cC1qR, gC1qR, C3aR, and C5aR, and storing complement proteins and regulators such as C3 and factor H. Platelets stimulate the classical and alternative pathways of complement, causing the accumulation of opsonin C3b as well as the release of anaphylatoxins C3a and C5a, which chemoattract innate immune cells (63, 64).

Among innate immune system receptors, TLRs are pattern recognition receptors that link various pathogen-shared molecules, including lipopolysaccharide (LPS), lipoproteins, and other bacterial wall constituents (65). As a consequence of expressing TLRs involved in the innate immunity response, platelets may contribute to the immune response and to infections by secreting a number of inflammatory mediators and pro-inflammatory factors (5, 66, 67).

Recent studies found significantly elevated levels of CXCL1, CXCL8, CXCL12, CCL2, CCL3, CCL5, EGF, VEGF, and PDGF-AB/BB in plasma and BAL fluid of patients with severe COVID-19, all of which can be found in platelet granules, indicating that platelets may play a role in hyperinflammation observed in patients with ARDS (68, 69). These factors mediate the inflammatory response by acting synergistically, through both autocrine and paracrine pathways.

There is a wealth of evidence suggesting that platelets modulate both the innate and adaptive immune systems. Platelets and platelet-derived microparticles are active in killing foreign pathogens in addition to directing a cascade of proteases and inhibitors to sites of vascular injury and inflammation (70). As well, secreted chemokines and cytokines (RANTES, IL-1β, and MCP-1) and platelet-expressed P-selectin promote leukocyte recruitment, adhesion, and transmigration at sites of vascular injury (Figure 1).

A CD40L-expressing platelet is capable of not only increasing inflammatory responses in the endothelium, but also triggering antigen-presenting cells (dendritic cells and macrophages), resulting in enhanced antigen delivery to T lymphocytes. As demonstrated in CD40L deficient mice infected with viruses, CD40L on activated platelets also facilitates B cell differentiation and Ig class switching (71, 72). Also, patients with immune thrombocytopenia have reduced levels of regulatory T (TReg) cells, and therapeutic increases in platelet counts restore TReg cell numbers and functions in such individuals (7375). The effect of TGFβ secreted by platelets in TReg formation is unclear, but since differentiation of TReg cells requires TGFβ, platelets may contribute to TReg formation.

Platelets in Bacterial and Viral Infections

In Bacterial Infections

A wide variety of bacteria can interact with platelets, including the Staphylococci family, Neisseria gonorrhea, Porphyromonas gingivalis, and Helicobacter pylori (7678). GPIIb/IIIa, FcγRIIa, and IgG receptors on platelets help them adhere and aggregate around bacteria, as well as fibrinogen and fibronectin (79, 80). TLR1, 2, 4, 6, and 9 within platelets enable them to bind bacteria, which will either cause platelets to secrete thrombocidins (antibacterial proteins within platelet α-granule, including thrombocidin 1 and 2) or aggregate around bacteria to trap them for elimination by phagocytes (20, 8184). Upon interacting with bacteria, platelets release antimicrobial compounds that are contained within their granules. Alpha toxin released by S. Aureus mediates the release of β-defensin from platelets, which is responsible for NET formation (85). Platelets binding to Gram-negative bacteria releases a PF4 with exposed heparin-like epitopes, increasing antibody binding to the surface of bacteria and possibly leading to neutrophil opsonization and phagocytosis (86, 87). Some Gram-negative bacteria, such as Yersinia pestis, are insensitive to mammalian TLR4s, making this mechanism crucial (88, 89). Based on in vivo experiments with another Gram-negative bacteria, Porphyromonas gingivalis, it has been shown that during infection, platelets interact with neutrophils forming heterotypic aggregates in a TLR2-dependant fashion and that TLR2 promotes the aggregation of platelets (90). These study findings indicate that platelets can trigger platelet thrombotic pathway and/or inflammatory pathway activation upon recognizing bacterial components (90).

In Viral Infections

On the other hand, platelets interact with various types of viruses and their phenotype may vary depending on the type of viral infection (91). Thrombocytopenia and even thrombosis can accompany viral infections (92). Viruses can be divided into those that have either DNA or RNA genomes. Furthermore, RNA viruses can be divided into double-stranded and single-stranded viruses. Several DNA viruses, such as the herpes simplex virus type 1 (HSV1), cytomegalovirus (CMV) and vaccinia, have been identified associated with platelets. However, it is unknown if these viruses can be internalized by platelets (9395). In contrast, RNA viruses such as HIV, hepatitis C virus (HCV), dengue, influenza, CVB, and EMCV have smaller sizes and are easily internalized by platelets (96101).

There was recently evidence that SARS-CoV-2, a single-stranded RNA virus, can increase platelet activity and formation of platelet-monocyte aggregates facilitated by TF expression on monocytes (55, 102105). Koupenova et al. (106) reported that SARS-CoV-2 promotes programmed cell death in platelets in another aspect of platelet-SARS-CoV-2 interaction. According to this study, RNA sequence analysis for SARS-CoV-2 shown by ARTIC v3 sequencing, transmission electron microscopy and immunofluorescence showed that SARS-CoV-2 virions internalized when attached to microparticles. As a consequence of such internalization, apoptosis, necroptosis, and EV release occur, which contribute to impaired immunity and thrombosis (106).

Several studies suggest that platelets may associate with SARS-CoV-2 RNA molecules, and that this event may be more likely to occur in older patients (55, 105, 107). However, Bury et al. did not detect viral RNA in platelets from COVID-19 patients (108). The disparity could be due to the size of the cohort which is significantly larger in the studies that detected traces of viral RNA in platelets of some COVID-19 patients.

Conclusion

Anucleated megakaryocyte-derived platelets play an important role in hemostasis and thrombosis. Platelets serve as additional mediators of inflammation beyond hemostasis and contribute to several aspects of immune response, including priming of other immune cells and integration of extrinsic immunological stimuli. Indeed, platelets initiate innate immunity as well as adaptive immunity, which is beneficial for host defenses at certain stages of infection. The functions of platelets are thus diversified and rooted firmly in their interactions with vascular and circulating immune cells. Ultimately, uncontrolled endothelial damage and inflammation caused by viral infection progression can result in enhanced platelet reactions which amplify thrombosis and inflammation leading to higher cardiovascular, cerebral and lung pathologic events.

Author Contributions

YZ and YM wrote and edited the manuscript, read, and agreed to the published version of the manuscript.

Funding

The authors acknowledge support from the Fonds de Recherche de l’Institut de Cardiologie de Montréal (FRICM) (Montreal, QC, Canada).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med. (2007) 357:2482–94. doi: 10.1056/NEJMra071014

PubMed Abstract | CrossRef Full Text | Google Scholar

2. El-Kadiry AE, Merhi Y. The role of the proteasome in platelet function. Int J Mol Sci. (2021) 22:3999. doi: 10.3390/ijms22083999

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Ridger VC, Boulanger CM, Angelillo-Scherrer A, Badimon L, Blanc-Brude O, Bochaton-Piallat ML, et al. Microvesicles in vascular homeostasis and diseases. position paper of the European society of cardiology (ESC) working group on atherosclerosis and vascular biology. Thromb Haemost. (2017) 117:1296–316. doi: 10.1160/TH16-12-0943

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Semple JW, Italiano JE, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. (2011) 11:264–74. doi: 10.1038/nri2956

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Kapur R, Zufferey A, Boilard E, Semple JW. Nouvelle cuisine: platelets served with inflammation. J Immunol. (2015) 194:5579–87.

Google Scholar

6. Morrell CN, Aggrey AA, Chapman LM, Modjeski KL. Emerging roles for platelets as immune and inflammatory cells. Blood. (2014) 123:2759–67. doi: 10.1182/blood-2013-11-462432

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Allaoui A, Khawaja AA, Badad O, Naciri M, Lordkipanidze M, Guessous F, et al. Platelet function in viral immunity and SARS-CoV-2 infection. Semin Thromb Hemost. (2021) 47:419–26. doi: 10.1055/s-0041-1726033

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Limami Y, Senhaji N, Zaid N, Khalki L, Naya A, Hajjaj-Hassouni N, et al. PKC-delta-dependent pathways contribute to the exacerbation of the platelet activity in Crohn’s disease. Semin Thromb Hemost. (2021) 48:246–50. doi: 10.1055/s-0041-1736571

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Lindemann S, Tolley ND, Dixon DA, McIntyre TM, Prescott SM, Zimmerman GA, et al. Activated platelets mediate inflammatory signaling by regulated interleukin 1beta synthesis. J Cell Biol. (2001) 154:485–90. doi: 10.1083/jcb.200105058

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Karas SP, Rosse WF, Kurlander RJ. Characterization of the IgG-Fc receptor on human platelets. Blood. (1982) 60:1277–82.

Google Scholar

11. Idzko M, Pitchford S, Page C. Role of platelets in allergic airway inflammation. J Allergy Clin Immunol. (2015) 135:1416–23. doi: 10.1016/j.jaci.2015.04.028

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Arman M, Krauel K. Human platelet IgG Fc receptor FcgammaRIIA in immunity and thrombosis. J. Thromb Haemost. (2015) 13:893–908. doi: 10.1111/jth.12905

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Auerbach DJ, Lin Y, Miao H, Cimbro R, Difiore MJ, Gianolini ME, et al. Identification of the platelet-derived chemokine CXCL4/PF-4 as a broad-spectrum HIV-1 inhibitor. Proc Natl Acad Sci USA. (2012) 109:9569–74. doi: 10.1073/pnas.1207314109

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Thomas MR, Storey RF. The role of platelets in inflammation. Thromb Haemost. (2015) 114:449–58. doi: 10.1160/TH14-12-1067

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Engelmann B, Massberg S. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol. (2013) 13:34–45. doi: 10.1038/nri3345

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Evangelista V, Manarini S, Sideri R, Rotondo S, Martelli N, Piccoli A, et al. Platelet/polymorphonuclear leukocyte interaction: P-selectin triggers protein-tyrosine phosphorylation-dependent CD11b/CD18 adhesion: role of PSGL-1 as a signaling molecule. Blood. (1999) 93:876–85.

Google Scholar

17. Scheuerer B, Ernst M, Durrbaum-Landmann I, Fleischer J, Grage-Griebenow E, Brandt E, et al. The CXC-chemokine platelet factor 4 promotes monocyte survival and induces monocyte differentiation into macrophages. Blood. (2000) 95:1158–66.

Google Scholar

18. Kasper B, Brandt E, Brandau S, Petersen F. Platelet factor 4 (CXC chemokine ligand 4) differentially regulates respiratory burst, survival, and cytokine expression of human monocytes by using distinct signaling pathways. J Immunol. (2007) 179:2584–91. doi: 10.4049/jimmunol.179.4.2584

PubMed Abstract | CrossRef Full Text | Google Scholar

19. von Hundelshausen P, Koenen RR, Sack M, Mause SF, Adriaens W, Proudfoot AE, et al. Heterophilic interactions of platelet factor 4 and RANTES promote monocyte arrest on endothelium. Blood. (2005) 105:924–30. doi: 10.1182/blood-2004-06-2475

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. (2007) 13:463–9. doi: 10.1038/nm1565

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Carestia A, Kaufman T, Rivadeneyra L, Landoni VI, Pozner RG, Negrotto S, et al. Mediators and molecular pathways involved in the regulation of neutrophil extracellular trap formation mediated by activated platelets. J Leukoc Biol. (2016) 99:153–62. doi: 10.1189/jlb.3A0415-161R

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Novotny J, Chandraratne S, Weinberger T, Philippi V, Stark K, Ehrlich A, et al. Histological comparison of arterial thrombi in mice and men and the influence of Cl-amidine on thrombus formation. PLoS One. (2018) 13:e0190728. doi: 10.1371/journal.pone.0190728

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Swystun LL, Liaw PC. The role of leukocytes in thrombosis. Blood. (2016) 128:753–62. doi: 10.1182/blood-2016-05-718114

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Massberg S, Brand K, Gruner S, Page S, Muller E, Muller I, et al. A critical role of platelet adhesion in the initiation of atherosclerotic lesion formation. J Exp Med. (2002) 196:887–96. doi: 10.1084/jem.20012044

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Sreeramkumar V, Adrover JM, Ballesteros I, Cuartero MI, Rossaint J, Bilbao I, et al. Neutrophils scan for activated platelets to initiate inflammation. Science. (2014) 346:1234–8. doi: 10.1126/science.1256478

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Merhi Y, Provost P, Chauvet P, Theoret JF, Phillips ML, Latour JG. Selectin blockade reduces neutrophil interaction with platelets at the site of deep arterial injury by angioplasty in pigs. Arterioscler Thromb Vasc Biol. (1999) 19:372–7. doi: 10.1161/01.atv.19.2.372

CrossRef Full Text | Google Scholar

27. Abou-Saleh H, Theoret JF, Yacoub D, Merhi Y. Neutrophil P-selectin-glycoprotein-ligand-1 binding to platelet P-selectin enhances metalloproteinase 2 secretion and platelet-neutrophil aggregation. Thromb Haemost. (2005) 94:1230–5. doi: 10.1160/TH05-05-0344

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Bienvenu JG, Tanguay JF, Theoret JF, Kumar A, Schaub RG, Merhi Y. Recombinant soluble p-selectin glycoprotein ligand-1-Ig reduces restenosis through inhibition of platelet-neutrophil adhesion after double angioplasty in swine. Circulation. (2001) 103:1128–34. doi: 10.1161/01.cir.103.8.1128

CrossRef Full Text | Google Scholar

29. Tanguay JF, Geoffroy P, Sirois MG, Libersan D, Kumar A, Schaub RG, et al. Prevention of in-stent restenosis via reduction of thrombo-inflammatory reactions with recombinant P-selectin glycoprotein ligand-1. Thromb Haemost. (2004) 91:1186–93. doi: 10.1160/TH03-11-0701

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Brinkmann V, Reichard U, Goosmann C, Fauler B, Uhlemann Y, Weiss DS, et al. Neutrophil extracellular traps kill bacteria. Science. (2004) 303:1532–5. doi: 10.1126/science.1092385

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Remijsen Q, Kuijpers TW, Wirawan E, Lippens S, Vandenabeele P, Vanden Berghe T. Dying for a cause: NETosis, mechanisms behind an antimicrobial cell death modality. Cell Death Differ. (2011) 18:581–8. doi: 10.1038/cdd.2011.1

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Kaplan MJ, Radic M. Neutrophil extracellular traps: double-edged swords of innate immunity. J Immunol. (2012) 189:2689–95. doi: 10.4049/jimmunol.1201719

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Noubouossie DF, Reeves BN, Strahl BD, Key NS. Neutrophils: back in the thrombosis spotlight. Blood. (2019) 133:2186–97. doi: 10.1182/blood-2018-10-862243

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Fuchs TA, Brill A, Duerschmied D, Schatzberg D, Monestier M, Myers DD Jr., et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci USA. (2010) 107:15880–5. doi: 10.1073/pnas.1005743107

PubMed Abstract | CrossRef Full Text | Google Scholar

35. von Bruhl ML, Stark K, Steinhart A, Chandraratne S, Konrad I, Lorenz M, et al. Massberg, Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med. (2012) 209:819–35. doi: 10.1084/jem.20112322

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Stakos DA, Kambas K, Konstantinidis T, Mitroulis I, Apostolidou E, Arelaki S, et al. Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur Heart J. (2015) 36:1405–14. doi: 10.1093/eurheartj/ehv007

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Wang Y, Luo L, Braun OO, Westman J, Madhi R, Herwald H, et al. Neutrophil extracellular trap-microparticle complexes enhance thrombin generation via the intrinsic pathway of coagulation in mice. Sci Rep. (2018) 8:4020. doi: 10.1038/s41598-018-22156-5

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Thomas GM, Brill A, Mezouar S, Crescence L, Gallant M, Dubois C, et al. Tissue factor expressed by circulating cancer cell-derived microparticles drastically increases the incidence of deep vein thrombosis in mice. J Thromb Haemost. (2015) 13:1310–9. doi: 10.1111/jth.13002

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Czaikoski PG, Mota JM, Nascimento DC, Sonego F, Castanheira FV, Melo PH, et al. Neutrophil extracellular traps induce organ damage during experimental and clinical sepsis. PLoS One. (2016) 11:e0148142. doi: 10.1371/journal.pone.0148142

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Lefrancais E, Mallavia B, Zhuo H, Calfee CS, Looney MR. Maladaptive role of neutrophil extracellular traps in pathogen-induced lung injury. JCI Insight. (2018) 3:e98178. doi: 10.1172/jci.insight.98178

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Qi H, Yang S, Zhang L. Neutrophil extracellular traps and endothelial dysfunction in atherosclerosis and thrombosis. Front Immunol. (2017) 8:928. doi: 10.3389/fimmu.2017.00928

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Shrestha B, Ito T, Kakuuchi M, Totoki T, Nagasato T, Yamamoto M, et al. Recombinant thrombomodulin suppresses histone-induced neutrophil extracellular trap formation. Front Immunol. (2019) 10:2535. doi: 10.3389/fimmu.2019.02535

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Kopytek M, Kolasa-Trela R, Zabczyk M, Undas A, Natorska J. NETosis is associated with the severity of aortic stenosis: links with inflammation. Int J Cardiol. (2019) 286:121–6. doi: 10.1016/j.ijcard.2019.03.047

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Laridan E, Martinod K, De Meyer SF. Neutrophil extracellular traps in arterial and venous thrombosis. Semin Thromb Hemost. (2019) 45:86–93. doi: 10.1055/s-0038-1677040

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Jurk K. Analysis of platelet function and dysfunction. Hamostaseologie. (2015) 35:60–72. doi: 10.5482/HAMO-14-09-0047

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Kim MH, Choi SY, An SY, Serebruany V. Validation of three platelet function tests for bleeding risk stratification during dual antiplatelet therapy following coronary interventions. Clin Cardiol. (2016) 39:385–90. doi: 10.1002/clc.22540

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Ferroni P, Riondino S, Vazzana N, Santoro N, Guadagni F, Davi G. Biomarkers of platelet activation in acute coronary syndromes. Thromb Haemost. (2012) 108:1109–23. doi: 10.1160/TH12-08-0550

PubMed Abstract | CrossRef Full Text | Google Scholar

48. van der Pol E, Boing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol Rev. (2012) 64:676–705. doi: 10.1124/pr.112.005983

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Webber AJ, Johnson SA. Platelet participation in blood coagulation aspects of hemostasis. Am J Pathol. (1970) 60:19–42.

Google Scholar

50. Crawford N. The presence of contractile proteins in platelet microparticles isolated from human and animal platelet-free plasma. Br J Haematol. (1971) 21:53–69. doi: 10.1111/j.1365-2141.1971.tb03416.x

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Sandberg H, Bode AP, Dombrose FA, Hoechli M, Lentz BR. Expression of coagulant activity in human platelets: release of membranous vesicles providing platelet factor 1 and platelet factor 3. Thromb Res. (1985) 39:63–79. doi: 10.1016/0049-3848(85)90122-7

CrossRef Full Text | Google Scholar

52. Tessandier N, Melki I, Cloutier N, Allaeys I, Miszta A, Tan S, et al. Platelets disseminate extracellular vesicles in lymph in rheumatoid arthritis. Arterioscler Thromb Vasc Biol. (2020) 40:929–42. doi: 10.1161/ATVBAHA.119.313698

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Gitz E, Pollitt AY, Gitz-Francois JJ, Alshehri O, Mori J, Montague S, et al. CLEC-2 expression is maintained on activated platelets and on platelet microparticles. Blood. (2014) 124:2262–70. doi: 10.1182/blood-2014-05-572818

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Boilard E, Pare G, Rousseau M, Cloutier N, Dubuc I, Levesque T, et al. Influenza virus H1N1 activates platelets through FcgammaRIIA signaling and thrombin generation. Blood. (2014) 123:2854–63. doi: 10.1182/blood-2013-07-515536

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Zaid Y, Puhm F, Allaeys I, Naya A, Oudghiri M, Khalki L, et al. Platelets can associate with SARS-Cov-2 RNA and are hyperactivated in COVID-19. Circ Res. (2020) 127:1404–18. doi: 10.1161/CIRCRESAHA.120.317703

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Boilard E, Nigrovic PA, Larabee K, Watts GF, Coblyn JS, Weinblatt ME, et al. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science. (2010) 327:580–3. doi: 10.1126/science.1181928

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Knijff-Dutmer EA, Koerts J, Nieuwland R, Kalsbeek-Batenburg EM, van de Laar MA. Elevated levels of platelet microparticles are associated with disease activity in rheumatoid arthritis. Arthritis Rheum. (2002) 46:1498–503. doi: 10.1002/art.10312

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Milasan A, Tessandier N, Tan S, Brisson A, Boilard E, Martel C. Extracellular vesicles are present in mouse lymph and their level differs in atherosclerosis. J Extracell Vesicles. (2016) 5:31427. doi: 10.3402/jev.v5.31427

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Mobarrez F, Vikerfors A, Gustafsson JT, Gunnarsson I, Zickert A, Larsson A, et al. Microparticles in the blood of patients with systemic lupus erythematosus (SLE): phenotypic characterization and clinical associations. Sci Rep. (2016) 6:36025. doi: 10.1038/srep36025

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Guervilly C, Bonifay A, Burtey S, Sabatier F, Cauchois R, Abdili E, et al. Dissemination of extreme levels of extracellular vesicles: tissue factor activity in patients with severe COVID-19. Blood Adv. (2021) 5:628–34. doi: 10.1182/bloodadvances.2020003308

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Cappellano G, Raineri D, Rolla R, Giordano M, Puricelli C, Vilardo B, et al. Circulating platelet-derived extracellular vesicles are a hallmark of Sars-Cov-2 infection. Cells. (2021) 10:10.3390/cells10010085.

Google Scholar

62. Boilard E. Extracellular vesicles and their content in bioactive lipid mediators: more than a sack of microRNA. J Lipid Res. (2018) 59:2037–46. doi: 10.1194/jlr.R084640

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Peerschke EI, Yin W, Ghebrehiwet B. Platelet mediated complement activation. Adv Exp Med Biol. (2008) 632:81–91. doi: 10.1007/978-0-387-78952-1_7

CrossRef Full Text | Google Scholar

64. Del Conde I, Cruz MA, Zhang H, Lopez JA, Afshar-Kharghan V. Platelet activation leads to activation and propagation of the complement system. J Exp Med. (2005) 201:871–9. doi: 10.1084/jem.20041497

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Janeway CA Jr., Medzhitov R. Innate immune recognition. Annu Rev Immunol. (2002) 20:197–216. doi: 10.1146/annurev.immunol.20.083001.084359

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Ebermeyer T, Cognasse F, Berthelot P, Mismetti P, Garraud O, Hamzeh-Cognasse H. Platelet innate immune receptors and TLRs: a double-edged sword. Int J Mol Sci. (2021) 22:7894. doi: 10.3390/ijms22157894

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Banerjee M, Huang Y, Joshi S, Popa GJ, Mendenhall MD, Wang QJ, et al. Platelets endocytose viral particles and are activated via TLR (toll-like receptor) signaling. Arterioscler Thromb Vasc Biol. (2020) 40:1635–50. doi: 10.1161/ATVBAHA.120.314180

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Zaid Y, Dore E, Dubuc I, Archambault AS, Flamand O, Laviolette M, et al. Chemokines and eicosanoids fuel the hyperinflammation within the lungs of patients with severe COVID-19. J Allergy Clin Immunol. (2021) 148:368.e–80.e. doi: 10.1016/j.jaci.2021.05.032

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Archambault AS, Zaid Y, Rakotoarivelo V, Turcotte C, Dore E, Dubuc I, et al. High levels of eicosanoids and docosanoids in the lungs of intubated COVID-19 patients. FASEB J. (2021) 35:e21666. doi: 10.1096/fj.202100540R

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Peerschke EI, Yin W, Ghebrehiwet B. Complement activation on platelets: implications for vascular inflammation and thrombosis. Mol Immunol. (2010) 47:2170–5. doi: 10.1016/j.molimm.2010.05.009

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Elzey BD, Schmidt NW, Crist SA, Kresowik TP, Harty JT, Nieswandt B, et al. Platelet-derived CD154 enables T-cell priming and protection against Listeria monocytogenes challenge. Blood. (2008) 111:3684–91. doi: 10.1182/blood-2007-05-091728

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Elzey BD, Sprague DL, Ratliff TL. The emerging role of platelets in adaptive immunity. Cell Immunol. (2005) 238:1–9. doi: 10.1016/j.cellimm.2005.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Ling Y, Cao X, Yu Z, Ruan C. Circulating dendritic cells subsets and CD4+Foxp3+ regulatory T cells in adult patients with chronic ITP before and after treatment with high-dose dexamethasome. Eur J Haematol. (2007) 79:310–6. doi: 10.1111/j.1600-0609.2007.00917.x

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Liu B, Zhao H, Poon MC, Han Z, Gu D, Xu M, et al. Abnormality of CD4(+)CD25(+) regulatory T cells in idiopathic thrombocytopenic purpura. Eur J Haematol. (2007) 78:139–43. doi: 10.1111/j.1600-0609.2006.00780.x

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Stasi R, Cooper N, Del Poeta G, Stipa E, Laura Evangelista M, Abruzzese E, et al. Analysis of regulatory T-cell changes in patients with idiopathic thrombocytopenic purpura receiving B cell-depleting therapy with rituximab. Blood. (2008) 112:1147–50. doi: 10.1182/blood-2007-12-129262

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Yeaman MR. Platelets in defense against bacterial pathogens. Cell Mol Life Sci. (2010) 67:525–44. doi: 10.1007/s00018-009-0210-4

PubMed Abstract | CrossRef Full Text | Google Scholar

77. Fitzgerald JR, Foster TJ, Cox D. The interaction of bacterial pathogens with platelets. Nat Rev Microbiol. (2006) 4:445–57. doi: 10.1038/nrmicro1425

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Hamzeh-Cognasse H, Damien P, Chabert A, Pozzetto B, Cognasse F, Garraud O. Platelets and infections - complex interactions with bacteria. Front Immunol. (2015) 6:82. doi: 10.3389/fimmu.2015.00082

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Fitzgerald JR, Loughman A, Keane F, Brennan M, Knobel M, Higgins J, et al. Fibronectin-binding proteins of Staphylococcus aureus mediate activation of human platelets via fibrinogen and fibronectin bridges to integrin GPIIb/IIIa and IgG binding to the FcgammaRIIa receptor. Mol Microbiol. (2006) 59:212–30. doi: 10.1111/j.1365-2958.2005.04922.x

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Loughman A, Fitzgerald JR, Brennan MP, Higgins J, Downer R, Cox D, et al. Roles for fibrinogen, immunoglobulin and complement in platelet activation promoted by Staphylococcus aureus clumping factor A. Mol Microbiol. (2005) 57:804–18. doi: 10.1111/j.1365-2958.2005.04731.x

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Beaulieu LM, Freedman JE. The role of inflammation in regulating platelet production and function: toll-like receptors in platelets and megakaryocytes. Thromb Res. (2010) 125:205–9. doi: 10.1016/j.thromres.2009.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Aslam R, Speck ER, Kim M, Crow AR, Bang KW, Nestel FP, et al. Platelet Toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-alpha production in vivo. Blood. (2006) 107:637–41. doi: 10.1182/blood-2005-06-2202

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Yeaman MR, Puentes SM, Norman DC, Bayer AS. Partial characterization and staphylocidal activity of thrombin-induced platelet microbicidal protein. Infect Immun. (1992) 60:1202–9. doi: 10.1128/iai.60.3.1202-1209.1992

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Krijgsveld J, Zaat SA, Meeldijk J, van Veelen PA, Fang G, Poolman B, et al. Thrombocidins, microbicidal proteins from human blood platelets, are C-terminal deletion products of CXC chemokines. J Biol Chem. (2000) 275:20374–81. doi: 10.1074/jbc.275.27.20374

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Kraemer BF, Campbell RA, Schwertz H, Cody MJ, Franks Z, Tolley ND, et al. Novel anti-bacterial activities of beta-defensin 1 in human platelets: suppression of pathogen growth and signaling of neutrophil extracellular trap formation. PLoS Pathog. (2011) 7:e1002355. doi: 10.1371/journal.ppat.1002355

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Krauel K, Weber C, Brandt S, Zahringer U, Mamat U, Greinacher A, et al. Platelet factor 4 binding to lipid A of Gram-negative bacteria exposes PF4/heparin-like epitopes. Blood. (2012) 120:3345–52. doi: 10.1182/blood-2012-06-434985

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Krauel K, Potschke C, Weber C, Kessler W, Furll B, Ittermann T, et al. Platelet factor 4 binds to bacteria, [corrected] inducing antibodies cross-reacting with the major antigen in heparin-induced thrombocytopenia. Blood. (2011) 117:1370–8. doi: 10.1182/blood-2010-08-301424

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Dziarski R. Deadly plague versus mild-mannered TLR4. Nat Immunol. (2006) 7:1017–9. doi: 10.1038/ni1006-1017

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Montminy SW, Khan N, McGrath S, Walkowicz MJ, Sharp F, Conlon JE, et al. Virulence factors of Yersinia pestis are overcome by a strong lipopolysaccharide response. Nat Immunol. (2006) 7:1066–73. doi: 10.1038/ni1386

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Blair P, Rex S, Vitseva O, Beaulieu L, Tanriverdi K, Chakrabarti S, et al. Stimulation of Toll-like receptor 2 in human platelets induces a thromboinflammatory response through activation of phosphoinositide 3-kinase. Circ Res. (2009) 104:346–54. doi: 10.1161/CIRCRESAHA.108.185785

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Zaid Y, Guessous F, Puhm F, Elhamdani W, Chentoufi L, Morris AC, et al. Platelet reactivity to thrombin differs between patients with COVID-19 and those with ARDS unrelated to COVID-19. Blood Adv. (2021) 5:635–9. doi: 10.1182/bloodadvances.2020003513

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Koupenova M, Clancy L, Corkrey HA, Freedman JE. Circulating platelets as mediators of immunity, inflammation, and thrombosis. Circ Res. (2018) 122:337–51. doi: 10.1161/CIRCRESAHA.117.310795

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Assinger A, Kral JB, Yaiw KC, Schrottmaier WC, Kurzejamska E, Wang Y, et al. Human cytomegalovirus-platelet interaction triggers toll-like receptor 2-dependent proinflammatory and proangiogenic responses. Arterioscler Thromb Vasc Biol. (2014) 34:801–9. doi: 10.1161/ATVBAHA.114.303287

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Forghani B, Schmidt NJ. Association of herpes simplex virus with platelets of experimentally infected mice. Arch Virol. (1983) 76:269–74. doi: 10.1007/BF01311111

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Bik T, Sarov I, Livne A. Interaction between vaccinia virus and human blood platelets. Blood. (1982) 59:482–7.

Google Scholar

96. Koupenova M, Vitseva O, MacKay CR, Beaulieu LM, Benjamin EJ, Mick E, et al. Platelet-TLR7 mediates host survival and platelet count during viral infection in the absence of platelet-dependent thrombosis. Blood. (2014) 124:791–802. doi: 10.1182/blood-2013-11-536003

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Youssefian T, Drouin A, Masse JM, Guichard J, Cramer EM. Host defense role of platelets: engulfment of HIV and Staphylococcus aureus occurs in a specific subcellular compartment and is enhanced by platelet activation. Blood. (2002) 99:4021–9. doi: 10.1182/blood-2001-12-0191

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Danon D, Jerushalmy Z, De Vries A. Incorporation of influenza virus in human blood platelets in vitro. Electron microscopical observation. Virology. (1959) 9:719–22. doi: 10.1016/0042-6822(59)90168-0

CrossRef Full Text | Google Scholar

99. de Almeida AJ, Campos-de-Magalhaes M, Brandao-Mello CE, de Oliveira RV, do Espirito-Santo MP, Yoshida CF, et al. Detection of hepatitis C virus in platelets: evaluating its relationship to antiviral therapy outcome. Hepatogastroenterology. (2009) 56:429–36.

Google Scholar

100. Noisakran S, Gibbons RV, Songprakhon P, Jairungsri A, Ajariyakhajorn C, Nisalak A, et al. Detection of dengue virus in platelets isolated from dengue patients. Southeast Asian J Trop Med Public Health. (2009) 40:253–62.

Google Scholar

101. Negrotto S, Jaquenod de Giusti C, Rivadeneyra L, Ure AE, Mena HA, Schattner M, et al. Platelets interact with Coxsackieviruses B and have a critical role in the pathogenesis of virus-induced myocarditis. J Thromb Haemost. (2015) 13:271–82. doi: 10.1111/jth.12782

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Zhang S, Liu Y, Wang X, Yang L, Li H, Wang Y, et al. SARS-CoV-2 binds platelet ACE2 to enhance thrombosis in COVID-19. J Hematol Oncol. (2020) 13:120. doi: 10.1186/s13045-020-00954-7

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Shen S, Zhang J, Fang Y, Lu S, Wu J, Zheng X, et al. SARS-CoV-2 interacts with platelets and megakaryocytes via ACE2-independent mechanism. J Hematol Oncol. (2021) 14:72. doi: 10.1186/s13045-021-01082-6

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Hottz ED, Azevedo-Quintanilha IG, Palhinha L, Teixeira L, Barreto EA, Pao CRR, et al. Platelet activation and platelet-monocyte aggregate formation trigger tissue factor expression in patients with severe COVID-19. Blood. (2020) 136:1330–41. doi: 10.1182/blood.2020007252

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Manne BK, Denorme F, Middleton EA, Portier I, Rowley JW, Stubben C, et al. Platelet gene expression and function in patients with COVID-19. Blood. (2020) 136:1317–29. doi: 10.1182/blood.2020007214

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Koupenova M, Corkrey HA, Vitseva O, Tanriverdi K, Somasundaran M, Liu P, et al. SARS-CoV-2 initiates programmed cell death in platelets. Circ Res. (2021) 129:631–46. doi: 10.1161/CIRCRESAHA.121.319117

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Zaid Y, Guessous F. The ongoing enigma of SARS-CoV-2 and platelet interaction. Res Pract Thromb Haemost. (2022) 6:e12642. doi: 10.1002/rth2.12642

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Bury L, Camilloni B, Castronari R, Piselli E, Malvestiti M, Borghi M, et al. Search for SARS-CoV-2 RNA in platelets from COVID-19 patients. Platelets. (2021) 32:284–7. doi: 10.1080/09537104.2020.1859104

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: platelets, immuno-thrombosis, thrombo-inflammation, NETosis, extracellular vesicles (EVs)

Citation: Zaid Y and Merhi Y (2022) Implication of Platelets in Immuno-Thrombosis and Thrombo-Inflammation. Front. Cardiovasc. Med. 9:863846. doi: 10.3389/fcvm.2022.863846

Received: 27 January 2022; Accepted: 04 March 2022;
Published: 25 March 2022.

Edited by:

Robert Campbell, The University of Utah, United States

Reviewed by:

Frederik Denorme, The University of Utah, United States
Nirav Dhanesha, The University of Iowa, United States

Copyright © 2022 Zaid and Merhi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Younes Zaid, y.zaid@um5r.ac.ma

Download