Take a look at the Recent articles

Platelet-derived vesicles in acute myocardial infarction

Alexander E Berezin

Internal Medicine Department, State Medical University of Zaporozhye, Ukraine

Medical Academy of Postgraduate Education, Zaporozhye, Ukraine

Alexander A Berezin

Internal Medicine Department, State Medical University of Zaporozhye, Ukraine

Medical Academy of Postgraduate Education, Zaporozhye, Ukraine

DOI: 10.15761/CRT.1000262

Article
Article Info
Author Info
Figures & Data

Abstract

Extracellular vesicles (EVs) are involved in the pathogenesis of CV diseases through several biological mechanisms that support microvascular inflammation, arterial stiffness, vascular calcification, atherosclerotic plaque shaping and rupture, endothelial dysfunction, hypercoagulation and thrombosis, cardiac remodelling, and kidney dysfunction. Platelet activation, which is associated with secretion of EV, is common for acute coronary syndrome (ACS) and myocardial infarction (MI). The aim of the mini review is to discuss the role of platelet-derived micro vesicles as predictive biomarker in coronary atherothrombotic events. It has found that elevated levels of EVs and altered signature of platelet-derived vesicles are biomarkers of endothelial dysfunction, accelerating atherosclerosis, pro-coagulation state, and poor clinical outcomes in acute MI. Platelet-derived vesicles are biomarker of endothelial dysfunction and pro-coagulative activity, which can be discussed a predictor of poor outcomes in ACS/MI, while the diagnostic and predictive roles of this biomarker require to be investigated in large clinical trials in the future.

Key words

myocardial infarction, extracellular vesicles, biomarkers, prognosis

Introduction

Extracellular vesicles (EVs) are defined as small sized (range of diameter is approximately 150 nm and less) exosomes that deliver biologically active molecules (peptides, RNAs, active molecules, growth factors, hormones, enzymes) from mother cells to recipient cells through binding, fusion or endocytosis appearing to be a core element in cell-to-cell cooperation [1,2]. They are involved in the pathogenesis of CV diseases through several biological mechanisms that support microvascular inflammation, arterial stiffness, vascular calcification, atherosclerotic plaque shaping and rupture, endothelial dysfunction, hypercoagulation and thrombosis, cardiac remodelling, and kidney dysfunction [3,4]. Additionally, via intracellular signaling cascades EVs play a pivotal role in tissue reparation, angiogenesis, and neovascularization [5,6]. In fact, EVs mediate autocrine and paracrine signals that are enabled to reconstructing the homeostatic micro-environment in heart and vessels [7]. EVs are released a wide spectrum of cells, including endothelial cells, cardiomyocytes, red blood cells, mononuclears / macrophages, as well as platelets. EVs, which are produced by endothelial precursors and mature endothelial cells, as well as mononuclears, are under investigations for long time respectively and there is evidence regarding predictive capability of altered circulating number of EVs in established CV disease including hypertension [8,9]. On contrary, the exact role of platelet-derived micro vesicles in pathogenesis of hypertension remains to be not fully discovered. The aim of the mini review is to discuss the role of platelet-derived micro vesicles as predictive biomarker in acute myocardial infacrtion.

Extracellular vesicles: definition

Extracellular vesicles (EVs) are defined as small sized (<1000 nm) cell membranous originated structures that secreted by various cells (platelets, red and white blood cells, endothelial cells, precursors / stem cells, cardiomyocytes, and even tumor cells) into human fluids and contained wide range of proteins, hormones, signalling molecules, lipids, mRNA, miRNA, and others [10,11]. There are at least two distinct subpopulations of EVs (exosomes and micro vesicles [MVs]), which distinguish each other in size, compounds, immune phenotypes and wide range of formation modes. Exosomes are sufficiently smaller to MVs (size averages are 30-120 nm and 40–1000 nm respectively) and are shaped by the endocytosis of multivesicular bodies and are shed from cells by exocytosis. On contrary, MVs are defined as membrane-bound vesicles, which are released from the plasma membrane. Additionally, surface-specific antigens, which are presented for both populations of EVs, are different, but they overlap between exosomes and MVs that does not allow differencing both populations of the particles. Physiologically, EVs are a cargo for molecule transfer, auto / paracrine signalling, cell-to-cell cooperation, playing a pivotal role in immune response, vascular integrity, cellular senescence, tissue reparation and development, angiogenesis and neovascularization [12,13]. In fact, EVs are able to mediate the exchange of appropriate information between various cells. EVs were found witness and biomarkers of several diseases, such as CV, autoimmune, infective, metabolic, renal, rheumatic, neurological, ocular diseases and malignancy [14-17].

Platelet-derived vesicles

The population of the platelet-derived vesicles is the largest in other types of EVs in the circulation. Indeed, the majority of the blood-borne EVs are thought to originate from the megakaryocytes, i.e. either from circulating platelets or directly from the platelet precursor cells, which reside in the bone marrow. Although coagulation, thrombosis, vascular senescence and permeability are most common and well established biological function of platelet-derived vesicles, the role of them in CV diseases has not exhausted a crucial impact on hemostasis. Elevated levels of platelet-derived vesicles were determined in patients with acute coronary syndrome, myocardial infarction, as well as heparin-induced thrombocytopenia, thrombotic complications, hemolytic uremic syndrome, while developing of abdominal obesity, diabetes mellitus, antiphospholipid syndrome, and sepsis was not associated with increased circulating level of platelet-derived vesicles [18]. Probably, this evidence might relate to the mechanisms regarding release of MPs from platelets. It has suggested that in various cases circulating platelets are likely to adhere to leukocytes or endothelial cells at the activation site and that the circulating platelet-derived MPs are likely to be a residue of activated platelets [19].

It has been found that upon activation, blood platelets are able to release two types of EVs, i.e. micro particles and exosomes. Platelet-derived micro particles are characterized by average of size of 70-100 nm and the phosphatidylserine (PS) presentation at their surface, which mediates their capabilities in coagulation and thrombosis [20]. On contrary, platelet-derived exosomes are characterized by their small size (<80 nm) and the presence of CD63 antigen, which is known as the tetraspanin that belongs to the transmembrane 4 superfamily [19,20]. CD63 plays a pivotal role in the role in the activation of cellular signalling cascades that trigger AkT, FAK-related non-kinase polypeptide / protein tyrosine kinase-2 (PTK-2) and mitogen-activated protein (MAP) kinases, attenuates an adhesion of circulating monocytes / macrophages onto endothelial cells through the regulation of P-selectin (CD62) trafficking, as well as promotes vascular endothelial growth factor receptor (VEGFR) signalling and internationalization [21]. All these factors are embedded into cell-to-cell cooperation and cell interactions with the extracellular matrix. Moreover, EVs expressing Ms4a2/FceRI on the surface are able to be triggers for degranulation of circulating cell in response to other stimuli [22]. Additionally, some EVs may express CD41 (Integrin alpha-IIb) on their surface, which in a complex with CD61 may be a receptor for thrombospondin, fibronectin, vitronectin, fibrinogen, and von Willebrand factor playing a crucial role in blood coagulation [22,23]. The populations of the platelet-derived EVs may distinguish each other in not just their size and immune phenotype, but yet they could have different in density (utilized as sucrose density gradient in ultracentrifugation), proteomic and lipidomic profiles [24] Labeling of platelet-derived EVs is reported in the Table 1.

Table 1. Labelling of platelet-derived EVs with surface CD markers

CD marker

Alternative name

Prominent feature

Cell labelling

Expression

Significance

CD9

Tetraspanin (Tspan 29)

Key regulator of intracellular signalling, proliferation, activation, survival, migration, invasion, cell adhesion, and diapedesis

Platelets, B cells, CD4+ T cells, CD8+ T cells, natural killer cells, granulocytes, monocytes and macrophages, and immature and mature dendritic cells

Low

None-specific lympho / hematopoietic marker

CD23

Fc epsilon RII (FcεRII)

Interaction with many ligands (IgE, CD21, MHC class II,integrins), exhibiting pleiotropic cytokine-like activities, sustaining the growth of activated B-cells, differentiation of myeloid precursors, thymocytes and bone marrow CD4+ T cells, degranulation of platelets

Platelets, T and B lymphocytes, polymorphonuclear leucocytes, monocytes, follicular dendritic cells, intestinal epithelial cells, bone marrow stromal cells

Low

None-specific lymphohematopoietic marker, diagnostic marker for neoplastic and autoimmune inflammatory disease

CD31

PECAM-1, GP IIa, hec7 antigen, EndoCAM

Key inhibitory receptor in circulating platelets and leukocytes, major endothelial mechanosensor, modulator of vascular permeability, and leukocyte trafficking, and cell-to-cell cooperation, core player in in thrombosis, inflammation and the immune response

Platelets, leucocytes, monocytes / mononuclears, endothelial cells, EPCs

High

A maker of endothelial integrity, vascular permeability, platelet degranulation, endothelialopathies in tumor angiogenesis and the growth and development of hemangiomas

CD36

GP-IV, Scarb3

Recognition of thrombospondine, fatty acids, collagen, adhesion of platelets

Monocytes, macrophages, platelets, adipose cells, endothelial cells, PECs, epithelial cells

High

Platelet collagen receptor

CD41

αIIb integrin, platelet GPIIb

Key regulator of growth and differentiation

Hematopoetic progenitors in embrio, specific to the megakaryocytic / platelet lineage

Moderate

Markers of all progenitors (megakaryocytic, granulomacrophagic, erythroid and pluripotent cells)

CD42 (a, b, c)

Integrin subunit αIIb, GPIIb

Regulator of platelet activation and adhesion

Platelets

High

Marker of platelet aggregation

CD49f

GPIc, integrin α6-chain, ITGA6, ITGA6B, VLA 6 alpha Subunit

Regulator of embryogenesis, cell adhesion and cell migration through T-cell receptor/CD3 complex

T cells, endothelial cells, epithelial cell, macrophages, monocytes; platelets and stem cell

High

Marker of blood cell adhesion and T-cell proliferation

CD51

Integrin subunit alpha V, vitronectin receptor

Trigger of cell adhesion and signal transduction, regulator of angiogenesis and cancer progression

T cells, endothelial cells, platelets

High

Marker of cell adhesion to sequences of von Willebrand factor, vitronectin, fibrinogen and thrombospondin

CD61

GP3A, integrin subunit beta 3

Regulator of cell adhesion and cell-surface mediated signalling

Endothelial cells, Macrophage, platelets

High

Marker of human megakaryocytic cell lineages

CD84

Cell surface antigen MAX.3, Ly-9B, SLAMF-5

Regulator of signalling and adhesion in lymphocytic cells and other cells

Thymus and spleen cells, cells of megakaryocytic lineages

Moderate

Marker of adhesion interactions between T lymphocytes and accessory cells, platelets

CD92

Choline Transporter like Protein 1

Regulator of cell growth and differentiation

Subset of bone marrow stromal cells, cells of osteogenic, megakaryocytic and adipogenic lineages

High

Marker of human bone marrow stromal cells, and cells of megakaryocytic and adipogenic lineages

CD110

MPL proto-oncogene, TPO-R,

Regulator of heamopoesis

Subset of stem cells and cells of megakaryocytic lineage as well as platelets

High

Marker of megakaryocytic lineage

CD147

BSG, EMMPRIN

Trigger of cell proliferation associated with the TGF-β/Smad4 signalling pathway, cell adhesion and T-cell activation

Endothelial cell, epithelial cells, lymphocytes, mononuclears, platelets

Low

Marker of cell growth, adhesion, and proliferation, malignancy, angiogenesis, vascular permeability

CD151

GP27, membrane glycoprotein SFA-1, platelet-endothelia-l tetraspan antigen-3

Regulator of the VCAM-1 activity during lymphocyte and platelet recruitment

Endothelial cells, platelets, T-cells

High

Marker of fibrosis, vascular inflammation and remodelling, cancinogenesis, vascular permeability, endothelial dysfunction

CD226

DNAM1, PTA1, TLiSA1, DNAX

An adhesion molecule involved in NK and T cell-mediated cytotoxicity

T cells, NK cells, platelets, mononuclears

Low

Triggering cytotoxicity and cytokine secretion by T and NK cells

GARP

-

A key regulator of network between Tregs and its targets

Activated Tregs, mature peripheral naive CD4+ T cells, platelets

Low

Marker of activated Tregs and degradated platelets

LAP

-

Phagosome maturation, stabilization of the cargo to prolong antigen presentation on MHC, recruiting LC3 molecules to phagosome membranes class II molecules

Macrophages, dendritic cells and platelets

Low

A marker of catabolic process involved during exogenous antigen processing, recognition of apoptotic, necrotic, or entotic cells, degradation of platelets

Abbreviation: MHC: major histocompatibility complex; GP: glycoprotein; PECAM-1: platelet/endothelial cell adhesion molecule 1; EPCs: endothelial progenitor cells; NK: nature killer; TPO-R: thrombopoietin receptor; BSG: basigin; EMMPRIN: extracellular matrix metalloproteinase inducer; TGF-β: transforming growth factor-beta; SLAMF-5: Signalling lymphocytic activation molecule 5; LAP: LC3-associated phagocytosis; GARP:glycoprotein A repetitions predominant.

Although there is difficulties in identification of distinction between both two types of EVs (micro particles and exosomes) in cargo potency, immune heterogeneity and size averaging of EV populations allows identifying them using several methods for their isolation and quantification, including conventional and cryo-enhanced electron microscopy, immuno-gold labelling, laser-scanning confocal microscopy, nanoparticle tracking analysis and flow cytometry [25,26] However, lack of generally accepted methods as a standard for determination, isolation and quantification of platelet-derived EVs, sufficiently limits our efforts in understanding their biological role in the pathogenesis of several states and diseases [27,28].

Platelet-derived vesicles in CV disease

There is large body of evidence regarding pivotal role of platelet-derived vesicles in the pathogenesis of CV diseases [29-31]. In a result of several triggers, such as ischemia, hypoxia, anemia, inflammation, shear stress, blood turbulence, platelets are activated, aggregated, then they changed their contain enriching phospholipids and GPs, prepare for vesiculation via shaping spaced architectonic and exocytosis and release vesicles into circulation. Regulation platelet vesiculation is mediated via platelet Ca(2+)-ATPases by cyclic AMP through the phosphorylation of the Rap1 protein.

Platelet-derived vesicles exhibit haemostatic properties, mediated aggregation of blood cells including whole platelets, promote coagulation, stabilize the vasculature and maintain endothelial cell barrier integrity [32]. Therefore, platelet-derived vesicles may demonstrate pro-atherogenic, pro-inflammatory, and immunomodulatory, even anticoagulant activity depending on triggers that induce their releasing and the expression of their surface certain molecules [33-35]. For instance, phospholipid-enriched platelet-derived vesicles that express Annexin V possess as pro-atherogenic and pro-coagulant particles [36]. On contrary Annexin V-negative vesicles were found rather anticoagulant than pro-coagulant particles. Therefore, platelets contain S100A8/A9 in membrane-enclosed vesicles and transfer it to target cells including endothelial cells and their precursors [37]. Probably S100A8/A9-rich platelet-derived vesicles can play a pivotal role in developing endothelial dysfunction upon manifestation of CV disease. Additionally, platelet-derived vesicles deliver oxidized lipids, P-selectin / P-selectin glycoprotein ligand 1 (PSGL-1), CD40 ligand (CD154) and induce B-cell synthesis and secretion of antigen-specific immunoglobulin G and potentiate the adaptive immune response through CD4+ cells [38-40] Although restored endothelial cell junctions and vascular integrity after thrombin releasing, the balance between promoting coagulation and mitigating endothelial dysfunction and vascular permeability has being turn to the side of potentiation of platelet activation status and pro-coagulant activity [41]. This effect appears to be harmful for majority patients with established CV diseases including asymptomatic atherosclerosis, acute coronary syndrome / myocardial infarction.

Platelet-derived vesicles in acute coronary syndrome and acute myocardial infarction

Acute coronary syndrome (ACS) and acute myocardial infarction (AMI) have been shown to be associated with an activation of various circulating cells, including platelets. Platelets are able to aggregate each other and with classical monocytes/macrophages shaping thrombus, embedding onto sub-cap space in a plaque, destroy a shoulder zone of the plaque and occlude the artery [42]. Being higher than in healthy individuals, aggregated platelets seem to consist of several cells (monocytes, macrophages, endothelial cells, and erythrocytes) and vesicles of various origin, such as phospholipid-rich platelet-derived extracellular vesicles expressed P-selectin (CD63), CD31, CD41a, and tissue factor (TF, CD142), elevated levels of which in peripheral blood strongly correspond to infarct size and in-hospital complications in AMI including mortality [43-45]. Moreover, ST-elevation AMI patients had higher levels of platelet-derived vesicles with immune phenotypes (CD61+/AV+, CD31+/AV+, CD42b+/AV+ and CD31+/CD42b+/AV+) compared to non-ST elevation AMI (STEMI) patients [46]. Yet, the investigator found that STEMI patients with NYHA class III heart failure (HF) had higher levels of circulating CD142+/AV+, CD14+/AV+ and CD14+/CD142+/AV+ endothelial cell-derived EVs as well as CD61+ platelet-derived vesicles than those in class I/II HF [46]. Thus, beyond biomarkers of cell activation, platelet-derived vesicles have numerous functional effects on the development of damaged vessel wall-induced arterial thrombi and blood thrombogenicity on areas of arterial damage contributing to atherothrombotic events. Indeed, phospholipids-rich platelets-derived vesicles have found the increased level of platelet-derived vesicles in survivors of AMI correlated well with thrombosis and soluble CD40 ligand (CD154) [47]. Authors concluded that the independent association between large platelet-derived vesicles and thrombin generation supports the concept that the formation of platelet-derived vesicles is important for increased coagulation activation in AMI patients [47]. Interestingly, the platelet-derived vesicles are discussed not the only factor directly mediating endothelial dysfunction and atherosclerosis, but they contribute to vascular reparation acting as factors of ischemic preconditioning on myocardial ischemia/reperfusion and vascular injury in correspondence with endothelial precursors [48]. Other studies reported that size of AMI, lactate dehydrogenase activity, oxidative stress activity and the number of apoptotic cardiac myocytes were strongly and inversely associated with a number of platelet-derived vesicles in blood collected from the infarct-dependent artery [49, 50]. Moreover, platelet-derived vesicles turn to be able to reduce the activity of caspase 3 and the expression of endoplasmic reticulum stress markers in the myocardium [49]. Additionally, endothelial precursors consolidate their interaction with platelets under dynamic flow conditions through secretion of platelet-derived vesicles and thereby mediate their regenerative potential [51-53]. Previously it has been reported that platelet-derived vesicles may modulate biological functions of hematopoietic cells and that they play an important but as yet not fully understood role in intercellular cross-talk in hematopoiesis and regeneration [54]. There is evidence that the platelet-derived vesicles can augment re-endothelialization capacity of circulating angiogenic cells [55]. All these facts support the hypothesis that angiopoietic capability of platelet-derived vesicles can modulate cardiac protection in ACS/AMI [56, 57] Taken together, these findings open new insight on cardiac protection in ACS/AMI based on an attenuation of endoplasmic reticulum stress apoptosis and angiogenesis through platelet-derived vesicles-endothelial precursors cooperation.

Conclusion: Elevated levels of platelet-derived vesicles are found in a wide range of thrombophilia-related condition and CV disorders including ACS/AMI. Platelet-derived vesicles are biomarker of endothelial dysfunction and pro-coagulative activity, which can be discussed a predictor of poor outcomes in ACS/AMI, while the diagnostic and predictive roles of this biomarker require to be investigated in large clinical trials in the future.

Financial support and sponsorship

None

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

References

  1. Shanmuganathan M, Vughs J, Noseda M, Emanueli C (2018) Exosomes: Basic Biology and Technological Advancements Suggesting Their Potential as Ischemic Heart Disease Therapeutics. Front Physiol 9: 1159.
  2. Berezin A, Zulli A, Kerrigan S, Petrovic D, Kruzliak P (2015) Predictive role of circulating endothelial-derived microparticles in cardiovascular diseases. Clin Biochem 48: 562-568.
  3. Nawaz M, Shah N, Zanetti BR, Maugeri M, Silvestre RN, et al. (2018) Extracellular Vesicles and Matrix Remodeling Enzymes: The Emerging Roles in Extracellular Matrix Remodeling, Progression of Diseases and Tissue Repair. Cells 7: E167.
  4. La Marca V, Fierabracci A. Insights into the Diagnostic Potential of Extracellular Vesicles and Their miRNA Signature from Liquid Biopsy as Early Biomarkers of Diabetic Micro/Macrovascular Complications. Int J Mol Sci 18: E1974.
  5. Berezin AE, Kremzer AA, Berezina TA, Martovitskaya YV (2015) Pattern of circulating microparticles in chronic heart failure patients with metabolic syndrome: Relevance to neurohumoral and inflammatory activation. BBA Clin 4: 69-75.
  6. Caporali A, Martello A, Miscianinov V, Maselli D, Vono R, et al. (2017) Contribution of pericyte paracrine regulation of the endothelium to angiogenesis. Pharmacol Ther 171: 56-64.
  7. De Toro J, Herschlik L, Waldner C, Mongini C (2015) Emerging roles of exosomes in normal and pathological conditions: new insights for diagnosis and therapeutic applications. Front Immunol 6: 203.
  8. Berezin AE (2017) Microparticles in Chronic Heart Failure. Adv Clin Chem 81: 1-41. [Crossref]
  9. Berezin AE, Kremzer AA, Cammarota G, Zulli A, Petrovic D, et al. (2016) Circulating endothelial-derived apoptotic microparticles and insulin resistance in non-diabetic patients with chronic heart failure. Clin Chem Lab Med 54: 1259-1267.
  10. Chen BY, Sung CW, Chen C, Cheng CM, Lin DP, et al. (2019) Advances in exosomes technology. Clin Chim Acta 493: 14-19. [Crossref]
  11. Juan T, Fürthauer M (2018) Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol 74: 66-77.
  12. Hervera A, Santos CX, De Virgiliis F, Shah AM, Di Giovanni S (2019) Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration. Trends Cell Biol.
  13. Mobarak H, Heidarpour M, Lolicato F, Nouri M, Rahbarghazi R, et al. (2019) Physiological impact of extracellular vesicles on female reproductive system; highlights to possible restorative effects on female age-related fertility. Biofactors 45: 293–303.
  14. Navarro A, Molins L, Marrades RM, Moises J, Viñolas N, et al. (2019) Exosome Analysis in Tumor-Draining Pulmonary Vein Identifies NSCLC Patients with Higher Risk of Relapse after Curative Surgery. Cancers (Basel) p. 2.
  15. Aslan C, Maralbashi S, Salari F, Kahroba H, Sigaroodi F, et al. (2019) Tumor-derived exosomes: Implication in angiogenesis and antiangiogenesis cancer therapy. J Cell Physiol.
  16. Yang L, Zhu J, Zhang C, Wang J, Yue F, et al. (2019) Stem cell-derived extracellular vesicles for myocardial infarction: a meta-analysis of controlled animal studies. Aging Albany NY.
  17. Bei Y, Das S, Rodosthenous RS, Holvoet P, Vanhaverbeke M, et al. (2017) Extracellular Vesicles in Cardiovascular Theranostics. Theranostics 7: 4168-4182.
  18. Kenari AN, Kastaniegaard K, Greening DW, Shambrook M, Stensballe A, et al. (2019) Exosome-mimetic nanovesicles contain distinct proteome and post-translational modified protein cargo, in comparison to exosomes. Proteomics p. e1800161.
  19. Vagner T, Chin A, Mariscal J, Bannykh S, Engman D, et al. (2019) Protein composition reflects extracellular vesicle heterogeneity. Proteomics p. e1800167.
  20. Aatonen M, Valkonen S, Böing A, Yuana Y, Nieuwland R, et al. (2017) Isolation of Platelet-Derived Extracellular Vesicles. Methods Mol Biol 1545: 177-188.
  21. Hedley BD, Llewellyn-Smith N, Lang S, Hsia CC, MacNamara N, et al. Combined accurate platelet enumeration and reticulated platelet determination by flow cytometry. Cytometry B Clin Cytom 88:330–337.
  22. Bennett JS (1990) The molecular biology of platelet membrane proteins. Semin Hematol 27: 186–204.
  23. Clemetson KJ, Clemetson JM (2008) Platelet GPIb complex as a target for anti-thrombotic drug development. Thromb Haemost 99: 473-479. [Crossref]
  24. Chevillet JR, Kang Q, Ruf IK, Briggs HA, Vojtech LN, et al. (2014) Quantitative and stoichiometric analysis of the microRNA content of exosomes. Proc Natl Acad Sci U S A 111: 14888-14893.
  25. van der Pol E, Boing AN, Harrison P, Sturk A, Nieuwland R (2012) Classification, Functions, and Clinical Relevance of Extracellular Vesicles. Pharmacological Reviews 64: 676–705.
  26. Liu C, Zhao J, Tian F, Chang J, Zhang W, et al. (2019) DNA and Aptamer Mediated Sorting and Analysis of Extracellular Vesicles. J Am Chem Soc 141: 3817-3821.
  27. Nolan JP, Jones JC (2017) Detection of platelet vesicles by flow cytometry. Platelets 28: 256-262. [Crossref]
  28. Lacroix R, Robert S, Poncelet P, Kasthuri R, Key N, et al. (2010) Standardization of platelet-derived microparticle enumeration by flow cytometry with calibrated beads: results of the International Society on Thrombosis and Haemostasis SSC Collaborative workshop. Journal of Thrombosis and Haemostasis 8: 2571–2574.
  29. Tao SC, Guo SC, Zhang CQ. Platelet-derived Extracellular Vesicles: An Emerging Therapeutic Approach. Int J Biol Sci 13: 828-834.
  30. Zaldivia MTK, Hering D, Marusic P, Sata Y, Lee R, et al. (2019) Successful Renal Denervation Decreases the Platelet Activation Status in Hypertensive Patients. Cardiovasc Res.
  31. Zaldivia MT, Rivera J, Hering D, Marusic P, Sata Y, et al. (2017) Renal Denervation Reduces Monocyte Activation and Monocyte-Platelet Aggregate Formation: An Anti-Inflammatory Effect Relevant for Cardiovascular Risk. Hypertension 69: 323-331.
  32. Barnes JN, Harvey RE, Miller KB, Jayachandran M, Malterer KR. et al. (2018) Cerebrovascular Reactivity and Vascular Activation in Postmenopausal Women With Histories of Preeclampsia. Hypertension 71: 110-117.
  33. VanWijk MJ, VanBavel E, Sturk A, Nieuwland R (2003) Microparticles in cardiovascular diseases. Cardiovasc Res 59: 277-287. [Crossref]
  34. Yano Y, Kambayashi J, Shiba E, Sakon M, Oiki E. et al. (1994) The role of protein phosphorylation and cytoskeletal reorganization in microparticle formation from the platelet plasma membrane. Biochem J 299: 303–308.
  35. Gemmel CH, Sefton MV, Yeo E (1993) Platelet-derived microparticle formation involves glycoprotein IIb-IIIa. Inhibition by RGDS and a Glanzmann’s thrombasthenia defect. J Biol Chem 268:14586-14589.
  36. Cauwenberghs S, Feijge MA, Harper AG, Sage SO, Curvers J, et al. (2006) Shedding of procoagulant microparticles from unstimulated platelets by integrin-mediated destabilization of actin cytoskeleton. FEBS Lett 580: 5313–5320.
  37. Nomura S, Komiyama Y, Miyake T, Miyazaki Y, Kido H. et al. (1994) Amyloid-protein precursor-rich platelet microparticles in thrombotic disease. Thromb Haemost 72: 519-522.
  38. Furie B, Furie BC (2004) Role of platelet P-selectin and microparticle PSGL-1 in thrombus formation. Trends Mol Med 10: 171-178. [Crossref]
  39. Barry OP, Practico D, Lawson JA, FitzGerald GA (1997) Transcellular activation of platelets and endothelial cells by bioactive lipids in platelet microparticles. J Clin Invest 99: 2118–2127.
  40.  Garcia BA, Smalley DM, Cho H, Shabanowitz J, Ley K, et al. (2005) The platelet microparticle proteome. J Proteome Res 4: 1516-1521. [Crossref]
  41. Sprague DL, Elzey BD, Crist SA, Waldschmidt TJ, Jensen RJ, et al. (2008) Platelet-mediated modulation of adaptive immunity: unique delivery of CD154 signal by platelet-derived membrane vesicles. Blood 111: 5028–5036.
  42. van der Zee PM, Biró E, Ko Y, de Winter RJ, Hack CE, et al. (2006) P-selectin- and CD63-exposing platelet microparticles reflect platelet activation in peripheral arterial disease and myocardial infarction. Clin Chem 52: 657-664.
  43. Loguinova M, Pinegina N, Kogan V, Vagida M, Arakelyan A, et al. (2018) Monocytes of Different Subsets in Complexes with Platelets in Patients with Myocardial Infarction. Thromb Haemost 118: 1969-1981. 
  44. Min PK, Kim JY, Chung KH, Lee BK, Cho M, et al. Local increase in microparticles from the aspirate of culprit coronary arteries in patients with ST-segment elevation myocardial infarction. Atherosclerosis 227: 323-328.
  45. Vagida MS, Arakelyan A, Lebedeva AM, Grivel JC, Shpektor AV, et al. (2016) Analysis of Extracellular Vesicles Using Magnetic Nanoparticles in Blood of Patients with Acute Coronary Syndrome. Biochemistry (Mosc) 81: 382-391.
  46. Jung C, Sörensson P, Saleh N, Arheden H, Rydén L, et al. Circulating endothelial and platelet derived microparticles reflect the size of myocardium at risk in patients with ST-elevation myocardial infarction. Atherosclerosis 221: 226-231.
  47. Chiva-Blanch G, Laake K, Myhre P, Bratseth V, Arnesen H, et al. (2017) Platelet-, monocyte-derived and tissue factor-carrying circulating microparticles are related to acute myocardial infarction severity. PLoS One 12: e0172558.
  48. Suades R, Padró T, Vilahur G, Badimon L (2012) Circulating and platelet-derived microparticles in human blood enhance thrombosis on atherosclerotic plaques. Thromb Haemost 108: 1208-1219.
  49. Michelsen AE, Brodin E, Brosstad F, Hansen JB (2008) Increased level of platelet microparticles in survivors of myocardial infarction. Scand J Clin Lab Invest 68: 386-389.
  50. Liu M, Wang Y, Zhu Q, Zhao J, Wang Y, et al. (2018) Protective effects of circulating microvesicles derived from ischemic preconditioning on myocardial ischemia/reperfusion injury in rats by inhibiting endoplasmic reticulum stress. Apoptosis 23: 436-448.
  51. Porto I, Biasucci LM, De Maria GL, Leone AM, Niccoli G, et al. (2012) Intracoronary microparticles and microvascular obstruction in patients with ST elevation myocardial infarction undergoing primary percutaneous intervention. Eur Heart J 33: 2928-2938.
  52. Alexandru N, Andrei E, Dragan E, Georgescu A (2015) Interaction of platelets with endothelial progenitor cells in experimental atherosclerosis: Role of transplanted endothelial progenitor cells and platelet microparticles. Biol Cell 107: 189-204.
  53. Berezin AE. Are Endothelial Cell-Derived Microparticles Predictive Biomarkers in Cardiovascular Diseases? Atherosclerosis: open Access 1: e101-e103.
  54. Mause SF, Ritzel E, Liehn EA, Hristov M, Bidzhekov K, et al. (2010) Platelet microparticles enhance the vasoregenerative potential of angiogenic early outgrowth cells after vascular injury. Circulation 122: 495-506.
  55. Baj-Krzyworzeka M, Majka M, Pratico D, Ratajczak J, Vilaire G, et al. (2002) Platelet-derived microparticles stimulate proliferation, survival, adhesion, and chemotaxis of hematopoietic cells. Exp Hematol 30: 450-459.
  56. Ohtsuka M, Sasaki K, Ueno T, Seki R, Nakayoshi T, et al. (2013) Platelet-derived microparticles augment the adhesion and neovascularization capacities of circulating angiogenic cells obtained from atherosclerotic patients. Atherosclerosis 227: 275-282.
  57. Chen BA, Zhong YJ, Huang CY, Li CP, Shi GY, et al. (2007) Effects of platelet-derived membrane microparticles on angiogenesis in chick chorioallantoic membranes. 15: 1070-1073.

Editorial Information

Editor-in-Chief

Article Type

Review Article

Publication history

Received date: April 30, 2019
Accepted date: May 23, 2019
Published date: May 27, 2019

Copyright

©2019 Berezin AE (2019) This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Citation

Berezin AE, Berezin AA (2019) Platelet-derived vesicles in acute myocardial infarction. Clin Res Trials 5: DOI: 10.15761/CRT.1000262

Corresponding author

Alexander E Berezin

Senior Consultant of Therapeutic Unit, Internal Medicine Department, State Medical University of Zaporozhye, 26, Mayakovsky av., Zaporozhye, Ukraine

E-mail : bhuvaneswari.bibleraaj@uhsm.nhs.uk

Table 1. Labelling of platelet-derived EVs with surface CD markers

CD marker

Alternative name

Prominent feature

Cell labelling

Expression

Significance

CD9

Tetraspanin (Tspan 29)

Key regulator of intracellular signalling, proliferation, activation, survival, migration, invasion, cell adhesion, and diapedesis

Platelets, B cells, CD4+ T cells, CD8+ T cells, natural killer cells, granulocytes, monocytes and macrophages, and immature and mature dendritic cells

Low

None-specific lympho / hematopoietic marker

CD23

Fc epsilon RII (FcεRII)

Interaction with many ligands (IgE, CD21, MHC class II,integrins), exhibiting pleiotropic cytokine-like activities, sustaining the growth of activated B-cells, differentiation of myeloid precursors, thymocytes and bone marrow CD4+ T cells, degranulation of platelets

Platelets, T and B lymphocytes, polymorphonuclear leucocytes, monocytes, follicular dendritic cells, intestinal epithelial cells, bone marrow stromal cells

Low

None-specific lymphohematopoietic marker, diagnostic marker for neoplastic and autoimmune inflammatory disease

CD31

PECAM-1, GP IIa, hec7 antigen, EndoCAM

Key inhibitory receptor in circulating platelets and leukocytes, major endothelial mechanosensor, modulator of vascular permeability, and leukocyte trafficking, and cell-to-cell cooperation, core player in in thrombosis, inflammation and the immune response

Platelets, leucocytes, monocytes / mononuclears, endothelial cells, EPCs

High

A maker of endothelial integrity, vascular permeability, platelet degranulation, endothelialopathies in tumor angiogenesis and the growth and development of hemangiomas

CD36

GP-IV, Scarb3

Recognition of thrombospondine, fatty acids, collagen, adhesion of platelets

Monocytes, macrophages, platelets, adipose cells, endothelial cells, PECs, epithelial cells

High

Platelet collagen receptor

CD41

αIIb integrin, platelet GPIIb

Key regulator of growth and differentiation

Hematopoetic progenitors in embrio, specific to the megakaryocytic / platelet lineage

Moderate

Markers of all progenitors (megakaryocytic, granulomacrophagic, erythroid and pluripotent cells)

CD42 (a, b, c)

Integrin subunit αIIb, GPIIb

Regulator of platelet activation and adhesion

Platelets

High

Marker of platelet aggregation

CD49f

GPIc, integrin α6-chain, ITGA6, ITGA6B, VLA 6 alpha Subunit

Regulator of embryogenesis, cell adhesion and cell migration through T-cell receptor/CD3 complex

T cells, endothelial cells, epithelial cell, macrophages, monocytes; platelets and stem cell

High

Marker of blood cell adhesion and T-cell proliferation

CD51

Integrin subunit alpha V, vitronectin receptor

Trigger of cell adhesion and signal transduction, regulator of angiogenesis and cancer progression

T cells, endothelial cells, platelets

High

Marker of cell adhesion to sequences of von Willebrand factor, vitronectin, fibrinogen and thrombospondin

CD61

GP3A, integrin subunit beta 3

Regulator of cell adhesion and cell-surface mediated signalling

Endothelial cells, Macrophage, platelets

High

Marker of human megakaryocytic cell lineages

CD84

Cell surface antigen MAX.3, Ly-9B, SLAMF-5

Regulator of signalling and adhesion in lymphocytic cells and other cells

Thymus and spleen cells, cells of megakaryocytic lineages

Moderate

Marker of adhesion interactions between T lymphocytes and accessory cells, platelets

CD92

Choline Transporter like Protein 1

Regulator of cell growth and differentiation

Subset of bone marrow stromal cells, cells of osteogenic, megakaryocytic and adipogenic lineages

High

Marker of human bone marrow stromal cells, and cells of megakaryocytic and adipogenic lineages

CD110

MPL proto-oncogene, TPO-R,

Regulator of heamopoesis

Subset of stem cells and cells of megakaryocytic lineage as well as platelets

High

Marker of megakaryocytic lineage

CD147

BSG, EMMPRIN

Trigger of cell proliferation associated with the TGF-β/Smad4 signalling pathway, cell adhesion and T-cell activation

Endothelial cell, epithelial cells, lymphocytes, mononuclears, platelets

Low

Marker of cell growth, adhesion, and proliferation, malignancy, angiogenesis, vascular permeability

CD151

GP27, membrane glycoprotein SFA-1, platelet-endothelia-l tetraspan antigen-3

Regulator of the VCAM-1 activity during lymphocyte and platelet recruitment

Endothelial cells, platelets, T-cells

High

Marker of fibrosis, vascular inflammation and remodelling, cancinogenesis, vascular permeability, endothelial dysfunction

CD226

DNAM1, PTA1, TLiSA1, DNAX

An adhesion molecule involved in NK and T cell-mediated cytotoxicity

T cells, NK cells, platelets, mononuclears

Low

Triggering cytotoxicity and cytokine secretion by T and NK cells

GARP

-

A key regulator of network between Tregs and its targets

Activated Tregs, mature peripheral naive CD4+ T cells, platelets

Low

Marker of activated Tregs and degradated platelets

LAP

-

Phagosome maturation, stabilization of the cargo to prolong antigen presentation on MHC, recruiting LC3 molecules to phagosome membranes class II molecules

Macrophages, dendritic cells and platelets

Low

A marker of catabolic process involved during exogenous antigen processing, recognition of apoptotic, necrotic, or entotic cells, degradation of platelets

Abbreviation: MHC: major histocompatibility complex; GP: glycoprotein; PECAM-1: platelet/endothelial cell adhesion molecule 1; EPCs: endothelial progenitor cells; NK: nature killer; TPO-R: thrombopoietin receptor; BSG: basigin; EMMPRIN: extracellular matrix metalloproteinase inducer; TGF-β: transforming growth factor-beta; SLAMF-5: Signalling lymphocytic activation molecule 5; LAP: LC3-associated phagocytosis; GARP:glycoprotein A repetitions predominant.