CC BY 4.0 · TH Open 2021; 05(04): e533-e542
DOI: 10.1055/a-1682-3415
Original Article

Combined Antiplatelet Therapy Reduces the Proinflammatory Properties of Activated Platelets

Alexandra C.A. Heinzmann
1   Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
,
Daniëlle M. Coenen
1   Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
2   Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, Kentucky, United States
,
Tanja Vajen
1   Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
3   Cardiovascular Research Laboratory, Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
,
1   Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
,
1   Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
› Author Affiliations
Funding This work was supported by the Netherlands Foundation for Scientific Research (ZonMW VIDI 016.126.358), the Landsteiner Foundation for Blood Transfusion Research (LSBR Nr. 1638) awarded to R.R.K., and by the Netherlands Organization for Scientific Research (NWO) (Vidi 91716421) and the Dutch Heart Foundation (2015T79) to D.M.C. and J.M.E.M.C.

Abstract

The cause of atherothrombosis is rupture or erosion of atherosclerotic lesions, leading to an increased risk of myocardial infarction or stroke. Here, platelet activation plays a major role, leading to the release of bioactive molecules, for example, chemokines and coagulation factors, and to platelet clot formation. Several antiplatelet therapies have been developed for secondary prevention of cardiovascular events, in which anticoagulant drugs are often combined. Besides playing a role in hemostasis, platelets are also involved in inflammation. However, it is unclear whether current antiplatelet therapies also affect platelet immune functions. In this study, the possible anti-inflammatory effects of antiplatelet medications on chemokine release were investigated using enzyme-linked immunosorbent assay and on the chemotaxis of THP-1 cells toward platelet releasates. We found that antiplatelet medication acetylsalicylic acid (ASA) led to reduced chemokine (CC motif) ligand 5 (CCL5) and chemokine (CXC motif) ligand 4 (CXCL4) release from platelets, while leukocyte chemotaxis was not affected. Depending on the agonist, αIIbβ3 and P2Y12 inhibitors also affected CCL5 or CXCL4 release. The combination of ASA with a P2Y12 inhibitor or a phosphodiesterase (PDE) inhibitor did not lead to an additive reduction in CCL5 or CXCL4 release. Interestingly, these combinations did reduce leukocyte chemotaxis. This study provides evidence that combined therapy of ASA and a P2Y12 or PDE3 inhibitor can decrease the inflammatory leukocyte recruiting potential of the releasate of activated platelets.

Key Findings

1. Combined therapy of ASA and αP2Y12 or PDE3 inhibitor decreases platelets' proinflammatory potential of leukocyte recruitment, which appeared to depend on platelet-derived CCL5.


2. Secretion triggered via PAR1/PAR4 is most affected by antiplatelet medications.


3. As single therapy, cangrelor and ASA are comparable in reducing proinflammatory chemokine release by platelets.


Author Contributions

A.C.A.H performed experiments, analyzed data, and drafted the manuscript; T.V and D.M.C performed experiments; J.M.E.M.C provided critical reagents and intellectual input; and R.R.K. supervised this study, obtained funding, and finalized the manuscript.


Supplementary Material



Publication History

Received: 18 June 2021

Accepted: 22 October 2020

Accepted Manuscript online:
28 October 2021

Article published online:
07 December 2021

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Herrington W, Lacey B, Sherliker P, Armitage J, Lewington S. Epidemiology of atherosclerosis and the potential to reduce the global burden of atherothrombotic disease. Circ Res 2016; 118 (04) 535-546
  • 2 Jackson SP. Arterial thrombosis–insidious, unpredictable and deadly. Nat Med 2011; 17 (11) 1423-1436
  • 3 Viles-Gonzalez JF, Fuster V, Badimon JJ. Atherothrombosis: a widespread disease with unpredictable and life-threatening consequences. Eur Heart J 2004; 25 (14) 1197-1207
  • 4 Smyth SS, McEver RP, Weyrich AS. et al; 2009 Platelet Colloquium Participants. Platelet functions beyond hemostasis. J Thromb Haemost 2009; 7 (11) 1759-1766
  • 5 da Costa Martins P, van den Berk N, Ulfman LH, Koenderman L, Hordijk PL, Zwaginga JJ. Platelet-monocyte complexes support monocyte adhesion to endothelium by enhancing secondary tethering and cluster formation. Arterioscler Thromb Vasc Biol 2004; 24 (01) 193-199
  • 6 Kuckleburg CJ, Yates CM, Kalia N. et al. Endothelial cell-borne platelet bridges selectively recruit monocytes in human and mouse models of vascular inflammation. Cardiovasc Res 2011; 91 (01) 134-141
  • 7 Schrottmaier WC, Mussbacher M, Salzmann M, Assinger A. Platelet-leukocyte interplay during vascular disease. Atherosclerosis 2020; 307: 109-120
  • 8 Vajen T, Mause SF, Koenen RR. Microvesicles from platelets: novel drivers of vascular inflammation. Thromb Haemost 2015; 114 (02) 228-236
  • 9 Konkoth A, Saraswat R, Dubrou C. et al. Multifaceted role of extracellular vesicles in atherosclerosis. Atherosclerosis 2021; 319: 121-131
  • 10 Maugeri N, Campana L, Gavina M. et al. Activated platelets present high mobility group box 1 to neutrophils, inducing autophagy and promoting the extrusion of neutrophil extracellular traps. J Thromb Haemost 2014; 12 (12) 2074-2088
  • 11 Vajen T, Koenen RR, Werner I. et al. Blocking CCL5-CXCL4 heteromerization preserves heart function after myocardial infarction by attenuating leukocyte recruitment and NETosis. Sci Rep 2018; 8 (01) 10647 DOI: 10.1038/s41598-018-29026-0.
  • 12 Gleissner CA. Macrophage phenotype modulation by CXCL4 in atherosclerosis. Front Physiol 2012; 3: 1 DOI: 10.3389/fphys.2012.00001.
  • 13 von Hundelshausen P, Schmitt MM. Platelets and their chemokines in atherosclerosis-clinical applications. Front Physiol 2014; 5: 294 DOI: 10.3389/fphys.2014.00294.
  • 14 Fernandez EJ, Lolis E. Structure, function, and inhibition of chemokines. Annu Rev Pharmacol Toxicol 2002; 42: 469-499
  • 15 Burkhart JM, Vaudel M, Gambaryan S. et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 2012; 120 (15) e73-e82
  • 16 Blanchet X, Cesarek K, Brandt J. et al. Inflammatory role and prognostic value of platelet chemokines in acute coronary syndrome. Thromb Haemost 2014; 112 (06) 1277-1287
  • 17 von Hundelshausen P, Koenen RR, Sack M. et al. Heterophilic interactions of platelet factor 4 and RANTES promote monocyte arrest on endothelium. Blood 2005; 105 (03) 924-930
  • 18 Koenen RR, von Hundelshausen P, Nesmelova IV. et al. Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice. Nat Med 2009; 15 (01) 97-103
  • 19 Coenen DM, Heinzmann ACA, Karel MFA, Cosemans JMEM, Koenen RR. The multifaceted contribution of platelets in the emergence and aftermath of acute cardiovascular events. Atherosclerosis 2021; 319: 132-141
  • 20 Gurbel PA, Fox KAA, Tantry US, Ten Cate H, Weitz JI. Combination antiplatelet and oral anticoagulant therapy in patients with coronary and peripheral artery disease. Circulation 2019; 139 (18) 2170-2185
  • 21 Piepoli MF, Hoes AW, Agewall S. et al; ESC Scientific Document Group. 2016 European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts) Developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR). Eur Heart J 2016; 37 (29) 2315-2381
  • 22 Tsigkou V, Siasos G, Rovos K, Tripyla N, Tousoulis D. Peripheral artery disease and antiplatelet treatment. Curr Opin Pharmacol 2018; 39: 43-52
  • 23 Sibbing D, Byrne RA, Bernlochner I, Kastrati A. High platelet reactivity and clinical outcome - fact and fiction. Thromb Haemost 2011; 106 (02) 191-202
  • 24 Müller KA, Chatterjee M, Rath D, Geisler T. Platelets, inflammation and anti-inflammatory effects of antiplatelet drugs in ACS and CAD. Thromb Haemost 2015; 114 (03) 498-518
  • 25 Denisov SS, Ramírez-Escudero M, Heinzmann ACA. et al. Structural characterization of anti-CCL5 activity of the tick salivary protein evasin-4. J Biol Chem 2020; 295 (42) 14367-14378
  • 26 Rosado JA, Meijer EM, Hamulyak K, Novakova I, Heemskerk JW, Sage SO. Fibrinogen binding to the integrin alpha(IIb)beta(3) modulates store-mediated calcium entry in human platelets. Blood 2001; 97 (09) 2648-2656
  • 27 Parker WAE, Storey RF. Novel approaches to P2Y12 inhibition and aspirin dosing. Platelets 2020; 32 (01) 7-14
  • 28 Floyd CN. Dual Antiplatelet therapy in coronary artery disease: comparison between ACC/AHA 2016 and ESC 2017 guidelines. Eur Cardiol 2020; 15: 1-3
  • 29 Coenen DM, Heinzmann ACA, Oggero S. et al. Inhibition of phosphodiesterase 3A by cilostazol dampens proinflammatory platelet functions. Cells 2021; 10 (08) 1998 DOI: 10.3390/cells10081998.
  • 30 Mauler M, Herr N, Schoenichen C. et al. Platelet serotonin aggravates myocardial ischemia/reperfusion injury via neutrophil degranulation. Circulation 2019; 139 (07) 918-931
  • 31 Cloutier N, Allaeys I, Marcoux G. et al. Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration. Proc Natl Acad Sci U S A 2018; 115 (07) E1550-E1559
  • 32 Konopatskaya O, Gilio K, Harper MT. et al. PKC alpha regulates platelet granule secretion and thrombus formation in mice. J Clin Invest 2009; 119 (02) 399-407
  • 33 van Holten TC, Bleijerveld OB, Wijten P. et al. Quantitative proteomics analysis reveals similar release profiles following specific PAR-1 or PAR-4 stimulation of platelets. Cardiovasc Res 2014; 103 (01) 140-146
  • 34 Coller BS. αIIbβ3: structure and function. J Thromb Haemost 2015; 13 (Suppl 1): S17-S25
  • 35 Lambrechts K, de Maistre S, Abraini JH, Blatteau JE, Risso JJ, Vallée N. Tirofiban, a glycoprotein IIb/IIIa antagonist, has a protective effect on decompression sickness in rats: is the crosstalk between platelet and leukocytes essential?. Front Physiol 2018; 9: 906 DOI: 10.3389/fphys.2018.00906.
  • 36 Kupatt C, Habazettl H, Hanusch P. et al. c7E3Fab reduces postischemic leukocyte-thrombocyte interaction mediated by fibrinogen. Implications for myocardial reperfusion injury. Arterioscler Thromb Vasc Biol 2000; 20 (10) 2226-2232
  • 37 Massberg S, Brand K, Grüner S. et al. A critical role of platelet adhesion in the initiation of atherosclerotic lesion formation. J Exp Med 2002; 196 (07) 887-896
  • 38 May AE, Kälsch T, Massberg S, Herouy Y, Schmidt R, Gawaz M. Engagement of glycoprotein IIb/IIIa (alpha(IIb)beta3) on platelets upregulates CD40L and triggers CD40L-dependent matrix degradation by endothelial cells. Circulation 2002; 106 (16) 2111-2117
  • 39 May AE, Neumann FJ, Gawaz M, Ott I, Walter H, Schömig A. Reduction of monocyte-platelet interaction and monocyte activation in patients receiving antiplatelet therapy after coronary stent implantation. Eur Heart J 1997; 18 (12) 1913-1920
  • 40 Neumann FJ, Zohlnhöfer D, Fakhoury L, Ott I, Gawaz M, Schömig A. Effect of glycoprotein IIb/IIIa receptor blockade on platelet-leukocyte interaction and surface expression of the leukocyte integrin Mac-1 in acute myocardial infarction. J Am Coll Cardiol 1999; 34 (05) 1420-1426
  • 41 Undas A, Brummel-Ziedins K, Mann KG. Why does aspirin decrease the risk of venous thromboembolism? On old and novel antithrombotic effects of acetyl salicylic acid. J Thromb Haemost 2014; 12 (11) 1776-1787
  • 42 Otto GP, Sossdorf M, Boettel J. et al. Effects of low-dose acetylsalicylic acid and atherosclerotic vascular diseases on the outcome in patients with severe sepsis or septic shock. Platelets 2013; 24 (06) 480-485
  • 43 Thomas MR, Outteridge SN, Ajjan RA. et al. Platelet P2Y12 inhibitors reduce systemic inflammation and its prothrombotic effects in an experimental human model. Arterioscler Thromb Vasc Biol 2015; 35 (12) 2562-2570
  • 44 Schäfer A, Fraccarollo D, Pförtsch S. et al. Clopidogrel improves endothelial function and NO bioavailability by sensitizing adenylyl cyclase in rats with congestive heart failure. Basic Res Cardiol 2011; 106 (03) 485-494
  • 45 Schrottmaier WC, Kral JB, Badrnya S, Assinger A. Aspirin and P2Y12 Inhibitors in platelet-mediated activation of neutrophils and monocytes. Thromb Haemost 2015; 114 (03) 478-489
  • 46 Thomas MR, Storey RF. Effect of P2Y12 inhibitors on inflammation and immunity. Thromb Haemost 2015; 114 (03) 490-497
  • 47 Coimbra LS, Steffens JP, Rossa Jr C, Graves DT, Spolidorio LC. Clopidogrel enhances periodontal repair in rats through decreased inflammation. J Clin Periodontol 2014; 41 (03) 295-302
  • 48 Harding SA, Sarma J, Din JN, Maciocia PM, Newby DE, Fox KA. Clopidogrel reduces platelet-leucocyte aggregation, monocyte activation and RANTES secretion in type 2 diabetes mellitus. Heart 2006; 92 (09) 1335-1337
  • 49 Meyer A, Weithaeuser A, Steffens D. et al. Inhibition of platelet function with clopidogrel is associated with a reduction of inflammation in patients with peripheral artery disease. Cardiovasc Revasc Med 2016; 17 (03) 169-175
  • 50 Siegel PM, Sander L, Fricke A. et al. P2Y12 receptor blockers are anti-inflammatory drugs inhibiting both circulating monocytes and macrophages including THP-1 cells. Sci Rep 2021; 11 (01) 17459 DOI: 10.1038/s41598-021-95710-3.
  • 51 Micklewright JJ, Layhadi JA, Fountain SJ. P2Y12 receptor modulation of ADP-evoked intracellular Ca2+ signalling in THP-1 human monocytic cells. Br J Pharmacol 2018; 175 (12) 2483-2491
  • 52 Kloss L, Dollt C, Schledzewski K. et al. ADP secreted by dying melanoma cells mediates chemotaxis and chemokine secretion of macrophages via the purinergic receptor P2Y12. Cell Death Dis 2019; 10 (10) 760 DOI: 10.1038/s41419-019-2010-6.
  • 53 Dickhout A, Tullemans BME, Heemskerk JWM, Thijssen VLJL, Kuijpers MJE, Koenen RR. Galectin-1 and platelet factor 4 (CXCL4) induce complementary platelet responses in vitro. PLoS One 2021; 16 (01) e0244736 DOI: 10.1371/journal.pone.0244736.
  • 54 Heinzmann ACA, Karel MFA, Coenen DM. et al. Complementary roles of platelet αIIbβ3 integrin, phosphatidylserine exposure and cytoskeletal rearrangement in the release of extracellular vesicles. Atherosclerosis 2020; 310: 17-25
  • 55 Chatterjee M, Huang Z, Zhang W. et al. Distinct platelet packaging, release, and surface expression of proangiogenic and antiangiogenic factors on different platelet stimuli. Blood 2011; 117 (14) 3907-3911
  • 56 Italiano Jr JE, Richardson JL, Patel-Hett S. et al. Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released. Blood 2008; 111 (03) 1227-1233
  • 57 Jonnalagadda D, Izu LT, Whiteheart SW. Platelet secretion is kinetically heterogeneous in an agonist-responsive manner. Blood 2012; 120 (26) 5209-5216
  • 58 Kamykowski J, Carlton P, Sehgal S, Storrie B. Quantitative immunofluorescence mapping reveals little functional coclustering of proteins within platelet α-granules. Blood 2011; 118 (05) 1370-1373
  • 59 Gresele P, Momi S, Falcinelli E. Anti-platelet therapy: phosphodiesterase inhibitors. Br J Clin Pharmacol 2011; 72 (04) 634-646
  • 60 Liu D, Richardson G, Benli FM. et al. Inflammageing in the cardiovascular system: mechanisms, emerging targets, and novel therapeutic strategies. Clin Sci (Lond) 2020; 134 (17) 2243-2262
  • 61 Jansen EE, Braun A, Jansen P, Hartmann M. Platelet-therapeutics to improve tissue regeneration and wound healing-physiological background and methods of preparation. Biomedicines 2021; 9 (08) 869 DOI: 10.3390/biomedicines9080869.