CC BY-NC-ND 4.0 · Thromb Haemost 2019; 119(08): 1295-1310
DOI: 10.1055/s-0039-1683409
Cellular Haemostasis and Platelets
Georg Thieme Verlag KG Stuttgart · New York

Lysophosphatidylcholine is a Major Component of Platelet Microvesicles Promoting Platelet Activation and Reporting Atherosclerotic Plaque Instability

Philipp Diehl
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center – University of Freiburg, University of Freiburg, Freiburg, Germany
,
Frederik Nienaber
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Maria T. K. Zaldivia
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Johannes Stamm
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Patrick M. Siegel
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
2   Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center – University of Freiburg, University of Freiburg, Freiburg, Germany
,
Natalie A. Mellett
3   Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Marius Wessinger
2   Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center – University of Freiburg, University of Freiburg, Freiburg, Germany
,
Xiaowei Wang
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
James D. McFadyen
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Nicole Bassler
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Gerhard Puetz
4   Institute for Clinical Chemistry and Laboratory Medicine, University of Freiburg, Freiburg, Germany
,
Nay M. Htun
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
David Braig
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
5   Department of Plastic and Hand Surgery, University of Freiburg, Freiburg, Germany
,
Jonathon Habersberger
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Thomas Helbing
2   Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center – University of Freiburg, University of Freiburg, Freiburg, Germany
,
Steffen U. Eisenhardt
5   Department of Plastic and Hand Surgery, University of Freiburg, Freiburg, Germany
,
Maria Fuller
6   Centre for Molecular Pathology, University of Adelaide, Adelaide, Australia
,
Christoph Bode
2   Department of Cardiology and Angiology I, Faculty of Medicine, Heart Center – University of Freiburg, University of Freiburg, Freiburg, Germany
,
Peter J. Meikle
3   Metabolomics Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Yung Chih Chen
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
,
Karlheinz Peter
1   Department of Atherothrombosis and Vascular Biology, Baker Heart and Diabetes Institute, Melbourne, Australia
› Institutsangaben
Funding P.D. was funded by scholarships from the German Research Foundation (DFG, DI 1623/1–1) and Monash University in Melbourne (Australia), J.D.M. and N.M.H. were funded by Royal Australasian College of Physicians research establishment fellowships, S.U.E. was funded by a professorial Heisenberg Fellowship from the German Research Foundation (DFG) and X.W. and Y.C.C. were funded by a fellowship from the Heart Foundation of Australia. P.J.M. and K.P. were funded by fellowships from the National Health and Medical Research Council (NHMRC) of Australia. The project was also supported in part by the Victorian Government's OIS Program.
Weitere Informationen

Publikationsverlauf

16. September 2018

25. Januar 2019

Publikationsdatum:
02. August 2019 (online)

Abstract

Background Microvesicles (MVs) are small cell-derived vesicles, which are mainly released by activated cells. They are part of a communication network delivering biomolecules, for example, inflammatory molecules, via the blood circulation to remote cells in the body. Platelet-derived MVs are known to induce vascular inflammation. Research on the mediators and mechanisms of their inflammatory effects has attracted major interest. We hypothesize that specific lipids are the mediators of vascular inflammation caused by platelet-derived MVs.

Methods and Results Liquid chromatography electrospray ionization–tandem mass spectrometry was used for lipid profiling of platelet-derived MVs. Lysophosphatidylcholine (LPC) was found to be a major component of platelet-derived MVs. Investigating the direct effects of LPC, we found that it induces platelet activation, spreading, migration and aggregation as well as formation of inflammatory platelet–monocyte aggregates. We show for the first time that platelets express the LPC receptor G2AR, which mediates LPC-induced platelet activation. In a mouse model of atherosclerotic plaque instability/rupture, circulating LPC was detected as a surrogate marker of plaque instability. These findings were confirmed by matrix-assisted laser desorption ionization imaging, which showed that the LPC concentration of human plaques was highest in vulnerable plaque regions.

Conclusion LPC is a major component of platelet-derived MVs and via its interaction with G2AR on platelets contributes to platelet activation, spreading, migration and aggregation and ultimately to vascular inflammation. Circulating LPC reports on atherosclerotic plaque instability in mice and is significantly increased in unstable areas of atherosclerotic plaques in both mice and humans, linking LPC to plaque instability.

Note

An abstract of this paper was presented at the ISTH Congress, July 6–10, 2019 in Melbourne, Australia.


Supplementary Material

 
  • References

  • 1 Yuana Y, Sturk A, Nieuwland R. Extracellular vesicles in physiological and pathological conditions. Blood Rev 2013; 27 (01) 31-39
  • 2 Zaldivia MTK, McFadyen JD, Lim B, Wang X, Peter K. Platelet-derived microvesicles in cardiovascular diseases. Front Cardiovasc Med 2017; 4: 74
  • 3 Ridger VC, Boulanger CM, Angelillo-Scherrer A. , et al; Position Paper of the European Society of Cardiology (ESC) Working Group on Atherosclerosis and Vascular Biology. Microvesicles in vascular homeostasis and diseases. Thromb Haemost 2017; 117 (07) 1296-1316
  • 4 Hugel B, Martínez MC, Kunzelmann C, Freyssinet JM. Membrane microparticles: two sides of the coin. Physiology (Bethesda) 2005; 20: 22-27
  • 5 Pluskota E, Woody NM, Szpak D. , et al. Expression, activation, and function of integrin alphaMbeta2 (Mac-1) on neutrophil-derived microparticles. Blood 2008; 112 (06) 2327-2335
  • 6 Diehl P, Fricke A, Sander L. , et al. Microparticles: major transport vehicles for distinct microRNAs in circulation. Cardiovasc Res 2012; 93 (04) 633-644
  • 7 Brown GT, McIntyre TM. Lipopolysaccharide signaling without a nucleus: kinase cascades stimulate platelet shedding of proinflammatory IL-1β-rich microparticles. J Immunol 2011; 186 (09) 5489-5496
  • 8 Mause SF, von Hundelshausen P, Zernecke A, Koenen RR, Weber C. Platelet microparticles: a transcellular delivery system for RANTES promoting monocyte recruitment on endothelium. Arterioscler Thromb Vasc Biol 2005; 25 (07) 1512-1518
  • 9 Bernal-Mizrachi L, Jy W, Jimenez JJ. , et al. High levels of circulating endothelial microparticles in patients with acute coronary syndromes. Am Heart J 2003; 145 (06) 962-970
  • 10 Bei Y, Das S, Rodosthenous RS. , et al. Extracellular vesicles in cardiovascular theranostics. Theranostics 2017; 7 (17) 4168-4182
  • 11 Vajen T, Benedikter BJ, Heinzmann ACA. , et al. Platelet extracellular vesicles induce a pro-inflammatory smooth muscle cell phenotype. J Extracell Vesicles 2017; 6 (01) 1322454
  • 12 Edelstein LC. The role of platelet microvesicles in intercellular communication. Platelets 2017; 28 (03) 222-227
  • 13 Vajen T, Mause SF, Koenen RR. Microvesicles from platelets: novel drivers of vascular inflammation. Thromb Haemost 2015; 114 (02) 228-236
  • 14 Burnier L, Fontana P, Kwak BR, Angelillo-Scherrer A. Cell-derived microparticles in haemostasis and vascular medicine. Thromb Haemost 2009; 101 (03) 439-451
  • 15 Mause SF, Weber C. Microparticles: protagonists of a novel communication network for intercellular information exchange. Circ Res 2010; 107 (09) 1047-1057
  • 16 Stegemann C, Pechlaner R, Willeit P. , et al. Lipidomics profiling and risk of cardiovascular disease in the prospective population-based Bruneck study. Circulation 2014; 129 (18) 1821-1831
  • 17 Braig D, Nero TL, Koch HG. , et al. Transitional changes in the CRP structure lead to the exposure of proinflammatory binding sites. Nat Commun 2017; 8: 14188
  • 18 Habersberger J, Strang F, Scheichl A. , et al. Circulating microparticles generate and transport monomeric C-reactive protein in patients with myocardial infarction. Cardiovasc Res 2012; 96 (01) 64-72
  • 19 Weir JM, Wong G, Barlow CK. , et al. Plasma lipid profiling in a large population-based cohort. J Lipid Res 2013; 54 (10) 2898-2908
  • 20 Elizondo E, Moreno E, Cabrera I. , et al. Liposomes and other vesicular systems: structural characteristics, methods of preparation, and use in nanomedicine. Prog Mol Biol Transl Sci 2011; 104: 1-52
  • 21 Greenberg SM, Rosenthal DS, Greeley TA, Tantravahi R, Handin RI. Characterization of a new megakaryocytic cell line: the Dami cell. Blood 1988; 72 (06) 1968-1977
  • 22 Schwarz M, Meade G, Stoll P. , et al. Conformation-specific blockade of the integrin GPIIb/IIIa: a novel antiplatelet strategy that selectively targets activated platelets. Circ Res 2006; 99 (01) 25-33
  • 23 Rikitake Y, Hirata K, Yamashita T. , et al. Expression of G2A, a receptor for lysophosphatidylcholine, by macrophages in murine, rabbit, and human atherosclerotic plaques. Arterioscler Thromb Vasc Biol 2002; 22 (12) 2049-2053
  • 24 Chen YC, Bui AV, Diesch J. , et al. A novel mouse model of atherosclerotic plaque instability for drug testing and mechanistic/therapeutic discoveries using gene and microRNA expression profiling. Circ Res 2013; 113 (03) 252-265
  • 25 Htun NM, Chen YC, Lim B. , et al. Near-infrared autofluorescence induced by intraplaque hemorrhage and heme degradation as marker for high-risk atherosclerotic plaques. Nat Commun 2017; 8 (01) 75
  • 26 Montague SJ, Andrews RK, Gardiner EE. Mechanisms of receptor shedding in platelets. Blood 2018; 132 (24) 2535-2545
  • 27 Li X, Fang P, Li Y. , et al. Mitochondrial reactive oxygen species mediate lysophosphatidylcholine-induced endothelial cell activation. Arterioscler Thromb Vasc Biol 2016; 36 (06) 1090-1100
  • 28 Qin B, Yang H, Xiao B. Role of microRNAs in endothelial inflammation and senescence. Mol Biol Rep 2012; 39 (04) 4509-4518
  • 29 Hackl M, Brunner S, Fortschegger K. , et al. miR-17, miR-19b, miR-20a, and miR-106a are down-regulated in human aging. Aging Cell 2010; 9 (02) 291-296
  • 30 Hergenreider E, Heydt S, Tréguer K. , et al. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nat Cell Biol 2012; 14 (03) 249-256
  • 31 Puddu P, Puddu GM, Cravero E, Muscari S, Muscari A. The involvement of circulating microparticles in inflammation, coagulation and cardiovascular diseases. Can J Cardiol 2010; 26 (04) 140-145
  • 32 Shantsila E, Kamphuisen PW, Lip GY. Circulating microparticles in cardiovascular disease: implications for atherogenesis and atherothrombosis. J Thromb Haemost 2010; 8 (11) 2358-2368
  • 33 Boulanger CM, Loyer X, Rautou PE, Amabile N. Extracellular vesicles in coronary artery disease. Nat Rev Cardiol 2017; 14 (05) 259-272
  • 34 Yuan Y, Li P, Ye J. Lipid homeostasis and the formation of macrophage-derived foam cells in atherosclerosis. Protein Cell 2012; 3 (03) 173-181
  • 35 Ketelhuth DF, Hansson GK. Cellular immunity, low-density lipoprotein and atherosclerosis: break of tolerance in the artery wall. Thromb Haemost 2011; 106 (05) 779-786
  • 36 Martinet W, Schrijvers DM, De Meyer GR. Necrotic cell death in atherosclerosis. Basic Res Cardiol 2011; 106 (05) 749-760
  • 37 Ojala PJ, Hermansson M, Tolvanen M. , et al. Identification of alpha-1 acid glycoprotein as a lysophospholipid binding protein: a complementary role to albumin in the scavenging of lysophosphatidylcholine. Biochemistry 2006; 45 (47) 14021-14031
  • 38 Sinauridze EI, Kireev DA, Popenko NY. , et al. Platelet microparticle membranes have 50- to 100-fold higher specific procoagulant activity than activated platelets. Thromb Haemost 2007; 97 (03) 425-434
  • 39 McGregor L, Martin J, McGregor JL. Platelet-leukocyte aggregates and derived microparticles in inflammation, vascular remodelling and thrombosis. Front Biosci 2006; 11: 830-837
  • 40 McEver RP, Cummings RD. Role of PSGL-1 binding to selectins in leukocyte recruitment. J Clin Invest 1997; 100 (11, Suppl): S97-S103
  • 41 Kaplan ZS, Jackson SP. The role of platelets in atherothrombosis. Hematology (Am Soc Hematol Educ Program) 2011; 2011: 51-61
  • 42 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
  • 43 Gazos-Lopes F, Oliveira MM, Hoelz LV. , et al. Structural and functional analysis of a platelet-activating lysophosphatidylcholine of Trypanosoma cruzi. PLoS Negl Trop Dis 2014; 8 (08) e3077
  • 44 Jy W, Mao WW, Horstman L, Tao J, Ahn YS. Platelet microparticles bind, activate and aggregate neutrophils in vitro. Blood Cells Mol Dis 1995; 21 (03) 217-231
  • 45 Gómez-Muñoz A, O'Brien L, Hundal R, Steinbrecher UP. Lysophosphatidylcholine stimulates phospholipase D activity in mouse peritoneal macrophages. J Lipid Res 1999; 40 (06) 988-993
  • 46 Huang YH, Schäfer-Elinder L, Wu R, Claesson HE, Frostegård J. Lysophosphatidylcholine (LPC) induces proinflammatory cytokines by a platelet-activating factor (PAF) receptor-dependent mechanism. Clin Exp Immunol 1999; 116 (02) 326-331
  • 47 Badimon L, Suades R, Arderiu G, Peña E, Chiva-Blanch G, Padró T. Microvesicles in atherosclerosis and angiogenesis: from bench to bedside and reverse. Front Cardiovasc Med 2017; 4: 77
  • 48 Hafiane A, Daskalopoulou SS. Extracellular vesicles characteristics and emerging roles in atherosclerotic cardiovascular disease. Metabolism 2018; 85: 213-222
  • 49 Huber J, Vales A, Mitulovic G. , et al. Oxidized membrane vesicles and blebs from apoptotic cells contain biologically active oxidized phospholipids that induce monocyte-endothelial interactions. Arterioscler Thromb Vasc Biol 2002; 22 (01) 101-107
  • 50 Hoefer IE, Steffens S, Ala-Korpela M. , et al; ESC Working Group Atherosclerosis and Vascular Biology. Novel methodologies for biomarker discovery in atherosclerosis. Eur Heart J 2015; 36 (39) 2635-2642
  • 51 Chatterjee M, Rath D, Schlotterbeck J. , et al. Regulation of oxidized platelet lipidome: implications for coronary artery disease. Eur Heart J 2017; 38 (25) 1993-2005
  • 52 Tan KT, Lip GY. The potential role of platelet microparticles in atherosclerosis. Thromb Haemost 2005; 94 (03) 488-492
  • 53 Lavi S, McConnell JP, Rihal CS. , et al. Local production of lipoprotein-associated phospholipase A2 and lysophosphatidylcholine in the coronary circulation: association with early coronary atherosclerosis and endothelial dysfunction in humans. Circulation 2007; 115 (21) 2715-2721
  • 54 Wilensky RL, Shi Y, Mohler III ER. , et al. Inhibition of lipoprotein-associated phospholipase A2 reduces complex coronary atherosclerotic plaque development. Nat Med 2008; 14 (10) 1059-1066
  • 55 Mayr M, Grainger D, Mayr U. , et al. Proteomics, metabolomics, and immunomics on microparticles derived from human atherosclerotic plaques. Circ Cardiovasc Genet 2009; 2 (04) 379-388
  • 56 Uchida Y. Recent advances in fluorescent angioscopy for molecular imaging of human atherosclerotic coronary plaque. J Atheroscler Thromb 2017; 24 (06) 539-551
  • 57 Ménégaut L, Masson D, Abello N. , et al. Specific enrichment of 2-arachidonoyl-lysophosphatidylcholine in carotid atheroma plaque from type 2 diabetic patients. Atherosclerosis 2016; 251: 339-347
  • 58 Stegemann C, Drozdov I, Shalhoub J. , et al. Comparative lipidomics profiling of human atherosclerotic plaques. Circ Cardiovasc Genet 2011; 4 (03) 232-242
  • 59 Gonçalves I, Edsfeldt A, Ko NY. , et al. Evidence supporting a key role of Lp-PLA2-generated lysophosphatidylcholine in human atherosclerotic plaque inflammation. Arterioscler Thromb Vasc Biol 2012; 32 (06) 1505-1512
  • 60 Mannheim D, Herrmann J, Versari D. , et al. Enhanced expression of Lp-PLA2 and lysophosphatidylcholine in symptomatic carotid atherosclerotic plaques. Stroke 2008; 39 (05) 1448-1455
  • 61 Herrmann J, Mannheim D, Wohlert C. , et al. Expression of lipoprotein-associated phospholipase A(2) in carotid artery plaques predicts long-term cardiac outcome. Eur Heart J 2009; 30 (23) 2930-2938
  • 62 Nishi K, Itabe H, Uno M. , et al. Oxidized LDL in carotid plaques and plasma associates with plaque instability. Arterioscler Thromb Vasc Biol 2002; 22 (10) 1649-1654
  • 63 Qin X, Qiu C, Zhao L. Lysophosphatidylcholine perpetuates macrophage polarization toward classically activated phenotype in inflammation. Cell Immunol 2014; 289 (1-2): 185-190
  • 64 Peter C, Waibel M, Radu CG. , et al. Migration to apoptotic “find-me” signals is mediated via the phagocyte receptor G2A. J Biol Chem 2008; 283 (09) 5296-5305
  • 65 Kraemer BF, Borst O, Gehring EM. , et al. PI3 kinase-dependent stimulation of platelet migration by stromal cell-derived factor 1 (SDF-1). J Mol Med (Berl) 2010; 88 (12) 1277-1288
  • 66 Stafford MJ, Watson SP, Pears CJ. PKD: a new protein kinase C-dependent pathway in platelets. Blood 2003; 101 (04) 1392-1399
  • 67 Prokazova NV, Zvezdina ND, Korotaeva AA. Effect of lysophosphatidylcholine on transmembrane signal transduction. Biochemistry (Mosc) 1998; 63 (01) 31-37
  • 68 Schmidt EM, Münzer P, Borst O. , et al. Ion channels in the regulation of platelet migration. Biochem Biophys Res Commun 2011; 415 (01) 54-60