Hamostaseologie 2023; 43(05): 338-347
DOI: 10.1055/a-2113-1134
Review Article

Flow Chamber Analyses in Cardiovascular Research: Impact of Platelets and the Intercellular Crosstalk with Endothelial Cells, Leukocytes, and Red Blood Cells

Kim Jürgen Krott
1   Department of Vascular- and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
,
Tobias Feige
1   Department of Vascular- and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
,
Margitta Elvers
1   Department of Vascular- and Endovascular Surgery, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
› Author Affiliations
Funding The study was supported by the Deutsche Forschungsgemeinschaft (DFG), grant number EL651/6-1; EL651/8-1 and Collaborative Research Centre TRR259 (Aortic Disease—grant no. 397484323) to M.E.

Abstract

Platelets are main drivers of thrombus formation. Besides platelet aggregate formation, platelets interact with different blood cells such as red blood and white blood cells (RBCs, WBCs) and endothelial cells (ECs), to promote thrombus formation and inflammation. In the past, the role of different proteins in platelet adhesion, activation, and aggregate formation has been analyzed using platelets/mice with a genetic loss of a certain protein. These knock-out mouse models have been investigated for changes in experimental arterial thrombosis or hemostasis. In this review, we focused on the Maastricht flow chamber, which is a very elegant tool to analyze thrombus formation under flow using whole blood or different blood cell components of genetically modified mice. Besides, the interaction of platelets with RBCs, WBCs, and ECs under flow conditions has been evaluated with regard to thrombus formation and platelet-mediated inflammation. Importantly, alterations in thrombus formation as emerged in the flow chamber frequently reflect arterial thrombosis in different mouse models. Thus, the results of flow chamber experiments in vitro are excellent indicators for differences in arterial thrombosis in vivo. Taken together, the Maastricht flow chamber can be used to (1) determine the severity of platelet alterations in different knock-out mice; (2) analyze differences in platelet adhesion, aggregation, and activation; (3) investigate collagen and non–collagen-dependent alterations of thrombus formation; and (4) highlight differences in the interaction of platelets with different blood/ECs. Thus, this experimental approach is a useful tool to increase our understanding of signaling mechanisms that drive arterial thrombosis and hemostasis.



Publication History

Received: 02 May 2023

Accepted: 16 June 2023

Article published online:
19 October 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Nagy M, van Geffen JP, Stegner D. et al. Comparative analysis of microfluidics thrombus formation in multiple genetically modified mice: link to thrombosis and hemostasis. Front Cardiovasc Med 2019; 6: 99
  • 2 Ruggeri ZM, Mendolicchio GL. Adhesion mechanisms in platelet function. Circ Res 2007; 100 (12) 1673-1685
  • 3 Stegner D, Nieswandt B. Platelet receptor signaling in thrombus formation. J Mol Med (Berl) 2011; 89 (02) 109-121
  • 4 van der Meijden PEJ, Heemskerk JWM. Platelet biology and functions: new concepts and clinical perspectives. Nat Rev Cardiol 2019; 16 (03) 166-179
  • 5 Versteeg HH, Heemskerk JW, Levi M, Reitsma PH. New fundamentals in hemostasis. Physiol Rev 2013; 93 (01) 327-358
  • 6 Auger JM, Kuijpers MJ, Senis YA, Watson SP, Heemskerk JW. Adhesion of human and mouse platelets to collagen under shear: a unifying model. FASEB J 2005; 19 (07) 825-827
  • 7 Nieswandt B, Brakebusch C, Bergmeier W. et al. Glycoprotein VI but not alpha2beta1 integrin is essential for platelet interaction with collagen. EMBO J 2001; 20 (09) 2120-2130
  • 8 Nieswandt B, Varga-Szabo D, Elvers M. Integrins in platelet activation. J Thromb Haemost 2009; 7 (Suppl. 01) 206-209
  • 9 Siljander PR, Munnix IC, Smethurst PA. et al. Platelet receptor interplay regulates collagen-induced thrombus formation in flowing human blood. Blood 2004; 103 (04) 1333-1341
  • 10 Varga-Szabo D, Pleines I, Nieswandt B. Cell adhesion mechanisms in platelets. Arterioscler Thromb Vasc Biol 2008; 28 (03) 403-412
  • 11 Bigalke B, Elvers M, Schönberger T, Gawaz M. Platelet and soluble glycoprotein VI - novel applications in diagnosis and therapy. Curr Drug Targets 2011; 12 (12) 1821-1830
  • 12 Nieswandt B, Watson SP. Platelet-collagen interaction: is GPVI the central receptor?. Blood 2003; 102 (02) 449-461
  • 13 Elvers M, Herrmann A, Seizer P. et al. Intracellular cyclophilin A is an important Ca(2+) regulator in platelets and critically involved in arterial thrombus formation. Blood 2012; 120 (06) 1317-1326
  • 14 Mammadova-Bach E, Nagy M, Heemskerk JWM, Nieswandt B, Braun A. Store-operated calcium entry in thrombosis and thrombo-inflammation. Cell Calcium 2019; 77: 39-48
  • 15 Senis YA, Mazharian A, Mori J. Src family kinases: at the forefront of platelet activation. Blood 2014; 124 (13) 2013-2024
  • 16 Guidetti GF, Canobbio I, Torti M. PI3K/Akt in platelet integrin signaling and implications in thrombosis. Adv Biol Regul 2015; 59: 36-52
  • 17 Harper MT, Poole AW. Diverse functions of protein kinase C isoforms in platelet activation and thrombus formation. J Thromb Haemost 2010; 8 (03) 454-462
  • 18 Pleines I, Eckly A, Elvers M. et al. Multiple alterations of platelet functions dominated by increased secretion in mice lacking Cdc42 in platelets. Blood 2010; 115 (16) 3364-3373
  • 19 Pleines I, Elvers M, Strehl A. et al. Rac1 is essential for phospholipase C-gamma2 activation in platelets. Pflugers Arch 2009; 457 (05) 1173-1185
  • 20 Pleines I, Hagedorn I, Gupta S. et al. Megakaryocyte-specific RhoA deficiency causes macrothrombocytopenia and defective platelet activation in hemostasis and thrombosis. Blood 2012; 119 (04) 1054-1063
  • 21 Elvers M, Stegner D, Hagedorn I. et al. Impaired alpha(IIb)beta(3) integrin activation and shear-dependent thrombus formation in mice lacking phospholipase D1. Sci Signal 2010; 3 (103) ra1
  • 22 Metz LM, Elvers M. Pannexin-1 activation by phosphorylation is crucial for platelet aggregation and thrombus formation. Int J Mol Sci 2022; 23 (09) 5059
  • 23 Hoermann H, Krueger I, Maurus N. et al. The proteoglycan biglycan modulates platelet adhesion and thrombus formation in a GPVI-dependent manner. Int J Mol Sci 2021; 22 (22) 12168
  • 24 Gowert NS, Krüger I, Klier M. et al. Loss of reelin protects mice against arterial thrombosis by impairing integrin activation and thrombus formation under high shear conditions. Cell Signal 2017; 40: 210-221
  • 25 Krueger I, Gremer L, Mangels L. et al. Reelin amplifies glycoprotein VI activation and alphaIIb beta3 integrin outside-in signaling via PLC gamma 2 and rho GTPases. Arterioscler Thromb Vasc Biol 2020; 40 (10) 2391-2403
  • 26 Agbani EO, Poole AW. Procoagulant platelets: generation, function, and therapeutic targeting in thrombosis. Blood 2017; 130 (20) 2171-2179
  • 27 Klatt C, Krüger I, Zey S. et al. Platelet-RBC interaction mediated by FasL/FasR induces procoagulant activity important for thrombosis. J Clin Invest 2018; 128 (09) 3906-3925
  • 28 Baaten CCFMJ, Meacham S, de Witt SM. et al. A synthesis approach of mouse studies to identify genes and proteins in arterial thrombosis and bleeding. Blood 2018; 132 (24) e35-e46
  • 29 Nieswandt B, Schulte V, Bergmeier W. et al. Long-term antithrombotic protection by in vivo depletion of platelet glycoprotein VI in mice. J Exp Med 2001; 193 (04) 459-469
  • 30 Navarro S, Starke A, Heemskerk JWM, Kuijpers MJE, Stegner D, Nieswandt B. Targeting of a conserved epitope in mouse and human GPVI differently affects receptor function. Int J Mol Sci 2022; 23 (15) 8610
  • 31 Kato K, Kanaji T, Russell S. et al. The contribution of glycoprotein VI to stable platelet adhesion and thrombus formation illustrated by targeted gene deletion. Blood 2003; 102 (05) 1701-1707
  • 32 Navarro S, Stegner D, Nieswandt B, Heemskerk JWM, Kuijpers MJE. Temporal roles of platelet and coagulation pathways in collagen- and tissue factor-induced thrombus formation. Int J Mol Sci 2022; 23 (01) 358
  • 33 Elvers M, Pozgaj R, Pleines I. et al. Platelet hyperreactivity and a prothrombotic phenotype in mice with a gain-of-function mutation in phospholipase Cgamma2. J Thromb Haemost 2010; 8 (06) 1353-1363
  • 34 May F, Hagedorn I, Pleines I. et al. CLEC-2 is an essential platelet-activating receptor in hemostasis and thrombosis. Blood 2009; 114 (16) 3464-3472
  • 35 Bourne JH, Smith CW, Jooss NJ. et al. CLEC-2 supports platelet aggregation in mouse but not human blood at arterial shear. Thromb Haemost 2022; 122 (12) 1988-2000
  • 36 Suzuki-Inoue K, Inoue O, Ding G. et al. Essential in vivo roles of the C-type lectin receptor CLEC-2: embryonic/neonatal lethality of CLEC-2-deficient mice by blood/lymphatic misconnections and impaired thrombus formation of CLEC-2-deficient platelets. J Biol Chem 2010; 285 (32) 24494-24507
  • 37 Bender M, May F, Lorenz V. et al. Combined in vivo depletion of glycoprotein VI and C-type lectin-like receptor 2 severely compromises hemostasis and abrogates arterial thrombosis in mice. Arterioscler Thromb Vasc Biol 2013; 33 (05) 926-934
  • 38 Thielmann I, Stegner D, Kraft P. et al. Redundant functions of phospholipases D1 and D2 in platelet α-granule release. J Thromb Haemost 2012; 10 (11) 2361-2372
  • 39 Borst O, Schmidt EM, Münzer P. et al. The serum- and glucocorticoid-inducible kinase 1 (SGK1) influences platelet calcium signaling and function by regulation of Orai1 expression in megakaryocytes. Blood 2012; 119 (01) 251-261
  • 40 Varga-Szabo D, Braun A, Kleinschnitz C. et al. The calcium sensor STIM1 is an essential mediator of arterial thrombosis and ischemic brain infarction. J Exp Med 2008; 205 (07) 1583-1591
  • 41 Münzer P, Borst O, Walker B. et al. Acid sphingomyelinase regulates platelet cell membrane scrambling, secretion, and thrombus formation. Arterioscler Thromb Vasc Biol 2014; 34 (01) 61-71
  • 42 Elvers M. RhoGAPs and rho GTPases in platelets. Hamostaseologie 2016; 36 (03) 168-177
  • 43 Fotinos A, Klier M, Gowert NS. et al. Loss of oligophrenin1 leads to uncontrolled Rho activation and increased thrombus formation in mice. J Thromb Haemost 2015; 13 (04) 619-630
  • 44 Baumgartner HR. The role of blood flow in platelet adhesion, fibrin deposition, and formation of mural thrombi. Microvasc Res 1973; 5 (02) 167-179
  • 45 Savage B, Saldívar E, Ruggeri ZM. Initiation of platelet adhesion by arrest onto fibrinogen or translocation on von Willebrand factor. Cell 1996; 84 (02) 289-297
  • 46 Estevez B, Shen B, Du X. Targeting integrin and integrin signaling in treating thrombosis. Arterioscler Thromb Vasc Biol 2015; 35 (01) 24-29
  • 47 Jurk K, Kehrel BE. Platelets: physiology and biochemistry. Semin Thromb Hemost 2005; 31 (04) 381-392
  • 48 Ruggeri ZM. Platelets in atherothrombosis. Nat Med 2002; 8 (11) 1227-1234
  • 49 Heemskerk JW, Sakariassen KS, Zwaginga JJ, Brass LF, Jackson SP, Farndale RW. Biorheology Subcommittee of the SSC of the ISTH. Collagen surfaces to measure thrombus formation under flow: possibilities for standardization. J Thromb Haemost 2011; 9 (04) 856-858
  • 50 Cosemans JM, Kuijpers MJ, Lecut C. et al. Contribution of platelet glycoprotein VI to the thrombogenic effect of collagens in fibrous atherosclerotic lesions. Atherosclerosis 2005; 181 (01) 19-27
  • 51 Munnix IC, Kuijpers MJ, Auger J. et al. Segregation of platelet aggregatory and procoagulant microdomains in thrombus formation: regulation by transient integrin activation. Arterioscler Thromb Vasc Biol 2007; 27 (11) 2484-2490
  • 52 Cosemans JMEM, Iserbyt BF, Deckmyn H, Heemskerk JWM. Multiple ways to switch platelet integrins on and off. J Thromb Haemost 2008; 6 (08) 1253-1261
  • 53 Jackson SP. The growing complexity of platelet aggregation. Blood 2007; 109 (12) 5087-5095
  • 54 Whelihan MF, Mann KG. The role of the red cell membrane in thrombin generation. Thromb Res 2013; 131 (05) 377-382
  • 55 Walker B, Towhid ST, Schmid E. et al. Dynamic adhesion of eryptotic erythrocytes to immobilized platelets via platelet phosphatidylserine receptors. Am J Physiol Cell Physiol 2014; 306 (03) C291-C297
  • 56 Gersh KC, Nagaswami C, Weisel JW. Fibrin network structure and clot mechanical properties are altered by incorporation of erythrocytes. Thromb Haemost 2009; 102 (06) 1169-1175
  • 57 Wohner N, Sótonyi P, Machovich R. et al. Lytic resistance of fibrin containing red blood cells. Arterioscler Thromb Vasc Biol 2011; 31 (10) 2306-2313
  • 58 Wohner N. Role of cellular elements in thrombus formation and dissolution. Cardiovasc Hematol Agents Med Chem 2008; 6 (03) 224-228
  • 59 Machovich R, Owen WG. The elastase-mediated pathway of fibrinolysis. Blood Coagul Fibrinolysis 1990; 1 (01) 79-90
  • 60 Kolev K, Machovich R. Molecular and cellular modulation of fibrinolysis. Thromb Haemost 2003; 89 (04) 610-621
  • 61 Berger G, Hartwell DW, Wagner DD. P-selectin and platelet clearance. Blood 1998; 92 (11) 4446-4452
  • 62 André P, Hartwell D, Hrachovinová I, Saffaripour S, Wagner DD. Pro-coagulant state resulting from high levels of soluble P-selectin in blood. Proc Natl Acad Sci U S A 2000; 97 (25) 13835-13840
  • 63 Massberg S, Grahl L, von Bruehl ML. et al. Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med 2010; 16 (08) 887-896
  • 64 Fuchs TA, Brill A, Duerschmied D. et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 2010; 107 (36) 15880-15885
  • 65 Fuchs TA, Brill A, Wagner DD. Neutrophil extracellular trap (NET) impact on deep vein thrombosis. Arterioscler Thromb Vasc Biol 2012; 32 (08) 1777-1783
  • 66 Sorvillo N, Cherpokova D, Martinod K, Wagner DD. Extracellular DNA NET-works with dire consequences for health. Circ Res 2019; 125 (04) 470-488
  • 67 Kulkarni S, Woollard KJ, Thomas S, Oxley D, Jackson SP. Conversion of platelets from a proaggregatory to a proinflammatory adhesive phenotype: role of PAF in spatially regulating neutrophil adhesion and spreading. Blood 2007; 110 (06) 1879-1886
  • 68 Morozova DS, Martyanov AA, Obydennyi SI. et al. Ex vivo observation of granulocyte activity during thrombus formation. BMC Biol 2022; 20 (01) 32
  • 69 Goel MS, Diamond SL. Adhesion of normal erythrocytes at depressed venous shear rates to activated neutrophils, activated platelets, and fibrin polymerized from plasma. Blood 2002; 100 (10) 3797-3803
  • 70 Noh J-Y, Lim KM, Bae ON. et al. Procoagulant and prothrombotic activation of human erythrocytes by phosphatidic acid. Am J Physiol Heart Circ Physiol 2010; 299 (02) H347-H355
  • 71 Hermand P, Gane P, Huet M. et al. Red cell ICAM-4 is a novel ligand for platelet-activated alpha IIbbeta 3 integrin. J Biol Chem 2003; 278 (07) 4892-4898
  • 72 Zwaal RF, Schroit AJ. Pathophysiologic implications of membrane phospholipid asymmetry in blood cells. Blood 1997; 89 (04) 1121-1132
  • 73 Reimers RC, Sutera SP, Joist JH. Potentiation by red blood cells of shear-induced platelet aggregation: relative importance of chemical and physical mechanisms. Blood 1984; 64 (06) 1200-1206
  • 74 Sprague RS, Ellsworth ML, Stephenson AH, Lonigro AJ. ATP: the red blood cell link to NO and local control of the pulmonary circulation. Am J Physiol 1996; 271 (6, Pt 2): H2717-H2722
  • 75 Elzey BD, Tian J, Jensen RJ. et al. Platelet-mediated modulation of adaptive immunity. A communication link between innate and adaptive immune compartments. Immunity 2003; 19 (01) 9-19
  • 76 Semple JW, Italiano Jr JE, Freedman J. Platelets and the immune continuum. Nat Rev Immunol 2011; 11 (04) 264-274
  • 77 Hu H, Varon D, Hjemdahl P, Savion N, Schulman S, Li N. Platelet-leukocyte aggregation under shear stress: differential involvement of selectins and integrins. Thromb Haemost 2003; 90 (04) 679-687
  • 78 Yan SL, Russell J, Granger DN. Platelet activation and platelet-leukocyte aggregation elicited in experimental colitis are mediated by interleukin-6. Inflamm Bowel Dis 2014; 20 (02) 353-362
  • 79 Gerdes N, Seijkens T, Lievens D. et al. Platelet CD40 exacerbates atherosclerosis by transcellular activation of endothelial cells and leukocytes. Arterioscler Thromb Vasc Biol 2016; 36 (03) 482-490
  • 80 Haselmayer P, Grosse-Hovest L, von Landenberg P, Schild H, Radsak MP. TREM-1 ligand expression on platelets enhances neutrophil activation. Blood 2007; 110 (03) 1029-1035
  • 81 Swystun LL, Liaw PC. The role of leukocytes in thrombosis. Blood 2016; 128 (06) 753-762
  • 82 Klier M, Gowert NS, Jäckel S, Reinhardt C, Elvers M. Phospholipase D1 is a regulator of platelet-mediated inflammation. Cell Signal 2017; 38: 171-181
  • 83 Bombeli T, Schwartz BR, Harlan JM. Adhesion of activated platelets to endothelial cells: evidence for a GPIIbIIIa-dependent bridging mechanism and novel roles for endothelial intercellular adhesion molecule 1 (ICAM-1), alphavbeta3 integrin, and GPIbalpha. J Exp Med 1998; 187 (03) 329-339
  • 84 Gawaz M, Langer H, May AE. Platelets in inflammation and atherogenesis. J Clin Invest 2005; 115 (12) 3378-3384
  • 85 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
  • 86 Romo GM, Dong JF, Schade AJ. et al. The glycoprotein Ib-IX-V complex is a platelet counterreceptor for P-selectin. J Exp Med 1999; 190 (06) 803-814
  • 87 Theilmeier G, Michiels C, Spaepen E. et al. Endothelial von Willebrand factor recruits platelets to atherosclerosis-prone sites in response to hypercholesterolemia. Blood 2002; 99 (12) 4486-4493
  • 88 Coenen DM, Mastenbroek TG, Cosemans JMEM. Platelet interaction with activated endothelium: mechanistic insights from microfluidics. Blood 2017; 130 (26) 2819-2828
  • 89 Dardik R, Savion N, Kaufmann Y, Varon D. Thrombin promotes platelet-mediated melanoma cell adhesion to endothelial cells under flow conditions: role of platelet glycoproteins P-selectin and GPIIb-IIIA. Br J Cancer 1998; 77 (12) 2069-2075
  • 90 Gowert NS, Klier M, Reich M. et al. Defective platelet activation and bleeding complications upon cholestasis in mice. Cell Physiol Biochem 2017; 41 (06) 2133-2149
  • 91 Reusswig F, Fazel Modares N, Brechtenkamp M. et al. Efficiently restored thrombopoietin production by Ashwell-Morell receptor and IL-6R induced janus kinase 2/signal transducer and activator of transcription signaling early after partial hepatectomy. Hepatology 2021; 74 (01) 411-427
  • 92 Urbahn MA, Kaup SC, Reusswig F. et al. Phospholipase D1 regulation of TNF-alpha protects against responses to LPS. Sci Rep 2018; 8 (01) 10006
  • 93 Li TR, Liu FQ. β-Amyloid promotes platelet activation and activated platelets act as bridge between risk factors and Alzheimer's disease. Mech Ageing Dev 2022; 207: 111725
  • 94 Jarre A, Gowert NS, Donner L. et al. Pre-activated blood platelets and a pro-thrombotic phenotype in APP23 mice modeling Alzheimer's disease. Cell Signal 2014; 26 (09) 2040-2050
  • 95 Canobbio I, Visconte C, Oliviero B. et al. Increased platelet adhesion and thrombus formation in a mouse model of Alzheimer's disease. Cell Signal 2016; 28 (12) 1863-1871
  • 96 Donner L, Fälker K, Gremer L. et al. Platelets contribute to amyloid-β aggregation in cerebral vessels through integrin αIIbβ3-induced outside-in signaling and clusterin release. Sci Signal 2016; 9 (429) ra52
  • 97 Donner L, Toska LM, Krüger I. et al. The collagen receptor glycoprotein VI promotes platelet-mediated aggregation of β-amyloid. Sci Signal 2020; 13 (643) eaba9872
  • 98 Nicoll JA, Yamada M, Frackowiak J, Mazur-Kolecka B, Weller RO. Cerebral amyloid angiopathy plays a direct role in the pathogenesis of Alzheimer's disease. Pro-CAA position statement. Neurobiol Aging 2004; 25 (05) 589-597 , discussion 603–604
  • 99 Gowert NS, Donner L, Chatterjee M. et al. Blood platelets in the progression of Alzheimer's disease. PLoS One 2014; 9 (02) e90523