Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Platelet biology and functions: new concepts and clinical perspectives

Abstract

Platelets — blood cells continuously produced from megakaryocytes mainly in the bone marrow — are implicated not only in haemostasis and arterial thrombosis, but also in other physiological and pathophysiological processes. This Review describes current evidence for the heterogeneity in platelet structure, age, and activation properties, with consequences for a diversity of platelet functions. Signalling processes of platelet populations involved in thrombus formation with ongoing coagulation are well understood. Genetic approaches have provided information on multiple genes related to normal haemostasis, such as those encoding receptors and signalling or secretory proteins, that determine platelet count and/or responsiveness. As highly responsive and secretory cells, platelets can alter the environment through the release of growth factors, chemokines, coagulant factors, RNA species, and extracellular vesicles. Conversely, platelets will also adapt to their environment. In disease states, platelets can be positively primed to reach a pre-activated condition. At the inflamed vessel wall, platelets interact with leukocytes and the coagulation system, interactions mediating thromboinflammation. With current antiplatelet therapies invariably causing bleeding as an undesired adverse effect, novel therapies can be more beneficial if directed against specific platelet responses, populations, interactions, or priming conditions. On the basis of these novel concepts and processes, we discuss several initiatives to target platelets therapeutically.

Key points

  • Multiomic approaches combined with functional testing of platelets have greatly advanced the understanding of genetic factors of platelet-related haemorrhagic disorders, but to a lesser extent the understanding of the causes of platelet hyper-reactivity.

  • Negative and positive platelet priming alter the threshold for platelet activation in the circulation, with consequences for diagnostic assays.

  • The diverse pathways of information transfer by platelets through release of bioactive molecules and extracellular vesicles are still incompletely understood.

  • Platelets contribute to thromboinflammatory processes by their capacity to interact functionally with the activated endothelium, leukocytes, and coagulation proteins; the mechanisms are multivariate.

  • Platelet populations and specific platelet responses are promising targets for new antithrombotic treatment of patients with cardiovascular disease.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Major signalling events and responses during platelet activation.
Fig. 2: Platelet alterations during ageing.
Fig. 3: Environmental and platelet factors influencing platelet heterogeneity in the thrombus.
Fig. 4: Coagulation pathways contributing to the heterogeneous nature of thrombus formation.
Fig. 5: Negative and positive priming factors influencing platelet responses.
Fig. 6: Platelets in vascular thromboinflammation.

Similar content being viewed by others

References

  1. Quach, M. E., Chen, W. & Li, R. Mechanisms of platelet clearance and translation to improve platelet storage. Blood 131, 1512–1521 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Lefrancais, E. et al. The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors. Nature 544, 105–109 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Stegner, D. et al. Thrombopoiesis is spatially regulated by the bone marrow vasculature. Nat. Commun. 8, 127 (2017).

    PubMed  PubMed Central  Google Scholar 

  4. Grozovsky, R., Giannini, S., Falet, H. & Hoffmeister, K. M. Regulating billions of blood platelets: glycans and beyond. Blood 126, 1877–1884 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Kaser, A. et al. Interleukin-6 stimulates thrombopoiesis through thrombopoietin: role in inflammatory thrombocytosis. Blood 98, 2720–2725 (2001).

    CAS  PubMed  Google Scholar 

  6. den Dekker, E. et al. Cell-to-cell variability in the differentiation program of human megakaryocytes. Biochim. Biophys. Acta 1643, 85–94 (2003).

    Google Scholar 

  7. Moreau, T. et al. Large-scale production of megakaryocytes from human pluripotent stem cells by chemically defined forward programming. Nat. Commun. 7, 11208 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Machlus, K. R. & Italiano, J. E. Jr. The incredible journey: From megakaryocyte development to platelet formation. J. Cell Biol. 201, 785–796 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Bender, M. et al. Microtubule sliding drives proplatelet elongation and is dependent on cytoplasmic dynein. Blood 125, 860–868 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Josefsson, E. C. et al. Platelet production proceeds independently of the intrinsic and extrinsic apoptosis pathways. Nat. Commun. 5, 3455 (2014).

    PubMed  Google Scholar 

  11. Semeniak, D. et al. Proplatelet formation is selectively inhibited by collagen type I through Syk-independent GPVI signaling. J. Cell Sci. 129, 3473–3484 (2016).

    CAS  PubMed  Google Scholar 

  12. Abbonante, V. et al. A new path to platelet production through matrix sensing. Haematologica 102, 1150–1160 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Shi, D. S. et al. Proteasome function is required for platelet production. J. Clin. Invest. 124, 3757–3766 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. McArthur, K., Chappaz, S. & Kile, B. T. Apoptosis in megakaryocytes and platelets: the life and death of a lineage. Blood 131, 605–610 (2018).

    CAS  PubMed  Google Scholar 

  15. Mason, K. D. et al. Programmed anuclear cell death delimits platelet life span. Cell 128, 1173–1186 (2007).

    CAS  PubMed  Google Scholar 

  16. Alhasan, A. A. et al. Circular RNA enrichment in platelets is a signature of transcriptome degradation. Blood 127, e1–e11 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Male, R., Moon, D. G., Garvey, J. S., Vannier, W. E. & Baldeschwieler, J. D. Organ distributions of liposome-loaded rat platelets. Biochem. Biophys. Res. Commun. 195, 276–281 (1993).

    CAS  PubMed  Google Scholar 

  18. Karpatkin, S. Heterogeneity of human platelets. I. Metabolic and kinetic evidence suggestive of young and old platelets. J. Clin. Invest. 48, 1073–1082 (1969).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Vicic, W. J. & Weiss, H. J. Evidence that platelet α-granules are a major determinant of platelet density: studies in storage pool deficiency. Thromb. Haemost. 50, 878–880 (1983).

    CAS  PubMed  Google Scholar 

  20. Savage, B., McFadden, P. R., Hanson, S. R. & Harker, L. A. The relation of platelet density to platelet age: survival of low- and high-density 111indium-labeled platelets in baboons. Blood 68, 386–393 (1986).

    CAS  PubMed  Google Scholar 

  21. Freson, K. et al. Platelet characteristics in patients with X-linked macrothrombocytopenia because of a novel GATA1 mutation. Blood 98, 85–92 (2001).

    CAS  PubMed  Google Scholar 

  22. Baaten, C. C. F. M. J., Ten Cate, H., van der Meijden, P. E. J. & Heemskerk, J. W. M. Platelet populations and priming in hematological diseases. Blood Rev. 31, 389–399 (2017).

    PubMed  Google Scholar 

  23. Heemskerk, J. W. M., Mattheij, N. & Cosemans, J. M. E. M. Platelet-based coagulation: different populations, different functions. J. Thromb. Haemost. 11, 2–11 (2013).

    CAS  PubMed  Google Scholar 

  24. Jackson, S. P. & Schoenwaelder, S. M. Procoagulant platelets — are they necrotic? Blood 116, 2011–2018 (2010).

    CAS  PubMed  Google Scholar 

  25. Mattheij, N. J. et al. Coated platelets function in platelet-dependent fibrin formation via integrin αIIbβ3 and transglutaminase factor XIII. Haematologica 101, 427–436 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Agbani, E. O. et al. Coordinated membrane ballooning and procoagulant spreading in human platelets. Circulation 132, 1414–1424 (2015).

    CAS  PubMed  Google Scholar 

  27. Vogler, M. et al. BCL2/BCL-XL inhibition induces apoptosis, disrupts cellular calcium homeostasis and prevents platelet activation. Blood 117, 7145–7154 (2011).

    CAS  PubMed  Google Scholar 

  28. Schubert, S., Weyrich, A. S. & Rowley, J. W. A tour through the transcriptional landscape of platelets. Blood 124, 493–502 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Pleines, I. et al. Extended platelet in vivo survival results in exhausted platelets. Blood 126, 416 (2015).

    Google Scholar 

  30. Pleines, I. et al. Intrinsic apoptosis circumvents the functional decline of circulating platelets but does not cause the storage lesion. Blood 132, 197–209 (2018).

    CAS  PubMed  Google Scholar 

  31. McManus, D. D. & Freedman, J. E. MicroRNAs in platelet function and cardiovascular disease. Nat. Rev. Cardiol. 12, 711–717 (2015).

    CAS  PubMed  Google Scholar 

  32. Rowley, J. W. et al. Dicer1-mediated miRNA processing shapes the mRNA profile and function of murine platelets. Blood 127, 1743–1751 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Clancy, L., Beaulieu, L. M., Tanriverdi, K. & Freedman, J. E. The role of RNA uptake in platelet heterogeneity. Thromb. Haemost. 117, 948–961 (2017).

    PubMed  Google Scholar 

  34. Burkhart, J. M. et al. The first comprehensive and quantitative analysis of human platelet protein composition allows the comparative analysis of structural and functional pathways. Blood 120, e73–82 (2012).

    CAS  PubMed  Google Scholar 

  35. Zeiler, M., Moser, M. & Mann, M. Copy number analysis of the murine platelet proteome spanning the complete abundance range. Mol. Cell. Proteom. 13, 3435–3445 (2014).

    CAS  Google Scholar 

  36. Solari, F. A. et al. Combined quantification of the global proteome, phosphoproteome and protein cleavage to characterize altered platelet functions in the human Scott syndrome. Mol. Cell. Proteom. 15, 3154–3169 (2016).

    CAS  Google Scholar 

  37. Schoenwaelder, S. M. et al. 14-3-3ζ regulates the mitochondrial respiratory reserve linked to platelet phosphatidylserine exposure and procoagulant function. Nat. Commun. 7, 12862 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Versteeg, H. H., Heemskerk, J. W. M., Levi, M. & Reitsma, P. S. New fundamentals in hemostasis. Physiol. Rev. 93, 327–358 (2013).

    CAS  PubMed  Google Scholar 

  39. Jackson, S. P. Arterial thrombosis: insidious, unpredictable and deadly. Nat. Med. 17, 1423–1436 (2011).

    CAS  PubMed  Google Scholar 

  40. Mastenbroek, T. G., van Geffen, J. P., Heemskerk, J. W. M. & Cosemans, J. M. E. M. Acute and persistent platelet and coagulant activities in atherothrombosis. J. Thromb. Haemost. 13 (Suppl. 1), S272–S280 (2015).

    CAS  PubMed  Google Scholar 

  41. Shekhonin, B. V., Domogatsky, S. P., Muzykantov, V. R., Idelson, G. L. & Rukosuev, V. S. Distribution of type I, III, IV and V collagen in normal and atherosclerotic human arterial wall: immunomorphological characteristics. Coll. Relat. Res. 5, 355–368 (1985).

    CAS  PubMed  Google Scholar 

  42. De Witt, S. M. et al. Identification of platelet function defects by multi-parameter assessment of thrombus formation. Nat. Commun. 5, 4257 (2014).

    PubMed  Google Scholar 

  43. Swieringa, F., Spronk, H. M. H., Heemskerk, J. W. M. & van der Meijden, P. E. J. Integrating platelet and coagulation activation in fibrin clot formation. Res. Pract. Thromb. Haemost. 2, 450–460 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Dubois, C., Panicot-Dubois, L., Merrill-Skoloff, G., Furie, B. & Furie, B. C. Glycoprotein VI-dependent and -independent pathways of thrombus formation in vivo. Blood 107, 3902–3906 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhu, S., Lu, Y., Sinno, T. & Diamond, S. L. Dynamics of thrombin generation and flux from clots during whole human blood flow over collagen/tissue factor surfaces. J. Biol. Chem. 291, 23027–23035 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Zilberman-Rudensko, J. et al. Coagulation factor XI promotes distal platelet activation and single platelet consumption in the bloom stream under shear flow. Arterioscler. Thromb. Vasc. Biol. 36, 510–517 (2016).

    Google Scholar 

  47. Morowski, M. et al. Only severe thrombocytopenia results in bleeding and defective thrombus formation in mice. Blood 121, 4938–4947 (2013).

    CAS  PubMed  Google Scholar 

  48. Boulaftali, Y., Hess, P. R., Kahn, M. L. & Bergmeier, W. Platelet immunoreceptor tyrosine-based activation motif (ITAM) signaling and vascular integrity. Circ. Res. 114, 1174–1184 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Van Gestel, M. et al. Real-time detection of activation patterns in individual platelets during thromboembolism in vivo: differences between thrombus growth and embolus formation. J. Vasc. Res. 39, 534–543 (2002).

    PubMed  Google Scholar 

  50. Stalker, T. J. et al. A systems approach to hemostasis: 3. Thrombus consolidation regulates intrathrombus transport and local thrombin activity. Blood 124, 1824–1831 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Brass, L. F. & Stalker, T. J. Minding the gaps—and the junctions, too. Circulation 125, 2414–2416 (2012).

    PubMed  PubMed Central  Google Scholar 

  52. Vaiyapuri, S. et al. Gap junctions and connexin hemichannels underpin hemostasis and thrombosis. Circulation 125, 2479–2491 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Swieringa, F., Kuijpers, M. J., Lamers, M. M., van der Meijden, P. E. J. & Heemskerk, J. W. M. Rate-limiting roles of the tenase complex of factors VIII and IX in platelet procoagulant activity and formation of platelet-fibrin thrombi under flow. Haematologica 100, 748–756 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Mammadova-Bach, E. et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood 126, 683–691 (2015).

    CAS  PubMed  Google Scholar 

  55. Van der Meijden, P. E. J. et al. Dual role of collagen in factor XII-dependent thrombus and clot formation. Blood 114, 881–890 (2009).

    PubMed  Google Scholar 

  56. Verhoef, J. J. et al. Polyphosphate nanoparticles on the platelet surface trigger contact system activation. Blood 129, 1707–1717 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Payne, H., Ponomaryov, T., Watson, S. P. & Brill, A. Mice with a deficiency in CLEC-2 are protected against deep vein thrombosis. Blood 129, 2013–2020 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Stefanini, L. et al. RASA3 s a critical inhibitor of RAP1-dependent platelet activation. J. Clin. Invest. 125, 1419–1432 (2015).

    PubMed  PubMed Central  Google Scholar 

  59. Golebiewska, E. M. et al. Syntaxin 8 regulates platelet dense granule secretion, aggregation, and thrombus stability. J. Biol. Chem. 290, 1536–1545 (2015).

    PubMed  Google Scholar 

  60. Mattheij, N. J. A. et al. Survival protein anoctamin-6 controls multiple platelet responses including phospholipid scrambling, swelling and protein cleavage. FASEB. J. 30, 727–737 (2016).

    CAS  PubMed  Google Scholar 

  61. Schaff, M. et al. Integrin α6β1 is the main receptor for vascular laminins and plays a role in platelet adhesion, activation, and arterial thrombosis. Circulation 128, 541–552 (2013).

    CAS  PubMed  Google Scholar 

  62. Bunimov, N., Fuller, N. & Hayward, C. P. Genetic loci associated with platelet traits and platelet disorders. Semin. Thromb. Hemost. 39, 291–305 (2013).

    CAS  PubMed  Google Scholar 

  63. Nurden, A. T. & Nurden, P. Inherited disorders of platelet function: selected updates. J. Thromb. Haemost. 13, S2–S9 (2015).

    CAS  PubMed  Google Scholar 

  64. Bianchi, E., Norfo, R., Pennucci, V., Zini, R. & Manfredini, R. Genomic landscape of megakaryopoiesis and platelet function defects. Blood 127, 1249–1259 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Tijssen, M. R. et al. Genome-wide analysis of simultaneous GATA1/2, RUNX1, FLI1, and SCL binding in megakaryocytes identifies hematopoietic regulators. Dev. Cell 20, 597–609 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Freson, K. & Turro, E. High-throughput sequencing approaches for diagnosing hereditary bleeding and platelet disorders. J. Thromb. Haemost. 15, 1262–1272 (2017).

    CAS  PubMed  Google Scholar 

  67. Simeoni, L. et al. A comprehensive high-throughput sequencing test for the diagnosis of inherited bleeding, thrombotic and platelet disorders. Blood 127, 2791–2803 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Bastida, J. M. et al. Introducing high-throughput sequencing into mainstream genetic diagnosis practice in inherited platelet disorders. Haematologica 103, 148–162 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Lentaigne, C. et al. Inherited platelet disorders: toward DNA-based diagnosis. Blood 127, 2814–2823 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Astle, W. J. et al. The allelic landscape of human blood cell trait variation and links to common complex disease. Cell 167, 1415–1429 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Gieger, C. et al. New gene functions in megakaryopoiesis and platelet formation. Nature 480, 201–207 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Petersen, R. et al. Platelet function is modified by common sequence variation in megakaryocyte super enhancer. Nat. Commun. 8, 16058 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Nagy, M. et al. Variable impairment of platelet functions in patients with severe, genetically linked immune deficiencies. Haematologica 103, 540–549 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Snoep, J. D. et al. The minor alleleof GP6 T13254C is associated with decreased platelet activation and a reduced risk of recurrent cardiovascular events and mortality: results from the SMILE-Platelets project. J. Thromb. Haemost. 8, 2377–2384 (2010).

    CAS  PubMed  Google Scholar 

  75. Williams, M. S. et al. Genetic regulation of platelet receptor expression and function: application in clinical practice and drug development. Arterioscler. Thromb. Vasc. Biol. 30, 2372–2384 (2010).

    CAS  Google Scholar 

  76. Joshi, S. & Whiteheart, S. W. The nuts and bolts of the platelet release reaction. Platelets 28, 129–137 (2017).

    CAS  PubMed  Google Scholar 

  77. Golebiewska, E. M. & Poole, A. W. Platelet secretion: from haemostasis to wound healing and beyond. Blood Rev. 29, 153–162 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Adam, F. et al. Kinesin-1 is a new actor involved in platelet secretion and thrombus stability. Arterioscler. Thromb. Vasc. Biol. 38, 1037–1051 (2018).

    CAS  PubMed  Google Scholar 

  79. Meng, R. et al. Defective release of α granule and lysosome contents from platelets in mouse Hermansky-Pudlak syndrome models. Blood 125, 1623–1632 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Sharda, A. et al. Defective PDI release from platelets and endothelial cells impairs thrombus formation in Hermansky-Pudlak syndrome. Blood 125, 1633–1642 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Battinelli, E. M., Markens, B. A. & Italiano, J. E. Jr. Release of angiogenesis regulatory proteins from platelet α granules: modulation of physiologic and pathologic angiogenesis. Blood 118, 1359–1369 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Sobota, J. A., Ferraro, F., Back, N., Eipper, B. A. & Mains, R. E. Not all secretory granules are created equal: partitioning of soluble content proteins. Mol. Biol. Cell 17, 5038–5052 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Eckly, A. et al. Respective contributions of single and compound granule fusion to secretion by activated platelets. Blood 128, 2538–2549 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. King, S. M. et al. Platelet dense-granule secretion plays a critical role in thrombosis and subsequent vascular remodeling in atherosclerotic mice. Circulation 120, 785–791 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Deppermann, C. et al. Gray platelet syndrome and defective thrombo-inflammation in Nbeal2-deficient mice. J. Clin. Invest. 123, 3331–3342 (2013).

    CAS  PubMed Central  Google Scholar 

  86. O’Donnell, V. B., Murphy, R. C. & Watson, S. P. Platelet lipidomics: modern day perspective on lipid discovery and characterization in platelets. Circ. Res. 114, 1185–1203 (2014).

    PubMed  PubMed Central  Google Scholar 

  87. Edelstein, L. C. The role of platelet microvesicles in intercellular communication. Platelets 28, 222–227 (2017).

    CAS  PubMed  Google Scholar 

  88. Melki, I., Tessandier, N., Zufferey, A. & Boilard, E. Platelet microvesicles in health and disease. Platelets 28, 214–221 (2017).

    CAS  PubMed  Google Scholar 

  89. Dinkla, S. et al. Platelet microparticles inhibit IL-17 production by regulatory T cells through P-selectin. Blood 127, 1976–1986 (2016).

    CAS  PubMed  Google Scholar 

  90. Duchez, A. C. et al. Platelet microparticles are internalized in neutrophils via the concerted activity of 12-lipoxygenase and secreted phospholipase A2-IIA. Proc. Natl Acad. Sci. USA 112, E3564–E3573 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Vasina, E. M. et al. Aging- and activation-induced platelet microparticles suppress apoptosis in monocytic cells and differentially signal to proinflammatory mediator release. Am. J. Blood Res. 3, 107–123 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Boilard, E. et al. Platelets amplify inflammation in arthritis via collagen-dependent microparticle production. Science 327, 580–583 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Best, M. G. et al. RNA-seq of tumor-educated platelets enables blood-based pan-cancer, multiclass, and molecular pathway cancer diagnostics. Cancer Cell 28, 666–676 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Gidlof, O. et al. Platelets activated during myocardial infarction release functional miRNA, which can be taken up by endothelial cells and regulate ICAM1 expression. Blood 121, 3908–3917 (2013).

    PubMed  Google Scholar 

  95. Michael, J. V. et al. Platelet microparticles infiltrating solid tumors transfer mi-RNAs that suppress tumor growth. Blood 130, 567–580 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Keularts, I. M., van Gorp, R. M., Feijge, M. A., Vuist, W. M. & Heemskerk, J. W. α2A-adrenergic receptor stimulation potentiates calcium release in platelets by modulating cAMP levels. J. Biol. Chem. 275, 1763–1772 (2000).

    CAS  PubMed  Google Scholar 

  97. Blair, T. A. et al. Phosphoinositide 3-kinases p110α and p110β have differential roles in insulin-like growth factor-1-mediated Akt phosphorylation and platelet priming. Arterioscler. Thromb. Vasc. Biol. 34, 1681–1688 (2014).

    CAS  PubMed  Google Scholar 

  98. Cosemans, J. M. E. M. et al. Potentiating roles for Gas6 and Tyro, Axl and Mer (TAM) receptors in human and murine platelet activation and thrombus stabilization. J. Thromb. Haemost. 8, 1797–1808 (2010).

    CAS  PubMed  Google Scholar 

  99. Kuijpers, M. J. et al. Platelet CD40L modulates thrombus growth via phosphatidylinositol 3-kinase β, and not via CD40 and IκB kinase α. Arterioscler. Thromb. Vasc. Biol. 35, 1374–1381 (2015).

    CAS  PubMed  Google Scholar 

  100. Westein, E. et al. Atherosclerotic geometries spatially confine and exacerbate pathological thrombus formation poststenosis in a von Willebrand factor-dependent manner. Proc. Natl Acad. Sci. USA 110, 1357–1362 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Blair, T. A., Moore, S. F. & Hers, I. Circulating primers enhance platelet function and induce resistance to antiplatelet therapy. J. Thromb. Haemost. 13, 1479–1493 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Swieringa, F., Kuijpers, M. J. E., Heemskerk, J. W. M. & van der Meijden, P. E. J. Targeting platelet receptor function in thrombus formation: the risk of bleeding. Blood Rev. 28, 9–21 (2014).

    CAS  PubMed  Google Scholar 

  103. Naseem, K. M. & Roberts, W. Nitric oxide at a glance. Platelets 22, 148–152 (2011).

    CAS  PubMed  Google Scholar 

  104. Tourdot, B. E. et al. 12-HETrE inhibits platelet reactivity and thrombosis in part through the prostacyclin receptor. Blood Adv. 1, 1124–1131 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Kraakman, M. J. et al. Neutrophil-derived S100 calcium-binding proteins A8/A9 promote reticulated thrombocytosis and atherogenesis in diabetes. J. Clin. Invest. 127, 2133–2147 (2017).

    PubMed  PubMed Central  Google Scholar 

  106. Von Hundelshausen, P. et al. Chemokine interactome mapping enables tailored intervention in acute and chronic inflammation. Sci. Transl Med. 9, 384 (2017).

    Google Scholar 

  107. Ferroni, P. et al. Biomarkers of platelet activation in acute coronary syndromes. Thromb. Haemost. 108, 1109–1123 (2012).

    PubMed  Google Scholar 

  108. Ho-Tin-Noe, B., Demers, M. & Wagner, D. D. How platelets safeguard vascular integrity. J. Thromb. Haemost. 9 (Suppl. 1), 56–65 (2011).

    PubMed  PubMed Central  Google Scholar 

  109. Chatterjee, M. & Gawaz, M. Platelet-derived CXCL12 (SDF-1α): basic mechanisms and clinical implications. J. Thromb. Haemost. 11, 1954–1967 (2013).

    CAS  PubMed  Google Scholar 

  110. Ho-Tin-Noe, B., Boulaftali, Y. & Camerer, E. Platelets and vascular integrity: how platelets prevent bleeding in inflammation. Blood 131, 277–288 (2018).

    CAS  PubMed  Google Scholar 

  111. Croce, K. & Libby, P. Intertwining of thrombosis and inflammation in atherosclerosis. Curr. Opin. Hematol. 14, 55–61 (2007).

    CAS  PubMed  Google Scholar 

  112. Nieswandt, B., Kleinschnitz, C. & Stoll, G. Ischaemic stroke: a thrombo-inflammatory disease? J. Physiol. 589, 4115–4123 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Maiocchi, S., Alwis, I., Wu, M. C. L., Yuan, Y. & Jackson, S. P. Thromboinflammatory functions of platelets in ischemia-reperfusion injury and its dysregulation in diabetes. Semin. Thromb. Hemost. 44, 102–113 (2018).

    CAS  PubMed  Google Scholar 

  114. Kleinschnitz, C. et al. Targeting platelets in acute experimental stroke: impact of glycoprotein Ib, VI, and IIb/IIIa blockade on infarct size, functional outcome, and intracranial bleeding. Circulation 115, 2323–2330 (2007).

    CAS  PubMed  Google Scholar 

  115. Bierings, R. & Voorberg, J. Up or out: polarity of VWF release. Blood 128, 154–155 (2016).

    CAS  PubMed  Google Scholar 

  116. Sreeramkumar, V. et al. Neutrophils scan for activated platelets to initiate inflammation. Science 346, 1234–1238 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Gerdes, N. et al. Platelet CD40 exacerbates atherosclerosis by transcellular activation of endothelial cells and leukocytes. Arterioscler. Thromb. Vasc. Biol. 36, 482–490 (2016).

    CAS  PubMed  Google Scholar 

  118. Wang, Y. et al. Leukocyte integrin Mac-1 regulates thrombosis via interaction with platelet GPIbα. Nat. Commun. 8, 15559 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Koenen, R. R. et al. Disrupting functional interactions between platelet chemokines inhibits atherosclerosis in hyperlipidemic mice. Nat. Med. 15, 97–103 (2009).

    CAS  PubMed  Google Scholar 

  120. Vajen, T., Mause, S. F. & Koenen, R. R. Microvesicles from platelets: novel drivers of vascular inflammation. Thromb. Haemost. 114, 228–236 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Ekdahl, K. N. et al. Thromboinflammation in therapeutic medicine. Adv. Exp. Med. Biol. 865, 3–17 (2015).

    CAS  PubMed  Google Scholar 

  122. Martinod, K. et al. Neutrophil histone modification by peptidylarginine deiminase 4 is critical for deep vein thrombosis in mice. Proc. Natl Acad. Sci. USA 110, 8674–8679 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Muller, K. A., Chatterjee, M., Rath, D. & Geisler, T. Platelets, inflammation and anti-inflammatory effects of antiplatelet drugs in ACS and CAD. Thromb. Haemost. 114, 498–518 (2015).

    PubMed  Google Scholar 

  124. Chatterjee, M. & Geisler, T. Inflammatory contribution of platelets revisited: new players in the arena of inflammation. Semin. Thromb. Hemost. 42, 205–214 (2016).

    CAS  PubMed  Google Scholar 

  125. Koupenova, M., Clancy, L., Corkrey, H. A. & Freedman, J. E. Circulating platelets as mediators of immunity, inflammation, and thrombosis. Circ. Res. 122, 337–351 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Virmani, R., Burke, A. P., Farb, A. & Kolodgie, F. D. Pathology of the vulnerable plaque. J. Am. Coll. Cardiol. 47, C13–C18 (2006).

    CAS  PubMed  Google Scholar 

  127. Hechler, B. & Gachet, C. Comparison of two murine models of thrombosis induced by atherosclerotic plaque injury. Thromb. Haemost. 105 (Suppl. 1), S3–12 (2011).

    CAS  PubMed  Google Scholar 

  128. Kuijpers, M. J. E. et al. Complementary roles of platelets and coagulation in thrombus formation on plaques acutely ruptured by targeted ultrasound treatment: a novel intravital model. J. Thromb. Haemost. 7, 152–161 (2009).

    CAS  PubMed  Google Scholar 

  129. Farb, A. et al. Coronary plaque erosion without rupture into a lipid core. A frequent cause of coronary thrombosis in sudden coronary death. Circulation 93, 1354–1363 (1996).

    CAS  PubMed  Google Scholar 

  130. Sato, Y. et al. Proportion of fibrin and platelets differs in thrombi on ruptured and eroded coronary atherosclerotic plaques in humans. Heart 91, 526–530 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Xing, L. et al. EROSION study (Effective Anti-Thrombotic Therapy Without Stenting: Intravascular Optical Coherence Tomography-Based Management in Plaque Erosion): a 1-year follow-up report. Circ. Cardiovasc. Interv. 10 (2017).

  132. Mackman, N. Triggers, targets and treatments for thrombosis. Nature 451, 914–918 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Olie, R. H., van der Meijden, P. E. J. & Ten Cate, H. The coagulation system in atherothrombosis: implications for new therapeutic strategies. Res. Pract. Thromb. Haemost. 2, 188–198 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Patrono, C. et al. Antiplatelet agents for the treatment and prevention of coronary atherothrombosis. J. Am. Coll. Cardiol. 70, 1760–1776 (2017).

    PubMed  Google Scholar 

  135. Halvorsen, S. et al. Aspirin therapy in primary cardiovascular disease prevention: a position paper of the European Society of Cardiology working group on thrombosis. J. Am. Coll. Cardiol. 64, 319–327 (2014).

    CAS  PubMed  Google Scholar 

  136. McFadyen, J. D., Schaff, M. & Peter, K. Current and future antiplatelet therapies: emphasis on preserving haemostasis. Nat. Rev. Cardiol. 15, 181–191 (2018).

    CAS  PubMed  Google Scholar 

  137. Cattaneo, M. P2Y12 receptors: structure and function. J. Thromb. Haemost. 13 (Suppl. 1), S10–S16 (2015).

    CAS  PubMed  Google Scholar 

  138. Claessen, B. E. et al. Stent thrombosis: a clinical perspective. JACC Cardiovasc. Interv. 7, 1081–1092 (2014).

    PubMed  Google Scholar 

  139. Torrado, J. et al. Restenosis, stent thrombosis, and bleeding complications: navigating between Scylla and Charybdis. J. Am. Coll. Cardiol. 71, 1676–1695 (2018).

    PubMed  Google Scholar 

  140. Levine, G. N. et al. 2016 ACC/AHA Guideline Focused Update on duration of dual antiplatelet therapy in patients with coronary artery disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 68, 1082–1115 (2016).

    PubMed  Google Scholar 

  141. Jones, B. M. et al. Matching patients with the ever-expanding range of TAVI devices. Nat. Rev. Cardiol. 14, 615–626 (2017).

    PubMed  Google Scholar 

  142. Nishimura, R. A. et al. 2017 AHA/ACC Focused Update of the 2014 AHA/ACC Guideline for the management of patients with valvular heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J. Am. Coll. Cardiol. 70, 252–289 (2017).

    PubMed  Google Scholar 

  143. Raheja, H. et al. Comparison of single versus dual antiplatelet therapy after TAVR: a systematic review and meta-analysis. Catheter Cardiovasc. Interv. 00, 1–9 (2018).

    Google Scholar 

  144. Baumann Kreuziger, L. M., Kim, B. & Wieselthaler, G. M. Antithrombotic therapy for left ventricular assist devices in adults: a systematic review. J. Thromb. Haemost. 13, 946–955 (2015).

    CAS  PubMed  Google Scholar 

  145. Bergmeijer, T. O. et al. Genome-wide and candidate gene approaches of clopidogrel efficacy using pharmacodynamic and clinical end points-Rationale and design of the International Clopidogrel Pharmacogenomics Consortium (ICPC). Am. Heart. J. 198, 152–159 (2018).

    CAS  PubMed  Google Scholar 

  146. Gilio, K. et al. Non-redundant roles of phosphoinositide 3-kinase isoforms α and β in glycoprotein VI-induced platelet signaling and thrombus formation. J. Biol. Chem. 284, 33750–33762 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Nylander, S., Wagberg, F., Andersson, M., Skarby, T. & Gustafsson, D. Exploration of efficacy and bleeding with combined phosphoinositide 3-kinase β inhibition and aspirin in man. J. Thromb. Haemost. 13, 1494–1502 (2015).

    CAS  PubMed  Google Scholar 

  148. Tullemans, B. M. E., Heemskerk, J. W. M. & Kuijpers, M. J. E. Acquired platelet antagonism: off-target antiplatelet effects of malignancy treatment with tyrosine kinase inhibitors. J. Thromb. Haemost. 16, 1–14 (2018).

    Google Scholar 

  149. Busygina, K. et al. Oral Bruton tyrosine kinase inhibitors selectively block atherosclerotic plaque-triggered thrombus formation in humans. Blood 131, 2605–2616 (2018).

    CAS  PubMed  Google Scholar 

  150. Moeckel, D. et al. Optimizing human apyrase to treat arterial thrombosis and limit reperfusion injury without increasing bleeding risk. Sci. Transl. Med. 6, 248ra105 (2014).

    PubMed  PubMed Central  Google Scholar 

  151. Tardif, J. C. et al. Effects of the P-selectin antagonist inclacumab on myocardial damage after percutaneous coronary intervention for non-ST-segment elevation myocardial infarction: results of the SELECT-ACS trial. J. Am. Coll. Cardiol. 61, 2048–2055 (2013).

    CAS  PubMed  Google Scholar 

  152. Pasalic, L. et al. Novel assay demonstrates that coronary artery disease patients have heightened procoagulant platelet response. J. Thromb. Haemost. 16, 1198–1210 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Eikelboom, J. W. et al. Rivaroxaban with or without aspirin in stable cardiovascular disease. N. Engl. J. Med. 377, 1319–1330 (2017).

    CAS  PubMed  Google Scholar 

  154. Bye, A. P., Unsworth, A. J. & Gibbins, J. M. Platelet signaling: a complex interplay between inhibitory and activatory networks. J. Thromb. Haemost. 14, 918–930 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank the Cardiovascular Centre (HVC) of Maastricht University Medical Centre, The Netherlands, for support. We thank C. Baaten and J. van Geffen (Maastricht University, The Netherlands) for their help in preparing the figures before submission.

Reviewer information

Nature Reviews Cardiology thanks E. Gardiner, M. Gawaz, and the other, anonymous reviewer for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

Both authors researched data for the article, discussed its content, wrote the manuscript, and reviewed and edited it before submission.

Corresponding authors

Correspondence to Paola E. J. van der Meijden or Johan W. M. Heemskerk.

Ethics declarations

Competing interests

P.E.J.v.d.M. is a consultant at Bayer AG. J.W.M.H. is a founder and shareholder of FlowChamber BV.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Fibrin coat

Fibrin-coated platelets are a subpopulation of phosphatidylserine-exposing platelets that bind fibrin via transglutaminase activity and activated integrin αIIbβ3. Fibrin is ‘coated’ on the platelet surface.

Membrane ballooning

Adherent platelets on a collagen surface form phosphatidylserine-exposing, balloon-like membrane structures as a result of salt and water entry into the platelets.

Procoagulant platelet

Platelet swollen to a balloon shape, with surface exposure of phosphatidylserine and displaying greatly increased capacity for coagulation factor activation.

Pseudopod formation

Cytoplasm-filled projection of the platelet membrane following platelet activation.

α-Granules

Platelet secretion granules containing multiple stored proteins including growth factors.

δ-Granules

Platelet secretion granules with dense appearance in electron microscopy, containing Ca2+-bound nucleotides (ADP, ATP, and polyphosphates).

Plateletcrit

Product of mean platelet volume and platelet count in blood.

Negative or positive platelet priming

Suppression or promotion of platelet activation by bioactive molecules in the blood.

Exhausted platelets

Also known as refractive platelets; platelets with reduced secretion capacity owing to previous activation.

Weibel–Palade bodies

Storage granules of endothelial cells that store ultralarge von Willebrand factor multimers.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

van der Meijden, P.E.J., Heemskerk, J.W.M. Platelet biology and functions: new concepts and clinical perspectives. Nat Rev Cardiol 16, 166–179 (2019). https://doi.org/10.1038/s41569-018-0110-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41569-018-0110-0

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing