Thromb Haemost 2019; 119(05): 744-757
DOI: 10.1055/s-0039-1679908
Cellular Haemostasis and Platelets
Georg Thieme Verlag KG Stuttgart · New York

Epithelial (E)-Cadherin is a Novel Mediator of Platelet Aggregation and Clot Stability

Vanessa M. Scanlon*
1   Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States
2   The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
,
Alexandra M. Teixeira*
3   Department of Pathology, Yale University, New Haven, Connecticut, United States
,
Tarun Tyagi
4   Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
,
Siying Zou
5   Department of Cell Biology, Yale University, New Haven, Connecticut, United States
,
Ping-Xia Zhang
1   Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States
2   The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
,
Carmen Jane Booth
6   Department of Comparative Medicine, Yale University, New Haven, Connecticut, United States
,
M. Anna Kowalska
7   Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
8   Institute of Medical Biology, Polish Academy of Sciences, Philadelphia, Pennsylvania, United States
,
Jialing Bao
9   Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
10   Department of Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
11   University of Pennsylvania, Philadelphia, Pennsylvania, United States
,
John Hwa
4   Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
,
Vincent Hayes
12   Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
,
9   Department of Pathology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
10   Department of Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
11   University of Pennsylvania, Philadelphia, Pennsylvania, United States
,
Mortimer Poncz
11   University of Pennsylvania, Philadelphia, Pennsylvania, United States
12   Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
,
Diane S. Krause
1   Department of Laboratory Medicine, Yale University, New Haven, Connecticut, United States
2   The Yale Stem Cell Center, Yale University, New Haven, Connecticut, United States
3   Department of Pathology, Yale University, New Haven, Connecticut, United States
5   Department of Cell Biology, Yale University, New Haven, Connecticut, United States
› Institutsangaben
Funding These studies were supported by NIH R01 DK094934, the Yale NIDDK-funded Cooperative Center of Excellence in Hematology (U54 DK106857), R01 DK086267, T32HL007974 and R01 HL121323.
Weitere Informationen

Publikationsverlauf

18. Mai 2018

10. Januar 2019

Publikationsdatum:
12. März 2019 (online)

Abstract

Cadherins play a major role in mediating cell–cell adhesion, which shares many parallels with platelet–platelet interactions during aggregate formation and clot stabilization. Platelets express epithelial (E)-cadherin, but its contribution to platelet function and/or platelet production is currently unknown. To assess the role of E-cadherin in platelet production and function in vitro and in vivo, we utilized a megakaryocyte-specific E-cadherin knockout mouse model. Loss of E-cadherin in megakaryocytes does not affect megakaryocyte maturation, platelet number or size. However, platelet dysfunction in the absence of E-cadherin is revealed when conditional knockout mice are challenged with acute antibody-mediated platelet depletion. Unlike wild-type mice that recover fully, knockout mice die within 72 hours post-antibody administration, likely from haemorrhage. Furthermore, conditional knockout mice have prolonged tail bleeding times, unstable clot formation, reduced clot retraction and reduced fibrin deposition in in vivo injury models. Murine platelet aggregation in vitro in response to thrombin and thrombin receptor activating peptide is compromised in E-cadherin null platelets, while aggregation in response to adenosine diphosphate (ADP) is not significantly different. Consistent with this, in vitro aggregation of primary human platelets in response to thrombin is decreased by an inhibitory E-cadherin antibody. Integrin activation and granule secretion in response to ADP and thrombin are not affected in E-cadherin null platelets, but Akt and glycogen synthase kinase 3β (GSK3β) activation are attenuated, suggesting a that E-cadherin contributes to aggregation, clot stabilization and retraction that is mediated by phosphoinositide 3-kinase/Akt/GSK3β signalling. In summary, E-cadherin plays a salient role in platelet aggregation and clot stability.

* These authors contributed equally to this work.


 
  • References

  • 1 Du X. Signaling and regulation of the platelet glycoprotein Ib-IX-V complex. Curr Opin Hematol 2007; 14 (03) 262-269
  • 2 Schoenwaelder SM, Ono A, Nesbitt WS, Lim J, Jarman K, Jackson SP. Phosphoinositide 3-kinase p110 beta regulates integrin alpha IIb beta 3 avidity and the cellular transmission of contractile forces. J Biol Chem 2010; 285 (04) 2886-2896
  • 3 Yang H, Reheman A, Chen P. , et al. Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo. J Thromb Haemost 2006; 4 (10) 2230-2237
  • 4 Nurden AT, Fiore M, Nurden P, Pillois X. Glanzmann thrombasthenia: a review of ITGA2B and ITGB3 defects with emphasis on variants, phenotypic variability, and mouse models. Blood 2011; 118 (23) 5996-6005
  • 5 Ni H, Denis CV, Subbarao S. , et al. Persistence of platelet thrombus formation in arterioles of mice lacking both von Willebrand factor and fibrinogen. J Clin Invest 2000; 106 (03) 385-392
  • 6 Reheman A, Yang H, Zhu G. , et al. Plasma fibronectin depletion enhances platelet aggregation and thrombus formation in mice lacking fibrinogen and von Willebrand factor. Blood 2009; 113 (08) 1809-1817
  • 7 Dunne E, Spring CM, Reheman A. , et al. Cadherin 6 has a functional role in platelet aggregation and thrombus formation. Arterioscler Thromb Vasc Biol 2012; 32 (07) 1724-1731
  • 8 Elrod JW, Park JH, Oshima T, Sharp CD, Minagar A, Alexander JS. Expression of junctional proteins in human platelets. Platelets 2003; 14 (04) 247-251
  • 9 Fong KP, Barry C, Tran AN. , et al. Deciphering the human platelet sheddome. Blood 2011; 117 (01) e15-e26
  • 10 Steele BM, Harper MT, Macaulay IC. , et al. Canonical Wnt signaling negatively regulates platelet function. Proc Natl Acad Sci U S A 2009; 106 (47) 19836-19841
  • 11 Larue L, Ohsugi M, Hirchenhain J, Kemler R. E-cadherin null mutant embryos fail to form a trophectoderm epithelium. Proc Natl Acad Sci U S A 1994; 91 (17) 8263-8267
  • 12 Tiedt R, Schomber T, Hao-Shen H, Skoda RC. Pf4-Cre transgenic mice allow the generation of lineage-restricted gene knockouts for studying megakaryocyte and platelet function in vivo. Blood 2007; 109 (04) 1503-1506
  • 13 Boussadia O, Kutsch S, Hierholzer A, Delmas V, Kemler R. E-cadherin is a survival factor for the lactating mouse mammary gland. Mech Dev 2002; 115 (1-2): 53-62
  • 14 Cheng EC, Luo Q, Bruscia EM. , et al. Role for MKL1 in megakaryocytic maturation. Blood 2009; 113 (12) 2826-2834
  • 15 Smith EC, Thon JN, Devine MT. , et al. MKL1 and MKL2 play redundant and crucial roles in megakaryocyte maturation and platelet formation. Blood 2012; 120 (11) 2317-2329
  • 16 Sharkis SJ, Cahill R, Ahmed A, Jedrzejczak WW, Sell KW. Genetic requirements for bone marrow transplantation for stem-cell-defective W/Wv mice. Transplant Proc 1979; 11 (01) 511-516
  • 17 Mahooti S, Graesser D, Patil S. , et al. PECAM-1 (CD31) expression modulates bleeding time in vivo. Am J Pathol 2000; 157 (01) 75-81
  • 18 Eslin DE, Zhang C, Samuels KJ. , et al. Transgenic mice studies demonstrate a role for platelet factor 4 in thrombosis: dissociation between anticoagulant and antithrombotic effect of heparin. Blood 2004; 104 (10) 3173-3180
  • 19 Neyman M, Gewirtz J, Poncz M. Analysis of the spatial and temporal characteristics of platelet-delivered factor VIII-based clots. Blood 2008; 112 (04) 1101-1108
  • 20 Falati S, Gross P, Merrill-Skoloff G, Furie BC, Furie B. Real-time in vivo imaging of platelets, tissue factor and fibrin during arterial thrombus formation in the mouse. Nat Med 2002; 8 (10) 1175-1181
  • 21 Weiler-Guettler H, Christie PD, Beeler DL. , et al. A targeted point mutation in thrombomodulin generates viable mice with a prethrombotic state. J Clin Invest 1998; 101 (09) 1983-1991
  • 22 Uchtmann K, Park ER, Bergsma A, Segula J, Edick MJ, Miranti CK. Homozygous loss of mouse tetraspanin CD82 enhances integrin αIIbβ3 expression and clot retraction in platelets. Exp Cell Res 2015; 339 (02) 261-269
  • 23 Tyagi T, Ahmad S, Gupta N. , et al. Altered expression of platelet proteins and calpain activity mediate hypoxia-induced prothrombotic phenotype. Blood 2014; 123 (08) 1250-1260
  • 24 Chen J, De S, Damron DS, Chen WS, Hay N, Byzova TV. Impaired platelet responses to thrombin and collagen in AKT-1-deficient mice. Blood 2004; 104 (06) 1703-1710
  • 25 Woulfe D, Jiang H, Morgans A, Monks R, Birnbaum M, Brass LF. Defects in secretion, aggregation, and thrombus formation in platelets from mice lacking Akt2. J Clin Invest 2004; 113 (03) 441-450
  • 26 O'Brien KA, Stojanovic-Terpo A, Hay N, Du X. An important role for Akt3 in platelet activation and thrombosis. Blood 2011; 118 (15) 4215-4223
  • 27 Chen X, Zhang Y, Wang Y. , et al. PDK1 regulates platelet activation and arterial thrombosis. Blood 2013; 121 (18) 3718-3726
  • 28 Morowski M, Vögtle T, Kraft P, Kleinschnitz C, Stoll G, Nieswandt B. Only severe thrombocytopenia results in bleeding and defective thrombus formation in mice. Blood 2013; 121 (24) 4938-4947
  • 29 Sproull M, Kramp T, Tandle A, Shankavaram U, Camphausen K. Serum amyloid A as a biomarker for radiation exposure. Radiat Res 2015; 184 (01) 14-23
  • 30 Jin J, Daniel JL, Kunapuli SP. Molecular basis for ADP-induced platelet activation. II. The P2Y1 receptor mediates ADP-induced intracellular calcium mobilization and shape change in platelets. J Biol Chem 1998; 273 (04) 2030-2034
  • 31 Hollopeter G, Jantzen HM, Vincent D. , et al. Identification of the platelet ADP receptor targeted by antithrombotic drugs. Nature 2001; 409 (6817): 202-207
  • 32 Zhang FL, Luo L, Gustafson E. , et al. ADP is the cognate ligand for the orphan G protein-coupled receptor SP1999. J Biol Chem 2001; 276 (11) 8608-8615
  • 33 Moore SF, van den Bosch MT, Hunter RW, Sakamoto K, Poole AW, Hers I. Dual regulation of glycogen synthase kinase 3 (GSK3)α/β by protein kinase C (PKC)α and Akt promotes thrombin-mediated integrin αIIbβ3 activation and granule secretion in platelets. J Biol Chem 2013; 288 (06) 3918-3928
  • 34 Laurent PA, Séverin S, Hechler B, Vanhaesebroeck B, Payrastre B, Gratacap MP. Platelet PI3Kβ and GSK3 regulate thrombus stability at a high shear rate. Blood 2015; 125 (05) 881-888
  • 35 Jiang L, Xu C, Yu S. , et al. A critical role of thrombin/PAR-1 in ADP-induced platelet secretion and the second wave of aggregation. J Thromb Haemost 2013; 11 (05) 930-940
  • 36 Prevost N, Woulfe D, Tognolini M, Brass LF. Contact-dependent signaling during the late events of platelet activation. J Thromb Haemost 2003; 1 (07) 1613-1627
  • 37 Guilford P, Hopkins J, Harraway J. , et al. E-cadherin germline mutations in familial gastric cancer. Nature 1998; 392 (6674): 402-405