Thromb Haemost 2023; 123(05): 501-509
DOI: 10.1055/s-0043-1761419
Cellular Haemostasis and Platelets

Decreased Platelet Reactivity and Function in a Mouse Model of Human Pancreatic Cancer

1   Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
,
1   Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
,
Steven P. Grover
1   Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
,
Wyatt J. Schug
2   Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
,
David S. Paul
2   Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
,
Wolfgang Bergmeier
2   Department of Biochemistry and Biophysics, UNC Blood Research Center, University of North Carolina at Chapel Hill, North Carolina, United States
,
Nigel Mackman
1   Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
› Author Affiliations
Funding The presented study was supported by grant from the NIH (N.M.: 1R35HL155657; W.B.: R35HL144976) and SENSHIN Medical Foundation (T.K.).


Abstract

Cancer patients have increased thrombosis and bleeding compared with the general population. Cancer is associated with activation of both platelets and coagulation. Mouse models have been used to study the dysregulation of platelets and coagulation in cancer. We established a mouse model of pancreatic cancer in which tissue factor-expressing human pancreatic tumors (BxPC-3) are grown in nude mice. Tumor-bearing mice have an activated coagulation system and increased venous thrombosis compared to control mice. We also showed that tumor-derived, tissue factor-positive extracellular vesicles activated platelets ex vivo and in vivo. In this study, we determined the effect of tumors on a platelet-dependent arterial thrombosis model. Unexpectedly, we observed significantly reduced carotid artery thrombosis in tumor-bearing mice compared to controls. In addition, we observed significantly increased tail bleeding in tumor-bearing mice compared to controls. These results suggested that the presence of the tumor affected platelets. Indeed, tumor-bearing mice exhibited a significant decrease in platelet count and an increase in mean platelet volume and percentage of reticulated platelets, findings that are consistent with increased platelet turnover. Levels of the platelet activation marker platelet factor 4 were also increased in tumor-bearing mice. We also observed decreased platelet receptor expression in tumor-bearing mice and reduced levels of active αIIb3 integrin in response to PAR4 agonist peptide and convulxin in platelets from tumor-bearing mice compared with platelets from control mice. In summary, our study suggests that in tumor-bearing mice there is chronic platelet activation, leading to thrombocytopenia, decreased receptor expression, and impaired platelet adhesive function.

Authors Contribution

T.K., D.S.P., W.B., and N.M. designed experiments. T.K., Y.H., D.S.P., and W.J.S. conducted experiments and analyzed data. All the authors interpreted data. T.K. and N.M. wrote the manuscript. Y.H., S.P.G., D.S.P., and W.B. edited the manuscript.




Publication History

Received: 20 April 2022

Accepted: 20 December 2022

Article published online:
30 January 2023

© 2023. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Khorana AA, Dalal M, Lin J, Connolly GC. Incidence and predictors of venous thromboembolism (VTE) among ambulatory high-risk cancer patients undergoing chemotherapy in the United States. Cancer 2013; 119 (03) 648-655
  • 2 Navi BB, Reiner AS, Kamel H. et al. Risk of arterial thromboembolism in patients with cancer. J Am Coll Cardiol 2017; 70 (08) 926-938
  • 3 Timp JF, Braekkan SK, Versteeg HH, Cannegieter SC. Epidemiology of cancer-associated venous thrombosis. Blood 2013; 122 (10) 1712-1723
  • 4 Hsu C, Patell R, Zwicker JI. The prevalence of thrombocytopenia in patients with acute cancer-associated thrombosis. Blood Adv 2022; (e-pub ahead of print). DOI: 10.1182/bloodadvances.2022008644.
  • 5 Prandoni P, Lensing AW, Piccioli A. et al. Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood 2002; 100 (10) 3484-3488
  • 6 Weitz JI, Haas S, Ageno W. et al; GARFIELD-VTE investigators. Cancer associated thrombosis in everyday practice: perspectives from GARFIELD-VTE. J Thromb Thrombolysis 2020; 50 (02) 267-277
  • 7 Riedl J, Pabinger I, Ay C. Platelets in cancer and thrombosis. Hamostaseologie 2014; 34 (01) 54-62
  • 8 Riedl J, Hell L, Kaider A. et al. Association of platelet activation markers with cancer-associated venous thromboembolism. Platelets 2016; 27 (01) 80-85
  • 9 Ay C, Simanek R, Vormittag R. et al. High plasma levels of soluble P-selectin are predictive of venous thromboembolism in cancer patients: results from the Vienna Cancer and Thrombosis Study (CATS). Blood 2008; 112 (07) 2703-2708
  • 10 Poruk KE, Firpo MA, Huerter LM. et al. Serum platelet factor 4 is an independent predictor of survival and venous thromboembolism in patients with pancreatic adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2010; 19 (10) 2605-2610
  • 11 Riedl J, Kaider A, Marosi C. et al. Decreased platelet reactivity in patients with cancer is associated with high risk of venous thromboembolism and poor prognosis. Thromb Haemost 2017; 117 (01) 90-98
  • 12 Levi M. Disseminated intravascular coagulation in cancer: an update. Semin Thromb Hemost 2019; 45 (04) 342-347
  • 13 Sallah S, Wan JY, Nguyen NP, Hanrahan LR, Sigounas G. Disseminated intravascular coagulation in solid tumors: clinical and pathologic study. Thromb Haemost 2001; 86 (03) 828-833
  • 14 Dvorak HF, Quay SC, Orenstein NS. et al. Tumor shedding and coagulation. Science 1981; 212 (4497): 923-924
  • 15 Yu JL, Rak JW. Shedding of tissue factor (TF)-containing microparticles rather than alternatively spliced TF is the main source of TF activity released from human cancer cells. J Thromb Haemost 2004; 2 (11) 2065-2067
  • 16 Geddings JE, Mackman N. Tumor-derived tissue factor-positive microparticles and venous thrombosis in cancer patients. Blood 2013; 122 (11) 1873-1880
  • 17 Mir Seyed Nazari P, Riedl J, Pabinger I, Ay C. The role of podoplanin in cancer-associated thrombosis. Thromb Res 2018; 164 (Suppl 1): S34-S39
  • 18 Geddings JE, Hisada Y, Boulaftali Y. et al. Tissue factor-positive tumor microvesicles activate platelets and enhance thrombosis in mice. J Thromb Haemost 2016; 14 (01) 153-166
  • 19 Hisada Y, Geddings JE, Ay C, Mackman N. Venous thrombosis and cancer: from mouse models to clinical trials. J Thromb Haemost 2015; 13 (08) 1372-1382
  • 20 Davila M, Amirkhosravi A, Coll E. et al. Tissue factor-bearing microparticles derived from tumor cells: impact on coagulation activation. J Thromb Haemost 2008; 6 (09) 1517-1524
  • 21 Wang JG, Geddings JE, Aleman MM. et al. Tumor-derived tissue factor activates coagulation and enhances thrombosis in a mouse xenograft model of human pancreatic cancer. Blood 2012; 119 (23) 5543-5552
  • 22 Hisada Y, Ay C, Auriemma AC, Cooley BC, Mackman N. Human pancreatic tumors grown in mice release tissue factor-positive microvesicles that increase venous clot size. J Thromb Haemost 2017; 15 (11) 2208-2217
  • 23 Thomas GM, Panicot-Dubois L, Lacroix R, Dignat-George F, Lombardo D, Dubois C. Cancer cell-derived microparticles bearing P-selectin glycoprotein ligand 1 accelerate thrombus formation in vivo. J Exp Med 2009; 206 (09) 1913-1927
  • 24 Thomas GM, Brill A, Mezouar S. et al. Tissue factor expressed by circulating cancer cell-derived microparticles drastically increases the incidence of deep vein thrombosis in mice. J Thromb Haemost 2015; 13 (07) 1310-1319
  • 25 Sasano T, Cho MS, Rodriguez-Aguayo C. et al. Role of tissue-factor bearing extracellular vesicles released from ovarian cancer cells in platelet aggregation in vitro and venous thrombosis in mice. Thrombosis Update 2021; 2: 100020
  • 26 Stark K, Schubert I, Joshi U. et al. Distinct pathogenesis of pancreatic cancer microvesicle-associated venous thrombosis identifies new antithrombotic targets in vivo. Arterioscler Thromb Vasc Biol 2018; 38 (04) 772-786
  • 27 Mezouar S, Darbousset R, Dignat-George F, Panicot-Dubois L, Dubois C. Inhibition of platelet activation prevents the P-selectin and integrin-dependent accumulation of cancer cell microparticles and reduces tumor growth and metastasis in vivo. Int J Cancer 2015; 136 (02) 462-475
  • 28 Hisada Y, Grover SP, Maqsood A. et al. Neutrophils and neutrophil extracellular traps enhance venous thrombosis in mice bearing human pancreatic tumors. Haematologica 2020; 105 (01) 218-225
  • 29 Owens III AP, Lu Y, Whinna HC, Gachet C, Fay WP, Mackman N. Towards a standardization of the murine ferric chloride-induced carotid arterial thrombosis model. J Thromb Haemost 2011; 9 (09) 1862-1863
  • 30 Schiviz A, Magirr D, Leidenmühler P, Schuster M, Muchitsch EM, Höllriegl W. Subcommittee on Animal Models of the Scientific and Standardization Committee of the International Society on Thrombosis and Hemostasis. Influence of genetic background on bleeding phenotype in the tail-tip bleeding model and recommendations for standardization: communication from the SSC of the ISTH. J Thromb Haemost 2014; 12 (11) 1940-1942
  • 31 Sommeijer DW, van Oerle R, Reitsma PH. et al. Analysis of blood coagulation in mice: pre-analytical conditions and evaluation of a home-made assay for thrombin-antithrombin complexes. Thromb J 2005; 3: 12
  • 32 Stefanini L, Paul DS, Robledo RF. et al. RASA3 is a critical inhibitor of RAP1-dependent platelet activation. J Clin Invest 2015; 125 (04) 1419-1432
  • 33 Grover SP, Mackman N. How useful are ferric chloride models of arterial thrombosis?. Platelets 2020; 31 (04) 432-438
  • 34 Palacios-Acedo AL, Mezouar S, Mège D, Crescence L, Dubois C, Panicot-Dubois L. P2RY12-inhibitors reduce cancer-associated thrombosis and tumor growth in pancreatic cancers. Front Oncol 2021; 11: 704945
  • 35 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
  • 36 Liu Y, Jennings NL, Dart AM, Du XJ. Standardizing a simpler, more sensitive and accurate tail bleeding assay in mice. World J Exp Med 2012; 2 (02) 30-36
  • 37 Bergmeier W, Rackebrandt K, Schröder W, Zirngibl H, Nieswandt B. Structural and functional characterization of the mouse von Willebrand factor receptor GPIb-IX with novel monoclonal antibodies. Blood 2000; 95 (03) 886-893
  • 38 Montague SJ, Andrews RK, Gardiner EE. Mechanisms of receptor shedding in platelets. Blood 2018; 132 (24) 2535-2545
  • 39 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
  • 40 Hisada Y, Mackman N. Mouse models of cancer-associated thrombosis. Thromb Res 2018; 164 (Suppl 1): S48-S53
  • 41 Mege D, Mezouar S, Dignat-George F, Panicot-Dubois L, Dubois C. Microparticles and cancer thrombosis in animal models. Thromb Res 2016; 140 (Suppl 1): S21-S26
  • 42 Mezouar S, Frère C, Darbousset R. et al. Role of platelets in cancer and cancer-associated thrombosis: experimental and clinical evidences. Thromb Res 2016; 139: 65-76
  • 43 Mwiza JMN, Lee RH, Paul DS. et al. Both G protein-coupled and immunoreceptor tyrosine-based activation motif receptors mediate venous thrombosis in mice. Blood 2022; 139 (21) 3194-3203