Thromb Haemost 2020; 120(03): 423-436
DOI: 10.1055/s-0040-1702229
Coagulation and Fibrinolysis
Georg Thieme Verlag KG Stuttgart · New York

Plasma Protein Signatures of a Murine Venous Thrombosis Model and Slc44a2 Knockout Mice Using Quantitative-Targeted Proteomics

1   Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
,
Sarah A. Michaud
2   UVic-Genome British Columbia Proteomics Centre, Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
,
Chrissta X. Maracle
1   Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
,
Henri H. Versteeg
1   Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
,
Christoph H. Borchers
2   UVic-Genome British Columbia Proteomics Centre, Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
3   Molecular Pathology, Jewish General Hospital Proteomics Centre, Lady Davis Institute, McGill University, Montreal, QC, Canada
4   Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
,
Bart J. M. van Vlijmen
1   Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
,
2   UVic-Genome British Columbia Proteomics Centre, Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
5   Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
6   Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
› Author Affiliations
Funding This work was supported by the Trombosestichting Nederland (2015–4) and Landsteiner Foundation for Blood Transfusion Research (1503). This work was supported by Genome Canada and Genome British Columbia's Genome Innovations Network (204PRO for operations; 214PRO for technology development), Genome Canada and Genome British Columbia's Genomics Technology Platform (264PRO), and Genome Canada and Genome British Columbia Bioinformatics and Computational Biology (282PQP).
Further Information

Publication History

18 October 2019

11 January 2020

Publication Date:
05 March 2020 (online)

Abstract

The plasma compartment of the blood holds important information on the risk to develop cardiovascular diseases such as venous thrombosis (VT). Mass spectrometry-based targeted proteomics with internal standards quantifies proteins in multiplex allowing generation of signatures associated with a disease or a condition. Here, to demonstrate the method, we investigate the plasma protein signatures in mice following the onset of VT, which was induced by RNA interference targeting the natural anticoagulants antithrombin and protein C. We then study mice lacking Slc44a2, which was recently characterized as a VT-susceptibility gene in human genome-wide association studies. We use a recently developed panel of 375 multiplexed mouse protein assays measured by mass spectrometry. A strong plasma protein siganture was observed when VT was induced. Discriminators included acute phase response proteins, and proteins related to erythrocyte function. In mice lacking Slc44a2, protein signature was primarily overruled by the difference between sexes and not by the absent gene. Upon separate analyses for males and females, we were able to establish a signature for Slc44a2 deficiency, in which glycosylation-dependent cell adhesion molecule-1 and thrombospondin-1 were shared by both sexes. The minimal impact of Slc44a2 deficiency on the measured plasma proteins suggests that the main effect of Slc44a2 on VT does not lay ultimately in the plasma compartment. This suggests further investigation into the role of this VT-susceptibility gene should perhaps also question the possible involvement in cellular mechanisms.

Authors' Contributions

J.T, B.J.M.V.V., and Y.M. performed experimental design. J.T. and B.J.M.V.V carried out sample collection. S.A.M and Y.M. performed the MRM experiments. J.T. and Y.M. carried out the data analysis. J.T., B.J.M.V.V., and Y.M. were responsible for results interpretation and drafted the manuscript. All authors commented on the manuscript drafts and contributed to the text.


Bart J. M. van Vlijmen and Yassene Mohammed contributed equally to the study.


Supplementary Material

 
  • References

  • 1 Furie B, Furie BC. Mechanisms of thrombus formation. N Engl J Med 2008; 359 (09) 938-949
  • 2 Rosendaal FR. Venous thrombosis: the role of genes, environment, and behavior. Hematology (Am Soc Hematol Educ Program) 2005; 1-12 . Doi: 10.1182/asheducation-2005.1.1
  • 3 Tripodi A. Levels of coagulation factors and venous thromboembolism. Haematologica 2003; 88 (06) 705-711
  • 4 Rosendaal FR. Causes of venous thrombosis. Thromb J 2016; 14 (Suppl. 01) 24
  • 5 Germain M, Chasman DI, de Haan H. , et al; Cardiogenics Consortium. Meta-analysis of 65,734 individuals identifies TSPAN15 and SLC44A2 as two susceptibility loci for venous thromboembolism. Am J Hum Genet 2015; 96 (04) 532-542
  • 6 Roumen-Klappe EM, den Heijer M, van Uum SH, van der Ven-Jongekrijg J, van der Graaf F, Wollersheim H. Inflammatory response in the acute phase of deep vein thrombosis. J Vasc Surg 2002; 35 (04) 701-706
  • 7 Fox EA, Kahn SR. The relationship between inflammation and venous thrombosis. A systematic review of clinical studies. Thromb Haemost 2005; 94 (02) 362-365
  • 8 Conway EM. Reincarnation of ancient links between coagulation and complement. J Thromb Haemost 2015; 13 (Suppl. 01) S121-S132
  • 9 Mosevoll KA, Lindås R, Tvedt TH, Bruserud Ø, Reikvam H. Altered plasma levels of cytokines, soluble adhesion molecules and matrix metalloproteases in venous thrombosis. Thromb Res 2015; 136 (01) 30-39
  • 10 Morelli VM, Lijfering WM, Bos MHA, Rosendaal FR, Cannegieter SC. Lipid levels and risk of venous thrombosis: results from the MEGA-study. Eur J Epidemiol 2017; 32 (08) 669-681
  • 11 Hinds DA, Buil A, Ziemek D. , et al; METASTROKE Consortium, INVENT Consortium. Genome-wide association analysis of self-reported events in 6135 individuals and 252 827 controls identifies 8 loci associated with thrombosis. Hum Mol Genet 2016; 25 (09) 1867-1874
  • 12 Bayat B, Tjahjono Y, Berghöfer H. , et al. Choline transporter-like protein-2: new von willebrand factor-binding partner involved in antibody-mediated neutrophil activation and transfusion-related acute lung injury. Arterioscler Thromb Vasc Biol 2015; 35 (07) 1616-1622
  • 13 Kommareddi P, Nair T, Kakaraparthi BN. , et al. Hair cell loss, spiral ganglion degeneration, and progressive sensorineural hearing loss in mice with targeted deletion of Slc44a2/Ctl2. J Assoc Res Otolaryngol 2015; 16 (06) 695-712
  • 14 Tilburg J, Adili R, Nair TS. , et al. Characterization of hemostasis in mice lacking the novel thrombosis susceptibility gene Slc44a2. Thromb Res 2018; 171: 155-159
  • 15 Geyer PE, Kulak NA, Pichler G, Holdt LM, Teupser D, Mann M. Plasma proteome profiling to assess human health and disease. Cell Syst 2016; 2 (03) 185-195
  • 16 Hüttenhain R, Choi M, Martin de la Fuente L. , et al. A targeted mass spectrometry strategy for developing proteomic biomarkers: a case study of epithelial ovarian cancer. Mol Cell Proteomics 2019; 18 (09) 1836-1850
  • 17 Niu L, Geyer PE, Wewer Albrechtsen NJ. , et al. Plasma proteome profiling discovers novel proteins associated with non-alcoholic fatty liver disease. Mol Syst Biol 2019; 15 (03) e8793
  • 18 Pimienta G, Heithoff DM, Rosa-Campos A. , et al. Plasma proteome signature of sepsis: a functionally connected protein network. Proteomics 2019; 19 (05) e1800389
  • 19 Wewer Albrechtsen NJ, Geyer PE, Doll S. , et al. Plasma proteome profiling reveals dynamics of inflammatory and lipid homeostasis markers after roux-En-Y gastric bypass surgery. Cell Syst 2018; 7 (06) 601-612.e3
  • 20 Terfve C, Sabidó E, Wu Y. , et al. System-wide quantitative proteomics of the metabolic syndrome in mice: genotypic and dietary effects. J Proteome Res 2017; 16 (02) 831-841
  • 21 Liumbruno GM, Franchini M. Proteomic analysis of venous thromboembolism: an update. Expert Rev Proteomics 2013; 10 (02) 179-188
  • 22 Lippi G, Favaloro EJ, Plebani M. Proteomic analysis of venous thromboembolism. Expert Rev Proteomics 2010; 7 (02) 275-282
  • 23 Bruzelius M, Iglesias MJ, Hong MG. , et al. PDGFB, a new candidate plasma biomarker for venous thromboembolism: results from the VEREMA affinity proteomics study. Blood 2016; 128 (23) e59-e66
  • 24 Ganesh SK, Sharma Y, Dayhoff J. , et al. Detection of venous thromboembolism by proteomic serum biomarkers. PLoS ONE 2007; 2 (06) e544
  • 25 Stachowicz A, Siudut J, Suski M. , et al. Optimization of quantitative proteomic analysis of clots generated from plasma of patients with venous thromboembolism. Clin Proteomics 2017; 14: 38
  • 26 Michaud SA, Sinclair NJ, Pětrošová H. , et al. Molecular phenotyping of laboratory mouse strains using 500 multiple reaction monitoring mass spectrometry plasma assays. Commun Biol 2018; 1: 78
  • 27 Ding J, Berryman DE, Jara A, Kopchick JJ. Age- and sex-associated plasma proteomic changes in growth hormone receptor gene-disrupted mice. J Gerontol A Biol Sci Med Sci 2012; 67 (08) 830-840
  • 28 Mohammed Y, van Vlijmen BJ, Yang J. , et al. Multiplexed targeted proteomic assay to assess coagulation factor concentrations and thrombosis-associated cancer. Blood Adv 2017; 1 (15) 1080-1087
  • 29 Mohammed Y, Pan J, Zhang S, Han J, Borchers CH. ExSTA: external standard addition method for accurate high-throughput quantitation in targeted proteomics experiments. Proteomics Clin Appl 2018; 12 (02) 12
  • 30 Addona TA, Abbatiello SE, Schilling B. , et al. Multisite assessment of the precision and reproducibility of multiple reaction monitoring-based measurements of proteins in plasma. Nat Biotechnol 2009; 27 (07) 633-641
  • 31 Safdar H, Cheung KL, Salvatori D. , et al. Acute and severe coagulopathy in adult mice following silencing of hepatic antithrombin and protein C production. Blood 2013; 121 (21) 4413-4416
  • 32 Heestermans M, Salloum-Asfar S, Streef T. , et al. Mouse venous thrombosis upon silencing of anticoagulants depends on tissue factor and platelets, not FXII or neutrophils. Blood 2019; 133 (19) 2090-2099
  • 33 Cleuren AC, Van der Linden IK, De Visser YP, Wagenaar GT, Reitsma PH, Van Vlijmen BJ. 17α-Ethinylestradiol rapidly alters transcript levels of murine coagulation genes via estrogen receptor α. J Thromb Haemost 2010; 8 (08) 1838-1846
  • 34 Schwenk JM, Omenn GS, Sun Z. , et al. The human plasma proteome draft of 2017: building on the human plasma peptideatlas from mass spectrometry and complementary assays. J Proteome Res 2017; 16 (12) 4299-4310
  • 35 Bhowmick P, Mohammed Y, Borchers CH. MRMAssayDB: an integrated resource for validated targeted proteomics assays. Bioinformatics 2018; 34 (20) 3566-3571
  • 36 Mohammed Y, Bhowmick P, Smith DS. , et al. PeptideTracker: a knowledge base for collecting and storing information on protein concentrations in biological tissues. Proteomics 2017; 17 (07) 17
  • 37 Mohammed Y, Domański D, Jackson AM. , et al. PeptidePicker: a scientific workflow with web interface for selecting appropriate peptides for targeted proteomics experiments. J Proteomics 2014; 106: 151-161
  • 38 Percy AJ, Chambers AG, Yang J, Hardie DB, Borchers CH. Advances in multiplexed MRM-based protein biomarker quantitation toward clinical utility. Biochim Biophys Acta 2014; 1844 (05) 917-926
  • 39 MacLean B, Tomazela DM, Shulman N. , et al. Skyline: an open source document editor for creating and analyzing targeted proteomics experiments. Bioinformatics 2010; 26 (07) 966-968
  • 40 Ward JH. Hierarchical grouping to optimize an objective function. J Am Stat Assoc 1963; 58: 236
  • 41 Murtagh F, Legendre P. Ward's hierarchical agglomerative clustering method: which algorithms implement ward's criterion?. J Classif 2014; 31: 274-295
  • 42 Zhang Q, Menon R, Deutsch EW. , et al. A mouse plasma peptide atlas as a resource for disease proteomics. Genome Biol 2008; 9 (06) R93
  • 43 Scheurer B, Rittner C, Schneider PM. Expression of the human complement C8 subunits is independently regulated by interleukin 1 beta, interleukin 6, and interferon gamma. Immunopharmacology 1997; 38 (1-2): 167-175
  • 44 Gruys E, Toussaint MJ, Niewold TA, Koopmans SJ. Acute phase reaction and acute phase proteins. J Zhejiang Univ Sci B 2005; 6 (11) 1045-1056
  • 45 Schreiber G, Tsykin A, Aldred AR. , et al. The acute phase response in the rodent. Ann N Y Acad Sci 1989; 557: 61-85
  • 46 Ohman-Hanson RA, Cree-Green M, Kelsey MM. , et al. Ethnic and sex differences in adiponectin: from childhood to adulthood. J Clin Endocrinol Metab 2016; 101 (12) 4808-4815
  • 47 Gaya da Costa M, Poppelaars F, van Kooten C. , et al. Age and Sex-associated changes of complement activity and complement levels in a healthy caucasian population. Front Immunol 2018; 9: 2664
  • 48 Prakash P, Kulkarni PP, Chauhan AK. Thrombospondin 1 requires von Willebrand factor to modulate arterial thrombosis in mice. Blood 2015; 125 (02) 399-406
  • 49 Bonnefoy A, Daenens K, Feys HB. , et al. Thrombospondin-1 controls vascular platelet recruitment and thrombus adherence in mice by protecting (sub)endothelial VWF from cleavage by ADAMTS13. Blood 2006; 107 (03) 955-964
  • 50 Lasky LA, Singer MS, Dowbenko D. , et al. An endothelial ligand for L-selectin is a novel mucin-like molecule. Cell 1992; 69 (06) 927-938
  • 51 Dwir O, Shimron F, Chen C, Singer MS, Rosen SD, Alon R. GlyCAM-1 supports leukocyte rolling in flow: evidence for a greater dynamic stability of L-selectin rolling of lymphocytes than of neutrophils. Cell Adhes Commun 1998; 6 (04) 349-370
  • 52 Wolberg AS. Fibrinogen and factor XIII: newly recognized roles in venous thrombus formation and composition. Curr Opin Hematol 2018; 25 (05) 358-364
  • 53 Byrnes JR, Duval C, Wang Y. , et al. Factor XIIIa-dependent retention of red blood cells in clots is mediated by fibrin α-chain crosslinking. Blood 2015; 126 (16) 1940-1948
  • 54 Aleman MM, Byrnes JR, Wang JG. , et al. Factor XIII activity mediates red blood cell retention in venous thrombi. J Clin Invest 2014; 124 (08) 3590-3600
  • 55 Conforto TL, Waxman DJ. Sex-specific mouse liver gene expression: genome-wide analysis of developmental changes from pre-pubertal period to young adulthood. Biol Sex Differ 2012; 3: 9
  • 56 Jurk K, Clemetson KJ, de Groot PG. , et al. Thrombospondin-1 mediates platelet adhesion at high shear via glycoprotein Ib (GPIb): an alternative/backup mechanism to von Willebrand factor. FASEB J 2003; 17 (11) 1490-1492
  • 57 von Brühl ML, Stark K, Steinhart A. , et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 2012; 209 (04) 819-835
  • 58 Maracle CXTJ, Zirka G, Morange PE, van Vlijmen BJM, Thomas GM. Slc44a2 deficient mice exhibit less severity of thrombosis in a stenosis model of DVT. International Society on Thrombosis & Hemostasis: Research and Practice in Thrombosis and Haemostasis Melboure, Australia, 2019 Australia. Available at: https://www.ahajournals.org/doi/10.1161/atvb.38.suppl_1.090 . Accessed January 29, 2020
  • 59 Maracle CXTJ, Zirka G, van den Heijkant L, Morange PE, van Vlijmen BJM, Thomas GM. Slc44a2 deficient mice exhibit less severity of thrombosis in a stenosis model of DVT. European congress on thrombosis and haemostasis, Marseille, France, 2018 Available at: https://www.ahajournals.org/doi/10.1161/atvb.38.suppl_1.090 . Accessed January 29, 2020
  • 60 Constantinescu-Bercu A, Grassi L, Frontini M, Salles-Crawley II, Woollard KJ, Crawley JTB. Activated αIIbβ3 on platelets mediates flow-dependent NETosis via SLC44A2. bioRxiv 2019; 373670 . Doi: https://doi.org/10.1101/373670