Thromb Haemost 2015; 114(05): 910-919
DOI: 10.1160/TH15-05-0410
Theme Issue Article
Schattauer GmbH

Genetics of Venous Thrombosis: update in 2015

Pierre-Emmanuel Morange
1   Laboratory of Haematology, La Timone Hospital, Marseille, France
2   Institut National pour la Santé et la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR_S) 1062, Nutrition Obesity and Risk of Thrombosis, Marseille, France
3   Aix-Marseille University, UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Marseille, France
,
Pierre Suchon
1   Laboratory of Haematology, La Timone Hospital, Marseille, France
2   Institut National pour la Santé et la Recherche Médicale (INSERM), Unité Mixte de Recherche en Santé (UMR_S) 1062, Nutrition Obesity and Risk of Thrombosis, Marseille, France
3   Aix-Marseille University, UMR_S 1062, Nutrition Obesity and Risk of Thrombosis, Marseille, France
,
David-Alexandre Trégouët
4   INSERM UMR_S 1166, Paris, France
5   Sorbonne Universités, Université Pierre et Marie Curie (UPMC Univ Paris 06), UMR_S 1166, Team Genomics & Pathophysiology of Cardiovascular Diseases, Paris, France
6   Institute for Cardiometabolism and Nutrition (ICAN), Paris, France
› Author Affiliations
Further Information

Publication History

Received: 15 May 2015

Accepted after minor revision: 14 July 2015

Publication Date:
06 December 2017 (online)

Summary

Venous thrombosis (VT) is a common multifactorial disease with a genetic component that was first suspected nearly 60 years ago. In this review, we document the genetic determinants of the disease, and update recent findings delivered by the application of high-throughput genotyping and sequencing technologies. To date, 17 genes have been robustly demonstrated to harbour genetic variations associated with VT risk: ABO, F2, F5, F9, F11, FGG, GP6, KNG1, PROC, PROCR, PROS1, SERPINC1, SLC44A2, STXBP5, THBD, TSPAN15 and VWF. The common polymorphisms are estimated to account only for a modest part (~5 %) of the VT heritability. Much remains to be done to fully disentangle the exact genetic (and epigenetic) architecture of the disease. A large suite of powerful tools and research strategies can be deployed on the large collections of patients that have already been assembled (and additional are ongoing).

* Some earlier investigations on genetic variants of candidate genes are not cited due to space contraints.


 
  • References[*]

  • 1 Jordan FL, Nandorff A. The familial tendency in thrombo-embolic disease. Acta Med Scand 1956; 156: 267-275.
  • 2 Egeberg O. Inherited antithrombin deficiency causing thrombophilia. Thromb Diath Haemorrh 195 13: 516-530.
  • 3 Souto JC. et al. Genetic determinants of hemostasis phenotypes in Spanish families. Circulation 2000; 101: 1546-1551.
  • 4 Larsen TB. et al. Major genetic susceptibility for venous thromboembolism in men: a study of Danish twins. Epidemiology 2003; 14: 328-332.
  • 5 Heit JA. et al. Familial segregation of venous thromboembolism. J Thromb Haemost 2004; 2: 731-736.
  • 6 Zöller B. et al. Age- and gender-specific familial risks for venous thromboembolism: a nationwide epidemiological study based on hospitalizations in Sweden. Circulation 2011; 124: 1012-1020.
  • 7 Jick H. et al. Venous thromboembolism disease and ABO blood type. A cooperative study. Lancet 1969; 1: 539-542.
  • 8 Griffin JH. et al. Deficiency of protein C in congenital thrombotic disease. J Clin Invest 1981; 68: 1370-1373.
  • 9 Schwarz HP. et al. Plasma protein S deficiency in familial thrombotic disease. Blood 1984; 64: 1297-1300.
  • 10 Clerget-Darpoux F. Overview of strategies for complex genetic diseases. Kidney Int 1998; 53: 1441-1445.
  • 11 Dahlbäck B. et al. Familial thrombophilia due to a previously unrecognized mechanism characterized by poor anticoagulant response to activated protein C: prediction of a cofactor to activated protein C. Proc Natl Acad Sci USA 1993; 90: 1004-1008.
  • 12 Bertina RM. et al. Mutation in blood coagulation factor V associated with resistance to activated protein C. Nature 1994; 369: 64-67.
  • 13 Smith NL. et al. Association of genetic variations with non fatal venous thrombosis in postmenopausal women. J Am Med Assoc 2007; 297: 489-498.
  • 14 Bezemer ID. et al. Updated analysis of gene variants associated with deep vein thrombosis. J Am Med Assoc 2010; 303: 421-422.
  • 15 Germain M. et al. Meta-analysis of 65, 734 individuals identifies TSPAN15 and SLC44A2 as two susceptibility loci for venous thromboembolism. Am J Hum Genet 2015; 96: 532-542.
  • 16 Poort SR. et al. A common genetic variation in the 3'-untranslated region of the prothrombin gene is associated with elevated plasma prothrombin levels and an increase in venous thrombosis. Blood 1996; 88: 3698-3703.
  • 17 Steams-Kurosawa DJ. et al. The endothelial cell protein C receptor augments protein C activation by the thrombin-thrombomodulin complex. Proc Natl Acad Sci USA 1996; 93: 10212-10216.
  • 18 Saposnik B. et al. A haplotype of the EPCR gene is associated with increased plasma levels of sEPCR and is a candidate risk factor for thrombosis. Blood 2004; 103: 1311-1318.
  • 19 Dennis J. et al. The endothelial protein C receptor (PROCR) Ser219Gly variant and risk of common thrombotic disorders: a HuGE review and meta-analysis of evidence from observational studies. Blood 2012; 119: 2392-2400.
  • 20 Uitte de Willige S. et al. Genetic variation in the fibrinogen gamma gene increases the risk for deep venous thrombosis by reducing plasma fibrinogen gamma' levels. Blood 2005; 106: 4176-4183.
  • 21 Zondervan Kt, Cardon LR. Designing candidate gene and genome-wide case-control association studies. Nat Protoc 2007; 2: 2492-2501.
  • 22 Welter D. et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res 2014; 42: D1001-1006.
  • 23 Bezemer ID. et al Gene variants associated with deep vein thrombosis. J Am Med Assoc 2008; 299: 1306-1314.
  • 24 Tregouet DA. et al. Common susceptibility alleles are unlikely to contribute as strongly as the FV and ABO loci to VTE risk: results from a GWAS approach. Blood 2009; 113: 5298-5303.
  • 25 Austin H. et al. New gene variants associated with venous thrombosis: a replication study in White and Black Americans. J Thromb Haemost 2011; 9: 489-495.
  • 26 Li Y. et al. Genetic variants associated with deep vein thrombosis: the F11 locus. J Thromb Haemost 2009; 7: 1802-1808.
  • 27 Germain M. et al. Genetics of venous thrombosis: insights from a new genome wide association study. PLoS One 2011; 6: e25581.
  • 28 Heit JA. et al. A genome-wide association study of venous thromboembolism acidentifies risk variants in chromosomes. J Thromb Haemost 2012; 10: 1521-1531.
  • 29 Tang W. et al. A genome-wide association study for venous thromboembolism: the extended cohorts for heart and aging research in genomic epidemiology(CHARGE) consortium. Genet Epidemiol 2013; 37: 512-521.
  • 30 Marchini J, Howie B. Genotype imputation for genome-wide association studies. Nat Rev Genet 2010; 11: 499-511.
  • 31 Manolio TA. et al. Finding the missing heritability of complex diseases. Nature 2009; 461: 747-753.
  • 32 Bruzelius M. et al. Influence of coronary artery disease-associated genetic variants on risk of venous thromboembolism. Thromb Res 2014; 134: 426-432.
  • 33 Smith NL. et al. Novel associations of multiple genetic loci with plasma levels of factor VII, factor VIII, and von Willebrand factor: The CHARGE (Cohorts for Heart and Aging Research in Genome Epidemiology) Consortium. Circulation 2010; 121: 1382-1392.
  • 34 Smith NL. et al. Genetic variation associated with plasma von Willebrand factor levels and the risk of incident venous thrombosis. Blood 2011; 117: 6007-6011.
  • 35 van Loon JE. et al. Effect of genetic variations in syntaxin-binding protein-5 and syntaxin-2 on von Willebrand factor concentration and cardiovascular risk. Circ Cardiovasc Genet 2010; 3: 507-512.
  • 36 Huang J. et al. Genome-wide association study for circulating tissue plasminogen activator levels and functional follow-up implicates endothelial STXBP5 and STX2. Arterioscler Thromb Vasc Biol 2014; 34: 1093-1101.
  • 37 Ye S. et al. Platelet secretion and hemostasis require syntaxin-binding protein STXBP5. J Clin Invest 2014; 124: 4517-4528.
  • 38 Zhu Q. et al. Syntaxin-binding protein STXBP5 inhibits endothelial exocytosis and promotes platelet secretion. J Clin Invest 2014; 124: 4503-4516.
  • 39 Morange PE. et al. Impact on venous thrombosis risk of newly discovered gene variants associated with FVIII and VWF plasma levels. J Thromb Haemost 2011; 9: 229-231.
  • 40 Houlihan LM. et al. Common variants of large effect in F12, KNG1, and HRG are associated with activated partial thromboplastin time. Am J Hum Genet 2010; 86: 626-631.
  • 41 Morange PE. et al. KNG1 Ile581Thr and susceptibility to venous thrombosis. Blood 2011; 117: 3692-3694.
  • 42 Sabater-Lleal M. et al. Multiethnic meta-analysis of genome-wide association studies in > 100 000 subjects identifies 23 fibrinogen-associated Loci but no strong evidence of a causal association between circulating fibrinogen and cardiovascular disease. Circulation 2013; 128: 1310-1324.
  • 43 Smith NL. et al. Genetic predictors of fibrin D-dimer levels in healthy adults. Circulation 2011; 123: 1864-1872.
  • 44 Tang W. et al. Genome-wide association study identifies novel loci for plasma levels of protein C: the ARIC study. Blood 2010; 116: 5032-5036.
  • 45 Oudot-Mellakh T. et al. Genome wide association study for plasma levels of natural anticoagulant inhibitors and protein C anticoagulant pathway: the MARTHA project. Br J Haematol 2012; 157: 230-239.
  • 46 Huang J. et al. Genome-wide association study for circulating levels of PAI-1 provides novel insights into its regulation. Blood 2012; 120: 4873-4881.
  • 47 Xu Z. et al. Tag SNP selection for candidate gene association studies using Hap-Map and gene resequencing data. Eur J Hum Genet 2007; 15: 1063-1070.
  • 48 McPherson R. A gene-centric approach to elucidating cardiovascular risk. Circ Cardiovasc Genet 2009; 2: 3-6.
  • 49 Evans DM. et al. To what extent do scans of non-synonymous SNPs complement denser genome-wide association studies ?. Eur J Hum Genet 2008; 16: 718-723.
  • 50 Cambien F, Tiret L. Genetics of cardiovascular diseases: from single mutations to the whole genome. Circulation 2007; 116: 1714-1724.
  • 51 The 1000 Genomes Project Consortium. A map of human genome variation from population-scale sequencing. Nature. 2010 467. 1061-1073.
  • 52 Germain M. et al. Caution in interpreting results from imputation analysis when linkage disequilibrium extends over a large distance: a case study on venous thrombosis. PLoS One 2012; 7: e38538.
  • 53 Berditchevski F. Complexes of tetraspanins with integrins: more than meets the eye. J Cell Sci 2001; 114: 4143-4151.
  • 54 Mair DC, Eastlund T. The pathophysiology and prevention of transfusion-related acute lung injury (TRALI): a review. Immunohematology 2010; 26: 161-173.
  • 55 Bayat B. 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 Epub ahead of print.
  • 56 Rocanin-Arjo A. et al. A meta-analysis of genome-wide association studies identifies ORM1 as a novel gene controlling thrombin generation potential. Blood 2014; 123: 777-785.
  • 57 Cohen W. et al. Risk assessment of venous thrombosis in families with known hereditary thrombophilia: the MARseilles-NImes prediction model. J Thromb Haemost 2014; 12: 138-146.
  • 58 Sørensen HT. et al. Familial risk of venous thromboembolism: a nationwide cohort study. J Thromb Haemost 2011; 9: 320-324.
  • 59 Couturaud F. et al. Factors that predict risk of thrombosis in relatives of patients with unprovoked venous thromboembolism. Chest 2009; 136: 1537-1545.
  • 60 Sadee W. et al. Missing heritability of common diseases and treatments outside the protein-coding exome. Hum Genet 2014; 133: 1199-1215.
  • 61 Zuk O. et al. Searching for missing heritability: designing rare variant association studies. Proc Natl Acad Sci USA 2014; 111: E455-E464.
  • 62 Guo Y. et al. Illumina human exome genotyping array clustering and quality control. Nat Protoc 2014; 9: 2643-2662.
  • 63 Lee S. et al. Rare-variant association analysis: study designs and statistical tests. Am J Hum Genet 2014; 95: 5-23.
  • 64 Zuo X. et al. Whole-exome SNP array identifies 15 new susceptibility loci for psoriasis. Nat Commun 2015; 6: 6793.
  • 65 Huyghe JR. et al. Exome array analysis identifies new loci and low-frequency variants influencing insulin processing and secretion. Nat Genet 2013; 45: 197-201.
  • 66 Wessel J. et al. Low-frequency and rare exome chip variants associate with fasting glucose and type 2 diabetes susceptibility. Nat Commun 2015; 6: 5897.
  • 67 Auer PL. et al. Rare and low-frequency coding variants in CXCR2 and other genes are associated with hematological traits. Nat Genet 2014; 46: 629-634.
  • 68 Peloso GM. et al. Association of low-frequency and rare coding-sequence variants with blood lipids and coronary heart disease in 56, 000 whites and blacks. Am J Hum Genet 2014; 94: 223-232.
  • 69 Holmen OL. et al. Systematic evaluation of coding variation identifies a candidate causal variant in TM6SF2 influencing total cholesterol and myocardial infarction risk. Nat Genet 2014; 46: 345-351.
  • 70 Kozlitina J. et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat Genet 2014; 46: 352-356.
  • 71 Metzker ML. Sequencing technologies – the next generation. Nat Rev Genet 2010; 11: 31-46.
  • 72 Koboldt DC. et al. The next-generation sequencing revolution and its impact on genomics. Cell 2013; 155: 27-38.
  • 73 Simioni P. et al. X-linked thrombophilia with a mutant factor IX (factor IX Padua). N Engl J Med 2009; 361: 1671-1675.
  • 74 Finn JD. et al. The efficacy and the risk of immunogenicity of FIX Padua (R338L) in hemophilia B dogs treated by AAV muscle gene therapy. Blood 2012; 120: 4521-4523.
  • 75 Miyawaki Y. et al. Thrombosis from a prothrombin mutation conveying anti-thrombin resistance. N Engl J Med 2012; 366: 2390-2396.
  • 76 Djordjevic V. et al. A novel prothrombin mutation in two families with prominent thrombophilia--the first cases of antithrombin resistance in a Caucasian population. J Thromb Haemost 2013; 11: 1936-1939.
  • 77 Tang L. et al. Common genetic risk factors for venous thrombosis in the Chinese population. Am J Hum Genet 2013; 92: 177-187.
  • 78 Nogami K. et al. Novel FV mutation (W1920R, FVNara) associated with serious deep vein thrombosis and more potent APC resistance relative to FVLeiden. Blood 2014; 123: 2420-2428.
  • 79 Castoldi E. FV and APC resistance: the plot thickens. Blood 2014; 123: 2288-2289.
  • 80 Albers CA. et al. Exome sequencing identifies NBEAL2 as the causative gene for gray platelet syndrome. Nat Genet 2011; 43: 735-737.
  • 81 Albers CA. et al. Compound inheritance of a low-frequency regulatory SNP and a rare null mutation in exon-junction complex subunit RBM8A causes TAR syndrome. Nat Genet 2012; 44: 435-439.
  • 82 Kunishima S. et al. ACTN1 mutations cause congenital macrothrombocytopnia. Am J Hum Genet 2013; 92: 431-438.
  • 83 Canault M. et al. Human CalDAG-GEFI gene (RASGRP2) mutation affects platelet function and causes severe bleeding. J Exp Med 2014; 211: 1349-1362.
  • 84 Noetzli L. et al. Germline mutations in ETV6 are associated with thrombocytopenia, red cell macrocytosis and predisposition to lymphoblastic leukemia. Nat Genet 2015; 47: 535-538.
  • 85 Do R. et al. Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature 2015; 518: 102-106.
  • 86 SIGMA Type 2 Diabetes Consortium Association of a low-frequency variant in HNF1A with type 2 diabetes in a Latino population. J Am Med Assoc 2014; 311: 2305-2314.
  • 87 Belkadi A. et al. Whole-genome sequencing is more powerful than whole-exome sequencing for detecting exome variants. Proc Natl Acad Sci USA 2015; 112: 5473-5478.
  • 88 Stankiewicz P, Lupski JR. Structural variation in the human genome and its role in disease. Annu Rev Med 2010; 61: 437-455.
  • 89 Ott J. et al. Genetic linkage analysis in the age of whole-genome sequencing. Nat Rev Genet 2015; 16: 275-284.
  • 90 Lupski JR. et al. Whole-genome sequencing in a patient with Charcot-Marie-Tooth neuropathy. N Engl J Med 2010; 362: 1181-1191.
  • 91 Jiang YH. et al. Detection of clinically relevant genetic variants in autism spectrum disorder by whole-genome sequencing. Am J Hum Genet 2013; 93: 249-263.
  • 92 Michaelson JJ. et al. Whole-genome sequencing in autism identifies hot spots for de novo germline mutation. Cell 2012; 151: 1431-1442.
  • 93 Guo G. et al. Whole-genome and whole-exome sequencing of bladder cancer identifies frequent alterations in genes involved in sister chromatid cohesion and segregation. Nat Genet 2013; 45: 1459-1463.
  • 94 Gartner JJ. et al. Whole-genome sequencing identifies a recurrent functional synonymous mutation in melanoma. Proc Natl Acad Sci USA 2013; 110: 13481-13486.
  • 95 Warde-Farley D. et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res 2010; 38 (Suppl) W214-20.
  • 96 Lotta LA. et al. Next-generation sequencing study finds an excess of rare, coding single-nucleotide variants of ADAMTS13 in patients with deep vein thrombosis. J Thromb Haemost 2013; 11: 1228-1239.
  • 97 Martini CH. et al. The effect of genetic variants in the thrombin activatable fibrinolysis inhibitor (TAFI) gene on TAFI-antigen levels, clot lysis time and the risk of venous thrombosis. Br J Haematol 2006; 134: 92-94.
  • 98 Buil A. et al. C4BPB/C4BPA is a new susceptibility locus for venous thrombosis with unknown protein S-independent mechanism: results from genome-wide association and gene expression analyses followed by case-control studies. Blood 2010; 115: 4644-4650.
  • 99 Zuk O. et al. The mystery of missing heritability: Genetic interactions create phantom heritability. Proc Natl Acad Sci USA 2012; 109: 1193-1198.
  • 100 Greliche N. et al. A genome-wide search for common SNP x SNP interactions on the risk of venous thrombosis. BMC Med Genet 2013; 14: 36.
  • 101 Monte E, Vondriska TM. Epigenomes: the missing heritability in human cardiovascular disease?. Proteomics Clin Appl 2014; 8: 480-487.
  • 102 Dick KJ. et al. DNA methylation and body-mass index: a genome-wide analysis. Lancet 2014; 383: 1990-1998.
  • 103 Gagnon F. et al. Robust validation of methylation levels association at CPT1A locus with lipid plasma levels. J Lipid Res 2014; 55: 1189-1191.
  • 104 Rocañín-Arjó A. et al. Thrombin generation potential and whole-blood DNA methylation. Thromb Res 2015; 135: 561-564.
  • 105 Lewis DA. et al. Whole blood gene expression profiles distinguish clinical phenotypes of venous thromboembolism. Thromb Res 2015; 135: 659-665.
  • 106 ISTH Steering Committee for World Thrombosis Day. Thrombosis: a major contributor to global disease burden. Thromb Haemost. 2014 112. 843-852.
  • 107 Bounameaux H, Rosendaal FR. Venous thromboembolism: why does ethnicity matter?. Circulation 2011; 123: 2189-2191.
  • 108 Rosendaal FR. Once and only once. Circulation 2010; 121: 1688-1690.
  • 109 de Haan HG. et al. Multiple SNP testing improves risk prediction of first venous thrombosis. Blood 2012; 120: 656-663.
  • 110 van Hylckama Vlieg A. et al. Genetic variations associated with recurrent venous thrombosis. Circ Cardiovasc Genet 2014; 7: 806-813.