Thromb Haemost 2017; 117(06): 1083-1092
DOI: 10.1160/TH16-09-0677
Cellular Haemostasis and Platelets
Schattauer GmbH

Whole exome sequencing in the Framingham Heart Study identifies rare variation in HYAL2 that influences platelet aggregation

John D. Eicher
1   Population Sciences Branch, National Heart, Lung, Blood Institute, National Institutes of Health, Framingham, Massachusetts, USA
2   The Framingham Heart Study, Framingham, Massachusetts, USA
,
Ming-Huei Chen
1   Population Sciences Branch, National Heart, Lung, Blood Institute, National Institutes of Health, Framingham, Massachusetts, USA
2   The Framingham Heart Study, Framingham, Massachusetts, USA
,
Achilleas N. Pitsillides
2   The Framingham Heart Study, Framingham, Massachusetts, USA
,
Honghuang Lin
3   Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
,
Narayanan Veeraraghavan
4   Rady Children’s Institute for Genomic Medicine, San Diego, California, USA
,
Jennifer A. Brody
5   Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
,
Ginger A. Metcalf
6   Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
,
Donna M. Muzny
6   Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
,
Richard A. Gibbs
6   Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
,
Diane M. Becker
7   GeneSTAR Research Program, Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
Lewis C. Becker
7   GeneSTAR Research Program, Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
8   GeneSTAR Research Program, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
Nauder Faraday
9   GeneSTAR Research Program, Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
Rasika A. Mathias
10   GeneSTAR Research Program, Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
Lisa R. Yanek
7   GeneSTAR Research Program, Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
Eric Boerwinkle
6   Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
7   GeneSTAR Research Program, Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
L. Adrienne Cupples
2   The Framingham Heart Study, Framingham, Massachusetts, USA
8   GeneSTAR Research Program, Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
,
Andrew D. Johnson
1   Population Sciences Branch, National Heart, Lung, Blood Institute, National Institutes of Health, Framingham, Massachusetts, USA
2   The Framingham Heart Study, Framingham, Massachusetts, USA
› Author Affiliations
Further Information

Publication History

Received: 01 September 2016

Accepted after major revision: 12 February 2017

Publication Date:
07 November 2017 (online)

Summary

Inhibition of platelet reactivity is a common therapeutic strategy in secondary prevention of cardiovascular disease. Genetic and environmental factors influence inter-individual variation in platelet reactivity. Identifying genes that contribute to platelet reactivity can reveal new biological mechanisms and possible therapeutic targets. Here, we examined rare coding variation to identify genes associated with platelet reactivity in a population-based cohort. To do so, we performed whole exome sequencing in the Framingham Heart Study and conducted single variant and gene-based association tests against platelet reactivity to collagen, adenosine diphosphate (ADP), and epinephrine agonists in up to 1,211 individuals. Single variant tests revealed no significant associations (p<1.44×10–7), though we observed a suggestive association with previously implicated MRVI1 (rs11042902, p = 1.95×10–7). Using gene-based association tests of rare and low-frequency variants, we found significant associations of HYAL2 with increased ADP-induced aggregation (p = 1.07×10–7) and GSTZ1 with increased epinephrine-induced aggregation (p = 1.62×10–6). HYAL2 also showed suggestive associations with epinephrine-induced aggregation (p = 2.64×10–5). The rare variants in the HYAL2 gene-based association included a missense variant (N357S) at a known N-glycosylation site and a nonsense variant (Q406*) that removes a glycophosphatidylinositol (GPI) anchor from the resulting protein. These variants suggest that improper membrane trafficking of HYAL2 influences platelet reactivity. We also observed suggestive associations of AR (p = 7.39×10–6) and MAPRE1 (p = 7.26×10–6) with ADP-induced reactivity. Our study demonstrates that gene-based tests and other grouping strategies of rare variants are powerful approaches to detect associations in population-based analyses of complex traits not detected by single variant tests and possible new genetic influences on platelet reactivity.

Supplementary Material to this article is available online at www.thrombosis-online.com.

 
  • References

  • 1 O’Donnell CJ, Larson MG, Feng D. et al. Genetic and environmental contributions to platelet aggregation: the Framingham heart study. Circulation 2001; 103: 3051-3056.
  • 2 Bray PF, Mathias RA, Faraday N. et al. Heritability of platelet function in families with premature coronary artery disease. J Thromb Haemost 2007; 5: 1617-1623.
  • 3 Johnson AD. The genetics of common variation affecting platelet development, function and pharmaceutical targeting. J Thromb Haemost 2011; 9 (Suppl. 01) 246-257.
  • 4 Davi G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med 2007; 357: 2482-2494.
  • 5 Hennekens CH, Dyken ML, Fuster V. Aspirin as a therapeutic agent in cardiovascular disease: a statement for healthcare professionals from the American Heart Association. Circulation 1997; 96: 2751-2753.
  • 6 Qayyum R, Becker LC, Becker DM. et al. Genome-wide association study of platelet aggregation in African Americans. BMC Genet 2015; 16: 58.
  • 7 Qayyum R, Snively BM, Ziv E. et al. A meta-analysis and genome-wide association study of platelet count and mean platelet volume in african americans. PLoS Genet 2012; 8: e1002491.
  • 8 Lewis JP, Ryan K, O’Connell JR. et al. Genetic variation in PEAR1 is associated with platelet aggregation and cardiovascular outcomes. Circ Cardiovasc Genet 2013; 6: 184-192.
  • 9 Johnson AD, Yanek LR, Chen MH. et al. Genome-wide meta-analyses identifies seven loci associated with platelet aggregation in response to agonists. Nat Genet 2010; 42: 608-613.
  • 10 Edelstein LC, Luna EJ, Gibson IB. et al. Human genome-wide association and mouse knockout approaches identify platelet supervillin as an inhibitor of thrombus formation under shear stress. Circulation 2012; 125: 2762-2771.
  • 11 Kauskot A, Vandenbriele C, Louwette S. et al. PEAR1 attenuates megakaryopoiesis via control of the PI3K/PTEN pathway. Blood 2013; 121: 5208-5217.
  • 12 Kauskot A, Di MM, Loyen S. et al. A novel mechanism of sustained platelet alphaIIb-beta3 activation via PEAR1. Blood 2012; 119: 4056-4065.
  • 13 Sun Y, Vandenbriele C, Kauskot A. et al. A Human Platelet Receptor Protein Micro-array Identifies the High Affinity Immunoglobulin E Receptor Subunit alpha (FcepsilonR1alpha) as an Activating Platelet Endothelium Aggregation Receptor 1 (PEAR1) Ligand. Mol Cell Proteomics 2015; 14: 1265-1274.
  • 14 Vandenbriele C, Kauskot A, Vandersmissen I. et al. Platelet endothelial aggregation receptor-1: a novel modifier of neoangiogenesis. Cardiovasc Res 2015; 108: 124-138.
  • 15 Gieger C, Radhakrishnan A, Cvejic A, Tang W, Porcu E, Pistis G. et al. New gene functions in megakaryopoiesis and platelet formation. Nature 2011; 480: 201-208.
  • 16 Eicher JD, Chami N, Kacprowski T, Normura A. Platelet-related variants identified by exome chip meta-analysis in 157,293 individuals. Am J Hum Genet 2016; 99: 40-55.
  • 17 Auer PL, Johnsen JM, Johnson AD. et al. Imputation of exome sequence variants into population- based samples and blood-cell-trait-associated loci in African Americans: NHLBI GO Exome Sequencing Project. Am J Hum Genet 2012; 91: 794-808.
  • 18 Auer PL, Teumer A, Schick U. et al. Rare and low-frequency coding variants in CXCR2 and other genes are associated with hematological traits. Nat Genet 2014; 46: 629-634.
  • 19 Psaty BM, O’Donnell CJ, Gudnason V. et al. Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium: Design of prospective meta-analyses of genome-wide association studies from 5 cohorts. Circ Cardiovasc Genet 2009; 2: 73-80.
  • 20 Bainbridge MN, Wang M, Wu Y. et al. Targeted enrichment beyond the consensus coding DNA sequence exome reveals exons with higher variant densities. Genome Biol 2011; 12: R68.
  • 21 Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009; 25: 1754-1760.
  • 22 Li H, Handsaker B, Wysoker A. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 2009; 25: 2078-2079.
  • 23 DePristo MA, Banks E, Poplin R. et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet 2011; 43: 491-498.
  • 24 Challis D, Yu J, Evani US. et al. An integrative variant analysis suite for whole exome next-generation sequencing data. BMC Bioinformatics 2012; 13: 8.
  • 25 Danecek P, Auton A, Abecasis G. et al. The variant call format and VCFtools. Bioinformatics 2011; 27: 2156-2158.
  • 26 Liu X, Wu C, Li C, Boerwinkle E. dbNSFP v3.0: A One-Stop Database of Functional Predictions and Annotations for Human Nonsynonymous and Splice-Site SNVs. Hum Mutat 2016; 37: 235-241.
  • 27 Chen MH, Yang Q. RVFam: an R package for rare variant association analysis with family data. Bioinformatics 2016; 32: 624-626.
  • 28 Li B, Leal SM. Methods for detecting associations with rare variants for common diseases: application to analysis of sequence data. Am J Hum Genet 2008; 83: 311-321.
  • 29 Madsen BE, Browning SR. A groupwise association test for rare mutations using a weighted sum statistic. PLoS Genet 2009; 5: e1000384.
  • 30 Pan W. Asymptotic tests of association with multiple SNPs in linkage disequilibrium. Genet Epidemiol 2009; 33: 497-507.
  • 31 Kircher M, Witten DM, Jain P. et al. A general framework for estimating the relative pathogenicity of human genetic variants. Nat Genet 2014; 46: 310-315.
  • 32 UniProt: a hub for protein information. Nucleic Acids Res 2015; 43: D204-D212.
  • 33 Li W, Cowley A, Uludag M. et al. The EMBL-EBI bioinformatics web and program-matic tools framework. Nucleic Acids Res 2015; 43: W580-W584.
  • 34 Eicher JD, Wakabayashi Y, Vitseva O. et al. Characterisation of the platelet transcriptome by RNA sequencing in patients with acute myocardial infarction. Platelets 2016; 27: 230-239.
  • 35 Mathias RA, Taub MA, Gignoux CR. et al. A continuum of admixture in the Western Hemisphere revealed by the African Diaspora genome. Nat Commun 2016; 7: 12522.
  • 36 Johnson AD, Handsaker RE, Pulit SL. et al. SNAP: a web-based tool for identification and annotation of proxy SNPs using HapMap. Bioinformatics 2008; 24: 2938-2939.
  • 37 Maclachlan A, Watson SP, Morgan NV. Inherited platelet disorders: Insight from platelet genomics using next-generation sequencing. Platelets 2016; 1-6.
  • 38 Chami N, Chen MH, Slater AJ. et al. Several new pleiotropic variants associated with red blood cell traits identified by exome genotyping. Am J Hum Genet 2016; 99: 8-21.
  • 39 Jadin L, Wu X, Ding H. et al. Skeletal and hematological anomalies in HYAL2-deficient mice: a second type of mucopolysaccharidosis IX?. FASEB J 2008; 22: 4316-4326.
  • 40 Onclinx C, Dogne S, Jadin L. et al. Deficiency in mouse hyaluronidase 2: a new mechanism of chronic thrombotic microangiopathy. Haematologica 2015; 100: 1023-1030.
  • 41 Albeiroti S, Ayasoufi K, Hill DR. et al. Platelet hyaluronidase-2: an enzyme that translocates to the surface upon activation to function in extracellular matrix degradation. Blood 2015; 125: 1460-1469.
  • 42 de la Motte C, Nigro J, Vasanji A. et al. Platelet-derived hyaluronidase 2 cleaves hyaluronan into fragments that trigger monocyte-mediated production of proinflammatory cytokines. Am J Pathol 2009; 174: 2254-2264.
  • 43 Goto Y, Niwa Y, Suzuki T. et al. N-glycosylation is required for secretion and enzymatic activity of human hyaluronidase1. FEBS Open Bio 2014; 4: 554-559.
  • 44 Lek M, Karczewski KJ, Minikel EV. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 2016; 536: 285-291.
  • 45 Lim CE, Matthaei KI, Blackburn AC. et al. Mice deficient in glutathione transferase zeta/maleylacetoacetate isomerase exhibit a range of pathological changes and elevated expression of alpha, mu, and pi class glutathione transferases. Am J Pathol 2004; 165: 679-693.
  • 46 Faraday N, Yanek LR, Vaidya D. et al. Leukocyte count is associated with increased platelet reactivity and diminished response to aspirin in healthy individuals with a family history of coronary artery disease. Thromb Res 2009; 124: 311-317.
  • 47 Hassel D, Dahme T, Erdmann J. et al. Nexilin mutations destabilize cardiac Z-disks and lead to dilated cardiomyopathy. Nat Med 2009; 15: 1281-1288.
  • 48 Faraday N, Goldschmidt-Clermont PJ, Bray PF. Gender differences in platelet GPIIb-IIIa activation. Thromb Haemost 1997; 77: 748-754.
  • 49 Becker DM, Segal J, Vaidya D. et al. Sex differences in platelet reactivity and response to low-dose aspirin therapy. J Am Med Assoc 2006; 295: 1420-1427.
  • 50 Kelton JG, Carter CJ, Santos A, Hirsh J. Sex related differences in platelet function: the effect of aspirin. Blood 1982; 59: 625-627.
  • 51 Kuliczkowski W, Filus A, Kuliczkowska-Plaksej J. et al. Androgen receptor polymorphism and platelet reactivity in healthy men. Thromb Res 2010; 126: e65-e67.
  • 52 Patel-Hett S, Richardson JL, Schulze H. et al. Visualisation of microtubule growth in living platelets reveals a dynamic marginal band with multiple microtubules. Blood 2008; 111: 4605-4616.
  • 53 Auer PL, Lettre G. Rare variant association studies: considerations, challenges and opportunities. Genome Med 2015; 7: 16.