Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jul 1.
Published in final edited form as: J Thromb Haemost. 2011 Jul;9(Suppl 1):246–257. doi: 10.1111/j.1538-7836.2011.04359.x

The Genetics of Common Variation affecting Platelet Development, Function and Pharmaceutical Targeting

Andrew D Johnson 1
PMCID: PMC3151008  NIHMSID: NIHMS299407  PMID: 21781261

Summary

Common variant effects on human platelet function and response to anti-platelet treatment have traditionally been studied using candidate gene approaches involving a limited number of variants and genes. These studies have often been undertaken in clinically defined cohorts. More recently, studies have applied genome-wide scans in larger population samples than prior candidate studies, in some cases scanning relatively healthy individuals. These studies demonstrate synergy with some prior candidate gene findings (e.g., GP6, ADRA2A) but also uncover novel loci involved in platelet function. Here, I summarise findings on common genetic variation influencing platelet development, function and therapeutics. Taken together, candidate gene and genome-wide studies begin to account for common variation in platelet function and provide information that may ultimately be useful in pharmacogenetic applications in the clinic. More than 50 loci have been identified with consistent associations with platelet phenotypes in ≥2 populations. Several variants are under further study in clinical trials relating to anti-platelet therapies. In order to have useful clinical applications, variants must have large effects on a modifiable outcome. Regardless of clinical applications, studies of common genetic influences, even of small effect, offer additional insights into platelet biology including the importance of intracellular signalling and novel receptors. Understanding of common platelet-related genetics remains behind parallel fields (e.g., lipids, blood pressure) due to challenges in phenotype ascertainment. Further work is necessary to discover and characterise loci for platelet function, and to assess whether these loci contribute to disease aetiologies or response to therapeutics.

Keywords: platelet, aggregation, polymorphism, genome, GWAS, SNP

Importance, measurement and heritability of platelet traits

Platelets play critical roles in thrombosis and haemostasis and are the targets of multiple pharmaceutical targeting strategies and endogenous activation pathways. Severe inherited disorders impacting platelet function have been described underlining the clinical relevance of platelet function (1;2). Evidence further indicates that successful modulation of platelet function provides significant survival benefit, and that variation in platelet volume or function may be an independent risk factor and predictor for disease or outcomes (312). Although studies of anti-platelet drug resistance have significant limitations in terms of phenotypic definition, there is general agreement that a significant portion of individuals exhibit anti-platelet resistance (13;14). This, along with the underlying importance of platelet biology and the severity of rare platelet disorders, has provided significant motivation for the study of platelet-related genetics.

This article reviews knowledge on common genetic influences on platelet development, function and pharmaceutical targeting, with particular emphasis on recent developments employing genomic technologies. In particular, a catalogue of >450 platelet-related genome-wide association study (GWAS) results is provided in Supplemental Table 1. Common platelet traits in genetic studies include assays of cell parameters usually performed in whole blood [e.g., platelet count (PLT) and mean platelet volume (MPV)], measurements of platelet function by aggregation response in whole blood or platelet rich plasma (PRP) to a variety of agonists (e.g., collagen), mimetics or stimulants (e.g., shear stress), and less direct measurements of platelet function (e.g., P-selectin levels, fibrinogen levels). Each measurement has potential limitations and focuses most on specific portions of the platelet life cycle or activation cascades.

One gold standard for platelet function testing is light aggregometry in PRP (15), but this measure has a drawback for genetic studies in being challenging to scale to the large sample numbers required to achieve significant statistical power in outbred populations. Cell counts, mainly PLT and MPV in whole blood, are more scalable, and thus have been more widely applied in large genetic studies. While PLT and MPV provide less specific information about platelet function, with modern methods they have reasonable coefficients of variation (16), and the largest genetic study to date on platelet function provided evidence for partial overlap with PLT/MPV loci indicating they may be good proxy measurements for discovering some loci that contribute to platelet function variability (17).

A high degree of heritability for platelet quantitative traits has been widely established, first with twin studies of PRP epinephrine-induced aggregation, PLT and MPV (18;19). Studies on the heritability (h2) of platelet measures are summarised in Table 1 with the general pattern for h2 being: MPV > PLT > platelet function tests. A large study of PRP aggregation in families in the Framingham Heart Study (FHS) established the heritability of platelet function to a range of agonists within a general population (20). The Johns Hopkins GeneStar (GS) study later provided further evidence of heritability in both European ancestry and African ancestry families, and to both whole blood (WB) and PRP responses with a range of measures, and in pre- (21) and post- (22) aspirin conditions. Animal model studies, particularly in families of baboons (23) and mouse crosses (24;25), confirm the importance of inherited alleles to platelet measures in other species.

Table 1.

Heritability studies for platelet-related traits.

Phenotype description Cohort description Heritability observations Citation
Epinephrine-induced PRP aggregation 32 male twin pairs (17 MZ, 15 DZ) PRP Epi=0.65 in MZ
PRP Epi=0.43 in DZ
Gaxiola 1984 (19)
PLT, MPV (Coulter) 92 male and female twin pairs PLT=0.86, MPV=0.88 Whitfield 1985 (18)
PLT 105 twin pairs n.s. Dal Colletto 1993 (81)
PLT (Coulter) 385 twin pairs (192 female, 193 male) PLT=0.80 Evans 1999 (35)
PLT (Bayer H3 RTX autoanalyzer) 712 twin pairs (234 MZ, 478 DZ) PLT=0.57 Garner 2000 (82)
Epinephrine, ADP and collagen induced PRP aggregation 1,041 siblings and 928 spouses ADP=0.44
Epinephrine=0.44
Collagen=0.62
O’Donnell 2001 (20)
PLT 6,111 Italian ancestry individuals in Sardinia PLT=0.64 Pilia 2006 (83)
Whole blood aggregation (ADP, Collagen, AA, Tbx), PFA closure epinephrine (PFA-100), PRP aggregation (ADP, Collagen, AA, Epinephrine), PLT, MPV 687 European ancestry, 321 African ancestry individuals in families PLT=0.60/0.67*
MPV=0.55/0.71
WB ADP=0.51/0.39
WB Collagen=0.28/n.s.
WB AA=0.58/n.s.
PFA closure(epi)=0.24/0.61
PRP Collagen=0.26/0.79
PRP Coll. Lag=0.50/0.47
PRP AA=0.38/n.s.
PRP Epi=0.36/0.76
PRP ADP=0.42/0.78
Bray 2007 (21)
Post-aspirin responses with or without adjustment of baseline measures: most as above in Bray et al., additionally TXB2 (ELISA), Tx-M (ELISA), beta-thromboglobulin release (ELISA) 1,144 European ancestry, 736 African ancestry individuals in families PRP Collagen=0.29/0.39*,
PRP Coll. Lag=0.23/n.s.
WB Collagen=0.27/0.28
PRP ADP=0.22/n.s.
WB ADP=0.20/n.s.
PRP Epi=0.33/n.s.
PFA closure(epi)=n.s./n.s.
TXB2=0.38/n.s.
Tx-M=n.s./0.35
B-thromboglobulin=0.33/n.s.
Faraday 2007 (22)
PLT, MPV, PCT, PDW 1,101 Italian ancestry individuals in S. Tyrol PLT=0.55, MPV=0.79, PCT=0.53, PDW=0.37 Marroni 2008 (84)
PLT, MPV 1,803 Italian ancestry ind. in Val Borbera PLT=0.60, MPV=0.73 Traglia 2009 (85)
PLT (Coulter) 12,517 individuals in Sardinia PLT=0.54 Biino 2011 (86)
*

Heritabilites are given for the two populations as European ancestry/African ancestry. Where multiple doses were studied the maximum heritability is given.

Heritabilities under a model fully adjusted for pre-aspirin treatment and other covariates are given. n.s. indicates the authors did not accept the hypothesis that the trait was significantly heritable.

PCT=plateletocrit, PDW=platelet distribution width

Early studies into the genetics of platelet traits

Early studies into the genetics of specific platelet-related genes largely stemmed from a focus on cells derived from alloimmune thrombocytopenias and efforts to identify the alloantibodies (26). Isolation and sequencing of cDNAs due to the anuclear nature of platelets led to the discovery of coding region polymorphisms. Early variant discovery was restricted by this approach, the nascent state of information on the human genome sequence and infeasibility of sequencing large populations for the coding regions and surrounding sequence. Most early studies focused on cell surface receptors and their ligands, a candidate gene approach, as these were among the most highly expressed RNAs and proteins with recognised roles in platelet function. A series of coding and proposed regulatory polymorphisms were further studied for their functional effects in association with diseases including venous thrombosis (VT), stroke and myocardial infarction (MI).

A thorough review of platelet candidate gene studies is beyond the scope of this review. Readers are directed to other recent reviews that cover this topic in more depth (2628). Overall, studies from the candidate gene era did not produce many results that were consistently replicated in independent populations for roles in either platelet function and/or disease. There are a variety of potential explanations for this observation including the heterogeneity in ex vivo assays used in platelet function studies. However, the modest successes of candidate gene studies in platelets are not inconsistent with the experiences of other genetics fields of this era (e.g., blood pressure genetics), and were likely due to limited statistical power in small population samples and the relatively incomplete knowledge of common alleles in human populations available in that time period.

There are a few notable exceptions of platelet candidate genes and polymorphisms that show relatively consistent positive results in the literature including the GP6 and ADRA2A receptors. Another important exception is CYP2C19, a cytochrome P-450 enzyme responsible for metabolism of the pro-drug form of clopidogrel, polymorphisms of which have been consistently associated with clopidogrel response. Some candidate genes have re-emerged in genome-wide studies (see below) providing a validated role for these genes in genetic variability (17;29). A similar pattern has been observed for blood pressure loci (30) and other traits (31;32), showing that the information gained from candidate gene studies remains valuable in the genome-wide era.

Genome-wide human linkage and animal QTL studies

First attempts at genome-wide genetic studies for platelet traits employed microsatellite studies to conduct linkage analysis in families, or quantitative trait locus (QTL) mapping in animals. Most linkage analyses use a moderate number of markers and search for cosegregation of markers in individuals’ within families with a disease or other trait, generating LOD scores or similar statistics. Results from linkage and QTL studies are summarised in Table 2. While these studies produced several genome-wide significant LOD scores, and even more suggestive signals, in most cases they did not lead to fine mapping, identification or replication of novel platelet function genes. An exception is a linkage study of PLT in Asian Indian pedigrees which fine-mapped putative functional mutations in THPO and GP9 (33). Remarkably, a recent GWAS for PLT in Japanese individuals had a strong peak signal at the same THPO variant (rs6141) identified by fine mapping of the linkage scan (34).

Table 2.

Human linkage and animal QTL studies for platelet traits.

Phenotype description Sample description Human or orthologous human
regions (LOD)
Citation
Animal studies
PLT (Sysmex XE9000 hematology analyzer) 1,126 F2 progeny from a CBA/CaH X QSi5 intercross Orthologous regions not given.
PLT (LODs 11.2, 6.8, 3.6, 3.5, 3.4)
Cheung 2004 (24)
PLT, MPV (Advia 120 whole blood analyzer) 279 F2 progeny each from crosses of NZW/LacJ X SM/J and C57BLKS/J X SM/J MPV – 12q12-13 (24.2)
MPV – 16q13-p21 (2.7)
PLT – 9q33-34 (3.5)
PLT – 11p15 (3.3)
PLT – 2p25 (3.2)
Peters 2005 (25)
PLT, MPV (Coulter) 582 baboons (410 female, 172 male) in 11 pedigrees PLT/MCV – 1p34.2-p36.23 (3.1)
PLT/Hb – 19p13.12-q13.41 (2.2)
PLT/MPV – 10q26.3 (2.0)
PLT/RDW – 3p23-p22.3 (1.6)
PLT/RDW – 10q26.3 (1.5)
MPV/RDW – 20p12.1-q11.22 (2.4)
MPV/RDW – 10q23.3 (2.0)
MPV/MCV – 9q34.2-q34.3 (1.7)
MPV/RBC – 8p21.3-p22 (1.6)
Bertin 2007 (23)
PLT, MPV, PDW (CD1700 whole blood analyzer) 1,033 F2 progeny of White Durox X Erhualian porcine crosses Orthologous regions not given.
PLT (F-val 12.1, 10.1)
PDW (F-val 21.7, 11.1, 9.6)
MPV (F-val 10.1)
Yang 2009 (87)
PLT, MPV, PDW, PCT (TEK-II mini automatic hemocyte analyser) 368 purebred piglet (Landrace, Large White, Songliao Black) Orthologous regions not given.
PLT (LR 17.89, 14.61)
PDW (LR 22.24, 19.12)
Gong 2010 (88)
Human studies
PLT (Coulter) 745 twin pairs (327 MZ, 418 DZ) PLT – 19q13.11-q13.32 (2.59)
PLT – 10q11.23-q22.1 (2.18)
PLT – 5q32-q33.2 (1.97)
PLT – 2q24.3-q31.1 (1.42)
PLT – 5q23.1 (1.26)
PLT – 15q11.2 (1.15)
Evans 2004 (35)
PLT (Bayer automated analyzer) 6 Asian Indian families (125 with PLT+markers) PLT – 3q23 (3.26)
PLT – 3q26.2 (2.52)
Garner 2006 (33)
Epinephrine, ADP and collagen induced PRP aggregation Up to 724 individuals in families ADP – 7q21.12 (2.0)
Collagen – Xp11.4 (2.0)
Yang 2007 (57)
Pre- and Post-aspirin responses with or without adjustment of baseline measures: most as above in Bray et al. Table 1, additionally Tx-M (ELISA) 1,231 European ancestry, 846 African ancestry individuals in families WB ADP post AA – 5q11.2 (3.6)
WB ADP post AA – 5q12.3 (3.6)
WB ADP post AA – 5q13.1 (2.2)
WB ADP post EA – 5q35.3 (2.2)
WB epi post EA – 3p14.2 (2.2)
WB epi post EA – 11q23.3 (2.2)
WB coll. pre AA – 1q23.3 (2.2)
WB coll. post EA – 4p12 (2.2)
WB coll. post EA – 4p14 (2.2)
WB coll. post EA – 4q12 (2.2)
WB coll. post EA – 6q25.3 (2.2)
Coll. lag pre AA – 19q13.41 (2.2)
Mathias 2010 (36)

MCV = mean corpuscular volume, Hb = hemoglobin, RDW = red cell distribution width, PDW = platelet distribution width, PCT = plateletocrit

There are a few notable consistencies across the human and animal studies. Human chromosome region 19q13.41 was encompassed in linkage peaks for PLT in twins (35), for collagen lag time response in African-Americans (36), and for an orthologous region in the baboon genome in a bivariate analysis of PLT and haemoglobin levels (23). This region contains GP6, the direct receptor for platelet activation by collagen, and a previous candidate gene with characterised functional coding alleles. This locus was further validated in a recent GWAS for PRP collagen lag time (17).

Regions orthologous to human chromosome 9q33-q34.3 were identified in mouse PLT studies (25) and in bivariate analysis of MPV and mean corpuscular volume in baboons (23). Notably, this large region of the human genome contains the human blood group antigen locus ABO. While this locus has not emerged in large human linkage or genome-wide association studies (GWAS) of PLT, MPV or platelet function to date, some candidate studies have indicated an effect of ABO blood group on particular platelet function measures (3739). Furthermore, a GWAS study of soluble P-selectin and ICAM-1 levels found the ABO locus strongly linked with these measures (40). The effects of ABO-type matching on platelet transfusions may be of potential important to clinical outcomes. A meta-analysis indicates that ABO-matched transfusions consistently result in higher outcome platelet counts (41), though further studies are needed to determine conclusively whether this is of definitive clinical benefit.

Functional genomics studies of platelet traits

Genome-wide genetic studies, transcriptomic (RNA) and proteomic studies have the potential to dramatically advance scientific understanding because they are relatively unbiased and current technologies probe a significant fraction of common human variation, expressed transcripts and proteins. Genome-wide genetic studies have flourished due to extensive maps of common genetic variation (e.g., the HapMap and the 1000 Genomes Project), as well as the stability of DNA and relative availability of samples in large biobanks and cross-sectional population studies. These studies, referred to as GWAS, employ hundreds of thousands or millions of SNP genotypes (or estimates) per individual to conduct large-scale genetic association analysis usually in population-based samples. Transcriptomic and proteomic studies are challenged more than DNA studies by sample availability, technical challenges in assay design and coverage, and the potential impact of confounder variables like tissue specificity and sample quality. In particular, due to the transient platelet life span, challenges in isolation of pure cell populations, and lower transcript and protein copy numbers such studies in platelets face major challenges. Nonetheless, several studies have shown value in non-genetic “omics” approaches to uncovering novel genetic candidates in platelet function.

Platelet endothelial aggregation receptor-1 (PEAR1) was a relatively unknown human gene and receptor until it was characterised in a proteomic screen for proteins that undergo tyrosine phosphorylation upon platelet-platelet cell contact (42). Subsequent candidate gene studies consistently found PEAR1 to be associated with platelet aggregation response to multiple, diverse agonists (43;44), and in both pre- and post-aspirin treated conditions (43). In the largest study of PEAR1 to date, the combined meta-analysis of PRP aggregation GWAS results from GS and FHS, we found that peak variants in intron 1 of PEAR1 are strongly associated with both ADP- and epinephrine-induced aggregation (17). Furthermore, we found that intron 1 variants were associated with PEAR1 protein levels in platelet lysates (45). PEAR1, brought to light by proteomics, may be an important novel platelet signalling regulator, a potential drug target and its variants may play a role in platelet-related response variability in the general population, all of which require further detailed study.

Efforts at transcriptome profiling in megakaryocytes and platelets have been undertaken in relatively small sample sizes and often in clinically affected samples, thus our knowledge of transcriptome profiles in the general population is still limited ([n=60 (46); [n=126] (47); [n=29] (48); [n=4 megakaryocyte samples] (49); [n=37] (50)). Despite limited sample sizes, these studies can provide important clues supporting platelet-related genetic signals, or suggest additional candidates for further study. Based on transcript profiling of 37 samples over normal ranges of platelet response, Goodall et al. prioritised several genes for further study, finding that COMMD7 and LRRFIP1 show evidence of genetic association with MI (50). In follow-up functional studies employing gene silencing and proteomics approaches LRRFIP1 demonstrated evidence for effects on thrombus formation and interaction with the platelet cytoskeleton (50).

Notably, of 63 transcripts identified as correlated with platelet responses to ADP and/or a collagen mimetic (50), several are transcribed from loci, or have similar function to loci recently implicated in GWAS for platelet aggregation in PRP (RGS18; ST3GAL3 vs. ST3GAL4; ATP6V0D2 vs. ATP6V1F from (17)) and in GWAS for MPV (DNM3 (51)). Targeted functional reports (e.g., RTPCR) also provide valuable information. In published MPV and PRP GWAS studies the integration of megakaryocyte transcriptome data (49) as well as targeted functional studies on particular genes shows some loci contain transcripts or proteins with known significant expression and/or function in platelets and/or megakaryocytes (17;51); see evidence codes “(R)NA”, “(P)rotein/roteomics”, “(S)ignaling” in Tables 3 and 4. These examples demonstrate that functional genomics screening experiments can provide valuable information to guide new genetic studies or aid in the interpretation of existing study results.

Table 3.

Genome-wide association (GWAS) results for platelet traits with consistency in two or more human population samples and association with 2 or more distinct traits in either the same or different studies.

Gene* Chr region Phenotype
association
Association P-
val
Pop. # in
PM
# in
PM
(PLT)
Further evidence
CYP2C19 10q23.33 Clopidogrel response and outcomes(29) 1.5e-13 EA, AA, AS, H >500 192 DM, FD, HS
PEAR1 1q23.1 PRP Epi, ADP(17), Aspirin PRP response(43), WB coll.(44) 4.9e-19, 3.8e-16, 3.8e-4, 2.6e-4 EA, AA 2 2 P(42;45)
PIK3CG 7q22.3 PRP Epi(17), MPV(51;56;56) 3.1e-9, 1.6e-33 EA 210 13 AM(59;60),R(56),S(59;60)
JMJD1C 10q21.2 PRP Epi(17), MPV(51), PRP ADP(54) 1.6e-8, 3.3e-21, p<0.05 EA, AA 10 0 MK(65)
MRVI1 11p15.4 PRP ADP, Epi(17) 2.0e-8, 7.6e-7 EA, AA 16 2 AM(58),R(89),S(58)
RGS18 1q31.2 PRP Epi, ADP(17) 6.8e-7, 4.0e-5 EA, AA 19 6 R(90–92),P(93), S(93)
TAOK1 17q11.2 MPV(51;55), PRP ADP(17) 1.4e-22, 6.9e-5 EA 3 0
P2RY12 3q25.1 PRP ADP, Epi(17) 8.2e-6, 9.5e-4 EA 453 395 AM, FD, HS, R, P, S
GP1Bα 17p13.2 PLT(34) 2.1e-12 AS 25 16 AM, FD, HS, R, P, S
PRP Epi(17) 0.03 EA
PIP5K1B 9q21.11 PRP ADP, Epi(17) 2.2e-7, 8.0e-6 EA 5 0
HMGB1L1 20q13.31 PRP ADP, Epi(17) 1.1e-6, 5.2e-6 EA 0 0
SETBP1 18q12.3 PRP Epi, ADP(17) 1.3e-6, 5.4e-6 EA 8 0
FBXL7 5p15.1 PRP Epi, ADP(17) 2.9e-6, 2.3e-5 EA 1 0
ATP6VOD2 8q21.2-q21.3 PRP Epi, ADP(17) 7.9e-6, 8.7e-6 EA 0 0
KIAA0802 18p11.22 PRP Epi, ADP(17) 9.6e-6, 7.2e-5 EA 0 0
FLJ39743 15q26.3 PRP Epi, ADP(17) 1.5e-5, 3.3e-5 EA 0 0
WBSCR17 7q11.22 PRP ADP, Epi(17) 1.7e-5, 6.3e-5 EA 3 0
CUBN 10p13 PRP ADP, Epi(17) 1.9e-5, 8.1e-5 EA 17 0
MIPOL1 14q13.3-q21.1 PRP Epi, ADP(17) 2.2e-5, 4.9e-5 EA 5 0
NUP210 3p25.1 PRP Epi, ADP(17) 4.2e-5, 8.8e-5 EA 44 0
SVIL 10p11.23 PRP Epi, ADP(17) 4.4e-5, 8.2e-5 EA, AA 17 0
THSD4 15q23 PRP ADP, Epi(17) 4.4e-5, 8.2e-5 EA 3 0 FD
STMN4 8p21.2 PRP ADP, Epi(17) 5.6e-5, 9.0e-5 EA 23 0
KLHL31 6p12.1 PRP Epi, ADP(17) 6.0e-5, 6.3e-5 EA 2 0
GMDS 6p25.3 PRP ADP, Epi(17) 6.1e-5, 8.1e-5 EA 0 0
ADAMTS2 5q35.3 PRP Epi, ADP(17) 6.8e-5, 7.1e-5 EA 24 0 FD
*

some regions contain >1 gene near the genetic signal. Either the closest gene or strongest candidate gene is given.

2 PubMed search was performed for each gene name on March 16, 2011. One search was only with gene name. The second search was with gene name AND (platelet OR megakaryocyte OR thrombosis OR hemostasis). The GWAS studies considered here are not counted among the results.

Population abbreviations are: EA=European ancestry, AA=African ancestry, AS=Asian ancestry, H=Hispanic. Further evidence abbreviations: AM=animal model, DM=drug metabolism function, FD=functional domain, HS=human study of platelet function/disease, MK=role in megakaryocyte development or function, P=protein or proteomics evidence for a role in platelets/MK, R=RNA gene expression evidence for role in platelets/MK, S=signaling role known in platelets/MK

Table 4.

Genome-wide association (GWAS) results for platelet traits with consistency in two or more human population samples with strong association with only one trait.

Gene* Chr region Phenotype
association
Association P-
val
Pop. # in
PM
# in PM
(PLT)
Further evidence
ADRA2A 10q25.2 PRP Epi(17) 3.2e-12 EA, AA >500 >500 FD, HS(94), R(95), P(96)
NEURL 10q24.33 PRP Epi(17) 2.5e-7 EA, AA 10 1
PRNP 20p13 PRP Epi(17) 1.7e-6 EA, AA >500 1 HS(97), P(98)
TRIM27 6p22.1 PRP Epi(17) 2.4e-6 EA, AA 54 0
SGCZ 8p22 PRP Epi(17) 4.5e-6 EA, AA 2 0
ST3GAL4 11q24.2 PRP Epi(17) 3.0e-6 EA, AA 5 1 AM(61;63), FD(62), HS(61)
LPAR1 9q31.3 PRP Epi(54) 5.8e-3 EA 11 0 FD, HS(99)
SHH 7q36.3 PRP ADP(17) 4.5e-8 EA, AA >500 34 MK(100)
MST151 10p13 PRP ADP(17) 6.7e-7 EA, AA 0 0
KCNQ1 11p15.5-p15.4 PRP ADP(17) 5.9e-6 EA, AA 200 0
RAPGEF2 4q32.1 PRP ADP(17) 9.1e-7 EA, AA 19 6 S(101;102)
JAK2 9p24.1 WB ADP(44), PRP ADP(54) 6.0e-4, p<0.05 EA >500 499 AM, FD, HS, R, P, S
COMMD7 20q11.21 WB ADP(50), PRP ADP(54) 1.2e-3, p<0.05 EA 0 0 AM, R(50)
GP6 19q13.42 PRP collagen lag time(17) 8.4e-14 EA, AA 237 17 FD, R(103), P(104)
PTPRD 9p24.1 PRP collagen lag time(17) 1.2e-7 EA, AA 200 3
HSD17B6 12q13.3 PRP collagen lag time(17) 1.1e-6 EA, AA 6 0
FCER1G 1q23.3 WB coll.(44), PRP coll. lag(17) 9.6e-6, 1.6e-5 EA 12 0
UGT1A10 2q37.1 PRP collagen lag time(17) 1.2e-5 EA, AA 126 0
RAP1B 12q15 PRP collagen lag time(17) 1.5e-5 EA, AA 150 52 AM(64), R(105;106), S(107), P(108)
MYO5B 18q21.1 PRP collagen(54) 3.0e-5 EA 34 0
WDR66 12q24.31 MPV(51;55) 2.7e-44 EA 0 0
ARHGEF3 3p21-p13 MPV(51;55) 5.5e-31 EA 6 0 R(109)
TMCC2 1q32.1 MPV(51) 1.4e-20 EA 1 0
BET1L 11p15.5 MPV(51) 1.3e-14 EA 10 0
DNM3 1q24.3 MPV(51) 2.1e-14 EA 6 1 MK(110), R(50)
EHD3 2p21 MPV(51) 3.2e-11 EA 21 0 R(51)
SIRPA 20p13 MPV(51) 7.7e-11 EA 193 2
CD226 18q22.3 MPV(51) 1.4e-10 EA 113 0 MK(111)
TPM1 15q22.1 MPV(51) 1.9e-8 EA 158 1 MK(112), R(51;51;112)
AK3 9p24.1-p24.3 PLT(51) 8.5e-17 EA 51 0
ATXN2 12q24 PLT(51) 2.2e-13 EA 23 0 R(51)
PTPN11 12q24 PLT(51) 7.7e-12 EA 200 108 HS(113), R(51), S(113;114)
THPO 3q27.1 PLT(34) 5.4e-11 AS 94 12 MK(115), FD, HS(33;116)
BAK1 6p21.3 PLT(51) 3.7e-10 EA >500 10
NRG3 10q23.1 PLT(54) 3.6e-5 EA 38 0
*

some regions contain >1 gene near the genetic signal. Either the closest gene or strongest candidate gene is given.

2 PubMed search was performed for each gene name on March 16, 2011. One search was only with gene name. The second search was with gene name AND (platelet OR megakaryocyte OR thrombosis OR hemostasis). The GWAS studies considered here are not counted among the results.

Population abbreviations are: EA=European ancestry, AA=African ancestry, AS=Asian ancestry. Further evidence abbreviations: AM=animal model, FD=functional domain, HS=human study of platelet function/disease, MK=role in megakaryocyte development or function, P=protein or proteomics evidence for a role in platelets/MK, R=RNA gene expression evidence for role in platelets/MK, S=signaling role known in platelets/MK

Human GWAS of platelet traits

A small number of human GWAS studies have been conducted for platelet traits (17;34;36;5157). The major findings from these studies with consistency in 2 or more population samples are summarised in Tables 3 and 4, along with supporting functional information and citations (e.g., transcriptome, proteome, platelet signalling pathways, animal model results, human disease associations). Table 3 contains loci with evidence for 2 or more platelet phenotypes. Table 4 contains loci mainly associated with a single platelet phenotype. Specific SNP results are not given in Tables 3 and 4 as these can vary across studies due to differences in SNP coverage, population and linkage disequilibrium. Readers interested in further SNP-specific details underlying results in Tables 3 and 4 are referred to Supplemental Table 1 (where >450 significant SNP associations and related SNP identifiers (rsIDs) and mappings are given with reference to the cited GWAS studies).

The first platelet-related GWAS was a moderate density 100,000 SNP scan in FHS which displayed modest findings for PRP aggregation traits with no replication attempted (n≤724, (57)). This study is largely super-ceded by a higher density 550,000 SNP scan in FHS in a larger sample (n≤2,753) with imputation to 2.33 million variants which was further combined in a meta-analysis for similar agonists with imputation-based results from the GS study (n≤2,075) (17). This study represents the only large GWAS study of platelet function, and identified regions strongly associated with PRP ADP response (PEAR1, MRVI1, SHH), PRP epinephrine response (ADRA2A, PEAR1, PIK3CG, JMJD1C) and collagen lag time (GP6). Discovery was conducted in 2 European ancestry samples with replication in 1 African ancestry sample. Thus, this study represents the first platelet-related GWAS study to compare results across ancestry groups. Notably, there is general consistency in signals and direction of effects among many of the loci in both European and African ancestry samples suggesting that most of the platelet function loci may be shared across ancestries (17).

We identified additional loci with consistent associations in 2 or more populations with replication evidence in African ancestry samples but which did not reach genome-significance thresholds. Several loci were associated with platelet responses to 2 or more agonists (Table 3) with the alleles having the same direction of effect in each case, including loci near or containing ADAMTS2, ATP6VOD2, CTCFL, CUBN, FBXL7, FLJ39743, GMDS, HMG1L1, KIAA0802, KLHL31, MIPOL1, MRVI1, NUP210, PEAR1, PCK1, PIP5K1B, PSKH2, RGS18, SETBP1, STMN4, SVIL, THSD4, and WBSCR17. Overall these results suggest many loci contribute to platelet function, with most contributing to a small proportion of the total population variance.

Many of the identified loci contain genes that were not previously studied in relation to platelet biology (see PubMed search column results for each gene in Tables 3 and 4), indicating the value of GWAS in uncovering novel targets for investigation. Other loci from this study are notable for past links to platelet function including the prion protein PRNP, small G-protein related platelet genes including RAP1B, RAPGEF2, RGS18, and PIK3CG, or for their role in animal models for thrombosis, bleeding or healing (MRVI1(58), PIK3CG (59;60), ST3GAL4 (6163), RAP1B (6164)), or megakaryocyte development or function (JMJD1C (65)). The GS cohort published results separately from GWAS of pre- and post-aspirin platelet responses in whole blood and PRP in European ancestry and African ancestry samples (36). These results suggest several potential loci of interest (e.g., the endopeptidase MME) but await attempts at replication.

Larger GWAS sample sizes have been achievable for PLT and MPV due to the easier nature of collecting these measures. The first published study conducted GWAS on 335,152 SNPs in 1,606 individuals with MPV measurements, and follow-up replication in up to 8,443 individuals, finding and replicating 3 loci (ARHGEF3, TAOK1, WDR66) that account for ~4–5% of the variance in MPV (55). By further investigation of WDR66 within previously collected microarray results for leukocyte samples, WDR66 transcript levels were inversely correlated with MPV suggesting a functional link. Soranzo et al. followed soon with a MPV GWAS in samples that were used in the replication phase of Meisinger et al., discovering and replicating an additional MPV locus containing PIK3CG, and providing evidence that this locus further influences platelet responses to a collagen mimetic as assessed by annexin V binding (56). Consultation with microarray data suggested that PIK3CG and another nearby transcript are expressed in megakaryocytes and platelets.

In late 2009 a meta-analysis of MPV and PLT GWAS with up to 4,627 discovery samples and imputation to 2.1 million SNPs and replication in up to 9,316 samples was published (51). This was the first study to conduct GWAS of PLT or to report a platelet-related GWAS that employed SNP imputation to increase genomic coverage. The authors replicated 12 loci for MPV, which included the 4 prior loci, and additionally found and replicated 4 novel loci for PLT (summarised in Tables 3 and 4). Nine of the 12 MPV loci were also associated with PLT; in each case the MPV-raising alleles were associated with an inverse PLT-lowering effect consistent with the inverse correlation of PLT and MPV.

An independent GWAS on up to 6,015 individuals for blood cell traits with imputation to 2.1 million variants found the HBS1L locus was strongly associated with PLT (52). Platelet count was the only platelet trait analysed and no PLT SNPs were carried forward to the replication stage. The PLT association with HBS1L is consistent with a prior candidate gene study in twins (66), and the strong pleiotropy of this locus including effects on PLT was also observed in prior GWAS (51;53). Given strong association of HBS1L with several blood counts it may not be highly specific to platelet function. Ferreira et al. identified several additional suggestive PLT loci in univariate and bivariate analyses including ARHGEF3, PHACTR1, and CUBN (52). The CUBN locus is notable since it was subsequently associated with both ADP and epinephrine PRP responses (17) and recently with albuminuria (67). The PHACTR1 locus is notable since it is one of the published MI GWAS regions (68).

Kamatani and colleagues conducted GWAS of blood measures including PLT in 14,806 Japanese individuals finding support for prior loci (HBS1L, SH2B3, BAK1, RCL1) and additionally noting strong associations at 3q27.1 (THPO) and 17p13.2 (GP1Bα) (34). The THPO locus encodes thrombopoietin, a known growth factor for MK cells, with the identified GWAS SNP (rs6141) near the translation stop site in the 3’UTR. This region and specific variant were also identified in a prior linkage scan for PLT in Asian Indian families (33). The peak SNP at GP1Bα (rs6065) encodes a Thr161Met change previously studied as a candidate functional SNP, which was also modestly associated in a GWAS of epinephrine-induced aggregation in PRP (17). Finally, a recent GWAS measured a range of >20 platelet traits in a cohort of children (n=75) with limited replication attempted (54). Despite a sample size that limited statistical power, they discovered and replicated 3 new loci (LPAR1 for PRP Epi, MYO5B for PRP collagen, NRG3 for PLT), as well as providing support for several prior loci.

Platelet gene stories new and old

In the dawning “omics” era of platelet studies we remarkably find that some of the strongest candidates of prior work now re-emerge among the strongest signals of “omics” work (e.g., ADRA2A, GP6, CYP2C19). A number of prior candidate genes also re-emerge with weaker, yet supportive evidence in a recent PRP platelet function GWAS (e.g., P2RY12, GP1bα) (17). With more than 50 loci for platelet traits identified, and more than 50 additional PLT/MPV loci promised soon (69), we have considerably advanced in identifying novel candidates for platelet function, as well as ear-marking others that have some prior evidence but have not been the focus of in depth genetic or functional studies. Some have criticised GWAS as not identifying functional variants, but there is evidence that several of the strongest variants from GWAS may be functional (e.g., GP6, THPO, GP1bα). Synergy between RNA, protein, animal model, candidate gene and GWAS studies is emerging to varying degrees to identify the likely causal genes within loci and in some cases to identify the potential functional variants.

Among the newly highlighted loci are suggested functional receptors (PEAR1, ST3GAL4, PRNP), potential regulators of cytoskeleton dynamics and platelet morphology (DNM3, TPM1, ARHGEF3), MK development (JMJD1C), and intracellular platelet signalling (PIK3CG, MRVI1 (also known as IRAG), RGS18, RAPGEF2, RAP1B, PTPN11). These candidates propel us beyond a focus on cell surface receptors and their ligands and invite new functional platelet and megakaryocyte studies even without respect to specific alleles. It remains to be seen if any of these loci provide new therapeutic targets, or contribute to a deeper understanding of common or rare thrombosis or haemostasis conditions.

Few of the loci discovered to date have genetic alleles with strongly validated roles in human disease. One exception is GP6, which has been independently associated with venous thrombosis in 2 separate studies (70;71). Notably, the peak GP6 SNP from the VT GWAS and a PRP collagen lag GWAS are the same nonsynonymous SNP which was previously characterised for functional effects. Another exception, CYP2C19, is associated with outcomes in anti-platelet treatment trials (29;7274), though with conflicting results in another set of trials, which could be due to demographic variation among the clinical populations (75). Beyond CYP2C19 there is no other compelling locus associated with response to aspirin or other anti-platelet therapies at this time (13;76), though ABCB1 may play a role (73;74). Finally, an extremely large linkage disequilibrium block on chromosome 12q24 has been linked to multiple phenotypes including PLT, blood pressure and coronary artery disease but the functional aetiology and causal gene(s) remains to be worked out (51).

Although most known platelet loci have not been convincingly linked to a platelet-related human condition, evidence from animal models does link some of them to thrombosis or bleeding effects (e.g., GP6, ST3GAL4, MRVI1, PIK3CG, ADRA2A, P2RY12, RAP1B). Human disease studies with larger samples and a deeper examination of results with moderate effect sizes may eventually reveal additional platelet-related genes that make modest contributions to disease risk or treatment.

Limitations of current findings and future directions

Current genomic findings are limited both by the constraints of the phenotypes available, the ability to ascertain large populations, and by our current understanding and assessment of human population variation. While variation maps have improved substantially, we are still beginning to survey alleles at the low allele frequency end of the spectrum. Future directions of research include resequencing of platelet loci for additional rare variants that may have larger effect sizes and potential clinical relevance (e.g., in affected families with plateletopathies), functional experiments aimed at uncovering the mechanisms underlying genetic associations, and further study of platelet-related variants in relevant clinical samples which provide adequate statistical power.

While the validation, functional and clinical study of individual variants remains important, locus level views of overlapping trait scans suggests extensive pleiotropy for some platelet-related regions (e.g., ABO, SH2B3, HBS1L). With a growing number of published GWAS and examples of pleiotropy this is an active area of future development since pleiotropic signals may provide important clues to the functions of genes in the associated loci (31;77). As more data become available data mining, pathway analysis and other bioinformatics approaches may also prove useful to characterising important loci for platelet function. We previously assembled a catalogue of results from 118 published GWAS demonstrating the potential to discover novel suggestive patterns (31).

Using a similar, updated catalogue of results from >700 GWAS (unpublished results) Supplemental Table 1 was created with >450 significant associations related to platelet traits from all available GWAS results. This provides a quick overview of loci consistent across studies (e.g., HBS1L) but could also suggest loci to prioritise for further study. For example, RAPGEF2 is seen to be associated with PRP response to ADP in (17) and clopidogrel response assessed by ADP in (29), though this locus has not yet been the focus of replication studies for clopidogrel response. Another potential example is observed when PRP aggregation GWAS, venous thrombosis GWAS and platelet gene expression results are considered. Three distinct loci that encode subunits of a V-type proton ATPase were highlighted: for PRP aggregation associations (ATP6V0D2, 8q21.2-q21.3, (17)), gene expression (ATP6V1F, 7q23.1, (50)), and in a GWAS follow-up study on venous thrombosis (ATP6V1B2, 8p21.2, (78)). These loci were not among the strongest in any of these studies individually. Notably, they are all subunits of a single transporter that has received little study. Evidence indicates this vacuolar ATPase is involved in bone homeostasis (79) and thus it may influence megakaryopoeisis or other platelet-related functions. Additional examples of such hypotheses are likely to emerge as further “omics” studies are published for platelet and haemostasis related traits.

It is important to note that the loci identified thus far which contain common variation contributing to variability in platelet traits individually account for a small proportion of the population variance. Large, well-designed prospective trials of variants are needed to determine their potential relevance in clinical practice. While CYP2C19 is of potential clinical significance, a position statement does not support routine genotyping or platelet function testing in practice (80).Thus, at this time none of the specific markers highlighted here should be considered to have clear and direct utility in clinical practice.

Supplementary Material

Supp Table S1

Acknowledgments

A. Johnson was supported by a National Institutes of Health Intramural Research Training Fellowship.

Footnotes

Addendum

A. Johnson drafted the manuscript and is solely responsible for its content.

References

  • 1.Salles II, Feys HB, Iserbyt BF, De Meyer SF, Vanhoorelbeke K, Deckmyn H. Inherited traits affecting platelet function. Blood Rev. 2008;22:155–172. doi: 10.1016/j.blre.2007.11.002. [DOI] [PubMed] [Google Scholar]
  • 2.Nurden P, Nurden AT. Congenital disorders associated with platelet dysfunctions. Thromb Haemost. 2008;99:253–263. doi: 10.1160/TH07-09-0568. [DOI] [PubMed] [Google Scholar]
  • 3.Yang A, Pizzulli L, Luderitz B. Mean platelet volume as marker of restenosis after percutaneous transluminal coronary angioplasty in patients with stable and unstable angina pectoris. Thromb Res. 2006;117:371–377. doi: 10.1016/j.thromres.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 4.Mayda-Domac F, Misirli H, Yilmaz M. Prognostic role of mean platelet volume and platelet count in ischemic and hemorrhagic stroke. J Stroke Cerebrovasc Dis. 2010;19:66–72. doi: 10.1016/j.jstrokecerebrovasdis.2009.03.003. [DOI] [PubMed] [Google Scholar]
  • 5.Martin JF, Bath PM, Burr ML. Influence of platelet size on outcome after myocardial infarction. Lancet. 1991;338:1409–1411. doi: 10.1016/0140-6736(91)92719-i. [DOI] [PubMed] [Google Scholar]
  • 6.Klovaite J, Benn M, Yazdanyar S, Nordestgaard BG. High platelet volume and increased risk of myocardial infarction: 39,531 participants from the general population. J Thromb Haemost. 2011;9:49–56. doi: 10.1111/j.1538-7836.2010.04110.x. [DOI] [PubMed] [Google Scholar]
  • 7.Huczek Z, Kochman J, Filipiak KJ, Horszczaruk GJ, Grabowski M, Piatkowski R, et al. Mean platelet volume on admission predicts impaired reperfusion and long-term mortality in acute myocardial infarction treated with primary percutaneous coronary intervention. J Am Coll Cardiol. 2005;46:284–290. doi: 10.1016/j.jacc.2005.03.065. [DOI] [PubMed] [Google Scholar]
  • 8.Endler G, Klimesch A, Sunder-Plassmann H, Schillinger M, Exner M, Mannhalter C, et al. Mean platelet volume is an independent risk factor for myocardial infarction but not for coronary artery disease. Br J Haematol. 2002;117:399–404. doi: 10.1046/j.1365-2141.2002.03441.x. [DOI] [PubMed] [Google Scholar]
  • 9.De Luca G, Santagostino M, Secco GG, Cassetti E, Giuliani L, Franchi E, et al. Mean platelet volume and the extent of coronary artery disease: results from a large prospective study. Atherosclerosis. 2009;206:292–297. doi: 10.1016/j.atherosclerosis.2009.02.008. [DOI] [PubMed] [Google Scholar]
  • 10.Chu SG, Becker RC, Berger PB, Bhatt DL, Eikelboom JW, Konkle B, et al. Mean platelet volume as a predictor of cardiovascular risk: a systematic review and meta-analysis. J Thromb Haemost. 2010;8:148–156. doi: 10.1111/j.1538-7836.2009.03584.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Breet NJ, van Werkum JW, Bouman HJ, Kelder JC, Ruven HJ, Bal ET, et al. Comparison of platelet function tests in predicting clinical outcome in patients undergoing coronary stent implantation. JAMA. 2010;303:754–762. doi: 10.1001/jama.2010.181. [DOI] [PubMed] [Google Scholar]
  • 12.Bath P, Algert C, Chapman N, Neal B. Association of mean platelet volume with risk of stroke among 3134 individuals with history of cerebrovascular disease. Stroke. 2004;35:622–626. doi: 10.1161/01.STR.0000116105.26237.EC. [DOI] [PubMed] [Google Scholar]
  • 13.Feher G, Feher A, Pusch G, Lupkovics G, Szapary L, Papp E. The genetics of antiplatelet drug resistance. Clin Genet. 2009;75:1–18. doi: 10.1111/j.1399-0004.2008.01105.x. [DOI] [PubMed] [Google Scholar]
  • 14.Faraday N, Becker DM, Becker LC. Pharmacogenomics of platelet responsiveness to aspirin. Pharmacogenomics. 2007;8:1413–1425. doi: 10.2217/14622416.8.10.1413. [DOI] [PubMed] [Google Scholar]
  • 15.Harrison P, Keeling D. Platelets. 2nd ed. Burlington, MA: Academic Press; 2007. Clinical Tests of Platelet Function; pp. 445–474. [Google Scholar]
  • 16.Briggs R, Harrison P, Machin S. Platelets. 2nd ed. Burlington, MA: Academic Press; 2007. Platelet Counting; pp. 475–483. [Google Scholar]
  • 17.Johnson AD, Yanek LR, Chen MH, Faraday N, Larson MG, Tofler G, et al. Genome-wide meta-analyses identifies seven loci associated with platelet aggregation in response to agonists. Nat Genet. 2010;42:608–613. doi: 10.1038/ng.604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Whitfield JB, Martin NG. Genetic and environmental influences on the size and number of cells in the blood. Genet Epidemiol. 1985;2:133–144. doi: 10.1002/gepi.1370020204. [DOI] [PubMed] [Google Scholar]
  • 19.Gaxiola B, Friedl W, Propping P. Epinephrine-induced platelet aggregation. A twin study. Clin Genet. 1984;26:543–548. doi: 10.1111/j.1399-0004.1984.tb01101.x. [DOI] [PubMed] [Google Scholar]
  • 20.O'Donnell CJ, Larson MG, Feng D, Sutherland PA, Lindpaintner K, Myers RH, et al. Genetic and environmental contributions to platelet aggregation: the Framingham heart study. Circulation. 2001;103:3051–3056. doi: 10.1161/01.cir.103.25.3051. [DOI] [PubMed] [Google Scholar]
  • 21.Bray PF, Mathias RA, Faraday N, Yanek LR, Fallin MD, Herrera-Galeano JE, et al. Heritability of platelet function in families with premature coronary artery disease. J Thromb Haemost. 2007;5:1617–1623. doi: 10.1111/j.1538-7836.2007.02618.x. [DOI] [PubMed] [Google Scholar]
  • 22.Faraday N, Yanek LR, Mathias R, Herrera-Galeano JE, Vaidya D, Moy TF, et al. Heritability of platelet responsiveness to aspirin in activation pathways directly and indirectly related to cyclooxygenase-1. Circulation. 2007;115:2490–2496. doi: 10.1161/CIRCULATIONAHA.106.667584. [DOI] [PubMed] [Google Scholar]
  • 23.Bertin A, Mahaney MC, Cox LA, Rogers J, VandeBerg JL, Brugnara C, et al. Quantitative trait loci for peripheral blood cell counts: a study in baboons. Mamm Genome. 2007;18:361–372. doi: 10.1007/s00335-007-9022-8. [DOI] [PubMed] [Google Scholar]
  • 24.Cheung CC, Martin IC, Zenger KR, Donald JA, Thomson PC, Moran C, et al. Quantitative trait loci for steady-state platelet count in mice. Mamm Genome. 2004;15:784–797. doi: 10.1007/s00335-004-2408-y. [DOI] [PubMed] [Google Scholar]
  • 25.Peters LL, Zhang W, Lambert AJ, Brugnara C, Churchill GA, Platt OS. Quantitative trait loci for baseline white blood cell count, platelet count, and mean platelet volume. Mamm Genome. 2005;16:749–763. doi: 10.1007/s00335-005-0063-6. [DOI] [PubMed] [Google Scholar]
  • 26.Afshar-Kharghan V, Vijayan K, Bray P. Platelets. 2nd ed. Burlington, MA: Academic Press; 2007. Platelet Polymorphisms; pp. 281–307. [Google Scholar]
  • 27.Kunicki TJ, Nugent DJ. The genetics of normal platelet reactivity. Blood. 2010;116:2627–2634. doi: 10.1182/blood-2010-04-262048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Williams MS, Weiss EJ, Sabatine MS, Simon DI, Bahou WF, Becker LC, et al. Genetic regulation of platelet receptor expression and function: application in clinical practice and drug development. Arterioscler Thromb Vasc Biol. 2010;30:2372–2384. doi: 10.1161/ATVBAHA.110.218131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Shuldiner AR, O'Connell JR, Bliden KP, Gandhi A, Ryan K, Horenstein RB, et al. Association of cytochrome P450 2C19 genotype with the antiplatelet effect and clinical efficacy of clopidogrel therapy. JAMA. 2009;302:849–857. doi: 10.1001/jama.2009.1232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Johnson A, Newton-Cheh C, Chasman D, Ehret G, Johnson T, Rose L, et al. Association of Hypertension Drug Target Genes With Blood Pressure and Hypertension in 86,588 Individuals. Hypertension. 2011;57:903–910. doi: 10.1161/HYPERTENSIONAHA.110.158667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Johnson AD, O'Donnell CJ. An open access database of genome-wide association results. BMC Med Genet. 2009;10:6. doi: 10.1186/1471-2350-10-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Siontis KC, Patsopoulos NA, Ioannidis JP. Replication of past candidate loci for common diseases and phenotypes in 100 genome-wide association studies. Eur J Hum Genet. 2010;18:832–837. doi: 10.1038/ejhg.2010.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Garner C, Best S, Menzel S, Rooks H, Spector TD, Thein SL. Two candidate genes for low platelet count identified in an Asian Indian kindred by genome-wide linkage analysis: glycoprotein IX and thrombopoietin. Eur J Hum Genet. 2006;14:101–108. doi: 10.1038/sj.ejhg.5201499. [DOI] [PubMed] [Google Scholar]
  • 34.Kamatani Y, Matsuda K, Okada Y, Kubo M, Hosono N, Daigo Y, et al. Genome-wide association study of hematological and biochemical traits in a Japanese population. Nat Genet. 2010;42:210–215. doi: 10.1038/ng.531. [DOI] [PubMed] [Google Scholar]
  • 35.Evans DM, Zhu G, Duffy DL, Montgomery GW, Frazer IH, Martin NG. Multivariate QTL linkage analysis suggests a QTL for platelet count on chromosome 19q. Eur J Hum Genet. 2004;12:835–842. doi: 10.1038/sj.ejhg.5201248. [DOI] [PubMed] [Google Scholar]
  • 36.Mathias RA, Kim Y, Sung H, Yanek LR, Mantese VJ, Hererra-Galeano JE, et al. A combined genome-wide linkage and association approach to find susceptibility loci for platelet function phenotypes in European American and African American families with coronary artery disease. BMC Med Genomics. 2010;3:22. doi: 10.1186/1755-8794-3-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cho YU, Jang S, Park CJ, Chi HS. Variables that affect platelet function analyzer-100 (PFA-100) closure times and establishment of reference intervals in Korean adults. Ann Clin Lab Sci. 2008;38:247–253. [PubMed] [Google Scholar]
  • 38.Feuring M, Ruf A, Schultz A, Wehling M. The PFA-100R cannot detect blood group-dependent inhibition of platelet function by eptifibatide or abciximab at therapeutic plasma concentrations. Platelets. 2010;21:176–182. doi: 10.3109/09537100903518260. [DOI] [PubMed] [Google Scholar]
  • 39.Sweeney JD, Labuzetta JW, Hoernig LA, Fitzpatrick JE. Platelet function and ABO blood group. Am J Clin Pathol. 1989;91:79–81. doi: 10.1093/ajcp/91.1.79. [DOI] [PubMed] [Google Scholar]
  • 40.Barbalic M, Dupuis J, Dehghan A, Bis JC, Hoogeveen RC, Schnabel RB, et al. Large-scale genomic studies reveal central role of ABO in sP-selectin and sICAM-1 levels. Hum Mol Genet. 2010;19:1863–1872. doi: 10.1093/hmg/ddq061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Shehata N, Tinmouth A, Naglie G, Freedman J, Wilson K. ABO-identical versus nonidentical platelet transfusion: a systematic review. Transfusion. 2009;49:2442–2453. doi: 10.1111/j.1537-2995.2009.02273.x. [DOI] [PubMed] [Google Scholar]
  • 42.Nanda N, Bao M, Lin H, Clauser K, Komuves L, Quertermous T, et al. Platelet endothelial aggregation receptor 1 (PEAR1), a novel epidermal growth factor repeat-containing transmembrane receptor, participates in platelet contact-induced activation. J Biol Chem. 2005;280:24680–24689. doi: 10.1074/jbc.M413411200. [DOI] [PubMed] [Google Scholar]
  • 43.Herrera-Galeano JE, Becker DM, Wilson AF, Yanek LR, Bray P, Vaidya D, et al. A novel variant in the platelet endothelial aggregation receptor-1 gene is associated with increased platelet aggregability. Arterioscler Thromb Vasc Biol. 2008;28:1484–1490. doi: 10.1161/ATVBAHA.108.168971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jones CI, Bray S, Garner SF, Stephens J, de BB, Angenent WG, et al. A functional genomics approach reveals novel quantitative trait loci associated with platelet signaling pathways. Blood. 2009;114:1405–1416. doi: 10.1182/blood-2009-02-202614. [DOI] [PubMed] [Google Scholar]
  • 45.Becker L, Herrera J, Yanek L, Yang X, Johnson A, Chen M-H, et al. A variant in Intron 1 of the platelet endothelial aggregation receptor-1 (PEAR1) gene is strongly associated with increased platelet aggregability. Circulation [120] 2009:S599. RefType: Abstract. [Google Scholar]
  • 46.Healy AM, Pickard MD, Pradhan AD, Wang Y, Chen Z, Croce K, et al. Platelet expression profiling and clinical validation of myeloid-related protein-14 as a novel determinant of cardiovascular events. Circulation. 2006;113:2278–2284. doi: 10.1161/CIRCULATIONAHA.105.607333. [DOI] [PubMed] [Google Scholar]
  • 47.Gnatenko DV, Zhu W, Xu X, Samuel ET, Monaghan M, Zarrabi MH, et al. Class prediction models of thrombocytosis using genetic biomarkers. Blood. 2010;115:7–14. doi: 10.1182/blood-2009-05-224477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kondkar AA, Bray MS, Leal SM, Nagalla S, Liu DJ, Jin Y, et al. VAMP8/endobrevin is overexpressed in hyperreactive human platelets: suggested role for platelet microRNA. J Thromb Haemost. 2010;8:369–378. doi: 10.1111/j.1538-7836.2009.03700.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Watkins NA, Gusnanto A, de BB, De S, Miranda-Saavedra D, Hardie DL, et al. A HaemAtlas: characterizing gene expression in differentiated human blood cells. Blood. 2009;113:e1–e9. doi: 10.1182/blood-2008-06-162958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Goodall AH, Burns P, Salles I, Macaulay IC, Jones CI, Ardissino D, et al. Transcription profiling in human platelets reveals LRRFIP1 as a novel protein regulating platelet function. Blood. 2010;116:4646–4656. doi: 10.1182/blood-2010-04-280925. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Soranzo N, Spector TD, Mangino M, Kuhnel B, Rendon A, Teumer A, et al. A genome-wide meta-analysis identifies 22 loci associated with eight hematological parameters in the HaemGen consortium. Nat Genet. 2009;41:1182–1190. doi: 10.1038/ng.467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ferreira MA, Hottenga JJ, Warrington NM, Medland SE, Willemsen G, Lawrence RW, et al. Sequence variants in three loci influence monocyte counts and erythrocyte volume. Am J Hum Genet. 2009;85:745–749. doi: 10.1016/j.ajhg.2009.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gudbjartsson DF, Bjornsdottir US, Halapi E, Helgadottir A, Sulem P, Jonsdottir GM, et al. Sequence variants affecting eosinophil numbers associate with asthma and myocardial infarction. Nat Genet. 2009;41:342–347. doi: 10.1038/ng.323. [DOI] [PubMed] [Google Scholar]
  • 54.Guerrero JA, Rivera J, Quiroga T, Martinez-Perez A, Anton-Garcia AI, Martinez C, et al. Novel loci involved on platelet function and platelet count identified by a genome-wide study performed in children. Haematologica. doi: 10.3324/haematol.2011.042077. (published May 5, 2011) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Meisinger C, Prokisch H, Gieger C, Soranzo N, Mehta D, Rosskopf D, et al. A genome-wide association study identifies three loci associated with mean platelet volume. Am J Hum Genet. 2009;84:66–71. doi: 10.1016/j.ajhg.2008.11.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Soranzo N, Rendon A, Gieger C, Jones CI, Watkins NA, Menzel S, et al. A novel variant on chromosome 7q22.3 associated with mean platelet volume, counts, and function. Blood. 2009;113:3831–3837. doi: 10.1182/blood-2008-10-184234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Yang Q, Kathiresan S, Lin JP, Tofler GH, O'Donnell CJ. Genome-wide association and linkage analyses of hemostatic factors and hematological phenotypes in the Framingham Heart Study. BMC Med Genet. 2007;8 Suppl 1:S12. doi: 10.1186/1471-2350-8-S1-S12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Antl M, von Bruhl ML, Eiglsperger C, Werner M, Konrad I, Kocher T, et al. IRAG mediates NO/cGMP-dependent inhibition of platelet aggregation and thrombus formation. Blood. 2007;7:552–559. doi: 10.1182/blood-2005-10-026294. [DOI] [PubMed] [Google Scholar]
  • 59.Schoenwaelder SM, Ono A, Sturgeon S, Chan SM, Mangin P, Maxwell MJ, et al. Identification of a unique co-operative phosphoinositide 3-kinase signaling mechanism regulating integrin alpha IIb beta 3 adhesive function in platelets. J Biol Chem. 2007;4:28648–28658. doi: 10.1074/jbc.M704358200. [DOI] [PubMed] [Google Scholar]
  • 60.Zhao M, Song B, Pu J, Wada T, Reid B, Tai G, et al. Electrical signals control wound healing through phosphatidylinositol-3-OH kinase-gamma and PTEN. Nature. 2006;442:457–460. doi: 10.1038/nature04925. [DOI] [PubMed] [Google Scholar]
  • 61.Ellies LG, Ditto D, Levy GG, Wahrenbrock M, Ginsburg D, Varki A, et al. Sialyltransferase ST3Gal-IV operates as a dominant modifier of hemostasis by concealing asialoglycoprotein receptor ligands. Proc Natl Acad Sci U S A. 2002;17:10042–10047. doi: 10.1073/pnas.142005099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Grewal PK, Uchiyama S, Ditto D, Varki N, Le DT, Nizet V, et al. The Ashwell receptor mitigates the lethal coagulopathy of sepsis. Nat Med. 2008;18:648–655. doi: 10.1038/nm1760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sorensen AL, Rumjantseva V, Nayeb-Hashemi S, Clausen H, Hartwig JH, Wandall HH, et al. Role of sialic acid for platelet life span: exposure of beta-galactose results in the rapid clearance of platelets from the circulation by asialoglycoprotein receptor-expressing liver macrophages and hepatocytes. Blood. 2009;16:1645–1654. doi: 10.1182/blood-2009-01-199414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chrzanowska-Wodnicka M, Smyth SS, Schoenwaelder SM, Fischer TH, White GC. Rap1b is required for normal platelet function and hemostasis in mice. J Clin Invest. 2005;27:680–687. doi: 10.1172/JCI22973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kitajima K, Kojima M, Kondo S, Takeuchi T. A role of jumonji gene in proliferation but not differentiation of megakaryocyte lineage cells. Exp Hematol. 2001;5:507–514. doi: 10.1016/s0301-472x(00)00686-x. [DOI] [PubMed] [Google Scholar]
  • 66.Menzel S, Jiang J, Silver N, Gallagher J, Cunningham J, Surdulescu G, et al. The HBS1L-MYB intergenic region on chromosome 6q23.3 influences erythrocyte, platelet, and monocyte counts in humans. Blood. 2007;110:3624–3626. doi: 10.1182/blood-2007-05-093419. [DOI] [PubMed] [Google Scholar]
  • 67.Boger CA, Chen MH, Tin A, Olden M, Kottgen A, Deboer IH, et al. CUBN is a gene locus for albuminuria. J Am Soc Nephrol. 2011;22:555–570. doi: 10.1681/ASN.2010060598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kathiresan S, Voight BF, Purcell S, Musunuru K, Ardissino D, Mannucci PM, et al. Genome-wide association of early-onset myocardial infarction with single nucleotide polymorphisms and copy number variants. Nat Genet. 2009;41:334–341. doi: 10.1038/ng.327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Gieger C, Soranzo N for the HaemGen consortium. Genome-wide meta-analysis identifies 67 loci associated with platelet count and platelet volume in the HaemGen consortium; American Society for Human Genetics meeting; 2010. Ref Type: Abstract. [Google Scholar]
  • 70.Bezemer ID, Bare LA, Doggen CJ, Arellano AR, Tong C, Rowland CM, et al. Gene variants associated with deep vein thrombosis. JAMA. 2008;299:1306–1314. doi: 10.1001/jama.299.11.1306. [DOI] [PubMed] [Google Scholar]
  • 71.Tregouet DA, Heath S, Saut N, Biron-Andreani C, Schved JF, Pernod G, 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. doi: 10.1182/blood-2008-11-190389. [DOI] [PubMed] [Google Scholar]
  • 72.Mega JL, Simon T, Collet JP, Anderson JL, Antman EM, Bliden K, et al. Reduced-function CYP2C19 genotype and risk of adverse clinical outcomes among patients treated with clopidogrel predominantly for PCI: a meta-analysis. JAMA. 2010;304:1821–1830. doi: 10.1001/jama.2010.1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Mega JL, Close SL, Wiviott SD, Shen L, Walker JR, Simon T, et al. Genetic variants in ABCB1 and CYP2C19 and cardiovascular outcomes after treatment with clopidogrel and prasugrel in the TRITON-TIMI 38 trial: a pharmacogenetic analysis. Lancet. 2010;376:1312–1319. doi: 10.1016/S0140-6736(10)61273-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wallentin L, James S, Storey RF, Armstrong M, Barratt BJ, Horrow J, et al. Effect of CYP2C19 and ABCB1 single nucleotide polymorphisms on outcomes of treatment with ticagrelor versus clopidogrel for acute coronary syndromes: a genetic substudy of the PLATO trial. Lancet. 2010;376:1320–1328. doi: 10.1016/S0140-6736(10)61274-3. [DOI] [PubMed] [Google Scholar]
  • 75.Pare G, Mehta SR, Yusuf S, Anand SS, Connolly SJ, Hirsh J, et al. Effects of CYP2C19 genotype on outcomes of clopidogrel treatment. N Engl J Med. 2010;363:1704–1714. doi: 10.1056/NEJMoa1008410. [DOI] [PubMed] [Google Scholar]
  • 76.Goodman T, Ferro A, Sharma P. Pharmacogenetics of aspirin resistance: a comprehensive systematic review. Br J Clin Pharmacol. 2008;66:222–232. doi: 10.1111/j.1365-2125.2008.03183.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Huang J, Johnson AD, O'Donnell CJ. PRIMe: a method for characterization and evaluation of pleiotropic regions from multiple genome-wide association studies. Bioinformatics. 2011;27:1201–1206. doi: 10.1093/bioinformatics/btr116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Morange PE, Bezemer I, Saut N, Bare L, Burgos G, Brocheton J, et al. A follow-up study of a genome-wide association scan identifies a susceptibility locus for venous thrombosis on chromosome 6p24.1. Am J Hum Genet. 2010;86:592–595. doi: 10.1016/j.ajhg.2010.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lee SH, Rho J, Jeong D, Sul JY, Kim T, Kim N, et al. v-ATPase V0 subunit d2-deficient mice exhibit impaired osteoclast fusion and increased bone formation. Nat Med. 2006;12:1403–1409. doi: 10.1038/nm1514. [DOI] [PubMed] [Google Scholar]
  • 80.Holmes DR, Jr, Dehmer GJ, Kaul S, Leifer D, O'Gara PT, Stein CM. ACCF/AHA clopidogrel clinical alert: approaches to the FDA "boxed warning": a report of the American College of Cardiology Foundation Task Force on clinical expert consensus documents and the American Heart Association endorsed by the Society for Cardiovascular Angiography and Interventions and the Society of Thoracic Surgeons. J Am Coll Cardiol. 2010;56:321–341. doi: 10.1016/j.jacc.2010.05.013. [DOI] [PubMed] [Google Scholar]
  • 81.Dal Colletto GM, Fulker DW, Barretto OC, Kolya M. Genetic and environmental effects on blood cells. Acta Genet Med Gemellol (Roma) 1993;42:245–252. doi: 10.1017/s000156600000324x. [DOI] [PubMed] [Google Scholar]
  • 82.Garner C, Tatu T, Reittie JE, Littlewood T, Darley J, Cervino S, et al. Genetic influences on F cells and other hematologic variables: a twin heritability study. Blood. 2000;95:342–346. [PubMed] [Google Scholar]
  • 83.Pilia G, Chen WM, Scuteri A, Orru M, Albai G, Dei M, et al. Heritability of cardiovascular and personality traits in 6,148 Sardinians. PLoS Genet. 2006;2:e132. doi: 10.1371/journal.pgen.0020132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Marroni F, Grazio D, Pattaro C, Devoto M, Pramstaller P. Estimates of genetic and environmental contribution to 43 quantitative traits support sharing of a homogeneous environment in an isolated population from South Tyrol, Italy. Hum Hered. 2008;65:175–182. doi: 10.1159/000109734. [DOI] [PubMed] [Google Scholar]
  • 85.Traglia M, Sala C, Masciullo C, Cverhova V, Lori F, Pistis G, et al. Heritability and demographic analyses in the large isolated population of Val Borbera suggest advantages in mapping complex traits genes. PLoS One. 2009;4:e7554. doi: 10.1371/journal.pone.0007554. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Biino G, Balduini CL, Casula L, Cavallo P, Vaccargiu S, Parracciani D, et al. Analysis of 12,517 inhabitants of a Sardinian geographic isolate reveals that predispositions to thrombocytopenia and thrombocytosis are inherited traits. Haematologica. 2011;96:96–101. doi: 10.3324/haematol.2010.029934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Yang S, Ren J, Yan X, Huang X, Zou Z, Zhang Z, et al. Quantitative trait loci for porcine white blood cells and platelet-related traits in a White Duroc x Erhualian F resource population. Anim Genet. 2009;40:273–278. doi: 10.1111/j.1365-2052.2008.01830.x. [DOI] [PubMed] [Google Scholar]
  • 88.Gong YF, Lu X, Wang ZP, Hu F, Luo YR, Cai SQ, et al. Detection of quantitative trait loci affecting haematological traits in swine via genome scanning. BMC Genet. 2010;11:56. doi: 10.1186/1471-2156-11-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Shaughnessy JD, Largaespada DA, Tian E, Fletcher CF, Cho BC, Vyas P, et al. Mrvi1, a common MRV integration site in BXH2 myeloid leukemias, encodes a protein with homology to a lymphoid-restricted membrane protein Jaw1. Oncogene. 1999;6:2069–2084. doi: 10.1038/sj.onc.1202419. [DOI] [PubMed] [Google Scholar]
  • 90.Gagnon AW, Murray DL, Leadley RJ. Cloning and characterization of a novel regulator of G protein signalling in human platelets. Cell Signal. 2002;9:595–606. doi: 10.1016/s0898-6568(02)00012-8. [DOI] [PubMed] [Google Scholar]
  • 91.Kim SD, Sung HJ, Park SK, Kim TW, Park SC, Kim SK, et al. The expression patterns of RGS transcripts in platelets. Platelets. 2006;10:493–497. doi: 10.1080/09537100600758123. [DOI] [PubMed] [Google Scholar]
  • 92.Yowe D, Weich N, Prabhudas M, Poisson L, Errada P, Kapeller R, et al. RGS18 is a myeloerythroid lineage-specific regulator of G-protein-signalling molecule highly expressed in megakaryocytes. Biochem J. 2001;8:109–118. doi: 10.1042/0264-6021:3590109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Garcia A, Prabhakar S, Hughan S, Anderson TW, Brock CJ, Pearce AC, et al. Differential proteome analysis of TRAP-activated platelets: involvement of DOK-2 and phosphorylation of RGS proteins. Blood. 2004;103:2088–2095. doi: 10.1182/blood-2003-07-2392. [DOI] [PubMed] [Google Scholar]
  • 94.Yabe M, Matsubara Y, Takahashi S, Ishihara H, Shibano T, Miyaki K, et al. Identification of ADRA2A polymorphisms related to shear-mediated platelet function. Biochem Biophys Res Commun. 2006;11:1001–1005. doi: 10.1016/j.bbrc.2006.06.180. [DOI] [PubMed] [Google Scholar]
  • 95.Small KM, Brown KM, Seman CA, Theiss CT, Liggett SB. Complex haplotypes derived from noncoding polymorphisms of the intronless alpha2A-adrenergic gene diversify receptor expression. Proc Natl Acad Sci U S A. 2006;13:5472–5477. doi: 10.1073/pnas.0601345103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Kambayashi J, Shinoki N, Nakamura T, Ariyoshi H, Kawasaki T, Sakon M, et al. Prevalence of impaired responsiveness to epinephrine in platelets among Japanese. Thromb Res. 1996;12:85–90. doi: 10.1016/0049-3848(95)00216-2. [DOI] [PubMed] [Google Scholar]
  • 97.Jones M, Peden AH, Yull H, Wight D, Bishop MT, Prowse CV, et al. Human platelets as a substrate source for the in vitro amplification of the abnormal prion protein (PrP) associated with variant Creutzfeldt-Jakob disease. Transfusion. 2009;14:376–384. doi: 10.1111/j.1537-2995.2008.01954.x. [DOI] [PubMed] [Google Scholar]
  • 98.Holada K, Glierova H, Simak J, Vostal JG. Expression of cellular prion protein on platelets from patients with gray platelet or Hermansky-Pudlak syndrome and the protein's association with alpha-granules. Haematologica. 2006;15:1126–1129. [PubMed] [Google Scholar]
  • 99.Rother E, Brandl R, Baker DL, Goyal P, Gebhard H, Tigyi G, et al. Subtype-selective antagonists of lysophosphatidic Acid receptors inhibit platelet activation triggered by the lipid core of atherosclerotic plaques. Circulation. 2003;108:741–747. doi: 10.1161/01.CIR.0000083715.37658.C4. [DOI] [PubMed] [Google Scholar]
  • 100.Martinez MC, Larbret F, Zobairi F, Coulombe J, Debili N, Vainchenker W, et al. Transfer of differentiation signal by membrane microvesicles harboring hedgehog morphogens. Blood. 2006;19:3012–3020. doi: 10.1182/blood-2006-04-019109. [DOI] [PubMed] [Google Scholar]
  • 101.Letschka T, Kollmann V, Pfeifhofer-Obermair C, Lutz-Nicoladoni C, Obermair GJ, Fresser F, et al. PKC-theta selectively controls the adhesion-stimulating molecule Rap1. Blood. 2008;21:4617–4627. doi: 10.1182/blood-2007-11-121111. [DOI] [PubMed] [Google Scholar]
  • 102.Severson EA, Lee WY, Capaldo CT, Nusrat A, Parkos CA. Junctional adhesion molecule A interacts with Afadin and PDZ-GEF2 to activate Rap1A, regulate beta1 integrin levels, and enhance cell migration. Mol Biol Cell. 2009;20:1916–1925. doi: 10.1091/mbc.E08-10-1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Joutsi-Korhonen L, Smethurst PA, Rankin A, Gray E, IJsseldijk M, Onley CM, et al. The low-frequency allele of the platelet collagen signaling receptor glycoprotein VI is associated with reduced functional responses and expression. Blood. 2003;22:4372–4379. doi: 10.1182/blood-2002-08-2591. [DOI] [PubMed] [Google Scholar]
  • 104.Trifiro E, Williams SA, Cheli Y, Furihata K, Pulcinelli FM, Nugent DJ, et al. The low-frequency isoform of platelet glycoprotein VIb attenuates ligand-mediated signal transduction but not receptor expression or ligand binding. Blood. 2009;23:1893–1899. doi: 10.1182/blood-2009-03-209510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Canobbio I, Stefanini L, Cipolla L, Ciraolo E, Gruppi C, Balduini C, et al. Genetic evidence for a predominant role of PI3Kbeta catalytic activity in. Blood. 2009;25:2193–2196. doi: 10.1182/blood-2009-03-208074. [DOI] [PubMed] [Google Scholar]
  • 106.Guidetti GF, Lova P, Bernardi B, Campus F, Baldanzi G, Graziani A, et al. The Gi-coupled P2Y12 receptor regulates diacylglycerol-mediated signaling in human platelets. J Biol Chem. 2008;24:28795–28805. doi: 10.1074/jbc.M801588200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Wang Z, Holly SP, Larson MK, Liu J, Yuan W, Chrzanowska-Wodnicka M, et al. Rap1b is critical for glycoprotein VI-mediated but not ADP receptor-mediated alpha2beta1 activation. J Thromb Haemost. 2009;26:693–700. doi: 10.1111/j.1538-7836.2009.03289.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Maguire PB, Foy M, Fitzgerald DJ. Using proteomics to identify potential therapeutic targets in platelets. Biochem Soc Trans. 2005;33:409–412. doi: 10.1042/BST0330409. [DOI] [PubMed] [Google Scholar]
  • 109.Arthur WT, Ellerbroek SM, Der CJ, Burridge K, Wennerberg K. XPLN, a guanine nucleotide exchange factor for RhoA and RhoB, but not RhoC. J Biol Chem. 2002;277:42964–42972. doi: 10.1074/jbc.M207401200. [DOI] [PubMed] [Google Scholar]
  • 110.Reems JA, Wang W, Tsubata K, Abdurrahman N, Sundell B, Tijssen MR, et al. Dynamin 3 participates in the growth and development of megakaryocytes. Exp Hematol. 2008;28:1714–1727. doi: 10.1016/j.exphem.2008.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kojima H, Kanada H, Shimizu S, Kasama E, Shibuya K, Nakauchi H, et al. CD226 mediates platelet and megakaryocytic cell adhesion to vascular endothelial cells. J Biol Chem. 2003;29:36748–36753. doi: 10.1074/jbc.M300702200. [DOI] [PubMed] [Google Scholar]
  • 112.Sun L, Gorospe JR, Hoffman EP, Rao AK. Decreased platelet expression of myosin regulatory light chain polypeptide (MYL9) and other genes with platelet dysfunction and CBFA2/RUNX1 mutation: insights from platelet expression profiling. J Thromb Haemost. 2007;30:146–154. doi: 10.1111/j.1538-7836.2006.02271.x. [DOI] [PubMed] [Google Scholar]
  • 113.Derbent M, Oncel Y, Tokel K, Varan B, Haberal A, Yazici AC, et al. Clinical and hematologic findings in Noonan syndrome patients with PTPN11 gene mutations. Am J Med Genet A. 2010;152A:2768–2774. doi: 10.1002/ajmg.a.33713. [DOI] [PubMed] [Google Scholar]
  • 114.Korporaal SJ, Koekman CA, Verhoef S, van der Wal DE, Bezemer M, Van EM, et al. Downregulation of platelet responsiveness upon contact with LDL by the protein-tyrosine phosphatases SHP-1 and SHP-2. Arterioscler Thromb Vasc Biol. 2009;29:372–379. doi: 10.1161/ATVBAHA.108.173278. [DOI] [PubMed] [Google Scholar]
  • 115.Kaushansky K, Broudy VC, Lin N, Jorgensen MJ, McCarty J, Fox N, et al. Thrombopoietin, the Mp1 ligand, is essential for full megakaryocyte development. Proc Natl Acad Sci U S A. 1995;92:3234–3238. doi: 10.1073/pnas.92.8.3234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wiestner A, Schlemper RJ, van der Maas AP, Skoda RC. An activating splice donor mutation in the thrombopoietin gene causes hereditary thrombocythaemia. Nat Genet. 1998;18:49–52. doi: 10.1038/ng0198-49. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Table S1

RESOURCES