Abstract
Background
Protease activated receptors (PAR) −1 and −4 are the principal receptors for thrombin-mediated platelet activation. Functional genetic variation has been described in the human PAR1 gene, but not in the PAR4 gene (F2RL3). We sought to identify variants in and around F2RL3 and to determine their association with perioperative myocardial injury (PMI) after coronary artery bypass graft surgery. We further explored possible mechanisms for F2RL3 SNP associations with PMI including altered receptor expression and platelet activation.
Methods and Results
Twenty-three single nucleotide polymorphisms (SNPs) in the F2RL3 gene region were genotyped in two phases in 934 Caucasian subjects. Platelets from 43 subjects (23 major allele, 20 risk allele) homozygous for rs773857 (SNP with the strongest association with PMI) underwent flow cytometry to assess PAR4 receptor number and response to activation by a specific PAR4 activating peptide (AYPGKF) measured by von Willebrand factor (vWf) binding and P-selectin release and PAC-1 binding. We identified a novel association of SNP rs773857 with PMI (OR 2.4, P=0.004). rs773857 risk allele homozygotes have significantly increased platelet counts and platelets showed a significant increase in P-selectin release after activation (P=0.004).
Conclusion
We conclude that rs773857 risk allele homozygotes are associated with risk for increased platelet count and hyperactivity.
Keywords: Cardiology- general, Platelets- disorders of platelets, Molecular genetics, surgery, Arterial thrombosis, prophylaxis, diagnosis, treatment
INTRODUCTION
An estimated 1 million people worldwide annually experience nonfatal perioperative myocardial injury (PMI) or death.[1] PMI is associated with increased postoperative length of intensive care unit and hospital stays, short and long-term morbidity and mortality, as well as increased healthcare resource utilization.[2-6]
After coronary artery plaque rupture, activated platelets play a crucial role in the propagation of thrombosis, and therefore the exacerbation of PMI. The exposure of coronary vascular subendothelium and plaque contents during coronary arterial plaque rupture, activates the hemostatic system and locally generates thrombin. In response, circulating platelets roll on exposed subendothelium, adhere and form aggregates. These processes involve platelet receptors such as von Willebrand factor receptor complex, granule secretion, and integrins such as αIIbβ3.[7]
Thrombin, an extracellular protease, activates platelets by irreversible cleavage of the N-terminus of the class of protease-activated receptors (PAR) −1, −3 and −4. PARs belong to a G protein-coupled receptor family and serve as cellular targets of thrombin signaling to platelets and endothelium, via cell surface expression. PAR1 and PAR4 are expressed on human platelets and are the principal receptors for thrombin mediated platelet activation. Functional genetic variation has been described in the human PAR1 gene (F2R), but not in the PAR4 gene (F2RL3).[8-10]
Functional variation in the human platelet genome affects cell adhesion, cell activation, and cell-to-cell contact interactions and has been implicated in cardiovascular disease, arterial thrombosis and myocardial infarction.[11-17] Furthermore, platelet reactivity is a significant predictor of cardiac outcomes after myocardial infarction.[18, 19] In coronary artery bypass grafting (CABG) and vascular surgery patient populations, genetic variation of platelet membrane receptor (glyco) proteins has been associated with PMI.[17, 20-22]
Our aim was to determine the relationship between platelet genetic variation in the thrombin-receptor F2RL3 gene and incidence of PMI in subjects undergoing primary CABG surgery. We hypothesized that F2RL3 variants may be associated with PMI through a causal mechanism of altered receptor expression, platelet activation, and thrombin signaling.
METHODS
Study Population
Two institutions (Brigham and Women's Hospital and the Texas Heart Institute) recruited subjects aged 20-90 years undergoing non-emergent primary CABG surgery with cardiopulmonary bypass (CPB), without other concurrent surgery (http://clinicaltrials.gov/show/NCT00281164) between August 2001 and September 2006. Subjects with a preoperative hematocrit < 25% or who received transfusion of leukocyte-rich blood products within 30 days before surgery were not enrolled. In order to avoid potential influence of population stratification on observed associations, analysis was restricted to subjects who self-reported four Caucasian grand-parental ancestry. Study protocols were approved by respective Institutional Review Boards, and participants were enrolled following informed written consent.
Demographic and Clinical Data Collection
At each site, patient demographics, perioperative risk factors, medications, and postoperative outcomes were recorded using study-specific case report forms. Blood samples were drawn before induction of general anesthesia and on the morning of postoperative day (POD) 1. Serum and plasma were stored in vapor phase liquid nitrogen until analysis. Cardiac troponin I (cTnI) was measured at a single blinded core facility using the sandwich immunoassay Triage® platform (Biosite Inc., San Diego, Calif).
Genetic Association Study
F2RL3 Genotyping
DNA was extracted from white blood cells using standard procedures. Genotyping was performed in two phases using the iPLEX Gold assay on a MassARRAY system (Sequenom Inc., San Diego, CA, USA) in accordance with the manufacturer's standard recommendations. Automated genotype calling was done with MassARRAY Typer 4. Genotype clustering was visually checked by an experienced evaluator. SNPs with a genotyping call rate less than 95%, with significant deviation from Hardy–Weinberg equilibrium (P<0.001 in controls) and nonrandom missingness (P<0.05) between cases and controls, were excluded from subsequent analysis. After exclusions and quality control, Phase 1 included 685 subjects and 23 single nucleotide polymorphisms (SNPs) (Supplemental Table I). The 23 candidate SNPs were selected utilizing publicly available information from NCBI (http://www.ncbi.nlm.nih.gov/), the HapMap[23], SeattleSNPs[24], and SNPper[25] to obtain comprehensive coverage of the F2RL3 gene and its flanking regions. SNPs representative of one haplotype block upstream and downstream of F2RL3 were chosen. Linkage-disequilibrium (LD) tagging SNPs with minor allele frequencies of 5% or greater in the HapMap Caucasian cohort were identified using Tagger.[26] Preference was given to the following criteria: (i) non-synonymous coding variation, (ii) promoter region variation, (iii) variation in the 3′ untranslated region, (iv) variation at splice junctions, (v) haplotype tagging SNPs and (vi) previously identified candidate SNPs. The F2RL3 gene does not have identified copy number variations or candidate microsatellite polymorphisms.
Phase 2 included genotyping of 10 of the original 23 SNPs in 934 subjects, which included all subjects from Phase 1 and an additional 249 subjects (Supplemental Table I). The 10 SNPs genotyped in Phase 2 were selected from the 23 Phase 1 SNPs using Tagger[26], based on their association with PMI and their LD to the SNPs from Phase 1 with the goal of including the most significant SNPs. All SNPs with a P-value <0.05 in univariate analysis and an LD r2>0.8 were included.
Functional Platelet Analysis
Functional platelet analyses were performed using whole blood drawn from 43 male subjects selected from the Phase 1 cohort. These subjects were homozygous for the major or minor alleles of SNP rs773857 (23 major allele, 20 risk allele), the F2RL3 locus SNP with the most significant association with PMI in both Phase 1 and Phase 2 assessments. To ensure platelet viability, recruitment was limited to those subjects living within 1.5 hours of the single laboratory where the functional analysis was performed as blood draws were performed at subjects’ homes for their convenience. Subjects were divided into homozygous major allele and homozygous risk allele and matched by age (± 10 years) and by their POD 1 cTnI level (above or below median) after their CABG surgery. Analysis was limited to male subjects to avoid confounding by gender in a population of 82% male subjects.
PRP (Platelet Rich Plasma) Preparation
All phlebotomy for the functional analyses was performed between 7:00am and 10:00am with a 21g needle using aseptic technique. 9ml of whole blood was collected in two buffered 0.105 M sodium citrate blood collection tubes (BD Vacutainer, Franklin Lakes, NJ) within 2 hours of first centrifugation. Human PRP was obtained by centrifugation of the blood at 100g for 20 minutes at room temperature. PRP preparation and all further analyses were performed blinded to patient genotype.
Flow cytometry
Platelet count was determined by flow cytometry using reference beads (SPHERO rainbow fluorescent beads, 5.5 μm diameter, Spherotech Inc., Libertyville, IL).[27] PAR1 and PAR4 receptors were detected on resting and activated platelets in PRP using monoclonal (IgG) anti human PAR1 (Santa Cruz Biotechnology Inc., Santa Cruz, CA) and PAR4 (Abnova, Taipei City, Taiwan) antibodies. Platelet surface receptor numbers were quantified using a Calibrator kit (Platelet Calibrator, Biocytex, Marseille, France). To quantify receptor expression, a calibration curve was constructed for each PRP sample analysis series using the specific PAR1 and 4 antibodies and the appropriate included negative isotype control. Antibody labeling of platelets was performed for 20 minutes at room temperature with 20 μg/μL of anti-human mAbs against PAR1 and PAR4 and appropriate isotype controls. Platelet samples were analyzed on a FACSCalibur flow cytometer (BD, Franklin Lakes, NJ). Five thousand events were acquired and data were analyzed using CellQuest® software (BD). Receptor numbers were derived from the calibration curve obtained, after subtracting the negative isotype control value.[9]
Platelet Activation
Freshly obtained PRP was activated via PAR1 or PAR4 receptors using 3μg/ml of Thrombin Receptor Activating Peptide (TRAP) specific for PAR1 (SFLLRN, obtained from Bachem, Bubendorf, Switzerland) or 18μg/ml TRAP specific for PAR4 (AYPGKF, obtained from Bachem) for 3 min at 37°C. Following stimulation platelets were stained with 5μg/ml of FITC-labeled anti human P-Selectin (CD62P, Becton Dickinson) or PE-labeled anti human αIIbβ3 (PAC-1, BD) mAbs, or with 5μg/ml of FITC-labeled anti human von Willebrand factor (vWf) polyclonal antibody (Dako, Carpinteria, CA) for 20 min at room temperature and directly analyzed on a FACSCalibur (BD), as described above. The appropriate isotype for each antibody were used as control.
Statistical analysis for SNP association
Potential population stratification between Northern vs. Southern European origins was examined using SNPs within the lactase gene (LCT - rs182549, rs2322659, rs3754689, rs3769005, rs4954490, rs4988235) known to vary in frequency along a European north-south cline. The results did not show significant heterogeneity; therefore, a pooled analysis across Northern and Southern European groups was performed.
Categorical and continuous demographic characteristics were compared between groups with likelihood ratio χ2 and Wilcoxon rank sum tests, respectively. A multivariable logistic regression analysis was used to test the association between PAR4 variants and PMI, defined as POD 1 cTnI in the top 10th percentile of the examined Phase 1 study cohort (cTnI >6.73 mg/L), a definition we have utilized before.[6] Clinical and demographic covariates associated with PMI after cardiac surgery in prior studies were included in the multivariate model with stepwise selection (age, gender, institution, preoperative statin, acetylsalicylic acid (ASA), or platelet inhibitor use, recent MI, preoperative creatinine, CPB time, and number of grafts). Permutation– based empirical p-values were used to adjust for SNP data distribution (point-wise) and multiple SNP association tests (family-wise).
PLINK (version 1.04),[28] SAS and SAS/Genetics (SAS Institute, Cary, NC), were used for genetic analyses. Hardy-Weinberg equilibrium was evaluated using an exact test. After application of genotype quality control criteria, univariate analyses were carried out for each SNP to test the null hypothesis of no association between marker polymorphism and PMI, based on additive, dominant and recessive genetic models. Tests of frequency association were estimated with chi-square statistic.
Statistical analysis for platelet activation
Gated and mean fluorescence intensity (MFI) quantitative flow cytometry data of platelet activation with PAR1 and PAR4 agonists was analyzed with linear regression with and without adjusting for age at the time of blood draw. In addition to the raw flow cytometry measurements, the control at time 0 and the baseline measurement subtracted from the raw measurements were also analyzed. To account for the variability of platelet counts among subjects, the flow cytometry measurements were normalized by the platelet counts from each individual.
To avoid potential false positive results driven by distribution outliers or departure from normality, the same analysis on the rank transformed[29] and quantile-normalized[30] flow cytometry data was also applied. A two-sided P < 0.05 was considered significant. Statistical analyses were performed using SAS version 9.1.3 (SAS Institute, Cary, NC).
Power analysis
Since no data on PAR4 receptor density exists in humans, prior published data on PAR1 receptor polymorphisms was used as the basis for the power analysis. Prior work comparing receptor numbers for the F2R intronic polymorphism (IVSn-14 A/T) had shown a mean PAR1 density on human platelets of 1297 ± 186 receptors/platelet for major allele homozygotes and 857 and 1022 for the two minor allele homozygotic individuals.[9, 10] A 20% variation in the number of PAR1 receptors for this polymorphism was assumed, which would also be observed in relation to our identified PAR4 variants. Therefore, the sample size to detect a difference of 150 receptors/platelet was estimated, with a standard deviation of 200 receptors/platelet, a type I error of 0.05 and a power of 80%. Within these assumptions, the required sample would be 42 subjects; 21 subjects for each of the two groups (homozygote major and minor alleles).
RESULTS
Of the 1100 eligible Caucasian subjects enrolled into the source cohort during the study period, 415 were excluded from analysis in Phase 1 for one or more of the following exclusion criteria: no CABG or no CPB (n=40), no aortic cross-clamp (n=80), emergency surgery (n=25), prior cardiac surgery (n=13), CPB not used (n=32), unplanned concurrent valve surgery performed (n=130), missing genotype (n=124), genotyping call rate less than 95% (n=38), leaving 685 subjects for Phase 1 analysis. Perioperative patient characteristics of the 685 subjects included in the study analysis are shown in Table I and are stratified by occurrence of PMI. For Phase 2, an additional 249 Caucasian subjects undergoing non-emergent, isolated primary CABG with CPB were genotyped, increasing the total number of subjects in Phase 2 analysis to 934.
Table I.
Patient characteristics, stratified by perioperative myocardial injury.
| Preoperative Characteristic | no PMI (N=616) | PMI (N=69) | P value |
|---|---|---|---|
| Age (years) | 65 ± 10 | 65 ± 10 | |
| Male gender | 82% (505) | 80% (55) | 0.62 |
| Institution | |||
| Institution 1 | 90% (505) | 10% (57) | |
| Institution 2 | 90% (111) | 10% (12) | 1 |
| Diabetes | |||
| Insulin dependent | 8% (52) | 9% (6) | |
| Non-insulin dependent | 21% (132) | 12% (8) | 0.15 |
| Hypertension | 76% (465) | 75% (52) | 1 |
| Hyp ercholesterol emi a | 76% (470) | 74% (50) | 0.65 |
| BMI (kg/ m2) | 29.2 ± 5.4 | 29.3 ± 5.5 | 0.97 |
| Preoperative creatinine (mg/dL) | 1.09 ± 0.3 | 1.12 ± 0.4 | 0.92 |
| Myocardial infarction ≤ 2 weeks preoperatively | 16% (98) | 36% (25) | <0.001 |
| LV ejection fraction (%) | 52 ± 13 | 47 ±12 | 0.002 |
| Coronary artery regions with > 50% stenosis | |||
| 1-2 regions | 29% (181) | 25% (17) | |
| 3 regions | 52% (323) | 54% (37) | |
| >4 regions | 18% (112) | 22% (15) | 0.63 |
|
Preoperative Medications | |||
| Aspirin | 75% (460) | 74% (51) | 0.88 |
| Non-aspirin platelet inhibitor | 17% (103) | 25% (17) | 0.13 |
| Statin preop | 77% (472) | 71% (49) | 0.30 |
| Intravenous heparin | 25% (152) | 39% (27) | 0.01 |
| Intravenous nitrate | 12% (73) | 22% (15) | 0.03 |
| Warfarin < 7 days preoperatively | 5% (31) | 3% (2) | 0.76 |
|
Preoperative laboratory data | |||
| Hematocrit (%) | 39.9 ± 4.6 | 39.7 ± 4.9 | 0.77 |
| Platelet count (106/mL) | 238 ± 70 | 234 ± 69 | 0.47 |
| cTnl (μ,g/L) | 0.16 ± 0.66 | 3.8 ± 10.5 | <0.001 |
|
Intraoperative Characteristics | |||
| Cardiopulmonary bypass time (minutes) | 95 ± 36 | 116 ± 44 | <0.001 |
| Aortic cross clamp time (minutes) | 71 ± 29 | 86 ± 36 | 0.002 |
| Lowest body temperature (°C) | 33 ± 2 | 32 ± 3 | 0.43 |
| Number of coronary grafts | |||
| 1-2 | 15% (19) | 12% (8) | |
| 3 | 47% (291) | 52% (36) | |
| ≥ 4 | 38% (234) | 36% (25) | 0.67 |
|
Postoperative laboratory data | |||
| Hematocrit POD1(%) | 29.5 ± 3.8 | 29.8 ± 3.6 | 0.42 |
| Platelet count POD3 (106/mL) | 171 ± 60 | 153 ± 54 | 0.02 |
| cTnI postop Day 1 (μg/L) | 1.87 ± 1.8 | 24.1 ± 16.3 | <0.001 |
PMI defined as cardiac Troponin I (cTnI) greater than 90% percentile on postoperative day 1 after primary CABG surgery
POD, post-operative day; LV, left ventricle; BMI, body mass index;
Clinical Predictors of Perioperative Myocardial Injury
In the Phase 1 SNP association study, subjects with PMI (n=69) were more likely to have had a recent myocardial infarction, higher preoperative cTnI, lower preoperative left ventricular ejection fraction, have received preoperative intravenous heparin or nitrates, longer CPB and aortic cross clamp times, and lower postoperative platelet counts on (Table I). After adjusting for other covariates in multivariable analysis, only recent myocardial infarction (within 2 weeks) and length of CPB were independently associated with PMI, accounting for 9% of the risk of PMI (Supplemental Table II).
Genomic Predictors of Perioperative Myocardial Injury
In Phase 1, only rs773857 had a significant association with PMI (point-wise empirical P <0.01), and approached significance after correcting for multiple SNP association tests (family-wise empirical P=0.057) (Supplemental Table III). In Phase 2 analysis, rs773857 remained highly significant after multiple comparison correction (OR 2.4; IQR [1.4-2.0]; family-wise empirical P=0.004).
F2RL3 rs773857 risk allele associated with increase in platelet number and increased platelet α-degranulation
No significant differences in demographic or clinical variables were identified between subjects homozygous for the major allele or risk allele of rs773857 (Table II). Subjects homozygous for the risk allele rs773857 had a higher median platelet count than the homozygous major allele group (3.6 × 107 /mL vs. 3.1 × 107 /mL, P=0.024) (Figure 1). No differences in platelet size or density (not shown) or in PAR1 and PAR4 receptor platelet surface expression were detected, as determined by flow cytometry between major allele or risk allele (Figure 1). Next, the function of major allele and risk allele platelets was investigated. P-selectin platelet surface expression was measured by flow cytometry to evaluate α-alpha-granule secretion. Although PAR1 and PAR4 receptor expression was normal in subjects with the risk allele, upon activation with the specific PAR4 agonist (AYPGKF) platelets obtained from patients with the F2RL3 rs773857 risk allele (PAR4) had more measurable surface P-Selectin, indicating that platelets obtained from the risk group released more α-granule content compared to major allele controls. These data were consistent in both non- and rank-transformed and non- and quantile-normalized univariate analysis (P=0.001) and after adjusting for age and platelet number (P=0.004) (Table III). No significant difference in αIIbβ3 activation or vWf binding was identified by flow cytometry using PAC-1 or anti vWf antibodies (Table III), although an increase in vWf binding was observed in the risk. No differences were observed between the groups when platelets were activated via the collagen receptor GPVI using convulxin. 95% of subjects were on anticoagulant medication (aspirin, clopidogrel, warfarin) at the time of blood draw, but no significant difference in functional analyses was identified between groups in the types or doses of anti-platelet or anticoagulation medications.
Table II.
Patient characteristics of 43 subjects selected for functional platelet analysis
| Major allele | Risk allele | P value | |
|---|---|---|---|
| (n = 23 ) | (n = 20) | ||
|
Preoperative Characteristic
| |||
| Age at enrollment | 66 ± 11 | 61 ± 9 | 0.08 |
| Age at blood draw | 72 ± 11 | 67 ± 9 | 0.08 |
| Hyp ercholesterolemia | 87% (20) | 80% (16) | 0.69 |
|
Diabetes | |||
| Insulin dependent | 4% (1) | 0% (0) | |
| Non-insulin dependent | 9% (2) | 35% (7) | 0.06 |
| Hypertension | 78% (18) | 65% (13) | 0.5 |
| BMI (kg/ m2) | 28.0 ± 2.5 | 28.2 ± 2.5 | 0.83 |
| Smoker past | 87% (20) | 90% (18) | 1 |
| Previous myocardial infarction | 35% (8) | 25% (5) | 0.53 |
| Myocardial infarction ≤ 2 weeks preoperatively | 9% (2) | 15% (3) | 0.65 |
| Preoperative LVEF % | 54 ± 10 | 53 ± 11 | 0.85 |
|
Coronary artery regions with > 50% stenosis | |||
| 1-2 regions | 26% (6) | 40% (8) | |
| 3 regions | 48% (11) | 55% (11) | |
| ≥4 regions | 26% (6) | 5% (1) | 0.16 |
|
Preoperative laboratory data | |||
| Blood Type (O - vs. other) | 48% (11) | 55% (11) | 0.76 |
| Hematocrit (%) | 39.7 ± 4.5 | 40.0 ± 3.6 | 0.81 |
| Creatinine (mg/dL) | 1.1 ± 0.1 | 1.1 ± 0.2 | 0.97 |
| Platelet count (106/mL) | 225 ± 39 | 250 ± 45 | 0.07 |
| cTnI (μ,g/L) | 0.1 ± 0.2 | 0.3 ± 0.6 | 0.84 |
| Platelet count POD3 (106/mL) | 172 ± 41 | 189 ± 45 | 0.43 |
| cTnI POD1 (ng/L) | 2.5 ± 4.8 | 1.8 ± 1.1 | 0.71 |
|
Intraoperative Characteristics | |||
| Cardiopulmonary bypass time (minutes) | 98 ± 24 | 106 ± 22 | 0.22 |
| Aortic cross clamp time (minutes) | 74 ± 16 | 83 ± 20 | 0.11 |
| Number of coronary grafts | |||
| 1-2 | 4% (1) | 10% (2) | |
| 3 | 48% (11) | 45% (9) | |
| > 4 | 48% (11) | 45% (9) | 0.72 |
BMI, body mass index; POD, post-operative day; LVEF, left ventricular ejection fraction; cardiac troponin I, cTnI
Figure 1. rs773857 genotype vs. platelet count and PAR4 receptor expression.
Shown are box-whisper plots of the median platelet count and PAR4 receptor expression with inter-quartile range and outliers of 23 patients with the F2RL3 rs773857 homozygous major allele and 20 patients with the homozygous risk genotype. * - P-value 0.02.
Table III.
Functional analysis: Release of markers upon PAR4 agonist stimulation
| PAR4 (F2RL3 rs773857) | ||||
|---|---|---|---|---|
| univariate | adjust for age | |||
| P-Selectin | Beta | P-value | Beta | P-value |
| transformed | 42.96 | 0.001 | 37.77 | 0.004 |
| non-transformed | 39.72 | 0.006 | 35.70 | 0.014 |
| PAC-1 | ||||
| transformed | −1.54 | 0.935 | −5.65 | 0.771 |
| non-transformed | −18.45 | 0.358 | −18.95 | 0.364 |
| vWF | ||||
| transformed | −33.72 | 0.200 | −42.18 | 0.119 |
| non-transformed | −29.44 | 0.297 | −35.16 | 0.230 |
DISCUSSION
In this study we report a novel association between SNP rs773857, a SNP in the region of the gene encoding the principal receptor for thrombin-mediated platelet activation PAR4, and PMI in subjects undergoing CABG surgery. Furthermore, platelets of subjects homozygous for this SNP showed a significant increase in P-selectin release after activation with a specific PAR4 agonist.
Role of Thrombin in PAR activation
Both F2R and F2RL3 are genes of cardiovascular relevance currently being investigated as drug targets for anticoagulation in coronary disease.[31-33] A recent study identified genome-wide significance for reduced DNA methylation in F2RL3 in heavy smokers, suggesting a causal link between F2RL3 and smoking-related cardiovascular pathology through its role in endothelial physiology and platelet activation .[34] Functional genetic variation has been described in the human F2R gene, but not in the F2RL3 gene, though both are targets of thrombin signaling.[8-10] In rodents, PAR4 activation leads to platelet aggregation, while F2RL3 knock-out mice display impaired coagulation, and platelets that do not respond to thrombin signaling.[35] Equally important, these F2RL3 deficient mice are protected from thrombosis in a mesenteric arteriolar thrombosis model,[35] and protected from cerebral ischemia/reperfusion injury in a transient middle cerebral artery occlusion model.[36] Rats treated with PAR4 antagonists experienced smaller infarct sizes after exposure to cardiac ischemia/reperfusion injury.[37] Of note however, differences exist between pharmacologic inhibition (anatagonists) and F2RL3 deficient (knock-out) mice. F2RL3 deficient mice had larger myocardial infarcts in an ischemia/reperfusion model while pharmacologic inhibition of PAR4 proved to be cardioprotective.[38]
Mechanism of Action of rs773857 Risk Allele
The bi-exonal gene F2RL3 for the PAR4 protein on chromosome 19 encompasses 64 SNPs, eight of which are in coding exons (Build 37.1, GRCh37).[25] Twenty-three SNPs that encompassed three haplotypes in and up to 9kb around the gene were genotyped. rs773857 has a minor allele frequency of 35% in Caucasians, and though located in CPAMD8, 17kb downstream from the closest coding exon in F2RL3, has high LD to SNPs in F2RL3 (r2 up to 0.82) (Figure 2). CPAMD8 is a large gene encompassing 130kb and 40 exons expressed on platelets and in cardiac tissue. The last 5 exons of CPAMD8 overlap with the last exon and the 3’-UTR region of F2RL3. It is therefore plausible, that our SNP of interest is in LD with a hereto-unidentified functional variant in F2RL3 responsible for increased thrombin receptor binding and platelet activation. Only two of the 23 genotyped SNPs are non-synonymous and upon further examination in SIFT (http://sift.jcvi.org/)[39] and SNPnexus[40], tools to examine the possible effects on the transcriptome and protein level, no damaging amino acid changes were identified. rs773857 is located within a 5kb LD block which contains five c-Myc transcription factor binding sites (TFBS) and 12 CTCF TFBS from ChIP-seq peaks on H1 human embryonic stem cells (Open Chromatin TFBS by ChIP-seq from ENCODE/Open Chrom (UT Austin) through the ENCODE Jan 2011 Freeze and CTCF Binding Sites by ChIP-seq from ENCODE/University of Washington).[41] The proto-oncogene C-myc encodes for a transcription factor that regulates expression of up to 15% of genes through enhancer box binding recruitment of histone acetyltransferases.[42] Myc also regulates chromatin structure by regulating histone acetylation in gene-rich areas and in regions far from known genes.[43] The transcriptional repressor CTCF, also known as chicken 11-Zn-finger transcription factor termed CCCTC- binding factor (CTCF), is an insulator protein with an extensive role in gene regulation.[44] CTCF can block the interaction between enhancers and promoters, often as a long-range regulatory element.[45] Thus, it is conceivable that the association with PMI and the effect on platelet activation is mediated through TFBS.
Figure 2. Linkage disequilibrium plot of F2RL3 region.
The number within each square is the correlation (r2) between intersecting SNPs. Correlations and block structure were estimated using HaploView software (version 4.0; http://www.broad.mit.edu/mpg/haploview/).
Limitations
These findings have not been validated in a separate cardiac surgical cohort or a non-surgical population. Even so, this study spans two institutions and multiple surgeons, and our subjects make up a homogenous Caucasian population.
The SNP rs773857 is located within an intron of the gene CPAMD8, which is expressed on platelets and in cardiac tissue. Given the location of the SNP, the association with PMI could be attributed to CPAMD8. However, platelet activation with the specific PAR4 agonist resulting in an increased α-granule release in patients with the rs773857 risk allele makes this unlikely.
Conclusion
We identified a novel association between the SNP rs773857, in proximity to the F2RL3 gene and also the principal receptor (PAR4) for thrombin-mediated platelet activation, and PMI in subjects undergoing CABG surgery. Patients homozygous for this SNP had increased platelet counts and their platelets showed a significant increase in α-granule release after activation with a specific PAR4 agonist.
Supplementary Material
Acknowledgments
The authors acknowledge the outstanding contributory efforts of the CABG Genomics research staff: James Gosnell, RN; Kujtim Bodinaku, MD; Adrienne Kizca, BS; Svetlana Gorbatov, MPH. The authors also thank all study participants.
Funding:
This study was supported in part by the Bayer® Fellowship in Blood Conservation (JDM), a Society of Cardiovascular Anesthesiologists Research Starter Grant (JDM), Biosite Inc., San Diego, CA, NIH R01HL098601 (SCB). This work was conducted with support from the Scholars in Clinical Science Program of Harvard Catalyst | The Harvard Clinical and Translational Science Center (Award #UL1 RR 025758 and financial contributions from Harvard University and its affiliated academic health care centers). The content is solely the responsibility of the authors and does not necessarily represent the official views of Harvard Catalyst, Harvard University and its affiliated academic health care centers, the National Center for Research Resources or the National Institutes of Health.
Footnotes
Clinical Trial Registration: http://clinicaltrials.gov/show/NCT00281164
Disclosures:
None
References
- 1.Devereaux PJ, Goldman L, Cook DJ, et al. Perioperative cardiac events in patients undergoing noncardiac surgery: a review of the magnitude of the problem, the pathophysiology of the events and methods to estimate and communicate risk. CMAJ. 2005;173:627–634. doi: 10.1503/cmaj.050011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Adabag AS, Rector T, Mithani S, et al. Prognostic significance of elevated cardiac troponin I after heart surgery. Ann Thorac Surg. 2007;83:1744–1750. doi: 10.1016/j.athoracsur.2006.12.049. [DOI] [PubMed] [Google Scholar]
- 3.Thielmann M, Massoudy P, Neuhauser M, et al. Prognostic value of preoperative cardiac troponin I in patients undergoing emergency coronary artery bypass surgery with non-ST-elevation or ST-elevation acute coronary syndromes. Circulation. 2006;114:I448–453. doi: 10.1161/CIRCULATIONAHA.105.001057. [DOI] [PubMed] [Google Scholar]
- 4.Chen JC, Kaul P, Levy JH, et al. Myocardial infarction following coronary artery bypass graft surgery increases healthcare resource utilization. Crit Care Med. 2007;35:1296–1301. doi: 10.1097/01.CCM.0000262403.08546.A2. [DOI] [PubMed] [Google Scholar]
- 5.Ausset S, Auroy Y, Lambert E, et al. Cardiac troponin I release after hip surgery correlates with poor long-term cardiac outcome. Eur J Anaesthesiol. 2008;25:158–164. doi: 10.1017/S0265021507001202. [DOI] [PubMed] [Google Scholar]
- 6.Muehlschlegel JD, Perry TE, Liu KY, et al. Troponin is superior to electrocardiogram and creatinine kinase MB for predicting clinically significant myocardial injury after coronary artery bypass grafting. Eur Heart J. 2009;30:1574–1583. doi: 10.1093/eurheartj/ehp134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Andrews RK, Berndt MC. Platelet physiology and thrombosis. Thromb Res. 2004;114:447–453. doi: 10.1016/j.thromres.2004.07.020. [DOI] [PubMed] [Google Scholar]
- 8.Smith SM, Judge HM, Peters G, et al. PAR-1 genotype influences platelet aggregation and procoagulant responses in patients with coronary artery disease prior to and during clopidogrel therapy. Platelets. 2005;16:340–345. doi: 10.1080/00207230500120294. [DOI] [PubMed] [Google Scholar]
- 9.Dupont A, Fontana P, Bachelot-Loza C, et al. An intronic polymorphism in the PAR-1 gene is associated with platelet receptor density and the response to SFLLRN. Blood. 2003;101:1833–1840. doi: 10.1182/blood-2002-07-2149. [DOI] [PubMed] [Google Scholar]
- 10.Ramstrom S, Oberg KV, Akerstrom F, et al. Platelet PAR1 receptor density--correlation to platelet activation response and changes in exposure after platelet activation. Thromb Res. 2008;121:681–688. doi: 10.1016/j.thromres.2007.06.010. [DOI] [PubMed] [Google Scholar]
- 11.Bray PF. Integrin polymorphisms as risk factors for thrombosis. Thromb Haemost. 1999;82:337–344. [PubMed] [Google Scholar]
- 12.Croft SA, Samani NJ, Teare MD, et al. Novel platelet membrane glycoprotein VI dimorphism is a risk factor for myocardial infarction. Circulation. 2001;104:1459–1463. doi: 10.1161/hc3801.096397. [DOI] [PubMed] [Google Scholar]
- 13.Muehlschlegel JD, Body SC. Impact of genetic variation on perioperative bleeding. Am J Hematol. 2008;83:732–737. doi: 10.1002/ajh.21205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sing CF, Stengard JH, Kardia SL. Genes, environment, and cardiovascular disease. Arterioscler Thromb Vasc Biol. 2003;23:1190–1196. doi: 10.1161/01.ATV.0000075081.51227.86. [DOI] [PubMed] [Google Scholar]
- 15.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. doi: 10.1161/01.cir.103.25.3051. [DOI] [PubMed] [Google Scholar]
- 16.Di Castelnuovo A, de Gaetano G, Benedetta Donati M, et al. Platelet glycoprotein IIb/IIIa polymorphism and coronary artery disease: implications for clinical practice. Am J Pharmacogenomics. 2005;5:93–99. doi: 10.2165/00129785-200505020-00002. [DOI] [PubMed] [Google Scholar]
- 17.Rinder CS, Mathew JP, Rinder HM, et al. Platelet PlA2 polymorphism and platelet activation are associated with increased troponin I release after cardiopulmonary bypass. Anesthesiology. 2002;97:1118–1122. doi: 10.1097/00000542-200211000-00013. [DOI] [PubMed] [Google Scholar]
- 18.Trip MD, Cats VM, van Capelle FJ, et al. Platelet hyperreactivity and prognosis in survivors of myocardial infarction. N Engl J Med. 1990;322:1549–1554. doi: 10.1056/NEJM199005313222201. [DOI] [PubMed] [Google Scholar]
- 19.Campo G, Valgimigli M, Gemmati D, et al. Value of platelet reactivity in predicting response to treatment and clinical outcome in patients undergoing primary coronary intervention: insights into the STRATEGY Study. J Am Coll Cardiol. 2006;48:2178–2185. doi: 10.1016/j.jacc.2005.12.085. [DOI] [PubMed] [Google Scholar]
- 20.Faraday N, Martinez EA, Scharpf RB, et al. Platelet gene polymorphisms and cardiac risk assessment in vascular surgical patients. Anesthesiology. 2004;101:1291–1297. doi: 10.1097/00000542-200412000-00008. [DOI] [PubMed] [Google Scholar]
- 21.Ollikainen E, Mikkelsson J, Perola M, et al. Platelet membrane collagen receptor glycoprotein VI polymorphism is associated with coronary thrombosis and fatal myocardial infarction in middle-aged men. Atherosclerosis. 2004;176:95–99. doi: 10.1016/j.atherosclerosis.2004.03.021. [DOI] [PubMed] [Google Scholar]
- 22.Bojesen SE, Juul K, Schnohr P, et al. Platelet glycoprotein IIb/IIIa Pl(A2)/Pl(A2) homozygosity associated with risk of ischemic cardiovascular disease and myocardial infarction in young men: the Copenhagen City Heart Study. J Am Coll Cardiol. 2003;42:661–667. doi: 10.1016/s0735-1097(03)00781-2. [DOI] [PubMed] [Google Scholar]
- 23.The International HapMap Project. Nature. 2003;426:789–796. doi: 10.1038/nature02168. [DOI] [PubMed] [Google Scholar]
- 24.SeattleSNPs . NHLBI Program for Genomic Applications. Seattle, WA.: [Google Scholar]
- 25.Riva A, Kohane IS. A SNP-centric database for the investigation of the human genome. BMC Bioinformatics. 2004;5:33. doi: 10.1186/1471-2105-5-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Barrett JC, Fry B, Maller J, et al. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics. 2005;21:263–265. doi: 10.1093/bioinformatics/bth457. [DOI] [PubMed] [Google Scholar]
- 27.Alugupalli KR, Michelson AD, Barnard MR, et al. Serial determinations of platelet counts in mice by flow cytometry. Thromb Haemost. 2001;86:668–671. [PubMed] [Google Scholar]
- 28.Purcell S, Neale B, Todd-Brown K, et al. PLINK: A Tool Set for Whole-Genome Association and Population-Based Linkage Analyses. Am J Hum Genet. 2007;81:559–575. doi: 10.1086/519795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.SAS Institute Inc. Base SAS® 9.1.3 Procedures Guide. SAS Institute Inc.; Cary, NC: 2006. [Google Scholar]
- 30.Bolstad BM, Irizarry RA, Astrand M, et al. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003;19:185–193. doi: 10.1093/bioinformatics/19.2.185. [DOI] [PubMed] [Google Scholar]
- 31.Wu CC, Wang WY, Wei CK, et al. Combined blockade of thrombin anion binding exosite-1 and PAR4 produces synergistic antiplatelet effect in human platelets. Thromb Haemost. 2011;105:88–95. doi: 10.1160/TH10-05-0305. [DOI] [PubMed] [Google Scholar]
- 32.Leger AJ, Covic L, Kuliopulos A. Protease-activated receptors in cardiovascular diseases. Circulation. 2006;114:1070–1077. doi: 10.1161/CIRCULATIONAHA.105.574830. [DOI] [PubMed] [Google Scholar]
- 33.Wu CC, Teng CM. Comparison of the effects of PAR1 antagonists, PAR4 antagonists, and their combinations on thrombin-induced human platelet activation. Eur J Pharmacol. 2006;546:142–147. doi: 10.1016/j.ejphar.2006.07.004. [DOI] [PubMed] [Google Scholar]
- 34.Breitling LP, Yang R, Korn B, et al. Tobacco-smoking-related differential DNA methylation: 27K discovery and replication. Am J Hum Genet. 2011;88:450–457. doi: 10.1016/j.ajhg.2011.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sambrano GR, Weiss EJ, Zheng YW, et al. Role of thrombin signalling in platelets in haemostasis and thrombosis. Nature. 2001;413:74–78. doi: 10.1038/35092573. [DOI] [PubMed] [Google Scholar]
- 36.Mao Y, Zhang M, Tuma RF, et al. Deficiency of PAR4 attenuates cerebral ischemia/reperfusion injury in mice. J Cereb Blood Flow Metab. 2010;30:1044–1052. doi: 10.1038/jcbfm.2009.283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Strande JL, Hsu A, Su J, et al. Inhibiting protease-activated receptor 4 limits myocardial ischemia/reperfusion injury in rat hearts by unmasking adenosine signaling. J Pharmacol Exp Ther. 2008;324:1045–1054. doi: 10.1124/jpet.107.133595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Antoniak S, Pawlinski R, Mackman N. Protease-activated receptors and myocardial infarction. IUBMB Life. 2011;63:383–389. doi: 10.1002/iub.441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kumar P, Henikoff S, Ng PC. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nature protocols. 2009;4:1073–1081. doi: 10.1038/nprot.2009.86. [DOI] [PubMed] [Google Scholar]
- 40.Chelala C, Khan A, Lemoine NR. SNPnexus: a web database for functional annotation of newly discovered and public domain single nucleotide polymorphisms. Bioinformatics. 2009;25:655–661. doi: 10.1093/bioinformatics/btn653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Euskirchen GM, Rozowsky JS, Wei CL, et al. Mapping of transcription factor binding regions in mammalian cells by ChIP: comparison of array- and sequencing-based technologies. Genome Res. 2007;17:898–909. doi: 10.1101/gr.5583007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Gearhart J, Pashos EE, Prasad MK. Pluripotency redux--advances in stem-cell research. N Engl J Med. 2007;357:1469–1472. doi: 10.1056/NEJMp078126. [DOI] [PubMed] [Google Scholar]
- 43.Cotterman R, Jin VX, Krig SR, et al. N-Myc regulates a widespread euchromatic program in the human genome partially independent of its role as a classical transcription factor. Cancer Res. 2008;68:9654–9662. doi: 10.1158/0008-5472.CAN-08-1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Fu Y, Sinha M, Peterson CL, et al. The insulator binding protein CTCF positions 20 nucleosomes around its binding sites across the human genome. PLoS Genet. 2008;4:e1000138. doi: 10.1371/journal.pgen.1000138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kim TH, Abdullaev ZK, Smith AD, et al. Analysis of the vertebrate insulator protein CTCF-binding sites in the human genome. Cell. 2007;128:1231–1245. doi: 10.1016/j.cell.2006.12.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.


