World Journal of Emergency Medicine, 2024, 15(3): 229-231 doi: 10.5847/wjem.j.1920-8642.2024.031

Research Letters

Effects of PEAR1 gene polymorphism on big endothelin-1 levels in Chinese patients with acute myocardial infarction after percutaneous coronary intervention

Yi Yao, Na Xu, Xiaofang Tang, Ce Zhang, Sida Jia, Jingjing Xu, Ying Song, Xueyan Zhao, Runlin Gao, Jinqing Yuan,

Center for Coronary Heart Disease, National Center for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China

Corresponding authors: Jinqing Yuan, Email:dr_jinqingyuan@sina.com

Received: 2023-10-6   Accepted: 2024-01-26  

PDF (138KB) Metadata Metrics Related articles Export EndNote| Ris| Bibtex  Favorite

Cite this article

Yi Yao, Na Xu, Xiaofang Tang, Ce Zhang, Sida Jia, Jingjing Xu, Ying Song, Xueyan Zhao, Runlin Gao, Jinqing Yuan. Effects of PEAR1 gene polymorphism on big endothelin-1 levels in Chinese patients with acute myocardial infarction after percutaneous coronary intervention. World Journal of Emergency Medicine, 2024, 15(3): 229-231 doi:10.5847/wjem.j.1920-8642.2024.031

Acute myocardial infarction (AMI) has been associated with poor prognosis, even after revascularization with percutaneous coronary intervention (PCI), likely due to coronary endothelial cell dysfunction and injury.[1,2] Endothelin-1 (ET-1), a peptide that serves as a vasoconstrictor of smooth muscle cell proliferation, can reflect endothelial cell functional states. Due to low circulation levels and short plasma half-life time, measuring plasma ET-1 levels is difficult. In contrast, big ET-1, a biological precursor of endothelin with longer plasma half-life time, is a more sensitive indicator of endothelial activation. Indeed, some studies have used big ET-1 levels as an indicator of endothelial cell function,[3] and higher big ET-1 levels were associated with stent thrombosis among PCI patients.[4] Thus, big ET-1 is more widely evaluated than ET-1 as an indicator of endothelial cell function.

Platelet endothelial aggregation receptor 1 (PEAR1) is a transmembrane protein that is highly expressed in endothelial cells and platelets.[5] Studies have demonstrated that it plays key roles in endothelial cells, such as inhibiting pulmonary microvascular endothelial cell proliferation,[6] and is also a novel angiogenesis modulator.[7] In addition to examining the action mechanism of PEAR1, other studies have explored the correlation between PEAR1 single nucleotide polymorphisms (SNPs) and cardiovascular disease (CVD) prognosis; one study revealed that Caucasians and African-Americans with PEAR1 SNP rs12041331 had higher adverse cardiovascular events.[8]

Based on these findings, we postulated that post-AMI endothelial cell function may be associated with PEAR1 gene polymorphism, resulting in different clinical outcomes. Therefore, this study aimed to investigate whether PEAR1 gene polymorphism could be related to differences in big ET-1 levels and possibly reflect endothelial cell function among Chinese patients with AMI post-PCI.

This was a single-center retrospective observational study in which patients were admitted to Fuwai Hospital of the Chinese Academy of Medical Sciences and Peking Union Medical College between June 2014 and May 2015 were continuously enrolled. The inclusion criterion was AMI based on the Third Universal Definition from the American Heart Association combined with PCI based on coronary angiography. The exclusion criteria were as follows: (1) no blood sample or big ET-1 levels available; (2) bleeding events within one year according to the Bleeding Academic Research Consortium (BARC) criteria (BARC types 2-5 considered as bleeding events); (3) oral anticoagulation therapy and/or intensified antiplatelet agents (e.g., using tirofiban/cilostazol instead of standard dual antiplatelet therapy); (4) contraindication to antiplatelet therapy.

Fifteen PEAR1 SNPs were selected; their location, function and hypothesized biological roles are shown in supplementary Table 1.[8-13] All the SNPs were genotyped using an improved multiple ligase detection method according to previous research.[12]

To measure plasma big ET-1 levels, blood was collected into ethylenediamine tetraacetic acid (EDTA) vacutainer tubes at 12-36 h after PCI and centrifuged to obtain plasma. The big ET-1 concentrations were measured via an enzyme-linked immunosorbent assay (Biomedica, Austria), and the normal reference value was < 0.25 pmol/L.

Categorical variables are reported as counts (percentages). Continuous variables with a normal distribution are presented as the mean±standard deviation (SD). The big ET-1 levels are presented as medians with the interquartile, as they had a non-normal distribution. The relationships between PEAR1 SNPs and the big ET-1 levels were analyzed in terms of a dominant model, including major allele homozygote and minor allele carried. The big ET-1 levels between the two afore-mentioned genotypes were compared using a rank-sum test. Generalized linear model analysis was also conducted to identify independent associations between PEAR1 SNPs and big ET-1 levels after adjusting for potential confounding factors. All the statistical analyses were performed using SPSS version 26.0. The P-value <0.05 was considered statistically significant.

A total of 292 AMI-PCI patients were enrolled, as shown in supplementary Figure 1. The level of big ET-1 was 0.35 (0.23-0.50) pmol/L. The clinical characteristics of patients are shown in supplementary Table 2.

According to the dominant model analysis, two of the 15 selected SNPs showed a significant correlation with big ET-1 levels. More specifically, compared to AMI-PCI patients with major allele homozygote, patients with the minor allele T of rs56260937 or the minor allele A of rs822442 had markedly lower big ET-1 levels (Table 1). After adjusting for confounding factors that may affect endothelial cell function, including age, sex, body mass index, diabetes mellitus, hypertension, hyperlipidemia, and current smoking, we established a generalized linear model and found that rs56260937 and rs822442 were independently related with a decrease in plasma big ET-1 levels (odds ratio [OR]=0.90, 95% confidence interval [95% CI]: 0.82-0.98; P=0.025; and OR=0.91, 95% CI: 0.82-0.99, P=0.038, respectively).

Table 1.   Effects of different PEAR1 SNPs on big ET-1 levels

  

PEAR1 SNPGenotypes (n)Big ET-1 (pmol/L, median [interquartile])OR (95% confidence intervals)P-value
rs11264579CC (158)0.34 (0.23-0.48)1.31 (0.74-2.32)0.206
CT+TT (134)0.37 (0.25-0.51)
rs11264580TT (129)0.37 (0.24-0.52)0.77 (0.43-1.36)0.263
TC+CC (163)0.34 (0.22-0.47)
rs11264581GG (101)0.34 (0.22-0.46)2.06 (0.92-4.61)0.278
GA+AA (191)0.35 (0.24-0.52)
rs12041331GG (116)0.35 (0.24-0.45)1.32 (0.70-2.47)0.718
GA+AA (176)0.34 (0.23-0.53)
rs12137505GG (91)0.38 (0.23-0.52)0.92 (0.51-1.65)0.765
GA+AA (201)0.34 (0.23-0.48)
rs12566888GG (111)0.35 (0.23-0.44)1.32 (0.70-2.49)0.502
GT+TT (181)0.35 (0.23-0.53)
rs2644592AA (105)0.37 (0.24-0.52)0.96 (0.54-1.70)0.364
AG+GG (187)0.34 (0.22-0.48)
rs2768759AA (255)0.34 (0.23-0.49)1.74 (0.91-3.33)0.115
AC+CC (37)0.40 (0.30-0.52)
rs3737224CC (128)0.37 (0.24-0.52)0.76 (0.43-1.35)0.279
CT+TT (164)0.34 (0.22-0.47)
rs41273215CC (130)0.37 (0.24-0.52)0.67 (0.37-1.22)0.194
CT+TT (162)0.34 (0.22-0.46)
rs4661012GG (80)0.39 (0.24-0.53)0.78 (0.44-1.41)0.261
GT+TT (212)0.34 (0.22-0.48)
rs56260937CC (137)0.39 (0.25-0.54)0.48 (0.24-0.95)0.030
CT+TT (155)0.33 (0.22-0.46)
rs57731889CC (107)0.35 (0.23-0.44)1.92 (0.89-4.12)0.567
CT+TT (185)0.34 (0.23-0.52)
rs822441GG (109)0.37 (0.24-0.49)1.08 (0.60-1.96)0.835
GC+CC (183)0.34 (0.23-0.52)
rs822442CC (145)0.39 (0.25-0.54)0.51 (0.26-0.96)0.031
CA+AA (147)0.33 (0.22-0.43)

PEAR1: platelet endothelial aggregation receptor 1; SNP: single nucleotide polymorphism; ET-1: endothelin-1; OR: odds ratio.

New window| CSV


The big ET-1 levels can reflect endothelial cell function, with higher levels associated with poorer cell function. Indeed, the big ET-1 level among Chinese patients with AMI has been documented to exceed the normal range (>0.25 pmol/L), which was in line with our findings that the big ET-1 level was 0.35 (0.23-0.50) pmol/L. Furthermore, rs56260937 and rs822442 were significantly associated with lower big ET-1 levels, although they were still above 0.25 pmol/L. More specifically, patients with the minor allele T of rs56260937 or the minor allele A of rs822442 had relatively lower big ET-1 levels (0.33 pmol/L) compared with those without these SNPs (0.39 pmol/L). This association was found to be independent after adjusting for confounding factors, indicating that the presence of these variations could explain differences in big ET-1 levels.

The study aimed to investigate whether PEAR1 gene polymorphism could affect endothelial cell function in AMI-PCI patients. Fisch et al[13] found that flow-mediated dilation of the brachial artery was significantly higher among Old Order Amish carriers of the PEAR1 rs12041331 A-allele, compared to those without this allele. Discrepancies regarding SNP findings may be due to the different races of our participants compared with those of Fisch et al, as well as the different methods to assess endothelial cell function. In our study, the carrier rate for minor allele T of rs56260937 (CT+TT type patients) and minor allele A of rs822442 (CA+AA type patients) was 53.1% and 50.3%, respectively, both of which were higher than that reported in the NCBI-SNP database for Asian populations, with 21%-39% for rs56260937 and 15%-38% for rs822442. However, in the Siberian population, the carrier rate of minor allele T of rs56260937 was similar to that found in our study, up to 56%. In the African population, the carrier rate of minor allele A of rs822442 was as high as 59.1%.

The rs56260937 and rs822442 SNPs are located in the exon 23 and exon 20 regions of the PEAR1 gene, respectively. The rs56260937 is a synonymous mutation without any associated amino acid sequence changes. Therefore, based on the observed gene associations, it is difficult to determine its effect on big ET-1 levels, although a possibility could be that this occurred through long-range interactions that could affect the regulation of PEAR1 protein expression.[14] The rs822442 is a non-synonymous mutation that leads to a change in the amino acid sequence from asparagine to lysine. This in turn may lead to changes of PEAR1 function and big ET-1 levels.

One limitation was that the samples were only obtained from Chinese patients receiving standard dual antiplatelet therapy, and the other limitation was that the big ET-1 was the only indicator measured in association with PEAR1 SNPs. Future studies should be conducted to explore the association between big ET-1, PEAR1 SNPs, endothelial cell dysfunction, and AMI prognosis, as well as to examine whether these findings are also applicable to other races.

In AMI-PCI patients, PEAR1 gene polymorphisms may be associated with lower big ET-1 levels, which possibly could lead to changes in endothelial cell function.

Funding: National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences (NCRC2020013) and CAMS Innovation Fund for Medical Sciences (2020-I2M-C&T-B-049).

Ethical approval: The study was approved by the Fuwai Hospital Institutional Ethical Review Board (No. 2021-1501). Written informed consent was obtained from all study participants. The study conformed to the principles outlined in the Declaration of Helsinki.

Conflicts of interest: None.

Contributors: YY and JQY are the guarantors of the paper, with responsibility for the integrity of the work, from inception to published article. NX and XFT contributed to acquisition of data. CZ and SDJ contributed to analysis and interpretation of data. YS contributed to conception and design of the work. JJX contributed to genetic analysis. XYZ and RLG contributed to revise article critically for important intellectual content. All authors have read and gave final approval of the version of the article to be published.

All the supplementary files in this paper are available at http://wjem.com.cn.

Reference

Tseng CN, Chen DY, Chang SH, Huang WK, Hsieh MJ, See LC.

Ambient temperature effect on acute myocardial infarction by risk factors: daily data from 2000 to 2017, Taiwan, China

JACC Asia, 2023; 3(2):228-38.

[Cited within: 1]

Wang GM, Li Y, Wu S, Zheng W, Ma JJ, Xu F, et al.

The combination of creatine kinase-myocardial band isoenzyme and point-of-care cardiac troponin/ contemporary cardiac troponin for the early diagnosis of acute myocardial infarction

World J Emerg Med. 2022; 13(3):163-8.

[Cited within: 1]

Hossain E, Islam K, Yeasmin F, Karim MR, Rahman M, Agarwal S, et al.

Elevated levels of plasma big endothelin-1 and its relation to hypertension and skin lesions in individuals exposed to arsenic

Toxicol Appl Pharmacol. 2012; 259(2):187-94.

[Cited within: 1]

Chen Y, Li JX, Song Y, Xu JJ, Tang XF, Jiang L, et al.

Plasma big endothelin-1 and stent thrombosis: an observational study in patients undergoing percutaneous coronary intervention in China

Thromb Res. 2017; 159:5-12.

DOI:S0049-3848(17)30491-7      PMID:28942357      [Cited within: 1]

Stent thrombosis (ST) is a rare but catastrophic complication of percutaneous coronary intervention, leading to poor prognosis. Endothelin-1 (ET-1) plays an important role in endothelial dysfunction and thrombogenesis. However, the impact of big ET-1 level on ST in patients with coronary stenting is unknown. We aimed to evaluate big ET-1 level as a potential predictor of ST in patients undergoing percutaneous coronary intervention.From January 2013 to December 2013, 8106 consecutive patients underwent successful coronary stent implantation and were prospectively enrolled in this study. Patients were stratified into three groups based on plasma big ET-1 level at admission.The incidence of definite and probable ST at 2years postoperatively was 0.84%; ST incidence was lowest in the low big ET-1 group (0.56%), highest in the high big ET-1 group (1.48%), and intermediate in the medium big ET-1 group (0.74%, log-rank p=0.001). Compared with the low big ET-1 group, the multivariate-adjusted hazard ratio (HR) for ST in the high big ET-1 group was 2.06 (95% confidence interval (CI) 1.14-3.73, p=0.017). In subgroup analyses, high big ET-1 level was independently associated with ST in patients with acute coronary syndrome (HR 2.29, 95% CI 1.03-5.06, p=0.041), but not in those with stable coronary artery disease (p=0.331), and tended to be associated with older age.Plasma big ET-1 level is a valuable independent predictor of ST in patients with coronary stents, especially in the acute coronary syndrome population.Copyright © 2017 Elsevier Ltd. All rights reserved.

Nanda NS, Bao M, Lin HN, 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(26):24680-9.

DOI:10.1074/jbc.M413411200      PMID:15851471      [Cited within: 1]

The present study was designed to identify novel membrane proteins that signal during platelet aggregation. Because one putative mechanism for signaling by a membrane protein involves phosphorylation, we used oligonucleotide-based microarray analyses and mass spectrometric proteomics techniques to specifically discover membrane proteins and also identify those proteins that become phosphorylated on tyrosine, threonine, or serine residues upon platelet aggregation. Surprisingly, both techniques converged to identify a novel membrane protein we have termed PEAR1 (platelet endothelial aggregation receptor 1). Sequence analysis of PEAR1 predicts a type-1 membrane protein, 15 extracellular epidermal growth factor-like repeats, and multiple cytoplasmic tyrosines. Analysis of the tissue distribution of PEAR1 showed that it was most highly expressed in platelets and endothelial cells. Upon platelet aggregation induced by physiological agonists, PEAR1 became phosphorylated on tyrosine (Tyr-925), and serine (Ser-953 and Ser-1029) residues. PEAR1 tyrosine phosphorylation was blocked by eptifibatide, an alpha(IIb)beta(3) antagonist, which inhibits platelet aggregation. Immune clustering of PEAR1 resulted in PEAR1 phosphorylation. Aggregation-induced PEAR1 tyrosine phosphorylation lead to the subsequent association with the ShcB adaptor protein. Platelet proximity induced by centrifugation also induced PEAR1 tyrosine phosphorylation, a reaction not inhibited by eptifibatide. These data suggest that PEAR1 is a novel platelet receptor that signals secondary to alpha(IIb)beta(3)-mediated platelet-platelet contacts.

Zhan QH, Ma XX, He ZZ.

PEAR1 suppresses the proliferation of pulmonary microvascular endothelial cells via PI3K/AKT pathway in ALI model

Microvasc Res. 2020; 128:103941.

[Cited within: 1]

Vandenbriele C, Kauskot A, Vandersmissen I, Criel M, Geenens R, Craps S, et al.

Platelet endothelial aggregation receptor-1: a novel modifier of neoangiogenesis

Cardiovasc Res. 2015; 108(1):124-38.

DOI:10.1093/cvr/cvv193      PMID:26156496      [Cited within: 1]

Platelet endothelial aggregation receptor-1 (PEAR1) is a cell membrane protein, expressed on platelets and endothelial cells (ECs). PEAR1 sustains αIIbβ3 activation in aggregating platelets and attenuates megakaryopoiesis via controlling the degree of Akt phosphorylation. Its role in EC biology is unknown. The aim of this study was to determine the expression of PEAR1 in the human endothelium of various tissues and to investigate its role in ECs in vitro and in angiogenesis, using Pear1(-/-) mice.PEAR1 is present on the membrane and on filo- and lamellipodia of human cultured ECs, and its expression coincides with CD31 in various tissues. PEAR1 expression is variable in ECs of different origin. Lentiviral knockdown of PEAR1 in cultured ECs doubled EC proliferation and significantly stimulated EC migration, in turn enhancing in vitro tube formation on matrigel through the Akt/PTEN-dependent p21/CDC2 pathway. Even when physiological blood vessel formation was unaffected in Pear1(-/-) mice, neoangiogenesis in these mice was significantly increased both in a hind limb ischaemia ligation model [4.7-fold increase in capillary density in the ligated limb of Pear1(-/-) mice compared with ligated limbs in wild-type (WT) mice] and in a skin wound-healing model, resulting in a two-fold faster wound closure in Pear1(-/-) mice compared with WT littermates.We established an inverse correlation between endothelial PEAR1 expression and vascular assembly both in vitro and in vivo. These findings identify PEAR1 as a novel modifier of neoangiogenesis.Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2015. For permissions please email: journals.permissions@oup.com.

Lewis JP, Ryan K, O’Connell JR, Horenstein RB, Damcott CM, Gibson Q, et al.

Genetic variation in PEAR1 is associated with platelet aggregation and cardiovascular outcomes

Circ Cardiovasc Genet. 2013; 6(2):184-92.

[Cited within: 2]

Nie XY, Li JL, Qin SB, Fu Y, Liang GK, Shi LW, et al.

Genetic mutations in PEAR1 associated with cardiovascular outcomes in Chinese patients with acute coronary syndrome

Thromb Res. 2018; 163:77-82.

[Cited within: 1]

Stimpfle F, Bauer M, Rath D, Schaeffeler E, Schwab M, Gawaz M, et al.

Variants of PEAR1 are associated with outcome in patients with ACS and stable CAD undergoing PCI

Front Pharmacol. 2018; 9:490.

DOI:10.3389/fphar.2018.00490      PMID:29867494      [Cited within: 1]

Introduction: Platelet endothelial aggregation receptor 1 (PEAR1) triggers platelet aggregation and is expressed in platelets and endothelial cells. Genome-wide association studies (GWAS) showed an association between platelet function and single-nucleotide polymorphisms (SNPs) in PEAR1.Methods: In 582 consecutive patients with stable coronary artery disease (CAD) or acute coronary syndrome (ACS) scheduled for PCI and treated with ASA and Clopidogrel, Prasugrel, or Ticagrelor, SNP analysis for rs12566888, rs2768759, rs41273215, rs3737224, and rs822442 was performed. During a follow-up period of 365 days after initial PCI, all patients were tracked for a primary endpoint, defined as a combined endpoint consisting of either time to death, myocardial infarction (MI) or ischemic stroke. All cause mortality, MI and ischemic stroke were defined as secondary endpoints.Results: Multivariable Cox model analysis for the primary endpoint revealed a significantly increased risk in homozygous PEAR1 rs2768759 minor allele carriers (hazard ratio, 3.16; 95% confidence interval, 1.4-7.13, p = 0.006) Moreover, PEAR1 rs12566888 minor allele carriers also showed an increased risk in all patients (hazard ratio, 1.69; 95% confidence interval, 0.87-3.27, p = 0 122), which was marginally significant in male patients (hazard ratio, 2.12; 95% confidence interval, 1.02-4.43, p = 0 045; n = 425).Conclusions: To the best of our knowledge, this is the first study showing that distinct genetic variants of PEAR1 are associated with cardiovascular prognosis in high risk patients undergoing PCI and treated with dual anti platelet therapy.

Yao Y, Tang XF, He C, Song Y, Xu JJ, Meng XM, et al.

Effect of PEAR1 genetic variants on 1-year outcomes in Chinese patients with acute myocardial infarction after percutaneous coronary intervention

J Atheroscler Thromb. 2018; 25(5):454-9.

DOI:10.5551/jat.39982      PMID:29212986      [Cited within: 1]

Platelet endothelial aggregation receptor-1 (PEAR1) is a platelet transmembrane protein that plays an important role on platelet aggregation. The aim of this study was to investigate whether PEAR1 genetic variations are associated with 1-year outcomes in Chinese patients with acute myocardial infarction after percutaneous coronary intervention.A total of 647 consecutive Chinese patients with acute myocardial infarction that underwent percutaneous coronary intervention and that were exposed to standard dual antiplatelet therapy with aspirin and clopidogrel were enrolled in this study. Six single nucleotide polymorphisms of PEAR1 were detected using the ligase detection reaction method. The follow-up period was 12 months.Overall, 66 (10.2%) adverse ischemic events occurred. Multivariate Cox regression analysis showed that carriage of the PEAR1 rs56260937 minor allele was an independent predictor of revascularization events (OR=2.15, 95% CI 1.12-4.15, p=0.022) after adjusting for confounding factors.Genetic testing for PEAR1 variants can be helpful in predicting adverse ischemic events in Chinese patients with acute myocardial infarction after percutaneous coronary intervention.

Yao Y, Tang XF, Zhang JH, He C, Ma YL, Xu JJ, et al.

Association of PEAR1 genetic variants with platelet reactivity in response to dual antiplatelet therapy with aspirin and clopidogrel in the Chinese patient population after percutaneous coronary intervention

Thromb Res. 2016; 141:28-34.

DOI:10.1016/j.thromres.2016.02.031      PMID:26962983      [Cited within: 2]

Platelet Endothelial Aggregation Receptor-1 (PEAR1) is a recently reported platelet transmembrane protein which plays an important role in platelet aggregation. The aim of this study was to investigate whether PEAR1 genetic variations were associated with platelet reactivity as assessed by adenosine diphosphate(ADP)-induced platelet aggregation in Chinese patients treated with aspirin and clopidogrel.Patients with coronary heart disease (CHD) who underwent percutaneous coronary intervention (PCI) were enrolled in the study. All patients were on dual antiplatelet therapy with aspirin and clopidogrel. ADP-induced platelet aggregation was measured by thromboelastography and defined as percent inhibition of platelet aggregation (IPA). Patients (n=204) with IPA <30% were identified as high on-treatment platelet reactivity (HPR). Patients (n=201) with IPA >70% were identified as low on-treatment platelet reactivity (LPR). Sixteen single nucleotide polymorphisms (SNPs) of PEAR1 were determined by a method of improved multiple ligase detection reaction.Among the 16 SNPs examined by univariate analysis, 5 SNPs were significantly associated with ADP-induced platelet aggregation. Minor allele C at rs11264580 (p=0.033), minor allele G at rs2644592 (p=0.048), minor allele T at rs3737224 (p=0.033) and minor allele T at rs41273215 (p=0.025) were strongly associated with HPR, whereas homozygous TT genotype at rs57731889 (p=0.009) was associated with LPR. Multivariate logistic regression analysis further revealed that the minor allele T at rs41273215 (p=0.038) was an independent predictor of HPR and the homozygous TT genotype at rs57731889 (p=0.003) was an independent predictor of LPR.PEAR1 genetic variations were strongly associated with ADP-induced platelet aggregation in Chinese patients with CHD treated with aspirin and clopidogrel. These genetic variations may contribute to the variability in platelet function. The utility of PEAR1 genetic variants in the assessment and prediction of cardiovascular risk warrants further investigation.Copyright © 2016 Elsevier Ltd. All rights reserved.

Fisch AS, Yerges-Armstrong LM, Backman JD, Wang H, Donnelly P, Ryan KA, et al.

Genetic variation in the platelet endothelial aggregation receptor 1 gene results in endothelial dysfunction

PLoS One. 2015; 10(9):e0138795.

[Cited within: 2]

Faraday N, Yanek LR, Yang XP, Mathias R, Herrera-Galeano JE, Suktitipat B, et al.

Identification of a specific intronic PEAR1 gene variant associated with greater platelet aggregability and protein expression

Blood. 2011; 118(12):3367-75.

DOI:10.1182/blood-2010-11-320788      PMID:21791418      [Cited within: 1]

Genetic variation is thought to contribute to variability in platelet function; however, the specific variants and mechanisms that contribute to altered platelet function are poorly defined. With the use of a combination of fine mapping and sequencing of the platelet endothelial aggregation receptor 1 (PEAR1) gene we identified a common variant (rs12041331) in intron 1 that accounts for ≤ 15% of total phenotypic variation in platelet function. Association findings were robust in 1241 persons of European ancestry (P = 2.22 × 10⁻⁸) and were replicated down to the variant and nucleotide level in 835 persons of African ancestry (P = 2.31 × 10⁻²⁷) and in an independent sample of 2755 persons of European descent (P = 1.64 × 10⁻⁵). Sequencing confirmed that variation at rs12041331 accounted most strongly (P = 2.07 × 10⁻⁶) for the relation between the PEAR1 gene and platelet function phenotype. A dose-response relation between the number of G alleles at rs12041331 and expression of PEAR1 protein in human platelets was confirmed by Western blotting and ELISA. Similarly, the G allele was associated with greater protein expression in a luciferase reporter assay. These experiments identify the precise genetic variant in PEAR1 associated with altered platelet function and provide a plausible biologic mechanism to explain the association between variation in the PEAR1 gene and platelet function phenotype.

/