Skip to main content

Abstract

Rationale:

Platelets shed microRNAs (miRNAs). Plasma miRNAs change on platelet inhibition. It is unclear whether plasma miRNA levels correlate with platelet function.

Objective:

To link small RNAs to platelet reactivity.

Methods and Results:

Next-generation sequencing of small RNAs in plasma revealed 2 peaks at 22 to 23 and 32 to 33 nucleotides corresponding to miRNAs and YRNAs, respectively. Among YRNAs, predominantly, fragments of RNY4 and RNY5 were detected. Plasma miRNAs and YRNAs were measured in 125 patients with a history of acute coronary syndrome who had undergone detailed assessment of platelet function 30 days after the acute event. Using quantitative real-time polymerase chain reactions, 92 miRNAs were assessed in patients with acute coronary syndrome on different antiplatelet therapies. Key platelet-related miRNAs and YRNAs were correlated with platelet function tests. MiR-223 (rp=0.28; n=121; P=0.002), miR-126 (rp=0.22; n=121; P=0.016), and other abundant platelet miRNAs and YRNAs showed significant positive correlations with the vasodilator-stimulated phosphoprotein phosphorylation assay. YRNAs, miR-126, and miR-223 were also among the small RNAs showing the greatest dependency on platelets and strongly correlated with plasma levels of P-selectin, platelet factor 4, and platelet basic protein in the population-based Bruneck study (n=669). A single-nucleotide polymorphism that facilitates processing of pri-miR-126 to mature miR-126 accounted for a rise in circulating platelet activation markers. Inhibition of miR-126 in mice reduced platelet aggregation. MiR-126 directly and indirectly affects ADAM9 and P2Y12 receptor expression.

Conclusions:

Levels of platelet-related plasma miRNAs and YRNAs correlate with platelet function tests in patients with acute coronary syndrome and platelet activation markers in the general population. Alterations in miR-126 affect platelet reactivity.

Introduction

MicroRNAs (miRNAs) are small noncoding RNAs with cell-type specific expression patterns that are released by cells into the circulation as part of membranous particles or protein complexes.1 Thus, miRNAs can be readily quantified by real-time polymerase chain reactions (qPCRs) in plasma and serum and have generated increasing interest as potential new biomarkers.2 Our group has previously identified platelet-related miRNA signatures that are predictive of cardiovascular events.3 In addition, we measured miRNAs in healthy volunteers and in patients with symptomatic atherosclerosis before and after initiation of dual antiplatelet therapy and demonstrated reduced plasma levels of platelet-related miRNAs on platelet inhibition.4
In This Issue, see p 367
Editorial, see p 374
Dual oral antiplatelet therapy (acetylsalicylic acid [ASA]+a P2Y12 inhibitor) is commonly used for the management of non–ST-segment–elevation acute coronary syndromes (ACS) and ST-segment–elevation myocardial infarction.5 ASA irreversibly inhibits cyclooxygenase 1 in platelets, thereby repressing thromboxane A2 (TxA2) synthesis and, consequently, platelet activation. Clopidogrel, prasugrel, and ticagrelor target the P2Y12 receptor for ADP. However, interindividual variability in the platelet response to clopidogrel has been reported. Prasugrel and ticagrelor exhibit a more consistent antiplatelet effect and have shown benefits over clopidogrel in patients with ACS but also increase the risk of bleeding.6,7 It is currently unclear whether plasma levels of platelet-related miRNAs correlate with the residual platelet activity in patients with ACS and how different antiplatelet agents alter miRNAs.
In this study, we used RNA sequencing to characterize small RNAs in plasma. Then, we compared the effect of different antiplatelet agents and explored the association of small RNAs (miRNAs and YRNAs) with platelet function tests in patients with ACS. Moreover, we correlated their plasma levels to platelet activation markers in the prospective, population-based Bruneck study3 and investigated whether a single-nucleotide polymorphism (SNP) that facilitates miR-126 processing8 alters circulating miR-126 levels and platelet reactivity. These epidemiological observations were complemented by preclinical studies, assessing platelet function in mice on treatment with antagomiRs directed against miR-126 and by mechanistic studies measuring miR-126 targets.

Methods

An expanded Methods section is available in the Online Data Supplement.

Next-Generation Sequencing

Small RNA libraries were generated from non-normalized RNA (ranging from 375 pg to 1 ng) extracted from equal volumes of platelet-poor plasma (PPP) and platelet-rich plasma (PRP) from healthy human volunteers. Before library preparation, RNA was spiked with equal amounts of C. elegans miR-39 star (cel-miR-39*) to assist in normalization. Libraries were prepared using the small RNA library preparation kit version 2.0 (Illumina Cambridge Ltd) according to manufacturer’s protocol with limited modifications. In brief, adapters were ligated to the 3′ end and 5′ end of RNA followed by cDNA preparation through reverse transcription. The cDNA was amplified with 12 cycles of PCR. The resulting library was purified using Agencourt Ampure XP beads (Beckman Coulter Inc). Libraries were quantified using Qubit (Life Technologies) and Bioanalyzer (Agilent). Because the total amount of libraries generated was limited, the libraries were mixed together in nonequimolar concentrations and sequenced on HiSeq2000 (Illumina Cambridge Ltd). Traces of the adapter (TGGAATTCTCGGGTGCCAAGG) at the end of the 22 nucleotides for miRNAs were removed using the Trimgalore software, allowing for 10% mismatch and discarding reads that, after adapter removal, became shorter than 15 nucleotides. More than 99.5% of the reads were reduced in size after adapter removal.

RNA Isolation, Reverse Transcription, and Preamplification

RNA isolation, reverse transcription reaction, and preamplification, as well as individual qPCRs for miRNAs, were performed as described previously.3,4

Custom-Designed qPCR Plates

The expression profile of 92 miRNAs was assessed using custom-made Exiqon locked nucleic acid (LNA) qPCR plates (Exiqon Life Sciences) as described previously.4

ACS Study Population and Sample Collection

Plasma samples were obtained from a patient-based cohort of 125 patients with a history of ACS 30 days previously (ST-segment–elevation myocardial infarction, non–ST-segment–elevation myocardial infarction, or unstable angina) who had undergone detailed assessment of platelet function.5 The Northern General Hospital, Sheffield, United Kingdom, is the only center providing percutaneous coronary intervention and cardiac surgery services to the surrounding population of ≈1.8 million people, and protocols for antiplatelet therapy are standardized and implemented synchronously across this region. This was a prospective observational study intended to phenotype patients with ACS, including assessment of the effects of changing patterns of P2Y12 inhibitor usage. Treatment algorithms for oral antiplatelet and other secondary prevention therapies in patients with ACS are described by Joshi et al.5 All the patients recruited for the study provided informed consent, and the study was approved by the local research ethics committee. The exclusion criteria were <18 years of age, serious comorbidities, end-stage renal failure, and pregnancy or suspected pregnancy. Patients enrolled in the study attended the Clinical Research Facility, Northern General Hospital, Sheffield, United Kingdom, at 30 days after the onset of ACS. Venepuncture with needle and syringe was used to obtain venous blood samples that were anticoagulated with 3.13% trisodium citrate dihydrate (citrate).

Human Platelet Function Testing

Light Transmittance Aggregometry

Citrate-anticoagulated blood was first centrifuged at 200 relative centrifugation force for 10 minutes from which PRP was extracted. The remaining blood was then centrifuged again at 1500 relative centrifugation force from which PPP was extracted. A platelet count was performed on the PRP, and no dilution with PPP was performed unless the platelet count was >400×109/L in which case PPP was used to dilute the PRP to a platelet count of 400×109/L. Platelet aggregation response to ADP was determined using light transmittance aggregometry (BioData PAP-8E optical aggregometer). The maximum platelet aggregation response to 20 μmol/L ADP or 2 mmol/L arachidonic acid (AA) was determined after 6 minutes.

VerifyNow P2Y12 Assay

Citrate-anticoagulated blood was analyzed using VerifyNow P2Y12 cartridges and VerifyNow analyzer (Accumetrics, Elizabethtown, KY) according to the manufacturer’s instructions. Platelet reaction units were recorded.

Vasodilator-Stimulated Phosphoprotein Phosphorylation Assay

Citrate-anticoagulated blood was analyzed using vasodilator-stimulated phosphoprotein (VASP) phosphorylation assay kits (BioCytex, France), and platelet reactivity index was determined, according to the manufacturer’s instructions. Fluorescence of samples was measured using an LSRII flow cytometer (Becton Dickinson).

Bruneck Cohort

The Bruneck study is a population-based, prospective survey of the epidemiology and pathogenesis of atherosclerosis and cardiovascular disease.3,9 The study protocol was reviewed and approved by the Ethics Committees of Verona and Bolzano, and all participants provided their written informed consent before entering the study. At the 1990 baseline evaluation, the study population comprised an age- and sex-stratified random sample of all inhabitants of Bruneck (125 men and 125 women from each of the fifth through eighth decades of age, all whites). Samples from the year 2000 follow-up were used for this study (n=669). As part of the 2000 follow-up, citrate plasma and serum samples were drawn after an overnight fast and 12 hours of abstinence from smoking. Samples were divided into aliquots and immediately stored at −80°C.

Platelet Spike-In

Human platelets and plasma were isolated from 4 healthy volunteers. Whole blood (16 mL) was drawn onto 4 mL of acid–citrate–dextrose (stock solution: 2.5 g sodium citrate, 2.0 g glucose, and 1.5 g citric acid, in 100 mL H2O) using a 21-gauge needle and 20-mL syringe and centrifuged at 190g for 30 minutes. All centrifugations were performed at room temperature without brake. The supernatant was transferred into a new tube. To prevent platelet activation, 10 nmol/L prostaglandin E1 (Sigma-Aldrich) and 10 μmol/L indomethacin (Sigma-Aldrich) were added. To deplete leukocytes, the supernatant was further centrifuged at 280g for 10 minutes. Leukocyte-depleted PRP (100 μL) was transferred to a 1.5-mL tube for RNA extraction. The remaining volume was transferred to a new tube and centrifuged at 1180g for 10 minutes to pellet platelets. The resulting supernatant was PPP. The platelet pellet was washed twice with modified Tyrode buffer (134 mmol/L NaCl, 2.9 mmol/L KCl, 0.34 mmol/L Na2HPO4, 12 mmol/L NaHCO3, 20 mmol/L HEPES, and 1 mmol/L MgCl2, pH 7.4; glucose (45 mg/50 mL) was added just before use, and the solution was warmed to 37°C in a water bath). Prostaglandin E1 and indomethacin were added during each wash at the concentrations mentioned above. The solution was centrifuged between washes at 1180g for 10 minutes. The final platelet pellet was resuspended in 1/20 of the volume of original PRP to obtain a 20× stock platelet solution. The 20× platelet solution was then spiked back into the PPP from the same donor to achieve 200%, 100%, 50%, and 5% spike-ins. Immediately after spiking PPP, QIAzol lysis reagent was added and RNA was extracted as described previously.3,4

Systemic Inhibition of MiR-126-3p

Male C57BL/6J mice (Harlan), aged 8 weeks, were treated with cholesterol-conjugated antagomiR constructs (Fidelity Systems, Gaithersburg, MD) or sterile PBS. Sequences were designed to target miR-126-3p (5′-C*G*CAUUAUUACUCACGGU* A*C*G*A*-Chol*T-3′) or to serve as nontargeting control (5′-A*A*GGC AAGCUGACCCUGAA*G*U*U*Chol*T-3′). Intraperitoneal injections were performed on day 0, 1, and 2, in a dose of 25 mg/kg for the platelet function assays in whole blood and 40 mg/kg for the aggregometry experiments carried out in PRP. On day 7, mice were anesthetized using pentobarbital before collection of blood from the inferior vena cava using syringes containing lepirudin (Refludan, 25 μg/mL; Celgene, Windsor, United Kingdom). PRP was isolated as previously described.10 Briefly, whole blood was diluted 1:1 with HEPES–Tyrode buffer (137 mmol/L NaCl, 20 mmol/L HEPES, 5.6 mmol/L glucose, 1 g/L BSA, 1 mmol/L MgCl2, 2.7 mmol/L KCl, and 3.3 mmol/L NaH2PO4) before centrifugation (100g, 8 minutes, retention time).

Platelet Function in Mice

Platelet function tests were carried out as described previously.11 Half-area 96-well microtiter plates (Greiner Bio-One, Stonehouse, United Kingdom) were precoated with hydrogenated gelatin (0.75% wt/vol; Sigma, United Kingdom) in PBS to block nonspecific activation of blood. Vehicle or agonist solution (4 μL) was then added to each well: AA (0.03–0.6 mmol/L; Sigma, Poole, United Kingdom), Horm collagen (0.1–3 μg/mL; Nycomed, Linz, Austria), the protease-activated receptor 4 (PAR-4)–activating peptide AYPGKF amide (PAR4-AP, 50–100 μmol/L; Bachem, Bubendorf, Switzerland), and the stable TxA2 mimetic U46619 (0.1–10 μmol/L; Cayman Chemical Company, Ann Arbor, MI). To each well, 35 μL of PRP or whole blood was added, and the plate was then placed onto a heated plate shaker (Bioshake IQ, Q Instruments, Jena, Germany) at 37°C for 5 minutes mixing at 1200 rpm. Where appropriate, light transmission of each well was determined using a 96-well plate reader (SunriseTM, Tecan, Männedorf, Switzerland) at 595 nm. Alternatively, samples were diluted 1:5 with an acid–citrate–dextrose solution (5 mmol/L glucose, 6.8 mmol/L trisodium citrate, and 3.8 mmol/L citric acid) before individual platelet counts of each were determined by flow cytometry. Platelets were labeled with allophycocyanin-conjugated anti-CD41 (clone eBioMWReg30) for 30 minutes before further dilution 1:50 in PBS containing 0.1% formalin (Sigma, United Kingdom), 0.1% dextrose, and 0.2% BSA and addition of 104 CountBright absolute counting beads (Life Technologies). Labeled, diluted blood was then analyzed using a FACSCalibur flow cytometer (BD Biosciences, Oxford, United Kingdom).

Results

Next-Generation Sequencing of Plasma MiRNAs

To characterize circulating small RNAs, libraries of small RNAs were generated from PRP and PPP for next-generation sequencing. The presence of platelets and the absence of leukocyte contamination in PRP samples were verified by qPCR for ITGA2B and CD45, respectively (Online Figure IA). The quality of base calls is represented by consecutive boxplots for each position on the 100-nucleotide read (Online Figure IB). The total number of digital reads correlated with the amount of RNA obtained from PPP and PRP (rp=0.94). Two peaks were observed across all 4 samples, 1 at 22 to 23 nucleotides corresponding to miRNAs and another centered around the 32 to 33 nucleotide range (Figure 1A), which aligned predominantly to RNY4, a noncoding RNA, on chromosome 7. The 25 most abundant small RNAs in each sample are shown in Figure 1B. At a threshold of 0.4 reads per million and without distinguishing between isomiRs, 224 miRNAs were consistently identified in all samples (Online Table I). There was a good correlation for the number of reads per miRNA (Online Figure IC). Coverage of RNY4 was predominantly observed on the 5′ end, with some additional fragments from the 3′ end. Sequences derived from other RNY genes were also detected, although at much lower read counts: for RNY1 and RNY5, like RNY4, mostly, 5′ fragments were found; for RNY3, the 3′ fragments were more abundant (Online Figure II). Two hundred three of 224 miRNAs and both RNY4 fragments were detected at higher levels in PRP than PPP, suggesting that most circulating small RNAs are present in platelets (Online Figure III).
Figure 1. Next-generation sequencing of small RNAs in plasma. A, Representative histogram of sequence lengths for platelet-poor plasma (PPP) after processing for adapter removal and lower bound on length. B, Summary of the top 25 small RNAs (reads per million total reads) in each sample. YRNA fragments are highlighted in bold. PRP indicates platelet-rich plasma.

Effect of Antiplatelet Therapy

We have previously reported that antiplatelet therapy reduces plasma levels of platelet-related miRNAs, including miR-126 and miR-223.4 Ninety-two miRNAs were measured using custom-made Exiqon LNA qPCR plates. The plate layout has been published previously4 and is shown in Online Table II. In a principal component analysis, the interindividual variability in plasma miRNA profiles decreased with prolonged platelet inhibition4 (Figure 2A), reinforcing the concept that platelet activity is an important determinant of the plasma miRNA pool. To explore how different antiplatelet agents affect plasma miRNAs, the same 92 miRNAs were screened in a closely matched cohort of patients with ACS who were either on ASA only (n=8), ASA+clopidogrel (n=8), ASA+prasugrel (n=8), or ASA+ticagrelor (n=8) for 30 days after the acute event. In the principal component analysis, no clear separation was obtained between the different antiplatelet agents, but the least interpatient variability was observed in the ASA+prasugrel group (Figure 2B).
Figure 2. MicroRNAs (miRNAs) and antiplatelet therapy. Principal component (PC) analyses based on screening of 92 plasma miRNAs using custom-made quantitative polymerase chain reaction plates. A, Decreasing variability of plasma miRNA profiles in healthy volunteers after 2 and 3 weeks of platelet inhibition (n=6 at 4 time points). B, Effects of different antiplatelet agents in patients with acute coronary syndrome (ACS; n=8 per group). The tables show the variances of the PC1 and PC2. An F test was used to calculate the differences in variance; P values reflect the difference of variance compared with baseline (A) or 75 mg acetylsalicylic acid (ASA; B). BD indicates twice daily; and OD, once daily.

Correlation to Platelet Function in Patients With ACS

Next, platelet-related miRNAs (Online Figure IV) and abundant plasma YRNA fragments were measured by individual qPCR assays in the ACS patient cohort who had undergone detailed assessment of platelet function (n=125; Online Table III). The correlations of small RNAs to measurements of platelet function are shown in Figure 3. No correlation was found between miRNA and YRNA levels and optical aggregometry in response to AA or ADP. In contrast, significant associations were obtained for the VASP phosphorylation assays, for example, miR-223 (rp=0.28; P=0.002), miR-24 (rp=0.25; P=0.006), miR-191 (rp=0.24; P=0.009), RNY4 3′ (rp=0.23; P=0.012), miR-126 (rp=0.22; P=0.016), and RNY4 5′ (rp=0.21; P=0.025). For these miRNAs and YRNA fragments, the correlations with the VerifyNow P2Y12 assay were of similar strength (rp=0.18–0.35), but the statistical power was weaker in view of low numbers, and nominal significance was only noted for miR-126 (rp=0.35; P=0.033). No associations were obtained for miR-93, miR-106a, miR-146b, and miR-150.
Figure 3. Pearson correlation forest plot depicting associations of 11 microRNAs (miRNAs) and 2 YRNA fragments to platelet counts and platelet function tests in patients with acute coronary syndrome (n=125). Measurements of RNAs by individual Taqman assays were normalized to exogenous Cel-miR-39. Note that fewer samples were measured with the VerifyNow (n=40) compared with the vasodilator-stimulated phosphoprotein (VASP) aggregation assay (n=121). AA indicates arachidonic acid; and PRU, P2Y12 reaction units.

Evidence for Platelet Origin

Although all candidate miRNAs were present in platelets (Online Figure IV), some reflected platelet activation better than others. To determine to what extent platelets may contribute to circulating miRNA and YRNA levels, we reconstituted PPP with washed platelets. Platelets were isolated from PRP and spiked back into PPP corresponding to 5%, 50%, 100%, or 200% of the initial volume (Figure 4A). For miR-126 and miR-223, a significant linear increase was observed with increasing platelet content (Figure 4B). As expected, the liver-specific miRNA, miR-122, was not affected by addition of platelets. Notably, miR-126 showed the greatest dependency on platelets in comparison with 21 other miRNAs (Figure 4C). This spike-in experiment provides further evidence for platelets being a major source of circulating miRNAs, including miR-126, which was previously implicated to be of endothelial origin.1214 Levels of RNY4 fragments were also strongly affected by addition of platelets (Figure 4C). Unlike miRNAs, however, RNY4 was not associated with argonaute-2 complexes in MEG-01 cells, a human megakaryoblastic cell line (Online Figure V).
Figure 4. Platelet spike-in experiment. A, Schematic summary of workflow. B, Dependence of miR-126, miR-223, and miR-122 levels on platelet spike-in. C, P values for the dependence of levels of 22 microRNAs and 2 RNY4 fragments on platelet spike-in. RNAs marked in bold were tested in the acute coronary syndrome cohort. The grey line represents the significance threshold for a P value of 0.05 after Bonferroni correction. B and C, Results shown are derived from linear mixed models featuring fixed effects for platelet spike-in in categorical form (B) or in continuous form (C), random intercepts for subjects, and a general (unconstrained) covariance structure. PPP indicates platelet-poor plasma; and PRP, platelet-rich plasma.

MiR-126 and Platelet Function in the General Population

MiRNAs and RNY4 fragments were measured in the Bruneck study (n=669) and correlated to the platelet activation markers15 platelet factor 4 (PF4), pro-platelet basic protein (PPBP), and P-selectin as quantified by ELISA (Figure 5A). Substantial positive correlations of miRNAs and RNY4 fragments with all 3 platelet-activation markers were observed in plasma. These were more pronounced for the platelet-specific proteins PF4 and PPBP than for P-selectin, which is shed from platelets and endothelial cells.16,17 The liver-specific miRNA, miR-122, and other miRNAs, such as miR-150, showed no or considerably weaker correlations with platelet activation markers (Figure 5A). Accordingly, there was a striking correlation between the platelet dependency of miRNAs in the spike experiment and the correlations of miRNAs with platelet activation markers in the general population (r=0.92–0.94; Figure 5B; Online Figure VI). RNY4 fragments were strongly correlated with platelet-derived plasma miRNAs (Figure 5C), suggesting a common platelet origin.
Figure 5. Small RNAs and platelet proteins in the Bruneck cohort. A, Association of plasma microRNAs (miRNAs) and YRNA fragments with 3 platelet activation markers (platelet factor 4 [PF4], pro-platelet basic protein [PPBP], and P-selectin [SELP]) in the population-based Bruneck study (n=669). B, Relationship between the dependency of small RNA levels on platelets in the spike-in experiment (x axis; Figure 4) and the correlation of small RNA levels with platelet-derived protein concentrations (PF4 and PPBP) in the general population (y axis; A) for 16 miRNAs and 2 RNY4 fragments. Lines are Deming regression lines, and r denotes Pearson correlation with 95% confidence interval. C, Association of plasma YRNA fragments with plasma miRNAs measured in the Bruneck cohort (n=669). A and C, Tile color codes for direction and magnitude of correlation, whereas tile text gives its sign and first 2 decimal digits.
Although circulating miRNAs have been shown to be affected by disease state, cardiovascular risk factors, and drug treatment, the influence of genetic variability, especially SNPs, on miRNA expression and function is poorly understood. Thus far, only 1 functional SNP has been described for miR-126 (Figure 6A): the primary sequence of human miR-126 contains a SNP (dbSNP: rs4636297) downstream of the pre-miR sequence.8 The genotype of this SNP has been shown to affect the processing of pri-miR-126: pri-miR-126 encoded by the major G allele is processed to a lesser extent than pri-miR-126 encoded by the minor A allele. In the Bruneck cohort (Online Table IV), there was a trend toward higher miR-126 levels in individuals homozygous for the minor allele (AA genotype), which facilitates processing of miR-126, compared with the GA+GG genotypes in serum (+4.4%; P=0.050) and in plasma (+6.9%; P=0.099; Online Figure VII). Importantly, the AA genotype was associated with higher plasma levels of platelet activation markers: PF4 (P=0.002), PPBP (P<0.001), and P-selectin (P=0.099; Figure 6B). To compare the effect on platelet proteins to other plasma proteins, we measured 219 proteins, using mass spectrometry for detection of high-abundant proteins (n=84) and proximity extension assays for detection of low-abundant proteins (n=132), as well as ELISA (n=3). Of 61 proteins whose levels are significantly associated with the AA genotype (Online Figure VIII), PPBP and PF4 showed the second and third highest fold change (mean ratios, AA versus GA+GG of 1.49 and 1.47, respectively; Online Table V). The proteins associated with the AA genotype also showed an enrichment of the gene ontology term annotation platelet activation (P=0.019; Online Figure VIII).
Figure 6. Association of rs4636297 with plasma markers of platelet activation. A, Schematic representation of pri-miR-126. Cleavage sites for Drosha/DGCR8 and Dicer are indicated in gray. The single-nucleotide polymorphism (SNP) rs4636297 is located downstream of the stem loop (pre-miR-126). Pri-miR-126 carrying the major G genotype is less efficiently processed than the minor A genotype. B, Plasma levels of platelet activation markers are associated with the rs4636297 genotype in the Bruneck cohort (n=628). P values are shown for 1-way ANOVA. PF4 indicates platelet factor 4; PPBP, pro-platelet basic protein; and SELP, P-selectin.

MiR-126 and Platelet Function in Mice

The genetic associations of a SNP within miR-126 with parameters of platelet function prompted us to further investigate the role of miR-126 in platelet activation. It has recently been reported that oligonucleotides with a phosphorothioate backbone modification (minimum length of 18 nt) can activate platelets.18 Thus, we incubated human PRP with fully phosphorothioate-modified LNAs, as well as antagomiRs with 2 and 4 phosphorothioate-modified nucleotides at the 5′ and 3′ ends, respectively. None of the tested oligonucleotides with cholesterol modification induced platelet aggregation (Online Figure IX). Next, C57BL/6J mice were injected with PBS, a control antagomiR or antagomiR-126-3p at 25 mg/kg IP, for 3 consecutive days and euthanized at day 7 (Figure 7A). The knockdown of miR-126 was confirmed in blood samples by qPCR (Online Figure X). Whole blood was treated with different concentrations of agonists: AA, collagen, PAR4-AP, and the TxA2 analog U46619 (Figure 7B). The aggregation response to AA and U46619 was significantly reduced in platelets from mice treated with antagomiR-126-3p. At antagomiR doses of 25 mg/kg, however, there was no effect on platelet aggregation in response to collagen or PAR4-AP. Thus, we repeated the experiment with a higher antagomiR concentration (40 mg/kg IP). Again, the inhibition of miR-126 was confirmed by qPCR (Online Figure X). To further address the effect of miR-126 in platelets and to minimize a potential influence of other cell types, PRP was used instead of whole blood for platelet aggregation. AntagomiRs against miR-126-3p blocked platelet aggregation induced by 0.6 mmol/L AA (Figure 7C) and led to a significant reduction of aggregation in response to 50 μmol/L PAR4-AP. The attenuated response to 0.3 μg/mL collagen failed to reach statistical significance. No differences in platelet aggregation were observed at higher agonist concentrations of PAR4-AP (100 μmol/L) and collagen (3 μg/mL; data not shown).
Figure 7. Platelet function in mice treated with antagomiR against miR-126-3p. A, 8-week-old male C57BL/6J mice were injected with PBS, control antagomiR, or antagomiR-126-3p for 3 consecutive days and euthanized on day 7 for platelet function tests. B, Platelet aggregation was measured in whole blood from mice injected intraperitoneally with 25 mg antagomiR/kg in response to different concentrations of the indicated agonists. Asterisks denote significant difference in a 2-way ANOVA with Bonferroni post test (*P<0.05; **P<0.01). C, Platelet aggregation was assessed in platelet-rich plasma (PRP) in mice injected intraperitoneally with 40 mg antagomiR/kg. Asterisks denote significant difference in a 1-way ANOVA with Bonferroni post test (*P<0.05). B and C, Data are shown as mean±SEM; n=4 per condition. AA, arachidonic acid; PAR4-AP, PAR-4–activating peptide AYPGKF amide; and TxA2, thromboxane A2.

Targets of MiR-126 in Platelets

To explore potential mechanisms, we measured the expression levels of known miR-126 targets with a role in platelet function alongside platelet-related genes (Online Table VI). We observed reduced expression of the P2Y12 receptor in whole blood of antagomiR-126-3p–treated mice (Figure 8A). In a human megakaryoblastic cell line (MEG-01), mimics or LNA inhibitors of miR-126-3p (Figure 8B and 8C; Online Figure XI) regulated ADAM9, a confirmed target of miR-126 that has been shown to affect collagen-induced platelet aggregation (Figure 8D).19 Thus, miR-126 alters gene expression in megakaryocytes.
Figure 8. Effects of miR-126 in mice and MEG-01 cells. A, Gene expression was analyzed in whole blood from mice treated with 25 or 40 mg/kg antagomiR; n=8 per group. Gene expression was normalized to Actb, Gapdh, and Ppia. Gene expression was analyzed in MEG-01 cells transfected with mimic-126 (B) or LNA-126 (C) and the respective controls. Gene expression was normalized to GAPDH and SP1. Validated targets of miR-126 are shown in green, predicted targets are shown in blue, and platelet-enriched genes are shown in orange. B and C, n=4 per condition. Graphs represent mean+SEM, asterisks denote statistical significance in a 2-way ANOVA with Bonferroni post test (*P<0.05; **P<0.01; ***P<0.001). D, Schematic illustration of potential miR-126–dependent effects on platelet function. Ctrl indicates control; and TxA2, thromboxane A2.

Discussion

In this study, miRNA measurements were performed in 669 subjects of a population-based study, as well as in 125 patients with ACS. The study correlates miRNAs with platelet activation markers in the general population and with the residual platelet activity in patients with ACS on antiplatelet therapy. Most but not all abundant platelet miRNAs were positively correlated with the VerifyNow P2Y12 and VASP assays, which are standardized assays for assessing the effects of P2Y12 inhibitors. In addition, we show an association of plasma YRNAs with platelets and that inhibition of miR-126 attenuates platelet aggregation in response to low but not high agonist concentrations.

Platelet Dependency of Small RNAs in Plasma

To demonstrate the platelet dependency of our candidate miRNAs, washed platelets were isolated from PRP and spiked back into PPP. This spike-in experiment rules out a cellular contamination, which can hamper direct comparisons between PRP and PPP and provide further evidence that the selected miRNAs, including miR-126, are genuine platelet miRNAs.4 Furthermore, we provide evidence that RNY4 fragments in plasma also originate from platelets. YRNA fragments are enriched in exosomes.20,21 Our next-generation sequencing data confirm that RNY4 fragments are abundant in plasma.22 Unlike miRNAs, however, RNY4 fragments were not present in the Ago2 (argonaute 2 protein) complexes of MEG-01 cells.23,24 Recently, YRNA fragments were implicated as biomarkers for coronary artery disease.25 Changes in circulating levels were attributed to apoptotic macrophages, which generate and secrete YRNA fragments.25 In contrast, our data suggest that platelets are a major source of circulating YRNA fragments, and cellular origin has to be taken into account if YRNA fragments are considered as potential biomarkers for cardiovascular disease. The function of YRNA fragments remains unknown, but it has been suggested that YRNA fragments, including RNY4 5′, act as small guide RNAs for tRNase ZL, forming a tRNA-like duplex with a target RNA, thereby enabling its hydrolysis.26 This mechanism has been shown for synthetic target RNAs in vitro, and it remains to be seen whether YRNA fragments can act as guide RNAs for tRNase ZL in vivo.

Platelet MiRNAs in Patients Post ACS

Plasma was taken 30 days after the acute event when the inflammation associated with the acute injury has receded and antiplatelet drugs had been administered for a month. Importantly, none of the patients with ACS had received heparin at the time of sampling.27,28 Levels of miR-126 and other platelet-related miRNAs showed a positive correlation with the VerifyNow P2Y12 and VASP phosphorylation assays. VerifyNow P2Y12 is a commercial assay that uses whole blood to monitor P2Y12 inhibition.29 The VASP assay is considered to be among the most specific assays to monitor P2Y12 inhibition because it does not rely on coactivation of the P2Y1 receptor by ADP.30 In contrast, optical aggregometry is performed on isolated platelets. Theoretically, aggregometry responses to ADP and the VASP, and Verify Now P2Y12 assays assess the same parameter: P2Y12 receptor activation. However, the variability of the aggregometry results in isolated platelets being higher compared with tests performed in whole blood may explain the loss of associations with plasma miRNAs. Previous studies have suggested that VerifyNow P2Y12 and VASP assays are more discriminating of clopidogrel response and its genetic influences compared with light transmittance aggregometry, and there are only moderate correlations between the different platelet function assays.30,31 Moreover, aspirin is such a potent inhibitor of the platelet response to AA that any variation between patients is expected to be low. Aspirin inhibits the production of TxA2. Clopidogrel and prasugrel act by blocking the platelet P2Y12 receptor.5,29 Thus, their mechanisms are complementary. Clopidogrel and prasugrel are both thienopyridine prodrugs that are converted via hepatic CYP (cytochrome P450 enzyme) isoenzymes to their active metabolite, but prasugrel is more efficiently converted to its active form and so achieves more reliable P2Y12 inhibition. Ticagrelor is a novel P2Y12 receptor antagonist, but its clinical profile, both in terms of efficacy and adverse events, differs from that of the thienopyridine prodrugs.32 Ticagrelor is also supposed to have a dual mode of action as its P2Y12 antagonism is complemented by inhibition of adenosine cell uptake via inhibition of the equilibrative nucleoside transporter 1, thereby increasing extracellular adenosine level and mediating adenosine-receptor activation.32

Platelet MiRNAs and Platelet Activation in the General Population

It is currently unclear to what extent platelet miRNAs are mechanistically involved in platelet activation. A SNP (rs4636297) in the miR-126 locus has previously been demonstrated to affect the expression of mature miR-126 in cells overexpressing the different variants of pri-miR-126. An effect on endogenous miR-126 levels has not been investigated thus far.8 In plasma and serum from the Bruneck cohort, we observed a trend toward higher levels of circulating miR-126 in individuals carrying the minor allele (AA genotype), which facilitates the processing of pri-miR-126. Importantly, the SNP genotype affected the plasma concentrations of 3 platelet activation markers: PF4 (P=0.002), PPBP (P<0.001), and P-selectin (P=0.099) were all positively correlated with the AA genotype. Moreover, for proteins showing an association with the AA genotype in a panel of 219 plasma proteins measured in the Bruneck cohort, there was a significant enrichment of proteins linked to platelet activation (P=0.019; Online Figure VIII). These genetic associations, however, await confirmation in independent cohorts. MiR-126 is abundant in endothelial cells and platelets.3 Although miR-126 is known to be an important regulator of endothelial cell function,12,33 its role in platelets is unknown. It is, therefore, conceivable that higher miR-126 levels in individuals with the AA genotype influence platelet activity either directly by changing platelet function or indirectly by affecting endothelial cells. Recently, the A allele of SNP rs4636297 has been shown to be associated with sight-threatening diabetic retinopathy in patients with type II diabetes mellitus,34 corroborating the relevance of genetic variability of miRNAs for susceptibility to disease.

MiR-126 and Platelet Aggregation in Mice

Despite numerous reports on miRNAs, surprisingly little is known about their function in platelets. MiR-223 targets the P2Y12 receptor.35 MiR-223–deficient mice form larger thrombi and have a delayed clot retraction compared with wild-type mice.36 Platelets of miR-223–deficient mice display increased aggregation in response to thrombin and collagen but not to fibronectin and the TxA2 analog U44619. MiR-96 regulates VAMP8/endobrevin, a protein involved in platelet granule secretion that is upregulated in hyper-reactive platelets.37 Our experiments demonstrate that inhibition of miR-126 in mice attenuates platelet aggregation in response to AA, U44619, and PAR4-AP. Collagen-induced platelet aggregation may also be affected, but we did not obtain statistical significance, probably because of low numbers. Platelet aggregation was reduced in whole blood and in PRP. Thus, the involvement of other cell types is less likely. Moreover, MEG-01 cells express miR-126 in abundance, and mimics of miR-126 reduced the expression of ADAM9, whereas inhibition of miR-126 had the opposite effect. ADAM9 is a predicted and experimentally confirmed target of miR-126 that attenuates the adhesion of platelets to collagen.19 Given its function as a protease of the ADAM family, it may alter the platelet response by cleaving membrane proteins. In whole blood from antagomiR-treated mice, ADAM9 was not differentially expressed. This is expected because ADAM9 is present in many other blood cells that lack miR-126 and therefore are not affected. Notably, the expression of the P2Y12 receptor was reduced in the blood of antagomiR-126-3p–treated mice. P2Y12 levels are much higher in platelets than in leukocytes38 and erythrocytes.39 Thus, the latter finding may, at least in part, explain why platelets from antagomiR-126-3p–treated mice display an attenuated aggregation response: the aggregation responses to strong platelet agonists, such as collagen, thrombin receptor–activating peptides, and particularly, either TxA2 (generated from AA) or its mimetic U46619, are amplified by ADP, which is released from the platelet dense granules in response to the agonists and activates the P2Y12 receptor. On the other hand, higher concentrations of these agonists or less effective P2Y12 inhibition or deficiency allow a more robust platelet aggregation response. P2Y12 activation may also contribute to the generation of TxA2 from AA.40

Study Limitations

Causality cannot be inferred from associations of miRNAs with platelet function tests in patients with ACS. Although recent studies implicated miR-126 in atherogenesis,41,42 and miR-223 has been used for categorization of patients as responder and nonresponder to the P2Y12 inhibitor clopidogrel,43 larger cohorts with prolonged follow-up are required to determine whether miRNA levels are associated with clinical outcomes in patients with ACS. Because the P2Y12 receptor plays such an important role in platelet reactivity, relationships between the extent of P2Y12 receptor inhibition and miRNA levels have to be further explored in future studies. Similarly, the reported association of a SNP for miR-126 with platelet activation markers requires replication in independent cohorts. MiR-126 is also abundant in endothelial cells, and indirect effects on platelets cannot be excluded. On the other hand, gene expression in MEG-01 cells may be subject to different regulation mechanisms than in primary megakaryocytes and platelets, that is, in undifferentiated MEG-01 cells, P2Y12 expression is not detectable.

Conclusions

Numerous studies have demonstrated the importance of platelet reactivity in the risk of clinical events, such as stent thrombosis after percutaneous coronary intervention or recurrent arterial thrombotic events after ACS. Exciting opportunities exist to further pursue platelet miRNAs as potential biomarkers for treatment response in patients with ACS.44 MiRNAs can be measured in frozen samples, which could offer a potential advantage compared with other platelet function tests currently available. Besides their biomarker potential, some miRNAs, such as miR-126 and miR-223,36 may also regulate platelet reactivity.

Acknowledgments

We thank Dr Ursula Mayr for assistance with the in vivo experiments.

Novelty and Significance

What Is Known?

Plasma microRNAs (miRNAs) are highly correlated.
Platelets contain and release miRNAs.
Platelet inhibition reduces miRNA levels in platelet-poor plasma.

What New Information Does This Article Contribute?

Besides miRNAs, YRNA fragments in plasma are also platelet derived.
Platelet miRNAs and YRNA fragments correlate with indices of platelet function in patients on dual antiplatelet therapy.
MiRNA-126 alters platelet activity.
This study provides evidence for platelets being a source of miRNAs and YRNA fragments in plasma. A strong platelet dependency of miRNAs and YRNA fragments was observed in a spike-in experiment. There was also a striking correlation of miRNAs and YRNA fragments with platelet activation markers in the general population. Plasma miRNA and YRNA levels are associated with residual platelet activity in patients on dual antiplatelet therapy. MiRNA-126, previously considered to be endothelial specific, is present in platelets and in a human megakaryoblastic cell line. A single-nucleotide polymorphism that facilitates processing of miRNA-126 increases plasma levels of platelet activation markers. Treatment with antagomiRs to miRNA-126 reduces platelet activation in mice. MiRNAs may not just be markers of platelet activity but also alter their function, most probably by influencing gene expression in megakaryocytes.

Footnote

Nonstandard Abbreviations and Acronyms

AA
arachidonic acid
ACS
acute coronary syndrome
ASA
acetylsalicylic acid
MiRNA
microRNA
PAR4-AP
PAR-4–activating peptide AYPGKF amide
PF4
platelet factor 4
PPBP
pro-platelet basic protein
PPP
platelet-poor plasma
PRP
platelet-rich plasma
qPCR
quantitative real-time polymerase chain reaction
SNP
single-nucleotide polymorphism
TxA2
thromboxane A2
VASP
vasodilator-stimulated phosphoprotein

Supplemental Material

File (305663r4_online.pdf)

References

1.
Olson EN. MicroRNAs as therapeutic targets and biomarkers of cardiovascular disease. Sci Transl Med. 2014;6:239.
2.
Zampetaki A, Willeit P, Drozdov I, Kiechl S, Mayr M. Profiling of circulating microRNAs: from single biomarkers to re-wired networks. Cardiovasc Res. 2012;93:555–562. doi: 10.1093/cvr/cvr266.
3.
Zampetaki A, Willeit P, Tilling L, Drozdov I, Prokopi M, Renard JM, Mayr A, Weger S, Schett G, Shah A, Boulanger CM, Willeit J, Chowienczyk PJ, Kiechl S, Mayr M. Prospective study on circulating MicroRNAs and risk of myocardial infarction. J Am Coll Cardiol. 2012;60:290–299. doi: 10.1016/j.jacc.2012.03.056.
4.
Willeit P, Zampetaki A, Dudek K, et al. Circulating microRNAs as novel biomarkers for platelet activation. Circ Res. 2013;112:595–600. doi: 10.1161/CIRCRESAHA.111.300539.
5.
Joshi RR, Hossain R, Morton AC, Ecob R, Judge HM, Wales C, Walker JV, Karunakaran A, Storey RF. Evolving pattern of platelet P2Y12 inhibition in patients with acute coronary syndromes. Platelets. 2014;25:416–422. doi: 10.3109/09537104.2013.836175.
6.
Wallentin L, Becker RC, Budaj A, et al.; PLATO Investigators. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–1057. doi: 10.1056/NEJMoa0904327.
7.
Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, Neumann FJ, Ardissino D, De Servi S, Murphy SA, Riesmeyer J, Weerakkody G, Gibson CM, Antman EM; TRITON-TIMI 38 Investigators. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482.
8.
Harnprasopwat R, Ha D, Toyoshima T, Lodish H, Tojo A, Kotani A. Alteration of processing induced by a single nucleotide polymorphism in pri-miR-126. Biochem Biophys Res Commun. 2010;399:117–122. doi: 10.1016/j.bbrc.2010.07.009.
9.
Kiechl S, Lorenz E, Reindl M, Wiedermann CJ, Oberhollenzer F, Bonora E, Willeit J, Schwartz DA. Toll-like receptor 4 polymorphisms and atherogenesis. N Engl J Med. 2002;347:185–192. doi: 10.1056/NEJMoa012673.
10.
Woulfe D, Jiang H, Morgans A, Monks R, Birnbaum M, Brass LF. Defects in secretion, aggregation, and thrombus formation in platelets from mice lacking Akt2. J Clin Invest. 2004;113:441–450. doi: 10.1172/JCI20267.
11.
Armstrong PC, Kirkby NS, Chan MV, Finsterbusch M, Hogg N, Nourshargh S, Warner TD. Novel whole blood assay for phenotyping platelet reactivity in mice identifies ICAM-1 as a mediator of platelet-monocyte interaction. Blood. 2015;126:e11–e18. doi: 10.1182/blood-2015-01-621656.
12.
Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15:261–271. doi: 10.1016/j.devcel.2008.07.002.
13.
Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, Weber M, Hamm CW, Röxe T, Müller-Ardogan M, Bonauer A, Zeiher AM, Dimmeler S. Circulating microRNAs in patients with coronary artery disease. Circ Res. 2010;107:677–684. doi: 10.1161/CIRCRESAHA.109.215566.
14.
Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, Mayr A, Weger S, Oberhollenzer F, Bonora E, Shah A, Willeit J, Mayr M. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res. 2010;107:810–817. doi: 10.1161/CIRCRESAHA.110.226357.
15.
Ferroni P, Riondino S, Vazzana N, Santoro N, Guadagni F, Davì G. Biomarkers of platelet activation in acute coronary syndromes. Thromb Haemost. 2012;108:1109–1123. doi: 10.1160/TH12-08-0550.
16.
Fijnheer R, Frijns CJ, Korteweg J, Rommes H, Peters JH, Sixma JJ, Nieuwenhuis HK. The origin of P-selectin as a circulating plasma protein. Thromb Haemost. 1997;77:1081–1085.
17.
Ferroni P, Martini F, Riondino S, La Farina F, Magnapera A, Ciatti F, Guadagni F. Soluble P-selectin as a marker of in vivo platelet activation. Clin Chim Acta. 2009;399:88–91. doi: 10.1016/j.cca.2008.09.018.
18.
Flierl U, Nero TL, Lim B, et al. Phosphorothioate backbone modifications of nucleotide-based drugs are potent platelet activators. J Exp Med. 2015;212:129–137. doi: 10.1084/jem.20140391.
19.
Cominetti MR, Martin AC, Ribeiro JU, Djaafri I, Fauvel-Lafève F, Crépin M, Selistre-de-Araujo HS. Inhibition of platelets and tumor cell adhesion by the disintegrin domain of human ADAM9 to collagen I under dynamic flow conditions. Biochimie. 2009;91:1045–1052. doi: 10.1016/j.biochi.2009.05.012.
20.
Tosar JP, Gámbaro F, Sanguinetti J, Bonilla B, Witwer KW, Cayota A. Assessment of small RNA sorting into different extracellular fractions revealed by high-throughput sequencing of breast cell lines. Nucleic Acids Res. 2015;43:5601–5616. doi: 10.1093/nar/gkv432.
21.
van Balkom BW, Eisele AS, Pegtel DM, Bervoets S, Verhaar MC. Quantitative and qualitative analysis of small RNAs in human endothelial cells and exosomes provides insights into localized RNA processing, degradation and sorting. J Extracell Vesicles. 2015;4:26760.
22.
Dhahbi JM, Spindler SR, Atamna H, Boffelli D, Mote P, Martin DI. 5’-YRNA fragments derived by processing of transcripts from specific YRNA genes and pseudogenes are abundant in human serum and plasma. Physiol Genomics. 2013;45:990–998. doi: 10.1152/physiolgenomics.00129.2013.
23.
Thomson DW, Pillman KA, Anderson ML, Lawrence DM, Toubia J, Goodall GJ, Bracken CP. Assessing the gene regulatory properties of Argonaute-bound small RNAs of diverse genomic origin. Nucleic Acids Res. 2015;43:470–481. doi: 10.1093/nar/gku1242.
24.
Nicolas FE, Hall AE, Csorba T, Turnbull C, Dalmay T. Biogenesis of Y RNA-derived small RNAs is independent of the microRNA pathway. FEBS Lett. 2012;586:1226–1230. doi: 10.1016/j.febslet.2012.03.026.
25.
Repetto E, Lichtenstein L, Hizir Z, et al. RNY-derived small RNAs as a signature of coronary artery disease. BMC Med. 2015;13:259. doi: 10.1186/s12916-015-0489-y.
26.
Ninomiya S, Kawano M, Abe T, Ishikawa T, Takahashi M, Tamura M, Takahashi Y, Nashimoto M. Potential small guide RNAs for tRNase ZL from human plasma, peripheral blood mononuclear cells, and cultured cell lines. PLoS One. 2015;10:e0118631. doi: 10.1371/journal.pone.0118631.
27.
Kaudewitz D, Lee R, Willeit P, McGregor R, Markus HS, Kiechl S, Zampetaki A, Storey RF, Channon KM, Mayr M. Impact of intravenous heparin on quantification of circulating microRNAs in patients with coronary artery disease. Thromb Haemost. 2013;110:609–615. doi: 10.1160/TH13-05-0368.
28.
Boeckel JN, Thomé CE, Leistner D, Zeiher AM, Fichtlscherer S, Dimmeler S. Heparin selectively affects the quantification of microRNAs in human blood samples. Clin Chem. 2013;59:1125–1127. doi: 10.1373/clinchem.2012.199505.
29.
Grove EL, Storey RF, Würtz M. Platelet function testing in atherothrombotic disease. Curr Pharm Des. 2012;18:5379–5391.
30.
Tantry US, Bliden KP, Wei C, Storey RF, Armstrong M, Butler K, Gurbel PA. First analysis of the relation between CYP2C19 genotype and pharmacodynamics in patients treated with ticagrelor versus clopidogrel: the ONSET/OFFSET and RESPOND genotype studies. Circ Cardiovasc Genet. 2010;3:556–566. doi: 10.1161/CIRCGENETICS.110.958561.
31.
Bouman HJ, Parlak E, van Werkum JW, Breet NJ, ten Cate H, Hackeng CM, ten Berg JM, Taubert D. Which platelet function test is suitable to monitor clopidogrel responsiveness? A pharmacokinetic analysis on the active metabolite of clopidogrel. J Thromb Haemost. 2010;8:482–488. doi: 10.1111/j.1538-7836.2009.03733.x.
32.
Gaglia MA Ticagrelor and the prevention of cardiovascular mortality: more than just platelet inhibition? Heart. 2014;100:1739–1740. doi: 10.1136/heartjnl-2014-306285.
33.
Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, Ivey KN, Bruneau BG, Stainier DY, Srivastava D. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 2008;15:272–284. doi: 10.1016/j.devcel.2008.07.008.
34.
McAuley AK, Dirani M, Wang JJ, Connell PP, Lamoureux EL, Hewitt AW. A genetic variant regulating miR-126 is associated with sight threatening diabetic retinopathy. Diab Vasc Dis Res. 2015;12:133–138. doi: 10.1177/1479164114560160.
35.
Landry P, Plante I, Ouellet DL, Perron MP, Rousseau G, Provost P. Existence of a microRNA pathway in anucleate platelets. Nat Struct Mol Biol. 2009;16:961–966. doi: 10.1038/nsmb.1651.
36.
Elgheznawy A, Shi L, Hu J, Wittig I, Laban H, Pircher J, Mann A, Provost P, Randriamboavonjy V, Fleming I. Dicer cleavage by calpain determines platelet microRNA levels and function in diabetes. Circ Res. 2015;117:157–165. doi: 10.1161/CIRCRESAHA.117.305784.
37.
Kondkar AA, Bray MS, Leal SM, Nagalla S, Liu DJ, Jin Y, Dong JF, Ren Q, Whiteheart SW, Shaw C, Bray PF. 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.
38.
Li D, Wang Y, Zhang L, Luo X, Li J, Chen X, Niu H, Wang K, Sun Y, Wang X, Yan Y, Chai W, Gartner TK, Liu J. Roles of purinergic receptor P2Y, G protein-coupled 12 in the development of atherosclerosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2012;32:e81–e89. doi: 10.1161/ATVBAHA.111.239095.
39.
Wang L, Olivecrona G, Götberg M, Olsson ML, Winzell MS, Erlinge D. ADP acting on P2Y13 receptors is a negative feedback pathway for ATP release from human red blood cells. Circ Res. 2005;96:189–196. doi: 10.1161/01.RES.0000153670.07559.E4.
40.
Armstrong PC, Leadbeater PD, Chan MV, Kirkby NS, Jakubowski JA, Mitchell JA, Warner TD. In the presence of strong P2Y12 receptor blockade, aspirin provides little additional inhibition of platelet aggregation. J Thromb Haemost. 2011;9:552–561. doi: 10.1111/j.1538-7836.2010.04160.x.
41.
Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med. 2011;17:1410–1422. doi: 10.1038/nm.2538.
42.
Zampetaki A, Mayr M. MicroRNAs in vascular and metabolic disease. Circ Res. 2012;110:508–522. doi: 10.1161/CIRCRESAHA.111.247445.
43.
Shi R, Ge L, Zhou X, Ji WJ, Lu RY, Zhang YY, Zeng S, Liu X, Zhao JH, Zhang WC, Jiang TM, Li YM. Decreased platelet miR-223 expression is associated with high on-clopidogrel platelet reactivity. Thromb Res. 2013;131:508–513. doi: 10.1016/j.thromres.2013.02.015.
44.
Mayr M, Zampetaki A, Willeit P, Willeit J, Kiechl S. MicroRNAs within the continuum of postgenomics biomarker discovery. Arterioscler Thromb Vasc Biol. 2013;33:206–214. doi: 10.1161/ATVBAHA.112.300141.

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research

Relative frequency of noncardiomyocytes in the mouse heart. Endothelial cells (red; isolectin B4), visualized in a confocal stack, are the most abundant. Fibroblasts (green; Col1A1GFPTg and nuclear PDGFRaGFP), pericytes (blue; NG2), and leukocytes (yellow; Cx3cr1-GFP, CD45, Mrc1, MHCII, and B220) comprise the remaining predominant cell populations. See related article, page 400.

Circulation Research
Pages: 420 - 432
PubMed: 26646931

Versions

You are viewing the most recent version of this article.

History

Received: 31 December 2014
Revision received: 1 December 2015
Accepted: 8 December 2015
Published online: 8 December 2015
Published in print: 5 February 2016

Permissions

Request permissions for this article.

Keywords

  1. acute coronary syndrome
  2. biomarkers
  3. blood platelet
  4. micro-RNAs
  5. polymorphism, single nucleotide

Subjects

Authors

Affiliations

Dorothee Kaudewitz*
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Philipp Skroblin*
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Lukas H. Bender
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Temo Barwari
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Peter Willeit
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Raimund Pechlaner
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Nicholas P. Sunderland
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Karin Willeit
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Allison C. Morton
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Paul C. Armstrong
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Melissa V. Chan
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Ruifang Lu
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Xiaoke Yin
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Filipe Gracio
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Katarzyna Dudek
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Sarah R. Langley
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Anna Zampetaki
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Emanuele de Rinaldis
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Shu Ye
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Timothy D. Warner
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Alka Saxena
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Stefan Kiechl
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Robert F. Storey
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).
Manuel Mayr
From the Cardiovascular Division, King’s British Heart Foundation Centre (D.K., P.S., L.H.B., T.B., N.P.S., R.L., X.Y., K.D., S.R.L., A.Z., M.M.) and Biomedical Research Centre (F.G., E.d.R., A.S.), King’s College London, London, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom (P.W.); Department of Neurology, Medical University Innsbruck, Innsbruck, Austria (P.W., R.P., K.W., S.K.); Cardiothoracic Centre, Sheffield Teaching Hospitals NHS Foundation Trust (A.C.M.) and Department of Cardiovascular Science (R.F.S.), University of Sheffield, Sheffield, United Kingdom; William Harvey Research Institute, Queen Mary University of London, London, United Kingdom (P.C.A., M.V.C., T.D.W.); and Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom (S.Y.).

Notes

In November 2015, the average time from submission to first decision for all original research papers submitted to Circulation Research was 15.52 days.
*
These authors contributed equally to this article.
The online-only Data Supplement is available with this article at http://circres.ahajournals.org/lookup/suppl/doi:10.1161/CIRCRESAHA.114.305663/-/DC1.
Correspondence to Manuel Mayr, MD, PhD, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Ln, London SE5 9NU, United Kingdom. E-mail [email protected]

Disclosures

King’s College London filed and licensed patent applications related to circulating microRNAs as biomarkers. R. F. Storey reports receiving research grants, honoraria, or consultancy fees from AstraZeneca, Aspen, ThermoFisher Scientific, Eli Lilly/Daiichi Sankyo, Merck, Accumetrics, Novartis, Correvio, PlaqueTec, Sanofi-Aventis, Medscape, Regeneron, Roche, and The Medicines Company. D. Kaudewitz was supported by a stipendium of the Studienstiftung des Deutschen Volkes. P. Willeit is an Erwin Schrödinger fellow in Epidemiology sponsored by the Austrian Science Fund (J 3679-B13). A. Zampetaki and M. Mayr are supported by an Intermediate Fellowship and a Senior Research Fellowship from the British Heart Foundation, respectively. The other authors report no conflicts.

Sources of Funding

This research was conducted with support from AstraZeneca UK Limited. This study was also supported by the National Institute for Health Research Biomedical Research Centre based at Guy’s and St Thomas’ National Health Service Foundation Trust and King’s College London in partnership with King’s College Hospital, the Fondation Leducq (MIRVAD), Diabetes UK, the Juvenile Diabetes Research Foundation, and by an excellence initiative (Competence Centers for Excellent Technologies) of the Austrian Research Promotion Agency FFG: Research Center of Excellence in Vascular Ageing—Tyrol, VASCage (K-Project no. 843536) funded by the BMVIT, BMWFW, the Wirtschaftsagentur Wien, and the Standortagentur Tirol.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Fragments derived from non-coding RNAs: how complex is genome regulation?, Genome, 67, 9, (292-306), (2024).https://doi.org/10.1139/gen-2023-0136
    Crossref
  2. microRNAs and thrombo-inflammation: relationship in sight, Current Opinion in Hematology, 31, 3, (140-147), (2024).https://doi.org/10.1097/MOH.0000000000000803
    Crossref
  3. Intraplatelet miRNA-126 regulates thrombosis and its reduction contributes to platelet inhibition, Cardiovascular Research, 120, 13, (1622-1635), (2024).https://doi.org/10.1093/cvr/cvae138
    Crossref
  4. Prothymosin α accelerates dengue virus-induced thrombocytopenia, iScience, 27, 1, (108422), (2024).https://doi.org/10.1016/j.isci.2023.108422
    Crossref
  5. RNA interference-based therapies for atherosclerosis: Recent advances and future prospects, RNA Therapeutics Part B, (1-43), (2024).https://doi.org/10.1016/bs.pmbts.2023.12.009
    Crossref
  6. MicroRNAs and Cardiovascular Disease Risk, Current Cardiology Reports, 26, 2, (51-60), (2024).https://doi.org/10.1007/s11886-023-02014-1
    Crossref
  7. Technically Challenging Percutaneous Interventions of Chronic Total Occlusions Are Associated with Enhanced Platelet Activation, Journal of Clinical Medicine, 12, 21, (6829), (2023).https://doi.org/10.3390/jcm12216829
    Crossref
  8. Platelets and the Atherosclerotic Process: An Overview of New Markers of Platelet Activation and Reactivity, and Their Implications in Primary and Secondary Prevention, Journal of Clinical Medicine, 12, 18, (6074), (2023).https://doi.org/10.3390/jcm12186074
    Crossref
  9. Ticagrelor Resistance in Cardiovascular Disease and Ischemic Stroke, Journal of Clinical Medicine, 12, 3, (1149), (2023).https://doi.org/10.3390/jcm12031149
    Crossref
  10. The Novel Role of Noncoding RNAs in Modulating Platelet Function: Implications in Activation and Aggregation, International Journal of Molecular Sciences, 24, 8, (7650), (2023).https://doi.org/10.3390/ijms24087650
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Association of MicroRNAs and YRNAs With Platelet Function
Circulation Research
  • Vol. 118
  • No. 3

Purchase access to this journal for 24 hours

Circulation Research
  • Vol. 118
  • No. 3
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Share article link

Share

Comment Response