Subscribe to RSS

DOI: 10.1055/a-2741-4754
The Role of Oxidative Stress and MicroRNAs in Platelet Activation and the Efficacy of Antiplatelet Therapy in Acute Myocardial Infarction
Authors
Funding Information This work was supported by the project National Institute for Research of Metabolic and Cardiovascular Diseases (Program EXCELES, ID Project No. LX22NPO5104), Funded by the European Union, Next Generation EU, and by the Charles University Research Program COOPERATIO – Cardiovascular Science.

Abstract
Acute myocardial infarction (AMI) remains a formidable challenge in cardiovascular medicine, necessitating effective antiplatelet therapy to mitigate adverse outcomes. Recent advances have underscored the pivotal role of oxidative stress and micro ribonucleic acids (miRNAs) in regulating platelet activation and modulating the efficacy of antiplatelet agents. This review comprehensively examines the current understanding of how oxidative stress influences platelet function and the regulatory mechanisms of miRNAs in this context. It discusses the dual role of oxidative stress in promoting and impairing platelet activity and its implications for miRNAs as critical modulators of platelet activation, including their potential utility as biomarkers and therapeutic targets. Furthermore, the interaction between oxidative stress, miRNA expression, and antiplatelet drugs is analyzed to elucidate their combined impact on AMI treatment. These insights provide potential pathways to optimize therapeutic strategies, ultimately improving patient outcomes in AMI management.
Keywords
oxidative stress - miRNA - platelet activation - antiplatelet therapy - ticagrelor - prasugrelIntroduction
In acute myocardial infarction (AMI), timely reperfusion of the ischemic myocardium improves patient prognosis. However, the abrupt restoration of blood flow and the consequent increase in oxygen levels induce myocardial ischemia–reperfusion injury (IRI), which exacerbates the inflammatory response and elevates the production of reactive oxygen species (ROS). Circulating platelets and leukocytes activate early during reperfusion; their activation is influenced by the duration of the preceding coronary occlusion and correlates with the severity of myocardial injury.[1] Platelets adhere to the damaged endothelium upon reperfusion and infiltrate the ischemic/reperfused (IR) myocardium. By releasing proinflammatory cytokines, chemokines, and ROS, activated platelets further potentiate IRI by forming microthrombi, platelet–leukocyte aggregates, and releasing numerous potent proinflammatory molecules within the affected myocardium.[2] Platelet aggregation and microthrombi formation can occlude microvasculature and impair tissue perfusion despite the restoration of macroscopic blood flow. The expansion of damage in the infarct zone due to IRI can make up to 50% of the final myocardial infarct size.[3] Various imaging studies show that platelets contribute significantly to cardiac reperfusion injury.[1] [4] Recent investigations suggest that microRNAs (miRNAs), combined with oxidative stress, may be important modulators of platelet function, potentially influencing outcomes following IRI.[5] [6] Nevertheless, a comprehensive overview of the mechanisms responsible for these processes remains absent. This review aims to elaborate on the potential interactions among oxidative stress, platelets, and platelet-derived miRNAs and evaluate their utility as diagnostic tools, predictors of outcome, and treatment targets in managing AMI.
Platelets and Oxidative Stress
Platelet adhesion and aggregation are central to hemostasis; however, they also contribute to pathological processes such as thrombosis and vessel occlusion, the key events in the development of an acute coronary syndrome (ACS). The oxidative environment in ACS affects platelet function through multiple mechanisms. ROS, produced by ischemic endothelial, vascular smooth muscle cells, cardiomyocytes, and leukocytes, are also generated within activated platelets, making them both producers and targets of oxidative stress.[7] [8] ROS are important signaling molecules, promoting megakaryocyte differentiation, and platelet production and activation.[9] At low to moderate levels, ROS enhance platelet adhesion, coagulation, and thrombus formation, increasing thromboembolic risk. However, high ROS levels induce platelet apoptosis, leading to thrombocytopenia and bleeding.[10]
ROS can sensitize or directly activate key platelet surface receptors such as glycoprotein IIb/IIIa, and thromboxane receptors, facilitating fibrinogen binding, collagen-induced activation, and platelet aggregation,[9] [11] [12] and P2Y12 and thromboxane receptors.[12] ROS also enhance intracellular calcium mobilization, a critical trigger for platelet activation and degranulation.[13] Additionally, oxidative stress causes glycation, oxidation, and lipid peroxidation of platelet membranes, altering integrity, fluidity, and receptor function, which promotes platelet activation and aggregation.[14] [15] These mechanisms contribute to platelet hyperreactivity and thrombus formation in myocardial IRI, leading to vascular occlusion and myocardial injury. ROS-induced platelet activation also plays important roles in several pathological conditions beyond cardiovascular disease—such as cancer-associated thrombosis,[16] inflammation,[17] autoimmune diseases,[18] and neurodegeneration.[19]
Sources of ROS in Platelets
ROS are produced in platelets by several sources, most notably nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX), cyclooxygenase (COX), mitochondrial electron transport chain (ETC), and xanthine oxidase (XO).[20] The primary function of NOX is to catalyze superoxide production from oxygen and NADPH. In human platelets, NOX-1 and NOX-2 are major contributors to production of ROS and key regulators of platelet activation.[21] [22] NOX isoforms are inactive at rest and are triggered by agonist stimulation, with NOX-1 predominantly responding to collagen and NOX-2 to thrombin. The dismutation of NOX-2-derived superoxide into hydrogen peroxide is the primary trigger for platelet-mediated thrombus consolidation. Hereditary NOX-2 deficiency reduces platelet recruitment and thrombus growth, highlighting its critical role in platelet activation.[23] NOX-2 also contributes to adverse cardiac remodeling and dysfunction after AMI.[24]
ROS activate COX enzymes, primarily COX-1, to produce thromboxane A2 (TXA2) in platelets. COX and peroxidase catalyze the formation of prostaglandin H2, the precursor of TXA2, from arachidonic acid. ROS, including superoxide and hydrogen peroxide, are metabolic byproducts of this process, contributing to the redox environment and influencing platelet function. TXA2 is a powerful platelet activator, enhancing aggregation and vascular constriction, exacerbating thrombotic events following AMI. In patients undergoing percutaneous coronary intervention (PCI), platelets demonstrated enhanced TXA2 production via ROS-dependent NOX-2.[23]
XO catalyzes the oxidation of hypoxanthine to xanthine and subsequently to uric acid, generating hydrogen peroxide and superoxide. Although XO is mainly synthesized in the liver and endothelial cells, platelets can internalize circulating XO or acquire it from endothelial microparticles, enabling localized ROS production. XO-derived ROS enhance platelet activation by modulating redox-sensitive signaling pathways, such as phosphatidylinositol 3-kinase (PI3K)/Akt, mitogen-activated protein kinase (MAPK), and protein kinase C (PKC). Elevated XO activity is seen in AMI, and it has been implicated in myocardial stunning after ischemia.[25] [26] Platelets contain functional mitochondria essential for adenosine triphosphate (ATP) production through oxidative phosphorylation and are significant sources of ROS. During mitochondrial respiration, ROS are byproducts of electron transport chain (ETC) activity, involving complexes I to V. Electrons supplied by nicotinamide adenine dinucleotide (NADH) and flavin adenine dinucleotide (FADH2) are transferred through the ETC, reducing oxygen to water at complex IV. Throughout this process, electron leakage predominantly occurs at complex I and complex III. Leaked electrons reduce molecular oxygen to form superoxide anion.[27] Elevated ROS levels can impair mitochondrial membrane potential, promoting further ROS generation in a positive feedback loop that can lead to mitochondrial dysfunction, platelet hyperreactivity, or apoptosis.
Modulation of Platelet Activity by ROS via Apoptosis and Ferroptosis
Apoptosis
Apoptosis is a regulated process leading to cell self-destruction. While traditional apoptosis involves nuclear DNA fragmentation, platelet apoptosis shares many morphological and biochemical features, primarily driven by mitochondrial signals.[28] ROS play a central role in platelet homeostasis: at low to moderate levels, they promote activation, while at high levels, they trigger apoptosis and ferroptosis. Platelets contain both pro- and anti-apoptotic proteins. During apoptosis, survival signals are suppressed and the pro-apoptotic signals trigger mitochondrial depolarization, cytochrome C release, and caspase cascade activation. Caspase-3 then drives phosphatidylserine (PS) externalization, promoting both clearance by phagocytes and a procoagulant platelet phenotype that enhances thrombin generation and clot stability.[29] [30] Excessive platelet apoptosis may cause thrombocytopenia, and mitochondrial apoptosis further amplifies ROS production in a feedback loop.[10] [31] Platelets may also undergo apoptosis via extrinsic pathways triggered by cytokines like tumor necrosis factor-α (TNF-α), linking inflammation to thrombosis, although this pathway in platelets remains underexplored.
Ferroptosis
Ferroptosis is a form of iron-dependent cell death, driven by ROS-mediated lipid peroxidation, and mechanistically differs from apoptosis and necrosis.[32] It occurs under oxidative stress when glutathione peroxidase (GPx) 4, a key enzyme against lipid peroxidation, is depleted.[33] This disrupts iron homeostasis, leading to increased cytosolic iron from ferritin and mitochondria, and the release of hemin from hemoglobin. Hemin activates heme oxygenase-1 (HO-1), producing ferrous iron, biliverdin, and carbon monoxide, further contributing to iron overload and platelet ferroptosis.[34] [35] Both ferroptosis and apoptosis contribute to platelet dysfunction and cardiomyocyte injury during AMI. Platelets exacerbate cardiomyocyte damage by reducing protective lipid species like stearic acid–phosphatidylcholine.[36] Melatonin, an antioxidant, inhibits hemin-induced ferroptosis in platelets, suggesting its potential in treating thrombotic and thrombocytopenic conditions.[35]
Platelet Antioxidant Systems
Endogenous Antioxidant Systems
Antioxidants maintain platelet redox homeostasis by neutralizing ROS and modulating redox-sensitive signaling. Platelet defenses comprise enzymatic antioxidants such as catalase (CT), superoxide dismutase (SOD), GPx, HO-1, and glutathione-S-transferase (GST), or thioredoxin (TRX), along with non-enzymatic molecules including glutathione (GSH).[37] Since platelet activation generates ROS that amplify further activation, antioxidants help limit cytotoxic effects and promote antithrombotic actions by scavenging ROS and enhancing nitric oxide (NO) bioavailability.
Exogenous Antioxidant Systems
Vascular antioxidant systems, similar to platelet mechanisms, include enzymes such as SOD, CT, GPx, GST, and HO-1, along with extracellular antioxidants like the GSH/GSSG pair and GPx3.[38] These systems help suppress ROS and reactive nitrogen species (RNS)-driven oxidative reactions that promote platelet activation, thus contributing to redox homeostasis. Plasma antioxidants, such as SOD and CT, neutralize ROS near the platelet membrane, preventing oxidative damage to membrane lipids and proteins. Other antioxidants, such as TRX and vitamin C, are actively transported into platelets via specific membrane transporters, while NO, a key antiplatelet and vasodilatory molecule, diffuses freely into platelets, modulating ROS levels to suppress activation and aggregation. The summary of platelet antioxidant systems is in [Supplementary Table S1] (available in the online version only).
Protective Roles of Platelets in Myocardial Reperfusion Injury
Although platelets are traditionally seen as contributors to myocardial IRI through their prothrombotic and proinflammatory roles, emerging evidence suggests they also have protective effects during reperfusion. These include the release of cardioprotective molecules, mitophagy, and interactions with inflammatory cells and the endothelium.
Release of Cardioprotective Molecules
Platelets release bioactive molecules such as adenine nucleotides, TXA2, serotonin, and transforming growth factor-β1 (TGF-β1), which stimulate endothelial NO production, promoting cardioprotection.[39] [40] TGF-β1, released during platelet activation, modulates inflammation and supports cardiac repair, its reduced expression being linked to higher mortality and AMI incidence.[41] Additionally, sphingosine-1-phosphate, released in the course of platelet activation and thrombus development, provides further cardioprotection.[42] Platelet-released stromal cell-derived factor-1α (CXCL12) promotes progenitor cell migration for tissue repair and enhances cardiac function after AMI by delaying cardiomyocyte death.[43]
Platelet Mitochondrial Uncoupling
Platelets rely on both glycolysis (60%) and oxidative phosphorylation (40%) for ATP production.[44] Mitochondrial function is crucial for energy, calcium homeostasis, and ROS signaling, all of which influence platelet activation and thrombus formation.[45] Mitochondrial uncoupling, a state when protons leak back into the mitochondrial matrix without generating ATP, reduces membrane potential and ROS production.[46] Although initially thought to indicate dysfunction, endogenous uncoupling proteins suggest it regulates several biological processes.[47] [48] Mild uncoupling can decrease platelet activation by limiting ATP and ROS production,[49] while excessive uncoupling leads to metabolic stress, ATP depletion, and compensatory glycolysis, which indirectly affects ROS levels. Mitochondrial uncoupling may be a potential target for modulating platelet function and decreasing thrombotic risk.[50]
Mitophagy
Mitophagy, a selective degradation pathway, clears damaged mitochondria to maintain mitochondrial quality under hypoxic stress.[51] During early stages of cardiac IRI, it removes dysfunctional mitochondria while preserving functional ones to support platelet activation. Later, sustained hypoxia triggers more extensive mitophagy, reducing mitochondrial content and platelet activation. This mechanism limits excessive platelet activation and reduces myocardial injury via negative feedback.[52]
Interactions with Leukocytes and Endothelium, Modulation of Inflammation
Although these interactions can promote injury, platelets may facilitate reparative leukocyte responses and vascular repair under controlled activation. Some platelet-derived extracellular vesicles (EVs) and microparticles have anti-inflammatory effects, which may help limit neutrophil infiltration and reduce endothelial damage.[53] The contribution of platelets to cardioprotection is not yet fully understood. In most pathological contexts, platelet overactivation exacerbates injury. Consequently, therapeutic strategies typically aim to inhibit platelet activity.
Platelet miRNAs
miRNAs are small non-coding RNAs that modulate gene expression by interacting with target mRNAs after transcription. They influence up to two-thirds of the human genome and are involved in numerous cellular processes.[54] Although platelets contain fewer miRNAs compared with nucleated cells, they carry approximately 30% of mature human miRNAs. Up to 70% of the extracellular miRNAs, including that circulating in the blood, are platelet-derived.[55] Circulating miRNAs are stable and act as paracrine mediators between platelets, inflammatory cells, endothelium, and cardiomyocytes. Circulating miRNAs can be transported: (a) within microvesicles or extracellular vesicles (EV) shed from plasma membrane; (b) membrane-bound small vesicles of endosomal origin—exosomes; (c) in complexes with Argonaute 2 protein; (d) carried by high-density lipoprotein, or to the RNA-binding protein nucleophosmin; or (e) released from cells in apoptotic bodies. Platelet-derived exosomes can be taken up by recipient cells including leukocytes or endothelial cells, participating in inflammation, thrombosis, and atherogenesis.[55] Platelets retain megakaryocyte-derived mRNAs, as well as possess the translational machinery (e.g., rough endoplasmic reticulum, polyribosomes), enabling de novo protein synthesis.[56] miRNAs regulate key platelet functions—including activation, aggregation, secretion, and adhesion—by modulating the translation of key proteins such as integrin/cytoskeletal receptors, platelet secretion machinery (e.g., VAMP8), and signaling intermediates.[5] [6] [57] Interestingly, it has been observed that during activation, nearly half of the more than 700 quantified platelet proteins were modulated, while the platelet transcriptome remained largely unchanged owing mostly to the alterations in miRNA expression.[58]
Platelet miRNAs and Oxidative Stress Interactions
Growing evidence highlights the dual role of miRNAs as both regulators and targets of oxidative stress-related pathways.[59] miRNAs can promote or inhibit ROS production by modulating transcription factors and genes involved in ROS generation and scavenging.[60] Similarly, ROS alter miRNA expression and activity at multiple levels. The complex interactions impact key processes involved in IRI such as platelet function, inflammation, angiogenesis, fibrosis, and ventricular remodeling.[61] [62]
Impact of ROS on miRNAs
Regulation of miRNAs Production at Transcriptional Level
miRNA expression is influenced by various stressors, including oxidative stress, which acts through transcription factors like nuclear factor kappa B (NF-κB) and nuclear factor E2-related factor 2 (Nrf2). In megakaryocytes, these changes shape the miRNA content of newly formed platelets.[59] [63] The regulation of miRNA transcription occurs not only in megakaryocytes but also in platelets. Although anucleate, platelets retain a functional transcriptional and posttranscriptional regulatory system including pathways typically found in nucleated cells, such as NF-κB, sirtuin 1 (SIRT), and Nrf2.[64] [65] NF-κB generally promotes a pro-thrombotic, inflammatory phenotype in response to oxidative stress, while SIRT and Nrf2 pathways exert protective effects by attenuating ROS-mediated damage.
For the overview of selected miRNAs and their impact on signaling pathways, see [Table 1].
|
miRNA |
Signaling pathway |
Effect on tissues |
Use as biomarker/predictor |
|---|---|---|---|
|
miRNA-223 |
Blocks the NF-κB and MAPK pathways, targets the NLRP3 and ICAM-1 gene and inhibits its expression[55] [125] |
Protects cardiomyocytes from oxidative stress-induced apoptosis, promotes proliferation, migration, induces fibrosis, inhibits endothelial inflammation[55] [125] |
Negatively correlated with risk of incident AMI[86] ↑ levels predicted CVD death in CAD patients[89] Associated with higher risk of ischemic endpoint within 30 days and 1 year after AMI[120] |
|
miRNA-126 |
Activates ERK1/2 signaling pathway, SIRT1/Nrf2 signaling pathway[74] |
Inhibits oxidative stress and reperfusion injury, promotes angiogenesis in MI patients[126]; decrease in the levels of pro-inflammatory cytokines |
Positively correlated with risk of incident AMI ↑ Levels predicted ↑ risk of MACE after PCI[126] Predictor of thrombotic events after AMI[120] |
|
miRNA-21 |
Deficiency activates NF-κB pathway, regulates TGF-β/Smad7 signaling[84] [127] |
↓ Apoptosis, reduces inflammation, preserves cardiac function, and limits adverse remodeling after AMI; continuous expression may lead to cardiac hypertrophy and obesity[127] [128] |
↑ Levels in patients with ACS, earlier than troponins[129] Strong predictive ability for MACE at 3 months following AMI[130] |
|
miRNA-24 |
Inhibits NF-κB/TNF-α pathway, activates Keap1/Nrf2, activates the Nrf2/heme oxygenase-1 signaling pathway[85] [131] |
Inhibition of oxidative stress, reduction of apoptosis in MI reperfusion, attenuates fibrosis following AMI, blocking increased angiogenesis and ↓ apoptosis post-AMI[85] [132] [133] |
↓ Levels in AMI, correlated with troponin[119] |
|
miRNA-197 |
Inhibits the TLR4/NF-κB/NLRP3 pathway[134] |
Mitigates inflammation and endothelial dysfunction[134] |
↑ Levels predicted CVD death in CAD patients[89] Negatively correlated with risk of incident AMI[86] |
|
miRNA-96 |
Suppresses caspase 1 expression, negatively targets IGF1R, Bax protein[135] [136] |
Inhibits pyroptosis in ischemia-reperfusion injury, ↓ endothelial inflammation, migration and tubule formation, suppresses apoptosis, regulates DNA repair, carcinogenic effect[136] |
↑ Levels in AMI mice[137] |
|
miRNA-150 |
Activates PI3K/Akt/eNOS pathway, ↓ expression of cleaved caspase 3 and Bax, ↑ expression of Bcl-2[138] |
↓ Apoptosis, reduces inflammation; ↑ ischemia-induced neovascularization, ↓ associated with maladaptive remodeling[138] [139] [140] |
↓ Levels associated with advanced heart failure, its severity and outcome[140] |
|
miRNA-191 |
Represses OXSR1 + vs MAPK/NF-κB signaling pathway[143] |
↓ Levels in AMI[145] |
Abbreviations: AMI, acute myocardial infarction; AP1, activator protein 1; Bax, bcl-2-like protein; CAD, coronary artery disease; CVD, cardiovascular disease; EBP, emopamil binding protein; ERK1/2, extracellular signal-regulated kinase ½; ICAM-1, intercellular adhesion molecule 1; IGF1R, insulin-like growth factor 1 receptor; Keap1, Kelch-like ECH-associated protein 1; MACE, major adverse cardiovascular events; MAPK, mitogen-activated protein kinase; miRNA, micro ribonucleic acid; NF-κB, nuclear factor kappa B; NLRP3, NOD-like receptor protein 3; OXSR1, oxidative stress responsive kinase 1; PCI, percutaneous coronary intervention; PI3K/Akt/eNOS, phosphoinositide 3-kinase/protein kinase B/ endothelial nitric oxide synthase; SIRT1/Nrf2, sirtuin 1/nuclear factor E2-related factor 2; TGF-β/Smad7, transforming growth factor β.
Regulation of miRNAs at Post-transcriptional Level
At the post-transcriptional level, cellular stress affects miRNA maturation. miRNAs are generated as primary transcripts (pri-miRNAs), converted into pre-miRNAs by the nuclear RNase III enzyme Drosha and its cofactor DGCR8, and then transported to the cytoplasm via exportin-5. In the cytoplasm, Dicer1, a member of the RNase family, shortens pre-miRNAs into 22-nucleotide duplexes, and one functional strand is assembled with Argonaute proteins (Ago1–Ago4) into the miRNA-induced silencing complex (miRISC), enabling miRNA to exert its post-transcriptional regulatory function.[66]
-
(a) ROS can disrupt miRNA processing by RNases such as DROSHA, DGCR8, or Dicer1, enhancing or suppressing miRNA maturation.[67] [68]
-
(b) Stress can impair pre-miRNA export from the nucleus, such as when exportin-5 becomes saturated, reducing mature miRNA levels.[63]
-
(c) Modifications of Ago2 proteins reduces its binding to target mRNA, leading to deficiencies in miRNA function.[69] Platelet-specific Ago2 deletion in mice increases platelet volume, clot retraction, and integrin expression.[70]
Impact of miRNAs on ROS Production and Clearance
Platelet miRNAs can reciprocally regulate transcriptional and signaling pathways, forming a feedback loop that impacts oxidative stress.[71] For example, miRNA-25 and miRNA-146a regulate NOX enzymes and mitochondrial ROS production.[72] Upregulation of miRNA-24 promotes endothelial regeneration following balloon injury by reducing oxidative stress via the Nrf2/HO-1 pathway and modulating antioxidant enzymes like SOD and GPx.[73] Overexpression of miRNA-126 decreases ROS and enhances SOD and GPx activity.[74] These mechanisms balance platelet activation and inhibition under oxidative conditions.
miRNAs and Their Roles in Regulation of Platelets and Other Cells
miRNAs are transferred bidirectionally between platelets and their environment.[53] [75] Under oxidative stress, activated endothelial cells, leukocytes, and megakaryocytes release extracellular vesicles containing miRNAs, which platelets can internalize. The internalization modulates signal-dependent translation processes and platelet function. Many circulating microRNA levels change with antiplatelet therapy, reflecting their role in platelet function.
miRNA-223 is one of the most prevalent and well-characterized miRNAs in platelets. It modulates platelet activation by targeting genes associated with inflammatory responses and integrin pathways, including the P2Y12 receptor and NF-κB pathway.[76] miRNA-223 influences platelet–leukocyte interactions via EV transfer and plays a significant role in macrophage function during inflammatory stimulation and atherosclerosis.[77] Additionally, miRNA-223 regulates intercellular adhesion molecule 1 (ICAM-1) expression, decreasing inflammation following thrombosis.[55] On the other hand, platelet miRNA-223 promotes myocardial IRI and ferroptosis in cardiomyocytes following AMI by affecting lipid content in cardiomyocyte membranes.[36]
miRNA-126 is one of the most highly expressed miRNAs in platelets and is closely associated with platelet activation markers like P-selectin.[78] Platelets serve as a primary contributor to circulating miRNA-126, which promotes thrombus formation.[79] In cardiomyocytes, miRNA-126 inhibits oxidative stress, inflammation, macrophage recruitment and fibrosis.[80]
miRNA-21, produced in platelets, is involved in protecting against myocardial IRI by suppressing apoptosis and cardiac remodeling.[81] [82] Inhibition of miRNA-21 lowers circulating platelet counts and decreases platelet release of TGF-β1.[83] Overexpression of miRNA-21 reduces hydrogen peroxide-induced ROS activity and cardiomyocyte apoptosis, playing a key role in ROS-mediated injury.[84]
Platelets and the vascular endothelium exhibit substantial expression of miRNA-24, with dysregulation linked to endothelial dysfunction and cardiovascular diseases. During IRI in mice hearts, miRNA-24–3p expression decreases, but miRNA mimics reduce cardiomyocyte apoptosis caused by ROS.[85] Blocking endothelial miRNA-24 reduces infarct size by inhibiting endothelial cell death and promoting vascular growth.[85]
miRNA-197 is highly expressed in platelets.[86] Although its function in platelet activation remains unclear, it might play a part in lipid abnormalities associated with metabolic syndrome, promoting cardiovascular disease (CVD) progression.[87] Lower blood levels of miRNA-197 are observed in individuals with type 2 diabetes, suggesting it could be a prognostic biomarker for future CVDs.[88] [89]
miRNA-96 regulates vesicle-associated membrane protein 8 (VAMP8)/endobrevin, a protein involved in platelet degranulation. The expression of VAMP8 differs in hyperreactive versus hyporeactive platelets. When switching from clopidogrel to ticagrelor, miRNA-96 levels increased.[57]
An overview of selected miRNAs, their relation to oxidative stress, and their potential as biomarkers is summarized in [Table 1], and impact of antiplatelet therapy on miRNA levels in [Table 2]. Interactions of platelet miRNAs and oxidative stress are depicted in [Fig. 1].
|
miRNA |
Dynamics on antithrombotic agents |
Thrombocyte receptor miRNA |
Impact on platelet function/correlation with other platelet reactivity assessment methods |
|---|---|---|---|
|
miRNA-223 |
↓ On ticagrelor vs. clopidogrel[105] [106] ↓ On prasugrel and prasugrel + ASA[5] [104] ↓ On ASA + clopidogrel in HTPR vs. LTPR[155] ↑ On ticagrelor vs. prasugrel[105] [107] and ticagrelor vs. clopidogrel[107] |
PAR1/PAR4, P2Y1/ P2Y12, SMOC1, Factor XIII |
Positive correlation with platelet function (ADP-induced PAG)[105] ↑ Platelet inhibition on dose escalation of ASA + prasugrel, assessed by plate aggregometry and thromboxane B2 + VerifyNow[5] ↓ Levels correlated with PRI (VASP) on ASA + clopidogrel, determined low responders[156] Correlated with ADP-stimulated P-selectin expression circulating levels[104] ↑ In high responders on clopidogrel compared with low responders (PAG by LTA)[156] |
|
miRNA-126 |
↑ On ticagrelor vs. prasugrel[107] ↓ On ASA[78] ↓ On ticagrelor vs. clopidogrel[105] [106] ↓ Prasugrel and prasugrel + ASA[5] ↓ On ASA + clopidogrel in HTPR vs. LTPR[155] |
PAR1/PAR4, P2Y1/ P2Y12, PLXNB2, ADAM9 |
Correlates with P-selectin expression[78] ↓ Levels on ticagrelor increasing platelet inhibition on dose escalation of ASA + prasugrel assessed by plate aggregometry and thromboxane B2 + VerifyNow[5] Attenuated platelet activity via downregulation of disintegrin and metalloproteinase-9 (ADAM9)[157] |
|
miRNA-21 |
↓ On ticagrelor vs. clopidogrel[105] ↑ On ticagrelor compared with prasugrel[105] [107] and ticagrelor vs. clopidogrel[107] |
PAR1/PAR4, P2Y1/ P2Y12, WASp |
Positive correlation with platelet function (ADP-induced PAG)[105] ↑ In high responders on clopidogrel compared with low responders (PAG by LTA)[158] Correlated with ADP-stimulated P-selectin expression circulating levels[104] |
|
miRNA-24 |
↓ On prasugrel vs. ASA[104] ↑ ASA + clopidogrel in clopidogrel resistance[159] |
PAR1/PAR4, P2Y1/ P2Y12 |
Correlated with ADP-stimulated P-selectin expression circulating levels[104] Inversely correlated with inhibition of platelet aggregation (VerifyNow)[159] |
|
miRNA-197 |
↓ On ticagrelor vs. clopidogrel[105] ↓ On prasugrel vs. ASA[104] |
PAR1/PAR4, P2Y1/ P2Y12 |
No correlation with ADP-stimulated P-selectin expression circulating levels[104] |
|
miRNA-96 |
↑ On ASA + ticagrelor vs. ASA + clopidogrel[106] |
VAMP8/endobrevin |
↑ Levels on ticagrelor compared with clopidogrel[106] |
|
miRNA-150 |
↓ On ticagrelor compared with clopidogrel[106] ↓ Prasugrel and prasugrel + ASA[5] ↑ On ticagrelor vs. prasugrel and clopidogrel[107] ↑ Clopidogrel + ASA in HTPR vs. LTPR[155] |
NA |
Increasing platelet inhibition on dose escalation of ASA + prasugrel assessed by plate aggregometry and thromboxane B2 additionally assessed by VerifyNow[5] |
|
miRNA-191 |
NA |
Increasing platelet inhibition on dose escalation of ASA + prasugrel assessed by PAG and thromboxane B2 additionally assessed by VerifyNow[5] Correlated with ADP-stimulated P-selectin expression |
Abbreviations: ADAM9, A disintegrin and a metalloprotease 9; ADP, adenosine diphosphate; ASA, acetylsalicylic acid; HTPR, high on-treatment platelet reactivity; LTA, light transmission aggregometry; LTPR, low on-treatment platelet reactivity; miRNA, micro ribonucleic acid; PAG, platelet aggregometry; PAR, protease-activated receptor; PLXNB2, plexin B2; SMOC1, secreted modular calcium-binding protein 1; VAMP8, vesicle-associated membrane protein 8; WASp, Wiskott-Aldrich syndrome protein.


Antiplatelet Therapy, Oxidative Stress, and miRNAs
Following an ACS, a combination of oral aspirin (acetylsalicylic acid [ASA]) and P2Y12 receptor antagonists represents the mainstay of antiplatelet therapy.
General Modes of Action of Antiplatelet Agents
Aspirin irreversibly inhibits platelet COX-1, blocking TXA2 synthesis via the arachidonic acid pathway. However, since it inhibits only a single pathway of platelet activation, aspirin alone is less effective for preventing thrombotic events following AMI compared with combination therapy.[90] Platelet activation involves multiple signaling pathways, particularly in the prothrombotic environment following AMI.
The P2Y12 receptor is pivotal for platelet aggregation by regulating adenosine diphosphate (ADP)-driven granule release and inhibiting prostacyclin signaling. Clopidogrel, a widely used P2Y12 antagonist in ACS and PCI, has limitations due to delayed activation, drug interactions, and metabolic variability due to undergoing metabolization via cytochrome (CYP) enzymes.[91] Newer P2Y12 inhibitors—prasugrel, ticagrelor, and cangrelor—offer faster, more reliable inhibition. Prasugrel, like clopidogrel, is a CYP-activated prodrug and acts irreversibly. In contrast, ticagrelor and cangrelor are direct-acting, reversible inhibitors with rapid onset. Overall, prasugrel and ticagrelor show greater potency and consistency than clopidogrel.[92] [93]
Impact of Oxidative Stress on Antiplatelet Agents' Efficacy
Oxidative stress increases platelet reactivity by enhancing surface receptors activation (e.g., GPIIb/IIIa, P2Y12) and promoting signaling pathways leading to aggregation. This hyperactivity reduces antiplatelet efficacy. ROS can bypass aspirin's action through COX-independent activation via isoprostanes,[14] [94] [95] or interfere with CYP activity,[96] reducing P2Y12 inhibitor activation. Oxidative stress is linked to high on-treatment platelet reactivity (HTPR) in individuals receiving clopidogrel.[97] In populations with elevated oxidative stress, such as diabetes, chronic kidney disease, or AMI, increased aspirin resistance, clopidogrel non-responsiveness, and higher thrombotic risk were observed despite receiving DAPT.[97] [98]
Impact of Antiplatelet Therapy on Oxidative Stress
Aspirin
There are emerging data regarding the antioxidant and anti-inflammatory properties of antiplatelet therapy. Aspirin exerts anti-inflammatory effects by inhibiting prostaglandin synthesis, a process that also produces ROS as metabolic byproducts.[99] Furthermore, aspirin induces the expression of HO-1, an enzyme with cytoprotective and antioxidant properties.[100] In a model of myocardial IRI, aspirin use resulted in a marked decrease in nitro-oxidative stress and enhanced function of the endothelium.[101]
P2Y12 Inhibitors
The P2Y12 receptor is present on platelets, as well as on leukocytes, vascular smooth muscle cells, or microglia. This broad distribution suggests that P2Y12 antagonists may also affect non-platelet P2Y12 receptors, potentially modulating vascular function and inflammatory responses.[102] Since inflammatory pathways are closely linked to oxidative stress, reducing inflammation may decrease ROS production, further limiting platelet activation. In addition to its role as a P2Y12 receptor antagonist, ticagrelor prevents adenosine uptake, enhancing adenosine-mediated effects. Adenosine, which protects the heart from IRI, inhibits platelet activation and suppresses ROS generation in reperfused tissues.[103] The mechanisms of action and effects of antiplatelet therapy on oxidative stress are summarized in [Table 3].
|
Agent |
Primary mechanism of action |
Impact of oxidative stress on efficacy |
Antioxidant/Anti-inflammatory effects |
Clinical notes |
|---|---|---|---|---|
|
Aspirin |
Irreversibly inhibits COX-1 → blocks TXA2 synthesis |
Bypasses COX-1 via isoprostanes[14] [95] [146]; aspirin resistance in DM, smokers, inflammation[147] [148] |
Inhibits prostaglandin synthesis and oxidative stress[99]; induces HO-1[100]; reduces nitro-oxidative stress[101] |
Foundational antiplatelet therapy; limited efficacy when used alone post-AMI[90] |
|
Clopidogrel |
Prodrug; irreversibly inhibits P2Y12 receptor via active metabolite (CYP activation required) |
Impaired CYP activation reduces efficacy[96]; HTPR more common in high ROS states[97] [98] |
Reduces lipid peroxidation; increases CT and total antioxidant capacity[149]; mitigates IRI[150] |
Commonly used; genetic variability in response; slower onset |
|
Prasugrel |
Prodrug; irreversibly inhibits P2Y12 receptor (faster, more potent than clopidogrel) |
Affected by CYP metabolism but less variability compared with clopidogrel |
Improves endothelial NO bioavailability[151]; reduces inflammation via neutrophil inhibition[151] |
Faster and more potent than clopidogrel; prodrug (CYP dependent) |
|
Ticagrelor |
Direct, reversible P2Y12 receptor inhibitor; also inhibits adenosine reuptake |
Unaffected by CYP; more consistent inhibition under oxidative stress |
Upregulates SOD,[152] reduces ROS; inhibits apoptosis via AKT/Nrf2/HO-1 and adenosine[153] [154] |
Rapid onset; effective in oxidative stress; adenosine-mediated cardioprotection[103] |
|
Cangrelor |
Intravenous, direct reversible P2Y12 receptor inhibitor with rapid onset/offset |
Likely less affected due to direct action (limited clinical ROS-specific data) |
Unknown—limited data on ROS-specific effects |
Used in acute PCI; rapid-on/off kinetics ideal for procedural use |
Abbreviations: AMI, acute myocardial infarction; COX, cyclooxygenase; CT, catalase; CYP, cytochrome; DM, diabetes mellitus; HO-1, heme oxygenase-1; HTPR, high on-treatment platelet reactivity; IRI, ischemia-reperfusion injury; NO, nitric oxide; PCI, percutaneous coronary intervention; ROS, reactive oxygen species; SOD, superoxide dismutase; TXA2, thromboxane A2.
Impact of Antiplatelet Agents on Platelet miRNAs
Aspirin
Antiplatelet therapy modifies the expression of miRNAs associated with oxidative stress and platelet function by regulating platelet activation and related intracellular signaling pathways. Plasma levels of several platelet miRNAs, including miRNA-223, miRNA-191, miRNA-126, miRNA-197, and miRNA-150, decrease upon platelet inhibition with aspirin.[78] [104]
P2Y12 Inhibitors
In mice, miRNA-126 inhibition reduced TXA2-dependent platelet aggregation and affected P2Y12 expression.[6] In patients, miRNA levels, including miRNA-126, miRNA-223, miRNA -21 and miRNA-150, correlated with the P2Y12 inhibitor used, with prasugrel and ticagrelor generally showing stronger suppression than clopidogrel.[105] [106] In contrast, Jӓger et al reported that ticagrelor was associated with higher levels of miRNA-150, miRNA-21, and miRNA-126 compared with clopidogrel and prasugrel, while drug cessation did not alter miRNA expression despite increased ADP-mediated platelet activation.[107] Dose-escalation studies confirmed that most platelet miRNA levels decline as platelet inhibition intensifies.[5] Some, such as miRNA-96, increase with stronger platelet inhibition.[106] Prasugrel, but not clopidogrel, reduced miRNA-223, miRNA-24, miRNA-191, and miRNA-197 expression in diabetic patients compared with ASA, suggesting prasugrel may provide superior antiplatelet efficacy, particularly in patients with diabetes.[104] [Table 2] shows selected miRNAs and their changes in response to antiplatelet therapy.
Clinical Use
Monitoring of Therapy
European Society of Cardiology treatment guidelines advocate the use of P2Y12 inhibitors such as ticagrelor or prasugrel instead of clopidogrel because of their greater potency, quicker onset of action, and reduced interindividual variability, though they carry an increased bleeding risk.[93] [108] Despite this, clopidogrel is still commonly used, though 4 to 30% of patients exhibit inadequate responses, increasing cardiovascular risk.[109] Monitoring DAPT strategies can help identify such patients and guide treatment adjustments to balance ischemic and bleeding risks.
Several methods assess platelet activity, but no single test is ideal for routine use.[110] [111] The vasodilator-stimulated phosphoprotein (VASP) phosphorylation assay is highly specific for P2Y12 inhibition, with minimal interference from aspirin or GPIIb/IIIa antagonists and is more precise than traditional light transmission aggregometry (LTA).[112] Other methods, like VerifyNow and multiplate, show modest correlation, with the best comparability between LTA and VASP.[113] [114] Biomarkers like platelet–leukocyte aggregates, CD40L, and sP-selectin may also assess platelet activation.[115] [116] Flow cytometry using monoclonal antibodies for platelet activation or adhesion markers like CD41 (GP IIb/IIIa), CD42b (GP Ib), CD40L, or CD63 can assess platelet activation.[117] However, methods like LTA are time-consuming and technically challenging, while sP-selectin may have limited detectability. Immunological methods can also be costly.
Monitoring oxidative stress markers and miRNAs involved in thrombotic processes, such as platelet activation and coagulation, can provide an alternative approach to assessing thrombotic risk and evaluating antithrombotic therapy efficacy. Increased or decreased levels of miRNAs like miRNA-126 and miRNA-223 correlate with thrombotic risk, while their change after antithrombotic treatment could indicate therapeutic success ([Table 2]).
Diagnostic Markers and Outcome Predictors
Studies have shown that miRNA-223, miRNA-19b, and miRNA-483–5p provide higher diagnostic accuracy than troponin for early AMI diagnosis.[118] Additionally, miRNA-24–1 and miRNA-545 were identified as potential diagnostic biomarkers for AMI.[119] miRNA-223–3p, miRNA-197, and miRNA-126–3p show potential as independent indicators of thrombosis and ischemic complications post-AMI.[89] [120] [121] When combined with the Framingham Risk Score, these miRNAs enhance risk stratification.[86] These findings suggest that combining these markers with those currently in use may enhance diagnostic and therapeutic accuracy, especially in high-risk populations. Additionally, using miRNA primers for quantitative real-time PCR is less time- and cost-intensive than developing specific antibodies or ELISAs.[122]
There are limitations to using miRNAs as biomarkers. Most miRNAs implicated in CVDs are not platelet-specific and are found in other cells, such as endothelium, leukocytes, cardiomyocytes, and hematopoietic stem cells.[123] Conditions like hypertension, hyperlipidemia, smoking, and diabetes can affect miRNA levels, including miRNA-126, miRNA-146a, miRNA-223, and miRNA-214, as well as oxidative stress markers. In diabetes, downregulation of miRNAs critical for platelet activation can lead to upregulation of P2Y12 receptors, reducing the efficacy of P2Y12-targeting antiplatelet drugs.[124] Oxidative modifications can also make platelets more resistant to endogenous antiplatelet mechanisms, increasing thrombotic risk. The complexity and diverse sources of miRNAs can complicate their use as platelet activity markers, but this may also offer valuable insights into underlying processes.
Conclusion
Oxidative stress significantly alters platelet function after AMI by promoting platelet activation and aggregation, while impairing endothelial function and reducing NO bioavailability. These changes highlight the importance of targeting oxidative stress in managing thrombotic complications post-AMI.
Antiplatelet therapy plays a key role in treating AMI and its complications. Aspirin and P2Y12 receptor inhibitors not only exhibit antiplatelet activity but also help mitigate oxidative stress, inflammation, and myocardial injury. Integrating oxidative stress markers and miRNA-based biomarkers holds promise for monitoring antithrombotic efficacy, assessing thrombotic risk, and evaluating treatment response.
miRNAs offer potential as therapeutic targets for modulating thrombotic processes. Manipulating miRNA expression or activity could enhance existing therapies, improve outcomes, reduce adverse effects, and enable personalized treatment strategies.
Contributors' Statement
T. V. and Z.M. designed the manuscript; T.V. wrote the manuscript. Both the authors critically revised and approved the final version of the manuscript.
Conflict of Interest
The authors declare that they have no conflict of interest.
-
References
- 1 Xu Y, Huo Y, Toufektsian MC. et al. Activated platelets contribute importantly to myocardial reperfusion injury. Am J Physiol Heart Circ Physiol 2006; 290 (02) H692-H699
- 2 Ziegler M, Wang X, Peter K. Platelets in cardiac ischaemia/reperfusion injury: a promising therapeutic target. Cardiovasc Res 2019; 115 (07) 1178-1188
- 3 Fröhlich GM, Meier P, White SK, Yellon DM, Hausenloy DJ. Myocardial reperfusion injury: looking beyond primary PCI. Eur Heart J 2013; 34 (23) 1714-1722
- 4 Ziegler M, Alt K, Paterson BM. et al. Highly sensitive detection of minimal cardiac ischemia using positron emission tomography imaging of activated platelets. Sci Rep 2016; 6 (01) 38161
- 5 Willeit P, Zampetaki A, Dudek K. et al. Circulating microRNAs as novel biomarkers for platelet activation. Circ Res 2013; 112 (04) 595-600
- 6 Kaudewitz D, Skroblin P, Bender LH. et al. Association of microRNAs and YRNAs with platelet function. Circ Res 2016; 118 (03) 420-432
- 7 Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res 2017; 120 (04) 713-735
- 8 Leo R, Praticò D, Iuliano L. et al. Platelet activation by superoxide anion and hydroxyl radicals intrinsically generated by platelets that had undergone anoxia and then reoxygenated. Circulation 1997; 95 (04) 885-891
- 9 Qiao J, Arthur JF, Gardiner EE, Andrews RK, Zeng L, Xu K. Regulation of platelet activation and thrombus formation by reactive oxygen species. Redox Biol 2018; 14: 126-130
- 10 Rakesh KS, Jagadish S, Vinayaka AC. et al. A new ibuprofen derivative inhibits platelet aggregation and ROS mediated platelet apoptosis. PLoS One 2014; 9 (09) e107182
- 11 Ye Y, Yang L, Leng M. et al. Luteolin inhibits GPVI-mediated platelet activation, oxidative stress, and thrombosis. Front Pharmacol 2023; 14: 1255069
- 12 Capra V, Bäck M, Angiolillo DJ, Cattaneo M, Sakariassen KS. Impact of vascular thromboxane prostanoid receptor activation on hemostasis, thrombosis, oxidative stress, and inflammation. J Thromb Haemost 2014; 12 (02) 126-137
- 13 Redondo PC, Jardin I, Hernández-Cruz JM, Pariente JA, Salido GM, Rosado JA. Hydrogen peroxide and peroxynitrite enhance Ca2+ mobilization and aggregation in platelets from type 2 diabetic patients. Biochem Biophys Res Commun 2005; 333 (03) 794-802
- 14 Bauer J, Ripperger A, Frantz S, Ergün S, Schwedhelm E, Benndorf RA. Pathophysiology of isoprostanes in the cardiovascular system: implications of isoprostane-mediated thromboxane A2 receptor activation. Br J Pharmacol 2014; 171 (13) 3115-3131
- 15 Bikulčienė I, Garjonytė N, Žėkas V. et al. Relationship between composition of fatty acid in platelet phospholipid membrane and markers of oxidative stress in healthy men and men after a myocardial infarction. Med Sci Monit Basic Res 2021; 27: e929634
- 16 Zhang X, Yu S, Li X. et al. Research progress on the interaction between oxidative stress and platelets: another avenue for cancer?. Pharmacol Res 2023; 191: 106777
- 17 Tyml K. Critical role for oxidative stress, platelets, and coagulation in capillary blood flow impairment in sepsis. Microcirculation 2011; 18 (02) 152-162
- 18 López-Pedrera C, Barbarroja N, Jimenez-Gomez Y, Collantes-Estevez E, Aguirre MA, Cuadrado MJ. Oxidative stress in the pathogenesis of atherothrombosis associated with anti-phospholipid syndrome and systemic lupus erythematosus: new therapeutic approaches. Rheumatology (Oxford) 2016; 55 (12) 2096-2108
- 19 Beura SK, Dhapola R, Panigrahi AR, Yadav P, Reddy DH, Singh SK. Redefining oxidative stress in Alzheimer's disease: targeting platelet reactive oxygen species for novel therapeutic options. Life Sci 2022; 306: 120855
- 20 Villalpando-Rodriguez GE, Gibson SB. Reactive oxygen species (ROS) regulates different types of cell death by acting as a rheostat. Oxid Med Cell Longev 2021; 2021: 9912436
- 21 Fuentes E, Gibbins JM, Holbrook LM, Palomo I. NADPH oxidase 2 (NOX2): a key target of oxidative stress-mediated platelet activation and thrombosis. Trends Cardiovasc Med 2018; 28 (07) 429-434
- 22 Krötz F, Sohn HY, Gloe T. et al. NAD(P)H oxidase-dependent platelet superoxide anion release increases platelet recruitment. Blood 2002; 100 (03) 917-924
- 23 Basili S, Pignatelli P, Tanzilli G. et al. Anoxia-reoxygenation enhances platelet thromboxane A2 production via reactive oxygen species-generated NOX2: effect in patients undergoing elective percutaneous coronary intervention. Arterioscler Thromb Vasc Biol 2011; 31 (08) 1766-1771
- 24 Looi YH, Grieve DJ, Siva A. et al. Involvement of Nox2 NADPH oxidase in adverse cardiac remodeling after myocardial infarction. Hypertension 2008; 51 (02) 319-325
- 25 Raghuvanshi R, Kaul A, Bhakuni P, Mishra A, Misra MK. Xanthine oxidase as a marker of myocardial infarction. Indian J Clin Biochem 2007; 22 (02) 90-92
- 26 Charlat MI, O'Neill PG, Egan JM. et al. Evidence for a pathogenetic role of xanthine oxidase in the “stunned” myocardium. Am J Physiol 1987; 252 (3 Pt 2): H566-H577
- 27 Lambert AJ, Brand MD. Superoxide production by NADH:ubiquinone oxidoreductase (complex I) depends on the pH gradient across the mitochondrial inner membrane. Biochem J 2004; 382 (Pt 2): 511-517
- 28 Leytin V. Apoptosis in the anucleate platelet. Blood Rev 2012; 26 (02) 51-63
- 29 McArthur K, Chappaz S, Kile BT. Apoptosis in megakaryocytes and platelets: the life and death of a lineage. Blood 2018; 131 (06) 605-610
- 30 Choo HJ, Saafir TB, Mkumba L, Wagner MB, Jobe SM. Mitochondrial calcium and reactive oxygen species regulate agonist-initiated platelet phosphatidylserine exposure. Arterioscler Thromb Vasc Biol 2012; 32 (12) 2946-2955
- 31 Ricci JE, Gottlieb RA, Green DR. Caspase-mediated loss of mitochondrial function and generation of reactive oxygen species during apoptosis. J Cell Biol 2003; 160 (01) 65-75
- 32 Stockwell BR, Friedmann Angeli JP, Bayir H. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017; 171 (02) 273-285
- 33 Liu JP, Cen SY, Xue Z. et al. A class of disulfide compounds suppresses ferroptosis by stabilizing GPX4. ACS Chem Biol 2022; 17 (12) 3389-3406
- 34 Miyamoto HD, Ikeda M, Ide T. et al. Iron overload via heme degradation in the endoplasmic reticulum triggers ferroptosis in myocardial ischemia-reperfusion injury. JACC Basic Transl Sci 2022; 7 (08) 800-819
- 35 NaveenKumar SK, Hemshekhar M, Kemparaju K, Girish KS. Hemin-induced platelet activation and ferroptosis is mediated through ROS-driven proteasomal activity and inflammasome activation: protection by melatonin. Biochim Biophys Acta Mol Basis Dis 2019; 1865 (09) 2303-2316
- 36 Miao S, Zhang Q, Ding W. et al. Platelet internalization mediates ferroptosis in myocardial infarction. Arterioscler Thromb Vasc Biol 2023; 43 (02) 218-230
- 37 Masselli E, Pozzi G, Vaccarezza M. et al. ROS in platelet biology: functional aspects and methodological insights. Int J Mol Sci 2020; 21 (14) 4866
- 38 Scioli MG, Storti G, D'Amico F. et al. Oxidative stress and new pathogenetic mechanisms in endothelial dysfunction: potential diagnostic biomarkers and therapeutic targets. J Clin Med 2020; 9 (06) 1995
- 39 Walsh TG, Poole AW. Platelets protect cardiomyocytes from ischaemic damage. TH Open 2017; 1 (01) e24-e32
- 40 Yang BC, Mehta JL. Platelet-derived adenosine contributes to the cardioprotective effects of platelets against ischemia-reperfusion injury in isolated rat heart. J Cardiovasc Pharmacol 1994; 24 (05) 779-785
- 41 Rath D, Chatterjee M, Müller I. et al. Platelet expression of transforming growth factor beta 1 is enhanced and associated with cardiovascular prognosis in patients with acute coronary syndrome. Atherosclerosis 2014; 237 (02) 754-759
- 42 Vito CD, Hadi LA, Navone SE. et al. Platelet-derived sphingosine-1-phosphate and inflammation: from basic mechanisms to clinical implications. Platelets 2016; 27 (05) 393-401
- 43 Geisler T, Fekecs L, Wurster T. et al. Association of platelet-SDF-1 with hemodynamic function and infarct size using cardiac MR in patients with AMI. Eur J Radiol 2012; 81 (04) e486-e490
- 44 Chacko BK, Kramer PA, Ravi S. et al. Methods for defining distinct bioenergetic profiles in platelets, lymphocytes, monocytes, and neutrophils, and the oxidative burst from human blood. Lab Invest 2013; 93 (06) 690-700
- 45 Tamura N, Goto S, Yokota H, Goto S. Contributing role of mitochondrial energy metabolism on platelet adhesion, activation and thrombus formation under blood flow conditions. Platelets 2022; 33 (07) 1083-1089
- 46 Miwa S, Brand MD. Mitochondrial matrix reactive oxygen species production is very sensitive to mild uncoupling. Biochem Soc Trans 2003; 31 (Pt 6): 1300-1301
- 47 Bouillaud F, Alves-Guerra MC, Ricquier D. UCPs, at the interface between bioenergetics and metabolism. Biochim Biophys Acta 2016; 1863 (10) 2443-2456
- 48 Bround MJ, Bers DM, Molkentin JDA. A 20/20 view of ANT function in mitochondrial biology and necrotic cell death. J Mol Cell Cardiol 2020; 144: A3-A13
- 49 Shiva S, Novelli EM, Bullock GC, Kenny E, Hill G, Corey CG. Mitochondrial uncoupling protein 2 is present in human platelets and regulates platelet activation. Blood 2014; 124 (21) 4147
- 50 Demine S, Renard P, Arnould T. Mitochondrial uncoupling: a key controller of biological processes in physiology and diseases. Cells 2019; 8 (08) 795
- 51 Zhang W, Chen C, Wang J, Liu L, He Y, Chen Q. Mitophagy in cardiomyocytes and in platelets: a major mechanism of cardioprotection against ischemia/reperfusion injury. Physiology (Bethesda) 2018; 33 (02) 86-98
- 52 Zhang W, Ren H, Xu C. et al. Hypoxic mitophagy regulates mitochondrial quality and platelet activation and determines severity of I/R heart injury. eLife 2016; 5: e21407
- 53 Szilágyi B, Fejes Z, Rusznyák Á. et al. Platelet microparticles enriched in miR-223 reduce ICAM-1-dependent vascular inflammation in septic conditions. Front Physiol 2021; 12: 658524
- 54 Kloosterman WP, Plasterk RHA. The diverse functions of microRNAs in animal development and disease. Dev Cell 2006; 11 (04) 441-450
- 55 Li J, Tan M, Xiang Q, Zhou Z, Yan H. Thrombin-activated platelet-derived exosomes regulate endothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb Res 2017; 154: 96-105
- 56 Weyrich AS, Lindemann S, Tolley ND. et al. Change in protein phenotype without a nucleus: translational control in platelets. Semin Thromb Hemost 2004; 30 (04) 491-498
- 57 Kondkar AA, Bray MS, Leal SM. et al. VAMP8/endobrevin is overexpressed in hyperreactive human platelets: suggested role for platelet microRNA. J Thromb Haemost 2010; 8 (02) 369-378
- 58 Cimmino G, Tarallo R, Nassa G. et al. Activating stimuli induce platelet microRNA modulation and proteome reorganisation. Thromb Haemost 2015; 114 (01) 96-108
- 59 Engedal N, Žerovnik E, Rudov A. et al. From oxidative stress damage to pathways, networks, and autophagy via microRNAs. Oxid Med Cell Longev 2018; 2018: 4968321
- 60 Climent M, Viggiani G, Chen YW, Coulis G, Castaldi A. MicroRNA and ROS crosstalk in cardiac and pulmonary diseases. Int J Mol Sci 2020; 21 (12) 4370
- 61 Kura B, Bacova BS, Kalocayova B, Sykora M, Slezak J. Oxidative stress-responsive microRNAs in heart injury. Int J Mol Sci 2020; 21 (01) 358
- 62 Schütte JP, Manke MC, Hemmen K. et al. Platelet-derived microRNAs regulate cardiac remodeling after myocardial ischemia. Circ Res 2023; 132 (07) e96-e113
- 63 Olejniczak M, Kotowska-Zimmer A, Krzyzosiak W. Stress-induced changes in miRNA biogenesis and functioning. Cell Mol Life Sci 2018; 75 (02) 177-191
- 64 Kumari S, Chaurasia SN, Nayak MK, Mallick RL, Dash D. Sirtuin inhibition induces apoptosis-like changes in platelets and thrombocytopenia. J Biol Chem 2015; 290 (19) 12290-12299
- 65 Spinelli SL, Casey AE, Pollock SJ. et al. Platelets and megakaryocytes contain functional nuclear factor-kappaB. Arterioscler Thromb Vasc Biol 2010; 30 (03) 591-598
- 66 Pu M, Chen J, Tao Z. et al. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell Mol Life Sci 2019; 76 (03) 441-451
- 67 Wiesen JL, Tomasi TB. Dicer is regulated by cellular stresses and interferons. Mol Immunol 2009; 46 (06) 1222-1228
- 68 Kozakowska M, Ciesla M, Stefanska A. et al. Heme oxygenase-1 inhibits myoblast differentiation by targeting myomirs. Antioxid Redox Signal 2012; 16 (02) 113-127
- 69 Quévillon Huberdeau M, Zeitler DM, Hauptmann J. et al. Phosphorylation of Argonaute proteins affects mRNA binding and is essential for microRNA-guided gene silencing in vivo . EMBO J 2017; 36 (14) 2088-2106
- 70 Lazar S. The Roles of AGO2 in Megakaryopoiesis, Protein Expression, and Platelet Reactivity [Ph.D. dissertation]. Temple University, PA; 2022. . ProQuest
- 71 Cheng X, Ku CH, Siow RCM. Regulation of the Nrf2 antioxidant pathway by microRNAs: new players in micromanaging redox homeostasis. Free Radic Biol Med 2013; 64: 4-11
- 72 Fierro-Fernández M, Miguel V, Lamas S. Role of redoximiRs in fibrogenesis. Redox Biol 2016; 7: 58-67
- 73 Zhang J, Cai W, Fan Z. et al. MicroRNA-24 inhibits the oxidative stress induced by vascular injury by activating the Nrf2/Ho-1 signaling pathway. Atherosclerosis 2019; 290: 9-18
- 74 Li J, Yang C, Wang Y. miR–126 overexpression attenuates oxygen–glucose deprivation/reperfusion injury by inhibiting oxidative stress and inflammatory response via the activation of SIRT1/Nrf2 signaling pathway in human umbilical vein endothelial cells. Mol Med Rep 2021; 23 (02) 165
- 75 Laffont B, Corduan A, Plé H. et al. Activated platelets can deliver mRNA regulatory Ago2•microRNA complexes to endothelial cells via microparticles. Blood 2013; 122 (02) 253-261
- 76 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 (09) 961-966
- 77 O'Connell RM, Zhao JL, Rao DS. MicroRNA function in myeloid biology. Blood 2011; 118 (11) 2960-2969
- 78 de Boer HC, van Solingen C, Prins J. et al. Aspirin treatment hampers the use of plasma microRNA-126 as a biomarker for the progression of vascular disease. Eur Heart J 2013; 34 (44) 3451-3457
- 79 Zhang LJ, Hu YX, Huang RZ. et al. Intraplatelet miRNA-126 regulates thrombosis and its reduction contributes to platelet inhibition. Cardiovasc Res 2024; 120 (13) 1622-1635
- 80 Chen J, Cui C, Yang X. et al. MiR-126 affects brain-heart interaction after cerebral ischemic stroke. Transl Stroke Res 2017; 8 (04) 374-385
- 81 Yang Q, Yang K, Li A. microRNA-21 protects against ischemia-reperfusion and hypoxia-reperfusion-induced cardiocyte apoptosis via the phosphatase and tensin homolog/Akt-dependent mechanism. Mol Med Rep 2014; 9 (06) 2213-2220
- 82 Gu GL, Xu XL, Sun XT. et al. Cardioprotective effect of microRNA-21 in murine myocardial infarction. Cardiovasc Ther 2015; 33 (03) 109-117
- 83 Barwari T, Eminaga S, Mayr U. et al. Inhibition of profibrotic microRNA-21 affects platelets and their releasate. JCI Insight 2018; 3 (21) e123335
- 84 Wei C, Li L, Kim IK, Sun P, Gupta S. NF-κB mediated miR-21 regulation in cardiomyocytes apoptosis under oxidative stress. Free Radic Res 2014; 48 (03) 282-291
- 85 Xiao X, Lu Z, Lin V. et al. MicroRNA miR-24-3p reduces apoptosis and regulates Keap1-Nrf2 pathway in mouse cardiomyocytes responding to ischemia/reperfusion injury. Oxid Med Cell Longev 2018; 2018: 7042105
- 86 Zampetaki A, Willeit P, Tilling L. et al. Prospective study on circulating microRNAs and risk of myocardial infarction. J Am Coll Cardiol 2012; 60 (04) 290-299
- 87 Karolina DS, Tavintharan S, Armugam A. et al. Circulating miRNA profiles in patients with metabolic syndrome. J Clin Endocrinol Metab 2012; 97 (12) E2271-E2276
- 88 Zampetaki A, Kiechl S, Drozdov I. et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res 2010; 107 (06) 810-817
- 89 Schulte C, Molz S, Appelbaum S. et al. miRNA-197 and miRNA-223 predict cardiovascular death in a cohort of patients with symptomatic coronary artery disease. PLoS One 2015; 10 (12) e0145930
- 90 Chen ZM, Jiang LX, Chen YP. et al; COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction Trial) collaborative group. Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005; 366 (9497) 1607-1621
- 91 Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL, Topol EJ. Variability in platelet responsiveness to clopidogrel among 544 individuals. J Am Coll Cardiol 2005; 45 (02) 246-251
- 92 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 (11) 1045-1057
- 93 Wiviott SD, Braunwald E, McCabe CH. et al; TRITON-TIMI 38 Investigators. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007; 357 (20) 2001-2015
- 94 Krötz F, Sohn HY, Pohl U. Reactive oxygen species: players in the platelet game. Arterioscler Thromb Vasc Biol 2004; 24 (11) 1988-1996
- 95 Cipollone F, Ciabattoni G, Patrignani P. et al. Oxidant stress and aspirin-insensitive thromboxane biosynthesis in severe unstable angina. Circulation 2000; 102 (09) 1007-1013
- 96 Bhattacharyya S, Sinha K, Sil PC. Cytochrome P450s: mechanisms and biological implications in drug metabolism and its interaction with oxidative stress. Curr Drug Metab 2014; 15 (07) 719-742
- 97 Caruso R, Rocchiccioli S, Gori AM. et al. Inflammatory and antioxidant pattern unbalance in “clopidogrel-resistant” patients during acute coronary syndrome. Mediators Inflamm 2015; 2015: 710123
- 98 Geisler T, Anders N, Paterok M. et al. Platelet response to clopidogrel is attenuated in diabetic patients undergoing coronary stent implantation. Diabetes Care 2007; 30 (02) 372-374
- 99 Prasad K, Lee P. Suppression of oxidative stress as a mechanism of reduction of hypercholesterolemic atherosclerosis by aspirin. J Cardiovasc Pharmacol Ther 2003; 8 (01) 61-69
- 100 Grosser N, Abate A, Oberle S. et al. Heme oxygenase-1 induction may explain the antioxidant profile of aspirin. Biochem Biophys Res Commun 2003; 308 (04) 956-960
- 101 Frydrychowski P, Michałek M, Bil-Lula I. et al. Cardioprotective effect of acetylsalicylic acid in the myocardial ischemia-reperfusion model on oxidative stress markers levels in heart muscle and serum. Antioxidants 2022; 11 (08) 1432
- 102 Adamski P, Koziński M, Ostrowska M. et al. Overview of pleiotropic effects of platelet P2Y12 receptor inhibitors - PubMed. Thromb Haemost 2014; 112 (02) 224-242
- 103 Nanhwan MK, Ling S, Kodakandla M, Nylander S, Ye Y, Birnbaum Y. Chronic treatment with ticagrelor limits myocardial infarct size: an adenosine and cyclooxygenase-2-dependent effect. Arterioscler Thromb Vasc Biol 2014; 34 (09) 2078-2085
- 104 Parker WAE, Schulte C, Barwari T. et al. Aspirin, clopidogrel and prasugrel monotherapy in patients with type 2 diabetes mellitus: a double-blind randomised controlled trial of the effects on thrombotic markers and microRNA levels. Cardiovasc Diabetol 2020; 19 (01) 3
- 105 Gumiężna K, Bednarek A, Sygitowicz G. et al. Platelet microRNAs as potential novel biomarkers for antiplatelet therapy with P2Y12 inhibitors and their association with platelet function. J Clin Med 2023; 13 (01) 63
- 106 Carino A, De Rosa S, Sorrentino S. et al. Modulation of circulating microRNAs levels during the switch from clopidogrel to ticagrelor. BioMed Res Int 2016; 2016: 3968206
- 107 Jäger B, Stojkovic S, Haller PM. et al. Course of platelet miRNAs after cessation of P2Y12 antagonists. Eur J Clin Invest 2019; 49 (08) e13149
- 108 Byrne RA, Rossello X, Coughlan JJ. et al; ESC Scientific Document Group. 2023 ESC Guidelines for the management of acute coronary syndromes. Eur Heart J 2023; 44 (38) 3720-3826
- 109 Nguyen TA, Diodati JG, Pharand C. Resistance to clopidogrel: a review of the evidence. J Am Coll Cardiol 2005; 45 (08) 1157-1164
- 110 Paniccia R, Priora R, Liotta AA, Abbate R. Platelet function tests: a comparative review. Vasc Health Risk Manag 2015; 11: 133-148
- 111 Roule V, Ardouin P, Repessé Y. et al. Point of care tests VerifyNow P2Y12 and INNOVANCE PFA P2Y compared to light transmittance aggregometry after fibrinolysis. Clin Appl Thromb Hemost 2018; 24 (07) 1109-1116
- 112 Sun J, Yang GH, Liu JX. et al. Discordance between VASP phosphorylation and platelet aggregation in defining high on-clopidogrel platelet reactivity after ST-segment elevation myocardial infarction. Clin Appl Thromb Hemost 2018; 24 (01) 47-54
- 113 Köhler D, Straub A, Weissmüller T. et al. Phosphorylation of vasodilator-stimulated phosphoprotein prevents platelet-neutrophil complex formation and dampens myocardial ischemia-reperfusion injury. Circulation 2011; 123 (22) 2579-2590
- 114 Dias JD, Pottgiesser T, Hartmann J, Duerschmied D, Bode C, Achneck HE. Comparison of three common whole blood platelet function tests for in vitro P2Y12 induced platelet inhibition. J Thromb Thrombolysis 2020; 50 (01) 135-143
- 115 Yun SH, Sim EH, Goh RY, Park JI, Han JY. Platelet activation: the mechanisms and potential biomarkers. BioMed Res Int 2016; 2016: 9060143
- 116 Pluta K, Porębska K, Urbanowicz T. et al. Platelet-leucocyte aggregates as novel biomarkers in cardiovascular diseases. Biology (Basel) 2022; 11 (02) 224
- 117 Serebruany VL, Midei MG, Meilman H, Malinin AI, Lowry DR. Platelet inhibition with prasugrel (CS-747) compared with clopidogrel in patients undergoing coronary stenting: the subset from the JUMBO study. Postgrad Med J 2006; 82 (968) 404-410
- 118 Li L, Li S, Wu M. et al. Early diagnostic value of circulating microRNAs in patients with suspected acute myocardial infarction. J Cell Physiol 2019; 234 (08) 13649-13658
- 119 Wang Q, Liu B, Wang Y, Bai B, Yu T, Chu XM. The biomarkers of key miRNAs and target genes associated with acute myocardial infarction. PeerJ 2020; 8: e9129
- 120 Hromadka M, Motovska Z, Hlinomaz O. et al. MiR-126-3p and MiR-223-3p as biomarkers for prediction of thrombotic risk in patients with acute myocardial infarction and primary angioplasty. J Pers Med 2021; 11 (06) 508
- 121 Yu XY, Chen JY, Zheng ZW. et al. Plasma miR-126 as a potential marker predicting major adverse cardiac events in dual antiplatelet-treated patients after percutaneous coronary intervention. EuroIntervention 2013; 9 (05) 546-554
- 122 Salic K, De Windt LJ. MicroRNAs as biomarkers for myocardial infarction. Curr Atheroscler Rep 2012; 14 (03) 193-200
- 123 Krammer TL, Mayr M, Hackl M. microRNAs as promising biomarkers of platelet activity in antiplatelet therapy monitoring. Int J Mol Sci 2020; 21 (10) 3477
- 124 Fejes Z, Póliska S, Czimmerer Z. et al. Hyperglycaemia suppresses microRNA expression in platelets to increase P2RY12 and SELP levels in type 2 diabetes mellitus. Thromb Haemost 2017; 117 (03) 529-542
- 125 Meng Q, Huo X, Sun H, Wang H, Luan Z, Wang S. miR-223 regulates myocardial ischemia-reperfusion damage via targeting NLRP3 in vitro and in vivo. Naunyn Schmiedebergs Arch Pharmacol 2021; 394 (11) 2187-2195
- 126 Duan S, Wang C, Xu X. et al. Peripheral serum exosomes isolated from patients with acute myocardial infarction promote endothelial cell angiogenesis via the miR-126-3p/TSC1/mTORC1/HIF-1α pathway. Int J Nanomedicine 2022; 17: 1577-1592
- 127 Jayawardena E, Medzikovic L, Ruffenach G, Eghbali M. Role of miRNA-1 and miRNA-21 in acute myocardial ischemia-reperfusion injury and their potential as therapeutic strategy. Int J Mol Sci 2022; 23 (03) 1512
- 128 Gu H, Liu Z, Li Y. et al. Serum-derived extracellular vesicles protect against acute myocardial infarction by regulating miR-21/PDCD4 signaling pathway. Front Physiol 2018; 9: 348
- 129 Oerlemans MIFJ, Mosterd A, Dekker MS. et al. Early assessment of acute coronary syndromes in the emergency department: the potential diagnostic value of circulating microRNAs. EMBO Mol Med 2012; 4 (11) 1176-1185
- 130 Xu L, Tian L, Yan Z, Wang J, Xue T, Sun Q. Diagnostic and prognostic value of miR-486-5p, miR-451a, miR-21-5p and monocyte to high-density lipoprotein cholesterol ratio in patients with acute myocardial infarction. Heart Vessels 2023; 38 (03) 318-331
- 131 Li C, Fang M, Lin Z, Wang W, Li X. MicroRNA-24 protects against myocardial ischemia-reperfusion injury via the NF-κB/TNF-α pathway. Exp Ther Med 2021; 22 (05) 1288
- 132 Fiedler J, Jazbutyte V, Kirchmaier BC. et al. MicroRNA-24 regulates vascularity after myocardial infarction. Circulation 2011; 124 (06) 720-730
- 133 Wang J, Huang W, Xu R. et al. MicroRNA-24 regulates cardiac fibrosis after myocardial infarction. J Cell Mol Med 2012; 16 (09) 2150-2160
- 134 Cheng J, Liu Q, Hu N. et al. Downregulation of hsa_circ_0068087 ameliorates TLR4/NF-κB/NLRP3 inflammasome-mediated inflammation and endothelial cell dysfunction in high glucose conditioned by sponging miR-197. Gene 2019; 709: 1-7
- 135 Bian H, Shan H, Chen T. Resveratrol ameliorates hypoxia/ischemia-induced brain injury in the neonatal rat via the miR-96/Bax axis. Childs Nerv Syst 2017; 33 (11) 1937-1945
- 136 Jin F, Jin L, Wei B. et al. miR-96-5p alleviates cerebral ischemia-reperfusion injury in mice by inhibiting pyroptosis via downregulating caspase 1. Exp Neurol 2024; 374: 114676
- 137 Wang J, Dong G, Chi W, Nie Y. MiR-96 promotes myocardial infarction-induced apoptosis by targeting XIAP. Biomed Pharmacother 2021; 138: 111208
- 138 Desjarlais M, Dussault S, Dhahri W, Mathieu R, Rivard A. MicroRNA-150 modulates ischemia-induced neovascularization in atherosclerotic conditions. Arterioscler Thromb Vasc Biol 2017; 37 (05) 900-908
- 139 Zhu XG, Zhang TN, Wen R, Liu CF. Overexpression of miR-150-5p alleviates apoptosis in sepsis-induced myocardial depression. BioMed Res Int 2020; 2020: 3023186
- 140 Scrutinio D, Conserva F, Passantino A, Iacoviello M, Lagioia R, Gesualdo L. Circulating microRNA-150-5p as a novel biomarker for advanced heart failure: a genome-wide prospective study. J Heart Lung Transplant 2017; 36 (06) 616-624
- 141 Li H, Zhang P, Li F. et al. Plasma miR-22-5p, miR-132-5p, and miR-150-3p are associated with acute myocardial infarction. BioMed Res Int 2019; 2019: 5012648
- 142 Li S, Guo LZ, Kim MH, Han JY, Serebruany V. Platelet microRNA for predicting acute myocardial infarction. J Thromb Thrombolysis 2017; 44 (04) 556-564
- 143 Qin Y, Wang G, Peng Z. MicroRNA-191-5p diminished sepsis-induced acute kidney injury through targeting oxidative stress responsive 1 in rat models. Biosci Rep 2019; 39 (08) BSR20190548
- 144 Du K, Zhao C, Wang L. et al. MiR-191 inhibit angiogenesis after acute ischemic stroke targeting VEZF1. Aging (Albany NY) 2019; 11 (09) 2762-2786
- 145 Li C, Chen X, Huang J, Sun Q, Wang L. Clinical impact of circulating miR-26a, miR-191, and miR-208b in plasma of patients with acute myocardial infarction. Eur J Med Res 2015; 20 (01) 58
- 146 Kranzhofer R, Ruef J. Aspirin resistance in coronary artery disease is correlated to elevated markers for oxidative stress but not to the expression of cyclooxygenase (COX) 1/2, a novel COX-1 polymorphism or the PlA(1/2) polymorphism. Platelets 2006; 17 (03) 163-169
- 147 Simpson SH, Abdelmoneim AS, Omran D, Featherstone TR. Prevalence of high on-treatment platelet reactivity in diabetic patients treated with aspirin. Am J Med 2014; 127 (01) 95.e1-95.e9
- 148 Li WJ, Zhang HY, Miao CL. et al. Cigarette smoking inhibits the anti-platelet activity of aspirin in patients with coronary heart disease. Chin Med J (Engl) 2011; 124 (10) 1569-1572
- 149 Khudair LK, Al-Ani AW. Correlation between oxidative stress and clopidogrel drug in Iraqi patients with coronary artery disease. Int J Health Sci (Qassim) 2022; 6 (S9): 1164-1174
- 150 Kanko M, Maral H, Akbas MH. et al. Protective effects of clopidogrel on oxidant damage in a rat model of acute ischemia. Tohoku J Exp Med 2005; 205 (02) 133-139
- 151 Rudolph TK, Fuchs A, Klinke A. et al. Prasugrel as opposed to clopidogrel improves endothelial nitric oxide bioavailability and reduces platelet-leukocyte interaction in patients with unstable angina pectoris: a randomized controlled trial. Int J Cardiol 2017; 248: 7-13
- 152 Fakhri YA, Al-Ani AW. Association of superoxide dismutase with ticagrelor treatment in patients with peripheral arterial disease. Iraqi J Sci 2025; 66 (05) 1812-1823
- 153 El-Mokadem BM, El-Abhar HS, Abdallah DM, Awad AS, Soubh AA. Epac-1/Rap-1 signaling pathway orchestrates the reno-therapeutic effect of ticagrelor against renal ischemia/reperfusion model. Biomed Pharmacother 2021; 139: 111488
- 154 Adamski P, Koziński M, Ostrowska M. et al. Overview of pleiotropic effects of platelet P2Y12 receptor inhibitors. Thromb Haemost 2014; 112 (02) 224-242
- 155 Liu J, Qin L, Wang Z. et al. Platelet-derived miRNAs as determinants of the antiplatelet response in clopidogrel-treated patients with ACS. Thromb Res 2020; 186: 71-74
- 156 Shi R, Zhou X, Ji WJ. et al. The emerging role of miR-223 in platelet reactivity: implications in antiplatelet therapy. BioMed Res Int 2015; 2015: 981841
- 157 Garcia A, Dunoyer-Geindre S, Zapilko V, Nolli S, Reny JL, Fontana P. Functional validation of microRNA-126-3p as a platelet reactivity regulator using human haematopoietic stem cells. Thromb Haemost 2019; 119 (02) 254-263
- 158 Peng L, Liu J, Qin L. et al. Interaction between platelet-derived microRNAs and CYP2C19*2 genotype on clopidogrel antiplatelet responsiveness in patients with ACS. Thromb Res 2017; 157: 97-102
- 159 Lin S, Xu X, Hu H. et al. The expression profile of platelet-derived miRNA in coronary artery disease patients with clopidogrel resistance. Pharmacol Res Perspect 2021; 9 (02) e00751
Correspondence
Publication History
Received: 27 December 2024
Accepted after revision: 16 October 2025
Article published online:
27 November 2025
© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
References
- 1 Xu Y, Huo Y, Toufektsian MC. et al. Activated platelets contribute importantly to myocardial reperfusion injury. Am J Physiol Heart Circ Physiol 2006; 290 (02) H692-H699
- 2 Ziegler M, Wang X, Peter K. Platelets in cardiac ischaemia/reperfusion injury: a promising therapeutic target. Cardiovasc Res 2019; 115 (07) 1178-1188
- 3 Fröhlich GM, Meier P, White SK, Yellon DM, Hausenloy DJ. Myocardial reperfusion injury: looking beyond primary PCI. Eur Heart J 2013; 34 (23) 1714-1722
- 4 Ziegler M, Alt K, Paterson BM. et al. Highly sensitive detection of minimal cardiac ischemia using positron emission tomography imaging of activated platelets. Sci Rep 2016; 6 (01) 38161
- 5 Willeit P, Zampetaki A, Dudek K. et al. Circulating microRNAs as novel biomarkers for platelet activation. Circ Res 2013; 112 (04) 595-600
- 6 Kaudewitz D, Skroblin P, Bender LH. et al. Association of microRNAs and YRNAs with platelet function. Circ Res 2016; 118 (03) 420-432
- 7 Förstermann U, Xia N, Li H. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res 2017; 120 (04) 713-735
- 8 Leo R, Praticò D, Iuliano L. et al. Platelet activation by superoxide anion and hydroxyl radicals intrinsically generated by platelets that had undergone anoxia and then reoxygenated. Circulation 1997; 95 (04) 885-891
- 9 Qiao J, Arthur JF, Gardiner EE, Andrews RK, Zeng L, Xu K. Regulation of platelet activation and thrombus formation by reactive oxygen species. Redox Biol 2018; 14: 126-130
- 10 Rakesh KS, Jagadish S, Vinayaka AC. et al. A new ibuprofen derivative inhibits platelet aggregation and ROS mediated platelet apoptosis. PLoS One 2014; 9 (09) e107182
- 11 Ye Y, Yang L, Leng M. et al. Luteolin inhibits GPVI-mediated platelet activation, oxidative stress, and thrombosis. Front Pharmacol 2023; 14: 1255069
- 12 Capra V, Bäck M, Angiolillo DJ, Cattaneo M, Sakariassen KS. Impact of vascular thromboxane prostanoid receptor activation on hemostasis, thrombosis, oxidative stress, and inflammation. J Thromb Haemost 2014; 12 (02) 126-137
- 13 Redondo PC, Jardin I, Hernández-Cruz JM, Pariente JA, Salido GM, Rosado JA. Hydrogen peroxide and peroxynitrite enhance Ca2+ mobilization and aggregation in platelets from type 2 diabetic patients. Biochem Biophys Res Commun 2005; 333 (03) 794-802
- 14 Bauer J, Ripperger A, Frantz S, Ergün S, Schwedhelm E, Benndorf RA. Pathophysiology of isoprostanes in the cardiovascular system: implications of isoprostane-mediated thromboxane A2 receptor activation. Br J Pharmacol 2014; 171 (13) 3115-3131
- 15 Bikulčienė I, Garjonytė N, Žėkas V. et al. Relationship between composition of fatty acid in platelet phospholipid membrane and markers of oxidative stress in healthy men and men after a myocardial infarction. Med Sci Monit Basic Res 2021; 27: e929634
- 16 Zhang X, Yu S, Li X. et al. Research progress on the interaction between oxidative stress and platelets: another avenue for cancer?. Pharmacol Res 2023; 191: 106777
- 17 Tyml K. Critical role for oxidative stress, platelets, and coagulation in capillary blood flow impairment in sepsis. Microcirculation 2011; 18 (02) 152-162
- 18 López-Pedrera C, Barbarroja N, Jimenez-Gomez Y, Collantes-Estevez E, Aguirre MA, Cuadrado MJ. Oxidative stress in the pathogenesis of atherothrombosis associated with anti-phospholipid syndrome and systemic lupus erythematosus: new therapeutic approaches. Rheumatology (Oxford) 2016; 55 (12) 2096-2108
- 19 Beura SK, Dhapola R, Panigrahi AR, Yadav P, Reddy DH, Singh SK. Redefining oxidative stress in Alzheimer's disease: targeting platelet reactive oxygen species for novel therapeutic options. Life Sci 2022; 306: 120855
- 20 Villalpando-Rodriguez GE, Gibson SB. Reactive oxygen species (ROS) regulates different types of cell death by acting as a rheostat. Oxid Med Cell Longev 2021; 2021: 9912436
- 21 Fuentes E, Gibbins JM, Holbrook LM, Palomo I. NADPH oxidase 2 (NOX2): a key target of oxidative stress-mediated platelet activation and thrombosis. Trends Cardiovasc Med 2018; 28 (07) 429-434
- 22 Krötz F, Sohn HY, Gloe T. et al. NAD(P)H oxidase-dependent platelet superoxide anion release increases platelet recruitment. Blood 2002; 100 (03) 917-924
- 23 Basili S, Pignatelli P, Tanzilli G. et al. Anoxia-reoxygenation enhances platelet thromboxane A2 production via reactive oxygen species-generated NOX2: effect in patients undergoing elective percutaneous coronary intervention. Arterioscler Thromb Vasc Biol 2011; 31 (08) 1766-1771
- 24 Looi YH, Grieve DJ, Siva A. et al. Involvement of Nox2 NADPH oxidase in adverse cardiac remodeling after myocardial infarction. Hypertension 2008; 51 (02) 319-325
- 25 Raghuvanshi R, Kaul A, Bhakuni P, Mishra A, Misra MK. Xanthine oxidase as a marker of myocardial infarction. Indian J Clin Biochem 2007; 22 (02) 90-92
- 26 Charlat MI, O'Neill PG, Egan JM. et al. Evidence for a pathogenetic role of xanthine oxidase in the “stunned” myocardium. Am J Physiol 1987; 252 (3 Pt 2): H566-H577
- 27 Lambert AJ, Brand MD. Superoxide production by NADH:ubiquinone oxidoreductase (complex I) depends on the pH gradient across the mitochondrial inner membrane. Biochem J 2004; 382 (Pt 2): 511-517
- 28 Leytin V. Apoptosis in the anucleate platelet. Blood Rev 2012; 26 (02) 51-63
- 29 McArthur K, Chappaz S, Kile BT. Apoptosis in megakaryocytes and platelets: the life and death of a lineage. Blood 2018; 131 (06) 605-610
- 30 Choo HJ, Saafir TB, Mkumba L, Wagner MB, Jobe SM. Mitochondrial calcium and reactive oxygen species regulate agonist-initiated platelet phosphatidylserine exposure. Arterioscler Thromb Vasc Biol 2012; 32 (12) 2946-2955
- 31 Ricci JE, Gottlieb RA, Green DR. Caspase-mediated loss of mitochondrial function and generation of reactive oxygen species during apoptosis. J Cell Biol 2003; 160 (01) 65-75
- 32 Stockwell BR, Friedmann Angeli JP, Bayir H. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 2017; 171 (02) 273-285
- 33 Liu JP, Cen SY, Xue Z. et al. A class of disulfide compounds suppresses ferroptosis by stabilizing GPX4. ACS Chem Biol 2022; 17 (12) 3389-3406
- 34 Miyamoto HD, Ikeda M, Ide T. et al. Iron overload via heme degradation in the endoplasmic reticulum triggers ferroptosis in myocardial ischemia-reperfusion injury. JACC Basic Transl Sci 2022; 7 (08) 800-819
- 35 NaveenKumar SK, Hemshekhar M, Kemparaju K, Girish KS. Hemin-induced platelet activation and ferroptosis is mediated through ROS-driven proteasomal activity and inflammasome activation: protection by melatonin. Biochim Biophys Acta Mol Basis Dis 2019; 1865 (09) 2303-2316
- 36 Miao S, Zhang Q, Ding W. et al. Platelet internalization mediates ferroptosis in myocardial infarction. Arterioscler Thromb Vasc Biol 2023; 43 (02) 218-230
- 37 Masselli E, Pozzi G, Vaccarezza M. et al. ROS in platelet biology: functional aspects and methodological insights. Int J Mol Sci 2020; 21 (14) 4866
- 38 Scioli MG, Storti G, D'Amico F. et al. Oxidative stress and new pathogenetic mechanisms in endothelial dysfunction: potential diagnostic biomarkers and therapeutic targets. J Clin Med 2020; 9 (06) 1995
- 39 Walsh TG, Poole AW. Platelets protect cardiomyocytes from ischaemic damage. TH Open 2017; 1 (01) e24-e32
- 40 Yang BC, Mehta JL. Platelet-derived adenosine contributes to the cardioprotective effects of platelets against ischemia-reperfusion injury in isolated rat heart. J Cardiovasc Pharmacol 1994; 24 (05) 779-785
- 41 Rath D, Chatterjee M, Müller I. et al. Platelet expression of transforming growth factor beta 1 is enhanced and associated with cardiovascular prognosis in patients with acute coronary syndrome. Atherosclerosis 2014; 237 (02) 754-759
- 42 Vito CD, Hadi LA, Navone SE. et al. Platelet-derived sphingosine-1-phosphate and inflammation: from basic mechanisms to clinical implications. Platelets 2016; 27 (05) 393-401
- 43 Geisler T, Fekecs L, Wurster T. et al. Association of platelet-SDF-1 with hemodynamic function and infarct size using cardiac MR in patients with AMI. Eur J Radiol 2012; 81 (04) e486-e490
- 44 Chacko BK, Kramer PA, Ravi S. et al. Methods for defining distinct bioenergetic profiles in platelets, lymphocytes, monocytes, and neutrophils, and the oxidative burst from human blood. Lab Invest 2013; 93 (06) 690-700
- 45 Tamura N, Goto S, Yokota H, Goto S. Contributing role of mitochondrial energy metabolism on platelet adhesion, activation and thrombus formation under blood flow conditions. Platelets 2022; 33 (07) 1083-1089
- 46 Miwa S, Brand MD. Mitochondrial matrix reactive oxygen species production is very sensitive to mild uncoupling. Biochem Soc Trans 2003; 31 (Pt 6): 1300-1301
- 47 Bouillaud F, Alves-Guerra MC, Ricquier D. UCPs, at the interface between bioenergetics and metabolism. Biochim Biophys Acta 2016; 1863 (10) 2443-2456
- 48 Bround MJ, Bers DM, Molkentin JDA. A 20/20 view of ANT function in mitochondrial biology and necrotic cell death. J Mol Cell Cardiol 2020; 144: A3-A13
- 49 Shiva S, Novelli EM, Bullock GC, Kenny E, Hill G, Corey CG. Mitochondrial uncoupling protein 2 is present in human platelets and regulates platelet activation. Blood 2014; 124 (21) 4147
- 50 Demine S, Renard P, Arnould T. Mitochondrial uncoupling: a key controller of biological processes in physiology and diseases. Cells 2019; 8 (08) 795
- 51 Zhang W, Chen C, Wang J, Liu L, He Y, Chen Q. Mitophagy in cardiomyocytes and in platelets: a major mechanism of cardioprotection against ischemia/reperfusion injury. Physiology (Bethesda) 2018; 33 (02) 86-98
- 52 Zhang W, Ren H, Xu C. et al. Hypoxic mitophagy regulates mitochondrial quality and platelet activation and determines severity of I/R heart injury. eLife 2016; 5: e21407
- 53 Szilágyi B, Fejes Z, Rusznyák Á. et al. Platelet microparticles enriched in miR-223 reduce ICAM-1-dependent vascular inflammation in septic conditions. Front Physiol 2021; 12: 658524
- 54 Kloosterman WP, Plasterk RHA. The diverse functions of microRNAs in animal development and disease. Dev Cell 2006; 11 (04) 441-450
- 55 Li J, Tan M, Xiang Q, Zhou Z, Yan H. Thrombin-activated platelet-derived exosomes regulate endothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb Res 2017; 154: 96-105
- 56 Weyrich AS, Lindemann S, Tolley ND. et al. Change in protein phenotype without a nucleus: translational control in platelets. Semin Thromb Hemost 2004; 30 (04) 491-498
- 57 Kondkar AA, Bray MS, Leal SM. et al. VAMP8/endobrevin is overexpressed in hyperreactive human platelets: suggested role for platelet microRNA. J Thromb Haemost 2010; 8 (02) 369-378
- 58 Cimmino G, Tarallo R, Nassa G. et al. Activating stimuli induce platelet microRNA modulation and proteome reorganisation. Thromb Haemost 2015; 114 (01) 96-108
- 59 Engedal N, Žerovnik E, Rudov A. et al. From oxidative stress damage to pathways, networks, and autophagy via microRNAs. Oxid Med Cell Longev 2018; 2018: 4968321
- 60 Climent M, Viggiani G, Chen YW, Coulis G, Castaldi A. MicroRNA and ROS crosstalk in cardiac and pulmonary diseases. Int J Mol Sci 2020; 21 (12) 4370
- 61 Kura B, Bacova BS, Kalocayova B, Sykora M, Slezak J. Oxidative stress-responsive microRNAs in heart injury. Int J Mol Sci 2020; 21 (01) 358
- 62 Schütte JP, Manke MC, Hemmen K. et al. Platelet-derived microRNAs regulate cardiac remodeling after myocardial ischemia. Circ Res 2023; 132 (07) e96-e113
- 63 Olejniczak M, Kotowska-Zimmer A, Krzyzosiak W. Stress-induced changes in miRNA biogenesis and functioning. Cell Mol Life Sci 2018; 75 (02) 177-191
- 64 Kumari S, Chaurasia SN, Nayak MK, Mallick RL, Dash D. Sirtuin inhibition induces apoptosis-like changes in platelets and thrombocytopenia. J Biol Chem 2015; 290 (19) 12290-12299
- 65 Spinelli SL, Casey AE, Pollock SJ. et al. Platelets and megakaryocytes contain functional nuclear factor-kappaB. Arterioscler Thromb Vasc Biol 2010; 30 (03) 591-598
- 66 Pu M, Chen J, Tao Z. et al. Regulatory network of miRNA on its target: coordination between transcriptional and post-transcriptional regulation of gene expression. Cell Mol Life Sci 2019; 76 (03) 441-451
- 67 Wiesen JL, Tomasi TB. Dicer is regulated by cellular stresses and interferons. Mol Immunol 2009; 46 (06) 1222-1228
- 68 Kozakowska M, Ciesla M, Stefanska A. et al. Heme oxygenase-1 inhibits myoblast differentiation by targeting myomirs. Antioxid Redox Signal 2012; 16 (02) 113-127
- 69 Quévillon Huberdeau M, Zeitler DM, Hauptmann J. et al. Phosphorylation of Argonaute proteins affects mRNA binding and is essential for microRNA-guided gene silencing in vivo . EMBO J 2017; 36 (14) 2088-2106
- 70 Lazar S. The Roles of AGO2 in Megakaryopoiesis, Protein Expression, and Platelet Reactivity [Ph.D. dissertation]. Temple University, PA; 2022. . ProQuest
- 71 Cheng X, Ku CH, Siow RCM. Regulation of the Nrf2 antioxidant pathway by microRNAs: new players in micromanaging redox homeostasis. Free Radic Biol Med 2013; 64: 4-11
- 72 Fierro-Fernández M, Miguel V, Lamas S. Role of redoximiRs in fibrogenesis. Redox Biol 2016; 7: 58-67
- 73 Zhang J, Cai W, Fan Z. et al. MicroRNA-24 inhibits the oxidative stress induced by vascular injury by activating the Nrf2/Ho-1 signaling pathway. Atherosclerosis 2019; 290: 9-18
- 74 Li J, Yang C, Wang Y. miR–126 overexpression attenuates oxygen–glucose deprivation/reperfusion injury by inhibiting oxidative stress and inflammatory response via the activation of SIRT1/Nrf2 signaling pathway in human umbilical vein endothelial cells. Mol Med Rep 2021; 23 (02) 165
- 75 Laffont B, Corduan A, Plé H. et al. Activated platelets can deliver mRNA regulatory Ago2•microRNA complexes to endothelial cells via microparticles. Blood 2013; 122 (02) 253-261
- 76 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 (09) 961-966
- 77 O'Connell RM, Zhao JL, Rao DS. MicroRNA function in myeloid biology. Blood 2011; 118 (11) 2960-2969
- 78 de Boer HC, van Solingen C, Prins J. et al. Aspirin treatment hampers the use of plasma microRNA-126 as a biomarker for the progression of vascular disease. Eur Heart J 2013; 34 (44) 3451-3457
- 79 Zhang LJ, Hu YX, Huang RZ. et al. Intraplatelet miRNA-126 regulates thrombosis and its reduction contributes to platelet inhibition. Cardiovasc Res 2024; 120 (13) 1622-1635
- 80 Chen J, Cui C, Yang X. et al. MiR-126 affects brain-heart interaction after cerebral ischemic stroke. Transl Stroke Res 2017; 8 (04) 374-385
- 81 Yang Q, Yang K, Li A. microRNA-21 protects against ischemia-reperfusion and hypoxia-reperfusion-induced cardiocyte apoptosis via the phosphatase and tensin homolog/Akt-dependent mechanism. Mol Med Rep 2014; 9 (06) 2213-2220
- 82 Gu GL, Xu XL, Sun XT. et al. Cardioprotective effect of microRNA-21 in murine myocardial infarction. Cardiovasc Ther 2015; 33 (03) 109-117
- 83 Barwari T, Eminaga S, Mayr U. et al. Inhibition of profibrotic microRNA-21 affects platelets and their releasate. JCI Insight 2018; 3 (21) e123335
- 84 Wei C, Li L, Kim IK, Sun P, Gupta S. NF-κB mediated miR-21 regulation in cardiomyocytes apoptosis under oxidative stress. Free Radic Res 2014; 48 (03) 282-291
- 85 Xiao X, Lu Z, Lin V. et al. MicroRNA miR-24-3p reduces apoptosis and regulates Keap1-Nrf2 pathway in mouse cardiomyocytes responding to ischemia/reperfusion injury. Oxid Med Cell Longev 2018; 2018: 7042105
- 86 Zampetaki A, Willeit P, Tilling L. et al. Prospective study on circulating microRNAs and risk of myocardial infarction. J Am Coll Cardiol 2012; 60 (04) 290-299
- 87 Karolina DS, Tavintharan S, Armugam A. et al. Circulating miRNA profiles in patients with metabolic syndrome. J Clin Endocrinol Metab 2012; 97 (12) E2271-E2276
- 88 Zampetaki A, Kiechl S, Drozdov I. et al. Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res 2010; 107 (06) 810-817
- 89 Schulte C, Molz S, Appelbaum S. et al. miRNA-197 and miRNA-223 predict cardiovascular death in a cohort of patients with symptomatic coronary artery disease. PLoS One 2015; 10 (12) e0145930
- 90 Chen ZM, Jiang LX, Chen YP. et al; COMMIT (ClOpidogrel and Metoprolol in Myocardial Infarction Trial) collaborative group. Addition of clopidogrel to aspirin in 45,852 patients with acute myocardial infarction: randomised placebo-controlled trial. Lancet 2005; 366 (9497) 1607-1621
- 91 Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL, Topol EJ. Variability in platelet responsiveness to clopidogrel among 544 individuals. J Am Coll Cardiol 2005; 45 (02) 246-251
- 92 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 (11) 1045-1057
- 93 Wiviott SD, Braunwald E, McCabe CH. et al; TRITON-TIMI 38 Investigators. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med 2007; 357 (20) 2001-2015
- 94 Krötz F, Sohn HY, Pohl U. Reactive oxygen species: players in the platelet game. Arterioscler Thromb Vasc Biol 2004; 24 (11) 1988-1996
- 95 Cipollone F, Ciabattoni G, Patrignani P. et al. Oxidant stress and aspirin-insensitive thromboxane biosynthesis in severe unstable angina. Circulation 2000; 102 (09) 1007-1013
- 96 Bhattacharyya S, Sinha K, Sil PC. Cytochrome P450s: mechanisms and biological implications in drug metabolism and its interaction with oxidative stress. Curr Drug Metab 2014; 15 (07) 719-742
- 97 Caruso R, Rocchiccioli S, Gori AM. et al. Inflammatory and antioxidant pattern unbalance in “clopidogrel-resistant” patients during acute coronary syndrome. Mediators Inflamm 2015; 2015: 710123
- 98 Geisler T, Anders N, Paterok M. et al. Platelet response to clopidogrel is attenuated in diabetic patients undergoing coronary stent implantation. Diabetes Care 2007; 30 (02) 372-374
- 99 Prasad K, Lee P. Suppression of oxidative stress as a mechanism of reduction of hypercholesterolemic atherosclerosis by aspirin. J Cardiovasc Pharmacol Ther 2003; 8 (01) 61-69
- 100 Grosser N, Abate A, Oberle S. et al. Heme oxygenase-1 induction may explain the antioxidant profile of aspirin. Biochem Biophys Res Commun 2003; 308 (04) 956-960
- 101 Frydrychowski P, Michałek M, Bil-Lula I. et al. Cardioprotective effect of acetylsalicylic acid in the myocardial ischemia-reperfusion model on oxidative stress markers levels in heart muscle and serum. Antioxidants 2022; 11 (08) 1432
- 102 Adamski P, Koziński M, Ostrowska M. et al. Overview of pleiotropic effects of platelet P2Y12 receptor inhibitors - PubMed. Thromb Haemost 2014; 112 (02) 224-242
- 103 Nanhwan MK, Ling S, Kodakandla M, Nylander S, Ye Y, Birnbaum Y. Chronic treatment with ticagrelor limits myocardial infarct size: an adenosine and cyclooxygenase-2-dependent effect. Arterioscler Thromb Vasc Biol 2014; 34 (09) 2078-2085
- 104 Parker WAE, Schulte C, Barwari T. et al. Aspirin, clopidogrel and prasugrel monotherapy in patients with type 2 diabetes mellitus: a double-blind randomised controlled trial of the effects on thrombotic markers and microRNA levels. Cardiovasc Diabetol 2020; 19 (01) 3
- 105 Gumiężna K, Bednarek A, Sygitowicz G. et al. Platelet microRNAs as potential novel biomarkers for antiplatelet therapy with P2Y12 inhibitors and their association with platelet function. J Clin Med 2023; 13 (01) 63
- 106 Carino A, De Rosa S, Sorrentino S. et al. Modulation of circulating microRNAs levels during the switch from clopidogrel to ticagrelor. BioMed Res Int 2016; 2016: 3968206
- 107 Jäger B, Stojkovic S, Haller PM. et al. Course of platelet miRNAs after cessation of P2Y12 antagonists. Eur J Clin Invest 2019; 49 (08) e13149
- 108 Byrne RA, Rossello X, Coughlan JJ. et al; ESC Scientific Document Group. 2023 ESC Guidelines for the management of acute coronary syndromes. Eur Heart J 2023; 44 (38) 3720-3826
- 109 Nguyen TA, Diodati JG, Pharand C. Resistance to clopidogrel: a review of the evidence. J Am Coll Cardiol 2005; 45 (08) 1157-1164
- 110 Paniccia R, Priora R, Liotta AA, Abbate R. Platelet function tests: a comparative review. Vasc Health Risk Manag 2015; 11: 133-148
- 111 Roule V, Ardouin P, Repessé Y. et al. Point of care tests VerifyNow P2Y12 and INNOVANCE PFA P2Y compared to light transmittance aggregometry after fibrinolysis. Clin Appl Thromb Hemost 2018; 24 (07) 1109-1116
- 112 Sun J, Yang GH, Liu JX. et al. Discordance between VASP phosphorylation and platelet aggregation in defining high on-clopidogrel platelet reactivity after ST-segment elevation myocardial infarction. Clin Appl Thromb Hemost 2018; 24 (01) 47-54
- 113 Köhler D, Straub A, Weissmüller T. et al. Phosphorylation of vasodilator-stimulated phosphoprotein prevents platelet-neutrophil complex formation and dampens myocardial ischemia-reperfusion injury. Circulation 2011; 123 (22) 2579-2590
- 114 Dias JD, Pottgiesser T, Hartmann J, Duerschmied D, Bode C, Achneck HE. Comparison of three common whole blood platelet function tests for in vitro P2Y12 induced platelet inhibition. J Thromb Thrombolysis 2020; 50 (01) 135-143
- 115 Yun SH, Sim EH, Goh RY, Park JI, Han JY. Platelet activation: the mechanisms and potential biomarkers. BioMed Res Int 2016; 2016: 9060143
- 116 Pluta K, Porębska K, Urbanowicz T. et al. Platelet-leucocyte aggregates as novel biomarkers in cardiovascular diseases. Biology (Basel) 2022; 11 (02) 224
- 117 Serebruany VL, Midei MG, Meilman H, Malinin AI, Lowry DR. Platelet inhibition with prasugrel (CS-747) compared with clopidogrel in patients undergoing coronary stenting: the subset from the JUMBO study. Postgrad Med J 2006; 82 (968) 404-410
- 118 Li L, Li S, Wu M. et al. Early diagnostic value of circulating microRNAs in patients with suspected acute myocardial infarction. J Cell Physiol 2019; 234 (08) 13649-13658
- 119 Wang Q, Liu B, Wang Y, Bai B, Yu T, Chu XM. The biomarkers of key miRNAs and target genes associated with acute myocardial infarction. PeerJ 2020; 8: e9129
- 120 Hromadka M, Motovska Z, Hlinomaz O. et al. MiR-126-3p and MiR-223-3p as biomarkers for prediction of thrombotic risk in patients with acute myocardial infarction and primary angioplasty. J Pers Med 2021; 11 (06) 508
- 121 Yu XY, Chen JY, Zheng ZW. et al. Plasma miR-126 as a potential marker predicting major adverse cardiac events in dual antiplatelet-treated patients after percutaneous coronary intervention. EuroIntervention 2013; 9 (05) 546-554
- 122 Salic K, De Windt LJ. MicroRNAs as biomarkers for myocardial infarction. Curr Atheroscler Rep 2012; 14 (03) 193-200
- 123 Krammer TL, Mayr M, Hackl M. microRNAs as promising biomarkers of platelet activity in antiplatelet therapy monitoring. Int J Mol Sci 2020; 21 (10) 3477
- 124 Fejes Z, Póliska S, Czimmerer Z. et al. Hyperglycaemia suppresses microRNA expression in platelets to increase P2RY12 and SELP levels in type 2 diabetes mellitus. Thromb Haemost 2017; 117 (03) 529-542
- 125 Meng Q, Huo X, Sun H, Wang H, Luan Z, Wang S. miR-223 regulates myocardial ischemia-reperfusion damage via targeting NLRP3 in vitro and in vivo. Naunyn Schmiedebergs Arch Pharmacol 2021; 394 (11) 2187-2195
- 126 Duan S, Wang C, Xu X. et al. Peripheral serum exosomes isolated from patients with acute myocardial infarction promote endothelial cell angiogenesis via the miR-126-3p/TSC1/mTORC1/HIF-1α pathway. Int J Nanomedicine 2022; 17: 1577-1592
- 127 Jayawardena E, Medzikovic L, Ruffenach G, Eghbali M. Role of miRNA-1 and miRNA-21 in acute myocardial ischemia-reperfusion injury and their potential as therapeutic strategy. Int J Mol Sci 2022; 23 (03) 1512
- 128 Gu H, Liu Z, Li Y. et al. Serum-derived extracellular vesicles protect against acute myocardial infarction by regulating miR-21/PDCD4 signaling pathway. Front Physiol 2018; 9: 348
- 129 Oerlemans MIFJ, Mosterd A, Dekker MS. et al. Early assessment of acute coronary syndromes in the emergency department: the potential diagnostic value of circulating microRNAs. EMBO Mol Med 2012; 4 (11) 1176-1185
- 130 Xu L, Tian L, Yan Z, Wang J, Xue T, Sun Q. Diagnostic and prognostic value of miR-486-5p, miR-451a, miR-21-5p and monocyte to high-density lipoprotein cholesterol ratio in patients with acute myocardial infarction. Heart Vessels 2023; 38 (03) 318-331
- 131 Li C, Fang M, Lin Z, Wang W, Li X. MicroRNA-24 protects against myocardial ischemia-reperfusion injury via the NF-κB/TNF-α pathway. Exp Ther Med 2021; 22 (05) 1288
- 132 Fiedler J, Jazbutyte V, Kirchmaier BC. et al. MicroRNA-24 regulates vascularity after myocardial infarction. Circulation 2011; 124 (06) 720-730
- 133 Wang J, Huang W, Xu R. et al. MicroRNA-24 regulates cardiac fibrosis after myocardial infarction. J Cell Mol Med 2012; 16 (09) 2150-2160
- 134 Cheng J, Liu Q, Hu N. et al. Downregulation of hsa_circ_0068087 ameliorates TLR4/NF-κB/NLRP3 inflammasome-mediated inflammation and endothelial cell dysfunction in high glucose conditioned by sponging miR-197. Gene 2019; 709: 1-7
- 135 Bian H, Shan H, Chen T. Resveratrol ameliorates hypoxia/ischemia-induced brain injury in the neonatal rat via the miR-96/Bax axis. Childs Nerv Syst 2017; 33 (11) 1937-1945
- 136 Jin F, Jin L, Wei B. et al. miR-96-5p alleviates cerebral ischemia-reperfusion injury in mice by inhibiting pyroptosis via downregulating caspase 1. Exp Neurol 2024; 374: 114676
- 137 Wang J, Dong G, Chi W, Nie Y. MiR-96 promotes myocardial infarction-induced apoptosis by targeting XIAP. Biomed Pharmacother 2021; 138: 111208
- 138 Desjarlais M, Dussault S, Dhahri W, Mathieu R, Rivard A. MicroRNA-150 modulates ischemia-induced neovascularization in atherosclerotic conditions. Arterioscler Thromb Vasc Biol 2017; 37 (05) 900-908
- 139 Zhu XG, Zhang TN, Wen R, Liu CF. Overexpression of miR-150-5p alleviates apoptosis in sepsis-induced myocardial depression. BioMed Res Int 2020; 2020: 3023186
- 140 Scrutinio D, Conserva F, Passantino A, Iacoviello M, Lagioia R, Gesualdo L. Circulating microRNA-150-5p as a novel biomarker for advanced heart failure: a genome-wide prospective study. J Heart Lung Transplant 2017; 36 (06) 616-624
- 141 Li H, Zhang P, Li F. et al. Plasma miR-22-5p, miR-132-5p, and miR-150-3p are associated with acute myocardial infarction. BioMed Res Int 2019; 2019: 5012648
- 142 Li S, Guo LZ, Kim MH, Han JY, Serebruany V. Platelet microRNA for predicting acute myocardial infarction. J Thromb Thrombolysis 2017; 44 (04) 556-564
- 143 Qin Y, Wang G, Peng Z. MicroRNA-191-5p diminished sepsis-induced acute kidney injury through targeting oxidative stress responsive 1 in rat models. Biosci Rep 2019; 39 (08) BSR20190548
- 144 Du K, Zhao C, Wang L. et al. MiR-191 inhibit angiogenesis after acute ischemic stroke targeting VEZF1. Aging (Albany NY) 2019; 11 (09) 2762-2786
- 145 Li C, Chen X, Huang J, Sun Q, Wang L. Clinical impact of circulating miR-26a, miR-191, and miR-208b in plasma of patients with acute myocardial infarction. Eur J Med Res 2015; 20 (01) 58
- 146 Kranzhofer R, Ruef J. Aspirin resistance in coronary artery disease is correlated to elevated markers for oxidative stress but not to the expression of cyclooxygenase (COX) 1/2, a novel COX-1 polymorphism or the PlA(1/2) polymorphism. Platelets 2006; 17 (03) 163-169
- 147 Simpson SH, Abdelmoneim AS, Omran D, Featherstone TR. Prevalence of high on-treatment platelet reactivity in diabetic patients treated with aspirin. Am J Med 2014; 127 (01) 95.e1-95.e9
- 148 Li WJ, Zhang HY, Miao CL. et al. Cigarette smoking inhibits the anti-platelet activity of aspirin in patients with coronary heart disease. Chin Med J (Engl) 2011; 124 (10) 1569-1572
- 149 Khudair LK, Al-Ani AW. Correlation between oxidative stress and clopidogrel drug in Iraqi patients with coronary artery disease. Int J Health Sci (Qassim) 2022; 6 (S9): 1164-1174
- 150 Kanko M, Maral H, Akbas MH. et al. Protective effects of clopidogrel on oxidant damage in a rat model of acute ischemia. Tohoku J Exp Med 2005; 205 (02) 133-139
- 151 Rudolph TK, Fuchs A, Klinke A. et al. Prasugrel as opposed to clopidogrel improves endothelial nitric oxide bioavailability and reduces platelet-leukocyte interaction in patients with unstable angina pectoris: a randomized controlled trial. Int J Cardiol 2017; 248: 7-13
- 152 Fakhri YA, Al-Ani AW. Association of superoxide dismutase with ticagrelor treatment in patients with peripheral arterial disease. Iraqi J Sci 2025; 66 (05) 1812-1823
- 153 El-Mokadem BM, El-Abhar HS, Abdallah DM, Awad AS, Soubh AA. Epac-1/Rap-1 signaling pathway orchestrates the reno-therapeutic effect of ticagrelor against renal ischemia/reperfusion model. Biomed Pharmacother 2021; 139: 111488
- 154 Adamski P, Koziński M, Ostrowska M. et al. Overview of pleiotropic effects of platelet P2Y12 receptor inhibitors. Thromb Haemost 2014; 112 (02) 224-242
- 155 Liu J, Qin L, Wang Z. et al. Platelet-derived miRNAs as determinants of the antiplatelet response in clopidogrel-treated patients with ACS. Thromb Res 2020; 186: 71-74
- 156 Shi R, Zhou X, Ji WJ. et al. The emerging role of miR-223 in platelet reactivity: implications in antiplatelet therapy. BioMed Res Int 2015; 2015: 981841
- 157 Garcia A, Dunoyer-Geindre S, Zapilko V, Nolli S, Reny JL, Fontana P. Functional validation of microRNA-126-3p as a platelet reactivity regulator using human haematopoietic stem cells. Thromb Haemost 2019; 119 (02) 254-263
- 158 Peng L, Liu J, Qin L. et al. Interaction between platelet-derived microRNAs and CYP2C19*2 genotype on clopidogrel antiplatelet responsiveness in patients with ACS. Thromb Res 2017; 157: 97-102
- 159 Lin S, Xu X, Hu H. et al. The expression profile of platelet-derived miRNA in coronary artery disease patients with clopidogrel resistance. Pharmacol Res Perspect 2021; 9 (02) e00751

