Next Article in Journal
Bladder Management Strategies for Urological Complications in Patients with Chronic Spinal Cord Injury
Previous Article in Journal
Reoperation for Recurrent and Persistent Cushing’s Disease without Visible MRI Findings
Previous Article in Special Issue
Short- and Mid-Term Outcomes of Stenting in Patients with Isolated Distal Internal Carotid Artery Stenosis or Post-Surgical Restenosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Clinical Application of Serum microRNAs in Atherosclerotic Coronary Artery Disease

by
Anna Kabłak-Ziembicka
1,2,*,
Rafał Badacz
1,3 and
Tadeusz Przewłocki
3,4
1
Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
2
Noninvasive Cardiovascular Laboratory, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
3
Department of Interventional Cardiology, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
4
Department of Cardiac and Vascular Diseases, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(22), 6849; https://doi.org/10.3390/jcm11226849
Submission received: 24 October 2022 / Revised: 15 November 2022 / Accepted: 18 November 2022 / Published: 20 November 2022
(This article belongs to the Special Issue The Prevention and Treatment of Atherosclerosis—Part II)

Abstract

:
MicroRNAs (miRs) are promising diagnostic, prognostic and therapeutic biomolecules for atherosclerotic cardiovascular disease. Atherosclerotic occlusive disease concerns a large population of patients, carrying the highest incidence of fatal and non-fatal adverse events, such as myocardial infarction, ischemic stroke, and limb ischemia, worldwide. Consistently, miRs are involved in regulation and pathogenesis of atherosclerotic coronary artery disease (CAD), acute coronary syndromes (ACS), both with ST-segment (STEMI) and non-ST segment elevation myocardial infarctions (NSTEMI), as well as cardiac remodeling and fibrosis following ACS. However, the genetic and molecular mechanisms underlying adverse outcomes in CAD are multifactorial, and sometimes difficult to interpret for clinicians. Therefore, in the present review paper we have focused on the clinical meaning and the interpretation of various miRs findings, and their potential application in routine clinical practice.

1. Introduction

microRNAs (miRs) are endogenous non-coding single-stranded RNAs of approximately 20 nucleotides in length that negatively regulate post-transcriptional gene functions [1]. Since their discovery in the later years of the 20th century, miRs have become potential genetic biomarkers, among many other markers, for atherosclerotic cardiovascular disease [2,3,4]. Atherosclerotic occlusive disease concerns a large population, carrying the highest incidence of fatal and non-fatal adverse events worldwide, including myocardial infarction (MI), ischemic stroke, renal or limb ischemia [5,6,7]. Consistently, miRs has been shown to be involved in the regulation and pathogenesis of atherosclerotic stable coronary artery disease (CAD), acute coronary syndromes (ACS), both with ST-segment (STEMI) and non-ST segment elevation myocardial infarction (NSTEMI), as well as ischemia/reperfusion (I/R) injury, left ventricular remodeling (LVR) and fibrosis following ACS.
The genetic and molecular mechanisms underlying adverse outcomes in CAD are multifactorial, and sometimes difficult to interpret for clinicians. miRs have features that make them a potential diagnostic, prognostic and therapeutic target. As they regulate gene expression at the post-transcriptional level, usually by binding to the 3′-untranslated regions of target mRNAs, leading to the inhibited translation, and/or inducing degradation of the target mRNA (Figure 1) [8]. In this mechanism, a single miR can act on several or even hundreds of mRNAs. Furthermore, circulating miRs in serum are resistant to lysis. They show huge stability properties against RNase, e.g., by hiding themselves in various microvesicles (microparticles, apoptotic bodies, etc.). Thus, miRs keep the reins on all the major physiologic and pathophysiologic processes.
In a cardiovascular setting, the deprivation of vessel physiological processes, as a consequence of the degradation of responsible mRNAs, leads to the initiation of atherosclerosis [1,2,9,10,11,12]. The loss of vital mRNAs protecting against endothelial dysfunction, oxidative stress, low-grade inflammation, and many others, results in the promotion of atherosclerosis. The latter eventually leads to adverse atherosclerosis-related cardiovascular events [13,14]. Additionally, miRs play a fundamental role in either plaque destabilization or rupture, eventually triggering acute atherosclerotic ischemic events [1,2,10,11]. Eventually, they are important regulators of LVR, fibrosis, and I/R injury [10,11].
However, as the total number of all human miRs identified so far is over 2000 (www.mirbase.org, accessed on 22 August 2022), there is huge pressure to identify miRs that are crucial for adverse events incidence, which may result in production of antidotes against ‘bad’ miRs, or agonists to increase the expression of ‘good’ miRs [12].
Therefore, in the present review paper we have focused on the clinical meaning and the interpretation of various miRs findings, and their potential application in a routine clinical practice. We addressed the expression levels of circulating miRs in patients with stable CAD, ACS (STEMI/NSTEMI), I/R injury, and LVR following cardiac ischemia.

2. Atherosclerotic Stable CAD

Atherosclerotic CAD is estimated to cause death in 7–7.5 million people annually [6,15]. However, ACS and sudden death are responsible for 1.8 million deaths annually [16].
A variety of miRs regulate multiple metabolic pathways responsible for atherosclerosis initiation, plaque formation, and finally plaque rupture [1,8,9,17]. As circulating miRs are origin-specific, they may be released from arterial endothelial cells (ECs: miR-17, 92a; -103; -126; -142; -652; etc.), cardiomyocytes (miR-1; -16; -23a; -133a/b; -208b; -423; -499, etc.), vascular smooth muscle cells (VSMCs: miR-22; -34a; -141; -143; -145; -132; -138; -150; -494; etc.), platelets (miR-34b; -34b; -126; -150; -191; -223, etc.), or macrophages (miR-33a/b; -146a/b; -155; -103; etc.) into a blood stream [1,17]. Some miRs are abundant in many cells, induced by shear stress miR-21 that is highly expressed in VSMCs, ECs, cardiac fibroblasts, cardiomyocytes, and platelets, and responsible for apoptosis and eNOS activity [18]. Among many candidate miRs, some are of special interest as potential diagnostic markers for significant CAD, ACS, and LVR, prognostic markers for cardiovascular events or even a promising therapeutic target in atherosclerosis [1,17,19]. Specific miRs addressed in this review, with the inclusion of their anti-, and/or pro-atherothrombotic regulatory functions, are presented in Figure 2.

2.1. miRs Diagnostic for CAD

In patients suspected of chronic coronary insufficiency, some of miRs may help to discriminate patients with or without significant CAD (Table 1). For example, the discriminating power of circulating miR-145, miR-182, miR-205, and miR-133a was found to be highly predictive of stable CAD with receiver operating characteristic curves (ROC) and area under the curve (AUC) values above 0.80 [20]. Cardiac muscle–enriched miRs, such as miR-133a, miR-134, miR-145, miR-122, and miR-370 are associated with the presence of CAD [19,21]. EC-enriched circulating levels of miR-126, miR-17, miR-92a and macrophage inflammation-associated miR-155 were significantly reduced in patients with CAD compared with healthy controls [21]. Similarly, the smooth muscle–enriched miR-145 was significantly reduced in CAD [22]. However, when patients presented with unstable angina and chest pain corresponding to vulnerable coronary plaques on a coronary angiography, several vascular and inflammation-associated miRs, previously reported as down-regulated in stable CAD, such as miR-21, miR-17, miR-20a, and miR-92a, were found to be up-regulated in unstable angina [21]. Thus, the role and clinical significance of individual miRs can be subject to variability depending on the stage of atherosclerotic disease, and its stable or unstable presentation.

2.1.1. Platelets Activity

Although platelets do not contain a nucleus and genomic DNA, they have been shown to contain subcellular organelles, as well as a small amount of poly(A) + RNA from their megakaryocyte progenitor cells [23,24]. It is estimated that between 15% and 32% of protein-coding genes are represented in the form of mRNAs in platelets [23].
There are some platelets-derived miRs that play a key role in thrombosis and hemostasis [25,26,27]. Some of them are responsible for platelet activation and are influenced by antiplatelet therapy [25,26,27,28,29,30,31]. Platelet activity, responsible for blood clotting, might be modified by miR-34b, miR-126-5p, miR-150, miR-223-3p, and miR-191 (Table 1) [25,26,27,28,29,30,31].
miR-34b-3p has be associated with thromboxane-mediated platelet aggregation through the cyclooxygenase 1 (COX-1) receptor. COX-1 is the therapeutic target of aspirin, which irreversibly binds and inactivates the enzyme [28]. Inhibition of miR-34b-3p in megakaryocytes increased their viability and decreased the expression of thromboxane synthase and thromboxane B2, a stable metabolite of TxA2 [25]. Thus, miR-34b-3p may facilitate the antiplatelet efficiency of aspirin through inhibiting thromboxane synthase [28].
miR-223, one of the most abundant miRNAs in megakaryocytes and platelets, targets the P2Y12 receptor [26,29]. miR-223-deficient mice form larger thrombi and have a delayed clot retraction compared to wild type mice [27]. Platelets of miR-223-deficient mice display increased aggregation in response to thrombin and collagen [27]. Some miRs (miR-126-3p and miR-223-3p) play a role in platelets activation in ACS, and their expression levels are modified by antiplatelets therapy with aspirin and P2Y12 receptor inhibitors [24,30]. Antiplatelet therapy significantly reduces levels of miR-223, miR-191, miR-126 and miR-150, that decreases platelet inhibition [28]. In line with this, an up-regulation of miR-126 is associated with a higher platelet susceptibility to aggregate, while administration of the antagomir against miR-126-3p reduces platelets aggregation (Table 1) [29].
Due to numerous limitations of the currently used platelet function test, studies position miRs as promising candidates to assess the platelet response to COX-1 and P2Y12 inhibitors, as they are stable in biological samples. However, their low levels may impose difficulties in their quantification [30,31].

2.1.2. Vascular Smooth Muscle Cells

VSMC proliferation plays a critical role in atherosclerosis [32]. At the beginning of atherothrombotic process, irregular VSMC proliferation promotes plaque formation, but in advanced plaques VSMCs are beneficial, promoting stability and preventing rupture of the fibrous cap [32,33].
VSMCs have two states: contractile or synthetic/proliferative [34,35,36]. The first is associated with maintaining vessels tone and elasticity, while the second characterizes arterial stiffening and promotes atherosclerotic growth [34,35]. Increasing arterial stiffness is a well-known risk factor for adverse cardiovascular events, including cardiovascular death (CVD) [37,38]. High expression of miR-22, miR-124, miR-132-3p, miR-138-5p, miR-141-3p, miR-145, and miR-150-5p is needed for the contractile phenotype of VSMCs [39]. Of those, an increased expression of miR-22, miR-124 and miR-145 can reverse VSMC phenotypic switching, favoring the contractile phenotype (Table 1) [40,41]. In models adding miR-145, or miR-22, the contractile function of VSMCs can be restored [40,41]. Importantly, miR-145 is responsible for the increase in plaque collagen content and enlarging the fibrous cap area, in line with reducing the necrotic core area [42,43]. In the study by Gao et al., reduced plasma miRNA-145 levels correlated with an increase in CAD severity (a SYNTAX score) [44].
Wang et al. demonstrated that the sustained release of miR-22 enhanced the contractile phenotype of VSMCs without interfering with the proliferation of ECs [45]. Moreover, miR-22 showed therapeutic potential against restenosis, as the miR-22-coated stents showed a significant capability to inhibit in-stent restenosis in minipigs (an animal study) (Table 1) [45]. Although more controversial, a similar role was proposed for anti-miR-21-eluting stents in rats, as miR-21 is abnormally expressed in patients with coronary restenosis [46,47]. These findings are very relevant in everyday clinical practice, as restenosis following endovascular interventions with stent implantation is a serious drawback [48]. It seems that miRs could help to overcome this clinical problem.
On the contrary, an increased expression of miR-34a, while a decreased expression of miR-143 promotes vascular senescence of VSMCs [34,49]. In a study enrolling 203 patients with stable CAD and 100 controls, miR-34a showed good diagnostic value for CAD presence with an AUC of 0.899 (p < 0.001), and was associated with a Gensini score in CAD patients (p < 0.001) [50].

2.1.3. Arterial Endothelial Cells

Vascular endothelium injury and dysfunction are pivotal in the atherosclerotic process and comprise components such as: inflammation, cells infiltration, EC apoptosis and neointimal formation. EC miRs are regulated upon shear stress and disturb laminar flow in the arteries [51]. ECs harbor large amounts of regulatory miRs, with the highest expression for miR-21-5p, miR-126-3p, and the family and clusters of let-7 miRNA, miR-17-92, and miR-221/222 [51,52,53].
Among them, miR-126 is a crucial regulator of atherosclerosis [53,54,55,56,57,58]. miR-126-5p promotes regenerative proliferation of ECs, and limits atherosclerosis [54,55,56]. Up-regulation of miR-126 is also required for angiogenesis during organismal development or the repair of injured arterial vasculature [53]. In patients with a down-regulated expression of miR-126, more severe and complex CAD has been observed [57,58]. In line with this, Wang et al. observed that the miR-126 expression was significantly down-regulated in CAD patients compared to control subjects, but up-regulated in patients presenting with ACS compared to the controls [58].
In contrast, increased miR-652-3p expression levels promoted atherosclerosis through the inhibition of the EC regeneration and repair following mechanical injury [59]. The same was observed for miR-142-3p in which high expression levels increased EC apoptosis and atherosclerotic development by up-regulating the expression of Rictor and activating the Akt/eNOS signaling pathway [60]. Treatment with the miR-142-3p antagomir attenuated endothelial apoptosis and retarded the progression of atherosclerosis in the aorta of ApoE-/- mice [60].
In addition, miR-103-3p stimulated inflammatory activation, and uptake of oxidized LDL-cholesterol promoting atherosclerotic growth [61,62]. Thus, a reduction in miR-103 levels, also results in the reduction in atherosclerosis and endothelial inflammation [61]. Similarly, miR-92a-3p is up-regulated in ECs in response to shear and oxidative stress, and oxidized LDL [63]. Overexpression of miR-92a promotes ox-LDL (including malondialdehyde-LDL)-induced apoptosis [64]. The latter was associated with high-risk plaques in the coronary arteries despite statin treatment [65]. Up-regulation of miR-92a and miR-486 could discriminate between patients with stable and vulnerable CAD [66]. Another, miR involved in lipid metabolism is miR-122 that is increased in the plasma of patients with significant CAD [67]. In 255 hyperlipidemia patients with or without CAD and 100 control patients with normal blood lipid levels, miR-122 and miR-370 levels were positively correlated with the severity of CAD quantified by the Gensini score [67]. Finally, miR-17-5p was highly expressed in ECs, but low in VSMCs. There was a negative correlation between miR-17 expression levels and CAD severity [68]. However, high miR-17 expression led to the inhibition of angiogenesis, which is desirable after ACS [69].

2.1.4. Macrophages

Macrophages contain several miRs, including, most prevalently, miR-146a-5p, miR-10a-5p, let-7 family members, miR-21a-5p, and miR-155-5p [52,70,71]. They are responsible for atherosclerotic progression, the uptake of lipoproteins, self-transformation to foam cells and their subsequent apoptosis, cholesterol crystal deposition and necrotic core formation, and mature lesions with a thrombogenic core [52]. Macrophages are involved in cholesterol homeostasis and are regulated by several miRs involved in lipid metabolism. In cholesterol homeostasis, inhibition by miR-33a lowers plasma cholesterol and reduces plaque burden, while miR-33b the increases level of plasma HDL-cholesterol [72].
Some miRs that take part in atherosclerotic lesion progression are activated by lipopolysaccharides [52]. One of them, miR-155-5p has an ambiguous role in atherosclerosis [71,73,74,75,76]. At the early stages of atherosclerosis, miR-155 reduce macrophage proliferation [73], while in advanced atherosclerotic lesions it promotes further formation of atherosclerotic lesions through mediation of pro-inflammatory processes [74]. In addition, miR-155 increases glucagon-like peptide 1 (GLP-1), which inhibits glucagon production, reduces adipose tissue, and improves glucose metabolism, but at the same time miR-155 is associated with increased lipid levels [75]. Dysregulated miR-155 levels play a crucial role in the pathogenesis of diabetes mellitus [76]. This ambiguous action of miR-155 limits its clinical application. Research studies using intravascular ultrasonography (IVUS) and optimal coherence therapy (OCT) have demonstrated that coronary lesions can be found at various growing stages at the same time [77]. In an individual patient, some atherosclerotic lesions can be limited to fatty streaks, while others can have features of a necrotic core, or even rupture, and they all are regulated by miRs [78].
Lipopolysaccharide-induced macrophage activation through the expression of miR-146b is another mechanism of atherosclerotic evolution [79,80,81,82,83]. miR-146a antagonizes the pro-inflammatory effects of miR-155 [79,80]. miR-146a has a critical role as an anti-inflammatory and athero-protective agent, constituting a ‘brake’ for inflammation through the inhibition of IL-6, IL-1β, IL-8 and TNF-α [81]. It inhibits oxidized LDL-induced lipid accumulation and inflammatory response via targeting of Toll-like receptor 4 [80]. miR-146a plays a major role in thrombo-inflammation, i.e., thrombosis associated with an inflammatory process [81]. Its rise after an MI is cardioprotective as it suppresses apoptosis, the inflammatory response, and fibrosis [82]. In contrast, low expression levels of miR-146a facilitate an increase in infarct size, apoptosis and I/R injury following ACS [83].

2.1.5. Cardiomyocytes

miRs contained within cardiomyocytes are released into blood stream as a result of cardiac cell ischemia, that leads to cell damage [84,85]. Among cardiomyocyte-enriched miRs, some deserve special attention, such as miR-1, miR-133a, miR-133b, miR-145, miR-208b, miR-223, and miR-499, and they all (if detected in serum) are considered markers of myocardial damage [1,12,19,84,85,86,87,88,89,90,91,92,93,94].
Their clinical significance results from the diagnostic properties to distinguish healthy individuals from those with significant CAD, unstable angina, or ACS (STEMI and NSTEMI) (Table 1) [1,12,19,19,84,85]. In a study by Guo et al., researchers analyzed a group of 300 patients with stable CAD compared to 100 healthy control individuals, the level of circulating miR-223 was highly predictive of the severity of CAD with an AUC of 0.933 [86]. In a study by Zhu et al., among the five examined miRs (miR-92a, miR-133a, miR-133b, miR-125b, and miR-21), only miR-133a was significantly increased in CAD patients, compared to the healthy control patients; however, at a low AUC value (0.597) [87].
In a study by Abdallah et al., researchers enrolled 73 patients with stable CAD and compared them to 73 control patients. miR-133a, miR-155 and miR-208a were down-regulated, while miR-182, miR-145, miR-21, miR-126, miR-200b, miR-146a, miR-205, miR-135b, miR-196b, and miR-223 were significantly up-regulated [88]. In this study, miR-133a, miR-182, miR-145, and miR-205 showed satisfactory diagnostic levels with AUC values above 0.8 [88].
Navickas et al. reviewed 19 studies to identify miRs that could be used as biomarkers in plasma/serum to diagnose patients with atherosclerosis, significant CAD and/or ACS [19]. Out of analyzed miRs, miR-1, miR-208, and miR-133a were observed as significant serum biomarkers of acute chest pain associated with cardiac ischemia [19]. While de Rosa et al. observed that the muscle-enriched miR-499 (20-fold), miR-133a (11-fold), and miR-208a (5-fold) were significantly elevated in the aorta of troponin-positive ACS patients, compared to patients with stable CAD [89].
Table 1. Summary of the most clinically utile miRs in atherosclerotic coronary artery disease, with the inclusion of the miRs role, place of origin, expression levels, and targets.
Table 1. Summary of the most clinically utile miRs in atherosclerotic coronary artery disease, with the inclusion of the miRs role, place of origin, expression levels, and targets.
Postulated RolemicroRNADown- vs. Up-
Regulated
Diagnostic/TherapeuticReference
 Expressed in many cells   
Highly expressed in VSMCs, ECs, cardiac fibroblasts, cardiomyocytes, and platelet apoptosis and eNOS activitymiR-21-5pUp D, up-regulated in CAD patients compared to controls (AUC: 0.767, p < 0.001)Abdallah H.Y., 2022 [88]
 Platelets   
Humans: collagen-induced platelet aggregation
Mice: expression of the P2Y12 receptor
miR-126-3pUpD, monitors P2Y12 inhibitionKaudewitz D., 2016 [26]
Responsive to antiplatelet therapymiR-126-3pUpT, an antagomir against miR-126-3p reduces platelets aggregationKaudewitz D., 2016 [26]
Marker of platelet activation, that targets the COX-1 receptor through the regulation of TXSmiR-34b-3pUpD, miR-34b-3p may facilitate the antiplatelet efficiency of aspirin through inhibiting TXSLiu W.W., 2009 [28]
Marker of response to clopidogrel, that targets the P2Y12 receptormiR-223-3pDownD, high on-clopidogrel platelet reactivityShi R., 2016 [29]
miRs released by platelets, that are responsive
to antiplatelet therapy
miR-126
miR-150
miR-191
miR-223
Up
Up
Up
Up
D, antiplatelet therapy significantly reduces their levelsCzajka P., 2021 [31]
 VSMCs   
High expression is needed to maintain a contractile phenotype of VSMCsmiR-22DownT, a stent with an miR-22 coating showed significant capability to inhibit in-stent restenosis (an animal study)Yang F., 2018 [40]
Mitigates atherosclerosis, VSMCs contractility, increases fibrous cap area, and reduces the necrotic core areamiR-145Down
Down
T, delivery of miR-145 may limit atherosclerotic plaque growth, and restore contractile levels in VSMCsPatel N., 2022 [42]
Down-regulation of miR-145 plays a critical role in the pathogenesis of atherosclerotic plaques, and neointimal hyperplasiamiR-145DownD, reduced plasma miR-145 levels correlate with an increase in CAD severity (SYNTAX score)Gao H., 2015 [44]
Induces VSMC senescence, promotes the expression of age-associated pro-inflammatory secretory factors, and increases the binding capacity of ox-LDL to macrophagesmiR-34aUpD, increased expression in CAD, compared to healthy controls (AUC: 0.899, p < 0.001), associated with Gensini score (p < 0.001)Li H., 2022 [50]
 Arterial endothelial cells   
Plays a crucial anti-atherogenic role by regulating the function of ECs and enhancing endothelial repairmiR-126-3pDownD, reduced expression is associated with more severe and complex CADLi H., 2016 [57]
Decreases size of atherosclerotic lesions, alleviates ox-LDL-induced EC injurymiR-126-3pDown
Up
D, decreased expression in CAD patients, compared to healthy controls, but up-regulated in ACSWang X., 2017, [58]
Induces EC apoptosis, development of atherosclerosismiR-142-3pUpT, down-regulation of miR-142-3p suppresses ECs apoptosisQin, B., 2018 [60]
Induces apoptosis and oxidative stress, and is pro-atheroscleroticmiR-92a-3p
miR-486
Up
Up
D, discriminate between stable and vulnerable CADNiculescu L.S., 2015 [66]
Lipid metabolismmiR-122Up D, increased in CAD patients, and with CAD severity (Gensini score)Gao W., 2012 [67]
Recovery of ischemic tissuemiR-17Down D, reduced expression is associated with more severe and complex CADChen J., 2015 [68]
Rate of apoptosis in ECs miR-17-5pUpT, inhibition of miR-17 suppresses apoptosis, hence, decreases infarct size area, and improves microcirculation of heart tissue, decreasing heart failure symptomsYang S., 2018 [69]
 Macrophages   
Regulator of cholesterol and fatty acid homeostasis, reverse cholesterol transport, increases HDL-cholesterol levelmiR-33a
miR-33b
Up
Up
T, inhibition of miR-33a facilitates atherosclerotic regressionPrice N.L., 2017 [72]
Inhibits oxidized LDL-induced lipid accumulation and inflammatory responsemiR-146aUpD, patients with stable CAD had 3.62-fold higher expression level, compared to controls (AUC:0.767)Abdallah H.Y., 2022 [88]
Reduced in diabetics, has a role in lipid metabolismmiR-155DownD, patients with stable CAD had 1.89-fold lower expression level, compared to controls (AUC:0.767)Abdallah H.Y., 2022 [88]
 Cardiomyocytes   
Indicates myocardial damagemiR-223-5pUpD, increased expression, compared to healthy control group, with a AUC of 0.933 for predicting CAD severityGuo J.F., 2018 [86]
Suppresses EC proliferation rate, viability, and migration activity involved in heart development, and indicates myocardial damagemiR-133aUpD, increased expression in CAD, compared to healthy controls, but with a low AUC of 0.597 correlates with Gensini score of CAD severity (r = 0.303, p = 0.007)Zhu L., 2017 [87]

Indicates myocardial damage
Indicates myocardial damage
Fibrous cap increase
Indicates myocardial damage
Fibrous cap thinning
Plaque neovascularization
Indicates myocardial damage

miR-133a
miR-182
miR-145
miR-205
miR-208a
miR-21
miR-126
miR-223

Down
Up
Up
Up
Down
Up
Up
Up
CAD vs. controls:
AUC: 0.863, p < 0.001
AUC: 0.959, p < 0.001
AUC: 0.836, p < 0.001
AUC: 0.959, p < 0.001
AUC: 0.616, p = 0.015
AUC: 0.767, p < 0.001
AUC: 0.767, p < 0.001
AUC: 0.616, p = 0.015
Abdallah H.Y., 2022 [88]
Induces angiogenesis and myocardial damage
Indicate myocardial damage
miR-1
miR-133a
Up
Up
D, increased expression, compared to a healthy control groupKuwabara Y., 2011 [90]
Expressed in myocardial cellsmiR-23aUpD, up-regulated, positive correlation with CAD severityLu H.Q., 2013 [91]
Cardiac myofibroblast differentiation, smooth muscle cell modulator, increases fibrous cap area, reduces necrotic coremiR-145DownD, reduced in patients with cardiac ischemiaZhang M., 2017 [92]
Indicates myocardial damage, cardiac hypertrophy
Protects against H2O2-induced apoptosis
miR-208b
miR-499
Up
Up
D, independent predictors of a high SYNTAX score
miR-208b: AUC: 0.775, p < 0.001
miR-499: AUC: 0.713, p < 0.001
Wang W., 2019 [93]
Increases foam cell formationmiR-23aUpD, correlates with CAD severity (Gensini score)Wang S., 2016 [94]
AUC: area under the curve; D: diagnostic; miR: microRNA, CAD: coronary artery disease; ECs: endothelial cells; T: therapeutic approach; TXS: thromboxane synthase; VSMC: vascular smooth muscle cells.
Of note, decreased miR-27b and miR-146 expression levels are postulated to be associated with a higher severity of coronary, lower extremity, and carotid atherosclerosis [95]. These miRs presented an AUC of ≥0.75 for predicting polyvascular atherosclerosis involving the three territories [95]. Polyvascular atherosclerosis is a major clinical issue, both due to its relatively high prevalence as well as increased risk of major adverse coronary and carotid events [96,97,98]. As miRs are abundantly present in a remarkably stable form and can be detected in peripheral circulation, they are natural biomarkers of atherosclerosis at any stage of its evolution, as well as the indicators of plaque vulnerability and acute ischemia [99,100].

3. Acute Coronary Syndrome

3.1. miRs Diagnostic in ACS

ACS is a result of the interplay between coronary artery in situ thrombus formation, vulnerable plaque features such as a lipid or a necrotic core, myocardial necrosis followed by fibrosis. As previously evidenced, several miRs follow the same kinetics as highly sensitive cardiac troponins (cTn), since they derive from myocardial necrosis [1,19].
For miRs to be considered as diagnostic markers for ACS, they must be quickly released, optimally preceding typical cardiac markers of myocardial necrosis, such as cTn, creatine kinase MB. Then, the potential miR must be characterized by high sensitivity and specificity for ACS, preferably with a power assessed with AUCs above 0.9. They should well-differentiate patients with ACS from those with stable CAD and healthy individuals (Table 2).
There is growing evidence that these criteria are fulfilled for miR-1, miR-133a and miR-133b that may have an advantage over other miRs, as their peak concentration has been documented to anticipate the peak cTn concentration, even at 2.5 h after the onset of chest pain [101]. Plasma miR-1 levels were shown to be significantly up-regulated in 93 ACS patients on admission compared to 66 healthy controls, and this decreased to similar level observed in healthy volunteers on discharge [102]. In a study by Long et al., circulating miR-1 and miR-126 in ACS patients significantly differed compared to healthy adults, with a peak change at 4 and 8 h since symptom onset, then the fold change gradually decreased over time [103]. Both miR-1 and miR-126 showed high sensitivity and specificity for ACS (Table 2). In line with this, Kazimierczyk et al. demonstrated that the concentration levels of serum miR-1 and miR-126 were higher in ACS patients on admission, compared to the controls [104]. Moreover, miR-1 correlated positively with the maximal cTn concentration (r = 0.59, p = 0.02), and negatively with the left ventricular ejection fraction (LVEF) (r = −0.76, p = 0.0004) [104]. Of note, in the work by Wang et al., and Widera et al., higher expression levels of miR-1, miR-133a, and miR-208a were found in patients with cardiac ischemia compared to healthy subjects [105,106]. In addition, Wang et al. and Zhang et al. found good accuracy for miR-499 for the early diagnosis of ACS [105,107]. Conversely, in a study by He et al., the AUC value of miR-126-3p performed better (AUC: 0.992, p < 0.001), compared to cTn (AUC 0.787, p < 0.001), and creatine kinase MB (AUC 0.863, p < 0.001) [108]. Additionally, Gidlöf et al. observed in 25 patients with STEMI an abrupt increase in miR-1, miR-133a, miR-208b and miR-499-5p with a peak within 12 h from the onset of chest pain. Moreover, expression levels of miR-208b correlated with peak cTn and the left ventricular ejection fraction [109]. In line with this, Su et al. identified miR-1 as an early marker of ACS with a similar diagnostic accuracy to cTn [110].
Xue et al. proposed a different set of miRs in the diagnosis of ACS [111]. In this study, the expression levels of plasma miR-17-5p, miR-126-5p, and miR-145-3p showed considerable diagnostic efficiency for ACS, as individual measurement, and in the combination [111]. Horvath et al. observed up-regulated levels of miR-24, miR-146a, miR-145, miR-151-3p, miR-323p, and miR-331 in STEMI compared to patients with stable CAD and healthy individuals [112]. Similarly, miR-223 and miR-191, markers of platelet activation, showed higher expression levels in patients with STEMI compared to healthy controls and stable CAD patients, indicating the presence of intracoronary thrombus as the trigger for ACS [112]. The ROC analysis confirmed the suitability of miR-331 and miR-151-3p as early biomarkers of STEMI (in a median of 2.25 hours since the onset of chest pain), while the markers of myocardial necrosis were still negative at the time of sampling [112].
Conflicting data presented by Meng et al. found decreased plasma levels of miR-143 and miR-145 in patients presenting with ACS compared to controls [113]. Both miRs were negatively correlated with Gensini score, and they showed good predictive value for the onset of ACS (miR-143: OR 0.087, 95% CI 0.026–0.384, p = 0.019, and miR-145: OR 0.179, 95% CI 0.08–0.399, p < 0.001) [113]. In line with this, data from the REGICOR registry comparing 500 samples from ACS patients matched with 500 samples from healthy controls showed that miR-143 was significantly associated with time-to-ACS (HR 0.56, 95% CI 0.38–0.82), p = 0.003) [114].
Less evidence exists for miR-23a-3p, although in a study by Bukauskas et al. miR-23a-3p showed relatively high predictive value for STEMI (AUC 0.806, 95% CI 0.694–0.917), compared to healthy individuals, and provided information on the 1-year mortality according to the GRACE and APACHE scales (p = 0.045, log-rank tests) [115]. Zhang et al. found that plasma levels of miR-21 were significantly higher in patients with AMI or angina compared to the controls. They also found a significant correlation between miR-21 and clinically established markers, including cTn and creatine kinase MB (p < 0.001) [116].
An interesting approach was presented by Kayvanpour et al. [117]. In their study, the authors developed a neural network model which incorporated 34 validated ACS miRs, showing excellent classification results with an accuracy of 0.96 (95% CI 0.96–0.97), sensitivity of 0.95, specificity of 0.96 and AUC of 0.99, compared to the one-point cTn value (accuracy of 0.89, sensitivity of 0.82, specificity of 0.96, and AUC of 0.96) [117].

Differences between STEMI and NSTEMI

Although, many studies have enrolled patient with ACS, including both STEMI and NSTEMI patients [1,19,102,103,105,106,107,108], some studies have addressed miR expression levels with respect to the type of ACS, Table 2 [100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,118,119,120,121,122,123]. Most evidence has been reported on STEMI and miRs, but much less for NSTEMI.
In a study by Bukauskas et al., higher expression levels were found for miR-23a, miR-30d, miR-146 in STEMI patients compared to healthy participants [115]. In a study by Biener et al. enrolling 137 NSTEMI patients and 905 patients admitted with chest pain (after exclusion of STEMI), higher expression levels were found for five miRs (miR-29a, miR-92a, miR-126, miR-132, and miR-133) [119]. However, the AUCs were disappointingly low, ranging between 0.577 and 0.656 for individual miRs, and 0.662 for the panel of the most predictive miRs [110]. Furthermore, in this study cTn changes had a higher predictive value for NSTEMI than miRs [110].
In a study by Liu et al. including 145 NSTEMI patients and 30 control subjects, the expression levels of miR-1, miR-133, miR-208, and miR-499 were analyzed [120]. The authors found that three out of the four analyzed miRs (miR-133, miR-208 and miR-499) demonstrated superior diagnostic accuracy than cTn (AUC: 0.778), Table 2 [120]. In contrast, Zhelankin et al. found increased plasma levels of miR-146a-5p and miR-21-5p was a general ACS circulating biomarkers and lower levels of miR-17-5p was a general biomarker of CAD [121]. In a study by Gacoń et al., the increased expression level of miR-134 in STEMI compared to NSTEMI patients was observed [122]. Interestingly in that study, patients with occluded compared with patient infarct-related coronary artery thrombosis had higher levels of miR-133a (fold change: 7.00), miR-133b (4.57), miR-34a (5.50), and miR-124 (2.55), providing a significant signature for acute plaque rupture with subsequent coronary artery thrombosis, irrespective of ACS type [122]. Thus, miR expression might indicate subjects in which a coronary angiography should be performed without further delay due to artery occlusion, and the increased risk of myocardial injury.
In 62 patients with unstable angina, Zhang et al. found that a decrease in miR-223 levels was the only independent predictor for platelet reactivity index-determined lower responders [123].
Table 2. microRNAs that are potentially diagnostic in acute coronary syndromes.
Table 2. microRNAs that are potentially diagnostic in acute coronary syndromes.
Study Groups, N of ParticipantsmicroRNADown vs. Up-RegulatedRationale for Use of Individual microRNAAUC, or OR (95% CI), p-ValueReference
STEMI/NSTEMI, 93
Healthy Controls, 66
miR-1UpD, early marker, up-regulated expression, compared to healthy control groupAUC: 0.774, p < 0.001Ai J., 2010 [102]
STEMI/NSTEMI, 17
Healthy Controls, 25
miR-1
miR-126-3p
Up
Down
D, early markers, changed expressions, compared to healthy control groupAUC: 0.92, p = 0.001
AUC: 0.860, p = 0.01
Long G., 2012 [103]
STEMI/NSTEMI, 33
Healthy Controls, 33
miR-1
miR-133a
miR-208a
miR-499
Up
Up
Up
Up
D, early markers, increased expressions, compared to healthy control groupAUC: 0.850, p = 0.001
AUC: 0.870, p = 0.01
AUC: 0.970, p = 0.001
AUC: 0.820, p = 0.01
Wang G.K., 2010 [105]
STEMI/NSTEMI, 142
Non-ACS chest pain, 100
Healthy Controls, 85
miR-499UpD, early marker of ACS, 1 h after onset of chest pain, correlated with CK-MB level and cTn, but not superior to cTn (AUC: 0.90) AUC: 0.860, p < 0.001Zhang L., 2015 [107]
STEMI/NSTEMI, 27
Healthy Controls, 30
miR-126-3pDownD, early marker, diagnostic effect superior to cTn (AUC 0.787) and CK-MB (AUC 0.863)AUC: 0.992, p < 0.001He Y., 2017 [108]
STEMI, 25
Healthy Controls, 11
miR-1
miR-133a
miR-208b
miR-499-5p
Up
Up
Up
Up
D, with a peak within 12 h from onset of chest pain, expression levels of miR-208b correlated with peak cTn and the LV ejection fractionAUC: 0.980, p < 0.001
AUC: 0.859, p = 0.007
AUC: 1.000, p < 0.001
AUC: 0.989, p < 0.001
Gidlöf O.; 2011 [109]
STEMI, 106
NSTEMI, 68
Non-ACS chest pain, 163
miR-1UpD, early marker of ACS within 3 h since onset of chest pain, similar AUC to cTn (AUC: 0.862, p < 0.001)AUC: 0.863, p < 0.001Su T., 2020 [110]
STEMI, 15
NSTEMI, 14
Healthy Controls, 21
miR-17-5p miR-126-5p miR-145-3pUp
Up
Up
D, within 4 h after the onset of chest pain AUC: 0.857, p < 0.001
AUC: 0.802, p < 0.001
AUC: 0.720, p = 0.01
Xue S., 2019 [111]
STEMI, 20
CAD, 20
Healthy Controls, 20
miR-151-3pUpD, proceeded release of necrotic markers, increased expression, compared to healthy controls and stable CADSTEMI vs. controls:
AUC: 0.758, p = 0.005
STEMI vs. CAD
AUC: 0.754, p = 0.006
Horvath M., 2020 [112]
STEMI, 20
CAD, 20
Healthy Controls, 20
miR-331UpD, proceeded release of necrotic markers, increased expression, compared to healthy and stable CADSTEMI vs. controls:
AUC: 0.790, p = 0.002
STEMI vs. CAD
AUC: 0.773, p = 0.003
Horvath M., 2020 [112]
STEMI/NSTEMI, 78
Unstable angina, 201
Healthy Controls, 65
miR-143
miR-145
Down
Down
D, down-regulated compared to controls, good predictive value for the onset of ACS0.087 (0.026–0.384), p = 0.019
0.179 (0.08–0.399), p < 0.001
Meng L., 2022 [113]
ACS, 500
Healthy Controls, 500
miR-143DownD, down-regulated compared to controls, good predictive value for the onset of ACS0.56 (0.38–0.82), p = 0.003Dégano I.R., 2020 [114]
STEMI, 62
Healthy Controls, 26
miR-23a-3p
miR-30d-5p
miR-146a-5p
Down
Down
Down
D, for STEMI vs. healthy controls;
p, correlated with GRACE and APACHE scores of in-hospital mortality, and 1-month survival
D, for STEMI vs. healthy controls
D, for STEMI vs. healthy controls
AUC: 0.806, p < 0.05
p = 0.045 (log-rank tests)
AUC 0.745, p <0.05
AUC 0.800, p < 0.05
Bukauskas T., 2019 [115]
NSTEMI, 137
Chest pain *, 905
miR-126 miR-133 miR-134Up
Up
Up
D, diagnostic for NSTEMI, but not superior to cTn (AUC: 0.937)AUC: 0.578, p = 0.003
AUC: 0.656, p < 0.001
AUC: 0.506, p = 0.032
Biener M., 2021 [119]
NSTEMI, 145
Healthy Controls, 30
miR-1
miR-133
miR-208
miR-499
Up
Up
Up
Up
D, for NSTEMI vs. healthy controls, miR-133, miR-208 and miR-499 superior to cTn (AUC: 0.778)AUC: 0.772, p < 0.05
AUC: 0.928, p < 0.05
AUC: 0.994, p < 0.05
AUC: 0.994, p < 0.05
Liu G., 2018 [120]
STEMI, 16
NSTEMI, 27
miR-134
miR-134
miR-124
miR-133b
Up
Up
Up
Up
D, for STEMI, but not superior to cTn
D, for occluded IRA
D, for occluded IRA
D, for occluded IRA
AUC: 0.725, p = 0.002
AUC: 0.686, p = 0.016
AUC: 0.787, p < 0.001
AUC: 0.704, p = 0.006
Gacoń J., 2016 [122]
NSTEMImiR-223-3pDownMarker of response to clopidogrel, targets P2Y12 receptor D, lower response to clopidogrel in NSTEMI0.111, (0.018–0.692), p = 0.019Zhang Y.Y., 2014 [123]
* excluded patients with STEMI; ACS: acute coronary syndrome; AUC: area under the curve; CAD: coronary artery disease; CI: confidence interval; CK-MB: creatine kinase MB; cTn: cardiac troponin, D: diagnostic; IRA: infarct related artery; LV: left ventricle; miR: microRNA, NSTEMI: non-ST elevation myocardial infarction, OR: odds ratio; STEMI: ST-elevation myocardial infarction.
In summary, miR-1, miR-133, and miR-499 have the greatest potential for diagnostic biomarkers of ACS, as they are detected in blood samples before cTns. A meta-analysis of twenty-six studies enrolling in total 1973 ACS patients and 1236 healthy controls, indicated miR-1, miR-133 and miR-499 to have the highest value as diagnostic biomarkers of ACS [115]. The pooled sensitivity for miR-1 in the diagnosis of ACS was 70% (95% CI: 0.66–0.74), specificity: 81% (95% CI: 0.78–0.85), and AUC of 84%. The values for miR-133 were 82% (95%CI: 0.77–0.86), 87% (95%CI: 0.82–0.90) and 92.9% respectively, while for miR-499 were 80% (0.77–0.83), 89% (0.86–0.92) and 89.8%, respectively [124].
However, as time since ACS diagnosis to coronary artery revascularization is critical in post-myocardial injury salvage, the potential utility of these miRs may still be questionable, as quick test results are required to proceed with the treatment.

3.2. MiRs Worth of Examination as They Might Be Prognostic for Outcomes

3.2.1. miRs Predictive of Plaque Instability and Myocardial Infarction in Patients with Stable CAD or after Index ACS

While some miRs are released as a result of ischemic injury, local and systemic inflammation, others provide information on adverse cardiovascular events, risk of ACS, myocardial I/R injury, or LVR (Table 3). Data regarding the clinical meaning of miRs as prognostic biomarkers in patients with stable CAD, or following ACS are sparse, often inconsistent, and/or not systematized. They address the need for further research. Therefore, we present some findings that may stimulate future research studies with systematized methodology using optimal multicenter study designs.
Wang et al., in a cohort study of 2812 subjects of the general population, found that out of five promising serum miRs (miR-10a-5p, miR-126-3p, miR-210-3p, miR-423-3p, and miR-92a-3p), miR-423-3p was able to precisely predict cardiac events such as ACS and subsequent ACS during a median follow-up of 6 years [125]. Importantly, adding miR-423-3p to the model of traditional risk factors improved the predictive power for ACS (AUCs: 0.806 vs. 0.782) [125]. Overexpression of miR-423 has been shown to have a role in hypoxia/reoxygenation following ACS, accompanying I/R injury, and promote cardiomyocyte apoptosis [126].
Amongst platelet-derived miRs, that are responsible for platelet activation and are influenced by antiplatelet therapy, miR-223-3p is one the most investigated in the context of cardiovascular outcomes. Adding miR-223-3p level into the model for calculating ischemic risk following ACS significantly increased the predictive accuracy for CVD, as well as the combined ischemic endpoint (CVD/re-MI/IS) within 30 days and one year (Table 3) [127]. Similarly, the miR-126-3p:miR-223-3p ratio resulted in the increased predictive power for CVD and combined end-point (Table 3) [127]. In a recently published research paper, Scărlătescu et al. found that in a group of 50 young patients with recent STEMI, that echocardiographic myocardial work indices along with miR-223-3p, miR-146a-5p, and miR-142-3p were clinically utile predictors of adverse cardiovascular outcomes [128]. In this study, 18% of the STEMI patients experienced adverse events including CVD, re-admission for heart failure, or required another cardiovascular intervention at 1-year follow-up, [128]. In a study by Schulte et al. enrolling 340 ACS patients and 533 patients with stable CAD, miR-223 was observed to predict CVD in the ACS group and, after age-adjustment, also in patients with stable CAD at 4-years follow-up [129]. In ACS patients, the prognostic power of miR-223 and miR-197 was even higher [129].
In another study including 1112 secondary care patients, six miRs were able to predict the risk of CVD during a median follow-up of 4 years [130]. Ziaee et al. suggested that inflammation-related miRs, such as: miR-146a, miR-342, and miR-145 may be useful biomarkers in predictive and preventive cardiology [131]. In line with this, STEMI patients with high levels of miR-146a had a higher risk of major adverse cardiovascular events (MACE) compared to those with low miR-146a levels (log-rank p = 0.034). The authors suggested that miR-146a could serve as a biomarker for the adverse prognosis of STEMI and MACE at 3 years following primary ACS [132].
Whereas Bukauskas et al. found that the expression levels of miR-23a-3p were correlated with in-hospital mortality risk scores, suggesting that a down-regulation of circulating miR-23a-3p levels may be associated with an increased severity of STEMI and a higher risk of CVD [115]. It is probable that the expression levels of miR-208 can also be taken into consideration as predictors of reduced 6-month survival following ACS [133]. Finally, as one ischemic event may increase the risk for secondary ischemic events, in a study by Badacz et al., expression levels of miR-1-3p, miR-16-5p and miR-122-5p during incident ischemia were identified as risk factors of secondary cardiovascular events [134]. In addition, miR-134 was observed to be an important prognostic factor of secondary adverse cardiovascular events in patients presenting with vulnerable plaques and diabetes [135].
Table 3. microRNAs that may be prognostic biomarkers in stable coronary artery disease and acute coronary syndrome for cardiovascular outcomes.
Table 3. microRNAs that may be prognostic biomarkers in stable coronary artery disease and acute coronary syndrome for cardiovascular outcomes.
Study GroupsmicroRNADown vs. Up-RegulatedPrognostic/TherapeuticStatistical Analysis
AUC, or HR/OR (95% CI), p-Value
Reference
Cardiovascular events     
2812 general population subjectsmiR-423-3p Upp, for ACS during a median follow-up of 6 y.Better model including miR-423 (AUC: 0.806) vs. traditional risk factors (AUC: 0.782)Wang X., 2020 [125]
62 STEMI patients
26 healthy controls
miR-23a-3pDownp, correlates with GRACE and APACHE scores of in-hospital mortality, and 1-month survivalAUC 0.806, p < 0.05
p = 0.045 (log-rank tests)
Bukauskas T., 2019 [115]
598 ACS patients randomized to ticagrelor vs. prasugrel treatmentmiR-223-3pUpp, CVD/re-MI/IS at 30d.
CVD/re-MI/IS at 1 y.
15.74 (2.07–119.9), p = 0.008
3.18 (1.40–7.19), p = 0.006
Hromadka M., 2021 [127]
598 ACS patients randomized to ticagrelor vs. prasugrel treatmentmiR-126 to miR-223 ratioLowp, CVD/re-MI/IS at 30d.
CVD/re-MI/IS at 1 y.
0.14 (0.03–0.61), p = 0.009
0.37 (0.17–0.82), p = 0.014
Hromadka M., 2021 [127]
50 STEMI patients
10 healthy controls
miR-223-3p miR-142-3p
miR-146a-5p
Up
Up
Up
p, for CVD/readmission for HF/new cardiovascular interventionAUC 0.832, p =0.002
AUC 0.732, p = 0.031
AUC 0.848, p = 0.001
Scărlătescu A.I., 2022 [128]
340 ACS patients
533 patients with stable CAD
miR-197
miR-223
miR-126
miR-197
miR-223
Up
Up
Up
Up
Up
p, for CVD after MI at 4 y.
p, for CVD after adjustment to age at 4 y.
2.24 (1.25; 4.01), p = 0.006
4.94 (1.42; 17.2), p = 0.012
3.47 (1.39; 8.66), p = 0.008
3.37 (1.35; 8.39), p = 0.009
3.54 (1.41; 8.92), p = 0.007
Schulte C., 2015 [129]
430 ACS patients
682 patients with stable CAD
miR-19b
miR-132
miR-140-3p
miR-150
miR-186
miR-210
Up
Up
Up
Up
Up
Up
p, for CVD at 4 years3.59 (1.27–10.15), 0.025
2.85 (1.33–6.08), 0.022
2.88 (1.36–6.09), 0.022
2.14 (1.21–3.79), 0.022
2.08 (1.18–3.66), 0.022
3.10 (1.12–8.55), 0.039
Karakas M., 2017 [130]
7 STEMI patients
7 healthy controls
miR-146aUp p, MACE at 3 years1.329 (1.06–1.664), p = 0.01Xiao S., 2021 [132]
21 ACS patients
8 healthy controls
miR-208bUpp, elevated miR-208b expression was associated with reduced 6-month survival 5.08 (1.13–22.82), p = 0.03Alavi-Moghaddam M., 2018 [133]
142 patients with ACS or ischemic carotid eventmiR-1-3pUpp, expression levels during incident ischemia are risk factors of CVD at 6 y.2.73 (1.22–6.12), p = 0.014Badacz R., 2021 [134]
Left ventricular remodeling     
80 patients with STEMImiR-1Upp, for LVEDV increase >10% at 6 months, better value in combination with CK-MB, Nt-pro-BNP and CMRmiR-1: AUC: 0.680
miR-1 + CMR + NT-pro-BNP+ CK-MB: AUC: 0.890
Ma Q., 2020 [136]
44 ACS patientsmiR-1
miR-21
miR-29a
Up
Up
Up
p, for absolute change
for LVEDV at 6 months
miR-1 and miR-29b correlated with lower infarct zone,
miR-29b correlated with absolute change in LVEDV
Grabmaier, U., 2017 [137]
14 patients with STEMImiR-30a-5pUpp, for LVEF < 50%, and NT-proBNP > 150pg/mL at 6 monthsAUC: 0.750 (0.58–0.92)Maciejak A., 2018 [138]
198 ACS patientsmiR-21
miR-146a
Up
Up
miR-21 and miR-146a are early
biomarkers of LVR
p, for LVEDV increase >20% at 1 year
miR-146a: AUC: 0.818
miR-21: AUC: 0.719
in combination they have a higher predictive power
Liu, X., 2015 [139]
359 patients with MImiR-34a
miR-208a
Up
Up
p, for CVD and LVEDV increase >10%
at 6 months
miR-34a: OR 17.91 (2.07–98.81),
p = 0.003
miR-208b: OR 4.18 (1.36–12.83),
p = 0.012
combination of the two miRs:
OR 18.73 (1.96–101.23), p < 0.001)
Lv P., 2014 [140]
113 MI patients
59 healthy controls
miR-150Downp, for HF and LVEF at 1-year, better value in combination with BNPHR: 1.233 (1.125–1.352)
miR-150 alone: AUC: 0.764
miR-150 plus BNP: AUC: 0.807
Lin X., 2019 [141]
12 ACS patients
12 healthy controls
miR-29aUpThe greater increase in miR-29a, the greater increase in LVEDV at 90 days post MIearly miR-29a increase correlates with a negative outcome of post-MI LVRZile M.R., 2011 [142]
Ischemia/reperfusion injury     
44 patients deceased (19 for ACS, 25 as SCD)
18 trauma victims
miR-1
miR-499
miR-208
Up
Up
Up
p, miR-1 and miR-499 are sensitive markers to diagnose SCD compared ACS, and miR-208 for ACS vs. controlsSCD vs. ACS (AUC: 0.917)
SCD vs. ACS (AUC: 0.898)
ACS vs. controls (AUC: 0.855)
Pinchi E., 2019 [143]
24 patients deceased for ACS
8 patients deceased in accidents
miR-1Upp, might play role in cardiac remodeling3.8-fold increase in miR-1 in remote myocardiumBoštjancic E., 2010 [144]
47 patients deceased for ACS, including 23 from VF
8 trauma victims
miR-133a/bDownp, may contribute to VFFor VF, 2.9-fold decrease in miR-133a/b level in remote myocardiumBoštjancic E., 2018 [145]
ACS: acute coronary syndrome; AUC: area under the curve; BNP: brain natriuretic peptide; CMR: cardiac magnetic resonance; CVD: cardiovascular death; HF: heart failure; HR: hazard ratio; IS: ischemic stroke; LVAD: left ventricular assist device; LVEDV: left ventricular end-diastolic volume; LVR: left ventricular remodeling; MACE: major adverse cardiovascular event; MI: myocardial infarction; OR: odds ratio; p: prognostic; SCD: sudden cardiac death; VF: ventricular fibrillation.

3.2.2. miRs Associated with Myocardial I/R Injury and LVR

The most effective strategy for treating ACS is early and rapid myocardial reperfusion via percutaneous coronary intervention (PCI) [1]. However, restoring blood flow to the ischemic myocardium can induce further damage, known as I/R injury [146]. I/R contributes to increased mortality and morbidity [147]. In addition, many STEMI patients present on admission with a high-thrombus burden, which is known to carry an increased risk of distal embolization by fragmented thrombus debris [148]. This results in an increase in infarct size, as thrombotic debris constitutes a biologically active material that may exacerbate local injury via endothelial cell inflammation, higher vascular permeability, vasodilating/vasoconstricting factor imbalance, and complement and coagulation system activation [149,150].
In addition, heart failure following ischemia-induced LVR is a frequent complication of ACS. it prevalance is estimated at 30%, and is associated with a worsening of cardiovascular outcomes [151]. It accounts for 40–60% of CVD at 5 years. Besides many traditional prognostic risk factors, miRs have emerged as important biomarkers of LVR and SCD. In patients who have suffered from ACS, LVR has been associated with miR-1, miR-21, miR-29a, miR-30a, miR-34a, miR-146a, miR-150, and miR-208a expression (Table 3) [136,137,138,139,140,141,142]. Lin et al. found that out of the most common LVR-related miRs (miR-29a, -133a, -150, -192, -194, -34a, -208b, and -499), expression levels of miR-150 were observed as a good predictor for LVR and heart failure [141].
Although many cardioprotective strategies against I/R injury have been proposed, none have shown a clinically significant improvement in STEMI patients [148]. Some success has been reported with intracoronary oxygen therapy [147], or modulation of miR expression, such as miR-1, miR-21, miR-34a, and miR-146 [146,152,153,154,155].
There are only a few human studies on miR expression and the risk of I/R injury (Table 3) [143,144,145]. They include research on cardiomyocyte-derived miRs from deceased patients, such as miR-1, miR-208, miR-499 and miR-133, that are associated with extensive apoptosis, increased infarct size, and proneness to ventricular arrhythmia after ACS [143,144,145].
Unfortunately, the studied miRs differ between human and culture/animal studies. In a porcine model, the levels of miRs from the miR-15 family were increased in I/R injury, which directly promoted cardiac ischemia, while miR-15 inhibition protected against the injury [156]. In rats, up-regulated miR-21 protected against atherosclerotic growth, and protected cardiomyocytes against ACS and reactive oxygen species (ROS)-induced injury by targeting the programmed cell death 4 (PDCD4) gene [146]. Consistently, exogenous miR-21 was cardioprotective and decreased infarct size [11]. Regretfully, the clinical effect of miR-21 in individual patients with recent ACS regarding I/R injury and LVR is under discussion, as miR-21 obtains its anti-apoptotic effect through many diverse routes [157,158]. In the acute phase of ACS, overexpression of miR-21 protects against I/R injury, as decreases cardiomyocyte apoptosis [157]. However, later on, miR-21 protects fibroblasts from apoptosis, which is inconvenient as this exaggerates scarring and increases LVR [157]. Zhang et al. revealed that higher levels of miR-21 during hospitalization for ACS were associated with an increased risk of hospitalization for heart failure (Table 3) [116].
Recently, new miRs have been proposed as potential therapeutic targets after ACS [159,160,161]. In animal models, miR-19a/19b, miR-93, and miR-144 protected cardiomyocytes against I/R injury and LVR following ACS [159,160,161]. Their delivery stimulated cardiac regeneration, reduced border zone fibrosis, inflammation and apoptosis. In contrast, as miR-1 and miR-34a increase apoptosis and inflammatory response, their inhibition would be beneficial after ACS [162,163,164].
In summary, the mechanism of I/R injury and LVR are complex and multifactorial, thus the potential therapeutic approach should be a panel of microRNA-mimics and antagomirs [162,163,164,165,166,167].

4. Conclusions

The knowledge on miRs is constantly growing, and although not all molecular mechanisms have been recognized, miRs have entered the era of clinical application and therapy. Some miRs can be used as ‘quick’ diagnostic tests for CAD presence and severity. miR-1, miR-133, and miR-499 have the greatest potential for diagnostic biomarker of ACS, as they can be detected in blood samples before troponins. Many miRs can be used as markers of prognosis, including the widely studied miR-21 for I/R injury and LVR risk.
Much more limitations concern therapeutic approach, although miR-mimics and antagomirs are already on the board (animal studies). At different stages of atherosclerosis, different miRs can be considered for therapy, as a variety of miRs regulate particular stages of atherosclerotic development. At the beginning of atherosclerosis, a promising target is miR-142-3p that may have a role in the prevention and treatment of atherosclerosis. In addition, miR-92a seems an interesting target as it is associated with plaque progression and instability. Caution must be paid to individual miRs which role varies depending on their source or stage of atherosclerosis, e.g., miR-21, miR-155, or miR-33a/b that regulate cholesterol homeostasis and oncogene expression. Of note, single miRs can inhibit target mRNAs irreversibly, as many are very stable in circulation. Thus, some miRs could be given locally to avoid their systemic action.
In conclusion, miRs repeatedly gather support as being valuable in clinical practice as diagnostic biomarkers for CAD and ACS, prognostic biomarkers for cardiovascular outcomes, and potential therapeutic agents. Some have already been used in phase II and III clinical studies on cancer. We may expect that in a near future they will also enter into clinical practice in cardiology. The research is still ongoing.

Author Contributions

Conceptualization, A.K.-Z.; methodology, A.K.-Z., R.B. and T.P.; writing—original draft preparation, A.K.-Z., R.B. and T.P.; writing—review and editing, A.K.-Z.; visualization, A.K.-Z. and T.P.; supervision, T.P.; project administration, A.K.-Z.; funding acquisition, A.K.-Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Jagiellonian University Medical College, grant no N41/DBS/000752.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tanase, D.M.; Gosav, E.M.; Ouatu, A.; Badescu, M.C.; Dima, N.; Ganceanu-Rusu, A.R.; Popescu, D.; Floria, M.; Rezus, E.; Rezus, C. Current Knowledge of MicroRNAs (miRNAs) in Acute Coronary Syndrome (ACS): ST-Elevation Myocardial Infarction (STEMI). Life 2021, 11, 1057. [Google Scholar] [CrossRef] [PubMed]
  2. Ren, J.; Zhang, J.; Xu, N.; Han, G.; Geng, Q.; Song, J.; Li, S.; Zhao, J.; Chen, H. Signature of circulating microRNAs as po-tential biomarkers in vulnerable coronary artery disease. PLoS ONE 2013, 8, e80738. [Google Scholar] [CrossRef] [Green Version]
  3. Badacz, R.; Przewlocki, T.; Gacoń, J.; Stępień, E.; Enguita, F.J.; Karch, I.; Żmudka, K.; Kabłak-Ziembicka, A. Circulating miRNA levels differ with respect to carotid plaque characteristics and symptom occurrence in patients with carotid artery stenosis and provide information on future cardiovascular events. Adv. Interv. Cardiol. 2018, 14, 75–84. [Google Scholar] [CrossRef] [PubMed]
  4. Kabłak-Ziembicka, A.; Przewłocki, T. Clinical Significance of Carotid Intima-Media Complex and Carotid Plaque As-sessment by Ultrasound for the Prediction of Adverse Cardiovascular Events in Primary and Secondary Care Patients. J. Clin. Med. 2021, 10, 4628. [Google Scholar] [CrossRef]
  5. Aboyans, V.; Ricco, J.B.; Bartelink, M.E.L.; Björck, M.; Brodmann, M.; Cohnert, T.; Collet, J.P.; Czerny, M.; De Carlo, M.; Debus, S.; et al. 2017 ESC Guidelines on the Diagnosis and Treatment of Peripheral Arterial Diseases, in collaboration with the European Society for Vascular Surgery (ESVS): Document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteries Endorsed by: The European Stroke Organization (ESO) The Task Force for the Diagnosis and Treatment of Peripheral Arterial Diseases of the European Society of Cardiol-ogy (ESC) and of the European Society for Vascular Surgery (ESVS). Eur. Heart J. 2018, 39, 763–816. [Google Scholar]
  6. Ralapanawa, U.; Sivakanesan, R. Epidemiology and the Magnitude of Coronary Artery Disease and Acute Coronary Syndrome: A Narrative Review. J. Epidemiol. Glob. Health 2021, 11, 169–177. [Google Scholar] [CrossRef]
  7. Przewłocki, T.; Kablak-Ziembicka, A.; Tracz, W.; Kozanecki, A.; Kopeć, G.; Rubiś, P.; Kostkiewicz, M.; Rosławiecka, A.; Rzeźnik, D.; Stompór, T. Renal artery stenosis in patients with coronary artery disease. Kardiol. Pol. 2008, 66, 856–862. [Google Scholar]
  8. Yates, L.A.; Norbury, C.J.; Gilbert, R.J. The Long and Short of MicroRNA. Cell 2013, 153, 516–519. [Google Scholar] [CrossRef] [Green Version]
  9. Creemers, E.E.; Tijsen, A.J.; Pinto, Y.M. Circulating microRNAs: Novel biomarkers and extracellular communicators in cardiovascular disease? Circ. Res. 2012, 110, 483–495. [Google Scholar] [CrossRef] [Green Version]
  10. Ghafouri-Fard, S.; Gholipour, M.; Taheri, M. Role of MicroRNAs in the Pathogenesis of Coronary Artery Disease. Front. Cardiovasc. Med. 2021, 8, 632392. [Google Scholar] [CrossRef]
  11. Cipollone, F.; Felicioni, L.; Sarzani, R.; Ucchino, S.; Spigonardo, F.; Mandolini, C.; Malatesta, S.; Bucci, M.; Mammarella, C.; Santovito, D.; et al. A unique Microrna signature associated with plaque instability in humans. Stroke 2011, 42, 2556–2563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Poredoš, P.; Šabovič, M.; Mijovski, M.B.; Nikolajević, J.; Antignani, P.L.; Paraskevas, K.I.; Mikhailidis, D.P.; Blinc, A. Inflammatory and Prothrombotic Biomarkers, DNA Polymorphisms, MicroRNAs and Personalized Medicine for Patients with Peripheral Arterial Disease. Int. J. Mol. Sci. 2022, 23, 12054. [Google Scholar] [CrossRef]
  13. Puz, P.; Lasek-Bal, A.; Warsz-Wianecka, A.; Kaźmierski, M. Prevalence of atherosclerotic stenosis of carotid and cerebral arteries in patients with stable or unstable coronary artery disease. Pol. Arch. Intern. Med. 2020, 130, 412–419. [Google Scholar] [CrossRef] [PubMed]
  14. Gacoń, J.; Przewłocki, T.; Podolec, J.; Badacz, R.; Pieniążek, P.; Mleczko, S.; Ryniewicz, W.; Żmudka, K.; Kabłak-Ziembicka, A. Prospective study on the prognostic value of repeated carotid intima-media thickness assessment in patients with coronary and extra coronary steno-occlusive arterial disease. Pol. Arch. Intern. Med. 2019, 129, 808–817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Vedanthan, R.; Seligman, B.; Fuster, V. Global perspective on acute coronary syndrome: A burden on the young and poor. Circ Res. 2014, 114, 1959–1975. [Google Scholar] [CrossRef] [Green Version]
  16. Stefan, J.; Bueno, H. Epidemiology of acute coronary syndromes. In The ESC Textbook of Cardiovascular Medicine, 3rd ed.; Camm, A.J., Ed.; The European Society of Cardiology Series; ESC Publications: Oxford, UK, 2018. [Google Scholar] [CrossRef]
  17. Sayed, D.; Abdellatif, M. MicroRNAs in Development and Disease. Physiol. Rev. 2011, 91, 827–887. [Google Scholar] [CrossRef]
  18. Weber, M.; Baker, M.B.; Moore, J.P.; Searles, C.D. MiR-21 is induced in endothelial cells by shear stress and modulates apoptosis and eNOS activity. Biochem. Biophys. Res. Commun. 2010, 393, 643–648. [Google Scholar] [CrossRef] [Green Version]
  19. Navickas, R.; Gal, D.; Laucevičius, A.; Taparauskaitė, A.; Zdanytė, M.; Holvoet, P. Identifying circulating microRNAs as biomarkers of cardiovascular disease: A systematic review. Cardiovasc. Res. 2016, 111, 322–337. [Google Scholar] [CrossRef] [Green Version]
  20. Nikas, J.B.; Low, W.C. ROC-supervised principal component analysis in connection with the diagnosis of diseases. Am. J. Transl. Res. 2011, 3, 180–196. [Google Scholar]
  21. Fichtlscherer, S.; De Rosa, S.; Fox, H.; Schwietz, T.; Fischer, A.; Liebetrau, C.; Weber, M.; Hamm, C.W.; Röxe, T.; Müller-Ardogan, M.; et al. Circulating Micrornas in patients with coronary artery disease. Circ. Res. 2010, 107, 677–684. [Google Scholar] [CrossRef] [Green Version]
  22. Zhang, C. MicroRNA-145 in vascular smooth muscle cell biology: A new therapeutic target for vascular disease. Cell Cycle 2009, 8, 3469–3473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gnatenko, D.V.; Dunn, J.J.; Schwedes, J.; Bahou, W.F. Transcript profiling of human platelets using microarray and serial analysis of gene expression (SAGE). Methods Mol. Biol. 2009, 496, 245–272. [Google Scholar] [PubMed]
  24. Landry, P.; Plante, I.; Ouellet, D.L.; Perron, M.P.; Rousseau, G.; Provost, P. Existence of a microRNA pathway in anucleate platelets. Nat. Struct. Mol. Biol. 2009, 16, 961–966. [Google Scholar] [CrossRef] [Green Version]
  25. Pordzik, J.; Pisarz, K.; De Rosa, S.; Jones, A.D.; Eyileten, C.; Indolfi, C.; Małek, L.; Postula, M. The Potential Role of Platelet-Related microRNAs in the Development of Cardiovascular Events in High-Risk Populations, Including Diabetic Patients: A Review. Front. Endocrinol. 2018, 9, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Kaudewitz, D.; Skroblin, P.; Bender, L.H.; Barwari, T.; Willeit, P.; Pechlaner, R.; Sunderland, N.P.; Willeit, K.; Morton, A.C.; Arm-strong, P.C.; et al. Association of MicroRNAs and YRNAs With Platelet Function. Circ Res. 2016, 118, 420–432. [Google Scholar] [CrossRef]
  27. Willeit, P.; Zampetaki, A.; Dudek, K.; Kaudewitz, D.; King, A.; Kirkby, N.S.; Crosby-Nwaobi, R.; Prokopi, M.; Drozdov, I.; Langley, S.R.; et al. Circulating micrornas as novel biomarkers for platelet activation. Circ. Res. 2013, 112, 595–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Liu, W.W.; Wang, H.; Chen, X.H.; Fu, S.W.; Liu, M.L. miR-34b-3p may promote antiplatelet efficiency of aspirin by inhibiting thromboxane synthase expression. Thromb. Haemost. 2019, 119, 1451–1460. [Google Scholar] [CrossRef]
  29. Shi, R.; Ge, L.; Zhou, X.; Ji, W.-J.; Lu, R.-Y.; Zhang, Y.-Y.; Zeng, S.; Liu, X.; Zhao, J.-H.; Zhang, W.-C.; et al. Decreased platelet miR-223 expression is associated with high on-clopidogrel platelet reactivity. Thromb. Res. 2013, 131, 508–513. [Google Scholar] [CrossRef]
  30. Krammer, T.L.; Mayr, M.; Hackl, M. microRNAs as promising biomarkers of platelet activity in antiplatelet therapy monitoring. Int. J. Mol. Sci. 2020, 21, 3477. [Google Scholar] [CrossRef]
  31. Czajka, P.; Fitas, A.; Jakubik, D.; Eyileten, C.; Gasecka, A.; Wicik, Z.; Siller-Matula, J.M.; Filipiak, K.J.; Postula, M. MicroRNA as Potential Biomarkers of Platelet Function on Antiplatelet Therapy: A Review. Front. Physiol. 2021, 12, 652579. [Google Scholar] [CrossRef]
  32. Wang, D.; Atanasov, A.G. The microRNAs Regulating Vascular Smooth Muscle Cell Proliferation: A Minireview. Int. J. Mol. Sci. 2019, 20, 324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Bennett, M.R.; Sinha, S.; Owens, G.K. Vascular Smooth Muscle Cells in Atherosclerosis. Circ. Res. 2016, 118, 692–702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Badi, I.; Burba, I.; Ruggeri, C.; Zeni, F.; Bertolotti, M.; Scopece, A.; Pompilio, G.; Raucci, A. MicroRNA-34a induces vascular smooth muscle cells senescence by SIRT1 downregulation and promotes the expression of age-associated pro-inflammatory secretory factors. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 1304–1311. [Google Scholar] [CrossRef] [PubMed]
  35. Ahmed, S.; Warren, D.T. Vascular smooth muscle cell contractile function and mechanotransduction. Vessel Plus 2018, 2, 36. [Google Scholar] [CrossRef] [Green Version]
  36. Nguyen, D.N.D.; Chilian, W.M.; Zain, S.M.; Daud, M.F.; Pung, Y.-F. MicroRNA regulation of vascular smooth muscle cells and its significance in cardiovascular diseases. Can. J. Physiol. Pharmacol. 2021, 99, 827–838. [Google Scholar] [CrossRef]
  37. Baran, J.; Kleczyński, P.; Niewiara, Ł.; Podolec, J.; Badacz, R.; Gackowski, A.; Pieniążek, P.; Legutko, J.; Żmudka, K.; Przewłocki, T.; et al. Importance of Increased Arterial Resistance in Risk Prediction in Patients with Cardiovascular Risk Factors and Degenerative Aortic Stenosis. J. Clin. Med. 2021, 10, 2109. [Google Scholar] [CrossRef]
  38. Arnăutu, S.F.; Morariu, V.I.; Arnăutu, D.A.; Tomescu, M.C. The Predictive Value of Carotid Artery Strain and Strain-Rate in Assessing the 3-Year Risk for Stroke and Acute Coronary Syndrome in Patients with Metabolic Syndrome. Rev. Cardiovasc. Med. 2022, 23, 146. [Google Scholar] [CrossRef]
  39. Zhang, J.; Starkuviene, V.; Erfle, H.; Wang, Z.; Gunkel, M.; Zeng, Z.; Sticht, C.; Kan, K.; Rahbari, N.; Keese, M. High-content analysis of microRNAs involved in the phenotype regulation of vascular smooth muscle cells. Sci. Rep. 2022, 12, 3498. [Google Scholar] [CrossRef]
  40. Yang, F.; Chen, Q.; He, S.; Yang, M.; Maguire, E.M.; An, W.; Afzal, T.A.; Luong, L.A.; Zhang, L.; Xiao, Q. miR-22 is a novel mediator of vascular smooth muscle cell phenotypic modulation and neointima formation. Circulation 2018, 137, 1824–1841. [Google Scholar] [CrossRef]
  41. Zhang, Y.-N.; Xie, B.-D.; Sun, L.; Chen, W.; Jiang, S.-L.; Liu, W.; Bian, F.; Tian, H.; Li, R.-K. Phenotypic switching of vascular smooth muscle cells in the ‘normal region’ of aorta from atherosclerosis patients is regulated by miR-145. J. Cell. Mol. Med. 2016, 20, 1049–1061. [Google Scholar] [CrossRef] [Green Version]
  42. Patel, N.; Chin, D.D.; Magee, G.A.; Chung, E.J. Therapeutic Response of miR-145 Micelles on Patient-Derived Vascular Smooth Muscle Cells. Front. Digit. Health 2022, 4, 836579. [Google Scholar] [CrossRef] [PubMed]
  43. Lovren, F.; Pan, Y.; Quan, A.; Singh, K.K.; Shukla, P.C.; Gupta, N.; Steer, B.M.; Ingram, A.J.; Gupta, M.; Al-Omran, M.; et al. MicroRNA-145 targeted therapy reduces atherosclerosis. Circulation 2012, 126, S81–S90. [Google Scholar] [CrossRef] [PubMed]
  44. Gao, H.; Guddeti, R.R.; Matsuzawa, Y.; Liu, L.-P.; Su, L.-X.; Guo, D.; Nie, S.-P.; Du, J.; Zhang, M. Plasma Levels of microRNA-145 Are Associated with Severity of Coronary Artery Disease. PLoS ONE 2015, 10, e0123477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wang, J.; Qian, H.-L.; Chen, S.-Y.; Huang, W.-P.; Huang, D.-N.; Hao, H.-Y.; Ren, K.-F.; Wang, Y.-B.; Fu, G.-S.; Ji, J. miR-22 eluting cardiovascular stent based on a self-healable spongy coating inhibits in-stent restenosis. Bioact. Mater. 2021, 6, 4686–4696. [Google Scholar] [CrossRef]
  46. Wang, D.; Deuse, T.; Stubbendorff, M.; Chernogubova, E.; Erben, R.G.; Eken, S.M.; Jin, H.; Li, Y.; Busch, A.; Heeger, C.H.; et al. Local MicroRNA modulation using a novel anti-miR-21-eluting stent effectively prevents experimental in-stent restenosis. Arter. Thromb. Vasc. Biol. 2015, 35, 1945–1953. [Google Scholar] [CrossRef] [Green Version]
  47. Dai, H.; Wang, J.; Shi, Z.; Ji, X.; Huang, Y.; Zhou, R. Predictive value of miRNA-21 on coronary restenosis after percuta-neous coronary intervention in patients with coronary heart disease. A protocol for systematic review and meta-analysis. Medicine 2021, 100, e24966. [Google Scholar] [CrossRef]
  48. Tekieli, Ł.; Musiałek, P.; Kablak-Ziembicka, A.; Trystuła, M.; Przewłocki, T.; Legutko, J.; Dzierwa, K.; Maciejewski, D.; Michalski, M.; Pieniążek, P. Severe, recurrent in-stent carotid restenosis: Endovascular approach, risk factors. Results from a prospective academic registry of 2637 consecutive carotid artery stenting procedures (TARGET-CAS). Adv. Interv. Cardiol. 2019, 15, 465–471. [Google Scholar] [CrossRef]
  49. Soriano-Arroquia, A.; McCormick, R.; Molloy, A.P.; McArdle, A.; Goljanek-Whysall, K. Age-related changes in miR-143-3p:Igfbp5 interactions affect muscle regeneration. Aging Cell 2016, 15, 361–369. [Google Scholar] [CrossRef] [Green Version]
  50. Li, H.; Chen, M.; Feng, Q.; Zhu, L.; Bai, Z.; Wang, B.; Guo, Z.; Hou, A.; Li, H. MicroRNA-34a in coronary heart disease: Correlation with disease risk, blood lipid, stenosis degree, inflammatory cytokines, and cell adhesion molecules. J. Clin. Lab. Anal. 2021, 36, e24138. [Google Scholar] [CrossRef]
  51. Suárez, Y.; Fernández-Hernando, C.; Yu, J.; Gerber, S.A.; Harrison, K.D.; Pober, J.S.; Iruela-Arispe, M.L.; Merkenschlager, M.; Sessa, W.C. Dicer-dependent endothelial microRNAs are necessary for postnatal angiogenesis. Proc. Natl. Acad. Sci. USA 2008, 105, 14082–14087. [Google Scholar] [CrossRef] [Green Version]
  52. Schober, A.; Maleki, S.S.; Nazari-Jahantigh, M. Regulatory Non-coding RNAs in Atherosclerosis. In Textbook: Prevention and Treatment of Atherosclerosis: Improving State-of-the-Art Management and Search for Novel Targets; Von Eckardstein, A., Binder, C.J., Eds.; Springer: Cham, Switzerland, 2022. [Google Scholar] [PubMed]
  53. Fish, J.E.; Santoro, M.M.; Morton, S.U.; Yu, S.; Yeh, R.-F.; Wythe, J.D.; Ivey, K.N.; Bruneau, B.G.; Stainier, D.Y.R.; Srivastava, D. miR-126 regulates angiogenic signaling and vascular integrity. Dev. Cell 2008, 15, 272–284. [Google Scholar] [CrossRef] [PubMed]
  54. Zernecke, A.; Bidzhekov, K.; Noels, H.; Shagdarsuren, E.; Gan, L.; Denecke, B.; Hristov, M.; Köppel, T.; Jahantigh, M.N.; Lutgens, E.; et al. Delivery of MicroRNA-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci. Signal. 2009, 2, ra81. [Google Scholar] [CrossRef] [PubMed]
  55. Schober, A.; Nazari-Jahantigh, M.; Wei, Y.; Bidzhekov, K.; Gremse, F.; Grommes, J.; Megens, R.T.; Heyll, K.; Noels, H.; Hristov, M.; et al. MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat. Med. 2014, 20, 368–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Tang, F.; Yang, T.-L. MicroRNA-126 alleviates endothelial cells injury in atherosclerosis by restoring autophagic flux via inhibiting of PI3K/Akt/mTOR pathway. Biochem. Biophys. Res. Commun. 2018, 495, 1482–1489. [Google Scholar] [CrossRef]
  57. Li, H.-Y.; Zhao, X.; Liu, Y.-Z.; Meng, Z.; Wang, D.; Yang, F.; Shi, Q.-W. Plasma MicroRNA-126-5p is Associated with the Complexity and Severity of Coronary Artery Disease in Patients with Stable Angina Pectoris. Cell. Physiol. Biochem. 2016, 39, 837–846. [Google Scholar] [CrossRef]
  58. Wang, X.; Lian, Y.; Wen, X.; Guo, J.; Wang, Z.; Jiang, S.; Hu, Y. Expression of miR-126 and its potential function in coronary ar-tery disease. Afr. Health Sci. 2017, 17, 474–480. [Google Scholar] [CrossRef] [Green Version]
  59. Huang, R.; Hu, Z.; Cao, Y.; Li, H.; Zhang, H.; Su, W.; Xu, Y.; Liang, L.; Melgiri, N.; Jiang, L. MiR-652-3p inhibition enhances endothelial repair and reduces atherosclerosis by promoting Cyclin D2 expression. eBioMedicine 2019, 40, 685–694. [Google Scholar] [CrossRef] [Green Version]
  60. Qin, B.; Shu, Y.; Long, L.; Li, H.; Men, X.; Feng, L.; Yang, H.; Lu, Z. MicroRNA-142-3p Induces Atherosclero-sis-Associated Endothelial Cell Apoptosis by Directly Targeting Rictor. Cell. Physiol. Biochem. 2018, 47, 1589–1603. [Google Scholar] [CrossRef]
  61. Hartmann, P.; Zhou, Z.; Natarelli, L.; Wei, Y.; Nazari-Jahantigh, M.; Zhu, M.; Grommes, G.; Steffens, S.; Weber, C.; Schober, A. Endothelial dicer promotes atherosclerosis and vascular inflammation by miRNA-103-mediated suppression of KLF4. Nat. Commun. 2016, 7, 10521. [Google Scholar] [CrossRef] [Green Version]
  62. Jiang, L.; Qiao, Y.; Wang, Z.; Ma, X.; Wang, H.; Li, J. Inhibition of microRNA-103 attenuates inflammation and endoplasmic re-ticulum stress in atherosclerosis through disrupting the PTEN-mediated MAPK signaling. J. Cell Physiol. 2020, 235, 380–393. [Google Scholar] [CrossRef]
  63. Chen, Z.; Wen, L.; Martin, M.; Hsu, C.-Y.; Fang, L.; Lin, F.-M.; Lin, T.-Y.; Geary, M.J.; Geary, G.G.; Zhao, Y.; et al. Oxidative stress activates endothelial innate immunity via sterol regulatory element binding protein 2 (SREBP2) transactivation of MicroRNA-92a. Circulation 2015, 131, 805–814. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, Y.; Miao, C.; Cui, J.; Bian, X. miR-92a-3p promotes ox-LDL induced-apoptosis in HUVECs via targeting SIRT6 and activating MAPK signaling pathway. Braz. J. Med. Biol. Res. 2021, 54, e9386. [Google Scholar] [CrossRef] [PubMed]
  65. Ichikawa, K.; Miyoshi, T.; Osawa, K.; Miki, T.; Ito, H. Increased Circulating Malondialdehyde-Modified Low-Density Lipoprotein Level Is Associated with High-Risk Plaque in Coronary Computed Tomography Angiography in Patients Receiving Statin Therapy. J. Clin. Med. 2021, 10, 1480. [Google Scholar] [CrossRef] [PubMed]
  66. Niculescu, L.S.; Simionescu, N.; Sanda, G.M.; Carnuta, M.G.; Stancu, C.S.; Popescu, A.C.; Popescu, M.; Vlad, A.; Dimulescu, D.R.; Simionescu, M.; et al. MiR-486 and miR-92a Identified in Circulating HDL Discriminate between Stable and Vulnerable Coronary Artery Disease Patients. PLoS ONE 2015, 10, e0140958. [Google Scholar] [CrossRef]
  67. Gao, W.; He, H.-W.; Wang, Z.-M.; Zhao, H.; Lian, X.-Q.; Wang, Y.-S.; Zhu, J.; Yan, J.-J.; Zhang, D.-G.; Yang, Z.-J.; et al. Plasma levels of lipometabolism-related miR-122 and miR-370 are increased in patients with hyperlipidemia and associated with coronary artery disease. Lipids Health Dis. 2012, 11, 55. [Google Scholar] [CrossRef] [Green Version]
  68. Chen, J.; Xu, L.; Hu, Q.; Yang, S.; Zhang, B.; Jiang, H. MiR-17-5p as circulating biomarkers for the severity of coronary atherosclerosis in coronary artery disease. Int. J. Cardiol. 2015, 197, 123–124. [Google Scholar] [CrossRef]
  69. Yang, S.; Fan, T.; Hu, Q.; Xu, W.; Yang, J.; Xu, C.; Zhang, B.; Chen, J.; Jiang, H. Downregulation of microRNA-17-5p improves cardiac function after myocardial infarction via attenuation of apoptosis in endothelial cells. Mol. Genet. Genomics 2018, 293, 883–894. [Google Scholar] [CrossRef]
  70. Canfran-Duque, A.; Rotllan, N.; Zhang, X.; Fernandez-Fuertes, M.; Ramirez-Hidalgo, C.; Araldi, E. Macrophage deficiency of miR-21 promotes apoptosis, plaque necrosis, and vascular inflammation during atherogenesis. EMBO Mol. Med. 2017, 9, 1244–1262. [Google Scholar] [CrossRef]
  71. Nazari-Jahantigh, M.; Wei, Y.; Noels, H.; Akhtar, S.; Zhou, Z.; Koenen, R.R.; Heyll, K.; Gremse, F.; Kiessling, F.; Grommes, J.; et al. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J. Clin. Investig. 2012, 122, 4190–4202. [Google Scholar] [CrossRef] [Green Version]
  72. Price, N.L.; Rotllan, N.; Canfrán-Duque, A.; Zhang, X.; Pati, P.; Arias, N.; Moen, J.; Mayr, M.; Ford, D.A.; Baldán, Á.; et al. Genetic Dissection of the Impact of miR-33a and miR-33b during the Progression of Atherosclerosis. Cell Rep. 2017, 21, 1317–1330. [Google Scholar] [CrossRef] [Green Version]
  73. Wei, Y.; Zhu, M.; Corbalán-Campos, J.; Heyll, K.; Weber, C.; Schober, A. Regulation of Csf1r and Bcl6 in macrophages mediates the stage-specific effects of MicroRNA-155 on atherosclerosis. Arter. Thromb. Vasc. Biol. 2015, 35, 796–803. [Google Scholar] [CrossRef] [PubMed]
  74. Androulidaki, A.; Iliopoulos, D.; Arranz, A.; Doxaki, C.; Schworer, S.; Zacharioudaki, V.; Margioris, A.N.; Tsichlis, P.N.; Tsatsanis, C. The kinase Akt1 controls macro-phage response to lipopolysaccharide by regulating microRNAs. Immunity 2009, 31, 220–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Rakipovski, G.; Rolin, B.; Nøhr, J.; Klewe, I.; Frederiksen, K.S.; Augustin, R.; Hecksher-Sørensen, J.; Ingvorsen, C.; Polex-Wolf, J.; Bjerre Knudsen, L. The GLP-1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE(−/−) and LDLr(−/−) mice by a mechanism that includes inflammatory pathways. JACC Basic Transl. Sci. 2018, 3, 844–857. [Google Scholar] [CrossRef] [PubMed]
  76. Jankauskas, S.; Gambardella, J.; Sardu, C.; Lombardi, A.; Santulli, G. Functional Role of miR-155 in the Pathogenesis of Diabetes Mellitus and Its Complications. Non-Coding RNA 2021, 7, 39. [Google Scholar] [CrossRef] [PubMed]
  77. Musialek, P.; Pieniazek, P.; Tracz, W.; Tekieli, L.; Przewlocki, T.; Kablak-Ziembicka, A.; Motyl, R.; Moczulski, Z.; Stepniewski, J.; Trystula, M.; et al. Safety of embolic protection device-assisted and unprotected intravascular ultrasound in evaluating carotid artery atherosclerotic lesions. Med. Sci. Monit. 2012, 18, MT7–MT18. [Google Scholar] [CrossRef] [Green Version]
  78. Chiorescu, R.M.; Mocan, M.; Inceu, A.I.; Buda, A.P.; Blendea, D.; Vlaicu, S.I. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int. J. Mol. Sci. 2022, 23, 13638. [Google Scholar] [CrossRef]
  79. Su, Y.L.; Wang, X.; Mann, M.; Adamus, T.P.; Wang, D.; Moreira, D.F.; Zhang, Z.; Ouyang, C.; He, X.; Zhang, B.; et al. Myeloid cell-targeted miR-146a mimic inhibits NF-kappaB-driven inflammation and leukemia progression in vivo. Blood 2020, 135, 167–180. [Google Scholar] [CrossRef] [Green Version]
  80. Yang, K.; He, Y.S.; Wang, X.Q.; Lu, L.; Chen, Q.J.; Liu, J.; Sun, Z.; Shen, W.F. MiR-146a inhibits oxidized low-density lip-oprotein-induced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett. 2011, 585, 854–860. [Google Scholar] [CrossRef] [Green Version]
  81. Arroyo, A.B.; Águila, S.; Fernández-Pérez, M.P.; Reyes-García, A.M.L.; Reguilón-Gallego, L.; Zapata-Martínez, L.; Vicente, V.; Mar-tínez, C.; González-Conejero, R. miR-146a in Cardiovascular Diseases and Sepsis: An Additional Burden in the Inflamma-tory Balance? Thromb. Haemost. 2021, 121, 1138–1150. [Google Scholar] [CrossRef]
  82. Pan, J.; Alimujiang, M.; Chen, Q.; Shi, H.; Luo, X. Exosomes derived from miR-146a-modified adipose-derived stem cells attenuate acute myocardial infarction−induced myocardial damage via downregulation of early growth response factor 1. J. Cell. Biochem. 2018, 120, 4433–4443. [Google Scholar] [CrossRef]
  83. Zhang, T.; Ma, Y.; Gao, L.; Mao, C.; Zeng, H.; Wang, X.; Sun, Y.; Gu, J.; Wang, Y.; Chen, K.; et al. MicroRNA-146a protects against myocardial ischaemia reperfusion injury by targeting Med1. Cell Mol. Biol. Lett. 2019, 24, 62. [Google Scholar] [CrossRef] [PubMed]
  84. Mir, R.; Elfaki, I.; Khullar, N.; Waza, A.; Jha, C.; Mir, M.; Nisa, S.; Mohammad, B.; Mir, T.; Maqbool, M.; et al. Role of Selected miRNAs as Diagnostic and Prognostic Biomarkers in Cardiovascular Diseases, Including Coronary Artery Disease, Myocardial Infarction and Atherosclerosis. J. Cardiovasc. Dev. Dis. 2021, 8, 22. [Google Scholar] [CrossRef] [PubMed]
  85. Singh, S.; de Ronde, M.W.J.; Kok, M.G.M.; Beijk, M.A.; De Winter, R.J.; van der Wal, A.C.; Sondermeijer, B.M.; Meijers, J.C.M.; E Creemers, E.; Pinto-Sietsma, S.-J. MiR-223-3p and miR-122-5p as circulating biomarkers for plaque instability. Open Heart 2020, 7, e001223. [Google Scholar] [CrossRef] [PubMed]
  86. Guo, J.-F.; Zhang, Y.; Zheng, Q.-X.; Zhang, Y.; Zhou, H.-H.; Cui, L.-M. Association between elevated plasma microRNA-223 content and severity of coronary heart disease. Scand. J. Clin. Lab. Investig. 2018, 78, 373–378. [Google Scholar] [CrossRef] [PubMed]
  87. Zhu, L. The correlations of circulating microRNA-133a with the risk and severity of coronary heart disease. Int. J. Clin. Exp. Med. 2017, 10, 972–978. [Google Scholar]
  88. Abdallah, H.Y.; Hassan, R.; Fareed, A.; Abdelgawad, M.; Mostafa, S.A.; Mohammed, E.A.-M. Identification of a circulating microRNAs biomarker panel for non-invasive diagnosis of coronary artery disease: Case–control study. BMC Cardiovasc. Disord. 2022, 22, 286. [Google Scholar] [CrossRef]
  89. De Rosa, S.; Fichtlscherer, S.; Lehmann, R.; Assmus, B.; Dimmeler, S.; Zeiher, A.M. Transcoronary Concentration Gradients of Circulating MicroRNAs. Circulation 2011, 124, 1936–1944. [Google Scholar] [CrossRef] [Green Version]
  90. Kuwabara, Y.; Ono, K.; Horie, T.; Nishi, H.; Nagao, K.; Kinoshita, M.; Watanabe, S.; Baba, O.; Kojima, Y.; Shizuta, S.; et al. Increased MicroRNA-1 and MicroRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ. Cardiovasc. Genet. 2011, 4, 446–454. [Google Scholar] [CrossRef]
  91. Lu, H.-Q.; Liang, C.; He, Z.-Q.; Fan, M.; Wu, Z.-G. Circulating miR-214 is associated with the severity of coronary artery disease. J. Geriatr. Cardiol. 2013, 10, 34–38. [Google Scholar] [CrossRef]
  92. Zhang, M.; Cheng, Y.-J.; Sara, J.D.; Liu, L.-J.; Zhao, X.; Gao, H. Circulating MicroRNA-145 is Associated with Acute Myocardial Infarction and Heart Failure. Chin. Med. J. 2017, 130, 51–56. [Google Scholar] [CrossRef]
  93. Wang, W.; Li, T.; Gao, L.; Li, Y.; Sun, Y.; Yao, H.-C. Plasma miR-208b and miR-499: Potential Biomarkers for Severity of Coronary Artery Disease. Dis. Markers 2019, 2019, 9842427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Wang, S.; He, W.; Wang, C. MiR-23a Regulates the Vasculogenesis of Coronary Artery Disease by Targeting Epidermal Growth Factor Receptor. Cardiovasc. Ther. 2016, 34, 199–208. [Google Scholar] [CrossRef] [PubMed]
  95. Pereira-Da-Silva, T.; Napoleão, P.; Costa, M.; Gabriel, A.; Selas, M.; Silva, F.; Enguita, F.; Ferreira, R.; Carmo, M. Circulating miRNAs Are Associated with the Systemic Extent of Atherosclerosis: Novel Observations for miR-27b and miR-146. Diagnostics 2021, 11, 318. [Google Scholar] [CrossRef]
  96. Przewłocki, T.; Kablak-Ziembicka, A.; Kozanecki, A.; Rzeźnik, D.; Pieniazek, P.; Musiałek, P.; Piskorz, A.; Sokołowski, A.; Rosławiecka, A.; Tracz, W. Polyvascular extracoronary atherosclerotic disease in patients with coronary artery disease. Kardiol. Pol. 2009, 67, 978–984. [Google Scholar]
  97. Vrsalovic, M.; Presecki, A.V.; Aboyans, V. Cardiac troponins predict mortality and cardiovascular outcomes in patients with peripheral artery disease: A systematic review and meta-analysis of adjusted observational studies. Clin. Cardiol. 2022, 45, 198–204. [Google Scholar] [CrossRef]
  98. Kabłak-Ziembicka, A.; Rosławiecka, A.; Badacz, R.; Sokołowski, A.; Rzeźnik, D.; Trystuła, M.; Musiałek, P.; Przewłocki, T. Simple clinical scores to predict blood pressure and renal function response to renal artery stenting for atherosclerotic renal artery stenosis. Pol. Arch. Intern. Med. 2020, 130, 953–959. [Google Scholar] [CrossRef]
  99. Faccini, J.; Ruidavets, J.-B.; Cordelier, P.; Martins, F.; Maoret, J.-J.; Bongard, V.; Ferrieres, J.; Roncalli, J.; Elbaz, M.; Vindis, C. Circulating miR-155, miR-145 and let-7c as diagnostic biomarkers of the coronary artery disease. Sci. Rep. 2017, 7, srep42916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Gacoń, J.; Badacz, R.; Stępień, E.; Karch, I.; Enguita, F.J.; Żmudka, K.; Przewłocki, T.; Kabłak-Ziembicka, A. Diagnostic and prognostic micro-RNAs in ischaemic stroke due to carotid artery stenosis and in acute coronary syndrome: A four-year prospective study. Kardiol. Pol. 2018, 76, 362–369. [Google Scholar] [CrossRef] [Green Version]
  101. D’Alessandra, Y.; Devanna, P.; Limana, F.; Straino, S.; Di Carlo, A.; Brambilla, P.G.; Rubino, M.; Carena, M.C.; Spazzafumo, L.; De Simone, M.; et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur. Heart J. 2010, 31, 2765–2773. [Google Scholar] [CrossRef] [Green Version]
  102. Ai, J.; Zhang, R.; Li, Y.; Pu, J.; Lu, Y.; Jiao, J.; Li, K.; Yu, B.; Li, Z.; Wang, R.; et al. Circulating microRNA-1 as a potential novel biomarker for acute myocardial infarction. Biochem. Biophys. Res. Commun. 2010, 391, 73–77. [Google Scholar] [CrossRef]
  103. Long, G.; Wang, F.; Duan, Q.; Chen, F.; Yang, S.; Gong, W.; Wang, Y.; Chen, C.; Wang, D.W. Human Circulating MicroRNA-1 and MicroRNA-126 as Potential Novel Indicators for Acute Myocardial Infarction. Int. J. Biol. Sci. 2012, 8, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Kazimierczyk, E.; Eljaszewicz, A.; Kazimierczyk, R.; Tynecka, M.; Zembko, P.; Tarasiuk, E.; Kaminski, K.; Sobkowicz, B.; Moniuszko, M.; Tycinska, A. Altered microRNA dynamics in acute coronary syndrome. Adv. Interv. Cardiol. 2020, 16, 287–293. [Google Scholar] [CrossRef] [PubMed]
  105. Wang, G.; Zhu, J.-Q.; Zhang, J.-T.; Li, Q.; Li, Y.; He, J.; Qin, Y.-W.; Jing, Q. Circulating microRNA: A novel potential biomarker for early diagnosis of acute myocardial infarction in humans. Eur. Heart J. 2010, 31, 659–666. [Google Scholar] [CrossRef] [PubMed]
  106. Widera, C.; Gupta, S.K.; Lorenzen, J.M.; Bang, C.; Bauersachs, J.; Bethmann, K.; Kempf, T.; Wollert, K.C.; Thum, T. Diagnostic and prognostic impact of six circulating microRNAs in acute coronary syndrome. J. Mol. Cell Cardiol. 2011, 51, 872–875. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, L.; Chen, X.; Su, T.; Li, H.; Huang, Q.; Wu, D.; Yang, C.; Han, Z. Circulating miR-499 are novel and sensitive biomarker of acute myocardial infarction. J. Thorac. Dis. 2015, 7, 303–308. [Google Scholar] [CrossRef] [Green Version]
  108. He, Y.; Zhong, J.; Huang, S.; Shui, X.; Kong, D.; Chen, C.; Lei, W. Elevated circulating miR-126-3p expression in patients with acute myocardial infarction: Its diagnostic value. Int. J. Clin. Exp. Pathol. 2017, 10, 11051–11056. [Google Scholar]
  109. Gidlöf, O.; Andersson, P.; van der Pals, J.; Götberg, M.; Erlinge, D. Cardiospecific microRNA Plasma Levels Correlate with Troponin and Cardiac Function in Patients with ST Elevation Myocardial Infarction, Are Selectively Dependent on Renal Elimination, and Can Be Detected in Urine Samples. Cardiology 2011, 118, 217–226. [Google Scholar] [CrossRef] [Green Version]
  110. Su, T.; Shao, X.; Zhang, X.; Yang, C.; Shao, X. Value of circulating miRNA-1 detected within 3 h after the onset of acute chest pain in the diagnosis and prognosis of acute myocardial infarction. Int. J. Cardiol. 2019, 307, 146–151. [Google Scholar] [CrossRef]
  111. Xue, S.; Liu, D.; Zhu, W.; Su, Z.; Zhang, L.; Zhou, C.; Li, P. Circulating MiR-17-5p, MiR-126-5p and MiR-145-3p Are Novel Biomarkers for Diagnosis of Acute Myocardial Infarction. Front. Physiol. 2019, 10, 123. [Google Scholar] [CrossRef] [Green Version]
  112. Horváth, M.; Horváthová, V.; Hájek, P.; Štěchovský, C.; Honěk, J.; Šenolt, L.; Veselka, J. MicroRNA-331 and microRNA-151-3p as biomarkers in patients with ST-segment elevation myocardial infarction. Sci. Rep. 2020, 10, 5845. [Google Scholar] [CrossRef] [Green Version]
  113. Meng, L.; Yu, X.; Han, H.; Jia, X.; Hu, B.; Zhang, L.; Wang, Z.; Zhang, W.; Zhong, M.; Zhu, H. Circulating miR-143 and miR-145 as promising biomarkers for evaluating severity of coronary artery stenosis in patients with acute coronary syndrome. Clin. Biochem. 2022, Online ahead of print. [Google Scholar] [CrossRef]
  114. Dégano, I.R.; Camps-Vilaró, A.; Subirana, I.; García-Mateo, N.; Cidad, P.; Muñoz-Aguayo, D.; Puigdecanet, E.; Nonell, L.; Vila, J.; Crepaldi, F.M.; et al. Association of Circulating microRNAs with Coronary Artery Disease and Usefulness for Reclassification of Healthy Individuals: The REGICOR Study. J. Clin. Med. 2020, 9, 1402. [Google Scholar] [CrossRef] [PubMed]
  115. Bukauskas, T.; Mickus, R.; Cereskevicius, D.; Macas, A. Value of Serum miR-23a, miR-30d, and miR-146a Biomarkers in ST-Elevation Myocardial Infarction. J. Pharmacol. Exp. Ther. 2019, 25, 3925–3932. [Google Scholar] [CrossRef] [PubMed]
  116. Zhang, Y.; Liu, Y.J.; Liu, T.; Zhang, H.; Yang, S.J. Plasma microRNA-21 is a potential diagnostic biomarker of acute myocardial infarction. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 323–329. [Google Scholar] [PubMed]
  117. Kayvanpour, E.; Gi, W.-T.; Sedaghat-Hamedani, F.; Lehmann, D.H.; Frese, K.S.; Haas, J.; Tappu, R.; Samani, O.S.; Nietsch, R.; Kahraman, M.; et al. microRNA neural networks improve diagnosis of acute coronary syndrome (ACS). J. Mol. Cell Cardiol. 2020, 151, 155–162. [Google Scholar] [CrossRef]
  118. Esa, J.A.W.N. Circulating Cell and Plasma microRNA Profiles Differ between Non-STSegment and ST-Segment-Elevation Myocardial Infarction. Fam. Med. Med. Sci. Res. 2013, 2, 108. [Google Scholar] [CrossRef] [Green Version]
  119. Biener, M.; Giannitsis, E.; Thum, T.; Bär, C.; Costa, A.; Andrzejewski, T.; Stoyanov, K.M.; Vafaie, M.; Meder, B.; A Katus, H.; et al. Diagnostic value of circulating microRNAs compared to high-sensitivity troponin T for the detection of non-ST-segment elevation myocardial infarction. Eur. Heart J. Acute Cardiovasc. Care 2021, 10, 653–660. [Google Scholar] [CrossRef] [PubMed]
  120. Liu, G.; Niu, X.; Meng, X.; Zhang, Z. Sensitive miRNA markers for the detection and management of NSTEMI acute myocardial infarction patients. J. Thorac. Dis. 2018, 10, 3206–3215. [Google Scholar] [CrossRef]
  121. Zhelankin, A.; Stonogina, D.; Vasiliev, S.; Babalyan, K.; Sharova, E.; Doludin, Y.; Shchekochikhin, D.; Generozov, E.; Akselrod, A. Circulating Extracellular miRNA Analysis in Patients with Stable CAD and Acute Coronary Syndromes. Biomolecules 2021, 11, 962. [Google Scholar] [CrossRef]
  122. Gacoń, J.; Kabłak-Ziembicka, A.; Stępień, E.; Enguita, F.J.; Karch, I.; Derlaga, B.; Żmudka, K.; Przewłocki, T. Decision-making microRNAs (miR-124, -133a/b, -34a and -134) in patients with occluded target vessel in acute coronary syndrome. Kardiol. Pol. 2016, 74, 280–288. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, Y.-Y.; Zhou, X.; Ji, W.-J.; Shi, R.; Lu, R.-Y.; Li, J.-L.; Yang, G.-H.; Luo, T.; Zhang, J.-Q.; Zhao, J.-H.; et al. Decreased circulating microRNA-223 level predicts high on-treatment platelet reactivity in patients with troponin-negative non-ST elevation acute coronary syndrome. J. Thromb. Thrombolysis 2013, 38, 65–72. [Google Scholar] [CrossRef]
  124. Wang, Q.; Ma, J.; Jiang, Z.; Wu, F.; Ping, J.; Ming, L. Identification of microRNAs as diagnostic biomarkers for acute myocardial infarction in Asian populations. Medicine 2017, 96, e7173. [Google Scholar] [CrossRef] [PubMed]
  125. Wang, X.; Dong, Y.; Fang, T.; Wang, X.; Chen, L.; Zheng, C.; Kang, Y.; Jiang, L.; You, X.; Gai, S.; et al. Circulating MicroRNA-423-3p Improves the Prediction of Coronary Artery Disease in a General Population―Six-Year Follow-up Results From the China-Cardiovascular Disease Study. Circ. J. 2020, 84, 1155–1162. [Google Scholar] [CrossRef] [PubMed]
  126. Zhu, X.; Lu, X. MiR-423-5p inhibition alleviates cardiomyocyte apoptosis and mitochondrial dysfunction caused by hypoxia/reoxygenation through activation of the wnt/β-catenin signaling pathway via targeting MYBL2. J. Cell Physiol. 2019, 234, 22034–22043. [Google Scholar] [CrossRef] [PubMed]
  127. Hromadka, M.; Motovska, Z.; Hlinomaz, O.; Kala, P.; Tousek, F.; Jarkovsky, J.; Beranova, M.; Jansky, P.; Svoboda, M.; Krepelkova, I.; 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, 508. [Google Scholar] [CrossRef]
  128. Scărlătescu, A.I.; Barbălată, T.; Sima, A.V.; Stancu, C.; Niculescu, L.; Micheu, M.M. miR-146a-5p, miR-223-3p and miR-142-3p as Potential Predictors of Major Adverse Cardiac Events in Young Patients with Acute ST Elevation Myocardial Infarction—Added Value over Left Ventricular Myocardial Work Indices. Diagnostics 2022, 12, 1946. [Google Scholar] [CrossRef]
  129. Schulte, C.; Molz, S.; Appelbaum, S.; Karakas, M.; Ojeda, F.; Lau, D.M.; Hartmann, T.; Lackner, K.J.; Westermann, D.; Schnabel, R.B.; et al. miRNA-197 and miRNA-223 Predict Cardiovascular Death in a Cohort of Pa-tients with Symptomatic Coronary Artery Disease. PLoS ONE 2015, 10, e0145930. [Google Scholar] [CrossRef] [Green Version]
  130. Karakas, M.; Schulte, C.; Appelbaum, S.; Ojeda, F.; Lackner, K.J.; Münzel, T.; Schnabel, R.B.; Blankenberg, S.; Zeller, T. Circulating microRNAs strongly predict cardiovascular death in patients with coronary artery disease—Results from the large AtheroGene study. Eur. Heart J. 2016, 38, 516–523. [Google Scholar] [CrossRef] [Green Version]
  131. Ziaee, S.; Hosseindokht, M.; Cheraghi, S.; Pourgholi, L.; Ahmadi, A.; Sadeghian, S.; Abbasi, S.H.; Davarpasand, T.; Boroumand, M. Predictive Inflammation-related microRNAs for Cardiovascular Events Following Early-Onset Coronary Artery Disease. Arch. Med. Res. 2020, 52, 69–75. [Google Scholar] [CrossRef]
  132. Xiao, S.; Xue, T.; Pan, Q.; Hu, Y.; Wu, Q.; Liu, Q.; Wang, X.; Liu, A.; Liu, J.; Zhu, H.; et al. MicroRNA-146a Serves as a Biomarker for Adverse Prognosis of ST-Segment Elevation Myocardial Infarction. Cardiovasc. Ther. 2021, 2021, 2923441. [Google Scholar] [CrossRef]
  133. Alavi-Moghaddam, M.; Chehrazi, M.; Alipoor, S.D.; Mohammadi, M.; Baratloo, A.; Mahjoub, M.P.; Movasaghi, M.; Garssen, J.; Adcock, I.M.; Mortaz, E. A Preliminary Study of microRNA-208b after Acute Myocardial Infarction: Impact on 6-Month Survival. Dis. Markers 2018, 2018, 2410451. [Google Scholar] [CrossRef] [Green Version]
  134. Badacz, R.; Kleczyński, P.; Legutko, J.; Żmudka, K.; Gacoń, J.; Przewłocki, T.; Kabłak-Ziembicka, A. Expression of miR-1-3p, miR-16-5p and miR-122-5p as Possible Risk Factors of Secondary Cardiovascular Events. Biomedicines 2021, 9, 1055. [Google Scholar] [CrossRef] [PubMed]
  135. Badacz, R.; Przewłocki, T.; Pieniążek, P.; Rosławiecka, A.; Kleczyński, P.; Legutko, J.; Żmudka, K.; Kabłak-Ziembicka, A. MicroRNA-134-5p and the Extent of Arterial Occlusive Disease Are Associated with Risk of Future Adverse Cardiac and Cerebral Events in Diabetic Patients Undergoing Carotid Artery Stenting for Symptomatic Carotid Artery Disease. Molecules 2022, 27, 2472. [Google Scholar] [CrossRef] [PubMed]
  136. Ma, Q.; Ma, Y.; Wang, X.; Li, S.; Yu, T.; Duan, W.; Wu, J.; Wen, Z.; Jiao, Y.; Sun, Z.; et al. Circulating miR-1 as a potential predictor of left ventricular remodeling following acute ST-segment myocardial infarction using cardiac magnetic reso-nance. Quant Imaging Med. Surg. 2020, 10, 1490–1503. [Google Scholar] [CrossRef] [PubMed]
  137. Grabmaier, U.; Clauss, S.; Gross, L.; Klier, I.; Franz, W.; Steinbeck, G.; Wakili, R.; Theiss, H.; Brenner, C. Diagnostic and prognostic value of miR-1 and miR-29b on adverse ventricular remodeling after acute myocardial infarction—The SITAGRAMI-miR analysis. Int. J. Cardiol. 2017, 244, 30–36. [Google Scholar] [CrossRef]
  138. Maciejak, A.; Kostarska-Srokosz, E.; Gierlak, W.; Dluzniewski, M.; Kuch, M.; Marchel, M.; Opolski, G.; Kiliszek, M.; Matlak, K.; Dobrzycki, S.; et al. Circulating miR-30a-5p as a prognostic biomarker of left ventricular dysfunction after acute myocardial infarction. Sci. Rep. 2018, 8, 9883. [Google Scholar] [CrossRef] [Green Version]
  139. Liu, X.; Dong, Y.; Chen, S.; Zhang, G.; Zhang, M.; Gong, Y.; Li, X. Circulating MicroRNA-146a and MicroRNA-21 Predict Left Ventricular Remodeling after ST-Elevation Myocardial Infarction. Cardiology 2015, 132, 233–241. [Google Scholar] [CrossRef]
  140. Lv, P.; Zhou, M.; He, J.; Meng, W.; Ma, X.; Dong, S.; Meng, X.; Zhao, X.; Wang, X.; He, F. Circulating miR-208b and miR-34a Are Associated with Left Ventricular Remodeling after Acute Myocardial Infarction. Int. J. Mol. Sci. 2014, 15, 5774–5788. [Google Scholar] [CrossRef]
  141. Lin, X.; Zhang, S.; Huo, Z. Serum Circulating miR-150 is a Predictor of Post-Acute Myocardial Infarction Heart Failure. Int. Heart J. 2019, 60, 280–286. [Google Scholar] [CrossRef] [Green Version]
  142. Zile, M.R.; Mehurg, S.M.; Arroyo, J.E.; Stroud, R.E.; DeSantis, S.M.; Spinale, F.G. Relationship between the temporal pro-file of plasma microRNA and left ventricular remodeling in patients after myocardial infarction. Circ. Cardiovasc. Genet. 2011, 4, 614–619. [Google Scholar] [CrossRef] [Green Version]
  143. Pinchi, E.; Frati, P.; Aromatario, M.; Cipolloni, L.; Fabbri, M.; La Russa, R.; Maiese, A.; Neri, M.; Santurro, A.; Scopetti, M.; et al. miR-1, miR-499 and miR-208 are sensitive markers to diagnose sudden death due to early acute myocardial infarction. J. Cell. Mol. Med. 2019, 23, 6005–6016. [Google Scholar] [CrossRef] [Green Version]
  144. Bostjancic, E.; Zidar, N.; Stajner, D.; Glavac, D. MicroRNA miR-1 is up-regulated in remote myocardium in patients with myocardial infarction. Folia Biol. 2010, 56, 27–31. [Google Scholar]
  145. Boštjančič, E.; Brandner, T.; Zidar, N.; Glavač, D.; Štajer, D. Down-regulation of miR-133a/b in patients with myocardial infarction correlates with the presence of ventricular fibrillation. Biomed. Pharmacother. 2018, 99, 65–71. [Google Scholar] [CrossRef] [PubMed]
  146. Jayawardena, E.; Medzikovic, L.; Ruffenach, G.; Eghbali, M. Role of miRNA-1 and miRNA-21 in Acute Myocardial Is-chemia-Reperfusion Injury and Their Potential as Therapeutic Strategy. Int. J. Mol. Sci. 2022, 23, 1512. [Google Scholar] [CrossRef] [PubMed]
  147. Naito, H.; Nojima, T.; Fujisaki, N.; Tsukahara, K.; Yamamoto, H.; Yamada, T.; Aokage, T.; Yumoto, T.; Osako, T.; Nakao, A. Therapeutic strategies for ischemia reperfusion injury in emergency medicine. Acute Med. Surg. 2020, 7, e501. [Google Scholar] [CrossRef] [PubMed]
  148. Zajdel, W.; Miszalski-Jamka, T.; Zalewski, J.; Legutko, J.; Żmudka, K.; Paszek, E. Cardiac Magnetic Resonance Shows Improved Outcomes in Patients with an ST-Segment Elevation Myocardial Infarction and a High Thrombus Burden Treated with Adjuvant Aspiration Thrombectomy. J. Clin. Med. 2022, 11, 5000. [Google Scholar] [CrossRef] [PubMed]
  149. Bellis, A.; Di Gioia, G.; Mauro, C.; Mancusi, C.; Barbato, E.; Izzo, R.; Trimarco, B.; Morisco, C. Reducing Cardiac Injury during ST-Elevation Myocardial Infarction: A Reasoned Approach to a Multitarget Therapeutic Strategy. J. Clin. Med. 2021, 10, 2968. [Google Scholar] [CrossRef]
  150. Sagris, M.; Theofilis, P.; Antonopoulos, A.S.; Oikonomou, E.; Paschaliori, C.; Galiatsatos, N.; Tsioufis, K.; Tousoulis, D. In-flammation in Coronary Microvascular Dysfunction. Int. J. Mol. Sci. 2021, 22, 13471. [Google Scholar] [CrossRef]
  151. Minicucci, M.F.; Azevedo-Gaiolla, P.S.; Polegato, B.; Paiva, S.; Zornoff, L.A.M. Heart failure after myocardial infarction: Clinical implications and treatment. Clin. Cardiol. 2011, 34, 410–414. [Google Scholar] [CrossRef]
  152. Pan, Z.; Sun, X.; Ren, J.; Li, X.; Gao, X.; Lu, C.; Zhang, Y.; Sun, H.; Wang, Y.; Wang, H.; et al. miR-1 exacerbates cardiac ischemia-reperfusion injury in mouse models. PLoS ONE 2012, 7, e50515. [Google Scholar] [CrossRef]
  153. Yin, C.; Wang, X.; Kukreja, R.C. Endogenous microRNAs induced by heat-shock reduce myocardial infarction following ischemia-reperfusion in mice. FEBS Lett. 2008, 582, 4137–4142. [Google Scholar] [CrossRef] [Green Version]
  154. Dehaini, H.; Awada, H.; El-Yazbi, A.; Zouein, F.A.; Issa, K.; Eid, A.A.; Ibrahim, M.; Badran, A.; Baydoun, E.; Pintus, G.; et al. MicroRNAs as Potential Pharmaco-Targets in Ischemia-Reperfusion Injury Compounded by Diabetes. Cells 2019, 8, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Seo, H.-H.; Lee, S.-Y.; Lee, C.Y.; Kim, R.; Kim, P.; Oh, S.; Lee, H.; Lee, M.Y.; Kim, J.; Kim, L.K.; et al. Exogenous miRNA-146a Enhances the Therapeutic Efficacy of Human Mesenchymal Stem Cells by Increasing Vascular Endothelial Growth Factor Secretion in the Ischemia/Reperfusion-Injured Heart. J. Vasc. Res. 2017, 54, 100–108. [Google Scholar] [CrossRef] [PubMed]
  156. Hullinger, T.G.; Montgomery, R.L.; Seto, A.G.; Dickinson, B.A.; Semus, H.M.; Lynch, J.M.; Dalby, C.M.; Robinson, K.; Stack, C.; Latimer, P.A.; et al. Inhibition of miR-15 protects against cardiac ischemic injury. Circ. Res. 2012, 110, 71–81. [Google Scholar] [CrossRef] [PubMed]
  157. Kura, B.; Kalocayova, B.; Devaux, Y.; Bartekova, M. Potential Clinical Implications of miR-1 and miR-21 in Heart Disease and Cardioprotection. Int. J. Mol. Sci. 2020, 21, 700. [Google Scholar] [CrossRef] [PubMed]
  158. Krzywińska, O.; Bracha, M.; Jeanniere, C.; Recchia, E.; Kędziora-Kornatowska, K.; Kozakiewicz, M. Meta-analysis of the potential role of miRNA-21 in cardiovascylar system function monitoring. BioMed Res. Int. 2020, 2020, 4525410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Gao, F.; Kataoka, M.; Liu, N.; Liang, T.; Huang, Z.-P.; Gu, F.; Ding, J.; Liu, J.; Zhang, F.; Ma, Q.; et al. Therapeutic role of miR-19a/19b in cardiac regeneration and protection from myocardial infarction. Nat. Commun. 2019, 10, 1802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Li, K.; Lin, T.; Chen, L.; Wang, N. MicroRNA-93 elevation after myocardial infarction is cardiac protective. Med. Hypotheses 2017, 106, 23–25. [Google Scholar] [CrossRef]
  161. Li, J.; Cai, S.X.; He, Q.; Zhang, H.; Friedberg, D.; Wang, F.; Redington, A.N. Intravenous miR-144 reduces left ventricular remodeling after myocardial infarction. Basic Res. Cardiol. 2018, 113, 36. [Google Scholar] [CrossRef]
  162. Sygitowicz, G.; Maciejak-Jastrzębska, A.; Sitkiewicz, D. MicroRNAs in the development of left ventricular remodeling and postmyocardial infarction heart failure. Pol. Arch. Intern. Med. 2020, 130, 59–65. [Google Scholar] [CrossRef] [Green Version]
  163. Maries, L.; Marian, C.; Sosdean, R.; Goanta, F.; Sirbu, I.O.; Anghel, A. MicroRNAs—The Heart of Post-Myocardial Infarction Remodeling. Diagnostics 2021, 11, 1675. [Google Scholar] [CrossRef]
  164. Bostan, M.-M.; Stătescu, C.; Anghel, L.; Șerban, I.-L.; Cojocaru, E.; Sascău, R. Post-Myocardial Infarction Ventricular Remodeling Biomarkers—The Key Link between Pathophysiology and Clinic. Biomolecules 2020, 10, 1587. [Google Scholar] [CrossRef] [PubMed]
  165. Sheikh, S.A.; Alduraywish, A.; Almaeen, A.; Alruwali, M.; Alruwaili, R.; Alomair, B.M.; Salma, U.; Hedeab, G.M.; Bugti, N.; Abdulhabeeb, I. Therapeutic Value of miRNAs in Coronary Artery Disease. Oxidative Med. Cell Longev. 2021, 2021, 8853748. [Google Scholar] [CrossRef]
  166. Peters, L.J.F.; Biessen, E.A.L.; Hohl, M.; Weber, C.; Van Der Vorst, E.P.C.; Santovito, D. Small Things Matter: Relevance of MicroRNAs in Cardiovascular Disease. Front. Physiol. 2020, 11, 793. [Google Scholar] [CrossRef] [PubMed]
  167. Christopher, A.F.; Kaur, R.P.; Kaur, G.; Kaur, A.; Gupta, V.; Bansal, P. MicroRNA therapeutics: Discovering novel targets and developing specific therapy. Perspect. Clin. Res. 2016, 7, 68–74. [Google Scholar] [CrossRef]
Figure 1. Basic scheme of classic miR biogenesis. The primary miRNA (Pri-miRNA) is produced in the cell nucleus through the transcription of a DNA strand mediated by RNA polymerase II. After transcription, Pri-miRNA is cleaved by the enzymatic complex DROSHA in a micro-RNA precursor (pre-miRNA). Pre-miRNA is exported to the cytoplasm by exportin-5 and cleaved by dicer (RNA degrading enzyme) and produces approximately 22 nucleotide RNA duplexes. A miRNA strand is transferred to the Argonaute complex (AGO), forming an RNA-induced silencing complex (RISC) and guides it to pair with the target messenger RNA (mRNA) through binding the miRNA seed sequence with the miRNA recognition site in the mRNA. miRs are secreted out of cells via exosomes (adapted from Creemers et al., 2012 [9]).
Figure 1. Basic scheme of classic miR biogenesis. The primary miRNA (Pri-miRNA) is produced in the cell nucleus through the transcription of a DNA strand mediated by RNA polymerase II. After transcription, Pri-miRNA is cleaved by the enzymatic complex DROSHA in a micro-RNA precursor (pre-miRNA). Pre-miRNA is exported to the cytoplasm by exportin-5 and cleaved by dicer (RNA degrading enzyme) and produces approximately 22 nucleotide RNA duplexes. A miRNA strand is transferred to the Argonaute complex (AGO), forming an RNA-induced silencing complex (RISC) and guides it to pair with the target messenger RNA (mRNA) through binding the miRNA seed sequence with the miRNA recognition site in the mRNA. miRs are secreted out of cells via exosomes (adapted from Creemers et al., 2012 [9]).
Jcm 11 06849 g001
Figure 2. Selected miRs that have anti- or pro-atherothrombotic impacts on the development of atherosclerotic plaques, acute coronary syndrome, and/or post-myocardial ischemic outcomes. Please note that specific miRs are important at different stages of atherosclerosis. (1) At early stages of atherosclerosis (from fatty streaks undetectable with standard imaging tools up to intima-media complex thickening), miRs enriched in ECs, VSMCs and macrophages play a key role. Anti-atherothrombotic and anti-inflammatory effects are achieved through many signaling pathways, with a major regulatory role in lipids metabolism and inflammation. (2) At advanced stages of atherosclerosis, with development of mature plaques (easily detectable with various imaging tools), there is a struggle between miRs stabilizing plaque (e.g., increasing fibrous cap thickness, lowering inflammation, or lipids uptake), and miRs destabilizing plaque (e.g., thinning fibrous cap, enlarging lipid and necrotic core, or developing angiogenesis within the plaque). (3) At the stage of plaque rupture and thrombosis, there is a huge role for ECs-, platelets- and VSMCs/cardiomyocytes-derived miRs (such as miR-223 promoting ECs apoptosis, or miR-126 promoting angiogenesis). (4) After ACS, cardiovascular outcomes are associated with myocardial injury and cardiac fibrosis, in which cardiomyocytes-, VSMCs-, ECs-, and fibroblasts-derived miRs play a crucial role. Please note that some miRs can have dual roles, or opposing (conflicting) roles, in different cells (such as miR-223 which aggravates myocardial fibrosis but protects against hypoxia-induced apoptosis and oxidative stress). * miRs that may be used as therapeutic agents are noted in bold in clear green, while miRs that possess antagonists to neutralize their negative effects are in clear orange. CVD: cardiovascular death; ECs: endothelial cells; I/R: ischemia/reperfusion; LVR: left ventricular remodeling; VSMC: vascular smooth muscle cells.
Figure 2. Selected miRs that have anti- or pro-atherothrombotic impacts on the development of atherosclerotic plaques, acute coronary syndrome, and/or post-myocardial ischemic outcomes. Please note that specific miRs are important at different stages of atherosclerosis. (1) At early stages of atherosclerosis (from fatty streaks undetectable with standard imaging tools up to intima-media complex thickening), miRs enriched in ECs, VSMCs and macrophages play a key role. Anti-atherothrombotic and anti-inflammatory effects are achieved through many signaling pathways, with a major regulatory role in lipids metabolism and inflammation. (2) At advanced stages of atherosclerosis, with development of mature plaques (easily detectable with various imaging tools), there is a struggle between miRs stabilizing plaque (e.g., increasing fibrous cap thickness, lowering inflammation, or lipids uptake), and miRs destabilizing plaque (e.g., thinning fibrous cap, enlarging lipid and necrotic core, or developing angiogenesis within the plaque). (3) At the stage of plaque rupture and thrombosis, there is a huge role for ECs-, platelets- and VSMCs/cardiomyocytes-derived miRs (such as miR-223 promoting ECs apoptosis, or miR-126 promoting angiogenesis). (4) After ACS, cardiovascular outcomes are associated with myocardial injury and cardiac fibrosis, in which cardiomyocytes-, VSMCs-, ECs-, and fibroblasts-derived miRs play a crucial role. Please note that some miRs can have dual roles, or opposing (conflicting) roles, in different cells (such as miR-223 which aggravates myocardial fibrosis but protects against hypoxia-induced apoptosis and oxidative stress). * miRs that may be used as therapeutic agents are noted in bold in clear green, while miRs that possess antagonists to neutralize their negative effects are in clear orange. CVD: cardiovascular death; ECs: endothelial cells; I/R: ischemia/reperfusion; LVR: left ventricular remodeling; VSMC: vascular smooth muscle cells.
Jcm 11 06849 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kabłak-Ziembicka, A.; Badacz, R.; Przewłocki, T. Clinical Application of Serum microRNAs in Atherosclerotic Coronary Artery Disease. J. Clin. Med. 2022, 11, 6849. https://doi.org/10.3390/jcm11226849

AMA Style

Kabłak-Ziembicka A, Badacz R, Przewłocki T. Clinical Application of Serum microRNAs in Atherosclerotic Coronary Artery Disease. Journal of Clinical Medicine. 2022; 11(22):6849. https://doi.org/10.3390/jcm11226849

Chicago/Turabian Style

Kabłak-Ziembicka, Anna, Rafał Badacz, and Tadeusz Przewłocki. 2022. "Clinical Application of Serum microRNAs in Atherosclerotic Coronary Artery Disease" Journal of Clinical Medicine 11, no. 22: 6849. https://doi.org/10.3390/jcm11226849

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop