Next Article in Journal
Inhibition of JAK1,2 Prevents Fibrotic Remodeling of Pulmonary Vascular Bed and Improves Outcomes in the Rat Model of Chronic Thromboembolic Pulmonary Hypertension
Next Article in Special Issue
Inter-Alpha Inhibitor Proteins Modify the Microvasculature after Exposure to Hypoxia–Ischemia and Hypoxia in Neonatal Rats
Previous Article in Journal
Genomic Diversity and Chromosomal Rearrangements in Neisseria gonorrhoeae and Neisseria meningitidis
Previous Article in Special Issue
Clot Morphology in Acute Ischemic Stroke Decision Making
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

microRNAs Associated with Carotid Plaque Development and Vulnerability: The Clinician’s Perspective

by
Rafał Badacz
1,2,
Tadeusz Przewłocki
2,3,
Jacek Legutko
1,2,
Krzysztof Żmudka
1,2 and
Anna Kabłak-Ziembicka
1,4,*
1
Department of Interventional Cardiology, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
2
Department of Interventional Cardiology, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
3
Department of Cardiac and Vascular Diseases, Institute of Cardiology, Jagiellonian University Medical College, św. Anny 12, 31-007 Kraków, Poland
4
Noninvasive Cardiovascular Laboratory, The John Paul II Hospital, Prądnicka 80, 31-202 Kraków, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(24), 15645; https://doi.org/10.3390/ijms232415645
Submission received: 22 November 2022 / Accepted: 7 December 2022 / Published: 9 December 2022
(This article belongs to the Special Issue Molecular Researches on Ischemic Stroke)

Abstract

:
Ischemic stroke (IS) related to atherosclerosis of large arteries is one of the leading causes of mortality and disability in developed countries. Atherosclerotic internal carotid artery stenosis (ICAS) contributes to 20% of all cerebral ischemia cases. Nowadays, atherosclerosis prevention and treatment measures aim at controlling the atherosclerosis risk factors, or at the interventional (surgical or endovascular) management of mature occlusive lesions. There is a definite lack of the established circulating biomarkers which, once modulated, could prevent development of atherosclerosis, and consequently prevent the carotid-artery-related IS. Recent studies emphasize that microRNA (miRNA) are the emerging particles that could potentially play a pivotal role in this approach. There are some research studies on the association between the expression of small non-coding microRNAs with a carotid plaque development and vulnerability. However, the data remain inconsistent. In addition, all major studies on carotid atherosclerotic plaque were conducted on cell culture or animal models; very few were conducted on humans, whereas the accumulating evidence demonstrates that it cannot be automatically extrapolated to processes in humans. Therefore, this paper aims to review the current knowledge on how miRNA participate in the process of carotid plaque formation and rupture, as well as stroke occurrence. We discuss potential target miRNA that could be used as a prognostic or therapeutic tool.

1. Introduction

Ischemic stroke (IS) is one of the leading causes of mortality and disability in developed countries [1]. Atherosclerotic internal carotid artery stenosis (ICAS) accounts for about 20% cases of cerebral ischemia [2]. The present diagnostic tools for carotid artery assessment are based on imaging studies, including carotid Doppler ultrasonography, computed tomography, magnetic resonance, or conventional invasive angiography with a use of intravascular ultrasound (IVUS), and optical coherence tomography (OCT) [3,4,5]. They display the degree of ICAS, as well as carotid plaque morphology [6].
The current guidelines position carotid endarterectomy (CEA) and carotid artery stenting (CS) as the established treatment methods for ICAS [7]. In addition to invasive treatment, the optimal medical approach, including cardiovascular risk factor-control, as well as pharmacotherapy (i.e., antiplatelet and antidiabetic agents, lipid and blood pressure lowering medication), should be introduced in order to reduce IS risk [8,9]. The optimal timing for the intervention on carotid artery is controversial [10,11]. According to guidelines, CEA or CS is recommended in recently symptomatic ICAS with stenosis severity above 50% lumen reduction [10,11], whereas the intervention on asymptomatic ICAS is recommended in high-grade stenosis, or in carotid plaques exceeding 60% lumen reduction when features of high-risk plaque for cerebral ischemia are present [10]. As IS can result from a fragmented plaque debris release with a subsequent embolization of cerebral arteries, plaque rupture followed by local carotid artery thrombosis, or hypoperfusion of cerebral structures, the mechanism of cerebral ischemia is complex [12,13,14,15]. Thus, as evidenced, plaque morphology and structure, in addition to the degree of carotid stenosis, play the pivotal role in the IS risk assessment and decision on the intervention [16].
The serious drawback of the aforementioned imaging tools is that they do not allow for the assessment of early stages of atherosclerosis, i.e., those that precede intima-media complex thickening and early fatty lesions incidences [17]. Unfortunately, current guidelines miss laboratory biomarkers which could predict the incidence of IS and thus target the high-risk group of patients with preemptive treatment, whereas early intervention upon the initiation of atherosclerosis seems very attractive [18]. Data show the important roles of pro-atherothrombotic and pro-inflammatory biomarkers, including cytokines (IL-1β, IL-6, TNFα), platelets, and macrophages activity [19,20,21].
Recent studies emphasize that microRNA (miRNA) are the emerging particles that could potentially play a pivotal role in this approach [22]. miRNAs are small, non-coding RNA nucleotides, having a length that is typically between 18 and 27 nucleotides that regulate post-transcriptional gene expression, by binding to the 3′- (more often), or to 5′-untranslated regions of mRNA, or exons [23]. The role of the miRNA has already been confirmed in the broad range of both physiological and pathological processes [24]. They are responsible for target gene expression regulation after the transcription process, either by inhibiting the translation or mRNA degradation [25]. The diagnostic and prognostic role of circulating miRNAs in ICAS leading to IS has been studied, however the conclusions remain inconsistent. This paper aims to review the current knowledge and assess the newest studies on how the miRNA participate in the process of carotid plaque formation and rupture, as well as stroke occurrence.

2. From Fatty Streaks and Foam Cells to Mature Plaque

Plaque formation initiates from stages that are not detectable by imaging tools [26]. First stages include endothelium dysfunction, accompanied by inflammation and modified low-density lipoprotein (LDL) retention in the intimal layer of the intima-media complex [27]. In the endothelium equilibrium, a great number of miRNAs are involved, including protective ones [28]. Their protective effect is achieved through many signaling pathways, however their major role is to prevent unfavorable lipid metabolism and reduce inflammation [28]. One of these miRNAs, miR-126, protects endothelial cells (ECs) through the suppression of NOTCH-1 inhibitor and activation of the vascular endothelial growth factor (VEGF) signaling (Table 1) [29,30]. At the beginning, miR-155 induces the downregulation of mitogen-activated protein 3 kinase 10 (MAP3K10), endothelin-1 (ET-1), and angiotensin II (ANG II) type I receptor [31,32]. The downregulation of ET-1 is important in many cardiovascular settings, as elevated levels of ET-1 are independently associated with increased cardiovascular mortality [33,34]. miR-146a and miR-125a decrease the lipid uptake in macrophages [35,36]. miR-146a also inhibits endothelial activation by increasing nitric oxide synthase (eNOS) expression [35]. miR-125 modulates extracellular vascular endothelial growth factor (VEGF) by manipulating macrophage soluble VEGF receptor-1 (sVEGFR1) production. This mechanism has a therapeutic potential in many diseases [36]. miR-206 and miR-223 regulate cholesterol synthesis through the reverse cholesterol transport from macrophages to the liver for excretion, attenuates pro-inflammatory cytokine production, and has a role in platelet activation [37,38,39,40,41].
Some miRs, such as miR-let-7g, also modulate ECs senescence by regulating anti-aging gene sirtuin 1 (SIRT1) and the insulin growth factor (IGF) 1 pathway [42]. In line with this, miR-143 is downregulated with advancing age and protects against vascular senescence [43,44]. On the contrary, miR-92a released from ECs stimulates macrophages to the pro-inflammatory responses and LDL uptake, which enhance atherosclerotic progression [85]. In mice, inhibition of miR-92a reduces endothelial inflammation and atheroma plaque size through the regulation of Kruppel-like factor 2 (KLF2) [47]. Similarly, miR-34a aggravates and accelerates vascular senescence through the downregulation of SIRT1 and AXL Receptor Tyrosine Kinase [48], whereas miR-34a inhibition by anti-miR-34a reduced vascular inflammation, senescence, and apoptosis [49,86]. In macrophages, ox-LDL increases miR-34a levels that target the cholesterol transporters’ ATP-binding cassette subfamily A member 1 (ABCA1), and ATP-binding cassette subfamily G member 1 (ABCG1) [48]. This alters lipid metabolism, while miR-34a enhanced secretion of inflammatory cytokines promotes inflammation facilitating atherosclerotic plaque formation [48,49]. ECs dysregulation is enhanced by the lipid accumulation due to disturbed reverse cholesterol transport of cholesterol efflux from macrophages to the liver [50]. This results in lipid accumulation in macrophages with formation of foam cells [48]. miR-206 and miR-233 promote cholesterol efflux to the liver, whereas miR-33a inhibits reverse transport (Table 1) [39,40,51,87]. miR-33a/b have been shown to act as post-transcriptional regulators of a lipid metabolism, and their pharmacological inhibition diminished atherosclerosis by raising plasma high-density lipoprotein levels [87]. Nguyen et al. demonstrated that chitosan nanoparticles containing miRs can be delivered to macrophages [88]. In mice, macrophages treated with miR-33-loaded nanoparticles showed decreased reverse cholesterol transport [88]. In contrast, when efflux-promoting miRs were delivered the efflux was improved [88].
miR-10a, miR-31, and miR-17-3p regulate inflammation modulating the expression of adhesion molecules in ECs, while miR-155 and miR-331 through the down-regulation of the anti-inflammatory suppressor of cytokine signaling 1 (SOCS1) protein [53,54,73,89,90]. During atherosclerosis development, miR-155 begins to stimulate atherosclerosis progression through repressing Bcl6 in macrophages, suppressing the expression of eNOS, and increasing pro-inflammatory NF-κB signaling [52].
There is a continuous crosstalk between ECs and vascular smooth muscle cells (VSMCs) [91]. ECs-derived miRNAs, like miR-126, miR-92a exert action on VSMCs, resulting in the VSMCs-enriched miRNAs release, that often have reciprocal unfavorable effects on ECs [92,93]. On atherosclerosis initiation, VSCMs migrates from the medial arterial wall into the intimal space, resulting in promotion of plaque formation [91]. VSCMs migration and proliferation is one of key stages in early atherosclerosis. miR-520 regulates VSMCs function by targeting RelA/p65. This way, decreasing cells migration and proliferation exerts a protective role in atherosclerosis [45]. Moreover, miR-520c-3p mimics may act as a promising therapeutic strategy for atherosclerosis [45]. The VSMCs equilibrium plays a great role in the inhibition of atherosclerosis. The maintenance of contractile phenotype prevents atherosclerosis [45,59,60,91,92,93,94]. Of the miRs capable of the contractile function recovery in VSMCs, miR-22 and miR143/145 are probably the most investigated ones, and are potential therapeutic targets [59,60,91,94]. Intravenous delivery of miR-143/145 extracellular vesicles blocked atherosclerotic lesion progression and exerted protective effects on intima-media complex [59,60], while miR-22 restores contractile phenotype of VSMCs without a negative impact on EC’s function [95]. In addition, miR-22 inhibits intima-media hyperplasia, which is important both for inhibition of atherosclerosis plaque growth, as well as in the restenosis following stent implantation [46,95].
Some miRNAs were investigated in the context of carotid plaques, including miR-520, miR-455, and miR-105. Some are common for many arterial territories, including miR-21, miR-27, miR-100, and miR-122 (Table 1) [45,55,56,57,58,61,62,63,64,65,66,67,68,69,70,71,72,74,75,76,77,78,79,80,81,82,83,84,96,97,98,99,100,101,102].
When the anti-atherothrombotic miRNAs are overbalanced by the pro-atherothrombotic miRNAs, we can steadily observe carotid intima-media complex thickening (CIMT), followed by the occurrence of focal non-calcified lesions [102]. Then, a formation of mature plaques composed of lipid and necrotic cores, fibrotic matrix, and calcifications are observed, accompanied by inflammation and angiogenesis. The first caution that should attract the attention of clinicians is CIMT [103,104,105]. CIMT, in ranges above the 75th percentile for age and gender, can even be observed in teenagers and young adults, particularly if accompanied by atherosclerosis risk factors, such as diabetes or familial hypercholesterolemia [106,107]. This parameter is well correlated with risk of cardiovascular events, such as cardiovascular death (CVD), IS, and myocardial infarction (MI) [103,104,105]. It has also a good predictive value for a presence of significant atherosclerosis in the other territories, e.g., coronary arteries [108,109]. Several miRNAs are associated with CIMT, including miR-22, miR-29a, miR-143/145, and miR-92a [46,70,110,111]. With increasing CIMT, atherosclerotic process accelerates. There is a huge role for metalloproteinases (MMP), such as MMP-2 and MMP-9, as they are associated with a promotion of plaque growth and CIMT increase, rather than a decrease in VSMCs contractility [111]. Interestingly, in advanced carotid plaques, migration and proliferation of VSMCs is beneficial, promoting the stability of the fibrous cap and prevention of plaque rupture [91,102]. This process in stimulated by the expression of miR-145 and miR-210 that drive the increase in plaque collagen content and a fibrous cap area, while at the same time reducing the necrotic core area [46,65,95].
In contrast, plaque instability is associated with increasing levels of MMP-2, MMP-9, the increasing size of plaque and the lipid and necrotic core, particularly when abundant in lipids [111,112,113,114]. MMP-9 is particularly important as it predicts future adverse cardiovascular events [71,111,112,113]. It was observed that MMP-9 is regulated by several miRNAs, including miR-92a, which is a predictor of plaque instability [114]. However, miR-92a is not necessarily always negative [115,116]. The upregulation of MMP-9 and the downregulation of TIMP3 in H2O2-induced VSMCs were observed to be reversed by mimicking miR-92a in addition to SIRT1 and siRNA, which may prevent a phenotypic change of VSMCs [115,116]. Other miRNAs associated with serum concentration of MMP-9 include miR-100, miR-155, miR-133a, and miR-223 [111,114]. These miRNAs are also linked to plaque instability and might be used as biomarkers of plaque conversion from a stable state into a vulnerable state [114,115,116,117,118,119].
Thus, it is of the utmost importance to identify carotid plaques that are likely to undergo transformation from the stable ones to vulnerable ones, prone to rupture and cause symptoms of cerebral ischemia. The research is ongoing for the identification of specific microRNAs that could prevent IS through the manipulation of their expression levels.

3. Degree of Carotid Plaque, Patients with Asymptomatic and Symptomatic ICAS: Prognostic miRNAs

One of the key issues addressed in recent studies was the role of miRNAs in differentiating patients with stable asymptomatic ICAS from those who may develop symptomatic ICAS, thus the identification of patients characterized with high risk of ICAS-related IS [114,119]. Alas, the majority of studies on carotid atherosclerosis describe carotid lesions that are non-occlusive, whereas the occlusive carotid artery lesions regarding miRNA expression profile and levels are under-investigated. Notably, ICAS causes at least a 50% stenosis, which increases the risk of carotid lesion-related IS [120]. miRNA as potential markers of carotid plaque that is non-obstructive compared to plaques causing a significant carotid stenosis (equal or greater than 50% lumen reduction) were investigated by Raskurazhew et al. (Table 2) [100]. In this study, the levels of miR-126-3p, miR-126-5p, miR-21-3p, and miR-29a-3p were higher in non-obstructive carotid plaques, meanwhile they were low in carotid plaques which caused lumen reduction of 50% or greater [100]. In contrast, levels of miR-33a were lower in early carotid plaques, while higher in asymptomatic ICAS exceeding 50% [100].
In a study by Zhu et al., low serum levels of miR-455 were highly diagnostic for a high-grade asymptomatic ICAS, with the AUC value of 0.992, a sensitivity of 82.9%, and a specificity of 90.8% [61]. The multivariate regression analysis demonstrated that levels of LDL-cholesterol (OR = 4.380, 95% CI = 1.101–17.419, p = 0.036) and miR-455-5p (OR = 0.154, 95% CI = 0.039–0.606, p = 0.007) were independently associated with the severity of ICAS. Moreover, miR-455 (HR = 0.128, 95% CI = 0.029–0.573, p = 0.007), likewise the degree of ICAS, occurred important risk factors for ICAS-related IS [61]. The Kaplan–Meier survival curves showed that low miR-455-5p expression was related to the incidence of IS during follow-up (log Rank P = 0.002).
In the study by Dolz et al., the authors focused on the progression of asymptomatic ICAS and the potential role of miRNAs as non-invasive biomarkers of carotid plaque instability and ICAS progression [121]. The analysis identified a different miRNA expression profile in RNA samples from patients with ICAS progression during the follow-up period, compared to the patients without ICAS progression. Progression of ICAS was associated with significantly higher expression levels of miR-199b-3p (0.056; IQR, 0.014–0.958 vs. 0.029; IQR, 0.002–1.139; p = 0.049), miR-130a-3p (0.022; IQR, 0.007–0.177 vs. 0.006; IQR, 0.001–0.197; p = 0.042) and miR-24-3p (0.126; IQR 0.025–1.978 vs. 0.053; IQR, 0.007–0.95; p = 0.028) [121]. However, in the multivariate analysis, only the overexpression of a proangiogenic miR-130a-3p was independently associated with the ICAS progression (ORs, 5.4; 95% CI, 1.03–28.2; p = 0.025) and IS risk [121].
In line with this, miR-637 was assessed as a potential biomarker of ICAS and IS occurrence [122]. The prospective analysis revealed that down-regulation of miR-637 was associated with the degree of ICAS, with a sensitivity of 85.6%, and a specificity of 83.3% for ICAS of at least 50% lumen narrowing [122]. During the 5 year follow-up, the Kaplan–Meier analysis showed that the lower miR-637 levels were associated with a higher incidence of ICAS-related IS (HR = 0.073, 95% CI = 0.017–0.313, p < 0.001), along with severe ICAS (HR = 0.144, 95% CI = 0.045–0.463, p = 0.001) [122]. The study proved that the miR-637 downregulation is responsible for cardiovascular events in patients with atherosclerosis [137] by promoting the proliferation and migration of VSMCs by regulating IGF-2 [138].
In another study by Zhu et al., the sensitivity and the specificity of miR-486 for a diagnosis of significant asymptomatic ICAS were 82.4% and 89.7%, respectively [123]. The results showed that HAEC apoptosis was weakened by increased miR-486-5p, reflecting its protective effect against vascular ECs injury [123]. Vascular ECs injury may promote the release of intracellular adhesion factors and then trigger the transformation of macrophages into foam cells. In vitro experimental results demonstrated that miR-486-5p can also inhibit the release of ICAM-1 in HAECs. Besides its regulatory role in the apoptosis of vascular ECs, the findings of the study also demonstrated the influence of miR-486-5p on inflammation and oxidative stress [139].
There is limited data on the miRNAs acting as triggers for symptom development in patients with a significant ICAS. In the study by Grosse et al., the levels of several popular circulating miRNAs (miR-92a, miR-126, miR-143, miR-145, miR-155, miR-210, miR-221, miR-222, and miR-342-3p) were analyzed in patients with symptomatic and asymptomatic ICAS that were referred to CEA [124]. The study has not found miRNAs diagnostic for symptomatic ICAS, except from the trend to difference for miR-92a in multivariate analysis [124]. The authors have not observed either the relationship between miRNAs expression levels and micro embolic signals that correspond to plaque fragility and distal embolization to cerebral arteries by debris released from the plaque [140].
The study by Luque et al. aimed to present the associations between atherosclerotic plaque instability and circulating miR-638 level [125]. The results showed significantly lower miR-638 levels in patients with symptomatic ICAS, as compared to asymptomatic ICAS [125]. The study demonstrated that miR-638 level was correlated with the IS incidence and IS recurrence, smoking habit, bilateral ICAS, coronary artery disease, and hypercholesterolemia [125]. The authors considered miR-638 as a potential marker associated with plaque vulnerability and IS.
In the study by Badacz et al., the differences in expression levels of circulating miR-133a, miR-124, and miR-134 were found in patients with symptomatic vs. asymptomatic ICAS [83]. The authors observed higher serum levels of miR-1 (p = 0.032) in a thrombotic plaque, as well as higher levels of miR-1 (p = 0.04) and miR-16-5p (p = 0.003) in patients with ulcerated plaques [83].
Similarly, in the study by Zhang et al., the expression levels of miR-106b-5p were analyzed in asymptomatic ICAS patients [126]. The expression of miR-106b-5p was associated with hypertension, dyslipidemia, and degree of ICAS. In ROC analysis, miR-106b-5p showed a good predictive value for significant ICAS compared to healthy individuals, with a sensitivity of 89.7% and a specificity of 83.6% [126]. Furthermore, patients with higher miR-106b-5p expression levels were more likely to suffer from IS during follow-up (log rank p = 0.020; HR 5.431, 95% CI: 1.592–18.520, p = 0.007).
The other study by Zhang et al. focused on miR-637 and its possible role as a biomarker for IS risk [122]. The multivariate analysis revealed that downregulation of miR-637 independently predicted future IS (HR = 0.073, 95%CI = 0.017–0.313, p < 0.001), thus making the analyzed miRs possible biomarkers in ICAS patients [122].
In line with this, Li et al. analyzed expression levels of miR-483-5p in patients with asymptomatic ICAS and evaluated its diagnostic and predictive value for IS [127]. The results revealed that patients with high miR-483-5p levels had higher a risk of IS (log-rank p = 0.011), as compared to patients with lower miR-483-5p expression levels (HR 2.670; 95% CI 1.099–6.484; p = 0.030) [127]. Similarly, miR-342-5p was proposed as an IS risk predictor at a 5 year follow-up by Zhou et al. [128].
Several more studies found possible predictive value of specific miRNAs for the onset of IS. Chen at al. stated that elevated miR-92a expression could differentiate patients with asymptomatic ICAS from healthy subjects, and was an independent predictive factor for the occurrence of IS (HR = 2.971, 95% CI = 1.230–7.173, p = 0.015) [129], while Zhou et al. observed miR-342-5p as a potential predictor of IS in asymptomatic patients with ICAS (HR = 5.512, 95% CI = 1.370–22.176, p = 0.016) [129].
Similar results were observed for miR-19a-3p by Liu et al. who stated that higher expression of serum miR-19a-3p (HR = 8.507, 95% CI = 2.239–32.328, p = 0.002) and degree of ICAS (HR = 3.695, 95% CI = 1.127–12.109, p = 0.031) were independent predictors for the onset of IS [130], whereas Lu et al. found miR-27b an important risk factor of IS (HR = 5.067, 95% CI = 1.170–21.943, p = 0 .030) [131].
Conversely, Li et al. observed that a downregulation of miR-206 was an independent risk factor for the development of IS during a 5 year follow-up (HR = 0.046, 95% CI = 0.005–0.431, p = 0.007) [132]. In the study by Liu et al., low miR-9-5p expression levels (HR = 0.239; 95% CI = 0.087–0.652, p = 0.005) were associated with IS [133].
Finally, the recent study by Badacz et al. analyzed cardiovascular outcomes in patients with diabetes who underwent intervention on ICAS [134]. During a follow-up period of 7 years, miR-134-5p occurred an independent risk factor of CVD/MI/recurrent IS (HR = 1.12; 95% CI = 1.05–1.21, p < 0.001) in diabetic patients, while miR-16 in patients without diabetes [134]. The authors suggested the diversity of miRNAs profile in patients with and without diabetes for cardiovascular outcomes. In line with this, Sardu et al. found miR-24 as a possible predictor of ischemic cardiovascular events and hospitalization for heart failure in pre-diabetic patients at a 2 year follow-up [141].
In addition to miRNA, there is probably a huge role for microparticles in vascular damage in diabetic patients [142]. Some of these macromolecules have angiogenetic properties or participate in modulation of vascular senescence or remodeling, taking part in the process of vascular aging [142]. The other miRNAs that could be potentially associated with plaque instability and cerebrovascular outcomes are: miR-21, miR-221, miR-133b, and miR-124 were independent predictors (Table 2) [135,136].

4. microRNA Expression Levels in Plaques Removed during CEA

Apart from circulating serum or plasma miRNAs, the carotid plaque can contain large amounts of miRNAs. These miRNAs often differ or have diverse expression levels in plaque, compared to systemic miRNAs [97]. Intra-plaque miRNAs play the role in the process of carotid plaque evolution and progression, leading to plaque rupture and IS, as well [97]. These miRNAs can be explored only when a biological material is available, e.g., after the excision of carotid plaque that takes place during CEA.
The study by Cipollone et al. covered the large-scale miRNA analysis of symptomatic and asymptomatic carotid plaques excised during CEA (Table 3) [97]. The results presented significant up-regulation of several miRNAs (out of 41 analyzed), miR-145 and miR-133a in particular, in symptomatic plaques as compared to asymptomatic ones [97].
In the study by Maitrias et al., seven miRNAs were dysregulated, including miR-100, miR-125a, miR-127, miR-133a, miR-145, and miR-221 in carotid plaques obtained during CEA in 30 symptomatic and asymptomatic patients [22]. The identified miRNA profile was corresponding to the inflammatory process leading to plaque destabilization. miR-100, miR-125a, and miR-127 were proven to modulate the vascular inflammation and angiogenesis, as well as oxidative stress [125,146,147,148]. Finally, miR-133a, and miR-145 are considered to play the key role in regulating the vascular remodeling and inflammation especially by controlling VSMCs proliferation and differentiation [149,150]. In the study by Bazan et al., the miR-221 and miR-222 expressions were evaluated in carotid plaques obtained from CEA in three groups of patients: with asymptomatic ICAS, symptomatic ICAS (neurologic symptoms within 5 days), and patients who underwent neurologic event earlier [143]. Patients in acute phases of neurologic symptoms related to ICAS exhibited a significant decrease in miR-221 and miR-222 expression as compared to the other groups [143,150]. This confirms the direct association of miRNAs dysregulation in the mechanisms of plaque instability [143].
Another promising potential biomarker of plaque instability in carotid atherosclerosis is miR-200c [144]. The miR-200c plaque concentration was up-regulated in carotid plaques that presented unstable features on imaging studies. Additionally, miR-200c positively correlated with instability biomarkers, such as COX2, IL-6 or MMP-9 [144]. miR-146a and pro-inflammatory cytokines in patients with severe ICAS were addressed in the study by Huang et al. [151]. The expression levels of miR-146a, IL-6, and TNF-α in the ICAS group were higher than those in the control group and positively correlated with the degree of ICAS and plaque vulnerability [151]. In the study by Wei et al., upregulation of miR-330-5p level was identified in symptomatic carotid plaques [145].
Thus, the complex process of plaque destabilization is attributed to both intra-plaque action of miRNAs and is simultaneously regulated by various circulating miRNAs.

5. Conclusions

MicroRNA have become a point of interest in recent years of scientific research including broad range of pathophysiological issues. Multiple recent studies proved miRNAs to be the important factor of atherosclerotic process, including coronary, peripheral, or carotid artery disease. miRNA seem to be directly associated with a patient’s atherosclerosis severity and functional prognosis, suggesting that miRNAs can be used as potential biomarkers for the diagnosis and prognosis evaluation of cardiovascular events. According to modern evidence, some miRNAs reveal a potential diagnostic or prognostic value in revealing the potential vulnerable plaque features or predicting future ischemic events.
Several miRNAs were proven to be a viable diagnostic tool in assessing the risk of carotid plaque destabilization, and as a result of plaque rupture, the risk of IS occurrence is high. Moreover, the studies have shown that different miRNAs were able to differentiate the symptomatic and asymptomatic course of ICAS.
However, the studies struggle with a wide range of miRNAs, missing identification of the narrow group of pivotal and key miRNAs responsible for the atherosclerosis processes, which may play the role of biomarkers in everyday routine practice. Therefore, dedicated study is necessary in order to establish which miRNAs could be used in carotid atherosclerosis.

Author Contributions

Conceptualization, T.P., R.B. and A.K.-Z.; Data curation, R.B., T.P., K.Ż., J.L. and A.K.-Z.; Formal analysis; T.P. and A.K.-Z.; Funding acquisition, A.K.-Z.; Investigation, R.B. and A.K.-Z., Methodology, R.B., A.K.-Z., K.Ż. and J.L.; Project administration, T.P.; Resources, A.K.-Z., R.B. and T.P.; Supervision, A.K.-Z. and T.P.; Validation, K.Ż. and J.L.; Visualization, R.B., K.Ż. and J.L.; Writing—Original draft, R.B., K.Ż., J.L., A.K.-Z. and T.P.; Writing, R.B.,T.P. and 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, to Anna Kabłak-Ziembicka (grant no N41/DBS/000752).

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to privacy.

Conflicts of Interest

The authors declare that there are no conflict of interest regarding the publication of this article.

References

  1. Petty, G.W.; Brown, R.D.; Whisnant, J.P.; Sicks, J.D.; O’Fallon, W.M.; Wiebers, D.O. Ischemic stroke subtypes: A population-based study of incidence and risk factors. Stroke 1999, 30, 2513–2516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Wityk, R.J.; Lehman, D.; Klag, M.; Coresh, J.; Ahn, H.; Litt, B. Race and sex differences in the distribution of cerebral atherosclerosis. Stroke 1996, 27, 1974–1980. [Google Scholar] [CrossRef] [PubMed]
  3. Musiałek, P.; Pieniążek, P.; Tracz, W.; Tekieli, L.; Przewłocki, T.; Kabłak-Ziembicka, A.; Motyl, R.; Moczulski, Z.; Step-Niewski, 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] [PubMed] [Green Version]
  4. Pierro, L.; Arrigo, A.; De Crescenzo, M.; Aragona, E.; Chiesa, R.; Castellano, R.; Catenaccio, B.; Bandello, F. Quantitative Optical Coherence Tomography Angiography Detects Retinal Perfusion Changes in Carotid Artery Stenosis. Front. Neurosci. 2021, 15, 640666. [Google Scholar] [CrossRef]
  5. Vista, F.P., 4th; Ngo, M.T.; Cho, S.B.; Kwak, H.S.; Chong, K.T. Carotid Artery Plaque Identification and Display System (MRI-CAPIDS) Using Opensource Tools. Diagnostics 2020, 10, 1111. [Google Scholar] [CrossRef]
  6. North American Symptomatic Carotid Endarterectomy Trial (NASCET) Steering Committee. North American symptomatic carotid endarterectomy trial: Methods, patient characteristics, and progress. Stroke 1991, 22, 711–720. [Google Scholar] [CrossRef] [Green Version]
  7. Naylor, A.R.; Rothwell, P.M.; Bell, P.R. Overview of the principal results and secondary analyses from the European and North American randomised trials of endarterectomy for symptomatic carotid stenosis. Eur. J. Vasc. Endovasc. Surg. 2003, 26, 115–129. [Google Scholar] [CrossRef] [Green Version]
  8. Kadoglou, N.P.; Khattab, E.; Velidakis, N.; Patsourakos, N.; Lambadiari, V. A new approach of statin therapy in carotid atherosclerosis: Targeting indices of plaque vulnerability on the top of lipid-lowering. A narrative review. Kardiol. Pol. 2022, 80, 880–890. [Google Scholar] [CrossRef]
  9. Parish, S.; Arnold, M.; Clarke, R.; Du, H.; Wan, E.; Kurmi, O.; Chen, Y.; Guo, Y.; Bian, Z.; Collins, R.; et al. Assessment of the Role of Carotid Atherosclerosis in the Association between Major Cardiovascular Risk Factors and Ischemic Stroke Subtypes. JAMA Netw. Open 2019, 2, e194873. [Google Scholar] [CrossRef]
  10. European Stroke Organisation; Tendera, M.; Aboyans, V.; Bartelink, M.L.; Baumgartner, I.; Clément, D.; Collet, J.P.; Cremonesi, A.; De Carlo, M.; Erbel, R.; et al. ESC Guidelines on the diagnosis and treatment of peripheral artery diseases. Document covering atherosclerotic disease of extracranial carotid and vertebral, mesenteric, renal, upper and lower extremity arteries: The Task Force on the Diagnosis and Treatment of Peripheral Artery Diseases of the European Society of Cardiology (ESC). Eur. Heart J. 2011, 32, 2851–2906. [Google Scholar]
  11. Bonati, L.H.; Kakkos, S.; Berkefeld, J.; de Borst, G.J.; Bulbulia, R.; Halliday, A.; van Herzeele, I.; Koncar, I.; McCabe, D.J.; Lal, A.; et al. European Stroke Organisation guideline on endarterectomy and stenting for carotid artery stenosis. Eur. Stroke J. 2021, 6, I-XLVII. [Google Scholar] [CrossRef] [PubMed]
  12. Badacz, R.; Przewłocki, T.; Karch, I.; Pieniążek, P.; Rosławiecka, A.; Mleczko, S.; Brzychczy, A.; Trystuła, M.; Żmudka, K.; Kabłak-Ziembicka, A. Low prevalence of collateral cerebral circulation in the circle of Willis in patients with severe carotid artery stenosis and recent ischemic stroke. Postep. Kardiol. Inter. 2015, 11, 312–317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Geiger, M.A.; Flumignan, R.L.G.; Sobreira, M.L.; Avelar, W.M.; Fingerhut, C.; Stein, S.; Guillaumon, A.T. Carotid Plaque Composition and the Importance of Non-Invasive in Imaging Stroke Prevention. Front. Cardiovasc. Med. 2022, 9, 885483. [Google Scholar] [CrossRef]
  14. Brinjikji, W.; Rabinstein, A.A.; Lanzino, G.; Murad, M.H.; Williamson, E.E.; DeMarco, J.K.; Huston, J., 3rd. Ultrasound Characteristics of Symptomatic Carotid Plaques: A Systematic Review and Meta-Analysis. Cereb. Dis. 2015, 40, 165–174. [Google Scholar] [CrossRef]
  15. Nogueira, R.C.; Beishon, L.; Bor-Seng-Shu, E.; Panerai, R.B.; Robinson, T.G. Cerebral Autoregulation in Ischemic Stroke: From Pathophysiology to Clinical Concepts. Brain Sci. 2021, 11, 511. [Google Scholar] [CrossRef]
  16. 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 Cardi-ology (ESC) and of the European Society for Vascular Surgery (ESVS). Eur. Heart J. 2018, 39, 763–816. [Google Scholar]
  17. Tarkin, J.M.; Dweck, M.R.; Evans, N.R.; Takx, R.A.; Brown, A.J.; Tawakol, A.; Fayad, Z.A.; Rudd, J.H. Imaging Atherosclerosis. Circ. Res. 2016, 118, 750–769. [Google Scholar] [CrossRef] [Green Version]
  18. Formanowicz, D.; Krawczyk, J.B.; Perek, B.; Lipski, D.; Tykarski, A. Management of high-risk atherosclerotic patients by statins may be supported by logistic model of intima-media thickening. J. Clin. Med. 2021, 10, 2876. [Google Scholar] [CrossRef]
  19. Kabłak-Ziembicka, A.; Przewłocki, T.; Sokołowski, A.; Tracz, W.; Podolec, P. Carotid intima-media thickness, hs-CRP and TNF-α are independently associated with cardiovascular event risk in patients with atherosclerotic occlusive disease. Atherosclerosis 2011, 214, 185–190. [Google Scholar] [CrossRef]
  20. Kondakov, A.; Lelyuk, V. Clinical Molecular Imaging for Atherosclerotic Plaque. J. Imaging 2021, 7, 211. [Google Scholar] [CrossRef]
  21. Ząbczyk, M.; Natorska, J.; Undas, A. Fibrin clot properties in atherosclerotic vascular disease: From pathophysiology to clinical outcomes. J. Clin. Med. 2021, 10, 2999. [Google Scholar] [CrossRef] [PubMed]
  22. Maitrias, P.; Metzinger-Le Meuth, V.; Nader, J.; Reix, T.; Caus, T.; Metzinger, L. MicroRNA deregulation in symptomatic carotid plaque. J. Vasc. Surg. 2015, 62, 1245–1250.e1. [Google Scholar] [CrossRef]
  23. Madrigal-Matute, J.; Rotllan, N.; Aranda, J.F.; Fernández-Hernando, C. MicroRNAs and atherosclerosis. Curr. Atheroscler. Rep. 2013, 15, 322. [Google Scholar] [CrossRef] [PubMed]
  24. De Franciscis, S.; Metzinger, L.; Serra, R. The discovery of novel genomic, transcriptomic, and proteomic biomarkers in cardiovascular and peripheral vascular disease: The state of the art. Biomed. Res. Int. 2016, 2016, 7829174. [Google Scholar] [CrossRef] [Green Version]
  25. Bartel, D.P. MicroRNAs: Target recognition and regulatory functions. Cell 2009, 136, 215–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Jebari-Benslaiman, S.; Galicia-García, U.; Larrea-Sebal, A.; Olaetxea, J.R.; Alloza, I.; Vandenbroeck, K.; Benito-Vicente, A.; Martín, C. Pathophysiology of Atherosclerosis. Int. J. Mol. Sci. 2022, 23, 3346. [Google Scholar] [CrossRef]
  27. Hermida, N.; Balligand, J.L. Low-Density Lipoprotein-Cholesterol-Induced Endothelial Dysfunction and Oxidative Stress: The Role of Statins. Antioxid. Redox Signal. 2014, 20, 1216–1237. [Google Scholar]
  28. Churov, A.; Summerhill, V.; Grechko, A.; Orekhova, V.; Orekhov, A. MicroRNAs as Potential Biomarkers in Atherosclerosis. Int. J. Mol. Sci. 2019, 20, 5547. [Google Scholar] [CrossRef] [Green Version]
  29. 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] [Green Version]
  30. Qu, M.J.; Pan, J.J.; Shi, X.J.; Zhang, Z.J.; Tang, Y.H.; Yang, G.Y. MicroRNA-126 is a prospective target for vascular disease. Neuroimmunol. Neuroinflammation 2018, 5, 10. [Google Scholar] [CrossRef] [Green Version]
  31. Zhu, J.; Chen, T.; Yang, L.; Li, Z.; Wong, M.M.; Zheng, X.; Pan, X.; Zhang, L.; Yan, H. Regulation of microRNA-155 in atherosclerotic inflammatory responses by targeting MAP3K10. PLoS ONE 2012, 7, e46551. [Google Scholar] [CrossRef] [PubMed]
  32. Sun, H.X.; Zeng, D.Y.; Li, R.T.; Pang, R.P.; Yang, H.; Hu, Y.L.; Zhang, Q.; Jiang, Y.; Huang, L.Y.; Tang, Y.B.; et al. Essential role of microRNA-155 in regulating endothelium-dependent vasorelaxation by targeting endothelial nitric oxide synthase. Hypertension 2012, 60, 1407–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Baran, J.; Niewiara, Ł.; Podolec, J.; Siedliński, M.; Józefczuk, E.; Bernacik, A.; Badacz, R.; Przewłocki, T.; Pieniążek, P.; Żmudka, K.; et al. Serum and Vascular Stiffness Associated with the Severity of Degenerative Aortic Valve Stenosis and Cardiovascular Outcomes. J. Cardiovasc. Dev. Dis. 2022, 9, 193. [Google Scholar] [CrossRef]
  34. Enevoldsen, F.C.; Sahana, J.; Wehland, M.; Grimm, D.; Infanger, M.; Krüger, M. Endothelin Receptor Antagonists: Status Quo and Future Perspectives for Targeted Therapy. J. Clin. Med. 2020, 9, 824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. 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 lipoproteininduced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett. 2011, 585, 854–860. [Google Scholar] [CrossRef] [Green Version]
  36. Melton, D.W.; Lei, X.; Gelfond, J.A.; Shireman, P.K. Dynamic macrophage polarization-specific miRNA patterns reveal increased soluble VEGF receptor 1 by miR-125a-5p inhibition. Physiol. Genom. 2016, 48, 345–360. [Google Scholar] [CrossRef] [Green Version]
  37. Shan, Z.; Qin, S.; Li, W.; Wu, W.; Yang, J.; Chu, M.; Li, X.; Huo, Y.; Schaer, G.L.; Wang, S. An Endocrine Genetic Signal between Blood Cells and Vascular Smooth Muscle Cells: Role of MicroRNA-223 in Smooth Muscle Function and Atherogenesis. J. Am. Coll. Cardiol. 2015, 65, 2526–2537. [Google Scholar] [CrossRef] [Green Version]
  38. 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] [Green Version]
  39. Vinod, M.; Chennamsetty, I.; Colin, S.; Belloy, L.; De Paoli, F.; Schaider, H.; Graier, W.F.; Frank, S.; Kratky, D.; Staels, B.; et al. MiR-206 Controls LXRα Expression and Promotes LXR-Mediated Cholesterol Efflux in Macrophages. Biochim. Biophys. Acta 2014, 1841, 827–835. [Google Scholar] [CrossRef] [Green Version]
  40. Vickers, K.C.; Landstreet, S.R.; Levin, M.G.; Shoucri, B.M.; Toth, C.L.; Taylor, R.C.; Palmisano, B.T.; Tabet, F.; Cui, H.L.; Rye, K.-A.; et al. MicroRNA-223 Coordinates Cholesterol Homeostasis. Proc. Natl. Acad. Sci. USA 2014, 111, 14518–14523. [Google Scholar] [CrossRef] [Green Version]
  41. 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]
  42. Ghafouri-Fard, S.; Khoshbakht, T.; Hussen, B.M.; Baniahmad, A.; Branicki, W.; Taheri, M.; Eghbali, A. Emerging Role of Non-Coding RNAs in Senescence. Front. Cell Dev. Biol. 2022, 10, 869011. [Google Scholar] [CrossRef] [PubMed]
  43. Hergenreider, E.; Heydt, S.; Tréguer, K.; Boettger, T.; Horrevoets, A.J.; Zeiher, A.M.; Scheffer, M.P.; Frangakis, A.S.; Yin, X.; Mayr, M.; et al. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nat. Cell Biol. 2012, 14, 249–256. [Google Scholar] [CrossRef]
  44. Gao, J.; Yang, S.; Wang, K.; Zhong, Q.; Ma, A.; Pan, X. Plasma miR-126 and miR-143 as Potential Novel Biomarkers for Cerebral Atherosclerosis. J. Stroke Cereb. Dis. 2019, 28, 38–43. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, J.; Hu, X.; Hu, X.; Gao, F.; Li, M.; Cui, Y.; Wei, X.; Qin, Y.; Zhang, C.; Zhao, Y.; et al. MicroRNA-520c-3p targeting of RelA/p65 suppresses atherosclerotic plaque formation. Int. J. Biochem. Cell Biol. 2021, 131, 105873. [Google Scholar] [CrossRef] [PubMed]
  46. 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, 13, 1824–1841. [Google Scholar] [CrossRef]
  47. Loyer, X.; Potteaux, S.; Vion, A.C.; Guérin, C.L.; Boulkroun, S.; Rautou, P.E.; Ramkhelawon, B.; Esposito, B.; Dalloz, M.; Paul, J.L.; et al. Inhibition of microRNA-92a prevents endothelial dysfunction and atherosclerosis in mice. Circ. Res. 2014, 114, 434–443. [Google Scholar] [CrossRef] [Green Version]
  48. Badi, I.; Mancinelli, L.; Polizzotto, A.; Ferri, D.; Zeni, F.; Burba, I.; Milano, G.; Brambilla, F.; Saccu, C.; Bianchi, M.E.; et al. miR-34a Promotes Vascular Smooth Muscle Cell Calcification by Downregulating SIRT1 (Sirtuin 1) and Axl (AXL Receptor Tyrosine Kinase). Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2079–2090. [Google Scholar] [CrossRef]
  49. Raucci, A.; Macrì, F.; Castiglione, S.; Badi, I.; Vinci, M.C.; Zuccolo, E. MicroRNA-34a: The bad guy in age-related vascular diseases. Cell. Mol. Life Sci. 2021, 78, 7355–7378. [Google Scholar] [CrossRef]
  50. 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]
  51. Rayner, K.J.; Sheedy, F.J.; Esau, C.C.; Hussain, F.N.; Temel, R.E.; Parathath, S.; van Gils, J.M.; Rayner, A.J.; Chang, A.N.; Suarez, Y.; et al. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J. Clin. Investig. 2011, 121, 2921–2931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Nazari-Jahantigh, M.; Wei, Y.; Noels, H.; Akhtar, S.; Zhou, Z.; Koenen, R.R. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J. Clin. Investig. 2012, 122, 4190–4202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yang, Y.; Yang, L.; Liang, X.; Zhu, G. MicroRNA-155 Promotes Atherosclerosis Inflammation via Targeting SOCS1. Cell. Physiol. Biochem. 2015, 36, 1371–1381. [Google Scholar] [CrossRef] [PubMed]
  54. Tan, L.; Liu, L.; Jiang, Z.; Hao, X. Inhibition of microRNA-17-5p reduces the inflammation and lipid accumulation, and up-regulates ATP-binding cassette transporterA1 in atherosclerosis. J. Pharmacol. Sci. 2019, 139, 280–288. [Google Scholar] [CrossRef]
  55. Zhou, J.; Wang, K.C.; Wu, W.; Subramaniam, S.; Shyy, J.Y.; Chiu, J.J.; Li, J.Y.; Chien, S. MicroRNA-21 targets peroxisome proliferators-activated receptor-alpha in an autoregulatory loop to modulate flow-induced endothelial inflammation. Proc. Natl. Acad. Sci. USA 2011, 108, 10355–10360. [Google Scholar] [CrossRef] [Green Version]
  56. Feng, J.; Li, A.; Deng, J.; Yang, Y.; Dang, L.; Ye, Y.; Li, Y.; Zhang, W. miR-21 attenuates lipopolysaccharide-induced lipid accumulation and inflammatory response: Potential role in cerebrovascular disease. Lipids Health Dis. 2014, 13, 27. [Google Scholar] [CrossRef] [Green Version]
  57. Qin, B.; Shu, Y.; Long, L.; Li, H.; Men, X.; Feng, L.; Yang, H.; Lu, Z. MicroRNA-142-3p Induces Atherosclerosis-Associated Endothelial Cell Apoptosis by Directly Targeting Rictor. Cell. Physiol. Biochem. 2018, 47, 1589–1603. [Google Scholar]
  58. Wei, Y.; Nazari-Jahantigh, M.; Chan, L.; Zhu, M.; Heyll, K.; Corbalán-Campos, J.; Hartmann, P.; Thiemann, A.; Weber, C.; Schober, A. The microRNA-342-5p fosters inflammatory macrophage activation through an Akt1- and microRNA-155-dependent pathway during atherosclerosis. Circulation 2013, 127, 1609–1619. [Google Scholar] [CrossRef] [Green Version]
  59. 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, 15, 836579. [Google Scholar] [CrossRef]
  60. 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] [Green Version]
  61. Zhu, B.; Liu, W.; Xu, Q.; Liu, H.L. Clinical Values and Underlying Mechanism Analysis of Serum miR-455-5p in Carotid Artery Stenosis. J. Inflamm. Res. 2022, 15, 3207–3217. [Google Scholar] [CrossRef] [PubMed]
  62. 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. 2009, 119, 1451–1460. [Google Scholar] [CrossRef] [PubMed]
  63. Kaudewitz, D.; Skroblin, P.; Bender, L.H.; Barwari, T.; Willeit, P.; Pechlaner, R.; Sunderland, N.P.; Willeit, K.; Morton, A.C.; Armstrong, P.C.; et al. Association of MicroRNAs and YRNAs with Platelet Function. Circ. Res. 2016, 118, 420–432. [Google Scholar] [CrossRef] [PubMed]
  64. Wang, X.; Lian, Y.; Wen, X.; Guo, J.; Wang, Z.; Jiang, S.; Hu, Y. Expression of miR-126 and its potential function in coronary artery disease. Afr. Health Sci. 2017, 17, 474–480. [Google Scholar] [CrossRef] [PubMed]
  65. Eken, S.M.; Jin, H.; Chernogubova, E.; Li, Y.; Simon, N.; Sun, C.; Korzunowicz, G.; Busch, A.; Backlund, A.; Osterholm, C.; et al. MicroRNA-210 Enhances Fibrous Cap Stability in Advanced Atherosclerotic Lesions. Circ. Res. 2017, 120, 633–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Hartmann, P.; Schober, A.; Weber, C. Chemokines and microRNAs in atherosclerosis. Cell. Mol. Life Sci. 2015, 72, 3253–3266. [Google Scholar] [CrossRef] [Green Version]
  67. Huang, R.; Hu, Z.; Cao, Y.; Li, H.; Zhang, H.; Su, W.; Xu, Y.; Liang, L.; Melgiri, N.D.; 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]
  68. Wang, J.; Bai, X.; Song, Q.; Fan, F.; Hu, Z.; Cheng, G.; Zhang, Y. miR-223 Inhibits Lipid Deposition and Inflammation by Suppressing Toll-Like Receptor 4 Signaling in Macrophages. Int. J. Mol. Sci. 2015, 16, 24965–24982. [Google Scholar] [CrossRef]
  69. Niculescu, L.S.; Simionescu, N.; Sanda, G.M.; Carnuta, M.G.; Stancu, C.S.; Popescu, A.C.; Popescu, M.R.; 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]
  70. Liu, P.; Su, J.; Song, X.; Wang, S. miR-92a regulates the expression levels of matrix metalloproteinase 9 and tissue inhibitor of metalloproteinase 3 via sirtuin 1 signaling in hydrogen peroxide-induced vascular smooth muscle cells. Mol. Med. Rep. 2018, 17, 1041–1048. [Google Scholar] [CrossRef] [Green Version]
  71. Shi, L.; Yu, C.; Tian, X.; Ma, C.; Wang, L.; Xia, D.; Cui, C.; Chen, X.; Jiang, T.; Gu, Y.; et al. Effect of microRNA-133a-3p/matrix metalloproteinase-9 axis on the growth of atherosclerotic vascular smooth muscle cells. Exp. Ther. Med. 2019, 18, 4356–4362. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, D.; Zhang, M.; Xie, W.; Lan, G.; Cheng, H.P.; Gong, D.; Huang, C.; Lv, Y.C.; Yao, F.; Tan, Y.L.; et al. MiR-486 regulates cholesterol efflux by targeting HAT1. Biochem. Biophys. Res. Commun. 2016, 472, 418–424. [Google Scholar] [CrossRef] [PubMed]
  73. Xiao, L.; Ming, H.; Tao, C.; Yuliang, W. The expression of SOCS is altered in atherosclerosis. Heart 2011, 97, A51. [Google Scholar] [CrossRef]
  74. 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] [PubMed]
  75. Pankratz, F.; Hohnloser, C.; Bemtgen, X.; Jaenich, C.; Kreuzaler, S.; Hoefer, I.; Pasterkamp, G.; Mastroianni, J.; Zeiser, R.; Smolka, C.; et al. MicroRNA-100 Suppresses Chronic Vascular Inflammation by Stimulation of Endothelial Autophagy. Circ. Res. 2018, 122, 417–432. [Google Scholar] [CrossRef] [PubMed]
  76. Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [Green Version]
  77. Wei, Y.; Zhu, M.; Corbalan-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. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 796–803. [Google Scholar]
  78. Tian, F.J.; An, L.N.; Wang, G.K.; Zhu, J.Q.; Li, Q.; Zhang, Y.Y.; Zeng, A.; Zou, J.; Zhu, R.F.; Han, X.S.; et al. Elevated microRNA-155 promotes foam cell formation by targeting HBP1 in atherogenesis. Cardiovasc. Res. 2014, 103, 100–110. [Google Scholar] [CrossRef] [PubMed]
  79. Chen, W.J.; Yu, F.; Di, M.; Li, M.; Chen, Y.; Zhang, Y.; Liu, X.; Huang, X.; Zhang, M. MicroRNA-124-3p inhibits collagen synthesis in atherosclerotic plaques by targeting prolyl 4-hydroxylase subunit alpha-1 (P4HA1) in vascular smooth muscle cells. Atherosclerosis 2018, 277, 98–107. [Google Scholar] [CrossRef]
  80. Liang, X.; Wang, L.; Wang, M.; Liu, Z.; Liu, X.; Zhang, B.; Liu, E.; Li, G. MicroRNA-124 inhibits macrophage cell apoptosis via targeting p38/MAPK signaling pathway in atherosclerosis development. Aging (Albany NY) 2020, 12, 13005–13022. [Google Scholar] [CrossRef]
  81. Lan, G.; Xie, W.; Li, L.; Zhang, M.; Liu, D.; Tan, Y.L.; Cheng, H.P.; Gong, D.; Huang, C.; Zheng, X.L.; et al. MicroRNA-134 actives lipoprotein lipase-mediated lipid accumulation and inflammatory response by targeting angiopoietin-like 4 in THP-1 macrophages. Biochem. Biophys. Res. Commun. 2016, 472, 410–417. [Google Scholar] [CrossRef] [PubMed]
  82. Ye, Q.; Tian, G.P.; Cheng, H.P.; Zhang, X.; Ou, X.; Yu, X.H.; Tan, R.Q.; Yang, F.Y.; Gong, D.; Huang, C.; et al. MicroRNA-134 Promotes the Development of Atherosclerosis Via the ANGPTL4/LPL Pathway in Apolipoprotein E Knockout Mice. J. Atheroscler. Thromb. 2018, 25, 244–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Badacz, R.; Przewłocki, 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. Postep. Kardiol. Inter. 2018, 14, 75–84. [Google Scholar] [CrossRef] [PubMed]
  84. 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]
  85. Chang, Y.J.; Li, Y.S.; Wu, C.C.; Wang, K.C.; Huang, T.C.; Chen, Z.; Chien, S. Extracellular MicroRNA-92a Mediates Endothelial Cell-Macrophage Communication. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 2492–2504. [Google Scholar] [CrossRef]
  86. Zhan, J.; Qin, S.; Lu, L.; Hu, X.; Zhou, J.; Sun, Y.; Yang, J.; Liu, Y.; Wang, Z.; Tan, N.; et al. miR-34a is a common link in both HIV- and antiretroviral therapy-induced vascular aging. Aging (Albany NY) 2016, 8, 3298–3310. [Google Scholar] [CrossRef] [Green Version]
  87. Ono, K. Functions of microRNA-33a/b and microRNA therapeutics. J. Cardiol. 2016, 67, 28–33. [Google Scholar] [CrossRef] [Green Version]
  88. Nguyen, M.A.; Wyatt, H.; Susser, L.; Geoffrion, M.; Rasheed, A.; Duchez, A.C.; Cottee, M.L.; Afolayan, E.; Farah, E.; Kahiel, Z.; et al. Delivery of MicroRNAs by Chitosan Nanoparticles to Functionally Alter Macrophage Cholesterol Efflux in Vitro and in Vivo. ACS Nano 2019, 13, 6491–6505. [Google Scholar] [CrossRef]
  89. 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. Genom. 2018, 293, 883–894. [Google Scholar] [CrossRef]
  90. Cao, R.Y.; Li, Q.; Miao, Y.; Zhang, Y.; Yuan, W.; Fan, L.; Liu, G.; Mi, Q.; Yang, J. The Emerging Role of MicroRNA-155 in Cardiovascular Diseases. Biomed. Res. Int. 2016, 2016, 9869208. [Google Scholar] [CrossRef] [Green Version]
  91. Méndez-Barbero, N.; Gutiérrez-Muñoz, C.; Blanco-Colio, L.M. Cellular Crosstalk between Endothelial and Smooth Muscle Cells in VascularWall Remodeling. Int. J. Mol. Sci. 2021, 22, 7284. [Google Scholar] [PubMed]
  92. Wang, C.; Wu, H.; Xing, Y.; Ye, Y.; He, F.; Yin, Q.; Li, Y.; Shang, F.; Shyy, J.Y.-J.; Yuan, Z.-Y. Endothelial-derived extracellular microRNA-92a promotes arterial stiffness by regulating phenotype changes of vascular smooth muscles cells. Sci. Rep. 2022, 12, 344. [Google Scholar] [CrossRef] [PubMed]
  93. Zhou, J.; Li, Y.S.; Nguyen, P.; Wang, K.C.; Weiss, A.; Kuo, Y.C.; Chiu, J.J.; Shyy, J.Y.; Chien, S. Regulation of vascular smooth muscle cell turnover by endothelial cell-secreted microRNA-126: Role of shear stress. Circ. Res. 2012, 113, 40–51. [Google Scholar]
  94. 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] [PubMed]
  95. Ji, R.; Cheng, Y.; Yue, J.; Yang, J.; Liu, X.; Chen, H.; Dean, D.B.; Zhang, C. MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ. Res. 2007, 100, 1579–1588. [Google Scholar] [CrossRef] [PubMed]
  96. Denecke, B.; Rostalsky, A.; Hristov, M.; Koeppel, T.A.; Bidzhekov, K.; Gan, L.; Zernecke, A.; Weber, C. microRNA expression signatures and parallels between monocyte subsets and atherosclerotic plaque in humans. Thromb. Haemost. 2012, 107, 619–625. [Google Scholar] [CrossRef]
  97. 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] [Green Version]
  98. Soeki, T.; Yamaguchi, K.; Niki, T.; Uematsu, E.; Bando, S.; Matsuura, T.; Ise, T.; Kusunose, K.; Hotchi, J.; Tobiume, T.; et al. Plasma microRNA-100 is associated with coronary plaque vulnerability. Circ. J. 2015, 79, 413–418. [Google Scholar] [CrossRef] [Green Version]
  99. Kabłak-Ziembicka, A.; Badacz, R.; Przewłocki, T. Clinical Application of Serum microRNAs in Atherosclerotic Coronary Artery Disease. J. Clin. Med. 2022, 11, 6849. [Google Scholar] [CrossRef]
  100. Raskurazhev, A.A.; Kuznetsova, P.I.; Shabalina, A.A.; Tanashyan, M.M. MicroRNA and Hemostasis Profile of Carotid Atherosclerosis. Int. J. Mol. Sci. 2022, 23, 10974. [Google Scholar] [CrossRef]
  101. Pereira-Da-Silva, T.; Napoleão, P.; Costa, M.; Gabriel, A.; Selas, M.; Silva, F.; Enguita, F.; Ferreira, R.; Carmo, M. Circulatin miRNAs Are Associated with the Systemic Extent of Atherosclerosis: Novel Observations for miR-27b and miR-146. Diagnostics 2021, 11, 318. [Google Scholar] [CrossRef]
  102. Mao, Z.; Wu, F.; Shan, Y. Identification of key genes and miRNAs associated with carotid atherosclerosis based on mRNA-seq data. Medicine 2018, 97, e9832. [Google Scholar] [CrossRef] [PubMed]
  103. Kabłak-Ziembicka, A.; Przewłocki, T. Clinical Significance of Carotid Intima-Media Complex and Carotid Plaque Assessment by Ultrasound for the Prediction of Adverse Cardiovascular Events in Primary and Secondary Care Patients. J. Clin. Med. 2021, 10, 4628. [Google Scholar] [CrossRef]
  104. Liu, D.; Du, C.; Shao, W.; Ma, G. Diagnostic Role of Carotid Intima-Media Thickness for Coronary Artery Disease: A Meta-Analysis. Biomed. Res. Int. 2020, 2020, 9879463. [Google Scholar] [CrossRef] [Green Version]
  105. Willeit, P.; Tschiderer, L.; Allara, E.; Reuber, K.; Seekircher, L.; Gao, L.; Liao, X.; Lonn, E.; Gerstein, H.C.; Yusuf, S.; et al. Carotid Intima-Media Thickness Progression as Surrogate Marker for Cardiovascular Risk: Meta-Analysis of 119 Clinical Trials Involving 100 667 Patients. Circulation 2020, 142, 621–642. [Google Scholar] [CrossRef] [PubMed]
  106. Peczyńska, J.; Klonowska, B.; Żelazowska-Rutkowska, B.; Polkowska, A.; Noiszewska, K.; Bossowski, A.; Głowińska -Olszewska, B. The Relationship between Selected Inflammation and Oxidative Stress Biomarkers and Carotid Intima-Media Thickness (IMT) Value in Youth with Type 1 Diabetes Co-Existing with Early Microvascular Complications. J. Clin. Med. 2022, 11, 4732. [Google Scholar] [PubMed]
  107. Kim, S.-A.; Park, K.H.; Woo, S.; Kim, Y.M.; Lim, H.J.; Park, W.-J. Vascular Alterations Preceding Arterial Wall Thickening in Overweight and Obese Children. J. Clin. Med. 2022, 11, 3520. [Google Scholar] [CrossRef]
  108. Kabłak-Ziembicka, A.; Przewłocki, T.; Tracz, W.; Podolec, P.; Stopa, I.; Kostkiewicz, M.; Mura, A.; Sadowski, J.; Kopeć, G. Prognostic value of carotid intima-media thickness in detection of coronary atherosclerosis in patients with calcified aortic valve stenosis. J. Ultrasound Med. 2005, 24, 461–467. [Google Scholar] [CrossRef] [PubMed]
  109. Kabłak-Ziembicka, A.; Przewłocki, T.; Tracz, W.; Pieniążek, P.; Musialek, P.; Sokołowski, A.; Drwila, R.; Rzeźnik, D. Assessment of carotid intima-media thickness in peri- and postmenopausal women with suspected coronary artery disease. Heart Vessels 2008, 23, 295–300. [Google Scholar]
  110. Liu, C.Z.; Zhong, Q.; Huang, Y.Q. Elevated Plasma miR-29a Levels Are Associated with Increased Carotid Intima-Media Thickness in Atherosclerosis Patients. Tohoku J. Exp. Med. 2017, 241, 183–188. [Google Scholar] [CrossRef] [Green Version]
  111. Olejarz, W.; Łacheta, D.; Kubiak-Tomaszewska, G. Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int. J. Mol. Sci. 2020, 21, 3946. [Google Scholar] [CrossRef] [PubMed]
  112. Lightbody, R.J.; Taylor, J.M.W.; Dempsie, Y.; Graham, A. MicroRNA sequences modulating inflammation and lipid accumulation in macrophage “foam” cells: Implications for atherosclerosis. World J. Cardiol. 2020, 12, 303–333. [Google Scholar] [PubMed]
  113. Solly, E.L.; Dimasi, C.G.; Bursill, C.A.; Psaltis, P.J.; Tan, J.T.M. MicroRNAs as Therapeutic Targets and Clinical Biomarkers in Atherosclerosis. J. Clin. Med. 2019, 8, 2199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Puig, N.; Jiménez-Xarrié, E.; Camps-Renom, P.; Benitez, S. Search for Reliable Circulating Biomarkers to Predict Carotid Plaque Vulnerability. Int. J. Mol. Sci. 2020, 21, 8236. [Google Scholar] [CrossRef]
  115. 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]
  116. Yingchun, X.; 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]
  117. Badacz, R.; Podolec, J.; Przewłocki, T.; Siedliński, M.; Józefczuk, E.; Oleksy, H.; Baran, J.; Pieniążek, P.; Żmudka, K.; Kabłak-Ziembicka, A. The role of chemokine CCL5/RANTES and metalloproteinase-9 as inflammatory modulators in symptomatic internal carotid artery stenosis. J. Physiol. Pharmacol. 2019, 70, 545–555. [Google Scholar] [CrossRef]
  118. Wang, C.; Zhang, Y.; Jiang, Z.; Bai, H.; Du, Z. miR-100 alleviates the inflammatory damage and apoptosis of H2O2-induced human umbilical vein endothelial cells via inactivation of Notch signaling by targeting MMP9. Vascular 2022, 30, 151–161. [Google Scholar] [CrossRef]
  119. Raju, S.; Fish, J.E.; Howe, K.L. MicroRNAs as sentinels and protagonists of carotid artery thromboembolism. Clin. Sci. 2020, 134, 169–192. [Google Scholar] [CrossRef]
  120. Joh, J.H.; Cho, S. Cardiovascular risk of carotid atherosclerosis: Global consensus beyond societal guidelines. Lancet Glob. Health 2020, 8, e625–e626. [Google Scholar]
  121. Dolz, S.; Górriz, D.; Tembl, J.I.; Sánchez, D.; Fortea, G.; Parkhutik, V.; Lago, A. Circulating MicroRNAs as Novel Biomarkers of Stenosis Progression in Asymptomatic Carotid Stenosis. Stroke 2017, 48, 10–16. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, T.; Liu, R. Dysregulation of miR-637 serves as a diagnostic biomarker in patients with carotid artery stenosis and predicts the occurrence of the cerebral ischemic event. Bioengineered 2021, 12, 8658–8665. [Google Scholar] [CrossRef] [PubMed]
  123. Zhu, B.; Liu, W.; Xu, Q.; Liu, H.L. MicroRNA-486-5p functions as a diagnostic marker for carotid artery stenosis and prevents endothelial dysfunction through inhibiting inflammation and oxidative stress. Bioengineered 2022, 13, 8667–8675. [Google Scholar] [CrossRef] [PubMed]
  124. Grosse, G.M.; Derda, A.A.; Stauss, R.D.; Neubert, L.; Jonigk, D.D.; Kühnel, M.P.; Gabriel, M.M.; Schuppner, R.; Wilhelmi, M.; Bär, C.; et al. Circulating microRNAs in Symptomatic and Asymptomatic Carotid Stenosis. Front. Neurol. 2021, 12, 755827. [Google Scholar] [CrossRef]
  125. Luque, A.; Farwati, A.; Krupinski, J.; Aran, J.M. Association between low levels of serum miR-638 and atherosclerotic plaque vulnerability in patients with high-grade carotid stenosis. J. Neurosurg. 2018, 131, 72–79. [Google Scholar] [CrossRef] [PubMed]
  126. Zhang, T.; Chen, Z.; Yang, X.; Fu, R.; Wang, J.; Xu, H. Circulating miR-106b-5p serves as a diagnostic biomarker for asymptomatic carotid artery stenosis and predicts the occurrence of cerebral ischemic events. Vasc. Med. 2020, 25, 436–442. [Google Scholar] [CrossRef]
  127. Li, R.; Jiang, L.; Wang, X. Aberrant expression of miR-483-5p in patients with asymptomatic carotid artery stenosis and its predictive value for cerebrovascular event occurrence. Exp. Ther. Med. 2021, 22, 1101. [Google Scholar] [CrossRef]
  128. Zhou, A.; Li, Y.; Wang, P.; Yu, P.; Lang, L. Circulating miR-342-5p serves as a diagnostic biomarker in patients with carotid artery stenosis and predicts the occurrence of the cerebral ischemic event. Ir. J. Med. Sci. 2022, 191, 713–718. [Google Scholar] [CrossRef]
  129. Chen, G.; Gao, J.; Sheng, Y.; Han, X.; Ji, X.; Zhao, M.; Wu, J. Diagnostic value of miR-92a in asymptomatic carotid artery stenosis patients and its ability to predict cerebrovascular events. Diagn. Pathol. 2020, 15, 74. [Google Scholar] [CrossRef]
  130. Liu, X.; Zheng, X.; Wang, Y.; Liu, J. Dysregulation Serum miR-19a-3p is a Diagnostic Biomarker for Asymptomatic Carotid Artery Stenosis and a Promising Predictor of Cerebral Ischemia Events. Clin. Appl. Thromb./Hemost. 2021, 27, 10760296211039287. [Google Scholar] [CrossRef]
  131. Lu, T.; Li, X.; Long, C.; Ji, W.; Jiang, L.; Tian, J. Circulating miR-27b as a Biomarker of the Development and Progression of Carotid Artery Stenosis. Clin. Appl. Thromb./Hemost. 2021, 27, 10760296211057903. [Google Scholar] [CrossRef] [PubMed]
  132. Li, D.; Pan, J. Diagnostic and Prognostic Value Analysis of miR-206 in Asymptomatic Carotid Artery Stenosis. Br. J. Biomed. Sci. 2022, 79, 10592. [Google Scholar] [CrossRef] [PubMed]
  133. Liu, H.; Zhou, J.; Jiang, W.; Wang, F. Analysis of the diagnostic and prognostic value of miR-9-5p in carotid artery stenosis. Bosn. J. Basic Med. Sci. 2021, 21, 724–729. [Google Scholar] [CrossRef]
  134. Badacz, R.; Przewłocki, T.; Pieniążek, P.; Rosławiecka, A.; Klec-zyński, 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]
  135. Tsai, P.C.; Liao, Y.C.; Wang, Y.S.; Lin, H.F.; Lin, R.T.; Juo, S.H. Serum microRNA-21 and microRNA-221 as potential biomarkers for cerebrovascular disease. J. Vasc. Res. 2013, 50, 346–354. [Google Scholar] [CrossRef]
  136. Wang, L.; Xu, L. Combined Value of Serum miR-124, TNF-α and IL-1β for Vulnerable Carotid Plaque in Acute Cerebral Infarction. J. Coll. Physicians Surg. Pak. 2020, 30, 385–388. [Google Scholar] [CrossRef]
  137. Xu, Y.; Zhao, L.; Liu, H.; Sun, B.; Zhao, X. Diagnostic value of miR-637 in patients with atherosclerosis and its predictive significance for the future cardiovascular events. Vascular 2021, 29, 704–710. [Google Scholar] [CrossRef]
  138. Yang, N.; Dong, B.; Song, Y.; Li, Y.; Kou, L.; Yang, J.; Qin, Q. Downregulation of miR-637 promotes vascular smooth muscle cell proliferation and migration via regulation of insulin-like growth factor-2. Cell. Mol. Biol. Lett. 2020, 25, 30. [Google Scholar] [CrossRef]
  139. Li, F.; Guo, X.; Chen, S.Y. Function and therapeutic potential of mesenchymal stem cells in atherosclerosis. Front. Cardiovasc. Med. 2017, 4, 32. [Google Scholar] [CrossRef]
  140. Badacz, R.; Kabłak-Ziembicka, A.; Urbańczyk-Zawadzka, M.; Banyś, R.P.; Musiałek, P.; Odrowąż-Pieniążek, P.; Trystuła, M.; Ścigalski, J.; Żmudka, K.; Przewłocki, T. Magnetic resonance imaging and clinical outcome in patients with symptomatic carotid artery stenosis after carotid artery revascularization. Postep. Kardiol. Inter. 2017, 13, 225–232. [Google Scholar] [CrossRef] [Green Version]
  141. Sardu, C.; Modugno, P.; Castellano, G.; Scisciola, L.; Barbieri, M.; Petrella, L.; Fanelli, M.; Macchia, G.; Caradonna, E.; Massetti, M.; et al. Atherosclerotic Plaque Fissuration and Clinical Outcomes in Pre-Diabetics vs. Normoglycemics Patients Affected by Asymptomatic Significant Carotid Artery Stenosis at 2 Years of Follow-Up: Role of microRNAs Modulation: The ATIMIR Study. Biomedicines 2021, 9, 401. [Google Scholar] [PubMed]
  142. Stępień, E.; Kabłak-Ziembicka, A.; Czyż, J.; Przewłocki, T.; Małecki, M. Microparticles, not only markers but also a therapeutic target in the early stage of diabetic retinopathy and vascular aging. Expert Opin. Ther. Targets 2012, 16, 677–688. [Google Scholar] [CrossRef] [PubMed]
  143. Bazan, H.A.; Hatfield, S.A.; O’Malley, C.B.; Brooks, A.J.; Lightell, D., Jr.; Woods, T.C. Acute Loss of miR-221 and miR-222 in the Atherosclerotic Plaque Shoulder Accompanies Plaque Rupture. Stroke 2015, 46, 3285–3287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Magenta, A.; Sileno, S.; D’Agostino, M.; Persiani, F.; Beji, S.; Paolini, A.; Camilli, D.; Platone, A.; Capogrossi, M.C.; Furgiuele, S. Atherosclerotic plaque instability in carotid arteries: miR-200c as a promising biomarker. Clin. Sci. 2018, 132, 2423–2436. [Google Scholar] [CrossRef] [PubMed]
  145. Wei, X.; Sun, Y.; Han, T.; Zhu, J.; Xie, Y.; Wang, S.; Wu, Y.; Fan, Y.; Sun, X.; Zhou, J.; et al. Upregulation of miR-330-5p is associated with carotid plaque’s stability by targeting Talin-1 in symptomatic carotid stenosis patients. BMC Cardiovasc. Disord. 2019, 19, 149. [Google Scholar] [CrossRef]
  146. Grundmann, S.; Hans, F.P.; Kinniry, S.; Heinke, J.; Helbing, T.; Bluhm, F.; Sluijter, J.P.; Hoefer, I.; Pasterkamp, G.; Bode, C.; et al. MicroRNA-100 regulates neovascularization by suppression of mammalian target of rapamycin in endothelial and vascular smooth muscle cells. Circulation 2011, 123, 999–1009. [Google Scholar]
  147. Santovito, D.; Mezzetti, A.; Cipollone, F. MicroRNAs and atherosclerosis: New actors for an old movie. Nutr. Metab. Cardiovasc. Dis. 2012, 22, 937–943. [Google Scholar] [CrossRef]
  148. Chen, T.; Huang, Z.; Wang, L.; Wang, Y.; Wu, F.; Meng, S.; Wang, C. MicroRNA125a-5p partly regulates the inflammatory response, lipid uptake, and ORP9 expression in oxLDL-stimulated monocyte/macrophages. Cardiovasc. Res. 2009, 83, 131–139. [Google Scholar]
  149. Torella, D.; Iaconetti, C.; Catalucci, D.; Ellison, G.M.; Leone, A.; Waring, C.D.; Bochicchio, A.; Vicinanza, C.; Aquila, I.; Curcio, A.; et al. MicroRNA-133 controls vascular smooth muscle cell phenotypic switch in vitro and vascular remodeling in vivo. Circ. Res. 2011, 109, 880–893. [Google Scholar] [CrossRef] [Green Version]
  150. Cordes, K.R.; Sheehy, N.T.; White, M.P.; Berry, E.C.; Morton, S.U.; Muth, A.N.; Lee, T.H.; Miano, J.M.; Ivey, K.N.; Srivastava, D. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 2009, 460, 705–710. [Google Scholar] [CrossRef] [Green Version]
  151. Huang, P.; He, X.Y.; Xu, M. The Role of miRNA-146a and Proinflammatory Cytokines in Carotid Atherosclerosis. Biomed. Res. Int. 2020, 2020, 6657734. [Google Scholar] [CrossRef] [PubMed]
Table 1. Critical miRNAs participating in atherosclerotic carotid artery lesions development: from fatty streaks to mature plaque: a therapeutic approach.
Table 1. Critical miRNAs participating in atherosclerotic carotid artery lesions development: from fatty streaks to mature plaque: a therapeutic approach.
Critical Stages in AtherosclerosismiRNAMechanismEffect of miRNA ActionTherapeutic Approach (HUVEC or Animal Studies)Ref.
Initiation and early atherosclerosis
‘Brakes’ of atherosclerosis
Promotes ECs proliferation and repair, protects ECsmiR-126-5psuppression of the Notch1 inhibitor Dlk1At non-predilection sites, high miR-126-5p levels in ECs confer a proliferative reserve that compensates for the antiproliferative effects of hyperlipidemiaT, injection of miR-126-5p rescued ECs proliferation at predilection sites and limited atherosclerosis[29]
Decreases atherosclerosis progressionmiR-155downregulation of MAP3K10
downregulation of ET-1 and ANG II type I receptor
Down-modulates inflammatory cytokine productionT, the miR-155 mimic decreased IL-6, MMP-9 and TNF-α secretions of oxLDL-induced macrophages[31,32]
Decreases lipid uptake in macrophages, inhibits endothelial activation miR-146aregulates TLR4, increases eNOS expression Inhibits ox-LDL and inflammatory response (decreases IL-6, -8, MMP-9)Overexpression may be useful[35]
Macrophage polarization MiR-125adownregulation of sVEGFR1Decreases lipid uptake in macrophages, modulates extracellular VEGF by manipulating sVEGFR1T, miR-125a-5p inhibition reduces VEGF through the increased sVEGFR1[36]
Increase reverse cholesterol transport from macrophages to the liver for excretionmiR-206
miR-223
promote efflux
promote efflux
crucial for the prevention of lipid accumulation and atherosclerosisT, these miRs can be efficiently delivered to macrophages via chitosan nanoparticles[39,40]
Prevents ECs senescence miR-let-7gStimulates anti-aging gene SIRT1, and IGF 1,
inhibits expression of LOX-1
exert anti-aging effects on ECsT, antagonizing endogenous
let-7 has induced cell
proliferation
[42]
Prevents ECs senescencemiR-143targets a network of transcription factors, including KLF4, myocardin, and Elk-1promotes differentiation and repress proliferation of VSMCsmicrovesicles containing miR-143 injected into mice could reduce the formation of atherosclerotic plaques[43,44]
Suppresses atherosclerotic plaque formationmiR-520targets RelA/p65regulates VSMCs decreasing migration and proliferationmiR-520c-3p agomir decreased atherosclerotic plaque size[45]
High expression is needed to maintain a contractile phenotype of VSMCsmiR-22multiple target genesinduce the phenotypic switch from synthetic to contractileT, the stent with the miR-22 coating showed significant capability to inhibit in-stent restenosis[46]
Promotors of atherosclerosis
Increases endothelial inflammationmiR-92aregulation of KLF2markedly enhanced by hypercholesterolemiaT, inhibition of miR-92a reduces endothelial inflammation and atheroma plaque size[47]
Vascular senescence, vascular calcifications
Altered lipid metabolism
Increases inflammatory
cytokines secretion of macrophages M1
miR-34ainhibition of SIRT1 and AXL receptor tyrosine kinase
targets cholesterol transporters: ABCA1 and ABCG1
through the nuclear hormone LXRα
aggravates and accelerates vascular senescence
increase the binding capacity
of oxLDL to macrophages
stimulate pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, IL-12, IL-23), and chemokines (CCL5, CCL8, CXCL2, CXCL4)
T, inhibition with antago-miR-34a [48,49]
Promotes cholesterol accumulation in macrophages, decreases reverse cholesterol transport miR-33aTargets hepatic ABCA1inhibit efflux, increases macrophages ox-LDL uptake, foam cells accumulationT, inhibition of miR-33a facilitates atherosclerosis regression[50,51]
Promotes atherosclerosismiR-155repressing Bcl6 in macrophages, suppress eNOSincreases pro-inflammatory NF-κB signaling, down-regulates the expression of eNOS and production of NOT, inhibition of miR-155 increased eNOS expression and NO production[32,52]
Increases apoptosis in ECs miR-17-5prepression of ABCA1 expression through directly binding to its 3′-UTRrate of apoptosis in ECsT, inhibition of miR-17 suppresses apoptosis, hence decrease infarct size area, and improves microcirculation of the heart tissue, decreasing heart failure symptoms[53,54]
Promotion of monocyte adhesion, proinflammatory
Lipid metabolism
miR-21targets PPAR α
targets TLR4 and NF-κB
enhances the expression of VCAM-1 and MCP-1 and the adhesion of monocytes to ECs
LPS-induced lipid accumulation and inflammatory response
in macrophages
Overexpression of miR-21 up-regulated AP-1 activation, which was attenuated by exogenous expression of PPARα
overexpression of miR-21 significantly decreased the secretion of IL-6 and increased IL-10 levels
[55,56]
Induces ECs apoptosis, development of atherosclerosismiR-142-3pup-regulation of Rictor and the Akt/eNOSatherosclerosis-associated
ECs apoptosis
T, the antagomir-142-3p attenuated endothelial apoptosis and retarded the atherosclerosis progression in the aorta of ApoE-/- mice[57]
Increase pro-inflammatory cytokines miR-342-5ptargets Akt1induces proinflammatory mediators such as NOS2 and II6 in macrophages via the upregulation of miR-155T, the miR-342-5p antagomir upregulated Akt1 expression and suppressed the expression of miR-155 and Nos2[58]
Mature plaques
Marker of response to clopidogrel, targets P2Y12 receptormiR-223-3ppossible P2Y12 site targetingon-clopidogrel platelet reactivitydecreased miR-223 expression
was a predictor of low responders
to clopidogrel
[41]
Plaque stabilization miR-145targets KLF4,5VSMCs contractility, increase fibrous cap area, reduce the necrotic core areaT, delivery of miR-145 may limit atherosclerotic plaque growth, and
restore contractile levels in VSMCs
[59,60]
Macrophage polarizationmiR-455targets SOCS3decreased expression leads to ECs injury induced by ox-LDLT, overexpression with miR-455
inhibits apoptosis, migration of VSMCs,
and lowers ox-LDL
[61]
Marker of platelet activation, targets COX-1 receptor through the regulation of TXSmiR-34b-3ptargets TBXAS1miR-34b-3p may regulate the platelet response by suppressing TBXAS1 expression and megakaryocyte proliferationT, miR-34b-3p may facilitate the
antiplatelet efficiency of aspirin
through inhibiting TBXAS1
[62]
Responsive to antiplatelet therapymiR-126-3paffects ADAM9 and P2Y12 receptor expressionIncreases platelets aggregationT, antagomiR against miR-126-3p reduces platelets aggregation[63]
Decreases size of atherosclerotic lesions, alleviate ox-LDL-induced ECs injury, angiogenesis and vascular integritymiR-126-3pactivation of VEGF and NF-kB signaling decreased expression in advanced carotid plaques with high discriminating value (AUC: 0.998) patients with severe carotid stenosis demonstrated down-regulation of miR-126[64]
Plaque stabilizationmiR-210targets the APC gene, affecting Wnt signaling and regulating VSMCs survivalenhances fibrous plaque stability in mature plaquesT, miR-210 mimics prevent carotid plaque rupture; modulating miR-210 improved
fibrous cap stability
[65]
Promotes atherosclerosis growthmiR-103-3ptargets KLF4stimulates inflammatory activation, and uptake of oxidized LDL cholesterolT, reduction in miR-103 levels results
in the reduction of atherosclerosis
and endothelial inflammation
[66]
Decreases ECs regeneration and repairmiR-652-3psuppression of the endothelial repair gene Ccnd2inhibits ECs regeneration and repair following mechanical injurydownregulates Ccnd2 in endothelial cells, lowering cell proliferation[67]
Plaque stabilizationmiR-223targets TLR4reduces foam cell
formation, and production of pro-inflammatory
cytokines
Overexpression decreases lipids
deposition and inflammation
[68]
Plaque instabilitymiR-92a-3pSIRT1, H2O2-induced changes in VSMCsincreased apoptosis, oxidative stress, CIMT, and pro-inflammatory MMP-9,miR-92a overexpression regulates
the expression levels of MMP-9
and TIMP3
[69,70]
Plaque instabilitymiR-133aMatrix metallopeptidase 9 inhibits the proliferation of VSMCs and induces apoptosisthe miR-133a-3p mimic inhibited proliferation and promoted VSMC cell apoptosis[71]
Promotes endothelial migration miR-486targets HAT1induces apoptosis and oxidative stress, pro-atherosclerotic, affects
endothelial migratory activity
Inhibition of miR-486 limits foam
cell formation by increasing
cholesterol efflux
[69,72]
Increases pro-inflammatory cytokinesmiR-331down-regulation
of SOCS1
a pro-inflammatory response in atherosclerotic plaquesmiR-331 suppression causes up-regulation of SOCS1 and anti-inflammatory mechanism in atherosclerosis[73,74]
Plaque stabilizationmiR-100down-regulation of E-selectin and VCAM-1miR-100 restrains vascular inflammation in vitro and in vivo by suppressing endothelial adhesion molecule expression and thereby attenuating leukocyte–endothelial interactionInhibition of miR-100 Stimulates Atherogenesis in Mice[75]
Plaque instabilitymiR-105transported via HDLoverexpression of miR-105 in patients with familial hypercholesterolemiaHDL can deliver miRNA-105 to recipient cells, contributing to altered gene expression[76]
Plaque instabilitymiR-155Targets the transcription factor HBP1Increase macrophages, foam cells content, and necrotic core in plaquesT, inhibition of miR-155 reduced necrotic core, apoptosis, and prevented progression of atherosclerosis [77,78]
Plaque instabilitymiR-124Targets P4HA1 Inhibits collagen synthesis in atherosclerotic plaquesOverexpression of miR-124 increased the expression of anti-inflammatory cytokines by binding p38 signaling pathway[79,80]
Plaque instabilitymiR-134ANGTPL4/LPLassociated with chronic inflammation (hs-CRP and TNF-α), cholesterol mass, and plaque vulnerability features on ultrasonography T, lower LPL activity with inhibitors of miR-134[81,82,83]
LipometabolismmiR-122inhibits AMPK and SIRT1 correlated with TC, TG, and LDL-C levelsserum level of miR-122
correlated with atherosclerotic
severity
[84]
ABCA1: ATP-binding cassette subfamily A member 1; ABCG1: ATP-binding cassette subfamily G member 1; Akt: protein kinase B; AMPK: Adenosine monophosphate–activated protein kinase; ANG II: angiotensin II; ANGTPL/LPL: Angiopoietin/lipoprotein lipase; CIMT: carotid intima-media thickness; Dlk1: delta-like 1 homolog; eNOS: nitric oxide synthase; ECs: endothelial cells; ET-1: Endothelin 1; H2O2: hydrogen peroxide; HAT1: Histone acetyl-transferase 1; HMG-box transcription factor 1; hs-CRP: high-sensitivity C-Reactive Protein; IGF: insulin growth factor; KLF: Kruppel-like factor; LXRα: Liver X receptor α; LPS: lipopolysaccharide; MAP3K10: mitogen-activated protein 3 kinase 10; MCP-1; monocyte chemotactic protein-1; MMP: metalloproteinase protein; n/d: no data available; NF-κB: nuclear factor-κB; P4HA1: prolyl 4-hydroxylase subunit alpha-1; PPARα: peroxisome proliferators-activated receptor-α, RelA/p65: REL-associated protein involved in NF-κB heterodimer formation p65 subunit; sVEGFR1: soluble VEGF receptor-1; SIRT1: sirtuin 1; SOCS1: suppressor of cytokine signaling 1; T: therapeutic approach; TBXAS1: thromboxane synthase thromboxane A synthase 1; TLR4: toll-like receptor 4; TNF-α: tumor necrosis factor alpha; VCAM-1: vascular cell adhesion molecule-1; VSMC: vascular smooth muscle cells; VEGF: vascular endothelial growth factor.
Table 2. Serum/plasma microRNAs that are potentially diagnostic for the degree of ICAS, can differentiate between patients with asymptomatic versus symptomatic ICAS, or are prognostic for risk of ICAS-related IS.
Table 2. Serum/plasma microRNAs that are potentially diagnostic for the degree of ICAS, can differentiate between patients with asymptomatic versus symptomatic ICAS, or are prognostic for risk of ICAS-related IS.
Study ParticipantsmicroRNADown- vs. Up-RegulatedRationale for the Use of
the miRNA
Statistical Analysis
AUC, or HR (95% CI),
p-Value
Ref.
30 patients with aICAS (≥50%)
20 patients with non-obstructive carotid plaques
miR-29a-3p
miR-33a-3p
miR-126-3p
miR-126-5p
Down
Up
Down
Down
Regulates VSMCs function, inflammation
Cholesterol metabolism
ECs repair and proliferation
ECs repair and proliferation
For 50% ICAS or greater:
AUC 0.79, p = 0.035
AUC 0.68, p < 0.001
AUC 0.78, p < 0.001
AUC 0.89, p < 0.001
[100]
70 patients with aICAS (≥50%)
65 patients with non-obstructive carotid plaques
miR-455DownNeuroprotective, anti-inflammatory, inhibits apoptosis and the migration of VSMCs, lowers ox-LDLFor 50% ICAS or greater:
AUC 0.927, p < 0.001
Risk factor for IS:
0.128 (0.029–0.573), p = 0.007
[61]
67 patients with sICAS (≥50%)
27 patients with aICAS (≥50%)
miR-124
miR-133a
miR-134
Up
Down
Up
associated with inhibited proliferation
of VSMCs and colagen syntesis, increased apoptosis
The expression levels of miR-124 (p = 0.036) and miR-134 (p = 0.02) were upregulated in sICAS, whereas miR-133a level was down-regulated (p = 0.043) [83]
65 patients with sICAS (≥50%)
27 patients with aICAS (≥50%)
miR-133bUpthe incidence of IS at 3 y.risk of IS:
2.25 (1.01–5.02), p = 0.047
[83]
60 patients with aICAS (≥50%)miR-130a-3pUpa proangiogenic miRNAFor ICAS progression:
5.4 (1.03–28.2); p = 0.025
[121]
97 patients with aICAS (≥50%)
90 healthy individuals
miR-637Downvascular-related diseases such as atherosclerosis, ischemic stroke, and hypertensionFor 50% ICAS or greater:
0.050 (0.014–0.174), p < 0.001
[122]
97 patients with aICAS (≥50%)
90 healthy controls
miR-637Downthe incidence of IS at 5 y.risk of IS:
0.073 (0.017–0.313), p < 0.001
[122]
91 patients with aICAS (≥50%)miR-486-5pDownassociation with the cardiovascular risk score; prediction of ICAS progressionFor 50% ICAS or greater:
AUC 0.921, p < 0.001
[123]
21 patients with sICAS (≥50%)
23 patients with aICAS (≥50%)
miR-92a
miR-126
miR-143
miR-145
miR-155
miR-210
miR-221
miR-222
miR-342-3p
Up
NS
NS
NS
NS
NS
NS
NS
NS
miRs potentially associated with atherosclerosisThe level of miR-92a was higher in sICAS compared to aICAS in T-student test (p = 0.046), but failed in multivariate analysis
no statistical difference for the other miRs
[124]
22 patients with sICAS (≥50%)
36 patients with aICAS
miR-638DownmiR expressed in VSMCs, proliferative vascular diseases, the incidence of IS at 5 y.Risk of IS:
AUC 0.66, p = 0.04
[125]
58 patients with aICAS (≥50%)
61 healthy controls
miR-106b-5pUpmiR as independent factor for the high degree of ICASFor 50% ICAS or greater:
6.582 (1.549–27.963), p = 0.011
[126]
58 patients with aICAS (≥50%)
61 healthy controls
miR-106b-5pUprole in the regulatory network of atherosclerotic gene expression, the incidence of IS at 5 y.Risk of IS:
5.431 (1.592–18.520), p = 0.007
[126]
128 patients with aICAS (≥50%)
76 healthy controls
miR-483-5pUpinhibitory effect on angiogenesis
the incidence of IS at 5 y.
risk of IS:
2.670 (1.099–6.484), p = 0.030
[127]
92 patients with aICAS (≥50%)
86 healthy controls
miR-342-5pUpthe incidence of IS at 5 y.risk of IS:
5.512 (1.370–22.176), p = 0.016
[128]
122 patients with aICAS (≥50%)
67 healthy controls
miR-92aUpplaque instability
the incidence of IS at 5 y.
risk of IS:
2.971 (1.230–7.173), p = 0.015
[129]
101 patients with aICAS (≥50%)
98 healthy controls
miR-19a-3pUpinflammation and apoptosis
the incidence of IS at 5 y.
risk of IS:
8.507 (2.239–32.328), p = 0.002
[130]
71 patients with aICAS (≥50%)
58 healthy controls
miR-27bUphigher expression in atherosclerosis
the incidence of IS at 5 y.
risk of IS:  
5.067 (1.170–21.943), p = 0.030
[131]
105 patients with aICAS (≥50%)
101 healthy controls
miR-206DownmiR correlation with ICAS degree
the incidence of IS at 5 y.
For 50% ICAS or greater:
0.336 (0.132–0.857), p = 0.022
risk of IS:
0.046 (0.005–0.431), p = 0.007
[132]
88 patients with aICAS (≥50%)
86 healthy controls
miR-9-5pDowndevelopment of atherosclerosis
the incidence of IS at 5 y.
risk of IS:
0.239 (0.087–0.652), p = 0.005
[133]
37 diabetic patients with sICAS (≥50%)
64 non-diabetic patients with sICAS (≥50%)
miR-134
miR-16
Up
Up
CVD/MI/re-IS in diabetic vs. non-diabetic patients at 7 y.miR-134 associated with risk in DM: 1.12 (1.05–1.21), p < 0.001
miR-16 associated with risk in non-DM: 1.01 (1.001–1.012), p = 0.016
[134]
167 patients with sICAS (≥50%)
66 patients with aICAS (≥50%)
157 healthy controls
miR-21
miR-221
Up
Down
plaque instability predictors
the incidence of IS at 5 y.
risk of IS:
miR-21: HR: 6.2, p < 0.001
miR-221: HR: 10.4, p < 0.001
[135]
73 patients with features of stable plaque on US
87 vulnerable plaques on US
miR-124Upplaque instability predictorAssociated with vulnerability
AUC 0.853, p < 0.001
[136]
aICAS: asymptomatic internal carotid artery stenosis; AUC: area under the curve; CI: confidence interval; CVD/MI/re-IS: cardiovascular death/myocardial infarction/recurrent ischemic stroke; HR: hazard ratio; NS: non-significant; OR: odds ratio; sICAS: symptomatic internal carotid artery stenosis; US: ultrasonography; y.: years.
Table 3. Intra-plaque microRNAs associated with carotid plaque vulnerability and rupture.
Table 3. Intra-plaque microRNAs associated with carotid plaque vulnerability and rupture.
Study GroupsmicroRNADown- vs.
Up-Regulated
Rationale for Use of Individual microRNAStatistical AnalysisReference
15 plaques from sICAS
15 plaques from aICAS
miR-100,
miR-125a,
miR-127,
miR-133a,
miR-145,
miR-221
Up
Up
Up
Up
Up
Up
microRNAs involved in plaque growth and instability
Difference between sICAS vs. aICAS:
p < 0.001
p < 0.001
p < 0.001
p < 0.001
p < 0.001
p < 0.001
[22]
22 plaques from sICAS
31 plaques from aICAS
miR-145
miR-133a
Uplarge-scale analysisDifference between sICAS vs. aICAS:
p = 0.027
p = 0.044
[97]
45 plaques from sICAS
31 plaques from aICAS
miR-221
miR-222
Down
Down
miRs promoting VSMC proliferation and intimal thickeningTime to neurological symptoms onset:
R2 = 0.44, p < 0.001
R2 = 0.45, p < 0.001
[143]
12 plaques from sICAS
10 plaques from aICAS
miR-200cUpinduction of endothelial dysfunction, ROS productionDifference between sICAS vs. aICAS:
p < 0.001
[144]
20 plaques from sICAS
Qualified as stable vs. unstable in histological assessment
miR-330-5pUpregulatory effect of miRNA-330-5p on Talin-1 mediatorDifference between sICAS vs. aICAS:
p < 0.05
[145]
aICAS: asymptomatic carotid artery stenosis; sICAS: symptomatic carotid artery stenosis; ROS: reactive oxygen species; CEA: carotid endarterectomy; VSMC: vascular smooth muscle cells; TNF-α: tumor necrosis factor alpha; IL-1β: interleukin 1-beta; AUC: area under the curve; CI: confidence interval.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Badacz, R.; Przewłocki, T.; Legutko, J.; Żmudka, K.; Kabłak-Ziembicka, A. microRNAs Associated with Carotid Plaque Development and Vulnerability: The Clinician’s Perspective. Int. J. Mol. Sci. 2022, 23, 15645. https://doi.org/10.3390/ijms232415645

AMA Style

Badacz R, Przewłocki T, Legutko J, Żmudka K, Kabłak-Ziembicka A. microRNAs Associated with Carotid Plaque Development and Vulnerability: The Clinician’s Perspective. International Journal of Molecular Sciences. 2022; 23(24):15645. https://doi.org/10.3390/ijms232415645

Chicago/Turabian Style

Badacz, Rafał, Tadeusz Przewłocki, Jacek Legutko, Krzysztof Żmudka, and Anna Kabłak-Ziembicka. 2022. "microRNAs Associated with Carotid Plaque Development and Vulnerability: The Clinician’s Perspective" International Journal of Molecular Sciences 23, no. 24: 15645. https://doi.org/10.3390/ijms232415645

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