Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 May 18.
Published in final edited form as: Adv Exp Med Biol. 2015;887:53–77. doi: 10.1007/978-3-319-22380-3_4

microRNAs Distinctively Regulate Vascular Smooth Muscle and Endothelial Cells: Functional Implications in Angiogenesis, Atherosclerosis, and In-Stent Restenosis

Gaetano Santulli 1
PMCID: PMC4871245  NIHMSID: NIHMS785782  PMID: 26662986

Abstract

Endothelial cells (EC) and vascular smooth muscle cells (VSMC) are the main cell types within the vasculature. We describe here how microRNAs (miRs)—noncoding RNAs that can regulate gene expression via translational repression and/or post-transcriptional degradation—distinctively modulate EC and VSMC function in physiology and disease. In particular, the specific roles of miR-126 and miR-143/145, master regulators of EC and VSMC function, respectively, are deeply explored. We also describe the mechanistic role of miRs in the regulation of the pathophysiology of key cardiovascular processes including angiogenesis, atherosclerosis, and in-stent restenosis post-angioplasty. Drawbacks of currently available therapeutic options are discussed, pointing at the challenges and potential clinical opportunities provided by miR-based treatments.

Keywords: Endothelium, Restenosis, VSMC, Neointima, Stent, DES, Circulation, Thrombosis, miR-126, miR-143/145, Diabetes, Myocardin, Myoregulin, Atherosclerosis, Angiogenesis, Inflammation, Angioplasty

Introduction

Endothelial cells (EC) and vascular smooth muscle cells (VSMC) constitute the main cell types within the vasculature [1, 2]. The endothelium forms the inner thin layer that represents an interface between circulating fluid in the lumen and the rest of the vessel wall. The endothelial monolayer lines the entire circulatory system, from the smallest capillaries to the heart [3]. The main functions of EC include regulation of vascular tone, fluid filtration, neutrophil recruitment, hormone trafficking, and hemostasis [46].

VSMC are able to contract or relax in response to various stimuli, and are thereby responsible for the redistribution of the blood within the body to areas where it is needed, including tissues with temporarily enhanced oxygen consumption [711]. Their main function is to regulate the caliber of the blood vessels: excessive vasoconstriction might lead to hypertension, whereas excessive vasodilation can induce hypotension (as in shock) [12, 13].

Coding or Noncoding: Whether “Tis Nobler in the Mind to Suffer…”

MicroRNAs (miRs) are small, generally noncoding RNAs, that regulate gene expression via post-transcriptional degradation or translational repression [14]. Unquestionably, miRs are fundamental regulators of numerous biological processes. See Chaps. 1 and 2 of this book for more detailed information on biological aspects of miRs.

More than 30,000 mature miR products have been identified (~200 in the human genome), and the number of published miR sequences continues to increase rapidly [15].

Importantly, some transcripts that originally appeared to be lncRNAs may actually encode micropeptides. For instance, a conserved micropeptide, named myoregulin, encoded by RNA that had been misannotated as noncoding, has been recently identified by Eric Olson [16]. The fact that putative noncoding RNA may actually harbor hidden micropeptides had been recently suggested by genome-wide analyses [17].

Consequently, scanning the microproteome embedded in “noncoding RNA,” whose coding capacity has been somehow ignored in gene annotations [18, 19], will certainly represent a main field of research in the next future, revisiting previously established annotations of noncoding RNAs and hopefully leading to the discovery of new mechanistically important micropeptides.

Fine Tuning of Vascular Biology via MicroRNA

Vascular endothelium plays a key role in regulating) vessel biology and homeostasis. Alteration of its function can be the basis of cardiovascular disorders including hypertension, atherosclerosis, and impairment of the angiogenetic processes [2022]. Several events are involved in angiogenesis, including EC division, selective degradation of the basal membrane, and the surrounding extracellular matrix, with the subsequent EC migration with the formation of neovessels [7].

Similar events take place during embryogenesis, however in this context blood vessels are generated from the differentiation of EC precursors, the angioblasts, which associate to eventually form primitive vessels in a process known as vasculogenesis [23]. Endothelial maturation is finely guided by signals from other cell types [23]. Indeed, development of the vascular architecture involves the strict association between EC and mural cells, including VSMC and pericytes [24]. Under physiological conditions, the communication between these cell types leads to the maturation and stabilization of the vessel. These processes involve the action of different growth factors and heterotypic cellular interactions [25].

miR-143/145 vs. miR-126, Master Regulators of VSMC and EC Function

miR-143/145

A major advance in the smooth muscle) biology field occurred with the report of the essential role of a bicistrionic unit encoding for miR-143 and miR-145 in the regulation of smooth muscle physiology [2641]. The miR-143/145 gene is located on chromosome 5 in humans and chromosome 18 in mice and rats. miR-143 and miR-145 are distinct in sequence, yet transcribed together as one primary cluster. Being controlled by serum response factor (together with myocardin) and Nkx2.5, expression of miR-143/145 seems to occur primarily in VSMCs and is critical for maintaining their contractile phenotype [2, 26]. Its expression pattern correlates with a differentiated phenotype as defined by alpha-smooth muscle actin, smooth muscle myosin heavy chain, and calponin expression [39, 42, 43].

The joint suppression of specific target mRNAs by miR-143/145 contributes to a contractile phenotype, as demonstrated in mice with genetic deficiency of miR-143/145, which display reduced vascular tone and blood pressure control [44]. miR-143/145−/− mice also have a tendency to develop neointimal lesions. Aortic VSMCs from the double knockout mouse lack myofilamentous cytoskeletal organization and have diffuse actin staining. A proteomic analysis of aortas from global miR-143/145−/− mice revealed angiotensin-converting enzyme (ACE-1) as a target of the miR-143/145 cluster. Strikingly, pharmacological inhibition by ACE inhibitors or angiotensin 1 receptor blockers partially reverses vascular dysfunction normalizing gene expression in miR-143/145−/− mice [45].

At the ultrastructural electron microscopic analysis, miR-143/145-deficient VSMC exhibit show greater presence of rough endoplasmic reticulum and less focal adhesions compared with wild-type cells. These differences have been associated with impaired contractility of femoral artery rings in response to angiotensin II and phenylephrine. Of note, the functional relevance of miR-143/145 in human vessel pathology had been suggested by the observed downregulation of the miR-143/145 cluster in human aortic aneurysms compared with normal aortic tissue [46].

As mentioned above, the cardiac muscle- and smooth muscle-specific transcriptional factor myocardin has been shown to up-regulate miR-143/145 expression [47, 48]. Similarly, the introduction of miR-143/145 potentiates myocardin-induced smooth muscle marker gene expression, calcium fluxes, and myofilament formation in response to endothelin-1. The repressor of myocardin-directed smooth muscle gene expression KLF4, which binds myocardin, has been identified as a potential target of miR-145 in murine VSMC [29]. However, human KLF4 mRNA contains no binding site for miR-143 and only a low-probability site for miR-145, whereas KLF5 (which has protein sequences highly conserved across human, mouse, and rat) contains a binding site for miR-145. In vivo experiments revealed that vascular injury causes downregulation of miR-145, which up-regulates KLF5 and inhibits myocardin, leading to VSMC dedifferentiation and increased neointimal lesion formation [26, 49]. Notably, PDGF receptor α, fascin, and protein kinase C ε, essential regulators of podosome formation, were revealed as direct targets of miR-143 and miR-145 [50].

Various reports revealed that cells can release miRs which can then exert their specific effects by modulating processes in recipient cells. Thus, miRs have intercellular signal transduction capabilities. Most recently, Climent and colleagues elegantly demonstrated that VSMC communicate with EC via miR-143 and miR-145: cell-to-cell VSMC/EC contacts induce the activation of miR-143/145 transcription in VSMC, promoting the transfer of these miRNAs to the endothelium [51]. In particular, VSMC can deliver miR-143/145 to EC via fine intercellular tubes, named membrane nanotubes or tunneling nanotubes [51]. Indeed, the level of miR-143/145, but not that of its precursor molecule (pri-miR-143/145), rose substantially in EC when these cells were cultured together with VSMC. A specific molecular pathway has been elucidated, in which secretion of transforming growth factor-β by EC stimulates the transfer of miR-143/145 from VSMC to EC, where VSMC-derived miR-143/145 represses hexokinase II and integrin β8 and thereby the angiogenic potential of the recipient cell [51]. Notably, the expression of miR-143/145 in EC could not be achieved by the simple transfer of conditioned medium or VSMC-derived exosomes and was not sensitive to gap junction uncoupling agents (both exosomes and gap junctions had been reported as potential routes for intercellular transfer of miRs). Instead, the transfer of miR-143/145 was sensitive to latrunculin A, an inhibitor of the formation of tunneling nanotubes, tiny membrane connections that cultured cells form among each other. The intercellular transfer of miRs through tunneling nanotubes had been previously reported in ovarian cancer [52]. High-resolution imaging allowed the direct visualization of tunneling nanotubes between EC and VSMC and the transport of miRs within them [51].

miR-126

miR-126 is encoded by) intron 7 of the EGF-like domain 7 (EGFL7) gene, a.k.a. vascular endothelial-statin (VE-statin), which is under the transcriptional control of the E-26 family of transcription factors ETS1/2 [5365]. In resting conditions, ETS1 is expressed at a very low level whereas during angiogenesis or re-endothelialization, it is transiently expressed at high levels. During replicative senescence an increased expression of ETS1 could induce the increasing of miR-126 expression. Interestingly, one of the main targets of mir-126 is the host gene EGFL7, which regulates the proper spatial organization of ECs within each sprout and influences their collective movement. The cardiovascular phenotype of EGFL7-deficient mice is recapitulated by the ablation of miR-126, causing ruptured blood vessels, multifocal hemorrhages, and systemic edema (~40 % of mir-126 KO mice die embryonically) [57].

miR-126 has been extensively studied in plasma and circulating cells because its expression is very high in EC [2], endothelial progenitor cells (EPCs), and platelets [59, 6680]. Most recently, miR-126 has been identified as an efficient marker in the detection and purification of EC [81]. This miR plays a critical role in modulating vascular development and homeostasis, targeting specific mRNAs including the Sprouty-related protein 1 (SPRED-1), CXCL12, SDF-1, and phosphoinositol-3 kinase regulatory subunit 2 (PIK3R2) [8287]. Confirming its key role in maintaining vascular integrity, amidst the other targets of miR-126 there is a key mediator of leukocyte adhesion and inflammation: vascular cell adhesion molecule 1 (VCAM-1). miR-126 has also been related with the endothelial dysfunction associated with the development of diabetes and its complications [88].

Circulating miR-126 acts as an intercellular messenger mainly released by EC and internalized primarily by monocytes and VSMC [57]. Its transfer modulation may be an important strategy to prevent or delay endothelial dysfunction [8892]. A significant increase in circulating miR-126 has been detected in patients with acute myocardial infarction and angina whereas miR-126 downregulation has been reported in plasma from patients with diabetes, heart failure, or cancer [14]. Hence, circulating miR-126 can be modulated by diverse stimuli inducing dissimilar cellular fates in different cell types [60, 85, 9397]. Most recently, Italian researchers demonstrated that circulating miR-126-3p (see Chap. 1 for miR nomenclature) can be considered a biomarker of physiological endothelial senescence in normoglycemic elderly subjects and appears to underlie a mechanism that may be disrupted in aged diabetic patients [98]. Intriguingly, diabetes mellitus leads to dysregulated activation of ETS, which blocks the functional activity of progenitor cells and their commitment toward the endothelial cell lineage [99].

The Phenotypic Switch of Vascular Smooth Muscle Cells: A Key Event in Atherosclerosis and Restenosis

VSMC can modulate their phenotype) in response to the environmental stimuli via a process characterized by decreased gene expression of VSMC contractile markers and increased proliferation, migration, and matrix synthesis [100]. Such a phenotypic switch represents one of the main cellular events underlying various VSMC-related pathological conditions, including hypertension, atherosclerosis, post-angioplasty restenosis, and angiogenesis. Unraveling the mechanisms involved in VSMC phenotypic switch is an important step toward better understanding the pathophysiology of these disorders and ultimately designing therapeutic agents for their treatment and prevention.

The switch between the contractile (differentiated, quiescent) and synthetic (dedifferentiated, proliferative) VSMC phenotypes are orchestrated via a synergistic molecular regulatory network. Overall, contractile smooth muscle proteins contain an evolutionarily conserved cis-elements, the CArG box (CC(A/T)6GG) in the promoter–enhancer region of the gene. This has been demonstrated for α-smooth muscle actin, desmin, smooth muscle myosin heavy chain, SM22-α, and calponin.

The serum response factor (SRF), a ubiquitously expressed transcription factor, binds the CArG box driving transcription. The interaction between SRF and CArG boxes in response to environmental changes is generally coordinated by the interaction of SRF with additional transcription factors, divided in co-activators (myocardin, PRX1, and GATA factors) and co-repressors (ELK-1, KLF4, YY1, and Foxo factors).

Virtually all of the VSMC-specific contractile protein genes alongside with many other genes involved in migration, proliferation, and extracellular matrix production during the process of VSMC phenotypic switch, containing CArG boxes within their promoters. The balance between those positive and negative SRF cofactors finely regulates the dynamic VSMC gene expression in response to environmental signals. Amidst these regulatory components, myocardin, which binds SRF in a 1:2 stoichiometric ratio inducing CArG-dependent VSMC gene transcription, is widely considered to be the master gene regulating VSMC differentiation.

The TGF-β and bone morphogenetic proteins (BMPs) also contribute to VSMC phenotypic switch by promoting VSMC contractility enhancing the expression of the contractile apparatus [101]. In particular, TGF-β superfamily of growth factors triggers VSMC differentiation by post-transcriptionally increasing the expression of a subset of miRs including miR-21. Treatment of VSMC with TGF-β and BMP4 results in change in the expression of miRs, by finely regulating DROSHA complex [102].

Intriguingly, the TGF-β signaling pathway is also under the control of miRs. Other studies demonstrated that the inhibition of miR-26a promote VSMC apoptosis and phenotypic switch to a contractile status while inhibiting proliferation and migration [103]. Intriguingly, miR-24 is involved into both TGF-β and PDGF-BB signaling pathways, which respectively represent the distinguishing trigger of VSMC differentiation and proliferation [104]: PDGF-BB induces miR-24 expression, which subsequently leads to Tribbles-like protein-3 (TRB3) downregulation by a post-transcriptional effect, with a decrease in BMP and TGFb signaling, promoting VSMC proliferation. On the contrary, inhibition of miR-24 enhances TRB3 expression and impairs VSMC proliferative activity [104]. Of note, TGF-β Signaling has been also implied in the mineralization of VSMC induced by oxidized LDL [105].

The specific roles of miRs in the regulation of VSMC phenotypic switch are summarized in Table 4.1 .

Table 4.1.

miRNAs involved in the VSMC phenotypic switch

Effect of injury on
miRNA level
Main targets References
Pro-contractile miRNAs (quiescent, differentiated state)
miR-1 Reduction PIM1, KLF4 [106, 107]
miR-10 Unknown HDAC4, MAP3K7, β-TRC [108, 109]
miR-15b Increase MAP2K4 [110]
miR-18a-5p Increase Syndecan4 [111]
miR-21 Increase PDCD4 [112, 113]
miR-132 Increase LRRFIP1 [114]
miR-133a Reduction MSN, SP-1 [115, 116]
miR-143/145 Reduction FRA1, Elk1, CALMK, KLF4,
ACE, KLF5, MRTFB, myocardin
[26, 42, 49]
miR-365 Reduction Cyclin D1 [117]
miR-424/322 Increase STIM1, calumenin [118]
miR-490-3p Reduction PAPP-A [119]
miR-638 Unknown NOR1 [120]
miR-663 Unknown JunB [121, 122]
Pro-synthetic miRNAs (proliferative, dedifferentiated state)
miR-21 Increase PTEN, Bcl-2 [102, 123, 124]
miR-24 Reduction TRB3 [104, 125, 126]
miR-26a Increase SMAD1/4 [103, 127, 128]
miR-31 Increase LATS2, CREG [129131]
miR-146 Increase HuR, NF-κB [132]
miR-221/222 Increase p27, p57, c-kit [102, 133135]

βTRC β transducin repeat-containing gene, CREG cellular repressor of E1A-stimulated genes, Elk1 ETS domain-containing protein-1, KLF4 Kruppel-like factor 4, Trb3 Tribbles-like protein-3, LATS2 large tumor suppressor homolog 2, LRRFIP1 leucine-rich repeat in Flightless-1 interacting protein-1, MAP2K4 mitogen-activated protein kinase kinase 4, MAP3K7 mitogen-activated kinase kinase kinase 7, MSN moesin, NOR1 orphan nuclear receptor, PAPP-A pregnancy-associated plasma protein A, PDCD4 Programmed Cell Death 4, SMAD small mother against decapentaplegic, STIM1 stromal-interacting molecule 1, TRB3 Tribbles-like protein-3

Several miRs have been shown to modulate VSMC proliferation and migration, key aspects in the pathogenesis of atherosclerosis. For instance, miR-133 [136] and miR-136 [137] promote whereas miR-365 [117] and miR-1298 (which has been shown to be regulated by DNA methylation [138]) inhibit VSMC proliferation and migration.

Importantly, the role of VSMC varies depending on the stage of atherosclerotic disease, playing a maladaptive role in early lesion development and progression and a beneficial adaptive role within the fibrous cap by stabilizing advanced plaques in the face of end-stage disease events such as plaque rupture [139]. Indeed, VSMC can synthesize components of the fibrous cap, which separates circulating blood from the thrombogenic plaque interior [140]. In humans, ruptured atherosclerotic plaques show less VSMC compared with stable lesions, indicating an active contribution of VSMC to plaque stability [139]. Promoting a quiescent VSMC phenotype might lead to increased fibrous cap integrity, and in this respect, miRs represent an opportunity for positive VSMC regulation (see Chap. 5 of this book for a systematic overview of miRs involved in the atherosclerotic process).

Mechanistic Importance of Endothelium in Restenosis and in Vascular Remodeling

Stent implantation and/or balloon angioplasty, interventions that are used routinely to treat coronary artery disease, lead to mechanical EC damage. A normal operative endothelium is crucial due to its participation in the regulation of vascular tone, alongside with its role in suppressing intimal hyperplasia by inhibiting inflammation, thrombus formation, and VSMC proliferation and migration [2, 141, 142]. Thus, the endothelium provides a selectively permeable barrier that protects against potentially detrimental circulating factors [143145]. Endothelial denudation and medial wall injury are generally considered the initial effects of angioplasty-induced injury [146149].

Given the essential role of EC in suppressing inflammation and thrombosis [150, 151] and overall in controlling vascular tone and function [4], the restoration of a healthy endothelial layer is an imperative therapeutic goal in order to prevent restenosis and to avoid the detrimental consequences of in-stent thrombosis [2, 152].

Re-endothelialization of injured arteries occurs naturally via outgrowth of local EC [153]. The recruitment of circulating bone marrow-derived endothelial progenitor cells in this process is controversial [154156], and the actual contribution of this cell population continues to be uncertain [157].

The effect of stent deployment on EC behavior remains poorly understood. Unquestionably, re-endothelialization of injured coronary arteries is affected by the presence of a stent since such a structure provides a nonphysiological surface for adhesion and generates perturbations in blood flow [158, 159].

The issue of EC repair has been somehow brought into sharp relief in the era of drug-eluting stents (DES), which can release cytostatic compounds that inhibit cell cycle progression [160, 161]. Albeit DES are associated with reduced restenosis rates via inhibition of VSMC proliferation [162164], they have also been linked to lethal late-stage thrombotic events, which are associated with EC injury [165167]. Ergo, there is an urgent need to develop new therapeutic interventions to promote EC repair in stented arteries and thereby reduce the incidence of late thrombosis and avoid serious risks associated with prolonged administration of systemic antiplatelet treatments [168, 169], as discussed in detail in the section “ Angioplasty, Stents, and miRs ” of this chapter.

The precise mechanisms of endothelial repair following angioplasty-related injury have been the focus of a plethora of studies. As mentioned above, the potential regenerative capacity of endothelial progenitor cells remains controversial [170172], and current research focuses on the complex interaction of circulating cells and mature vessel-wall residual EC. In this context, the emerging functional role of microparticles, tiny membrane fragments of activated and apoptotic cells, has been recently investigated: in brief, EC injury triggers the release of EC-derived microparticles, which act as important carriers of bioactive molecules playing crucial roles in cell–cell cross talk. Indeed, microparticles can trigger antiapoptotic effects on EC, and are able to transfer microRNAs, such as miR-126, to target EC, ultimately enhancing endothelial repair mechanisms [170].

Under hyperglycemic conditions, EC microparticles exhibit reduced regenerative capacity, suggesting that hyperglycemia not only directly harms the endothelium, but also indirectly promotes vascular damage by altering endogenous vascular regeneration mechanisms [170]. Analysis of microRNA-126 level in patients with stable coronary artery disease confirmed that diabetes mellitus reduces microRNA-126 expression in circulating microparticles. Moreover, genetic downregulation of microRNA-126 reduces endothelial microparticle-mediated EC repair both in vivo and in vitro [170, 173].

The endothelium plays a fundamental role in angiogenesis [2022, 25, 174] and numerous studies investigated the role of miRs in regulating this process. For simplicity, the most important miRs heretofore implicated in EC-mediated angiogenetic process are reported in Table 4.2, alongside with their target gene(s) and function(s).

Table 4.2.

Proangiogenic and antiangiogenic miRNAs

Targets Main effect References
Pro-angiogenic miRNAs
miR-9 SOCS5 Enhanced EC migration [175]
miR-10a MAP3K7; HOXA1;
bTRC
Anti-inflammatory [109, 176, 177]
miR-23/27 SEMA6A; SPROUTY2 Pro-angiogenic [178]
miR-126 PI3KR2; SPRED1 Pro-angiogenic [57, 89, 98,
179182]
VCAM1; SDF1 Anti-inflammatory
miR-130 HOXA5, GAX Migration and
proliferation
miR-210 EFNA3 Pro-angiogenic [183]
miR-217 SIRT1-FOXO/eNOS Pro-angiogenic [184]
miR-424 HIF-1α Chemotaxis,
proliferation
[185]
Anti-angiogenic miRNAs
miR-17 JAK-1 Anti-angiogenic [186]
miR-21 RhoB Antiproliferative [187]
PPARγ Pro-inflammatory [188]
miR-24 GATA-2; Anti-angiogenic [189]
PAK4 Pro-apoptotic [190]
miR-92a SIRT1; ITGA5 Anti-angiogenic [191]
KLF4 and MKK4 Inhibited
re-endothelialization
[192]
miR-200 Ets-1; IL-8; CXCL1 Anti-angiogenic [193, 194]
miR-221/222 STA5a; c-KIT; eNOS Anti-angiogenic [195197]
miR-492 eNOS Anti-angiogenic [198]
14q32 miR cluster
(329, 487b, 494, 495)
Multiple
neovascularization genes
Anti-angiogenic [199]

EFNA3 Ephrin-A3, eNOS Endothelial Nitric Oxide Synthase, JAK1 Janus kinase 1, KLF4 Kruppel-like factor 4, PPAR-γ Peroxisome proliferator-activated receptor gamma, RhoB Ras homolog gene family, member B, SIRT1 sirtuin (silent mating type information regulation 2 homolog) 1, SOCS5 suppressor of cytokine signaling 5, PI3KR2 phosphoinositol-3 kinase regulatory subunit 2, SEMA6A Semaphorin-6A, VCAM-1 vascular cell adhesion molecule 1

Angioplasty, Stents, and miRs

Coronary artery disease represents a leading cause of mortality worldwide [200203]. Percutaneous coronary intervention (PCI) is one of the most commonly performed interventions [204], representing the main option for revascularization in a series of cardiovascular disorders, from unstable angina and myocardial infarction, to multivascular disease [204].

Milestones in Interventional Cardiology

Recurrent lumen narrowing has been a substantial limitation of open and percutaneous methods of arterial reconstruction from their inception.

The term restenosis, commonly used since 1950 in reference to recurrent cardiac valvular stenosis [205], was later adopted to define lumen re-narrowing after open arterial reconstruction such as carotid endarterectomy.

With the development of peripheral angioplasty by Dotter in the 1960s [206] and then the application of percutaneous approach to angioplasty of renal, coronary, and iliac arteries by Gruentzig and colleagues [207, 208] the problem of restenosis grew exponentially: as angioplasty became widely adopted in the 1980s, restenosis after the intervention was reported in up to 60 % of patients [209, 210].

A major breakthrough in the field was the introduction of bare metal stents (BMS), which revolutionized interventional cardiology preventing the elastic recoil of the treated vessels [211] and, more importantly, significantly reducing the phenomenon of restenosis, as demonstrated by two milestone prospective randomized clinical trials [212, 213]: compared with angioplasty, stenting massively reduced restenosis, reaching percentages of 22 %.

However, the major drawback of this procedure is the induction of proliferation/migration and subsequent accumulation of VSMC [214], macrophages [215], and lymphocytes [216] in the arterial wall, eventually leading to restenosis [217].

Once the molecular mechanisms underlying the restenosis process were better comprehended (and mainly attributable to the proliferation of VSMC), DES were introduced in the clinical scenario, in order to deliver in situ a drug that can inhibit cell proliferation [218, 219].

Nowadays, millions of procedures to intervene on occlusive vascular lesions are performed worldwide each year (~700,000 angioplasties are performed annually in the USA) and 70–90 % of all angioplasty patients receive a stent, inserted permanently at the site of the vascular blockage to form an internal scaffolding that keeps the angioplastied vessel from closing.

The introduction of DES significantly reduced rates of restenosis [218, 220, 221]. Though, concerns have been raised over the long-term safety of the DES, with particular reference to stent thrombosis, essentially due to impaired re-endothelization caused by the nonselective antiproliferative properties of DES [222224]. As such, when the obstacle of restenosis seemed finally overcome, enthusiasm and euphoria were tempered by epidemiologic data reporting that DES did not ameliorate mortality rates when compared to BMS [225]. Basic research revealed that the essential cause of the increased mortality observed in patients receiving DES, despite the achieved prevention of restenosis, was mainly attributable to the fact that the antiproliferative drugs eluted by the stents were nonselective, inhibiting not only the proliferation and migration of the “bad” cells responsible for restenosis (VSMC), but also the growth and mobility of the “good” EC, utterly indispensable for the healing of the vessel following the stent implantation [226]. The lack of endothelial coverage eventually leads to an increased risk of thrombosis, with catastrophic clinical consequences for the patients.

Drugs eluted by the stents currently available in clinical practice (Fig. 4.1) are not able to differentiate EC from VSMC, T-cells or macrophages [227229], and the inhibition of proliferation and migration affects all these cellular types [228, 230, 231], leading to an increased risk for late thrombosis, due to delayed/incomplete re-endothelization [204, 214, 232]. Thus, impaired endothelial coverage after angioplasty prolongs the window of vulnerability to thrombosis, requiring thereby a prolonged dual antiplatelet therapy.

Fig. 4.1.

Fig. 4.1

Chemical modifications of the macrocyclic ring in order to obtain drugs with potent antiproliferative and immunosuppressive effects, including rapamycin (sirolimus), everolimus, zotarolimus, umirolimus (biolimus A9), novolimus, and myolimus. Purple: FKBP12 (a.k.a. Calstabin1) binding site; Green: mTOR binding site

Several vasculoprotective approaches have been proposed to overcome the restenosis problem after angioplasty, preserving endothelial function [233235]. However, vascular restenosis and thrombosis continue to be a major problem of interventional cardiology, limiting the effectiveness of revascularization procedures. The ideal eluting stent should display a selective antiproliferative effect on VSMC, macrophages, and T-lymphocytes, without affecting EC [204].

Since EC injury is a fundamental element in the pathophysiology of atherogenesis, understanding EC repair is of critical importance in order to develop novel therapeutic strategies to preserve endothelial integrity and vascular health. In this sense, miRs and their intrinsic properties represent a wonderful opportunity to obtain a specific attenuation of neointimal formation.

Several miRs have been implicated in modification of vessel restenosis after interventional endothelial injury: Antisense knockdown of miR-21, which is moderately increased after vessel injury [123], can blunt the formation of neointimal lesions in response to balloon injury of the carotid artery. Inducing miR-221 in VSMCs on PDGF-β stimulation causes p27Kip1 inhibition [236], thereby increasing VSMC proliferation contributing to the formation of a neointimal lesion after arterial injury. Overexpression of miR-145 promotes neointima formation in response to balloon injury [237].

The fundamental role of miRs in the restenosis process has been also confirmed by Baker and colleagues, who identified multiple miRs, including miR-21, miR-146, and miR142-3p, aberrantly expressed in stented pig arteries. Using a mouse vascular stent model, they demonstrate that the knockout of miR-21 can attenuate neointimal formation post-stenting modulating inflammation and VSMC response [238].

Harnessing the EC-specific expression of miR-126, we were able to obtain in one fell swoop both the inhibition of restenosis, attacking VSMC, and the prevention of restenosis and thrombosis, preserving the endothelial function [2].

A major challenge in the field is the delivery of miR-based therapies. In preclinical studies, antagomiRs and miR mimetics have been successfully [239243] delivered systemically (intravenously injected) showing beneficial effects on cardiac remodeling; however, they are preferentially targeted to the liver, spleen, and kidney. The specific application of miR-based agents to the vasculature, for instance during cardiac catheterization for angioplasty, can be considered as an effective therapeutic strategy.

Potential alternatives [93, 244250] to the direct intravascular delivery (which ideally could be combined to the new generation bioresorbable stents with biodegradable scaffolds [251255]) include the stabilization of the miR-based agent (various chemical modifications of the nucleotides can enhance stability in vivo, for instance by using cholesterol-conjugated, 2′-O-methyl-modified antagomiRs) or the conjugation to targeting molecules including antibodies, peptides, or other bioactive molecules, which may promote the specific homing of the miR-based drug to the site of the injury.

Conclusion

Accumulating evidence establishes that miRs are becoming one of the most fascinating areas of biology, given their fundamental roles in several pathophysiological processes. The relative role of different miRs in vascular biology as direct or indirect post-transcriptional regulators of genes implied in structural remodeling, inflammation, angiogenesis, atherosclerosis, in-stent restenosis, and thrombosis indicate that miRs may serve as promising drug targets or potential biomarkers in prevention and management of vascular disorders.

Acknowledgements

Dr. Gaetano Santulli is supported by the National Institutes of Health (K99DK107895) and by the American Heart Association (AHA 15SDG25300007).

References

  • 1.Iaccarino G, Ciccarelli M, Sorriento D, Cipolletta E, Cerullo V, Iovino GL, et al. AKT participates in endothelial dysfunction in hypertension. Circulation. 2004;109:2587–93. doi: 10.1161/01.CIR.0000129768.35536.FA. [DOI] [PubMed] [Google Scholar]
  • 2.Santulli G, Wronska A, Uryu K, Diacovo TG, Gao M, Marx SO, et al. A selective microRNA-based strategy inhibits restenosis while preserving endothelial function. J Clin Invest. 2014;124:4102–14. doi: 10.1172/JCI76069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cimpean A, Ribatti D, Raica M. A historical appraisal of angiogenesis assays since Judah Folkman and before…. In: Santulli G, editor. Angiogenesis: insights from a systematic overview. Nova Science; New York: 2013. pp. 31–50. [Google Scholar]
  • 4.Santulli G, Cipolletta E, Sorriento D, Del Giudice C, Anastasio A, Monaco S, et al. CaMK4 gene deletion induces hypertension. J Am Heart Assoc. 2012;1:e001081. doi: 10.1161/JAHA.112.001081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Santulli G, Trimarco B, Iaccarino G. G-protein-coupled receptor kinase 2 and hypertension: molecular insights and pathophysiological mechanisms. High Blood Press Cardiovasc Prev. 2013;20:5–12. doi: 10.1007/s40292-013-0001-8. [DOI] [PubMed] [Google Scholar]
  • 6.Sorriento D, Ciccarelli M, Santulli G, Campanile A, Altobelli GG, Cimini V, et al. The G-protein-coupled receptor kinase 5 inhibits NFkappaB transcriptional activity by inducing nuclear accumulation of IkappaB alpha. Proc Natl Acad Sci U S A. 2008;105:17818–23. doi: 10.1073/pnas.0804446105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Santulli G. Angiopoietin-like proteins: a comprehensive look. Front Endocrinol. 2014;5:4. doi: 10.3389/fendo.2014.00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Santulli G. β-blockers in diabetic patients with heart failure. JAMA Intern Med. 2015;175(4):657. doi: 10.1001/jamainternmed.2014.8009. [DOI] [PubMed] [Google Scholar]
  • 9.Santulli G. Effects of low-carbohydrate and low-fat diets. Ann Intern Med. 2015;162:392. doi: 10.7326/L15-5057-3. [DOI] [PubMed] [Google Scholar]
  • 10.Perino A, Ghigo A, Ferrero E, Morello F, Santulli G, Baillie GS, et al. Integrating cardiac PIP(3) and cAMP signaling through a PKA anchoring function of p110gamma. Mol Cell. 2011;42:84–95. doi: 10.1016/j.molcel.2011.01.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Santulli G. Adrenal signaling in heart failure: something more than a distant ship’s smoke on the horizon. Hypertension. 2014;63:215–6. doi: 10.1161/HYPERTENSIONAHA.113.02382. [DOI] [PubMed] [Google Scholar]
  • 12.van Lieshout JJ, Wieling W, Karemaker JM, Eckberg DL. The vasovagal response. Clin Sci. 1991;81:575–86. doi: 10.1042/cs0810575. [DOI] [PubMed] [Google Scholar]
  • 13.Santulli G, Ciccarelli M, Trimarco B, Iaccarino G. Physical activity ameliorates cardiovascular health in elderly subjects: the functional role of the beta adrenergic system. Front Physiol. 2013;4:209. doi: 10.3389/fphys.2013.00209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wronska A, Kurkowska-Jastrzebska I, Santulli G. Application of microRNAs in diagnosis and treatment of cardiovascular disease. Acta Physiol (Oxf) 2015;213:60–83. doi: 10.1111/apha.12416. [DOI] [PubMed] [Google Scholar]
  • 15.Kozomara A, Griffiths-Jones S. miRBase: annotating high confidence microRNAs using deep sequencing data. Nucleic Acids Res. 2014;42:D68–73. doi: 10.1093/nar/gkt1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Anderson DM, Anderson KM, Chang CL, Makarewich CA, Nelson BR, McAnally JR, et al. A micropeptide encoded by a putative long noncoding RNA regulates muscle performance. Cell. 2015;160:595–606. doi: 10.1016/j.cell.2015.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Andrews SJ, Rothnagel JA. Emerging evidence for functional peptides encoded by short open reading frames. Nat Rev Genet. 2014;15:193–204. doi: 10.1038/nrg3520. [DOI] [PubMed] [Google Scholar]
  • 18.Carninci P, Kasukawa T, Katayama S, Gough J, Frith MC, Maeda N, et al. The transcriptional landscape of the mammalian genome. Science. 2005;309:1559–63. doi: 10.1126/science.1112014. [DOI] [PubMed] [Google Scholar]
  • 19.Lee LJ, Hughes TR, Frey BJ. How many new genes are there? Science. 2006;311:1709–11. doi: 10.1126/science.311.5768.1709b. author reply-1711. [DOI] [PubMed] [Google Scholar]
  • 20.Santulli G, Ciccarelli M, Palumbo G, Campanile A, Galasso G, Ziaco B, et al. In vivo properties of the proangiogenic peptide QK. J Transl Med. 2009;7:41. doi: 10.1186/1479-5876-7-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ciccarelli M, Santulli G, Campanile A, Galasso G, Cervero P, Altobelli GG, et al. Endothelial alpha1-adrenoceptors regulate neo-angiogenesis. Br J Pharmacol. 2008;153:936–46. doi: 10.1038/sj.bjp.0707637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Santulli G, Basilicata MF, De Simone M, Del Giudice C, Anastasio A, Sorriento D, et al. Evaluation of the anti-angiogenic properties of the new selective alphaVbeta3 integrin antagonist RGDechiHCit. J Transl Med. 2011;9:7. doi: 10.1186/1479-5876-9-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Berthod F. Fibroblasts and endothelial cells: the basic angiogenic unit. In: Santulli G, editor. Angiogenesis: insights from a systematic overview. Nova Science; New York: 2013. pp. 145–58. [Google Scholar]
  • 24.Yamamoto S, Niida S, Azuma E, Yanagibashi T, Muramatsu M, Huang TT, et al. Inflammation-induced endothelial cell-derived extracellular vesicles modulate the cellular status of pericytes. Sci Rep. 2015;5:8505. doi: 10.1038/srep08505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Matarese A, Santulli G. Angiogenesis in chronic obstructive pulmonary disease: a translational appraisal. Transl Med UniSa. 2012;3:49–56. [PMC free article] [PubMed] [Google Scholar]
  • 26.Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, et al. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature. 2009;460:705–10. doi: 10.1038/nature08195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Iio A, Nakagawa Y, Hirata I, Naoe T, Akao Y. Identification of non-coding RNAs embracing microRNA-143/145 cluster. Mol Cancer. 2010;9:136. doi: 10.1186/1476-4598-9-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boucher JM, Peterson SM, Urs S, Zhang C, Liaw L. The miR-143/145 cluster is a novel transcriptional target of Jagged-1/Notch signaling in vascular smooth muscle cells. J Biol Chem. 2011;286:28312–21. doi: 10.1074/jbc.M111.221945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Davis-Dusenbery BN, Chan MC, Reno KE, Weisman AS, Layne MD, Lagna G, et al. Down-regulation of Kruppel-like factor-4 (KLF4) by microRNA-143/145 is critical for modulation of vascular smooth muscle cell phenotype by transforming growth factor-beta and bone morphogenetic protein 4. J Biol Chem. 2011;286:28097–110. doi: 10.1074/jbc.M111.236950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Batliner J, Buehrer E, Fey MF, Tschan MP. Inhibition of the miR-143/145 cluster attenuated neutrophil differentiation of APL cells. Leuk Res. 2012;36:237–40. doi: 10.1016/j.leukres.2011.10.006. [DOI] [PubMed] [Google Scholar]
  • 31.Iio A, Takagi T, Miki K, Naoe T, Nakayama A, Akao Y. DDX6 post-transcriptionally down-regulates miR-143/145 expression through host gene NCR143/145 in cancer cells. Biochim Biophys Acta. 2013;1829:1102–10. doi: 10.1016/j.bbagrm.2013.07.010. [DOI] [PubMed] [Google Scholar]
  • 32.Medrano S, Sequeira-Lopez ML, Gomez RA. Deletion of the miR-143/145 cluster leads to hydronephrosis in mice. Am J Pathol. 2014;184:3226–38. doi: 10.1016/j.ajpath.2014.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Dahan D, Ekman M, Larsson-Callerfelt AK, Turczynska K, Boettger T, Braun T, et al. Induction of angiotensin-converting enzyme after miR-143/145 deletion is critical for impaired smooth muscle contractility. Am J Physiol Cell Physiol. 2014;307:C1093–101. doi: 10.1152/ajpcell.00250.2014. [DOI] [PubMed] [Google Scholar]
  • 34.Kent OA, McCall MN, Cornish TC, Halushka MK. Lessons from miR-143/145: the importance of cell-type localization of miRNAs. Nucleic Acids Res. 2014;42:7528–38. doi: 10.1093/nar/gku461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chivukula RR, Shi G, Acharya A, Mills EW, Zeitels LR, Anandam JL, et al. An essential mesenchymal function for miR-143/145 in intestinal epithelial regeneration. Cell. 2014;157:1104–16. doi: 10.1016/j.cell.2014.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kojima S, Enokida H, Yoshino H, Itesako T, Chiyomaru T, Kinoshita T, et al. The tumor-suppressive microRNA-143/145 cluster inhibits cell migration and invasion by targeting GOLM1 in prostate cancer. J Hum Genet. 2014;59:78–87. doi: 10.1038/jhg.2013.121. [DOI] [PubMed] [Google Scholar]
  • 37.Zhao W, Zheng XL, Peng DQ, Zhao SP. Myocyte enhancer factor 2A regulates hydrogen peroxide-induced senescence of vascular smooth muscle cells via microRNA-143. J Cell Physiol. 2015;230:2202–11. doi: 10.1002/jcp.24948. [DOI] [PubMed] [Google Scholar]
  • 38.Bhattachariya A, Dahan D, Ekman M, Boettger T, Braun T, Sward K, et al. Spontaneous activity and stretch-induced contractile differentiation are reduced in vascular smooth muscle of miR-143/145 knockout mice. Acta Physiol (Oxf) 2015 doi: 10.1111/apha.12536. doi: 10.1111/apha.12536. [DOI] [PubMed] [Google Scholar]
  • 39.Vengrenyuk Y, Nishi H, Long X, Ouimet M, Savji N, Martinez FO, et al. Cholesterol loading reprograms the MicroRNA-143/145-myocardin axis to convert aortic smooth muscle cells to a dysfunctional macrophage-like phenotype. Arterioscler Thromb Vasc Biol. 2015;35:535–46. doi: 10.1161/ATVBAHA.114.304029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Koo S, Martin G, Toussaint LG. MicroRNA-145 promotes the phenotype of human glioblastoma cells selected for invasion. Anticancer Res. 2015;35:3209–15. [PubMed] [Google Scholar]
  • 41.Avgeris M, Mavridis K, Tokas T, Stravodimos K, Fragoulis EG, Scorilas A. Uncovering the clinical utility of miR-143, miR-145 and miR-224 for predicting the survival of bladder cancer patients following treatment. Carcinogenesis. 2015;36:528–37. doi: 10.1093/carcin/bgv024. [DOI] [PubMed] [Google Scholar]
  • 42.Riches K, Alshanwani AR, Warburton P, O’Regan DJ, Ball SG, Wood IC, et al. Elevated expression levels of miR-143/5 in saphenous vein smooth muscle cells from patients with Type 2 diabetes drive persistent changes in phenotype and function. J Mol Cell Cardiol. 2014;74:240–50. doi: 10.1016/j.yjmcc.2014.05.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sala F, Aranda JF, Rotllan N, Ramirez CM, Aryal B, Elia L, et al. MiR-143/145 deficiency attenuates the progression of atherosclerosis in Ldlr−/−mice. Thromb Haemost. 2014;112:796–802. doi: 10.1160/TH13-11-0905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Engelhardt S, Leierseder S. Coinciding functions for miR-145 in vascular smooth muscle and cardiac fibroblasts. J Mol Cell Cardiol. 2013;65:105–7. doi: 10.1016/j.yjmcc.2013.10.002. [DOI] [PubMed] [Google Scholar]
  • 45.Boettger T, Beetz N, Kostin S, Schneider J, Kruger M, Hein L, et al. Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. J Clin Invest. 2009;119:2634–47. doi: 10.1172/JCI38864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Elia L, Quintavalle M, Zhang J, Contu R, Cossu L, Latronico MV, et al. The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell Death Differ. 2009;16:1590–8. doi: 10.1038/cdd.2009.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang D, Chang PS, Wang Z, Sutherland L, Richardson JA, Small E, et al. Activation of cardiac gene expression by myocardin, a transcriptional cofactor for serum response factor. Cell. 2001;105:851–62. doi: 10.1016/s0092-8674(01)00404-4. [DOI] [PubMed] [Google Scholar]
  • 48.Wang DZ, Li S, Hockemeyer D, Sutherland L, Wang Z, Schratt G, et al. Potentiation of serum response factor activity by a family of myocardin-related transcription factors. Proc Natl Acad Sci U S A. 2002;99:14855–60. doi: 10.1073/pnas.222561499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Liu X, Cheng Y, Yang J, Qin S, Chen X, Tang X, et al. Flank sequences of miR-145/143 and their aberrant expression in vascular disease: mechanism and therapeutic application. J Am Heart Assoc. 2013;2:e000407. doi: 10.1161/JAHA.113.000407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Quintavalle M, Elia L, Condorelli G, Courtneidge SA. MicroRNA control of podosome formation in vascular smooth muscle cells in vivo and in vitro. J Cell Biol. 2010;189:13–22. doi: 10.1083/jcb.200912096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Climent M, Quintavalle M, Miragoli M, Chen J, Condorelli G, Elia L. TGFbeta triggers miR-143/145 transfer from smooth muscle cells to endothelial cells, thereby modulating vessel stabilization. Circ Res. 2015;116:1753–64. doi: 10.1161/CIRCRESAHA.116.305178. [DOI] [PubMed] [Google Scholar]
  • 52.Thayanithy V, Dickson EL, Steer C, Subramanian S, Lou E. Tumor-stromal cross talk: direct cell-to-cell transfer of oncogenic microRNAs via tunneling nanotubes. Transl Res. 2014;164:359–65. doi: 10.1016/j.trsl.2014.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, et al. miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell. 2008;15:272–84. doi: 10.1016/j.devcel.2008.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Guo C, Sah JF, Beard L, Willson JK, Markowitz SD, Guda K. The noncoding RNA, miR-126, suppresses the growth of neoplastic cells by targeting phosphatidylinositol 3-kinase signaling and is frequently lost in colon cancers. Genes Chromosomes Cancer. 2008;47:939–46. doi: 10.1002/gcc.20596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kuhnert F, Mancuso MR, Hampton J, Stankunas K, Asano T, Chen CZ, et al. Attribution of vascular phenotypes of the murine Egfl7 locus to the microRNA miR-126. Development. 2008;135:3989–93. doi: 10.1242/dev.029736. [DOI] [PubMed] [Google Scholar]
  • 56.Musiyenko A, Bitko V, Barik S. Ectopic expression of miR-126*, an intronic product of the vascular endothelial EGF-like 7 gene, regulates prostein translation and invasiveness of prostate cancer LNCaP cells. J Mol Med (Berl) 2008;86:313–22. doi: 10.1007/s00109-007-0296-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, et al. The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell. 2008;15:261–71. doi: 10.1016/j.devcel.2008.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhang J, Du YY, Lin YF, Chen YT, Yang L, Wang HJ, et al. The cell growth suppressor, mir-126, targets IRS-1. Biochem Biophys Res Commun. 2008;377:136–40. doi: 10.1016/j.bbrc.2008.09.089. [DOI] [PubMed] [Google Scholar]
  • 59.Zhao C, Li Y, Zhang M, Yang Y, Chang L. miR-126 inhibits cell proliferation and induces cell apoptosis of hepatocellular carcinoma cells partially by targeting Sox2. Hum Cell. 2015;28:91–9. doi: 10.1007/s13577-014-0105-z. [DOI] [PubMed] [Google Scholar]
  • 60.Khella HW, Scorilas A, Mozes R, Mirham L, Lianidou E, Krylov SN, et al. Low expression of miR-126 is a prognostic marker for metastatic clear cell renal cell carcinoma. Am J Pathol. 2015;185:693–703. doi: 10.1016/j.ajpath.2014.11.017. [DOI] [PubMed] [Google Scholar]
  • 61.Yang Y, Song KL, Chang H, Chen L. Decreased expression of microRNA-126 is associated with poor prognosis in patients with cervical cancer. Diagn Pathol. 2014;9:220. doi: 10.1186/s13000-014-0220-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bijkerk R, van Solingen C, de Boer HC, de Vries DK, Monge M, van Oeveren-Rietdijk A, et al. Silencing of miRNA-126 in kidney ischemia reperfusion is associated with elevated SDF-1 levels and mobilization of Sca-1+/Lin− progenitor cells. Microrna. 2014;3:144–9. doi: 10.2174/2211536604666150121000340. [DOI] [PubMed] [Google Scholar]
  • 63.Lonvik K, Sorbye SW, Nilsen MN, Paulssen RH. Prognostic value of the MicroRNA regulators dicer and Drosha in non-small-cell lung cancer: co-expression of Drosha and miR-126 predicts poor survival. BMC Clin Pathol. 2014;14:45. doi: 10.1186/1472-6890-14-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Liu R, Gu J, Jiang P, Zheng Y, Liu X, Jiang X, et al. DNMT1-microRNA126 epigenetic circuit contributes to esophageal squamous cell carcinoma growth via ADAM9-EGFR-AKT signaling. Clin Cancer Res. 2015;21:854–63. doi: 10.1158/1078-0432.CCR-14-1740. [DOI] [PubMed] [Google Scholar]
  • 65.Jiang L, Tao C, He A, He X. Overexpression of miR-126 sensitizes osteosarcoma cells to apoptosis induced by epigallocatechin-3-gallate. World J Surg Oncol. 2014;12:383. doi: 10.1186/1477-7819-12-383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Liu B, Peng XC, Zheng XL, Wang J, Qin YW. MiR-126 restoration down-regulate VEGF and inhibit the growth of lung cancer cell lines in vitro and in vivo. Lung Cancer. 2009;66:169–75. doi: 10.1016/j.lungcan.2009.01.010. [DOI] [PubMed] [Google Scholar]
  • 67.Feng R, Chen X, Yu Y, Su L, Yu B, Li J, et al. miR-126 functions as a tumour suppressor in human gastric cancer. Cancer Lett. 2010;298:50–63. doi: 10.1016/j.canlet.2010.06.004. [DOI] [PubMed] [Google Scholar]
  • 68.Harnprasopwat R, Ha D, Toyoshima T, Lodish H, Tojo A, Kotani A. Alteration of processing induced by a single nucleotide polymorphism in pri-miR-126. Biochem Biophys Res Commun. 2010;399:117–22. doi: 10.1016/j.bbrc.2010.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Meister J, Schmidt MH. miR-126 and miR-126*: new players in cancer. ScientificWorldJournal. 2010;10:2090–100. doi: 10.1100/tsw.2010.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Oglesby IK, Bray IM, Chotirmall SH, Stallings RL, O’Neill SJ, McElvaney NG, et al. miR-126 is downregulated in cystic fibrosis airway epithelial cells and regulates TOM1 expression. J Immunol. 2010;184:1702–9. doi: 10.4049/jimmunol.0902669. [DOI] [PubMed] [Google Scholar]
  • 71.Sun Y, Bai Y, Zhang F, Wang Y, Guo Y, Guo L. miR-126 inhibits non-small cell lung cancer cells proliferation by targeting EGFL7. Biochem Biophys Res Commun. 2010;391:1483–9. doi: 10.1016/j.bbrc.2009.12.098. [DOI] [PubMed] [Google Scholar]
  • 72.Chen JJ, Zhou SH. Mesenchymal stem cells overexpressing MiR-126 enhance ischemic angiogenesis via the AKT/ERK-related pathway. Cardiol J. 2011;18:675–81. doi: 10.5603/cj.2011.0032. [DOI] [PubMed] [Google Scholar]
  • 73.Collison A, Herbert C, Siegle JS, Mattes J, Foster PS, Kumar RK. Altered expression of microRNA in the airway wall in chronic asthma: miR-126 as a potential therapeutic target. BMC Pulm Med. 2011;11:29. doi: 10.1186/1471-2466-11-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cui W, Li Q, Feng L, Ding W. MiR-126-3p regulates progesterone receptors and involves development and lactation of mouse mammary gland. Mol Cell Biochem. 2011;355:17–25. doi: 10.1007/s11010-011-0834-1. [DOI] [PubMed] [Google Scholar]
  • 75.Donnem T, Lonvik K, Eklo K, Berg T, Sorbye SW, Al-Shibli K, et al. Independent and tissue-specific prognostic impact of miR-126 in nonsmall cell lung cancer: coexpression with vascular endothelial growth factor-A predicts poor survival. Cancer. 2011;117:3193–200. doi: 10.1002/cncr.25907. [DOI] [PubMed] [Google Scholar]
  • 76.Li XM, Wang AM, Zhang J, Yi H. Down-regulation of miR-126 expression in colorectal cancer and its clinical significance. Med Oncol. 2011;28:1054–7. doi: 10.1007/s12032-010-9637-6. [DOI] [PubMed] [Google Scholar]
  • 77.Li Z, Chen J. In vitro functional study of miR-126 in leukemia. Methods Mol Biol. 2011;676:185–95. doi: 10.1007/978-1-60761-863-8_13. [DOI] [PubMed] [Google Scholar]
  • 78.Ren G, Kang Y. A one-two punch of miR-126/126* against metastasis. Nat Cell Biol. 2013;15:231–3. doi: 10.1038/ncb2703. [DOI] [PubMed] [Google Scholar]
  • 79.Schmidt Y, Simunovic F, Strassburg S, Pfeifer D, Stark GB, Finkenzeller G. miR-126 regulates platelet-derived growth factor receptor-alpha expression and migration of primary human osteoblasts. Biol Chem. 2015;396:61–70. doi: 10.1515/hsz-2014-0168. [DOI] [PubMed] [Google Scholar]
  • 80.Togliatto G, Trombetta A, Dentelli P, Gallo S, Rosso A, Cotogni P, et al. Unacylated ghrelin induces oxidative stress resistance in a glucose intolerance and peripheral artery disease mouse model by restoring endothelial cell miR-126 expression. Diabetes. 2015;64:1370–82. doi: 10.2337/db14-0991. [DOI] [PubMed] [Google Scholar]
  • 81.Miki K, Endo K, Takahashi S, Funakoshi S, Takei I, Katayama S, et al. Efficient detection and purification of cell populations using synthetic microRNA switches. Cell Stem Cell. 2015;16:699–711. doi: 10.1016/j.stem.2015.04.005. [DOI] [PubMed] [Google Scholar]
  • 82.Feng X, Tan W, Cheng S, Wang H, Ye S, Yu C, et al. Upregulation of microRNA-126 in hepatic stellate cells may affect pathogenesis of liver fibrosis through the NF-kappaB pathway. DNA Cell Biol. 2015;34(7):470–80. doi: 10.1089/dna.2014.2760. [DOI] [PubMed] [Google Scholar]
  • 83.Rohde JH, Weigand JE, Suess B, Dimmeler S. A universal aptamer chimera for the delivery of functional microRNA-126. Nucleic Acid Ther. 2015;25:141–51. doi: 10.1089/nat.2014.0501. [DOI] [PubMed] [Google Scholar]
  • 84.Shibayama Y, Kondo T, Ohya H, Fujisawa S, Teshima T, Iseki K. Upregulation of microRNA-126-5p is associated with drug resistance to cytarabine and poor prognosis in AML patients. Oncol Rep. 2015;33:2176–82. doi: 10.3892/or.2015.3839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hu J, Zeng L, Huang J, Wang G, Lu H. miR-126 promotes angiogenesis and attenuates inflammation after contusion spinal cord injury in rats. Brain Res. 2015;1608:191–202. doi: 10.1016/j.brainres.2015.02.036. [DOI] [PubMed] [Google Scholar]
  • 86.Nakano M, Fukushima Y, Yokota S, Fukami T, Takamiya M, Aoki Y, et al. CYP2A7 pseudogene transcript affects CYP2A6 expression in human liver by acting as a decoy for miR-126. Drug Metab Dispos. 2015;43:703–12. doi: 10.1124/dmd.115.063255. [DOI] [PubMed] [Google Scholar]
  • 87.Meng Q, Wang W, Yu X, Li W, Kong L, Qian A, et al. Upregulation of microRNA-126 contributes to endothelial progenitor cell function in deep vein thrombosis via its target PIK3R2. J Cell Biochem. 2015;116(8):1613–23. doi: 10.1002/jcb.25115. [DOI] [PubMed] [Google Scholar]
  • 88.Mocharla P, Briand S, Giannotti G, Dorries C, Jakob P, Paneni F, et al. AngiomiR-126 expression and secretion from circulating CD34(+) and CD14(+) PBMCs: role for proangiogenic effects and alterations in type 2 diabetics. Blood. 2013;121:226–36. doi: 10.1182/blood-2012-01-407106. [DOI] [PubMed] [Google Scholar]
  • 89.Meng S, Cao JT, Zhang B, Zhou Q, Shen CX, Wang CQ. Downregulation of microRNA-126 in endothelial progenitor cells from diabetes patients, impairs their functional properties, via target gene Spred-1. J Mol Cell Cardiol. 2012;53:64–72. doi: 10.1016/j.yjmcc.2012.04.003. [DOI] [PubMed] [Google Scholar]
  • 90.Prattichizzo F, Giuliani A, Ceka A, Rippo MR, Bonfigli AR, Testa R, et al. Epigenetic mechanisms of endothelial dysfunction in type 2 diabetes. Clin Epigenetics. 2015;7(1):56. doi: 10.1186/s13148-015-0090-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Tian HS, Zhou QG, Shao F. Relationship between arterial atheromatous plaque morphology and platelet-associated miR-126 and miR-223 expressions. Asian Pac J Trop Med. 2015;8:309–14. doi: 10.1016/S1995-7645(14)60336-9. [DOI] [PubMed] [Google Scholar]
  • 92.Zhang T, Li L, Shang Q, Lv C, Wang C, Su B. Circulating miR-126 is a potential biomarker to predict the onset of type 2 diabetes mellitus in susceptible individuals. Biochem Biophys Res Commun. 2015;463(1–2):60–3. doi: 10.1016/j.bbrc.2015.05.017. [DOI] [PubMed] [Google Scholar]
  • 93.Endo-Takahashi Y, Negishi Y, Nakamura A, Ukai S, Ooaku K, Oda Y, et al. Systemic delivery of miR-126 by miRNA-loaded bubble liposomes for the treatment of hindlimb ischemia. Sci Rep. 2014;4:3883. doi: 10.1038/srep03883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Potus F, Graydon C, Provencher S, Bonnet S. Vascular remodeling process in pulmonary arterial hypertension, with focus on miR-204 and miR-126 (2013 Grover Conference series) Pulm Circ. 2014;4:175–84. doi: 10.1086/675980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yamaguchi T, Iijima T, Wakaume R, Takahashi K, Matsumoto H, Nakano D, et al. Underexpression of miR-126 and miR-20b in hereditary and nonhereditary colorectal tumors. Oncology. 2014;87:58–66. doi: 10.1159/000363303. [DOI] [PubMed] [Google Scholar]
  • 96.Yan T, Cui K, Huang X, Ding S, Zheng Y, Luo Q, et al. Assessment of therapeutic efficacy of miR-126 with contrast-enhanced ultrasound in preeclampsia rats. Placenta. 2014;35:23–9. doi: 10.1016/j.placenta.2013.10.017. [DOI] [PubMed] [Google Scholar]
  • 97.Goerke SM, Kiefer LS, Stark GB, Simunovic F, Finkenzeller G. miR-126 modulates angiogenic growth parameters of peripheral blood endothelial progenitor cells. Biol Chem. 2015;396:245–52. doi: 10.1515/hsz-2014-0259. [DOI] [PubMed] [Google Scholar]
  • 98.Olivieri F, Bonafe M, Spazzafumo L, Gobbi M, Prattichizzo F, Recchioni R, et al. Age- and glycemia-related miR-126-3p levels in plasma and endothelial cells. Aging. 2014;6:771–87. doi: 10.18632/aging.100693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.McAuley AK, Dirani M, Wang JJ, Connell PP, Lamoureux EL, Hewitt AW. A genetic variant regulating miR-126 is associated with sight threatening diabetic retinopathy. Diab Vasc Dis Res. 2015;12:133–8. doi: 10.1177/1479164114560160. [DOI] [PubMed] [Google Scholar]
  • 100.Schwartz SM. Smooth muscle migration in atherosclerosis and restenosis. J Clin Invest. 1997;100:S87–9. [PubMed] [Google Scholar]
  • 101.Lagna G, Ku MM, Nguyen PH, Neuman NA, Davis BN, Hata A. Control of phenotypic plasticity of smooth muscle cells by bone morphogenetic protein signaling through the myocardin-related transcription factors. J Biol Chem. 2007;282:37244–55. doi: 10.1074/jbc.M708137200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Davis BN, Hilyard AC, Lagna G, Hata A. SMAD proteins control DROSHA-mediated microRNA maturation. Nature. 2008;454:56–61. doi: 10.1038/nature07086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Leeper NJ, Raiesdana A, Kojima Y, Chun HJ, Azuma J, Maegdefessel L, et al. MicroRNA-26a is a novel regulator of vascular smooth muscle cell function. J Cell Physiol. 2011;226:1035–43. doi: 10.1002/jcp.22422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Chan MC, Hilyard AC, Wu C, Davis BN, Hill NS, Lal A, et al. Molecular basis for antagonism between PDGF and the TGFbeta family of signalling pathways by control of miR-24 expression. EMBO J. 2010;29:559–73. doi: 10.1038/emboj.2009.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Yan J, Stringer SE, Hamilton A, Charlton-Menys V, Gotting C, Muller B, et al. Decorin GAG synthesis and TGF-beta signaling mediate Ox-LDL-induced mineralization of human vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2011;31:608–15. doi: 10.1161/ATVBAHA.110.220749. [DOI] [PubMed] [Google Scholar]
  • 106.Nasser MW, Datta J, Nuovo G, Kutay H, Motiwala T, Majumder S, et al. Down-regulation of micro-RNA-1 (miR-1) in lung cancer. Suppression of tumorigenic property of lung cancer cells and their sensitization to doxorubicin-induced apoptosis by miR-1. J Biol Chem. 2008;283:33394–405. doi: 10.1074/jbc.M804788200. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 107.Xie C, Huang H, Sun X, Guo Y, Hamblin M, Ritchie RP, et al. MicroRNA-1 regulates smooth muscle cell differentiation by repressing Kruppel-like factor 4. Stem Cells Dev. 2011;20:205–10. doi: 10.1089/scd.2010.0283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Huang H, Xie C, Sun X, Ritchie RP, Zhang J, Chen YE. miR-10a contributes to retinoid acid-induced smooth muscle cell differentiation. J Biol Chem. 2010;285:9383–9. doi: 10.1074/jbc.M109.095612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Fang Y, Shi C, Manduchi E, Civelek M, Davies PF. MicroRNA-10a regulation of proinflammatory phenotype in athero-susceptible endothelium in vivo and in vitro. Proc Natl Acad Sci U S A. 2010;107:13450–5. doi: 10.1073/pnas.1002120107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kim S, Kang H. miR-15b induced by platelet-derived growth factor signaling is required for vascular smooth muscle cell proliferation. BMB Rep. 2013;46:550–4. doi: 10.5483/BMBRep.2013.46.11.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kee HJ, Kim GR, Cho SN, Kwon JS, Ahn Y, Kook H, et al. miR-18a-5p microRNA increases vascular smooth muscle cell differentiation by downregulating syndecan4. Korean Circ J. 2014;44:255–63. doi: 10.4070/kcj.2014.44.4.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cheng Y, Zhang C. MicroRNA-21 in cardiovascular disease. J Cardiovasc Transl Res. 2010;3:251–5. doi: 10.1007/s12265-010-9169-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Young MR, Santhanam AN, Yoshikawa N, Colburn NH. Have tumor suppressor PDCD4 and its counteragent oncogenic miR-21 gone rogue? Mol Interv. 2010;10:76–9. doi: 10.1124/mi.10.2.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Choe N, Kwon JS, Kim JR, Eom GH, Kim Y, Nam KI, et al. The microRNA miR-132 targets Lrrfip1 to block vascular smooth muscle cell proliferation and neointimal hyperplasia. Atherosclerosis. 2013;229:348–55. doi: 10.1016/j.atherosclerosis.2013.05.009. [DOI] [PubMed] [Google Scholar]
  • 115.Kinoshita T, Nohata N, Fuse M, Hanazawa T, Kikkawa N, Fujimura L, et al. Tumor suppressive microRNA-133a regulates novel targets: moesin contributes to cancer cell proliferation and invasion in head and neck squamous cell carcinoma. Biochem Biophys Res Commun. 2012;418:378–83. doi: 10.1016/j.bbrc.2012.01.030. [DOI] [PubMed] [Google Scholar]
  • 116.Qiu T, Zhou X, Wang J, Du Y, Xu J, Huang Z, et al. MiR-145, miR-133a and miR-133b inhibit proliferation, migration, invasion and cell cycle progression via targeting transcription factor Sp1 in gastric cancer. FEBS Lett. 2014;588:1168–77. doi: 10.1016/j.febslet.2014.02.054. [DOI] [PubMed] [Google Scholar]
  • 117.Kim MH, Ham O, Lee SY, Choi E, Lee CY, Park JH, et al. MicroRNA-365 inhibits the proliferation of vascular smooth muscle cells by targeting cyclin D1. J Cell Biochem. 2014;115:1752–61. doi: 10.1002/jcb.24841. [DOI] [PubMed] [Google Scholar]
  • 118.Merlet E, Atassi F, Motiani RK, Mougenot N, Jacquet A, Nadaud S, et al. miR-424/322 regulates vascular smooth muscle cell phenotype and neointimal formation in the rat. Cardiovasc Res. 2013;98:458–68. doi: 10.1093/cvr/cvt045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sun Y, Chen D, Cao L, Zhang R, Zhou J, Chen H, et al. MiR-490-3p modulates the proliferation of vascular smooth muscle cells induced by ox-LDL through targeting PAPP-A. Cardiovasc Res. 2013;100:272–9. doi: 10.1093/cvr/cvt172. [DOI] [PubMed] [Google Scholar]
  • 120.Li P, Liu Y, Yi B, Wang G, You X, Zhao X, et al. MicroRNA-638 is highly expressed in human vascular smooth muscle cells and inhibits PDGF-BB-induced cell proliferation and migration through targeting orphan nuclear receptor NOR1. Cardiovasc Res. 2013;99:185–93. doi: 10.1093/cvr/cvt082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Li P, Zhu N, Yi B, Wang N, Chen M, You X, et al. MicroRNA-663 regulates human vascular smooth muscle cell phenotypic switch and vascular neointimal formation. Circ Res. 2013;113:1117–27. doi: 10.1161/CIRCRESAHA.113.301306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Korff T, Pfisterer L, Schorpp-Kistner M. miR-663 and the miRaculous vascular smooth muscle phenotypic switch. Circ Res. 2013;113:1102–5. doi: 10.1161/CIRCRESAHA.113.302578. [DOI] [PubMed] [Google Scholar]
  • 123.Ji R, Cheng Y, Yue J, Yang J, Liu X, Chen H, et al. MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ Res. 2007;100:1579–88. doi: 10.1161/CIRCRESAHA.106.141986. [DOI] [PubMed] [Google Scholar]
  • 124.Silvestri P, Rigattieri S, Loschiavo P. Does the effect of microRNAs in vascular neointimal formation depend on cell cycle phase? Circ Res. 2008;102(9):e101. doi: 10.1161/CIRCRESAHA.108.175661. author reply e102. [DOI] [PubMed] [Google Scholar]
  • 125.Davis-Dusenbery BN, Wu C, Hata A. Micromanaging vascular smooth muscle cell differentiation and phenotypic modulation. Arterioscler Thromb Vasc Biol. 2011;31:2370–7. doi: 10.1161/ATVBAHA.111.226670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Talasila A, Yu H, Ackers-Johnson M, Bot M, van Berkel T, Bennett MR, et al. Myocardin regulates vascular response to injury through miR-24/-29a and platelet-derived growth factor receptor-beta. Arterioscler Thromb Vasc Biol. 2013;33:2355–65. doi: 10.1161/ATVBAHA.112.301000. [DOI] [PubMed] [Google Scholar]
  • 127.Icli B, Wara AK, Moslehi J, Sun X, Plovie E, Cahill M, et al. MicroRNA-26a regulates pathological and physiological angiogenesis by targeting BMP/SMAD1 signaling. Circ Res. 2013;113:1231–41. doi: 10.1161/CIRCRESAHA.113.301780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Bai Y, Wang L, Sun L, Ye P, Hui R. Circulating microRNA-26a: potential predictors and therapeutic targets for non-hypertensive intracerebral hemorrhage. Med Hypotheses. 2011;77:488–90. doi: 10.1016/j.mehy.2011.06.017. [DOI] [PubMed] [Google Scholar]
  • 129.Liu X, Cheng Y, Chen X, Yang J, Xu L, Zhang C. MicroRNA-31 regulated by the extracellular regulated kinase is involved in vascular smooth muscle cell growth via large tumor suppressor homolog 2. J Biol Chem. 2011;286:42371–80. doi: 10.1074/jbc.M111.261065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Liu X, Sempere LF, Ouyang H, Memoli VA, Andrew AS, Luo Y, et al. MicroRNA-31 functions as an oncogenic microRNA in mouse and human lung cancer cells by repressing specific tumor suppressors. J Clin Invest. 2010;120:1298–309. doi: 10.1172/JCI39566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wang J, Yan CH, Li Y, Xu K, Tian XX, Peng CF, et al. MicroRNA-31 controls phenotypic modulation of human vascular smooth muscle cells by regulating its target gene cellular repressor of E1A-stimulated genes. Exp Cell Res. 2013;319:1165–75. doi: 10.1016/j.yexcr.2013.03.010. [DOI] [PubMed] [Google Scholar]
  • 132.Cheng HS, Sivachandran N, Lau A, Boudreau E, Zhao JL, Baltimore D, et al. MicroRNA-146 represses endothelial activation by inhibiting pro-inflammatory pathways. EMBO Mol Med. 2013;5:949–66. doi: 10.1002/emmm.201202318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Togliatto G, Trombetta A, Dentelli P, Rosso A, Brizzi MF. MIR221/MIR222-driven post-transcriptional regulation of P27KIP1 and P57KIP2 is crucial for high-glucose- and AGE-mediated vascular cell damage. Diabetologia. 2011;54:1930–40. doi: 10.1007/s00125-011-2125-5. [DOI] [PubMed] [Google Scholar]
  • 134.Kothapalli D, Castagnino P, Rader DJ, Phillips MC, Lund-Katz S, Assoian RK. Apolipoprotein E-mediated cell cycle arrest linked to p27 and the Cox2-dependent repression of miR221/222. Atherosclerosis. 2013;227:65–71. doi: 10.1016/j.atherosclerosis.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Mackenzie NC, Staines KA, Zhu D, Genever P, Macrae VE. miRNA-221 and miRNA-222 synergistically function to promote vascular calcification. Cell Biochem Funct. 2014;32:209–16. doi: 10.1002/cbf.3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Gao S, Wassler M, Zhang L, Li Y, Wang J, Zhang Y, et al. MicroRNA-133a regulates insulin-like growth factor-1 receptor expression and vascular smooth muscle cell proliferation in murine atherosclerosis. Atherosclerosis. 2014;232:171–9. doi: 10.1016/j.atherosclerosis.2013.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zhang CF, Kang K, Li XM, Xie BD. MicroRNA-136 promotes vascular muscle cell proliferation through the ERK1/2 pathway by targeting PPP2R2A in atherosclerosis. Curr Vasc Pharmacol. 2014;13(3):405–12. doi: 10.2174/1570161112666141118094612. [DOI] [PubMed] [Google Scholar]
  • 138.Hu W, Wang M, Yin H, Yao C, He Q, Yin L, et al. MicroRNA-1298 is regulated by DNA methylation and affects vascular smooth muscle cell function by targeting connexin 43. Cardiovasc Res. 2015;107:534–45. doi: 10.1093/cvr/cvv160. [DOI] [PubMed] [Google Scholar]
  • 139.Weissberg PL, Clesham GJ, Bennett MR. Is vascular smooth muscle cell proliferation beneficial? Lancet. 1996;347:305–7. doi: 10.1016/s0140-6736(96)90472-9. [DOI] [PubMed] [Google Scholar]
  • 140.O’Sullivan JF, Martin K, Caplice NM. Microribonucleic acids for prevention of plaque rupture and in-stent restenosis: “a finger in the dam”. J Am Coll Cardiol. 2011;57:383–9. doi: 10.1016/j.jacc.2010.09.029. [DOI] [PubMed] [Google Scholar]
  • 141.Vanhoutte PM. Endothelium-derived free radicals: for worse and for better. J Clin Invest. 2001;107:23–5. doi: 10.1172/JCI11832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Harrison DG. Cellular and molecular mechanisms of endothelial cell dysfunction. J Clin Invest. 1997;100:2153–7. doi: 10.1172/JCI119751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Iaccarino D, Politi L, Rossi R, Sgura F, Monopoli D, Modena MG, et al. Rationale and study design of the OISTER trial: optical coherence tomography evaluation of stent struts re-endothelialization in patients with non-ST-elevation acute coronary syndromes—a comparison of the intrEpide tRapidil eluting stent vs. taxus drug-eluting stent implantation. J Cardiovasc Med (Hagerstown) 2010;11:536–43. doi: 10.2459/JCM.0b013e32833499c4. [DOI] [PubMed] [Google Scholar]
  • 144.Mei H, Campbell JM, Paddock CM, Lertkiatmongkol P, Mosesson MW, Albrecht RM, et al. Regulation of endothelial cell barrier function by antibody-driven affinity modulation of PECAM-1. J Biol Chem. 2014;289(30):20836–44. doi: 10.1074/jbc.M114.557454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Pannekoek WJ, Post A, Bos JL. Rap1 signaling in endothelial barrier control. Cell Adh Migr. 2014;8(2):100–7. doi: 10.4161/cam.27352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gadang V, Konaniah E, Hui DY, Jaeschke A. Mixed-lineage kinase 3 deficiency promotes neointima formation through increased activation of the RhoA pathway in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2014;34:1429–36. doi: 10.1161/ATVBAHA.114.303439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Popma JJ, Topol EJ. Factors influencing restenosis after coronary angioplasty. Am J Med. 1990;88:16N–24. [PubMed] [Google Scholar]
  • 148.Faxon DP, Sanborn TA, Haudenschild CC. Mechanism of angioplasty and its relation to restenosis. Am J Cardiol. 1987;60:5B–9. doi: 10.1016/0002-9149(87)90476-0. [DOI] [PubMed] [Google Scholar]
  • 149.Chaabane C, Otsuka F, Virmani R, Bochaton-Piallat ML. Biological responses in stented arteries. Cardiovasc Res. 2013;99:353–63. doi: 10.1093/cvr/cvt115. [DOI] [PubMed] [Google Scholar]
  • 150.De Caterina R, Massaro M, Scoditti E, Annunziata CM. Pharmacological modulation of vascular inflammation in atherothrombosis. Ann N Y Acad Sci. 2010;1207:23–31. doi: 10.1111/j.1749-6632.2010.05784.x. [DOI] [PubMed] [Google Scholar]
  • 151.Esmon CT. Inflammation and thrombosis. J Thromb Haemost. 2003;1:1343–8. doi: 10.1046/j.1538-7836.2003.00261.x. [DOI] [PubMed] [Google Scholar]
  • 152.Rhee JW, Wu JC. Advances in nanotechnology for the management of coronary artery disease. Trends Cardiovasc Med. 2013;23:39–45. doi: 10.1016/j.tcm.2012.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Otsuka F, Finn AV, Yazdani SK, Nakano M, Kolodgie FD, Virmani R. The importance of the endothelium in atherothrombosis and coronary stenting. Nat Rev Cardiol. 2012;9:439–53. doi: 10.1038/nrcardio.2012.64. [DOI] [PubMed] [Google Scholar]
  • 154.Hagensen MK, Raarup MK, Mortensen MB, Thim T, Nyengaard JR, Falk E, et al. Circulating endothelial progenitor cells do not contribute to regeneration of endothelium after murine arterial injury. Cardiovasc Res. 2012;93:223–31. doi: 10.1093/cvr/cvr278. [DOI] [PubMed] [Google Scholar]
  • 155.Tsuzuki M. Bone marrow-derived cells are not involved in reendothelialized endothelium as endothelial cells after simple endothelial denudation in mice. Basic Res Cardiol. 2009;104:601–11. doi: 10.1007/s00395-009-0021-7. [DOI] [PubMed] [Google Scholar]
  • 156.Cevese A. Totipotent stem cells could do everything … or else nothing: the case of vascular reendothelialization. Cardiovasc Res. 2012;93:211–2. doi: 10.1093/cvr/cvr342. [DOI] [PubMed] [Google Scholar]
  • 157.Pearson JD. Endothelial progenitor cells—hype or hope? J Thromb Haemost. 2009;7:255–62. doi: 10.1111/j.1538-7836.2008.03214.x. [DOI] [PubMed] [Google Scholar]
  • 158.Jimenez JM, Prasad V, Yu MD, Kampmeyer CP, Kaakour AH, Wang PJ, et al. Macro- and microscale variables regulate stent haemodynamics, fibrin deposition and thrombomodulin expression. J R Soc Interface. 2014;11(94):20131079. doi: 10.1098/rsif.2013.1079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Alexander RW. Getting stents to go with the flow. J Clin Invest. 2004;113:1532–4. doi: 10.1172/JCI22000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Liuzzo JP, Ambrose JA, Coppola JT. Sirolimus- and taxol-eluting stents differ towards intimal hyperplasia and re-endothelialization. J Invasive Cardiol. 2005;17:497–502. [PubMed] [Google Scholar]
  • 161.Simard T, Hibbert B, Ramirez FD, Froeschl M, Chen YX, O’Brien ER. The evolution of coronary stents: a brief review. Can J Cardiol. 2014;30:35–45. doi: 10.1016/j.cjca.2013.09.012. [DOI] [PubMed] [Google Scholar]
  • 162.Palmerini T, Biondi-Zoccai G, Della Riva D, Stettler C, Sangiorgi D, D’Ascenzo F, et al. Stent thrombosis with drug-eluting and bare-metal stents: evidence from a comprehensive network meta-analysis. Lancet. 2012;379:1393–402. doi: 10.1016/S0140-6736(12)60324-9. [DOI] [PubMed] [Google Scholar]
  • 163.Di Lorenzo E, Carbone G, Sauro L, Casafina A, Capasso M, Sauro R. Bare-metal stents versus drug-eluting stents for primary angioplasty: long-term outcome. Curr Cardiol Rep. 2011;13:459–64. doi: 10.1007/s11886-011-0207-0. [DOI] [PubMed] [Google Scholar]
  • 164.Wessely R. New drug-eluting stent concepts. Nat Rev Cardiol. 2010;7:194–203. doi: 10.1038/nrcardio.2010.14. [DOI] [PubMed] [Google Scholar]
  • 165.Huang KN, Grandi SM, Filion KB, Eisenberg MJ. Late and very late stent thrombosis in patients with second-generation drug-eluting stents. Can J Cardiol. 2013;29:1488–94. doi: 10.1016/j.cjca.2013.04.001. [DOI] [PubMed] [Google Scholar]
  • 166.Siddiqi OK, Faxon DP. Very late stent thrombosis: current concepts. Curr Opin Cardiol. 2012;27:634–41. doi: 10.1097/HCO.0b013e3283587c7e. [DOI] [PubMed] [Google Scholar]
  • 167.McFadden EP, Stabile E, Regar E, Cheneau E, Ong AT, Kinnaird T, et al. Late thrombosis in drug-eluting coronary stents after discontinuation of antiplatelet therapy. Lancet. 2004;364:1519–21. doi: 10.1016/S0140-6736(04)17275-9. [DOI] [PubMed] [Google Scholar]
  • 168.Rossini R, Baroni M, Musumeci G, Gavazzi A. Oral antiplatelet therapy after drug-eluting stent implantation: adherence and side-effects. J Cardiovasc Med (Hagerstown) 2013;14:81–90. doi: 10.2459/JCM.0b013e328356a545. [DOI] [PubMed] [Google Scholar]
  • 169.Dehmer GJ, Smith KJ. Drug-eluting coronary artery stents. Am Fam Physician. 2009;80:1245–51. [PubMed] [Google Scholar]
  • 170.Jansen F, Yang X, Hoelscher M, Cattelan A, Schmitz T, Proebsting S, et al. Endothelial microparticle-mediated transfer of MicroRNA-126 promotes vascular endothelial cell repair via SPRED1 and is abrogated in glucose-damaged endothelial microparticles. Circulation. 2013;128:2026–38. doi: 10.1161/CIRCULATIONAHA.113.001720. [DOI] [PubMed] [Google Scholar]
  • 171.Deb A, Patterson C. Hard luck stories: the reality of endothelial progenitor cells continues to fall short of the promise. Circulation. 2010;121:850–2. doi: 10.1161/CIR.0b013e3181d4c360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Hagensen MK, Shim J, Thim T, Falk E, Bentzon JF. Circulating endothelial progenitor cells do not contribute to plaque endothelium in murine atherosclerosis. Circulation. 2010;121:898–905. doi: 10.1161/CIRCULATIONAHA.109.885459. [DOI] [PubMed] [Google Scholar]
  • 173.Jansen F, Yang X, Proebsting S, Hoelscher M, Przybilla D, Baumann K, et al. MicroRNA expression in circulating microvesicles predicts cardiovascular events in patients with coronary artery disease. J Am Heart Assoc. 2014;3:e001249. doi: 10.1161/JAHA.114.001249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Santulli G. Angiogenesis: insights from a systematic overview. Nova Science; New York: 2013. [Google Scholar]
  • 175.Zhuang G, Wu X, Jiang Z, Kasman I, Yao J, Guan Y, et al. Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012;31:3513–23. doi: 10.1038/emboj.2012.183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Qin B, Yang H, Xiao B. Role of microRNAs in endothelial inflammation and senescence. Mol Biol Rep. 2012;39:4509–18. doi: 10.1007/s11033-011-1241-0. [DOI] [PubMed] [Google Scholar]
  • 177.Kumar S, Kim CW, Simmons RD, Jo H. Role of flow-sensitive microRNAs in endothelial dysfunction and atherosclerosis: mechanosensitive athero-miRs. Arterioscler Thromb Vasc Biol. 2014;34:2206–16. doi: 10.1161/ATVBAHA.114.303425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Zhou Q, Gallagher R, Ufret-Vincenty R, Li X, Olson EN, Wang S. Regulation of angiogenesis and choroidal neovascularization by members of microRNA-23~27~24 clusters. Proc Natl Acad Sci U S A. 2011;108:8287–92. doi: 10.1073/pnas.1105254108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Nicoli S, Standley C, Walker P, Hurlstone A, Fogarty KE, Lawson ND. MicroRNA-mediated integration of haemodynamics and Vegf signalling during angiogenesis. Nature. 2010;464:1196–200. doi: 10.1038/nature08889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Ye X, Hemida MG, Qiu Y, Hanson PJ, Zhang HM, Yang D. MiR-126 promotes coxsackievirus replication by mediating cross-talk of ERK1/2 and Wnt/beta-catenin signal pathways. Cell Mol Life Sci. 2013;70:4631–44. doi: 10.1007/s00018-013-1411-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Taverna S, Amodeo V, Saieva L, Russo A, Giallombardo M, De Leo G, et al. Exosomal shuttling of miR-126 in endothelial cells modulates adhesive and migratory abilities of chronic myelogenous leukemia cells. Mol Cancer. 2014;13:169. doi: 10.1186/1476-4598-13-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Liu LY, Wang W, Zhao LY, Guo B, Yang J, Zhao XG, et al. Mir-126 inhibits growth of SGC-7901 cells by synergistically targeting the oncogenes PI3KR2 and Crk, and the tumor suppressor PLK2. Int J Oncol. 2014;45:1257–65. doi: 10.3892/ijo.2014.2516. [DOI] [PubMed] [Google Scholar]
  • 183.Wang Z, Yin B, Wang B, Ma Z, Liu W, Lv G. MicroRNA-210 promotes proliferation and invasion of peripheral nerve sheath tumor cells targeting EFNA3. Oncol Res. 2013;21:145–54. doi: 10.3727/096504013X13841340689573. [DOI] [PubMed] [Google Scholar]
  • 184.Menghini R, Casagrande V, Cardellini M, Martelli E, Terrinoni A, Amati F, et al. MicroRNA 217 modulates endothelial cell senescence via silent information regulator 1. Circulation. 2009;120:1524–32. doi: 10.1161/CIRCULATIONAHA.109.864629. [DOI] [PubMed] [Google Scholar]
  • 185.Kim J, Kang Y, Kojima Y, Lighthouse JK, Hu X, Aldred MA, et al. An endothelial apelin-FGF link mediated by miR-424 and miR-503 is disrupted in pulmonary arterial hypertension. Nat Med. 2013;19:74–82. doi: 10.1038/nm.3040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Katz G, Pobezinsky LA, Jeurling S, Shinzawa M, Van Laethem F, Singer A. T cell receptor stimulation impairs IL-7 receptor signaling by inducing expression of the microRNA miR-17 to target Janus kinase 1. Sci Signal. 2014;7:ra83. doi: 10.1126/scisignal.2005221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Jin C, Zhao Y, Yu L, Xu S, Fu G. MicroRNA-21 mediates the rapamycin-induced suppression of endothelial proliferation and migration. FEBS Lett. 2013;587:378–85. doi: 10.1016/j.febslet.2012.12.021. [DOI] [PubMed] [Google Scholar]
  • 188.Zhou J, Wang KC, Wu W, Subramaniam S, Shyy JY, Chiu JJ, et al. MicroRNA-21 targets peroxisome proliferators-activated receptor-alpha in an autoregulatory loop to modulate flow-induced endothelial inflammation. Proc Natl Acad Sci U S A. 2011;108:10355–60. doi: 10.1073/pnas.1107052108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Fiedler J, Jazbutyte V, Kirchmaier BC, Gupta SK, Lorenzen J, Hartmann D, et al. MicroRNA-24 regulates vascularity after myocardial infarction. Circulation. 2011;124:720–30. doi: 10.1161/CIRCULATIONAHA.111.039008. [DOI] [PubMed] [Google Scholar]
  • 190.Zhou Q, Anderson C, Zhang H, Li X, Inglis F, Jayagopal A, et al. Repression of choroidal neovascularization through actin cytoskeleton pathways by microRNA-24. Mol Ther. 2014;22:378–89. doi: 10.1038/mt.2013.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Ohyagi-Hara C, Sawada K, Kamiura S, Tomita Y, Isobe A, Hashimoto K, et al. miR-92a inhibits peritoneal dissemination of ovarian cancer cells by inhibiting integrin alpha5 expression. Am J Pathol. 2013;182:1876–89. doi: 10.1016/j.ajpath.2013.01.039. [DOI] [PubMed] [Google Scholar]
  • 192.Chen Z, Wen L, Martin M, Hsu CY, Fang L, Lin FM, et al. Oxidative stress activates endothelial innate immunity via sterol regulatory element binding protein 2 (SREBP2) transactivation of microRNA-92a. Circulation. 2015;131:805–14. doi: 10.1161/CIRCULATIONAHA.114.013675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Chan YC, Khanna S, Roy S, Sen CK. miR-200b targets Ets-1 and is down-regulated by hypoxia to induce angiogenic response of endothelial cells. J Biol Chem. 2011;286:2047–56. doi: 10.1074/jbc.M110.158790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Pecot CV, Rupaimoole R, Yang D, Akbani R, Ivan C, Lu C, et al. Tumour angiogenesis regulation by the miR-200 family. Nat Commun. 2013;4:2427. doi: 10.1038/ncomms3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Liu X, Cheng Y, Yang J, Xu L, Zhang C. Cell-specific effects of miR-221/222 in vessels: molecular mechanism and therapeutic application. J Mol Cell Cardiol. 2012;52:245–55. doi: 10.1016/j.yjmcc.2011.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Nicoli S, Knyphausen CP, Zhu LJ, Lakshmanan A, Lawson ND. miR-221 is required for endothelial tip cell behaviors during vascular development. Dev Cell. 2012;22:418–29. doi: 10.1016/j.devcel.2012.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Poliseno L, Tuccoli A, Mariani L, Evangelista M, Citti L, Woods K, et al. MicroRNAs modulate the angiogenic properties of HUVECs. Blood. 2006;108:3068–71. doi: 10.1182/blood-2006-01-012369. [DOI] [PubMed] [Google Scholar]
  • 198.Patella F, Leucci E, Evangelista M, Parker B, Wen J, Mercatanti A, et al. MiR-492 impairs the angiogenic potential of endothelial cells. J Cell Mol Med. 2013;17:1006–15. doi: 10.1111/jcmm.12085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Welten SM, Bastiaansen AJ, de Jong RC, de Vries MR, Peters EA, Boonstra MC, et al. Inhibition of 14q32 microRNAs miR-329, miR-487b, miR-494, and miR-495 increases neovascularization and blood flow recovery after ischemia. Circ Res. 2014;115:696–708. doi: 10.1161/CIRCRESAHA.114.304747. [DOI] [PubMed] [Google Scholar]
  • 200.Santulli G. Coronary heart disease risk factors and mortality. JAMA. 2012;307:1137. doi: 10.1001/jama.2012.323. [DOI] [PubMed] [Google Scholar]
  • 201.Santulli G. Epidemiology of cardiovascular disease in the 21st century: updated numbers and updated facts. J Cardiovasc Dis (JCvD) 2013;1:1–2. [Google Scholar]
  • 202.Weintraub WS, Daniels SR, Burke LE, Franklin BA, Goff DC, Jr, Hayman LL, et al. Value of primordial and primary prevention for cardiovascular disease: a policy statement from the American Heart Association. Circulation. 2011;124:967–90. doi: 10.1161/CIR.0b013e3182285a81. [DOI] [PubMed] [Google Scholar]
  • 203.Lloyd-Jones D, Adams RJ, Brown TM, Carnethon M, Dai S, De Simone G, et al. Heart disease and stroke statistics–2010 update: a report from the American Heart Association. Circulation. 2010;121:e46–215. doi: 10.1161/CIRCULATIONAHA.109.192667. [DOI] [PubMed] [Google Scholar]
  • 204.Stefanini GG, Holmes DR., Jr Drug-eluting coronary-artery stents. N Engl J Med. 2013;368:254–65. doi: 10.1056/NEJMra1210816. [DOI] [PubMed] [Google Scholar]
  • 205.Rao PS, Thapar MK, Kutayli F. Causes of restenosis after balloon valvuloplasty for valvular pulmonary stenosis. Am J Cardiol. 1988;62:979–82. doi: 10.1016/0002-9149(88)90909-5. [DOI] [PubMed] [Google Scholar]
  • 206.Friedman SG. Charles dotter: interventional radiologist. Radiology. 1989;172:921–4. doi: 10.1148/172.3.921. [DOI] [PubMed] [Google Scholar]
  • 207.Gruentzig AR. Percutaneous transluminal coronary angioplasty. Semin Roentgenol. 1981;16:152–3. doi: 10.1016/0037-198x(81)90049-3. [DOI] [PubMed] [Google Scholar]
  • 208.Meier B, Gruentzig AR, Goebel N, Pyle R, von Gosslar W, Schlumpf M. Assessment of stenoses in coronary angioplasty. Inter- and intraobserver variability. Int J Cardiol. 1983;3:159–69. doi: 10.1016/0167-5273(83)90032-3. [DOI] [PubMed] [Google Scholar]
  • 209.Nobuyoshi M, Kimura T, Nosaka H, Mioka S, Ueno K, Yokoi H, et al. Restenosis after successful percutaneous transluminal coronary angioplasty: serial angiographic follow-up of 229 patients. J Am Coll Cardiol. 1988;12:616–23. doi: 10.1016/s0735-1097(88)80046-9. [DOI] [PubMed] [Google Scholar]
  • 210.Holmes DR, Jr, Vlietstra RE, Smith HC, Vetrovec GW, Kent KM, Cowley MJ, et al. Restenosis after percutaneous transluminal coronary angioplasty (PTCA): a report from the PTCA registry of the National Heart, Lung, and Blood Institute. Am J Cardiol. 1984;53:77C–81. doi: 10.1016/0002-9149(84)90752-5. [DOI] [PubMed] [Google Scholar]
  • 211.Sigwart U, Puel J, Mirkovitch V, Joffre F, Kappenberger L. Intravascular stents to prevent occlusion and restenosis after transluminal angioplasty. N Engl J Med. 1987;316:701–6. doi: 10.1056/NEJM198703193161201. [DOI] [PubMed] [Google Scholar]
  • 212.Serruys PW, de Jaegere P, Kiemeneij F, Macaya C, Rutsch W, Heyndrickx G, et al. A comparison of balloon-expandable-stent implantation with balloon angioplasty in patients with coronary artery disease. Benestent Study Group. N Engl J Med. 1994;331:489–95. doi: 10.1056/NEJM199408253310801. [DOI] [PubMed] [Google Scholar]
  • 213.Fischman DL, Leon MB, Baim DS, Schatz RA, Savage MP, Penn I, et al. A randomized comparison of coronary-stent placement and balloon angioplasty in the treatment of coronary artery disease. Stent Restenosis Study Investigators. N Engl J Med. 1994;331:496–501. doi: 10.1056/NEJM199408253310802. [DOI] [PubMed] [Google Scholar]
  • 214.Marx SO, Totary-Jain H, Marks AR. Vascular smooth muscle cell proliferation in restenosis. Circ Cardiovasc Interv. 2011;4:104–11. doi: 10.1161/CIRCINTERVENTIONS.110.957332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Park YM, Febbraio M, Silverstein RL. CD36 modulates migration of mouse and human macrophages in response to oxidized LDL and may contribute to macrophage trapping in the arterial intima. J Clin Invest. 2009;119:136–45. doi: 10.1172/JCI35535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Suzuki N, Angiolillo DJ, Monteiro C, Shuja S, Futamatsu H, Kawaguchi R, et al. Variable histological and ultrasonic characteristics of restenosis after drug-eluting stents. Int J Cardiol. 2008;130:444–8. doi: 10.1016/j.ijcard.2007.08.143. [DOI] [PubMed] [Google Scholar]
  • 217.Jukema JW, Verschuren JJW, Ahmed TAN, Quax PH. Restenosis after PCI. Part 1: pathophysiology and risk factors. Nat Rev Cardiol. 2012;9:53–62. doi: 10.1038/nrcardio.2011.132. [DOI] [PubMed] [Google Scholar]
  • 218.Marks AR. Sirolimus for the prevention of in-stent restenosis in a coronary artery. N Engl J Med. 2003;349:1307–9. doi: 10.1056/NEJMp038141. [DOI] [PubMed] [Google Scholar]
  • 219.Santulli G, Totary-Jain H. Tailoring mTOR-based therapy: molecular evidence and clinical challenges. Pharmacogenomics. 2013;14:1517–26. doi: 10.2217/pgs.13.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Moses JW, Leon MB, Popma JJ, Fitzgerald PJ, Holmes DR, O’Shaughnessy C, et al. Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery. N Engl J Med. 2003;349:1315–23. doi: 10.1056/NEJMoa035071. [DOI] [PubMed] [Google Scholar]
  • 221.Stone GW, Ellis SG, Cox DA, Hermiller J, O’Shaughnessy C, Mann JT, et al. A polymer-based, paclitaxel-eluting stent in patients with coronary artery disease. N Engl J Med. 2004;350:221–31. doi: 10.1056/NEJMoa032441. [DOI] [PubMed] [Google Scholar]
  • 222.Rollini F, Aprile A, Politi L, Sangiorgi GM. Evaluation of re-endothelization extent at mid-term follow-up after drug eluting balloon plus bare metal stent implantation during primary coronary angioplasty: insight from OCT imaging. Minerva Cardioangiol. 2011;59:109–12. [PubMed] [Google Scholar]
  • 223.Jakabcin J, Bystron M, Spacek R, Veselka J, Kvasnak M, Kala P, et al. The lack of endothelization after drug-eluting stent implantation as a cause of fatal late stent thrombosis. J Thromb Thrombolysis. 2008;26:154–8. doi: 10.1007/s11239-007-0080-2. [DOI] [PubMed] [Google Scholar]
  • 224.Tamburino C, Ussia GP, Zimarino M, Galassi AR, De Caterina R. Early restenosis after drug-eluting stent implantation: a putative role for platelet activation. Can J Cardiol. 2007;23:57–9. doi: 10.1016/s0828-282x(07)70214-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Wijns WC, Krucoff MW. Increased mortality after implantation of first generation drug-eluting stents: seeing the smoke, where is the fire? Eur Heart J. 2006;27:2737–9. doi: 10.1093/eurheartj/ehl378. [DOI] [PubMed] [Google Scholar]
  • 226.Joner M, Finn AV, Farb A, Mont EK, Kolodgie FD, Ladich E, et al. Pathology of drug-eluting stents in humans: delayed healing and late thrombotic risk. J Am Coll Cardiol. 2006;48:193–202. doi: 10.1016/j.jacc.2006.03.042. [DOI] [PubMed] [Google Scholar]
  • 227.Cassese S, Kastrati A. New-generation drug-eluting stents for patients with myocardial infarction. JAMA. 2012;308:814–5. doi: 10.1001/2012.jama.10465. [DOI] [PubMed] [Google Scholar]
  • 228.Moss SC, Lightell DJ, Jr, Marx SO, Marks AR, Woods TC. Rapamycin regulates endothelial cell migration through regulation of the cyclin-dependent kinase inhibitor p27Kip1. J Biol Chem. 2010;285:11991–7. doi: 10.1074/jbc.M109.066621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Lange RA, Hillis LD. Second-generation drug-eluting coronary stents. N Engl J Med. 2010;362:1728–30. doi: 10.1056/NEJMe1001069. [DOI] [PubMed] [Google Scholar]
  • 230.Sun J, Marx SO, Chen HJ, Poon M, Marks AR, Rabbani LE. Role for p27(Kip1) in vascular smooth muscle cell migration. Circulation. 2001;103:2967–72. doi: 10.1161/01.cir.103.24.2967. [DOI] [PubMed] [Google Scholar]
  • 231.Poon M, Marx SO, Gallo R, Badimon JJ, Taubman MB, Marks AR. Rapamycin inhibits vascular smooth muscle cell migration. J Clin Invest. 1996;98:2277–83. doi: 10.1172/JCI119038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Wenaweser P, Daemen J, Zwahlen M, van Domburg R, Juni P, Vaina S, et al. Incidence and correlates of drug-eluting stent thrombosis in routine clinical practice. 4-year results from a large 2-institutional cohort study. J Am Coll Cardiol. 2008;52:1134–40. doi: 10.1016/j.jacc.2008.07.006. [DOI] [PubMed] [Google Scholar]
  • 233.Kipshidze N, Dangas G, Tsapenko M, Moses J, Leon MB, Kutryk M, et al. Role of the endothelium in modulating neointimal formation: vasculoprotective approaches to attenuate restenosis after percutaneous coronary interventions. J Am Coll Cardiol. 2004;44:733–9. doi: 10.1016/j.jacc.2004.04.048. [DOI] [PubMed] [Google Scholar]
  • 234.Yu PJ, Ferrari G, Pirelli L, Gulkarov I, Galloway AC, Mignatti P, et al. Vascular injury and modulation of MAPKs: a targeted approach to therapy of restenosis. Cell Signal. 2007;19:1359–71. doi: 10.1016/j.cellsig.2007.03.002. [DOI] [PubMed] [Google Scholar]
  • 235.Torella D, Gasparri C, Ellison GM, Curcio A, Leone A, Vicinanza C, et al. Differential regulation of vascular smooth muscle and endothelial cell proliferation in vitro and in vivo by cAMP/PKA-activated p85alphaPI3K. Am J Physiol Heart Circ Physiol. 2009;297:H2015–25. doi: 10.1152/ajpheart.00738.2009. [DOI] [PubMed] [Google Scholar]
  • 236.Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A. Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem. 2009;284:3728–38. doi: 10.1074/jbc.M808788200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Cheng Y, Liu X, Yang J, Lin Y, Xu DZ, Lu Q, et al. MicroRNA-145, a novel smooth muscle cell phenotypic marker and modulator, controls vascular neointimal lesion formation. Circ Res. 2009;105:158–66. doi: 10.1161/CIRCRESAHA.109.197517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.McDonald RA, Halliday CA, Miller AM, Diver LA, Dakin RS, Montgomery J, et al. Reducing in-stent restenosis: therapeutic manipulation of miRNA in vascular remodeling and inflammation. J Am Coll Cardiol. 2015;65:2314–27. doi: 10.1016/j.jacc.2015.03.549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.van Rooij E, Sutherland LB, Thatcher JE, DiMaio JM, Naseem RH, Marshall WS, et al. Dysregulation of microRNAs after myocardial infarction reveals a role of miR-29 in cardiac fibrosis. Proc Natl Acad Sci U S A. 2008;105:13027–32. doi: 10.1073/pnas.0805038105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Care A, Catalucci D, Felicetti F, Bonci D, Addario A, Gallo P, et al. MicroRNA-133 controls cardiac hypertrophy. Nat Med. 2007;13:613–8. doi: 10.1038/nm1582. [DOI] [PubMed] [Google Scholar]
  • 241.Lin Z, Murtaza I, Wang K, Jiao J, Gao J, Li PF. miR-23a functions downstream of NFATc3 to regulate cardiac hypertrophy. Proc Natl Acad Sci U S A. 2009;106:12103–8. doi: 10.1073/pnas.0811371106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Bonauer A, Carmona G, Iwasaki M, Mione M, Koyanagi M, Fischer A, et al. MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice. Science. 2009;324:1710–3. doi: 10.1126/science.1174381. [DOI] [PubMed] [Google Scholar]
  • 243.Thum T, Gross C, Fiedler J, Fischer T, Kissler S, Bussen M, et al. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature. 2008;456:980–4. doi: 10.1038/nature07511. [DOI] [PubMed] [Google Scholar]
  • 244.Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature. 2005;438:685–9. doi: 10.1038/nature04303. [DOI] [PubMed] [Google Scholar]
  • 245.Weiler J, Hunziker J, Hall J. Anti-miRNA oligonucleotides (AMOs): ammunition to target miRNAs implicated in human disease? Gene Ther. 2006;13:496–502. doi: 10.1038/sj.gt.3302654. [DOI] [PubMed] [Google Scholar]
  • 246.Stenvang J, Kauppinen S. MicroRNAs as targets for antisense-based therapeutics. Expert Opin Biol Ther. 2008;8:59–81. doi: 10.1517/14712598.8.1.59. [DOI] [PubMed] [Google Scholar]
  • 247.Baumann V, Winkler J. miRNA-based therapies: strategies and delivery platforms for oligonucleotide and non-oligonucleotide agents. Future Med Chem. 2014;6:1967–84. doi: 10.4155/fmc.14.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Zhang Y, Wang Z, Gemeinhart RA. Progress in microRNA delivery. J Control Release. 2013;172:962–74. doi: 10.1016/j.jconrel.2013.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Luck ME, Muljo SA, Collins CB. Prospects for therapeutic targeting of MicroRNAs in human immunological diseases. J Immunol. 2015;194:5047–52. doi: 10.4049/jimmunol.1403146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Muthiah M, Park IK, Cho CS. Nanoparticle-mediated delivery of therapeutic genes: focus on miRNA therapeutics. Expert Opin Drug Deliv. 2013;10:1259–73. doi: 10.1517/17425247.2013.798640. [DOI] [PubMed] [Google Scholar]
  • 251.Puricel S, Arroyo D, Corpataux N, Baeriswyl G, Lehmann S, Kallinikou Z, et al. Comparison of everolimus- and biolimus-eluting coronary stents with everolimus-eluting bioresorbable vascular scaffolds. J Am Coll Cardiol. 2015;65:791–801. doi: 10.1016/j.jacc.2014.12.017. [DOI] [PubMed] [Google Scholar]
  • 252.Sato K, Latib A, Panoulas VF, Kawamoto H, Naganuma T, Miyazaki T, et al. Procedural feasibility and clinical outcomes in propensity-matched patients treated with bioresorbable scaffolds vs new-generation drug-eluting stents. Can J Cardiol. 2015;31:328–34. doi: 10.1016/j.cjca.2014.12.001. [DOI] [PubMed] [Google Scholar]
  • 253.Raungaard B, Jensen LO, Tilsted HH, Christiansen EH, Maeng M, Terkelsen CJ, et al. Zotarolimus-eluting durable-polymer-coated stent versus a biolimus-eluting biodegradable-polymer-coated stent in unselected patients undergoing percutaneous coronary intervention (SORT OUT VI): a randomised non-inferiority trial. Lancet. 2015;385:1527–35. doi: 10.1016/S0140-6736(14)61794-3. [DOI] [PubMed] [Google Scholar]
  • 254.Karanasos A, Simsek C, Gnanadesigan M, van Ditzhuijzen NS, Freire R, Dijkstra J, et al. OCT assessment of the long-term vascular healing response 5 years after everolimus-eluting bioresorbable vascular scaffold. J Am Coll Cardiol. 2014;64:2343–56. doi: 10.1016/j.jacc.2014.09.029. [DOI] [PubMed] [Google Scholar]
  • 255.Muthiah M, Islam MA, Cho CS, Hwang JE, Chung IJ, Park IK. Substrate-mediated delivery of microRNA-145 through a polysorbitol-based osmotically active transporter suppresses smooth muscle cell proliferation: implications for restenosis treatment. J Biomed Nanotechnol. 2014;10:571–9. doi: 10.1166/jbn.2014.1737. [DOI] [PubMed] [Google Scholar]

RESOURCES