Next Article in Journal
Effects of Zinc Oxide Nanoparticle Exposure on Human Glial Cells and Zebrafish Embryos
Previous Article in Journal
Systematic Development of Vanadium Catalysts for Sustainable Epoxidation of Small Alkenes and Allylic Alcohols
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Insights into the Reparative Angiogenesis after Myocardial Infarction

by
Marta Martín-Bórnez
1,2,†,
Débora Falcón
1,2,†,
Rosario Morrugares
1,2,3,
Geraldine Siegfried
4,
Abdel-Majid Khatib
4,
Juan A. Rosado
5,
Isabel Galeano-Otero
1,2,* and
Tarik Smani
1,2,*
1
Group of Cardiovascular Pathophysiology, Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013 Seville, Spain
2
Department of Medical Physiology and Biophysics, Faculty of Medicine, University of Seville, 41009 Seville, Spain
3
Department of Cell Biology, Physiology and Immunology, Universidad de Córdoba, 14071 Córdoba, Spain
4
RyTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615 Pessac, France
5
Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers (IMPB), University of Extremadura, 10003 Caceres, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(15), 12298; https://doi.org/10.3390/ijms241512298
Submission received: 3 July 2023 / Revised: 23 July 2023 / Accepted: 29 July 2023 / Published: 1 August 2023
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)

Abstract

:
Myocardial infarction (MI) causes massive loss of cardiac myocytes and injury to the coronary microcirculation, overwhelming the limited capacity of cardiac regeneration. Cardiac repair after MI is finely organized by complex series of procedures involving a robust angiogenic response that begins in the peri-infarcted border area of the infarcted heart, concluding with fibroblast proliferation and scar formation. Efficient neovascularization after MI limits hypertrophied myocytes and scar extent by the reduction in collagen deposition and sustains the improvement in cardiac function. Compelling evidence from animal models and classical in vitro angiogenic approaches demonstrate that a plethora of well-orchestrated signaling pathways involving Notch, Wnt, PI3K, and the modulation of intracellular Ca2+ concentration through ion channels, regulate angiogenesis from existing endothelial cells (ECs) and endothelial progenitor cells (EPCs) in the infarcted heart. Moreover, cardiac repair after MI involves cell-to-cell communication by paracrine/autocrine signals, mainly through the delivery of extracellular vesicles hosting pro-angiogenic proteins and non-coding RNAs, as microRNAs (miRNAs). This review highlights some general insights into signaling pathways activated under MI, focusing on the role of Ca2+ influx, Notch activated pathway, and miRNAs in EC activation and angiogenesis after MI.

1. Introduction

Coronary artery disease remains one of the main causes of mortality worldwide, being myocardial infarction (MI) the most responsible for these deaths [1,2]. In recent years, remarkable advances in treatments for MI have been achieved thanks to successful and prompt strategies of revascularization, which have led to an improvement in patients’ outcomes and survival [3]. However, adverse consequences resulting from oxygen deficiency due to the impact of ischemia, the loss of cardiomyocytes, and the resulting hypertrophy and fibrosis inevitably provoke the progress of the MI disease toward heart failure [4]. Hence, it remains urgent to search for new therapies capable to efficiently re-establish perfusion and provide oxygen and nutrients to mitigate ischemic damages. During MI, ischemia and healing cardiac tissue trigger a cascade of signaling reactions involving an increase in reactive oxygen species (ROS) and intracellular Ca2+ levels, a dysregulation of endothelial NO synthase (eNOS) [5,6,7], and the activation of hypoxia-induced factor 1α (HIF-1α) required for blood vessels formation from pre-existing vasculature, an action carried out by endothelial cells (ECs) known as angiogenesis [8], as summarized in Figure 1. In addition to the role of ECs in post-infraction angiogenesis, many reports demonstrated that most cell populations of the heart, including cardiomyocytes, macrophages, fibroblasts, and monocytes, participate in this process because they express a high amount of vascular endothelial growth factor (VEGF), the proangiogenic factor par excellence, up to one week after the ischemic event, and other growth factors. Complementarily, ECs close to the infarcted region show an increase in VEGF receptor 2 (VEGFR2) expression [9,10,11], which would explain ECs activation and the starting of new blood vessel formation. Other co-factors might be taken into consideration in cardiac angiogenesis given the high amount of research demonstrating an increase in pro-angiogenic paracrine/autocrine signals after MI due to the action of fibroblast growth factor 2 (FGF2), hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), insulin growth factor-1 (IGF-1) [12,13,14,15,16], hormones (estradiol, estrogen, etc.) [17,18], interleukins 2, 6, 17 (IL2, IL6, IL17), transforming necrosis factor α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) [19,20,21,22]. Likewise, other studies showed an exacerbated traffic of vesicles released by different heart cell populations whose contents include proteins and non-coding RNA, such as microRNAs (miRNAs) [23,24]. All these molecular players trigger a plethora of signaling pathways and changes in the free intracellular Ca2+ concentrations ([Ca2+]i), among other signaling, which fine-tune and orchestrate the complex process of angiogenesis, as illustrated in Figure 1 and recently reviewed [25]. However, the large number of signaling pathways participating in angiogenesis after MI makes it difficult to determine a clear mechanism for an optimal angiogenic response, as discussed recently [10,26]. This review provides an overview of the recent literature showing the role of different signaling pathways and miRNAs in ECs activation and angiogenesis after MI.

2. Reparative Role of Mature and Endothelial Progenitor Cells in the Infarcted Heart

Two different phenotypes of ECs can be distinguished in this process [27]. Tip cells are highly migrative and poorly proliferative vessel wall stalk cells, which have high proliferative and poor migrative rates [28]. Beyond these two phenotypes, a recent study using a single-cell RNA sequencing approach in mice infarcted hearts suggested the presence of up to ten clusters of heart ECs population with different gene expression signatures, suggesting the functional richness of these cells [29]. New capillary formation under MI starts from the border zone and continues to the infarcted necrotic zone, which is considered a reparative process because a correctly harmonized post-ischemic angiogenesis has been associated with a better prognosis of the disease [27,29]. Indeed, early neovascularization of an infarcted heart is supposed to reduce the infarct size and limit the risk of adverse cardiac remodeling, as well as improve cardiomyocytes’ survival and heart function, as demonstrated in the microstructure of rabbit cardiac tissue [30]. However, mature resident ECs are terminally differentiated cells with limited proliferative and migrative potentials; therefore, their role in myocardial infarction angiogenesis is still under debate [31]. However, ECs can be stimulated by a plethora of endothelial progenitor cells (EPCs)-released pro-angiogenic factors such as VEGF, FGF, or IGF, to promote new capillary formation [32]. In the meantime, EPCs are considered the precursors of blood vessels due to their robust angiogenic potential since they have high proliferative and migrative skills required for new blood vessel formation (for review, [33]). The definition of endothelial progenitors has varied widely in the literature [32], but at least two clear subgroups of EPCs have been implicated in post-ischemic neovascularization, myeloid angiogenic cells (MACs), previously known as early outgrowth cells, and endothelial colony-forming cells (ECFCs), also referred to as late EPCs or late outgrowth EPCs [33]. MACs are not able to differentiate into adult ECs, and consequently, they are not capable of forming new vessels themselves. MACs theoretically act by secreting pro-angiogenic mediators to stimulate ECs and recruit more circulating EPCs (Figure 2). For instance, stromal cell-derived factor 1 (SDF-1), through the activation of G protein-coupled receptor CXCR4, mediated bone marrow progenitor cells recruitment to ischemic cardiac tissue during permanent ligation of coronary artery in mice [34]. SDF-1 is a well-studied chemotactic cytokine whose levels increase in the infarcted zone [35], enhancing the homing of mobilized progenitor cells to areas of SDF-1 production. By contrast, ECFCs can differentiate into new mature ECs, promoting angiogenesis [36,37], which could be the most suitable cellular substrate to induce reparative angiogenesis in MI. An early study showed that the administration of ECFCs in a porcine model of AMI decreased the infarct size and increased microvessel density, which attenuated the adverse myocardial remodeling [38]. Another recent study demonstrated that the transplantation of ECFCs combined with mesenchymal stem cells (MSCs) into MI mice with permanent ligation of the left descendent coronary artery significantly improved cardiac function and alleviated the ischemic cardiac damage by increasing capillary density 30 days after AMI, although the underlying signaling pathways of this combined cell-therapy has not been addressed [39]. Likewise, a previous report indicated that the injection of ECFCs and MSCs into the infarcted heart of MI mice promoted a regenerative effect seemingly mediated by the upregulation of multiple paracrine factors associated with angiogenesis such as Ang-2, FGF-2, IGF-1, and SDF-1α [40].
Interestingly, single-cell transcriptome analysis confirmed that resident cardiac but not myeloid EPCs are the main ones responsible for ECs clonal expansion after the ischemic event [41]. In the same vein, the use of hematopoietic and vessel wall EPCs from male patients whose bone marrow transplant was owned by a female donor demonstrated that only endothelial lineages EPCs produced mature ECs [42]. Thus, although circulating EPCs seems to be a promising therapy to directly improve neovascularization after MI [43], nowadays, they are being used mainly as a vector to carry out microRNAs (miRNAs) and other mediators involved in angiogenesis [44,45]. Recently, a nice study revealed that an Ionic solution of calcium silicate bioceramics regulated extracellular vesicles (eVs) synthesis in EPCs, which, when delivered in gelatin methacryloyl-polyethylene glycol microsphere, promoted angiogenesis and improved cardiac function after MI [46]. This study further demonstrated that stimulated EPCs eVs promoted angiogenesis by transferring highly expressed miR-126a-3p to cardiac ECs, which stimulated SDF-1α and CXCR4 expression. Therefore, this study supports the idea that EPCs might require extra stimulation to secrete high-yield bioactive eVs that need to be delivered to the infarct site using stable hydrogel microspheres to effectively promote angiogenesis and heart recovery after MI.

3. Signaling Pathways Participating in the Post-Ischemic Angiogenesis

The role of Notch in angiogenesis cardiac repair is still under debate. Notch pathway plays an important role in both healthy and pathological conditions since it controls angiogenesis and endothelial sprouting, among other processes. Key proteins of this pathway, namely Notch1, Notch4, Jagged1, delta-like 1 and 4 (DLL1 and DLL4), are expressed in ECs preserving their homeostasis and normal function [47]. Many studies demonstrated the role of Notch in neovascularization, stem cell differentiation and fibrosis mitigation after MI [48,49]. Notch1 and associated proteins, Hes1 and Jagged1, are significantly in-creased up to 4 days after MI in cardiomyocytes [50]. Likewise, Notch is associated with MI recovery by stimulating RBP-J signaling pathway, thus limiting ventricular remodeling, and improving cardiac function [51], and improving cardioprotection [52], but also promoting angiogenic processes, as discussed recently [48,52,53,54,55]. For instance, the ad-ministration of an adenovirus overexpressing the Notch intracellular domain (NICD) in rat’s infarcted heart increased VEGF staining and angiogenesis, improving cardiac function [52]. Recently, neovascularization 24h after MI was induced by the administration of extracellular vesicles derived from cardiac MSCs overexpressing NICD-overexpressing in mice [49].
Interestingly, Notch is not a solo player in post-infarction angiogenesis, other signaling pathways synergistically work together with Notch, as VEGF-A [5] or HIF-1α, during MI [56]. Indeed, HIF-1α stimulated MSCs secretion of Jagged1-containing exosomes after MI, activating ECs and inducing angiogenesis, in vitro, as assessed by capillary tube formation, as well as in vivo using a Matrigel assay in athymic nude mice [8]. Notch pathway also crosstalk with the phosphoinositide 3-kinase (PI3K)/Akt signaling pathways after MI, since Notch was activated by PI3K/Akt signaling, meanwhile, Notch itself enhanced the expression of PI3K/Akt signaling in adult myocardium following MI, suggesting a positive survival feedback mechanism between Notch and Akt signaling [50]. Interestingly, main downstream effectors of Akt signaling, eNOS, VEGF, mTOR or FOXO [57,58,59] con-tributed to myocardial angiogenesis. For example, VEGF induced Akt and FOXO3a phosphorylation, limiting its transcriptional activity and enhancing cardiomyocyte survival and native angiogenesis in the post-ischemic environment [60,61].
In addition to those widely studied classical pathways others like Wnt/β-Catenin and JAK/STAT also play a role in post-infarction angiogenesis. As reviewed recently, Wnt signaling is triggered after MI injury, being related to inflammation and angiogenesis [62]. In fact, protein levels of members of Wnt family (Wnt2, Wnt4, Wnt10b, and Wnt11) are increased 5 days following MI in perivascular smooth muscle actin-positive cells and subepicardial ECs, seemingly through endothelial-to-mesenchymal transition mechanism [63]. Moreover, using Wnt signaling reporter in modified mice and rats, it was demonstrated that canonical Wnt signaling increased in the border zone after MI [63,64,65]. In addition, genetically enhanced Wnt10b expression in cardiomyocytes of transgenic mouse boosted arterial formation and attenuated fibrosis in cardiac tissue after the injury [66]. In the case of the role of JAK/STAT3 pathway in neovascularization under MI, STAT3 cardiac specific KO mice exhibited reduced myocardial capillary density and more susceptibility to myocardial ischemia/reperfusion injury, even though no variations in VEGF expression was observed [67]. By contrast, STAT3 activation by Astragaloside IV, a Chinese drug herb that alleviates ischemic heart failure enhanced CD31+ stained cells (EPCs) in ischemic hearts and stimulated HUVEC-tube formation [68]. Similarly, the administration of hyaluronic acid oligosaccharides into MI mice promoted neovessels formation and improved cardiac function as assessed 28 days post-intervention. These beneficial effects were associated with the activation of chemokines expression implicated in macrophage polarization, and the stimulation of MAPK and JAK/STAT signaling pathway for myocardial function reconstruction, as revealed by transcriptomic analyses [69]. These studies suggest that JAK/STAT signaling pathway has a role in cardiac remodeling after cardiac infarction by controlling angiogenesis among others cellular processes as reviewed recently [25].

4. Reactive Oxygen Species Regulation of Angiogenesis

Oxidative stress can be both a cause and consequence of physiological or pathological angiogenesis regulation [70,71]. A recent study used a novel conditional binary transgenic mouse overexpressing the mitochondrial antioxidant isoform of superoxide dismutase in an ECs specific manner to demonstrate that they have increased coronary capillary and arteriolar density in the post-MI ischemic region and improved left ventricle function in MI mice model [72]. This study showed that specific reduction in ECs mitochondrial ROS induced mitochondrial complex I biogenesis, increased proteins associated with oxidative phosphorylation pathways such as COX6A1, NDUFA9, NDUFB1, NDUFV3, NDUVB3 and NDUVB7 in coronary ECs, and induced coronary angiogenesis. By contrast, early reports suggested that a well-controlled oxidative stress is beneficial for angiogenesis during tissue repair because low levels of ROS seemingly promote the production of cytokines and growth factors, such as VEGF through HIF-1α in the injured myocardium, which handled angiogenesis by cell survival, proliferation, and ECs apoptosis regulation [70,71]. ROS production in ECs can be induced by VEGF via NADPH oxidase through Nox2 subunit regulation. In this way, Nox2 knockout (KO) mice showed significant downregulation of pro-angiogenic genes and worse prognosis after MI, as compared to control mice [73]. These data suggest a fine-tune regulation of angiogenesis under oxidative stress.

5. Ca2+ Signaling in Angiogenesis

Ca2+ ion is an important second messenger that regulates many primary functions of ECs, as proliferation, migration, control of permeability, the synthesis and release of vasoactive factors and angiogenesis [74,75,76]. Different studies have demonstrated the contribution of several cationic channels which permeate Ca2+ to angiogenesis, such as transient receptor potential (TRP) and store operated Ca2+ (SOC) channels, using different approaches and transgenic mice [77,78,79,80,81]. Concretely, many studies demonstrated that TRPC and protein related to SOC entry, like Orai1 [82], Orai3 [83], SARAF [78], STIM1 and 2 [82,84] are involved in ECs activation and angiogenesis, but their role in MI angiogenesis has not been revealed yet. Compelling evidence demonstrated that proangiogenic growth factors, such as VEGF, increased [Ca2+]i through the activation of non-excitable Ca2+ channels, modulating the signal transduction pathway leading to angiogenesis [78,85]. In the context of MI, significant increase of Orai isoforms and TRP channels has been observed in peri-infarcted hearts, 1 week after the intervention in rats [6,86]. A recent study demonstrated the role of TRPC1 in post-ischemic angiogenesis using EC-specific TRPC1 KO mice (TRPC1EC−/−) [77]. TRPC1EC−/− mice showed more depressed cardiac function than TRPC1fl/fl mice, as evaluated by a reduced ejection fraction and fractional shortening. Interestingly, the staining of ECs with anti-CD31 confirmed a significant decrease in capillary density in the infarct area of the hearts of TRPC1EC−/− mice. Moreover, the expression of HIF-1α induced by dimethyloxaloylglycine stimulated TRPC1 expression in primary mouse coronary artery ECs and improved cardiac function of TRPC1fl/fl, as compared to TRPC1EC−/− mice after MI, which was associated with an increased capillary density, decreased infarct size and improved ejection fraction. Altogether, this study concludes that TRPC1 stimulation of angiogenesis contributes to the functional recovery of post-infarcted heart [77]. Similarly, TRPC5 channel has been proposed as fair therapeutic candidate that may recover ischemic tissue through angiogenesis [87], because TRPC5 expression together with Orai1 was increased after MI, forming SOC channels [6]. TRPC5 role in angiogenesis has been proved in hind limb ischemia model [88], and an animal model of spinal cord ischemia/reperfusion (I/R) injury [89]. Nevertheless, the impact of TRPC5 on the improvement of myocardial angiogenesis has not been demonstrated. Therefore, the specific role of Ca2+ entry through different isoforms of TRP and SOC channels in the context of reparative angiogenesis after MI still need to be verified.

6. miRNAs as Regulators of Post-Ischemic Angiogenesis

miRNAs play an important role as epigenetic regulators of endothelial function, therefore they were explored as potential therapeutic targets for promoting angiogenesis to improve cardiac repair, as reviewed recently [90]. miRNAs are small non-coding RNAs (∼22 nucleotide) that regulate gene expression by binding mainly to the 3’UTR region of target mRNA. By forming imperfect base-pairing with target mRNA, miRNAs can either cleave the mRNA directly or repress its translation, allowing them to fine-tune gene expression, playing crucial roles in cellular processes such as post-MI angiogenesis [91]. Traffic of microvesicles released from the different heart cell populations containing miRNAs has been detected, supporting cell-to-cell communication under MI [92,93]. In this section, we will summarize the current state of research on miRNA-mediated regulation of angiogenesis following MI through their regulation of key pro-angiogenic signaling pathway as summarized in Table 1.
One of the main gene regulated by miRNA in angiogenesis is HIF-1α which expression is known to increase after myocardial ischemia and regulates the expression of pro-angiogenic genes. An interesting study used rat model for MI to demonstrate that rodent miR-322, homolog of human miR-424, was upregulated in the peri-infarct cardiac tissues [94]. Additional experiments in ECs under hypoxia showed that miR-424 regulated the expression of HIF-1α through its proteasomal degradation process. ECs stimulation by hypoxia increased C/EBPα in conjunction with RUNX-1 to activate the PU.1 promoter, which stimulated the increase of miR-424 level. Moreover, miR-424 targeted CUL2 which destabilized the VCBCR U3-ligase complex, causing translocation of HIF-1α to the nucleus of ECs that stimulated pro-angiogenic genes transcription as VEGF, glucose transporter 1 (GLUT1), and erythropoietin (EPO) [94]. Interestingly, miRNAs might also inhibit angiogenesis through HIF-1α during MI. A recent study suggested that upon the insult of ischemia, exosomes including miR-19a-3p were secreted from cardiomyocytes that are absorbed by ECs, which inhibited their proliferation via HIF-1α downregulation. At the same time, the level of miR-19a-3p were increased in serum of patients with acute MI, and in culture medium of cardiac myocytes stressed with H2O2 protocol to mimic hypoxia [95]. In contrast, downregulation of miR-19a-3p promoted ECs survival and proliferation, and improved heart function by restoring HIF-1α expression. Significant increase in the expression of ECs markers, CD31, was observed in antagomiR-19a-3p transfected mice’s hearts, as compared with non-transfected group after MI, suggesting that the administration of antagomiR-19a-3p enhanced angiogenesis and improved heart function in MI mice by modulating HIF-1α expression [95]. Therefore, these studies suggested that HIF-1α can be modulated by miRNAs in ECs either positively or negatively which will affect the onset of angiogenesis after MI.
VEGF pathway is also regulated by miRNAs in heart, as reviewed elsewhere [96]. Independent studies focused on miR-126 as the main regulator of physiological angiogenesis since it has been demonstrated that miR-126 represses negative regulators of VEGF pathway, such as sprouty-related protein 1 (SPRED1) and PI3K regulatory subunit 2 (PIK3R2). Therefore, miR-126 action enhanced ECs response to VEGF, improving angiogenesis [97]. Its implication in angiogenesis following MI also has been demonstrated by the intramyocardial injection of MSCs overexpressing miR-126 into infracted area, which enhanced microvessel formation and increased blood flow to the infarcted and peri-infarcted zones. This study also determined that this treatment increased the level of proteins involved in VEGF pathway and Notch signaling in MSCs, specifically VEGF, basic FGF and DLL4 [98]. Furthermore, the transplantation of EPCs overexpressing miR-126-3p in an ischemic cardiomyopathy rat model created by left coronary artery ligation, stimulated angiogenesis, and improved cardiac function, which correlated with the alteration of pro-angiogenic cytokine expression, as VEGF-A, IL-10, IL-3, IGF-1, and angiogenin [99]. Interestingly, an early study has shown that miR-126 and miR-130a are downregulated in angiogenic MACs from patients with chronic heart failure (CHF) [100]. Meanwhile, the administration of MACs from patients with CHF transfected with miR-126 and miR-130a in nude mice with MI improved cardiac contractility and enhanced capillary density, indicating efficient neovascularization and cardiac repair under these conditions. In vitro, those MACs transfected with miR-126 or miR-130a mimics markedly enhanced their capacity to promote HUVECs-tube formation, an action associated with SPRED1 downregulation [100]. miR-130a seems also involved in the regulation of VEGF and PI3K/Akt signaling pathways. In fact, the administration of lentivirus expressing miR-130a into mouse hearts seven days before they were subjected to MI attenuated cardiac dysfunction and improved remodeling, which was associated with an enhanced microvascular density. Using in vitro wound healing assay, it was confirmed that miR-130a stimulated HUVEC migration, involving PTEN suppression, PIP3/Akt pathway activation, and VEGF levels’ increase [101]. VEGF is also a target gene of miR-375, which levels are increased both in rodent models of MI and in patients with heart failure. Knockdown of miR-375 enhanced VEGF expression in the infarcted myocardium, attenuated MI-induced inflammation, cardiomyocyte apoptosis and restored left ventricle function and remodeling. Moreover, the effect of miR-375 knockdown increased PDK-1 expression and downstream survival signaling AKT in cardiac ECs, cardiac myocytes, and macrophages [102]. A recent study, provided evidence showing that significant downregulation of miR-21 in post-MI hearts of mouse model injected with cardiac stromal cells from patients with heart failure. Importantly, restoring miR-21-5p expression contributed to heart repair by enhancing angiogenesis and cardiomyocyte survival through PTEN inhibition, which triggered the activation of Akt kinase activity and promoted the expression of VEGF in ECs [103]. PTEN signaling pathway activation of angiogenesis was also regulated by miR-499-5p stimulated by Tanshionone IIA, the main active monomer compound of Danshen, injected in the peri-infarcted mice heart, which improved cardiac function after MI by increasing the expression of VEGF and angiotensin-1, activating angiogenesis [104]. Many other miRNAs boosted neovascularization by targeting other non-classical path-ways in ECs. For example, miR-378 enhanced angiogenic capacity of CD34+ cells by targeting suppressor of fused (SuFu) and Fus-1 expression, which trended to upregulate pro-angiogenesis factors [105]. By contrast, other miRNAs have anti-angiogenic role and appear reduced after MI such as miR-185-5p, miR-143 and miR-92a, which target CatK, IGF signaling pathway and integrin subunit alpha5 (ITGA5), respectively [106,107,108,109], or miR-24 that regulates PAK4, GATA2 [110], and eNOS [111], suggesting the use of encapsuled antagomiR as therapeutic strategy to improve angiogenesis and heart function after MI.
In several studies, exosomes have been used as carriers delivering miRNAs, as mentioned above. Compelling evidence have shown that secreted exosomes from myocardial cells containing miR-125b, miR-126, miR-25-3p, miR-144, or miR-146a are associated with ischemic cardiac repair [23,24,112,113,114]. For example, exosomes derived from the peripheral serum of patients with acute MI containing significant amount of miR-126-3p, as compared to exosome isolated from healthy volunteers [115], efficiently increased ECs proliferation and microvessel sprouting by the activation of mTORC1/HIF-1α axis [115]. Similarly, exosomes derived from DEXs dendritic cells, which infiltrated into infarcted area following MI, overexpress miR-494-3p that enhanced cardiac microvascular ECs tube formation in vitro and angiogenesis, as demonstrated by significant increase of CD31+ marked cells in the infarcted myocardium [116]. Furthermore, the injection of exosomes derived from cardiac progenitor cells containing miR-322 in MI mice reduced the infarct size, as well as increased capillary density in heart tissue. These exosomes increased HUVEC tube formation and migration by increasing Nox2-derived ROS in vitro [117]. In a similar way, MSC-derived extracellular vesicles (MSC-EVs) enriched with miR-210 inhibited cardiac fibrosis, activated ECs and increased the number of capillaries in the peri-infarct regions of the post-MI mice heart [118]. The effect of MSC-EVs overexpressing miR-210 was induced by the inhibition of Efna3, a GPI-anchored membrane protein, as demonstrated in HUVEC [118]. This cardioprotective role of miR-210 was supported by another study which confirmed that overexpressing miR-210 promoted angiogenesis in adult transgenic mice following MI [119]. Another report investigated cardiac telocyte interstitial cells and ECs communication demonstrating that the delivery of cardiac telocytes to infarcted hearts increased vessels density thanks to the release of exosomal miR-21-5p, which modulated cell death inducing p53 target 1 (Cdip1) gene and downregulates caspase-3 expression [120].
Table 1. miRNAs, targets genes, and pathways involved in post-myocardial infarction angiogenesis activated in endothelial cells. ↑: promotes; ↓: prevents. CatK: cathepsin K; CUL2: cullin 2; MACs: Myeloid angiogenic cells; Efna3: ephrin A3; eNOS: endothelial NO synthase; HIF-1α: hypoxia-inducible factor 1α; IGF-IR: insulin-like growth factor receptor 1; PDK-1: phosphoinositide-dependent protein kinase 1; PIK3R2: PI3K regulatory subunit 2; PTEN: phosphatase and tensin homolog; PAK4: p21 activated kinases 4; ITGA5: integrin subunit α 5; miRNA: microRNA; MSCs: mesenchymal stem cells; MSC-EVs: mesenchymal stem cells extra vesicles; SPRED1: sprouty-related protein 1; SuFu: suppressor of fused; VEGF: vascular endothelial growth factor.
Table 1. miRNAs, targets genes, and pathways involved in post-myocardial infarction angiogenesis activated in endothelial cells. ↑: promotes; ↓: prevents. CatK: cathepsin K; CUL2: cullin 2; MACs: Myeloid angiogenic cells; Efna3: ephrin A3; eNOS: endothelial NO synthase; HIF-1α: hypoxia-inducible factor 1α; IGF-IR: insulin-like growth factor receptor 1; PDK-1: phosphoinositide-dependent protein kinase 1; PIK3R2: PI3K regulatory subunit 2; PTEN: phosphatase and tensin homolog; PAK4: p21 activated kinases 4; ITGA5: integrin subunit α 5; miRNA: microRNA; MSCs: mesenchymal stem cells; MSC-EVs: mesenchymal stem cells extra vesicles; SPRED1: sprouty-related protein 1; SuFu: suppressor of fused; VEGF: vascular endothelial growth factor.
miRNAReleased or Delivered byTarget GenesPromotes/
Prevents
References
miR-322/miR-424ECsCUL2/VEGF/GLUT1/EPO/HIF-1α[94]
miR-19a-3pCardiomyocytesHIF-1α[95]
miR-126-3pMSCs/exosomesPIK3R2/SPRED1/VEGF/bFGF/DLL4/
mTORC1/HIF-1α
[97,98]
EPCsVEGF-A/IL-10/IL-3/IGF-1/angiogenin[99,115]
miR126/miR-130aMACsSPRED1[100]
miR-130aLentivirusVEGF/HoxA5/AKT[101]
PTEN
miR-375LNA anti-miR-375PDK-1/AKT[102]
miR-21-5pCardiac stromal cellsPTEN/AKT↓/↑[103]
miR-499-5pTanshinone IIA administrationPTEN/VEGF/angiotensin-1[104]
miR-210MSC-EVs; transgenic miceEfna3/VEGF[118,119]
miR-378CD34+ progenitor cellsSuFu/Fus-1[105]
miR-185-5pECsCatK[107]
miR-143CardiomyocytesIGF-1R[108]
miR-92aECsITGA5[109]
miR-24encapsuled antagomiRPAK4, GATA, eNOS[110,111]
Altogether, these studies suggest that pro- or anti-angiogenic miRNAs target multiple signaling pathways involved in angiogenesis. Nevertheless, the reason why the ischemic insult can stimulate miRNAs with the opposite effect is still undiscovered, which is worth to be clarified.

7. Conclusions

Neovascularization within the infarcted tissue is an integral critical component of the cardiac remodeling process. The formation of the new dense capillary network would favor gas exchange, nutrient diffusion, and waste removal, which would attenuate cardiac myocyte dysfunction in the peri-infarcted zone. Understanding the complex mechanisms behind angiogenesis after MI may create multiple therapeutic opportunities. As summarized in Figure 1, in this review, we briefly highlight multiple signaling events implicated in this process, including classical pathways Notch, Wnt, and PI3K, and we discussed the role of endothelial progenitor cells [Ca2+]i and miRNAs that regulate a wide range of genes implicated in EC proliferation, migration, and tube formation, the hallmarks of angiogenesis. Several miRNAs might be attractive candidates for modulating certain therapeutic targets in MI, although more work is still required to explore new targets and to develop efficient delivery vehicles into the infarcted heart.

Author Contributions

Conceptualization, T.S., M.M.-B., D.F. and I.G.-O.; writing—original draft preparation, I.G.-O., M.M.-B., D.F. and R.M.; writing—review and editing, T.S., G.S., A.-M.K. and J.A.R.; supervision, T.S.; funding acquisition, T.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by Grants: PID2019-104084GB-C22 funded by MCIN/ AEI /10.13039/501100011033, and by the Andalusian Government [grants number: ProyExcel_00530; US-1381135], and FEDER Funds. RM was supported by the “Margarita Salas” postdoctoral grant, convened by Universidad de Córdoba (UCO), and funded by the Ministerio de Universidades, Spain, and the European Union-Next Generation EU. Debora Falcon is recipient of contract from PAIDI 2020 program financed by the European Social Fund and the Andalusian Government.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Isabel Galeano-Otero is awarded by the Postdoctoral Fellowship “Josy Reiffers” of “La Fondation Bergonié”.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

CHF: chronic heart failure; DEXs: dendritic cells; ECs: endothelial cells; MACs: Myeloid angiogenic cells; eNOS: endothelial nitric oxide synthase; EPC: endothelial progenitor cells; HUVEC: human umbilical vein endothelial cells; HIF-1α: hypoxia-induced factor 1α; IGF-1: insulin growth factor-1; IL: interleukin; [Ca2+]i: intracellular Ca2+ concentration, MSCs: mesenchymal stem cells; miRNAs: microRNAs; MI: myocardial infarction; NF-κβ: nuclear factor κβ; PDGF: platelet-derived growth factor; SRF: serum response factor; SOCE: store-operated Ca2+ entry; TNF-α: transforming necrosis factor α; TRP: transient receptor potential; TRPC: TRP canonical; VEGF-A: vascular endothelial growth factor A; VEGFR1: VEGF receptor 1; VEGFR2: VEGF receptor 2.

References

  1. Brown, J.C.; Gerhardt, T.E.; Kwon, E. Risk Factors for Coronary Artery Disease. Risk Factors in Coronary Artery Disease; CRC Press: Boca Raton, FL, USA, 2006; pp. 1–219. [Google Scholar] [CrossRef]
  2. Cardiovascular Diseases. Available online: https://www.who.int/health-topics/cardiovascular-diseases#tab=tab_1 (accessed on 8 December 2022).
  3. Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disease and Stroke Statistics-2022 Update: A Report from the American Heart Association. Circulation 2022, 145, e153–e639. [Google Scholar] [CrossRef] [PubMed]
  4. Li, J.; Zhao, Y.; Zhu, W. Targeting Angiogenesis in Myocardial Infarction: Novel Therapeutics (Review). Exp. Ther. Med. 2021, 23, 64. [Google Scholar] [CrossRef]
  5. Zou, J.; Fei, Q.; Xiao, H.; Wang, H.; Liu, K.; Liu, M.; Zhang, H.; Xiao, X.; Wang, K.; Wang, N. VEGF-A Promotes Angiogenesis after Acute Myocardial Infarction through Increasing ROS Production and Enhancing ER Stress-Mediated Autophagy. J. Cell Physiol. 2019, 234, 17690–17703. [Google Scholar] [CrossRef] [PubMed]
  6. Domínguez-Rodríguez, A.; Mayoral-Gonzalez, I.; Avila-Medina, J.; de Rojas-de Pedro, E.S.R.; Calderón-Sánchez, E.; Díaz, I.; Hmadcha, A.; Castellano, A.; Rosado, J.A.; Benitah, J.P.; et al. Urocortin-2 Prevents Dysregulation of Ca2+ Homeostasis and Improves Early Cardiac Remodeling After Ischemia and Reperfusion. Front. Physiol. 2018, 9, 813. [Google Scholar] [CrossRef] [PubMed]
  7. Nauli, S.M. Endothelial Nitric Oxide Synthase (ENOS) and the Cardiovascular System: In Physiology and in Disease States. Am. J. Biomed. Sci. Res. 2022, 15, 155–179. [Google Scholar] [CrossRef]
  8. Gonzalez-King, H.; García, N.A.; Ontoria-Oviedo, I.; Ciria, M.; Montero, J.A.; Sepúlveda, P. Hypoxia Inducible Factor-1α Potentiates Jagged 1-Mediated Angiogenesis by Mesenchymal Stem Cell-Derived Exosomes. Stem Cells 2017, 35, 1747–1759. [Google Scholar] [CrossRef]
  9. Ferraro, B.; Leoni, G.; Hinkel, R.; Ormanns, S.; Paulin, N.; Ortega-Gomez, A.; Viola, J.R.; de Jong, R.; Bongiovanni, D.; Bozoglu, T.; et al. Pro-Angiogenic Macrophage Phenotype to Promote Myocardial Repair. J. Am. Coll. Cardiol. 2019, 73, 2990–3002. [Google Scholar] [CrossRef]
  10. Kobayashi, K.; Maeda, K.; Takefuji, M.; Kikuchi, R.; Morishita, Y.; Hirashima, M.; Murohara, T. Dynamics of Angiogenesis in Ischemic Areas of the Infarcted Heart. Sci. Rep. 2017, 7, 7156. [Google Scholar] [CrossRef]
  11. Loyer, X.; Zlatanova, I.; Devue, C.; Yin, M.; Howangyin, K.Y.; Klaihmon, P.; Guerin, C.L.; Khelouf, M.; Vilar, J.; Zannis, K.; et al. Intra-Cardiac Release of Extracellular Vesicles Shapes Inflammation Following Myocardial Infarction Short Communication. Circ. Res. 2018, 123, 100–106. [Google Scholar] [CrossRef]
  12. Virag, J.A.I.; Rolle, M.L.; Reece, J.; Hardouin, S.; Feigl, E.O.; Murry, C.E. Fibroblast Growth Factor-2 Regulates Myocardial Infarct Repair: Effects on Cell Proliferation, Scar Contraction, and Ventricular Function. Am. J. Pathol. 2007, 171, 1431–1440. [Google Scholar] [CrossRef]
  13. Ono, K.; Matsumori, A.; Shioi, T.; Furakawa, Y.; Sasayama, S. Enhanced Expression of Hepatocyte Growth Factor/c-Met by Myocardial Ischemia and Reperfusion in a Rat Model. Circulation 1997, 95, 2552–2558. [Google Scholar] [CrossRef] [PubMed]
  14. Tomita, N.; Morishita, R.; Taniyama, Y.; Koike, H.; Aoki, M.; Shimizu, H.; Matsumoto, K.; Nakamura, T.; Kaneda, Y.; Ogihara, T. Angiogenic Property of Hepatocyte Growth Factor Is Dependent on Upregulation of Essential Transcription Factor for Angiogenesis, Ets-1. Circulation 2003, 107, 1411–1417. [Google Scholar] [CrossRef] [PubMed]
  15. Zhao, W.; Zhao, T.; Huang, V.; Chen, Y.; Ahokas, R.A.; Sun, Y. Platelet-Derived Growth Factor Involvement in Myocardial Remodeling Following Infarction. J. Mol. Cell Cardiol. 2011, 51, 830. [Google Scholar] [CrossRef] [PubMed]
  16. Dobrucki, L.W.; Tsutsumi, Y.; Kalinowski, L.; Dean, J.; Gavin, M.; Sen, S.; Mendizabal, M.; Sinusas, A.J.; Aikawa, R. Analysis of Angiogenesis Induced by Local IGF-1 Expression after Myocardial Infarction Using MicroSPECT-CT Imaging. J. Mol. Cell Cardiol. 2011, 48, 1071–1079. [Google Scholar] [CrossRef] [PubMed]
  17. Iwakura, A.; Shastry, S.; Luedemann, C.; Hamada, H.; Kawamoto, A.; Kishore, R.; Zhu, Y.; Qin, G.; Silver, M.; Thorne, T.; et al. Estradiol Enhances Recovery After Myocardial Infarction by Augmenting Incorporation of Bone Marrow–Derived Endothelial Progenitor Cells into Sites of Ischemia-Induced Neovascularization via Endothelial Nitric Oxide Synthase–Mediated Activation of Matrix Metalloproteinase-9. Circulation 2006, 113, 1605–1614. [Google Scholar] [CrossRef] [PubMed]
  18. Barnabas, O.; Wang, H.; Gao, X.M. Role of Estrogen in Angiogenesis in Cardiovascular Diseases. J. Geriatr. Cardiol. 2013, 10, 377–382. [Google Scholar] [CrossRef]
  19. Bouchentouf, M.; Williams, P.; Forner, K.A.; Cuerquis, J.; Michaud, V.; Paradis, P.; Schiffrin, E.L.; Galipeau, J. Interleukin-2 Enhances Angiogenesis and Preserves Cardiac Function Following Myocardial Infarction. Cytokine 2011, 56, 732–738. [Google Scholar] [CrossRef]
  20. Mayfield, A.E.; Kanda, P.; Nantsios, A.; Parent, S.; Mount, S.; Dixit, S.; Ye, B.; Seymour, R.; Stewart, D.J.; Davis, D.R. Interleukin-6 Mediates Post-Infarct Repair by Cardiac Explant-Derived Stem Cells. Theranostics 2017, 7, 4850–4861. [Google Scholar] [CrossRef]
  21. Kishore, R.; Tkebuchava, T.; Sasi, S.P.; Silver, M.; Gilbert, H.Y.; Yoon, Y.S.; Park, H.Y.; Thorne, T.; Losordo, D.W.; Goukassian, D.A. Tumor Necrosis Factor-α Signaling via TNFR1/P55 Is Deleterious Whereas TNFR2/P75 Signaling Is Protective in Adult Infarct Myocardium. Adv. Exp. Med. Biol. 2011, 691, 433–448. [Google Scholar] [CrossRef]
  22. Xia, Y.; Frangogiannis, N.G. MCP-1/CCL2 as a Therapeutic Target in Myocardial Infarction and Ischemic Cardiomyopathy. Inflamm. Allergy Drug Targets 2007, 6, 101–107. [Google Scholar] [CrossRef]
  23. Wang, W.; Zheng, Y.; Wang, M.; Yan, M.; Jiang, J.; Li, Z. Exosomes Derived MiR-126 Attenuates Oxidative Stress and Apoptosis from Ischemia and Reperfusion Injury by Targeting ERRFI1. Gene 2019, 690, 75–80. [Google Scholar] [CrossRef]
  24. Zhu, L.P.; Tian, T.; Wang, J.Y.; He, J.N.; Chen, T.; Pan, M.; Xu, L.; Zhang, H.X.; Qiu, X.T.; Li, C.C.; et al. Hypoxia-Elicited Mesenchymal Stem Cell-Derived Exosomes Facilitates Cardiac Repair through MiR-125b-Mediated Prevention of Cell Death in Myocardial Infarction. Theranostics 2018, 8, 6163–6177. [Google Scholar] [CrossRef] [PubMed]
  25. Zhang, Q.; Wang, L.; Wang, S.; Cheng, H.; Xu, L.; Pei, G.; Wang, Y.; Fu, C.; Jiang, Y.; He, C.; et al. Signaling Pathways and Targeted Therapy for Myocardial Infarction. Signal Transduct. Target. Ther. 2022, 7, 78. [Google Scholar] [CrossRef] [PubMed]
  26. Johnson, T.; Zhao, L.; Manuel, G.; Taylor, H.; Liu, D. Approaches to Therapeutic Angiogenesis for Ischemic Heart Disease. J. Mol. Med. 2019, 97, 141–151. [Google Scholar] [CrossRef] [PubMed]
  27. Zheng, J.; Chen, P.; Zhong, J.; Cheng, Y.; Chen, H.; He, Y.; Chen, C. HIF-1α in Myocardial Ischemia-Reperfusion Injury. Mol. Med. Rep. 2021, 23, 352. [Google Scholar] [CrossRef]
  28. Blanco, R.; Gerhardt, H. VEGF and Notch in Tip and Stalk Cell Selection. Cold Spring Harb. Perspect. Med. 2013, 3, a006569. [Google Scholar] [CrossRef]
  29. Wu, X.; Reboll, M.R.; Korf-Klingebiel, M.; Wollert, K.C. Angiogenesis after Acute Myocardial Infarction. Cardiovasc. Res. 2021, 117, 1257–1273. [Google Scholar] [CrossRef]
  30. Seidel, T.; Edelmann, J.-C.; Sachse, F.B. Analyzing Remodeling of Cardiac Tissue: A Comprehensive Approach Based on Confocal Microscopy and 3D Reconstructions. Annu. Biomed. Eng. 2016, 44, 1436–1448. [Google Scholar] [CrossRef]
  31. Jujo, K.; Ii, M.; Losordo, D.W. Endothelial Progenitor Cells in Neovascularization of Infarcted Myocardium. J. Mol. Cell Cardiol. 2008, 45, 530–544. [Google Scholar] [CrossRef]
  32. Ratajczak, J.; Kucia, M.; Mierzejewska, K.; Marlicz, W.; Pietrzkowski, Z.; Wojakowski, W.; Greco, N.J.; Tendera, M.; Ratajczak, M.Z. Paracrine Proangiopoietic Effects of Human Umbilical Cord Blood-Derived Purified CD133+ Cells-Implications for Stem Cell Therapies in Regenerative Medicine. Stem Cells Dev. 2013, 22, 422–430. [Google Scholar] [CrossRef]
  33. Huang, H.; Huang, W. Regulation of Endothelial Progenitor Cell Functions in Ischemic Heart Disease: New Therapeutic Targets for Cardiac Remodeling and Repair. Front. Cardiovasc. Med. 2022, 9, 896782. [Google Scholar] [CrossRef] [PubMed]
  34. Cheng, M.; Huang, K.; Zhou, J.; Yan, D.; Tang, Y.L.; Zhao, T.C.; Miller, R.J.; Kishore, R.; Losordo, D.W.; Qin, G. A Critical Role of Src Family Kinase in SDF-1/CXCR4-Mediated Bone-Marrow Progenitor Cell Recruitment to the Ischemic Heart. J. Mol. Cell Cardiol. 2015, 81, 49–53. [Google Scholar] [CrossRef] [PubMed]
  35. Abbott, J.D.; Huang, Y.; Liu, D.; Hickey, R.; Krause, D.S.; Giordano, F.J. Stromal Cell-Derived Factor-1α Plays a Critical Role in Stem Cell Recruitment to the Heart after Myocardial Infarction but Is Not Sufficient to Induce Homing in the Absence of Injury. Circulation 2004, 110, 3300–3305. [Google Scholar] [CrossRef] [PubMed]
  36. Medina, R.J.; Barber, C.L.; Sabatier, F.; Dignat-George, F.; Melero-Martin, J.M.; Khosrotehrani, K.; Ohneda, O.; Randi, A.M.; Chan, J.K.Y.; Yamaguchi, T.; et al. Endothelial Progenitors: A Consensus Statement on Nomenclature. Stem Cells Transl. Med. 2017, 6, 1316–1320. [Google Scholar] [CrossRef]
  37. Balducci, V.; Faris, P.; Balbi, C.; Costa, A.; Negri, S.; Rosti, V.; Bollini, S.; Moccia, F. The Human Amniotic Fluid Stem Cell Secretome Triggers Intracellular Ca2+ Oscillations, NF-ΚB Nuclear Translocation and Tube Formation in Human Endothelial Colony-Forming Cells. J. Cell Mol. Med. 2021, 25, 8074–8086. [Google Scholar] [CrossRef]
  38. Dubois, C.; Liu, X.; Claus, P.; Marsboom, G.; Pokreisz, P.; Vandenwijngaert, S.; Dépelteau, H.; Streb, W.; Chaothawee, L.; Maes, F.; et al. Differential Effects of Progenitor Cell Populations on Left Ventricular Remodeling and Myocardial Neovascularization After Myocardial Infarction. J. Am. Coll. Cardiol. 2010, 55, 2232–2243. [Google Scholar] [CrossRef]
  39. Tripathi, H.; Domingues, A.; Donahue, R.; Cras, A.; Guerin, C.L.; Gao, E.; Levitan, B.; Ratajczak, M.Z.; Smadja, D.M.; Abdel-Latif, A.; et al. Combined Transplantation of Human MSCs and ECFCs Improves Cardiac Function and Decrease Cardiomyocyte Apoptosis After Acute Myocardial Infarction. Stem Cell Rev. Rep. 2023, 19, 573–577. [Google Scholar] [CrossRef]
  40. Kim, S.W.; Jin, H.L.; Kang, S.M.; Kim, S.; Yoo, K.J.; Jang, Y.; Kim, H.O.; Yoon, Y.S. Therapeutic Effects of Late Outgrowth Endothelial Progenitor Cells or Mesenchymal Stem Cells Derived from Human Umbilical Cord Blood on Infarct Repair. Int. J. Cardiol. 2016, 203, 498–507. [Google Scholar] [CrossRef]
  41. Li, Z.; Solomonidis, E.G.; Meloni, M.; Taylor, R.S.; Duffin, R.; Dobie, R.; Magalhaes, M.S.; Henderson, B.E.P.; Louwe, P.A.; D’Amico, G.; et al. Single-Cell Transcriptome Analyses Reveal Novel Targets Modulating Cardiac Neovascularization by Resident Endothelial Cells Following Myocardial Infarction. Eur. Heart J. 2019, 40, 2507–2520. [Google Scholar] [CrossRef]
  42. Fujisawa, T.; Tura-Ceide, O.; Hunter, A.; Mitchell, A.; Vesey, A.; Medine, C.; Gallogly, S.; Hadoke, P.W.F.; Keith, C.; Sproul, A.; et al. Endothelial Progenitor Cells Do Not Originate from the Bone Marrow. Circulation 2019, 140, 1524–1526. [Google Scholar] [CrossRef]
  43. Lin, Y.; Weisdorf, D.J.; Solovey, A.; Hebbel, R.P. Origins of Circulating Endothelial Cells and Endothelial Outgrowth from Blood. J. Clin. Investig. 2000, 105, 71–77. [Google Scholar] [CrossRef] [PubMed]
  44. Huang, Y.; Chen, L.; Feng, Z.; Chen, W.; Yan, S.; Yang, R.; Xiao, J.; Gao, J.; Zhang, D.; Ke, X. EPC-Derived Exosomal MiR-1246 and MiR-1290 Regulate Phenotypic Changes of Fibroblasts to Endothelial Cells to Exert Protective Effects on Myocardial Infarction by Targeting ELF5 and SP1. Front. Cell Dev. Biol. 2021, 9, 647763. [Google Scholar] [CrossRef] [PubMed]
  45. Ke, X.; Yang, R.; Wu, F.; Wang, X.; Liang, J.; Hu, X.; Hu, C. Exosomal MiR-218-5p/MiR-363-3p from Endothelial Progenitor Cells Ameliorate Myocardial Infarction by Targeting the P53/JMY Signaling Pathway. Oxid. Med. Cell Longev. 2021, 2021, 5529430. [Google Scholar] [CrossRef]
  46. Yu, B.; Li, H.; Zhang, Z.; Chen, P.; Wang, L.; Fan, X.; Ning, X.; Pan, Y.; Zhou, F.; Hu, X.; et al. Extracellular Vesicles Engineering by Silicates-Activated Endothelial Progenitor Cells for Myocardial Infarction Treatment in Male Mice. Nat. Commun. 2023, 14, 2094. [Google Scholar] [CrossRef]
  47. Mack, J.J.; Mosqueiro, T.S.; Archer, B.J.; Jones, W.M.; Sunshine, H.; Faas, G.C.; Briot, A.; Aragón, R.L.; Su, T.; Romay, M.C.; et al. NOTCH1 Is a Mechanosensor in Adult Arteries. Nat. Commun. 2017, 8, 1620. [Google Scholar] [CrossRef] [PubMed]
  48. Huang, Z.; Rivas, B.; Agoulnik, A.I. NOTCH1 Gain of Function in Germ Cells Causes Failure of Spermatogenesis in Male Mice. PLoS ONE 2013, 8, e71213. [Google Scholar] [CrossRef] [PubMed]
  49. Xuan, W.; Khan, M.; Ashraf, M. Extracellular Vesicles from Notch Activated Cardiac Mesenchymal Stem Cells Promote Myocyte Proliferation and Neovasculogenesis. Front. Cell Dev. Biol. 2020, 8, 11. [Google Scholar] [CrossRef]
  50. Gude, N.A.; Emmanuel, G.; Wu, W.; Cottage, C.T.; Fischer, K.; Quijada, P.; Muraski, J.A.; Alvarez, R.; Rubio, M.; Schaefer, E.; et al. Activation of Notch-Mediated Protective Signaling in the Myocardium. Circ. Res. 2008, 102, 1025–1035. [Google Scholar] [CrossRef]
  51. He, Y.; Pang, S.; Huang, J.; Zhu, K.; Tong, J.; Tang, Y.; Ma, G.; Chen, L. Blockade of RBP-J-Mediated Notch Signaling Pathway Exacerbates Cardiac Remodeling after Infarction by Increasing Apoptosis in Mice. Biomed. Res. Int. 2018, 2018, 5207031. [Google Scholar] [CrossRef]
  52. Zhou, X.-L.; Zhu, R.-R.; Liu, S.; Xu, H.; Xu, X.; Wu, Q.-C.; Liu, J.-C. Notch Signaling Promotes Angiogenesis and Improves Cardiac Function after Myocardial Infarction. J. Cell Biochem. 2018, 119, 7105–7112. [Google Scholar] [CrossRef]
  53. Li, Y.; Hiroi, Y.; Liao, J.K. Notch Signaling Negatively Regulates Cardiac Differentiation (Nemir, 2006). Trends Cardiovasc. Med. 2010, 20, 228–231. [Google Scholar] [CrossRef] [PubMed]
  54. Fan, J.; Shen, W.; Lee, S.R.; Mathai, A.E.; Zhang, R.; Xu, G.; Gillies, M.C. Targeting the Notch and TGF-β Signaling Pathways to Prevent Retinal Fibrosis in Vitro and in Vivo. Theranostics 2020, 10, 7956–7973. [Google Scholar] [CrossRef]
  55. Crovella, S.; Ticarico, P.M.; Kachanova, O.; Lobov, A.; Malashicheva, A. The Role of the Notch Signaling Pathway in Recovery of Cardiac Function after Myocardial Infarction. Int. J. Mol. Sci. 2022, 23, 12509. [Google Scholar] [CrossRef]
  56. Wang, K.; Ding, R.; Ha, Y.; Jia, Y.; Liao, X.; Wang, S.; Li, R.; Shen, Z.; Xiong, H.; Guo, J.; et al. Hypoxia-Stressed Cardiomyocytes Promote Early Cardiac Differentiation of Cardiac Stem Cells through HIF-1 α/Jagged1/Notch1 Signaling. Acta Pharm. Sin. B 2018, 8, 795–804. [Google Scholar] [CrossRef]
  57. Sciarretta, S.; Forte, M.; Frati, G.; Sadoshima, J. New Insights into the Role of MTOR Signaling in the Cardiovascular System. Circ. Res. 2018, 122, 489–505. [Google Scholar] [CrossRef] [PubMed]
  58. Kazakov, A.; Hall, R.; Jagoda, P.; Bachelier, K.; Müller-Best, P.; Semenov, A.; Lammert, F.; Böhm, M.; Laufs, U. Inhibition of Endothelial Nitric Oxide Synthase Induces and Enhances Myocardial Fibrosis. Cardiovasc. Res. 2013, 100, 211–221. [Google Scholar] [CrossRef]
  59. Apte, R.S.; Chen, D.S.; Ferrara, N. VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 2019, 176, 1248–1264. [Google Scholar] [CrossRef] [PubMed]
  60. Meloni, M.; Caporali, A.; Graiani, G.; Lagrasta, C.; Katare, R.; van Linthout, S.; Spillmann, F.; Campesi, I.; Madeddu, P.; Quaini, F.; et al. Nerve Growth Factor Promotes Cardiac Repair Following Myocardial Infarction. Circ. Res. 2010, 106, 1275–1284. [Google Scholar] [CrossRef] [PubMed]
  61. Ronnebaum, S.M.; Patterson, C. The FoxO Family in Cardiac Function and Dysfunction. Annu. Rev. Physiol. 2009, 72, 81–94. [Google Scholar] [CrossRef]
  62. Fu, W.-B.; Wang, W.E.; Zeng, C.-Y. Wnt Signaling Pathways in Myocardial Infarction and the Therapeutic Effects of Wnt Pathway Inhibitors. Acta Pharmacol. Sin. 2019, 40, 9–12. [Google Scholar] [CrossRef]
  63. Aisagbonhi, O.; Rai, M.; Ryzhov, S.; Atria, N.; Feoktistov, I.; Hatzopoulos, A.K. Experimental Myocardial Infarction Triggers Canonical Wnt Signaling and Endothelial-to-Mesenchymal Transition. DMM Dis. Models Mech. 2011, 4, 469–483. [Google Scholar] [CrossRef] [PubMed]
  64. Oerlemans, M.I.F.J.; Goumans, M.J.; van Middelaar, B.; Clevers, H.; Doevendans, P.A.; Sluijter, J.P.G. Active Wnt Signaling in Response to Cardiac Injury. Basic. Res. Cardiol. 2010, 105, 631–641. [Google Scholar] [CrossRef]
  65. Matteucci, M.; Casieri, V.; Gabisonia, K.; Aquaro, G.D.; Agostini, S.; Pollio, G.; Diamanti, D.; Rossi, M.; Travagli, M.; Porcari, V.; et al. Magnetic Resonance Imaging of Infarct-Induced Canonical Wingless/Integrated (Wnt)/β-Catenin/T-Cell Factor Pathway Activation, In Vivo. Cardiovasc. Res. 2016, 112, 645–655. [Google Scholar] [CrossRef] [PubMed]
  66. Paik, D.T.; Rai, M.; Ryzhov, S.; Sanders, L.N.; Aisagbonhi, O.; Funke, M.J.; Feoktistov, I.; Hatzopoulos, A.K. Wnt10b Gain-of-Function Improves Cardiac Repair by Arteriole Formation and Attenuation of Fibrosis. Circ. Res. 2015, 117, 804–816. [Google Scholar] [CrossRef] [PubMed]
  67. Hilfiker-Kleiner, D.; Hilfiker, A.; Fuchs, M.; Kaminski, K.; Schaefer, A.; Schieffer, B.; Hillmer, A.; Schmiedl, A.; Ding, Z.; Podewski, E.; et al. Signal Transducer and Activator of Transcription 3 Is Required for Myocardial Capillary Growth, Control of Interstitial Matrix Deposition, and Heart Protection from Ischemic Injury. Circ. Res. 2004, 95, 187–195. [Google Scholar] [CrossRef] [PubMed]
  68. Sui, Y.B.; Wang, Y.; Liu, L.; Liu, F.; Zhang, Y.Q. Astragaloside IV Alleviates Heart Failure by Promoting Angiogenesis through the JAK-STAT3 Pathway. Pharm. Biol. 2019, 57, 48–54. [Google Scholar] [CrossRef]
  69. Wang, N.; Liu, C.; Wang, X.; He, T.; Li, L.; Liang, X.; Wang, L.; Song, L.; Wei, Y.; Wu, Q.; et al. Hyaluronic Acid Oligosaccharides Improve Myocardial Function Reconstruction and Angiogenesis against Myocardial Infarction by Regulation of Macrophages. Theranostics 2019, 9, 1980–1992. [Google Scholar] [CrossRef] [PubMed]
  70. Zhao, W.; Zhao, T.; Chen, Y.; Ahokas, R.A.; Sun, Y. Reactive Oxygen Species Promote Angiogenesis in the Infarcted Rat Heart. Int. J. Exp. Pathol. 2009, 90, 621–629. [Google Scholar] [CrossRef]
  71. Kim, Y.W.; Byzova, T.V. Oxidative Stress in Angiogenesis and Vascular Disease. Blood 2014, 123, 625–631. [Google Scholar] [CrossRef]
  72. Teixeira, R.B.; Pfeiffer, M.; Zhang, P.; Shafique, E.; Rayta, B.; Karbasiafshar, C.; Ahsan, N.; Sellke, F.W.; Abid, M.R. Reduction in Mitochondrial ROS Improves Oxidative Phosphorylation and Provides Resilience to Coronary Endothelium in Non-Reperfused Myocardial Infarction. Basic. Res. Cardiol. 2023, 118, 3. [Google Scholar] [CrossRef]
  73. Thirunavukkarasu, M.; Adluri, R.S.; Juhasz, B.; Samuel, S.M.; Zhan, L.; Kaur, A.; Maulik, G.; Sanchez, J.A.; Hager, J.; Maulik, N. Novel Role of NADPH Oxidase in Ischemic Myocardium: A Study with Nox2 Knockout Mice. Funct. Integr. Genomics 2012, 12, 501–514. [Google Scholar] [CrossRef]
  74. Dalal, P.J.; Muller, W.A.; Sullivan, D.P. Endothelial Cell Calcium Signaling during Barrier Function and Inflammation. Am. J. Pathol. 2020, 190, 535–542. [Google Scholar] [CrossRef]
  75. Tran, Q.K.; Ohashi, K.; Watanabe, H. Calcium Signalling in Endothelial Cells. Cardiovasc. Res. 2000, 48, 13–22. [Google Scholar] [CrossRef] [PubMed]
  76. Filippini, A.; D’Amore, A.; D’Alessio, A. Calcium Mobilization in Endothelial Cell Functions. Int. J. Mol. Sci. 2019, 20, 4525. [Google Scholar] [CrossRef]
  77. Wen, X.; Peng, Y.; Gao, M.; Zhu, Y.; Zhu, Y.; Yu, F.; Zhou, T.; Shao, J.; Feng, L.; Ma, X. Endothelial Transient Receptor Potential Canonical Channel Regulates Angiogenesis and Promotes Recovery After Myocardial Infarction. J. Am. Heart Assoc. 2022, 11, e023678. [Google Scholar] [CrossRef] [PubMed]
  78. Galeano-Otero, I.; Del Toro, R.; Khatib, A.M.; Rosado, J.A.; Ordóñez-Fernández, A.; Smani, T. SARAF and Orai1 Contribute to Endothelial Cell Activation and Angiogenesis. Front. Cell Dev. Biol. 2021, 9, 639952. [Google Scholar] [CrossRef]
  79. Moccia, F.; Tanzi, F.; Munaron, L. Endothelial Remodelling and Intracellular Calcium Machinery. Curr. Mol. Med. 2014, 14, 457–480. [Google Scholar] [CrossRef] [PubMed]
  80. Andrikopoulos, P.; Eccles, S.A.; Yaqoob, M.M. Coupling between the TRPC3 Ion Channel and the NCX1 Transporter Contributed to VEGF-Induced ERK1/2 Activation and Angiogenesis in Human Primary Endothelial Cells. Cell Signal 2017, 37, 12–30. [Google Scholar] [CrossRef]
  81. Smani, T.; Shapovalov, G.; Skryma, R.; Prevarskaya, N.; Rosado, J.A. Functional and Physiopathological Implications of TRP Channels. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2015, 1853, 1772–1782. [Google Scholar] [CrossRef]
  82. Abdullaev, I.F.; Bisaillon, J.M.; Potier, M.; Gonzalez, J.C.; Motiani, R.K.; Trebak, M. Stim1 and Orai1 Mediate CRAC Currents and Store-Operated Calcium Entry Important for Endothelial Cell Proliferation. Circ. Res. 2008, 103, 1289–1299. [Google Scholar] [CrossRef]
  83. Li, J.; Bruns, A.F.; Hou, B.; Rode, B.; Webster, P.J.; Bailey, M.A.; Appleby, H.L.; Moss, N.K.; Ritchie, J.E.; Yuldasheva, N.Y.; et al. Orai3 Surface Accumulation and Calcium Entry Evoked by Vascular Endothelial Growth Factor. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 1987–1994. [Google Scholar] [CrossRef]
  84. Ye, J.; Wei, J.; Luo, Y.; Deng, Y.; Que, T.; Zhang, X.; Liu, F.; Zhang, J.; Luo, X. Epstein-Barr Virus Promotes Tumor Angiogenesis by Activating STIM1-Dependent Ca2+ Signaling in Nasopharyngeal Carcinoma. Pathogens 2021, 10, 1275. [Google Scholar] [CrossRef]
  85. Simons, M.; Gordon, E.; Claesson-Welsh, L. Mechanisms and Regulation of Endothelial VEGF Receptor Signalling. Nat. Rev. Mol. Cell Biol. 2016, 17, 611–625. [Google Scholar] [CrossRef]
  86. Falcón, D.; Galeano-Otero, I.; Calderón-Sánchez, E.; del Toro, R.; Martín-Bórnez, M.; Rosado, J.A.; Hmadcha, A.; Smani, T. TRP Channels: Current Perspectives in the Adverse Cardiac Remodeling. Front. Physiol. 2019, 10, 159. [Google Scholar] [CrossRef]
  87. Du, S.L.; Jia, Z.Q.; Zhong, J.C.; Wang, L.F. TRPC5 in Cardiovascular Diseases. Rev. Cardiovasc. Med. 2021, 22, 127–135. [Google Scholar] [CrossRef]
  88. Zhu, Y.; Gao, M.; Zhou, T.; Xie, M.; Mao, A.; Feng, L.; Yao, X.; Wong, W.T.; Ma, X. The TRPC5 Channel Regulates Angiogenesis and Promotes Recovery from Ischemic Injury in Mice. J. Biol. Chem. 2019, 294, 28–37. [Google Scholar] [CrossRef] [PubMed]
  89. Shen, N.; Wang, L.; Wu, Y.; Liu, Y.; Pei, H.; Xiang, H. Adeno-Associated Virus Packaged TRPC5 Gene Therapy Alleviated Spinal Cord Ischemic Reperfusion Injury in Rats. Neuroreport 2020, 31, 29–36. [Google Scholar] [CrossRef] [PubMed]
  90. Fadaei, S.; Zarepour, F.; Parvaresh, M.; Motamedzadeh, A.; Tamehri Zadeh, S.S.; Sheida, A.; Shabani, M.; Hamblin, M.R.; Rezaee, M.; Zarei, M.; et al. Epigenetic Regulation in Myocardial Infarction: Non-Coding RNAs and Exosomal Non-Coding RNAs. Front. Cardiovasc. Med. 2022, 9, 1014961. [Google Scholar] [CrossRef]
  91. Smani, T.; Mayoral-González, I.; Galeano-Otero, I.; Gallardo-Castillo, I.; Rosado, J.A.; Ordoñez, A.; Hmadcha, A. Chapter 15: Non-Coding RNAs and Ischemic Cardiovascular Diseases. In Non-Coding RNAs in Cardiovascular Diseases, Advances in Experimental Medicine and Biology; Xiao, J., Ed.; Springer: Singapore, 2020. [Google Scholar]
  92. Kesidou, D.; da Costa Martins, P.A.; de Windt, L.J.; Brittan, M.; Beqqali, A.; Baker, A.H. Extracellular Vesicle MiRNAs in the Promotion of Cardiac Neovascularisation. Front. Physiol. 2020, 11, 579892. [Google Scholar] [CrossRef] [PubMed]
  93. Cheng, M.; Yang, J.; Zhao, X.; Zhang, E.; Zeng, Q.; Yu, Y.; Yang, L.; Wu, B.; Yi, G.; Mao, X.; et al. Circulating Myocardial MicroRNAs from Infarcted Hearts Are Carried in Exosomes and Mobilise Bone Marrow Progenitor Cells. Nat. Commun. 2019, 10, 959. [Google Scholar] [CrossRef]
  94. Ghosh, G.; Subramanian, I.V.; Adhikari, N.; Zhang, X.; Joshi, H.P.; Basi, D.; Chandrashekhar, Y.S.; Hall, J.L.; Roy, S.; Zeng, Y.; et al. Hypoxia-Induced MicroRNA-424 Expression in Human Endothelial Cells Regulates HIF-α Isoforms and Promotes Angiogenesis. J. Clin. Investig. 2010, 120, 4141–4154. [Google Scholar] [CrossRef]
  95. Gou, L.; Xue, C.; Tang, X.; Fang, Z. Inhibition of Exo-MiR-19a-3p Derived from Cardiomyocytes Promotes Angiogenesis and Improves Heart Function in Mice with Myocardial Infarction via Targeting HIF-1α. Aging 2020, 12, 23609–23618. [Google Scholar] [CrossRef]
  96. Fernandes, T.; Baraúna, V.G.; Negrão, C.E.; Ian Phillips, M.; Oliveira, E.M. Aerobic Exercise Training Promotes Physiological Cardiac Remodeling Involving a Set of MicroRNAs. Am. J. Physiol. Heart Circ. Physiol. 2015, 309, H543–H552. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, S.; Aurora, A.B.; Johnson, B.A.; Qi, X.; McAnally, J.; Hill, J.A.; Richardson, J.A.; Bassel-Duby, R.; Olson, E.N. The Endothelial-Specific MicroRNA MiR-126 Governs Vascular Integrity and Angiogenesis. Dev. Cell 2008, 15, 261–271. [Google Scholar] [CrossRef]
  98. Huang, F.; Zhu, X.; Hu, X.Q.; Fang, Z.F.; Tang, L.; Lu, X.L.; Zhou, S.H. Mesenchymal Stem Cells Modified with MiR-126 Release Angiogenic Factors and Activate Notch Ligand Delta-like-4, Enhancing Ischemic Angiogenesis and Cell Survival. Int. J. Mol. Med. 2013, 31, 484–492. [Google Scholar] [CrossRef]
  99. Li, H.; Liu, Q.; Wang, N.; Xu, Y.; Kang, L.; Ren, Y.; Zhu, G. Transplantation of Endothelial Progenitor Cells Overexpressing MiR-126-3p Improves Heart Function in Ischemic Cardiomyopathy. Circ. J. 2018, 82, 2332–2341. [Google Scholar] [CrossRef]
  100. Jakob, P.; Doerries, C.; Briand, S.; Mocharla, P.; Kränkel, N.; Besler, C.; Mueller, M.; Manes, C.; Templin, C.; Baltes, C.; et al. Loss of AngiomiR-126 and 130a in Angiogenic Early Outgrowth Cells from Patients with Chronic Heart Failure: Role for Impaired in Vivo Neovascularization and Cardiac Repair Capacity. Circulation 2012, 126, 2962–2975. [Google Scholar] [CrossRef] [PubMed]
  101. Lu, C.; Wang, X.; Ha, T.; Hu, Y.; Liu, L.; Zhang, X.; Yu, H.; Miao, J.; Kao, R.; Kalbfleisch, J.; et al. Attenuation of Cardiac Dysfunction and Remodeling of Myocardial Infarction by MicroRNA-130a Is Mediated by Suppression of PTEN and Activation of PI3K Dependent Signaling. J. Mol. Cell Cardiol. 2015, 89, 87–97. [Google Scholar] [CrossRef]
  102. Garikipati, V.N.S.; Verma, S.K.; Jolardarashi, D.; Cheng, Z.; Ibetti, J.; Cimini, M.; Tang, Y.; Khan, M.; Yue, Y.; Benedict, C.; et al. Therapeutic Inhibition of MiR-375 Attenuates Post-Myocardial Infarction Inflammatory Response and Left Ventricular Dysfunction via PDK-1-AKT Signalling Axis. Cardiovasc. Res. 2017, 113, 938–949. [Google Scholar] [CrossRef]
  103. Qiao, L.; Hu, S.; Liu, S.; Zhang, H.; Ma, H.; Huang, K.; Li, Z.; Su, T.; Vandergriff, A.; Tang, J.; et al. MicroRNA-21-5p Dysregulation in Exosomes Derived from Heart Failure Patients Impairs Regenerative Potential. J. Clin. Investig. 2019, 129, 2237–2250. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, X.; Wu, C. Tanshinone IIA Improves Cardiac Function via Regulating MiR-499-5p Dependent Angiogenesis in Myocardial Ischemic Mice. Microvasc. Res. 2022, 143, 104399. [Google Scholar] [CrossRef]
  105. Templin, C.; Volkmann, J.; Emmert, M.Y.; Mocharla, P.; Müller, M.; Kraenkel, N.; Ghadri, J.R.; Meyer, M.; Styp-Rekowska, B.; Briand, S.; et al. Increased Proangiogenic Activity of Mobilized CD34+ Progenitor Cells of Patients with Acute ST-Segment-Elevation Myocardial Infarction: Role of Differential MicroRNA-378 Expression. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 341–349. [Google Scholar] [CrossRef]
  106. Bellera, N.; Barba, I.; Rodriguez-Sinovas, A.; Ferret, E.; Asín, M.A.; Gonzalez-Alujas, M.T.; Pérez-Rodon, J.; Esteves, M.; Fonseca, C.; Toran, N.; et al. Single Intracoronary Injection of Encapsulated Antagomir-92a Promotes Angiogenesis and Prevents Adverse Infarct Remodeling. J. Am. Heart Assoc. Cardiovasc. Cerebrovasc. Dis. 2014, 3, e000946. [Google Scholar] [CrossRef]
  107. Li, C.C.; Qiu, X.T.; Sun, Q.; Zhou, J.P.; Yang, H.J.; Wu, W.Z.; He, L.F.; Tang, C.E.; Zhang, G.G.; Bai, Y.P. Endogenous Reduction of MiR-185 Accelerates Cardiac Function Recovery in Mice Following Myocardial Infarction via Targeting of Cathepsin K. J. Cell Mol. Med. 2019, 23, 1164–1173. [Google Scholar] [CrossRef] [PubMed]
  108. Geng, T.; Song, Z.Y.; Xing, J.X.; Wang, B.X.; Dai, S.P.; Xu, Z.S. Exosome Derived from Coronary Serum of Patients with Myocardial Infarction Promotes Angiogenesis Through the MiRNA-143/IGF-IR Pathway. Int. J. Nanomed. 2020, 15, 2647–2658. [Google Scholar] [CrossRef]
  109. Daniel, J.M.; Penzkofer, D.; Teske, R.; Dutzmann, J.; Koch, A.; Bielenberg, W.; Bonauer, A.; Boon, R.A.; Fischer, A.; Bauersachs, J.; et al. Editor’s Choice: Inhibition of MiR-92a Improves Re-Endothelialization and Prevents Neointima Formation Following Vascular Injury. Cardiovasc. Res. 2014, 103, 564–572. [Google Scholar] [CrossRef] [PubMed]
  110. Bang, C.; Fiedler, J.; Thum, T. Cardiovascular Importance of the MicroRNA-23/27/24 Family. Microcirculation 2012, 19, 208–214. [Google Scholar] [CrossRef]
  111. Meloni, M.; Marchetti, M.; Garner, K.; Littlejohns, B.; Sala-Newby, G.; Xenophontos, N.; Floris, I.; Suleiman, M.S.; Madeddu, P.; Caporali, A.; et al. Local Inhibition of MicroRNA-24 Improves Reparative Angiogenesis and Left Ventricle Remodeling and Function in Mice with Myocardial Infarction. Mol. Ther. 2013, 21, 1390–1402. [Google Scholar] [CrossRef] [PubMed]
  112. Peng, Y.; Zhao, J.L.; Peng, Z.Y.; Xu, W.F.; Yu, G.L. Exosomal MiR-25-3p from Mesenchymal Stem Cells Alleviates Myocardial Infarction by Targeting pro-Apoptotic Proteins and EZH2. Cell Death Dis. 2020, 11, 317. [Google Scholar] [CrossRef]
  113. Wen, Z.; Mai, Z.; Zhu, X.; Wu, T.; Chen, Y.; Geng, D.; Wang, J. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Cardiomyocyte Apoptosis in Hypoxic Conditions through MicroRNA144 by Targeting the PTEN/AKT Pathway. Stem Cell Res. Ther. 2020, 11, 36. [Google Scholar] [CrossRef]
  114. Pan, J.; Alimujiang, M.; Chen, Q.; Shi, H.; Luo, X. Exosomes Derived from MiR-146a-Modified Adipose-Derived Stem Cells Attenuate Acute Myocardial Infarction−induced Myocardial Damage via Downregulation of Early Growth Response Factor 1. J. Cell Biochem. 2019, 120, 4433–4443. [Google Scholar] [CrossRef]
  115. Duan, S.; Wang, C.; Xu, X.; Zhang, X.; Su, G.; Li, Y.; Fu, S.; Sun, P.; Tian, J. Peripheral Serum Exosomes Isolated from Patients with Acute Myocardial Infarction Promote Endothelial Cell Angiogenesis via the MiR-126-3p/TSC1/MTORC1/HIF-1α Pathway. Int. J. Nanomed. 2022, 17, 1577–1592. [Google Scholar] [CrossRef]
  116. Liu, H.; Zhang, Y.; Yuan, J.; Gao, W.; Zhong, X.; Yao, K.; Lin, L.; Ge, J. Dendritic Cell-Derived Exosomal MiR-494-3p Promotes Angiogenesis Following Myocardial Infarction. Int. J. Mol. Med. 2021, 47, 315–325. [Google Scholar] [CrossRef]
  117. Youn, S.W.; Li, Y.; Kim, Y.M.; Sudhahar, V.; Abdelsaid, K.; Kim, H.W.; Liu, Y.; Fulton, D.J.R.; Ashraf, M.; Tang, Y.; et al. Modification of Cardiac Progenitor Cell-Derived Exosomes by MiR-322 Provides Protection against Myocardial Infarction through Nox2-Dependent Angiogenesis. Antioxidants 2019, 8, 18. [Google Scholar] [CrossRef]
  118. Wang, N.; Chen, C.; Yang, D.; Liao, Q.; Luo, H.; Wang, X.; Zhou, F.; Yang, X.; Yang, J.; Zeng, C.; et al. Mesenchymal Stem Cells-Derived Extracellular Vesicles, via MiR-210, Improve Infarcted Cardiac Function by Promotion of Angiogenesis. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2085–2092. [Google Scholar] [CrossRef] [PubMed]
  119. Arif, M.; Pandey, R.; Alam, P.; Jiang, S.; Sadayappan, S.; Paul, A.; Ahmed, R.P.H. MicroRNA-210-Mediated Proliferation, Survival, and Angiogenesis Promote Cardiac Repair Post Myocardial Infarction in Rodents. J. Mol. Med. 2017, 95, 1369–1385. [Google Scholar] [CrossRef] [PubMed]
  120. Liao, Z.; Chen, Y.; Duan, C.; Zhu, K.; Huang, R.; Zhao, H.; Hintze, M.; Pu, Q.; Yuan, Z.; Lv, L.; et al. Cardiac Telocytes Inhibit Cardiac Microvascular Endothelial Cell Apoptosis through Exosomal MiRNA-21-5p-Targeted Cdip1 Silencing to Improve Angiogenesis Following Myocardial Infarction. Theranostics 2021, 11, 268–291. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Signaling pathways activated in Endothelial Cells (ECs) after Myocardial Infarction (MI). During MI, ischemic cardiac tissue evokes the activation of HIF-1α, ROS, eNOS, Wnt, and JAK/STAT pathways in EC, which trigger the transcription of pro-angiogenic genes. Different heart cell populations (EC, EPC, cardiomyocytes, fibroblasts, macrophages) produce a high number of autocrine signals, including cytokines, growth factors such as VEGF, and non-coding RNAs, miRNAs, which regulate EC functions to rescue myocardium healing. VEGF induces the increase in intracellular Ca2+ concentration via store-operated Ca2+ entry (SOCE) through Orai and TRPC channels, which activates different transcription factors essential to ECs activity. VEGF increases DLL4 expression in EC, polarizing it to tip cell phenotype. In neighbor ECs, DLL4 activates Notch1 signaling, promoting the EC switch to stalk phenotype. Altogether, these events promote an increase in EC permeability, as well as in their proliferation, migration, and tube formation ability, hallmarks of angiogenesis. EPC: endothelial progenitor cells; ER: endoplasmic reticulum; DAG: directed acyclic graphs; DLL4: delta-like 4; eNOS: endothelial nitric oxide synthase; GF: growth factors; IP3: 1,4,5-trisphosphate; IP3R: IP3 receptor; miRNAs: microRNAs; NICD: Notch intracellular domain; PLC: phospholipase C; ROS: reactive oxygen species; SERCA: sarcoendoplasmic reticulum (SR) calcium transport ATPase; VEGF: vascular endothelial growth factor; VEGFR1: VEGF receptor 1; VEGFR2: VEGF receptor 2.
Figure 1. Signaling pathways activated in Endothelial Cells (ECs) after Myocardial Infarction (MI). During MI, ischemic cardiac tissue evokes the activation of HIF-1α, ROS, eNOS, Wnt, and JAK/STAT pathways in EC, which trigger the transcription of pro-angiogenic genes. Different heart cell populations (EC, EPC, cardiomyocytes, fibroblasts, macrophages) produce a high number of autocrine signals, including cytokines, growth factors such as VEGF, and non-coding RNAs, miRNAs, which regulate EC functions to rescue myocardium healing. VEGF induces the increase in intracellular Ca2+ concentration via store-operated Ca2+ entry (SOCE) through Orai and TRPC channels, which activates different transcription factors essential to ECs activity. VEGF increases DLL4 expression in EC, polarizing it to tip cell phenotype. In neighbor ECs, DLL4 activates Notch1 signaling, promoting the EC switch to stalk phenotype. Altogether, these events promote an increase in EC permeability, as well as in their proliferation, migration, and tube formation ability, hallmarks of angiogenesis. EPC: endothelial progenitor cells; ER: endoplasmic reticulum; DAG: directed acyclic graphs; DLL4: delta-like 4; eNOS: endothelial nitric oxide synthase; GF: growth factors; IP3: 1,4,5-trisphosphate; IP3R: IP3 receptor; miRNAs: microRNAs; NICD: Notch intracellular domain; PLC: phospholipase C; ROS: reactive oxygen species; SERCA: sarcoendoplasmic reticulum (SR) calcium transport ATPase; VEGF: vascular endothelial growth factor; VEGFR1: VEGF receptor 1; VEGFR2: VEGF receptor 2.
Ijms 24 12298 g001
Figure 2. Endothelial Progenitor Cells (EPCs) are essential in post-ischemic angiogenesis. Both types of EPCs, Myeloid angiogenic cells (MACs) and endothelial colony forming (ECFCs), are actively involved in reparative neovascularization after myocardial infarction (MI). MACs are key regulators for EPCs recruitment as they secrete a wide range of pro-angiogenic factors, as well as microRNAs (miRNAs), to stimulate cardiac endothelial cells (ECs). Circulating and resident ECFCs can differentiate into mature ECs and take part in new blood vessel formation.
Figure 2. Endothelial Progenitor Cells (EPCs) are essential in post-ischemic angiogenesis. Both types of EPCs, Myeloid angiogenic cells (MACs) and endothelial colony forming (ECFCs), are actively involved in reparative neovascularization after myocardial infarction (MI). MACs are key regulators for EPCs recruitment as they secrete a wide range of pro-angiogenic factors, as well as microRNAs (miRNAs), to stimulate cardiac endothelial cells (ECs). Circulating and resident ECFCs can differentiate into mature ECs and take part in new blood vessel formation.
Ijms 24 12298 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Martín-Bórnez, M.; Falcón, D.; Morrugares, R.; Siegfried, G.; Khatib, A.-M.; Rosado, J.A.; Galeano-Otero, I.; Smani, T. New Insights into the Reparative Angiogenesis after Myocardial Infarction. Int. J. Mol. Sci. 2023, 24, 12298. https://doi.org/10.3390/ijms241512298

AMA Style

Martín-Bórnez M, Falcón D, Morrugares R, Siegfried G, Khatib A-M, Rosado JA, Galeano-Otero I, Smani T. New Insights into the Reparative Angiogenesis after Myocardial Infarction. International Journal of Molecular Sciences. 2023; 24(15):12298. https://doi.org/10.3390/ijms241512298

Chicago/Turabian Style

Martín-Bórnez, Marta, Débora Falcón, Rosario Morrugares, Geraldine Siegfried, Abdel-Majid Khatib, Juan A. Rosado, Isabel Galeano-Otero, and Tarik Smani. 2023. "New Insights into the Reparative Angiogenesis after Myocardial Infarction" International Journal of Molecular Sciences 24, no. 15: 12298. https://doi.org/10.3390/ijms241512298

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

Article Metrics

Back to TopTop