Next Article in Journal
Quassinoids from Twigs of Harrisonia perforata (Blanco) Merr and Their Anti-Parkinson’s Disease Effect
Next Article in Special Issue
A Meta-Analysis Approach to Gene Regulatory Network Inference Identifies Key Regulators of Cardiovascular Diseases
Previous Article in Journal
Computational Insight into the Nature and Strength of the π-Hole Type Chalcogen∙∙∙Chalcogen Interactions in the XO2∙∙∙CH3YCH3 Complexes (X = S, Se, Te; Y = O, S, Se, Te)
Previous Article in Special Issue
The Role of Long Non-Coding RNAs in Cardiovascular Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Intercellular Communication Mediated by Exosomal ncRNAs in Cardiovascular Disease

1
College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China
2
Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(22), 16197; https://doi.org/10.3390/ijms242216197
Submission received: 23 July 2023 / Revised: 28 September 2023 / Accepted: 29 September 2023 / Published: 11 November 2023

Abstract

:
Cardiovascular diseases are a leading cause of worldwide mortality, and exosomes have recently gained attention as key mediators of intercellular communication in these diseases. Exosomes are double-layered lipid vesicles that can carry biomolecules such as miRNAs, lncRNAs, and circRNAs, and the content of exosomes is dependent on the cell they originated from. They can be involved in the pathophysiological processes of cardiovascular diseases and hold potential as diagnostic and monitoring tools. Exosomes mediate intercellular communication, stimulate or inhibit the activity of target cells, and affect myocardial hypertrophy, injury and infarction, ventricular remodeling, angiogenesis, and atherosclerosis. Exosomes can be released from various types of cells, including endothelial cells, smooth muscle cells, cardiomyocytes, fibroblasts, platelets, adipocytes, immune cells, and stem cells. In this review, we highlight the communication between different cell-derived exosomes and cardiovascular cells, with a focus on the roles of RNAs. This provides new insights for further exploring targeted therapies in the clinical management of cardiovascular diseases.

1. Introduction

Cardiovascular disease (CVD) includes diseases of the heart muscle, blood vessels, or interstitial tissues. The prevalence of CVD has risen dramatically over the past two decades, making it one of the major issues of global concern and imposing an enormous social and economic burden. In 2012, approximately 17.5 million people died from CVD, and this number is expected to increase to 23 million by 2030. As the heterogeneity and complexity observed in CVD progression increases, the need for specific and accurate diagnosis of the disease state becomes more urgent. In this context, it is therefore of great interest to identify new targets to evaluate pathological responses or new diagnostic therapies for the early detection of people at risk of CVD. The number and characteristics of exosomes change according to the pathophysiological state of the disease and, therefore, can serve to some extent as biomarkers for diagnosis and monitoring of the disease. The identification of biomarkers with high sensitivity and specificity is necessary in the diagnosis and treatment of CVD. The study of exosomes has become a frontier and hotspot in the diagnosis and treatment of CVD.
Exosomes, as a new molecular platform for chemical signaling, have played a great role in recent years in a variety of diseases including cardiovascular diseases and tumors [1,2,3]. Exosomes are vesicle-like substances secreted by living cells with a double-membrane structure, and their diameters generally range from 30 nm to 150 nm. All cells, including those of eukaryotic and prokaryotic cells, are capable of releasing exosomes both under physiological and pathological conditions [4]. Exosomes have a similar topology to cells and contain substances such as DNA, RNA, proteins, lipids, small molecule metabolites, and cell surface proteins [5]. The reasons for cells producing exosomes and the physiological functions of exosomes are still not particularly well understood, and more research is needed to prove this. Previously, it was believed that the main function of exosomes was to remove excess or nonessential cellular components from the cell to maintain cellular homeostasis. In recent years, studies have shown that cells can selectively accumulate cellular components in exosomes through targeted and mechanism-specific-driven processes, which suggests an important role for exosomes in regulating intercellular communication [6]. In 2007, it was first discovered that exosomes could transport miRNA between cells, and this finding revealed an important transport mechanism for the functional role of ncRNAs [7]. Several studies have confirmed that exosomes are important tools for the extracellular transport of miRNA and lncRNA [8,9,10,11]. The ncRNAs carried by exosomes are highly stable in body fluids and have the potential to serve as biomarkers. There is increasing evidence that ncRNAs in exosomes are not only diverse and abundant but also highly associated with a variety of diseases such as cardiovascular diseases, central nervous system diseases, and malignant tumors [12,13,14,15,16,17]. In view of this, exosomal ncRNAs have a broad prospect in the field of disease diagnosis.
The exosomes are important bioinformatic carriers that facilitate intercellular communication and participate in the pathophysiological processes of various cardiovascular diseases. Exosomes are released by different types of cells in response to various biological and chemical stimuli, including oxidative stress [18], low pH [19], and hypoxia [20]. More importantly, these findings have introduced a new approach in cardiology, where exosomes are recognized as a new mechanism of communication between the heart and other organs. A study has explored that the exosomes involved in atherosclerosis originated from various cell types, with the majority being derived from leukocytes. Specifically, macrophages account for 29 ± 5%, lymphocytes for 15 ± 3%, granulocytes for 8 ± 1%, red blood cells for 27 ± 4%, smooth muscle cells for 13 ± 4%, and endothelial cells for 8 ± 2% [21].

2. Biology of Exosomes

2.1. The Biogenesis and Uptake of Exosomes

The origin, synthesis, and secretion of exosomes involve the following process: the cell membrane of the parent cell forms early endosomes through endocytosis or “inward budding” (Figure 1). These early endosomes gradually mature into late endosomes and multivesicular bodies (MVBs) within the cell (Figure 1). The precursor of extracellular vesicles exists as intraluminal vesicles (ILVs) inside MVBs (Figure 1); subsequently, MVBs fuse with the cell membrane and excrete ILVs into the extracellular space through exocytosis, forming exosomes (Figure 1) [22]. As a result, their composition and structure are more complex, and they may even contain components engulfed from the extracellular matrix or culture medium. Exosomes contain proteins such as membrane transport proteins (GTPases, Annexins, Flotillin), heat shock proteins (HSP90-70), transmembrane proteins (CD81, CD63, CD9), and proteins associated with MVB biogenesis (ALIX and TSG101). They also have higher levels of membrane cholesterol and diacylglycerol [23]. The biogenesis of exosomes is regulated by two mechanisms. One is the endosomal sorting complex required for transport (ESCRT)-dependent pathway and the other is the ESCRT-independent pathway. The former involves more than 30 proteins organized into four families (ESCRT-0, I, II, III), while the latter involves proteins such as neutral sphingomyelinase 2 (nSMase2) and CD63.

2.2. Mechanisms of Exosome Internalization

Compared to the relatively clear “linear process” of exosome generation, the fate of exosomes is significantly complex and uncertain. If exosomes are recognized and “captured” by neighboring tissue cells, they will be taken up and utilized by the neighboring cells. If the adjacent cells do not recognize and capture the exosomes, the exosomes are transported through the circulatory system to more distant cells or tissues. The uptake and utilization of exosomes by both adjacent and distant tissue cells are mainly through several different mechanisms of action [24,25].
(1) The ligand on the membrane surface of exosomes binds to the receptor on the membrane of the recipient cells, activating the receptor-mediated signaling pathway, and the activated recipient cells ingest the contents into the cell through endocytosis. For example, exosomes derived from dendritic cells containing MHC–peptide complexes can activate T cells through homologous T-cell receptors [26]; (2) Recipient cells directly internalize exosomes through endocytosis, allowing the contents of the exosomes to be released into the cells. Some components of exosomes participate in new multivesicular body biosynthesis processes; (3) Direct fusion of the exosome membrane with the cell membrane releases the exosome contents into the cytoplasm. Since exosomes carry a number of lipid and protein components, the interaction of these exosomal membrane proteins with receptors present in the membrane of the recipient cells may trigger responses leading to cellular changes. Thus, through these processes, exosomes can interact with recipient cells to trigger the release of cargo or the induction of signaling cascades that ultimately lead to changes in cellular activity or function [27,28].
Exosomes are also primarily characterized by their ability to enter the recipient cells and directly transfer active substances protected by the lipid bilayer membrane to the target cells. The mechanisms by which exosomes are internalized by the recipient cells are numerous and controversial. Currently suggested mechanisms include cell-specific uptake, non-specific protein interactions, endocytosis, and membrane fusion [29]. Most studies suggest that the uptake of exosomes is non-specific. On the one hand, tumor-cell-derived exosomes can be taken up by multiple cells [25], and on the other hand, exosome-mediated material delivery can cross species. For example, exosomes can transfer functional mRNAs and miRNAs from mouse to human mast cells and mouse proteins can be detected in human mast cells [7].
The fate of exosomes is highly uncertain and influenced by various factors. There is both directed migration towards specific destinations, such as receptor-mediated specific binding, as well as aimless or random wandering. They can be engulfed non-specifically by the circulatory system and reticuloendothelial system, or their destinations can be influenced by the local microenvironment and cellular status of recipient cells. Furthermore, studies have shown that exosomes can interact with specific target cells based on their cargo and origin, efficiently releasing their contents to induce phenotypic changes in target cells [30]. Due to their ability to effectively deliver bioactive substances or artificially loaded molecular cargos, exosomes have earned a reputation as ”natural carriers”. Therefore, in-depth exploration of the generation process, secretion conditions, and biological functions is crucial in understanding their impact on the physiological and pathological processes of recipient cells, as well as their clinical applications.
Nowadays, the secretion of exosomes has expanded to various cell types, and their significance in intercellular communication in both normal and pathological states has been well documented. Moreover, different cell types regulate exosome biogenesis according to their physiological state and release exosomes with specific lipid, protein, and nucleic-acid compositions.

3. ncRNAs and Cardiovascular Diseases

With the continuous development and advancement of molecular biology technology, the role of genes and RNA in cardiovascular diseases has been increasingly emphasized. Some studies have shown that ncRNAs are abundantly expressed in the cardiovascular system, and changes in ncRNA expression levels have been found in the occurrence and development of many cardiovascular diseases. ncRNAs include transfer ribonucleic acid (tRNAs), ribosome ribonucleic acid (rRNAs), long non-coding ribonucleic acid (lncRNAs), circular ribonucleic acid (circRNAs), small nuclear ribonucleic acid (snRNAs) and micro-ribonucleic acid (miRNAs), as well as other RNAs with unknown functions. It is now well established that lncRNAs, circRNAs, and miRNAs are closely associated with the development of CVDs and have been recognized as new biomarkers and potential therapeutic targets for a variety of diseases, including CVDs.
Recent studies have proposed approximately 50 miRNAs associated with essential hypertension and more than 30 miRNAs associated with heart failure and myocardial infarction, many of which could serve as promising biomarkers. miR-21 was expressed in many cell types associated with the cardiovascular system, including vascular smooth muscle cells, vascular endothelial cells, cardiomyocytes, cardiac fibroblasts, and blood [31]. miR-21 expression was closely associated with the occurrence and development of hypertension [32]. In addition, studies of cardiac dysfunction after myocardial infarction have shown that miR-21 can be involved in the occurrence of hypertension and myocardial fibrosis through the TGF-β/smad7 signaling pathway [32]. miR-19a/19b expression was up-regulated in patients with heart failure after myocardial infarction, and miR-19a/19b increased cardiac proliferation and regeneration, suggesting that there might be a compensatory mechanism for the stress response [33].
Similar to miRNAs, a large number of lncRNAs are involved in the critical regulation of a variety of cardiac diseases, highlighting their role in the occurrence and development of cardiovascular disease. It was shown that lncRNA H19 promoted the development of atherosclerosis by promoting the MAPK and NF-kB signaling pathways [34]. lncRNA Chaer was significantly down-regulated in hypoxia-treated cardiomyocytes and in the hearts of myocardial infarction [35]. In vitro overexpression of lncRNA Chaer reduced hypoxia-induced cardiomyocyte apoptosis; conversely, silencing of lncRNA Chaer promoted cardiomyocyte apoptosis [35]. In vivo overexpression of lncRNA Chaer slowed myocyte apoptosis, reduced the infarcted area, and improved cardiac function in infarcted mice, suggesting that lncRNA Chaer has a protective effect against myocardial infarction [35]. lncRNA Nron expression was elevated in the peripheral blood of myocardial infarcted patients and in hypoxia-stimulated H9c2 cells [36]. Knockdown of lncRNA Nron promoted cell viability and inhibited apoptosis in hypoxia-stimulated H9c2 cells; meanwhile, knockdown of lncRNA Nron significantly attenuated cardiac injury and improved cardiac function in myocardial infarction mice [36].
The role of circRNA in various cardiovascular diseases has also been reported. Exogenous expression of circANRIL in a rat model of coronary atherosclerosis exerted beneficial effects by lowering the levels of total cholesterol, triglycerides, ox-LDL, and pro-inflammatory and pro-apoptotic markers’ expression in endothelial cells, thereby playing a beneficial role [37]. circRNA circ_0003204 inhibited the progression of atherosclerosis through the miR-370-3p/TGFβR2/phosph-SMAD3 axis [38]. circSNRK was significantly down-regulated in myocardial infarction rats, and overexpression of circSNRK in cardiomyocytes inhibited apoptosis and promoted cell proliferation. Overexpression of circSNRK in the heart after myocardial infarction reduced cardiomyocyte apoptosis, promoted cardiomyocyte proliferation, enhanced angiogenesis, and improved cardiac function. Overall, circSNRK promotes cardiac survival and functional recovery after myocardial infarction [39].

4. Exosome-Mediated Intercellular Communication

Since intercellular communication is critical for maintaining tissue homeostasis and preventing disease, understanding exosome-mediated intercellular communication may provide important insights into the development of vascular pathogenesis. Over short distances, direct cell-to-cell contact can generate cellular crosstalk, whereas long-distance communication can be mediated by cytokines or hormones. Recently, a novel communication pathway mediated by exosomes has emerged as a newly discovered means of intercellular communication. Transfer of exosome contents to receptor cells has been described in a variety of cells and these exosomes act as regulators of intercellular communication between neighboring and distal cells [40]. Furthermore, the microenvironmental stimuli can influence the quantity and types of exosome contents through induction of molecular enrichment or depletion [41]. Therefore, their potential use as diagnostic, prognostic, and therapeutic markers in physiological and pathological processes has garnered immense interest [42,43,44,45]. The role of exosomes as key regulators in intravascular homeostasis and cardiovascular disease progression has been emphasized by recent studies [46,47,48,49,50,51].

4.1. The Role of Exosomal ncRNA from Vascular Smooth Muscle Cells in Cardiovascular Disease

The composition of the human medium and large arterial vessel wall includes a variety of cells such as endothelial cells (ECs), immune macrophages, and vascular smooth muscle cells (SMCs). The structural and functional integrity of these cells is an important guarantee of the maintenance of the normal physiological function of blood vessels. Among the various cells, ECs and SMCs especially, perform very complicated intercellular communication. They are the main components of the vascular wall and exhibit a high degree of plasticity. The communication between ECs and VSMCs can be carried out in an indirect (biochemical) manner [52]. Exosomes originating from VSMCs can inhibit vascular regeneration through an intermediary mediation of miR-16 to down-regulate vascular endothelial growth factor levels and inhibit vascular regeneration in breast cancer cells [53]. Research has explored the role of exosome-mediated transfer of miR-155 from VSMCs to ECs in inducing endothelial injury and promoting atherosclerosis [54]. The study reveals that VSMCs release exosomes carrying miR-155, which can be taken up by neighboring ECs. This transfer leads to endothelial injury characterized by compromised barrier function, increased permeability, and enhanced endothelial cell apoptosis [54].

4.2. The Role of Exosomal ncRNA from Endothelial Cells in Cardiovascular Disease

Abnormal proliferation of VSMCs plays an important role in the development of diabetic vascular complications. Under high glucose (HG) conditions, ECs act as the first barrier to injury stimuli, triggering multiple responses, including ECs and VSMCs crosstalk. The results showed that exosomes secreted by mouse aortic endothelial cells (MAEC) promoted HG-induced proliferation and inhibited apoptosis of VSMCs. MAEC exosomes exposed to HG could transfer circHIPK3 enriched in MAEC exosomes to VSMCs [55]. Exosomal circHIPK3 promoted VSMC proliferation and inhibited VSMC apoptosis. circHIPK3 was sponge-wiped with miR-106a-5p to deregulate its inhibition of Foxo1 expression. The increased expression of Foxo1 served as a transcription factor to promote the expression of Vcam1, which promoted the uptake of MAEC-derived exosomes by VSMCs [55].
In the presence of atherosclerosis, the amount of microRNA-92a-3p in EC exosomes was significantly increased [56]. MicroRNA-92a-3p encapsulated by EC exosomes could translocate to recipient cells and regulate target genes through a thbs1-dependent mechanism [56]. Oxidized low-density lipoprotein-treated VSMCs released exosomes encapsulating miR-505 [57]. These exosomes are ingested and internalized into macrophages, targeting and inhibiting SIRT3 in neutrophils, thereby inducing elevated levels of reactive oxygen species in neutrophils, triggering an inflammatory response, and exacerbating atherosclerosis development [57]. It was found that exosomal miRNAs mediated signaling between cardiomyocytes and ECs [58]. Prolactin hydrolyzed fragments could mediate signaling between cardiomyocytes and ECs by stimulating ECs to release exosomes containing miR-146a [58]. Acquisition of these exosomes by cardiomyocytes resulted in a decrease in metabolic activity [58]. Specifically, the researchers discovered that miRNA-143/145 played a key role in the development of atherosclerosis. miRNA-143/145 was transferred from ECs to SMCs via exosomes, mediating intercellular communication. This communication could inhibit the proliferation and migration of SMCs and maintain the integrity of the vascular endothelium, thus exerting a protective effect on the process of atherosclerosis [59].
The increased expression of lncRNA-LINC00174 in exosomes derived from mouse primary aortic ECs directly interacted with SRSF1 to suppress the expression of p53, thereby inhibiting p53-mediated autophagy and cell apoptosis, reducing ischemia-reperfusion-induced myocardial injury [59]. Knockdown of circNPHP4 inhibited heterotypic adhesion between monocytes and coronary artery endothelial cells and reduced the expression of ICAM-1 and VCAM-1. Studies on the potential mechanisms showed that circNPHP4 affected the expression of miR-1231 and its target gene EGFR. Overexpression of miR-1231 abrogated the inhibitory effect of circNPHP4 on heterotypic adhesion [60]. The article comprehensively elucidated the abnormal communication between ECs and macrophages mediated by METTL3 through the regulation of miR-93 in exosomes in smoking-induced emphysema [61]. Smoking induced the accumulation of METTL3, which in turn promoted the maturation of miR-93 in exosomes. By targeting the negative regulation of DUSP2, the JNK/MMPs pathway was activated, leading to the degradation of elastin and the development of emphysema [61].

4.3. The Role of Exosomal ncRNA from Cardiac Fibroblasts in Cardiovascular Disease

Cardiac fibroblasts (CFs) play an important role in the regulation of cardiac physiology during injury [62]. Emerging evidence suggests that exosomes released by CFs are one of the major components leading to cardiomyocyte hypertrophy [63,64,65,66]. Recently, several published papers have confirmed the direct involvement of fibroblasts in ec-mediated angiogenesis [67,68]. The study investigated the role of CF-derived exosomes microRNA in cardiomyocyte hypertrophy. Evaluation of miRNA content in exosomes derived from CFs revealed a relatively high abundance of miRNAs. Among them, miR-21_3p (miR-21*) could be taken up by cardiomyocytes to silence SORBS2 or PDLIM5, leading to the induction of cardiomyocyte hypertrophy [69]. In addition, CF-exo miR-23a-3p could be taken up by cardiomyocytes, resulting in the inhibition of SLC7A11 expression and the promotion of ferroptosis, ultimately contributing to the development of atrial fibrillation [70].
The role of myofibroblast-derived exosomes in inducing cardiac EC dysfunction was explored [71]. It was shown that miR-200a-3q secreted by TGF-β-activated fibroblasts targeted the VEGF-A/PIGF signaling pathway, leading to a decrease in vascular formation ability, impaired migration capability, and increased vascular permeability in cardiac ECs [71]. A research article on miRNA-423-3p derived from CF exosomes regulating cardioprotective effects after ischemia is reported [72]. The study discovered that miRNA-423-3p in CF-derived exosomes was able to attenuate cardiac damage induced by ischemia-reperfusion injury and improve cardiac function by translocating into cardiomyocytes and targeting the downstream effector RAP2C [72]. The study aimed to investigate the role of circular RNAs (circRNAs) derived from M2 macrophages (M2Ms) exosomes in the development of myocardial fibrosis. The results showed that the circRNAbe3a derived from M2Ms exosomes promoted the proliferation, migration, and phenotypic transformation of CFs by directly targeting the miR-138-5p/RhoC axis, which might exacerbate myocardial fibrosis after acute myocardial infarction [73].

4.4. The Role of Exosomal ncRNA from Cardiomyocyte in Cardiovascular Disease

Myocardial infarction (MI) is the leading cause of congestive heart failure and death. Hypoxia is an important trigger factor for myocardial remodeling during the development of heart disease. The mechanism by which exosomes derived from cardiomyocytes promote cardiac fibrosis through interactions between cardiomyocytes and fibroblasts was investigated. The study found that lncRNA AK139128 produced by cardiomyocytes under hypoxic conditions could be delivered to CFs through exosomes, regulating their apoptosis and proliferation processes [74]. Cardiomyocytes secreted exosomes containing miR-208a into fibroblasts, which promoted fibroblast proliferation and differentiation toward myofibroblasts by targeting Dyrk2 [75]. The contribution of miR-210-3p from myocyte-derived exosomes to atrial fibrosis in patients with atrial fibrillation was covered [76]. The findings indicated that miR-210-3p inhibited GPD1L in atrial fibroblasts, thereby regulating the PI3K/AKT signaling pathway and promoting cell proliferation and collagen synthesis [76]. By inhibiting miR-210-3p in these exosomes, the extent of atrial fibrosis could be attenuated [76]. Meanwhile, the study found that the exosome released by cardiomyocytes contained miRNA-92a, and that the exosome was able to deliver miR-92a to postischemic myofibroblasts, alleviating smad7-mediated repression of αSMA transcription and triggering phenotypic transformation towards myofibroblasts [77]. The study investigated the mechanism by which cardiomyocytes mediated anti-angiogenesis in type-2 diabetic rats by releasing exosomes for delivery to endothelial cells [78]. The results showed that cardiomyocytes transferred exosomes miR-320 to ECs, leading to the down-regulation of target genes (IGF-1, Hsp20, and Ets2), thereby inhibiting ECs proliferation, migration, and angiogenesis [78].
In 2019, the specific expression pattern of circular RNAs (circRNAs) in exosomes from cardiac tissues during ischemia/reperfusion (I/R) injury was first revealed, providing important evidence for the role of circRNAs and exosomes in cardiac I/R pathology [79]. It was found that exosomes released from hypoxia preconditioned cardiomyocytes were enriched in circHIPK3, which was delivered to cardiac microvascular ECs and inhibited IGF-1 expression by binding to miR-29a to reduce oxidative stress-induced injury and protected cardiac microvascular ECs [80]. On the other hand, the circHIPK3 in these exosomes could increase the expression of VEGFA by inhibiting the activity of miR-29a [81]. This, in turn, accelerated cell cycle progression and proliferation of cardiac ECs, ultimately facilitating angiogenesis [81].
lncRNA-AK139128, up-regulated in hypoxia preconditioned cardiomyocyte-derived exosomes, promoted cardiac fibroblast apoptosis, and inhibited their proliferation and migration [74]. lncRNA-MALAT1 was up-regulated in exosomes derived from cardiomyocytes treated with hyperbaric oxygen, inhibiting the expression of miR-92a and relieving miR-92a-mediated inhibition of KLF2 and CD31 expression after myocardial infarction, thus, enhancing neovascularization and significantly reducing the infarct size [82]. Furthermore, exosomes enriched with lncRNA-ZFAS1 from cardiomyocytes have been shown to worsen cardiac fibrosis in a mouse model of chronic kidney disease through the activation of the Wnt4/β-catenin signaling pathway [83].

4.5. The Role of Exosomal ncRNA from Stem Cells in Cardiovascular Disease

Embryonic stem cells (ESCs) hold great promise for cardiac regeneration and have the ability to produce exosomes, and the miR-290-295 cluster, especially miR-294, is significantly enriched in ESCs exosomes [84]. The delivery of miR-294 to the heart mediated the activation of endogenous repair mechanisms, enhancing cardiac function [84]. A study evaluated the protective effects of mesenchymal stem cell (MSC)-derived exosomes on cardiomyocytes in an animal model of doxorubicin-induced cardiomyopathy [85]. This protection was achieved through the modulation of the miR-199a-3p-Akt-Sp1/p53 signaling pathway [85]. Exosomes derived from MSCs have been widely reported to have a protective effect against myocardial infarction. It was discovered that blocking exosomal miRNA-153-3p derived from bone marrow mesenchymal stem cells could activate the VEGF/VEGFR2/PI3K/Akt/eNOS pathway, thereby alleviating hypoxia-induced myocardial and microvascular damage and improving myocardial function [86]. A study has identified that exosomes derived from iPS carried various cardioprotective miRNAs, including miRNA-21 and miRNA-210 [87]. Delivery of these miRNAs modulated multiple target genes, inhibited cardiomyocyte apoptosis, and promoted cell survival and improvement of myocardial function [87]. It provides a novel therapeutic strategy of utilizing exosomes to deliver miRNAs for cardioprotection and improving prognosis in ischemic heart disease [87].
LncRNA-UCA1 expression was found to be up-regulated in hypoxia-treated MSC exosomes. In vitro experiments demonstrated that exosomal lncRNA-UCA1 mediated the protective effect on hypoxic cardiomyocytes through the miR-873-5p/interlocked apoptosis inhibitory protein (XIAP) axis [88]. MSC-derived exosomal lncRNA KLF3-AS1 ameliorated the focal death of infarcted cardiomyocytes, inhibited the release of pro-inflammatory cytokines, and ultimately reduced the area of cardiac infarction by competitively inhibiting the down-regulation of silencing information regulatory factor 1 (SIRT1) by miR-138-5p [89]. Increased lncRNA H19 in MSC-derived exosomes pretreated with atorvastatin up-regulated the expression of VEGF and ICAM-1 and attenuated the apoptosis of infarcted myocardium by acting on miR-675 [90]. Meanwhile, it down-regulated the levels of IL-6 and TNF-α, inhibited the excessive inflammatory response, and ultimately improved the cardiac function of infarcted hearts [90]. The expression of lncRNA-NEAT1 in human adipose MSC-derived exosomes pretreated with macrophage migration inhibitory factor (MIF) was up-regulated, which protected cardiomyocytes from H2O2-induced apoptosis by competitively inhibiting miR-142-3p and restored the expression of FOXO1 [91]. Additionally, lncRNA-UCA1 in exosomes derived from human umbilical cord mesenchymal stem cells up-regulated the anti-apoptotic factor Bcl-2 through competitive binding with miR-143, inhibiting excessive apoptosis and autophagy in the hearts of rats subjected to ischemia-reperfusion injury [92].
The study investigated whether exosomes derived from umbilical cord mesenchymal stem cells (UMSCs) can repair the heart after myocardial infarction (MI) by delivering circRNAs. The results showed that circRNA-0001273 significantly inhibited apoptosis of myocardial cells under ischemic conditions and promoted myocardial infarction repair, providing a promising reference for clinical treatment [93]. The exosomes derived from circRNA_0002113-deficient MSCs could regulate the RUNX1 nuclear relocation by sponge miR-188-3p to inhibit myocardial infarction [94]. The expression of circHIPK3 in ischemic muscles was reduced, while treatment with exosomes derived from umbilical cord mesenchymal stem cells (UMSC-Exo) significantly increased circHIPK3 expression and improved blood perfusion [95]. Functional studies demonstrated that miR-421/FOXO3a is a direct target of circHIPK3 [95].

4.6. The Role of Exosomal ncRNA from Adipocyte in Cardiovascular Disease

Adipocytes have been shown as a major source of exosomes containing miRNAs [96,97,98]. The results showed that exosomes released from visceral adipocytes carried high levels of miR-27b-3p, and these exosomes could activate ECs by translocating miR-27b-3p and promote endothelial inflammation and atherosclerosis by activating the NF-κB pathway through down-regulation of PPARα [99]. Exosomes released from epididymal white adipose tissue were enriched in miR-23a-3p [100]. These exosomes promoted myocardial fibrosis by translocating miR-23a-3p to fibroblasts, which were converted to myofibroblasts [100]. Meanwhile, it has been suggested that adipocyte-derived exosomes deliver lncRNA-SNHG9 to ECs, which in turn suppresses the expression of the TNF receptor-associated death domain (TRADD), thereby reducing endothelial inflammation and apoptosis [101]. This provides a potential molecular mechanism for the maintenance of endothelial function [101]. Research established that microRNA-31 presented in exosomes derived from adipose-derived stem cells could be penetrated to cardiomyocytes and inhibited FIH1 expression, thus encouraging the activation of HIF-1α with enhancing cell survival ability, promotion of angiogenesis, and inflammatory reaction reduction, consequently diminishing the impact of myocardial infarction [102]. The exosomes released from highly obese adipocytes were enriched in miR-802-5p, which inhibited the expression of HSP60, altered the pathways associated with insulin signaling in cardiac myocytes, and led to the development of insulin resistance [103]. This finding not only deepens the understanding of the mechanism of insulin resistance but also provides new clues for further exploration of the treatment of obesity-related heart diseases [103].

4.7. The Role of Exosomal ncRNA from Immune Cells in Cardiovascular Disease

Expansion and activation of CD4+ T cells in the heart have been identified as contributors to pathological cardiac remodeling. The researchers found that the miR-142-3p in exosomes released by activated CD4+ T cells inhibited the expression of APC, the negative regulator of the WNT signaling pathway, promoting post-ischemic ventricular remodeling [104]. Researchers have found that the crosstalk between cardiomyocytes and macrophages is mediated by the expression of miR-34a-5p/PNUTS signaling pathway in exosomes to exert pro-aging effects [105].
Arteriosclerosis is a common cardiovascular disease associated with chronic inflammation and ES dysfunction. Recent research has found that neutrophil microvesicles, by delivering miR-155 to ECs, drive the development of atherosclerosis [106]. Experimental results demonstrated that miR-155 in exosomes downregulated the expression of SoS1, a gene associated with fibroblast proliferation, and activated inflammatory pathways, thereby regulating the fibrotic response during cardiac injury [107]. Up-regulation of lncRNA 39868 expression in neutrophil-derived exosomes promoted the expression of p-AKT and Bcl-xL by acting on PDGFD and reduced the production of NADPH oxidase 2 (NOX2) and reactive oxygen species (ROS), alleviating ischemia-reperfusion-induced myocardial oxidative stress injury and improving heart function [108].

5. Exosome Therapy

The functional delivery of RNA molecules is an emerging vaccination and therapeutic approach, and mRNA delivery holds great clinical potential. Although naked mRNA delivery has been reported, its efficacy is limited by high plasma RNA degradation rates, low cellular uptake, and nucleic-acid-induced inflammatory responses. Formulating RNA into lipid nanoparticles (LNPs) has been developed as a method to overcome these limitations but LNPs and other synthetic nanoparticles are exogenous entities associated with a range of side effects. Exosomes are the only biologically normal nanovesicles, and compared to LNPs that can cause significant cellular toxicity, exosomes have shown no adverse effects in vitro or in vivo at any tested dose. However, the targeting ability of natural exosomes is limited, which partially restricts their application in cardiovascular disease treatment [109]. Constructing exosomes as drug delivery vehicles in a specific way can improve cardiac targeting and improve the stability of exosomes in the circulation [110]. Methods for engineering exosomes mainly include passive loading, active loading, and biotechnological modifications of donor cells. Each method has its own advantages and disadvantages, and the appropriate method should be selected based on the source, characteristics, and practical application of the exosome to effectively target delivery and stabilize its therapeutic effects. Passive loading involves co-incubation of the target drug with exosomes. Loading efficiency mainly depends on the concentration gradient and hydrophobicity of the drug. Compared to hydrophilic drugs, highly concentrated hydrophobic drugs have higher loading efficiency [111]. The advantage of passive loading is that it does not disrupt the integrity of the exosome membrane but the disadvantage is that the drug payload is lower compared to other loading methods.
Active loading involves directly targeting exosomes using techniques such as electroporation, ultrasound, liposomal freeze-thaw, and mechanical extrusion, etc. These methods are conducive to delivering various types of RNA to target tissues and regulate gene expression in target cells [112]. Some researchers have collected exosomes from cardiac progenitor cells and loaded them with miR-322 using electroporation [113]. Injection of these loaded exosomes into a myocardial infarction mouse model resulted in reduced infarct size, reduced fibrosis, and increased angiogenesis compared to mice injected with miR-322-unloaded exosomes. M2 macrophage-derived exosomes (M2-Exos) exhibited inflammation-targeting ability, and their active loading via electroporation significantly alleviated atherosclerosis [114]. Since exosomes are secreted by donor cells, biotechnological modification of donor cells can be utilized to change their genetic characteristics and enable the integration of molecular components naturally into budding vesicles, and substances internalized into the cell interior can be packaged into secreted exosomes. It has been reported that exosomes derived from MSCs can effectively promote cardiac injury repair. To further enhance the therapeutic effects of these exosomes, Wang and Mentkowski engineered donor cells via lentiviral transduction to produce ischemic myocardium-targeting peptide-lamp2b and cardiomyocyte-specific peptide-lamp2b fusion-expressing exosomes [109,115]; however, the targeting peptide-lamp2b fusion protein related to exosomes was found to be prone to degradation by nuclear proteases during exosome biogenesis. To overcome peptide loss, a glycosylation motif at different positions of the targeting peptide-lamp2b fusion protein was inserted to protect the peptide from degradation and increase the expression of the lamp2b fusion protein in cells and exosomes [116].
The advantages of exosomes over other carriers include biocompatibility and a lower likelihood of triggering innate and adaptive immune responses [117]. Clinical studies have demonstrated that exosomes secreted by dendritic cells can stimulate the immune system and, therefore, can be used as antitumor vaccines [118,119]. Exosome-encapsulated adeno-associated viral vectors are more efficient and less immunogenic than free vectors in delivering gene-therapeutic substances to recipient cells [120]. Manipulation of exosomes in vitro to load specific cargo (such as siRNAs, mRNAs, proteins, and miRNAs) and then using them as drugs or for bioengineering purposes have been reported [121,122]. Exosomes are naturally occurring nanocarriers that can maintain the biological activity of their cargo in living organisms. They possess low immunogenicity and high safety, making them promising for drug delivery. In addition, exosomes can circulate to all compartments in the body and have good potential for non-hepatic targeting of nucleic acid drug delivery. Currently, although preliminary results have been taken, there are several obstacles that need to be overcome for exosomes to become effective nucleic acid carriers, such as improving circulation time and in vivo distribution, enhancing organ-specific targeting, addressing safety concerns related to exosomes derived from tumors, and achieving stable large-scale production. The development of the exosomes industry is still at a very early and budding stage, offering tremendous development opportunities but also facing difficulties in clinical translation and inconsistent evaluation systems.

6. Conclusions

Exosomes are an emerging area of research in cardiovascular disease. Currently, the study of exosomes highly relies on the methods used for isolation and purification, which impose higher requirements on the extraction steps and laboratory equipment. Several ncRNAs have been described in the context of cardiovascular disease specifically for mediating cellular communication (Table 1). The different small molecules carried by exosomes from different sources hold the potential to become therapeutic targets for future cardiovascular diseases and provide a basis for precision medicine (Figure 2). However, further research and exploration are still needed in this regard.

Author Contributions

Conceived and designed the review, X.Z., W.L. and H.C.; wrote the manuscript, X.Z.; drew pictures of the manuscript, S.S.; revise manuscript, G.R. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National 14th Five-Year Key Research and Development Project (No. 2021YFD1600702) and the National Natural Science Foundation Project (No. 32160775).

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  2. Zhu, L.; Sun, H.T.; Wang, S.; Huang, S.L.; Zheng, Y.; Wang, C.Q.; Hu, B.Y.; Qin, W.; Zou, T.T.; Fu, Y.; et al. Isolation and characterization of exosomes for cancer research. J. Hematol. Oncol. 2020, 13, 152. [Google Scholar] [CrossRef] [PubMed]
  3. Jia, G.; Sowers, J.R. Targeting endothelial exosomes for the prevention of cardiovascular disease. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165833. [Google Scholar] [CrossRef] [PubMed]
  4. Wortzel, I.; Dror, S.; Kenific, C.M.; Lyden, D. Exosome-Mediated Metastasis: Communication from a Distance. Dev. Cell. 2019, 49, 347–360. [Google Scholar] [CrossRef]
  5. Qiao, L.; Hu, S.; Huang, K.; Su, T.; Li, Z.; Vandergriff, A.; Cores, J.; Dinh, P.U.; Allen, T.; Shen, D.; et al. Tumor cell-derived exosomes home to their cells of origin and can be used as Trojan horses to deliver cancer drugs. Theranostics 2020, 10, 3474–3487. [Google Scholar] [CrossRef] [PubMed]
  6. Li, S.P.; Lin, Z.X.; Jiang, X.Y.; Yu, X.Y. Exosomal cargo-loading and synthetic exosome-mimics as potential therapeutic tools. Acta Pharmacol. Sin. 2018, 39, 542–551. [Google Scholar] [CrossRef]
  7. Valadi, H.; Ekström, K.; Bossios, A.; Sjöstrand, M.; Lee, J.J.; Lötvall, J.O. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 2007, 9, 654–659. [Google Scholar] [CrossRef]
  8. Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef]
  9. Rodríguez, M.; Bajo-Santos, C.; Hessvik, N.P.; Lorenz, S.; Fromm, B.; Berge, V.; Sandvig, K.; Linē, A.; Llorente, A. Identification of non-invasive miRNAs biomarkers for prostate cancer by deep sequencing analysis of urinary exosomes. Mol. Cancer 2017, 16, 156. [Google Scholar] [CrossRef]
  10. Işın, M.; Uysaler, E.; Özgür, E.; Köseoğlu, H.; Şanlı, Ö.; Yücel, Ö.B.; Gezer, U.; Dalay, N. Exosomal lncRNA-p21 levels may help to distinguish prostate cancer from benign disease. Front. Genet. 2015, 6, 168. [Google Scholar]
  11. Dong, L.; Lin, W.; Qi, P.; Xu, M.D.; Wu, X.; Ni, S.; Huang, D.; Weng, W.W.; Tan, C.; Sheng, W.; et al. Circulating Long RNAs in Serum Extracellular Vesicles: Their Characterization and Potential Application as Biomarkers for Diagnosis of Colorectal Cancer. Cancer Epidemiol. Biomark. Prev. 2016, 25, 1158–1166. [Google Scholar] [CrossRef] [PubMed]
  12. Li, M.; Zeringer, E.; Barta, T.; Schageman, J.; Cheng, A.; Vlassov, A.V. Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2014, 369, 20130502. [Google Scholar] [CrossRef] [PubMed]
  13. Chandy, M.; Rhee, J.W.; Ozen, M.O.; Williams, D.R.; Pepic, L.; Liu, C.; Zhang, H.; Malisa, J.; Lau, E.; Demirci, U.; et al. Atlas of Exosomal microRNAs Secreted From Human iPSC-Derived Cardiac Cell Types. Circulation 2020, 142, 1794–1796. [Google Scholar] [CrossRef]
  14. Boulanger, C.M.; Loyer, X.; Rautou, P.E.; Amabile, N. Extracellular vesicles in coronary artery disease. Nat. Rev. Cardiol. 2017, 14, 259–272. [Google Scholar] [CrossRef]
  15. Yu, W.; Hurley, J.; Roberts, D.; Chakrabortty, S.K.; Enderle, D.; Noerholm, M.; Breakefield, X.O.; Skog, J.K. Exosome-based liquid biopsies in cancer: Opportunities and challenges. Ann. Oncol. 2021, 32, 466–477. [Google Scholar] [CrossRef]
  16. Jin, T.; Gu, J.; Li, Z.; Xu, Z.; Gui, Y. Recent Advances on Extracellular Vesicles in Central Nervous System Diseases. Clin. Interv. Aging 2021, 16, 257–274. [Google Scholar] [CrossRef]
  17. Li, S.; Li, Y.; Chen, B.; Zhao, J.; Yu, S.; Tang, Y.; Zheng, Q.; Li, Y.; Wang, P.; He, X.; et al. exoRBase: A database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 2018, 46, D106–D112. [Google Scholar] [CrossRef] [PubMed]
  18. Eldh, M.; Ekström, K.; Valadi, H.; Sjöstrand, M.; Olsson, B.; Jernås, M.; Lötvall, J. Exosomes communicate protective messages during oxidative stress; possible role of exosomal shuttle RNA. PLoS ONE 2010, 5, e15353. [Google Scholar] [CrossRef]
  19. Parolini, I.; Sargiacomo, M.; Lisanti, M.P.; Peschle, C. Signal transduction and glycophosphatidylinositol-linked proteins (lyn, lck, CD4, CD45, G proteins, and CD55) selectively localize in Triton-insoluble plasma membrane domains of human leukemic cell lines and normal granulocytes. Blood 1996, 87, 3783–3794. [Google Scholar] [CrossRef]
  20. King, H.W.; Michael, M.Z.; Gleadle, J.M. Hypoxic enhancement of exosome release by breast cancer cells. BMC Cancer 2012, 12, 421. [Google Scholar] [CrossRef]
  21. Leroyer, A.S.; Isobe, H.; Lesèche, G.; Castier, Y.; Wassef, M.; Mallat, Z.; Binder, B.R.; Tedgui, A.; Boulanger, C.M. Cellular origins and thrombogenic activity of microparticles isolated from human atherosclerotic plaques. J. Am. Coll. Cardiol. 2007, 49, 772–777. [Google Scholar] [CrossRef] [PubMed]
  22. Hessvik, N.P.; Llorente, A. Current knowledge on exosome biogenesis and release. Cell Mol. Life Sci. 2018, 75, 193–208. [Google Scholar] [CrossRef]
  23. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  24. Chicón-Bosch, M.; Tirado, O.M. Exosomes in Bone Sarcomas: Key Players in Metastasis. Cell 2020, 9, 241. [Google Scholar] [CrossRef]
  25. Svensson, K.J.; Christianson, H.C.; Wittrup, A.; Bourseau-Guilmain, E.; Lindqvist, E.; Svensson, L.M.; Mörgelin, M.; Belting, M. Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1. J. Biol. Chem. 2013, 288, 17713–17724. [Google Scholar] [CrossRef]
  26. Tkach, M.; Kowal, J.; Zucchetti, A.E.; Enserink, L.; Jouve, M.; Lankar, D.; Saitakis, M.; Martin-Jaular, L.; Théry, C. Qualitative differences in T-cell activation by dendritic cell-derived extracellular vesicle subtypes. EMBO J. 2017, 36, 3012–3028. [Google Scholar] [CrossRef]
  27. Munich, S.; Sobo-Vujanovic, A.; Buchser, W.J.; Beer-Stolz, D.; Vujanovic, N.L. Dendritic cell exosomes directly kill tumor cells and activate natural killer cells via TNF superfamily ligands. Oncoimmunology 2012, 1, 1074–1083. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, S.; Zhan, Y.; Luo, J.; Feng, J.; Lu, J.; Zheng, H.; Wen, Q.; Fan, S. Roles of exosomes in the carcinogenesis and clinical therapy of non-small cell lung cancer. Biomed. Pharmacother. 2019, 111, 338–346. [Google Scholar] [CrossRef]
  29. Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell Vesicles 2014, 3, 24641. [Google Scholar] [CrossRef]
  30. Mathieu, M.; Martin-Jaular, L.; Lavieu, G.; Théry, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 2019, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
  31. Kontaraki, J.E.; Marketou, M.E.; Zacharis, E.A.; Parthenakis, F.I.; Vardas, P.E. Differential expression of vascular smooth muscle-modulating microRNAs in human peripheral blood mononuclear cells: Novel targets in essential hypertension. J. Hum. Hypertens. 2014, 28, 510–516. [Google Scholar] [CrossRef] [PubMed]
  32. Yuan, J.; Chen, H.; Ge, D.; Xu, Y.; Xu, H.; Yang, Y.; Gu, M.; Zhou, Y.; Zhu, J.; Ge, T.; et al. Mir-21 Promotes Cardiac Fibrosis After Myocardial Infarction Via Targeting Smad7. Cell Physiol. Biochem. 2017, 42, 2207–2219. [Google Scholar] [CrossRef] [PubMed]
  33. Gao, F.; Kataoka, M.; Liu, N.; Liang, T.; Huang, Z.P.; Gu, F.; Ding, J.; Liu, J.; Zhang, F.; Ma, Q.; et al. Therapeutic role of miR-19a/19b in cardiac regeneration and protection from myocardial infarction. Nat. Commun. 2019, 10, 1802. [Google Scholar] [CrossRef] [PubMed]
  34. Pan, J.X. LncRNA H19 promotes atherosclerosis by regulating MAPK and NF-kB signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 322–328. [Google Scholar]
  35. He, Z.; Zeng, X.; Zhou, D.; Liu, P.; Han, D.; Xu, L.; Bu, T.; Wang, J.; Ke, M.; Pan, X.; et al. LncRNA Chaer Prevents Cardiomyocyte Apoptosis From Acute Myocardial Infarction Through AMPK Activation. Front. Pharmacol. 2021, 12, 649398. [Google Scholar] [CrossRef]
  36. Deng, X.; Liu, Y.; Xu, Z.; Wang, Z. lncRNA Nuclear Factor of Activated T Cells Knockdown Alleviates Hypoxia/Reoxygenation-induced Cardiomyocyte Apoptosis by Upregulating HIF-1α Expression. J. Cardiovasc. Pharmacol. 2022, 79, 479–488. [Google Scholar] [CrossRef]
  37. Holdt, L.M.; Stahringer, A.; Sass, K.; Pichler, G.; Kulak, N.A.; Wilfert, W.; Kohlmaier, A.; Herbst, A.; Northoff, B.H.; Nicolaou, A.; et al. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat. Commun. 2016, 7, 12429. [Google Scholar] [CrossRef]
  38. Zhang, S.; Song, G.; Yuan, J.; Qiao, S.; Xu, S.; Si, Z.; Yang, Y.; Xu, X.; Wang, A. Circular RNA circ_0003204 inhibits proliferation, migration and tube formation of endothelial cell in atherosclerosis via miR-370-3p/TGFβ R2/phosph-SMAD3 axis. J. Biomed. Sci. 2020, 27, 11. [Google Scholar] [CrossRef]
  39. Zhu, Y.; Zhao, P.; Sun, L.; Lu, Y.; Zhu, W.; Zhang, J.; Xiang, C.; Mao, Y.; Chen, Q.; Zhang, F. Overexpression of circRNA SNRK targets miR-103-3p to reduce apoptosis and promote cardiac repair through GSK3β/β-catenin pathway in rats with myocardial infarction. Cell Death Discov. 2021, 7, 84. [Google Scholar] [CrossRef]
  40. de Jong, O.G.; Verhaar, M.C.; Chen, Y.; Vader, P.; Gremmels, H.; Posthuma, G.; Schiffelers, R.M.; Gucek, M.; van Balkom, B.W. Cellular stress conditions are reflected in the protein and RNA content of endothelial cell-derived exosomes. J. Extracell Vesicles. 2012, 1, 10. [Google Scholar] [CrossRef]
  41. Xu, Z.; Chen, Y.; Ma, L.; Chen, Y.; Liu, J.; Guo, Y.; Yu, T.; Zhang, L.; Zhu, L.; Shu, Y. Role of exosomal non-coding RNAs from tumor cells and tumor-associated macrophages in the tumor microenvironment. Mol. Ther. 2022, 30, 3133–3154. [Google Scholar] [CrossRef] [PubMed]
  42. Loyer, X.; Vion, A.C.; Tedgui, A.; Boulanger, C.M. Microvesicles as cell-cell messengers in cardiovascular diseases. Circ. Res. 2014, 114, 345–353. [Google Scholar] [CrossRef] [PubMed]
  43. Jansen, F.; Nickenig, G.; Werner, N. Extracellular Vesicles in Cardiovascular Disease: Potential Applications in Diagnosis, Prognosis, and Epidemiology. Circ. Res. 2017, 120, 1649–1657. [Google Scholar] [CrossRef] [PubMed]
  44. D’Ascenzo, F.; Femminò, S.; Ravera, F.; Angelini, F.; Caccioppo, A.; Franchin, L.; Grosso, A.; Comità, S.; Cavallari, C.; Penna, C.; et al. Extracellular vesicles from patients with Acute Coronary Syndrome impact on ischemia-reperfusion injury. Pharmacol. Res. 2021, 170, 105715. [Google Scholar] [CrossRef] [PubMed]
  45. Femminò, S.; D’Ascenzo, F.; Ravera, F.; Comità, S.; Angelini, F.; Caccioppo, A.; Franchin, L.; Grosso, A.; Thairi, C.; Venturelli, E.; et al. Percutaneous Coronary Intervention (PCI) Reprograms Circulating Extracellular Vesicles from ACS Patients Impairing Their Cardio-Protective Properties. Int. J. Mol. Sci. 2021, 22, 10270. [Google Scholar] [CrossRef] [PubMed]
  46. Su, S.A.; Xie, Y.; Fu, Z.; Wang, Y.; Wang, J.A.; Xiang, M. Emerging role of exosome-mediated intercellular communication in vascular remodeling. Oncotarget 2017, 8, 25700–25712. [Google Scholar] [CrossRef] [PubMed]
  47. Aliotta, J.M.; Pereira, M.; Wen, S.; Dooner, M.S.; Del Tatto, M.; Papa, E.; Goldberg, L.R.; Baird, G.L.; Ventetuolo, C.E.; Quesenberry, P.J.; et al. Exosomes induce and reverse monocrotaline-induced pulmonary hypertension in mice. Cardiovasc. Res. 2016, 110, 319–330. [Google Scholar] [CrossRef]
  48. Zhao, L.; Luo, H.; Li, X.; Li, T.; He, J.; Qi, Q.; Liu, Y.; Yu, Z. Exosomes Derived from Human Pulmonary Artery Endothelial Cells Shift the Balance between Proliferation and Apoptosis of Smooth Muscle Cells. Cardiology 2017, 137, 43–53. [Google Scholar] [CrossRef]
  49. Deng, L.; Blanco, F.J.; Stevens, H.; Lu, R.; Caudrillier, A.; McBride, M.; McClure, J.D.; Grant, J.; Thomas, M.; Frid, M.; et al. MicroRNA-143 Activation Regulates Smooth Muscle and Endothelial Cell Crosstalk in Pulmonary Arterial Hypertension. Circ. Res. 2015, 117, 870–883. [Google Scholar] [CrossRef]
  50. Kuwabara, Y.; Ono, K.; Horie, T.; Nishi, H.; Nagao, K.; Kinoshita, M.; Watanabe, S.; Baba, O.; Kojima, Y.; Shizuta, S.; et al. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ. Cardiovasc. Genet. 2011, 4, 446–454. [Google Scholar] [CrossRef]
  51. Farooqi, A.A.; Desai, N.N.; Qureshi, M.Z.; Librelotto, D.R.N.; Gasparri, M.L.; Bishayee, A.; Nabavi, S.M.; Curti, V.; Daglia, M. Exosome biogenesis, bioactivities and functions as new delivery systems of natural compounds. Biotechnol. Adv. 2018, 36, 328–334. [Google Scholar] [CrossRef] [PubMed]
  52. Jansen, F.; Stumpf, T.; Proebsting, S.; Franklin, B.S.; Wenzel, D.; Pfeifer, P.; Flender, A.; Schmitz, T.; Yang, X.; Fleischmann, B.K.; et al. Intercellular transfer of miR-126-3p by endothelial microparticles reduces vascular smooth muscle cell proliferation and limits neointima formation by inhibiting LRP6. J. Mol. Cell Cardiol. 2017, 104, 43–52. [Google Scholar] [CrossRef] [PubMed]
  53. Lee, J.K.; Park, S.R.; Jung, B.K.; Jeon, Y.K.; Lee, Y.S.; Kim, M.K.; Kim, Y.G.; Jang, J.Y.; Kim, C.W. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS ONE 2013, 8, e84256. [Google Scholar] [CrossRef] [PubMed]
  54. Zheng, B.; Yin, W.N.; Suzuki, T.; Zhang, X.H.; Zhang, Y.; Song, L.L.; Jin, L.S.; Zhan, H.; Zhang, H.; Li, J.S.; et al. Exosome-Mediated miR-155 Transfer from Smooth Muscle Cells to Endothelial Cells Induces Endothelial Injury and Promotes Atherosclerosis. Mol. Ther. 2017, 25, 1279–1294. [Google Scholar] [CrossRef]
  55. Wang, S.; Shi, M.; Li, J.; Zhang, Y.; Wang, W.; Xu, P.; Li, Y. Endothelial cell-derived exosomal circHIPK3 promotes the proliferation of vascular smooth muscle cells induced by high glucose via the miR-106a-5p/Foxo1/Vcam1 pathway. Aging 2021, 13, 25241–25255. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, Y.; Li, Q.; Hosen, M.R.; Zietzer, A.; Flender, A.; Levermann, P.; Schmitz, T.; Frühwald, D.; Goody, P.; Nickenig, G.; et al. Atherosclerotic Conditions Promote the Packaging of Functional MicroRNA-92a-3p Into Endothelial Microvesicles. Circ. Res. 2019, 124, 575–587. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, L.; Hu, L.; Li, Q.; Ma, J.; Li, H. Exosome-encapsulated miR-505 from ox-LDL-treated vascular endothelial cells aggravates atherosclerosis by inducing NET formation. Acta Biochim. Biophys. Sin. 2019, 51, 1233–1241. [Google Scholar] [CrossRef]
  58. Halkein, J.; Tabruyn, S.P.; Ricke-Hoch, M.; Haghikia, A.; Nguyen, N.Q.; Scherr, M.; Castermans, K.; Malvaux, L.; Lambert, V.; Thiry, M.; et al. MicroRNA-146a is a therapeutic target and biomarker for peripartum cardiomyopathy. J. Clin. Investig. 2013, 123, 2143–2154. [Google Scholar] [CrossRef]
  59. Hergenreider, E.; Heydt, S.; Tréguer, K.; Boettger, T.; Horrevoets, A.J.; Zeiher, A.M.; Scheffer, M.P.; Frangakis, A.S.; Yin, X.; Mayr, M.; et al. Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nat. Cell Biol. 2012, 14, 249–256. [Google Scholar] [CrossRef]
  60. Su, Q.; Lv, X.W.; Xu, Y.L.; Cai, R.P.; Dai, R.X.; Yang, X.H.; Zhao, W.K.; Kong, B.H. Exosomal LINC00174 derived from vascular endothelial cells attenuates myocardial I/R injury via p53-mediated autophagy and apoptosis. Mol. Ther. Nucleic Acids 2021, 23, 1304–1322. [Google Scholar] [CrossRef]
  61. Xiong, F.; Mao, R.; Zhang, L.; Zhao, R.; Tan, K.; Liu, C.; Xu, J.; Du, G.; Zhang, T. CircNPHP4 in monocyte-derived small extracellular vesicles controls heterogeneous adhesion in coronary heart atherosclerotic disease. Cell Death Dis. 2021, 12, 948. [Google Scholar] [CrossRef] [PubMed]
  62. Xia, H.; Wu, Y.; Zhao, J.; Li, W.; Lu, L.; Ma, H.; Cheng, C.; Sun, J.; Xiang, Q.; Bian, T.; et al. The aberrant cross-talk of epithelium-macrophages via METTL3-regulated extracellular vesicle miR-93 in smoking-induced emphysema. Cell Biol. Toxicol. 2022, 38, 167–183. [Google Scholar] [CrossRef] [PubMed]
  63. Travers, J.G.; Kamal, F.A.; Robbins, J.; Yutzey, K.E.; Blaxall, B.C. Cardiac Fibrosis: The Fibroblast Awakens. Circ. Res. 2016, 118, 1021–1040. [Google Scholar] [CrossRef] [PubMed]
  64. Wu, L.; Belasco, J.G. Examining the influence of microRNAs on translation efficiency and on mRNA deadenylation and decay. Methods Enzymol. 2008, 449, 373–393. [Google Scholar] [PubMed]
  65. Kellouche, S.; Mourah, S.; Bonnefoy, A.; Schoëvaert, D.; Podgorniak, M.P.; Calvo, F.; Hoylaerts, M.F.; Legrand, C.; Dosquet, C. Platelets, thrombospondin-1 and human dermal fibroblasts cooperate for stimulation of endothelial cell tubulogenesis through VEGF and PAI-1 regulation. Exp. Cell Res. 2007, 313, 486–499. [Google Scholar] [CrossRef] [PubMed]
  66. Paunescu, V.; Bojin, F.M.; Tatu, C.A.; Gavriliuc, O.I.; Rosca, A.; Gruia, A.T.; Tanasie, G.; Bunu, C.; Crisnic, D.; Gherghiceanu, M.; et al. Tumour-associated fibroblasts and mesenchymal stem cells: More similarities than differences. J. Cell Mol. Med. 2011, 15, 635–646. [Google Scholar] [CrossRef]
  67. Tille, J.C.; Pepper, M.S. Mesenchymal cells potentiate vascular endothelial growth factor-induced angiogenesis in vitro. Exp. Cell Res. 2002, 280, 179–191. [Google Scholar] [CrossRef]
  68. Berthod, F.; Germain, L.; Tremblay, N.; Auger, F.A. Extracellular matrix deposition by fibroblasts is necessary to promote capillary-like tube formation in vitro. J. Cell Physiol. 2006, 207, 491–498. [Google Scholar] [CrossRef]
  69. Bang, C.; Batkai, S.; Dangwal, S.; Gupta, S.K.; Foinquinos, A.; Holzmann, A.; Just, A.; Remke, J.; Zimmer, K.; Zeug, A.; et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J. Clin. Investig. 2014, 124, 2136–2146. [Google Scholar] [CrossRef]
  70. Liu, D.; Yang, M.; Yao, Y.; He, S.; Wang, Y.; Cao, Z.; Chen, H.; Fu, Y.; Liu, H.; Zhao, Q. Cardiac Fibroblasts Promote Ferroptosis in Atrial Fibrillation by Secreting Exo-miR-23a-3p Targeting SLC7A11. Oxid. Med. Cell Longev. 2022, 2022, 3961495. [Google Scholar] [CrossRef]
  71. Ranjan, P.; Kumari, R.; Goswami, S.K.; Li, J.; Pal, H.; Suleiman, Z.; Cheng, Z.; Krishnamurthy, P.; Kishore, R.; Verma, S.K. Myofibroblast-Derived Exosome Induce Cardiac Endothelial Cell Dysfunction. Front. Cardiovasc. Med. 2021, 8, 676267. [Google Scholar] [CrossRef] [PubMed]
  72. Luo, H.; Li, X.; Li, T.; Zhao, L.; He, J.; Zha, L.; Qi, Q.; Yu, Z. microRNA-423-3p exosomes derived from cardiac fibroblasts mediates the cardioprotective effects of ischaemic post-conditioning. Cardiovasc. Res. 2019, 115, 1189–1204. [Google Scholar] [CrossRef] [PubMed]
  73. Wang, Y.; Li, C.; Zhao, R.; Qiu, Z.; Shen, C.; Wang, Z.; Liu, W.; Zhang, W.; Ge, J.; Shi, B. CircUbe3a from M2 macrophage-derived small extracellular vesicles mediates myocardial fibrosis after acute myocardial infarction. Theranostics 2021, 11, 6315–6333. [Google Scholar] [CrossRef] [PubMed]
  74. Wang, L.; Zhang, J. Exosomal lncRNA AK139128 Derived from Hypoxic Cardiomyocytes Promotes Apoptosis and Inhibits Cell Proliferation in Cardiac Fibroblasts. Int. J. Nanomed. 2020, 15, 3363–3376. [Google Scholar] [CrossRef]
  75. Yang, J.; Yu, X.; Xue, F.; Li, Y.; Liu, W.; Zhang, S. Exosomes derived from cardiomyocytes promote cardiac fibrosis via myocyte-fibroblast cross-talk. Am. J. Transl. Res. 2018, 10, 4350–4366. [Google Scholar]
  76. Hao, H.; Yan, S.; Zhao, X.; Han, X.; Fang, N.; Zhang, Y.; Dai, C.; Li, W.; Yu, H.; Gao, Y.; et al. Atrial myocyte-derived exosomal microRNA contributes to atrial fibrosis in atrial fibrillation. J. Transl. Med. 2022, 20, 407. [Google Scholar] [CrossRef]
  77. Wang, X.; Morelli, M.B.; Matarese, A.; Sardu, C.; Santulli, G. Cardiomyocyte-derived exosomal microRNA-92a mediates post-ischemic myofibroblast activation both in vitro and ex vivo. ESC Heart Fail. 2020, 7, 284–288. [Google Scholar] [CrossRef]
  78. Wang, X.; Huang, W.; Liu, G.; Cai, W.; Millard, R.W.; Wang, Y.; Chang, J.; Peng, T.; Fan, G.C. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. J. Mol. Cell Cardiol. 2014, 74, 139–150. [Google Scholar] [CrossRef]
  79. Ge, X.; Meng, Q.; Zhuang, R.; Yuan, D.; Liu, J.; Lin, F.; Fan, H.; Zhou, X. Circular RNA expression alterations in extracellular vesicles isolated from murine heart post ischemia/reperfusion injury. Int. J. Cardiol. 2019, 296, 136–140. [Google Scholar] [CrossRef]
  80. Wang, Y.; Zhao, R.; Liu, W.; Wang, Z.; Rong, J.; Long, X.; Liu, Z.; Ge, J.; Shi, B. Exosomal circHIPK3 Released from Hypoxia-Pretreated Cardiomyocytes Regulates Oxidative Damage in Cardiac Microvascular Endothelial Cells via the miR-29a/IGF-1 Pathway. Oxid. Med. Cell Longev. 2019, 2019, 7954657. [Google Scholar] [CrossRef]
  81. Wang, Y.; Zhao, R.; Shen, C.; Liu, W.; Yuan, J.; Li, C.; Deng, W.; Wang, Z.; Zhang, W.; Ge, J.; et al. Exosomal CircHIPK3 Released from Hypoxia-Induced Cardiomyocytes Regulates Cardiac Angiogenesis after Myocardial Infarction. Oxid. Med. Cell Longev. 2020, 2020, 8418407. [Google Scholar] [CrossRef] [PubMed]
  82. Xia, W.; Chen, H.; Xie, C.; Hou, M. Long-noncoding RNA MALAT1 sponges microRNA-92a-3p to inhibit doxorubicin-induced cardiac senescence by targeting ATG4a. Aging 2020, 12, 8241–8260. [Google Scholar] [CrossRef]
  83. Kilchert, C.; Wittmann, S.; Vasiljeva, L. The regulation and functions of the nuclear RNA exosome complex. Nat. Rev. Mol. Cell Biol. 2016, 17, 227–239. [Google Scholar] [CrossRef] [PubMed]
  84. Khan, M.; Nickoloff, E.; Abramova, T.; Johnson, J.; Verma, S.K.; Krishnamurthy, P.; Mackie, A.R.; Vaughan, E.; Garikipati, V.N.; Benedict, C.; et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ. Res. 2015, 117, 52–64. [Google Scholar] [CrossRef] [PubMed]
  85. Lee, J.Y.; Chung, J.; Byun, Y.; Kim, K.H.; An, S.H.; Kwon, K. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Protect Cardiomyocytes from Doxorubicin-Induced Cardiomyopathy by Upregulating Survivin Expression via the miR-199a-3p-Akt-Sp1/p53 Signaling Pathway. Int. J. Mol. Sci. 2021, 22, 7102. [Google Scholar] [CrossRef] [PubMed]
  86. Ning, W.; Li, S.; Yang, W.; Yang, B.; Xin, C.; Ping, X.; Huang, C.; Gu, Y.; Guo, L. Blocking exosomal miRNA-153-3p derived from bone marrow mesenchymal stem cells ameliorates hypoxia-induced myocardial and microvascular damage by targeting the ANGPT1-mediated VEGF/PI3k/Akt/eNOS pathway. Cell Signal. 2021, 77, 109812. [Google Scholar] [CrossRef]
  87. Wang, Y.; Zhang, L.; Li, Y.; Chen, L.; Wang, X.; Guo, W.; Zhang, X.; Qin, G.; He, S.H.; Zimmerman, A.; et al. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int. J. Cardiol. 2015, 192, 61–69. [Google Scholar] [CrossRef]
  88. Huang, P.; Wang, L.; Li, Q.; Tian, X.; Xu, J.; Xu, J.; Xiong, Y.; Chen, G.; Qian, H.; Jin, C.; et al. Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc. Res. 2020, 116, 353–367. [Google Scholar] [CrossRef]
  89. Mao, Q.; Liang, X.L.; Zhang, C.L.; Pang, Y.H.; Lu, Y.X. LncRNA KLF3-AS1 in human mesenchymal stem cell-derived exosomes ameliorates pyroptosis of cardiomyocytes and myocardial infarction through miR-138-5p/Sirt1 axis. Stem. Cell Res. Ther. 2019, 10, 393. [Google Scholar] [CrossRef]
  90. Harrell, C.R.; Jovicic, N.; Djonov, V.; Arsenijevic, N.; Volarevic, V. Mesenchymal Stem Cell-Derived Exosomes and Other Extracellular Vesicles as New Remedies in the Therapy of Inflammatory Diseases. Cells 2019, 8, 1605. [Google Scholar] [CrossRef]
  91. Liu, R.; Tang, A.; Wang, X.; Chen, X.; Zhao, L.; Xiao, Z.; Shen, S. Inhibition of lncRNA NEAT1 suppresses the inflammatory response in IBD by modulating the intestinal epithelial barrier and by exosome-mediated polarization of macrophages. Int. J. Mol. Med. 2018, 42, 2903–2913. [Google Scholar] [CrossRef] [PubMed]
  92. Sun, Z.; Yang, S.; Zhou, Q.; Wang, G.; Song, J.; Li, Z.; Zhang, Z.; Xu, J.; Xia, K.; Chang, Y.; et al. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Mol. Cancer 2018, 17, 82. [Google Scholar] [CrossRef] [PubMed]
  93. Li, C.X.; Song, J.; Li, X.; Zhang, T.; Li, Z.M. Circular RNA 0001273 in exosomes derived from human umbilical cord mesenchymal stem cells (UMSCs) in myocardial infarction. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 10086–10095. [Google Scholar]
  94. Tian, T.; Li, F.; Chen, R.; Wang, Z.; Su, X.; Yang, C. Therapeutic Potential of Exosomes Derived From circRNA_0002113 Lacking Mesenchymal Stem Cells in Myocardial Infarction. Front. Cell Dev. Biol. 2022, 9, 779524. [Google Scholar] [CrossRef] [PubMed]
  95. Yan, B.; Zhang, Y.; Liang, C.; Liu, B.; Ding, F.; Wang, Y.; Zhu, B.; Zhao, R.; Yu, X.Y.; Li, Y. Stem cell-derived exosomes prevent pyroptosis and repair ischemic muscle injury through a novel exosome/circHIPK3/ FOXO3a pathway. Theranostics 2020, 10, 6728–6742. [Google Scholar] [CrossRef]
  96. Chen, Y.; Buyel, J.J.; Hanssen, M.J.; Siegel, F.; Pan, R.; Naumann, J.; Schell, M.; van der Lans, A.; Schlein, C.; Froehlich, H.; et al. Exosomal microRNA miR-92a concentration in serum reflects human brown fat activity. Nat. Commun. 2016, 7, 11420. [Google Scholar] [CrossRef]
  97. Ogawa, R.; Tanaka, C.; Sato, M.; Nagasaki, H.; Sugimura, K.; Okumura, K.; Nakagawa, Y.; Aoki, N. Adipocyte-derived microvesicles contain RNA that is transported into macrophages and might be secreted into blood circulation. Biochem. Biophys. Res. Commun. 2010, 398, 723–729. [Google Scholar] [CrossRef]
  98. Thomou, T.; Mori, M.A.; Dreyfuss, J.M.; Konishi, M.; Sakaguchi, M.; Wolfrum, C.; Rao, T.N.; Winnay, J.N.; Garcia-Martin, R.; Grinspoon, S.K.; et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 2017, 542, 450–455. [Google Scholar] [CrossRef]
  99. Tang, Y.; Yang, L.J.; Liu, H.; Song, Y.J.; Yang, Q.Q.; Liu, Y.; Qian, S.W.; Tang, Q.Q. Exosomal miR-27b-3p secreted by visceral adipocytes contributes to endothelial inflammation and atherogenesis. Cell Rep. 2023, 42, 111948. [Google Scholar] [CrossRef]
  100. Su, M.; Li, W.; Yuan, Y.; Liu, S.; Liang, C.; Liu, H.E.; Zhang, R.; Liu, Y.; Sun, L.I.; Wei, Y.; et al. Epididymal white adipose tissue promotes angiotensin II-induced cardiac fibrosis in an exosome-dependent manner. Transl. Res. 2022, 248, 51–67. [Google Scholar] [CrossRef]
  101. Song, Y.; Li, H.; Ren, X.; Li, H.; Feng, C. SNHG9, delivered by adipocyte-derived exosomes, alleviates inflammation and apoptosis of endothelial cells through suppressing TRADD expression. Eur. J. Pharmacol. 2020, 872, 172977. [Google Scholar] [CrossRef] [PubMed]
  102. Zhu, D.; Wang, Y.; Thomas, M.; McLaughlin, K.; Oguljahan, B.; Henderson, J.; Yang, Q.; Chen, Y.E.; Liu, D. Exosomes from adipose-derived stem cells alleviate myocardial infarction via microRNA-31/FIH1/HIF-1α pathway. J. Mol. Cell Cardiol. 2022, 162, 10–19. [Google Scholar] [CrossRef] [PubMed]
  103. Wen, Z.; Li, J.; Fu, Y.; Zheng, Y.; Ma, M.; Wang, C. Hypertrophic Adipocyte-Derived Exosomal miR-802-5p Contributes to Insulin Resistance in Cardiac Myocytes Through Targeting HSP60. Obesity 2020, 28, 1932–1940. [Google Scholar] [CrossRef] [PubMed]
  104. Cai, L.; Chao, G.; Li, W.; Zhu, J.; Li, F.; Qi, B.; Wei, Y.; Chen, S.; Zhou, G.; Lu, X.; et al. Activated CD4+ T cells-derived exosomal miR-142-3p boosts post-ischemic ventricular remodeling by activating myofibroblast. Aging 2020, 12, 7380–7396. [Google Scholar] [CrossRef] [PubMed]
  105. Xia, W.; Chen, H.; Chen, D.; Ye, Y.; Xie, C.; Hou, M. PD-1 inhibitor inducing exosomal miR-34a-5p expression mediates the cross talk between cardiomyocyte and macrophage in immune checkpoint inhibitor-related cardiac dysfunction. J. Immunother. Cancer 2020, 8, e001293. [Google Scholar] [CrossRef] [PubMed]
  106. Gomez, I.; Ward, B.; Souilhol, C.; Recarti, C.; Ariaans, M.; Johnston, J.; Burnett, A.; Mahmoud, M.; Luong, L.A.; West, L.; et al. Neutrophil microvesicles drive atherosclerosis by delivering miR-155 to atheroprone endothelium. Nat. Commun. 2020, 11, 214. [Google Scholar] [CrossRef]
  107. Wang, C.; Zhang, C.; Liu, L.; Xi, A.; Chen, B.; Li, Y.; Du, J. Macrophage-Derived mir-155-Containing Exosomes Suppress Fibroblast Proliferation and Promote Fibroblast Inflammation during Cardiac Injury. Mol. Ther. 2017, 25, 192–204. [Google Scholar] [CrossRef]
  108. Zhai, T.Y.; Cui, B.H.; Zhou, Y.; Xu, X.Y.; Zou, L.; Lin, X.; Zhu, X.S.; Zhang, S.W.; Xie, W.L.; Cheng, Y.Y.; et al. Exosomes Released from CaSR-Stimulated PMNs Reduce Ischaemia/Reperfusion Injury. Oxid. Med. Cell Longev. 2021, 2021, 3010548. [Google Scholar] [CrossRef]
  109. Wang, X.; Chen, Y.; Zhao, Z.; Meng, Q.; Yu, Y.; Sun, J.; Yang, Z.; Chen, Y.; Li, J.; Ma, T.; et al. Engineered Exosomes with Ischemic Myocardium-Targeting Peptide for Targeted Therapy in Myocardial Infarction. J. Am. Heart Assoc. 2018, 7, e008737. [Google Scholar] [CrossRef]
  110. Tominaga, N.; Yoshioka, Y.; Ochiya, T. A novel platform for cancer therapy using extracellular vesicles. Adv. Drug Deliv. Rev. 2015, 95, 50–55. [Google Scholar] [CrossRef]
  111. Raimondo, S.; Giavaresi, G.; Lorico, A.; Alessandro, R. Extracellular Vesicles as Biological Shuttles for Targeted Therapies. Int. J. Mol. Sci. 2019, 20, 1848. [Google Scholar] [CrossRef] [PubMed]
  112. Shahabipour, F.; Banach, M.; Sahebkar, A. Exosomes as nanocarriers for siRNA delivery: Paradigms and challenges. Arch. Med. Sci. 2016, 12, 1324–1326. [Google Scholar] [CrossRef]
  113. 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] [PubMed]
  114. Wu, G.; Zhang, J.; Zhao, Q.; Zhuang, W.; Ding, J.; Zhang, C.; Gao, H.; Pang, D.W.; Pu, K.; Xie, H.Y. Molecularly Engineered Macrophage-Derived Exosomes with Inflammation Tropism and Intrinsic Heme Biosynthesis for Atherosclerosis Treatment. Angew Chem. Int. Ed. Engl. 2020, 59, 4068–4074. [Google Scholar] [CrossRef] [PubMed]
  115. Mentkowski, K.I.; Lang, J.K. Exosomes engineered to express a cardiomyocyte binding peptide demonstrate improved cardiac retention in vivo. Sci. Rep. 2019, 9, 10041. [Google Scholar] [CrossRef] [PubMed]
  116. Hung, M.E.; Leonard, J.N. Stabilization of exosome-targeting peptides via engineered glycosylation. J. Biol. Chem. 2015, 290, 8166–8172. [Google Scholar] [CrossRef]
  117. Manole, M.D.; Kochanek, P.M.; Fink, E.L.; Clark, R.S. Postcardiac arrest syndrome: Focus on the brain. Curr. Opin. Pediatr. 2009, 21, 745–750. [Google Scholar] [CrossRef]
  118. Heusch, G.; Kleinbongard, P.; Böse, D.; Levkau, B.; Haude, M.; Schulz, R.; Erbel, R. Coronary microembolization: From bedside to bench and back to bedside. Circulation 2009, 120, 1822–1836. [Google Scholar] [CrossRef]
  119. Dezfulian, C.; Shiva, S.; Alekseyenko, A.; Pendyal, A.; Beiser, D.G.; Munasinghe, J.P.; Anderson, S.A.; Chesley, C.F.; Vanden Hoek, T.L.; Gladwin, M.T. Nitrite therapy after cardiac arrest reduces reactive oxygen species generation, improves cardiac and neurological function, and enhances survival via reversible inhibition of mitochondrial complex I. Circulation 2009, 120, 897–905. [Google Scholar] [CrossRef]
  120. Bonauer, A.; Carmona, G.; Iwasaki, M.; Mione, M.; Koyanagi, M.; Fischer, A.; Burchfield, J.; Fox, H.; Doebele, C.; Ohtani, K.; et al. MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice. Science 2009, 324, 1710–1713. [Google Scholar] [CrossRef]
  121. Chu, C.H.; Huang, C.Y.; Lu, M.C.; Lin, J.A.; Tsai, F.J.; Tsai, C.H.; Chu, C.Y.; Kuo, W.H.; Chen, L.M.; Chen, L.Y. Enhancement of AG1024-induced H9c2 cardiomyoblast cell apoptosis via the interaction of IGF2R with Galpha proteins and its downstream PKA and PLC-beta modulators by IGF-II. Chin. J. Physiol. 2009, 52, 31–37. [Google Scholar] [CrossRef] [PubMed]
  122. Nishida, T.; Yu, J.D.; Minamishima, S.; Sips, P.Y.; Searles, R.J.; Buys, E.S.; Janssens, S.; Brouckaert, P.; Bloch, K.D.; Ichinose, F. Protective effects of nitric oxide synthase 3 and soluble guanylate cyclase on the outcome of cardiac arrest and cardiopulmonary resuscitation in mice. Crit. Care Med. 2009, 37, 256–262. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Structure, composition, secretion, and cellular entry of exosomes. Note: (A) means early endosomes, (B) means multivesicular bodies (MVBs), (C) means late endosomes and (D) means recipient cells.
Figure 1. Structure, composition, secretion, and cellular entry of exosomes. Note: (A) means early endosomes, (B) means multivesicular bodies (MVBs), (C) means late endosomes and (D) means recipient cells.
Ijms 24 16197 g001
Figure 2. Schematic diagram of intercellular communication mediated by exosomal ncRNAs in cardiovascular disease.
Figure 2. Schematic diagram of intercellular communication mediated by exosomal ncRNAs in cardiovascular disease.
Ijms 24 16197 g002
Table 1. ncRNAs and Cardiovascular Diseases.
Table 1. ncRNAs and Cardiovascular Diseases.
Exosomal ncRNATargetCell Type/CommunicationRefs.
miR-16VEGFVascular smooth muscle cells–Endothelial cells[53]
miR-155miR-155Vascular smooth muscle cells–Endothelial cells[54]
circHIPK3miR-106a-5pMouse aortic endothelial cells–Vascular smooth muscle cells[55]
miR-92a-3pTHBS1Endothelial cells–recipient Endothelial cells[56]
miR-505SIRT3Vascular endothelial cells–Macrophages[57]
miR-146aErbb4, Notch1, Irak1Endothelial cells–Cardiomyocytes[58]
miRNA-143/145UnknownEndothelial cells–Smooth muscle cells[59]
LINC00174SRSF1Vascular endothelial cells–Myocardial cells[60]
circNPHP4miR-1231-EGFRMonocytes–Coronary artery endothelial cells[61]
miR-93MMP12Macrophages–Bronchial epithelial cells[62]
miR-21_3pSORBS2, PDLIM5Cardiac fibroblasts–Cardiomyocytes[69]
miR-23a-3pSLC7A11Cardiac fibroblasts–Cardiomyocytes[70]
miR-200a-3qVEGF-A/PIGFFibroblasts–Cardiac endothelial cells[71]
miR-423-3pRAP2CCardiac fibroblasts–Cardiomyocytes[72]
circRNAbe3amiR-138-5p/RhoCM2 macrophages–Cardiac fibroblasts[73]
lncRNA AK139128UnknownCardiomyocytes–Fibroblasts[74]
miR-208aDyrk2Cardiomyocytes–Fibroblasts[75]
miR-210-3pGPD1L-PI3K/AKT Myocytes–Fibroblasts[76]
miR-92asmad7Cardiomyocytes–Myofibroblasts[77]
miR-320IGF-1, Hsp20, Ets2Cardiomyocytes–Endothelial cells[78]
circHIPK3miR-29a-IGF-1Cardiomyocytes–Cardiac microvascular endothelial cells[80,81]
lncRNA-AK139128UnknownCardiomyocytes–Cardiac fibroblasts[74]
lncRNA-MALAT1miR-92a-3p/ATG4aStem cells–Cardiomyocytes[82]
miR-294UnknownEmbryonic stem cells–Cardiomyocytes[84]
miR-199a-3pAkt-Sp1/p53 signaling pathwayMesenchymal stem cells–Cardiomyocytes[85]
miRNA-153-3pVEGF/VEGFR2/PI3K/Akt/eNOS pathwayMesenchymal stem cells–Cardiomyocytes[86]
miRNA-21, miRNA-210UnknownInduced pluripotent stem cells–Cardiomyocytes[87]
lncRNA-UCA1miR-873-5p/XIAPMesenchymal stem cells–Cardiomyocytes[88]
lncRNA KLF3-AS1miR-138-5p/SIRT1Mesenchymal stem cells–Cardiomyocytes[89]
lncRNA H19miR-675/VEGF, ICAM-1Mesenchymal stem cells–Cardiomyocytes[90]
lncRNA-NEAT1miR-142-3p/FOXO1Adipose mesenchymal stem cells–Macrophages[91]
lncRNA-UCA1miR-143/Bcl-2Human umbilical cord mesenchymal stem cells–Cardiomyocytes[92]
circRNA-0001273UnknownUmbilical cord mesenchymal stem cells–Myocardial cells[93]
circRNA_0002113miR-188-3p/RUNX1 axisMesenchymal stem cells–Myocardial cells[94]
circHIPK3miR-421/FOXO3aUmbilical cord mesenchymal stem cells[95]
miR-27b-3pPPARαVisceral adipocytes–Endothelial cells[99]
miR-23a-3pRAP1Epididymal white adipose tissue–Myocardial fibrosis[100]
lncRNA-SNHG9TRADDAdipocytes–Endothelial cells[101]
miRNA-31FIH1Adipose-derived stem cells–Cardiomyocytes[102]
miR-802-5pHSP60Adipocytes–Cardiac myocytes[103]
miR-142-3pWNT signaling pathwayCD4+ T cells–Cardiac myofibroblasts[104]
miR-34a-5pPNUTS signaling pathwayCardiomyocytes–Macrophages[105]
miR-155VEGF signaling pathwayNeutrophil microvesicles–Endothelial cells[106]
miR-155Son of Sevenless 1Macrophages–Fibroblasts[107]
lncRNA 39868PDGFDNeutrophils[108]
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

Zhang, X.; Sun, S.; Ren, G.; Liu, W.; Chen, H. Advances in Intercellular Communication Mediated by Exosomal ncRNAs in Cardiovascular Disease. Int. J. Mol. Sci. 2023, 24, 16197. https://doi.org/10.3390/ijms242216197

AMA Style

Zhang X, Sun S, Ren G, Liu W, Chen H. Advances in Intercellular Communication Mediated by Exosomal ncRNAs in Cardiovascular Disease. International Journal of Molecular Sciences. 2023; 24(22):16197. https://doi.org/10.3390/ijms242216197

Chicago/Turabian Style

Zhang, Xiaoyan, Shengjie Sun, Gang Ren, Wujun Liu, and Hong Chen. 2023. "Advances in Intercellular Communication Mediated by Exosomal ncRNAs in Cardiovascular Disease" International Journal of Molecular Sciences 24, no. 22: 16197. https://doi.org/10.3390/ijms242216197

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