Next Article in Journal
Optimization of Tissue Digestion Methods for Characterization of Photoaged Skin by Single Cell RNA Sequencing Reveals Preferential Enrichment of T Cell Subsets
Previous Article in Journal
Post-Anesthesia Cognitive Dysfunction in Mice Is Associated with an Age-Related Increase in Neuronal Intracellular [Ca2+]—Neuroprotective Effect of Reducing Intracellular [Ca2+]: In Vivo and In Vitro Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases

by
Sheila Caño-Carrillo
,
Juan Manuel Castillo-Casas
,
Diego Franco
and
Estefanía Lozano-Velasco
*
Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain
*
Author to whom correspondence should be addressed.
Cells 2024, 13(3), 265; https://doi.org/10.3390/cells13030265
Submission received: 29 December 2023 / Revised: 28 January 2024 / Accepted: 30 January 2024 / Published: 31 January 2024

Abstract

:
Effective intercellular communication is essential for cellular and tissue balance maintenance and response to challenges. Cellular communication methods involve direct cell contact or the release of biological molecules to cover short and long distances. However, a recent discovery in this communication network is the involvement of extracellular vesicles that host biological contents such as proteins, nucleic acids, and lipids, influencing neighboring cells. These extracellular vesicles are found in body fluids; thus, they are considered as potential disease biomarkers. Cardiovascular diseases are significant contributors to global morbidity and mortality, encompassing conditions such as ischemic heart disease, cardiomyopathies, electrical heart diseases, and heart failure. Recent studies reveal the release of extracellular vesicles by cardiovascular cells, influencing normal cardiac function and structure. However, under pathological conditions, extracellular vesicles composition changes, contributing to the development of cardiovascular diseases. Investigating the loading of molecular cargo in these extracellular vesicles is essential for understanding their role in disease development. This review consolidates the latest insights into the role of extracellular vesicles in diagnosis and prognosis of cardiovascular diseases, exploring the potential applications of extracellular vesicles in personalized therapies, shedding light on the evolving landscape of cardiovascular medicine.

1. Introduction

Intercellular communication is an essential activity for the maintenance of cellular and tissue homeostasis as well as responding to pathological processes. For this purpose, cells have different mechanisms depending on the distance of cell-to-cell communication requirements. Short cellular cross talk is mediated by direct cell-to-cell contact or secretion of soluble factors, whereas long range communication is driven through ligand–receptor interactions such as cytokine and/or hormone release [1,2]. However, in recent decades a novel mechanism of communication has emerged, where extracellular vesicles (EVs) are implicated [2]. EVs are constitutively produced by most of the cell types and consist in a lipidic bilayer membrane that encloses biological contents derived from the original cell, i.e., proteins, nucleic acids and lipids that can alter the biology of the distant cell [3,4]. The interest in EV biology has grown exponentially and many different types of EVs have been classified depending on their size, biogenesis and their biophysical properties [5,6] (Figure 1). There are two primary types of EVs, exosomes and ectosomes, whose classification is determined by their formation process. The genesis of these vesicles is contingent upon the assembly of local microdomains within endocytic membranes for exosomes and the plasma membrane for ectosomes, the latter subclassified into microvesicles and apoptotic bodies [7,8].
Exosomes are small EVs ranging from 30 to 100 nm. They are formed by inward budding of the early endosome membrane which subsequently matures into multivesicular bodies (MVBs) [9,10,11,12]. MVBs are involved in protein sorting, recycling, storage, transport and exosome release into the extracellular space [11,13,14,15,16]. Exosomes were originally considered as a cellular mechanism to eliminate cell debris; however, since few years ago, they are considered as principal mediators of cell-to-cell communication, cell and tissue maintenance and disease progression [17]. Moreover, exosomes can act as antigen-presenting vesicles participating in the stimulation of immune response [18,19]. Microvesicles (MVs) are EVs with a range from 200 nm to 1 µm which are directly shed as an outward budding of the plasma membrane [10,11,12,13,20,21]. Similarly to exosomes, MVs are involved in cell-to-cell communication between healthy and diseased cells [22,23]. Finally, apoptotic bodies are released into the extracellular space as blebs of cells undergoing apoptosis, and they have been reported as the bigger EVs, with a size range from 1 to 4 µm [13,24]. These apoptotic bodies are formed when the cytoskeleton is separated from the cellular plasma membrane due to increased hydrostatic pressure after the cell contracts [25]. The composition of the apoptotic bodies are organelles, chromatin and glycosylated proteins, in contrast to exosomes and MVs [13,26,27] (Figure 1).
As mentioned previously, most cell types release EVs of different sizes, composition and subcellular origin, and moreover, these EVs can be found in different body fluids such as plasma, saliva and urine [28,29,30]. Therefore, due to their presence in liquid biopsies, EVs are considered as potential disease biomarkers. Understanding the mechanisms by which molecular cargos are loaded into the EVs will provide the key to completely understanding the role of EVs in cellular communication during disease development and progression [31,32,33,34] (Figure 1).
Cardiovascular disease (CVD) is one of the leading causes of morbidity and mortality around the world [35]. CVDs cover a wide range of pathological conditions where the most common types are ischemic heart disease, structural cardiomyopathies, electrical heart diseases and finally heart failure (HF) [36]. There have been major steps forward in diagnosis and prognosis of CVDs that have improved patients’ survival and their quality of life, but there is still much work to do for CVD prevention and palliation, given the continued high mortality rate [37]. Several studies have recently indicated that small EVs can be released by cardiovascular cells, such as cardiomyocytes (CM), endothelial cells (EC), cardiac fibroblasts (CF), platelets, smooth muscle cells (SMCs), leucocytes, monocytes and macrophages [38]. These small EVs play important biological roles in the maintenance of normal cardiac structure and function, whereas small EVs are able to change their composition under pathological conditions, thus contributing to the development of CVDs [39,40,41].
In this review, we summarize the current state-of-the-art small EVs in the prognosis and diagnosis of CVDs, as well as the possible applications of small EVs in personalized therapies.

2. Small EVs in the Pathological Process of the Myocardial Infarction

Myocardial infarction (MI) is one of the most common CVDs, representing the main leading cause of death in the world [42]. Pathologically, MI is defined as the death of CMs due to a prolonged lack of oxygen in a specific area of the myocardium, initiating an apoptotic process leading to necrosis of the cardiac muscle and subsequently to other cardiac diseases such as arrhythmias or HF [43,44,45,46] (Figure 2A). The main etiology of MI is associated with the rupture of the atherosclerotic plate [47], although other causes, such as coronary artery embolism or coronary vasospasm have been also described [48]. After MI and the consequent death of CMs, autophagy and inflammatory processes begin as a strategy to remove the damaged tissue, allowing the replacement of the necrotic area with fibrotic tissue [46,49,50,51,52,53,54]. Furthermore, mechanisms of revascularization are also activated despite the low regenerative capacity of the heart after cardiac injury [55,56]. Currently, several studies show that exosomes play an important role in post-MI processes and participate in cellular communication regulating cardiac remodeling after MI [57,58,59]. For this reason, many authors focus their attention on the main mechanisms that regulate the production and content of these exosomes using transcriptomic and proteomic approaches, highlighting their detection as biomarkers after MI and thus their plausible therapeutic use [60,61,62]. The following paragraphs summarize the most important features of these exosomes in the MI context.

2.1. Small Extracellular Vesicle Transcriptomic Analyses in Myocardial Infarction

The discovery of new functions of exosomes has allowed the use of these small EVs as a potential tool to identify molecules that can be used as biomarkers after MI [63]. Currently, the main diagnostic biomarker for MI is cardiac troponin (T/I) [64], even though other more sensitive biomarkers have emerged, for example, non-coding RNAs, which allow for an earlier diagnosis [65,66]. Within non-coding RNAs, an increasing number of transcriptomic studies identified miRNAs in exosomes after MI [67,68,69]. Some analyses of circulating exosomes in the serum of MI patients have revealed a large number of miRNAs that are deregulated. For example, miR-203, miR-4516, or miR-183, which regulate the activity of several protein kinases in CMs, are upregulated, confirming their identification as MI biomarkers [67,70]. Another transcriptomic study identifies the deregulation of miRNA levels in exosomes on a large scale, identifying around 500 upregulated and downregulated miRNAs in MI exosomes, highlighting miR-6718 and miR-4329 [68]. Guo et al. (2021) reveal that, in addition to differential expression in exosomal miRNAs identifying up to 18 miRNAs as biomarkers, exosomes differ in size, being smaller in MI patients [69] (Table 1).
Some miRNAs have been widely studied due to their essential role in post-MI inflammatory, fibrotic and angiogenic processes, i.e., miR-126 or miR-155, which are upregulated in exosomes of MI patients, and miR-21 or miR-146a-5p, which are downregulated [71,72,73]. Specifically, miR-146a-5p is associated with inflammation after MI by regulating M1 macrophage polarization through the regulation of TNF Receptor-Associated Factor 6 (TRAF6) [74]. Regarding angiogenesis, another transcriptomic analysis of exosomes derived from MI patients revealed up to 40 differentially expressed miRNAs, highlighting the downregulation of miR-143. In vitro assays demonstrated that this downregulation promotes angiogenesis through the insulin-like growth factor 1 receptor and nitric oxide (IGF-IR/NO) signaling pathway [75]. In contrast, in vitro assays in CMs after ischemia/reperfusion (I/R) describe exosomes with high levels of miR-143 and miR-222, both enhancing angiogenesis and cardiac remodeling [76]. In addition, in vitro and in vivo assays in necrotic dendritic cells revealed the presence of eight upregulated miRNAs related to the regulation of angiogenesis, in which miR-494 is the most significant due its role in promoting revascularization after injury [77]. A similar result was demonstrated by Duan et al. (2022), where the analysis of exosomes extracted from the peripheral serum of patients with MI identified high levels of miR-126-3p, which promotes angiogenesis through regulation of the miR-126-3p/TSC1/mTORC1/HIF-1α signaling pathway [78] (Table 1).
The protective role of some exosomal miRNAs was also confirmed in ferroptosis after MI. Low levels of miR-26b-5p were described in exosomes derived from MI patients, which can reduce ferroptosis by positively regulating Solute Carrier Family 7 Member 11 (SLC7A11) [79]. Finally, other studies also associate miRNA transport in exosomes derived from MI patients with subsequent apoptosis [80]. Sun et al. (2022) revealed 52 differentially expressed miRNAs by transcriptomic analysis, highlighting the upregulation of miR-133a-3p, miR-151a-5p, miR-199b-5p, miR-374b-5p, miR-503-5p, and miR-708-5p. Concretely, they focused on miR-503-5p because of its ability to promote apoptosis through the regulation of Peroxisome Proliferator-activated Receptor Gamma Coactivator-1β (Ppargc-1β) and Sirtuin 3 (SIRT3) in CMs [80] (Table 1).
Although miRNAs are the most frequent non-coding RNAs studied in exosomes as MI biomarkers, long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) are also analyzed. For lncRNAs, sequencing profiles allowed us to identify 518 differentially expressed in post-MI exosomes, highlighting ENST00000556899.1 and ENST00000575985.1, which are upregulated, thus supporting their involvement in the regulation of post-MI processes [81]. More specifically, other lncRNAs like TUG1 or HCG15 have also been detected as being upregulated in MI exosomes, and in vitro and in vivo assays show that these lncRNAs are implicated in the inhibition of angiogenesis and cell viability through the regulation of HIF-1α/VEGF-α and NF-κβ/p65, respectively [82,83]. Despite being smaller in number, circular RNAs have been reported to have a significant advantage over linear RNAs because they exhibit better stability and can therefore be identified as biomarkers of MI for a longer period of time [84]. For this reason, some transcriptomic studies focused on the analysis of circRNAs in MI exosomes, highlighting, for example, the upregulation of circ_0020887 and circ_0009590 in the exosomes patients with ST segment elevation on electrocardiogram [85]. Another circRNA that also increases its expression in exosomes after MI is circITGB1. Within in vivo assays using a mouse model, these exosomes activate dendritic cells and exacerbate cardiac damage and inflammation through the miR-342-3p/NFAM1 pathway [86]. In vitro assays have also been reported in CMs subjected to hypoxia where high levels of circ_HIPK3 and circ_SLC8A1 are detected [87,88]. These circ_HIPK3-loaded exosomes target cardiac microvascular endothelial cells (CMVECs), protecting them from oxidative stress through the regulation of the miR-29a/VEGFA and miR-33a-5p/IRS1 pathways [89,90]. On the other hand, high levels of exosomal circSLC8A1 promote the inflammatory process and oxidative stress in other CMs, leading to their apoptosis through the regulation of the miR-214-5p/TEAD1 axis [88] (Table 1).
Table 1. Summary of ncRNAs associated with myocardial infarction (↑ upregulated and ↓ downregulated).
Table 1. Summary of ncRNAs associated with myocardial infarction (↑ upregulated and ↓ downregulated).
AssayDE ncRNAsMain ncRNAsSampleRef.
miRNAs
Liu et al. (2023)↑ miR-4516
↑ miR-203
miR-4516, miR-203Plasma
(MI patient)
[67]
Zhao et al. (2019)85 miRNAsmiR-183, miR-92, miR-4709, miR-550, miR-223Plasma
(MI patient)
[70]
Chen et al. (2021)↑ 544 miRNAs
↓ 518 miRNAs
miR-6718, miR-4329Serum
(MI patient)
[68]
Guo et al. (2021)↑ 138 miRNAs
↓ 208 miRNAs
miR-143-3p, miR-23b-5p, miR-106b-5p, miR-33a-5pPlasma
(MI patient)
[69]
Ling et al. (2020)↑ miR-126
↓ miR-21
miR-126
miR-21
Serum
(MI patient)
[71]
Geng et al. (2020)↑ 20 miRNAs
↓ 20 miRNAs
miR-143Serum
(MI patient)
[75]
Ribeiro-Rodrigues et al. (2017)↑ 3 miRNAs
↓ 4 miRNAs
miR-143, miR-222H9c2
primary CMs
[76]
Liu et al. (2020)↑ 8 miRNAsmiR-494-3pMI mouse model[77]
Sun et al. (2022)↑ 12 miRNAs
↓ 11 miRNAs
miR-503Peripheral blood
MI mouse model
[80]
LncRNAs
Zheng et al. (2020)↑ 245 lncRNAs
↓ 273 lncRNAs
ENST00000556899.1
ENST00000575985.1
Plasma
(MI patient)
[81]
Dang et al. (2023)↑ TUG1TUG1Plasma
(MI patient)
MI mouse model
[82]
Lin et al. (2021)↑ 29 lncRNAs
↓ 36 lncRNAs
HCG15Serum
(MI patient)
[83]
circRNAs
Wang et al. (2023)428 circRNAscirc_0020887, circ_0009590Plasma
(MI patient)
[85]
Zhu et al. (2022)↑ 10 circRNAs
↓ 10 circRNAs
circ_ITGB1, circ_SLC7A1, circ_ATG5, circ_POLR1APlasma
(MI patient)
[86]

2.2. Small Extracellular Vesicle Proteomic Analyses in Myocardial Infarction

Just as many transcriptomic studies have identified essential biomarkers for MI; there are also proteomic analyses that reveal the presence of distinct proteins in post-MI exosomes that can be used as biomarkers for MI diagnosis [91]. Xie et al. (2022) analyzed the protein profile within the exosomes of post-MI patients, identifying 72 differentially expressed proteins. Notably, three proteins exhibited elevated levels: Plasminogen (PLG), Complement Component C8 Beta (C8B) and Thrombin (F2) [91]. Furthermore, additional research emphasizes the presence of proteins within these exosomes that are involved in the inflammatory process and cardiac remodeling, such as Phosphatase and Tensin Homolog (PTEN) or Matrix Metalloproteinase-9 (MMP-9) [71,92]. In the post-fibrotic process after MI, some proteins present in exosomes have also been described, such as the transcriptional cofactor Limb Bud And Heart Development Protein Homolog (LBH). These exosomes produced by damaged CMs are taken up by CFs and activate Crystallin Alpha B (CRYAB), promoting further proliferation of CFs and their differentiation into myofibroblasts [59]. Post-MI exosomes have also been reported to carry proteins that promote cardiac repair and remodeling after damage, for example, Clusterin or Profilin 2 (PFN2) [93,94]. The levels of PFN2 are elevated in exosomes produced by ECs following MI. In both in vitro and in vivo assays, these exosomes demonstrate an increase in angiogenesis and cardiac improvement after damage through regulation of the PI3K/PNF2/ERK axis [94].
Finally, other studies highlight the importance of exosomes as biomarkers for MI diagnosis. This research conducted a proteomic study by comparing plasma from control and MI patients and analyzing the protein profile in post-MI exosomes. These results revealed 11 proteins that were deregulated in the exosomes compared to control plasma. However, three of these proteins, Chymotrypsin C (CTRC), Proto-oncogene Tyrosine-protein Kinase SRC (SRC), and C-C Motif Chemokine Ligand 17 (CCL17), did not exhibit downregulation when comparing their levels in serum between MI and control patients. Therefore, this analysis justified the need to also analyze post-MI exosomes to obtain additional diagnostic information that are not available only from plasma samples [95].

2.3. Mechanistic Insights into Small Extracellular Vesicle Related with Myocardial Infarction

As discussed in previous paragraphs, exosomes are involved in the regulation of the activation/inhibition of the main processes that occur after MI, such as inflammation, fibrosis, angiogenesis or apoptosis, as well as allowing communication between cells to coordinate these processes [57,58,59]. After MI, M1 macrophages are activated and increase the production of exosomes (M1-exos) with miR-155, which deliver this miRNA to CFs, thereby enhancing the inflammatory and fibrotic process [96]. M1-exos with high levels of miR-155 are also implicated in the inhibition of angiogenesis since these M1-exos can also be transferred to ECs and reduce angiogenesis via Sirt1/AMPKα2 and RAC1/PAK2 signaling after MI [97]. Macrophages are not the only cells that can release exosomes carrying miRNAs involved in post-MI process regulation. In vivo assays in a mouse MI model show that after MI, T CD4+ cells are activated and promote the synthesis of exosomes in which miR-142-3p is upregulated and promotes myofibroblast differentiation in ECs via miR-142-3p/APC/Wnt signaling [98]. Exosomes are involved in paracrine regulatory processes, as damaged CMs release exosomes after MI that will act on neighboring cells such as other CMs or ECs. This exosomal function is confirmed by the study of Gou et al. (2016), which showed that infarcted CMs generate exosomes that exhibit highly expressed miR-19a-3p, which are delivered to ECs and inhibit angiogenesis regulating Hypoxia-inducible factor 1-alpha (HIF-1α) [99]. Furthermore, post-MI CMs can generate exosomes after cardiac damage carrying miR-328-3p, which increase the apoptotic process through upregulation of Caspase 3 (Casp3) [100]. CFs also receive these exosomes produced by the CMs after MI, increasing their proliferation and the myofibroblast differentiation due to the high levels of miRNAs such us miR-208 or miR-92a [101,102,103]. Cell-to-cell communication via exosomes also takes place in the opposite direction, i.e., ECs can generate exosomes carrying miR-503 and promote CM apoptosis after MI [80]. Another example of miRNA that can be transferred between CMs after MI is miR-30a, whose levels are upregulated in exosomes extracted from the serum of MI patients. In vitro assays show that after hypoxia, these exosomes regulate the autophagy process between hypoxic CMs by upregulating genes such as Beclin-1 (BECN1), Autophagy Related 12 (Atg12), or Microtubule Associated Protein 1 Light Chain 3 Alpha (LC3I/II) [104]. Exosomes, carrying low levels of miR-342-3p as a consequence of MI, contribute to the regulation of both autophagy and apoptosis. Both in vitro and in vivo assays reveal that miR-342-3p regulates apoptosis and autophagy through the SRY-Box Transcription Factor 6 (SOX6) and Transcription Factor EB (TFEB), respectively [105]. A similar process, ferroptosis, also plays a relevant role in exosome regulation in post-MI processes. After MI, ferroptotic CMs release exosomes with low levels of miR-106b-3p, which promotes the activation of the Wnt pathway and increases the polarization of M1 macrophages, enhancing the inflammatory process after damage [106].
Other authors focused on the differential expression of proteins involved in the biogenesis, uptake or polarization of exosomes generated after MI. One example is the high level of CD44 in MI, which is involved in the synthesis of exosomes after MI through the positive regulation of Fibroblast Growth Factor Receptor 2 (FGFR2), as well as the subsequent uptake of these exosomes by ECs [107]. Regarding the ability of exosomes to migrate and the factors that trigger their polarization to the region of interest, it has been reported that dendritic cells increase the expression of C-C Motif Chemokine Receptor 7 (CCR7) after damage, and they generate exosomes containing both CCR7 and its ligands. These exosomes can target the spleen to activate CD4+ T cells, which produce anti-inflammatory cytokines that promote cardiac remodeling [108,109]. In addition, an external inflammatory stimulus, such as a decrease in the anti-inflammatory cytokine IL-10, can modify the protein content of exosomes. One protein that modifies its levels in exosomes is Integrin Linked Kinase (ILK), whose high levels promote the activation of NF-κβ, enhancing the inflammatory response and decreasing angiogenesis in the cells that receive these exosomes [110,111].

2.4. Therapeutic Approaches

Although many studies describe the potential use of exosomes as biomarkers for the diagnosis of MI, several studies additionally analyzed the use of exosomes as tools to transport specific molecules that promote cardiac repair after MI, thus becoming a widely used strategy in recent years [112,113,114,115]. The most frequent strategy is based on the use of exosomes derived from different mesenchymal stem cells (MSCs) that carry a specific molecule (RNA or protein) involved in the processes of inflammation, fibrosis or angiogenesis, among others [116,117,118].
Currently, there are numerous studies that use exosomes derived from different cell types to transport a specific non-coding RNA to the damaged area after MI (Table 2). One of the most widely used miRNAs as a therapeutic tool due to its role in inflammation and fibrosis is miR-21. Several studies extract exosomes derived from different cell types such as MSCs, cardiac telocytes (CTs), or even serum from control individuals loaded with miR-21, using these exosomes as a treatment for MI in both in vitro and in vivo assays [119,120,121]. Administration of these miR-21-loaded exosomes shows an improvement in cardiac function after MI due to the positive regulation of the angiogenic process and the inhibition of CM apoptosis and fibrosis through the regulation of PTEN and p53/Cdip1/Casp3 signaling pathways, among others [119,121,122,123] (Figure 2A). MSC-derived exosomes, both under control and hypoxic conditions and carrying high levels of miR-125b, have also been used as a therapy against CM apoptosis after MI [124,125]. One of the most commonly used types of MSCs to obtain exosomes, administered as a treatment for MI, are those derived from human umbilical cord mesenchymal stem cells (HUCMSCs) [126,127]. These cells have been used to obtain exosomes that act as a vehicle to transport miRNAs such as miR-23, miR-133 or miR-223 to the infarcted area, which promote cardiac repair by activating angiogenesis and reducing inflammation, ferroptosis or fibrosis [128,129,130] (Figure 2). Other studies use this strategy to produce exosomes derived from cardiosphere-derived cells (CDCs) or MSCs which carry miR-181, regulating macrophage polarization and reducing inflammation when they are administered after MI [131,132]. There is also evidence that the therapeutic use of MSC-derived exosomes carrying specific proteins such as Itchy E3 Ubiquitin Protein Ligase (ITCH), Fibronectin Type III Domain Containing 5 (FNDC5) or Stromal Cell-Derived Factor 1 (SDF1) promote cardiac repair after their administration [133,134,135] (Figure 2A) (Table 2).
Another treatment strategy post MI involves the modification of gene expression in the cells from which exosomes are subsequently extracted. This modification leads to changes in the content of these exosomes, offering a targeted approach for therapeutic interventions [136,137,138]. Several studies report that the overexpression of genes such as Hypoxia Inducible Factor 1 Subunit Alpha (HIF1-α) in MSCs results in a modification of the content of these exosomes, making them able to ameliorate cardiac damage after MI by promoting angiogenesis and reducing fibrosis, apoptosis or inflammation [139,140]. Overexpression of GATA-Binding Protein 4 (GATA4) in the cells from which exosomes are extracted has also been widely used to modify the exosome content for use as a treatment after MI [141,142,143]. He et al. (2018) revealed that overexpression of GATA4 in bone marrow mesenchymal stem cells (BMSCs) results in the production of exosomes that enhance myocyte precursor differentiation and reduce apoptosis after MI. This effect is due to a shift in the protein pattern carried by these exosomes, in which eight proteins associated with differentiation and six related to apoptosis have been identified with differential expression [141]. Finally, the reparative capacity of neonatal serum-derived exosomes has also been reported, identifying up to 28 ligands that can promote angiogenesis when these exosomes are administered after MI [144] (Table 2). Therefore, the understanding of molecular and structural aspects of exosomes has paved the development of a promising tool for MI treatment.
Table 2. Summary of ncRNAs for therapeutically approaches in myocardial infarction (↑ upregulated and ↓ downregulated).
Table 2. Summary of ncRNAs for therapeutically approaches in myocardial infarction (↑ upregulated and ↓ downregulated).
miRNAs
Source of ExosomesIn Vitro
Essay
In Vivo ModelmiRNASignalingBiological EffectsRefs.
MSCs
BMSCs
H9c2
HUVECs
MI (LAD)miR-210AIFM3/PI3K/AKT
AIFM3/p53
Ephrin, Casp8
↑ CMs viability
↓ Fibrosis, apoptosis
[145,146,147,148]
ADSCs-MI (LAD)miR-205-↑ Angiogenesis
↓ CM apoptosis, cardiac fibrosis
[149]
MSCs
BMSCs
NMCMsMI (LAD)miR-125-5p
miR-125b-5p
p53/BAK1
p53/Bnip3
↑ Cardiac function
↓ CM apoptosis,
autophagy
[124,125]
CTs
MSCs
plasma
BMSCs
CMECs H9c2
HUVECs MRCMs
MI (LAD)miR-21-5p
miR-21
miR-21a-5p
p53/Cdip1/Casp3
BTG2
PDCD4
PTEN
↑ Angiogenesis, CM proliferation
↓ Apoptosis,
fibrosis
[119,120,122]
MSCsCMsMI (LAD)miR-25-3pFASL/PTEN
EZH2/H3K27me3
↓ Apoptosis[150]
EPDCsH9c2
CMs
MI (LAD)miR-27a
miR-100
miR-30a
miR-30c
-↑ Proliferation[151]
MSCsECs
H9c2
-miR-153-3pAngpt1/Vegf/
Vegfr2/PI3k/Akt/
eNOs
↑ Angiogenesis
↓ Apoptosis
[152]
ADSCsHMVECMI (LAD)miR-31FIH1/HIF-1α↑ Angiogenesis[153]
CDCs
HUCMSCs
Macrophages
PBMCs
MI (LAD)miR-181b
miR-181a
PKCδ
c-Fos
↑ Macrophague polarization, cardiac protection
↓ Inflammation
[131,132]
BMSCsH9c2MI (LAD)miR-143-3pCHK2/Beclin2↓ Apoptosis,
autophagy
[154]
ADSCsH9c2MI (LAD)miR-93-5pAtg7/TLR4↑ Cardiac protection
↓ Inflammation
[155]
Serum
ADSCs
HUVECs
H9c2
MI (LAD)miR-126-3p
miR-126
TSC1/mTORC1/
HIF-1α
↑ Angiogenesis
↓ Inflammation,
fibrosis
[78,156]
BMSCsH9c2MI (LAD)miR-338MAP3K2/JNK↑ Cardiac function
↓ Apoptosis
[157]
MSCsHUVECsMI (LAD)miR-132RASA1↑ Angiogenesis[126]
BMSCs-MImiR-29b-3pADAMTS16↑ Angiogenesis
↓ Fibrosis
[158]
ADSCsCMsMI (LAD)miR-671TGFBR2/Smad2↓ Fibrosis, inflammation, apoptosis[159]
BMSCs
HUCMSCs
HUVECs
H9c2MI (LAD)miR-24
miR-24-3p
Plcb3/NF-κβ
CCR2
↑ M2 macrophague polarization
↓ CM apoptosis
[160,161]
M2 macrophage-MI (LAD)miR-1271-5pSOX6↓ Apoptosis[162]
BMSCsH9c2MI (LAD)miR-30eLOX1/NF-κβ-p65/Casp9↓ Apoptosis,
fibrosis
[163]
EPCsCFsMI (LCA)miR-1246
miR-1290
EFL5/CD31/
VEGFR2/α-SMA
SP1/CD31/VEGFR2/
α-SMA
↑ Angiogenesis
↓ Fibrosis
[164]
HUCMSCsCMs
HUVECs
MI (LAD)miR-214-3pPTEN/AKT↑ Angiogenesis
↓ Apoptosis
[165]
ADSCs
CMs
H9c2
M1 macrophages
MI (LAD)miR-146-aEGR1/TLR4/
NF-κβ
TRAF6
↓ Apoptosis,
inflammation,
fibrosis
[74,166]
BMSCs
HUCMSCs
MSCs
HL-1
H9c2
NRCMs
MI (LAD)miR-19a/b
miR-19
SOX6/AKT
JNK3/Casp3
PTEN/AKT
↓ Fibrosis,
CM apoptosis
[167,168]
MSCs-MI (LAD)miR-590-3pHoxp, Homer1,Cdk1,Cdk8↑ CM
proliferation
[169]
MSCsCMECsMI (LAD)miR-543Col4a1↑ Proliferation,
angiogenesis
[170]
CPCsHUVECsMI (LAD)miR-322NOX2↑ Angiogenesis[171]
BMSCs-MI (LAD)miR-301-↓ Autophagy[172]
BMSCsCMsMI (LAD)miR-183-5pFOXO1↓ Apoptosis,
oxidative stress
[173]
BMSCs
HUCMSCs
NRCMs
HUVECs
MI (LAD)miR-133
miR-133a-3p
Sanil1
AKT
↑ Angiogenesis
↓ Inflammation,
fibrosis, apoptosis
[130,174]
PlasmaH9c2MI (LAD)miR-342-3pSOX6
TFEB
↓ Apoptosis,
autophagy
[105]
EPCs-MI (LCA)miR-218-5p
miR-363-3p
p53/JMY↓ Fibrosis[175]
BMSCsH9c2MI (LAD)miR-455-3pMEKK1/MEKK4/
JNK
↓ Apoptosis[176]
BMSCs
MSCs
NMVM
HUVECs
RAW264.7
MI (LCA, LAD)miR-182-5p
miR-182
TLR4/NF-κβ
GSDMD
↑ M1 → M2
polarization
↓ Inflammation,
pyroptosis
[177,178,179]
PlasmaHUVECs
HEK293Ts
Carotid artery injurymiR-193a-5pACVR1↓ Oxidative stress[180]
BMSCs
cCFU-Fs
H9c2
HUVECs
MI (LAD)miR-221-3pPTEN/AKT↑ Angiogenesis
↓ Fibrosis,
apotosis
[142,181]
HUCMSCs
hiPSC-Ecs
Ac16
hiPSC-CMs
MI (LAD)miR-100-5pFOXO3/NLRP3
PP-1β/SERCA-2a
↑ Ca+2 homeostasis
↓ Inflammation,
pyroptosis
[182,183]
BMSCsCMsMI (LAD)miR-22Mecp2↓ Apoptosis,
fibrosis
[184]
BMSCsNRCFsMI (LAD)miR-212-5pNLRC5/VEGF/TGF-β/SMAD↓ Fibrosis[185]
HUCMSCs-MI (LAD)miR-200b-3pBCL2L11/NLRP1↓ Apoptosis,
inflammation
[186]
HUCMSCsHUVECsMI (LAD)miR-423-5PEFNA3↑ Angiogenesis,
migration
↓ Fibrosis
[187]
HUCMSCsHUVECs
H9c2
MI (LCA)miR-223p53/S100A9↑ Angiogenesis
↓ Fibrosis
[128]
M2 macrophageHL-1MI (LAD)miR-378a-3pELAV1/NLRP3/
Caspase1/
GSDMD
↓ Pyroptosis[188]
MSCsNMCMsMI (LAD)miR-150-5pTXNIP↓ Apoptosis[189]
PlasmaH9c2
HEK293T
MI (LAD)miR-130a-3pATG16L1↓ Inflammation,
autophagy
[190]
ADSCsNMCMsMI (LCA)miR-224-5pTXNIP↓ Apoptosis,
necrosis
[191]
iPSC derived CMsiCMsMI (LAD)miR-106a-363 (cluster)Notch3↑ Proliferation[192]
MSCsNRCMs
NRCFs
MI (LAD)miR-4732-↑ Angiogenesis
↓ Fibrosis,
apotosis
[193]
HUCMSCsCMsMI (LAD)miR-23a-3pDMT1↓ Ferroptosis[129]
mESs
MEFs
H9c2
HUVECs
MI (LAD)miR-294-↑ Angiogenesis, proliferation[194]
lncRNAs
Source of ExosomesIn Vitro
Essay
In Vivo ModellncRNASignalingBiological EffectsRefs.
PlasmaHUVECs
HMVECs
MI (LAD)KLF3-AS1miR-138-5p↓ Apoptosis,
pyroptosis
[195]
Cardiac myocites
hPSC-CVPC
NRCMs
HUVECs
MI (LAD)MALAT1miR-92a/KLF2
miR-497
↑ Angiogenesis[196,197]
ECsCMsMI (LAD)LINC00174SRSF1/p53↓ Apoptosis,
autophagy
[198]
CMsCFsMI (LAD)AK139128-↓ Fibrosis[199]
MSCsH9c2MI (LADUCA1miR-873/XIAP↓ Apoptosis[200]
MSCs-MITARIDTcf21/Smad3/TGF-β↓ Fibrosis[201]
circRNAs
Source of ExosomesIn Vitro
Essay
In Vivo ModellncRNASignalingBiological EffectsRefs.
BMSCs MI (LAD)circ_002113miR-188-3p/RUNX1↓ Apoptosis[202]
HUCMSCsH9c2MI (LAD)circ_0001273-↓ Apoptosis[203]
CMs
Ac16
ECs
Ac16
MI (LAD)circ_HIPK3miR-29a/VEGFA
miR-33a-5p/IRS1
↑ Angiogenesis
↓ Apoptosis
[89,90]
ADSCsHL-1-circ_0001747miR-199b/MCL1↑ Proliferation,
viability
↓ Inflammation,
apoptosis
[204]
ECsCMsMI (LAD)circWhsc1Trim59/Stat3/Cyclin B2↑ Proliferation
↓ Fibrosis
[205]
PlasmaVSMCs
CMECs
MI (LAD)circCEBPZOSmiR-1178-3p/PDPK1↑ Angiogenesis[206]

3. Small EVs in the Pathological Process of Cardiomyopathies

Cardiomyopathies are defined as “a myocardial disorders in which the heart muscle is structurally and functionally abnormal, in the absence of coronary artery disease, hypertension, valvular disease, and congenital heart disease, however, sufficient to cause the observed myocardial abnormality” [207]. Cardiomyopathies can be classified into primaries or secondaries. Primary cardiomyopathies are mostly idiopathic, leading to heart failure and sudden death, while secondary cardiomyopathies develop in response to several extrinsic factors such as hypertension, metabolic disorders, drug-induced myopathy, ischemic heart disease and coronary artery disease [208,209,210].

3.1. Hypertrophic Cardiomyopathy

Hypertrophic cardiomyopathy (HCM) is one of the most common cardiac genetic conditions, with a prevalence greater than 1 in 500 in the general adult population [211]. This inherited disorder is characterized by left ventricular hypertrophy (>15 mm for adults), that cannot be only attributed to abnormal load conditions [212] (Figure 2D). Essential histopathological features include myocyte hypertrophy and disarray alongside heightened myocardial fibrosis; the combination of these hallmarks leads to left ventricular outflow track obstruction, impaired diastolic function and cardiac arrhythmias [213]. Genetically, HCM is an autosomal-dominant disorder caused by mutations in genes encoding for contractile and structural proteins of the cardiac muscle sarcomere apparatus [214]. Genetic analysis has improved our knowledge about the molecular bases of HCM, enabling clinicians to make an early identification prior to the onset of cardiac disease.

3.1.1. Small Extracellular Vesicle Transcriptomic Analyses in Hypertrophic Cardiomyopathy

In this pathological scenario, James et al. (2021) [215] performed a transcriptomic analysis on small EVs derived from human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with or without the c.ACTC1G301A mutation. This model of HCM [216] was chosen for this study as it had previously been shown to recapitulate many key disease phenotypes including abnormal contractility, Ca2+ sensitivity/handling, arrhythmogenesis and hypertrophic brain natriuretic peptide signaling. Transcriptomic analysis of HCM small EVs has shown that CMs alter their EV cargo when HCM sarcomeric mutations are present. To be more precise, they observed differences in snoRNA cargo within HCM-released small EVs that specifically is altered when HCM hiPSC-CMs were subjected to an increased workload. In total, 12 snoRNAs were identified including 10 SNORDs (SNORD6, SNOTRD116-23, SNORD116-25, SNORD116-29, SNORD18A, SNORD42A, SNORD43, SNORD58C, SNORD60, and SNORD 101) and 2 SNORAs (SNORA3B and SNORA20). The functional role of these snoRNAs is related with post-translational modifications and alternative splicing processes differentially regulated in HCM (Table 3).

3.1.2. Mechanistic Insights into Small Extracellular Vesicle Related with Hypertrophic Cardiomyopathy

Some years ago, Tian et al. (2018) demonstrated an upregulation of miRNA-27a levels in both infarcted myocardial tissue and systemic circulation in a rodent model of MI. The identified miRNA-27a exhibited a propensity for incorporation into small EVs, contributing to oxidative stress and promoting hypertrophic gene expression via modulation of the Nuclear factor (erythroid-derived 2)-like 2/Kelch-like ECH-associated protein 1 (Nrf2/keap1) signaling pathway [217]. Moreover, clinical studies corroborated increased miRNA-27a levels in failing hearts and systemic circulation, suggesting its potential utility as a diagnostic and prognostic biomarker for HF [218,219,220]. Furthermore, the same lab evidenced that miR-27a* exhibited resistance to degradation and mirrored the expression pattern of miRNA-27a in chronic HF. This miR-27a* is packaged into small EVs and taken up by CM-targeting Z-line-associated protein PDLIM5, thereby contributing to hypertrophic gene expression [221].

3.1.3. Therapeutic Approaches

A recent study has evidenced that YF1, a derived non-coding RNA from cardiosphere-derived cell exosomes alleviates cardiomyocyte hypertrophy, inflammation, and fibrosis associated with HCM in transgenic mice harboring a clinically relevant mutation in cardiac troponin I (cTnIGly146) [222] (Figure 2D).
Table 3. Summary of ncRNAs associated with cardiomyopathies (↑ upregulated and ↓ downregulated).
Table 3. Summary of ncRNAs associated with cardiomyopathies (↑ upregulated and ↓ downregulated).
AssayDE ncRNAsMain ncRNAsSampleRef.
Hypertrophic cardiomyopathy >> snoRNAs
James et al. (2021)12 snoRNASNORD6, SNOTRD116-23, SNORD116-25, SNORD116-29, SNORD18A, SNORD42A, SNORD43, SNORD58C, SNORD60, SNORD 101, SNORA3B and SNORA20)hiPSC-CMs[215]
Dilated cardiomyopathy >> miRNAs
Zhang et al. (2023)↑ 48 miRNAs
↓ 44 miRNAs
miR-423-5p, hsa-miR-185-5p, hsa-miR-150-5p, hsa-miR-10a-5p_R-1, hsa-miR-1304-3p_1ss13CA, sa-miR-3138_L-5R+2Plasma
(DCM patient)
[223]

3.2. Dilated Cardiomyopathy

Dilated cardiomyopathy (DCM), with an incidence of 1 in 2500 individuals, is defined as the presence of left ventricular dilation along with systolic dysfunction [208,224]. Moreover, DCM is frequently associated with an increased likelihood of severe arrhythmias, which suggests the pathological affection of the cardiac conducting system. Finally, with disease progression, the right ventricle and diastolic function are affected, leading to HF and death [225] (Figure 2E). Some genes are associated with the initiation, progression and pathology of DCM; nonetheless, while these genes seem to be linked to DCM, only a limited number directly contribute to the onset of DCM owing to genetic variations [226,227].

3.2.1. Small Extracellular Vesicle Transcriptomic Analyses in Dilated Cardiomyopathy

Zhang et al. (2023) [223] performed high-throughput sequencing in plasma exosomes of DCM patients with chronic heart failure (CHF) and healthy controls, and a total of 3687 miRNAs were detected in these biological samples. However, only 92 miRNAs were significantly differentially expressed between the two groups; 48 miRNAs were upregulated and 44 miRNAs were downregulated [223]. Six of these miRNAs have been identified as significant contributors to the development of DCM through diverse mechanisms, such as the regulation of fibrosis (miR-423-5p, hsa-miR-185-5p, hsa-miR-150-5p, hsa-miR-10a-5p_R-1) [228,229,230,231,232,233], hypertrophy (hsa-miR-150-5p) [231,232], inflammation (hsa-miR-1304-3p_1ss13CA, hsa-miR-150-5p) [231,232,234], oxidative stress (hsa-miR-1304-3p_1ss13CA) [234], angiogenesis (hsa-miR-150-5p) [235] and mitochondrial function (sa-miR-3138_L-5R+2) [236] (Table 3).

3.2.2. Small Extracellular Vesicle Proteomic Analyses in Dilated Cardiomyopathy

Bayes-Genis laboratory explored the proteomic signature of plasma-derived small EVs obtained from DCM patients and healthy controls. A total of 176 proteins (74.6%) were shared by controls and DCM patients, whereas 51 proteins were exclusive for the DCM group and 7 proteins were exclusive for the control group [237]. They observed that some proteins were generally over-represented in the cargo proteome of circulating DCM small EVs compared with control small EVs. These included fibrinogen, crucially associated with a high risk of cardiovascular disease due to its contribution to endothelial injury, plasma viscosity and thrombus formation [237,238,239,240,241,242]; serotransferrin, related with anemia as comorbidity in HF patients [237,243,244,245,246,247]; protease inhibitor α-1-antitrypsin (AAT) as a putative biomarker for the evaluation of disease status [248,249]; and several apolipoproteins [237]. Gene ontology analysis evidenced that proteins associated with stress as well as with protein activation were found to be more abundant in DCM small EVs when compared to control samples [237].

3.2.3. Mechanistic Insights into Small Extracellular Vesicle Related with Dilated Cardiomyopathy

Wu et al. (2018) [250] analyzed three different serum exosomal miRNAs, exo-miR-92b-5p, exo-miR-192-5p and exo-miR-320a, in patients with DCM and acute HF (AHF) vs. healthy volunteers. In the study, exo-miR-92b-5p was increased in DCM-AHF patients compared to control, and was finally considered as a potential biomarker that potentially predicts DCM-AHF in patients [250]. Recent research with angiotensin II-stimulated hiPSCs differentiated cardiomyocytes has evidenced that miR-218-5p is upregulated in the DCM-Exos. This microRNA has been identified as a critical contributor to fibrogenesis through the activation of Tgf-β signaling after the suppression of TNFAIP3 [251].

3.2.4. Therapeutic Approaches

Several labs have reported the therapeutic role of small EVs in DCM. Vandergriff et al. (2015) [252] analyzed the therapeutic role of cardiac stem cell-derived exosomes (CSC-exo) in a mouse model of doxorubicin-induced DCM. Systemic delivery of human CSC-exo in mice showed improved heart function via echocardiography, as well as decreased apoptosis and fibrosis [252]. Sun et al. (2018) [253] proved that mesenchymal stem cell-derived exosomes (MSC-Exos) alleviate inflammatory cardiomyopathy by improving the inflammatory microenvironment of the myocardium, especially by regulating the activity of macrophages in a mouse model of DCM [253]. Ni et al. (2020) [254] evidenced that trophoblast stem cell-derived exosomes (TSC-exos) could alleviate DOX-induced cardiac injury via the let-7i/YAP pathway. They observed an improvement of cardiac function and decreased inflammatory responses, accompanied by downregulated YAP signaling [254] (Figure 2E). Zhang et al. (2022) [255] evidenced that small EVs derived from KLF2-overexpressing endothelial cells reduced cardiac inflammation and ameliorated left ventricular dysfunction in DCM mice by targeting the CCR2 protein to inhibit Ly6Chigh monocyte mobilization from the bone marrow [255].

3.3. Diabetic Cardiomyopathy

The prevalence of diabetes mellitus (DM) is approximately 9.3% of the world population [256]. In this vast group, HF has emerged as the most common cardiovascular complication of diabetes [257]. Diabetic cardiomyopathy (DmCM) is a myocardial-specific complication that is associated with coronary microvascular dysfunction and increases the risk of HF in patients with diabetes [258] (Figure 2F). DmCM is characterized by left ventricle dysfunction, CM apoptosis and interstitial fibrosis developed in the absence of coronary artery disease, valvular disease and/or hypertension [259,260].

3.3.1. Mechanistic Insights into Small Extracellular Vesicle Related with Diabetic Cardiomyopathy

Some years ago, Gonzalo-Calvo et al. (2017) [261] evidenced that circulating miR-1 and miR-133a levels are actively released from CM exosomes in response to lipid overload and are robustly associated with myocardial steatosis in type 2 diabetes patients [261]. Moreover, microRNAs which are encapsulated within exosomes offer a stable source of information to study the role of miRs associated with HF in diabetic hearts with preserved ejection fraction (HFpEF). Huang et al. (2022) [262] evidenced the association of exosomal miR-30d-5p and miR-126a-5p with diabetic HFpEF [262]. It has been shown that circulating miR-30d downregulation reduces the cardioprotective role of miR-30d in HF [263,264,265], whereas miR-126a downregulation decreases cardiac microvessel density and impairs ventricular function [266].
In addition, recent research evidenced that exosomes deliver Mst1 protein between cardiac microvascular endothelial cells (CMECs) and CM, playing a pivotal role in the development of DmCM. In this scenario, the increase in Mst1 in CM inhibits cell autophagy, enhancing the apoptotic CM ratio and, moreover, affecting glucose metabolism which leads to insulin resistance that finally contributes to DmCM and impaired cardiac function [267]. The same lab has recently demonstrated that exosomes mediate the interaction between CMECs and CFs. They observed that exosomes derived from CMECs under high glucose were rich in TGF-β1 mRNA, which significantly promoted the activation of CFs. This condition aggravates perivascular and interstitial fibrosis in mice with DmCM [268].

3.3.2. Therapeutic Approaches

Heat shock protein (Hsp) response is a cellular intrinsic defense mechanism [269]. The expression of these proteins in type 1 and type 2 diabetes are decreased, contributing to diabetes-induced organ damage [270]. In this scenario, it has been evidenced that CM exosomes derived from a transgenic mouse model with cardiac-specific overexpression of Hsp20 protected against in vitro hyperglycemia-triggered cell death, as well as in vivo STZ-induced cardiac adverse remodeling. Thus, Hsp20-engineered exosomes might be a novel therapeutic agent for DmCM (Figure 2D). Moreover, Lin et al. (2019) [271] evidenced that mesenchymal stem cell (MSC)-derived exosomes significantly increased the levels of fatty acid transporters (FATPs) and fatty acid beta oxidase (FA-β-oxidase), whereas TGF-β1 and Smad2 mRNAs levels were significantly reduced. Such molecular regulation indicates that MSC-derived exosomes improve DM-induced myocardial injury and fibrosis via inhibition of the TGF-β1/Smad2 signaling pathway [271]. Similarly, parasympathetic ganglionic neuron-derived exosomes (PGN-exos) are able to inhibit apoptosis, improve cell viability and restore levels of anti-apoptotic protein Bcl-2 in diabetes-induced H9c2 cells [272]. Finally, recent data support the notion that ginsenoside RG1 (RG1)-induced MSCs secrete exosomes that can alleviate DmCM. Mechanistically, exosomes derived from RG1-induced MSCs transferred circNOTCH1 into macrophages, activating the NOTCH signaling pathway through the regulatory axis consisting of circNOTCH1, miR-495-3p and NOTCH1 [273] (Figure 2F). All these findings may contribute to the development of new therapeutic approaches for DmCM.

4. Small EVs in the Pathological Process of Atrial Fibrillation

Atrial fibrillation (AF) is the most common electrical disorder in humans. With a 2% prevalence in the general population, the incidence of AF raises to almost 10% in the elderly (+80 y). AF substantially contributes to morbidity and mortality by significantly altering the quality of life and, moreover, increasing the risk of embolic stroke and HF [274,275]. The risk of developing AF is enhanced by distinct cardiac and medical conditions, such as hypertension, cardiomyopathy, valvular dysfunction or obstructive sleep apnea [276,277].
AF is characterized by an irregular electrical pattern in the atrial chambers, lacking the P wave in the ECG (Figure 2C). Clinically, AF can be classified according its temporal duration in three distinct types; paroxysmal, persistent and permanent AF [276,277]. AF is generally considered as paroxysmal AF when the fibrillatory episodes self-terminate within seven days. Paroxysmal AF may progress to persistent and finally chronic or permanent states that fail to self-terminate. In the last decade, an increasing number of studies have reported the contribution of extracellular vesicles to the pathophysiology of AF. In the following paragraphs, we will summarize the current state-of-the-art information regarding the transcriptomic and proteomic EV analyses in different AF conditions, as well as an array of studies in which the role of discrete molecules is studied. Finally, the first insights of a therapeutic approach to revert AF conditions using EV therapy are envisioned.

4.1. Small Extracellular Vesicle Transcriptomic Analyses in Atrial Fibrillation

Several studies have investigated the concentration of small EVs in AF, providing evidence of an increased number of small EVs in AF vs. sinus rhythm controls [278,279]. Gene expression analyses have also been performed, most of them focusing on microRNA differential expression [280,281,282,283,284,285,286], while others demonstrated differentially lncRNA [287,288], circRNA [289] and protein [290,291,292] loading. Four different studies analyzed the differential expression of microRNAs in AF vs. non-AF patients. Siwaponanan et al. (2022) [280] revealed that nineteen microRNAs were significantly higher in AF vs. non-AF, and six were subsequently validated (miR-106b-3p, miR-590-5p, miR-339-3p, miR-378-3p, miR-328-3p and miR-532-3p). Zhu et al. (2022) [283] screened by high-throughput sequencing analysis and subsequently verified by qRT-PCR the differential expression of exosomal miRNAs, identifying that miR-124-3p, miR-378d, miR-2110 and miR-3180-3p were remarkably upregulated, while miR-223-5p, miR-574-3p, miR-125a-3p and miR-1299 were downregulated. Similarly, Wei et al. (2020) [284] searched for differences in the exosomal miRNAs between AF and normal sinus rhythm (SR) patients by combining high-throughput sequencing results and real-time PCR. A total of 20 microRNAs were initially identified, among which miR-92b-3p, miR-1306-5p and miR-let-7b-3p were differentially validated. While most of these authors highlight that these microRNAs can constitute promising biomarkers to assess AF in patients, there are limited mechanistic insights into the functional role of these microRNAs in AF pathophysiology, as only miR-124-3p has been reported to modulate the Wnt/β-catenin signaling pathway via AXIN1 in CFs [283] (Table 4).
Three additional studies have investigated the exosomal microRNA differential loading in patients in distinct phases of AF, i.e., paroxysmal, persistent and permanent AF. Mun et al. (2019) [285] analyzed exosomes from the serum of patients’ supraventricular tachycardia (SVT) as the controls, and paroxysmal AF and persistent AF patients through microRNA microarray analysis. Forty-five miRNAs were significantly higher in patients with persistent AF, but not in patients with paroxysmal AF as compared to control. Similarly, Wang et al. (2019) [282] identified 39 differentially expressed exosomal microRNAs from plasma of persistent AF and SR patient. Four of them were subsequently validated, i.e., miR-483-5p, miR-142-5p, miR-223-3p and miR-223-5p, and multivariable logistic analyses demonstrated that one of them, i.e., miR-483-5p, was independently correlated with AF. Finally, Hao et al. (2022) [281] also analyzed the differential expression of microRNAs in isolated exosomes from atrial myocytes and patient serum, and particularly focused on the contribution of miR-210 in AF, directly targeting GPD1L, thus regulating atrial fibrosis via the PI3K/AKT signaling pathway (Table 4). Interestingly, there are scarce microRNA similarities between the distinct studies, suggesting a lack of reproducibility. Such low reproducibility might be attributed to other parameters than AF per se, such as variations in sample collection (serum vs. plasma, systemic vs. intracardiac blood sampling) as well as differences in the microRNA platforms employed for microRNA differential discovery. It is important to note that such variations should be minimized during the validation steps, particularly when using RT-qPCR. Thus, protocol standardization would be desirable to further ascertain the global applicability of these microRNAs as AF biomarkers.
In addition, the exosomal microRNA cargo has also been investigated in AF-associated cardiovascular diseases, such as stroke. Xie et al. (2023) [286] conducted an analysis of serum exosomes from healthy individuals with SR, AF and AF-ischemic stroke patients by deep sequencing. They identified that miR-641 and miR-30e-5p were significantly upregulated in AF-ischemic stroke patients. These authors suggest that these microRNAs can be considered biomarkers of ischemic stroke in AF patients. However, no mechanistic insights are provided in regard to how these microRNAs are related to this CVD (Table 4).
Different studies have provided evidence that the deposition of epicardial adipose tissue (EAT) surrounding the heart is intimately linked with the onset of AF [293,294]. Furthermore, several studies have demonstrated that EAT can release signaling molecules that may contribute to the initiation of such electrophysiological disorders [295,296]. Therefore, different studies have investigated the EAT-exosomal cargo in AF vs. non-AF patients. These studies have placed particular emphasis on the differential expression of microRNAs [297], lncRNAs [287,288] and circRNAs [289]. While gene ontology analyses of the differentially expressed non-coding RNAs revealed functional categories, such as metabolism and stress response, which might contribute to the pathogenesis of AF, no further insights are reported about the mechanistic link between these non-coding RNAs and AF pathophysiology.

4.2. Small Extracellular Vesicle Proteomic Analyses in Atrial Fibrillation

To date, only three studies have investigated the protein content of small EVs in an AF context [290,291,292]. Ni et al. (2021) [292] identified differentially expressed exosomal proteins in AF and non-AF patients, highlighting that bioinformatic analyses revealed enrichment in proteins involved in anticoagulation, complement system and protein folding. Shaihov-Teper et al. (2021) [291] studied the role of EAT-derived small EVs in the pathogenesis of AF. By generating culture explants from patients with AF, these authors uncovered more secreted small EVs with greater amounts of proinflammatory and profibrotic cytokines in AF patients compared to those without AF. Mechanistically, they demonstrated that while EAT-derived small EVs from patients with and without AF shortened the action potential duration of induced pluripotent stem cell-derived CMs, only those from AF patients induced sustained reentry. Additionally, Weiss et al. (2021) [290] explored the differences in circulating small EV proteomic profiles in rivaroxaban-treated non-valvular AF patients as compared with matched warfarin controls. These authors demonstrated that circulating small EV profiles were fundamentally altered, with a decrease in highly pro-inflammatory protein expression and complement factors, along with increased expression of negative regulators of inflammatory pathways. Therefore, these data suggest the notion that differential protein cargo in small EVs is also relevant in the AF setting.

4.3. Mechanistic Insights into Small Extracellular Vesicle Related with Atrial Fibrillation

Different molecular cascades have been implicated as triggering factors of AF. In this context, the contribution of small EVs to AF has been reported to play a fundamental role in inducing fibrosis [298,299,300,301,302], inflammation [302,303,304,305], oxidative stress [302] and apoptosis [306]. Within the context of AF-atrial fibrosis, both microRNAs and lncRNAs have been identified. Exosomes containing miR-23a-3p, derived from CFs, can target SCL7A11, contributing to AF pathophysiology by increasing ferroptosis [299]. Additionally, exosomes containing lncRNA LINC00636 modulate miR-450a-2-3p and, thus, MAPK1, leading to improved cardiac fibrosis in AF [298]. Xu et al. (2022) [300] reported that exosomes, derived from MSCs, overexpressing Nrf2, inhibited cardiac fibrosis in AF, while Xu et al. (2021) implicated miR-324-3p-regulating Tgf-ß1 and, consequently, fibroblast proliferation in AF. In the context of inflammation, the lncRNA LRON was found to promote M2 macrophage polarization, from atrial myocytes, by delivering miR-23a, thus decreasing atrial fibrosis [304], while the exosomal-loaded lncRNA PVT1 regulates M1 macrophage polarization from angiotensin II-treated CMs [303]. Curiously, rapid atrial pacing modulates Kca3.1 function, leading to the promotion of proinflammatory exosome secretion with the activation of the Akt/RAb27a signaling pathways [305]. Importantly, Chen et al. (2021) [302] reported that EV containing MIAT can influence multiple triggering signals leading to AF, such as atrial fibrosis, inflammation and oxidative stress. This process is modulated by CXCL10 regulation via miR-485-5p. Finally, in the context of AF, CM apoptosis can also be modulated by the delivery of miR-148a exosomes derived from BMSCs, modulating SMOC2 expression [306].
Besides the biological processes involved in AF, additional evidence is emerging. For instance, Yan et al. (2021) [307] reported that lncRNA XIST, shuttled by adipose tissue-derived mesenchymal stem cell-derived small EVs, suppresses myocardial pyroptosis in AF by disrupting miR-214-3p-mediated Arl2 inhibition. In addition, Li et al. (2020) [308] revealed that myofibroblast-derived exosomes containing miR-21-3p were capable of modulating Cav1.2, thus contributing to electrical remodeling in AF. In sum, evidence is emerging on the functional role of discrete EV-mediated non-coding RNAs contributing to the pathophysiology of AF.

4.4. Therapeutic Approaches

The importance of small EVs in cell–cell communication in homeostasis and diseases is undoubtedly emerging, but most importantly, their therapeutic application is also emerging. Seminal work by Parent et al. (2023) [309] reported a randomized controlled trial in which the prevention of AF, after open-chest surgery, was evaluated, yielding promising results. In this context, injection of small EVs at the time of open-chest surgery shows prominent anti-inflammatory effects and effectively prevented AF due to sterile pericarditis (Figure 2C).

5. Small EVs in the Pathological Process of Heart Failure

Heart failure (HF) is a complex heterogeneous clinical syndrome, representing the terminal stage of several CVDs and associated with a high mortality. It affects approximately 1% to 2% of the adult population [310,311]. This syndrome is produced by an impairment of ventricular filing or an ejection of blood associated with symptoms of dyspnea, fatigue and in some cases, pulmonary edema, increased sympathetic activity and circulation redistribution [310,312] (Figure 2B). The main risk of HF is its constant progression. Therefore, the exploration of biomarkers for early diagnosis and identification of potential therapeutic targets is essential in the ongoing effort to fight against HF. The discovery of small EVs, including exosomes, gives us new opportunities to identify diagnostic and therapeutic biomarkers [313,314]. In the following paragraphs, we summarize the potential of small EVs in HF as diagnosis biomarkers and their use in therapy.

5.1. Small Extracellular Vesicle Transcriptomic Analyses in Heart Failure

It has been widely documented that the number of small EVs is increased in the serum of patients with HF. The origin of these small EVs is not exclusive from the damaged myocardium, since hallmarks present on the surface of the small EVs reveal that they are also derived from ECs as well as from immune cells, contributing to the development of this syndrome, promoting the inflammatory process [315,316,317,318,319,320]. Small EVs’ cargo can thus be a source of HF diagnostic biomarkers. As previously said, many cardiac diseases end in HF; one of these is DCM. In order to find biomarkers for DCM diagnosis, Zhang et al. (2023) performed isolation of exosomes in patients with chronic heart failure (CHF) caused by DCM. By next generation sequencing, they identified 92 microRNAs differentially expressed, and gene ontology analysis revealed that these microRNAs are related to oxytocin signalling, Hippo signalling and Ras signalling, pointing them out as candidates for CHF diagnosis [223]. As mentioned in previous paragraphs, diabetes mellitus is related with complications in HF with preserved ejection fraction (HFpEF). In a diabetic rat model with HFpEF, circulating exosomes decreased their miR-30d-5p and miR-126a-5p expression levels, suggesting that they can be used as biomarkers for HF [262]. In this scenario, other authors proposed several microRNAs presents in exosomes from patients with HF as biomarkers for diagnosis such as miR-27a, miR-34a, miR-92b, miR-146, miR-194, miR-425 and miR-744 [250,321,322,323,324] (Table 5). Other biomarkers for HF are piRNAs, which are ncRNAs 24-32 nucleotides in length associated with PIWI proteins, repressing transposable elements, and thus keeping the integrity of the germinal cell line. The analysis of piRNA cargo in exosomes from serum in patients with HF by RNAseq identified 585 upregulated piRNAs and 4623 downregulated piRNAs, highlighting hsa-piR-02009 and hsa-piR-006426 as the most downregulated piRNAs, suggesting them as potential HF biomarkers [325] (Table 5). Following ncRNAs, circRNAs can be a promising source of biomarkers. Han et al. (2020) performed a screening of differentially expressed cirRNAs in HF-EXO by next generation sequencing, identifying 56 differentially expressed circRNAs, further suggesting that hsa-circ-0097435 can be used as a biomarker involved in myocardial cell injury [326] (Table 5).

5.2. Small Extracellular Vesicle Proteomic Analyses in Heart Failure

The study of proteomic signatures in small EVs from the plasma of patients with HF is also a significant tool for HF diagnosis. As HF is associated with others pathologies, a proteomic study performed by the isolation of small EVs from the plama of patients with HF, dyspnea and renal dysfunction shows that these small EVs bear Cystatin C and CD14 that can be used as biomarkers for diagnosis [329].

5.3. Mechanistic Insights into Small Extracellular Vesicle Related with Heart Failure

As previously pointed out, the great challenge in HF is its unstoppable progression, and in this progression, small EVs have an important role, controlling the cell-to-cell communication participating in several processes such as cell adhesion, apoptosis, immune response and vascular function [319]. miR-22-3p was previously known as an HF biomarker present in exosomes [327]. Functionally, miR-22-3p targets FURIN, and thus induces the expression of apoptotic-related genes in HF [330]. Transforming growth factor beta (TGF-β) is a cytokine upregulated in several cardiac diseases [331]. The isolation of exosomes from fibroblasts treated with TGF-β identified 50 genes differentially expressed related with cardiac hypertrophy. Cardiomyocytes treated with these exosomes expressed 40 of these differentially expressed genes, inducing an HF phenotype, thus pointing out fibroblasts as targets for HF treatment [332]. Sympathetic hyperactivity also plays an important role in the progression of CHF. Inflammation in the rostral ventrolateral medulla (RMLV), a key region for sympathetic control, excites the activity of neurons and promotes an increase in sympathetic outflow. In a CHF rat model, the levels of circulating microRNAs in exosomes vs. microRNA levels in the RVLM were compared, and 59 DE-microRNAs and 5 overlapping microRNAs were identified. Three of these overlapping microRNAs were miR-214-3p that was upregulated in exosomes and in RVLM, while let7g-5p and let7i were downregulated in exosomes and in RVLM of CHF rats. In vitro studies in PC12 cells showed that miR-214-3p enhanced the inflammatory response, and let7g-5p and let7i-p reduced neuroinflamation, suggesting that the circulating exosomes enhanced the inflammatory response in the RVLM, contributing to sympathetic hyperactivity in CHF [328]. NF-E2-related factor 2 (NRF2) mediated sympathetic activation, and its expression was decreased in HF. Importantly, cardiac-EVs enriched with microRNAs mediate the cross talk between heart- and brain-targeting NRF2, thus producing an unbalance in the sympathetic outflow [333].

5.4. Therapeutic Approaches

Heart transplantation stands out as the most effective treatment against HF. However, its effectiveness is impaired by the reduced number of donors and the difficulty of organ preservation. Currently, cold static storage represents the main method of preservation, but its disadvantages limit the overall success of heart transplantation [334]. During cold ischemia preservation, the myocardial function of the transplanted heart is impaired and cell death increases. Of note, as mentioned in the previous paragraphs on therapeutic approaches, small EVs are deemed as promising therapeutic agents that could be used to avoid heart transplantation, and in this regard, several labs have conducted scientific studies in the context of HF. MSC-EVs therapy exerts a cardioprotective effect on the heart, improving its myocardial function and decreasing cell death [335]. Additional evidences reported that mesenchymal stem cell exosome (MSC-EXO) delivery produces a cardioprotective effect, promoting angiogenesis, and decreasing fibrosis and apoptosis by deactivating the Hippo-YAP pathway in HF [336,337]. The efficiency of MSC-EXO treatment can be improved with the administration of agomiR-125a-5p. Mechanistically, miR-125a-5p has KLF13, TGFB1 and DAAM1 as targets genes, involved in macrophages, CF and EC function, respectively. Moreover, agomiR-125a-5p treatment increased M2 macrophage polarization, promoted angiogenesis and attenuated fibroblast proliferation in a mouse and porcine model of I/R [338]. Treatment with BMSCs-EVs improved cardiac function and angiogenesis and alleviated fibrosis and inflammation in a rat model with acute myocardial infarction-induced HF via the delivery of BMP2 [339] (Figure 2). Exosomes derived from embryonic stem cells (ESCs-EXOs) are also able to induce myocardial angiogenesis in a transverse aortic constriction HF model through the FGF2 signalling pathway by attenuating myocardial damage [340]. Small EVs secreted by induced pluripotent stem cell-derived cardiovascular progenitors have a cardioprotective effect, making them a potential treatment for CHF with their cargo enriched in 16 microRNAs associated with tissue repair pathways [341]. Another source of small EVs suitable for HF treatment are small EVs from umbilical cord mesenchymal stem cells (HucMSC-EVs); these small EVs reduce oxidative stress and cell apoptosis through miR-100-5p, which has NOX4 as a target [342]. Small EVs from ECs with Krüppel-like factor 2 (KLF2) overexpression can be used as a treatment against DCM-HF; KLF2 exerts an anti-inflammatory effect, ameliorating left ventricular dysfunction in DCM mice. KLF2 targets CCR2 protein, preventing monocyte mobilization from the bone marrow [255]. Human trophoblast stem cell-derived exosomes (TSC-Exos) showed cardioprotective properties in a mice model of HF induced by doxycycline; the administration of TSC-Exos and miR-200b inhibitor decreases CM apoptosis, maintains the integrity of the mitochondria and improves cardiac function through an increase in Zeb1, which is a target of miR-200b and has an antiapoptotic effect [343,344] (Figure 2B).
In summary, HF is a complex syndrome with a difficult treatment process, and small EVs play a role in the development of the disease but can also be used as therapeutic or diagnosis tools.

6. Conclusions and Perspectives

Cell-to-cell communication represents a key biological process in cellular and tissue homeostasis that is mediated by different molecular mechanisms (see recent reviews [1,2,3]). In recent years, exosome-mediated cell-to-cell communication has emerged as a fundamental process contributing to cellular homeostasis. Multiple lines of evidence have also been reported, indicating that such exosome-mediated cellular communication is altered and impaired in pathological conditions, particularly in oncogenesis [4,11]. More recently, impaired exosome signaling has also been reported in distinct cardiovascular pathological contexts, including ischemic [67,68,69], electrical [280,281,282,283,284,285,286] and structural [215,223] cardiopathies. It is important to highlight in this context that increasing evidence is reported about the functional role of exosomes in distinct ischemic diseases, such as MI and coronary artery diseases, as well as in structural heart diseases such as DCM and HCM, as reviewed in this study. However, our understanding of the functional role of exosomes in electrical cardiovascular alterations is mostly confined to AF, while scarce or no evidence is yet reported in other relevant electrophysiological defects, such as Brugada, LQT and SQT syndromes, respectively. This might be indeed caused by the lower prevalence of these cardiac defects as compared to cardiac structural defects. In the next coming years, we will, therefore, witness increasing evidence of the functional role of exosomes in other cardiovascular diseases, such as valvular heart diseases and electrical cardiac wiring defects.
Exosome cargo selectively includes a large array of distinct biologically active molecules, such as lipids, proteins, coding and non-coding RNAs (microRNAs, lncRNAs and circRNAs) [1,2,3]. Evidence of the differential and selective distribution of combinations of these molecules, derived from distinct cardiovascular cell sources, i.e., cardiomyocytes, endothelial and/or epicardial cells and infiltrated immune cells, has been largely documented [298,299,300,301,302,303,304,305]. Such differential distribution, particularly of microRNAs, lncRNAs and circRNAs, in distinct pathological conditions has provided molecular hallmarks that can be exploited as sensitive biomarkers for the early diagnosis and prediction of distinct cardiovascular pathological conditions. Curiously, while our current identification of these biomarkers is greatly increasing, only very few of them are consistently validated in different studies. Such discrepancies might be derived, among other causes, from the variability of the exosome sample collection (plasma, serum, biopsies), their subsequent isolation procedures (ultracentrifugation, column binding) and their discovery/validation strategies (microarrays, next generation sequencing). Thus, there is an urgent need to invest in providing standardized pipelines that can pave the path for the discovery of robust biomarkers that can speed up their use into the clinical arena.
Curiously, while numerous reports highlight the fitness of distinct exosome-contained non-coding RNAs as cardiovascular disease biomarkers, the molecular mechanisms underlying their contribution to such pathology are, in most cases, scarce or even absent. Therefore, additional studies should be performed to increase our understanding of these molecular pathways. Such information will pave the path to design novel strategies by which exosome cargos can be modified, thus enabling the customization of cell–cell communication pathway modulation. Such cellular and molecular approaches will allow, in the coming future, the generation of personalized exosome cargos, which will selectively modulate specific signaling pathway responses, therefore opening novel therapeutic roads. As mentioned previously, exosomes are secreted by most eukaryotic cells, including embryonic stem cells (ESC). These ESC exosomes represent an innovative cell-free approach to harnessing the robust regenerative capabilities of ESC without the inherent risks associated with direct transplantation of ESC, such as the potential for teratoma formation [194,345]. Finally, while exosomes hold great promise for clinical applications, some associated challenges need to be overcome. In this context, it is crucial to emphasize the significance of the exosome delivery route, as it directly influences the therapeutic effectiveness of exosomes. Different administration methods for exosome delivery exist, ranging from less invasive techniques such as intranasal, inhalation and oral administration, to more invasive approaches like intravenous, intraperitoneal, intramyocardial and intraventricular injection. Numerous researchers suggest that intramyocardial injection stands out as the optimal delivery method for cardiac therapy. This is attributed to its ability to achieve the highest concentration of exosomes in the cardiac muscle, thereby preventing excessive accumulation in other organs [194,346,347,348]. The determination of optimal dosages for exosomes treatments is challenging, primarily influenced by delivery methodologies, and further by exosomes’ inherent characteristics such as their short half-life and Z-potential (aggregation index) [349]. Last but not least, for exosome treatments to transition into clinical practice, it is imperative to define biopharmaceutical safety standards and ensure cost-effectiveness.
To conclude, exosome-mediated cell-to-cell communication holds promise in understanding and treating cardiac diseases, offering potential biomarkers for early diagnosis. However, overcoming challenges related to standardized collection methods, molecular understanding, and therapeutic optimization is crucial for the successful clinical integration of exosome cardiac treatments.

Author Contributions

Conceptualization, E.L.-V.; writing—original draft preparation, S.C.-C., J.M.C.-C., D.F. and E.L.-V.; writing—review and editing, S.C.-C., J.M.C.-C., D.F. and E.L.-V.; visualization, E.L.-V.; supervision, E.L.-V. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by grants of the Ministry of Education and Science of the Spanish Government to D.F. (PID2022-138163OB-C32) and by grant-in-aid (CTS-466) of the Junta de Andalucía Regional Council.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Camussi, G.; Deregibus, M.-C.; Bruno, S.; Grange, C.; Fonsato, V.; Tetta, C. Exosome/microvesicle-mediated epigenetic reprogramming of cells. Am. J. Cancer Res. 2011, 1, 98–110. [Google Scholar]
  2. Lee, Y.; El Andaloussi, S.; Wood, M.J.A. Exosomes and microvesicles: Extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 2012, 21, R125–R134. [Google Scholar] [CrossRef]
  3. Zappulli, V.; Friis, K.P.; Fitzpatrick, Z.; Maguire, C.A.; Breakefield, X.O. Extracellular vesicles and intercellular communication within the nervous system. J. Clin. Investig. 2016, 126, 1198–1207. [Google Scholar] [CrossRef]
  4. Yang, M.; Wu, S.Y. The Advances and Challenges in Utilizing Exosomes for Delivering Cancer Therapeutics. Front. Pharmacol. 2018, 9, 735. [Google Scholar] [CrossRef]
  5. Witwer, K.W.; Buzás, E.I.; Bemis, L.T.; Bora, A.; Lässer, C.; Lötvall, J.; Nolte-‘t Hoen, E.N.; Piper, M.G.; Sivaraman, S.; Skog, J.; et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J. Extracell. Vesicle 2013, 2, 20360. [Google Scholar] [CrossRef]
  6. Van Der Pol, E.; Coumans, F.; Varga, Z.; Krumrey, M.; Nieuwland, R. Innovation in detection of microparticles and exosomes. J. Thromb. Haemost. 2013, 11, 36–45. [Google Scholar] [CrossRef]
  7. Meldolesi, J. Exosomes and Ectosomes in Intercellular Communication. Curr. Biol. 2018, 28, R435–R444. [Google Scholar] [CrossRef]
  8. Li, G.; Chen, T.; Dahlman, J.; Eniola-Adefeso, L.; Ghiran, I.C.; Kurre, P.; Lam, W.A.; Lang, J.K.; Marbán, E.; Martín, P.; et al. Current challenges and future directions for engineering extracellular vesicles for heart, lung, blood and sleep diseases. J. Extracell. Vesicle 2023, 12, 12305. [Google Scholar] [CrossRef]
  9. Robbins, P.D.; Morelli, A.E. Regulation of immune responses by extracellular vesicles. Nat. Rev. Immunol. 2014, 14, 195–208. [Google Scholar] [CrossRef]
  10. Yáñez-Mó, M.; Siljander, P.R.-M.; Andreu, Z.; Bedina Zavec, A.; Borràs, F.E.; Buzas, E.I.; Buzas, K.; Casal, E.; Cappello, F.; Carvalho, J.; et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicle 2015, 4, 27066. [Google Scholar] [CrossRef]
  11. Bebelman, M.P.; Smit, M.J.; Pegtel, D.M.; Baglio, S.R. Biogenesis and function of extracellular vesicles in cancer. Pharmacol. Ther. 2018, 188, 1–11. [Google Scholar] [CrossRef]
  12. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef]
  13. Borges, F.T.; Reis, L.A.; Schor, N. Extracellular vesicles: Structure, function, and potential clinical uses in renal diseases. Braz. J. Med. Biol. Res. 2013, 46, 824–830. [Google Scholar] [CrossRef]
  14. Simons, M.; Raposo, G. Exosomes—Vesicular carriers for intercellular communication. Curr. Opin. Cell Biol. 2009, 21, 575–581. [Google Scholar] [CrossRef]
  15. Théry, C.; Ostrowski, M.; Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 2009, 9, 581–593. [Google Scholar] [CrossRef]
  16. Mathivanan, S.; Simpson, R.J. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics 2009, 9, 4997–5000. [Google Scholar] [CrossRef]
  17. Doyle, L.M.; Wang, M.Z. Overview of Extracellular Vesicles, Their Origin, Composition, Purpose, and Methods for Exosome Isolation and Analysis. Cells 2019, 8, 727. [Google Scholar] [CrossRef]
  18. Bobrie, A.; Colombo, M.; Raposo, G.; Théry, C. Exosome Secretion: Molecular Mechanisms and Roles in Immune Responses. Traffic 2011, 12, 1659–1668. [Google Scholar] [CrossRef]
  19. Chaput, N.; Théry, C. Exosomes: Immune properties and potential clinical implementations. Semin. Immunopathol. 2011, 33, 419–440. [Google Scholar] [CrossRef]
  20. Robbins, P.D.; Dorronsoro, A.; Booker, C.N. Regulation of chronic inflammatory and immune processes by extracellular vesicles. J. Clin. Investig. 2016, 126, 1173–1180. [Google Scholar] [CrossRef]
  21. Zaborowski, M.P.; Balaj, L.; Breakefield, X.O.; Lai, C.P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. BioScience 2015, 65, 783–797. [Google Scholar] [CrossRef]
  22. Rak, J. Microparticles in Cancer. Semin. Thromb. Hemost. 2010, 36, 888–906. [Google Scholar] [CrossRef]
  23. Hood, J.L.; San, R.S.; Wickline, S.A. Exosomes Released by Melanoma Cells Prepare Sentinel Lymph Nodes for Tumor Metastasis. Cancer Res. 2011, 71, 3792–3801. [Google Scholar] [CrossRef]
  24. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  25. Wickman, G.; Julian, L.; Olson, M.F. How apoptotic cells aid in the removal of their own cold dead bodies. Cell Death Differ. 2012, 19, 735–742. [Google Scholar] [CrossRef]
  26. Théry, C.; Boussac, M.; Véron, P.; Ricciardi-Castagnoli, P.; Raposo, G.; Garin, J.; Amigorena, S. Proteomic Analysis of Dendritic Cell-Derived Exosomes: A Secreted Subcellular Compartment Distinct from Apoptotic Vesicles. J. Immunol. 2001, 166, 7309–7318. [Google Scholar] [CrossRef]
  27. Escrevente, C.; Keller, S.; Altevogt, P.; Costa, J. Interaction and uptake of exosomes by ovarian cancer cells. BMC Cancer 2011, 11, 108. [Google Scholar] [CrossRef] [PubMed]
  28. Arraud, N.; Linares, R.; Tan, S.; Gounou, C.; Pasquet, J.-M.; Mornet, S.; Brisson, A.R. Extracellular vesicles from blood plasma: Determination of their morphology, size, phenotype and concentration. J. Thromb. Haemost. 2014, 12, 614–627. [Google Scholar] [CrossRef]
  29. Sun, Y.; Xia, Z.; Shang, Z.; Sun, K.; Niu, X.; Qian, L.; Fan, L.-Y.; Cao, C.-X.; Xiao, H. Facile preparation of salivary extracellular vesicles for cancer proteomics. Sci. Rep. 2016, 6, 24669. [Google Scholar] [CrossRef]
  30. Ranghino, A.; Dimuccio, V.; Papadimitriou, E.; Bussolati, B. Extracellular vesicles in the urine: Markers and mediators of tissue damage and regeneration. Clin. Kidney J. 2015, 8, 23–30. [Google Scholar] [CrossRef]
  31. Théry, C. Diagnosis by extracellular vesicles. Nature 2015, 523, 161–162. [Google Scholar] [CrossRef]
  32. Thompson, A.G.; Gray, E.; Heman-Ackah, S.M.; Mäger, I.; Talbot, K.; Andaloussi, S.E.; Wood, M.J.; Turner, M.R. Extracellular vesicles in neurodegenerative disease—Pathogenesis to biomarkers. Nat. Rev. Neurol. 2016, 12, 346–357. [Google Scholar] [CrossRef]
  33. Turpin, D.; Truchetet, M.-E.; Faustin, B.; Augusto, J.-F.; Contin-Bordes, C.; Brisson, A.; Blanco, P.; Duffau, P. Role of extracellular vesicles in autoimmune diseases. Autoimmun. Rev. 2016, 15, 174–183. [Google Scholar] [CrossRef]
  34. 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]
  35. Benjamin, E.J.; Blaha, M.J.; Chiuve, S.E.; Cushman, M.; Das, S.R.; Deo, R.; De Ferranti, S.D.; Floyd, J.; Fornage, M.; Gillespie, C.; et al. Heart Disease and Stroke Statistics—2017 Update: A Report From the American Heart Association. Circulation 2017, 135, e146–e603. [Google Scholar] [CrossRef]
  36. WHO, Fact Sheet about Cardiovascular Diseases. 2016. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 8 December 2023).
  37. Gartz, M.; Strande, J.L. Examining the Paracrine Effects of Exosomes in Cardiovascular Disease and Repair. J. Am. Heart Assoc. 2018, 7, e007954. [Google Scholar] [CrossRef]
  38. Chistiakov, D.; Orekhov, A.; Bobryshev, Y. Cardiac Extracellular Vesicles in Normal and Infarcted Heart. Int. J. Mol. Sci. 2016, 17, 63. [Google Scholar] [CrossRef]
  39. Pironti, G.; Strachan, R.T.; Abraham, D.; Mon-Wei Yu, S.; Chen, M.; Chen, W.; Hanada, K.; Mao, L.; Watson, L.J.; Rockman, H.A. Circulating Exosomes Induced by Cardiac Pressure Overload Contain Functional Angiotensin II Type 1 Receptors. Circulation 2015, 131, 2120–2130. [Google Scholar] [CrossRef]
  40. Sahoo, S.; Losordo, D.W. Exosomes and cardiac repair after myocardial infarction. Circ. Res. 2014, 114, 333–344. [Google Scholar] [CrossRef]
  41. Sluijter, J.P.G.; Verhage, V.; Deddens, J.C.; Van Den Akker, F.; Doevendans, P.A. Microvesicles and exosomes for intracardiac communication. Cardiovasc. Res. 2014, 102, 302–311. [Google Scholar] [CrossRef]
  42. Roth, G.A.; Mensah, G.A.; Johnson, C.O.; Addolorato, G.; Ammirati, E.; Baddour, L.M.; Barengo, N.C.; Beaton, A.Z.; Benjamin, E.J.; Benziger, C.P.; et al. Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019. J. Am. Coll. Cardiol. 2020, 76, 2982–3021. [Google Scholar] [CrossRef]
  43. Saleh, M.; Ambrose, J.A. Understanding myocardial infarction. F1000Res 2018, 7, 1378. [Google Scholar] [CrossRef]
  44. Jenča, D.; Melenovský, V.; Stehlik, J.; Staněk, V.; Kettner, J.; Kautzner, J.; Adámková, V.; Wohlfahrt, P. Heart failure after myocardial infarction: Incidence and predictors. ESC Heart Fail. 2021, 8, 222–237. [Google Scholar] [CrossRef]
  45. Börschel, C.S.; Schnabel, R.B. The imminent epidemic of atrial fibrillation and its concomitant diseases—Myocardial infarction and heart failure—A cause for concern. Int. J. Cardiol. 2019, 287, 162–173. [Google Scholar] [CrossRef]
  46. Thygesen, K.; Alpert, J.S.; Jaffe, A.S.; Chaitman, B.R.; Bax, J.J.; Morrow, D.A.; White, H.D. Fourth universal definition of myocardial infarction (2018). J. Am. Coll. Cardiol. 2018, 72, 2231–2264. [Google Scholar] [CrossRef]
  47. Golforoush, P.; Yellon, D.M.; Davidson, S.M. Mouse models of atherosclerosis and their suitability for the study of myocardial infarction. Basic Res. Cardiol. 2020, 115, 73. [Google Scholar] [CrossRef]
  48. Scheen, A. De l’athérosclérose à l’athérothrombose. Rev. Med. Liege 2018, 73, 224–228. [Google Scholar]
  49. Anderson, J.L.; Morrow, D.A. Acute Myocardial Infarction. N. Engl. J. Med. 2017, 376, 2053–2064. [Google Scholar] [CrossRef]
  50. Mitsis, A.; Gragnano, F. Myocardial Infarction with and without ST-segment Elevation: A Contemporary Reappraisal of Similarities and Differences. Curr. Cardiol. Rev. 2021, 17, e230421189013. [Google Scholar] [CrossRef]
  51. Wang, X.; Guo, Z.; Ding, Z.; Mehta, J.L. Inflammation, Autophagy, and Apoptosis after Myocardial Infarction. J. Am. Heart Assoc. 2018, 7, e008024. [Google Scholar] [CrossRef]
  52. Moreira, D.M.; Da Silva, R.L.; Vieira, J.L.; Fattah, T.; Lueneberg, M.E.; Gottschall, C.A.M. Role of Vascular Inflammation in Coronary Artery Disease: Potential of Anti-inflammatory Drugs in the Prevention of Atherothrombosis: Inflammation and Anti-Inflammatory Drugs in Coronary Artery Disease. Am. J. Cardiovasc. Drugs 2015, 15, 1–11. [Google Scholar] [CrossRef]
  53. 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]
  54. Talman, V.; Ruskoaho, H. Cardiac fibrosis in myocardial infarction—From repair and remodeling to regeneration. Cell Tissue Res. 2016, 365, 563–581. [Google Scholar] [CrossRef]
  55. He, L.; Huang, X.; Kanisicak, O.; Li, Y.; Wang, Y.; Li, Y.; Pu, W.; Liu, Q.; Zhang, H.; Tian, X.; et al. Preexisting endothelial cells mediate cardiac neovascularization after injury. J. Clin. Investig. 2017, 127, 2968–2981. [Google Scholar] [CrossRef]
  56. 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]
  57. Loyer, X.; Zlatanova, I.; Devue, C.; Yin, M.; Howangyin, K.-Y.; Klaihmon, P.; Guerin, C.L.; Kheloufi, M.; Vilar, J.; Zannis, K.; et al. Intra-Cardiac Release of Extracellular Vesicles Shapes Inflammation Following Myocardial Infarction. Circ. Res. 2018, 123, 100–106. [Google Scholar] [CrossRef]
  58. Pan, W.; Zhu, Y.; Meng, X.; Zhang, C.; Yang, Y.; Bei, Y. Immunomodulation by Exosomes in Myocardial Infarction. J. Cardiovasc. Trans. Res. 2019, 12, 28–36. [Google Scholar] [CrossRef]
  59. Xu, Y.; Wu, A.; Chen, J.; Song, X.; Chen, M.; Liu, Q. Limb-Bud and Heart (LBH) Upregulation in Cardiomyocytes under Hypoxia Promotes the Activation of Cardiac Fibroblasts via Exosome Secretion. Mediat. Inflamm. 2022, 2022, 8939449. [Google Scholar] [CrossRef]
  60. Nian, W.; Fu, C. Exosomes in Myocardial Infarction: Therapeutic Potential and Clinical Application. J. Cardiovasc. Trans. Res. 2023, 16, 87–96. [Google Scholar] [CrossRef]
  61. Gong, Z.; Wen, M.; Zhang, W.; Yu, L.; Huang, C.; Xu, Y.; Xia, Z.; Xu, M.; Xu, J.; Liang, Q.; et al. Plasma exosomes induce inflammatory immune response in patients with acute myocardial infarction. Arch. Physiol. Biochem. 2023, 129, 1168–1176. [Google Scholar] [CrossRef]
  62. Zhang, Z.; Xu, Y.; Cao, C.; Wang, B.; Guo, J.; Qin, Z.; Lu, Y.; Zhang, J.; Zhang, L.; Wang, W.; et al. Exosomes as a messager to regulate the crosstalk between macrophages and cardiomyocytes under hypoxia conditions. J. Cell. Mol. Medi 2022, 26, 1486–1500. [Google Scholar] [CrossRef]
  63. Cheng, M.; An, S.; Li, J. Identifying key genes associated with acute myocardial infarction. Medicine 2017, 96, e7741. [Google Scholar] [CrossRef]
  64. Keller, T.; Zeller, T.; Peetz, D.; Tzikas, S.; Roth, A.; Czyz, E.; Bickel, C.; Baldus, S.; Warnholtz, A.; Fröhlich, M.; et al. Sensitive Troponin I Assay in Early Diagnosis of Acute Myocardial Infarction. N. Engl. J. Med. 2009, 361, 868–877. [Google Scholar] [CrossRef]
  65. Ai, J.; Zhang, R.; Li, Y.; Pu, J.; Lu, Y.; Jiao, J.; Li, K.; Yu, B.; Li, Z.; Wang, R.; et al. Circulating microRNA-1 as a potential novel biomarker for acute myocardial infarction. Biochem. Biophys. Res. Commun. 2010, 391, 73–77. [Google Scholar] [CrossRef]
  66. Wang, G.-K.; Zhu, J.-Q.; Zhang, J.-T.; Li, Q.; Li, Y.; He, J.; Qin, Y.-W.; Jing, Q. Circulating microRNA: A novel potential biomarker for early diagnosis of acute myocardial infarction in humans. Eur. Heart J. 2010, 31, 659–666. [Google Scholar] [CrossRef]
  67. Liu, P.; Wang, S.; Li, K.; Yang, Y.; Man, Y.; Du, F.; Wang, L.; Tian, J.; Su, G. Exosomal microRNA-4516, microRNA-203 and SFRP1 are potential biomarkers of acute myocardial infarction. Mol. Med. Rep. 2023, 27, 124. [Google Scholar] [CrossRef]
  68. Chen, S.; Fang, H.; Liu, R.; Fang, Y.; Wu, Z.; Xie, P. miR-6718-5p and miR-4329 can be used as potential biomarkers for acute myocardial infarction. J. Card. Surg. 2021, 36, 3721–3728. [Google Scholar] [CrossRef]
  69. Guo, M.; Li, R.; Yang, L.; Zhu, Q.; Han, M.; Chen, Z.; Ruan, F.; Yuan, Y.; Liu, Z.; Huang, B.; et al. Evaluation of exosomal miRNAs as potential diagnostic biomarkers for acute myocardial infarction using next-generation sequencing. Ann. Transl. Med. 2021, 9, 219. [Google Scholar] [CrossRef]
  70. Zhao, X.; Jia, Y.; Chen, H.; Yao, H.; Guo, W. Plasma-derived exosomal miR-183 associates with protein kinase activity and may serve as a novel predictive biomarker of myocardial ischemic injury. Exp. Ther. Med. 2019, 18, 179–187. [Google Scholar] [CrossRef]
  71. Ling, H.; Guo, Z.; Shi, Y.; Zhang, L.; Song, C. Serum Exosomal MicroRNA-21, MicroRNA-126, and PTEN Are Novel Biomarkers for Diagnosis of Acute Coronary Syndrome. Front. Physiol. 2020, 11, 654. [Google Scholar] [CrossRef]
  72. Hu, J.; Huang, C.-X.; Rao, P.-P.; Zhou, J.-P.; Wang, X.; Tang, L.; Liu, M.-X.; Zhang, G.-G. Inhibition of microRNA-155 attenuates sympathetic neural remodeling following myocardial infarction via reducing M1 macrophage polarization and inflammatory responses in mice. Eur. J. Pharmacol. 2019, 851, 122–132. [Google Scholar] [CrossRef]
  73. Hu, C.; Liao, J.; Huang, R.; Su, Q.; He, L. MicroRNA-155-5p in serum derived-exosomes promotes ischaemia–reperfusion injury by reducing CypD ubiquitination by NEDD4. ESC Heart Fail. 2023, 10, 1144–1157. [Google Scholar] [CrossRef]
  74. Chen, C.; Cai, S.; Wu, M.; Wang, R.; Liu, M.; Cao, G.; Dong, M.; Yiu, K.-H. Role of Cardiomyocyte-Derived Exosomal MicroRNA-146a-5p in Macrophage Polarization and Activation. Dis. Markers 2022, 2022, 2948578. [Google Scholar] [CrossRef]
  75. 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]
  76. Ribeiro-Rodrigues, T.M.; Laundos, T.L.; Pereira-Carvalho, R.; Batista-Almeida, D.; Pereira, R.; Coelho-Santos, V.; Silva, A.P.; Fernandes, R.; Zuzarte, M.; Enguita, F.J.; et al. Exosomes secreted by cardiomyocytes subjected to ischaemia promote cardiac angiogenesis. Cardiovasc. Res. 2017, 113, 1338–1350. [Google Scholar] [CrossRef]
  77. 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. 2020, 47, 315–325. [Google Scholar] [CrossRef]
  78. 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]
  79. Li, H.; Ding, J.; Liu, W.; Wang, X.; Feng, Y.; Guan, H.; Chen, Z. Plasma exosomes from patients with acute myocardial infarction alleviate myocardial injury by inhibiting ferroptosis through miR-26b-5p/SLC7A11 axis. Life Sci. 2023, 322, 121649. [Google Scholar] [CrossRef]
  80. Sun, P.; Wang, C.; Mang, G.; Xu, X.; Fu, S.; Chen, J.; Wang, X.; Wang, W.; Li, H.; Zhao, P.; et al. Extracellular vesicle-packaged mitochondrial disturbing miRNA exacerbates cardiac injury during acute myocardial infarction. Clin. Transl. Med. 2022, 12, e779. [Google Scholar] [CrossRef]
  81. Zheng, M.; Liu, X.; Han, R.; Yuan, W.; Sun, K.; Zhong, J.; Yang, X. Circulating exosomal long non-coding RNAs in patients with acute myocardial infarction. J. Cell. Mol. Med. 2020, 24, 9388–9396. [Google Scholar] [CrossRef]
  82. Dang, Y.; Hua, W.; Zhang, X.; Sun, H.; Zhang, Y.; Yu, B.; Wang, S.; Zhang, M.; Kong, Z.; Pan, D.; et al. Anti-angiogenic effect of exo-LncRNA TUG1 in myocardial infarction and modulation by remote ischemic conditioning. Basic Res. Cardiol. 2023, 118, 1. [Google Scholar] [CrossRef]
  83. Lin, B.; Chen, X.; Lu, C.; Xu, J.; Qiu, Y.; Liu, X.; Song, H.; Chen, A.; Xiong, J.; Wang, K.; et al. Loss of exosomal LncRNA HCG15 prevents acute myocardial ischemic injury through the NF-κB/p65 and p38 pathways. Cell Death Dis. 2021, 12, 1007. [Google Scholar] [CrossRef] [PubMed]
  84. Wang, W.; Wang, Y.; Piao, H.; Li, B.; Huang, M.; Zhu, Z.; Li, D.; Wang, T.; Xu, R.; Liu, K. Circular RNAs as potential biomarkers and therapeutics for cardiovascular disease. PeerJ 2019, 7, e6831. [Google Scholar] [CrossRef] [PubMed]
  85. Wang, G.; Wang, C.; Huang, Z.; Sun, S.; Chen, Y. Exosomal circ-0020887 and circ-0009590 as novel biomarkers for the diagnosis and prediction of short-term adverse cardiovascular outcomes in STEMI patients. Open Med. 2023, 18, 20230807. [Google Scholar] [CrossRef]
  86. Zhu, J.; Chen, Z.; Peng, X.; Zheng, Z.; Le, A.; Guo, J.; Ma, L.; Shi, H.; Yao, K.; Zhang, S.; et al. Extracellular Vesicle-Derived circITGB1 Regulates Dendritic Cell Maturation and Cardiac Inflammation via miR-342-3p/NFAM1. Oxidative Med. Cell. Longev. 2022, 2022, 8392313. [Google Scholar] [CrossRef] [PubMed]
  87. 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. Oxidative Med. Cell. Longev. 2019, 2019, 7954657. [Google Scholar] [CrossRef]
  88. Lan, Z.; Wang, T.; Zhang, L.; Jiang, Z.; Zou, X. CircSLC8A1 Exacerbates Hypoxia-Induced Myocardial Injury via Interacting with MiR-214-5p to Upregulate TEAD1 Expression. Int. Heart J. 2022, 63, 591–601. [Google Scholar] [CrossRef]
  89. 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. Oxidative Med. Cell. Longev. 2020, 2020, 8418407. [Google Scholar] [CrossRef]
  90. Fan, M.; Liang, T.; Xie, F.; Ma, P.; Li, J. Exosomal circ_HIPK3 reduces apoptosis in H2O2-induced AC16 cardiomyocytes through miR-33a-5p/IRS1 axis. Transpl. Immunol. 2023, 80, 101862. [Google Scholar] [CrossRef]
  91. Xie, Y.; Zhang, H.; Huang, T. Quantitative proteomics reveal three potential biomarkers for risk assessment of acute myocardial infarction. Bioengineered 2022, 13, 4939–4950. [Google Scholar] [CrossRef]
  92. Chen, Z.; Yan, Y.; Wu, J.; Qi, C.; Liu, J.; Wang, J. Expression level and diagnostic value of exosomal NEAT1/miR-204/MMP-9 in acute ST-segment elevation myocardial infarction. IUBMB Life 2020, 72, 2499–2507. [Google Scholar] [CrossRef]
  93. Foglio, E.; Puddighinu, G.; Fasanaro, P.; D’Arcangelo, D.; Perrone, G.A.; Mocini, D.; Campanella, C.; Coppola, L.; Logozzi, M.; Azzarito, T.; et al. Exosomal clusterin, identified in the pericardial fluid, improves myocardial performance following MI through epicardial activation, enhanced arteriogenesis and reduced apoptosis. Int. J. Cardiol. 2015, 197, 333–347. [Google Scholar] [CrossRef]
  94. Li, Z.; Huo, X.; Chen, K.; Yang, F.; Tan, W.; Zhang, Q.; Yu, H.; Li, C.; Zhou, D.; Chen, H.; et al. Profilin 2 and Endothelial Exosomal Profilin 2 Promote Angiogenesis and Myocardial Infarction Repair in Mice. Front. Cardiovasc. Med. 2022, 9, 781753. [Google Scholar] [CrossRef]
  95. Gidlöf, O.; Evander, M.; Rezeli, M.; Marko-Varga, G.; Laurell, T.; Erlinge, D. Proteomic profiling of extracellular vesicles reveals additional diagnostic biomarkers for myocardial infarction compared to plasma alone. Sci. Rep. 2019, 9, 8991. [Google Scholar] [CrossRef]
  96. 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]
  97. Liu, S.; Chen, J.; Shi, J.; Zhou, W.; Wang, L.; Fang, W.; Zhong, Y.; Chen, X.; Chen, Y.; Sabri, A.; et al. M1-like macrophage-derived exosomes suppress angiogenesis and exacerbate cardiac dysfunction in a myocardial infarction microenvironment. Basic Res. Cardiol. 2020, 115, 22. [Google Scholar] [CrossRef]
  98. 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]
  99. 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]
  100. Huang, J.; Wang, F.; Sun, X.; Chu, X.; Jiang, R.; Wang, Y.; Pang, L. Myocardial infarction cardiomyocytes-derived exosomal miR-328-3p promote apoptosis via Caspase signaling. Am. J. Transl. Res. 2021, 13, 2365–2378. [Google Scholar]
  101. 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]
  102. Jiang, W.; Song, Q.; Lu, Z.; Wang, S.; Liu, T.; Wang, X.; Wang, M. Myocardial Infarction-Associated Extracellular Vesicle-Delivered miR-208b Affects the Growth of Human Umbilical Vein Endothelial Cells via Regulating CDKN1A. BioMed Res. Int. 2021, 2021, 9965639. [Google Scholar] [CrossRef]
  103. 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, 285–289. [Google Scholar] [CrossRef]
  104. Yang, Y.; Li, Y.; Chen, X.; Cheng, X.; Liao, Y.; Yu, X. Exosomal transfer of miR-30a between cardiomyocytes regulates autophagy after hypoxia. J. Mol. Med. 2016, 94, 711–724. [Google Scholar] [CrossRef]
  105. Wang, B.; Cao, C.; Han, D.; Bai, J.; Guo, J.; Guo, Q.; Li, D.; Zhang, J.; Zhang, Z.; Wang, Y.; et al. Dysregulation of miR-342-3p in plasma exosomes derived from convalescent AMI patients and its consequences on cardiac repair. Biomed. Pharmacother. 2021, 142, 112056. [Google Scholar] [CrossRef]
  106. Sun, S.; Wu, Y.; Maimaitijiang, A.; Huang, Q.; Chen, Q. Ferroptotic cardiomyocyte-derived exosomes promote cardiac macrophage M1 polarization during myocardial infarction. PeerJ 2022, 10, e13717. [Google Scholar] [CrossRef]
  107. Zhang, Q.; Chen, L.; Huang, L.; Cheng, H.; Wang, L.; Xu, L.; Hu, D.; He, C.; Fu, C.; Wei, Q. CD44 promotes angiogenesis in myocardial infarction through regulating plasma exosome uptake and further enhancing FGFR2 signaling transduction. Mol. Med. 2022, 28, 145. [Google Scholar] [CrossRef]
  108. Zhang, Y.; Gao, W.; Yuan, J.; Zhong, X.; Yao, K.; Luo, R.; Liu, H. CCR7 Mediates Dendritic-Cell-Derived Exosome Migration and Improves Cardiac Function after Myocardial Infarction. Pharmaceutics 2023, 15, 461. [Google Scholar] [CrossRef]
  109. Liu, H.; Gao, W.; Yuan, J.; Wu, C.; Yao, K.; Zhang, L.; Ma, L.; Zhu, J.; Zou, Y.; Ge, J. Exosomes derived from dendritic cells improve cardiac function via activation of CD4+ T lymphocytes after myocardial infarction. J. Mol. Cell. Cardiol. 2016, 91, 123–133. [Google Scholar] [CrossRef]
  110. Yue, Y.; Garikipati, V.N.S.; Verma, S.K.; Goukassian, D.A.; Kishore, R. Interleukin-10 Deficiency Impairs Reparative Properties of Bone Marrow-Derived Endothelial Progenitor Cell Exosomes. Tissue Eng. Part A 2017, 23, 1241–1250. [Google Scholar] [CrossRef] [PubMed]
  111. Yue, Y.; Wang, C.; Benedict, C.; Huang, G.; Truongcao, M.; Roy, R.; Cimini, M.; Garikipati, V.N.S.; Cheng, Z.; Koch, W.J.; et al. Interleukin-10 Deficiency Alters Endothelial Progenitor Cell–Derived Exosome Reparative Effect on Myocardial Repair via Integrin-Linked Kinase Enrichment. Circ. Res. 2020, 126, 315–329. [Google Scholar] [CrossRef]
  112. Zhang, Y.-Q.; Hong, L.; Jiang, Y.-F.; Hu, S.-D.; Zhang, N.-N.; Xu, L.-B.; Li, H.-X.; Xu, G.-D.; Zhou, Y.-F.; Sun, K.-Y. hAECs and their exosomes improve cardiac function after acute myocardial infarction in rats. Aging 2021, 13, 15032–15043. [Google Scholar] [CrossRef]
  113. Deng, S.; Zhou, X.; Ge, Z.; Song, Y.; Wang, H.; Liu, X.; Zhang, D. Exosomes from adipose-derived mesenchymal stem cells ameliorate cardiac damage after myocardial infarction by activating S1P/SK1/S1PR1 signaling and promoting macrophage M2 polarization. Int. J. Biochem. Cell Biol. 2019, 114, 105564. [Google Scholar] [CrossRef]
  114. Hu, H.; Wu, J.; Cao, C.; Ma, L. Exosomes derived from regulatory T cells ameliorate acute myocardial infarction by promoting macrophage M2 polarization. IUBMB Life 2020, 72, 2409–2419. [Google Scholar] [CrossRef]
  115. Liu, L.; Jin, X.; Hu, C.-F.; Li, R.; Zhou, Z.; Shen, C.-X. Exosomes Derived from Mesenchymal Stem Cells Rescue Myocardial Ischaemia/Reperfusion Injury by Inducing Cardiomyocyte Autophagy Via AMPK and Akt Pathways. Cell Physiol. Biochem. 2017, 43, 52–68. [Google Scholar] [CrossRef]
  116. Zhu, D.; Liu, S.; Huang, K.; Wang, Z.; Hu, S.; Li, J.; Li, Z.; Cheng, K. Intrapericardial Exosome Therapy Dampens Cardiac Injury via Activating Foxo3. Circ. Res. 2022, 131, e135–e150. [Google Scholar] [CrossRef]
  117. Jiao, W.; Hao, J.; Xie, Y.; Meng, M.; Gao, W. EZH2 mitigates the cardioprotective effects of mesenchymal stem cell-secreted exosomes against infarction via HMGA2-mediated PI3K/AKT signaling. BMC Cardiovasc. Disord. 2022, 22, 95. [Google Scholar] [CrossRef]
  118. Xu, R.; Zhang, F.; Chai, R.; Zhou, W.; Hu, M.; Liu, B.; Chen, X.; Liu, M.; Xu, Q.; Liu, N.; et al. Exosomes derived from pro-inflammatory bone marrow-derived mesenchymal stem cells reduce inflammation and myocardial injury via mediating macrophage polarization. J. Cell. Mol. Med. 2019, 23, 7617–7631. [Google Scholar] [CrossRef] [PubMed]
  119. 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]
  120. Gu, H.; Liu, Z.; Li, Y.; Xie, Y.; Yao, J.; Zhu, Y.; Xu, J.; Dai, Q.; Zhong, C.; Zhu, H.; et al. Serum-Derived Extracellular Vesicles Protect Against Acute Myocardial Infarction by Regulating miR-21/PDCD4 Signaling Pathway. Front. Physiol. 2018, 9, 348. [Google Scholar] [CrossRef]
  121. Wei, Z.; Chen, Z.; Zhao, Y.; Fan, F.; Xiong, W.; Song, S.; Yin, Y.; Hu, J.; Yang, K.; Yang, L.; et al. Mononuclear phagocyte system blockade using extracellular vesicles modified with CD47 on membrane surface for myocardial infarction reperfusion injury treatment. Biomaterials 2021, 275, 121000. [Google Scholar] [CrossRef] [PubMed]
  122. Zhang, J.; Lu, Y.; Mao, Y.; Yu, Y.; Wu, T.; Zhao, W.; Zhu, Y.; Zhao, P.; Zhang, F. IFN-γ enhances the efficacy of mesenchymal stromal cell-derived exosomes via miR-21 in myocardial infarction rats. Stem. Cell Res. Ther. 2022, 13, 333. [Google Scholar] [CrossRef]
  123. Luther, K.M.; Haar, L.; McGuinness, M.; Wang, Y.; Lynch Iv, T.L.; Phan, A.; Song, Y.; Shen, Z.; Gardner, G.; Kuffel, G.; et al. Exosomal miR-21a-5p mediates cardioprotection by mesenchymal stem cells. J. Mol. Cell. Cardiol. 2018, 119, 125–137. [Google Scholar] [CrossRef]
  124. Zhu, L.-P.; Tian, T.; Wang, J.-Y.; He, J.-N.; Chen, T.; Pan, M.; Xu, L.; Zhang, H.; 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]
  125. Xiao, C.; Wang, K.; Xu, Y.; Hu, H.; Zhang, N.; Wang, Y.; Zhong, Z.; Zhao, J.; Li, Q.; Zhu, D.; et al. Transplanted Mesenchymal Stem Cells Reduce Autophagic Flux in Infarcted Hearts via the Exosomal Transfer of miR-125b. Circ. Res. 2018, 123, 564–578. [Google Scholar] [CrossRef] [PubMed]
  126. Ma, T.; Chen, Y.; Chen, Y.; Meng, Q.; Sun, J.; Shao, L.; Yu, Y.; Huang, H.; Hu, Y.; Yang, Z.; et al. MicroRNA-132, Delivered by Mesenchymal Stem Cell-Derived Exosomes, Promote Angiogenesis in Myocardial Infarction. Stem. Cells Int. 2018, 2018, 3290372. [Google Scholar] [CrossRef] [PubMed]
  127. Wang, X.; Zhao, Y.; Sun, L.; Shi, Y.; Li, Z.; Zhao, X.; Xu, C.; Ji, H.; Wang, M.; Xu, W.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells improve myocardial repair via upregulation of Smad7. Int. J. Mol. Med. 2018, 41, 3063–3072. [Google Scholar] [CrossRef]
  128. Yang, M.; Liao, M.; Liu, R.; Zhang, Q.; Zhang, S.; He, Y.; Jin, J.; Zhang, P.; Zhou, L. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles loaded with miR-223 ameliorate myocardial infarction through P53/S100A9 axis. Genomics 2022, 114, 110319. [Google Scholar] [CrossRef]
  129. Song, Y.; Wang, B.; Zhu, X.; Hu, J.; Sun, J.; Xuan, J.; Ge, Z. Human umbilical cord blood–derived MSCs exosome attenuate myocardial injury by inhibiting ferroptosis in acute myocardial infarction mice. Cell Biol. Toxicol. 2021, 37, 51–64. [Google Scholar] [CrossRef]
  130. Zhu, W.; Sun, L.; Zhao, P.; Liu, Y.; Zhang, J.; Zhang, Y.; Hong, Y.; Zhu, Y.; Lu, Y.; Zhao, W.; et al. Macrophage migration inhibitory factor facilitates the therapeutic efficacy of mesenchymal stem cells derived exosomes in acute myocardial infarction through upregulating miR-133a-3p. J. Nanobiotechnol. 2021, 19, 61. [Google Scholar] [CrossRef]
  131. De Couto, G.; Gallet, R.; Cambier, L.; Jaghatspanyan, E.; Makkar, N.; Dawkins, J.F.; Berman, B.P.; Marbán, E. Exosomal MicroRNA Transfer Into Macrophages Mediates Cellular Postconditioning. Circulation 2017, 136, 200–214. [Google Scholar] [CrossRef] [PubMed]
  132. Wei, Z.; Qiao, S.; Zhao, J.; Liu, Y.; Li, Q.; Wei, Z.; Dai, Q.; Kang, L.; Xu, B. miRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced inflammatory response after myocardial ischemia-reperfusion injury. Life Sci. 2019, 232, 116632. [Google Scholar] [CrossRef] [PubMed]
  133. Li, X.; Hu, X.; Chen, Q.; Jiang, T. Bone marrow mesenchymal stem cell-derived exosomes carrying E3 ubiquitin ligase ITCH attenuated cardiomyocyte apoptosis by mediating apoptosis signal-regulated kinase-1. Pharmacogenet. Genom. 2023, 33, 117–125. [Google Scholar] [CrossRef]
  134. Ning, H.; Chen, H.; Deng, J.; Xiao, C.; Xu, M.; Shan, L.; Yang, C.; Zhang, Z. Exosomes secreted by FNDC5-BMMSCs protect myocardial infarction by anti-inflammation and macrophage polarization via NF-κB signaling pathway and Nrf2/HO-1 axis. Stem Cell Res. Ther. 2021, 12, 519. [Google Scholar] [CrossRef]
  135. Gong, X.; Liu, H.; Wang, S.; Liang, S.; Wang, G. Exosomes derived from SDF1-overexpressing mesenchymal stem cells inhibit ischemic myocardial cell apoptosis and promote cardiac endothelial microvascular regeneration in mice with myocardial infarction. J. Cell. Physiol. 2019, 234, 13878–13893. [Google Scholar] [CrossRef]
  136. 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]
  137. Liu, D.; Gu, G.; Gan, L.; Yan, W.; Zhang, Z.; Yao, P.; Zhu, D.; Lau, W.B.; Xie, D.; Wu, S.; et al. Identification of a CTRP9 C-Terminal polypeptide capable of enhancing bone-derived mesenchymal stem cell cardioprotection through promoting angiogenic exosome production. Redox Biol. 2021, 41, 101929. [Google Scholar] [CrossRef]
  138. Liu, X.; Li, X.; Zhu, W.; Zhang, Y.; Hong, Y.; Liang, X.; Fan, B.; Zhao, H.; He, H.; Zhang, F. Exosomes from mesenchymal stem cells overexpressing MIF enhance myocardial repair. J. Cell. Physiol. 2020, 235, 8010–8022. [Google Scholar] [CrossRef]
  139. Wang, Q.; Zhang, L.; Sun, Z.; Chi, B.; Zou, A.; Mao, L.; Xiong, X.; Jiang, J.; Sun, L.; Zhu, W.; et al. HIF-1α overexpression in mesenchymal stem cell-derived exosome-encapsulated arginine-glycine-aspartate (RGD) hydrogels boost therapeutic efficacy of cardiac repair after myocardial infarction. Mater. Today Bio 2021, 12, 100171. [Google Scholar] [CrossRef]
  140. Sun, J.; Shen, H.; Shao, L.; Teng, X.; Chen, Y.; Liu, X.; Yang, Z.; Shen, Z. HIF-1α overexpression in mesenchymal stem cell-derived exosomes mediates cardioprotection in myocardial infarction by enhanced angiogenesis. Stem Cell Res. Ther. 2020, 11, 373. [Google Scholar] [CrossRef] [PubMed]
  141. He, J.-G.; Li, H.-R.; Han, J.-X.; Li, B.-B.; Yan, D.; Li, H.-Y.; Wang, P.; Luo, Y. GATA-4-expressing mouse bone marrow mesenchymal stem cells improve cardiac function after myocardial infarction via secreted exosomes. Sci. Rep. 2018, 8, 9047. [Google Scholar] [CrossRef]
  142. Hao, C.; Lu, Z.; Zhao, Y.; Chen, Z.; Shen, C.; Ma, G.; Chen, L. Overexpression of GATA4 enhances the antiapoptotic effect of exosomes secreted from cardiac colony-forming unit fibroblasts via miRNA221-mediated targeting of the PTEN/PI3K/AKT signaling pathway. Stem Cell Res. Ther. 2020, 11, 251. [Google Scholar] [CrossRef] [PubMed]
  143. Yu, B.; Kim, H.W.; Gong, M.; Wang, J.; Millard, R.W.; Wang, Y.; Ashraf, M.; Xu, M. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. Int. J. Cardiol. 2015, 182, 349–360. [Google Scholar] [CrossRef]
  144. Li, X.; Lian, Y.; Wu, Y.; Ye, Z.; Feng, J.; Zhao, Y.; Guo, X.; Kang, J. Neonatal Plasma Exosomes Contribute to Endothelial Cell-Mediated Angiogenesis and Cardiac Repair after Acute Myocardial Infarction. Int. J. Mol. Sci. 2023, 24, 3196. [Google Scholar] [CrossRef] [PubMed]
  145. Cheng, H.; Chang, S.; Xu, R.; Chen, L.; Song, X.; Wu, J.; Qian, J.; Zou, Y.; Ma, J. Hypoxia-challenged MSC-derived exosomes deliver miR-210 to attenuate post-infarction cardiac apoptosis. Stem Cell Res. Ther. 2020, 11, 224. [Google Scholar] [CrossRef]
  146. Zhu, J.; Lu, K.; Zhang, N.; Zhao, Y.; Ma, Q.; Shen, J.; Lin, Y.; Xiang, P.; Tang, Y.; Hu, X.; et al. Myocardial reparative functions of exosomes from mesenchymal stem cells are enhanced by hypoxia treatment of the cells via transferring microRNA-210 in an nSMase2-dependent way. Artif. Cells Nanomed. Biotechnol. 2017, 46, 1659–1670. [Google Scholar] [CrossRef]
  147. Won Kim, H.; Haider, H.K.; Jiang, S.; Ashraf, M. Ischemic Preconditioning Augments Survival of Stem Cells via miR-210 Expression by Targeting Caspase-8-associated Protein 2. J. Biol. Chem. 2009, 284, 33161–33168. [Google Scholar] [CrossRef]
  148. Fasanaro, P.; D’Alessandra, Y.; Di Stefano, V.; Melchionna, R.; Romani, S.; Pompilio, G.; Capogrossi, M.C.; Martelli, F. MicroRNA-210 Modulates Endothelial Cell Response to Hypoxia and Inhibits the Receptor Tyrosine Kinase Ligand Ephrin-A3. J. Biol. Chem. 2008, 283, 15878–15883. [Google Scholar] [CrossRef]
  149. Wang, T.; Li, T.; Niu, X.; Hu, L.; Cheng, J.; Guo, D.; Ren, H.; Zhao, R.; Ji, Z.; Liu, P.; et al. ADSC-derived exosomes attenuate myocardial infarction injury by promoting miR-205-mediated cardiac angiogenesis. Biol. Direct. 2023, 18, 6. [Google Scholar] [CrossRef]
  150. 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]
  151. Del Campo, C.V.; Liaw, N.Y.; Gunadasa-Rohling, M.; Matthaei, M.; Braga, L.; Kennedy, T.; Salinas, G.; Voigt, N.; Giacca, M.; Zimmermann, W.-H.; et al. Regenerative potential of epicardium-derived extracellular vesicles mediated by conserved miRNA transfer. Cardiovasc. Res. 2022, 118, 597–611. [Google Scholar] [CrossRef]
  152. 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]
  153. 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]
  154. Chen, G.; Wang, M.; Ruan, Z.; Zhu, L.; Tang, C. Mesenchymal stem cell-derived exosomal miR-143-3p suppresses myocardial ischemia-reperfusion injury by regulating autophagy. Life Sci. 2021, 280, 119742. [Google Scholar] [CrossRef]
  155. Liu, J.; Jiang, M.; Deng, S.; Lu, J.; Huang, H.; Zhang, Y.; Gong, P.; Shen, X.; Ruan, H.; Jin, M.; et al. miR-93-5p-Containing Exosomes Treatment Attenuates Acute Myocardial Infarction-Induced Myocardial Damage. Mol. Ther.-Nucleic Acids 2018, 11, 103–115. [Google Scholar] [CrossRef]
  156. Luo, Q.; Guo, D.; Liu, G.; Chen, G.; Hang, M.; Jin, M. Exosomes from MiR-126-Overexpressing Adscs Are Therapeutic in Relieving Acute Myocardial Ischaemic Injury. Cell Physiol. Biochem. 2017, 44, 2105–2116. [Google Scholar] [CrossRef]
  157. Fu, D.L.; Jiang, H.; Li, C.Y.; Gao, T.; Liu, M.R.; Li, H.W. MicroRNA-338 in MSCs-derived exosomes inhibits cardiomyocyte apoptosis in myocardial infarction. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 10107–10117. [Google Scholar]
  158. Zheng, J.; Zhang, X.; Cai, W.; Yang, Y.; Guo, T.; Li, J.; Dai, H. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal microRNA-29b-3p Promotes Angiogenesis and Ventricular Remodeling in Rats with Myocardial Infarction by Targeting ADAMTS16. Cardiovasc. Toxicol. 2022, 22, 689–700. [Google Scholar] [CrossRef]
  159. Wang, X.; Zhu, Y.; Wu, C.; Liu, W.; He, Y.; Yang, Q. Adipose-Derived Mesenchymal Stem Cells-Derived Exosomes Carry MicroRNA-671 to Alleviate Myocardial Infarction through Inactivating the TGFBR2/Smad2 Axis. Inflammation 2021, 44, 1815–1830. [Google Scholar] [CrossRef]
  160. Zhang, C.S.; Shao, K.; Liu, C.W.; Li, J.C.; Yu, B.T. Hypoxic preconditioning BMSCs-exosomes inhibit cardiomyocyte apoptosis after acute myocardial infarction by upregulating microRNA-24. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6691–6699. [Google Scholar] [PubMed]
  161. Qiao, S.; Zhang, W.; Yin, Y.; Wei, Z.; Chen, F.; Zhao, J.; Sun, X.; Mu, D.; Xie, J.; Xu, B. Extracellular vesicles derived from Krüppel-Like Factor 2-overexpressing endothelial cells attenuate myocardial ischemia-reperfusion injury by preventing Ly6C high monocyte recruitment. Theranostics 2020, 10, 11562–11579. [Google Scholar] [CrossRef]
  162. Long, R.; Gao, L.; Li, Y.; Li, G.; Qin, P.; Wei, Z.; Li, D.; Qian, C.; Li, J.; Yang, G. M2 macrophage-derived exosomes carry miR-1271-5p to alleviate cardiac injury in acute myocardial infarction through down-regulating SOX6. Mol. Immunol. 2021, 136, 26–35. [Google Scholar] [CrossRef]
  163. Pu, L.; Kong, X.; Li, H.; He, X. Exosomes released from mesenchymal stem cells overexpressing microRNA-30e ameliorate heart failure in rats with myocardial infarction. Am. J. Transl. Res. 2021, 13, 4007–4025. [Google Scholar]
  164. 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]
  165. Zhu, W.; Wang, Q.; Zhang, J.; Sun, L.; Hong, X.; Du, W.; Duan, R.; Jiang, J.; Ji, Y.; Wang, H.; et al. Exosomes derived from mir-214-3p overexpressing mesenchymal stem cells promote myocardial repair. Biomater. Res. 2023, 27, 77. [Google Scholar] [CrossRef]
  166. Pan, J.; Alimujiang, M.; Chen, Q.; Shi, H.; Luo, X. Exosomes derived from miR-146a-modified adiposederived stem cells attenuate acute myocardial infarction− induced myocardial damage via downregulation of early growth response factor 1. J. Cell Biochem. 2018, 120, 4433–4443. [Google Scholar] [CrossRef] [PubMed]
  167. Wang, S.; Li, L.; Liu, T.; Jiang, W.; Hu, X. miR-19a/19b-loaded exosomes in combination with mesenchymal stem cell transplantation in a preclinical model of myocardial infarction. Regen. Med. 2020, 15, 1749–1759. [Google Scholar] [CrossRef] [PubMed]
  168. Huang, L.; Yang, L.; Ding, Y.; Jiang, X.; Xia, Z.; You, Z. Human umbilical cord mesenchymal stem cells-derived exosomes transfers microRNA-19a to protect cardiomyocytes from acute myocardial infarction by targeting SOX6. Cell Cycle 2020, 19, 339–353. [Google Scholar] [CrossRef]
  169. Wang, Y.; Ding, N.; Guan, G.; Liu, G.; Huo, D.; Li, Y.; Wei, K.; Yang, J.; Cheng, P.; Zhu, C. Rapid Delivery of Hsa-miR-590-3p Using Targeted Exosomes to Treat Acute Myocardial Infarction through Regulation of the Cell Cycle. J. Biomed. Nanotechnol. 2018, 14, 968–977. [Google Scholar] [CrossRef]
  170. Yang, M.; Liu, X.; Jiang, M.; Li, J.; Tang, Y.; Zhou, L. miR-543 in human mesenchymal stem cell–derived exosomes promotes cardiac microvascular endothelial cell angiogenesis after myocardial infarction through COL4A1. IUBMB Life 2021, 73, 927–940. [Google Scholar] [CrossRef] [PubMed]
  171. Youn, S.-W.; Li, Y.; Kim, Y.-M.; Sudhahar, V.; Abdelsaid, K.; Kim, H.; Liu, Y.; Fulton, D.; 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]
  172. Li, Y.; Yang, R.; Guo, B.; Zhang, H.; Zhang, H.; Liu, S.; Li, Y. Exosomal miR-301 derived from mesenchymal stem cells protects myocardial infarction by inhibiting myocardial autophagy. Biochem. Biophys. Res. Commun. 2019, 514, 323–328. [Google Scholar] [CrossRef]
  173. Mao, S.; Zhao, J.; Zhang, Z.-J.; Zhao, Q. MiR-183-5p overexpression in bone mesenchymal stem cell-derived exosomes protects against myocardial ischemia/reperfusion injury by targeting FOXO1. Immunobiology 2022, 227, 152204. [Google Scholar] [CrossRef]
  174. Chen, Y.; Zhao, Y.; Chen, W.; Xie, L.; Zhao, Z.-A.; Yang, J.; Chen, Y.; Lei, W.; Shen, Z. MicroRNA-133 overexpression promotes the therapeutic efficacy of mesenchymal stem cells on acute myocardial infarction. Stem Cell Res. Ther. 2017, 8, 268. [Google Scholar] [CrossRef]
  175. 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. In Review; 2021 Jan. Available online: https://www.researchsquare.com/article/rs-146749/v1 (accessed on 2 December 2023).
  176. Wang, Y.; Shen, Y. Exosomal miR-455-3p from BMMSCs prevents cardiac ischemia-reperfusion injury. Hum. Exp. Toxicol. 2022, 41, 096032712211025. [Google Scholar] [CrossRef] [PubMed]
  177. Sun, C.; Li, W.; Li, Y.; Chen, J.; An, H.; Zeng, G.; Wang, T.; Guo, Y.; Wang, C. MiR-182-5p Mediated by Exosomes Derived From Bone Marrow Mesenchymal Stem Cell Attenuates Inflammatory Responses by Targeting TLR4 in a Mouse Model of Myocardial Infraction. Immune Netw. 2022, 22, e49. [Google Scholar] [CrossRef]
  178. Yue, R.; Lu, S.; Luo, Y.; Zeng, J.; Liang, H.; Qin, D.; Wang, X.; Wang, T.; Pu, J.; Hu, H. Mesenchymal stem cell-derived exosomal microRNA-182-5p alleviates myocardial ischemia/reperfusion injury by targeting GSDMD in mice. Cell Death Discov. 2022, 8, 202. [Google Scholar] [CrossRef]
  179. Zhao, J.; Li, X.; Hu, J.; Chen, F.; Qiao, S.; Sun, X.; Gao, L.; Xie, J.; Xu, B. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovasc. Res. 2019, 115, 1205–1216. [Google Scholar] [CrossRef]
  180. Cao, C.; Wang, B.; Tang, J.; Zhao, J.; Guo, J.; Guo, Q.; Yue, X.; Zhang, Z.; Liu, G.; Zhang, H.; et al. Circulating exosomes repair endothelial cell damage by delivering miR-193a-5p. J. Cell. Mol. Med. 2021, 25, 2176–2189. [Google Scholar] [CrossRef]
  181. Sun, L.; Zhu, W.; Zhao, P.; Zhang, J.; Lu, Y.; Zhu, Y.; Zhao, W.; Liu, Y.; Chen, Q.; Zhang, F. Down-Regulated Exosomal MicroRNA-221—3p Derived From Senescent Mesenchymal Stem Cells Impairs Heart Repair. Front. Cell Dev. Biol. 2020, 8, 263. [Google Scholar] [CrossRef]
  182. Liang, C.; Liu, Y.; Xu, H.; Huang, J.; Shen, Y.; Chen, F.; Luo, M. Exosomes of Human Umbilical Cord MSCs Protect Against Hypoxia/Reoxygenation-Induced Pyroptosis of Cardiomyocytes via the miRNA-100-5p/FOXO3/NLRP3 Pathway. Front. Bioeng. Biotechnol. 2021, 8, 615850. [Google Scholar] [CrossRef] [PubMed]
  183. Li, H.; Wang, L.; Ma, T.; Liu, Z.; Gao, L. Exosomes secreted by endothelial cells derived from human induced pluripotent stem cells improve recovery from myocardial infarction in mice. Stem Cell Res. Ther. 2023, 14, 278. [Google Scholar] [CrossRef]
  184. Feng, Y.; Huang, W.; Wani, M.; Yu, X.; Ashraf, M. Ischemic Preconditioning Potentiates the Protective Effect of Stem Cells through Secretion of Exosomes by Targeting Mecp2 via miR-22. PLoS ONE 2014, 9, e88685. [Google Scholar] [CrossRef]
  185. Wu, Y.; Peng, W.; Fang, M.; Wu, M.; Wu, M. MSCs-Derived Extracellular Vesicles Carrying miR-212-5p Alleviate Myocardial Infarction-Induced Cardiac Fibrosis via NLRC5/VEGF/TGF-β1/SMAD Axis. J. Cardiovasc. Trans. Res. 2022, 15, 302–316. [Google Scholar] [CrossRef] [PubMed]
  186. Wan, J.; Lin, S.; Yu, Z.; Song, Z.; Lin, X.; Xu, R.; Du, S. Protective Effects of MicroRNA-200b-3p Encapsulated by Mesenchymal Stem Cells–Secreted Extracellular Vesicles in Myocardial Infarction Via Regulating BCL2L11. J. Am. Heart Assoc. 2022, 11, e024330. [Google Scholar] [CrossRef] [PubMed]
  187. Gao, T.; Fan, H.; Wang, J.; Wang, R. Extracellular vesicles derived from human umbilical cord mesenchymal stem cells stimulate angiogenesis in myocardial infarction via the microRNA-423-5p/EFNA3 axis. PWKI 2022, 18, 373–391. [Google Scholar] [CrossRef]
  188. Yuan, W.; Liang, X.; Liu, Y.; Wang, H. Mechanism of miR-378a-3p enriched in M2 macrophage-derived extracellular vesicles in cardiomyocyte pyroptosis after MI. Hypertens. Res. 2022, 45, 650–664. [Google Scholar] [CrossRef]
  189. Ou, H.; Teng, H.; Qin, Y.; Luo, X.; Yang, P.; Zhang, W.; Chen, W.; Lv, D.; Tang, H. Extracellular vesicles derived from microRNA-150-5p-overexpressing mesenchymal stem cells protect rat hearts against ischemia/reperfusion. Aging 2020, 12, 12669–12683. [Google Scholar] [CrossRef]
  190. Yu, S.; Tang, X.; Zheng, T.; Li, S.; Ren, H.; Wu, H.; Peng, F.; Gong, L. Plasma-derived extracellular vesicles transfer microRNA-130a-3p to alleviate myocardial ischemia/reperfusion injury by targeting ATG16L1. Cell Tissue Res. 2022, 389, 99–114. [Google Scholar] [CrossRef] [PubMed]
  191. Mao, C.-Y.; Zhang, T.-T.; Li, D.-J.; Zhou, E.; Fan, Y.-Q.; He, Q.; Wang, C.-Q.; Zhang, J.-F. Extracellular vesicles from hypoxia-preconditioned mesenchymal stem cells alleviates myocardial injury by targeting thioredoxin-interacting protein-mediated hypoxia-inducible factor-1α pathway. WJSC 2022, 14, 183–199. [Google Scholar] [CrossRef]
  192. Jung, J.-H.; Ikeda, G.; Tada, Y.; Von Bornstädt, D.; Santoso, M.R.; Wahlquist, C.; Rhee, S.; Jeon, Y.-J.; Yu, A.C.; O’brien, C.G.; et al. miR-106a–363 cluster in extracellular vesicles promotes endogenous myocardial repair via Notch3 pathway in ischemic heart injury. Basic Res. Cardiol. 2021, 116, 19. [Google Scholar] [CrossRef]
  193. Sánchez-Sánchez, R.; Gómez-Ferrer, M.; Reinal, I.; Buigues, M.; Villanueva-Bádenas, E.; Ontoria-Oviedo, I.; Hernándiz, A.; González-King, H.; Peiró-Molina, E.; Dorronsoro, A.; et al. miR-4732-3p in Extracellular Vesicles From Mesenchymal Stromal Cells Is Cardioprotective During Myocardial Ischemia. Front. Cell Dev. Biol. 2021, 9, 734143. [Google Scholar] [CrossRef] [PubMed]
  194. Khan, M.; Nickoloff, E.; Abramova, T.; Johnson, J.; Verma, S.K.; Krishnamurthy, P.; Mackie, A.R.; Vaughan, E.; Garikipati, V.N.S.; 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]
  195. 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]
  196. Shyu, K.; Wang, B.; Fang, W.; Pan, C.; Lin, C. Hyperbaric oxygen-induced long non-coding RNA MALAT1 exosomes suppress MicroRNA-92a expression in a rat model of acute myocardial infarction. J. Cell. Mol. Med. 2020, 24, 12945–12954. [Google Scholar] [CrossRef] [PubMed]
  197. Wu, Q.; Wang, J.; Tan, W.L.W.; Jiang, Y.; Wang, S.; Li, Q.; Yu, X.; Tan, J.; Liu, S.; Zhang, P.; et al. Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis. Cell Death Dis. 2020, 11, 354. [Google Scholar] [CrossRef]
  198. 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]
  199. 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]
  200. Sun, L.; Zhu, W.; Zhao, P.; Wang, Q.; Fan, B.; Zhu, Y.; Lu, Y.; Chen, Q.; Zhang, J.; Zhang, F. Long noncoding RNA UCA1 from hypoxia-conditioned hMSC-derived exosomes: A novel molecular target for cardioprotection through miR-873-5p/XIAP axis. Cell Death Dis. 2020, 11, 696. [Google Scholar] [CrossRef]
  201. Zhu, D.; Liu, S.; Huang, K.; Li, J.; Xuan, M.; Li, Z.; Cheng, K. Intrapericardial long non-coding RNA-Tcf21 antisense RNA inducing demethylation administration promotes cardiac repair. Eur. Heart J. 2023, 44, 1748–1760. [Google Scholar] [CrossRef]
  202. 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]
  203. 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]
  204. Zhou, D.; Dai, Z.; Ren, M.; Yang, M. Adipose-Derived Stem Cells-Derived Exosomes with High Amounts of Circ_0001747 Alleviate Hypoxia/Reoxygenation-Induced Injury in Myocardial Cells by Targeting MiR-199b-3p/MCL1 Axis. Int. Heart J. 2022, 63, 356–366. [Google Scholar] [CrossRef] [PubMed]
  205. Wei, G.; Li, C.; Jia, X.; Xie, J.; Tang, Z.; Jin, M.; Chen, Q.; Sun, Y.; He, S.; Li, X.; et al. Extracellular vesicle-derived CircWhsc1 promotes cardiomyocyte proliferation and heart repair by activating TRIM59/STAT3/Cyclin B2 pathway. J. Adv. Res. 2023, 53, 199–218. [Google Scholar] [CrossRef] [PubMed]
  206. Yu, L.; Liang, Y.; Zhang, M.; Yang, P.C.; Hinek, A.; Mao, S. Extracellular vesicle-derived circCEBPZOS attenuates postmyocardial infarction remodeling by promoting angiogenesis via the miR-1178-3p/PDPK1 axis. Commun. Biol. 2023, 6, 133. [Google Scholar] [CrossRef] [PubMed]
  207. Elliott, P.; Andersson, B.; Arbustini, E.; Bilinska, Z.; Cecchi, F.; Charron, P.; Dubourg, O.; Kuhl, U.; Maisch, B.; McKenna, W.J.; et al. Classification of the cardiomyopathies: A position statement from the european society of cardiology working group on myocardial and pericardial diseases. Eur. Heart J. 2007, 29, 270–276. [Google Scholar] [CrossRef] [PubMed]
  208. Richardson, P.; McKenna, W.; Bristow, M.; Maisch, B.; Mautner, B.; O’Connell, J.; Olsen, E.; Thiene, G.; Goodwin, J.; Gyarfas, I.; et al. Report of the 1995 World Health Organization/International Society and Federation of Cardiology Task Force on the Definition and Classification of Cardiomyopathies. Circulation 1996, 93, 841–842. [Google Scholar] [CrossRef]
  209. Jenni, R. Echocardiographic and pathoanatomical characteristics of isolated left ventricular non-compaction: A step towards classification as a distinct cardiomyopathy. Heart 2001, 86, 666–671. [Google Scholar] [CrossRef]
  210. Maron, B.J.; Towbin, J.A.; Thiene, G.; Antzelevitch, C.; Corrado, D.; Arnett, D.; Moss, A.J.; Seidman, C.E.; Young, J.B. Contemporary Definitions and Classification of the Cardiomyopathies: An American Heart Association Scientific Statement From the Council on Clinical Cardiology, Heart Failure and Transplantation Committee; Quality of Care and Outcomes Research and Functional Genomics and Translational Biology Interdisciplinary Working Groups; and Council on Epidemiology and Prevention. Circulation 2006, 113, 1807–1816. [Google Scholar] [CrossRef]
  211. Semsarian, C.; Ingles, J.; Maron, M.S.; Maron, B.J. New Perspectives on the Prevalence of Hypertrophic Cardiomyopathy. J. Am. Coll. Cardiol. 2015, 65, 1249–1254. [Google Scholar] [CrossRef]
  212. Medical Masterclass contributors; Firth, J. Cardiology: Hypertrophic cardiomyopathy. Clin. Med. 2019, 19, 61–63. [Google Scholar] [CrossRef]
  213. Halliday, B.P.; Prasad, S.K. The Interstitium in the Hypertrophied Heart. JACC Cardiovasc. Imaging 2019, 12, 2357–2368. [Google Scholar] [CrossRef] [PubMed]
  214. Maron, B.J.; Maron, M.S. Hypertrophic cardiomyopathy. Lancet 2013, 381, 242–255. [Google Scholar] [CrossRef] [PubMed]
  215. James, V.; Nizamudeen, Z.A.; Lea, D.; Dottorini, T.; Holmes, T.L.; Johnson, B.B.; Arkill, K.P.; Denning, C.; Smith, J.G.W. Transcriptomic Analysis of Cardiomyocyte Extracellular Vesicles in Hypertrophic Cardiomyopathy Reveals Differential snoRNA Cargo. Stem Cells Dev. 2021, 30, 1215–1227. [Google Scholar] [CrossRef] [PubMed]
  216. Smith, J.G.W.; Owen, T.; Bhagwan, J.R.; Mosqueira, D.; Scott, E.; Mannhardt, I.; Patel, A.; Barriales-Villa, R.; Monserrat, L.; Hansen, A.; et al. Isogenic Pairs of hiPSC-CMs with Hypertrophic Cardiomyopathy/LVNC-Associated ACTC1 E99K Mutation Unveil Differential Functional Deficits. Stem Cell Rep. 2018, 11, 1226–1243. [Google Scholar] [CrossRef]
  217. Tian, C.; Gao, L.; Zimmerman, M.C.; Zucker, I.H. Myocardial infarction-induced microRNA-enriched exosomes contribute to cardiac Nrf2 dysregulation in chronic heart failure. Am. J. Physiol.-Heart Circ. Physiol. 2018, 314, H928–H939. [Google Scholar] [CrossRef]
  218. Matkovich, S.J.; Van Booven, D.J.; Youker, K.A.; Torre-Amione, G.; Diwan, A.; Eschenbacher, W.H.; Dorn, L.E.; Watson, M.A.; Margulies, K.B.; Dorn, G.W. Reciprocal Regulation of Myocardial microRNAs and Messenger RNA in Human Cardiomyopathy and Reversal of the microRNA Signature by Biomechanical Support. Circulation 2009, 119, 1263–1271. [Google Scholar] [CrossRef]
  219. Kuosmanen, S.M.; Hartikainen, J.; Hippeläinen, M.; Kokki, H.; Levonen, A.-L.; Tavi, P. MicroRNA Profiling of Pericardial Fluid Samples from Patients with Heart Failure. PLoS ONE 2015, 10, e0119646. [Google Scholar] [CrossRef]
  220. Ovchinnikova, E.S.; Schmitter, D.; Vegter, E.L.; Ter Maaten, J.M.; Valente, M.A.E.; Liu, L.C.Y.; Van Der Harst, P.; Pinto, Y.M.; De Boer, R.A.; Meyer, S.; et al. Signature of circulating microRNAs in patients with acute heart failure. Eur. J. Heart Fail. 2016, 18, 414–423. [Google Scholar] [CrossRef]
  221. Tian, C.; Hu, G.; Gao, L.; Hackfort, B.T.; Zucker, I.H. Extracellular vesicular MicroRNA-27a* contributes to cardiac hypertrophy in chronic heart failure. J. Mol. Cell. Cardiol. 2020, 143, 120–131. [Google Scholar] [CrossRef] [PubMed]
  222. Huang, F.; Na, N.; Ijichi, T.; Wu, X.; Miyamoto, K.; Ciullo, A.; Tran, M.; Li, L.; Ibrahim, A.; Marbán, E.; et al. Exosomally derived Y RNA fragment alleviates hypertrophic cardiomyopathy in transgenic mice. Mol. Ther.-Nucleic Acids 2021, 24, 951–960. [Google Scholar] [CrossRef]
  223. Zhang, L.; Zhang, G.; Lu, Y.; Gao, J.; Qin, Z.; Xu, S.; Wang, Z.; Xu, Y.; Yang, Y.; Zhang, J.; et al. Differential expression profiles of plasma exosomal microRNAs in dilated cardiomyopathy with chronic heart failure. J. Cell. Mol. Med. 2023, 27, 1988–2003. [Google Scholar] [CrossRef]
  224. Cohn, J.N.; Bristow, M.R.; Chien, K.R.; Colucci, W.S.; Frazier, O.H.; Leinwand, L.A.; Lorell, B.H.; Moss, A.J.; Sonnenblick, E.H.; Walsh, R.A.; et al. Report of the National Heart, Lung, and Blood Institute Special Emphasis Panel on Heart Failure Research. Circulation 1997, 95, 766–770. [Google Scholar] [CrossRef]
  225. Masarone, D.; Kaski, J.P.; Pacileo, G.; Elliott, P.M.; Bossone, E.; Day, S.M.; Limongelli, G. Epidemiology and Clinical Aspects of Genetic Cardiomyopathies. Heart Fail. Clin. 2018, 14, 119–128. [Google Scholar] [CrossRef]
  226. Sweet, M.E.; Taylor, M.R.; Mestroni, L. Diagnosis, prevalence, and screening of familial dilated cardiomyopathy. Expert Opin. Orphan Drugs 2015, 3, 869–876. [Google Scholar] [CrossRef]
  227. Harakalova, M.; Kummeling, G.; Sammani, A.; Linschoten, M.; Baas, A.F.; Van Der Smagt, J.; Doevendans, P.A.; Van Tintelen, J.P.; Dooijes, D.; Mokry, M.; et al. A systematic analysis of genetic dilated cardiomyopathy reveals numerous ubiquitously expressed and muscle-specific genes. Eur. J. Heart Fail. 2015, 17, 484–493. [Google Scholar] [CrossRef]
  228. Bursa, F.; Yellowlees, A.; Bishop, A.; Beckett, A.; Hallis, B.; Matheson, M. Estimation of ELISA results using a parallel curve analysis. J. Immunol. Methods 2020, 486, 112836. [Google Scholar] [CrossRef]
  229. Lin, R.; Rahtu-Korpela, L.; Szabo, Z.; Kemppi, A.; Skarp, S.; Kiviniemi, A.M.; Lepojärvi, E.S.; Halmetoja, E.; Kilpiö, T.; Porvari, K.; et al. MiR-185-5p regulates the development of myocardial fibrosis. J. Mol. Cell. Cardiol. 2022, 165, 130–140. [Google Scholar] [CrossRef] [PubMed]
  230. Saincher, R.; Pentapati, K.; Gadicherla, S. Effect of audio-visual treatment information on hemodynamic parameters during the transalveolar extraction of mandibular third molars: A randomized clinical trial. J. Int. Soc. Prev. Communit. Dent. 2019, 9, 21. [Google Scholar] [CrossRef]
  231. Zahid, M.; Rinas, U. Guidelines for Small-Scale Production and Purification of Hepatitis B Surface Antigen Virus-Like Particles from Recombinant Pichia pastoris. In Recombinant Protein Production in Yeast; Gasser, B., Mattanovich, D., Eds.; Methods in Molecular Biology; Springer: New York, NY, USA, 2019; Volume 1923, pp. 309–322. ISBN 978-1-4939-9023-8. [Google Scholar]
  232. Sigterman, T.A.; Bolt, L.J.J.; Snoeijs, M.G.; Krasznai, A.G.; Heijboer, R.; Schurink, G.-W.H.; Bouwman, L.H. Radiation Exposure during Percutaneous Transluminal Angioplasty for Symptomatic Peripheral Arterial Disease. Ann. Vasc. Surg. 2016, 33, 167–172. [Google Scholar] [CrossRef] [PubMed]
  233. Linardi, D.; Walpoth, B.; Mani, R.; Murari, A.; Tessari, M.; Hoxha, S.; Anderloni, M.; Decimo, I.; Dolci, S.; Nicolato, E.; et al. Slow versus fast rewarming after hypothermic circulatory arrest: Effects on neuroinflammation and cerebral oedema. Eur. J. Cardio-Thorac. Surg. 2020, 58, 792–800. [Google Scholar] [CrossRef] [PubMed]
  234. Muranishi, Y.; Tanaka, N.; Kono, T.; Miyahara, R. A case of intrapulmonary lymphangioma surrounded by pulmonary hilar structures. Respir. Investig. 2020, 58, 506–509. [Google Scholar] [CrossRef] [PubMed]
  235. Dover, E.N.; Beck, R.; Huang, M.C.; Douillet, C.; Wang, Z.; Klett, E.L.; Stýblo, M. Arsenite and methylarsonite inhibit mitochondrial metabolism and glucose-stimulated insulin secretion in INS-1 832/13 β cells. Arch. Toxicol. 2018, 92, 693–704. [Google Scholar] [CrossRef] [PubMed]
  236. Kohanski, M.A.; Palmer, J.N.; Adappa, N.D. Indications and endonasal treatment of petrous apex cholesterol granulomas. Curr. Opin. Otolaryngol. Head Neck Surg. 2019, 27, 54–58. [Google Scholar] [CrossRef]
  237. Roura, S.; Gámez-Valero, A.; Lupón, J.; Gálvez-Montón, C.; Borràs, F.E.; Bayes-Genis, A. Proteomic signature of circulating extracellular vesicles in dilated cardiomyopathy. Lab. Investig. 2018, 98, 1291–1299. [Google Scholar] [CrossRef]
  238. Lowe, G.D. Fibrinogen Assays for Cardiovascular Risk Assessment. Clin. Chem. 2010, 56, 693–695. [Google Scholar] [CrossRef] [PubMed]
  239. Danesh, J.; Lewington, S.; Thompson, S.G.; Lowe, G.D.O.; Collins, R.; Kostis, J.B.; Wilson, A.C.; Folsom, A.R.; Wu, K.; Benderly, M.; et al. Plasma fibrinogen level and the risk of major cardiovascular diseases and nonvascular mortality: An individual participant meta-analysis. JAMA 2005, 294, 1799–1809. [Google Scholar] [CrossRef] [PubMed]
  240. The Emerging Risk Factors Collaboration. C-Reactive Protein, Fibrinogen, and Cardiovascular Disease Prediction. N. Engl. J. Med. 2012, 367, 1310–1320. [Google Scholar] [CrossRef]
  241. Appiah, D.; Schreiner, P.J.; MacLehose, R.F.; Folsom, A.R. Association of Plasma γ′ Fibrinogen with Incident Cardiovascular Disease: The Atherosclerosis Risk in Communities (ARIC) Study. ATVB 2015, 35, 2700–2706. [Google Scholar] [CrossRef]
  242. Becatti, M.; Marcucci, R.; Bruschi, G.; Taddei, N.; Bani, D.; Gori, A.M.; Giusti, B.; Gensini, G.F.; Abbate, R.; Fiorillo, C. Oxidative Modification of Fibrinogen Is Associated with Altered Function and Structure in the Subacute Phase of Myocardial Infarction. ATVB 2014, 34, 1355–1361. [Google Scholar] [CrossRef]
  243. Klip, I.T.; Comin-Colet, J.; Voors, A.A.; Ponikowski, P.; Enjuanes, C.; Banasiak, W.; Lok, D.J.; Rosentryt, P.; Torrens, A.; Polonski, L.; et al. Iron deficiency in chronic heart failure: An international pooled analysis. Am. Heart J. 2013, 165, 575–582.e3. [Google Scholar] [CrossRef]
  244. Comín-Colet, J.; Ruiz, S.; Cladellas, M.; Rizzo, M.; Torres, A.; Bruguera, J. A Pilot Evaluation of the Long-term Effect of Combined Therapy with Intravenous Iron Sucrose and Erythropoietin in Elderly Patients with Advanced Chronic Heart Failure and Cardio-Renal Anemia Syndrome: Influence on Neurohormonal Activation and Clinical Outcomes. J. Card. Fail. 2009, 15, 727–735. [Google Scholar] [CrossRef]
  245. Jankowska, E.A.; Rozentryt, P.; Witkowska, A.; Nowak, J.; Hartmann, O.; Ponikowska, B.; Borodulin-Nadzieja, L.; Von Haehling, S.; Doehner, W.; Banasiak, W.; et al. Iron Deficiency Predicts Impaired Exercise Capacity in Patients with Systolic Chronic Heart Failure. J. Card. Fail. 2011, 17, 899–906. [Google Scholar] [CrossRef]
  246. Enjuanes, C.; Klip, I.T.; Bruguera, J.; Cladellas, M.; Ponikowski, P.; Banasiak, W.; Van Veldhuisen, D.J.; Van Der Meer, P.; Jankowska, E.A.; Comín-Colet, J. Iron deficiency and health-related quality of life in chronic heart failure: Results from a multicenter European study. Int. J. Cardiol. 2014, 174, 268–275. [Google Scholar] [CrossRef]
  247. Van Veldhuisen, D.J.; Anker, S.D.; Ponikowski, P.; Macdougall, I.C. Anemia and iron deficiency in heart failure: Mechanisms and therapeutic approaches. Nat Rev Cardiol 2011, 8, 485–493. [Google Scholar] [CrossRef] [PubMed]
  248. Gettins, P.G.W. Serpin Structure, Mechanism, and Function. Chem. Rev. 2002, 102, 4751–4804. [Google Scholar] [CrossRef] [PubMed]
  249. Izquierdo, I.; Rosa, I.; Bravo, S.B.; Guitián, E.; Pérez-Serra, A.; Campuzano, O.; Brugada, R.; Mangas, A.; García, Á.; Toro, R. Proteomic identification of putative biomarkers for early detection of sudden cardiac death in a family with a LMNA gene mutation causing dilated cardiomyopathy. J. Proteom. 2016, 148, 75–84. [Google Scholar] [CrossRef] [PubMed]
  250. Wu, T.; Chen, Y.; Du, Y.; Tao, J.; Zhou, Z.; Yang, Z. Serum Exosomal MiR-92b-5p as a Potential Biomarker for Acute Heart Failure Caused by Dilated Cardiomyopathy. Cell Physiol. Biochem. 2018, 46, 1939–1950. [Google Scholar] [CrossRef] [PubMed]
  251. Fu, X.; Mishra, R.; Chen, L.; Arfat, M.Y.; Sharma, S.; Kingsbury, T.; Gunasekaran, M.; Saha, P.; Hong, C.; Yang, P.; et al. Exosomes mediated fibrogenesis in dilated cardiomyopathy through a MicroRNA pathway. iScience 2023, 26, 105963. [Google Scholar] [CrossRef] [PubMed]
  252. Vandergriff, A.; Huang, K.; Shen, D.; Hu, S.; Hensley, M.T.; Caranasos, T.G.; Qian, L.; Cheng, K. Targeting regenerative exosomes to myocardial infarction using cardiac homing peptide. Theranostics 2018, 8, 1869–1878. [Google Scholar] [CrossRef]
  253. Sun, X.; Shan, A.; Wei, Z.; Xu, B. Intravenous mesenchymal stem cell-derived exosomes ameliorate myocardial inflammation in the dilated cardiomyopathy. Biochem. Biophys. Res. Commun. 2018, 503, 2611–2618. [Google Scholar] [CrossRef]
  254. Ni, J.; Liu, Y.; Wang, K.; Wu, M.; Kang, L.; Sha, D.; Xu, B.; Gu, R. Trophoblast Stem-Cell-Derived Exosomes Improve Doxorubicin-Induced Dilated Cardiomyopathy by Modulating the let-7i/YAP Pathway. Mol. Ther.-Nucleic Acids 2020, 22, 948–956. [Google Scholar] [CrossRef]
  255. Zhang, W.; Chen, Z.; Qiao, S.; Chen, S.; Zheng, H.; Wei, X.; Li, Q.; Xu, B.; Huang, W. The effects of extracellular vesicles derived from Krüppel-Like Factor 2 overexpressing endothelial cells on the regulation of cardiac inflammation in the dilated cardiomyopathy. J. Nanobiotechnol. 2022, 20, 76. [Google Scholar] [CrossRef]
  256. World Health Organization G. The top 10 Causes of Death [Updated 201]. WHO. 2020. Available online: https://www.who.int/news-room/fact-sheets/detail/the-top-10-causes-of-death (accessed on 8 December 2023).
  257. Kannel, W.B. Diabetes and Cardiovascular Disease: The Framingham Study. JAMA 1979, 241, 2035. [Google Scholar] [CrossRef] [PubMed]
  258. Jia, G.; Whaley-Connell, A.; Sowers, J.R. Diabetic cardiomyopathy: A hyperglycaemia- and insulin-resistance-induced heart disease. Diabetologia 2018, 61, 21–28. [Google Scholar] [CrossRef] [PubMed]
  259. Seferović, P.M.; Paulus, W.J. Clinical diabetic cardiomyopathy: A two-faced disease with restrictive and dilated phenotypes. Eur. Heart J. 2015, 36, 1718–1727. [Google Scholar] [CrossRef] [PubMed]
  260. Wang, L.; Cai, Y.; Jian, L.; Cheung, C.W.; Zhang, L.; Xia, Z. Impact of peroxisome proliferator-activated receptor-α on diabetic cardiomyopathy. Cardiovasc. Diabetol. 2021, 20, 2. [Google Scholar] [CrossRef] [PubMed]
  261. De Gonzalo-Calvo, D.; Van Der Meer, R.W.; Rijzewijk, L.J.; Smit, J.W.A.; Revuelta-Lopez, E.; Nasarre, L.; Escola-Gil, J.C.; Lamb, H.J.; Llorente-Cortes, V. Serum microRNA-1 and microRNA-133a levels reflect myocardial steatosis in uncomplicated type 2 diabetes. Sci. Rep. 2017, 7, 47. [Google Scholar] [CrossRef] [PubMed]
  262. Huang, J.-P.; Chang, C.-C.; Kuo, C.-Y.; Huang, K.-J.; Sokal, E.M.; Chen, K.-H.; Hung, L.-M. Exosomal microRNAs miR-30d-5p and miR-126a-5p Are Associated with Heart Failure with Preserved Ejection Fraction in STZ-Induced Type 1 Diabetic Rats. Int. J. Mol. Sci. 2022, 23, 7514. [Google Scholar] [CrossRef] [PubMed]
  263. Watson, C.J.; Gupta, S.K.; O’Connell, E.; Thum, S.; Glezeva, N.; Fendrich, J.; Gallagher, J.; Ledwidge, M.; Grote-Levi, L.; McDonald, K.; et al. MicroRNA signatures differentiate preserved from reduced ejection fraction heart failure. Eur. J. Heart Fail. 2015, 17, 405–415. [Google Scholar] [CrossRef] [PubMed]
  264. Vegter, E.L.; Van Der Meer, P.; De Windt, L.J.; Pinto, Y.M.; Voors, A.A. MicroRNAs in heart failure: From biomarker to target for therapy. Eur. J Heart Fail. 2016, 18, 457–468. [Google Scholar] [CrossRef]
  265. Li, J.; Salvador, A.M.; Li, G.; Valkov, N.; Ziegler, O.; Yeri, A.; Yang Xiao, C.; Meechoovet, B.; Alsop, E.; Rodosthenous, R.S.; et al. Mir-30d Regulates Cardiac Remodeling by Intracellular and Paracrine Signaling. Circ. Res. 2021, 128, E1–E23. [Google Scholar] [CrossRef] [PubMed]
  266. Potus, F.; Ruffenach, G.; Dahou, A.; Thebault, C.; Breuils-Bonnet, S.; Tremblay, È.; Nadeau, V.; Paradis, R.; Graydon, C.; Wong, R.; et al. Downregulation of MicroRNA-126 Contributes to the Failing Right Ventricle in Pulmonary Arterial Hypertension. Circulation 2015, 132, 932–943. [Google Scholar] [CrossRef]
  267. Hu, J.; Wang, S.; Xiong, Z.; Cheng, Z.; Yang, Z.; Lin, J.; Wang, T.; Feng, X.; Gao, E.; Wang, H.; et al. Exosomal Mst1 transfer from cardiac microvascular endothelial cells to cardiomyocytes deteriorates diabetic cardiomyopathy. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2018, 1864, 3639–3649. [Google Scholar] [CrossRef] [PubMed]
  268. Zhang, Y.; Zhu, Z.; Wang, T.; Dong, Y.; Fan, Y.; Sun, D. TGF-β1-containing exosomes from cardiac microvascular endothelial cells mediate cardiac fibroblast activation under high glucose conditions. Biochem. Cell Biol. 2021, 99, 693–699. [Google Scholar] [CrossRef]
  269. Richter, K.; Haslbeck, M.; Buchner, J. The Heat Shock Response: Life on the Verge of Death. Mol. Cell 2010, 40, 253–266. [Google Scholar] [CrossRef]
  270. Wang, X.; Gu, H.; Huang, W.; Peng, J.; Li, Y.; Yang, L.; Qin, D.; Essandoh, K.; Wang, Y.; Peng, T.; et al. Hsp20-Mediated Activation of Exosome Biogenesis in Cardiomyocytes Improves Cardiac Function and Angiogenesis in Diabetic Mice. Diabetes 2016, 65, 3111–3128. [Google Scholar] [CrossRef]
  271. Lin, Y.; Zhang, F.; Lian, X.-F.; Peng, W.-Q.; Yin, C.-Y. Mesenchymal stem cell-derived exosomes improve diabetes mellitus-induced myocardial injury and fibrosis via inhibition of TGF-β1/Smad2 signaling pathway. Cell Mol. Biol. (Noisy-le-grand) 2019, 65, 123–126. [Google Scholar] [CrossRef]
  272. Singla, R.; Garner, K.H.; Samsam, M.; Cheng, Z.; Singla, D.K. Exosomes derived from cardiac parasympathetic ganglionic neurons inhibit apoptosis in hyperglycemic cardiomyoblasts. Mol. Cell Biochem. 2019, 462, 1–10. [Google Scholar] [CrossRef]
  273. Zhen, J.; Bai, J.; Liu, J.; Men, H.; Yu, H. Ginsenoside RG1-induced mesenchymal stem cells alleviate diabetic cardiomyopathy through secreting exosomal circNOTCH1 to promote macrophage M2 polarization. Phytother. Res. 2023. [Google Scholar] [CrossRef] [PubMed]
  274. Wijesurendra, R.S.; Casadei, B. Mechanisms of atrial fibrillation. Heart 2019, 105, 1860–1867. [Google Scholar] [CrossRef] [PubMed]
  275. Kornej, J.; Börschel, C.S.; Benjamin, E.J.; Schnabel, R.B. Epidemiology of Atrial Fibrillation in the 21st Century: Novel Methods and New Insights. Circ. Res. 2020, 127, 4–20. [Google Scholar] [CrossRef]
  276. Sagris, M.; Vardas, E.P.; Theofilis, P.; Antonopoulos, A.S.; Oikonomou, E.; Tousoulis, D. Atrial Fibrillation: Pathogenesis, Predisposing Factors, and Genetics. Int. J. Mol. Sci. 2021, 23, 6. [Google Scholar] [CrossRef]
  277. Elliott, A.D.; Middeldorp, M.E.; Van Gelder, I.C.; Albert, C.M.; Sanders, P. Epidemiology and modifiable risk factors for atrial fibrillation. Nat. Rev. Cardiol. 2023, 20, 404–417. [Google Scholar] [CrossRef]
  278. Thulin, Å.; Lindbäck, J.; Granger, C.B.; Wallentin, L.; Lind, L.; Siegbahn, A. Extracellular vesicles in atrial fibrillation and stroke. Thromb. Res. 2020, 193, 180–189. [Google Scholar] [CrossRef]
  279. Mørk, M.; Andreasen, J.J.; Rasmussen, L.H.; Lip, G.Y.H.; Pedersen, S.; Bæk, R.; Jørgensen, M.M.; Kristensen, S.R. Elevated blood plasma levels of tissue factor-bearing extracellular vesicles in patients with atrial fibrillation. Thromb. Res. 2019, 173, 141–150. [Google Scholar] [CrossRef]
  280. Siwaponanan, P.; Kaewkumdee, P.; Phromawan, W.; Udompunturak, S.; Chomanee, N.; Udol, K.; Pattanapanyasat, K.; Krittayaphong, R. Increased expression of six-large extracellular vesicle-derived miRNAs signature for nonvalvular atrial fibrillation. J. Transl. Med. 2022, 20, 4. [Google Scholar] [CrossRef]
  281. 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]
  282. Wang, S.; Min, J.; Yu, Y.; Yin, L.; Wang, Q.; Shen, H.; Yang, J.; Zhang, P.; Xiao, J.; Wang, Z. Differentially expressed miRNAs in circulating exosomes between atrial fibrillation and sinus rhythm. J. Thorac. Dis. 2019, 11, 4337–4348. [Google Scholar] [CrossRef]
  283. Zhu, P.; Li, H.; Zhang, A.; Li, Z.; Zhang, Y.; Ren, M.; Zhang, Y.; Hou, Y. MicroRNAs sequencing of plasma exosomes derived from patients with atrial fibrillation: miR-124-3p promotes cardiac fibroblast activation and proliferation by regulating AXIN1. J. Physiol. Biochem. 2022, 78, 85–98. [Google Scholar] [CrossRef]
  284. Wei, Z.; Bing, Z.; Shaohuan, Q.; Yanran, W.; Shuo, S.; Bi, T.; Feiyu, Z.; Heng, Z.; Qin, G.; Pinfang, K. Expression of miRNAs in plasma exosomes derived from patients with atrial fibrillation. Clin. Cardiol. 2020, 43, 1450–1459. [Google Scholar] [CrossRef] [PubMed]
  285. Mun, D.; Kim, H.; Kang, J.-Y.; Park, H.; Park, H.; Lee, S.-H.; Yun, N.; Joung, B. Expression of miRNAs in circulating exosomes derived from patients with persistent atrial fibrillation. FASEB J. 2019, 33, 5979–5989. [Google Scholar] [CrossRef]
  286. Xie, Y.; Zhou, L.; Yan, M.; Zhao, W.; Hu, Z. Differentiation of Atrial Fibrillation and Atrial Fibrillation-Associated Ischemic Stroke Based on Serum Exosome miRNA-Seq. Cardiology 2023, 148, 150–160. [Google Scholar] [CrossRef]
  287. Zhao, L.; Ma, Z.; Guo, Z.; Zheng, M.; Li, K.; Yang, X. Analysis of long non-coding RNA and mRNA profiles in epicardial adipose tissue of patients with atrial fibrillation. Biomed. Pharmacother. 2020, 121, 109634. [Google Scholar] [CrossRef]
  288. Kang, J.-Y.; Mun, D.; Kim, H.; Yun, N.; Joung, B. Serum exosomal long noncoding RNAs as a diagnostic biomarker for atrial fibrillation. Heart Rhythm. 2022, 19, 1450–1458. [Google Scholar] [CrossRef]
  289. Zheng, H.; Peng, Y.; Wang, P.; Su, P.; Zhao, L. The integrative network of circRNA, miRNA and mRNA of epicardial adipose tissue in patients with atrial fibrillation. Am. J. Transl. Res. 2022, 14, 6550–6562. [Google Scholar]
  290. Weiss, L.; Keaney, J.; Szklanna, P.B.; Prendiville, T.; Uhrig, W.; Wynne, K.; Kelliher, S.; Ewins, K.; Comer, S.P.; Egan, K.; et al. Nonvalvular atrial fibrillation patients anticoagulated with rivaroxaban compared with warfarin exhibit reduced circulating extracellular vesicles with attenuated pro-inflammatory protein signatures. J. Thromb. Haemost. 2021, 19, 2583–2595. [Google Scholar] [CrossRef]
  291. Shaihov-Teper, O.; Ram, E.; Ballan, N.; Brzezinski, R.Y.; Naftali-Shani, N.; Masoud, R.; Ziv, T.; Lewis, N.; Schary, Y.; Levin-Kotler, L.-P.; et al. Extracellular Vesicles From Epicardial Fat Facilitate Atrial Fibrillation. Circulation 2021, 143, 2475–2493. [Google Scholar] [CrossRef]
  292. Ni, H.; Pan, W.; Jin, Q.; Xie, Y.; Zhang, N.; Chen, K.; Lin, T.; Lin, C.; Xie, Y.; Wu, J.; et al. Label-free proteomic analysis of serum exosomes from paroxysmal atrial fibrillation patients. Clin. Proteom. 2021, 18, 1. [Google Scholar] [CrossRef]
  293. Conte, M.; Petraglia, L.; Cabaro, S.; Valerio, V.; Poggio, P.; Pilato, E.; Attena, E.; Russo, V.; Ferro, A.; Formisano, P.; et al. Epicardial Adipose Tissue and Cardiac Arrhythmias: Focus on Atrial Fibrillation. Front. Cardiovasc. Med. 2022, 9, 932262. [Google Scholar] [CrossRef]
  294. Goudis, C.A.; Vasileiadis, I.E.; Liu, T. Epicardial adipose tissue and atrial fibrillation: Pathophysiological mechanisms, clinical implications, and potential therapies. Curr. Med. Res. Opin. 2018, 34, 1933–1943. [Google Scholar] [CrossRef] [PubMed]
  295. Kira, S.; Abe, I.; Ishii, Y.; Miyoshi, M.; Oniki, T.; Arakane, M.; Daa, T.; Teshima, Y.; Yufu, K.; Shimada, T.; et al. Role of angiopoietin-like protein 2 in atrial fibrosis induced by human epicardial adipose tissue: Analysis using an organo-culture system. Heart Rhythm. 2020, 17, 1591–1601. [Google Scholar] [CrossRef]
  296. Li, B.; Po, S.S.; Zhang, B.; Bai, F.; Li, J.; Qin, F.; Liu, N.; Sun, C.; Xiao, Y.; Tu, T.; et al. Metformin regulates adiponectin signalling in epicardial adipose tissue and reduces atrial fibrillation vulnerability. J. Cell Mol. Med. 2020, 24, 7751–7766. [Google Scholar] [CrossRef] [PubMed]
  297. Bai, C.; Liu, Y.; Zhao, Y.; Ye, Q.; Zhao, C.; Liu, Y.; Wang, J. Circulating exosome-derived miR-122-5p is a novel biomarker for prediction of postoperative atrial fibrillation. J. Cardiovasc. Transl. Res. 2022, 15, 1393–1405. [Google Scholar] [CrossRef]
  298. Liu, L.; Luo, F.; Lei, K. Exosomes Containing LINC00636 Inhibit MAPK1 through the miR-450a-2-3p Overexpression in Human Pericardial Fluid and Improve Cardiac Fibrosis in Patients with Atrial Fibrillation. Mediat. Inflamm. 2021, 2021, 9960241. [Google Scholar] [CrossRef] [PubMed]
  299. 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] [PubMed]
  300. Xu, L.; Fan, Y.; Wu, L.; Zhang, C.; Chu, M.; Wang, Y.; Zhuang, W. Exosomes from Bone Marrow Mesenchymal Stem Cells with Overexpressed Nrf2 Inhibit Cardiac Fibrosis in Rats with Atrial Fibrillation. Cardiovasc. Ther. 2022, 2022, 2687807. [Google Scholar] [CrossRef] [PubMed]
  301. Xu, J.; Lei, S.; Sun, S.; Zhang, W.; Zhu, F.; Yang, H.; Xu, Q.; Zhang, B.; Li, H.; Zhu, M.; et al. MiR-324-3p Regulates Fibroblast Proliferation via Targeting TGF-β1 in Atrial Fibrillation. Int. Heart J. 2020, 61, 1270–1278. [Google Scholar] [CrossRef]
  302. Chen, Y.; Chen, X.; Li, H.; Li, Y.; Cheng, D.; Tang, Y.; Sang, H. Serum extracellular vesicles containing MIAT induces atrial fibrosis, inflammation and oxidative stress to promote atrial remodeling and atrial fibrillation via blockade of miR-485-5p-mediated CXCL10 inhibition. Clin. Transl. Med. 2021, 11, e482. [Google Scholar] [CrossRef] [PubMed]
  303. Cao, F.; Li, Z.; Ding, W.; Yan, L.; Zhao, Q. Angiotensin II-Treated Cardiac Myocytes Regulate M1 Macrophage Polarization via Transferring Exosomal PVT1. J. Immunol. Res. 2021, 2021, 1994328. [Google Scholar] [CrossRef]
  304. Li, J.; Zhang, Q.; Jiao, H. LncRNA NRON promotes M2 macrophage polarization and alleviates atrial fibrosis through suppressing exosomal miR-23a derived from atrial myocytes. J. Formos. Med. Assoc. 2021, 120, 1512–1519. [Google Scholar] [CrossRef]
  305. Liu, D.; Chen, H.; Fu, Y.; Yao, Y.; He, S.; Wang, Y.; Cao, Z.; Wang, X.; Yang, M.; Zhao, Q. KCa3.1 Promotes Proinflammatory Exosome Secretion by Activating AKT/Rab27a in Atrial Myocytes during Rapid Pacing. Cardiovasc. Ther. 2023, 2023, 3939360. [Google Scholar] [CrossRef]
  306. Zhang, W.; Man, Y.; Chen, Z. microRNA-148a in Exosomes Derived from Bone Marrow Mesenchymal Stem Cells Alleviates Cardiomyocyte Apoptosis in Atrial Fibrillation by Inhibiting SMOC2. Mol. Biotechnol. 2022, 64, 1076–1087. [Google Scholar] [CrossRef]
  307. Yan, B.; Liu, T.; Yao, C.; Liu, X.; Du, Q.; Pan, L. LncRNA XIST shuttled by adipose tissue-derived mesenchymal stem cell-derived extracellular vesicles suppresses myocardial pyroptosis in atrial fibrillation by disrupting miR-214-3p-mediated Arl2 inhibition. Lab. Investig. 2021, 101, 1427–1438. [Google Scholar] [CrossRef]
  308. Li, S.; Gao, Y.; Liu, Y.; Li, J.; Yang, X.; Hu, R.; Liu, J.; Zhang, Y.; Zuo, K.; Li, K.; et al. Myofibroblast-Derived Exosomes Contribute to Development of a Susceptible Substrate for Atrial Fibrillation. Cardiology 2020, 145, 324–332. [Google Scholar] [CrossRef] [PubMed]
  309. Parent, S.; Vaka, R.; Risha, Y.; Ngo, C.; Kanda, P.; Nattel, S.; Khan, S.; Courtman, D.; Stewart, D.J.; Davis, D.R. Prevention of atrial fibrillation after open-chest surgery with extracellular vesicle therapy. JCI Insight 2023, 8, e163297. [Google Scholar] [CrossRef] [PubMed]
  310. Tanai, E.; Frantz, S. Pathophysiology of Heart Failure. In Comprehensive Physiology; Wiley: Hoboken, NJ, USA, 2015; pp. 187–214. [Google Scholar]
  311. Xue, R.; Tan, W.; Wu, Y.; Dong, B.; Xie, Z.; Huang, P.; He, J.; Dong, Y.; Liu, C. Role of Exosomal miRNAs in Heart Failure. Front. Cardiovasc. Med. 2020, 7, 592412. [Google Scholar] [CrossRef]
  312. Snipelisky, D.; Chaudhry, S.-P.; Stewart, G.C. The Many Faces of Heart Failure. Card. Electrophysiol. Clin. 2019, 11, 11–20. [Google Scholar] [CrossRef]
  313. Han, C.; Yang, J.; Sun, J.; Qin, G. Extracellular vesicles in cardiovascular disease: Biological functions and therapeutic implications. Pharmacol. Ther. 2022, 233, 108025. [Google Scholar] [CrossRef]
  314. Zheng, D.; Huo, M.; Li, B.; Wang, W.; Piao, H.; Wang, Y.; Zhu, Z.; Li, D.; Wang, T.; Liu, K. The Role of Exosomes and Exosomal MicroRNA in Cardiovascular Disease. Front. Cell Dev. Biol. 2021, 8, 616161. [Google Scholar] [CrossRef]
  315. Vilella-Figuerola, A.; Cordero, A.; Mirabet, S.; Muñoz-García, N.; Suades, R.; Padró, T.; Badimon, L. Platelet-Released Extracellular Vesicle Characteristics Differ in Chronic and in Acute Heart Disease. Thromb. Haemost. 2023, 123, 892–903. [Google Scholar] [CrossRef] [PubMed]
  316. Matan, D.; Mobarrez, F.; Löfström, U.; Corbascio, M.; Ekström, M.; Hage, C.; Lyngå, P.; Persson, B.; Eriksson, M.; Linde, C.; et al. Extracellular vesicles in heart failure—A study in patients with heart failure with preserved ejection fraction or heart failure with reduced ejection fraction characteristics undergoing elective coronary artery bypass grafting. Front. Cardiovasc. Med. 2022, 9, 952974. [Google Scholar] [CrossRef]
  317. Suades, R.; Vilella-Figuerola, A.; Padró, T.; Mirabet, S.; Badimon, L. Red Blood Cells and Endothelium Derived Circulating Extracellular Vesicles in Health and Chronic Heart Failure: A Focus on Phosphatidylserine Dynamics in Vesiculation. Int. J. Mol. Sci. 2023, 24, 11824. [Google Scholar] [CrossRef] [PubMed]
  318. Pfeifer, P.; Zietzer, A.; Hölscher, M.; Jehle, J.; Nickenig, G.; Werner, N.; Gestrich, C.; Jansen, F. Transverse aortic constriction-induced heart failure leads to increased levels of circulating microparticles. Int. J. Cardiol. 2022, 347, 54–58. [Google Scholar] [CrossRef] [PubMed]
  319. Vilella-Figuerola, A.; Padró, T.; Roig, E.; Mirabet, S.; Badimon, L. New factors in heart failure pathophysiology: Immunity cells release of extracellular vesicles. Front. Cardiovasc. Med. 2022, 9, 939625. [Google Scholar] [CrossRef] [PubMed]
  320. Ye, W.; Tang, X.; Yang, Z.; Liu, C.; Zhang, X.; Jin, J.; Lyu, J. Plasma-derived exosomes contribute to inflammation via the TLR9-NF-κB pathway in chronic heart failure patients. Mol. Immunol. 2017, 87, 114–121. [Google Scholar] [CrossRef]
  321. Matsumoto, S.; Sakata, Y.; Suna, S.; Nakatani, D.; Usami, M.; Hara, M.; Kitamura, T.; Hamasaki, T.; Nanto, S.; Kawahara, Y.; et al. Circulating p53-Responsive MicroRNAs Are Predictive Indicators of Heart Failure after Acute Myocardial Infarction. Circ. Res. 2013, 113, 322–326. [Google Scholar] [CrossRef]
  322. Wang, L.; Liu, J.; Xu, B.; Liu, Y.-L.; Liu, Z. Reduced exosome miR-425 and miR-744 in the plasma represents the progression of fibrosis and heart failure. Kaohsiung J. Med. Sci. 2018, 34, 626–633. [Google Scholar] [CrossRef]
  323. Beg, F.; Wang, R.; Saeed, Z.; Devaraj, S.; Masoor, K.; Nakshatri, H. Inflammation-associated microRNA changes in circulating exosomes of heart failure patients. BMC Res. Notes 2017, 10, 751. [Google Scholar] [CrossRef]
  324. Xie, Y.; Hang, J.Z.; Zhang, N.; Liu, G. Clinical Significance of MiR-27a Expression in Serum Exosomes in Patients with Heart Failure. Cell. Mol. Biol. 2022, 67, 324–331. [Google Scholar] [CrossRef]
  325. Yang, J.; Xue, F.-T.; Li, Y.-Y.; Liu, W.; Zhang, S. Exosomal piRNA sequencing reveals differences between heart failure and healthy patients. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7952–7961. [Google Scholar] [CrossRef]
  326. Han, J.; Zhang, L.; Hu, L.; Yu, H.; Xu, F.; Yang, B.; Zhang, R.; Zhang, Y.; An, Y. Circular RNA-Expression Profiling Reveals a Potential Role of Hsa_circ_0097435 in Heart Failure via Sponging Multiple MicroRNAs. Front. Genet. 2020, 11, 212. [Google Scholar] [CrossRef]
  327. Galluzzo, A.; Gallo, S.; Pardini, B.; Birolo, G.; Fariselli, P.; Boretto, P.; Vitacolonna, A.; Peraldo-Neia, C.; Spilinga, M.; Volpe, A.; et al. Identification of novel circulating microRNAs in advanced heart failure by next-generation sequencing. ESC Heart Fail. 2021, 8, 2907–2919. [Google Scholar] [CrossRef]
  328. Xiao, Y.-C.; Wang, W.; Gao, Y.; Li, W.-Y.; Tan, X.; Wang, Y.-K.; Wang, W.-Z. The Peripheral Circulating Exosomal microRNAs Related to Central Inflammation in Chronic Heart Failure. J. Cardiovasc. Transl. Res. 2022, 15, 500–513. [Google Scholar] [CrossRef]
  329. Verbree-Willemsen, L.; Zhang, Y.; Ibrahim, I.; Ooi, S.B.S.; Wang, J.-W.; Mazlan, M.I.; Kuan, W.S.; Chan, S.-P.; Peelen, L.M.; Grobbee, D.E.; et al. Extracellular vesicle Cystatin C and CD14 are associated with both renal dysfunction and heart failure. ESC Heart Fail. 2020, 7, 2240–2249. [Google Scholar] [CrossRef]
  330. Lu, W.; Liu, X.; Zhao, L.; Yan, S.; Song, Q.; Zou, C.; Li, X. MiR-22-3p in exosomes increases the risk of heart failure after down-regulation of FURIN. Chem. Biol. Drug Des. 2023, 101, 550–567. [Google Scholar] [CrossRef]
  331. BUJAK, M.; FRANGOGIANNIS, N. The role of TGF-β signaling in myocardial infarction and cardiac remodeling. Cardiovasc. Res. 2007, 74, 184–195. [Google Scholar] [CrossRef]
  332. Basma, H.; Johanson, A.N.; Dhar, K.; Anderson, D.; Qiu, F.; Rennard, S.; Lowes, B.D. TGF-β induces a heart failure phenotype via fibroblasts exosome signaling. Heliyon 2019, 5, e02633. [Google Scholar] [CrossRef] [PubMed]
  333. Tian, C.; Gao, L.; Rudebush, T.L.; Yu, L.; Zucker, I.H. Extracellular Vesicles Regulate Sympatho-Excitation by Nrf2 in Heart Failure. Circ. Res. 2022, 131, 687–700. [Google Scholar] [CrossRef] [PubMed]
  334. Messer, S.; Large, S. Resuscitating heart transplantation: The donation after circulatory determined death donor. Eur. J. Cardio-Thorac. Surg. 2016, 49, 1–4. [Google Scholar] [CrossRef] [PubMed]
  335. Wang, M.; Yan, L.; Li, Q.; Yang, Y.; Turrentine, M.; March, K.; Wang, I. -wen Mesenchymal stem cell secretions improve donor heart function following ex vivo cold storage. J. Thorac. Cardiovasc. Surg. 2022, 163, e277–e292. [Google Scholar] [CrossRef] [PubMed]
  336. van den Hoogen, P.; de Jager, S.C.A.; Mol, E.A.; Schoneveld, A.S.; Huibers, M.M.H.; Vink, A.; Doevendans, P.A.; Laman, J.D.; Sluijter, J.P.G. Potential of mesenchymal- and cardiac progenitor cells for therapeutic targeting of B-cells and antibody responses in end-stage heart failure. PLoS ONE 2019, 14, e0227283. [Google Scholar] [CrossRef] [PubMed]
  337. Ren, Y.; Wu, Y.; He, W.; Tian, Y.; Zhao, X. Exosomes secreted from bone marrow mesenchymal stem cells suppress cardiomyocyte hypertrophy through Hippo-YAP pathway in heart failure. Genet. Mol. Biol. 2023, 46, e20220221. [Google Scholar] [CrossRef] [PubMed]
  338. Gao, L.; Qiu, F.; Cao, H.; Li, H.; Dai, G.; Ma, T.; Gong, Y.; Luo, W.; Zhu, D.; Qiu, Z.; et al. Therapeutic delivery of microRNA-125a-5p oligonucleotides improves recovery from myocardial ischemia/reperfusion injury in mice and swine. Theranostics 2023, 13, 685–703. [Google Scholar] [CrossRef] [PubMed]
  339. Xuan, L.; Fu, D.; Zhen, D.; Wei, C.; Bai, D.; Yu, L.; Gong, G. Extracellular vesicles derived from human bone marrow mesenchymal stem cells protect rats against acute myocardial infarction-induced heart failure. Cell Tissue Res. 2022, 389, 23–40. [Google Scholar] [CrossRef] [PubMed]
  340. Pang, Y.; Ma, M.; Wang, D.; Xia, J.; Wang, X.; Hou, L.; Wang, Z.; Li, X. Embryonic Stem Cell-Derived Exosomes Attenuate Transverse Aortic Constriction Induced Heart Failure by Increasing Angiogenesis. Front. Cardiovasc. Med. 2021, 8, 638771. [Google Scholar] [CrossRef] [PubMed]
  341. Harane, N.E.; Kervadec, A.; Bellamy, V.; Pidial, L.; Neametalla, H.J.; Perier, M.-C.; Correa, B.L.; Thiébault, L.; Cagnard, N.; Duché, A.; et al. Acellular therapeutic approach for heart failure: In vitro production of extracellular vesicles from human cardiovascular progenitors. Eur. Heart J. 2018, 39, 1835–1847. [Google Scholar] [CrossRef]
  342. Zhong, Z.; Tian, Y.; Luo, X.; Zou, J.; Wu, L.; Tian, J. Extracellular Vesicles Derived From Human Umbilical Cord Mesenchymal Stem Cells Protect Against DOX-Induced Heart Failure through the miR-100-5p/NOX4 Pathway. Front. Bioeng. Biotechnol. 2021, 9, 703241. [Google Scholar] [CrossRef]
  343. Duan, J.; Liu, X.; Shen, S.; Tan, X.; Wang, Y.; Wang, L.; Kang, L.; Wang, K.; Wei, Z.; Qi, Y.; et al. Trophoblast Stem-Cell-Derived Exosomes Alleviate Cardiotoxicity of Doxorubicin via Improving Mfn2-Mediated Mitochondrial Fusion. Cardiovasc. Toxicol. 2023, 23, 23–31. [Google Scholar] [CrossRef]
  344. Ni, J.; Liu, Y.; Kang, L.; Wang, L.; Han, Z.; Wang, K.; Xu, B.; Gu, R. Human trophoblast-derived exosomes attenuate doxorubicin-induced cardiac injury by regulating miR-200b and downstream Zeb1. J. Nanobiotechnol. 2020, 18, 171. [Google Scholar] [CrossRef] [PubMed]
  345. Zhu, D.; Cheng, K. Cardiac Cell Therapy for Heart Repair: Should the Cells Be Left Out? Cells 2021, 10, 641. [Google Scholar] [CrossRef] [PubMed]
  346. Amirzadeh Gougheri, K.; Ahmadi, A.; Ahmadabadi, M.G.; Babajani, A.; Yazdanpanah, G.; Bahrami, S.; Hassani, M.; Niknejad, H. Exosomal Cargo: Pro-angiogeneic, anti-inflammatory, and regenerative effects in ischemic and non-ischemic heart diseases—A comprehensive review. Biomed. Pharmacother. 2023, 168, 115801. [Google Scholar] [CrossRef] [PubMed]
  347. Shao, L.; Zhang, Y.; Lan, B.; Wang, J.; Zhang, Z.; Zhang, L.; Xiao, P.; Meng, Q.; Geng, Y.; Yu, X.; et al. MiRNA-Sequence Indicates That Mesenchymal Stem Cells and Exosomes Have Similar Mechanism to Enhance Cardiac Repair. BioMed Res. Int. 2017, 2017, 4150705. [Google Scholar] [CrossRef] [PubMed]
  348. Ibrahim, A.G.E.; Cheng, K.; Marbán, E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef]
  349. Tzng, E.; Bayardo, N.; Yang, P.C. Current challenges surrounding exosome treatments. Extracell. Vesicle 2023, 2, 100023. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of the biological process of the genesis of extracellular vesicles, including the major classification due their size, content and functional impact.
Figure 1. Schematic representation of the biological process of the genesis of extracellular vesicles, including the major classification due their size, content and functional impact.
Cells 13 00265 g001
Figure 2. Schematic representation of the most representative small EV therapy in the treatment of different of myocardial infarction (A), heart failure (B), atrial fibrillation (C), hypertrophic cardiomyopathy (D), dilated cardiomyopathy (E) and diabetic cardiomyopathy (F). (miRNA red, protein green, lncRNA blue, circRNA yellow).
Figure 2. Schematic representation of the most representative small EV therapy in the treatment of different of myocardial infarction (A), heart failure (B), atrial fibrillation (C), hypertrophic cardiomyopathy (D), dilated cardiomyopathy (E) and diabetic cardiomyopathy (F). (miRNA red, protein green, lncRNA blue, circRNA yellow).
Cells 13 00265 g002
Table 4. Summary of ncRNAs associated with atrial fibrillation (↑ upregulated and ↓ downregulated).
Table 4. Summary of ncRNAs associated with atrial fibrillation (↑ upregulated and ↓ downregulated).
StudyDE ncRNAsMain ncRNAsSampleRef.
microRNAs
Siwaponanan et al. (2022)↑ 19 miRNAsmiR-106b-3p, miR-590-5p, miR-339-3p, miR-378-3p, miR-328-3p, and miR-532-3plarge EVs
AF vs. non-AF
[280]
Zhu et al. (2022)↑ 13 miRNAs
↓ 27 miRNAs
miR-124-3p, miR-378d, miR-2110, and miR-3180-3p
miR-223-5p, miR-574-3p, miR-125a-3p, and miR-1299
plasma
exosomes
AF vs. non-AF
[283]
Wei et al. (2020)↑ 33 miRNAs
↓ 117 miRNAs
miR-92b-3p, miR-1306-5p, let-7b-3pplasma
exosomes
AF vs. non-AF
[284]
Mun et al. (2019)↑ 45 miRNAs miR-103a, miR-107,miR-320d, miR-486,let-7bserum
paroxysmal vs. persistent AF
[285]
Wang et al. (2019)↑ 21 miRNAs
↓ 18 miRNAs
miR-483-5p, miR-142-5p, miR-223-3pplasma
exosomes
AF vs. non-AF
[282]
Hao et al. (2022)↑ 41 miRNAs
↓ 50 miRNAs
miR-210atrial myocytes, serum
exosomes
[281]
Xie et al. (2023)↑ 31 miRNAs
↓ 37 miRNAs
miR-641
miR-30e-5p
plasma exosomes
ischemic AF stroke
[286]
Table 5. Summary of ncRNAs associated with heart failure (↑ upregulated and ↓ downregulated).
Table 5. Summary of ncRNAs associated with heart failure (↑ upregulated and ↓ downregulated).
AssayDE ncRNAsMain ncRNAsSampleRef.
miRNAs
Zhang et al. (2023)50 miRNAs ↑
48 miRNAs ↓
miR-103a-3p
miR-148a-3p
Plasma
(CDM patient)
[223]
Huang et al. (2022)2 miRNAs ↑miR-30d-5p
miR-126a-5p
HFpEF rat model[262]
Xie et al. (2022)miR-27a ↑miR-27aSerum
(HF patient)
[324]
Beg et al. (2017)2 miRNAs ↑miR-486
miR-146a
Plasma
(HF patient)
[323]
Wang et al. (2018)2 miRNAs ↓miR-425
miR-744
Plasma
(HF patient)
[322]
Matsumoto et al. (2013)2 miRNAs ↑miR-194
miR-34a
Serum
(Hf patient)
[321]
Wu et al. (2018)miR-92b-5p ↑miR-92b-5pSerum
(Hf patient)
[250]
Galluzzo et al. (2021)20 miRNAs ↑
12 miRNAs ↓
miR-22-3pPlasma
(HF patient)
[327]
Xiao et al. (2022)5 miRNAs ↑
13 miRNAs ↓
miR-214-3p
let-7i-5p
let-7g-5p
HF rat model[328]
piRNAs
Yang et al. (2018)585 piRNAs ↑
4623 piRNAs ↓
piR-02009
piR-006426
Serum
(HF patient)
[325]
circRNAs
Han et al. (2020)29 circRNAs ↑
27 circRNAs ↓
circ-0097435Blood
(HF patient)
[326]
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

Caño-Carrillo, S.; Castillo-Casas, J.M.; Franco, D.; Lozano-Velasco, E. Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases. Cells 2024, 13, 265. https://doi.org/10.3390/cells13030265

AMA Style

Caño-Carrillo S, Castillo-Casas JM, Franco D, Lozano-Velasco E. Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases. Cells. 2024; 13(3):265. https://doi.org/10.3390/cells13030265

Chicago/Turabian Style

Caño-Carrillo, Sheila, Juan Manuel Castillo-Casas, Diego Franco, and Estefanía Lozano-Velasco. 2024. "Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases" Cells 13, no. 3: 265. https://doi.org/10.3390/cells13030265

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