Introduction
Cardiovascular disease is the leading cause of death worldwide, accounting for almost a third of deaths.1 Although advances in research and cardiac care have increased survival, there is still a need to develop new strategies for prevention, intervention, and palliation, given the continued high‐mortality rate. Extracellular vesicles (EVs) hold tremendous potential to monitor and treat cardiovascular disease because of their capabilities for analysis and modulation. EVs also compose a critical component of intercellular signaling by transporting molecular cargo from donor cells to recipient cells, thereby modulating gene expression and influencing the phenotype of the recipient cell.2, 3, 4, 5, 6, 7 Most studies have documented this intercellular communication in vitro, but emerging evidence suggests it also occurs in vivo.8, 9, 10, 11 In this review, we focus on the role of exosomes, a subset of EVs, as mediators of intercellular signaling in the heart. Exosomes are constitutively released from several cardiac cell types including cardiomyocytes, cardiac fibroblasts, endothelial cells, inflammatory cells, and resident stem cells12 and may be responsible for the communication among these cell types under both physiological and pathological conditions.13, 14, 15 Exosomes, when injected into the heart, are well known to mimic the cardioprotective and restorative properties of various progenitor or stem cells including cardiac progenitor cells (CPCs),16 mesenchymal stem cells (MSCs),17, 18 hematopoietic stem cells,19 and induced pluripotent stem cells (iPSCs).20 Despite the importance of exosomes in paracrine signaling, less is known about the function of secreted exosomes and their paracrine actions among the different cell types of the heart.
The iPSCs have evolved into a crucial model system to understand human cardiovascular disease. Protocols have been developed to differentiate patient‐specific iPSCs into several cell types within the heart such as cardiomyocytes,21, 22, 23 endothelial cells,24, 25 and smooth muscle cells.26 As a result, iPSC‐derived cell studies have elucidated mechanisms involved in the pathogenesis of long QT syndrome,27 dilated cardiomyopathy,28 dystrophic cardiomyopathy,29, 30 hypertrophy,31 and diabetic cardiomyopathy,32 and thus their exosomes may be useful in addressing critical questions regarding intercellular communication in cardiac function and disease. In this review, we discuss the role of exosomes in the understanding of cardiovascular disease as well as the potential of iPSC‐derived cells to model the exosome‐specific paracrine effects.
Nomenclature of EVs
The field of EV research has been hampered not only by lack of standardized nomenclature but also by lack of criteria to distinguish, isolate, and identify the different subtypes of EVs. The term EV encompasses several subtypes of generated cells and expelled vesicles that are enclosed by a membrane bilayer. These include exosomes, microvesicles, and apoptotic bodies (Figure 1). The subtypes can be differentiated by their size, content, and route of intracellular formation. Exosomes are between 25 and 200 nm in diameter and are formed inside multivesicular bodies within the endocytic pathway (Figure 2).33 Here, the endosomal sorting complex required for transport facilitates the inward budding of the plasma membrane, which fills the exosome with cytoplasmic contents and retains membrane proteins specific to the cell of origin.34, 35, 36, 37 At this point, multivesicular bodies may either dock and fuse with the plasma membrane to release exosomes into the intracellular space or deliver their contents to the lysosome for degradation (Figure 2).38, 39 Microvesicles are 100 to 1000 nm in diameter and are formed by the outward budding and pinching of the plasma membrane,40 whereas apoptotic bodies (>1000 nm) are much larger and are formed by apoptotic cell membrane blebbing.41
Figure 1.

Extracellular vesicle subtypes. Extracellular vesicles are divided into 3 categories based on size, contents, and route of formation. Exosomes, the smallest, originate within multivesicular bodies and carry RNAs, proteins, and lipids. Microvesicles, the next largest, form through outward pinching off of the plasma membrane and also contain RNAs, proteins, and lipids. Apoptotic bodies are formed from dying cells as the plasma membrane blebs to recycle contents. Apoptotic bodies are variable in size and contain cell debris, genomic DNA (gDNA), and proteins.
Figure 2.

The formation and fates of multivesicular bodies. During endocytosis, the plasma membrane (PM) buds inward, filling with cytoplasmic contents and forming the early endosome. Surface proteins may be retained throughout endocytosis. The early endosome matures into the late endosome. This membrane further buds inward with the aid of the endosomal sorting complex required for transport (ESCRT), forming intraluminal vesicles. Once filled with these vesicles (now known as exosomes), the late endosome becomes the multivesicular body, which may either deliver its contents to the lysosome for degradation or fuse with the PM to secrete exosomes.
Exosomal Composition
Exosomes contain lipids, proteins, RNAs, and microRNAs (miRs).5, 6 The composition of exosomes not only differs from microvesicles and apoptotic bodies4 but also varies depending on cell type of origin, disease status, or stimuli during formation.42, 43, 44, 45 The exosome composition and internal cargo determine the outcome of the intercellular communication.46 The field of exosome research is continuously expanding. Resources for obtaining up‐to‐date literature on exosome biology, roles in disease, and composition can be found in Table 1.
Table 1.
Database Resources for Exosome Research
| Name | Website Address | Information |
|---|---|---|
| Exocarta | http://www.exocarta.org/ | Proteins, RNA, lipids |
| Vesiclepedia | http://www.microvesicles.org/ | Proteins, RNA, lipids |
| EVPedia | http://student4.postech.ac.kr/evpedia2_xe/xe/ | Proteins, RNA, lipids |
| EV Track | http://evtrack.org/ | Database of EV publications |
| Plasma proteome database | http://www.plasmaproteomedatabase.org/ | EV proteins from plasma |
EV indicates extracellular vesicle.
The lipid composition includes ceramide, sphingomyelin, phosphatidylcholine, and phosphatidylserine47, 48 within the membrane bilayer and cholesterol inside the exosomes. The organization of the lipids participates in the biological activity and can, in part, determine the fate of the exosome.49 Exosome‐specific proteins include the ubiquitously expressed tetraspanins CD81, CD9, CD63, CD82, HSP70 (heat shock 70‐kDa protein), HSP60, and Alix (apoptosis‐linked gene 2‐interacting protein X); annexins and cytoskeletal proteins; and unique cargo proteins.50, 51 Moreover, exosomes contain RNAs and miRs that can change depending on the exosome subtype, cell type, or disease state.34, 52, 53 The sorting of most proteins, RNAs, and lipids into exosomes is not well understood46 and is an area of current investigation.
Exosome Uptake
Exosomes are internalized by their destination cells through multiple mechanisms. A potential route is endocytosis, which requires an initial surface‐binding event either by direct interaction of a receptor with its ligand or through adhesion molecules such as integrins. Another route, membrane fusion of exosomes, has also been demonstrated in a minority of studies.54, 55 Other exosomal surface proteins such as CD47 may modulate the uptake process. For example, CD47, a thrombospondin 1 receptor, is present on the surface of EVs derived from Jurkat T cells and can alter gene expression and functional signaling in endothelial cells.56, 57 Whether other microenvironmental factors modulate exosome uptake is largely unknown.
Isolation Techniques for Exosomes
To study exosome‐mediated intercellular communication, exosomes need to be isolated and purified. A gold standard isolation method is lacking. Numerous isolation methods are available, but they result in an exosome populations with different yields, purity, and size.58 In addition, proper preanalytical processing is important to minimize variability,59 as the activation of cells during sample collection and handling has the potential to release additional exosomes. The isolation of exosomes from blood is especially problematic. The most common method of isolation is graded ultracentrifugation, and the exosomes can be purified further by the addition of a sucrose density gradient. Precipitation is also a common method that is commercially available through kits, although specificity is poor. Several studies in the literature examining the effects of exosomes may have used heterogeneous EV populations because of these limitations. Only size‐exclusion chromatography results in the purification of a homozygous population. Lai et al used this method to purify a fraction of exosomes from MSCs.18 These purified exosomes had a hydrodynamic radius of 55 to 65 nm and reduced infarct size when injected into an isolated heart model of ischemia and reperfusion injury. However, several studies which show EV‐associated cardioprotection are presumably a mixed population of exosomes and/or microvesicles isolated from progenitor or stem cells.60 This includes a study that showed MSC‐conditioned medium containing both exosomes and microvesicles decreased infarct size following ischemic injury.60 Consequently, it becomes less clear which homogenous population is critical when studying exosome‐mediated intercellular communication.
Methods to Identify and Characterize Exosomes
The small size of exosomes makes their identification and characterization challenging; therefore, most studies use a combination of techniques to characterize the population of exosomes being studied. Lotvall et al described at set of minimum standards to meet for publication of scientific analyses involving exosomes and other vesicles.61
Nanoparticle tracking analysis using NanoSight (Malvern Instruments) provides a rapid method to characterize nanoparticles.33 Particle size distribution and count is determined in real time using light scattering and Brownian motion of suspended samples. It allows the analysis of particles between 20 and 2000 nm and provides high‐resolution particle size distribution profiles and concentration measurements. Despite the ease of use to characterize different samples, it gives the most accurate readings between 50 and 1000 nm; however, it is not specific to exosomes and counts every particle within its size range including membrane fragments and other cellular debris.62
Electron microscopy, with its superior resolution ability, is an important element of exosome characterization.33 Exosomes are visualized by their characteristic negative staining with uranyl acetate and methylcellulose using transmission electron microscopy. They are identified as double‐membrane “cup‐shaped” vesicles that are also in the correct size range. The cup‐shape description is most likely not their true form but rather an artifact of the procedure and should not be taken as a definitive feature of exosomes.62, 63
For flow cytometry, the exosome size falls below the size threshold at which routine flow cytometers can accurately distinguish particles from noise.64 This limitation is overcome by binding exosomes to latex beads either directly or indirectly through exosome‐specific antibodies.65 The bound exosomes can then be stained with other fluorophore‐coupled antibodies. In this manner, surface proteins can be characterized, but the particle number may not be accurate because it is unknown how many exosomes bind to each bead.
The ability of exosomes to be taken up into cells can be determined by labeling the exosomes with lipophilic membrane‐binding dyes such as PKH67, DiD, or CellMask.66 These lipophilic dyes have also been shown to bind different types of lipoprotein particles such as low‐density lipoprotein that are also taken up by the cell and thus may create artifacts. Consequently, it is important to purify exosomes to minimize contamination with lipoproteins and to use the appropriate controls to assess for artifactual uptake.66 For certain studies, these issues can be overcome through labeling of specific vesicles by transfecting the parent cells with a vesicle‐targeted fluorescent probe or reporter protein.67
Biological Roles for Exosomes in Cardiovascular Disease and Repair
Several studies suggest the role of exosomes as potential disease biomarkers. In one study, an exosomal miR was detected before any elevated levels of traditional disease biomarkers such as creatine kinase or troponin, highlighting the exosomes' biomarker potential for an early, sensitive, and specific diagnostic tool for cardiomyopathy.68 Additional investigations have been exploring the use of exosomes as a delivery vehicle for various therapies,69, 70 given their ability to easily cross biological membranes.71 Importantly, exosomes are being examined for their roles in various cardiomyopathies such as hypertrophy,72, 73 heart failure,74 myocardial infarction, and ischemic injury.68, 75, 76, 77 These studies are discussed in more detail later and summarized in Table 2. It should be noted that the limitations of these studies include (1) inconsistent methods of exosome isolation that may lead to a mixed population of EVs being tested and (2) most of the mechanistic effects of exosomes being studied in in vitro models.
Table 2.
Study Summary
| Study | Exosome Source | Exosome Prep | In Vitro Effects | In Vivo Effects |
|---|---|---|---|---|
| Barile 201478 | Isolated CPCs |
|
Decreases apoptosis in HL‐1 cells | Improved cardiac function and angiogenesis |
| Xiao et al 201679 |
Sca‐1+ CPCs +/− H2O2 |
ExoQuick TC (System Biosciences) | H2O2–exosomes: Decrease oxidative stress in H9C2 myoblasts through miR‐21 transfer | … |
| Ibrahim et al 201476 | Cardiospheres | ExoQuick TC (System Biosciences) 3 sizes of exosomes |
|
|
| Aminzadeh et al 201880 | Cardiospheres | … | … | Exosomes injected in the heart improve skeletal muscle function. |
| Sahoo et al 201181 | MSCs | Ultracentrifugation | Increases HUVEC tube length, viability, and proliferation | |
| Yu et al 201582 | MSCs | ExoQuick Kit | Increase survival of cardiomyocytes exposed to hypoxia by decreasing apoptosis and preserving the mitochondria membrane potential |
|
| Ribeiro‐Rodrigues 201783 | H9C2 myoblasts +/− ischemia mimic conditions | Ultracentrifugation and sucrose gradient | Ischemia‐derived exosomes:
|
Ischemia‐derived exosomes:
|
| Wang et al 201484 | Diabetic GK rat cardiomyocytes |
Ultracentrifugation ExoQuick Kit Transwell coculture |
Inhibit proliferation, migration, and tube formation of cardiac ECs by transfer of miR‐320 | |
| Davidson et al 201885 |
|
Ultracentrifugation | … | … |
| Vicencio et al 201575 | Plasma from rats and healthy humans | Ultracentrifugation | HL‐1 cells increase in mitochondrial membrane potential and cell death after hypoxia and reoxygenation | Decreased infarct size in a rat subject to ischemia–reperfusion injury |
| Pironti et al 201513 |
|
Ultracentrifugation |
|
ATIR enriched exosomes confer blood pressure responses to angiotensin II in AT1R knockout mice |
| Bang et al 201472 | Neonatal rat cardiac fibroblasts |
Ultracentrifugation Transwell direct‐transfer experiments |
Induces hypertrophy of neonatal cardiomyocytes | … |
| Datta et al 201786 | Hypertrophied cardiomyocytes | Ultracentrifugation | Modulates STAT‐3 signaling in cardiac fibroblasts | … |
| Loyer et al 201810 |
Heart tissues Myoectomy tissue from humans |
Centrifuges at 20 500g to pellet large EVs and then exosomes isolated from Exosome Isolation Kit (Exiquon) to isolate small EVs | Used to stimulate cardiac‐derived monocytes | Local generation of EVs in the heart following MI |
| Bian et al 201477 | Hypoxic MSC in vitro | Ultracentrifugation (100 000g); frozen (mixture of exosomes and MVs (47–180 nm) | HUVECs for proliferation, migration, and tube formation | Promotes angiogenesis and preserves cardiac performance in an acute MI model |
| Garcia et al 20167 | Cultured neonatal murine cardiomyocytes and H9C2 cells +/− glucose starvation | Transwell direct transfer experiments | Exosomes contain glucose transporters and transfer the functional transporters to HUVECs and primary ECs | |
| Hu et al 201811 | Cardiomyocytes |
|
Exosomes from injured cardiomyocytes increase BMSC apoptosis | Exosomes from injured cardiomyocytes accelerate BMSC injury |
AT1R indicates angiotensin II type 1 receptor; BMSC, bone marrow–derived mesenchymal stem cell; CPC, cardiac progenitor cell; DM, diabetes mellitus; EC, endothelial cell; EV, extracellular vesicle; HEK293T, human embryonic kidney 293 cell line containing the SV40 large T antigen; HUVEC, human umbilical vein endothelial cell; MI, myocardial infarction; miR, microRNA; MSC, mesenchymal stem cells; MV, microvesicles; Sca‐1, stem cells antigen‐1; STAT3, signal transducer and activator of transcription 3; TAC, transverse aortic constriction.
Cardioprotective Effects of Progenitor and Stem Cell–Derived Exosomes
Several studies examining the roles of CPCs, cardiospheres, and MSC‐secreted exosomes have found their involvement in cardioprotective paracrine effects.87 CPC‐derived EVs, in which exosomes were a major component, decreased apoptosis in HL‐1 cardiomyocytes 48 hours after serum starvation. The EVs were enriched in miR‐132 and miR‐210 and transferred these miRs to the cells; this was in part responsible for the antiapoptotic effects of the exosomes.78 Furthermore, CPC‐derived EVs increased myocardial viability, decreased cardiomyocyte apoptosis, and increased blood vessel density when injected into the myocardium in vivo.78 The same effects were not seen using control exosomes isolated from dermal fibroblasts. Oxidative stress upregulates miR‐21 within the exosomes isolated from CPCs,79 and the transfer of exosomal miR‐21 to H9C2 myoblasts decreases oxidative stress‐induced apoptosis in vitro.79
Cardiospheres are multicellular clusters that contain progenitors of cardiomyocytes, endothelial cells, and smooth muscle cells.88 Exosomes derived from cardiospheres have shown cardioprotective benefits in vitro by enhancing angiogenesis and cardiomyocyte proliferation and decreasing apoptosis.76 Cardiosphere‐derived exosomes decreased cardiac infarct size in vivo via the delivery of miR‐146a76 and stimulated protective antioxidant and prosurvival signaling pathways.80 Furthermore, Aminzadeh et al demonstrated that cardiosphere exosomes are capable of exerting crosstalk among multiple cell types; exosomes injected in the heart led to improved skeletal muscle function in mdx mice, a mouse model of Duchenne muscular dystrophy.80
MSCs originate in the bone marrow, and MSC‐derived exosomes influence both endothelial cells and cardiomyocytes. MSC‐derived exosomes modulate endothelial cells by stimulating angiogenesis and increasing viability, proliferation, and tube formation81 and modulate cardiomyocytes by decreasing apoptosis and preserving mitochondrial membrane potential through the enrichment of miR‐19a.82
Understanding the Paracrine Effects Exerted by Exosomes Secreted From Cardiac Cells in Cardiovascular Disease
Proper cardiovascular function depends on the coordinated interplay and communication between smooth muscle cells, endothelial cells, fibroblasts, immune cells, and cardiomyocytes. Exosomes contribute to this intracellular communication.5, 6 Despite exosomes representing an important mechanism for intracellular communication in general, little is known about the exosome regulation of cardiomyocytes and other noncardiomyocyte cell types within the healthy and diseased heart. Figure 3 summarizes current findings regarding the effects elicited by exosomes secreted from the different cardiac cell types in cardiovascular disease.
Figure 3.

Paracrine effects in disease exerted by cardiovascular exosomes. Cardiovascular secreted exosomes exert diverse effects on their destination cells, considering the cell type of origin, exosomal contents, and the conditions of release. Cardiac fibroblast–derived exosomes have been associated with pathology because of their transference of microRNA (miR) 21, which led to the induction of cardiac hypertrophy.70 The effects of cardiomyocyte‐derived exosomes were either proangiogenic82 or antiangiogenic,89 dependent on the conditions of release, which affected their miR cargo. Cardiomyocyte‐secreted exosomes were also found to be involved in the transfer of functional angiotensin II type 1 receptors (AT1Rs) under conditions of stress12 and involved in HSP90 (heat shock 90‐kDa protein)–dependent regulation of collagen synthesis in fibroblasts in models of hypertrophy.83 Serum and plasma exosomes arise from a variety of cell sources, and the diversity of their effects results from the disease status and conditions of release. Diabetic serum exosomes fail to stimulate ERK1/2‐protective signaling in cardiomyocytes,90 whereas plasma exosomes from healthy controls could stimulate ERK1/2 (Extracellular signal‐regulated kinase 1 and 2) via TLR4 (Toll‐like receptor 4) and HSP27.73
Several studies suggest exosomes may change composition under pathological conditions. Recent studies have shown that miR profiles differ in plasma exosomes from spontaneously hypertensive rats compared with the normotensive Wistar–Kyoto rats.89 Likewise, the protein content of exosomes differ among Zucker lean, Zucker fatty, and Sprague–Dawley rats.90 In myocardial infarction, in which the heart is subject not only to hypoxic conditions but also to inflammation, cardiomyocytes increase the secretion of exosomes and change the composition of their cargo. H9C2 myoblasts subject to hypoxic conditions secrete exosomes enriched for miRs related to cell proliferation and differentiation including miR‐143 and miR‐222. In corollary experiments, the exosomes derived from the hypoxic cells induced endothelial proliferation, migration, and survival under stress.83
Exosomes do not always impart favorable effects on recipient cells and may be neutral to detrimental in exerting their effects. Exosomes released from diabetic GK rat cardiomyocytes inhibit the proliferation, migration, and tube formation of endothelial cell lines by a mechanism involving transfer of exosomal‐associated miR‐320.84 Furthermore, diabetic conditions render exosomes ineffective in activating survival pathways in cardiomyocytes,85 whereas in the absence of diabetes mellitus, cardioprotective effects were seen to activate the TLR‐4 (Toll‐like receptor 4)–ERK1/2 (Extracellular signal‐regulated kinase 1 and 2) pathway.75 HEK293T (human embryonic kidney 293 cells containing the SV40 large T antigen) cells overexpressing angiotensin II type 1 receptors (AT1Rs), when subject to osmotic stretch in vitro, secrete exosomes enriched for AT1Rs.13 Likewise, in mice subject to transverse aortic constriction to induced pressure overload, the exosomes released into the serum had a 100‐fold increase in AT1R density.13 These AT1R‐enriched exosomes transferred the functional AT1R to recipient wild‐type HEK293 cells in vitro and to the heart and skeletal muscle of AT1R‐knockout mice in vivo.13 Exosomes have also been shown to mediate cross‐talk between cardiomyocytes and cardiac fibroblasts in cardiac hypertrophy. Cardiac fibroblast–derived exosomes enriched in miR‐21‐3p have been shown to be potential regulators of cardiomyocyte hypertrophy.72 Likewise, exosomes secreted from hypertrophied cardiomyocytes contain HSP90 and IL‐6 (interleukin 6) to maintain STAT3 (signal transducer and activator of transcription 3)–fibrotic pathway signaling.86 This study, however, also showed differential regulation of collagen expression in the cardiac fibroblast in vitro versus in vivo, which points toward the potential paracrine effects resulting from crosstalk among different cell types. It will be important for future studies to compare in vivo findings with in vitro systems to confirm which cell type is releasing exosomes to exert specific effects.
It has recently been shown that exosomes derived from injured cardiomyocytes may impede the survival of bone marrow–derived MSCs in the infarcted heart.11 Exosomes isolated from cultured cardiomyocytes treated with H2O2 could induce bone marrow–derived MSCs to undergo apoptosis in vitro. In corollary in vivo experiments, Ras‐Related Protein Rab27a‐knockout mice were engineered to have reduced exosome release from cardiac tissues. After left anterior descending artery ligation in the Rab27a‐knockout mouse heart, mouse bone marrow–derived MSCs were injected into the border zone of the infarct. These transplanted bone marrow–derived MSCs showed 3‐fold increased survival after 4 days.11 The accumulative evidence suggests that EVs, including exosomes, when produced by cells under adverse conditions, not only contribute to the disease pathology but also may be detrimental toward cardiac regenerative therapies. Nevertheless, the full extent of EV‐mediated intracellular communication within the heart and how neighboring or distant cells can secrete EVs to influence the cardiac structure and function are largely unknown. The Rab27a‐knockout mouse and the continued development of cell‐type promoter CD63–GFP (green fluorescent protein) transgenic animals91, 92 are steps toward addressing these knowledge gaps.
iPSCs as a Tool to Understand Intercellular Communication in Disease
The extracellular spaces of the myocardium contain exosomes from a mixed cellular source, making it difficult to determine the influence of cell‐type–specific exosomes from one cell on its neighboring cell. Consequently, in vitro studies are critical to understanding specific cell‐type exosome function in health and disease. An important consideration in the interpretation of all exosome studies is the type of model system used in the investigation. In vivo, there is likely to be a mixture of EVs from multiple cell types present in the extracellular space, whereas in vitro, only cell‐type–specific EVs can be examined individually. Furthermore, the addition of exogenous exosomes (in vitro or in vivo) may not be physiologically accurate, which is critical in the interpretation of results.
iPSCs as disease models can be used to model intercellular communication between neighboring cell types in the heart by studying the effects of cell‐type–specific exosomes. Numerous studies have validated the use of iPSCs as a model system to study genetic cardiac diseases such as long QT syndrome,27, 93 LEOPARD syndrome,94 and Duchenne muscular dystrophy.29, 95 In addition, there is growing interest in using iPSC‐derived cells to study environmental modifiers of disease. The iPSC‐derived cells can be exposed to various conditions to precipitate oxidative, metabolic, or endoplasmic reticular stress or subjected to neurohormones, growth factors, or hypoxia to mimic certain disease states. For example, a diabetic environment can be created by supplementing the culture media with glucose, endothelin 1, and cortisol.32
Exosomes are also modulators of the disease process. The relevant recipient cell of interest can be cardiomyocytes, endothelial cells, smooth muscle cells, or cardiac fibroblasts. Exosomes can be isolated from one cell type and be applied to another cell type to assess phenotypic changes. For instance, iPSC‐derived endothelial cells exposed to exosomes isolated from iPSC‐derived cardiomyocytes can be assessed for changes in gene expression and various phenotypes such as tube formation, wound healing, senescence, and low‐density lipoprotein uptake (Figure 4). The contributions of different genetic backgrounds or gene mutations on exosome cargo and the resulting phenotypic effects of exosomes on recipient iPSC‐derived cells can also be studied using patient‐specific iPSCs. Furthermore, 3‐dimensional organoid models have been developed to investigate EV‐mediated intercellular communication.67 This model is based on coculturing recipient cells with donor cells containing a CD63–GFP reporter under control of a cell‐type–specific promoter. In this manner, EV‐mediated intercellular communication can be investigated under conditions closer to the physiological environment.
Figure 4.

Using induced pluripotent stem cells (iPSCs) to study exosomal communication between cardiovascular cell types. Patient‐ or disease‐specific iPSCs can be differentiated into multiple cell types including cardiomyocytes, endothelial cells, or smooth muscle cells. These derived cells can be used to collect and profile exosomes (exos) or to assess cell‐type–specific exosomal effects on recipient cell function. iCM indicates iPSC‐derived cardiomyoycte; iEC, iPSC‐derived endothelial cell; iSMC, iPSC‐derived smooth muscle cell; LDL, low‐density lipoprotein.
Exosome cargo can be compared from wild‐type and diseased iPSC lines and derived cells. Our laboratory and others have validated the use of iPSC‐derived cell lines as a model for cardiovascular disease29, 95 and showed that exosomes derived from these cell lines may play a role in the disease process related to their cargo. The miRs elicit functional effects in cells typically by inhibiting targeted gene expression. Dysregulation of miRs is associated with dilated cardiomyopathy.96 In Figure 5, we show that miR cargo differs not only between wild‐type and Duchenne muscular dystrophy iPSC‐derived cardiomyocytes but also between Duchenne muscular dystrophy iPSC‐derived cardiomyocytes and skeletal myocytes. Furthermore, cell‐type–specific exosome cargo can be compared under basal conditions along with different types of stress conditions such as hypoxic, metabolic, oxidative, and hyperglycemic stresses. Malik et al showed that pathologic changes to the environment such as hypoxia and ethanol change protein content and reactive oxygen species decrease exosome production from cardiac myocytes.44
Figure 5.

Example of using induced pluripotent stem cell (iPSC)–derived cells to investigate cell‐ or disease‐specific exosome cargo. Exosomes were isolated from wild‐type (WT) and dystrophin‐deficient (Dys)‐iPSC‐derived cardiac and skeletal myocyte‐conditioned media using an isolation reagent by Thermo Fisher. A, Isolated exosome size (50 nm) and morphology was confirmed by electron microscopy. B, WT and Dys cardiac and skeletal myocyte exosomes display differential microRNA (miR) profiles, as shown by polymerase chain reaction array analysis. hsa indicates homo sapiens; iCM, iPSC‐derived cardiomyocytes; iSkM, iPSC‐derived skeletal muscle cells; let, part of the lethal‐7 gene family.
The iPSC‐derived cells can also be used to decipher the function of exosomes compared with other EVs. Experiments using GW4869, a specific noncompetitive inhibitor of neutral spingomyelinase, is commonly used to inhibit the secretion of exosomes; however, it blocks the secretion of EVs in general. Consequently, the in vitro and in vivo findings with the compound can be conflicting. For instance, serum EVs isolated from the Duchenne muscular dystrophy (mdx) mice decrease apoptosis in C2C12 myocytes; however, mdx mice treated with GW4869 show improved muscle strength in vivo.97 In addition, GW4869 has been shown to exert cardioprotective effects against sepsis‐induced cardiac dysfunction.98 Others have found cardioprotection with the addition of serum EVs.75, 99 A consideration for the interpretation of results with the use of serum EVs and their global blockade with inhibitors is that serum EVs contain exosomes, microvesicles, and apoptotic bodies, all originating from a variety of cellular sources, so it can be difficult to pinpoint which vesicle type and source is responsible for the effects observed without purifying the EV subtype of interest and doing isolated cell studies. Because stimuli and originating cell type affect the contents of exosomes, it is likely that the same cell type can release both protective and pathogenic exosomes depending on its status at the time of release.
Conclusions
Exosomes play an important role in the function of the heart under normal and disease conditions. Whereas exosomes harvested from stem cells in vitro may protect against cardiac injury and promote repair, the exosomes secreted from the endogenous cardiac cells may play a more ominous role in cardiac disease progression. However, we are just beginning to understand how the exosomal cargo changes under different conditions and how this affects the signaling pathways and phenotypic behavior of recipient cells. More studies are needed to illuminate the mechanisms that influence the sorting of exosomes and the diverse paracrine effects exosomes have in cardiovascular disease processes. Harnessing the potential of the translational power of exosomes by understanding the fundamental roles of exosomes in cardiovascular disease will provide insights for the discovery of novel disease mechanisms and the development of novel therapeutics.
Sources of Funding
This work is supported by the National Institutes of Health and the National Heart, Lung, and Blood Institute (K08HL11148 and R01134932) grants awarded to Strande.
Disclosures
None.
J Am Heart Assoc. 2018;7:e007954 DOI: 10.1161/JAHA.117.007954.29858362
References
- 1. Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, Isasi CR, Jiménez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Mackey RH, Matsushita K, Mozaffarian D, Mussolino ME, Nasir K, Neumar RW, Palaniappan L, Pandey DK, Thiagarajan RR, Reeves MJ, Ritchey M, Rodriguez CJ, Roth GA, Rosamond WD, Sasson C, Towfighi A, Tsao CW, Turner MB, Virani SS, Voeks JH, Willey JZ, Wilkins JT, Wu JH, Alger HM, Wong SS, Muntner P; American Heart Association Statistics Committee and Stroke Statistics Subcommittee . Heart disease and stroke statistics—2017 update: a report from the American Heart Association. Circulation. 2017;135:e146–e603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Maia J, Caja S, Strano Moraes MC, Couto N, Costa‐Silva B. Exosome‐based cell‐cell communication in the tumor microenvironment. Front Cell Dev Biol. 2018;6:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Krämer‐Albers E‐M, Hill AF. Extracellular vesicles: interneural shuttles of complex messages. Curr Opin Neurobiol. 2016;39:101–107. [DOI] [PubMed] [Google Scholar]
- 4. Crescitelli R, Lässer C, Szabó TG, Kittel A, Eldh M, Dianzani I, Buzás EI, Lötvall J. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J Extracell Vesicles. 2013;2:20677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Ludwig AK, Giebel B. Exosomes: small vesicles participating in intercellular communication. Int J Biochem Cell Biol. 2012;44:11–15. [DOI] [PubMed] [Google Scholar]
- 6. Gyorgy B, Szabo TG, Pasztoi M, Pal Z, Misjak P, Aradi B, Laszlo V, Pallinger E, Pap E, Kittel A, Nagy G, Falus A, Buzas EI. Membrane vesicles, current state‐of‐the‐art: emerging role of extracellular vesicles. Cell Mol Life Sci. 2011;68:2667–2688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Garcia NA, Moncayo‐Arlandi J, Sepulveda P, Diez‐Juan A. Cardiomyocyte exosomes regulate glycolytic flux in endothelium by direct transfer of GLUT transporters and glycolytic enzymes. Cardiovasc Res. 2016;109:397–408. [DOI] [PubMed] [Google Scholar]
- 8. van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, Prabhakar S, Mardini O, Crommentuijn MH, Skog J, Krichevsky AM, Stemmer‐Rachamimov A, Mempel TR, El Khoury J, Hickman SE, Breakefield XO. Directly visualized glioblastoma‐derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016;18:58–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Kojima R, Bojar D, Rizzi G, Hamri GC, El‐Baba MD, Saxena P, Auslander S, Tan KR, Fussenegger M. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson's disease treatment. Nat Commun. 2018;9:1305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Loyer X, Zlatanova I, Devue C, Yin M, Howangyin K‐Y, Klaihmon P, Guerin CL, Kheloufi M, Vilar J, Zannis K, Fleischmann BK, Hwang DW, Park J, Lee H, Menasche P, Silvestre J‐S, Boulanger CM. Intra‐cardiac release of extracellular vesicles shapes inflammation following myocardial infarction. Circ Res. 2018;122:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Hu M, Guo G, Huang Q, Cheng C, Xu R, Li A, Liu N, Liu S. The harsh microenvironment in infarcted heart accelerates transplanted bone marrow mesenchymal stem cells injury: the role of injured cardiomyocytes‐derived exosomes. Cell Death Dis. 2018;9:357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Chistiakov DA, Orekhov AN, Bobryshev YV. Cardiac extracellular vesicles in normal and infarcted heart. Int J Mol Sci. 2016;17:E63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Pironti G, Strachan RT, Abraham D, Mon‐Wei YuS, Chen M, Chen W, Hanada K, Mao L, Watson LJ, Rockman HA. Circulating exosomes induced by cardiac pressure overload contain functional angiotensin II type 1 receptors. Circulation. 2015;24:2120–2130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Sahoo S, Losordo DW. Exosomes and cardiac repair after myocardial infarction. Circ Res. 2014;114:333–344. [DOI] [PubMed] [Google Scholar]
- 15. Sluijter JP, Verhage V, Deddens JC, van den Akker F, Doevendans PA. Microvesicles and exosomes for intracardiac communication. Cardiovasc Res. 2014;102:302–311. [DOI] [PubMed] [Google Scholar]
- 16. Gray WD, French KM, Ghosh‐Choudhary S, Maxwell JT, Brown ME, Platt MO, Searles CD, Davis ME. Identification of therapeutic covariant microRNA clusters in hypoxia‐treated cardiac progenitor cell exosomes using systems biology. Circ Res. 2015;116:255–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Liu L, Jin X, Hu CF, Li R, Zhou Z, Shen CX. 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. [DOI] [PubMed] [Google Scholar]
- 18. Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, Salto‐Tellez M, Timmers L, Lee CN, El Oakley RM, Pasterkamp G, de Kleijn DP, Lim SK. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214–222. [DOI] [PubMed] [Google Scholar]
- 19. Mackie AR, Klyachko E, Thorne T, Schultz KM, Millay M, Ito A, Kamide CE, Liu T, Gupta R, Sahoo S, Misener S, Kishore R, Losordo DW. Sonic hedgehog‐modified human CD34+ cells preserve cardiac function after acute myocardial infarction. Circ Res. 2012;111:312–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wang Y, Zhang L, Li Y, Chen L, Wang X, Guo W, Zhang X, Qin G, He SH, Zimmerman A, Liu Y, Kim IM, Weintraub NL, Tang Y. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int J Cardiol. 2015;192:61–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB, Azarin SM, Raval KK, Zhang J, Kamp TJ, Palecek SP. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc Natl Acad Sci USA. 2012;109:E1848–E1857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lian X, Zhang J, Azarin SM, Zhu K, Hazeltine LB, Bao X, Hsiao C, Kamp TJ, Palecek SP. Directed cardiomyocyte differentiation from human pluripotent stem cells by modulating Wnt/beta‐catenin signaling under fully defined conditions. Nat Protoc. 2013;8:162–175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Zhang J, Klos M, Wilson GF, Herman AM, Lian X, Raval KK, Barron MR, Hou L, Soerens AG, Yu J, Palecek SP, Lyons GE, Thomson JA, Herron TJ, Jalife J, Kamp TJ. Extracellular matrix promotes highly efficient cardiac differentiation of human pluripotent stem cells: the matrix sandwich method. Circ Res. 2012;111:1125–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Lippmann ES, Azarin SM, Kay JE, Nessler RA, Wilson HK, Al‐Ahmad A, Palecek SP, Shusta EV. Derivation of blood‐brain barrier endothelial cells from human pluripotent stem cells. Nat Biotechnol. 2012;30:783–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Orlova VV, van den Hil FE, Petrus‐Reurer S, Drabsch Y, Ten Dijke P, Mummery CL. Generation, expansion and functional analysis of endothelial cells and pericytes derived from human pluripotent stem cells. Nat Protoc. 2014;9:1514–1531. [DOI] [PubMed] [Google Scholar]
- 26. Dash Biraja C, Jiang Z, Suh C, Qyang Y. Induced pluripotent stem cell‐derived vascular smooth muscle cells: methods and application. Biochem J. 2015;465:185–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Itzhaki I, Maizels L, Huber I, Zwi‐Dantsis L, Caspi O, Winterstern A, Feldman O, Gepstein A, Arbel G, Hammerman H, Boulos M, Gepstein L. Modelling the long QT syndrome with induced pluripotent stem cells. Nature. 2011;471:225. [DOI] [PubMed] [Google Scholar]
- 28. Streckfuss‐Bömeke K, Tiburcy M, Fomin A, Luo X, Li W, Fischer C, Özcelik C, Perrot A, Sossalla S, Haas J, Vidal RO, Rebs S, Khadjeh S, Meder B, Bonn S, Linke WA, Zimmermann W‐H, Hasenfuss G, Guan K. Severe DCM phenotype of patient harboring RBM20 mutation S635A can be modeled by patient‐specific induced pluripotent stem cell‐derived cardiomyocytes. J Mol Cell Cardiol. 2017;113:9–21. [DOI] [PubMed] [Google Scholar]
- 29. Afzal MZ, Reiter M, Gastonguay C, McGivern JV, Guan X, Ge ZD, Mack DL, Childers MK, Ebert AD, Strande JL. Nicorandil, a nitric oxide donor and ATP‐sensitive potassium channel opener, protects against dystrophin‐deficient cardiomyopathy. J Cardiovasc Pharmacol Ther. 2016;21:549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Lin B, Li Y, Han L, Kaplan AD, Ao Y, Kalra S, Bett GC, Rasmusson RL, Denning C, Yang L. Modeling and study of the mechanism of dilated cardiomyopathy using induced pluripotent stem cells derived from individuals with Duchenne muscular dystrophy. Dis Model Mech. 2015;8:457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Han L, Li Y, Tchao J, Kaplan AD, Lin B, Li Y, Mich‐Basso J, Lis A, Hassan N, London B, Bett GC, Tobita K, Rasmusson RL, Yang L. Study familial hypertrophic cardiomyopathy using patient‐specific induced pluripotent stem cells. Cardiovasc Res. 2014;104:258–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Drawnel FM, Boccardo S, Prummer M, Delobel F, Graff A, Weber M, Gerard R, Badi L, Kam‐Thong T, Bu L, Jiang X, Hoflack JC, Kiialainen A, Jeworutzki E, Aoyama N, Carlson C, Burcin M, Gromo G, Boehringer M, Stahlberg H, Hall BJ, Magnone MC, Kolaja K, Chien KR, Bailly J, Iacone R. Disease modeling and phenotypic drug screening for diabetic cardiomyopathy using human induced pluripotent stem cells. Cell Rep. 2014;9:810–821. [DOI] [PubMed] [Google Scholar]
- 33. Emanueli C, Shearn AIU, Angelini GD, Sahoo S. Exosomes and exosomal miRNAs in cardiovascular protection and repair. Vascul Pharmacol. 2015;71:24–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Oksvold MP, Kullmann A, Forfang L, Kierulf B, Li M, Brech A, Vlassov AV, Smeland EB, Neurauter A, Pedersen KW. Expression of B‐cell surface antigens in subpopulations of exosomes released from B‐cell lymphoma cells. Clin Ther. 2014;36:847–862.e1. [DOI] [PubMed] [Google Scholar]
- 35. Gruenberg J. The endocytic pathway: a mosaic of domains. Nat Rev Mol Cell Biol. 2001;2:721–730. [DOI] [PubMed] [Google Scholar]
- 36. Mayers JR, Audhya A. Vesicle formation within endosomes: an ESCRT marks the spot. Commun Integr Biol. 2012;5:50–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Henne WM, Buchkovich NJ, Emr SD. The ESCRT pathway. Dev Cell. 2011;21:77–91. [DOI] [PubMed] [Google Scholar]
- 38. Futter CE, Pearse A, Hewlett LJ, Hopkins CR. Multivesicular endosomes containing internalized EGF‐EGF receptor complexes mature and then fuse directly with lysosomes. J Cell Biol. 1996;132:1011–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Stahl PD, Barbieri MA. Multivesicular bodies and multivesicular endosomes: the “ins and outs” of endosomal traffic. Sci STKE. 2002;2002:pe32. [DOI] [PubMed] [Google Scholar]
- 40. Cocucci E, Racchetti G, Meldolesi J. Shedding microvesicles: artefacts no more. Trends Cell Biol. 2009;19:43–51. [DOI] [PubMed] [Google Scholar]
- 41. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide‐ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ, Lotvall JO. Exosome‐mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 2007;9:654–659. [DOI] [PubMed] [Google Scholar]
- 43. Blomme A, Fahmy K, Peulen O, Costanza B, Fontaine M, Struman I, Baiwir D, de Pauw E, Thiry M, Bellahcene A, Castronovo V, Turtoi A. Myoferlin is a novel exosomal protein and functional regulator of cancer‐derived exosomes. Oncotarget. 2016;7:83669–83683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Malik ZA, Kott KS, Poe AJ, Kuo T, Chen L, Ferrara KW, Knowlton AA. Cardiac myocyte exosomes: stability, HSP60, and proteomics. Am J Physiol Heart Circ Physiol. 2013;304:H954–H965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Genneback N, Hellman U, Malm L, Larsson G, Ronquist G, Waldenstrom A, Morner S. Growth factor stimulation of cardiomyocytes induces changes in the transcriptional contents of secreted exosomes. J Extracell Vesicles. 2013;2:20167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Villarroya‐Beltri C, Baixauli F, Gutierrez‐Vazquez C, Sanchez‐Madrid F, Mittelbrunn M. Sorting it out: regulation of exosome loading. Semin Cancer Biol. 2014;28:3–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Skotland T, Sandvig K, Llorente A. Lipids in exosomes: current knowledge and the way forward. Prog Lipid Res. 2017;66:30–41. [DOI] [PubMed] [Google Scholar]
- 48. Subra C, Grand D, Laulagnier K, Stella A, Lambeau G, Paillasse M, De Medina P, Monsarrat B, Perret B, Silvente‐Poirot S, Poirot M, Record M. Exosomes account for vesicle‐mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 2010;51:2105–2120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Record M, Carayon K, Poirot M, Silvente‐Poirot S. Exosomes as new vesicular lipid transporters involved in cell‐cell communication and various pathophysiologies. Biochim Biophys Acta. 2014;1841:108–120. [DOI] [PubMed] [Google Scholar]
- 50. Gupta S, Knowlton AA. HSP60 trafficking in adult cardiac myocytes: role of the exosomal pathway. Am J Physiol Heart Circ Physiol. 2007;292:H3052–H3056. [DOI] [PubMed] [Google Scholar]
- 51. Thery C, Boussac M, Veron 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. [DOI] [PubMed] [Google Scholar]
- 52. Palma J, Yaddanapudi SC, Pigati L, Havens MA, Jeong S, Weiner GA, Weimer KM, Stern B, Hastings ML, Duelli DM. MicroRNAs are exported from malignant cells in customized particles. Nucleic Acids Res. 2012;40:9125–9138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Tauro BJ, Greening DW, Mathias RA, Mathivanan S, Ji H, Simpson RJ. Two distinct populations of exosomes are released from LIM1863 colon carcinoma cell‐derived organoids. Mol Cell Proteomics. 2013;12:587–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Montecalvo A, Larregina AT, Shufesky WJ, Stolz DB, Sullivan ML, Karlsson JM, Baty CJ, Gibson GA, Erdos G, Wang Z, Milosevic J, Tkacheva OA, Divito SJ, Jordan R, Lyons‐Weiler J, Watkins SC, Morelli AE. Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. Blood. 2012;119:756–766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Valapala M, Vishwanatha JK. Lipid raft endocytosis and exosomal transport facilitate extracellular trafficking of annexin A2. J Biol Chem. 2011;286:30911–30925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kaur S, Elkahloun AG, Singh SP, Arakelyan A, Roberts DD. A function‐blocking CD47 antibody modulates extracellular vesicle‐mediated intercellular signaling between breast carcinoma cells and endothelial cells. J Cell Commun Signal. 2018;12:157–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Kaur S, Singh SP, Elkahloun AG, Wu W, Abu‐Asab MS, Roberts DD. CD47‐dependent immunomodulatory and angiogenic activities of extracellular vesicles produced by T cells. Matrix Biol. 2014;37:49–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Xu R, Greening DW, Zhu HJ, Takahashi N, Simpson RJ. Extracellular vesicle isolation and characterization: toward clinical application. J Clin Invest. 2016;126:1152–1162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Coumans FAW, Brisson AR, Buzas EI, Dignat‐George F, Drees EEE, El‐Andaloussi S, Emanueli C, Gasecka A, Hendrix A, Hill AF, Lacroix R, Lee Y, van Leeuwen TG, Mackman N, Mager I, Nolan JP, van der Pol E, Pegtel DM, Sahoo S, Siljander PRM, Sturk G, de Wever O, Nieuwland R. Methodological guidelines to study extracellular vesicles. Circ Res. 2017;120:1632–1648. [DOI] [PubMed] [Google Scholar]
- 60. Timmers L, Lim SK, Arslan F, Armstrong JS, Hoefer IE, Doevendans PA, Piek JJ, El Oakley RM, Choo A, Lee CN, Pasterkamp G, de Kleijn DP. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 2007;1:129–137. [DOI] [PubMed] [Google Scholar]
- 61. Lotvall J, Hill AF, Hochberg F, Buzas EI, Di Vizio D, Gardiner C, Gho YS, Kurochkin IV, Mathivanan S, Quesenberry P, Sahoo S, Tahara H, Wauben MH, Witwer KW, Thery C. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Szatanek R, Baj‐Krzyworzeka M, Zimoch J, Lekka M, Siedlar M, Baran J. The methods of choice for extracellular vesicles (EVs) characterization. Int J Mol Sci. 2017;18:1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Erdbrügger U, Lannigan J. Analytical challenges of extracellular vesicle detection: a comparison of different techniques. Cytometry A. 2015;89:123–134. [DOI] [PubMed] [Google Scholar]
- 64. Dragovic RA, Gardiner C, Brooks AS, Tannetta DS, Ferguson DJ, Hole P, Carr B, Redman CW, Harris AL, Dobson PJ, Harrison P, Sargent IL. Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis. Nanomedicine. 2011;7:780–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Suarez H, Gamez‐Valero A, Reyes R, Lopez‐Martin S, Rodriguez MJ, Carrascosa JL, Cabanas C, Borras FE, Yanez‐Mo M. A bead‐assisted flow cytometry method for the semi‐quantitative analysis of Extracellular Vesicles. Sci Rep. 2017;7:11271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Takov K, Yellon DM, Davidson SM. Confounding factors in vesicle uptake studies using fluorescent lipophilic membrane dyes. J Extracell Vesicles. 2017;6:1388731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Sadovska L, Zandberga E, Sagini K, Jekabsons K, Riekstina U, Kalnina Z, Llorente A, Line A. A novel 3D heterotypic spheroid model for studying extracellular vesicle‐mediated tumour and immune cell communication. Biochem Biophys Res Commun. 2018;495:1930–1935. [DOI] [PubMed] [Google Scholar]
- 68. Kuwabara Y, Ono K, Horie T, Nishi H, Nagao K, Kinoshita M, Watanabe S, Baba O, Kojima Y, Shizuta S, Imai M, Tamura T, Kita T, Kimura T. Increased microRNA‐1 and microRNA‐133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ Cardiovasc Genet. 2011;4:446–454. [DOI] [PubMed] [Google Scholar]
- 69. Alvarez‐Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJ. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–345. [DOI] [PubMed] [Google Scholar]
- 70. Tang K, Zhang Y, Zhang H, Xu P, Liu J, Ma J, Lv M, Li D, Katirai F, Shen GX, Zhang G, Feng ZH, Ye D, Huang B. Delivery of chemotherapeutic drugs in tumour cell‐derived microparticles. Nat Commun. 2012;3:1282. [DOI] [PubMed] [Google Scholar]
- 71. Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles across biological membranes: current perspectives and future challenges. Acta Pharm Sin B. 2016;4:287–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Bang C, Batkai S, Dangwal S, Gupta SK, Foinquinos A, Holzmann A, Just A, Remke J, Zimmer K, Zeug A, Ponimaskin E, Schmiedl A, Yin X, Mayr M, Halder R, Fischer A, Engelhardt S, Wei Y, Schober A, Fiedler J, Thum T. Cardiac fibroblast‐derived microRNA passenger strand‐enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest. 2014;124:2136–2146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Lyu L, Wang H, Li B, Qin Q, Qi L, Nagarkatti M, Nagarkatti P, Janicki JS, Wang XL, Cui T. A critical role of cardiac fibroblast‐derived exosomes in activating renin angiotensin system in cardiomyocytes. J Mol Cell Cardiol. 2015;89:268–279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Yamaguchi T, Izumi Y, Nakamura Y, Yamazaki T, Shiota M, Sano S, Tanaka M, Osada‐Oka M, Shimada K, Miura K, Yoshiyama M, Iwao H. Repeated remote ischemic conditioning attenuates left ventricular remodeling via exosome‐mediated intercellular communication on chronic heart failure after myocardial infarction. Int J Cardiol. 2015;15:239–246. [DOI] [PubMed] [Google Scholar]
- 75. Vicencio JM, Yellon DM, Sivaraman V, Das D, Boi‐Doku C, Arjun S, Zheng Y, Riquelme JA, Kearney J, Sharma V, Multhoff G, Hall AR, Davidson SM. Plasma exosomes protect the myocardium from ischemia‐reperfusion injury. J Am Coll Cardiol. 2015;65:1525–1536. [DOI] [PubMed] [Google Scholar]
- 76. Ibrahim AG, Cheng K, Marbán E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Reports. 2014;5:606–619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Bian S, Zhang L, Duan L, Wang X, Min Y, Yu H. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J Mol Med. 2014;92:387–397. [DOI] [PubMed] [Google Scholar]
- 78. Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu LM, Torre T, Siclari F, Moccetti T, Vassalli G. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res. 2014;103:530–541. [DOI] [PubMed] [Google Scholar]
- 79. Xiao J, Pan Y, Li XH, Yang XY, Feng YL, Tan HH, Jiang L, Feng J, Yu XY. Cardiac progenitor cell‐derived exosomes prevent cardiomyocytes apoptosis through exosomal miR‐21 by targeting PDCD4. Cell Death Dis. 2016;7:e2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Aminzadeh MA, Rogers RG, Fournier M, Tobin RE, Guan X, Childers MK, Andres AM, Taylor DJ, Ibrahim A, Ding X, Torrente A, Goldhaber JM, Lewis M, Gottlieb RA, Victor RA, Marban E. Exosome‐mediated benefits of cell therapy in mouse and human models of duchenne muscular dystrophy. Stem Cell Reports. 2018;10:942–955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Sahoo S, Klychko E, Thorne T, Misener S, Schultz KM, Millay M, Ito A, Liu T, Kamide C, Agrawal H, Perlman H, Qin G, Kishore R, Losordo DW. Exosomes from human CD34(+) stem cells mediate their proangiogenic paracrine activity. Circ Res. 2011;107:724–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Yu B, Kim HW, Gong M, Wang J, Millard RW, 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;1:349–360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Ribeiro‐Rodrigues TM, Laundos TL, Pereira‐Carvalho R, Batista‐Almeida D, Pereira R, Coelho‐Santos V, Silva AP, Fernandes R, Zuzarte M, Enguita FJ, Costa MC, Pinto‐do‐Ó P, Pinto MT, Gouveia P, Ferreira L, Mason JC, Pereira P, Kwak BR, Nascimento DS, Girão H. Exosomes secreted by cardiomyocytes subjected to ischaemia promote cardiac angiogenesis. Cardiovasc Res. 2017;113:1338–1350. [DOI] [PubMed] [Google Scholar]
- 84. Wang X, Huang W, Liu G, Cai W, Millard RW, Wang Y, Chang J, Peng T, Fan G‐C. Cardiomyocytes mediate anti‐angiogenesis in type 2 diabetic rats through the exosomal transfer of miR‐320 into endothelial cells. J Mol Cell Cardiol. 2014;74:139–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Davidson SM, Riquelme JA, Takov K, Vicencio JM, Boi‐Doku C, Khoo V, Doreth C, Radenkovic D, Lavandero S, Yellon DM. Cardioprotection mediated by exosomes is impaired in the setting of type II diabetes but can be rescued by the use of non‐diabetic exosomes in vitro. J Cell Mol Med. 2018;22:141–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Datta R, Bansal T, Rana S, Datta K, Datta Chaudhuri R, Chawla‐Sarkar M, Sarkar S. Myocyte‐derived Hsp90 modulates collagen upregulation via biphasic activation of STAT‐3 in fibroblasts during cardiac hypertrophy. Mol Cell Biol. 2017;37:e00611–e00616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Gallet R, Dawkins J, Valle J, Simsolo E, de Couto G, Middleton R, Tseliou E, Luthringer D, Kreke M, Smith RR, Marban L, Ghaleh B, Marban E. Exosomes secreted by cardiosphere‐derived cells reduce scarring, attenuate adverse remodelling, and improve function in acute and chronic porcine myocardial infarction. Eur Heart J. 2017;38:201–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Smith RR, Barile L, Cho HC, Leppo MK, Hare JM, Messina E, Giacomello A, Abraham MR, Marban E. Regenerative potential of cardiosphere‐derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007;115:896–908. [DOI] [PubMed] [Google Scholar]
- 89. Liu X, Yuan W, Yang L, Li J, Cai J. miRNA profiling of exosomes from spontaneous hypertensive rats using next‐generation sequencing. Cardiovasc Transl Res. 2018;76:1770. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 90. Wider J, Undyala VVR, Whittaker P, Woods J, Chen X, Przyklenk K. Remote ischemic preconditioning fails to reduce infarct size in the Zucker fatty rat model of type‐2 diabetes: role of defective humoral communication. Basic Res Cardiol. 2018;113:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Yoshimura A, Kawamata M, Yoshioka Y, Katsuda T, Kikuchi H, Nagai Y, Adachi N, Numakawa T, Kunugi H, Ochiya T, Tamai Y. Generation of a novel transgenic rat model for tracing extracellular vesicles in body fluids. Sci Rep. 2016;6:31172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Yoshimura A, Adachi N, Matsuno H, Kawamata M, Yoshioka Y, Kikuchi H, Odaka H, Numakawa T, Kunugi H, Ochiya T, Tamai Y. The Sox2 promoter‐driven CD63‐GFP transgenic rat model allows tracking of neural stem cell‐derived extracellular vesicles. Dis Model Mech. 2018;11:dmm028779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Egashira T, Yuasa S, Suzuki T, Aizawa Y, Yamakawa H, Matsuhashi T, Ohno Y, Tohyama S, Okata S, Seki T, Kuroda Y, Yae K, Hashimoto H, Tanaka T, Hattori F, Sato T, Miyoshi S, Takatsuki S, Murata M, Kurokawa J, Furukawa T, Makita N, Aiba T, Shimizu W, Horie M, Kamiya K, Kodama I, Ogawa S, Fukuda K. Disease characterization using LQTS‐specific induced pluripotent stem cells. Cardiovasc Res. 2012;95:419–429. [DOI] [PubMed] [Google Scholar]
- 94. Carvajal‐Vergara X, Sevilla A, D'Souza SL, Ang YS, Schaniel C, Lee DF, Yang L, Kaplan AD, Adler ED, Rozov R, Ge Y, Cohen N, Edelmann LJ, Chang B, Waghray A, Su J, Pardo S, Lichtenbelt KD, Tartaglia M, Gelb BD, Lemischka IR. Patient‐specific induced pluripotent stem‐cell‐derived models of LEOPARD syndrome. Nature. 2010;465:808–812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Guan X, Mack DL, Moreno CM, Strande JL, Mathieu J, Shi Y, Markert CD, Wang Z, Liu G, Lawlor MW, Moorefield EC, Jones TN, Fugate JA, Furth ME, Murry CE, Ruohola‐Baker H, Zhang Y, Santana LF, Childers MK. Dystrophin‐deficient cardiomyocytes derived from human urine: new biologic reagents for drug discovery. Stem Cell Res. 2014;12:467–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Rao PK, Toyama Y, Chiang HR, Gupta S, Bauer M, Medvid R, Reinhardt F, Liao R, Krieger M, Jaenisch R, Lodish HF, Blelloch R. Loss of cardiac microRNA‐mediated regulation leads to dilated cardiomyopathy and heart failure. Circ Res. 2009;105:585–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Matsuzaka Y, Tanihata J, Komaki H, Ishiyama A, Oya Y, Rüegg U, Takeda S, Hashido K. Characterization and functional analysis of extracellular vesicles and muscle‐abundant miRNAs (miR‐1, miR‐133a, and miR‐206) in C2C12 myocytes and mdx mice. PLoS One. 2016;12:e0167811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Essandoh K, Yang L, Wang X, Huang W, Qin D, Hao J, Wang Y, Zingarelli B, Peng T, Fan GC. Blockade of exosome generation with GW4869 dampens the sepsis‐induced inflammation and cardiac dysfunction. Biochim Biophys Acta. 2015;11:2362–2371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Giricz Z, Varga ZV, Baranyai T, Sipos P, Pálóczi K, Kittel Á, Buzás E, Ferdinandy P. Cardioprotection by remote ischemic preconditioning of the rat heart is mediated by extracellular vesicles. J Mol Cell Cardiol. 2014;68:75–78. [DOI] [PubMed] [Google Scholar]
