Potential clinical applications of exosomes in the diagnosis, treatment, and prognosis of cardiovascular diseases: a narrative review
Review Article

Potential clinical applications of exosomes in the diagnosis, treatment, and prognosis of cardiovascular diseases: a narrative review

Xuyang Chen1,2, Qi Luo2

1Joint Program of Nanchang University and Queen Mary University of London, Queen Mary School, Medical Department, Nanchang University, Nanchang, China; 2Department of Histology and Embryology, Nanchang University School of Basic Medical Sciences, Nanchang, China

Contributions: (I) Conception and design: Q Luo; (II) Administrative support: Q Luo; (III) Provision of study materials or patients: X Chen; (IV) Collection and assembly of data: X Chen; (V) Data analysis and interpretation: X Chen; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Qi Luo. Department of Histology and Embryology, Nanchang University School of Basic Medical Sciences, Nanchang, China. Email: jxnculq2018@163.com.

Background and Objective: Cardiovascular diseases (CVDs) have been one of the most common threats to human health in recent decades. At present, despite many diagnostic, prognostic and therapeutic methods being applied in the clinic, the prevalence of CVDs continues to rise. Therefore, new discovery is needed and exosomes have received extensive attention. Exosomes are extracellular vesicles that enable communication between cells. They are widely distributed in biofluids, suggesting that they may be useful in CVD diagnosis and prognosis. Furthermore, exosomes are ideal drug transporters with relatively high transport efficiency and the capability to target different kinds of tissues. However, the present research concentrates, for the most part, on mechanistic studies with less attention to clinical applications.

Methods: More than 150 relevant scientific articles from databases like PubMed, Web of Science were screened and analysed for this narrative review. Data of clinical trials are collected from clinicaltrials.gov.

Key Content and Findings: In this review, we concentrate on different exosomes and CVDs, and we summarize the physiological and pathological roles of CVD-related exosomes. We focused on the role exosomes may have as biomarkers of CVDs, therapeutic opportunities, and possible hurdles to the clinical application of exosomes, aiming to provide a useful reference for its translational use in the CVD field.

Conclusions: Specific changes in exosome cargos (mainly miRNAs and proteins) are in accordance with the occurrence and development of CVDs including acute myocardial infarction (AMI), arrhythmia, coronary artery disease (CAD), heart failure (HF) and cardiomyopathy, therefore meaningful for diagnosis and prognosis of CVDs. For exosome related therapeutic methods, potential ways consist of direct administration of exosomes, targeting on exosome synthesis, processing and release, and working as adjuvants. All in all, exosomes are expected to serve as meaningful tools in the diagnosis, treatment and prognosis of CVDs.

Keywords: Exosomes; cardiovascular diseases (CVDs); miRNA; clinical applications


Submitted Dec 24, 2021. Accepted for publication Mar 07, 2022.

doi: 10.21037/atm-22-619


Introduction

Cardiovascular diseases (CVDs) remain among of the greatest sources of mortality and morbidity in developed countries (1). Despite significant investment in research and therapy, the overall outcomes remain suboptimal. At present, although biomarkers like cardiac troponins, creatine kinase, and brain natriuretic peptides have been widely employed in CVD diagnosis and prognosis, their value may be limited by factors such as age, genetic background, lifestyle, heart associated diseases, etc. (2). Besides, regardless of numerous options for CVD treatment, including angiotensin-converting enzyme inhibitors, sartans, beta-blockers, calcium channel blockers, statins, etc., the prevalence of CVDs keeps an increasing trend still (1). Thereby, new discovery is essential and attention is given to exosomes.

Exosomes are vesicles originating from endosomes, and they are approximately 40 to 160 nm (average 100 nm) in diameter (3). The membrane of an exosome is roughly the same as a plasma membrane, consisting of a phospholipid bilayer. Under certain physiological or pathological stimulation, the plasma membrane invaginates to form endocytic vesicles, which mainly contain proteins, lipids, small molecules, and ions. After invaginating into the cytoplasm, many endocytic vesicles may fuse together and constitute early-sorting endosomes (ESEs). Then, ESEs endocytose mitochondria, the endoplasmic reticulum, the Golgi apparatus, and nucleic acids to develop into late-sorting endosomes (LSEs), which invaginate again to form multiple intraluminal vesicles (ILVs) under the control of protein complexes called endosomal sorting complexes required for transport (ESCRT). Because LSEs containing ILVs resemble multivesicular vesicles, they are called multivesicular bodies (MVBs). MVBs have two main fates as follows: they are degraded by fusion with autophagosomes and lysosomes, and they are then recycled by cells; or they dock on a cell membrane and release ILVs by exocytosis. The ILVs released by exocytosis are called exosomes (4,5). The contents of exosomes can be different from the contents of their parental cells, indicating that the sorting and packing mechanisms are specific (6,7). Sorting processes are fulfilled by particular sequence motifs, posttranscriptional modifications, or subcellular localization (8).

Exosomes function to transmit information between cells (9). Exosomes can transfer substances, including proteins, lipids, metabolites, RNA, mitochondrial DNA, and even activated receptors, to recipient cells, which affects the signaling pathways of the recipient cells, consequently modulating protein expression and phenotype (3,7,10,11). Because exosomes have a small diameter, they can be transported through body fluids, and their phospholipid bilayer protects internal molecules from degradation. Compared to the direct transfer of signaling molecules, exosomes are relatively more reliable for sending molecules to target cells (12-14). Additionally, exosomes can home to target tissues or cells and penetrate biological barriers, such as the blood–brain barrier. In certain situations, exosomes are taken up by receptor-mediated endocytosis, which has a high degree of accuracy and can facilitate the consecutive and stable transport of exosome cargo, suggesting that exosomes may function in drug delivery (15,16). In animal models of CVDs and in cell culture models, exosomes mitigate injury. Not surprisingly, exosome-based therapy has been proposed as a novel therapeutic option in CVDs (10,17,18).

Additionally, exosomes and exosome cargos are detectable in the blood, supporting a possible role as biomarkers of CVDs (19). Specific molecular patterns are associated with CVDs, and exosomes may provide real-time information for these changes. As a diagnostic and prognostic method, an exosome test is minimally invasive and highly efficient, and it can be used alone or combined with traditional methods to increase the sensitivity and specificity of traditional methods. Indeed, exosome profiles and cargo are being investigated as biomarkers for the diagnosis and prognosis of diseases, such as acute myocardial infarction (AMI), arrhythmia, coronary artery disease (CAD), and heart failure (HF). Of translational importance, exosomes are already employed in the diagnosis of some tumors, such as the ExoDx™ Lung (ALK) test, thereby validating the potential clinical application of exosomes in CVDs. We present the following article in accordance with the Narrative Review reporting checklist (available at https://atm.amegroups.com/article/view/10.21037/atm-22-619/rc).


Methods

We reviewed many updated review articles on exosomes. Then, the blank field with few exosome applications, CVDs, was screened out and literature about exosomes in CVDs was searched up to October 2021. More than 100 relevant scientific articles were reviewed, screened and analysed. Also, clinical trials were searched and analysed (literature and data collected from databases including PubMed, Web of Science and clinicaltrials.gov). The search strategy summary is shown in Table 1 and the detailed search strategy of PubMed is shown in Table S1 as an example.

Table 1

The search strategy summary

Items Specification
Date of search October 15th, 2021
Databases and other sources searched PubMed, Web of Science and clinicaltrials.gov
Search terms used Keywords: exosome, cardiovascular disease, biomarker, clinical trial, therapy
Timeframe 1987–2021
Inclusion and exclusion criteria All kinds of studies are included; language restricted to English
Selection process Xuyang Chen and Qi Luo conducted the selection together; selecting literature from aspects including correlation with subjects, time of publication and experimental design

Discussion

Roles of CVD-related exosomes

The development of CVDs is a complex process. Numerous molecules are secreted by disordered cells, which subsequently induce responses throughout the body. Exosomes are secreted by distinct kinds of cells, either as an initial response to disorders or subsequent responses to preceding signals (20). The exosomes discussed in this section are critical to various processes of CVD development as representative mechanisms for regulation induced by exosomes, but some are not directly connected with CVDs. Additionally, important elements are discussed to clarify the roles of exosomes in CVD diagnosis, treatment, prognosis, and other potential clinical tasks.

Exosomes derived from platelets

Platelet-derived exosomes contain coagulation proteins, anticoagulation proteins, bioactive lipids, and cytokines (21). These exosomes are known to promote and inhibit inflammation, thrombosis, tissue repair, angiogenesis, endothelial apoptosis, myocardial dysfunction, and atherosclerosis (AS) (22-26). Platelet-rich plasma exosomes can encapsulate growth factors, including platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β), vascular endothelial growth factor (VEGF), and basic fibroblast growth factor (bFGF) (26). In this way, these exosomes promote the proliferation and migration of endothelial cells by regulating the PI3K/Akt and MAPK-Erk signaling pathways to promote angiogenesis. Additionally, platelet-derived exosomes activate Yes-associated protein (YAP) and stimulate fibroblast proliferation, fibroblast migration, and collagen synthesis, collectively enhancing angiogenesis (26).

Platelet exosomes also contain miRNAs, including miR-21, miR-191, miR-233, miR-320, and miR-339 (27). These miRNAs affect platelet aggregation and blood coagulation, which in turn affect AS formation. miR-191, miR-233, and miR-320 in platelet-derived exosomes are anti-inflammatory and cardioprotective; these miRNAs decrease the release of inflammatory factors by inhibiting the expression of intercellular adhesion molecule-1 (ICAM-1), which suppresses AS (25,28,29). Furthermore, platelet-derived exosomes impede AS formation by inhibiting CD36 expression in macrophages, which prevents macrophages from phagocytosing oxidized low-density lipoprotein and cholesterol to decrease foam cell formation (30).

Exosomes derived from stem cells

Many types of stem cells produce exosomes that function to lessen CVDs in benchtop and animal studies. Hypoxic preconditioning increases the exosome secretion of umbilical cord (UC)-mesenchymal stem cells (MSCs). Treatment of H9C2 cells with UC-MSC-derived exosomes limits hypoxia- and serum depletion-mediated apoptosis. In addition, treatment with these exosomes downregulates LC3B-II/I and beclin-1 but upregulates P62, p-Akt/Akt, and p-mTOR/mTOR (31). Adipose-derived stem cell (ADSC) exosomes overexpressing miR-126 downregulate inflammatory factors and alleviate myocardial injury (32). Bone marrow stromal cell-derived exosomes protect cardiomyocytes from ischemia reperfusion injury. Exosome miR-486-5p inhibits cardiomyocyte apoptosis by suppressing PTEN expression and activating the PI3K/AKT signaling pathway (33).

Exosomes derived from cardiac cells

Most heart cells, including cardiomyocytes, cardiac fibroblasts, cardiac endothelial cells, and cardiac progenitor cells, secrete exosomes and modulate cardiac function (10). Cardiomyocyte-derived exosomes vary depending upon the clinical scenario.

Goto-Kakizaki type 2 diabetic rat cardiomyocyte exosomes inhibit the proliferation, migration, and tube-like formation of murine cardiac endothelial cells (MCECs) through miR-320. In contrast, exosomes derived from Wistar rat cardiomyocytes are proangiogenic (34). Additionally, cardiomyocyte-derived exosomes are crucial to miR-92a-mediated postinfarct cardiac fibroblast activation (35).

Cardiac progenitor cells (CPCs) have been studied in relation to heart repair and regeneration. miR-210 found in CPC-derived exosomes inhibits cardiomyocyte apoptosis-induced angiogenesis and improves function (36-38). CPC-derived exosomes also improve right ventricular heart failure and reduce postinfarction scar formation (39,40). Treatment with CPC-derived exosomes attenuates doxorubicin- and trastuzumab-induced cardiac oxidative stress, in part, via miR-146a-5p (41). Finally, CPC-derived exosomes containing the matricellular protein, periostin, promote cardiomyocyte cell cycle reentry (42). Cardiac fibroblasts secrete miRNA-enriched exosomes containing miR-21-3p/miR-21, which stimulate the production of angiotensin II (Ang II) and promote cardiomyocyte hypertrophy (6). In a feedback manner, Ang II, via endogenous receptor signaling, increases cardiac fibroblast-derived exosome release. These exosomes increase renin, angiotensinogen, AT1R, and AT2R but decrease angiotensin-converting enzyme 2, and they also stimulate cell hypertrophy via mitogen-activated protein kinases (MAPKs) and the Akt pathway (43).

Exosomes derived from macrophages

In mice, cardiac miR-155 expression is increased after myocardial infarction. Pri-miR-155 and miR-155 are present in macrophage-derived exosomes and cardiac fibroblasts. Exosomes containing Pri-mir-155 and miR-155 suppress Son of Sevenless 1, and they inhibit cardiac fibroblast proliferation. Additionally, these exosomes increase inflammation by decreasing the expression of suppressor of cytokine signaling 1 (SOCS1). In vivo, mir-155-deficient mice have a lower rate of cardiac rupture and better function than control mice; fibroblast proliferation and collagen production are elevated, and cardiac inflammation is reduced postinfarction (44). Macrophage foam cell-derived exosomes transport proteins, such as integrins, into vascular smooth muscle cells (VSMCs), activating the extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) pathways, and facilitating cell migration and adhesion to promote atherosclerosis (45).


Exosomes as biomarkers in CVD diagnosis and prognosis

The secretion of exosomes is an essential phenomenon that exists in both physiological and pathological processes. Different processes determine the surface molecules and contents of exosomes. Therefore, blood exosomes may mimic the health and disease of specific cell types and organs. Additionally, blood-borne exosomes are a convenient method to assess otherwise inaccessible tissues because they provide useful information for identifying and classifying individual risk conditions in patients, diagnosing and monitoring disease conditions, effectively adjusting treatments, and estimating prognosis. Thus, exosomes and their contents are viewed as biomarkers (46). Indeed, studies have suggested that exosomes are noninvasive biomarkers for CVDs (47,48).

Exosomes carry miRNAs, indicating that they participate in the circulation and presentation of miRNAs (49). The stable existence of miRNAs in body fluids is promoted by miRNAs binding with carriers, such as exosomes, which offer protection from degradation (50). Studies that have focused on miRNA biomarkers in CVD diagnosis and prognosis are summarized in Table 2.

Table 2

Important miRNAs for CVD diagnosis and prognosis

Disease miRNA Regulation Purpose Ref.
Acute myocardial infarction (AMI) miR-1 Diagnostic (51-55)
Prognostic: death (53)
Prognostic: LVEF (55)
miR-133a/b Diagnostic (52-54,56-60)
Prognostic: death (53)
Prognostic: left ventricle remodeling (59)
miR-208a/b Diagnostic (53,57,60-64)
Prognostic: death (53)
Prognostic: left ventricle remodeling (63)
miR-423-5p Diagnostic (54,59)
Prognostic: left ventricle remodeling (59)
miR-499 Diagnostic (52,54,60,61,65)
miR-126 Prognostic: AMI (66)
miR-21 Diagnostic (54,55)
miR-29b Diagnostic (55)
miR-106a-5p Prognostic: AMI (67)
miR-223 Prognostic: AMI (66)
Arrhythmia: tachycardia miR-1 Diagnostic (59)
Arrhythmia: atrial fibrillation (AF) miR-29b Diagnostic (68)
miR-150 Diagnostic (69)
miR-328 Diagnostic (70)
Prognostic: AF (70)
Arrhythmia: postoperative atrial fibrillation (POAF) miR-23a Prognostic: POAF (71)
miR-26a Prognostic: POAF (71)
Coronary artery disease (CAD) miR-1 Diagnostic (72)
miR-133a/b Diagnostic (72,73)
miR-208a/b Diagnostic (73)
miR-122 Diagnostic (72)
miR-126 Diagnostic (72,74)
Prognostic: major adverse cardiovascular event (74)
miR-134 Diagnostic (75)
miR-199a Diagnostic (72,74)
Prognostic: major adverse cardiovascular event (74)
miR-126 Diagnostic (73)
miR-145 Diagnostic (73)
miR-146a Diagnostic (76)
miR-30c/d Diagnostic (76)
Heart failure (HF) miR-1 Diagnostic (77)
miR-133a/b Diagnostic (77)
miR-208a/b Diagnostic (77)
miR-423-5p Diagnostic (78,79)
miR-499 Diagnostic (77)
miR-145 Diagnostic/prognostic: CRT (80)
miR-26a Diagnostic: response to CRT (80)
miR-1254 Diagnostic (81,82)
Diagnostic/prognostic: death and HF hospitalization (81)
miR-1306-5p Diagnostic (81,82)
Diagnostic/prognostic: death and HF hospitalization (81)
miR-106a-5p Diagnostic/prognostic: mortality (83)
miR-146a Diagnostic (84)
miR-199a Diagnostic/prognostic: mortality (83)
miR-26a Diagnostic/prognostic: mortality (83)
miR-27a Diagnostic/prognostic: mortality (83)
miR-30c/d Diagnostic (83,84)
Prognostic: mortality (83)
miR-328 Diagnostic (84)
miR-30d Diagnostic (85)
Prognostic: response to CRT (85)
Hypertrophic cardiomyopathy miR-199a Diagnostic (86)
miR-21 Diagnostic (87)
miR-27a Diagnostic (86)
miR-30c/d Diagnostic (87)
Takotsubo cardiomyopathy miR-1 Diagnostic (88)
miR-133a/b Diagnostic (88)
miR-26a Diagnostic (88)

↑, upregulated; ↓, downregulated. CVD, cardiovascular disease; AMI, acute myocardial infarction; LVEF, left ventricular ejection fraction; AF, atrial fibrillation; POAF, postoperative atrial fibrillation; CAD, coronary artery disease; CRT, cardiac resynchronization therapy; HF, heart failure.

Compared to miRNAs, exosome proteins are less studied as biomarkers.

In individuals with cerebrovascular disease, endothelial-derived exosomes show increased levels of vascular proteins, such as VCAM-1, von Willebrand factor, platelet-derived growth factor (PDGF)-BB, angiopoietin-1, and lysyl oxidase-2, in addition to cerebrovascular-selective proteins, including glucose transporter 1, permeability-glycoprotein, and large neutral amino acid transporter 1. In platelet-derived exosomes, PDGF-AA, platelet glycoprotein VI, integrin-linked kinase-1, high mobility group box-1 protein, CXCL4 (chemokine), and antiangiogenic thrombospondin-1 are increased. Elevation of these atherosclerosis-promoting proteins underscores the possible role of exosomes in tracking atherosclerotic cerebrovascular disease (89).


Exosomes as a CVD treatment method

The therapeutic effects of exosomes in treating CVDs are well documented in preclinical studies. The exosomes discussed in this section have the potential to be used in exosome therapies for CVDs. However, safety issues remain a concern in human trials due to the potential for off-target effects.

Potential therapeutic methods with direct administration of exosomes

The studies below consider exosomes of identified origin but indefinite cargo or identified and edited single cargos (Table 3). Intravenous or local injection of exosomes is the primary mode of administration.

Table 3

Potential therapeutic methods with direct administration of exosomes

Exosome origin Substance CVD Effect Model type Ref
MSC miR125b-5p MI Promoted repair of ischemic myocardium by limiting apoptosis Mouse exosome, mouse model (90)
MSC Reduced infarct size, facilitated functional recovery and increased neoangiogenesis Human exosome, mouse model (91,92)
MSC miR-25-3p Reduced cardiomyocyte apoptosis and inflammatory responses Mouse exosome, mouse model (in vivo), human primary cardiomyocytes (in vitro) (93)
GATA-4 overexpression MSC miR-19a Restored cardiac contractile function and decreased infarct size Mouse exosome, mouse model (94)
Human CD34+ cell Stimulated angiogenesis Human exosome, mouse model (95)
CD34Shh cell Shh protein Decreased infarct size and elevated border zone capillary density Human exosome, mouse model (96)
endothelial stem cell miR-294 Promoted CPC survival and proliferation Mouse exosome, mouse model (97)
MSC miR-133b Stroke Promoted neurogenesis, angiogenesis, and neurite remodeling Mouse exosome, mouse model (98)
M2 macrophage (Loaded with hexyl 5-aminolevulinate hydrochloride) AS Alleviated AS Mouse exosome, mouse model (99)
MSC miR-223 Sepsis Decreased inflammation and cell death Mouse exosome, mouse model (100)
Cardiomyocyte overexpressing heat shock protein 20 Cardiomyocyte Hypertrophy Improved the myocardial function of diabetic mice, preventing diabetes-caused cardiac injury Mouse exosome, mouse model (101)

CVD, cardiovascular disease; MSC, mesenchymal stem cell; MI, myocardial infarction; CPC, cardiac progenitor cell; AS, atherosclerosis.

Therapy for post-MI regeneration

The injured myocardium presents a variety of responses to injury, including necrosis, inflammation, apoptosis, remodeling, and fibrosis. In AMI, myocardial secretion of exosomes increases with increasing ischemia (102). In peri-infarcted areas, the paracrine effects of non-injured myocardium-derived exosomes may reprogram cardiomyocytes and rescue the peri-infarcted region by transferring molecules, such as RNAs and peptides, to alleviate necrosis, inflammation, apoptosis, remodeling, and fibrosis (103). In these situations, myocardial exosomes can activate the release of stem cells and exosomes derived from bone marrow, which, in a feedback fashion, can repair and restore damaged tissue (20).

In most animal studies, stem cell-derived exosomes are administered via intramyocardial delivery. Hypoxic MSC-derived exosomes promote repair of ischemic myocardium by limiting apoptosis. Moreover, miR-125b-5p mediates this effect (mouse exosomes in a mouse model) (90). Human CD34+ cell exosomes (which are a type of MSC) show a paracrine effect, stimulating angiogenesis (95). Similarly, CD34Shh cell-derived exosomes preserve murine cardiac function after acute myocardial infarction (human exosomes in a mouse model) (96). Following ischemia and reperfusion, MSC-derived exosomes increase ATP levels, reduce oxidative stress, and activate the PI3K/Akt pathway to enhance cardiomyocyte viability. Infarct size is reduced by 45% in animals treated with exosomes (human exosomes in a mouse model) (91). Moreover, combinatorial treatment with microvesicles and exosomes reduces infarct size, facilitates functional recovery, and increases neoangiogenesis (human exosomes in a mouse model) (92).

GATA-4 overexpression of MSC-derived exosomes restores cardiac contractile function and decreases infarct size effects mediated, in part, by anti-apoptotic miR-19a (mouse exosomes in a mouse model) (94).

Murine endothelial stem cell-derived exosomes modulate the cardiomyocyte and CPC-based repair program in the heart, and these exosomes promote CPC survival and proliferation. A significant increase in the miR290-295 cluster has been observed in exosomes, and miR-294 mediates this salutary effect (mouse exosomes in a mouse model) (97). MSC-derived exosomes mediate the transfer of miR-25-3p and ameliorate MI by targeting pro-apoptotic proteins and EZH2 (mouse exosomes in a mouse model; and human primary cardiomyocytes in vitro) (93).

Human CPC-derived exosomes inhibit cardiomyocyte apoptosis and promote cardiac function post-MI (human exosomes in a mouse model) (104).

Following direct injection, cardiosphere-derived exosomes improve cardiac function, promote angiogenesis, and inhibit apoptosis (human exosomes in a mouse model); miR-146a is enriched in these exosomes and may contribute to the beneficial effects (105). CPC-derived exosomes with high levels of GATA4-responsive miR-451 inhibit cardiomyocyte apoptosis (mouse exosomes in a mouse model) (106).

Therapy for stroke, AS, septic cardiomyopathy, and cardiomyocyte hypertrophy

In rats with experimentally induced stroke, intravenous administration of MSC-derived exosomes improves function and promotes neurogenesis, angiogenesis, and neurite remodeling via miR-133b (mouse exosomes in a mouse model) (98,107).

M2 macrophage-derived exosomes loaded with hexyl 5-aminolevulinate hydrochloride homes to areas of neural inflammation. Exosomes function in an anti-inflammatory manner to alleviate AS. In addition, exosomes are capable of imaging and tracking AS by fluorescence (mouse exosomes in a mouse model) (99).

In sepsis, MSC-derived exosome transfer of miR-223 to cardiomyocytes and macrophages decreases inflammation and cell death (mouse exosomes in a mouse model) (100).

Exosomes derived from heat shock protein 20 (Hsp20)-overexpressing cardiomyocytes significantly decrease cell hypertrophy and improve the myocardial function of diabetic mice (mouse exosomes in a mouse model) (101).

Potential therapeutic methods target exosome synthesis, processing, and release

Regulation of exosome synthesis, process, and release are important areas of fundamental research. However, such studies are hampered by the fine line between beneficial physiological roles and harmful pathological roles of exosomes. It can be difficult to clarify those relationships. Compared to the direct administration of exosomes with defined components, exosome regulation methods need additional verification before clinical applications.

Because certain exosomes exacerbate CVDs (108), one therapeutic approach is to limit the secretion of so-called detrimental exosomes. Ang II increases the levels of miR-21-3p in cardiac fibroblast-derived exosomes and promotes cardiomyocyte hypertrophy (6). Conversely, the exosome inhibitors, GW4869 and DMA, block the release of these exosomes to limit Ang II-mediated myocardial hypertrophy and cardiac fibrosis (rat exosomes in a rat model) (43).

Apart from inhibition, some drugs are applicable in cell or animal models as regulators of the release or composition of exosomes. Simvastatin regulates the release of cardiomyocyte-derived exosomes and alleviates Ang II-induced cardiac fibrosis. Additionally, in cardiomyocyte-derived exosomes after treatment with simvastatin, decorin is increased, and periostin is decreased (human exosomes, in vitro) (109). Additionally, carvedilol increases ABCA1 levels in serum exosomes interfering with NF-κB and Akt signaling to limit atherosclerosis (mouse exosomes in a mouse model) (110).

Exosomes as an adjuvant to improve the therapeutic effects of other drugs

Combination with other therapies to improve the therapeutic effect is another option for the therapeutic role of exosomes. Intramyocardial delivery of bone marrow MSC-derived exosomes followed by MSC transplantation into the heart improves cardiac function, decreases infarct size, and increases neovascularization compared to animals treated with MSCs alone (rat exosomes in a rat AMI model) (111).


Clinical trials of exosome therapy

Preclinical studies have shown the beneficial therapeutic effects of exosomes in CVDs (90-101). Nevertheless, clinical investigations of exosome-based therapy are rare.

The NCT03384433 clinical trial (recruiting) entitled, “Allogenic Mesenchymal Stem Cell Derived Exosome in Patients with Acute Ischemic Stroke”, aims to evaluate the effects of allogenic MSC-derived exosomes in the treatment of acute ischemic stroke. Therapeutic exosomes are enriched with miR-124 by miR-124 transfection via stereotaxic administration. The NCT03478410 clinical trial entitled, “Role of Exosomes Derived from Epicardial Fat in Atrial Fibrillation”, aims to assess the biomarker and therapeutic potential of epicardial fat-derived exosomes in patients with atrial fibrillation. The NCT04356300 clinical trial entitled, “Exosome of Mesenchymal Stem Cells for Multiple Organ Dysfunction Syndrome (MODS) After Surgical Repair of Acute Type A Aortic Dissection”, aims to evaluate the safety and therapeutic efficiency of MSC-derived exosomes after surgical repair for acute type A aortic dissection. The NCT04356300 clinical trial has two parts. First, 150 mg of MSC-derived exosomes will be administered i.v. immediately after ascending aortic replacement followed by continuous administration of 150 mg of exosomes daily.


Conclusions

Exosomes are generated from different cells under specific pathophysiological conditions, and they function as carriers of signaling molecules, such as miRNAs and proteins. Exosomes are involved in CVDs by transporting specific exosomal cargo, either promoting or limiting disease.

Exosome cargo is specific to the situation and cell or origin, providing opportunities for developing novel diagnostic and prognostic methods. Studies have indicated that changes in exosomal cargo, such as miRNAs, are of significance in several pathologies, including AMI, arrhythmia, AF, CAD, HF, and cardiomyopathy, thus suggesting a role in the diagnosis of these conditions. Although many clinical studies concentrate on diagnostic and prognostic roles of exosome contents, shortcomings including relatively small cohorts of patients, different methods of contents extraction, and lack of consistency and standardization are major obstacles in front of its clinical applications (112). Taking all those factors into consideration and making a consensus on the standard experimental design at the start of a study might be one of the possible solutions.

For exosomes therapy, despite a large amount of published data from cell and rodent studies, exosome-based therapies are only now being tested in clinical trials. As safety is the major concern of clinical applications, preventing off-target effects and determining appropriate doses are of vital importance. Validation is needed before translation and wide use.

Finally, no matter for diagnosis, treatment or prognosis, lack of sufficient knowledge about the function and mechanisms of CVD-related exosomes will be hurdles for its development and application. Therefore further basic science researches on exosomes are indispensable for boosting the progress of exosome applications.


Acknowledgments

Funding: This work was supported by the National Natural Science Foundation of China (No. NSFC81860057).


Footnote

Reporting Checklist: Both authors have completed the Narrative Review reporting checklist. Available at https://atm.amegroups.com/article/view/10.21037/atm-22-619/rc

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://atm.amegroups.com/article/view/10.21037/atm-22-619/coif). The authors report that this work was supported by the National Natural Science Foundation of China (No. NSFC81860057). The authors have no other conflicts of interest to declare

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Virani SS, Alonso A, Benjamin EJ, et al. Heart Disease and Stroke Statistics-2020 Update: A Report From the American Heart Association. Circulation 2020;141:e139-596. [Crossref] [PubMed]
  2. Enroth S, Johansson A, Enroth SB, et al. Strong effects of genetic and lifestyle factors on biomarker variation and use of personalized cutoffs. Nat Commun 2014;5:4684. [Crossref] [PubMed]
  3. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science 2020;367:eaau6977. [Crossref] [PubMed]
  4. van Niel G, D'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol 2018;19:213-28. [Crossref] [PubMed]
  5. Yu H, Wang Z. Cardiomyocyte-Derived Exosomes: Biological Functions and Potential Therapeutic Implications. Front Physiol 2019;10:1049. [Crossref] [PubMed]
  6. Bang C, Batkai S, Dangwal S, et al. Cardiac fibroblast-derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J Clin Invest 2014;124:2136-46. [Crossref] [PubMed]
  7. Valadi H, Ekström K, Bossios A, et al. Exosome-mediated transfer of mRNAs and mi-croRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007;9:654-9. [Crossref] [PubMed]
  8. Fritz JV, Heintz-Buschart A, Ghosal A, et al. Sources and Functions of Extracellular Small RNAs in Human Circulation. Annu Rev Nutr 2016;36:301-36. [Crossref] [PubMed]
  9. Zhang Y, Bi J, Huang J, et al. Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications. Int J Nanomedicine 2020;15:6917-34. [Crossref] [PubMed]
  10. Barile L, Moccetti T, Marbán E, et al. Roles of exosomes in cardioprotection. Eur Heart J 2017;38:1372-9. [PubMed]
  11. Bhatnagar S, Schorey JS. Exosomes released from infected macrophages contain My-cobacterium avium glycopeptidolipids and are proinflammatory. J Biol Chem 2007;282:25779-89. [Crossref] [PubMed]
  12. Sun HJ, Zhu XX, Cai WW, et al. Functional roles of exosomes in cardiovascular disorders: a systematic review. Eur Rev Med Pharmacol Sci 2017;21:5197-206. [PubMed]
  13. Witwer KW, Buzás EI, Bemis LT, et al. Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles 2013; [Crossref] [PubMed]
  14. Lai CP, Breakefield XO. Role of exosomes/microvesicles in the nervous system and use in emerging therapies. Front Physiol 2012;3:228. [Crossref] [PubMed]
  15. Pinky Gupta S. Mesenchymal Stem Cell Derived Exosomes: a Nano Platform for Therapeutics and Drug Delivery in Combating COVID-19. Stem Cell Rev Rep 2021;17:33-43. [Crossref] [PubMed]
  16. Yang B, Chen Y, Shi J. Exosome Biochemistry and Advanced Nanotechnology for Next-Generation Theranostic Platforms. Adv Mater 2019;31:e1802896. [Crossref] [PubMed]
  17. Femminò S, Penna C, Margarita S, et al. Extracellular vesicles and cardiovascular system: Biomarkers and Cardioprotective Effectors. Vascul Pharmacol 2020;135:106790. [Crossref] [PubMed]
  18. Pegtel DM, Gould SJ. Exosomes. Annu Rev Biochem 2019;88:487-514. [Crossref] [PubMed]
  19. Hoefer IE, Steffens S, Ala-Korpela M, et al. Novel methodologies for biomarker discovery in atherosclerosis. Eur Heart J 2015;36:2635-42. [Crossref] [PubMed]
  20. Huang P, Tian X, Li Q, et al. New strategies for improving stem cell therapy in ischemic heart disease. Heart Fail Rev 2016;21:737-52. [Crossref] [PubMed]
  21. Barry OP, Praticò D, Savani RC, et al. Modulation of monocyte-endothelial cell interactions by platelet microparticles. J Clin Invest 1998;102:136-44. [Crossref] [PubMed]
  22. Wang Y, Xie Y, Zhang A, et al. Exosomes: An emerging factor in atherosclerosis. Biomed Pharmacother 2019;115:108951. [Crossref] [PubMed]
  23. Azevedo LC, Janiszewski M, Pontieri V, et al. Platelet-derived exosomes from septic shock patients induce myocardial dysfunction. Crit Care 2007;11:R120. [Crossref] [PubMed]
  24. Gambim MH, do Carmo Ade O, Marti L, et al. Platelet-derived exosomes induce endothelial cell apoptosis through peroxynitrite generation: experimental evidence for a novel mechanism of septic vascular dysfunction. Crit Care 2007;11:R107. [Crossref] [PubMed]
  25. Li J, Tan M, Xiang Q, et al. Thrombin-activated platelet-derived exosomes regulate en-dothelial cell expression of ICAM-1 via microRNA-223 during the thrombosis-inflammation response. Thromb Res 2017;154:96-105. [Crossref] [PubMed]
  26. Guo SC, Tao SC, Yin WJ, et al. Exosomes derived from platelet-rich plasma promote the re-epithelization of chronic cutaneous wounds via activation of YAP in a diabetic rat model. Theranostics 2017;7:81-96. [Crossref] [PubMed]
  27. Tan M, Yan HB, Li JN, et al. Thrombin Stimulated Platelet-Derived Exosomes Inhibit Platelet-Derived Growth Factor Receptor-Beta Expression in Vascular Smooth Muscle Cells. Cell Physiol Biochem 2016;38:2348-65. [Crossref] [PubMed]
  28. Gidlöf O, van der Brug M, Ohman J, et al. Platelets activated during myocardial infarction release functional miRNA, which can be taken up by endothelial cells and regulate ICAM1 expression. Blood 2013;121:3908-17, S1-26.
  29. Chistiakov DA, Orekhov AN, Bobryshev YV. Extracellular vesicles and atherosclerotic disease. Cell Mol Life Sci 2015;72:2697-708. [Crossref] [PubMed]
  30. Srikanthan S, Li W, Silverstein RL, et al. Exosome poly-ubiquitin inhibits platelet activation, downregulates CD36 and inhibits pro-atherothombotic cellular functions. J Thromb Haemost 2014;12:1906-17. [Crossref] [PubMed]
  31. Liu H, Sun X, Gong X, et al. Human umbilical cord mesenchymal stem cells derived ex-osomes exert antiapoptosis effect via activating PI3K/Akt/mTOR pathway on H9C2 cells. J Cell Biochem 2019;120:14455-64. [Crossref] [PubMed]
  32. Luo Q, Guo D, Liu G, et al. Exosomes from MiR-126-Overexpressing Adscs Are Thera-peutic in Relieving Acute Myocardial Ischaemic Injury. Cell Physiol Biochem 2017;44:2105-16. [Crossref] [PubMed]
  33. Sun X-H, Wang X, Zhang Y, et al. Exosomes of bone-marrow stromal cells inhibit cardiomyocyte apoptosis under ischemic and hypoxic conditions via miR-486-5p targeting the PTEN/PI3K/AKT signaling pathway. Thromb Res 2019;177:23-32. [Crossref] [PubMed]
  34. Wang X, Huang W, Liu G, et al. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic rats through the exosomal transfer of miR-320 into endothelial cells. J Mol Cell Cardiol 2014;74:139-50. [Crossref] [PubMed]
  35. Wang X, Morelli MB, Matarese A, et al. Cardiomyocyte-derived exosomal microRNA-92a mediates post-ischemic myofibroblast activation both in vitro and ex vivo. ESC Heart Fail 2020;7:284-8. [Crossref] [PubMed]
  36. Shi YF, Liu N, Li YX, et al. Insulin protects H9c2 rat cardiomyoblast cells against hydrogen peroxide-induced injury through upregulation of microRNA-210. Free Radic Res 2015;49:1147-55. [Crossref] [PubMed]
  37. Muralimanoharan S, Guo C, Myatt L, et al. Sexual dimorphism in miR-210 expression and mitochondrial dysfunction in the placenta with maternal obesity. Int J Obes (Lond) 2015;39:1274-81. [Crossref] [PubMed]
  38. Mutharasan RK, Nagpal V, Ichikawa Y, et al. microRNA-210 is upregulated in hypoxic cardiomyocytes through Akt- and p53-dependent pathways and exerts cytoprotective effects. Am J Physiol Heart Circ Physiol 2011;301:H1519-30. [Crossref] [PubMed]
  39. Trac D, Hoffman JR, Bheri S, et al. Predicting Functional Responses of Progenitor Cell Exosome Potential with Computational Modeling. Stem Cells Transl Med 2019;8:1212-21. [Crossref] [PubMed]
  40. Ciullo A, Biemmi V, Milano G, et al. Exosomal Expression of CXCR4 Targets Cardioprotective Vesicles to Myocardial Infarction and Improves Outcome after Systemic Administration. Int J Mol Sci 2019;20. [PubMed]
  41. Milano G, Biemmi V, Lazzarini E, et al. Intravenous administration of cardiac progenitor cell-derived exosomes protects against doxorubicin/trastuzumab-induced cardiac toxicity. Cardiovasc Res 2020;116:383-92. [PubMed]
  42. Balbi C, Milano G, Fertig TE, et al. An exosomal-carried short periostin isoform induces cardiomyocyte proliferation. Theranostics 2021;11:5634-49. [Crossref] [PubMed]
  43. Lyu L, Wang H, Li B, et al. A critical role of cardiac fibroblast-derived exosomes in activating renin angiotensin system in cardiomyocytes. J Mol Cell Cardiol 2015;89:268-79. [Crossref] [PubMed]
  44. Wang C, Zhang C, Liu L, et al. Macrophage-Derived mir-155-Containing Exosomes Suppress Fibroblast Proliferation and Promote Fibroblast Inflammation during Cardiac Injury. Mol Ther 2017;25:192-204. [Crossref] [PubMed]
  45. Niu C, Wang X, Zhao M, et al. Macrophage Foam Cell-Derived Extracellular Vesicles Promote Vascular Smooth Muscle Cell Migration and Adhesion. J Am Heart Assoc 2016; [Crossref] [PubMed]
  46. He C, Zheng S, Luo Y, et al. Exosome Theranostics: Biology and Translational Medicine. Theranostics 2018;8:237-55. [Crossref] [PubMed]
  47. Viereck J, Thum T. Circulating Noncoding RNAs as Biomarkers of Cardiovascular Disease and Injury. Circ Res 2017;120:381-99. [Crossref] [PubMed]
  48. Lu M, Yuan S, Li S, et al. The Exosome-Derived Biomarker in Atherosclerosis and Its Clinical Application. J Cardiovasc Transl Res 2019;12:68-74. [Crossref] [PubMed]
  49. Weber JA, Baxter DH, Zhang S, et al. The microRNA spectrum in 12 body fluids. Clin Chem 2010;56:1733-41. [Crossref] [PubMed]
  50. Li Q, Shao Y, Zhang X, et al. Plasma long noncoding RNA protected by exosomes as a potential stable biomarker for gastric cancer. Tumour Biol 2015;36:2007-12. [Crossref] [PubMed]
  51. Cheng Y, Tan N, Yang J, et al. A translational study of circulating cell-free microRNA-1 in acute myocardial infarction. Clin Sci (Lond) 2010;119:87-95. [Crossref] [PubMed]
  52. D'Alessandra Y, Devanna P, Limana F, et al. Circulating microRNAs are new and sensitive biomarkers of myocardial infarction. Eur Heart J 2010;31:2765-73. [Crossref] [PubMed]
  53. Widera C, Gupta SK, Lorenzen JM, et al. Diagnostic and prognostic impact of six circulating microRNAs in acute coronary syndrome. J Mol Cell Cardiol 2011;51:872-5. [Crossref] [PubMed]
  54. Olivieri F, Antonicelli R, Lorenzi M, et al. Diagnostic potential of circulating miR-499-5p in elderly patients with acute non ST-elevation myocardial infarction. Int J Cardiol 2013;167:531-6. [Crossref] [PubMed]
  55. Grabmaier U, Clauss S, Gross L, et al. Diagnostic and prognostic value of miR-1 and miR-29b on adverse ventricular remodeling after acute myocardial infarction - The SITAGRAMI-miR analysis. Int J Cardiol 2017;244:30-6. [Crossref] [PubMed]
  56. Kuwabara Y, Ono K, Horie T, et al. Increased microRNA-1 and microRNA-133a levels in serum of patients with cardiovascular disease indicate myocardial damage. Circ Cardiovasc Genet 2011;4:446-54. [Crossref] [PubMed]
  57. Gidlöf O, Andersson P, van der Pals J, et al. Cardiospecific microRNA plasma levels correlate with troponin and cardiac function in patients with ST elevation myocardial infarction, are selectively dependent on renal elimination, and can be detected in urine samples. Cardiology 2011;118:217-26. [Crossref] [PubMed]
  58. Wang F, Long G, Zhao C, et al. Plasma microRNA-133a is a new marker for both acute myocardial infarction and underlying coronary artery stenosis. J Transl Med 2013;11:222. [Crossref] [PubMed]
  59. Bauters C, Kumarswamy R, Holzmann A, et al. Circulating miR-133a and miR-423-5p fail as biomarkers for left ventricular remodeling after myocardial infarction. Int J Cardiol 2013;168:1837-40. [Crossref] [PubMed]
  60. Li YQ, Zhang MF, Wen HY, et al. Comparing the diagnostic values of circulating mi-croRNAs and cardiac troponin T in patients with acute myocardial infarction. Clinics (Sao Paulo, Brazil) 2013;68:75-80. [Crossref] [PubMed]
  61. Liu X, Fan Z, Zhao T, et al. Plasma miR-1, miR-208, miR-499 as potential predictive biomarkers for acute myocardial infarction: An independent study of Han population. Exp Gerontol 2015;72:230-8. [Crossref] [PubMed]
  62. Wang GK, Zhu JQ, Zhang JT, et al. Circulating microRNA: a novel potential biomarker for early diagnosis of acute myocardial infarction in humans. Eur Heart J 2010;31:659-66. [Crossref] [PubMed]
  63. Lv P, Zhou M, He J, et al. Circulating miR-208b and miR-34a are associated with left ventricular remodeling after acute myocardial infarction. Int J Mol Sci 2014;15:5774-88. [Crossref] [PubMed]
  64. Corsten MF, Dennert R, Jochems S, et al. Circulating MicroRNA-208b and MicroRNA-499 reflect myocardial damage in cardiovascular disease. Circ Cardiovasc Genet 2010;3:499-506. [Crossref] [PubMed]
  65. Adachi T, Nakanishi M, Otsuka Y, et al. Plasma microRNA 499 as a biomarker of acute myocardial infarction. Clin Chem 2010;56:1183-5. [Crossref] [PubMed]
  66. Zampetaki A, Willeit P, Tilling L, et al. Prospective study on circulating MicroRNAs and risk of myocardial infarction. J Am Coll Cardiol 2012;60:290-9. [Crossref] [PubMed]
  67. Bye A, Røsjø H, Nauman J, et al. Circulating microRNAs predict future fatal myocardial infarction in healthy individuals - The HUNT study. J Mol Cell Cardiol 2016;97:162-8. [Crossref] [PubMed]
  68. Dawson K, Wakili R, Ordög B, et al. MicroRNA29: a mechanistic contributor and potential biomarker in atrial fibrillation. Circulation 2013;127:1466-75, 1475e1-28.
  69. McManus DD, Tanriverdi K, Lin H, et al. Plasma microRNAs are associated with atrial fibrillation and change after catheter ablation (the miRhythm study). Heart Rhythm 2015;12:3-10. [Crossref] [PubMed]
  70. McManus DD, Lin H, Tanriverdi K, et al. Relations between circulating microRNAs and atrial fibrillation: data from the Framingham Offspring Study. Heart Rhythm 2014;11:663-9. [Crossref] [PubMed]
  71. Harling L, Lambert J, Ashrafian H, et al. Elevated serum microRNA 483-5p levels may predict patients at risk of post-operative atrial fibrillation. Eur J Cardiothorac Surg 2017;51:73-8. [Crossref] [PubMed]
  72. D'Alessandra Y, Carena MC, Spazzafumo L, et al. Diagnostic potential of plasmatic Mi-croRNA signatures in stable and unstable angina. PLoS One 2013;8:e80345. [Crossref] [PubMed]
  73. Fichtlscherer S, De Rosa S, Fox H, et al. Circulating microRNAs in patients with coronary artery disease. Circ Res 2010;107:677-84. [Crossref] [PubMed]
  74. Jansen F, Yang X, Proebsting S, et al. MicroRNA expression in circulating microvesicles predicts cardiovascular events in patients with coronary artery disease. J Am Heart Assoc 2014;3:e001249. [Crossref] [PubMed]
  75. Liu W, Ling S, Sun W, et al. Circulating microRNAs correlated with the level of coronary artery calcification in symptomatic patients. Sci Rep 2015;5:16099. [Crossref] [PubMed]
  76. Wagner J, Riwanto M, Besler C, et al. Characterization of levels and cellular transfer of circulating lipoprotein-bound microRNAs. Arterioscler Thromb Vasc Biol 2013;33:1392-400. [Crossref] [PubMed]
  77. Akat KM, Moore-McGriff DV, Morozov P, et al. Comparative RNA-sequencing analysis of myocardial and circulating small RNAs in human heart failure and their utility as bi-omarkers. Proc Natl Acad Sci U S A 2014;111:11151-6. [Crossref] [PubMed]
  78. Tijsen AJ, Creemers EE, Moerland PD, et al. MiR423-5p as a circulating biomarker for heart failure. Circ Res 2010;106:1035-9. [Crossref] [PubMed]
  79. Goren Y, Kushnir M, Zafrir B, et al. Serum levels of microRNAs in patients with heart failure. Eur J Heart Fail 2012;14:147-54. [Crossref] [PubMed]
  80. Marfella R, Di Filippo C, Potenza N, et al. Circulating microRNA changes in heart failure patients treated with cardiac resynchronization therapy: responders vs. non-responders. Eur J Heart Fail 2013;15:1277-88. [Crossref] [PubMed]
  81. Bayés-Genis A, Lanfear DE, de Ronde MWJ, et al. Prognostic value of circulating microRNAs on heart failure-related morbidity and mortality in two large diverse cohorts of general heart failure patients. Eur J Heart Fail 2018;20:67-75. [Crossref] [PubMed]
  82. van Boven N, Kardys I, van Vark LC, et al. Serially measured circulating microRNAs and adverse clinical outcomes in patients with acute heart failure. Eur J Heart Fail 2018;20:89-96. [Crossref] [PubMed]
  83. Ovchinnikova ES, Schmitter D, Vegter EL, et al. Signature of circulating microRNAs in patients with acute heart failure. Eur J Heart Fail 2016;18:414-23. [Crossref] [PubMed]
  84. Watson CJ, Gupta SK, O'Connell E, et al. MicroRNA signatures differentiate preserved from reduced ejection fraction heart failure. Eur J Heart Fail 2015;17:405-15. [Crossref] [PubMed]
  85. Melman YF, Shah R, Danielson K, et al. Circulating MicroRNA-30d Is Associated With Response to Cardiac Resynchronization Therapy in Heart Failure and Regulates Cardiomyocyte Apoptosis: A Translational Pilot Study. Circulation 2015;131:2202-16. [Crossref] [PubMed]
  86. Roncarati R, Viviani Anselmi C, Losi MA, et al. Circulating miR-29a, among other up-regulated microRNAs, is the only biomarker for both hypertrophy and fibrosis in patients with hypertrophic cardiomyopathy. J Am Coll Cardiol 2014;63:920-7. [Crossref] [PubMed]
  87. Fang L, Ellims AH, Moore XL, et al. Circulating microRNAs as biomarkers for diffuse myocardial fibrosis in patients with hypertrophic cardiomyopathy. J Transl Med 2015;13:314. [Crossref] [PubMed]
  88. Jaguszewski M, Osipova J, Ghadri JR, et al. A signature of circulating microRNAs differentiates takotsubo cardiomyopathy from acute myocardial infarction. Eur Heart J 2014;35:999-1006. [Crossref] [PubMed]
  89. Goetzl EJ, Schwartz JB, Mustapic M, et al. Altered cargo proteins of human plasma endothelial cell-derived exosomes in atherosclerotic cerebrovascular disease. FASEB J 2017;31:3689-94. [Crossref] [PubMed]
  90. Zhu LP, Tian T, Wang JY, 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;8:6163-77. [Crossref] [PubMed]
  91. Arslan F, Lai RC, Smeets MB, et al. Mesenchymal stem cell-derived exosomes increase ATP levels, decrease oxidative stress and activate PI3K/Akt pathway to enhance myocardial viability and prevent adverse remodeling after myocardial ischemia/reperfusion injury. Stem Cell Res 2013;10:301-12. [Crossref] [PubMed]
  92. Bian S, Zhang L, Duan L, et al. Extracellular vesicles derived from human bone marrow mesenchymal stem cells promote angiogenesis in a rat myocardial infarction model. J Mol Med (Berl) 2014;92:387-97. [Crossref] [PubMed]
  93. Peng Y, Zhao JL, Peng ZY, et al. Exosomal miR-25-3p from mesenchymal stem cells alleviates myocardial infarction by targeting pro-apoptotic proteins and EZH2. Cell Death Dis 2020;11:317. [Crossref] [PubMed]
  94. Yu B, Kim HW, Gong M, et al. Exosomes secreted from GATA-4 overexpressing mesenchymal stem cells serve as a reservoir of anti-apoptotic microRNAs for cardioprotection. Int J Cardiol 2015;182:349-60. [Crossref] [PubMed]
  95. Sahoo S, Klychko E, Thorne T, et al. Exosomes from human CD34(+) stem cells mediate their proangiogenic paracrine activity. Circ Res 2011;109:724-8. [Crossref] [PubMed]
  96. Mackie AR, Klyachko E, Thorne T, et al. Sonic hedgehog-modified human CD34+ cells preserve cardiac function after acute myocardial infarction. Circ Res 2012;111:312-21. [Crossref] [PubMed]
  97. Khan M, Nickoloff E, Abramova T, et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res 2015;117:52-64. [Crossref] [PubMed]
  98. Xin H, Li Y, Liu Z, et al. MiR-133b promotes neural plasticity and functional recovery after treatment of stroke with multipotent mesenchymal stromal cells in rats via transfer of exosome-enriched extracellular particles. Stem Cells 2013;31:2737-46. [Crossref] [PubMed]
  99. Wu G, Zhang J, Zhao Q, et al. Molecularly Engineered Macrophage-Derived Exosomes with Inflammation Tropism and Intrinsic Heme Biosynthesis for Atherosclerosis Treatment. Angew Chem Int Ed Engl 2020;59:4068-74. [Crossref] [PubMed]
  100. Wang X, Gu H, Qin D, et al. Exosomal miR-223 Contributes to Mesenchymal Stem Cell-Elicited Cardioprotection in Polymicrobial Sepsis. Sci Rep 2015;5:13721. [Crossref] [PubMed]
  101. Wang X, Gu H, Huang W, et al. Hsp20-Mediated Activation of Exosome Biogenesis in Cardiomyocytes Improves Cardiac Function and Angiogenesis in Diabetic Mice. Diabetes 2016;65:3111-28. [Crossref] [PubMed]
  102. Lazar E, Benedek T, Korodi S, et al. Stem cell-derived exosomes - an emerging tool for myocardial regeneration. World J Stem Cells 2018;10:106-15. [Crossref] [PubMed]
  103. Walsh DG, Kaplan LR, Burney RE, et al. Use of tissue plasminogen activator in the emergency department for acute myocardial infarction. Ann Emerg Med 1987;16:243-7. [Crossref] [PubMed]
  104. Barile L, Lionetti V, Cervio E, et al. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovasc Res 2014;103:530-41. [Crossref] [PubMed]
  105. Ibrahim AG, Cheng K, Marbán E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Reports 2014;2:606-19. [Crossref] [PubMed]
  106. Chen L, Wang Y, Pan Y, et al. Cardiac progenitor-derived exosomes protect ischemic myocardium from acute ischemia/reperfusion injury. Biochem Biophys Res Commun 2013;431:566-71. [Crossref] [PubMed]
  107. Xin H, Li Y, Cui Y, et al. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab 2013;33:1711-5. [Crossref] [PubMed]
  108. Anderson HC, Mulhall D, Garimella R. Role of extracellular membrane vesicles in the pathogenesis of various diseases, including cancer, renal diseases, atherosclerosis, and arthritis. Lab Invest 2010;90:1549-57. [Crossref] [PubMed]
  109. Kuo HF, Hsieh CC, Wang SC, et al. Simvastatin Attenuates Cardiac Fibrosis via Regulation of Cardiomyocyte-Derived Exosome Secretion. J Clin Med 2019;8:794. [Crossref] [PubMed]
  110. Chen SJ, Tsui PF, Chuang YP, et al. Carvedilol Ameliorates Experimental Atherosclerosis by Regulating Cholesterol Efflux and Exosome Functions. Int J Mol Sci 2019;20:5202. [Crossref] [PubMed]
  111. Huang P, Wang L, Li Q, et al. Combinatorial treatment of acute myocardial infarction using stem cells and their derived exosomes resulted in improved heart performance. Stem Cell Res Ther 2019;10:300. [Crossref] [PubMed]
  112. Moldovan L, Batte KE, Trgovcich J, et al. Methodological challenges in utilizing miRNAs as circulating biomarkers. J Cell Mol Med 2014;18:371-90. [Crossref] [PubMed]
Cite this article as: Chen X, Luo Q. Potential clinical applications of exosomes in the diagnosis, treatment, and prognosis of cardiovascular diseases: a narrative review. Ann Transl Med 2022;10(6):372. doi: 10.21037/atm-22-619

Download Citation