IMR Press / RCM / Volume 23 / Issue 9 / DOI: 10.31083/j.rcm2309310
Open Access Review
Mesenchymal Stem Cell-Derived Exosomes in Cardioprotection: A Novel Application to Prevent Myocardial Injury
Show Less
1 Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, 130021 Changchun, Jilin, China
2 Department of Thoracic Surgery, The First Hospital of Jilin University, 130021 Changchun, Jilin, China
*Correspondence: l_w01@jlu.edu.cn (Wei Liu); wei_shixiong@163.com (Shixiong Wei)
These authors contributed equally.
Academic Editor: Vincenzo Lionetti
Rev. Cardiovasc. Med. 2022, 23(9), 310; https://doi.org/10.31083/j.rcm2309310
Submitted: 29 May 2022 | Revised: 3 July 2022 | Accepted: 19 July 2022 | Published: 13 September 2022
(This article belongs to the Special Issue Translational Perioperative Cardioprotection)
Copyright: © 2022 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Perioperative myocardial injury is a common complication caused by major surgery. Many pharmacological and nonpharmacological studies have investigated perioperative cardioprotection. However, the methods are insufficient to meet the increasing clinical needs for cardioprotection. The application of Mesenchymal Stem Cell-Derived Exosomes (MSC-Exos) is a novel cell-free therapeutic strategy and has significantly benefitted patients suffering from various diseases. In this review, we comprehensively analyzed the application of MSC-Exos to prevent myocardial infarction/injury by regulating inflammatory reactions, inhibiting cardiomyocyte apoptosis and autophagy, promoting angiogenesis, and mediating cardiac remodeling. Finally, we assessed the therapeutic effects and the challenges associated with the application of MSC-Exos from a clinical perspective.

Keywords
mesenchymal stem cell
exosomes
cardioprotection
myocardial injury
1. Introduction

In an aging population, many perioperative patients suffer from cerebro-cardiovascular diseases, which result in high morbidity and mortality due to perioperative myocardial infarction (PMI) during anesthesia and surgery. PMI is a severe cardiovascular complication and contributes to non-fatal myocardial infarction, non-fatal cardiac arrest, and perioperative cardiac death in around 500,000~900,000 individuals, and also increases the risk of death due to cardiovascular complications every year in the first six months after major non-cardiac surgery [1, 2]. Irreversible short-term and long-term adverse outcomes caused by PMI increase the clinical need for perioperative cardioprotection during major surgery.

Perioperative cardioprotection has been applied for many years in cardiac and non-cardiac surgery and consists of pharmacological treatments, including beta-blockers, statins, alpha-2 agonists, aspirin, inhalation anesthetics, noble gases, and opioids [3], and nonpharmacological treatments, such as ischemic preconditioning (IPC), remote ischemic preconditioning (RIPC), and remote ischemic postconditioning (RIPostC) [4]. However, perioperative cardioprotection in cardiac and non-cardiac surgery remains a debated topic. Recently, mesenchymal stem cell therapy, which depends on the ability of self-renewal and secretion of regenerative cytokines, has been incorporated into the main therapeutic approaches in the regenerative medicine of cardiovascular diseases [5]. However, the problem of storage and transportation, and the risks of inducing tumorigenesis and deformity need to be addressed [6]. Exosomes primarily contribute to the efficacy of stem cells and are stable, easily stored, and not rejected by the immune system [7]. Mesenchymal stem cell-derived exosomes (MSC-Exos) were developed as a kind of novel cell-free therapy. They preserve the main biological features and functions of the parent cells and exhibit a strong cardioprotective effect [8]. We reviewed the studies related to MSC-Exos to improve the treatment of myocardial ischemia and investigated their ability to provide perioperative cardioprotection.

2. Mechanisms Underlying Perioperative Myocardial Injury

PMI is a kind of myocardial ischemia that mainly occurs during or a few days after surgery and might occur due to the usage of intense analgesia. Nearly 80% of patients sustaining PMI only show symptoms based on cardiac troponin but lack other typical ischemic symptoms, such as chest pain and changes in the ECG [9, 10]. Few PMI patients present atherosclerotic plaque rupture with thrombus formation and distal embolization. The flow-mediated hypoperfusion and supply-demand imbalance of oxygen promote PMI [11, 12].

3. The Biological Characteristics of Mesenchymal Stem Cell-Derived Exosomes

Mesenchymal stem cells are found in many tissues, including adipose tissue, bone marrow, placenta, heart, peripheral blood, and umbilical cord [13]. They can regenerate by dividing and differentiating into several kinds of cells [14]. The application of MSCs in cardiovascular diseases has advanced considerably [5]. Exosomes, containing RNA, DNA, proteins, and lipids, are nano-sized lipid bilayer vesicles of endosomal compartments [15]. The biogenesis of exosomes is shown in Fig. 1. Besides having various exosome biogenesis-related functional proteins, MSC-derived exosomes contain surface markers, such as CD9, CD44, CD63, CD73, CD81, and CD90, specific markers of MSCs, proteins that act as signaling molecules [16, 17], and more than 850 unique gene products and miRNAs [18, 19]. Certain RNA cargos (mRNA and microRNA) that are sorted into MSC-derived exosomes are important for angiogenesis, cell differentiation, cell proliferation, cell survival, tissue remodeling, and immune system modulation [20, 21]. According to the results of RNA sequencing, MSC-Exos, isolated from different tissues, were found to have various species of tRNA [22] that affected the differences in the clinical efficacy of MSC-Exos. The five most abundant miRNAs in adipose-derived MSC (ASC) exosomes are miR-486–5p, miR-10a-5p, miR-10b-5p, miR-191–5p, and miR-222–3p. In bone marrow-derived MSCs (BMSCs), exosomes contain miR-143–3p, miR-10b-5p, miR-486–5p, miR-22–3p, and miR-21–5p. The miRNA sequencing data showed that the cardioprotection provided by endometrial MSCs was better than that provided by BMSCs and adipose-derived MSCs [23]; the cardioprotection-related miRNAs were upregulated (miR-29 and miR-24), while the cardiac-damage related miRNAs were downregulated (miR-21 and miR-15) [8, 24, 25].

Fig. 1.

The biogenesis of MSC-Exos. First, the fusion of endocytic vesicles forms the early endosome. Then, early endosomes transform into multivesicular bodies. Finally, multivesicular bodies fuse with the plasma membrane to release exosomes via membrane budding. The MVBs might be transported to the Golgi for recycling endosomes and delivered to lysosomes for degradation.

4. Cardioprotection of Mesenchymal Stem Cell-Derived Exosomes
4.1 MSC-Exos Regulate Inflammatory Reactions

The inflammatory cascade plays a pivotal role in the myocardial ischemia-reperfusion (I/R) process [26]. The local inflammation induces pro-inflammatory cytokines and promotes cell proliferation and apoptosis [27, 28]. In turn, monocytes and macrophages secrete angiogenic cytokines and anti-inflammatory cytokines to promote injury repair [29]. MSC-Exo, the main efficient component of MSCs, participates in immune regulation [30]. Based on the myocardial I/R mouse model, Zhao and Fatih Arslan discovered that bone marrow-derived MSC-Exos could attenuate neutrophil infiltration [31, 32, 33], increase the concentration of the anti-inflammatory cytokine IL-10, and decrease the concentration of the pro-inflammatory cytokine IL-6 in the heart tissues of mice. More importantly, MSC-Exos promote the polarization of macrophages from the MI phenotype to the M2 phenotype by exchanging miR-182 to downregulate TLR4 and inhibit the relevant downstream signaling pathway (TLR4/NF-kB), while as the sequence of signaling cascade PI3K/Akt signaling pathway was activated, in vivo and in vitro [31]. MSC-Exos can increase the proportion of M2 macrophages by upregulating IL-10 and downregulating IL-6 via miR-21–5p, which reduces the inflammatory reaction in heart tissues [34]. MSC-Exos can deliver miR-182–5p and downregulate Gasdermin D to reduce the inflammatory cytokines (e.g., IL-1β and IL-18) released in the inflammasome of NLRP3 [35]. MSC-Exos enriched with miRNA-181a can attenuate inflammatory cell infiltration by targeting c-Fos, along with the upregulation of IL-10 and Treg cells and the downregulation of TNF-ɑ and IL-6 [36]. The basic mechanism is summarized in Fig. 2.

Fig. 2.

MSC-Exos regulate inflammatory reactions in receptor cells. The MSC-Exos enter the receptor cells and mediate the PI3K/AKT and TLR4/NF-kB signaling pathways, and the release of IL-1β, IL-6, IL-10, and IL-18 by transferring related microRNAs.

4.2 MSC-Exos Inhibit Cardiomyocyte Apoptosis and Autophagy

Inappropriate apoptosis in ischemia strongly influences myocardial injury [37, 38, 39]. The phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway plays a pivotal role in myocardial cell apoptosis, which can be reversed by enhancer of zeste homolog 2 (EZH2) [40]. In hypoxia, bone marrow-derived MSC-Exos can ameliorate cardiomyocyte apoptosis [41]. Phosphatase and tensin homolog deleted on chromosome ten (PTEN), the target mRNA of miR-144, miR21, and miR-141, is downregulated in a hypoxic environment, which is reversed by bone marrow-derived MSC-Exos in a dose-dependent manner, and activates the downstream PTEN/p-AKT and PTEN/β-catenin signaling pathways [33, 42, 43]. In the mouse myocardial injury model induced by sepsis, a significant abundance of miR-141 was found in bone marrow-derived MSC-Exo-treated mouse myocardial tissues. Exosomal miR-141 targeted PTEN and activated β-catenin to alleviate myocardial injury. MiR-144 enriched in the bone marrow-derived MSC-Exo decreased PTEN expression, increased p-AKT expression, and prevented the apoptosis of H9C2 cells [42]. In turn, exosomes secreted from MSCs in a hypoxic environment enhanced the function of anti-apoptotic effects. MiR-125b increased the expression of the p53 and BAK1 mRNA [41]. Upregulating miR-221–3p and miR-146a-5p also inhibited the apoptosis of cardiomyocytes [44, 45]. MSC-Exos pretreated with macrophage migration inhibitory factor showed a strong cardioprotective effect. The transfer of lncRNA-NEAT1 between MSC-Exos and cardiomyocytes directly targeted miR-142–3p and regulated the expression of Forkhead Box O1 (FOXO1). Additionally, exosomal miR-183–5p could also target FOXO1, which can protect cardiomyocytes from apoptosis and cellular senescence effectively [46, 47, 48]. H9c2 cells treated with human umbilical cord MSC-Exo (hMSC-Exo) showed higher cell viability and inhibition of apoptosis and autophagy. High levels of Bcl-2 facilitate cardioprotection [49, 50, 51, 52]. In the studies conducted by Gu, et al. [50] and Zou, et al. [53] a high concentration of MSC-Exo enhanced the BCL-2/BAX ratio; thus, preventing the apoptosis of cardiomyocytes, increased the expression of Beclin-1, pAMPK, LC3II/I, and ATG13 and decreased the expression of P62 and Apaf1, activating the AMPK/mTOR-mediated autophagy flux pathway. However, according to a study by Li, et al. [54] exosomal miR-29c from bone marrow MSCs downregulated the LC3II/I ratio and the level of P62. Additionally, targeting PTEN activated the downstream AKT/mTOR signaling pathway, which prevented excessive autophagy in the myocardium. Activation of the CHK2-Beclin2 pathway regulated autophagy and attenuated the apoptosis of cardiomyocytes, which is targeted by exosomal miR-143–3p [55]. Additionally, the miR-143/Bcl-2/Beclin-1 axis is another pathway for decreasing cell apoptosis and inhibiting autophagy that is competitively bound by lncRNA UCA1 derived from hMSC-Exo [52]. In another Doxorubicin-induced myocardial injury model, miR-199a-3p enriched in MSC-Exo activated Akt; thus, inducing the expression of Sp1 and inhibiting the activation of p53, along with the overexpression of survivin to reduce apoptosis [51]. The main signaling pathways are shown in Fig. 3.

Fig. 3.

MSC-Exos inhibit the apoptosis and autophagy of cardiomyocytes. MSC-Exos enter the receptor cells that mediate the mTOR signaling pathway and increase the BCL-2/BAX ratio, thus regulating the expression of FOXO1 and p53.

4.3 MSC-Exos Promote Angiogenesis

Myocardial injuries occur due to the dysfunction of angiogenesis and restriction of blood supply [56]. MSC-Exo has a robust proangiogenic ability, both in vivo and in vitro [57]. The GO analysis and the Panther pathway analysis aimed at the MSC-Exo proteome revealed canonical angiogenesis-related pathways, such as Fibroblast Growth Factor (FGF), Epidermal growth factor receptor (EGFR), Platelet-derived Growth Factor (PDGF), and cadherin [58]. In a study, Sun showed that MSC-Exo with abundant HIF-1a can increase the mRNA and protein levels of proangiogenic factors (e.g., VEGF and PDGF) and enhance neovessel formation to provide cardioprotection [59]. Hypoxic conditions can enhance this function [60]. Wang et al. [44] found that these proangiogenic effects were induced by the upregulation of miRNA-221–3p. In vivo, MSC-Exos were administered to ischemic limbs via intramuscular injection. Laser Doppler Perfusion Imaging showed that blood perfusion in limb ischemia was restored by nearly 85%, and the bioinformatics analysis suggested that proangiogenic effects might be induced by miR-7116–5p [61]. In the human umbilical vein endothelial cell model, MSC-Exo influenced capillary tube formation and promoted angiogenesis [60]. Although the mechanism is unclear, MSC-Exo can treat mouse hearts with a higher capillary density, which can protect the myocardium from ischemic injury [62, 63]. Intriguingly, Hemin (a potent heme oxygenase-1 inducer)-treated MSC-Exo had a superior effect in enhancing the capillary density compared to MSC-Exo [48]. Hemin pretreatment can upregulate miR-183–5p in MSC-Exo. Exosomal miR-183–5p can partially regulate the HMGB1/ERK pathway and inhibit ischemia-induced cardiomyocyte senescence to enhance the cardioprotective effects by regulating mitochondrial fission. Several experiments have confirmed that MSC-Exo can deliver miR-543 to reduce the expression of COL4A1 and lead to the proliferation, migration, invasion, and angiogenesis of cardiac microvascular endothelial cells [64].

4.4 MSC-Exo Participates in Cardiac Remodeling by Mediating Fibrosis

Reactive fibrosis, followed by the loss of cardiomyocytes, occurs in most myocardial injuries and contributes to the remodeling of post-myocardial injury [65, 66]. Collagen I promoted myocardial fibrosis in myocardial injury [67]. MSC-Exo can alleviate myocardial fibrosis and improve cardiac function more effectively than MSC [8, 40, 68, 69]. In the epithelial-mesenchymal transition (EMT) process, epithelial cells are gradually transformed into mesenchymal cells. EMT facilitates the pathogenesis of fibrosis [70]. MSC-Exo can downregulate EZH2 and upregulate High Mobility Group AT-Hook 2 (HMGA2); thus, activating the PI3K/AKT pathway that can delay EMT and fibrosis in myocardial tissues, increase the left ventricular end-diastolic internal diameter (Dd), and end-systolic internal diameter (Sd), and increase the cardiac function [40]. In diabetic patients, MSC-Exo can reduce fibrosis and damage to the myocardial tissue by inhibiting the TGF-β1/Smad2 signaling pathway to decrease the expression of Smad2 and TGF-β1 proteins. Moreover, MSC-Exo can increase the level of fatty acid transporters and fatty acid beta oxidase [71]. Arslan, et al. [32] found that MSC-Exo can also preserve the structure and function of the left ventricle by activating the PI3K/Akt pathway, elevating the level of ATP and NADH, and attenuating oxidative stress. Additionally, the renin-angiotensin (RAS) system helps to improve the index of cardiac function and cardiac remodeling. MSC-Exo maintains the balance of the RAS system, promotes the translation from Ang II to Ang 1–7, and provides constant myocardial protection [72]. The mechanism of cardiac remodeling facilitated by MSC-Exos is shown in Fig. 4. The characteristics and molecular mechanisms of all the related studies mentioned above are shown in Table 1 (Ref. [31, 32, 33, 34, 35, 36, 40, 41, 42, 43, 44, 45, 46, 47, 54, 55, 61, 64, 71, 72]).

Fig. 4.

The mechanism of cardiac remodeling is mediated by MSC-Exos. MSC-Exos help to regulate the PI3K/Akt signaling pathway, the conversion of Ang II to Ang 1–7, and the expression of EZH2 and HMGA2.

Table 1.The characteristics and molecular mechanisms of the related studies.
Study Design Myocardial injury model Intervention Result Mediator Signalling pathways
Zhao J 2019 [31] mice Ligating LCA bone marrow-derived MSC-Exo Converting macrophages to M2 phenotype and alleviating cardiac inflammation miR-182 TLR4/NF-κB/PI3K/Akt
Arslan F 2013 [32] mice Ligating LCA huES9.E1 derived MSC-Exo reducing WBC count activate adenosine receptors PI3K/Akt
Pei Y 2021 [33] mice cecalligation puncture induced myocardial impairment bone marrow-derived MSC-Exo reducing the inflammatory infiltration and cell apoptosis miR-141 PTEN/β-catenin
Shen D 2021 [34] mice Ligating LCA C57BL/6 mouse derived MSC-Exo promote the polarization of macrophages to the M2 phenotype miR-21-5p Not given
Yue R 2022 [35] mice Ligating LAD bone marrow-derived MSC-Exo Reducing GSDMD-dependent cell pyroptosis and inflammation miR-182-5p Not given
Wei Z 2019 [36] mice Ligating LAD human umbilical cord blood-derived MSC-Exo Reducing inflammatory cell infiltration miRNA-181a TNF-α and IL-6
Jiao W 2022 [40] rat Ligating LAD bone marrow-derived MSC-Exo Reducing fibrosis EZH2 PI3K/AKT
Zhu LP 2018 [41] mice Ligating LAD bone marrow-derived MSC-Exo ameliorating cardiomyocyte apoptosis miR-125b p53 and BAK1
Wen Z 2020 [42] H9C2 CMCs of rat cardiac origin Cells were incubated in the hypoxic container for 48 h at 37 C in a CO2 incubator bone marrow-derived MSC-Exo protect H9C2 cells from apoptosis MiRNA144 PTEN/AKT
Shi B 2018 [43] cardiac stem cells of the rat CSCs are treated with 100 μM H2O2 for 2 h bone marrow-derived MSC-Exo protection against oxidative stress-triggered cell death miR-21 PTEN/PI3K/Akt
Wang Q 2021 [44] rat Ligating LAD human umbilical cord blood-derived MSC-Exo Promoting the survival and angiogenesis in cardiomyocytes miR-221-3p Not given
Liu C 2021 [45] mice Cecal Ligation and Puncture bone marrow-derived MSC-Exo protect cardiomyocytes of inflammation model miR-146a-5p MYBL1
Chen H 2020 [46] cells Human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes human adipose-derived MSC-Exo protecting cardiomyocytes from apoptosis miR-142-3p LncRNA-NEAT1/miR-142-3p/FOXO1
Mao S 2022 [47] rat Ligating LAD bone marrow-derived MSC-Exo protecting cardiomyocytes from apoptosis miR-183-5p FOXO1
Li T 2020 [54] mice Ligating LAD bone marrow-derived MSC-Exo regulating autophagy under cardiac injury miRNA-29c PTEN/AKT/mTOR
Chen G 2021 [55] rat H9c2 cells were administrated to established the cellular hypoxia-reoxygenation model bone marrow-derived MSC-Exo Reducing cell apoptosis miR-143-3p CHK2-Beclin2
Ju C 2018 [61] mice Ligating LAD cardiac derived MSC-Exo Promoting cardiomyocyte proliferation, and preserves heart function Not given Not given
Yang M 2021 [64] rat Ligating LAD Human mesenchymal stem cells derived exosome Promoting cardiac microvascular endothelial cell angiogenesis miR-543 COL4A1
Lin Y 2019 [71] rat diabetes mellitus-induced myocardial injury myocardial injury bone marrow-derived MSC-Exo Reducing myocardial injury and fibrosis Not given TGF-β1/Smad2
Xiao M 2021 [72] rat H9c2 cells bone marrow-derived MSC-Exo Improving cardiac remodeling and cardiac function Not given renin-angiotensin system
Note: left anterior descending coronary artery LCA; left anterior descending LAD.
5. Discussion

Exosomes are endocytic vesicles that play a key role in communication between cells. The biogenesis, uptake, composition, and physiological features have been discussed in previous reviews [73, 74, 75]. Although the exact mechanism is unknown, exosomes are extracellular nanovesicles mainly involved in cardioprotection. In a prospective clinical study executed in Policlinico Hospital of Bari and “G. Monasterio” Foundation of Massa showed that distinct exosomal proteins playing their roles of cardioprotection in older cardiac surgery patients regardless of surgery type [76]. Lucio Barile proved cardiac progenitor cells (CPC) derived exosome possessed the capacity to reduce cardiomyocyte apoptosis, enhance angiogenesis, and improve LV ejection fraction in the rat myocardial infarction model [77]. In-depth study revealed that pregnancy-associated plasma protein-A existed in CPC derived exosomes played a significant role in reducing scar size and improving ventricular function in rats’ permanent coronary occlusion model [78]. The data of Valentina Casieri’s research indicated ticagrelor can be leveraged to modulate release of anti-hypoxic exosomes from resident human cardiac-derived mesenchymal progenitor cells (hCPCs) [79]. It is remarkable that, recently, MSC-Exos were also shown to provide effective cardioprotection as a cell-free treatment [80]. In our review, we comprehensively analyzed the feasibility of the application of MSC-Exos in perioperative cardioprotection, as it can regulate inflammatory reaction [30], mediate cardiomyocyte apoptosis and autophagy [81], promote angiogenesis [57, 58, 60, 82], and improve cardiac remodeling [32].

Some clinical research organizations conducted a series of exosome-related clinical trials. In a study, Dai, et al. [83] reported that ascites-derived exosomes (Aex) were administered in the immunotherapy of colorectal cancer in phase I clinical trials. Aex combined with Granulocyte-macrophage Colony Stimulating Factor (GM-CSF) was shown to have strong antitumor effects. Subsequent clinical trials showed that Dendritic cell-derived exosomes (Dex) have strong antitumor effects in melanoma and non-small cell lung cancer [84, 85, 86]. From a clinical perspective, MSCs have beneficial curative effects in some non-neoplastic diseases. The phase II/III clinical pilot studies in Sahel Teaching Hospital showed that MSC-Exos applied to grade III-IV chronic kidney diseases can inhibit inflammatory immune reactions and improve kidney function [87]. Moreover, clinical trials on bronchopulmonary dysplasia, macular holes, type 1 diabetes, and acute ischemic stroke are underway. Due to large inter-individual variability and technological limitations, MSC-Exos have not been widely applied in clinical treatment. Fortunately, other applications of exosomes in oncologic therapy have verified the safety and effectiveness of MSC exosomal therapy.

The bioactive cargoes in MSC-Exos are also being investigated. Several studies have shown that exosomal miRNAs and proteins are responsible for the cardiovascular protection and repair of MSC-Exos [21]. Exosomal miRNA is an important bioactive cargo in MSC-Exo and is transferred to the recipient cells and specifically combined with the complementary mRNA target; thus, it can regulate the expression of related genes. The result of miRNA analysis based on the NanoString platform showed that the predictable top 23 miRNAs of human bone marrow-derived MSC-Exo targeting 5481 genes enriched in the PDGF, TGF-β, and Wnt signaling pathways were associated with angiogenesis and tissue remodeling [88, 89, 90]. Determining the exact mechanism of action and the specific target genes of these miRNAs is important for the clinical application of exosomes. Many well-constructed models have shown that modified exosomes can provide perioperative cardioprotection efficiently [82]. Because of unresolved confounding factors (e.g., complex exosomal component, complicated isolation process, elaborate exosome-loading mechanism, etc.), modification of the MSC-Exo based on bioengineering has not been performed. According to the identity of the specific bioactive cargo and research on the mechanism of biogenesis of exosomes, enhancing the function of MSC-Exo via genetic manipulation needs to be investigated in future studies for clinical application. Some studies have shown that lentiviral transfection and virus-free electroporation can be used to develop bioengineered exosomes [21, 91] with higher efficacy. Through this method, a low dose of exosomes can be used to achieve superior effects, thus compensating for the limitations of exosome isolation. To summarize, optimal ways for harvesting, modifying, and applying exosomes need to be investigated to reduce perioperative myocardial injuries in cardiac and non-cardiac surgeries.

6. Conclusions

MSC-Exos regulate inflammatory reactions, inhibit cardiomyocyte apoptosis and autophagy, promote angiogenesis, and mediate cardiac remodeling to prevent myocardial injury. MSC-Exos show therapeutic potential for ischemic cardiac injury and have a good application prospect in Cardioprotection. However, exosomes alone are not enough to reverse cardiac dysfunction after myocardial injury. Further study of the molecular mechanism can better guide the clinical transformation.

Abbreviations

MSC-Exo, Mesenchymal Stem Cell-Derived Exosome; PMI, perioperative myocardial infarction; IPC, ischemic preconditioning; RIPC, remote ischemic preconditioning; RIPostC, remote ischemic postconditioning; I/R, ischemia-reperfusion; PI3K, phosphatidylinositol 3-kinase; Aex, ascites-derived exosomes; hMSC-Exo, human umbilical cord MSC-Exo; EMT, Epithelial to mesenchymal transition; Sd, end-systolic internal diameter; Dd, end-diastolic internal diameter; RAS, renin-angiotensin; BMSC, bone marrow-derived MSCs; ASC, adipose-derived MSCs; FOXO1, forkhead box O1; PTEN, phosphatase and tensin homolog deleted on chromosome ten; GM-CSF, granulocyte-macrophage colony stimulating factor; Dex, dendritic cell-derived exosomes; FGF, fibroblast growth factor; EGFR, Epidermal growth factor receptor; PDGF, platelet-derived growth factor; VEGF, vascular endothelial growth factor; HMGA2, high mobility group at-hook 2.

Author Contributions

WL and SW designed the research study. JL and SY performed the research and wrote the manuscript. MT and XG provided help and advice on figures. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

The figures were created in BioRender.com. We would like to express our gratitude to all those who helped us during the writing of this manuscript. Thanks to all the peer reviewers for their opinions and suggestions.

Funding

This research was funded by the Development Center for Medical Science & Technology, National Health Commission of the People’s Republic of China (Grant No: WA2020RW18); Wu Jieping Medical Foundation (Grant No: 320.6750.19092-1); Jilin Province Scientific and Technological Department, International Scientific and Technological Cooperation Project (20190701043GH).

Conflict of Interest

The authors declare no conflict of interest.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

References
[1]
Devereaux PJ, Goldman L, Cook DJ, Gilbert K, Leslie K, Guyatt GH. Perioperative cardiac events in patients undergoing noncardiac surgery: a review of the magnitude of the problem, the pathophysiology of the events and methods to estimate and communicate risk. Canadian Medical Association Journal. 2005; 173: 627–634.
[2]
Mangano DT. Long-term cardiac prognosis following noncardiac surgery. the Study of Perioperative Ischemia Research Group. The Journal of the American Medical Association. 1992; 268: 233–239.
[3]
Wong SSC, Irwin MG. Peri-operative cardiac protection for non-cardiac surgery. Anaesthesia. 2016; 71: 29–39.
[4]
Roth S, Torregroza C, Huhn R, Hollmann MW, Preckel B. Perioperative Cardioprotection: Clinical Implications. Anesthesia & Analgesia. 2020; 131: 1751–1764.
[5]
Bagno L, Hatzistergos KE, Balkan W, Hare JM. Mesenchymal Stem Cell-Based Therapy for Cardiovascular Disease: Progress and Challenges. Molecular Therapy. 2018; 26: 1610–1623.
[6]
Behr B, Ko SH, Wong VW, Gurtner GC, Longaker MT. Stem Cells. Plastic and Reconstructive Surgery. 2010; 126: 1163–1171.
[7]
Baglio SR, Pegtel DM, Baldini N. Mesenchymal stem cell secreted vesicles provide novel opportunities in (stem) cell-free therapy. Frontiers in Physiology. 2012; 3: 359.
[8]
Shao L, Zhang Y, Lan B, Wang J, Zhang Z, Zhang L, et al. MiRNA-Sequence Indicates that Mesenchymal Stem Cells and Exosomes have Similar Mechanism to Enhance Cardiac Repair. BioMed Research International. 2017; 2017: 4150705.
[9]
Devereaux PJ, Chan MT, Alonso-Coello P, Walsh M, Berwanger O, Villar JC, et al. Association between postoperative troponin levels and 30-day mortality among patients undergoing noncardiac surgery. The Journal of the American Medical Association. 2012; 307: 2295–2304.
[10]
Botto F, Alonso-Coello P, Chan MT, Villar JC, Xavier D, Srinathan S, et al. Myocardial injury after noncardiac surgery: a large, international, prospective cohort study establishing diagnostic criteria, characteristics, predictors, and 30-day outcomes. Anesthesiology. 2014; 120: 564–578.
[11]
Thygesen K, Alpert JS, Jaffe AS, Simoons ML, Chaitman BR, White HD, et al. Third universal definition of myocardial infarction. Journal of the American College of Cardiology. 2012; 60: 1581–1598.
[12]
Biccard BM, Rodseth RN. The pathophysiology of peri-operative myocardial infarction. Anaesthesia. 2010; 65: 733–741.
[13]
Hass R, Kasper C, Böhm S, Jacobs R. Different populations and sources of human mesenchymal stem cells (MSC): a comparison of adult and neonatal tissue-derived MSC. Cell Communication and Signaling. 2011; 9: 12.
[14]
Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al. Multilineage Potential of Adult Human Mesenchymal Stem Cells. Science. 1999; 284: 143–147.
[15]
Balaj L, Lessard R, Dai L, Cho Y, Pomeroy SL, Breakefield XO, et al. Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences. Nature Communications. 2011; 2: 180.
[16]
Deng H, Sun C, Sun Y, Li H, Yang L, Wu D, et al. Lipid, Protein, and MicroRNA Composition within Mesenchymal Stem Cell-Derived Exosomes. Cellular Reprogramming. 2018; 20: 178–186.
[17]
Kim H, Choi D, Yun SJ, Choi S, Kang JW, Jung JW, et al. Proteomic Analysis of Microvesicles Derived from Human Mesenchymal Stem Cells. Journal of Proteome Research. 2012; 11: 839–849.
[18]
Lai RC, Tan SS, Teh BJ, Sze SK, Arslan F, de Kleijn DP, et al. Proteolytic Potential of the MSC Exosome Proteome: Implications for an Exosome-Mediated Delivery of Therapeutic Proteasome. International Journal of Proteomics. 2012; 2012: 971907.
[19]
Chen TS, Lai RC, Lee MM, Choo ABH, Lee CN, Lim SK. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Research. 2010; 38: 215–224.
[20]
Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology. 2007; 9: 654–659.
[21]
Ferguson SW, Wang J, Lee CJ, Liu M, Neelamegham S, Canty JM, et al. The microRNA regulatory landscape of MSC-derived exosomes: a systems view. Scientific Reports. 2018; 8: 1419.
[22]
Baglio SR, Rooijers K, Koppers-Lalic D, Verweij FJ, Pérez Lanzón M, Zini N, et al. Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Research & Therapy. 2015; 6: 127.
[23]
Wang K, Jiang Z, Webster KA, Chen J, Hu H, Zhou Y, et al. Enhanced Cardioprotection by Human Endometrium Mesenchymal Stem Cells Driven by Exosomal MicroRNA-21. Stem Cells Translational Medicine. 2017; 6: 209–222.
[24]
Maegdefessel L, Spin JM, Raaz U, Eken SM, Toh R, Azuma J, et al. MiR-24 limits aortic vascular inflammation and murine abdominal aneurysm development. Nature Communications. 2014; 5: 5214.
[25]
Qian L, Van Laake LW, Huang Y, Liu S, Wendland MF, Srivastava D. MiR-24 inhibits apoptosis and represses Bim in mouse cardiomyocytes. Journal of Experimental Medicine. 2011; 208: 549–560.
[26]
Hausenloy DJ, Yellon DM. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. Journal of Clinical Investigation. 2013; 123: 92–100.
[27]
Takeuchi O, Akira S. Pattern Recognition Receptors and Inflammation. Cell. 2010; 140: 805–820.
[28]
Schett G, Neurath MF. Resolution of chronic inflammatory disease: universal and tissue-specific concepts. Nature Communications. 2018; 9: 3261.
[29]
Ben-Mordechai T, Holbova R, Landa-Rouben N, Harel-Adar T, Feinberg MS, Abd Elrahman I, et al. Macrophage Subpopulations are Essential for Infarct Repair with and without Stem Cell Therapy. Journal of the American College of Cardiology. 2013; 62: 1890–1901.
[30]
Zhang B, Yin Y, Lai RC, Tan SS, Choo ABH, Lim SK. Mesenchymal Stem Cells Secrete Immunologically Active Exosomes. Stem Cells and Development. 2014; 23: 1233–1244.
[31]
Zhao J, Li X, Hu J, Chen F, Qiao S, Sun X, et al. Mesenchymal stromal cell-derived exosomes attenuate myocardial ischaemia-reperfusion injury through miR-182-regulated macrophage polarization. Cardiovascular Research. 2019; 115: 1205–1216.
[32]
Arslan F, Lai RC, Smeets MB, Akeroyd L, Choo A, Aguor ENE, 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 Research. 2013; 10: 301–312.
[33]
Pei Y, Xie S, Li J, Jia B. Bone marrow-mesenchymal stem cell-derived exosomal microRNA-141 targets PTEN and activates β-catenin to alleviate myocardial injury in septic mice. Immunopharmacology and Immunotoxicology. 2021; 43: 584–593.
[34]
Shen D, He Z. Mesenchymal stem cell-derived exosomes regulate the polarization and inflammatory response of macrophages via miR-21-5p to promote repair after myocardial reperfusion injury. Annals of Translational Medicine. 2021; 9: 1323–1323.
[35]
Yue R, Lu S, Luo Y, Zeng J, Liang H, Qin D, et al. Mesenchymal stem cell-derived exosomal microRNA-182-5p alleviates myocardial ischemia/reperfusion injury by targeting GSDMD in mice. Cell Death Discovery. 2022; 8: 202.
[36]
Wei Z, Qiao S, Zhao J, Liu Y, Li Q, Wei Z, et al. MiRNA-181a over-expression in mesenchymal stem cell-derived exosomes influenced inflammatory response after myocardial ischemia-reperfusion injury. Life Sciences. 2019; 232: 116632.
[37]
Elmore S. Apoptosis: a Review of Programmed Cell Death. Toxicologic Pathology. 2007; 35: 495–516.
[38]
Poon IKH, Lucas CD, Rossi AG, Ravichandran KS. Apoptotic cell clearance: basic biology and therapeutic potential. Nature Reviews Immunology. 2014; 14: 166–180.
[39]
Renehan AG. What is apoptosis, and why is it important? British Medical Journal. 2001; 322: 1536–1538.
[40]
Jiao W, Hao J, Xie Y, Meng M, Gao W. EZH2 mitigates the cardioprotective effects of mesenchymal stem cell-secreted exosomes against infarction via HMGA2-mediated PI3K/AKT signaling. BMC Cardiovascular Disorders. 2022; 22: 95.
[41]
Zhu L, Tian T, Wang J, He J, Chen T, Pan M, et al. Hypoxia-elicited mesenchymal stem cell-derived exosomes facilitates cardiac repair through miR-125b-mediated prevention of cell death in myocardial infarction. Theranostics. 2018; 8: 6163–6177.
[42]
Wen Z, Mai Z, Zhu X, Wu T, Chen Y, Geng D, et al. Mesenchymal stem cell-derived exosomes ameliorate cardiomyocyte apoptosis in hypoxic conditions through microRNA144 by targeting the PTEN/AKT pathway. Stem Cell Research & Therapy. 2020; 11: 36.
[43]
Shi B, Wang Y, Zhao R, Long X, Deng W, Wang Z. Bone marrow mesenchymal stem cell-derived exosomal miR-21 protects C-kit+ cardiac stem cells from oxidative injury through the PTEN/PI3K/Akt axis. PLoS ONE. 2018; 13: e0191616.
[44]
Wang Q, Zhang L, Sun Z, Chi B, Zou A, Mao L, et al. HIF-1α overexpression in mesenchymal stem cell-derived exosome-encapsulated arginine-glycine-aspartate (RGD) hydrogels boost therapeutic efficacy of cardiac repair after myocardial infarction. Materials Today Bio. 2021; 12: 100171.
[45]
Liu C, Xue J, Xu B, Zhang A, Qin L, Liu J, et al. Exosomes Derived from miR-146a-5p-Enriched Mesenchymal Stem Cells Protect the Cardiomyocytes and Myocardial Tissues in the Polymicrobial Sepsis through Regulating MYBL1. Stem Cells International. 2021; 2021: 1530445.
[46]
Chen H, Xia W, Hou M. LncRNA-NEAT1 from the competing endogenous RNA network promotes cardioprotective efficacy of mesenchymal stem cell-derived exosomes induced by macrophage migration inhibitory factor via the miR-142-3p/FOXO1 signaling pathway. Stem Cell Research & Therapy. 2020; 11: 31.
[47]
Mao S, Zhao J, Zhang Z, Zhao Q. MiR-183-5p overexpression in bone mesenchymal stem cell-derived exosomes protects against myocardial ischemia/reperfusion injury by targeting FOXO1. Immunobiology. 2022; 227: 152204.
[48]
Zheng H, Liang X, Han Q, Shao Z, Zhang Y, Shi L, et al. Hemin enhances the cardioprotective effects of mesenchymal stem cell-derived exosomes against infarction via amelioration of cardiomyocyte senescence. Journal of Nanobiotechnology. 2021; 19: 332.
[49]
Jiang X, Lew K, Chen Q, Richards AM, Wang P. Human Mesenchymal Stem Cell-derived Exosomes Reduce Ischemia/Reperfusion Injury by the Inhibitions of Apoptosis and Autophagy. Current Pharmaceutical Design. 2018; 24: 5334–5341.
[50]
Gu X, Li Y, Chen K, Wang X, Wang Z, Lian H, et al. Exosomes derived from umbilical cord mesenchymal stem cells alleviate viral myocarditis through activating AMPK/mTOR‐mediated autophagy flux pathway. Journal of Cellular and Molecular Medicine. 2020; 24: 7515–7530.
[51]
Lee JY, Chung J, Byun Y, Kim KH, An SH, Kwon K. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Protect Cardiomyocytes from Doxorubicin-Induced Cardiomyopathy by Upregulating Survivin Expression via the miR-199a-3p-Akt-Sp1/p53 Signaling Pathway. International Journal of Molecular Sciences. 2021; 22: 7102.
[52]
Diao L, Zhang Q. Transfer of lncRNA UCA1 by hUCMSCs-derived exosomes protects against hypoxia/reoxygenation injury through impairing miR-143-targeted degradation of Bcl-2. Aging. 2021; 13: 5967–5985.
[53]
Zou L, Ma X, Lin S, Wu B, Chen Y, Peng C. Bone marrow mesenchymal stem cell-derived exosomes protect against myocardial infarction by promoting autophagy. Experimental and Therapeutic Medicine. 2019; 18: 2574–7582.
[54]
Li T, Gu J, Yang O, Wang J, Wang Y, Kong J. Bone Marrow Mesenchymal Stem Cell-Derived Exosomal miRNA-29c Decreases Cardiac Ischemia/Reperfusion Injury through Inhibition of Excessive Autophagy via the PTEN/Akt/mTOR Signaling Pathway. Circulation Journal. 2020; 84: 1304–1311.
[55]
Chen G, Wang M, Ruan Z, Zhu L, Tang C. Mesenchymal stem cell-derived exosomal miR-143-3p suppresses myocardial ischemia-reperfusion injury by regulating autophagy. Life Sciences. 2021; 280: 119742.
[56]
Jia S, Yao Y, Song Y, Tang X, Zhao X, Gao R, et al. Two-Year Outcomes after Left Main Coronary Artery Percutaneous Coronary Intervention in Patients Presenting with Acute Coronary Syndrome. Journal of Interventional Cardiology. 2020; 2020: 6980324.
[57]
Bian X, Ma K, Zhang C, Fu X. Therapeutic angiogenesis using stem cell-derived extracellular vesicles: an emerging approach for treatment of ischemic diseases. Stem Cell Research & Therapy. 2019; 10: 158.
[58]
Anderson JD, Johansson HJ, Graham CS, Vesterlund M, Pham MT, Bramlett CS, et al. Comprehensive Proteomic Analysis of Mesenchymal Stem Cell Exosomes Reveals Modulation of Angiogenesis via Nuclear Factor-KappaB Signaling. Stem Cells. 2016; 34: 601–613.
[59]
Sun J, Shen H, Shao L, Teng X, Chen Y, Liu X, et al. HIF-1α overexpression in mesenchymal stem cell-derived exosomes mediates cardioprotection in myocardial infarction by enhanced angiogenesis. Stem Cell Research & Therapy. 2020; 11: 373.
[60]
Almeria C, Weiss R, Roy M, Tripisciano C, Kasper C, Weber V, et al. Hypoxia Conditioned Mesenchymal Stem Cell-Derived Extracellular Vesicles Induce Increased Vascular Tube Formation in vitro. Frontiers in Bioengineering and Biotechnology. 2019; 7: 292.
[61]
Ju C, Li Y, Shen Y, Liu Y, Cai J, Liu N, et al. Transplantation of Cardiac Mesenchymal Stem Cell-Derived Exosomes for Angiogenesis. Journal of Cardiovascular Translational Research. 2018; 11: 429–437.
[62]
Ju C, Shen Y, Ma G, Liu Y, Cai J, Kim I, et al. Transplantation of Cardiac Mesenchymal Stem Cell-Derived Exosomes Promotes Repair in Ischemic Myocardium. Journal of Cardiovascular Translational Research. 2018; 11: 420–428.
[63]
Teng X, Chen L, Chen W, Yang J, Yang Z, Shen Z. Mesenchymal Stem Cell-Derived Exosomes Improve the Microenvironment of Infarcted Myocardium Contributing to Angiogenesis and Anti-Inflammation. Cellular Physiology and Biochemistry. 2015; 37: 2415–2424.
[64]
Yang M, Liu X, Jiang M, Li J, Tang Y, Zhou L. miR-543 in human mesenchymal stem cell-derived exosomes promotes cardiac microvascular endothelial cell angiogenesis after myocardial infarction through COL4A1. IUBMB Life. 2021; 73: 927–940.
[65]
Weber KT, Janicki JS, Shroff SG, Pick R, Chen RM, Bashey RI. Collagen remodeling of the pressure-overloaded, hypertrophied nonhuman primate myocardium. Circulation Research. 1988; 62: 757–765.
[66]
van den Borne SWM, Isobe S, Verjans JW, Petrov A, Lovhaug D, Li P, et al. Molecular Imaging of Interstitial Alterations in Remodeling Myocardium after Myocardial Infarction. Journal of the American College of Cardiology. 2008; 52: 2017–2028.
[67]
de Haas HJ, Arbustini E, Fuster V, Kramer CM, Narula J. Molecular Imaging of the Cardiac Extracellular Matrix. Circulation Research. 2014; 114: 903–915.
[68]
Chen F, Li X, Zhao J, Geng J, Xie J, Xu B. Bone marrow mesenchymal stem cell-derived exosomes attenuate cardiac hypertrophy and fibrosis in pressure overload induced remodeling. In Vitro Cellular & Developmental Biology - Animal. 2020; 56: 567–576.
[69]
Zhang Z, Yang J, Yan W, Li Y, Shen Z, Asahara T. Pretreatment of Cardiac Stem Cells with Exosomes Derived from Mesenchymal Stem Cells Enhances Myocardial Repair. Journal of the American Heart Association. 2016; 5: e002856
[70]
Sisto M, Ribatti D, Lisi S. Organ Fibrosis and Autoimmunity: The Role of Inflammation in TGFβ-Dependent EMT. Biomolecules. 2021; 11: 310.
[71]
Lin Y, Zhang F, Lian XF, Peng WQ, Yin CY. Mesenchymal stem cell-derived exosomes improve diabetes mellitus-induced myocardial injury and fibrosis via inhibition of TGF-β1/Smad2 signaling pathway. Cellular and Molecular Biology (Noisy-le-grand). 2019; 65: 123–126.
[72]
Xiao M, Zeng W, Wang J, Yao F, Peng Z, Liu G, et al. Exosomes Protect against Acute Myocardial Infarction in Rats by Regulating the Renin-Angiotensin System. Stem Cells and Development. 2021; 30: 622–631.
[73]
He C, Zheng S, Luo Y, Wang B. Exosome Theranostics: Biology and Translational Medicine. Theranostics. 2018; 8: 237–255.
[74]
Yang D, Zhang W, Zhang H, Zhang F, Chen L, Ma L, et al. Progress, opportunity, and perspective on exosome isolation - efforts for efficient exosome-based theranostics. Theranostics. 2020; 10: 3684–3707.
[75]
Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020; 367: eaau6977.
[76]
Carrozzo A, Casieri V, Di Silvestre D, Brambilla F, De Nitto E, Sardaro N, et al. Plasma exosomes characterization reveals a perioperative protein signature in older patients undergoing different types of on-pump cardiac surgery. GeroScience. 2021; 43: 773–789.
[77]
Barile L, Lionetti V, Cervio E, Matteucci M, Gherghiceanu M, Popescu LM, et al. Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction. Cardiovascular Research. 2014; 103: 530–541.
[78]
Barile L, Cervio E, Lionetti V, Milano G, Ciullo A, Biemmi V, et al. Cardioprotection by cardiac progenitor cell-secreted exosomes: role of pregnancy-associated plasma protein-A. Cardiovascular Research. 2018; 114: 992–1005.
[79]
Casieri V, Matteucci M, Pasanisi EM, Papa A, Barile L, Fritsche-Danielson R, et al. Ticagrelor Enhances Release of Anti-Hypoxic Cardiac Progenitor Cell-Derived Exosomes through Increasing Cell Proliferation in Vitro. Scientific Reports. 2020; 10: 2494.
[80]
Pashoutan Sarvar D, Shamsasenjan K, Akbarzadehlaleh P. Mesenchymal Stem Cell-Derived Exosomes: New Opportunity in Cell-Free Therapy. Advanced Pharmaceutical Bulletin. 2016; 6: 293–299.
[81]
Liu Z, Xu Y, Wan Y, Gao J, Chu Y, Li J. Exosomes from adipose-derived mesenchymal stem cells prevent cardiomyocyte apoptosis induced by oxidative stress. Cell Death Discovery. 2019; 5: 79.
[82]
Xue C, Shen Y, Li X, Li B, Zhao S, Gu J, et al. Exosomes Derived from Hypoxia-Treated Human Adipose Mesenchymal Stem Cells Enhance Angiogenesis through the PKA Signaling Pathway. Stem Cells and Development. 2018; 27: 456–465.
[83]
Dai S, Wei D, Wu Z, Zhou X, Wei X, Huang H, et al. Phase I clinical trial of autologous ascites-derived exosomes combined with GM-CSF for colorectal cancer. Molecular Therapy. 2008; 16: 782–790.
[84]
Escudier B, Dorval T, Chaput N, André F, Caby MP, Novault S, et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of thefirst phase I clinical trial. Journal of Translational Medicine. 2005; 3: 10.
[85]
Morse MA, Garst J, Osada T, Khan S, Hobeika A, Clay TM, et al. A phase I study of dexosome immunotherapy in patients with advanced non-small cell lung cancer. Journal of Translational Medicine. 2005; 3: 9.
[86]
Besse B, Charrier M, Lapierre V, Dansin E, Lantz O, Planchard D, et al. Dendritic cell-derived exosomes as maintenance immunotherapy after first line chemotherapy in NSCLC. OncoImmunology. 2016; 5: e1071008.
[87]
Nassar W, El-Ansary M, Sabry D, Mostafa MA, Fayad T, Kotb E, et al. Umbilical cord mesenchymal stem cells derived extracellular vesicles can safely ameliorate the progression of chronic kidney diseases. Biomaterials Research. 2016; 20: 21.
[88]
Bujak M, Frangogiannis NG. The role of TGF-beta signaling in myocardial infarction and cardiac remodeling. Cardiovascular Research. 2007; 74: 184–195.
[89]
Fan D, Takawale A, Lee J, Kassiri Z. Cardiac fibroblasts, fibrosis and extracellular matrix remodeling in heart disease. Fibrogenesis & Tissue Repair. 2012; 5: 15.
[90]
Gallini R, Lindblom P, Bondjers C, Betsholtz C, Andrae J. PDGF-A and PDGF-B induces cardiac fibrosis in transgenic mice. Experimental Cell Research. 2016; 349: 282–290.
[91]
Yu B, Gong M, Wang Y, Millard RW, Pasha Z, Yang Y, et al. Cardiomyocyte protection by GATA-4 gene engineered mesenchymal stem cells is partially mediated by translocation of miR-221 in microvesicles. PLoS ONE. 2013; 8: e73304.
Share
Back to top