Next Article in Journal
Genome-Wide Investigation of the NAC Transcription Factor Family in Apocynum venetum Revealed Their Synergistic Roles in Abiotic Stress Response and Trehalose Metabolism
Next Article in Special Issue
PPAR Alpha Activation by Clofibrate Alleviates Ischemia/Reperfusion Injury in Metabolic Syndrome Rats by Decreasing Cardiac Inflammation and Remodeling and by Regulating the Atrial Natriuretic Peptide Compensatory Response
Previous Article in Journal
A Closer Look at Opioid-Induced Adrenal Insufficiency: A Narrative Review
Previous Article in Special Issue
Anti-Fibrotic Potential of Angiotensin (1-7) in Hemodynamically Overloaded Rat Heart
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury

1
Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
2
Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(5), 4577; https://doi.org/10.3390/ijms24054577
Submission received: 22 January 2023 / Revised: 19 February 2023 / Accepted: 21 February 2023 / Published: 26 February 2023
(This article belongs to the Special Issue Cellular and Molecular Biology of Heart Diseases)

Abstract

:
Induced pluripotent stem cell (iPSC) therapy brings great hope to the treatment of myocardial injuries, while extracellular vesicles may be one of the main mechanisms of its action. iPSC-derived small extracellular vesicles (iPSCs-sEVs) can carry genetic and proteinaceous substances and mediate the interaction between iPSCs and target cells. In recent years, more and more studies have focused on the therapeutic effect of iPSCs-sEVs in myocardial injury. IPSCs-sEVs may be a new cell-free-based treatment for myocardial injury, including myocardial infarction, myocardial ischemia–reperfusion injury, coronary heart disease, and heart failure. In the current research on myocardial injury, the extraction of sEVs from mesenchymal stem cells induced by iPSCs was widely used. Isolation methods of iPSCs-sEVs for the treatment of myocardial injury include ultracentrifugation, isodensity gradient centrifugation, and size exclusion chromatography. Tail vein injection and intraductal administration are the most widely used routes of iPSCs-sEV administration. The characteristics of sEVs derived from iPSCs which were induced from different species and organs, including fibroblasts and bone marrow, were further compared. In addition, the beneficial genes of iPSC can be regulated through CRISPR/Cas9 to change the composition of sEVs and improve the abundance and expression diversity of them. This review focused on the strategies and mechanisms of iPSCs-sEVs in the treatment of myocardial injury, which provides a reference for future research and the application of iPSCs-sEVs.

1. Introduction

Cardiovascular disease (CVD) is the leading cause of global morbidity and mortality [1,2], with a 50% increase in associated mortality over the last 30 years [3]. In view of the heavy social burden, there is an urgent need for effective prevention and control measures. At present, surgery and drugs are the standard methods for the treatment of CVD, but they cannot promote the regeneration of damaged myocardial tissue [4]. The myocardial injury caused by a large number of cardiomyocyte apoptoses is irreversible [5]. Induced pluripotent stem cells (iPSCs) are reprogrammed cells that have features similar to embryonic stem cells, such as self-regeneration without restriction and differentiation into different tissue or cell types [6,7,8]. Compared with embryonic stem cells, they have abundant sources and have no ethical issues. Moreover, iPSCs induced by autologous cells can also reduce the risk of immune rejection and can be used as a potential treatment for CVD [9,10].
Like other cell therapies, iPSCs also have disadvantages such as low cell survival, retention and implantation rates of cells [11]. Recently, many studies have confirmed that stem cells play a therapeutic role in CVD mainly by inducing paracrine/autocrine growth factors, immunomodulators, and other bioactive molecules stored in their extracellular vesicles (EVs) [12,13,14]. EVs can be classified into apoptotic bodies (50~1000 nm in diameter), microvesicles (MVs) (100~1000 nm), and exosomes (40~160 nm, average ~100 nm) based on their origin [15]. With respect to the biogenesis of EVs, apoptotic bodies are released by dying cells, which are seldomly used for study possibly due to their large and uneven particle size. MVs are formed by the direct outward budding of plasma membranes. The specific process of exosome biogenesis is recognized as a “swallow and spit” process [16] (Figure 1A). Given that the latest MISEV guidelines suggest the use of “EVs” to generally denote a heterogeneous extracellular vesicle population, and “exosomes” are defined as small extracellular vesicles (sEVs), in this review, we focus on exosomes.
In the myocardial infarction (MI), myocardial ischemia–reperfusion injury (MIRI), and heart failure (HF) models, studies using stem cell EVs have shown that they can improve cardiac contractile function in the long term by reducing the initial infarct size, promoting angiogenesis, reducing fibrosis, and remodeling [17]. EVs derived from stem cells regulate gene expression by transferring different substances (including protein, DNA, mRNA, microRNA (miRNA), long-stranded non-coding RNA (lncRNA), and circular RNA (circRNA)) to achieve targeted regulation between cells [18,19], and they have the advantages of high biocompatibility, circulatory stability, and low immunogenicity [20], which open up a new field for resolving the obstacles of stem cell therapy (Figure 1B).
IPSC-derived extracellular vesicles (iPSCs-EVs) can play a therapeutic role similar to that of iPSCs, and iPSCs-EVs are easier to store and transport [21]. At the same time, some limitations of cell therapy, such as embolism and tumor occurrence, are avoided [22,23]. According to the comparison of EVs secreted from mesenchymal stem cells (MSCs) and iPSCs, it was found that while iPSC-EVs enclose proteins that modulate RNA and microRNA stability and protein sorting, MSC-derived EVs are rich in proteins that organize the extracellular matrix, regulate locomotion, and influence cell–substrate adhesion. Moreover, compared to their respective cells, iPSC-EVs share 76.63% of proteins with iPSCs [24], including proteins involved in angiogenesis signaling pathways (VEGF, TGFB1, and Angiogenin) [25], proteins related to membrane organization and the wound-healing process (HSPA5, RAB10, and CLIC1) [26], and proteins involved in cardiac development and cardiac mechanical and electrical function (GSTM, ARGBP2, CDH11, and ACTA2) [27].

2. Isolation of sEVs from Induced Pluripotent Stem Cells

The efficacious extraction of iPSC-derived extracellular vesicles (iPSC-sEVs) is a prerequisite for them to play a therapeutic role. How the high yield, high purity, and high biological activity of small extracellular vesicles can be obtained is directly related to future research and applications [28,29]. At present, many techniques for isolating sEVs have been developed, which depend to a large extent on the physical and chemical properties of sEVs, and the choice of methods should also take into account specific research needs. The isolation methods of iPSC-sEVs for the treatment of CVD include ultracentrifugation (UCF) [30,31], size-exclusion chromatography (SEC) [32], polymer-based precipitation [33], affinity capture [34], magnetic [35,36] and anion-exchange-based methods [37], or a combination of the aforementioned methods [38]. In this mini review, we will introduce three of the most common ones in detail.
UCF is the “gold standard” for isolating sEVs and the most commonly used technology [39]. The substances with different densities and sizes are separated by using different centrifugal forces and velocities (Figure 2A). First, larger cells, cell debris, and dead cells are removed by low-speed centrifugation [40]; then, resuspension with PBS is performed, and finally, ultracentrifugation is carried out to remove contaminated proteins to obtain granular exosomes [41]. The temperature of the whole centrifugation process is kept at 4 °C to ensure that proteases, DNA enzymes, and ribonucleases are inactivated [42]. The concentration of exosomes is determined using an enhanced BCA protein analysis kit [43,44] or nanoparticle tracking analysis, which is an optical particle tracking method developed to determine the concentration and size distribution of particles [45]. In addition, Western blotting can provide useful information on the size of the different proteins [46]. ELISA is another established technique for protein quantification and could be executed in multiple different assay formats [47]. Unlike Western blotting and ELISA, which quantify targeted proteins on a relatively small scale, mass spectrometry enables high-throughput peptide profiling [48]. Additionally, small EVs can be characterized by observation under a transmission electron microscope (TEM) [49]. Moreover, a TEM can also be coupled with immunogold labeling (immuno-EM) to provide molecular characterization [50]. UCF has advantages of simple operation, low cost, and repeatability, and it is suitable for large volume samples [51]. Dong et al. [52] found that when exosomes were separated from plasma, UCF had the highest separation purity.
However, UCF is time-consuming, and different individual operations will also lead to different results [53]. In particular, repeated ultra-high-speed centrifugation has adverse effects on the quality and quantity of exosomes [54,55]. Their structural and biological integrity may also be damaged [56]. The appearance of the isodensity gradient centrifugation method is an improvement of UCF. By constructing a density gradient medium (gradually increasing from the top to the bottom of the centrifuge tube), exosomes and the corresponding isodensity area settle together under the effect of centrifugal force, thus removing most of the contaminated proteins [57] (Figure 2B).
SEC is a widely recognized method that uses polymers to form porous stationary phases in chromatographic columns. Exosomes are separated according to the different path lengths of molecules or particles with different sizes [58] (Figure 2C). Compared with UCF, the exosomes separated via SEC have more complete physical structures and biological functions [59] and are suitable for various biological fluids [60]. However, the products obtained via the SEC method may be contaminated by a large number of proteins with low purity, which means the method is suitable for samples with small size and high yield.

3. Drug Delivery of iPSC-sEVs in the Repair of Myocardial Injury

EVs can transfer encapsulated proteins and genetic information to recipient cells and act as information messengers between cells [61]. They are natural biologics with autologous origin, while they also maintain cargo integrity and stability. Furthermore, exosomal membranes contain certain proteins that have binding affinities to specific receptors on the surface of the recipient cells. EV uptake may occur through three mechanisms: endocytosis, ligand–receptor uptake, and fusion [62]. Upon binding to a specific target cell, EVs have the ability to initiate intracellular signaling via receptor–ligand interactions, undergo internalization via endocytosis and/or phagocytosis, or even fuse with the target cell’s membrane, resulting in the transfer of their contents to the cytosol of the recipient cell. These processes ultimately lead to the modification of the physiological state of the recipient cell [63].
Rab GTP enzymes such as Rab11, Rab35, Rab27a, and Rab27b participate in the production of exosomes through vesicle budding [64,65,66]. The expression of exosomal markers such as CD63 was shown to be reduced by the silencing of Rab27a and Rab27b [67,68]. To demonstrate in vivo EV transfer between cells, a few groups have recently developed clever modifications of EVs, allowing their behavior and target cells to be tracked in vivo. For example, Lai et al. combined Gaussia luciferase with metabolic biotinylation to create a sensitive EV reporter for multimode imaging, showing that the dynamic processing of EVs has an accurate spatio-temporal resolution [69]. In order to further evaluate the accuracy of time and space, Lai et al. also designed optical reporters to label multiple EV populations, and they found that EV-borne mRNA transfer between cells and the process is dynamic and multidirectional [70]. IPSC-sEVs contain mRNAs which participate in a variety of biological processes of cell proliferation, promoting angiogenesis and paracrine response [71]. In addition to proteins and mRNAs, miRNAs and other non-coding RNAs are also possible active EVs cargoes. The miRNA secreted in sEVs can be functionally delivered to target cells, resulting in the direct modulation of their mRNA targets [72]. Mendel et al. reported miRNAs to be present in both iPSCs and iPSC-sEVs; they found miR-19b, miR-20a, miR-126-3p, miR-130a-3p and miR-210-3p were reportedly involved in the promotion of angiogenesis, adaptation to hypoxic stress, and regulation of cell cycles [73].
Exosomes are also highly engineerable, and the strategies include genetic engineering and chemical modification [74,75]. The engineering of exosomal surface proteins confers cell and tissue specificity [76]. The surface molecules anchored on exosomes from different cell sources vary, which endows them with selectivity for specific recipient cells. Bobis-Wozowicz et al. showed that iPSC-sEVs are able to transfer bioactive molecules delivered to human cardiac mesenchymal stromal cells and were found to exert protective effects by affecting the transcriptomes and proteomic profiles of the recipient cells [77]. Additionally, iPSC-sEVs combined with small-molecule RNA (miR-499) induce myocardial differentiation and improve cardiac function through the wnt/β-catenin signaling pathway in rats [78]. Jung et al. found that exosomal cargo containing miR-106a-363 improved the murine LV ejection fraction and reduced the myocardial fibrosis of the injured myocardium [79]. For the application of an in vivo model, 15–100 μg is the commonly used dose for the treatment of mouse or rat models [44], while 2–40 μg/mL is the commonly used intervention dose for in vitro studies [80].
Consequently, EVs from gene-edited patient-specific iPSCs can be directed to the specific lesions of each individual patient to promote the salvage of the existing injured cells. IPSC-sEVs hold potential for a wide spectrum of beneficial effects on cell function recovery to restore the myocardial injury by simulating and activating the endogenous repair, consisting of the native transfer of proteins, mRNAs, and miRNAs (Table 1). EVs represent the most feasible approach to translate the enormous potential of pluripotent stem cell biology.

4. Mechanism of iPSC-sEVs in the Repair of Myocardial Injury

Studies have found that iPSC-sEVs play a protective role in the treatment of CVD by regulating apoptosis, inflammation, and fibrosis, as well as promoting angiogenesis [94,95,96]. These are achieved through cell-to-cell communication, which is promoted by substances such as miRNA, small molecules, and proteins (Figure 3).

4.1. MI

The death of many CMs after MI leads to strong inflammation. IPSC-sEVs show angiogenesis and anti-inflammatory potential in the cell therapy of MI [97]. Angiogenesis is the main mechanism of improving left ventricular function through cell therapy after ischemic myocardial injury, which indicates that iPSC-sEVs are a potential target for MI therapy [98]. More and more studies have shown that exosomes derived from iPSCs can promote endogenous repair and enhance cardiac function after MI [79,99]. Takeda et al. [83] isolated exosomes from human iPSCs and administered them successively in the ischemic myocardial model of mice, which showed that iPSC-sEVs significantly improved myocardial injury after MI by reducing apoptosis and fibrosis. In vitro studies have also shown that angiogenesis and anti-apoptotic effects depend on the increased survival of CMs derived from iPSCs, and exosomes from iPSC-derived CMs (iPSC-CMs) improve myocardial recovery without increasing the probability of arrhythmogenic complications [100]. Gao et al. [101] demonstrated that exosomes from human iPSC-CMs also have cardioprotective effects in a swine MI model according to the ejection fraction, wall stress, myocardial bioenergetics, and cardiac hypertrophy. In vitro studies also showed their angiogenic and anti-apoptotic effects depending on increased endothelial cell tube formation and the survival of CMs derived from hiPSCs.

4.2. MIRI

IPSCs-sEVs can promote myocardial regeneration in MIRI, partly due to its ability to shuttle between cells, which contains a large amount of miRNA, especially miR-146a [102]. MiR-146a inhibits IRAK1 and TNF receptor-related factor 6 to reduce the activation of NF-κβ to increase cardiac function and reduce myocardial fibrosis after MIRI [103]. Furthermore, miR-21 has been proved to have beneficial effects on damaged myocardium [104,105]. MiR-21 reduces cardiomyocyte apoptosis by regulating the expression of PDCD4 and AKT pathways [106]. IPSC-sEVs are also involved in regulating signaling pathways such as WNT [107], which partially remuscularize the injured region, restore cardiac function, and reduce fibrosis in the infarcted hearts of rats by regulating actin cytoskeleton and immunogenicity. IPSC-sEVs have anti-apoptotic and antioxidant effects [108]. For example, iPSC-EVs can protect H9c2 cells from H2O2-induced oxidative stress by inhibiting the activation of caspase3/7. The intramyocardial injection of iPSC-sEVs before reperfusion can protect against MIRI. Furthermore, IPSC-sEVs deliver cardioprotective miRNAs, including nanog-regulated miR-21 and HIF-1α-regulated miR-210 [82].

4.3. Coronary Heart Disease

Coronary heart disease (CAD) is caused by coronary artery stenosis or obstruction due to atherosclerosis. According to the current view, oxidative stress, endothelial dysfunction, and inflammation are the three key factors for the occurrence and development of CAD [109,110]. Many studies have focused on the use of natural drugs and biodegradable synthetic materials for scaffolds. However, recent studies have combined the use of EVs derived from iPSCs, providing a promising solution for vascular tissue engineering [111]. IPSC-sEVs participate in paracrine and autocrine communication between cardiovascular cells through miRNAs and other mediators [112]. EVs released from iPSCs have been shown to have myocardial protective effects, which can improve the survival rate of CMs. This process is achieved by inducing macrophage polarization and reducing the transcription level of protein kinase by miR-181b [113]. Wang et al. [114] pointed out that iPSC-sEVs can increase type III collagen and fibronectin, increase vascular permeability, optimize the vascular environment, and improve cardiac function. More and more studies have confirmed that EVs from mesenchymal stem cells (MSCs) are effective drug carriers for the treatment of CAD, but their application is hindered by donor variation and traditional tissue-derived MSC expansion limitations [102,115]. While small EVs prepared from standardized MSCs derived from iPSCs (iMSC-sEVs) have unlimited scalability and have the ability to target CAD therapy [116], some studies show that they have a better protein structure than iPSC-sEVs, providing more possibilities for the prevention and treatment of CAD [117,118].

4.4. HF

The lost myocardium after MI is usually replaced by non-contractile scar tissue, which can lead to congestive heart failure (HF). As CMs are terminally differentiated cells with minimal regenerative capacity, heart transplantation is the gold standard for the treatment of HF, which faces the obstacles of the shortage of donor hearts, complications after transplantation, and the long-term failure of the transplanted heart [119]. Tian et al. [120] reviewed that the regulation of miRNAs rich in iPSCs-sEVs on Nrf2 and antioxidant proteins in the heart and brain mediates cardiac function and sympathetic excitation during HF. It is speculated that the targeted uptake ability of receptor cells can be increased when engineering exosomes with specific miRNAs or antagomirs is used to treat HF. Qiao et al. [121] confirmed that iPSC-sEVs alleviate cardiac dysfunction by regulating the Akt pathway through miR-21-5p. In recent years, lncRNA has become a key regulator of biological processes involved in the progression of HF [122]. Viereck et al. [123] focused on the potential of highly conservative lncRNAH19 and found that its expression was down-regulated in HF. The iMSC-sEVs also play an important role in heart failure. Hou et al. [124] found that iMSC-sEVs protected endothelial cells from oxidative stress by activating the Akt/Nrf2/HO-1 signaling pathway in HF models.

5. Challenges in the Treatment of CVD with IPSC-sEVs

It has been confirmed that iPSC-sEVs promote heart repair after MI, which means they are superior to iPSCs [21]. EVs provide a feasible alternative cell-free therapy in iPSC medicine. Because of their low immunogenicity, they does not seek a host immune response, so there is no need to match donor and recipient [125,126]. However, there are still many problems with the treatment of EVs, such as their production, stability, half-life, and delivery efficiency. Therefore, it is particularly necessary to comprehensively analyze the chemical and functional characteristics of the EVs and to study their physiological characteristics, diversity, and transport mode.
Chandy et al. [127] drew a map of microRNAs in cardiac extracellular secretions derived from human iPSCs. Human iPSCs were differentiated into iPSC-CMs, iPSC-ECs and iPSC-CFs, and the EVs were isolated. Their miRNA content was sequenced and compared with the source cells. Interestingly, only a part of cells miRNAs was found to be secreted in the EVs and was cell-specific. A comparative analysis showed a decrease in miR-22 expression in exosomes from cardiac-fibroblast-derived hiPSCs compared with dermal-fibroblast-derived hiPSC exosomes [27]. Future research needs to conduct in-depth sequencing analyses to understand the role of other non-coding RNAs in mediating the improvement of cardiac function. In addition, since iPSC-sEVs carry miRNA and each miRNA has multiple target genes, it is also necessary to prevent the occurrence of adverse non-target effects.
IPSCs differentiate into CMs, which are equivalent to fetal CMs, and lack the electrophysiological and ultrastructural characteristics of mature CMs [128,129], such as fully functional seromuscular reticular structure and transverse canal system. After differentiation, the maximum contractility was lower, calcium storage and circulation decreased, and the mitochondrial function was immature [130]. In addition, the EVs’ function was also affected. The current research is mainly focused on using the paracrine function of iPSCs to play a role, rather than ensuring they differentiate into therapeutic cells [131,132,133]. Even so, the content and level of iPSC-sEVs will change after serum starvation and hypoxia treatment [134,135], which makes clinical treatment more difficult. Nachlas et al. [136] highlight the importance of a 3D culture environment to influence cell phenotype and function. In addition, 3D-printed cardiac patches and personalized hydrogel can help iPSCs’ further maturation [136,137,138]. Furthermore, gene editing technology can be used to achieve the richness of iPSC cells [26,139]. For instance, CRISPR/Cas9 is used for gene editing based on homologous recombination to obtain mutation-corrected iPSCs so that the pathogenic mutation can be corrected without preserving the genetic footprint [26].
However, if these molecules are to be used in clinical therapy, the standard procedures for purifying exosomes need to be optimized. Overall, small EVs play critical roles in cell–cell communication through endocytosis, phagocytosis, and membrane fusion. EV uptake was found to correlate with intracellular and microenvironmental acidity [140,141], suggesting that the microenvironment influences the delivery efficiency of EVs. In the case of factors operating at the intracellular level, delivery into the correct cellular compartments while maintaining the stability, integrity, and biological potency of these factors remains challenging.
Furthermore, the content of exosomes can be modified by stress preconditioning [142], serum deprivation [143], or the genetic modification and epigenetic reprogramming of iPSCs [144,145,146]. Recent studies show that exosomes can cross the BBB (blood–brain barrier), and a leaky BBB state in mental disorders (such as stress, depression, and schizophrenia) may be initiated by exosomes released from cells being influenced by this disease state [147]. Chronic stress can cause immune disorders and inflammatory responses. Moreover, exosomal components are strongly influenced by inflammatory signals such as LPS, tumor necrosis factor (TNF)-α [148], and interferon (IFN)-γ [149]. They could modulate the therapeutic efficacy via the regulation of differential gene expressions [150,151] and largely influence the effect of iPSC-sEV treatment.

6. Prospects and Conclusions

The potential of IPSC-sEVs in the treatment of CVD is exciting. Compared with cells, EVs cannot self-replicate, which reduces tumor toxicity. The future application of IPSC-sEVs is likely to be combined with other drugs or systems. With the development of front-line technologies, including scRNA-seq, multi-omics, genome editing, and machine learning, they possess great potential for the analysis of exosome contents and their transfer specificity [152]. Exosomes can be endogenously modified by the genetic modification of production cells to produce cells overexpressing desired therapeutic substances that are eventually incorporated into exosomes upon secretion [153]. Alternatively, exosomes can be loaded exogenously using various techniques, such as sonication [154], membrane permeabilization [155], and extrusion [156]. Therefore, in order to ensure their safety and effectiveness, a number of challenges must be addressed, including the characteristics of the content, specific molecular mechanisms for disease treatment, and biosafety as a drug delivery system. In short, more basic research and new technologies are needed to fully realize the therapeutic potential of exosomes derived from iPSCs and accelerate their clinical application.

Author Contributions

W.-T.M. and H.-D.G. conceived the structure of the manuscript. W.-T.M. wrote the manuscript and constructed the figures. H.-D.G. reviewed and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (82174120, 81970991), Natural Science Foundation of Shanghai (No. 21ZR1463100), Shanghai Talent Development Funding Scheme (No. 2019090), and Program of Shanghai Academic Research Leader (22XD1423400).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

References

  1. Kovacic, J.C.; Dimmeler, S.; Harvey, R.P.; Finkel, T.; Aikawa, E.; Krenning, G.; Baker, A.H. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 190–209. [Google Scholar] [CrossRef] [PubMed]
  2. Li, H.; Sureda, A.; Devkota, H.P.; Pittalà, V.; Barreca, D.; Silva, A.S.; Tewari, D.; Xu, S.; Nabavi, S.M. Curcumin, the golden spice in treating cardiovascular diseases. Biotechnol. Adv. 2020, 38, 107343. [Google Scholar] [CrossRef] [PubMed]
  3. Liu, C.; Du, L.; Wang, S.; Kong, L.; Zhang, S.; Li, S.; Zhang, W.; Du, G. Differences in the prevention and control of cardiovascular and cerebrovascular diseases. Pharmacol. Res. 2021, 170, 105737. [Google Scholar] [CrossRef]
  4. Pala, R.; Pattnaik, S.; Busi, S.; Nauli, S.M. Nanomaterials as Novel Cardiovascular Theranostics. Pharmaceutics 2021, 13, 348. [Google Scholar] [CrossRef]
  5. Terashvili, M.; Bosnjak, Z.J. Stem Cell Therapies in Cardiovascular Disease. J. Cardiothorac. Vasc. Anesthesia 2019, 33, 209–222. [Google Scholar] [CrossRef] [PubMed]
  6. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130. [Google Scholar] [CrossRef]
  7. Gao, P.; Liu, S.; Wang, X.; Ikeya, M. Dental applications of induced pluripotent stem cells and their derivatives. Jpn. Dent. Sci. Rev. 2022, 58, 162–171. [Google Scholar] [CrossRef]
  8. Takahashi, K.; Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [Green Version]
  9. Deuse, T.; Hu, X.; Gravina, A.; Wang, D.; Tediashvili, G.; De, C.; Thayer, W.O.; Wahl, A.; Garcia, J.V.; Reichenspurner, H.; et al. Hypoimmunogenic derivatives of induced pluripotent stem cells evade immune rejection in fully immunocompetent allogeneic recipients. Nat. Biotechnol. 2019, 37, 252–258. [Google Scholar] [CrossRef]
  10. Doss, M.X.; Sachinidis, A. Current Challenges of iPSC-Based Disease Modeling and Therapeutic Implications. Cells 2019, 8, 403. [Google Scholar] [CrossRef] [Green Version]
  11. Crow, D. Could iPSCs Enable "Off-the-Shelf" Cell Therapy? Cell 2019, 177, 1667–1669. [Google Scholar] [CrossRef]
  12. Gao, L.; Wang, L.; Wei, Y.; Krishnamurthy, P.; Walcott, G.P.; Menasché, P.; Zhang, J. Exosomes secreted by hiPSC-derived cardiac cells improve recovery from myocardial infarction in swine. Sci. Transl. Med. 2020, 12, eaay1318. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, L.; Michowski, W.; Kolodziejczyk, A.; Sicinski, P. The cell cycle in stem cell proliferation, pluripotency and differentiation. Nat. Cell Biol. 2019, 21, 1060–1067. [Google Scholar] [CrossRef] [PubMed]
  14. van der Pol, E.; Böing, A.N.; Harrison, P.; Sturk, A.; Nieuwland, R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacol. Rev. 2012, 64, 676–705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Veerman, R.E.; Teeuwen, L.; Czarnewski, P.; Güclüler Akpinar, G.; Sandberg, A.; Cao, X.; Pernemalm, M.; Orre, L.M.; Gabrielsson, S.; Eldh, M. Molecular evaluation of five different isolation methods for extracellular vesicles reveals different clinical applicability and subcellular origin. J. Extracell. Vesicles 2021, 10, e12128. [Google Scholar] [CrossRef] [PubMed]
  16. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  17. Davidson, S.M.; Yellon, D.M. Exosomes and cardioprotection—A critical analysis. Mol. Asp. Med. 2018, 60, 104–114. [Google Scholar] [CrossRef]
  18. Maqsood, M.; Kang, M.; Wu, X.; Chen, J.; Teng, L.; Qiu, L. Adult mesenchymal stem cells and their exosomes: Sources, characteristics, and application in regenerative medicine. Life Sci. 2020, 256, 118002. [Google Scholar] [CrossRef]
  19. Yaghoubi, Y.; Movassaghpour, A.; Zamani, M.; Talebi, M.; Mehdizadeh, A.; Yousefi, M. Human umbilical cord mesenchymal stem cells derived-exosomes in diseases treatment. Life Sci. 2019, 233, 116733. [Google Scholar] [CrossRef]
  20. Peng, L.; Chen, Y.; Shi, S.; Wen, H. Stem cell-derived and circulating exosomal microRNAs as new potential tools for diabetic nephropathy management. Stem Cell Res. Ther. 2022, 13, 25. [Google Scholar] [CrossRef]
  21. Adamiak, M.; Cheng, G.; Bobis-Wozowicz, S.; Zhao, L.; Kedracka-Krok, S.; Samanta, A.; Karnas, E.; Xuan, Y.T.; Skupien-Rabian, B.; Chen, X.; et al. Induced Pluripotent Stem Cell (iPSC)-Derived Extracellular Vesicles Are Safer and More Effective for Cardiac Repair Than iPSCs. Circ. Res. 2018, 122, 296–309. [Google Scholar] [CrossRef] [PubMed]
  22. Goldenson, B.H.; Hor, P.; Kaufman, D.S. iPSC-Derived Natural Killer Cell Therapies—Expansion and Targeting. Front. Immunol. 2022, 13, 841107. [Google Scholar] [CrossRef] [PubMed]
  23. Mo, J.; Anastasaki, C.; Chen, Z.; Shipman, T.; Papke, J.; Yin, K.; Gutmann, D.H.; Le, L.Q. Humanized neurofibroma model from induced pluripotent stem cells delineates tumor pathogenesis and developmental origins. J. Clin. Investig. 2021, 131, e139807. [Google Scholar] [CrossRef]
  24. La Greca, A.; Solari, C.; Furmento, V.; Lombardi, A.; Biani, M.C.; Aban, C.; Moro, L.; García, M.; Guberman, A.S.; Sevlever, G.E.; et al. Extracellular vesicles from pluripotent stem cell-derived mesenchymal stem cells acquire a stromal modulatory proteomic pattern during differentiation. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef] [Green Version]
  25. Hu, G.W.; Li, Q.; Niu, X.; Hu, B.; Liu, J.; Zhou, S.M.; Guo, S.C.; Lang, H.L.; Zhang, C.Q.; Wang, Y.; et al. Exosomes secreted by human-induced pluripotent stem cell-derived mesenchymal stem cells attenuate limb ischemia by promoting angiogenesis in mice. Stem Cell Res. Ther. 2015, 6, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sun, R.; Liu, Y.; Lu, M.; Ding, Q.; Wang, P.; Zhang, H.; Tian, X.; Lu, P.; Meng, D.; Sun, N.; et al. ALIX increases protein content and protective function of iPSC-derived exosomes. J. Mol. Med. 2019, 97, 829–844. [Google Scholar] [CrossRef]
  27. Kurtzwald-Josefson, E.; Zeevi-Levin, N.; Rubchevsky, V.; Bechar Erdman, N.; Schwartz Rohaker, O.; Nahum, O.; Hochhauser, E.; Ben-Avraham, B.; Itskovitz-Eldor, J.; Aravot, D.; et al. Cardiac Fibroblast-Induced Pluripotent Stem Cell-Derived Exosomes as a Potential Therapeutic Mean for Heart Failure. Int. J. Mol. Sci. 2020, 21, 7215. [Google Scholar] [CrossRef]
  28. Alzhrani, G.N.; Alanazi, S.T.; Alsharif, S.Y.; Albalawi, A.M.; Alsharif, A.A.; Abdel-Maksoud, M.S.; Elsherbiny, N. Exosomes: Isolation, characterization, and biomedical applications. Cell Biol. Int. 2021, 45, 1807–1831. [Google Scholar] [CrossRef]
  29. Gandham, S.; Su, X.; Wood, J.; Nocera, A.L.; Alli, S.C.; Milane, L.; Zimmerman, A.; Amiji, M.; Ivanov, A.R. Technologies and Standardization in Research on Extracellular Vesicles. Trends Biotechnol. 2020, 38, 1066–1098. [Google Scholar] [CrossRef] [PubMed]
  30. Chen, B.Y.; Sung, C.W.; Chen, C.; Cheng, C.M.; Lin, D.P.; Huang, C.T.; Hsu, M.Y. Advances in exosomes technology. Clin. Chim. Acta 2019, 493, 14–19. [Google Scholar] [CrossRef]
  31. Iwai, K.; Minamisawa, T.; Suga, K.; Yajima, Y.; Shiba, K. Isolation of human salivary extracellular vesicles by iodixanol density gradient ultracentrifugation and their characterizations. J. Extracell. Vesicles 2016, 5, 30829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Böing, A.N.; van der Pol, E.; Grootemaat, A.E.; Coumans, F.A.; Sturk, A.; Nieuwland, R. Single-step isolation of extracellular vesicles by size-exclusion chromatography. J. Extracell. Vesicles 2014, 3, 23430. [Google Scholar] [CrossRef]
  33. Brown, P.N.; Yin, H. Polymer-Based Purification of Extracellular Vesicles. Methods Mol. Biol. 2017, 1660, 91–103. [Google Scholar] [CrossRef] [PubMed]
  34. Wijerathne, H.; Witek, M.A.; Jackson, J.M.; Brown, V.; Hupert, M.L.; Herrera, K.; Kramer, C.; Davidow, A.E.; Li, Y.; Baird, A.E.; et al. Affinity enrichment of extracellular vesicles from plasma reveals mRNA changes associated with acute ischemic stroke. Commun. Biol. 2020, 3, 613. [Google Scholar] [CrossRef]
  35. Wan, Y.; Cheng, G.; Liu, X.; Hao, S.J.; Nisic, M.; Zhu, C.D.; Xia, Y.Q.; Li, W.Q.; Wang, Z.G.; Zhang, W.L.; et al. Rapid magnetic isolation of extracellular vesicles via lipid-based nanoprobes. Nat. Biomed. Eng. 2017, 1, 0058. [Google Scholar] [CrossRef] [Green Version]
  36. Kim, Y.; Zhao, X. Magnetic Soft Materials and Robots. Chem. Rev. 2022, 122, 5317–5364. [Google Scholar] [CrossRef]
  37. Seo, N.; Nakamura, J.; Kaneda, T.; Tateno, H.; Shimoda, A.; Ichiki, T.; Furukawa, K.; Hirabayashi, J.; Akiyoshi, K.; Shiku, H. Distinguishing functional exosomes and other extracellular vesicles as a nucleic acid cargo by the anion-exchange method. J. Extracell. Vesicles 2022, 11, e12205. [Google Scholar] [CrossRef]
  38. Nordin, J.Z.; Lee, Y.; Vader, P.; Mäger, I.; Johansson, H.J.; Heusermann, W.; Wiklander, O.P.; Hällbrink, M.; Seow, Y.; Bultema, J.J.; et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomedicine 2015, 11, 879–883. [Google Scholar] [CrossRef] [Green Version]
  39. Staubach, S.; Bauer, F.N.; Tertel, T.; Börger, V.; Stambouli, O.; Salzig, D.; Giebel, B. Scaled preparation of extracellular vesicles from conditioned media. Adv. Drug Deliv. Rev. 2021, 177, 113940. [Google Scholar] [CrossRef]
  40. Zhu, L.; Sun, H.T.; Wang, S.; Huang, S.L.; Zheng, Y.; Wang, C.Q.; Hu, B.Y.; Qin, W.; Zou, T.T.; Fu, Y.; et al. Isolation and characterization of exosomes for cancer research. J. Hematol. Oncol. 2020, 13, 152. [Google Scholar] [CrossRef]
  41. Zhu, L.; Kalimuthu, S.; Gangadaran, P.; Oh, J.M.; Lee, H.W.; Baek, S.H.; Jeong, S.Y.; Lee, S.W.; Lee, J.; Ahn, B.C. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics 2017, 7, 2732–2745. [Google Scholar] [CrossRef]
  42. Wang, J.; Ma, P.; Kim, D.H.; Liu, B.F.; Demirci, U. Towards Microfluidic-Based Exosome Isolation and Detection for Tumor Therapy. Nano Today 2021, 37, 101066. [Google Scholar] [CrossRef] [PubMed]
  43. Bo, Y.; Yang, L.; Liu, B.; Tian, G.; Li, C.; Zhang, L.; Yan, Y. Exosomes from human induced pluripotent stem cells-derived keratinocytes accelerate burn wound healing through miR-762 mediated promotion of keratinocytes and endothelial cells migration. J. Nanobiotechnol. 2022, 20, 291. [Google Scholar] [CrossRef] [PubMed]
  44. Marzano, M.; Bejoy, J.; Cheerathodi, M.R.; Sun, L.; York, S.B.; Zhao, J.; Kanekiyo, T.; Bu, G.; Meckes, D.G., Jr.; Li, Y. Differential Effects of Extracellular Vesicles of Lineage-Specific Human Pluripotent Stem Cells on the Cellular Behaviors of Isogenic Cortical Spheroids. Cells 2019, 8, 993. [Google Scholar] [CrossRef] [Green Version]
  45. Gardiner, C.; Ferreira, Y.J.; Dragovic, R.A.; Redman, C.W.; Sargent, I.L. Extracellular vesicle sizing and enumeration by nanoparticle tracking analysis. J. Extracell. Vesicles 2013, 2, 19671. [Google Scholar] [CrossRef] [PubMed]
  46. Lobb, R.J.; Becker, M.; Wen, S.W.; Wong, C.S.; Wiegmans, A.P.; Leimgruber, A.; Möller, A. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 2015, 4, 27031. [Google Scholar] [CrossRef] [PubMed]
  47. Ueda, K.; Ishikawa, N.; Tatsuguchi, A.; Saichi, N.; Fujii, R.; Nakagawa, H. Antibody-coupled monolithic silica microtips for highthroughput molecular profiling of circulating exosomes. Sci. Rep. 2014, 4, 6232. [Google Scholar] [CrossRef] [Green Version]
  48. Pocsfalvi, G.; Stanly, C.; Vilasi, A.; Fiume, I.; Capasso, G.; Turiák, L.; Buzas, E.I.; Vékey, K. Mass spectrometry of extracellular vesicles. Mass Spectrom. Rev. 2016, 35, 3–21. [Google Scholar] [CrossRef]
  49. de Rus Jacquet, A.; Tancredi, J.L.; Lemire, A.L.; DeSantis, M.C.; Li, W.P.; O’Shea, E.K. The LRRK2 G2019S mutation alters astrocyte-to-neuron communication via extracellular vesicles and induces neuron atrophy in a human iPSC-derived model of Parkinson’s disease. eLife 2021, 10, e73062. [Google Scholar] [CrossRef]
  50. Street, J.M.; Barran, P.E.; Mackay, C.L.; Weidt, S.; Balmforth, C.; Walsh, T.S.; Chalmers, R.T.; Webb, D.J.; Dear, J.W. Identification and proteomic profiling of exosomes in human cerebrospinal fluid. J. Transl. Med. 2012, 10, 5. [Google Scholar] [CrossRef] [Green Version]
  51. Chen, J.; Li, P.; Zhang, T.; Xu, Z.; Huang, X.; Wang, R.; Du, L. Review on Strategies and Technologies for Exosome Isolation and Purification. Front. Bioeng. Biotechnol. 2021, 9, 811971. [Google Scholar] [CrossRef] [PubMed]
  52. Dong, L.; Zieren, R.C.; Horie, K.; Kim, C.J.; Mallick, E.; Jing, Y.; Feng, M.; Kuczler, M.D.; Green, J.; Amend, S.R.; et al. Comprehensive evaluation of methods for small extracellular vesicles separation from human plasma, urine and cell culture medium. J. Extracell. Vesicles 2020, 10, e12044. [Google Scholar] [CrossRef]
  53. Koh, Y.Q.; Almughlliq, F.B.; Vaswani, K.; Peiris, H.N.; Mitchell, M.D. Exosome enrichment by ultracentrifugation and size exclusion chromatography. Front. Biosci. (Landmark Ed.) 2018, 23, 865–874. [Google Scholar] [CrossRef] [Green Version]
  54. Li, S.; Yi, M.; Dong, B.; Tan, X.; Luo, S.; Wu, K. The role of exosomes in liquid biopsy for cancer diagnosis and prognosis prediction. Int. J. Cancer 2021, 148, 2640–2651. [Google Scholar] [CrossRef]
  55. Yu, D.; Li, Y.; Wang, M.; Gu, J.; Xu, W.; Cai, H.; Fang, X.; Zhang, X. Exosomes as a new frontier of cancer liquid biopsy. Mol. Cancer 2022, 21, 56. [Google Scholar] [CrossRef]
  56. Northrop-Albrecht, E.J.; Taylor, W.R.; Huang, B.Q.; Kisiel, J.B.; Lucien, F. Assessment of extracellular vesicle isolation methods from human stool supernatant. J. Extracell. Vesicles 2022, 11, e12208. [Google Scholar] [CrossRef]
  57. Onódi, Z.; Pelyhe, C.; Terézia Nagy, C.; Brenner, G.B.; Almási, L.; Kittel, Á.; Manček-Keber, M.; Ferdinandy, P.; Buzás, E.I.; Giricz, Z. Isolation of High-Purity Extracellular Vesicles by the Combination of Iodixanol Density Gradient Ultracentrifugation and Bind-Elute Chromatography From Blood Plasma. Front. Physiol. 2018, 9, 1479. [Google Scholar] [CrossRef] [Green Version]
  58. Liu, Y.; Xu, C.; Gao, T.; Chen, X.; Wang, J.; Yu, P.; Mao, L. Sizing Single Particles at the Orifice of a Nanopipette. ACS Sens. 2020, 5, 2351–2358. [Google Scholar] [CrossRef]
  59. Guo, J.; Wu, C.; Lin, X.; Zhou, J.; Zhang, J.; Zheng, W.; Wang, T.; Cui, Y. Establishment of a simplified dichotomic size-exclusion chromatography for isolating extracellular vesicles toward clinical applications. J. Extracell. Vesicles 2021, 10, e12145. [Google Scholar] [CrossRef]
  60. Karimi, N.; Cvjetkovic, A.; Jang, S.C.; Crescitelli, R.; Hosseinpour Feizi, M.A.; Nieuwland, R.; Lötvall, J.; Lässer, C. Detailed analysis of the plasma extracellular vesicle proteome after separation from lipoproteins. Cell. Mol. Life Sci. 2018, 75, 2873–2886. [Google Scholar] [CrossRef] [Green Version]
  61. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
  62. Fitzner, D.; Schnaars, M.; van Rossum, D.; Krishnamoorthy, G.; Dibaj, P.; Bakhti, M.; Regen, T.; Hanisch, U.K.; Simons, M. Selective transfer of exosomes from oligodendrocytes to microglia by macropinocytosis. J. Cell Sci. 2011, 124, 447–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Tkach, M.; Théry, C. Communication by Extracellular Vesicles: Where We Are and Where We Need to Go. Cell 2016, 164, 1226–1232. [Google Scholar] [CrossRef] [Green Version]
  64. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 2010, 12, 19–30. [Google Scholar] [CrossRef] [Green Version]
  65. Hsu, C.; Morohashi, Y.; Yoshimura, S.; Manrique-Hoyos, N.; Jung, S.; Lauterbach, M.A.; Bakhti, M.; Grønborg, M.; Möbius, W.; Rhee, J.; et al. Regulation of exosome secretion by Rab35 and its GTPase-activating proteins TBC1D10A-C. J. Cell Biol. 2010, 189, 223–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chutna, O.; Gonçalves, S.; Villar-Piqué, A.; Guerreiro, P.; Marijanovic, Z.; Mendes, T.; Ramalho, J.; Emmanouilidou, E.; Ventura, S.; Klucken, J.; et al. The small GTPase Rab11 co-localizes with α-synuclein in intracellular inclusions and modulates its aggregation, secretion and toxicity. Hum. Mol. Genet. 2014, 23, 6732–6745. [Google Scholar] [CrossRef] [Green Version]
  67. Sung, B.H.; Ketova, T.; Hoshino, D.; Zijlstra, A.; Weaver, A.M. Directional cell movement through tissues is controlled by exosome secretion. Nat. Commun. 2015, 6, 7164. [Google Scholar] [CrossRef] [Green Version]
  68. Bobrie, A.; Krumeich, S.; Reyal, F.; Recchi, C.; Moita, L.F.; Seabra, M.C.; Ostrowski, M.; Théry, C. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 2012, 72, 4920–4930. [Google Scholar] [CrossRef] [Green Version]
  69. Lai, C.P.; Mardini, O.; Ericsson, M.; Prabhakar, S.; Maguire, C.; Chen, J.W.; Tannous, B.A.; Breakefield, X.O. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 2014, 8, 483–494. [Google Scholar] [CrossRef] [Green Version]
  70. Lai, C.P.; Kim, E.Y.; Badr, C.E.; Weissleder, R.; Mempel, T.R.; Tannous, B.A.; Breakefield, X.O. Visualization and tracking of tumour extracellular vesicle delivery and RNA translation using multiplexed reporters. Nat. Commun. 2015, 6, 7029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Lu, M.; Peng, L.; Ming, X.; Wang, X.; Cui, A.; Li, Y.; Wang, X.; Meng, D.; Sun, N.; Xiang, M.; et al. Enhanced wound healing promotion by immune response-free monkey autologous iPSCs and exosomes vs. their allogeneic counterparts. EBioMedicine 2019, 42, 443–457. [Google Scholar] [CrossRef] [Green Version]
  72. Pegtel, D.M.; Cosmopoulos, K.; Thorley-Lawson, D.A.; van Eijndhoven, M.A.; Hopmans, E.S.; Lindenberg, J.L.; de Gruijl, T.D.; Würdinger, T.; Middeldorp, J.M. Functional delivery of viral miRNAs via exosomes. Proc. Natl. Acad. Sci. USA 2010, 107, 6328–6333. [Google Scholar] [CrossRef] [Green Version]
  73. Mendell, J.T. miRiad roles for the miR-17-92 cluster in development and disease. Cell 2008, 133, 217–222. [Google Scholar] [CrossRef] [Green Version]
  74. Armstrong, J.P.; Holme, M.N.; Stevens, M.M. Re-Engineering Extracellular Vesicles as Smart Nanoscale Therapeutics. ACS Nano 2017, 11, 69–83. [Google Scholar] [CrossRef] [Green Version]
  75. Luo, R.; Liu, M.; Tan, T.; Yang, Q.; Wang, Y.; Men, L.; Zhao, L.; Zhang, H.; Wang, S.; Xie, T.; et al. Emerging Significance and Therapeutic Potential of Extracellular vesicles. Int. J. Biol. Sci. 2021, 17, 2476–2486. [Google Scholar] [CrossRef]
  76. Xitong, D.; Xiaorong, Z. Targeted therapeutic delivery using engineered exosomes and its applications in cardiovascular diseases. Gene 2016, 575, 377–384. [Google Scholar] [CrossRef]
  77. Bobis-Wozowicz, S.; Kmiotek, K.; Sekula, M.; Kedracka-Krok, S.; Kamycka, E.; Adamiak, M.; Jankowska, U.; Madetko-Talowska, A.; Sarna, M.; Bik-Multanowski, M.; et al. Human Induced Pluripotent Stem Cell-Derived Microvesicles Transmit RNAs and Proteins to Recipient Mature Heart Cells Modulating Cell Fate and Behavior. Stem Cells 2015, 33, 2748–2761. [Google Scholar] [CrossRef]
  78. Zhang, L.L.; Liu, J.J.; Liu, F.; Liu, W.H.; Wang, Y.S.; Zhu, B.; Yu, B. MiR-499 induces cardiac differentiation of rat mesenchymal stem cells through wnt/β-catenin signaling pathway. Biochem. Biophys. Res. Commun. 2012, 420, 875–881. [Google Scholar] [CrossRef]
  79. Jung, J.H.; Ikeda, G.; Tada, Y.; von Bornstädt, D.; Santoso, M.R.; Wahlquist, C.; Rhee, S.; Jeon, Y.J.; Yu, A.C.; O’Brien, C.G.; et al. miR-106a-363 cluster in extracellular vesicles promotes endogenous myocardial repair via Notch3 pathway in ischemic heart injury. Basic Res. Cardiol. 2021, 116, 19. [Google Scholar] [CrossRef]
  80. Luo, Y.; Gao, D.; Wang, P.; Lou, C.; Li, T.; Niu, W.; Gao, Y. Optimized culture methods for isolating small extracellular vesicles derived from human induced pluripotent stem cells. J. Extracell. Vesicles 2021, 10, e12065. [Google Scholar] [CrossRef]
  81. Zhu, D.; Li, Z.; Huang, K.; Caranasos, T.G.; Rossi, J.S.; Cheng, K. Minimally invasive delivery of therapeutic agents by hydrogel injection into the pericardial cavity for cardiac repair. Nat. Commun. 2021, 12, 1412. [Google Scholar] [CrossRef]
  82. Wang, Y.; Zhang, L.; Li, Y.; Chen, L.; Wang, X.; Guo, W.; Zhang, X.; Qin, G.; He, S.H.; Zimmerman, A.; et al. Exosomes/microvesicles from induced pluripotent stem cells deliver cardioprotective miRNAs and prevent cardiomyocyte apoptosis in the ischemic myocardium. Int. J. Cardiol. 2015, 192, 61–69. [Google Scholar] [CrossRef] [Green Version]
  83. Takeda, M.; Kanki, Y.; Masumoto, H.; Funakoshi, S.; Hatani, T.; Fukushima, H.; Izumi-Taguchi, A.; Matsui, Y.; Shimamura, T.; Yoshida, Y.; et al. Identification of Cardiomyocyte-Fated Progenitors from Human-Induced Pluripotent Stem Cells Marked with CD82. Cell Rep. 2018, 22, 546–556. [Google Scholar] [CrossRef] [Green Version]
  84. Ellis, B.W.; Ronan, G.; Ren, X.; Bahcecioglu, G.; Senapati, S.; Anderson, D.; Handberg, E.; March, K.L.; Chang, H.C.; Zorlutuna, P. Human Heart Anoxia and Reperfusion Tissue (HEART) Model for the Rapid Study of Exosome Bound miRNA Expression As Biomarkers for Myocardial Infarction. Small 2022, 18, e2201330. [Google Scholar] [CrossRef]
  85. Jiang, X.; Sucharov, J.; Stauffer, B.L.; Miyamoto, S.D.; Sucharov, C.C. Exosomes from pediatric dilated cardiomyopathy patients modulate a pathological response in cardiomyocytes. Am. J. Physiol. Circ. Physiol. 2017, 312, H818–H826. [Google Scholar] [CrossRef]
  86. Turner, A.; Aggarwal, P.; Matter, A.; Olson, B.; Gu, C.C.; Hunt, S.C.; Lewis, C.E.; Arnett, D.K.; Lorier, R.; Broeckel, U. Donor-specific phenotypic variation in hiPSC cardiomyocyte-derived exosomes impacts endothelial cell function. Am. J. Physiol. Circ. Physiol. 2021, 320, H954–H968. [Google Scholar] [CrossRef]
  87. Srivastava, D.; Ivey, K.N. Potential of stem-cell-based therapies for heart disease. Nature 2006, 441, 1097–1099. [Google Scholar] [CrossRef]
  88. Gartz, M.; Darlington, A.; Afzal, M.Z.; Strande, J.L. Exosomes exert cardioprotection in dystrophin-deficient cardiomyocytes via ERK1/2-p38/MAPK signaling. Sci. Rep. 2018, 8, 16519. [Google Scholar] [CrossRef] [Green Version]
  89. Lee, W.H.; Chen, W.Y.; Shao, N.Y.; Xiao, D.; Qin, X.; Baker, N.; Bae, H.R.; Wei, T.T.; Wang, Y.; Shukla, P.; et al. Comparison of Non-Coding RNAs in Exosomes and Functional Efficacy of Human Embryonic Stem Cell- versus Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cells 2017, 35, 2138–2149. [Google Scholar] [CrossRef] [Green Version]
  90. Chen, M.; Chen, J.; Huang, W.; Li, C.; Luo, H.; Xue, Z.; Xiao, Y.; Wu, Q.; Chen, C. Exosomes from human induced pluripotent stem cells derived mesenchymal stem cells improved myocardial injury caused by severe acute pancreatitis through activating Akt/Nrf2/HO-1 axis. Cell Cycle 2022, 21, 1578–1589. [Google Scholar] [CrossRef]
  91. Zhang, J.; Guan, J.; Niu, X.; Hu, G.; Guo, S.; Li, Q.; Xie, Z.; Zhang, C.; Wang, Y. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J. Transl. Med. 2015, 13, 49. [Google Scholar] [CrossRef] [Green Version]
  92. El Harane, N.; Kervadec, A.; Bellamy, V.; Pidial, L.; Neametalla, H.J.; Perier, M.-C.; Lima Correa, B.; Thiébault, L.; Cagnard, N.; Duché, A.; et al. Acellular therapeutic approach for heart failure: In vitro production of extracellular vesicles from human cardiovascular progenitors. Eur. Heart J. 2018, 39, 1835–1847. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Mayourian, J.; Ceholski, D.K.; Gorski, P.A.; Mathiyalagan, P.; Murphy, J.F.; Salazar, S.I.; Stillitano, F.; Hare, J.M.; Sahoo, S.; Hajjar, R.J.; et al. Exosomal microRNA-21-5p Mediates Mesenchymal Stem Cell Paracrine Effects on Human Cardiac Tissue Contractility. Circ. Res. 2018, 122, 933–944. [Google Scholar] [CrossRef]
  94. Jung, J.H.; Fu, X.; Yang, P.C. Exosomes Generated From iPSC-Derivatives: New Direction for Stem Cell Therapy in Human Heart Diseases. Circ. Res. 2017, 120, 407–417. [Google Scholar] [CrossRef] [Green Version]
  95. Yuan, X.; Li, D.; Chen, X.; Han, C.; Xu, L.; Huang, T.; Dong, Z.; Zhang, M. Extracellular vesicles from human-induced pluripotent stem cell-derived mesenchymal stromal cells (hiPSC-MSCs) protect against renal ischemia/reperfusion injury via delivering specificity protein (SP1) and transcriptional activating of sphingosine kinase 1 and inhibiting necroptosis. Cell Death Dis. 2017, 8, 3200. [Google Scholar] [CrossRef] [Green Version]
  96. Dougherty, J.A.; Kumar, N.; Noor, M.; Angelos, M.G.; Khan, M.; Chen, C.A.; Khan, M. Extracellular Vesicles Released by Human Induced-Pluripotent Stem Cell-Derived Cardiomyocytes Promote Angiogenesis. Front. Physiol. 2018, 9, 1794. [Google Scholar] [CrossRef] [Green Version]
  97. Zununi Vahed, S.; Barzegari, A.; Zuluaga, M.; Letourneur, D.; Pavon-Djavid, G. Myocardial infarction and gut microbiota: An incidental connection. Pharmacol. Res. 2018, 129, 308–317. [Google Scholar] [CrossRef]
  98. Zhao, M.; Nakada, Y.; Wei, Y.; Bian, W.; Chu, Y.; Borovjagin, A.V.; Xie, M.; Zhu, W.; Nguyen, T.; Zhou, Y.; et al. Cyclin D2 Overexpression Enhances the Efficacy of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Myocardial Repair in a Swine Model of Myocardial Infarction. Circulation 2021, 144, 210–228. [Google Scholar] [CrossRef]
  99. Lozano, J.; Rai, A.; Lees, J.G.; Fang, H.; Claridge, B.; Lim, S.Y.; Greening, D.W. Scalable Generation of Nanovesicles from Human-Induced Pluripotent Stem Cells for Cardiac Repair. Int. J. Mol. Sci. 2022, 23, 14334. [Google Scholar] [CrossRef]
  100. Sharma, A.; Zhang, Y.; Buikema, J.W.; Serpooshan, V.; Chirikian, O.; Kosaric, N.; Churko, J.M.; Dzilic, E.; Shieh, A.; Burridge, P.W.; et al. Stage-specific Effects of Bioactive Lipids on Human iPSC Cardiac Differentiation and Cardiomyocyte Proliferation. Sci. Rep. 2018, 8, 6618. [Google Scholar] [CrossRef] [Green Version]
  101. Gao, L.; Gregorich, Z.R.; Zhu, W.; Mattapally, S.; Oduk, Y.; Lou, X.; Kannappan, R.; Borovjagin, A.V.; Walcott, G.P.; Pollard, A.E.; et al. Large Cardiac Muscle Patches Engineered From Human Induced-Pluripotent Stem Cell-Derived Cardiac Cells Improve Recovery From Myocardial Infarction in Swine. Circulation 2018, 137, 1712–1730. [Google Scholar] [CrossRef] [PubMed]
  102. Ibrahim, A.G.; Cheng, K.; Marbán, E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Rep. 2014, 2, 606–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Wang, X.; Ha, T.; Liu, L.; Zou, J.; Zhang, X.; Kalbfleisch, J.; Gao, X.; Williams, D.; Li, C. Increased expression of microRNA-146a decreases myocardial ischaemia/reperfusion injury. Cardiovasc. Res. 2013, 97, 432–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wang, J.; Liew, O.W.; Richards, A.M.; Chen, Y.T. Overview of MicroRNAs in Cardiac Hypertrophy, Fibrosis, and Apoptosis. Int. J. Mol. Sci. 2016, 17, 749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Zhu, H.; Fan, G.C. Extracellular/circulating microRNAs and their potential role in cardiovascular disease. Am. J. Cardiovasc. Dis. 2011, 1, 138–149. [Google Scholar] [PubMed]
  106. Huang, W.; Tian, S.S.; Hang, P.Z.; Sun, C.; Guo, J.; Du, Z.M. Combination of microRNA-21 and microRNA-146a Attenuates Cardiac Dysfunction and Apoptosis During Acute Myocardial Infarction in Mice. Mol. Ther. Nucleic Acids 2016, 5, e296. [Google Scholar] [CrossRef]
  107. Fang, Y.H.; Wang, S.P.H.; Gao, Z.H.; Wu, S.N.; Chang, H.Y.; Yang, P.J.; Liu, P.Y.; Liu, Y.W. Efficient Cardiac Differentiation of Human Amniotic Fluid-Derived Stem Cells into Induced Pluripotent Stem Cells and Their Potential Immune Privilege. Int. J. Mol. Sci. 2020, 21, 2359. [Google Scholar] [CrossRef] [Green Version]
  108. Taheri, B.; Soleimani, M.; Fekri Aval, S.; Esmaeili, E.; Bazi, Z.; Zarghami, N. Induced pluripotent stem cell-derived extracellular vesicles: A novel approach for cell-free regenerative medicine. J. Cell. Physiol. 2019, 234, 8455–8464. [Google Scholar] [CrossRef]
  109. Deanfield, J.E.; Halcox, J.P.; Rabelink, T.J. Endothelial function and dysfunction: Testing and clinical relevance. Circulation 2007, 115, 1285–1295. [Google Scholar] [CrossRef]
  110. Sun, X.; Feinberg, M.W. Regulation of endothelial cell metabolism: Just go with the flow. Arter. Thromb. Vasc. Biol. 2015, 35, 13–15. [Google Scholar] [CrossRef] [Green Version]
  111. Jouda, H.; Larrea Murillo, L.; Wang, T. Current Progress in Vascular Engineering and Its Clinical Applications. Cells 2022, 11, 493. [Google Scholar] [CrossRef] [PubMed]
  112. van Rooij, E.; Olson, E.N. MicroRNA therapeutics for cardiovascular disease: Opportunities and obstacles. Nat. Rev. Drug Discov. 2012, 11, 860–872. [Google Scholar] [CrossRef] [PubMed]
  113. de Couto, G.; Gallet, R.; Cambier, L.; Jaghatspanyan, E.; Makkar, N.; Dawkins, J.F.; Berman, B.P.; Marbán, E. Exosomal MicroRNA Transfer Into Macrophages Mediates Cellular Postconditioning. Circulation 2017, 136, 200–214. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, X.; Han, Z.; Yu, Y.; Xu, Z.; Cai, B.; Yuan, Y. Potential Applications of Induced Pluripotent Stem Cells for Cardiovascular Diseases. Curr. Drug Targets 2019, 20, 763–774. [Google Scholar] [CrossRef]
  115. de Godoy, M.A.; Saraiva, L.M.; de Carvalho, L.R.P.; Vasconcelos-Dos-Santos, A.; Beiral, H.J.V.; Ramos, A.B.; Silva, L.R.P.; Leal, R.B.; Monteiro, V.H.S.; Braga, C.V.; et al. Mesenchymal stem cells and cell-derived extracellular vesicles protect hippocampal neurons from oxidative stress and synapse damage induced by amyloid-β oligomers. J. Biol. Chem. 2018, 293, 1957–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Ding, Q.; Sun, R.; Wang, P.; Zhang, H.; Xiang, M.; Meng, D.; Sun, N.; Chen, A.F.; Chen, S. Protective effects of human induced pluripotent stem cell-derived exosomes on high glucose-induced injury in human endothelial cells. Exp. Ther. Med. 2018, 15, 4791–4797. [Google Scholar] [CrossRef] [Green Version]
  117. Gupta, S.; Krishnakumar, V.; Soni, N.; Rao, E.P.; Banerjee, A.; Mohanty, S. Comparative proteomic profiling of Small Extracellular vesicles derived from iPSCs and tissue specific mesenchymal stem cells. Exp. Cell Res. 2022, 420, 113354. [Google Scholar] [CrossRef]
  118. Barreca, M.M.; Cancemi, P.; Geraci, F. Mesenchymal and Induced Pluripotent Stem Cells-Derived Extracellular Vesicles: The New Frontier for Regenerative Medicine? Cells 2020, 9, 1163. [Google Scholar] [CrossRef]
  119. Hunt, S.A. Current status of cardiac transplantation. JAMA 1998, 280, 1692–1698. [Google Scholar] [CrossRef] [Green Version]
  120. Tian, C.; Gao, L.; Zucker, I.H. Regulation of Nrf2 signaling pathway in heart failure: Role of extracellular vesicles and non-coding RNAs. Free. Radic. Biol. Med. 2021, 167, 218–231. [Google Scholar] [CrossRef]
  121. Qiao, L.; Hu, S.; Liu, S.; Zhang, H.; Ma, H.; Huang, K.; Li, Z.; Su, T.; Vandergriff, A.; Tang, J.; et al. microRNA-21-5p dysregulation in exosomes derived from heart failure patients impairs regenerative potential. J. Clin. Investig. 2019, 129, 2237–2250. [Google Scholar] [CrossRef] [PubMed]
  122. Bär, C.; Chatterjee, S.; Falcão Pires, I.; Rodrigues, P.; Sluijter, J.P.G.; Boon, R.A.; Nevado, R.M.; Andrés, V.; Sansonetti, M.; de Windt, L.; et al. Non-coding RNAs: Update on mechanisms and therapeutic targets from the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc. Res. 2020, 116, 1805–1819. [Google Scholar] [CrossRef] [PubMed]
  123. Viereck, J.; Bührke, A.; Foinquinos, A.; Chatterjee, S.; Kleeberger, J.A.; Xiao, K.; Janssen-Peters, H.; Batkai, S.; Ramanujam, D.; Kraft, T.; et al. Targeting muscle-enriched long non-coding RNA H19 reverses pathological cardiac hypertrophy. Eur. Heart J. 2020, 41, 3462–3474. [Google Scholar] [CrossRef] [PubMed]
  124. Hou, J.R.; Wang, Y.H.; Zhong, Y.N.; Che, T.T.; Hu, Y.; Bao, J.; Meng, N. Protective Effect of Flavonoids from a Deep-Sea-Derived Arthrinium sp. against ox-LDL-Induced Oxidative Injury through Activating the AKT/Nrf2/HO-1 Pathway in Vascular Endothelial Cells. Mar. Drugs 2021, 19, 712. [Google Scholar] [CrossRef] [PubMed]
  125. Karnas, E.; Sekuła-Stryjewska, M.; Kmiotek-Wasylewska, K.; Bobis-Wozowicz, S.; Ryszawy, D.; Sarna, M.; Madeja, Z.; Zuba-Surma, E.K. Extracellular vesicles from human iPSCs enhance reconstitution capacity of cord blood-derived hematopoietic stem and progenitor cells. Leukemia 2021, 35, 2964–2977. [Google Scholar] [CrossRef]
  126. Upadhya, R.; Madhu, L.N.; Rao, S.; Shetty, A.K. Proficiency of Extracellular Vesicles From hiPSC-Derived Neural Stem Cells in Modulating Proinflammatory Human Microglia: Role of Pentraxin-3 and miRNA-21-5p. Front. Mol. Neurosci. 2022, 15, 845542. [Google Scholar] [CrossRef]
  127. Chandy, M.; Rhee, J.W.; Ozen, M.O.; Williams, D.R.; Pepic, L.; Liu, C.; Zhang, H.; Malisa, J.; Lau, E.; Demirci, U.; et al. Atlas of Exosomal microRNAs Secreted From Human iPSC-Derived Cardiac Cell Types. Circulation 2020, 142, 1794–1796. [Google Scholar] [CrossRef]
  128. Goversen, B.; van der Heyden, M.A.G.; van Veen, T.A.B.; de Boer, T.P. The immature electrophysiological phenotype of iPSC-CMs still hampers in vitro drug screening: Special focus on I(K1). Pharmacol. Ther. 2018, 183, 127–136. [Google Scholar] [CrossRef]
  129. Ronaldson-Bouchard, K.; Ma, S.P.; Yeager, K.; Chen, T.; Song, L.; Sirabella, D.; Morikawa, K.; Teles, D.; Yazawa, M.; Vunjak-Novakovic, G. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 2018, 556, 239–243. [Google Scholar] [CrossRef]
  130. Karbassi, E.; Fenix, A.; Marchiano, S.; Muraoka, N.; Nakamura, K.; Yang, X.; Murry, C.E. Cardiomyocyte maturation: Advances in knowledge and implications for regenerative medicine. Nat. Rev. Cardiol. 2020, 17, 341–359. [Google Scholar] [CrossRef]
  131. Chow, M.; Boheler, K.R.; Li, R.A. Human pluripotent stem cell-derived cardiomyocytes for heart regeneration, drug discovery and disease modeling: From the genetic, epigenetic, and tissue modeling perspectives. Stem Cell Res. Ther. 2013, 4, 97. [Google Scholar] [CrossRef] [Green Version]
  132. Hollý, D.; Klein, M.; Mazreku, M.; Zamborský, R.; Polák, Š.; Danišovič, Ľ.; Csöbönyeiová, M. Stem Cells and Their Derivatives-Implications for Alveolar Bone Regeneration: A Comprehensive Review. Int. J. Mol. Sci. 2021, 22, 11746. [Google Scholar] [CrossRef] [PubMed]
  133. Yu, L.; Yang, Y.; Zhang, B.; Bai, X.; Fei, Q.; Zhang, L. Rapid human-derived iPSC osteogenesis combined with three-dimensionally printed Ti6Al4V scaffolds for the repair of bone defects. J. Cell. Physiol. 2020, 235, 9763–9772. [Google Scholar] [CrossRef] [PubMed]
  134. Bec, N.; Bonhoure, A.; Henry, L.; Berry, L.; Larroque, C.; Coux, O.; Stoebner, P.E.; Vidal, M. Proteasome 19S RP and translation preinitiation complexes are secreted within exosomes upon serum starvation. Traffic 2019, 20, 516–536. [Google Scholar] [CrossRef] [Green Version]
  135. Bister, N.; Pistono, C.; Huremagic, B.; Jolkkonen, J.; Giugno, R.; Malm, T. Hypoxia and extracellular vesicles: A review on methods, vesicular cargo and functions. J. Extracell. Vesicles 2020, 10, e12002. [Google Scholar] [CrossRef] [PubMed]
  136. Nachlas, A.L.Y.; Li, S.; Jha, R.; Singh, M.; Xu, C.; Davis, M.E. Human iPSC-derived mesenchymal stem cells encapsulated in PEGDA hydrogels mature into valve interstitial-like cells. Acta Biomater. 2018, 71, 235–246. [Google Scholar] [CrossRef]
  137. Noor, N.; Shapira, A.; Edri, R.; Gal, I.; Wertheim, L.; Dvir, T. 3D Printing of Personalized Thick and Perfusable Cardiac Patches and Hearts. Adv. Sci. 2019, 6, 1900344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Tabei, R.; Kawaguchi, S.; Kanazawa, H.; Tohyama, S.; Hirano, A.; Handa, N.; Hishikawa, S.; Teratani, T.; Kunita, S.; Fukuda, J.; et al. Development of a transplant injection device for optimal distribution and retention of human induced pluripotent stem cell–derived cardiomyocytes. J. Heart Lung Transplant. 2019, 38, 203–214. [Google Scholar] [CrossRef]
  139. Qi, X.; Zhang, J.; Yuan, H.; Xu, Z.; Li, Q.; Niu, X.; Hu, B.; Wang, Y.; Li, X. Exosomes Secreted by Human-Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Repair Critical-Sized Bone Defects through Enhanced Angiogenesis and Osteogenesis in Osteoporotic Rats. Int. J. Biol. Sci. 2016, 12, 836–849. [Google Scholar] [CrossRef] [Green Version]
  140. Tian, T.; Wang, Y.; Wang, H.; Zhu, Z.; Xiao, Z. Visualizing of the cellular uptake and intracellular trafficking of exosomes by live-cell microscopy. J. Cell. Biochem. 2010, 111, 488–496. [Google Scholar] [CrossRef]
  141. Feng, D.; Zhao, W.L.; Ye, Y.Y.; Bai, X.C.; Liu, R.Q.; Chang, L.F.; Zhou, Q.; Sui, S.F. Cellular internalization of exosomes occurs through phagocytosis. Traffic 2010, 11, 675–687. [Google Scholar] [CrossRef] [PubMed]
  142. Chen, H.; Xue, R.; Huang, P.; Wu, Y.; Fan, W.; He, X.; Dong, Y.; Liu, C. Modified Exosomes: A Good Transporter for miRNAs within Stem Cells to Treat Ischemic Heart Disease. J. Cardiovasc. Transl. Res. 2022, 15, 514–523. [Google Scholar] [CrossRef] [PubMed]
  143. Holm, F.; Ström, S.; Inzunza, J.; Baker, D.; Strömberg, A.M.; Rozell, B.; Feki, A.; Bergström, R.; Hovatta, O. An effective serum- and xeno-free chemically defined freezing procedure for human embryonic and induced pluripotent stem cells. Hum. Reprod. 2010, 25, 1271–1279. [Google Scholar] [CrossRef] [Green Version]
  144. Ben Jehuda, R.; Shemer, Y.; Binah, O. Genome Editing in Induced Pluripotent Stem Cells using CRISPR/Cas9. Stem Cell Rev. Rep. 2018, 14, 323–336. [Google Scholar] [CrossRef] [PubMed]
  145. Hockemeyer, D.; Jaenisch, R. Induced Pluripotent Stem Cells Meet Genome Editing. Cell Stem Cell 2016, 18, 573–586. [Google Scholar] [CrossRef] [Green Version]
  146. Xu, H.; Wang, B.; Ono, M.; Kagita, A.; Fujii, K.; Sasakawa, N.; Ueda, T.; Gee, P.; Nishikawa, M.; Nomura, M.; et al. Targeted Disruption of HLA Genes via CRISPR-Cas9 Generates iPSCs with Enhanced Immune Compatibility. Cell Stem Cell 2019, 24, 566–578.e567. [Google Scholar] [CrossRef] [Green Version]
  147. Saeedi, S.; Israel, S.; Nagy, C.; Turecki, G. The emerging role of exosomes in mental disorders. Transl. Psychiatry 2019, 9, 122. [Google Scholar] [CrossRef] [Green Version]
  148. Yang, H.; Feng, R.; Fu, Q.; Xu, S.; Hao, X.; Qiu, Y.; Feng, T.; Zeng, Z.; Chen, M.; Zhang, S. Human induced pluripotent stem cell-derived mesenchymal stem cells promote healing via TNF-α-stimulated gene-6 in inflammatory bowel disease models. Cell Death Dis. 2019, 10, 718. [Google Scholar] [CrossRef] [Green Version]
  149. Workman, M.J.; Troisi, E.; Targan, S.R.; Svendsen, C.N.; Barrett, R.J. Modeling Intestinal Epithelial Response to Interferon-γ in Induced Pluripotent Stem Cell-Derived Human Intestinal Organoids. Int. J. Mol. Sci. 2020, 22, 288. [Google Scholar] [CrossRef]
  150. Dai, S.; Mo, Y.; Wang, Y.; Xiang, B.; Liao, Q.; Zhou, M.; Li, X.; Li, Y.; Xiong, W.; Li, G.; et al. Chronic Stress Promotes Cancer Development. Front. Oncol. 2020, 10, 1492. [Google Scholar] [CrossRef]
  151. Eckerling, A.; Ricon-Becker, I.; Sorski, L.; Sandbank, E.; Ben-Eliyahu, S. Stress and cancer: Mechanisms, significance and future directions. Nat. Rev. Cancer 2021, 21, 767–785. [Google Scholar] [CrossRef] [PubMed]
  152. Katoh, M. Therapeutics targeting angiogenesis: Genetics and epigenetics, extracellular miRNAs and signaling networks (Review). Int. J. Mol. Med. 2013, 32, 763–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Cui, X.; Fu, Q.; Wang, X.; Xia, P.; Cui, X.; Bai, X.; Lu, Z. Molecular mechanisms and clinical applications of exosomes in prostate cancer. Biomark. Res. 2022, 10, 56. [Google Scholar] [CrossRef]
  154. Polanco, J.C.; Hand, G.R.; Briner, A.; Li, C.; Götz, J. Exosomes induce endolysosomal permeabilization as a gateway by which exosomal tau seeds escape into the cytosol. Acta Neuropathol. 2021, 141, 235–256. [Google Scholar] [CrossRef]
  155. Sridharan, B.; Lim, H.G. Exosomes and ultrasound: The future of theranostic applications. Mater. Today. Bio 2023, 19, 100556. [Google Scholar] [CrossRef] [PubMed]
  156. Fan, J.; Lee, C.S.; Kim, S.; Chen, C.; Aghaloo, T.; Lee, M. Generation of Small RNA-Modulated Exosome Mimetics for Bone Regeneration. ACS Nano 2020, 14, 11973–11984. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Biogenesis of EVs and schematic of exosomal molecular compositions. (A). EVs contain apoptotic bodies, microvesicles, and exosomes. Apoptotic bodies are formed by membrane folding, invagination, and shedding with organelles and nuclear debris. MVs are formed by the direct outward budding of plasma membranes. As for exosomes, at the very beginning, the invagination of the plasma membrane forms a cup-shaped structure termed early endosome containing cell surface proteins and other biological substances. Early endosomes then develop into late endosomes, which invaginate to form multivesicular bodies (MVBs) that finally fuse with the plasma membrane and release the exosomes. (B). Exosome contains various important biomarkers, such as proteins, lipids, and miRNAs.
Figure 1. Biogenesis of EVs and schematic of exosomal molecular compositions. (A). EVs contain apoptotic bodies, microvesicles, and exosomes. Apoptotic bodies are formed by membrane folding, invagination, and shedding with organelles and nuclear debris. MVs are formed by the direct outward budding of plasma membranes. As for exosomes, at the very beginning, the invagination of the plasma membrane forms a cup-shaped structure termed early endosome containing cell surface proteins and other biological substances. Early endosomes then develop into late endosomes, which invaginate to form multivesicular bodies (MVBs) that finally fuse with the plasma membrane and release the exosomes. (B). Exosome contains various important biomarkers, such as proteins, lipids, and miRNAs.
Ijms 24 04577 g001
Figure 2. Exosome separation diagram. (A). Ultracentrifugation. First, the samples are centrifuged at 300× g, 2000× g, and 10,000× g to remove larger cells, cell debris, and dead cells. Secondly, the exosomes are isolated via ultracentrifugation twice at a speed of more than 100,000× g. (B). Isopycnic density gradient centrifugation. Impurities are firstly removed via low-speed centrifugation, and then, the separated samples are added to the constructed density medium (3%, 35%, 45%, and 90%) for separation. (C). Size-exclusion chromatography.
Figure 2. Exosome separation diagram. (A). Ultracentrifugation. First, the samples are centrifuged at 300× g, 2000× g, and 10,000× g to remove larger cells, cell debris, and dead cells. Secondly, the exosomes are isolated via ultracentrifugation twice at a speed of more than 100,000× g. (B). Isopycnic density gradient centrifugation. Impurities are firstly removed via low-speed centrifugation, and then, the separated samples are added to the constructed density medium (3%, 35%, 45%, and 90%) for separation. (C). Size-exclusion chromatography.
Ijms 24 04577 g002
Figure 3. Biogenesis and information exchange of exosomes. The invagination of the plasma membrane forms a cup-shaped structure, which includes proteins on the cell surface and some components in the extracellular environment, such as proteins, lipids and metabolites, making up the early endosomes. Early endosomes then develop into late endosomes and invaginate to form intraluminal vesicles, and the cytoplasmic components also enter intraluminal vesicles, and then, late endosomes form multivesicular body. Finally, multivesicular body fuses with the plasma membrane and the exosomes are released. The receptor cells mainly interact with the exosomes through three ways: (1) the exosomes bind to the receptors on the cell membrane; (2) the exosomes fuse directly with the cell membrane to release the contents; (3) the exosomes directly enter the cytoplasm in a complete form through cellular pinocytosis or phagocytosis.
Figure 3. Biogenesis and information exchange of exosomes. The invagination of the plasma membrane forms a cup-shaped structure, which includes proteins on the cell surface and some components in the extracellular environment, such as proteins, lipids and metabolites, making up the early endosomes. Early endosomes then develop into late endosomes and invaginate to form intraluminal vesicles, and the cytoplasmic components also enter intraluminal vesicles, and then, late endosomes form multivesicular body. Finally, multivesicular body fuses with the plasma membrane and the exosomes are released. The receptor cells mainly interact with the exosomes through three ways: (1) the exosomes bind to the receptors on the cell membrane; (2) the exosomes fuse directly with the cell membrane to release the contents; (3) the exosomes directly enter the cytoplasm in a complete form through cellular pinocytosis or phagocytosis.
Ijms 24 04577 g003
Table 1. The drug delivery of iPSC-sEVs in various disease models.
Table 1. The drug delivery of iPSC-sEVs in various disease models.
Cell SourcesCharacterizationModelsTherapeutic EffectsCargosReference
miPSCsTEMMIMitigate cardiac remodeling and
improve cardiac functions
post myocardial infarction
[81]
miPSCsWB (CD63, Tsg101)MIRIPrevent cardiomyocyte apoptosis
in ischemic myocardium
miR21,
HIF-1α-regulated miR210
[82]
miPSCsEM, FCM,
RT-PCR
MIRIImprove LV function and
enhance angiogenesis
global miRNA and proteomic profiling performed[21]
hiPSCsFCM, BCAMIReduce fibrosis in
infarcted mice hearts
CD82[83]
hiPSCsTEM, NTAMIFacilitate cardiac repair through
circulating miRNAs
circulating
miRNAs
[84]
hiPSCsNTA, WB (CD63)HFInvolved in the remodeling process
and observed in primary cardiomyocytes
miRNA
mRNA
[85]
hiPSCsTEM,
WB (CD63, CD9)
Endothelial cell
in vitro
Improve cardiac function and repairmiRNA[86]
hiPSCsTEM, NTAH9c2
in vitro
Protect against oxidative-stress-induced apoptosismiRNA[87]
hiCMsWB (CD63, CD81)Dys-iCMs
In vitro
Decrease reactive oxygen species
and delay mitochondrial permeability
[88]
hiCMsTEM,
WB (CD63, CD81)
MIFacilitate cardiac repair and avoid immune rejectionmiRNA,
LncRNA
[89]
hiCMsWB (CD81, CD63,
flotillin-1, TSTG101)
MIImprove recovery from
myocardial infarction in swine
[12]
hiMSCsEM, NanoFCMMIPromote cell viability through
activating the Akt/Nrf2/HO-1 axis and improve
cardiac function
[90]
hiMSCsTEM, immunoblotRat skin
wound model
Promote collagen synthesis and angiogenesis [91]
hiMSCsEM, NTAHFImprove cardiac function and increased
EF relative to baseline values
miRNA[92]
hiMSCsTEM, NTA,
RT-PCR
Ischemic Adult
Human Cardiomyocytes
Alter cardiac tissue-level remodelingmiR21-5p[93]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Meng, W.-T.; Guo, H.-D. Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. Int. J. Mol. Sci. 2023, 24, 4577. https://doi.org/10.3390/ijms24054577

AMA Style

Meng W-T, Guo H-D. Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. International Journal of Molecular Sciences. 2023; 24(5):4577. https://doi.org/10.3390/ijms24054577

Chicago/Turabian Style

Meng, Wan-Ting, and Hai-Dong Guo. 2023. "Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury" International Journal of Molecular Sciences 24, no. 5: 4577. https://doi.org/10.3390/ijms24054577

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop