Open access peer-reviewed chapter

Mesenchymal Stem Cell-Derived Exosomes for Myocardial Infarction Treatment

Written By

Huifeng Zheng, Yimei Hong, Bei Hu, Xin Li and Yuelin Zhang

Submitted: 24 February 2023 Reviewed: 01 March 2023 Published: 26 March 2023

DOI: 10.5772/intechopen.110736

From the Edited Volume

Exosomes - Recent Advances From Bench to Bedside

Edited by Sherin Saheera

Chapter metrics overview

121 Chapter Downloads

View Full Metrics

Abstract

Myocardial infarction (MI) is a major cause of morbidity and mortality in modern society. Over the past decades, mesenchymal stem cell (MSCs)-based therapy has shown promising results in the treatment of MI due to their unique properties of multi-differentiation ability, immune-privileged phenotype and paracrine activity. Recently, MSC-derived exosomes (MSC-EXO) have been proposed as a promising therapeutic strategy for MI with their ability to inhibit cardiomyocyte apoptosis and stimulate vascular angiogenesis. They also aid immunoregulation and rejuvenation of cardiomyocyte senescence by transporting their unique content such as proteins, lipids, and miRNAs. Compared with MSC transplantation, MSC-EXO administration has shown several advantages, including lower toxicity and immunogenicity and no risk of tumor formation. Nonetheless the potential mechanisms underlying MSC-EXO-based therapy for MI are not fully understood. In addition, lack of modification of MSC-EXOs can impact therapeutic efficacy. It is vital to optimize MSC-EXO and enhance their therapeutic efficacy for MI. We summarize the recent advances regarding biological characteristics, therapeutic potential and mechanisms, and optimal approaches to the use of MSC-EXOs in the treatment of MI.

Keywords

  • mesenchymal stem cells
  • exosome
  • myocardial infarction
  • treatment
  • therapeutic effect

1. Introduction

Myocardial infarction (MI) results in irreversible loss of cardiomyocytes due to a restricted blood supply and is the major cause of morbidity and mortality worldwide. It has been estimated to account for 80% of deaths in patients with ischemic heart disease worldwide, and its prevalence continues to increase every year leading to a significant medical, social, and financial burden [1]. Despite the availability of advanced surgical interventions and medications including primary percutaneous coronary intervention, angiotensin-converting enzyme drugs and β-blockers, there remains no effective means to prevent cardiomyocyte loss due to myocardial ischemia [2]. The only cure for this devastating disease is heart transplantation but this is restricted by its high cost, a shortage of donor hearts, and the occurrence of immune rejection following transplantation [3]. Exploration of novel therapies for left ventricle remodeling and dysfunction following infarction is urgently needed.

Over the past decades, stem cell-based therapy has become a promising strategy to treat MI with significant progress made in animal studies and clinical trials [4, 5, 6]. Among all types of stem cell under investigation, mesenchymal stem cells (MSCs) have garnered huge interest due to their easy isolation, high reproductive activity, differentiation capability and immunomodulatory properties [7, 8]. MSCs can be isolated from multiple tissues or cells including bone marrow, adipose tissue, umbilical cord blood and even pluripotent stem cells [9, 10, 11, 12]. There is accumulating evidence that MSCs are promising candidates for MI treatment [13, 14, 15]. More importantly, it is now widely accepted that the cardioprotective effects of MSC-based therapy in MI are due to their strong paracrine effects, rather than trans-differentiation ability [7, 16, 17, 18]. Therefore, researchers are increasingly huge interested in the therapeutic efficacy of MSC-derived bioactive molecules, especially exosome (EXO), that are considered major components of the paracrine effect in MSC-based therapy [19, 20]. EXO, a subgroup of extracellular vesicles (EVs), are 40–160 nm diameter membrane-bound vesicles that can be found in almost all biological fluids. It has been well documented that MSC-EXO exert their cardioprotective effects in MI by delivering diverse biological molecules, including non-coding RNA, DNA, lipids and proteins [21, 22, 23, 24]. More importantly, compared with MSC transplantation, MSC-EXO have several advantages such as easier storage and transplantation, less immune rejection, minimum risk of immunogenicity and no risk of tumor formation [25]. We discuss the current understanding of the biological characteristics, therapeutic effects and potential mechanisms of MSC-based therapy in MI. We also highlight the current challenges and potential approaches to improve the efficacy and production of MSC-EXO in regenerative medicine to guide their future clinical application.

Advertisement

2. Characterization and Isolation of MSC-EXO

EVs are bilayer lipid membrane-bound subcellular vesicles released by all types of cells and present in all body fluids. According to MISEV2018, EVs are divided into “small EVs” (sEVs, <100 nm or <200 nm) and “medium/large EVs”(m/lEVs, >200 nm) respectively [26]. EXO are sEVs approximately 40–160 nm in diameter (100 nm on average) and the main subclass of EVs [27]. The biogenesis of EXO begins with inward budding to form an early endosome. Finally, EXO are built when multi vesicular bodies (MVBs, late endosomes) fuse with plasma membrane and are secreted into the extracellular space [28, 29]. MSC-EXO express EXO-specific markers CD9, CD63, CD81, Alix and Tsg101 as well as MSC surface markers including CD29, CD44, CD90 and CD73. Among these, CD29 and CD44 have been identified previously as the specific biomarkers for MSC-EXO [30, 31]. The size and concentration of EXO can be characterized by nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM) [32]. Recently, plasmonic scattering microscopy has been applied to image exosomes and analyze biomarkers [33].

It is difficult to show whole landscape of EXO dispersed in solution. Therefore, purification of EXO is of importance for EXO definition. EXO are distributed throughout body fluids and this represents a challenge to their isolation. EXO are secreted into body fluids such as blood, urine, saliva, lymph, breast milk, cerebrospinal fluid and pericardial fluid etc. [34]. EXO components reflects the state of the original cell. Different methods of isolation of EXO varies from various body fluids. Meanwhile, the extraction result differs from types of biological fluid. Which was optimal remains controversial [35]. Isolation of abundant EXO can help in the assessment of their biological functions [36]. Several recent alternative methods ranging from conventional to newly developed techniques to isolate and purify EXO are summarized in Table 1. Different methods for EXO isolation have different advantages and disadvantages. During isolation, ultracentrifugation and density gradient centrifugation are the most commonly used techniques [47]. Currently, several new methods have been established to facilitate high-throughput and high-purity manufacture of EXO. The characterization and isolation of MSC-EXO are summarized in Figure 1.

No.Methods of EXO isolationAdvantages and disadvantagesRef.
1Ultracentrifugation
  1. Most widely used

  2. Gold standard for exosome separation

  3. A series of speed centrifugation

  4. Time consuming

[37]
2Density gradient centrifugation
  1. Sorts:

    Sucrose density gradient

    Iodixanol density gradient

    Optiprep density gradient

  2. Improve purity of exosomes

  3. Sucrose density gradient cannot effectively separate EXO and retroviruses

  4. Time consuming and complex procedure

[38]
3Chromatography (size-based isolation techniques)
  1. Sorts:

    Mini-size exclusion chromatography (mini-SEC)

    Size exclusion chromatography

  2. Quick, easy, small material consumption

  3. May be applied with other particles of similar size

[39]
4Ultrafiltration (size-based isolation techniques)
  1. Uses ultrafiltration membranes with different molecular weight cutoffs (MWCO)

  2. Low cost and high enrichment efficiency

  3. Low purity and non-specific binding of EXO

[40]
5Tangential flow filtration (size-based isolation techniques)
  1. Using a cutoff TFF cartridge

  2. Fluid flows tangentially across the surface, avoiding filter cake formation

  3. Fast and efficient

  4. Volume is limited by the instrument dead volume

[41]
6Polymer-based precipitation separation
  1. Uses polyethylene glycol (PEG) as a medium

  2. Easy to operate with short analysis time

  3. Polymer is difficult to remove

[42]
7Immunoaffinity chromatography (IAC)
  1. Based on the specific binding of antibodies and ligands

  2. Storage conditions of EXO are relatively harsh and are not suitable for large-scale separation of EXO

[43]
8Microfluidic technologies
  1. Sorts

    Physical-property-based microfluidics

    Immunoaffinity-based microfluidics

  2. Miniaturization, integration, high-throughput capacity, low-time consumption

  3. Specialized equipment needed

[44]
9Deterministic lateral displacement separation
  1. Uses tilted pillar arrays that generate a fluid bifurcation and a unique number of streamlines between the gaps

  2. Low separation throughput; particle adhesion and clogging; complex and bulky experimental setup

[45]
10Acoustic fluid separation
  1. Uses ultrasound waves to exert radiation forces on particles

  2. Highly controllable, and versatile

  3. The device is relatively low in a single channel microfluidic device

[46]

Table 1.

Methods of MSC-EXO isolation.

Figure 1.

Characterization and isolation of MSC-EXO.

Advertisement

3. The bioactive constituents of MSC-EXO for MI treatment

MSC-EXO exert their benefits in various diseases by enclosing and transporting a vast array of molecules [48]. It has been demonstrated that exosomal components are almost dependent on the source cell and cellular conditions [25, 49, 50]. Generally, EXO contain multiple characteristic molecules with typical physiological functions [51, 52, 53]. MSC-EXO comprise a variety of substances, including many kinds of proteins and a lot of noncoding RNA, including microRNAs (miRNAs) and long noncoding RNAs (lncRNA) [54]. These components can act as paracrine factors, mediating cell-to-cell signaling and communication. More importantly, they can be used as prognostic and diagnostic markers [55, 56].

3.1 Exosomal miRNAs in MSC-EXO for MI treatment

miRNAs are endogenous and 19–25 nucleotides in size. They can be isolated from cells, tissues and body fluids [57]. By pairing to the mRNAs of protein-coding genes, miRNAs play an important role in regulating post-transcriptional silencing of target genes [58, 59]. There is accumulating evidence that miRNAs are enriched in MSC-EXO and are the major bioactive constituents [60, 61, 62]. In the last few decades, the cardioprotective role of MSC-derived exosomal miRNAs has attracted huge attention [63]. It has been well documented that many MSC-derived exosomal miRNAs have beneficial functions in MI treatment [64]. Importantly, several potential mechanisms have been identified such as promotion of angiogenesis, reduction of cell death and an antifibrotic effect [65]. Enhanced angiogenesis is one of the important repair mechanisms underlying MSC-EXO-based therapy for MI [66, 67, 68]. Through direct miRNAs transfer, MSC-EXO convey their proangiogenic signals to injured cardiomyocytes [69]. Previous study has shown that silenced MSC-derived exosomal miR-210 largely lost its proangiogenic effect. Further experimental study revealed that exosomal miR-210 improves angiogenesis of MSC-EXO via targeting of Efna3 [70]. Zhu et al. demonstrated that macrophage migration inhibitory factor (MIF) could enhance the pro-angiogenic effect of MSC-EXO by enhancing the level of miR-133a-3p via regulation of the AKT signaling pathway [71]. miR-221 is one of the most studied miRNAs. A recent study reported that up-regulated exosomal miR-221-3p derived from senescent MSCs improved their ability of angiogenesis, migration and proliferation, and suppressed apoptosis by regulating the PTEN/AKT pathway [72]. Ma et al. revealed that miR-132-electroporated MSC-EXO could promote angiogenesis both in vitro and in vivo by downregulating RASA1 [23]. These studies show that MSC-EXO improve angiogenesis by transmitting miRNAs via various biological signaling pathway following MI.

There is increasing evidence that ameliorating cardiomyocyte death is another major mechanism by which EXO restore cardiac function following MI. MSC-EXO reduce myocardial cell death via multiple mechanisms including an anti-apoptosis action, inhibition of pyroptosis and an anti-inflammatory effect [73]. Apoptosis is programmed cell death that is strongly associated with myocardial ischemia [74]. Previous studies have proved that MSCs have an anti-apoptotic effect through secretion of exosomes enriched in miRNAs [75]. Hypoxia-elicited MSC-EXO (Hypo-EXO) facilitates cardiac repair by preventing cell death in MI via delivery of miR-125b. Mechanistically, miR-125b-5p suppresses apoptosis of cardiomyocytes by downregulating the expression of apoptotic genes p53 and BAK1 [63]. Another study demonstrated that EXO derived from miR-146a-modified adipose-MSCs attenuated MI via inhibition of apoptosis, the inflammatory response, and fibrosis in a rat model of AMI by targeting early growth response factor 1(EGR1) [76]. Wang et al. reported that adipose-MSC-EXO carrying miR-671 reduced the apoptosis of cardiomyocytes and alleviated myocardial fibrosis and inflammation via inactivation of the TGFBR2/Smad2 Axis [77]. miR-153-3p plays an important role in modulating cell proliferation, apoptosis and angiogenesis. It has been illustrated that EXO-miR-153-3p significantly reduces apoptosis of endothelial cells and cardiomyocytes and promotes their viability. By targeting ANGPT1, miR-153-3p can regulate the VEGF/VEGFR2/PI3K/AKT/eNOS pathways to prevent hypoxic damage to endothelial cells and cardiomyocytes [78]. Furthermore, a growing number of studies have shown that stem cell-derived exosomal miRNAs, such as miR-150-5p, miR-126, and miR-486-5p, demonstrate antiapoptotic activity in MI treatment [79, 80, 81]. These findings indicate that the anti-apoptotic effect of MSC-EXO can be partly ascribed to the delivery of some anti-apoptotic miRNAs.

Autophagy is a self-destructive process during which a cell degrades and recycles unnecessary or dysfunctional cellular components [82]. Autophagy is involved in promoting cell death and exacerbates myocardial dysfunction following severe ischemic stress. There is accumulating evidence that MSC-EXO reduce cell death by mediating autophagy. Xiao et al. determined that MSC-EXO reduced autophagic flux in infarcted hearts via exosomal transfer of miR-125b by interfering with p53/Bnip3 signaling and protected cardiomyocytes against damage [83]. Liu et al. showed that miR-93-5p-enhanced ADSC-EXO had a greater cardioprotective effect by suppressing hypoxia-induced autophagy and inflammatory cytokine expression via targeting of Atg7 and Toll-like receptor 4 (TLR4), respectively [84]. Furthermore, Li et al. reported that exosomal miR-301 derived from MSCs protected against MI by inhibiting myocardial autophagy [85]. In addition, MSC-exosomal miRNAs exerted a cardioprotective effect in MI by attenuating cardiac fibrosis. Inflammation and subsequent fibrosis are important pathological reactions that result in scar formation post-MI. Human umbilical cord MSCs-EXO containing miR-29b have been shown to prevent cardiac fibrosis following MI, leading to a reduction in infarct size and improved cardiac function in a mouse model of MI [86]. Moreover, miR-671 carried by adipose-derived MSC-EXO has been proven to also reduce myocardial fibrosis and inflammation both in vitro and in vivo [77]. The roles of MSC-exosomal miRNA and the potential mechanism for MI treatment are summarized in Table 2.

ModelSources of EXORelated-effectorsBiological effectsInvolved pathwayRef.
MI mouse with LAD ligationBM-MSCsmiR-210AngiogenesisEfna3[65]
MI rat with LAD ligationUC-MSCsmiR-133-3pAngiogenesis
Anti-apoptosis
Anti-fibrosis
P-AKT[66]
MI rat with LAD ligationBM-MSCsmiR-221-3pAngiogenesis
Anti-apoptosis
PTEN/AKT pathway[67]
MI mouse with LAD
ligation
BM-MSCsmiR-132Angiogenesis increase tube formation enhance neovascularizationRASA1[18]
MI mouse with LAD ligationBM-MSCsmiR-125bAnti-apoptosisP53 and BAK1[58]
MI rat with LAD ligationAD-MSCsmiR-146Anti-apoptosis
Anti-inflammation
Anti-fibrosis
EGR1/TLR4/NFκB[71]
MI rat with LAD ligationAD-MSCsmiR-671Anti-fibrosis
Anti-inflammation
TGFBR2/Smad2[72]
Vitro modelBM-MSCsmiR-153-3pAnti-apoptosis
Angiogenesis
ANGPT1-VEGF/PI3k/AKT/eNOS[73]
MI mouse with LAD ligationBM-MSCsmiR-150-5pAnti-apoptosisBax[74]
MI rat with LAD ligationAD-MSCsmiR-126Anti-apoptosis
Anti-inflammation
Anti-fibrosis
Angiogenesis
_[75]
MI rat with LAD ligationBM-MSCsmiR-486-5pAnti-apoptosisPTEN/PI3K/AKT[76]
MI mouse with LAD ligationBM-MSCsmiR-125bDecreasing autophagic fluxp53/Bnip3[78]
MI rat with LAD ligationBM-MSCsmiR-301Inhibiting myocardial autophagy[80]
MI mouse with LAD ligationUC-MSCsmiR-29bAnti-fibrosis[81]

Table 2.

MSC-exosomal miRNAs for MI treatment.

3.2 Exosomal lncRNAs in MSC-EXO for MI treatment

LncRNAs are defined as RNA transcripts >200 nucleotides without protein-coding potential. lncRNAs play important roles in regulating a variety of biological processes. Recent studies have shown that they participate in the initiation and progression of MI through regulation of gene expression at the epigenetic, transcriptional and post-transcriptional levels [87]. Moreover, MSC-derived exosomal lncRNAs have been shown to have cardioprotective effects for MI. LncRNA KLF3-AS1 in human MSC-EXO ameliorated pyroptosis of cardiomyocytes in a rat model of MI via regulation of the miR-138-5p/Sirt1 axis [88]. A recent study has illustrated that hypoxia promoted MSCs to secret lncRNA-UCA1-enriched EXO that had a cardioprotective effect via the lncRNA-UCA1/miR-873-5p/XIAP axis. Furthermore, exosomal lncRNA-UCA1 in human plasma may be considered a potential noninvasive biomarker for the diagnosis of AMI [89]. Similarly, Huang et al. showed that Atorvastatin pretreatment enhanced the therapeutic efficacy of MSC-EXO in a rat MI model via up-regulation of LncRNA H19 by promoting endothelial cell function [90]. The roles of MSC-exosomal LncRNA and their potential mechanism in MI treatment are summarized in Table 3.

ModelSources of EXORelated-effectorsBiological effectsInvolved pathwayRef.
MI rats with LAD ligationhMSCsLncRNA KLF3-AS1Amelioration of pyroptosismiR-138-5p/Sirt1[83]
MI rats with LAD ligationhMSCsLncRNA-UCA1Anti-apoptosismiR-873-5p/XIAP[84]
MI rats with LAD ligationBM-MSCsLncRNA H19Anti-apoptosis
Angiogenesis
Anti-inflammation
Anti-fibrosis
miR-675, VEGF and ICAM-1[85]

Table 3.

MSC-exosomal LncRNAs for MI treatment.

3.3 Exosomal proteins in MSC-EXO for MI treatment

MSC-EXOs further elicit benefit by delivering their cargo of potentially therapeutic proteins to recipient cells [91]. To date, nearly two thousand proteins in MSC-EXO have been identified [92, 93, 94, 95, 96]. Like miRNAs and lncRNAs, proteins in MSC-EXO have the potential to protect cardiomyocytes against injury following MI. Proteins in MSC-EXO whose role is basic cellular function, include common proteins, enzymes and signaling molecules [97]. One study suggested that hucMSC-EXO protected myocardial cells against apoptosis and promoted cell proliferation and angiogenesis by improving the expression of Bcl-2 family [98]. EXO secreted from CXCR4 overexpressing MSCs have been shown to promote cardiomyocyte survival and angiogenesis in ischemic hearts following MI via the AKT signaling pathway [99]. Deng et al. reported that EXO from AD-MSCs could ameliorate cardiac damage following MI by activating S1P/SK1/S1PR1 signaling and promoting macrophage M2 polarization [100]. The roles of MSC-exosomal proteins and their potential mechanism for MI treatment are summarized in Table 4.

ModelSources of EXORelated-effectorsBiological effectsInvolved pathwayRef.
MI rats with LAD ligationUC-MSCsBcl-2 family, Ki67Anti-fibrosis angiogenesis[98]
MI rat with LAD ligationBM-MSCsDMBT1Promotes angiogenesisPI3K-AKT/GSK3β/β-catenin/VEGF[99]
MI rat with LAD ligationAD-MSCsS1P, SK1, S1PR1Anti-apoptosis anti-fibrosis anti-inflammation promotes macrophage M2 polarizationS1P/SK1/S1PR1[100]

Table 4.

MSC-Exosomal proteins for MI treatment.

Taken together, although current knowledge is limited, it can be inferred that various proteins carried by MSC-EXO protect ischemic cardiomyocytes through different mechanisms.

Advertisement

4. Potential strategies to improve the therapeutic efficacy of MSC-EXO for MI

Although MSC-EXO-based therapy has shown promising results in MI, their therapeutic efficacy is heavily restricted by the low production and concentration of biological molecules released by EXO derived from MSCs under routine culture conditions. The production and biological components of MSC-EXO vary depending on the different external stimuli surrounding MSCs and MSC status. Therefore, modifying and optimizing exosomal content in MSC-EXO in vitro prior to transplantation to enhance their therapeutic efficacy for MI is vital. Over the past decades, several novel strategies, including altering culture conditions and pretreatment with pharmacological compounds and molecules, have been explored to generate modified MSC-EXO with greater benefits for MI treatment [56, 101]. More importantly, genetic modification of MSCs has had a great impact on the release of MSC-EXO, directly modulating their therapeutic efficacy. The influence of these factors on production and function of MSC-EXO will be discussed in the following sections. Different strategies to improve the therapeutic effects of MSC-EXO in MI are summarized in Figure 2.

Figure 2.

Different strategies to improve the therapeutic effects of MSC-EXO in MI.

4.1 MSC-EXO generated from different culture conditions

The status of MSCs is largely dependent on culture conditions. Changes to culture conditions may influence MSC-EXO content and its biological functions. As a key impact on MSC culture, oxygen concentration plays a critical role in the regulation of gene expression, exon splicing, and phenotype of MSCs [102]. Therefore, oxygen gradients control MSC functions and generate different biological functions of MSC-EXO. MSCs survive under hypoxic conditions after transplantation into the ischemic heart and then release EXO to exert their benefit. Nonetheless MSCs are usually cultured under normoxic conditions in vitro. Therefore, the EXO released from MSCs under normoxic conditions in vitro and under hypoxic conditions in vivo carry different biological molecules with correspondingly different therapeutic effects. It has been reported that transplantation of MSCs under hypoxic conditions results in an enhanced therapeutic effect for MI [103, 104], indicating that hypoxic preconditioning may be a potential approach to prime MSC-EXO for MI treatment. Accumulating evidence shows that EXO from hypoxia-primed MSCs used to treat MI are superior to EXO from MSCs cultured under normoxic conditions [63, 105, 106]. Hypoxic pre-conditioning can enrich some specific miRNAs in the MSC-EXO that protect against MI by promoting angiogenic potential, attenuating inflammation and ameliorating apoptosis of cardiomyocytes [101]. It has been documented that hypoxic pre-conditioning of MSC-EXO elicits better therapeutic efficacy for MI by reducing the apoptosis of cardiomyocytes via upregulation of miR-210 that targets AIFM3 protein [75]. Zhang et al. showed that EXO isolated from hypoxic MSCs improved myocardial function in a rat model of myocardial ischemia-reperfusion injury by suppressing oxidative stress and the inflammatory response via delivery of miR-98-5p [107]. More importantly, EXO derived from MSCs stably overexpressing hypoxia inducible factor (HIF)-1α displayed an increased angiogenic capacity, partially due to the high level of Jagged1. This may have potential applications for MI treatment [108]. Indeed, transplantation of EXO collected from HIF-1α overexpressing MSCs improved heart function by promoting angiogenic formation in a rat model of MI [109]. Apart from hypoxic conditions, culture medium with different types of serum influence the characteristics of MSCs, modulating the efficacy of MSC-EXO-based therapies. Compared with normal serum, MSCs cultured with serum collected from the blood of mice with middle cerebral artery occlusion robustly demonstrated an upregulated level of miR-20a in their EXO [110]. Whether culturing MSCs with special serum can improve the efficacy of MSC-EXO for MI remains to be determined. Recently, it has been reported that the production of MSC-EXO can be augmented using a 3D porous scaffold structure instead of the traditional 2D culture in plastic plates, providing a novel strategy to optimize MSC-EXO for MI treatment [111]. Therefore, exploring suitable culture conditions for MSCs will not only improve the yield of EXO but also modify the therapeutic components of the EXO, ultimately enhancing their efficacy for MI treatment.

4.2 MSC-EXO generated following preconditioning with pharmacological compounds and biomolecules of MSCs

There is accumulating evidence that preconditioning with pharmacological agents and biomolecules robustly improves the therapeutic efficacy of MSCs in MI by enhancing MSC survival and paracrine effects [112, 113, 114, 115]. These results prompted us to determine whether pharmacological preconditioning could be a novel approach to enhance the cardioprotective effects of MSC-EXO. Our group has shown that compared with MSC-EXO, EXO isolated from MSCs pretreated with hemin, a potent heme oxygenase-1 (HO-1) inducer, exhibited better cardioprotection for MI via inhibition of cardiomyocyte senescence by elevating the level of miR-183-5p [116]. Huang et al. demonstrated that EXO obtained from atorvastatin-pretreated MSCs had greatly enhanced therapeutic efficacy for MI treatment in terms of promoting angiogenesis and inhibiting inflammation [90]. In addition to pharmacological agents, preconditioning with specific biomolecules can contribute to the secretion of MSC-EXO. EXO derived from interferon-gamma (IFN-γ)-treated MSCs exhibited more potent cardioprotective function in a rat model of MI by increasing angiogenesis and inhibiting cardiomyocyte apoptosis through upregulation of miR-21 [117]. Interestingly, Xiao et al. found that compared with MSC-EXO, EXO derived from MSCs pretreated with ischemic rat heart extracts enriched with IL-22 promoted the angiogenic capacity of human umbilical vein endothelial cells, indicating a novel preconditioning approach to optimize MSC-EXO for MI treatment [99]. These reports confirm that preconditioning with pharmacological compounds or biomolecules can alter the surrounding microenvironment of the culture conditions of MSCs and influence their paracrine effects, ultimately affecting the action of their derived EXO.

4.3 MSC-EXO isolated from genetically modified MSCs

Genetic modification of MSCs via knockdown or overexpression of some RNAs or proteins is another efficient approach to improve the therapeutic effect of MSC-EXO. Our previous study showed that compared with MSC-EXO, administration of EXO secreted by MSCs transduced with macrophage migration inhibitory factor, a proinflammatory cytokine, exhibited a better therapeutic efficacy for MI by downregulating cardiomyocyte mitochondrial fragmentation, reactive oxygen species generation, and apoptosis [118]. A recent report revealed that EXO collected from stromal-derived factor 1-overexpressing MSCs intravenously administered in a mouse model displayed enhanced heart protection by inhibiting apoptosis and autophagy of myocardial cells and increasing angiogenesis by the regulating PI3K signaling pathway [119]. In another study, EXO from MSCs transduced with lentiviral CXCR4 promoted restoration of cardiac function in a rat model of MI by ameliorating cardiomyocyte apoptosis and increasing angiogenesis via upregulation of IGF-1α and p-AKT levels and downregulation of active caspase 3 level [120]. As discussed above, miRNAs are important biological components that play a pivotal role in the cardioprotective effect of MSC-EXO in MI [121, 122, 123]. Therefore, overexpression of miRNAs in MSCs can enhance the efficacy of MSC-EXO for MI treatment. Direct injection of MSC-EXO with miR-183-5p overexpression has been shown to result in better cardiac function via suppression of apoptosis and oxidative stress of cardiomyocytes by targeting FOXO1 [124]. Administration of EXO derived from miR-129-5p-modified MSCs displayed enhanced cardiac function following MI in mice by downregulating apoptosis of cardiomyocytes and production of inflammatory cytokines via targeting of HMGB1 [125]. Moreover, EXO derived from miR-126-overexpressing adipose-MSCs demonstrated better beneficial effects by inhibiting cardiac fibrosis and inflammatory cytokine expression and increasing angiogenesis [80]. Thus, genetically modified MSC-EXO have been considered an effective means by which to enhance their cardioprotective effects in MI.

Advertisement

5. Limitations and challenges of MSC-EXO-based therapy for MI

Despite several significant advantages over MSCs, there remain some limitations and challenges to the clinical application of MSC-EXO for MI treatment. First, the rapid clearance of MSC-EXO from ischemic heart tissue after transplantation limits the beneficial effects for MI. An optimum delivery route for administration of MSC-EXO is unavailable. Currently, intramyocardial transplantation is the most efficacious. Exploration of alternative approaches to optimize retention and engraftment of MSC-EXO in the ischemic heart is urgently needed. Second, although the biological components in MSC-EXO, including miRNAs, lncRNA, recombinant proteins, and cytokines, have been intensively investigated, the exact mechanisms underlying MSC-EXO-based therapy for MI require further investigation. Third, MSC-EXO are currently isolated mainly depending on their vesicle size. Different sizes of MSC-EXO may contain different components with corresponding different therapeutic outcomes for MI. A more accurate isolation and purification method for MSC-EXO should be adopted. Fourth, multiple harmful and unwanted biological components in MSC-EXO may restrict their efficiency. Several strategies to modify and remove unwanted components are under investigation. Finally, although classic high-speed centrifugation is the most common method used for MSC-EXO isolation, it is limited by the disadvantages of low production of EXO, high heterogeneity and non-scalability. A scalable isolation protocol for mass production of homogenous MSC-EXO for clinical application is needed.

Advertisement

6. Conclusion

Over the past decades, administration of MSC-EXO has been shown to attenuate cardiac remodeling and improve heart function recovery following MI by inhibiting cardiomyocyte apoptosis, stimulating vascular angiogenesis, immunoregulation and rejuvenating cardiomyocyte senescence. Although the great potential of MSC-EXO therapy for heart function recovery has been clearly demonstrated, the therapeutic role of MSC-EXO in MI is extremely complex. Many issues remain to be carefully addressed and evaluated including the need for a high quality isolation protocol, delivery routes, and optimum EXO dose. In addition, potential risks must be carefully evaluated prior to translation into clinical trials. MSC-EXO-based therapy is still in its infancy and most experimental studies have been in a small animal model. The therapeutic efficacy of MSC-EXO should be evaluated in a porcine model or pre-clinical large animal model. This may provide further evidence to support clinical translation of MSC-EXO-based therapy to humans. Despite the unresolved issues, with the advanced development and technical breakthroughs in EXO research, it is hoped that clinical translation of MSC-EXO to promote cardiac regeneration and repair will soon be a reality for patients with MI.

Advertisement

Acknowledgments

This research was in part supported by the Natural Science Foundation for Distinguished Scholarship of Guangdong Province of China (2022B1515020104 to Y. Zhang), the Distinguished Scholarship of Guangdong Provincial People’s Hospital (KY0120220132 to Y. Zhang), National Natural Science Grant of China (No. 82270253 to Y. Zhang, No. 82072225 to X. Li), Natural Science Foundation of Chongqing (No. cstc2020jycj-msxmX0301 to H. Zheng) and NSFC Incubation Program of GDPH (KY012021167 to Y. Hong).

Advertisement

Conflict of interest

The authors confirm that they have no conflicts of interest.

Advertisement

Other declarations

The authors thank Ms. S Aglionby for editing the manuscript.

References

  1. 1. GBD 2016 Mortality Collaborators. Global, regional, and national under-5 mortality, adult mortality, age-specific mortality, and life expectancy, 1970-2016: A systematic analysis for the global burden of disease study 2016. Lancet. 2017;390:1084-1150. DOI: 10.1016/s0140-6736(17)31833-0
  2. 2. Heusch G, Gersh BJ. The pathophysiology of acute myocardial infarction and strategies of protection beyond reperfusion: A continual challenge. European Heart Journal. 2017;38:774-784. DOI: 10.1093/eurheartj/ehw224
  3. 3. Kittleson MM, Kobashigawa JA. Cardiac transplantation: Current outcomes and contemporary controversies. JACC Heart Failure. 2017;5:857-868. DOI: 10.1016/j.jchf.2017.08.021
  4. 4. Feric NT, Radisic M. Strategies and challenges to myocardial replacement therapy. Stem Cells Translational Medicine. 2016;5:410-416. DOI: 10.5966/sctm.2015-0288
  5. 5. Raziyeva K, Smagulova A, Kim Y, Smagul S, Nurkesh A, Saparov A. Preconditioned and genetically modified stem cells for myocardial infarction treatment. International Journal of Molecular Sciences. 2020;21:7301. DOI: 10.3390/ijms21197301
  6. 6. Parizadeh SM, Jafarzadeh-Esfehani R, Ghandehari M, Parizadeh MR, Ferns GA, Avan A, et al. Stem cell therapy: A novel approach for myocardial infarction. Journal of Cellular Physiology. 2019;234:16904-16912. DOI: 10.1002/jcp.28381
  7. 7. Sid-Otmane C, Perrault LP, Ly HQ. Mesenchymal stem cell mediates cardiac repair through autocrine, paracrine and endocrine axes. Journal of Translation Medecine. 2020;18:336. DOI: 10.1186/s12967-020-02504-8
  8. 8. Sun SJ, Lai WH, Jiang Y, Zhen Z, Wei R, Lian Q , et al. Immunomodulation by systemic administration of human-induced pluripotent stem cell-derived mesenchymal stromal cells to enhance the therapeutic efficacy of cell-based therapy for treatment of myocardial infarction. Theranostics. 2021;11:1641-1654. DOI: 10.7150/thno.46119
  9. 9. 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. DOI: 10.1126/science.284.5411.143
  10. 10. Caplan AI. Mesenchymal stem cells. Journal of Orthopaetic Research. 1991;9:641-650. DOI: 10.1002/jor.1100090504
  11. 11. Lian QZ, Lye E, Yeo KS, Tan EKW, Salto-Tellez M, Liu TM, et al. Derivation of clinically compliant mscs from CD105+, CD24- differentiated human escs. Stem Cells. 2007;25:425-436. DOI: 10.1634/stemcells.2006-0420
  12. 12. Lian QZ, Zhang YL, Zhang J, Zhang HK, Wu X, Zhang Y, et al. Functional mesenchymal stem cells derived from human induced pluripotent stem cells attenuate limb ischemia in mice. Circulation. 2010;121:1113-1123. DOI: 10.1161/circulationaha.109.898312
  13. 13. Chen Z, Chen L, Zeng C, Wang WE. Functionally improved mesenchymal stem cells to better treat myocardial infarction. Stem Cells International. 2018;2018:7045245. DOI: 10.1155/2018/7045245
  14. 14. Liang P, Ye F, Hou CC, Pi L, Chen F. Mesenchymal stem cell therapy for patients with ischemic heart failure- past, present, and future. Current Stem Cell Research & Therapy. 2021;16:608-621. DOI: 10.2174/1574888x15666200309144906
  15. 15. Yong KW, Choi JR, Mohammadi M, Mitha AP, Sanati-Nezhad A, Sen A. Mesenchymal stem cell therapy for ischemic tissues. Stem Cells International. 2018;2018:8179075. DOI: 10.1155/2018/8179075
  16. 16. Yan W, Abu-El-Rub E, Saravanan S, Kirshenbaum LA, Arora RC, Dhingra S. Inflammation in myocardial injury: Mesenchymal stem cells as potential immunomodulators. American Journal of Physiology Heart and Circulatory Physiology. 2019;317:H213-H225. DOI: 10.1152/ajpheart.00065.2019
  17. 17. Zhu F, Chen YH, Li JJ, Yang ZY, Lin Y, Jiang BX, et al. Human umbilical cord mesenchymal stem cell-derived exosomes attenuate myocardial infarction injury via mir-24-3p-promoted m2 macrophage polarization. Advanced Biology (Weinh). 2022;6:e2200074. DOI: 10.1002/adbi.202200074
  18. 18. Suzuki E, Fujita D, Takahashi M, Oba S, Nishimatsu H. Therapeutic effects of mesenchymal stem cell-derived exosomes in cardiovascular disease. Advances in Experimental Medicine and Biology. 2017;998:179-185. DOI: 10.1007/978-981-10-4397-0_12
  19. 19. Ferreira JR, Teixeira GQ , Santos SG, Barbosa MA, Almeida-Porada G, Gonçalves RM. Mesenchymal stromal cell secretome: Influencing therapeutic potential by cellular pre-conditioning. Frontiers in Immunology. 2018;9:2837. DOI: 10.3389/fimmu.2018.02837
  20. 20. Chen LJ, Qu JJ, Mei QH, Chen X, Fang YX, Chen L, et al. Small extracellular vesicles from menstrual blood-derived mesenchymal stem cells (menscs) as a novel therapeutic impetus in regenerative medicine. Stem Cell Research & Therapy. 2021;12:433. DOI: 10.1186/s13287-021-02511-6
  21. 21. Meng H, Cheng WT, Wang L, Chen SQ , Teng Y, Lu ZW, et al. Mesenchymal stem cell exosomes in the treatment of myocardial infarction: A systematic review of preclinical in vivo studies. Journal of Cardiovascular Translational Research. 2022;15:317-339. DOI: 10.1007/s12265-021-10168-y
  22. 22. Pu L, Kong X, Li H, He X. Exosomes released from mesenchymal stem cells overexpressing microrna-30e ameliorate heart failure in rats with myocardial infarction. American Journal of Translational Research. 2021;13:4007-4025
  23. 23. Ma T, Chen Y, Chen Y, Meng Q , Sun J, Shao L, et al. Microrna-132, delivered by mesenchymal stem cell-derived exosomes, promote angiogenesis in myocardial infarction. Stem Cells International. 2018;2018:3290372. DOI: 10.1155/2018/3290372
  24. 24. Lazar E, Benedek T, Korodi S, Rat N, Lo J, Benedek I. Stem cell-derived exosomes-an emerging tool for myocardial regeneration. World Journal of Stem Cells. 2018;10:106-115. DOI: 10.4252/wjsc.v10.i8.106
  25. 25. 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. DOI: 10.1155/2017/4150705
  26. 26. ISEV 2018 Collaborators. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. Journal of Extracellular Vesicles. 2018;(7):1535750. DOI: 10.1080/20013078.2018.1535750
  27. 27. Kalluri R, Lebleu VS. The biology, function, and biomedical applications of exosomes. Science. 2020;367:eaau6977. DOI: 10.1126/science.aau6977
  28. 28. Abbaszadeh H, Ghorbani F, Derakhshani M, Movassaghpour A, Yousefi M. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles: A novel therapeutic paradigm. Journal of Cellular Physiology. 2020;235:706-717. DOI: 10.1002/jcp.29004
  29. 29. Chen S, Sun F, Qian H, Xu W, Jiang J. Preconditioning and engineering strategies for improving the efficacy of mesenchymal stem cell-derived exosomes in cell-free therapy. Stem Cells International. 2022;2022:1779346. DOI: 10.1155/2022/1779346
  30. 30. Cheng L, Zhang K, Wu S, Cui M, Xu T. Focus on mesenchymal stem cell-derived exosomes: Opportunities and challenges in cell-free therapy. Stem Cells International. 2017;2017:6305295. DOI: 10.1155/2017/6305295
  31. 31. 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;18:2574-2582. DOI: 10.3892/etm.2019.7874
  32. 32. 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. DOI: 10.1038/s41598-018-19581-x
  33. 33. Zhang P, Jiang J, Zhou X, Kolay J, Wang R, Wan Z, et al. Label-free imaging and biomarker analysis of exosomes with plasmonic scattering microscopy. Chemical Science. 2022;13:12760-12768. DOI: 10.1039/d2sc05191e
  34. 34. Yakubovicha EI, Polischouka AG, Evtushenkoa VI. Principles and problems of exosome isolation from biological fluids. Biochemistry (Moscow), Supplement Series A: Membrane and Cell Biology. 2022;16:115-126. DOI: 10.1134/S1990747822030096
  35. 35. Skalnikova HK, Bohuslavova B, Turnovcova K, Juhasova J, Juhas S, Rodinova M, et al. Isolation and characterization of small extracellular vesicles from porcine blood plasma, cerebrospinal fluid, and seminal plasma. Proteomes. 2019;7:17. DOI: 10.3390/proteomes7020017
  36. 36. Chen J, Li P, Zhang T, Xu Z, Huang X, Wang R, et al. Review on strategies and technologies for exosome isolation and purification. Frontiers in Bioengineering and Biotechnology. 2021;9:811971. DOI: 10.3389/fbioe.2021.811971
  37. 37. Klymiuk MC, Balz N, Elashry MI, Heimann M, Wenisch S, Arnhold S. Exosomes isolation and identification from equine mesenchymal stem cells. BMC Veterinary Research. 2019;15:42. DOI: 10.1186/s12917-019-1789-9
  38. 38. Ramasubramanian L, Jyothi H, Goldbloom-Helzner L, Light BM, Kumar P, Carney RP, et al. Development and characterization of bioinspired lipid raft nanovesicles for therapeutic applications. ACS Applied Materials Interfaces. 2022;14:54458-54477. DOI: 10.1021/acsami.2c13868
  39. 39. Zhang Y, Bi J, Huang J, Tang Y, Du S, Li P. Exosome: A review of its classification, isolation techniques, storage, diagnostic and targeted therapy applications. International Journal of Nanomedicine. 2020;15:6917-6934. DOI: 10.2147/IJN.S264498
  40. 40. Sridharan B, Lim HG. Exosomes and ultrasound: The future of theranostic applications. Materials Today Bio. 2023;19:100556. DOI: 10.1016/j.mtbio.2023.100556
  41. 41. Busatto S, Vilanilam G, Ticer T, Lin WL, Dickson DW, Shapiro S, et al. Tangential flow filtration for highly efficient concentration of extracellular vesicles from large volumes of fluid. Cell. 2018;7:273. DOI: 10.3390/cells7120273
  42. 42. Brown PN, Yin H. Polymer-based purification of extracellular vesicles. Methods in Molecular Biology. 2017;1660:91-103. DOI: 10.1007/978-1-4939-7253-1_8
  43. 43. Sidhom K, Obi PO, Saleem A. A review of exosomal isolation methods: Is size exclusion chromatography the best option? International Journal of Molecular Science. 2020;21:6466. DOI: 10.3390/ijms21186466
  44. 44. Gholizadeh S, Shehata Draz M, Zarghooni M, Sanati-Nezhad A, Ghavami S, Shafiee H, et al. Microfluidic approaches for isolation, detection, and characterization of extracellular vesicles: Current status and future directions. Biosensors & Bioelectronics. 2017;91:588-605. DOI: 10.1016/j.bios.2016.12.062
  45. 45. Salafi T, Zhang Y, Zhang Y. A review on deterministic lateral displacement for particle separation and detection. Nanomicro Letters. 2019;11:77. DOI: 10.1007/s40820-019-0308-7
  46. 46. Lee K, Shao H, Weissleder R, Lee H. Acoustic purification of extracellular microvesicles. ACS Nano. 2015;9:2321-2327. DOI: 10.1021/nn506538f
  47. 47. Song J, Song B, Yuan L, Yang G. Multiplexed strategies toward clinical translation of extracellular vesicles. Theranostics. 2022;12:6740-6761. DOI: 10.7150/thno.75899
  48. 48. Tenchov R, Sasso JM, Wang XM, Liaw WS, Chen CA, Zhou QA. Exosomes-nature’s lipid nanoparticles, a rising star in drug delivery and diagnostics. ACS Nano. 2022;16:17802-17846. DOI: 10.1021/acsnano.2c08774
  49. 49. Ferguson SW, Nguyen J. Exosomes as therapeutics: The implications of molecular composition and exosomal heterogeneity. Journal of Controlled Release. 2016;228:179-190. DOI: 10.1016/j.jconrel.2016.02.037
  50. 50. Takeuchi R, Katagiri W, Endo S, Kobayashi T. Exosomes from conditioned media of bone marrow-derived mesenchymal stem cells promote bone regeneration by enhancing angiogenesis. PLoS One. 2019;14:e0225472. DOI: 10.1371/journal.pone.0225472
  51. 51. Saheera S, Jani VP, Witwer KW, Kutty S. Extracellular vesicle interplay in cardiovascular pathophysiology. American Journal of Physiology-Heart and Circulatory Physiology. 2021;320:H1749-H1H61. DOI: 10.1152/ajpheart.00925.2020
  52. 52. Saheera S, Potnuri AG, Krishnamurthy P. Nano-vesicle (mis)communication in senescence-related pathologies. Cell. 2020;9:1974. DOI: 10.3390/cells9091974
  53. 53. Patil M, Saheera S, Dubey PK, Kahn-Krell A, Kumar GP, Singh S, et al. Novel mechanisms of exosome-mediated phagocytosis of dead cells in injured heart. Circulation Research. 2021;129:1006-1020. DOI: 10.1161/CIRCRESAHA.120.317900
  54. 54. Lee BC, Kang I, Yu KR. Therapeutic features and updated clinical trials of mesenchymal stem cell (msc)-derived exosomes. Journal of Clinical Medicine. 2021;10:711. DOI: 10.3390/jcm10040711
  55. 55. Giebel B, Kordelas L, Borger V. Clinical potential of mesenchymal stem/stromal cell-derived extracellular vesicles. Stem Cell Investigation. 2017;4:84. DOI: 10.21037/sci.2017.09.06
  56. 56. Wang X, Tang Y, Liu Z, Yin Y, Li Q , Liu G, et al. The application potential and advance of mesenchymal stem cell-derived exosomes in myocardial infarction. Stem Cells International. 2021;2021:5579904. DOI: 10.1155/2021/5579904
  57. 57. Lu TX, Rothenberg ME. Microrna. The Journal of Allergy Clinical Immunology. 2018;141:1202-1207. DOI: 10.1016/j.jaci.2017.08.034
  58. 58. Cai Y, Yu X, Hu S, Yu J. A brief review on the mechanisms of mirna regulation. Genomics, Proteomics & Bioinformatics. 2009;7:147-154. DOI: 10.1016/S1672-0229(08)60044-3
  59. 59. Fabian MR, Sonenberg N. The mechanics of mirna-mediated gene silencing: A look under the hood of mirisc. Nature Structural & Molecular Biology. 2012;19:586-593. DOI: 10.1038/nsmb.2296
  60. 60. Jahangiri B, Khalaj-Kondori M, Asadollahi E, Purrafee Dizaj L, Sadeghizadeh M. Msc-derived exosomes suppress colorectal cancer cell proliferation and metastasis via mir-100/mtor/mir-143 pathway. International Journal of Pharmaceutics. 2022;627:122214. DOI: 10.1016/j.ijpharm.2022.122214
  61. 61. Cheng C, Chen X, Wang Y, Cheng W, Zuo X, Tang W, et al. MSCs-derived exosomes attenuate ischemia-reperfusion brain injury and inhibit microglia apoptosis might via exosomal mir-26a-5p mediated suppression of cdk6. Molecular Medicine. 2021;27:67. DOI: 10.1186/s10020-021-00324-0
  62. 62. Xu J, Chen P, Yu C, Shi Q , Wei S, Li Y, et al. Hypoxic bone marrow mesenchymal stromal cells-derived exosomal mir-182-5p promotes liver regeneration via foxo1-mediated macrophage polarization. FASEB Journal. 2022;36:e22553. DOI: 10.1096/fj.202101868RRR
  63. 63. Zhu LP, Tian T, Wang JY, He JN, 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. DOI: 10.7150/thno.28021
  64. 64. 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. DOI: 10.1161/JAHA.115.002856
  65. 65. Tan SJO, Floriano JF, Nicastro L, Emanueli C, Catapano F. Novel applications of mesenchymal stem cell-derived exosomes for myocardial infarction therapeutics. Biomolecules. 2020;10:707. DOI: 10.3390/biom10050707
  66. 66. Chen B, Luo L, Wei X, Gong D, Li Z, Li S, et al. M1 bone marrow-derived macrophage-derived extracellular vesicles inhibit angiogenesis and myocardial regeneration following myocardial infarction via the malat1/microrna-25-3p/cdc42 axis. Oxidative Medicine and Cellular Longevity. 2021;2021:9959746. DOI: 10.1155/2021/9959746
  67. 67. Zheng Y, Lin J, Liu D, Wan G, Gu X, Ma J. Nogo-b promotes angiogenesis and improves cardiac repair after myocardial infarction via activating notch1 signaling. Cell Death & Disease. 2022;13:306. DOI: 10.1038/s41419-022-04754-4
  68. 68. Zhou XL, Zhu RR, Liu S, Xu H, Xu X, Wu QC, et al. Notch signaling promotes angiogenesis and improves cardiac function after myocardial infarction. Journal of Cellular Biochemistry. 2018;119:7105-7112. DOI: 10.1002/jcb.27032
  69. 69. Liu HB, Zhang YM, Yuan J, Gao J, Zhong X, Yao K, et al. Bdendritic cell-derived exosomal mir-494-3p promotes angiogenesis following myocardial infarction. International Journal of Molecular Medicine. 2021;47:315-325. DOI: 10.3892/ijmm.2020.4776
  70. 70. Wang N, Chen C, Yang D, Liao Q , Luo H, Wang X, et al. Mesenchymal stem cells-derived extracellular vesicles, via mir-210, improve infarcted cardiac function by promotion of angiogenesis. Biochimica et Biophysica Acta Molecular Basis of Disease. 2017;1863:2085-2092. DOI: 10.1016/j.bbadis.2017.02.023
  71. 71. Zhu W, Sun L, Zhao P, Liu Y, Zhang J, Zhang Y, et al. Macrophage migration inhibitory factor facilitates the therapeutic efficacy of mesenchymal stem cells derived exosomes in acute myocardial infarction through upregulating mir-133a-3p. Journal of Nanobiotechnology. 2021;19:61. DOI: 10.1186/s12951-021-00808-5
  72. 72. Sun L, Zhu W, Zhao P, Zhang J, Lu Y, Zhu Y, et al. Down-regulated exosomal microrna-221 - 3p derived from senescent mesenchymal stem cells impairs heart repair. Frontiers in Cell and Developmental Biology. 2020;8:263. DOI: 10.3389/fcell.2020.00263
  73. 73. Fang J, Zhang Y, Chen D, Zheng Y, Jiang J. Exosomes and exosomal cargos: A promising world for ventricular remodeling following myocardial infarction. International Journal of Nanomedicine. 2022;17:4699-4719. DOI: 10.2147/IJN.S377479
  74. 74. Anversa P, Cheng W, Liu Y, Leri A, Redaelli G, Kajstura J. Apoptosis and myocardial infarction. Basic Research in Cardiology. 1998;93:8-12
  75. 75. Cheng H, Chang S, Xu R, Chen L, Song X, Wu J, et al. Hypoxia-challenged msc-derived exosomes deliver mir-210 to attenuate post-infarction cardiac apoptosis. Stem Cell Research & Therapy. 2020;11:224. DOI: 10.1186/s13287-020-01737-0
  76. 76. Pan J, Alimujiang M, Chen Q , Shi H, Luo X. Exosomes derived from mir-146a-modified adipose-derived stem cells attenuate acute myocardial infarction-induced myocardial damage via downregulation of early growth response factor 1. Journal of Cellular Biochemistry. 2019;120:4433-4443. DOI: 10.1002/jcb.27731
  77. 77. Wang X, Zhu Y, Wu C, Liu W, He Y, Yang Q. Adipose-derived mesenchymal stem cells-derived exosomes carry microrna-671 to alleviate myocardial infarction through inactivating the tgfbr2/smad2 axis. Inflammation. 2021;44:1815-1830. DOI: 10.1007/s10753-021-01460-9
  78. 78. Ning W, Li S, Yang W, Yang B, Xin C, Ping X, et al. Blocking exosomal mirna-153-3p derived from bone marrow mesenchymal stem cells ameliorates hypoxia-induced myocardial and microvascular damage by targeting the angpt1-mediated vegf/pi3k/akt/enos pathway. Cellular Signalling. 2021;77:109812. DOI: 10.1016/j.cellsig.2020.109812
  79. 79. Wu Z, Cheng S, Wang S, Li W, Liu J. Bmscs-derived exosomal microrna-150-5p attenuates myocardial infarction in mice. International Immunopharmacology. 2021;93:107389. DOI: 10.1016/j.intimp.2021.107389
  80. 80. Luo Q , Guo D, Liu G, Chen G, Hang M, Jin M. Exosomes from mir-126-overexpressing adscs are therapeutic in relieving acute myocardial ischaemic injury. Cellular Physiology and Biochemistry. 2017;44:2105-2116. DOI: 10.1159/000485949
  81. 81. Sun XH, Wang X, Zhang Y, Hui J. Exosomes of bone-marrow stromal cells inhibit cardiomyocyte apoptosis under ischemic and hypoxic conditions via mir-486-5p targeting the pten/pi3k/akt signaling pathway. Thrombosis Research. 2019;177:23-32. DOI: 10.1016/j.thromres.2019.02.002
  82. 82. Wang X, Dai Y, Ding Z, Khaidakov M, Mercanti F, Mehta JL. Regulation of autophagy and apoptosis in response to angiotensin ii in hl-1 cardiomyocytes. Biochemical and Biophysical Research Communications. 2013;440:696-700. DOI: 10.1016/j.bbrc.2013.09.131
  83. 83. Xiao C, Wang K, Xu Y, Hu H, Zhang N, Wang Y, et al. Transplanted mesenchymal stem cells reduce autophagic flux in infarcted hearts via the exosomal transfer of mir-125b. Circulation Research. 2018;123:564-578. DOI: 10.1161/CIRCRESAHA.118.312758
  84. 84. Liu J, Jiang M, Deng S, Lu J, Huang H, Zhang Y, et al. Mir-93-5p-containing exosomes treatment attenuates acute myocardial infarction-induced myocardial damage. Molecular Therapy Nucleic Acids. 2018;11:103-115. DOI: 10.1016/j.omtn.2018.01.010
  85. 85. Li Y, Yang R, Guo B, Zhang H, Zhang H, Liu S, et al. Exosomal mir-301 derived from mesenchymal stem cells protects myocardial infarction by inhibiting myocardial autophagy. Biochemical Biophysical Research Communications. 2019;514:323-328. DOI: 10.1016/j.bbrc.2019.04.138
  86. 86. Yuan J, Yang H, Liu C, Shao L, Zhang H, Lu K, et al. Microneedle patch loaded with exosomes containing microrna-29b prevents cardiac fibrosis after myocardial infarction. Advanced Healthcare Materials. 2023:e2202959. DOI: 10.1002/adhm.202202959
  87. 87. Zheng X, Hermann DM, Bahr M, Doeppner TR. The role of small extracellular vesicles in cerebral and myocardial ischemia-molecular signals, treatment targets, and future clinical translation. Stem Cells. 2021;39:403-413. DOI: 10.1002/stem.3329
  88. 88. Mao Q , Liang XL, Zhang CL, Pang YH, Lu YX. Lncrna klf3-as1 in human mesenchymal stem cell-derived exosomes ameliorates pyroptosis of cardiomyocytes and myocardial infarction through mir-138-5p/sirt1 axis. Stem Cell Research & Therapy. 2019;10:393. DOI: 10.1186/s13287-019-1522-4
  89. 89. Sun L, Zhu W, Zhao P, Wang Q , Fan B, Zhu Y, et al. Long noncoding rna uca1 from hypoxia-conditioned hmsc-derived exosomes: A novel molecular target for cardioprotection through mir-873-5p/xiap axis. Cell Death & Disease. 2020;11:696. DOI: 10.1038/s41419-020-02783-5
  90. 90. Huang P, Wang L, Li Q , Tian X, Xu J, Xu J, et al. Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding rna h19. Cardiovascular Research. 2020;116:353-367. DOI: 10.1093/cvr/cvz139
  91. 91. Toh WS, Lai RC, Zhang B, Lim SK. MSC exosome works through a protein-based mechanism of action. Biochemical Society Transactions. 2018;46:843-853. DOI: 10.1042/BST20180079
  92. 92. 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. DOI: 10.1002/stem.2298
  93. 93. Kim HS, Choi DY, Yun SJ, Choi SM, Kang JW, Jung JW, et al. Proteomic analysis of microvesicles derived from human mesenchymal stem cells. Journal of Proteome Research. 2012;11:839-849. DOI: 10.1021/pr200682z
  94. 94. Tan SS, Chen TS, Tan KH, Lim SK. An overview of the proteomic and mirna cargo in msc-derived exosomes. Mesenchymal Stem Cell Derived Exosomes. 2015:21-36. DOI: 10.1016/B978-0-12-800164-6.00002-2
  95. 95. Collino F, Pomatto M, Bruno S, Lindoso RS, Tapparo M, Sicheng W, et al. Exosome and microvesicle-enriched fractions isolated from mesenchymal stem cells by gradient separation showed different molecular signatures and functions on renal tubular epithelial cells. Stem Cell Reviews and Reports. 2017;13:226-243. DOI: 10.1007/s12015-016-9713-1
  96. 96. 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. DOI: 10.1155/2012/971907
  97. 97. 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. DOI: 10.1089/cell.2017.0047
  98. 98. Zhao Y, Sun X, Cao W, Ma J, Sun L, Qian H, et al. Exosomes derived from human umbilical cord mesenchymal stem cells relieve acute myocardial ischemic injury. Stem Cells International. 2015;2015:761643. DOI: 10.1155/2015/761643
  99. 99. Xiao Y, Zhang Y, Li Y, Peng N, Liu Q , Qiu D, et al. Exosomes derived from mesenchymal stem cells pretreated with ischemic rat heart extracts promote angiogenesis via the delivery of dmbt1. Cell Transplantation. 2022;31:9636897221102898. DOI: 10.1177/09636897221102898
  100. 100. Deng S, Zhou X, Ge Z, Song Y, Wang H, Liu X, et al. Exosomes from adipose-derived mesenchymal stem cells ameliorate cardiac damage after myocardial infarction by activating s1p/sk1/s1pr1 signaling and promoting macrophage m2 polarization. The International Journal of Biochemestry & Cell Biology. 2019;114:105564. DOI: 10.1016/j.biocel.2019.105564
  101. 101. Ahmed L, Al-Massri K. New approaches for enhancement of the efficacy of mesenchymal stem cell-derived exosomes in cardiovascular diseases. Tissue Engineering and Regenerative Medicine. 2022;19:1129-1146. DOI: 10.1007/s13770-022-00469-x
  102. 102. Hu X, Wu R, Shehadeh LA, Zhou Q , Jiang C, Huang X, et al. Severe hypoxia exerts parallel and cell-specific regulation of gene expression and alternative splicing in human mesenchymal stem cells. BMC Genomics. 2014;15:303. DOI: 10.1186/1471-2164-15-303
  103. 103. Ahmad B, Skorska A, Wolfien M, Sadraddin H, Lemcke H, Vasudevan P, et al. The effects of hypoxic preconditioned murine mesenchymal stem cells on post-infarct arrhythmias in the mouse model. International Journal of Molecular Sciences. 2022;23:8843. DOI: 10.3390/ijms23168843
  104. 104. Song HF, Li B, Guo R, He S, Peng ZX, Qu JY, et al. Hypoxic preconditioned aged bmscs accelerates mi injury repair by modulating inflammation, oxidative stress and apoptosis. Biochemical and Biophysical Research Communication. 2022;627:45-51. DOI: 10.1016/j.bbrc.2022.08.039
  105. 105. Zhang CS, Shao K, Liu CW, Li CJ, Yu BT. Hypoxic preconditioning bmscs-exosomes inhibit cardiomyocyte apoptosis after acute myocardial infarction by upregulating microrna-24. European Review for Medical and Pharmacological Sciences. 2019;23:6691-6699. DOI: 10.26355/eurrev_201908_18560
  106. 106. Zhu JY, Lu K, Zhang N, Zhao Y, Ma QC, Shen J, et al. Myocardial reparative functions of exosomes from mesenchymal stem cells are enhanced by hypoxia treatment of the cells via transferring microrna-210 in an nsmase2-dependent way. Artificial Cells, Nanomedicine, and Biotechnology. 2018;46:1659-1670. DOI: 10.1080/21691401.2017.1388249
  107. 107. Zhang LY, Wei QX, Liu XM, Zhang T, Wang SY, Zhou LL, et al. Exosomal microrna-98-5p from hypoxic bone marrow mesenchymal stem cells inhibits myocardial ischemia-reperfusion injury by reducing tlr4 and activating the pi3k/akt signaling pathway. International Immunopharmacology. 2021;101:107592. DOI: 10.1016/j.intimp.2021.107592
  108. 108. Gonzalez-King H, García NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepúlveda P. Hypoxia inducible factor-1α potentiates jagged 1-mediated angiogenesis by mesenchymal stem cell-derived exosomes. Stem Cells. 2017;35:1747-1759. DOI: 10.1002/stem.2618
  109. 109. Sun JC, Shen H, Shao LB, Teng XM, Chen YQ , Liu X, et al. Hif-1α overexpression in mesenchymal stem cell-derived exosomes mediates cardioprotection in myocardial infarction by enhanced angiogenesis. Stem Cell Research and Therapy. 2020;11:373. DOI: 10.1186/s13287-020-01881-7
  110. 110. Kim EH, Kim DH, Kim HR, Kim SY, Kim HH, Bang OY. Stroke serum priming modulates characteristics of mesenchymal stromal cells by controlling the expression mirna-20a. Cell Transplantation. 2016;25:1489-1499. DOI: 10.3727/096368916x690430
  111. 111. Phan J, Kumar P, Hao D, Gao K, Farmer D, Wang AJ. Engineering mesenchymal stem cells to improve their exosome efficacy and yield for cell-free therapy. Journal of Extracellular Vesicles. 2018;7:1522236. DOI: 10.1080/20013078.2018.1522236
  112. 112. Chen GN, Liang XT, Han Q , Mai C, Shi LL, Shao Z, et al. Apelin-13 pretreatment promotes the cardioprotective effect of mesenchymal stem cells against myocardial infarction by improving their survival. Stem Cells International. 2022;2022:3742678. DOI: 10.1155/2022/3742678
  113. 113. Deng R, Liu YM, He HW, Zhang H, Zhao CL, Cui Z, et al. Haemin pre-treatment augments the cardiac protection of mesenchymal stem cells by inhibiting mitochondrial fission and improving survival. Journal of Cellular and Molecular Medicine. 2020;24:431-440. DOI: 10.1111/jcmm.14747
  114. 114. Xiong YY, Tang RJ, Xu JY, Jiang WY, Gong ZT, Zhang LL, et al. Tongxinluo-pretreated mesenchymal stem cells facilitate cardiac repair via exosomal transfer of mir-146a-5p targeting irak1/nf-κb p65 pathway. Stem Cell Research and Therapy. 2022;13:289. DOI: 10.1186/s13287-022-02969-y
  115. 115. Pittenger MF, Eghtesad S, Sanchez PG, Liu XY, Wu ZJ, Chen L, et al. Msc pretreatment for improved transplantation viability results in improved ventricular function in infarcted hearts. International Journal of Molecular Science. 2022;23:694. DOI: 10.3390/ijms23020694
  116. 116. Zheng HF, Liang XT, Han Q , Shao Z, Zhang YX, Shi LL, 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. DOI: 10.1186/s12951-021-01077-y
  117. 117. Zhang J, Lu Y, Mao YM, Yu Y, Wu TY, Zhao W, et al. Ifn-γ enhances the efficacy of mesenchymal stromal cell-derived exosomes via mir-21 in myocardial infarction rats. Stem Cell Research and Therapy. 2022;13:333. DOI: 10.1186/s13287-022-02984-z
  118. 118. Liu XL, Li X, Zhu WW, Zhang YL, Hong YM, Liang XT, et al. Exosomes from mesenchymal stem cells overexpressing mif enhance myocardial repair. Journal of Cellular Physiology. 2020;235:8010-8022. DOI: 10.1002/jcp.29456
  119. 119. Gong XH, Liu H, Wang SJ, Liang SW, Wang GG. Exosomes derived from sdf1-overexpressing mesenchymal stem cells inhibit ischemic myocardial cell apoptosis and promote cardiac endothelial microvascular regeneration in mice with myocardial infarction. Journal of Cellular Physiology. 2019;234:13878-13893. DOI: 10.1002/jcp.28070
  120. 120. Kang K, Ma R, Cai W, Huang W, Paul C, Liang J, et al. Exosomes secreted from cxcr4 overexpressing mesenchymal stem cells promote cardioprotection via akt signaling pathway following myocardial infarction. Stem Cells International. 2015;2015:659890. DOI: 10.1155/2015/659890
  121. 121. Small EM, Olson EN. Pervasive roles of micrornas in cardiovascular biology. Nature. 2011;469:336-342. DOI: 10.1038/nature09783
  122. 122. Liu N, Olson EN. Microrna regulatory networks in cardiovascular development. Developmental Cell. 2010;18:510-525. DOI: 10.1016/j.devcel.2010.03.010
  123. 123. Bridges MC, Daulagala AC, Kourtidis A. Lnccation: Lncrna localization and function. Journal of Cell Biology. 2021;220:e202009045. DOI: 10.1083/jcb.202009045
  124. 124. Mao S, Zhao J, Zhang ZJ, 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. DOI: 10.1016/j.imbio.2022.152204
  125. 125. Wang S, Dong J, Li L, Wu R, Xu L, Ren Y, et al. Exosomes derived from mir-129-5p modified bone marrow mesenchymal stem cells represses ventricular remolding of mice with myocardial infarction. Journal of Tissue Engineering and Regenerative Medicine. 2022;16:177-187. DOI: 10.1002/term.3268

Written By

Huifeng Zheng, Yimei Hong, Bei Hu, Xin Li and Yuelin Zhang

Submitted: 24 February 2023 Reviewed: 01 March 2023 Published: 26 March 2023